1600 John F. Kennedy Blvd. Ste 1800 Philadelphia, PA 19103-2899
TEXTBOOK OF PEDIATRIC RHEUMATOLOGY Copyright © 2005, Elsevier Inc.
ISBN 1-4160-0246-4
All rights reserved. No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Permissions may be sought directly from Elsevier's Health Sciences Rights Department in Philadelphia, PA, USA: phone: (+1) 215 239 3804, fax: (+1) 215 239 3805, e-mail:
[email protected]. You may also complete your request on-line via the Elsevier homepage (http://www.elsevier.com), by selecting ‘Customer Support' and then ‘Obtaining Permissions’.
Notice Knowledge and best practice in this field are constantly changing. As new research and experience broaden our knowledge, changes in practice, treatment and drug therapy may become necessary or appropriate. Readers are advised to check the most current information provided (i) on procedures featured or (ii) by the manufacturer of each product to be administered, to verify the recommended dose or formula, the method and duration of administration, and contraindications. It is the responsibility of the practitioner, relying on their own experience and knowledge of the patient, to make diagnoses, to determine dosages and the best treatment for each individual patient, and to take all appropriate safety precautions. To the fullest extent of the law, neither the Publisher nor the Editors assume any liability for any injury and/or damage to persons or property arising out or related to any use of the material contained in this book.
Library of Congress Cataloging-in-Publication Data Textbook of pediatric rheumatology / [edited by] James T. Cassidy, Ross E. Petty–5th ed / associate editors, Ronald M. Laxer, Carol B. Lindsley p.; cm. Includes bibliographical references and index ISBN 1-4160-0246-4 1. Pediatric rheumatology 2. Rheumatism in children. I. Cassidy, James T. II Petty, Ross E. [DNLM: 1. Rheumatic Disease–Child 2. Arthritis–Child 3. Connective Tissue Diseases–Child 4. Vasculitis–Child. WE 544 T355 2006] RJ482.R48.C37 2006 618.922723-dc22
Publishing Director: Kim Murphy Developmental Editor: Janine Kusza Project Manager: David Saltzberg Marketing Manager: Megan Carr
Printed in the United States Last digit is the print number: 9
8
7
6
5
4
3
2
1
To Nan, Beryl, Edda and Bart
\
CONTRIBUTING AUTHORS
'\,
KhaIed Alsaeld, MD Chief of Pediatric Rheumatology, Department of Pediatrics, Kuwait University, Faculty Medicine, Safat, Kuwait
Frank Dressler, MD Lecturer, Department of Pediatrics, Hannover Medical School, Assistentzarzt, Kinderklinik der Medizinischen, Hochschule Hannover, Hannover, Germany
Balu H. Athreya, MD Division of Rheumatology, Alfred 1. duPont Hospital for Children, Wilmington, Department of Pediatrics, Wilmington, Delaware, USA
Oaran M. Duffy, MB, BCH, M5c Associate Professor of Paediatrics and Director, Division of Paediatric Rheumatology, Montreal Children's Hospital, and McGill University, Montreal, Quebec, Canada
Ella M. Ayoub, MD (deceased) Distinguished Service Professor, Department of Pediatrics, University of Florida, College of Medicine, Gainesville, Florida, USA
Fernanda Faldnl, MD Department of Pediatrics, University of Florence and Oespedale Meyer, Florence, Italy
Susannah Brydges, PHD Fellow, Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
Ruben Burgos-Vargas, MD Professor of Medicine, Universidad Nacional, Auton6ma de Mexico, Mexico DF James T. Cassidy, MD Professor Emeritus of Child Health and Internal Medicine, University of Missouri School of Medicine, Columbia, Missouri, and Professor Emeritus of Pediatrics and Communicable Diseases and Internal Medicine, University of Michigan, Medical School, Ann Arbor, Michigan, USA Rolando Omaz, MD Dirigente Medico, Department of Pediatrics, Fondazione Policlinico Mangiagalli, University of Milan, Milan, Italy Robert A. Colbert, MD, PHD Associate Professor of Pediatrics, Associate Director, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
Marco Gattorno, MD Department of Pediatrics, University of Genoa, Genoa, Italy Edward H. Giannini, MSc DR PH Professor of Pediatrics, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA David N. Glass, MD Professor of Pediatrics, Director, Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA Judith G. Hall, OC, MD Professor Emeritus, Medical Genetics and Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada Robert M. Hamilton, MD Professor, Department of Paediatrics, University of Toronto; and Section Head, Electrophysiology, Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, Canada Hans-Iko Huppertz, MD Professor of Pediatrics, Head, and Director, Children's Hospital CProfessor-Hess-Kinderklinik), Bremen, Germany
xl
xII
CONTRIBUTING AUTHORS
Daniel L. Kastner, MD, PHD Chief, Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA Wletse Kuls, MD, PhD Professor of Pediatrics, University of Utrecht and Department of Pediatrics, University Medical Center, Utrecht, The Netherlands Ronald M. Laxer, MD, FRCPC Professor of Paediatrics and Medicine, University of Toronto; and Vice President, Clinical and Academic Affairs and Staff Rheumatologist, The Hospital for Sick Children, Toronto, Ontario, Canada Carol B. Lindsley, MD Professor, Department of Pediatrics, Chief, Pediatric Rheumatology, University of Kansas Medical Center, Kansas City, Kansas, USA Daniel J. Lovell, MD, MPH Professor of Pediatrics, University of Cincinnati College of Medicine; Associate Director, William G. Rowe Division of Rheumatology, Children's Hospital Medical Center, Cincinnati, Ohio, USA Peter N. Malleson, MBBS Professor of Pediatrics, Division of Pediatric Rheumatology, University of British Columbia, British Columbia Children's Hospital, Vancouver, British Columbia, Canada Alberto Martini, MD Professor and Head, Department of Pediatrics, Gaslini Children's Hospital, University of Genoa, Genoa, Italy Audrey M. Nelson, MD Emeritus Professor of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
Anne-Marie Prieur, MD Hopital Necker Enfants Malades, Paris, France L1eke A.M. Sanders, MD, PhD Associate Professor of Pediatric Immunology, University of Utrecht and Department of Pediatrics, University Medical Center, Utrecht, The Netherlands David D. Sherry, MD Professor of Pediatrics, University of Pennsylvania, Director, Clinical Rheumatology, Attending, Pain Management, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA Earl D. Silverman, MD, FRCPC Professor of Pediatrics and Immunology, University of Toronto, Division of Rheumatology, The Hospital for Sick Children, Toronto, Ontario, Canada Taunton R. Southwood, MD, BM, BS Professor of Paediatric Rheumatology, Department of Rheumatology, University of Birmingham Medical School, Birmingham Children's Hospital, Birmingham, United Kingdom Dawn Spence, RN, MSN, PNP Division of Rheumatology, The Hospital for Sick Children, Toronto, Ontario, Canada Robert P. Sundel, MD Assistant Professor of Pediatrics, Harvard Medical School, Director of Rheumatology, Children's Hospital Boston, Boston, Massachusetts, USA Janltzla V6zquez-Mellado, MD, PhD Professor of Medicine, Universidad Nacional Aut6noma de Mexico; Senior Investigator, Rheumatology Service, Hospital General de Mexico, Mexico City, Mexico
Benedlcte Neven Unite d'Immunologie-Hematologie et Rhumatologie, Hopital Necker Enfants Malades, Paris, France
Nlco M. Wulffraat, MD, PhD Associate Professor in Pediatric Immunology, University of Utrecht and Department of Pediatrics, University Medical Center, Utrecht, The Netherlands
Seza Ozen, MD Professor, Department of Pediatrics, Hacettepe University 02, Faculty of Medicine, Ankara, Turkey
Francesco Zullan, MD Professor, Chief, Division of Pediatric Rheumatology, Universita di Padova, Padua, Italy
Ross E. Petty, MD, PHD Professor of Pediatrics, University of British Columbia, Division of Rheumatology, Department of Pediatrics, Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
FOREWORD
In the beginning, we were a handful of naive but eager and explorative young physicians of disparate training, background, and temperament. We joined together with a vision of doing whatever was necessary to find better ways to study the rheumatic diseases of childhood and adolescence, and to treat our patients more effectively in a comprehensive continuum of care. Our successes resulted from consistent and collegial cooperation in pursuit of these common goals. With the first pediatric rheumatology centers in the u.s. in Ann Arbor, Boston, Dallas, Houston, Los Angeles, New York City, and Seattle, expansion had occurred to 27 by 1976 at the time of the First Park City Meeting, to 77 today, with a similar development of centers of excellence in clinical care and research in pediatric rheumatology around the globe on every continent. Over the years, the acquisition of spe-
cific knowledge in this field has developed at an astounding, accelerated pace. To a major extent the scientific and clinical progress of these decades and its publication has been organized and codified in this Textbook ofPediatric Rheumatology, beginning with the first edition in 1982. It is therefore our distinct privilege to join together to dedicate this fifth edition not only to that early small group of clinics and to their expansion in numbers and effectiveness, but even more to the children with rheumatic diseases, to their families, and to the now thousands of healthcare professionals endeavoring to ameliorate or cure these diseases and to improve the lifestyles and career opportunities of their patients. EARL
J.
BREWER,
JOSEPH
E.
MD, FAAP, MACR MD, MACR
LEVINSON,
xIII
PREFACE
The first edition of the Textbook of Pediatric Rheumatology was published in 1982. The intervening years have encompassed an enormous increase in our understanding of the pathophysiologic mechanisms operative in the genesis of pediatric rheumatic diseases, much of which is specific for the discipline. It was not so many years ago that the rheumatologic literature reflected data primarily from studies of adults. These recent and gratifying developments in scientific studies specific to the younger age groups have afforded us opportunities for better diagnosis and treatment of children with rheumatic disorders. Yet much remains to be accomplished. The present edition continues the tradition of incremental reassessment of authorship established by its predecessors, and represents a complete revision of the original text. We trust it will be a testament to the clinical scope and science of pediatric rheumatology, its codification as a distinct specialty, and the gratifying maturity that it has achieved. It is our belief that the advances in the practice and science of pediatric rheumatology described herein are increasingly recognized as an integral and essential element of the clinical, investigative, and educational programs of academic institutions of medical training and research. This edition was reorganized in order to provide a comprehensive source of information regarding the rheumatic diseases of childhood during the last five years for specialists concerned with the care of these children, and for those in primary fields of medicine in order to facilitate early diagnosis and treatment through timely referral. Significant advances in practice and knowledge have been incorporated including exhaustive reviews of the major clinical syndromes resulting in the rheumatic and inflammatory diseases of childhood and investigative efforts directed toward validation of their classification. The chapters on health care assessment, the conduct of clinical trials, and the design and statistical analysis of therapeutic investigations have been carefully revised. Most importantly, the text continues to reflect the ongoing therapeutic revolution in rheumatology involving biologic modification of the cytokine network which reqUired a reworking and considerable enlargement of the chapter on pharmacology. Chapters on the periodic
fever syndromes and the genetic profiles of related neonatal inflammatory disorders have been added. In the process of documenting these innumerable additions to our knowledge, the cited references have been extensively updated, retaining only those regarded as "classics" or having historic importance, and incorporating recent publications that specifically contribute to understanding the pathophysiology and clinical presentation of the over 100 rheumatic "diseases" that afflict children. The majority of these publications are now specific to children, in contrast to the first edition where much had to be assumed from adult studies. In fact, publications in this field are now so extensive that one can only suggest the depth of the investigations. To give due credit to all would require substantial additional text. It should be noted that much of the material summarized in previous editions has been deleted by acknowledging duplicative studies and tenets now universally accepted. Reviews where appropriate have been emphasized. Thirty-four of our colleagues have been enlisted in these efforts in what is now an international, multiauthored text. The contributions of these authors represent expert appraisals of specific fields of clinical concentration. We are incredibly grateful for their enthusiastic cooperation in this endeavor. Their efforts have clarified areas of immunogenetics that emphasize the extensive advancement in knowledge that will ultimately result from sequencing the human genome, immunologic mechanisms of inflammatory disease, neuroendocrine dysregulation, and developmental defects contributing to the inflammatory arthropathies. Much revision has also resulted from careful and exhaustive reviews of previous editions by John Bohnsack, Hermine Brunner, Edward Giannini, Carol Lindsley, David Sherry, and Carol Wallace. In addition to our colleagues, we are deeply indebted to our families and spousal support throughout this process of revision spanning more than three years, without whose patience and understanding this new edition would not have been possible. A transition in editorial direction, with the appointment of two associate editors who have been contributors to this and previous editions, has been of enormous aid in the preparation of this
xv
xvi
PREFACE
text, and in assuring the continuation of the contributions that it has brought to the specialty. We have also given our last farewells to two former contributors and dear friends who have now left us: Carol G. Ragsdale and Elia M. Ayoub. We trust that the fifth edition of the Textbook ofPediatric Rheumatology will aid physicians caring for children with rheumatic diseases to interpret the complex web of symptoms, signs, and laboratory abnormalities that are characteristic of these disorders, and their often inherent ambiguity; will inspire students of medicine to recognize
the challenges and excitement of this pediatric discipline; and ultimately will ensure the provision of prompt and optimal care to the hundreds of thousands of children and their families around the world who endure the pain and limitations imposed by these disorders. T. CASSIDY, MD PEITY, MD, PHD
JAMES
Ross E.
RONALD CAROL
B.
M. LAxER, MD LINDSLEY, MD
C HAP T E R
1
INTRODUCTION TO THE STUDY OF RHEUMATIC DISEASES IN CHILDREN Ja.s T. Cassidy and Ross E. Petty
~
The rheumatic diseases are chronic multisystem disorders that represent clinical manifestations of acute and chronic inflammation of the connective tissues of the musculoskeletal system, blood vessels, and skin. They range from arthritis limited to one joint, to widespread inflammation of joints, muscles, skin, blood vessels, and organs as diverse as the eye, lung, brain, and bone marrow. Homo sapiens has been afflicted with a variety of these disorders for thousands of years. Chronic arthritis-like changes have been identified in the spines of prehistoric Egyptian mummies circa 8000 BC,I and ankylosing spondylitis may be the most ancient of the defined rheumatic diseases, having been documented in skeletal remains from medieval Europe. 2 However, rheumatoid arthritis may be a more recent development, at least in Europe, where it was first described clinically in the 19th century, although it may have originated much earlier among aboriginals of North America. 3 Rheumatic diseases are worldwide in distribution, although there are notable differences in the frequency of some diseases in different racial groups. Considering the early age at onset of many rheumatic diseases of childhood, as well as their chronicity, they constitute an important burden for society.
EVOLUTION OF TERMINOLOGY In 1883, Barlow introduced a discussion on "Rheumatism and its allies in childhood" as follows: "The fundamental difficulty in discussing rheumatism consists in defining what we mean by it.,,4 The term rheumatism is derived from the Greek rheumatismos: a flux. The first use of this term has been ascribed to Galen, 5 to describe inflammatory or degenerative diseases of the joints, bones, muscles, or bursae. Baillou 0558-1616) first employed the word rheumatism to distinguish acute arthritis from gout. 6 The noun "rheumatism" currently has no precise meaning and has generally fallen into disuse. The adjective rheumatic originally referred to acute rheumatic fever and today has the connotation of "inflammatory."
2
Rheumatic fever per se was distinguished from acute gouty arthritis only in the 17th century by Sydenham in England7 ; he later described chorea. The word arthritis is derived from the Greek arthron, meaning joint. It entered the English language about 1544, when it was used to refer only to gout with the suffix -itis, meaning inflammation (-ites, originally an adjectival inflection, gradually evolved to -itis as a suffix to imply an inflammatory disorder). The term arthralgia indicates joint pain without objective evidence of inflammation. Garrod first proposed the designation rheumatoid arthritis to differentiate this disorder from gout and rheumatism. 8 However, it is likely that Hippocrates recognized these diseases in his innumerable observations on the origin of disease from natural phenomena, separate from sacred or philosophical causation or superstition. 9 In the absence of specific knowledge of etiology or pathogenesis, it is not surprising that the terminology used to describe and classify rheumatic diseases, including those of childhood, continues to evolve. Discrepancies between the American College of Rheumatology criteria for classification of juvenile rheumatoid arthritis (JRA)1O and those of the European League Against Rheumatism for juvenile chronic arthritis (JCA)ll exemplify this point. The problem of definition and classification has been a persistent one and is addressed again in the proposed criteria for juvenile idiopathic arthritis (JIA) of the International League of Associations for Rheumatology (see Chapter 9).12 Terminology for the other rheumatic diseases affecting children has been less controversial; however, validated classification criteria are often lacking. In large part, terms used to describe similar diseases in adults have been adopted without consideration for age-related differences in disorders such as systemic lupus erythematosus (SLE), scleroderma, dermatomyositis, and some of the vasculitides. Proposed criteria for the first three of these are now being validated by international studies. Diagnostic or classification criteria have been developed for "new" diseases such as Kawasaki disease, Lyme disease, and parvovirus Bl9-associated arthritis. Recent
C HAP T E R
I
INTRODUCTION TO THE STUDY OF RHEUMATIC DISEASES IN CHILDREN
genetic discoveries, as in the periodic febrile syndromes, have led to a reclassification of these disorders. In recent decades, chronic noninflammatory pain syndromes have become major clinical components of the field of rheumatology in its broadest sense in both children and adults; criteria for these disorders in children and adolescents have been adopted or modified from those used in adults.
THE RHEUMATIC DISEASES OF CHILDHOOD Pediatric rheumatology is a clinical discipline that embraces the study of inflammatory and noninflammatory disorders of the connective tissues and joints in children. Its boundaries continue to expand, and although inflammatory disorders of joints, muscles, and connective tissues constitute the core of the discipline, the differential diagnosis of these diseases necessitates a broad knowledge of potentially confusing disorders, the majority of which are included in chapters of this textbook. A summary of the history of arthritis in children has been published by Hayem. 13 The first book on pediatrics written by an Englishman, which contains the first English-language reference to rheumatism in children, is the 1545 text by Phaire, 1be Regiment ofLife Whereunto Is Added a Treatise of the Pestilence, with 1be Boke of Chytdren. In this work, the author refers to the "stifnes or starckenes of the limmes" resulting from exposure of a child to cold,14.15 a complaint that may not represent any specific rheumatic disease, and also says, "Here to doo them good that haue most nede, that is to saye children." In 1864, Cornil described a woman in whom polyarthritis had developed when she was 12 years 01d. 16 The disease pursued a chronic relapsing course and terminated in her death from uremic coma at the age of 28 years. Necropsy documented myocardial degeneration, nephrotic syndrome, and ankylosis of some joints, with synovial proliferation and marked destruction of cartilage in others. This girl may have had amyloidosis complicating chronic polyarticular arthritis. In 1873, Bouchut described chronic rheumatism in six children,17 and in 1870 .Moncorvo l8 diagnosed childhood arthritis in one of his own patients in Brazil and in eight patients from the literature. West's 1881 Lectures on the Diseases ofInfancy and Childhood!9 noted that "chronic rheumatic arthritis in children is a rare occurrence." In 1883, Barlow, a mentor to Still at the Hospital for Sick Children, Great Ormond Street, London, chaired a discussion on rheumatism in childhood. 4 In the report of this meeting, the term rheumatism was used to describe poststreptococcal disease, including acute rheumatic fever. Barlow recognized clearly the extent and complexity of these disorders: "For there are in children many affections of joints, and of structures around joints, which do not suppurate, and yet are not rheumatic; and there is much rheumatism in children which does not affect joints." What we would today call toxic synovitis of the hip, acute pyogenic arthritis, syphilitic arthritis, hemophiliac arthropathy, Henoch-Schonlein purpura, poststreptococcal arthritis, and acute rheumatic fever,
3
including carditis, arthritis, nodules, erythema marginatum, and chorea, are all identifiable in this paper. This physician was careful to exclude rickets and scurvy because he considered that the joint itself was not primarily involved in these conditions. In 1891, Diamant-Berger published a detailed account of chronic arthritis in 38 children whom he had seen or whose cases had been documented in the literature. 2o He noted the heterogeneity of onset, its predominance in girls, and involvement of the cervical spine and temporomandibular joints as well as ocular inflammation. He also stated that the prognosis in children was generally better than for chronic arthritis in adults. Five years later, Still described 22 cases of acute and chronic arthritis in children, almost all of whom were observed at the Hospital for Sick Children. 21 This treatise, written under the mentorship of Barlow,22 documented the clinical characteristics and the differing modes of onset in these children. Still was the first English physician to confine his practice to diseases of children and the first Professor of Paediatrics at King's College Hospital Medical School, London. Unfortunately, after his classic study, he rarely returned to the field of pediatric rheumatology, although his scholarly work comprised 108 papers and five books, including the History of Paediatrics (931) and Common Diseases in Children. Also in 1896, Koplick 23 described the first American child with chronic arthritis. Although these descriptions of arthritis in childhood rank as the most important milestones in the early development of pediatric rheumatology, other rheumatic diseases began to be identified in children in the 19th century. The clinical characteristics of leukocytoclastic vasculitis were described by Schonlein24 and Henoch25 in the mid-1800s. Juvenile dermatomyositis was identified by Unvericht in 1877,26 although it was not until the 1960s that significant experience with this disease was reported. SLE has been recognized in children at least since 1904. 27 The original description of scleroderma was in a 17-year-old girl,28 but the disease was rarely diagnosed thereafter in a child until the early 1960s. Ankylosing spondylitis was also perhaps first identified in a child 29 ; it was certainly known to occur in childhood in the 19505,30 but specific studies of the disorder in children did not emerge until the late 1960s. 31.32 More recent additions to the family of pediatric rheumatic diseases include Kawasaki disease, which was reported in some detail in 1967,33 although its clinical characteristics (in infants dying of "polyarteritis nodosa") were described by Munro-Faure in 1959. 34 Other rheumatic diseases, such as neonatal-onset multisystem inflammatory disease or NOMID (also called chronic infantile neurologic, cutaneous, and articular [CINCA] syndrome), neonatal lupus, and Lyme disease, have recently been identified. Noninflammatory musculoskeletal pain syndromes are more recent additions to the expanding list of disorders that cause musculoskeletal pain and dysfunction in children and adolescents. Very little further information about these rheumatic diseases was published until the last half of the 20th century. This development coincided with the availability of penicillin and the retreat of acute rheumatic fever in
4
C HAP T E R
I
INTRODUCTION TO THE STUDY OF RHEUMATIC DISEASES IN CHILDREN
Europe and North America. Until the mid-1940s this disease was the major rheumatic disease, and it still is in many areas of the developing world. With its control, however, attention was shifted to the chronic nonstreptococcal rheumatic diseases, principally chronic arthritis. Today, the specialty of pediatric rheumatology is concerned with a diverse group of disorders (Table 1-1), most of which are manifestations of systemic disorders and require the greatest expertise for prompt diagnosis and optimal management. Of all of the specialties, rheumatology, one of the most stimulating and challenging areas in all of medicine, may deal with the broadest spectrum of disease, both organ-specific and systemic. It is sometimes considered a "gray area" of medicine because there are few useful diagnostic tests, pathognomonic clinical signs are sparse, and therapy often lacks specificity. This specialty requires a diagnostic and therapeutic approach that embraces the "whole" child and family unit, patience, careful observation over long periods, and a heightened ability to tolerate ambiguity and uncertainty. Sometimes only the passage of time makes a diagnosis evident.
EMERGENCE OF THE SPECIALTY OF PEDIATRIC RHEUMATOLOGY The roots of contemporary pediatric rheumatology are found in pediatrics, internal medicine, immunology, and orthopedics; however, as experience with rheumatic diseases in children accumulates, it is apparent that many aspects of these disorders demand a uniquely pediatric approach. Furthermore, many of the diseases, or their complications, are confined to the childhood and adolescent population but have lasting effects on health and socioeconomic well-being or cause disability well into adulthood. These considerations first emerged in the United Kingdom in particular, where the study of rheumatic disease in children was established in earnest after World War II by the founding in 1947 of the Rheumatism Research Unit at the Canadian Red Cross Memorial Hospital in Taplow, Berkshire, England. 3S .36 Initially the Taplow unit dealt almost exclusively with rheumatic fever. As the frequency of this disease declined in England during the mid-20th century, its focus shifted to
I!:.
TABlE I-I
the other chronic rheumatic diseases and laid the foundation for the specialty of pediatric rheumatology. Pediatric rheumatology began to develop in the United States in the early 1940s. Hospitals for the treatment and rehabilitation of children with rheumatic fever were established after World War II in Chicago (La Rabida) and New York City (Irvington House) that were later expanded in scope to care for children with other rheumatic diseases and chronic illnesses. Establishment of clinical and academic centers of pediatric rheumatology in the United States and subsequently in many other countries marked the "coming of age" of the specialty. The American College of Rheumatology established the Pediatric Council in 1975; its first Conference on the Rheumatic Diseases of Children was held in Park City, Utah, in 1976. The first edition of this textbook was published in 1982. Reminiscences of six of the pioneers of pediatric rheumatology (Stillman, Hanson, Levinson, Ansell, Brewer, and Stoeber) are recommended to the interested reader. 37--42
FACTORS THAT MAY MODIFY RHEUMATIC DISEASES IN CHILDREN One of the fundamental questions that has pervaded research in pediatric rheumatology is the extent to which the rheumatic diseases of childhood are the same as, or different from, rheumatic diseases in adults. It is important to differentiate semantic from biologic similarities: The fact that two diseases bear the same name or share some physical findings, abnormalities in laboratory markers, or even treatments (e.g., JRA and adult rheumatoid arthritis) does not necessarily imply that they are the same disorder biologically. Historically, the pediatric diseases were usually named in ignorance of epidemiology, genetics, or biology. Any distinction based on age at onset alone is arbitrary and unlikely to represent biologic truth. There is no age at which these rheumatic diseases abruptly change from one to the other, yet age-related associations are often evident. For example, there are characteristic age-atonset distributions for the individual chronic arthritides. 43 The spectrum of int1ammatory myositis is very different in the child or adolescent than in the adult. Morphea is a common form of scleroderma in the child, whereas in the
Frequt'll(y ollhe Major Pedialri( (onnedive Tisslle Ubeases in 11,300 Children
Disease Juvenile rheumatoid arthritis Connective tissue diseases Spondyloarthropathy and reactive arthritis Psoriatic arthropathy Infectious arthritis and osteomyelitis Malignancy/hematologic Chronic pain syndromes Hypermobility and overuse syndromes Other diseases
Number of eases 7,368 3,861 2,973 173 1.620 290 4,483 2,745 34,216
%
12.8 6.7
5.1 0.3 2.8 0.5 7.8 4.8 59.3
57,729 diagnoses from 48.934 consecutive patients with definite diagnoses entered into the Pediatric Rheumatic. Disease Registry of the Pediatric Rheumatology Database Research Group. 1992-2002. Courtesy of Suzanne Bowyer. M.D., Pediatric Rheumatology Database Research Group.
C HAP T E R
I
INTRODUCTION TO THE STUDY OF RHEUMATIC DISEASES IN CHILDREN
adult, systemic scleroderma predominates. There is little doubt that some rheumatic diseases occur only in young children (e.g., oligoarthritis with uveitis, Kawasaki disease), and others affect almost exclusively adults (e.g., gout, osteoarthritis). Neonatal lupus, and rarely other rheumatic diseases, represent the effect of maternal autoimmunity on the fetus. In diseases such as SLE and the vasculitides, clinical and laboratory characteristics in children and adults are very similar, if not identical in many cases. However, the reasons that one individual has childhood onset of disease, whereas another has onset in adulthood, are not known. More commonly, however, diseases with similar names and similar biologic characteristics occur in both pediatric and adult populations (e.g., scleroderma, ankylosing spondylitis, SLE). Age-related manifestations of these diseases are well recognized. Some of the prominent differences between children and adults that may modify the clinical expression of a rheumatic disease are presented in Table 1-2. Most, if not all, rheumatic diseases are characterized by disordered immunity, and often there is a genetic predisposition that is expressed clinically early in life. This is particularly true for the inherited dysplasias of bone and cartilage and for biochemical disorders such as mucopolysaccharidoses, hemophilia, and the periodic febrHe syndromes. The influence of age on expression of diseases associated with specific histocompatibility antigens is becoming better understood44 ; however, the strongest major histocompatibility-complex disease association remains that of human leukocyte antigen B27 with ankylosing spondylitis, a disease that begins in childhood in only about 10% of cases. The fact that this genetic predisposition does not more frequently result in childhood onset may reflect yet other factors that differentiate the child from the adult, such as the extent of environmental antigenic experience and the ability of the immune system to respond to its accumulating impact year after year. One argument in support of the significance of antigenic memory and immune reactivity might be exemplified by oligoarthritis. This disease has a narrow age-at-onset distribution: In the majority of children, onset occurs between the ages of 1 and 4 years. 38 Another striking age restriction is apparent in Kawasaki disease, which in North America affects most frequently the 1- to 3-year-old age group.45 One might speculate that these diseases reflect the effect of age at initial exposure to an environmental pathogen such as a virus or bacterium, together with the absence of specific protective immunity, in a genetically predisposed individual.
,1 • TABLE 1-2 Factors rlMt Potentially Modify Expression of Rheumati< Diseases in Childhood
Early expression of genetically determined abnormalities Immaturity and relative inexperience of the immune system Limited antigenic exposure Immaturity of the skeleton and the potential for growth and development Imn:laturity of the gonads and variable hormonal intluences
5
The degree and manner in which skeletal maturity influences the expression of rheumatic diseases is poorly understood. It seems probable, however, that physical and biochemical differences between younger and older cartilage and bone influence the effect of an inflammatory process involving these structures. Because physes are not fused in the growing child with arthritis, local growth abnormalities (e.g., leg-length inequality) may occur. Short stature is a frequent result of widespread arthritis in the child, but it is not an expected development in the skeletally mature adult with arthritis. The anatomy of the blood supply to the physis and epiphysis in the infant is reflected in this age group's predisposition to septic arthritis as a complication of osteomyelitis. The impact of gonadal immaturity on the expression of rheumatic diseases is unclear. SLE occurs predominantly in the pubertal and postpubertal age range. In some studies of SLE,46 girls and boys were affected with equal frequency in early childhood, but in adulthood, women developed this disorder 8 to 9 times more frequently than men. That these observations may reflect the role of reproductive hormones in pathogenesis is supported by studies of adult men with Klinefelter's syndrome cLe., males with two X chromosomes and one Y chromosome), in whom the frequency of SLE is high,47,48 and by studies of lupus-like disease in mice. 49 In contrast, men are much more likely than women to develop ankylosing spondylitis, a characteristic that does not appear to be significantly affected by age.
CHILDHOOD RHEUMATIC DISEASES: EXTENT OF THE PROBLEM It has been difficult to establish the extent of childhood
rheumatic diseases in defined populations with any accuracy. 50 In many of the most densely populated areas of the world, incidence and prevalence data for these disorders do not exist. In the developed world, inconsistencies of definition and classification, the rarity of occurrence for many of these diseases, and brevity of follow-up, have prevented accumulation of any substantial body of epidemiologic data. Two fundamental questions require answers: (1) How many children and adolescents have each of the identifiable rheumatic diseases; and (2) What is the functional outcome for these children? Estimates of the relative frequencies of adult-onset and childhood-onset rheumatic diseases are in Table 1-3. Three national registries from 1996 provided some comparative insights concerning the relative prevalence of the rheumatic diseases of childhood in the United States? Canada,52 and the United Kingdom53 (Table 1-4). Conspicuously under-represented are children with acute rheumatic fever, which is the major rheumatic disease of childhood in much of the world. A study by Manners and Diepeveen54 in Western Australia documented that many cases of chronic arthritis in children went undiagnosed and untreated. If this is the reality of identification in a developed area of the world with readily available expert medical care, the proportion of such children in geographic areas where medical care is less accessible may be even higher. In Finland, Kunnamo and colleagues55
6
C HAP T E R
II
TABLE. I 3
I
INTRODUCTION TO THE STUDY OF RHEUMATIC DISEASES IN CHILDREN
FIl'clueiKY ollhe Rheullldtic [)isedses in Adults dnd Iheir Onset in Childhood
No, Affected
Sex RaUo
Rheumatic Disease In Adults
(xl()5)
(F:M)
Ethnic RaUo (Whlte:Black)
Peak Age at Onset (yr)
Qlldhood Onset (0/0)
Ankylosing spondylitis Rheumatoid arthritis Systemic lupus erythematosus Dermatomyositis/polymyositis Scleroderma Polyarteritis
SOD-WOO 800 50
1:5 3:1 10:6 11:8 4:1 1:1
White> Black Equal 1:4 1:3 Black> White Equal
Young adult Increases with age (20-30) 15-40 45-60 Increases with age 00-50) 40-60
5 18 20 3 Rare'
1.~3.s
24-29 6,3
10
'Except for Kawasaki disease, Modified from data in ref. SO and Chaplers 9-24.
surveyed all children younger than 16 years of age who had swelling or limitation of motion of a joint, walked with a limp, or had hip pain as determined by a primary care physician, pediatrician, or orthopedic surgeon. All of these patients were subsequently examined by a single group of pediatric rheumatologists. Overall, the incidence of arthritis was estimated to be 109 per 100,000 children per year. Transient synovitis of the hip accounted for 48%; other acute transient arthritis CHenoch-Schbnlein purpura, serum sickness) 24%; chronic arthritis, 17%; septic arthritis, 6%; and reactive arthritis, 5%. Connective tissue diseases such as SLE were not identified in this survey. Determination of outcome and the lifelong burden of a pediatric rheumatic disease requires much more precise and comprehensive data than those currently available. The impact of childhood rheumatic diseases on life expectancy, their contribution to morbidity and costs of medical care, and their effect on quality of life are all prognostic parameters of importance about which there is little information even in North Amer:ica and Europe, and no information whatsoever on the global scene. Nevertheless, there can be little doubt that a child who, for example, has arthritis beginning at the age of 2 or 3 years will carry a lifelong burden in one or more of these areas. The expense and inconvenience for other members of the family are also significant.
ADVANCES IN PEDIATRIC RHEUMATOLOGY In the last half century, there have been dramatic advances in understanding the nature of inflammation, the cells and molecules that mediate it, and the therapeutic pOSSibility of specifically regulating the aberrant
I.
TABLE 1-4
immunoinflammatory response. The genetics of rheumatic diseases and, more recently, of the polymorphisms of inflammatory mediators, are pointing the way to therapeutic manipulations at an even more fundamental level, that of the gene. Although this approach is still only a hope for the future, it has been successful experimentally in animal models of arthritis. Currently, specific therapeutic modulation of certain mediators of inflammation is possible, and in particular blockade of tumor necrosis factor-a CTNF-a) has demonstrated great benefit in children with chronic arthritis. It may be premature to consider further advances in understanding of the childhood rheumatic diseases, because so much more must be illuminated. Nevertheless, it is evident that mortality from diseases such as chronic arthritis complicated by amyloidosis, dermatomyositis, and SLE has been dramatically reduced since the 1970s. Disability associated with many rheumatic diseases has been minimized: Wheelchair dependence is now rare, the need for aids to ambulation uncommon, and significant leg-length inequalities increasingly infrequent. Visual outcome in children with uveitis is improved. But much more progress is necessary and should be demanded and expected. Morbidity and mortality, although diminished, still remain serious threats to the child with SLE, vasculitis, or scleroderma, among other disorders. Although there have been major improvements in short- and medium-term outcomes of these and other rheumatic diseases, long-term outcomes are still often disappointing. For example, half of the children with chronic arthritis have active disease 10 years after onset, and children with SLE accumulate visceral damage with the passage of time, with an enduring impact on quality of life, despite much better control of acute lifethreatening events.
Reldtive Frequencies 01 Rheullldtic Disedses in I)edidtrl< Rhellllidtoloyy Clinic s in North Alllericd dnd the United KlIlydolll
U.S.A.S1 Juvenile rheumatoid arthritis/juvenile chronic arthritis Mechanical!orthopedic Vasculitis Systemic lupus erythematosus Juvenile dermatomyositis Systemic scleroderma Rheumatic fever/poststreptococcal arthritis Total
33.1 34.9 10.2 7,1 5,2 0,9 8.6 100.0
CANADASZ 50.0 40,6 3.0 3,9 1.6 0,2 0,7 100,0
U.K." 61.7 32.6 1.9 1.3
2,3 0,2
o 100.0
C HAP T E R
I
INTRODUCTION TO THE STUDY OF RHEUMATIC DISEASES IN CHILDREN
The reasons for these improvements in outcomes are multiple; chief among them are the establishment of a body of knowledge and expertise and involvement of a multidisciplinary team of health professionals in diagnosis and care. Improved application of old techniques and the development of new approaches have been important contributors to improved prognosis. Therapeutic landmarks of importance to the child with a rheumatic disease must include the introduction of cortisone for treatment of rheumatoid arthritis: Its influence on pediatric rheumatology has been profound. Intra-articular steroid use has improved management of oligoarthritis in children, just as methotrexate has radically improved the course and outcome of childhood polyarthritis. More judicious use of glucocorticoids and cytotoxic drugs has minimized toxicity and maximized effectiveness in diseases such as SLE and dermatomyositis. Biologic agents acting against TNF-lX and interleukin1 count among the major advances of the last decade. Although not curative, these agents and their successors promise to revolutionize management of the severe rheumatic diseases of childhood. Identification of the cause of Lyme disease and the pathogenesis of neonatal lupus, and the aggressive treatment of Kawasaki disease with intravenous immunoglobulin, represent landmarks of progress. Collaborative clinical trials led by the Pediatric Rheumatology Collaborative Study Group (PRCSG) and, more recently, the Pediatric Rheumatology International Trials Organisation (PRINTO) and the Childhood Arthritis and Rheumatology Research Alliance (CAARA), have facili~ted the study of therapeutic interventions in chronic arthritis as well as in the rarer connective tissue diseases. Recognition of the appropriateness of patient and family involvement at all stages of decision-making and care has enabled individualized treatment options and improved compliance. Family support organizations such as the American Juvenile Arthritis Organization in the United States and similar groups abroad have helped to promote education and research and to provide psychosocial support for patients and families.
REFERENCES 1. Ruffer MA. Rietti A: On osseous lesions in ancient Egyptians. J Pathol Bacterial 16: 439. 1912. 2. Kramer C: A case of ankylosing spondylitc, in mediaeval Geneva. Ossa 8: 115, 1982. 3, Rothschild BM. Woods RJ: Symmetrical erosive disease in Archaic Indians: the origin of rheumatoid arthritis in the New World? Semin Arthritis Rheum 19: 27~284, 1990. 4. Barlow T: 51st Annual Meeting of the British Medical Association; Section of Diseases of Children. BM] 2: 509-519, 1883. 5. Dieppe P: Did Galen describe rheumatoid arthritis? Ann Rheum Dis 47: 84-85, 1988. 6. Baillou G: De Liber de Arthritede. Paris, 1591. Quoted by Delpeuch G: La Goulle et Ie Rhumatisme, Paris. 1900. 7. Sydenham T: A Treatise of the Gout and Dropsy. London, 1683. 8. Garrod AB: The Nature and Treatment of Gout and Rheumatic Gout. London. Walton and Maberly, 1859. (Cited by Baethge BA.] Rheumatol19: 185, 1992.) 9. Adams F: The Genuine Works of Hippocrates. Translated from the Greek. London, The Sydenham Society, 1849. (Published as the Loeb Classical Libraty translation of the works of Hippocrates, Harvard University Press. Cambridge. MA. reprinred in 1967-68; also reprinted by The Classics of Medicine Library. Gryphon Editions, Ltd. Birmingham, AL. 1985.) 10. Brewer E] ]r, Bass ]C, Cassidy]T, et al: Criteria for the classification of juvenile rheumatoid arthritis. Bull Rheum Dis 23: 712-719, 1972. II. European League Against Rheumatism: EULAR Bulletin No.4: Nomenclature and Classification of Arthritis in Children. Basel, National Zeitung AG, 1977.
7
12. Petty RE, Southwood TR, Manners p. et al: International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol 31: 390-392, 2004. 13. Hayem F: The history of chronic joint diseases in children. Rev Rhum Engl Ed 66: 499-504, 1999. 14. Phaire T: The Regiment of Life, Whereunto is Added a Treatise of the Pestilence, with The Boke of Chyldren. Newly Corrected and Enlarged. London, Edw. Whitechurch, 1545. (Reprinted 1955 by E. & S. Livingston Ltd., London, p. 31.) 15. Bywaters EG: The history of pediatric rheumatology. Arthritis Rheum 20: 145-152. 1977. 16. Cornil MV: Memoire sur des coincidences pathologiques du rhumatisme articulaire chronique. C R Soc Bioi (Paris) Series 4, 3: 3, 1864, 17. Bouchut E: Traite pratique des Malades des Enfanrs. Ed. 6. Paris. 1875. 18. Moncorvo: Du Rhumatisme Chronique Noueux Des EnI'ants, Paris, 1880. 19. West C: Lectures on the Diseases of Infancy and Childhood. Ed. 7, Philadelphia, 1881. 20. Diamant-Berger M-S: Du Rhumatisme Noueux (Polyarthrite Deformante) Chez les Enfants. Paris, Lecrosnier et Babe. Libraires -Editeurs, 1891. (Reprinted by Editions Louis Pariente, Paris, 1988,) 21. Still GF: On a form of chronic joint disease in children. Med-Chirurg Trans 80: 47-59, 1897, (Reprinted in Arch Dis Child 132: 195, 1978.) 22. Keen ]H: George Frederic Still-Registrar. Great Ormond Street Children's Hospital. Br] Rheumatol 37: 1247, 1998. 23. Koplick H: Arthritis deformans in a child seven years old. Arch Pediatr 13: 161, 1896. 24, Schonlein ]L: Allgemeine und specielle Pathologie und Therapie. Nach dessen Vorlesungen niedergeschrieben und hrsg. von einigen seiner Zuhi'lrer. ed. 3. Ellinger, Herisau Lit Compt. Wurzburg, 1837. 25, Henoch EHH: Ober eine eigenthumliche Form von Purpura. Berl Klin Worchschr 11: 641, lR74. (Translated and reprinted in Am] Dis Child 128: 78. 1974,) 26. Unvericht H: Uber cine eigenrumliche Form von acuter Muskelentzundung mit einem der Trichinose ahnelnden Krankheitsbilde. Munch Med Wochenschr 34: 488, 1887. 27, Osler W: On the visceral manifestations of the erythema group of skin diseases, Am] Med Sci 127: I, 1904. 28. Watson R: An account of an extraordinary disease of the skin. and its cure. Extracted from the Italian of Carlo Crusio: accompanied by a letter of the Abbe Nollet, F.R.S. to Mr. William Watson, F.R.S, Philos Trans R Soc Lond 48: 579, 1754. (Cited by Rodnan GP, Benedeck GT: An historical account of the study of progressive systemic sclerosis [diffuse scleroderma). Ann Intern Med 57: 305, 1968.1 29, Travers B: Curious case of anchylosis of a great part of the vertebral column, probably produced by an ossification of the intervertebral substance. Lancet 5: 254, 1814, (Cited by Bywaters EGL. In Moll ]MH: Ankylosing Spondylitis. Edinburgh. Churchill Livingstone, 1980. p. 14.1 30, Hart FD, Maclagan NF: Ankylosing spondylitis: a review of 184 cases. Ann Rheum Dis 14: 77-83, 1955. 31. Schaller ], Bitnum S, Wedgwood R]: Ankylosing spondylitis with childhood onset.] Pediatr 74: 505-516, 1969. 32. Ladd ]R. Cassidy JT. Martel W: Juvenile ankylosing spondylitis. Arthritis Rheum 14: 579-590, 1971. 33, Kawasaki T: [Acute febrile mucocutaneous syndrome with lymphoid involvement with specific desquamation of the fingers and toes in children!. Arerugi 16: 178-222, 1967, 34, Munro-Faure H: Necrotising arteritis of the coronary vessels in infancy; case report and review of the literature. Pediatrics 23: 914-926, 1959. 35, Ansell BM, Bywaters EGL, Spencer PE, et al: Ansell BM: Looking back 1947-1985. The Canadian Red Cross Memorial Hospital. Taplow, United Kingdom, 1997, 36, Smythe HA: Historical vignette. Professor Eric Bywaters, 1910-2003. Memories of Taplow.] Rheumatol 31: 601-604, 2004, 37. Stillman ]S: The history of pediatric rheumatology in the United States, Rheum Dis Clin North Am 13: 143-147, 1987. 38, Hanson V: Pediatric rheumatology: A personal perspective. Rheum Dis Clin North Am 13: 155-159. 1987. 39. Levinson JE: Rellections of a pediatric rheumatologist. Rheum Dis Clin North Am 13: 149-154, 1987, 40. Ansell BM: Taplow reminiscences.] Rheumatol 19 (Suppl 33): 105-107, 1992, 41. Brewer E] .Ir: The last thirty and the next ten years.] Rheumatol19 (SuppI33): 108, 1992. 42. Stoeber E: Zur Geschichte Der Kinderklinik und Rheumakinderklinik in Garmisch-Partenkirchen: 1952-1986: Garmisch-Partenkirchen, Umschloggestalrung Christa J. Burges, 1986. 43, Sullivan DB, Cassidy]T, Petty RE: Pathogenic implications of age of onset in juvenile rheumatoid arthritis. Arthritis Rheum 18: 251-255, 1975. 44. Murray K], Moroldo MB, Donnelly P, et al: Age-specific effects of juvenile rheumatoid arthritis-associated HLA alleles, Arthritis Rheum 42: 1843-1853. 1999. 45, Wortmann OW, Nelson AM: Kawasaki syndrome. Rheum Dis Clin North Am 16: 363-375, 1990. 46. Cassidy JT, Sullivan DB, Perty RE, et al: Lupus nephritis and encephalopathy: prognosis in 58 children. Arthritis Rheum 20: 315-322, 1977, 47, Ortiz-Neu C, LeRoy EC: The coincidence of Klinefelter's syndrome and systemic lupus erythematosus. Arthritis Rheum 12: 241-246. 1969.
8
CHAPTER
I
INTRODUCTION TO THE STUDY OF RHEUMATIC DISEASES IN CHILDREN
48. Jimenez-Balderas r], Tapia-Serrano R, Fonseca ME, et al: High frequency of association of rheumatic/autoimmune diseases and untreated male hypogonadism with severe testicular dysfunction. Arthritis Res 3: 362-367, 2001. 49. Roubinian J. Talal N, SHteri PK, et al: Sex hormone modulation of autoimmunity in NZBINZW mice. Arthritis Rheum 22: 1162-1169, 1979. 50. Lawrence RC, Helmick CG, Arnett FC, et al: Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States. Arthritis Rheum 41: 778-799, 1998. 51. Bowyer S, Roettcher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group. J Rheumatol 23: 1968-1974, 1996.
52. Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic dbeases: results from the Canadian Pediatric Rheumatology Association Disease Registry. J Rheumatol 23: 1981-1987, 1996. 53. Symmons DPM, Jones M, Osborne J, et al: Pediatric rheumatology in the United Kingdom: data from the Britbh Pediatric Rheumatology Group National Diagnostic Register. J Rheumatol 23: 1975-1980, 1996. 54. Manners PJ, Diepeveen DA: Prevalence of juvenile chronic arthritis in a population of 12-year-old children in urban Australia. Pediatrics 98: 84--90, 1996. 55. Kunnamo 1, Kallio P, Pelkonen P: Incidence of arthritis in urban Finnish children: A prospective study. Arthritis Rheum 29: 1232-1238, 1986.
2
C HAP T E R
STRUCTURE AND FUNCTION Ross E. Petty and James T. Cassidy
~
Rheumatic diseases frequently affect many different organ systems, but inflammation of the structures of the musculoskeletal system-particularly joints, connective tissues, and muscles-is common to almost all. 1 An appreciation of some of the fundamental aspects of the development, structure, and biochemistry of the connective tissues and the components of the musculoskeletal system is important to the study of pediatric rheumatology. This chapter is intended as an overview of selected aspects of the anatomy and biochemistry of tissues that are particularly important to the study of rheumatic diseases of childhood and as a stimulus for further study.
JOINTS Oassltlcatlon of Joints Joints may be classified as ji'brous, cartilaginous, or synovial (Table 2-1).2 Fibrous joints (synarthroses) are those in which little or no motion occurs and in which the bones are separated by fibrous connective tissue. Cartilaginous joints (amphiarthroses) are those in which little or no motion occurs but the bones are separated by cartilage. Synovial joints (diarthroses) are those in which considerable motion occurs and a joint space lined with a synovial membrane is present between the bones. It is the synovial joint that is the site of inflammation in most of the chronic arthritides of childhood.
the "cavity" regresses and becomes filled with fibrous tissue. 7 The synOVial lining forms subsequent to cavitation, and the development of other structures, such as bursae, intra-articular fat, tendons, muscle, and capsule, quickly ensues. The whole process takes place between the fourth and seventh weeks of gestation, except for the temporomandibular joint, which develops much later. 8
Anatomy of Synovial Joints The anatomy of a typical synovial joint is illustrated in Figure 2-1. The bones of such joints are almost always covered by hyaline cartilage. 9 The synovial membrane attaches at the cartilage-bone junction so that the entire joint "space" is covered by either hyaline cartilage or synovium. The temporomandibular joint is unusual in that the articular surface of the condyle is covered by fibrocartilage rather than by hyaline cartilage. In some synovial joints, intra-articular fibrocartilaginous structures are present. For example, a disk (or meniscus) separates the temporomandibular joint into two spaces; the knee joint contains two menisci that separate the articular surfaces of the tibia and femur; and the triangular fibrocartilage of the wrist joins the distal radioulnar surfaces. Other intra-articular structures include the anterior and posterior cruciate ligaments of the knee, the interosseous ligaments of the talocalcaneal joint, and the triangular ligament of the femoral head. These structures are actually extrasynovial, although they cross through the joint space.
Development of Dlarthrocllal Joints In the fetus, diarthrodial joints develop by the differentiation of a noncartilaginous interzone within the cartilage core of the limb bud. Subsequently, cavitation occurs in this location, resulting in the formation of a joint "space. "3 The most important signals for joint morphogenesis are provided by the cartilage-derived morphogenetic protein 1 (CDMPl) and the bone mOl'phogenetic proteins (BMPs).4,5 The joint "cavity" is occupied at first by hyaluronic acid-rich joint fluid secreted by fibroblast-like cells lining the synovial membrane. Continued development of the diarthrodial joint depends on fetal movement, 6 which induces formation of cartilage and synovial membrane, and without which
ARTICULAR CARTILAGE The hyaline cartilage, which covers subchondral bone, facilitates relatively frictionless motion and absorbs the compressive forces generated by weight-bearing.9--15 The cartilage is firmly fixed to subchondral bone, and its margins blend with the synovial membrane and the periosteum of the metaphysis of the bone. In children, hyaline cartilage is white or slightly blue and is somewhat compressible. It is composed of chondrocytes within an extracellular matrix (ECM) and becomes progressively less cellular throughout the period of growth; the cell volume in adult articular cartilage is less than 2%.16 The
9
10
1'111
C HAP T E R
lABlE
2-)
2
STRUCTURE AND FUNCTION
(Iassifi
Type
Motion
Charaderlstlcs
Examples
Disease Target
Fibrous Cartilaginous Synovial
No No Yes
Bones separated by flbrous connection tissue Bones separated by cartilage Bones separated by joint space lined with synovial membrane
Sutures of skull Symphysis pubis Joints of extremities
None Ankylosing spondylitis Juvenile rheumatoid arthritis
matrix consists of collagen fibers, which contribute tensile strength, and ground substance composed of water and proteoglycan, which contributes resistance to compression. 17-20
Cartilage Zones Articular cartilage is organized into four zones (Fig. 2-2).9 Zones 1, 2, and 3 represent a continuum from the most superficial area of zone 1, in which the long axes of the chondrocytes and collagen fibers are parallel to the surface; through zone 2, where the chondrocytes become rounder and the collagen fibers are oblique; to zone 3, in which the chondrocytes tend to be arranged in columns perpendicular to the surface. The tidemark, a line that stains blue with hematoxylin and eosin, separates zone 3
Quadriceps femoris m.
"
from zone 4 and represents the level at which calcification of the matrix occurs. Chondrocytes in each of the cartilage zones differ not only in appearance but in metabolic activity, gene expression, and response to stimuli. 21 In the child, end-capillaries proliferate in zone 4, eventually leading to replacement of this area by bone. This is probably the manner in which the chondrocytes are nourished, although in the adult, constituent replacement (through the exchange of synovial fluid with cartilage matrix) may play the predominant role.
Chondrocytes Chondrocytes are the sole cellular constituents of cartilage. Their terminal differentiation determines the character of the cartilage (hyaline, fibrous, or elastic). This
",
......
Patellar tendon'''''
... ,
.
Hyaline cartilage'..
• Figure Z-l Sagittal section of the knee. The distinguishing features of the diarthrodial joint are shown, including bone, hyaline cartilage, synovial space, fibrocartilage, capsule, bursae, ligaments, tendons, muscles, and the vascular and nervous supply.The rheumatic diseases affect all these structures individually or in concert.
C HAP T E R
CE:>
CD
~
<:» t::E:> ~
CD CD CD CD
<0
CD
<0
CJ)
~ CD <:?)
C!)
~
Q:>
c:Et ez:o CD CD C2> CI> <:il c::J)
G)G>
(0
IZone 1
e> (;') <::>
G<;)
e•
G> C!)Q)~ e ~ GO €> 1;"\ Qle G> G C!) 'U E:l ~ (!) €> ~
e
0
e
11
superficial zone protein Oubricin) which is important in maintaining relatively frictionless joint motion. Synthesis of this protein is defective in the camptodactyl-arthropathycoxa vara-pericarditis syndrome 24 (see Chapter 40).
Extracellular Matrix Zone 3
0
III'I~I ~~~i1~'I~
STRUCTURE AND FUNCTION
Zone 2
2
Zone 4
~~"trr~~iI~ ~~~~~~~~YII~~»J
The ECM consists of collagen fibers, which contribute tensile strength, water, diverse structural and regulatory proteins, and proteoglycans (mainly aggrecan), which together contribute resistance to compressiony-20 The ECM is heterogeneous and can be subdivided into three compartments. A thin inner rim of aggrecan-rich matrix surrounds the chondroeytes and lacks crosslinked collagen. An outer rim contains fine collagen fibrils. The remainder of the ECM consists primarily of aggrecan, which binds via the link protein to hyaluronan (Fig. 2_3).21 The endoskeleton of hyaline cartilage consists of a network of collagen fibrils, 90% of which are type II collagen, with minor components of collagen types IX and X. 16
. .....re
Proteoglycans
complex process has been recently reviewed. 22 Chondroeytes in articular cartilage persist and do not ordinarily divide after skeletal maturity is attained. 23 Those in the epiphyseal growth plate differentiate to facilitate endochondral ossification, after which they may undergo apoptosis or become osteoblasts. 23 Chondrocytes are responsible for the synthesis of the two major constituents of the matrix, collagen and proteoglycan, as well as enzymes that degrade matrix components (collagenase, neutral proteinases, and cathepsins).23 This dual function places the chondrocyte in the role of regulating cartilage synthesis and degradation. Immediately surrounding the chondrocyte is the pericellular region, which contains type VI collagen and the proteoglycans decorin and aggrecan. 16 Chondrocytes in zone 1 produce
Proteoglycans are macromolecules consisting of a protein core to which are attached 50 to 100 unbranched glycosaminoglycans (chondroitin sulfate and O-linked keratan sulfate).25-27 At least five different protein cores have been defined. The principal proteoglycan of hyaline cartilage is called aggrecan. Its attachment to hyaluronan is stabilized by link protein to form large proteoglycan aggregates with molecular weights of several million (see Figure 2_3).16,17,19 With increasing age, the size of the proteoglycan aggregates increases, the protein and keratan sulfate content increases, and the chondroitin sulfate content decreases. 2R ,29 Chondroitin sulfate chains also become shorter with increasing age, and the position of the sulfated moiety changes, from a combination of 4sulfated and 6-sulfated N-acetylgalactosamine at birth to mainly 6-sulfated N-acetylgalactosamine in the adult,3lH2 Dermatan sulfate and chondroitin-4-sulfate are the principal mucopolysaccharides in skin, tendon, and aorta; heparan sulfate is present in basal lamina.
Z-Z Organization of articular cartilage. In zone 1, adjacent to the joint space, the chondroc.ytes are flattened. In zone 2. the chondrocytes are more roun~d, and in zone 3 they are arranged in perpendicular columns. The tide mark separates zone 3 from zone 4, which is impregnated with calcium salts. Bone is beneath zone 4. (Courtesy of J. R. Petty.)
• f1ture Z-3
The structure of the proteoglycan aggregate of cartilage. The proteoglycan monomer consists of a core protein (a) of variable length that contains three globular domains: G1 (located at the aminoterminus and containing the hyaluronate binding region) G2, and G3. Link protein (e) stabilizes the aggregate by binding simultaneously to the hyaluronate chain (d) and Gl. Glycosaminoglycan molecules are attached to the core protein in specific regions: keratan sulfate (b) and chondroitin sulfate (c).
12
C HAP T E R
2
STRUCTURE AND FUNCTION
Collagens Collagens, the most abundant structural proteins of connective tissues, are glycoproteins with high proline and hydroxyproline content.33--35 Many are tough, fibrous proteins that provide structural strength to the tissues of the hody.36 There are at least 21 different types of collagen divided into seven subclasses (Table 2_2).37-39 Types I, II, and III are among the most common proteins in humans. Type II collagen, the principal constituent that accounts for more than half the dry weight of cartilage, is a trimer of three identical a-helical chains. Collagen types III, VI, IX through XII, and XIV are all present in minute quantities in the mature cartilage matrix. 33 The content of types IX and XI collagen is greater in young animals (20%) than in mature animals (3%).33 Collagen synthesis is minimal in the mature animal. The degree of stable cross-linking of collagen fibers increases with advancing age up to the fourth decade of life. 40 This results in increased resistance to pepsin degradation and may contribute to the increased rigidity and decreased tensile strength of old cartilage. 41 Collagen undergoes extensive changes in primary and tertiary structure after it is secreted from the fibroblast into the extracellular space as a triple-helical procollagen (Fig. 2-4),42 Specific peptidases cleave the amino and carhoxyl extension peptides, yielding collagen molecules
u: II
TABI E l l
that in some types form cross-links and fibrils via lysyl and hydroxylysyl residues. Glycosylation also occurs at this post-translational stage. Collagen genes are named for the type of collagen (e.g., COLI) and the fibril (e.g., Al) and encode the large triple-helical domain common to human collagens. Mutations in the collagen genes 43--46 account for human diseases such as Ehlers-Danlos syndrome and osteogenesis imperfecta (see Chapter 40):j7--49
Other Connective Tissue Constituents In addition to collagens, a number of specialized tissues derived from embryonic mesoderm contribute to connective tissue structures other than cartilage. Elastin occurs in association with collagen in many tissues, especially in the walls of blood vessels and in certain ligaments.50 Fibers of elastin lack the tensile strength of collagens but can stretch and then return to their original length. Elastin is produced by fibroblasts and smooth muscle cells. Fibronectin is a dimeric glycoprotein with a molecular weight of 450,000 that acts as an attachment protein in the ECM. 51 It is produced by many different cell types, including macrophages, dedifferentiated chondrocytes, and fibroblasts, and has the ability to bind to collagens, proteoglycans, fibrinogen, and actin, as well as to cell surfaces and bacteria. Fibronectin
IYI'es 01 (oUdl/en
Subclass and Type
Composition
Duue Distribution
Fibril-forming collagens Type I Type II Type III Type V Type XI
0.1(0, 0.2(0 o.10I) 0.1 (III) 0.1 (V) , o.2CY), o.3(V) 0.1 (XI) , o.2(XI), o.3(XI)
Most connective tissues; abundant in bone, skin, and tendon Cartilage, intervertebral disk, vitreous humor Most connective tissues, particularly skin, lung, blood vessels Tissues containing type I collagen, quantitatively minor component Cartilage, intervertebral disk, vitreous humor
Network-forming collagens o.ICIV), o.20V) , o.30V), o.40V) , Type IV o.5(N), o.60V) 0.1 (VIII), o.2(VIJI) Type VIII 0.1 (X) Type X
Several tissues, especially endothelium Hypertrophic cartilage
FACIT collagens Type IX Type XII Type XIV Type XVI Type XIX Type XX Type XXI
Cartilage, intervertebral disk, vitreous humor Tissues containing type I collagen Tissues containing type I collagen Several tissues Rhabdomyosarcoma cells Corneal epithelium Fetal blood vessel walls
a. ICIX), o.2CIX) , o.3CIX) 0.1 (XII) 0.1 (XIV) 0.1 (XVI) o.1(XIX) o.1(XX) 0.1 (XXI)
Basement membranes
Beaded filament-forming collagen Type VI o.1CYI), o.2(VI), o.3(Vn
Most connective tissues
Collagen of anchoring fibrils Type VII o.1(VIJ)
Skin, oral mucosa, cervix, cornea
Collagens with a transmembrane domain Type XIII 0.1 (XIII) Type XVII a. 1(XVII)
Endomysium, perichondrium, placenta, mucosa of the intestine, meninges Skin, cornea
Collagen types XV and xvm Type XV 0.1 (XV) Type XVIII 0.1 (XVIII)
Many tissues, especially skeletal and heart muscle, placenta Many tissues, especially kidney, liver, lung
FAClT, fihril-associated collagens with interrupted triple-helices. From Ala-Kakko L, Prockop OJ: Collagen and elastin. In Harris ED Jr, et al (eds): Kelley's Textbook of Rheumatology, 7th ed. Philadelphia, Elsevier, 2005. pp. 35--47.
C HAP T E R
mRNA for specific procollagen chains
~
Translation on RER
~
Hydroxylation by 3-proline hydroxylase, 4-proline hydroxylase, and Iysyl hydroxylase
~
Glycosylation of hydoxylysine by galactosyl transferase and addition of glucose by glucosyltransferase
~
Removal of N-terminal signal peptide
~
Release of completed chains from ribosomes
~
Formation of disulfide crosslinks between chains
~
Formation of triple helix procollagen
~
2
STRUCTURE AND FUNCTION
13
alytic groupS'4: the metalloproteinases and serine proteinases, which are active at neutral to slightly alkaline pH, and the cysteine and aspartic proteinases, which are most active at acid pH (Table 2-3). The metalloproteinases, which are activated by calcium and stabilized by zinc ions, consist of at least 10 well-characterized enzymes. 55 They are active in the degradation or remodeling of collagens and are known to be synthesized by rheumatoid synovium. Collagenases are inhibited naturally by a?-macroglobulin and by the tissue inhibitor of metalloprot'i!inase (77MP). Strome(vsin is a neutral proteinase synthesized by cultured fibroblasts and synovium. Other members of this family, the gelatinases, are secreted by many cells in culture and are active in the remodeling of collagen-containing tissues. The serine proteinases are a family of endopeptidases that participate in matrix degradation either directly or by activating precursors of the metalloproteinases. They include many of the enzymes of pathways involving coagulation, fibrinolysis, complement activation, and kinin generation: plasmin, plasminogen activator, kallikrein, and elastase. Serine proteinase inhibitors constitute 10% of the plasma proteins. The cysteine proteinases that degrade ECM include cathepsins Band L, which are lysosomal enzymes associated with inflammatory reactions. The aspartic proteinases are primarily lysosomal proteinases active at acid pH. Cathepsin D is the major representative of this family that degrades proteoglycans and is present in the lysosomes of most cells.
Packaging of procollagen into vesicles
~
Fusion of vesicles with cell membrane, removal of C-terminal nonhelical extensions and part of N-terminal nonhelical regions
~
Collagen
• ,.... 2-4 Schematic representation of collagen biosynthesis.Triplehelical procollagen is secreted from the fibroblast. Specific procollagenases produce collagen by deaving the ends of the molecules.The collagen molecules (except for type IV collagen) then form fibrils that undergo cross-linking to form collagen fibers. (Adapted from Nimni ME: Collagen: structure, function, and metabolism in normal and fibrotic tissues. Semin Arthritis Rheum 13: 1, 1983.)
is present in plasma and as an insoluble matrix throughout loose connective tissues, especially between basement membranes and cells, Laminin is a major constituent of the basement membrane together with type IV collagen.'2 Reticulin may be an embryonic form of type III collagen. It is present as a fine branching network of fibers widespread in spleen, liver, bone marrow, and lymph nodes,
Protelnases for Collagen and Cartilage The proteinases (endopeptidases) are proteolytic enzymes that are active in homeostatic remodeling of the ECM during health and in its degradation during inflammatlon.,3 These enzymes occur both intracellularly and extrllcellularly in tissue fluids and plasma and have been clasfified into four categories based on functional cat-
SYNOVIUM Synovial Membrane The synovial membrane is a vascular connective tissue structure that lines the capsules of all diarthrodial joints and has important intra-articular regulatory functions. 56,57 The synovium consists of specialized fibroblasts,'8 one to three cells in depth, overlying a loose meshwork of type I collagen fibers containing blood vessels, lymphatics, fat pads, unmyelinated nerves, and isolated cells such as mast cells. 59,60 There is no basement membrane separating the joint space from the subsynovial tissues, The synovial membrane is discontinuous, and within the joint space there are so-called bare areas between the edge of the cartilage and the attachment of the synovial membrane to the periosteum of the metaphysis. 6! These bare areas are especially vulnerable to damage (erosion) by inflamed synovium (pannus) in inflammatory joint diseases, Folds, or villi, of synovium provide for unrestricted motion of the joint and for augmented absorptive area. The synoviocytes are of two predominant types, a subdivision that may reflect different functional states rather than different origins. SynOVial A cells are capable of phagocytosis and pinocytosis, have numerous microfilopodia and a prominent Golgi apparatus, and synthesize hyaluronic acid. Synovial B cells are more fibroblast-like, have a prominent rough endoplasmic reticulum, and synthesize fibronectin, laminin, and types I and III collagen as well as enzymes (collagenase, neutral proteinases) and catabolin.
14
C HAP T E R
2
[ . fABLE 2- 3
STRUCTURE AND FUNCTION
Proteinas('s and Inhibitors for (ollagen and (a,tildge Substrdtes
Enzyme
Substrate
Inhibitor
Metalloproteinases Collagenase Gelatinase Stromelysin
Various collagens and GAGs Denatured collagens Fibronectin, GAG, elastin, collagens
TIMP TIMP TIMP
Serine proteinases Plasmin Elastase Cathepsin G Plasminogen activator
Metalloproteinases Various collagens and GAGs GAGs, type II collagen, elastin, TIMP Proplasminogen
a 2-Antiplasmin 0. 1- Plasminogen inactivator aI-Plasminogen inactivator
Cysteine proteinases Cathepsin B Cathepsin L
Type II collagen, GAGs, link protein Type I collagen, GAGs, link protein, elastin
Cystatins Cystatins
Aspartic proteinases Cathepsin D
GAGs, type II collagen
0.2-Macroglobulin
GAGs, glycosaminoglycans; TIMP, tissue inhibitor of metalloproteinase.
Synovial Fluid
VASCULAR SUPPLY
SynOVial fluid, present in very small quantities in normal synovial joints, has two functions: lubrication and nutrition. 62 ,63 Synovial fluid is a combination of a filtrate of plasma that enters the joint space from the subsynovial capillaries and hyaluronic acid, which is secreted by the synoviocytes. Hyaluronic acid provides the high viscosity of synovial fluid and, with water, its lubricating properties. 64 Concentrations of small molecules (electrolytes, glucose) are similar to those in plasma, but larger molecules (e.g., complement components) are present in low concentrations relative to plasma unless an inflammatory state alters vasopermeability. Notably absent from synovial fluid are elements of the coagulation pathway (fibrinogen, prothrombin, factors V and VlI, tissue thromboplastin, and antithrombin).65 As a result, normal synovial fluid is resistant to clotting. There appears to be free exchange of small molecules between synovial fluid of the joint space and water bound to collagen and proteoglycan of cartilage, Characteristics of normal synovial fluid are listed in Table 2-4. 66-74
The arterial supply to the diaphysis and metaphysis of a long bone arises from a nutrient artery that penetrates the diaphysis and terminates in the child in end-arteries at the epiphyseal plate.75 The epiphyseal blood supply arises from juxta-articular arteries that also supply the synovium via a complex network of arterial and alteriovenous anastomoses and capillary beds, which William Harvey called the circulus articularis vasculosus. Not until growth has ceased and the epiphyseal plate has ossified is there arte-
Synovial Strudures Synovium lines bursae and tendon sheaths as well as joints,9 Bursae facilitate frictionless movement between surfaces, such as subcutaneous tissue and bone, or between two tendons. Bursae located near synovial joints frequently communicate with the joint space. This is particularly evident at the shoulder, where the subscapular bursa or recess communicates with the glenohumeral joint; and around the knee, where the suprapatellar pouch, the posterior femoral recess, and occasionally other bursae communicate with the knee joint. Tendon sheaths lined with synovial cells are prominent around tendons as they pass under the extensor retinaculum at the wrist and at the ankle. Although they are closely associated with joints, tendon sheaths do not communicate with the synovial space.
"' ..'
TABLE 2-4
Charaderlstlc Volume pH Retiltive viscosity CI,HCO, Na, K, Ca, Mg Glucose Total protein Albumin
Normal Synovial Fluid
Reference Mean or Representative Value· No.
0.13-3.5 mL (adult knee) 7,3-7.4 235 Slightly higher than serum Slightly lower than serum Serum value ± 10% 1.7-2,1 g/dL 1.2 g/dL 0,17 g/elL 0.1 0,15 g/elL 0.2 0,23 g/dL ~ 0,38 g/dL Y Immunoglobulin G 13% of serum value Immunoglobulin M 5% of serum value Immunoglobulin E 220/0 of serum value 3% of serum value a.,-Macroglobulin 24% of semm vallie Transferrin Ceruloplasmin 16% of serum value 30-50% of plasma value CH,,, 300 mg/dL Hyaluronic acid 7,1 mg/dL Cholesterol Phospholipid 13.8 mg/dL
66 70 66 66 66 67 73 74
71 72
71 69 68 74 74
CH,o' total hemolytic complement assay. 'These values were obtained from various adult synovial lluid studie". Similar data are not available for cbildren.
C HAP T E R
rial communication between the metaphyseal and epiphyseal-synovial circulations, a phenomenon of importance in explaining the predisposition of the immature diaphysis to infection and aseptic necrosis after trauma (Fig. 2-5).
CONNECTIVE TISSUE STRUCTURES Connective tissues proVide the supporting structures of the body (bone, periosteum, cartilage); permit the action of muscles through tendons, ligaments and fasciae; support internal organs (dermis, capsules, serosal membranes, basement membranes); and provide support for blood vessels, lymphatics, and the bronchopulmonary tree.
2
STRUCTURE AND FUNCTION
15
Entheses An enthesis is the site of attachment of tendon, ligament, fascia, or capsule to bone. Unlike tendon or ligament, the enthesis is an active metabolic site, particularly in the child. It includes the peritenon, which is continuous with the periosteum; collagen fibers of the tendon or ligament which insert into the bone (Sharpey 's fibers); the adjoining cartilage; and bone not covered by periosteum. 77 In 1998, a highly informative commentary was published on the strongest tendon in the body, the Achilles tendon, and its enthesis. 78 Entheses have been the subject of a recent extensive review. 79
SKELETAL MUSCLE
Tendons Tendons are specialized connective tissue structures that, via the enthesis, attach muscle to bone. 76 They contain, in addition to water, type I collagen and small amounts of elastin and type III collagen, the latter forming the epitenon and endotenon. The type III collagen fibers are densely packed in a parallel configuration in a proteoglycan matrix containing elongated fibroblasts.
Upments and Fasciae Ligaments and fasciae join bone to bone and, like tendons, are composed of type I collagen. So-called elastic ligalllents, such as the ligamenta flava and ligamentum nuchae, predominantly contain elastin.
b
Anatomy Skeletal muscle makes up approximately 40% of the adult body mass and consists of about 640 separate muscles that support the skeleton and permit movement and locomotion. Skeletal muscle is formed during embryogenesis from mesodermal stem cells that differentiate into the various types of muscle, bone, and connective tissue. A skeletal muscle is surrounded by the connective tissue epimysium. Within the muscle, fascicles are covered by connective tissue perimysium. 8o Each fascicle contains many individual muscle fibers, which are the basic structural units of skeletal muscle (Fig. 2-6). Muscle fibers are elongated, multinucleated cells surrounded by connective tissue endomysium (reticulin, collagen) that is richly supplied with capillaries. Within each fiber are a large number of myofibrils, consisting of highly organized interdigitated myofilaments of actin and myosin: 81 Each myofilament has approximately 180 myosin molecules with a molecular weight of 500,000, a long tail, and a double head. The myofilament is composed of the myosin tails; the myosin heads project in a spiral arrangement. Lying parallel to the myosin molecules are actin filaments (F-actin) composed of globular subunits of G-actin with a molecular weight of 42,000. Two actin filaments are coiled around each other as a helix, with a second protein, tropomyosin E, lying in the groove. A regulatory protein, troponin, is located at intervals along this structure. This complex structure is demonstrable by light or electron microscopy as striations. Creatine kinase is bound to the myosin filaments at regular intervals. 82
Muscle Contraction
:r • ~re Z-5 Blood supply to the epiphysis and metaphysis. End-arteries (a) at the epiphyseal plate arise from the medullary arteries. Juxta-articular arteries (b) supply epiphysis and synovium. (Courtesy of lR. Petty.)
The functional ability of muscle to produce coordinated movements is governed by the conversion of chemical to mechanical energy by actomyosin. 83 ,B4 Calcium diffusion in the myoplasm and binding to thin-filament regulatory proteins are stimulated by the action potential of the a-motor neuron. Variation in the properties of the various types of motor fibers and motor units, and recruitment of motor units, result in the specific patterns of movement. The properties of the motor unit are influenced by the genetic makeup of the individual, muscular conditioning, and the presence
16
C HAP T E R
2
STRUCTURE AND FUNCTION
of any disease that results in joint pain or immobilization, or metabolic, hormonal, or nutritional disturbances. 85 A
Types of Muscle Fibers Muscle fibers constitute 85% of muscle tissue. There are two major types of fibers, which differ in structure and biochemistry (Table 2_5).86-89 Most muscles contain both types. Type I (slow) fibers are narrower, have poorly defined myofibrils, are irregular in size, have thick Z bands, and are rich in mitochondria and oxidative enzymes but poor in phosphorylases. Type II (fastJfibers have fewer mitochondria and are poor in oxidative enzymes but rich in phosphorylases and glycogen. Type I and II muscle fibers can be differentiated histochemically (Fig. 2-7). Muscles differ in the proportions of each
B
c
D
--
--
• •
•
E
F
Troponin
Actin
t
Tropomyosin 8
• Figure 2-6 Schematic representation of the anatomy of skeletal muscle: a, fascicle; b, fiber; c, myofibrils; d, actin and myosin; eand f, enlargement of actin and myosin filaments showing the actin filaments coiled around each other and associated with tropomyosin Blying in the groove. (Courtesy of I.R. Petty.)
II.
TABLE 2-5
• Figure 2-7 Frozen section of normal skeletal muscle stained with adenosine triphosphatase, pH 9.2. Type I fibers are pale, and type II fibers are dark. Magnification x 800. (Courtesy of Dr. M. Norman.)
Classifhalioll of Muscle Fiber Types
Characteristic
Type I
Type IIA
be liB
TYPe lie
Size Color Myoglobin content Mitochondria Blood supply ATPase (pH 4.4) ATPase (pH 10.6) Lipid Glycogen Metabolic characteristics Oxidative (aerobic) Glycolytic (anaerobic) Function Contraction time Resistance to fatigue
Moderate Red High Many
Small White Medium Intermediate
Large White Low Few
Small White High Intermediate
+++
+
+
+
High Low High Low
Low High Low High
Low High Low High
Variable
High Moderate
Intermediate High
Low High
High High
Slow and sustained High
Fast twitch Moderate
Fast twitch Low
Moderate twitch Moderate
ATPase. adenosine triphosphatase.
C HAP T E R
fiber type, For example, the diaphragm contains predominantly "slow" fibers, and small muscles contain predominantly "fast" fibers, Muscle conditioning leads to adaptations in the contractile and structural proteins and fiber species within the genetic potential of the individual. Strength training results in hypertrophy of type IIB fibers, and endurance training leads to metabolic alterations in type I and type IrA fibers. B5 .90-92
REFERENCES 1. Edwards JC. Morris V: Joint physiology: relevant to the rheumatologist? Br .1 Rheumatol 37: 121-125. 1998. 2. S()koloff L, Boland JH: The Musculoskeletal System. Baltimore, Williams & Wilkins, 1975. 3. Archer CW, Dowthwaite GP, Francis-West P: Development of synovial joints. Uirth Defects Res Part C Embryo Today 69: 144-155, 2003. 4. Edwards q, Francis-West PH: Bone morphogenetic proteins in the development and healing of synOVial joints. Semin Arthritis Rheum 31: 33-42, 2001. 5. Reddi AH: Cartilage morphogenetic proteins: role in joint development, homaeostasis, and regeneration. Aru> Rheum Dis 62 (Suppl 11): ii73-ii78, 2003. 6. Drachman DB, Sokoloff L: The role of movement in embryonic joint development. Dev Bioi 14: 401, 1966. 7. Pitsillides AA: Identifying and characterizing the joint cavity-forming cell. Cell Biochem Funct 21: 235-240, 2003. 8, Ohnuki H: On the formation of the temporomandibular joint cavity in the human fetus. Kaibogaku Zasshi 75: 325-336, 2000. 9. Ghadially FN: Fine structure of joints. In Sokoloff L (ed): The Joints and Synovial Fluid. New York, Academic Press, 1978. 10. Silver FH: Biological Materials: Structure, Mechanical Properties, and Modeling of Soft Tissues. New York, New York University Press, 1987. 11. Mow V, Rosenwasser M: Articular cartilage: biomechanics. In Woo SLY, Buckwalter JA (eds): Injury and Repair of the Musculoskeletal Soft Tissues. Park Ridge, IL, American Academy of Orthopaedic Surgeons, 1988, p. 427, 12. Hall BK, Newman SA (eds): Cartilage: Molecular Aspects. Boca Raton, FL, eRC Press, 1991. 13. Jasin HE: Structure and function of the articular cartilage surface, Scand .1 Rheumatol Suppl 10: 51-55, 1995. 14. Dijkgraaf LC, de Bont LG, Baering G, et al: Normal cartilage structure, biocnemistry, and metabolism: a review of the literature. .1 Oral Maxillofac Surg 53: 924-929, 1995. 15. Guilak F, Jones WR, Ting-Beall HI', et al: The deformation behavior and mechanical properties of chondrocytes in articular cartilage, Osteoarthritis Cartilage 7: 59--70, 1999. 16. Poole AR, Kohima T. Yasuda T, et al: Composition and structure of articular cartilage, Clin Orthop 391S: S26-533, 2001. 17. J-Ieinegard 0, Oldberg A: Structure and biology of cartilage and bone matrix honcollagenous macromolecules. FASEBJ 3: 2042-2051, 1989. 18. Bruckner P, van der Rest M: Structure and function of cartilage collagens. Microsc Res Tech 28: 378--384, 1994. 19. Cohen NP, Foster RJ, Mow VC: Composition and dynamics of articular cartilage: structure, function, and maintaining healthy state. .1 Orthop Sports Phys Ther 28: 203-215, 1998. 20. Mow VC, Wang CC, Hung CT: The extracellular matrix, interstitial fluid and i<'ms as a mechanical signal transducer in articular cartilage. Osteoarthritis Cartilage 7: 41-58, 1999. 21. Chubinskaya S. Kuettner KE: Regulation of osteogenic proteins by chondrocytes. 1m .1 Biochem Cell Bioi 35: 1323-1340, 2003. 22. Shum L, Nuckolls G: The life cycle of chondrocytes in the developing skeleton [review). Arthritis Res 4: 94-106, 2002. Available at: http://arthritisresearch.com/content/4/2/094 (accessed November 21, 2004). 23. Archer CW, Francis-West P. The chondrocyte, Int .1 Biochem Cell Bioi 35: 401-404, 2003. 24. Warman ML: Human genetic insights into skeletal development, growth and homeostasis. Clin Orthop 379: S4Q-S5425, 2000. 25. Lander AD: Proteoglycans: master regulators of molecular encounter? Matrix Bioi 17: 465-472, 1998, 26. liirk DE. Silver FH, Trelstad RL: Matrix assembly. In Hay ED (ed): Cell Biology of the Extracellular Matrix. New York. Plenum Press, 1991, p. 221. 27. Goetinck P, Winterbottom N: Proteoglycans: modular macromolecules of the extracellular matrix. In Goldsmith L (ed): Biochemistry and Physiology of the Skin. New York, Oxford University Press, 1991. 28. Gallagher JT: The extended family of proteoglycans: social residents of the pericellular zone. Curr Opin Cell Bioi 1: 1201-1218, 1989. 29. 1I:>zzo RV: Matrix proteoglycans: from molecular design to cellular function, Annu Rev Biochem 67: 609--652, 1998, 30. Bayliss MT, Ali SY: Age-related changes in the composition and structure of human articular-cartilage proteoglycans, BiochemJ 176: 683--693,1978.
2
STRUCTURE AND FUNCTION
17
31. RougWey PJ, White RJ: Age-related changes in the structure of the proteoglycan subunits from human articular cartilage. .1 Bioi Chern 255: 217-224, 1980. 32. Roughley PJ: Age-associated changes in cartilage matrix. Clin Orthop 391S: S153- S160, 2001. 33. Eyre 0, Articular cartilage and changes in arthritis: collagen of articular cartilage. Arthritis Res 4: 30-35, 2002, Available at: http:// arthritis-research.com/content/4/1/030 (accessed November 21, 2004). 34. Nimni ME: Collagen: structure, function, and metabolism in normal and fibrotic tissues. Semin Arthritis Rheum 13: 1-86, 1983, 35, van der Rest M, Garrone R: Collagen family of proteins. FASEB .1 5: 2814-2823, 1991. 36. Boskey AL, Wright TM, Blank RD: Collagen and bone strength, J Bone Miner Res 14: 330-335, 1999. 37, Uitto .1, Murray LW, Blumberg B, et al: Biochemistry of collagen in diseases [UCLA conference). Aru> Intern Med 105: 740-756, 1986. 38. Koch M, Foley JE, Hahn R, et al: e:t 1 (XX) collagen, a new member of the collagen subfamily, fibril associated collagens with interrupted triple helices. .1 Bioi Chern 276: 23120-23126, 2000. 39. Chou MY, Li HC: Genomic organization and characterization of the human type XXI collagen (COL21Al) gene. Genomics 79: 395-401, 2002. 40. Weiss JB, Sedowofia K, Jones C: Collagen degradation: a defended multienzyme system. In Viidik A, Vuust .1 (eds): Biology of Collagen. London, Academic Press, 1978, p. 113. 41. Muir H, Bullough 1', Maroudas A: The distribution of collagen in human articular cartilage with some of its physiological implications. .1 Bone Joint Surg Br 52: 554-563, 1970. 42. Piez K: Molecular and aggregate structures in the collagens. In Piez KA, Reddi AH (eds): Extracellular Matrix Biochemistry, New York, Elsevier, 1984. p. 1. 43. Prockop OJ, Kivirikko K1: Heritable diseases of collagen. N Engl J Med 311: 376-386, 1984. 44. Kainulainen K, Pulkkinen L, Savolainen A. et al: Location on chromosome 15 of the gene defect causing Marfan syndrome. N EnglJ Med 323: 935-939, 1990. 45. Dietz HC, Cutting GR, Pyeritz RE, et al: Marfan syndrome caused by a recurrent de novo missense mutation in the fibrillin gene. Nature 352: 337-339, 1991. 46. Lee B, Godfrey M, Vitale E, et al: Linkage of Marfan syndrome and a phenotypically related disorder to two different fibrillin genes. Nature 352: 330-334, 1991. 47. Prockop OJ, Kivirikko KI: Collagens: molecular biology, diseases, and potentials for therapy. Annu Rev Biochem 64: 403-434. 1995. 48. Raff ML, Byers PH: Joint hypennobility syndromes. Curr Opin Rheumatol 8: 459-466, 1996. 49. Prahalad S, Colbert RA: Genetic diseases with rheumatic manifestations in children. Curr Opin Rheumatol 10: 488--493, 1998. 50, Sandberg LB, Soskel NT, Leslie JG: Elastin structure, biosynthesis. and relation to disease states. N EnglJ Med 304: 566-579, 1981. 51. Ruoslahti E, Engvall E, Hayman EG: Fibronectin: current concepts of its structure and functions. Coil Relat Res 1: 95-128, 1981. 52, Beck K, Hunter I, Engel J: Structure and function of laminin: anatomy of a multidomain glycoprotein. FASEBJ 4: 148--160, 1990. 53. Bond JS, Butler PE: Intracellular proteases. Annu Rev Biochem 56: 333-364, 1987, 54. Cawston TE: Proteinases and inhibitors. Br Med Bull 51: 385-401, 1995. 55. Woessner .JFJ: Matrix metalloproteinases and their inhibitors in connective tissue remodeling. FASEBJ 5: 2145-2154. 1991. 56. Edwards J: Second international meeting on synovium: cell biology, physiology and pathology. Aru> Rheum Dis 54: 389--391. 1995. 57. FitzGerald 0, Bresnihan B: Synovial membrane cellularity and vascularity. Ann Rheum Dis 54: 511-515. 1995. 58. EdwardsJC: Synovial intimal fibroblasts, Ann Rheum Dis 54: 395-397,1995, 59. Revell PA, al-Saffar N, Fish S, et al: Extracellular matrix of the synovial intimal cell layer. Ann Rheum Dis 54: 404-407, 1995. 60. Athanasou NA: Synovial macrophages. Ann Rheum Dis 54: 392-394, 1995. 61. Muller-Ladner U, Gay RE, Gay S: Molecular biology of cartilage and bone destruction. Curr Opin Rheumatol 10: 212-219. 1998. 62. McCutchen CW: Lubrication of joints. In Sokoloff L (ed): The Joints and Synovial Fluid. New York, Academic Press, 1978, p. 437. 63. Simkin PA: Synovial perfusion and synovial fluid solutes. Ann Rheum Dis 54: 424-428, 1995. 64. LevickJR, MCDonaldJN: Fluid movement across synovium in healthy joints: role of synovial fluid macromolecules. Ann Rheum Dis 54: 417-423, 1995. 65, Cho MH, Neuhaus OW: Absence of blood cloning substances from synovial fluid. Thromb Diath Haemorrh 5: 496, 1960. 66. Ropes MW, Bauer W: Synovial Fluid Changes in Joint Disease. Cambridge, MA, Harvard University Press, 1953, 67. Ropes MW, Muller AF, Bauer W: The entrance of glucose and other sugars into joints. Arthritis Rheum 3: 496--514. 1960. 68. Hamerman 0, Schuster H: Hyaluronate in normal human synovial fluid. .1 CUn Invest 37: 57-64, 1958. 69. Pekin TJ, Zvaifler NJ: Hemolytic complement in synovial fluid. .1 Clin Invest 43: 1372-1382, 1964. 70. Cummings NA, Nordby GL: Measurement of synovial fluid pH in normal and arthritic knees. Arthritis Rheum 9: 47-56, 1966.
18
C HAP T E R
2
STRUCTURE AND FUNCTION
71. Kushner I, Somerville JA: Permeability of human synovial membrane to plasma proteins: relationship to molecular size and inflammation. Arthritis Rheum 14: 560-570, 1971. 72. Hunder GG, Gleich GJ: Immunoglobulin E (JgE) levels in serum and synovial fluid in rheumatoid arthritis. Arthritis Rheum 1974; 17: 955-963. 73 Rose NR, de Marcario EC, Fahey lL, et al (eds): Manual of Clinical Laboratory Immunology, 4th ed. washington, DC, American Society for Microbiology, 1992. 74. Gatter RA, Schumacher HR: A Practical Handbook of Joint Fluid Analysis, 2nd ed. Philadelphia, Lea & Febiger, 1992. 75. Liew M, Dick We: The anatomy and physiology of blood flow in a diarthrodial joint. Clin Rheum Dis 7: 131-148, 1981. 76. CanosoJJ: Bursae, tendons and ligaments. Clin Rheum Dis 7: 189-221, 1981. 77. Niepel GA, Sit'aj S: Enthesopathy. Clin Rheum Dis 5: 857-872, 1979. 78. Canoso JJ: The premiere enthesis. J Rheumatol 25: 1254-1256, 1998. 79. Benjamin M, Kumai T, Milz S, et al: The skeletal attachment of tendons: tendon "entheses." Compar Biochem Physiol A 133: 931-945, 2002. 80. Goldman YE, Dantzig JA: Harris ED Jr, et al (eds): Keiley's Textbook of Rheumatology, 7th ed. Philadelphia, Elsevier, 2005, pp. 82-94. 81. Gowitzke BA, Milner M: Scientific Basis of Human Movement. Baitimore, Williams & Wilkins, 1988, p. 144. 82. Turner DC, Wallimann T, Eppenberger HM: A protein that binds specifically to the M-line of skeletal muscle Ls identified as the muscle form of creatine kinase. Proc Natl Acad Sci USA 70: 702-705, 1973.
83. Squire J: The Structural Basis of Muscular Contraction. New York, Plenum Press, 1981. 84. Kelly AM, Rubinstein NA: Development of neuromuscular specialization. Med Sci Sports Exerc 18: 292-298, 1986. 85. Faulkner JA, White TP: Adaptations of skeletal muscle to physical activity. In Bouchard C, Shephard RJ, Stephens T, et al (eds): Exercise, Fitness, and Health. Champaign, IL, Human Kinetics, 1990, p. 265. 86. Heffner RR .Ir (ed): Muscle Pathology. New York, Churchill Livingstone, 1984. 87. Stockdale FE: Mechanisms of formation of muscle fiber types. Cell Strucr Funct 22: 37-43, 1997. 88. Staron RS: Human skeletal muscle fiber types: delineation, development. and distribution. Can.l App! Physiol 22: 307-327, 1997. 89. Zhang M, Koishi K, Mclennan IS: Skeletal muscle fibre types: detection methods and embryonic detenninants. Histol Histopathol 13: 201-207, 1998. 90. Booth FW, Tseng BS, Fluck M, et al: Molecular and cellular adaptation of muscle in response to physical training. Acta Physiol Scand 162: 343-350. 1998. 91. Hargreaves M: 1997 Sir William Refshauge Lecture. Skeletal muscle glucose metabolism during exercise: implications for health ,lOd performance. .I Sci Med Sport 1: 195-202, 1998. 92. Taylor AW, Bachman L: The effects of endurance training on muscle fibre types and enzyme activities. Can.l Appl Physiol 24: 41-53, 1999.
3
( HAP T E R
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE Marco Gattomo and Alberto Martini
;If
The irrunune system, H the function of which is to protect against infections, comprises two branches: a more primitive one called innate (natural, native) immunity and a highly sophisticated one called adaptive (specific) immunity. Innate and adaptive immunity are not two separate compartments but an integrated system of host defenses, sharing bidirectional interactions fundamental to both the inductive phase and the effector phase of the immune response. The cells of the immune system originate from the pluripotent hematopoietic stem cells that give rise to stem cells of more limited potential (lymphoid and myeloid precursors) (Fig. 3-1). The immune system functions by means of a complex network of cellular interactions that involve cell surface proteins and soluble mediators such as cytokines.
INNATE IMMUNITY The principal components of innate immunity are (1) physical and chemical barriers such as epithelia and antimicrobial substances produced at epithelial surfaces, (2) circulating effectors proteins such as the complement components and cytokines, and (3) cells with innate phagocytic activity: neutrophils, macrophages, and natural klller (NK) cells. Phagocyte surface receptors recognize highly conserved structures characteristic of microbial pathogens that are not present in mammalian cells. The binding of microbial structures to these receptors triggers the cell to engulf the bacterium and induces cytokines, chemokines, and costimulators that recruit and activate antigen-specific lymphocytes and initiate adaptive irrunune responses. Thus, innate irrununity not only represents an early effective defense mechanism against infection but also provides the "warning" of the presence of an infection against which a subsequent adaptive immune response has to be mounted. 4 The pivotal role of this compartment in the effector phase of the irrunune response is discussed later. Innate immunity represents an effective first line of defense against infections. However, its capacity to recognize infectious agents is limited. Moreover, innate immunity lacks of one of the most important characteris-
tics of the adaptive immunity: the capacity to memorize and to respond more vigorously to repeated exposure to the same infectious agent.
Cells Involved in Innate Immunity Phagocytes The cells of the phagocyte system originate from a common lineage in the bone marrow, circulate in the blood in inactive form, and are recruited and activated in the peripheral tissues in case of infection, tissue injury, or other pro-inflammatory stimuli. Monocytes are the classic example of immature circulating phagocytes; they are characterized by a granular cytoplasm with many phagocytic vacuoles and lysosomes. Once they enter the tissues, monocytes mature into macrophages. Macrophages are present in all tissues, where they act as "sentinels" together with dendritic cells (Des). One of their major roles is to recognize and respond to microbes and to amplify the response against a potentially harmful stimulus. Depending on the tissues in which they are found, macrophages are known by a number of different names: Kupffer cells in the liver, microglial cells in the central nervous system, and alveolar macrophages in the airways. These cells are the prototype of the effector cells of innate immunity. Once activated, macrophages initiate a number of crucial events, which include phagocytosis and destruction of ingested microbes and production of pro-inflammatory cytokines and other mediators of inflammation, that lead to further recruitment of cells of innate immunity (monocytes, neutrophils) and provide signals to T and B cells of adaptive immunity. Neutrophils, the other major group of phagocytes, are the most abundant type of circulating leukocytes. The cytoplasm is characterized by the presence of two types of granules. The so-called specific granules contain a number of enzymes, such as lysozyme, elastase, and collagenase. The azurophilic granules are lysosomes containing enzymes and microbicidal substances. Neutrophils are the first cells that enter the site of infection, and they represent the prevalent cell type in the early phases of
19
20
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
• Figure J-l Lineage of cells involved in the immune and inflammatory responses. Pluripotent hemapoietic stem cell (HSC) give rise to precursors for different hematopoietic lineages. The first step of HSC commitment generates common myeloid and common lymphoid progenitors. Most of the dendritic cells (DC) appear to be related to the mononuclear phagocyte lineage. Asubset of them is thought to be of lymphoid origin (plasmacytoid DC or interferon-produdng cells, IPC) (see also ref. 11). Development of natural killer (NK) cells is still under investigation; direct origin from HSC is assumed (see also ref. 18).
the inflammatory response. Within 1 or 2 days, neutrophils are almost completely replaced by newly recruited monocyte-macrophages, which represent the dominant effector cells in the later stages of inflammation.
Phagocyte Activation and Effector Functions Neutrophils and macrophages can be activated in several ways. The presence on their surface of multiple receptors
allows them to act as initiators as well as final effectors of the immune response (Fig. 3-2). These include receptors for components of infectious agents as well as receptors for molecules of the immune system (e.g., complement, cytoldnes, chemokines, immunoglobulins). As mentioned earlier, phagocyte surface receptors recognize structures, including nucleic acids and complex lipids and carbohydrates, that are characteristic of microbial pathogens but are not present on mammalian cells' (Table 3-1), Most of these substances are essential for the survival of invading
• Figure 3-2 Modalities of macrophage activation. A, Recognition of conserved molecular constituents of microbes (in the figure, lipopolysaccharide (LPSI) by spedfic receptors (I.e.,Toll-like receptors, mannose receptor, scavenger receptor). 8,T cell-mediated activation via interferon-y (IFN-y) and CD40CD40-ligand (CD40L) interaction. e, Recognition of antibodies, immune complexes, and complement by the membrane receptors for the Fe fragment of immunoglobulins and complement receptors. The main effector soluble mediators produced after macrophage activation are also shown.
C HAP T E R
._'i
lIIi
IABLE. 3-1
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
E:xalllple~ 01 Palhogen-As~ocialed
LPS Unmethylated DNA with CpG motifs Terminal mannose residues LPS,dsRNA dsRNA ssRNA N-formylmethionine residues
Molecular Pallerns (PAMPs) and Respective Pattern Recognition
21
Re(eplor~ (PRR~)
Origin
PIR
Main Effector Function
Gram-negative bacteria Bacterial DNA
Toll-like receptors, CD14 Toll-like receptors
Microbial glycoprotein and glycolipids
Macrophage mannose receptor Plasma mannose-binding lectin
Bacteria, viruses Viral Viral Bacteria
Macrophage scavenger receptor Toll-like receptors Toll-like receptors Chemokine receptors
Macrophage activation Macrophage, B cell, and plasmacytoid cell activation Phagocytosis Complement activation, opsonization Phagocytosis IFN type I production IFN type I production Neutrophil and macrophage activation and migration
dsRNA, double-stranded RNA; IFN, interferon; LPS, lipopolysaccharide: ssRNA, single-stranded RNA.
microbes and are defined as pathogen-associated molecular patterns (PAMPs), The receptors of the innate immune system that recognize PAMPs are called patternrecognition receptors (PRRs). PAMPs may significantly differ among the various classes of microorganisms, but they ;Ire invariant among microorganisms of a given class. This characteristic allows a limited number of germ-line encoded PRRs to detect the presence of different microbial infections. Because PAMPs are essential for microbial survival, mutations or deletions of PAMPs are lethal. This greatly reduces the possibility that microbes could escape recognition by the innate system by undergoing PAMP mutations.
• fIIIn 3-3 Structure ofTolI-like receptors (TlR) and signaling pathway. Activation ofTlR4: lipopolysaccharide (lPS) binds to the CD14 molec.ule present on phagocyte surface and to a circulating protein, called M02. Then, the lPS-CDI4 complex associates with TlR4 for subsequent intracellular signaling. The lipopolysaccharidebinding protein (lPB) is a circulating protein that binds to lPS in the blood or in ~racellular flUid, forming a complex that facilitates lPS binding to CD14.TlR2 binds to peptidoglycan and forms an heterodimer with TlR6 through MAlP-2. After activation, the intraceliularTolVll-l receptor (TIR) domain triggers the signaling pathway that results in generation of NF-lCB (nudear factor-lCB) transcription factor and activation of activation protein-l (AP-l). IRAK,ll-l receptor-associated kinase; TRAF, tumor necrosis factor receptor-associated factor.
Many classes of PRRs are present on the surface of macrophages and Des, where they act as "tissue sentinels" through the continuous monitoring of peripheral tissues for the possible invasion of microbial pathogens (see Table 3-1). The Toll-like receptors (TLRs) are a large class of PRRs characterized by an extracellular leucinerich repeat (LRR) domain and an intracellular Toll/interleukin-1 CIL-1) receptor (TIR) domain (Fig. 3-3). There are at least 10 different classes of TIRs involved in responses to widely divergent types of PAMPs (Table 3_2).5,6 In some cases, recognition of PAMPs and the consequent transmission of intracellular signals for phagocyte activation require the presence of accessory proteins
22
II I
C HAP T E R
1ABLE 3 l
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
loll-I ikl' Rl'( l'plIUS (fI Rs)
TLR
Ugand
Microbial Source
TLRI
Lipopeptides Atypical LPS Zymosan Peptidoglycans Lipotelchoic acids Lipoarabinomannan HSP70 (host) dsRNA LPS Lipoprotein HSP60 Fusion protein Flagellin Lipoprotein MALP-2 ssRNA ssRNA DNA with CpG motifs Unknown
Bacteria, mycobacteria Leptospira interrogans Fungi Gram-positive bacteria Gram-positive bacteria Mycobacteria
TLR2
TLR3 TLR4
TLR5 TLR6
TLR7 TLR8
TLR9 TLRlO
Vinlses Gram-negative bacteria Many pathogens Chlamydia pneumoniae Respiratory syncytial vinls Bacteria Mycoplasma Viruses Viruses Bacteria, protozoans
dsRNA, double-stranded RNA; HSP, heat-shock protein; LPS, lipopolysaccharide; MALP-2, mycoplasma-derived macrophage-activating 2-KDa lipopeptide; RSV, respiratory syncytial virus; ssRNA, single-stranded RNA.
(see Fig, 3-3). It is still debated whether TLRs may also recognize endogenous ligands, such as heat-shock proteins produced during stressing events, or extracellular matrix components, including fibronectin, hyaluronic acid, and heparin sulphate, produced in response to tissue injury (see Table 3-2).6,7
The binding of microbes to phagocytes through PRRs initiates the process of phagocytosis of the invading microorganism and its subsequent killing in phagolysosomes. In fact, activation of phagocytes through PRRs induces multiple effector molecules such as inducible nitric oxide synthase (NOS) and other antimicrobial peptides that can directly destroy microbial pathogens (Fig. 3-4). At the same time, peptides are generated from microbial proteins and presented to T cells to initiate the adaptive immune response (see later discussion), Moreover, the activation of signal transduction pathways by PRRs leads to the induction of genes that regulate both innate responses (inflammatory cytokines, growth factors, chemokines, proteolytic enzymes) and adaptive responses (major histocompatibility complex, costimulatory molecules) of the host to invading microbes. Other modes of macrophage activation include stimulation by type 1 T helper cell (ThO-oriented lymphocytes through the production of interferon-y (IFN-y) (see later discussion). Macrophages and neutrophils are also activated by immune complexes and complement fragments through binding with immunoglobulin and complement receptors expressed on their surface (see Fig. 3-2), This is the primary pathway of activation of neutrophils, which represent the first circulating leukocytes recmited at the site of inflammation.
Alternative Phagocyte Activation Macrophages playa cmcial role in the clearance of apoptotic cells, a function that is essential in normal home-
• Figure 3-4
Phagocytosis and intracellular destruction of microbes. A, The pathogen-associated molecular pattem present on a microbe is recognized by the pattem-recognition receptor expressed on the phagocyte membrane. 8, The microbe is intemalized into a phagosome. C, The phagosome fuses with cytoplasm Iysosomes to form a phagolysosome, in which the microbe is killed by reactive oxygen intermediates (ROI) and nitric oxide (NO).
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
ostasis. During the process of phagocytosis of apoptotic cells, tissue macrophages undergo alternative activation, S stimulated by the binding of apoptotic cells to surface receptors, such as ~J-glycoprotein 1 receptor and the vitronectin receptor (avp/ The alternative activation of macrophages leads to the production of a number of anti-inflammatory molecules, including transforming growth factor-~ (TGF-~), IL-1O, and IL-l receptor antagonist (IL-IRa), that are crucial components in the suppression of acute inflammation and protect the host from tissue damage. 9 IL-lO inhibits production of pro-inflammatory cytokines, suppresses the release of reactive oxygen intermediates, and downregulates the expression of major histocompatibility complex (MHC) class II and costimulatory molecules. IL-IRa binds competitively to the IL-l receptor present on macrophages, thus inhibiting the pro-inflammatory action of IL-1. TGF-~ inhibits the production of another pro-inflammatory cytokine, tumor necrosis factor (TNF), and promotes the expression of matrix proteins by macrophages, contributing to extracellular matrix deposition, wound healing, and fibrosis. The alternative activation of macrophages is also stimulated by Th2 cytokines, IL-4, and IL-13 (see later discussion).
Dendritic Cells DCs are specialized antigen-presenting cells that originate from the bone marrow and playa critical role in the processing and presentation of antigen to T cells during the adaptive immune response. lO ,ll DCs should be considered as a bridge between innate and adaptive immunity. At their immature stage of development, DCs act as sentinels in the epithelia of peripheral tissues (skin, gastrointestinal, and respiratory systems), continuously sampling the antigenic environment. These cells are morphologically identified by their extensive membrane projections. Their prototypes are the Langerhans cells of the epidermis. Recognition of microbial or viral products through the same PRRs that are present on the surface of phagocytes initiates the migration of DCs to lymph nodes, where they mature (express costimulatory molecules) to present antigen to T cells. 12 ,13 The role of DCs in antigen presentation and in the regulation of the immune response is described in more detail in the discussion of the adaptive immune response. Recently, another type of DC (plasmacytoid DC) of probable lymphoid origin was identified. 14 Differences in function between myeloid DCs and plasmacytoid DCs are still unclear. Plasmacytoid DCs have the unique capacity to secrete large amounts of type I IFN (aI~) in response to certain viruses and other microbial stimuli (they are also called plasmacytoid interferon-producing cells). It was shown that monocytes isolated from the blood of patients with systemic lupus erythematosus (5LE), but not those from healthy individuals, act as DCs and that their activation is driven by circulating IFN-a, which may come from plasmacytoid DCS.15 This and other experimental and clinical observations point to a possible pivotal role for this cell subpopulation in the pathogenesis of some autoimmune disorders. 16 Although DCs are the most potent APCs able to activate naIve T cells, it is now clear that DCs may also be
23
tolerogenic through various mechanisms that include induction of T cell anergy, T cell apoptosis, T regulatory cells and production of immunoregulatory cytokines by T cells (see later discussion). This tolerogenic phenotype is displayed by myeloid-derived immature DCs and by immature and mature plasmacytoid DCs. The induction of tolerance through DC manipulation is an exciting possibility for future treatments in autoimmune diseases and transplantation. 17
Natural Killer Cells NK cells are large lymphocytes characterized by the presence of numerous cytoplasmic granules containing proteolytic proteins (perforin, granzymes). They lack antigen-specific receptors (e.g., immunoglobulins, T cell receptors) but are able to kill abnormal cells such as some tumor cells and virus-infected cells. IS The term "natural killer" refers to their capacity to rapidly kill target cells without the need of activation by other cell types or soluble molecules (as is the case for CD8+ cytotoxic T lymphocytes [CTLs]). Activation of NK cells is regulated through activating and inhibitory cell surface receptors. 19 The inhibitory receptors bind to self class I MHC molecules, which are expressed on most cell types (Fig. 3-5A). The ligands for activating receptors are only partially known. The engagement of both inhibitory and activating receptors results in a dominant effect of the inhibitory receptors. The infection of host cells, especially by viruses, leads to the loss of class I MHC from their surfaces and exposes these cells to the exclusive activity of activating receptors (see Fig. 3-5A,B),20 Once activated, NK cells release the contents of their granules. Perforin creates pores in target cell membranes, and granzymes enter into the cells through the perforin pores, inducing the death of target cells by apoptosis, with the same mechanism of cytolysis used by CDW cns (see later discussion). Other important activities of NK cells include their ability to recognize (via Fc receptors) and destroy antibody-coated cells (a process called antibody-dependent cell-mediated cytotoxicity, or ADCC) and their ability to produce high amounts of IFN-y, a potent stimulator of macrophage activity. In turn, activated macrophages produce IL-12, a potent inducer of NK-cell IFN-y production and cytolytic activity (see Fig. 3-5C,D).
Fibroblasts Fibroblasts play an active role in the effector arm of the inflammatory response. Connective tissue contains a mixture of distinct fibroblast lineages, including mature fibroblasts with a lesser capacity of transformation and immature fibroblasts (mesenchymal fibroblasts) that are capable of differentiating into several different cell lineages. Fibroblast precursors with a multipotent character also circulate in blood; because of their similarity with stromal cells of bone marrow, they are called mesenchymal stem cells. During inflammation, the pro-inflammatory cytokines produced by tissue macrophages activate mature tissue fibroblasts to produce cytokines, chemokines, prostaglandins (PGE 2), and proteolytic enzymes (Le.,
24
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
• Figure 3-5 Functional properties of natural killer (NK) cells. A, The inhibitory receptors bind to self class I major histocompatibility complex (MHO molecules, which are normally expressed by most cell types. 8, Infection of the host cells by virus leads to the loss of surface class I MHC and exposes the infected cells to the exclusive activity of activating receptors and subsequent killing. C, NK cells recognize antibodycoated cells through the Fc receptors expressed on their surface and subsequently kill the infected cell. D, NK cells respond to interleukin-12 (IL-12) produced by macrophages during phagocytosis and secrete interferon-y (IFN-y), which, in tum, activates macrophage to kill phagocytosed microbes.
metalloproteinases). The failure to switch off activated tissue fibroblasts has been proposed as a mechanism leading to chronic inflammation, through the persistent overexpression of chemokine and pro-inflammatory cytokines and consequent continuous recruitment of leukocytes. 21
Molecules of Innate Immunity The Complement System The complement system consists of several normally inactive plasma proteins that, after activation, interact to generate a number of products that mediate important effector functions, including promotion of phagocytosis and lysis of the cells on which complement is activated and stimulation of inflammation. Activation of complement involves sequential proteolytic steps that generate an enzymatic cascade similar to that of the coagulation system. 22 There are three major pathways of complement activation (Fig. 3-6). The alternative pathway involves the direct binding of one of the complement proteins, C3b, to cells. The classic pathway involves a more sophisticated mode of activation, in which Cl binds to the CH 2 domains of immunoglobulin G CIgG) or to CH 3 domains of IgM that have bound antigen (see later discussion). The same proteins involved in the classic pathway can be activated in the absence of antibodies by a plasma protein (mannose-binding lectin) that binds to mannose residues on microbial glycoproteins and glycolipids; this
is known as the lectin pathway. The three pathways of complement activation converge in a central complement component, C3, which is cleaved into two fragments. The larger fragment (C3b) becomes covalently attached to cells, where it acts as opsonin to stimulate phagocytosis and activates C5 with subsequent generation of C5b. C5b initiates the formation of a complex of the complement proteins C6, C7, C8, and C9 that is assembled on cell membranes (membrane attack complex, MAC), forming a pore that causes lysis of the target cell. During complement activation, smaller complement fragments (C3a, C4a, C5a) are released into the circulation. Also known as anaphylatoxins, they exert several pro-inflammatory effects, including activation of mast cells and neutrophils and increased vascular permeability!2 Another function of complement is to bind to antigen-antibody complexes, thus promoting their solubilization and clearance by phagocytes. Recently, complement has been shown to have a role in the clearance of apoptotic blebs by the phagocytic system. 23 ,24 The biologic activities of complement are mediated by the binding of complement fragments to membrane receptors. Receptors for the fragments of C3 are best characterized (Table 3-3). Type 1 complement receptor (CRl, CD35) is expressed by almost all blood cells and promotes phagocytosis of C3b-coated cells. CRI expressed on erythrocytes binds to circulating immune complexes with attached C3b. In this way, circulating erythrocytes are able to transport immune complexes to the liver and spleen, where they are removed from erythrocyte surface and cleared. Type 2 complement receptor (CR2, CD2l) is
C HAP T E R
3
THE 1M M U N E
S Y S T E MAN D
THE IN F LAM MAT 0 RY RES P 0 N S E
25
• R.-. 3-6
The complement cascade. The alternative pathway is activated when Ob binds to the microbial cell wall, after spontaneous cleavage of free circulating 0. Thereafter, Ob binds to factor B, forming a ObBb convertase. The dassic pathway is Initiated by the binding of the tril1ilolecular complex Cl (Clq, Clr, C1s) to antigen-antibody compleKes. The same proteins involved in the classic pathway can be activated in the absence of antibodies by a plasma protein called mannose-binding lectin (MBL), which binds to mannose residues on microbial glycoproteins and glycolipids (lectin pathway). Subsequent binding with the C4b and C2a subunits leads to the formation of C4b2a convertase. The three pathways of complement activation converge at 0, which is cleaved into two fragments. The larger fragment (Ob) activates CS with subsequent generation of CSb; CSb initiates the formation of a complex of the complement proteins C6, C7, CS, and C9 that is assembled on the cell membrane (membrane attack complex,
MAO.
present on B lymphocytes and follicular DCs of lymph node germinal centers. Its main function is to act as coreceptor for B cell activation by antigen (see later discussion) and to stimulate the trapping of antigen-antibody • ~. fABLE 3 3
(omplement Re(eptors
Receptor
Cell 'JYpes
Ugands
Function
CRl (C035)
Band T cells Erythrocytes Monocytes, macrophages Eosinophils FDCs, Neutrophils B cells FDCs Upper airways epithelium
C3b, C4b, iC3b
C3b and C4b decay Clearance of immune complexes Phagocytosis
C3d, C3dg, iC3b
Activation of B cells (coreceptor) Antigen presentation in germinal centers Receptor for EBV Phagocytosis Adhesion to endothelium (via ICAM)
CR2 (CD21)
CR3 (CD llb/ CD1S)
CR4 (COllc/ CDlS)
Macrophages Neutrophils. NK cells Dendritic cells, FDCs Macrophages Neutrophils, NK cells Dendritic cells
iC3b,ICAM
iC3b
Phagocytosis
CD, cluster of differentiation; CR. complement receptor; EBV, Epstein-Barr virus; FOC. follicular dendritic cells; lCAM. intracellular adhesion molecule: NK. natural killer.
complexes in germinal centers. Type 3 and type 4 complement receptors are members of the integrin family and are expressed by the cells of innate immunity (neutrophils, NK cells, mononuclear phagocytes), The binding of CR3 or CR4 promotes the activation of these cells and the phagocytosis of cells opsonized with C3h. Complement deficiencies are associated with a number of pathologic conditions (see Chapter 33). Genetic deficiencies of classic pathway components (Clq, Clr, C2, and C4) may cause a disease that resembles SLE,24 This may be related to the role of early complement components in the clearance of apoptotic cells and circulating immune complexes. The deficiency of C3 is associated with serious pyogenic infections. Defects of the terminal complement components (CS-C9) are associated with an increased risk of disseminated Neisseria infections, including Neisseria meningitidis. Activation of the complement cascade is regulated by a number of circulating and cell membrane proteins that prevent activation on normal host cells and limit the duration of complement activation on microbial cells and antigen-antibody complexes. The CI inhibitor (CI INH) regulates the proteolytic activity of CI, the initiator of the classic pathway of complement activation. Deficiency of this protein causes an autosomal dominant inherited disease called hereditary angioneurotic edema. A number of membrane proteins (membrane cofactor protein, MCPj type 1 complement receptor, CRl; decay-accelerating factor, DAF) and the plasma protein, factor H, prevent the activation of C3b if it is deposited on the surfaces of normal mammalian cells, The deficiency of an enzyme required for linkage of DAF with the cell membrane causes a disease called paroxysmal nocturnal hemoglobinuria, which is characterized by recurrent attack of intravascular hemolysis due to unregulated complement activation on the surface of erythrocytes. Similarly, the rare deficiency of factor H is characterized by an
26
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
excess alternative pathway activation leading to C3 consumption and glomerulonephritis, Recent experimental observations have shown a role for complement components in the induction of regulatory T cells secreting anti-inflammatory cytokines such as TGF-~ and IL10,25,26 These data suggest a broader role of the complement system in the regulation of the immune response.
Other Circulating Proteins A number of circulating proteins behave as secreted PRRs (see earlier discussion) in their ability to specifically recognize microbial PAMPs and promote innate immunity, Mannose-binding lectin (MBL) belongs to a family of proteins (collectin) with a collagen-like domain separated by a neck region from a calcium-dependent (C-· type) lectin. 27 Like the mannose receptor expressed on the surface of macrophages, MBL binds carbohydrates with terminal mannose and fucose that are typically found on glycoproteins on the surface of bacterial but not mammalian cells. MBL is structurally similar to the C1q component of the complement system, binds the C1q receptor present on phagocytes, and may activate complement. Thus, MBL is able to opsonize microbes and induce phagocyte activation via C1q receptor. C-reactive protein (CRP) and serum amyloid protein (SAP) are plasma proteins belonging to the family of pentraxins. 28 They are abundantly produced during the acute phase of inflammation by the liver. They bind to phosphorylcholine present on the microbial membranes. Moreover, they are also able to activate the classic complement pathway and to act as opsonins for neutrophils. The lipopolysaccharide-binding protein (LPB) is a circulating protein that binds to LPS in the blood or extracellular fluid, forming a complex that facilitates LPS binding to CD14 (see earlier discussion). Other circulating proteins that participate in innate immunity include defensins, which are diverse members of a large family of antimicrobial peptides that contribute to the antimicrobial action of granulocytes, mucosal host defense in the small intestine. and epithelial host defense in the skin and elsewhere. 29
Cytoklnes and Chemoklnes Cytokines and chemokines are proteins secreted by cells of the innate and adaptive immune systems in response to a possible harmful exogenous stimulus (microbes, antigen) that mediate and regulate the immune and inflammatory response. Although some cytokines are produced in sufficient quantity to circulate and exert exocrine actions, they usually act locally in an autocrine or paracrine fashion. Their functions are mediated by cellular receptors belonging to a limited number of families (Table 3-4), All cytokine receptors consist of one or more transmembrane proteins whose extracellular portions are responsible for cytokine binding and whose cytoplasmic portions mediate the triggering of the intracellular signaling pathway (see Fig. 3-4). Cytokines may be classified in four main categories: (1) cytokines of innate immunity, which are produced mainly by mononuclear phagocytes in response to an infectious agent (Table 3-5); (2) cytokines of adaptive immunity, which are produced by T lymphocytes after specific recognition of foreign antigens (Table 3-6); (3) growth factors, which are produced by bone marrow stromal cells and leukocytes, stimulate the differentiation and proliferation of immature leukocytes, and sustain phenomena of angiogenesis; and (4) chemokines, a large family of cytokines produced by various cell types, which stimulate and regulate leukocyte migration (see also Table 3-12). Cytokines are the main functional molecular bridge between innate and adaptive immunity, and they are potent effectors in both systems, The functional characteristics of cytokines of innate immunity are outlined in more detail later in this chapter; the cytokines of adaptive immunity are described in the following section.
ADAPTIVE IMMUNITY The main characteristics of adaptive immunity are the capacity to recognize antigens with a high level of molecular specificity and the ability to remember and respond more promptly and more vigorously to subsequent exposure to the same antigens (immunologic memory). Any
Family
Structure
Spedfldty
Type I cytokines receptor (hematopoietin family)
Conserved cysteine residues + WSXWS sequence Common ~ chain Common y chain Common gpI30 subunit Conserved cysteine residues, no WSXWS sequence Conserved cysteine-rich extracellular domains Extracellular immunoglobulin domains Seven a-helical transmembrane domains
Erythropoietin, growth hormone, IL-13
Type II cytokines receptor TNF-receptor family Immunoglobulin superfamily receptors Chemokine receptors
IL-3, IL-S, GM-CSF IL-2, IL-IS, IL-7, IL-4, IL-9 IL-6, IL-ll IFN-a, -~, -y, IL-lO TNF, LT, C040, Fas, C027, NGF IL-I, M-CSF, stem cell factors Chemokines
CD. cluster of differentiation; GM-CSF, granulocyte-macrophage colony-stimulating factor; IFN. interferon; IL, interleukin; LT, Iymphotoxin; M-CSF. macrophage colony-stimulating factor; NGF, nerve growth factor; TNF, tumor necrosis factor.
3
C HAP T E R
'-
,11 •
rABLE 3- 5
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
(ytokines of Innate Immunity
CytGIcIne
Size and Form
Recepton
Main Cell Source
Main Biologic Effects
IL-Hx IL-I~
17 kD, monomer 33 kD (precursors)
CD121a, b (IL-l Rl, IL-l RlD
Macrophages, endothelial cells, epithelial cells
TNF-o.
17 kD homotrimer
p55, p75 (TNFRl, TNFRlI)
Macrophages, T cells, NK cells
Homotrimer
p55, p75 (TNFRl, TNFRII) ?
T cells, B cells
Fever Activation of endothelial cells and macrophage Acute phase reactants Fever Activation of endothelial cells, macrophage, and neutrophils Acute phase reactants Apoptosis Cachexia Killing, endothelial activation
CDl18 (IFNAR2)
Leukocytes Fibroblasts
IL-6R, gp130
T cells, macrophages, endothelial cells
IL-lO Ro. CRF2-4 IL-12 R~l IL-12 R~2 IL-15 R (CDI22)
Macrophages, T cells (Th2, Treg) DC, B cells Macrophages Macrophages and other non T cells Macrophage
TNP-~
(LT-o.)
27
MIF
Monomer
Type I IFNs IL-6
IFN-o.: 15-21 kD IFN-~: 20-25 kD monomers 19-26 kD, monomer
lL-lO
34-40 kD homodimer
IL-12 IL-15
Heterodimer of 35 and 40 kD· 13 kD, monomer
IL-18
17 kD, monomer
IL-23
Heterodimer of 19 and 40 kD·
T cells
IL-l Rrp (0. chain) AcPL (~ chain) IL-12 R~1 IL-23R
DC, B cells Macrophages
Macrophage activation Steroid resistance Antiviral response Activation of NK cells Fever Activation of endothelial cells Acute phase reactants B cell proliferation Suppression of macrophage function Differentiation of Th 1 cells Synthesis of IFN-y by T cells and NK cells NK cells and T cell proliferation Synthesis of IFN-y by T cells and NK cells Sustain survival and function of Thl cells
., IL-ll and IL-23 share the same p40 subunit. AcPL, accessory protein-like; CD, cluster of differentiation; CRF, Class" cytokine receptor family; lPN, interferon; IL, interleukin; LT, lymphotoxin; MIF, macrophage inhibitory factor; NK, natural killer; TNF, tumor necrosis factor; R, receptor; DC: dendritic cells; Thl, type I helper T cell; Th2, type 2 helper T cell; Treg, regulatory Teell .
• ~. IABLE 3--6
Cytokines of Adaptive Immunity
Cytaldne
Size and Fonn
Recepton
Main Cell Source
Main Biologic Effects
IL-2
14-17 kD, monomer
T cells
IL-4
18 kD, monomer
CD25 (0. chain) CD122 (~ chain) CD132 (y chain) CD124
IL-5
45 kD, homodimer
CD125
T cells (Th2)
IL-13
15 kD, monomer
IL-13 R
T cells (Th2)
IFN-y
50 kD, homodimer
CD119 (IFNGR2)
T cells, NK cells
LT-P TGF-~
Trimerizes with LT-o. 25 kD, homodimer
Proliferation and activation of T cells, NK cells Proliferation of B cells and antibody synthesis Fas-mediated apoptosis Isotype switching to IgE Th2 differentiation Inhibition of IFN-y-mediated macrophage activation Thl suppression Activation and proliferation of eosinophils B cell proliferation and 19A production B cell proliferation Isotype switching to IgE Inhibition of macrophage activation Macrophage activation Ig class switching to opsonizing and complement-fixing IgG antibodies Thl differentiation Th2 suppression Lymph node development Inhibition of proliferation and effector function of T cells Inhibition of B cell proliferation
T cells (Th2)
T cells, B cells T cells (Treg), macrophages
CD, cluster of differentiation; IFN, interferon; Ig, immunoglobulin; IL, interleukin; LT, lymphotoxin; NK, natural killer; TGF, transforming growth factor; Thl, type 1 helper T cells; Th2, type 2 helper T cells; Treg, regulatory T cells.
28
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
substance that is capable of being recognized by the adaptive immune system is called an antigen. Lymphocytes, the cells responsible for adaptive immunity, are the only cells capable of recognizing different soluble or membrane-bound antigenic determinants by specific antigen receptors. The adaptive immune response can be divided into three phases. The recognition phase consists of the binding of antigens to specific lymphocyte receptors. The activation phase is the sequence of events induced in lymphocytes after specific antigen recognition. Lymphocytes first proliferate and then differentiate into either effector cells (which are aimed at eliminating the antigen), or memory cells (which survive and are ready to respond promptly to antigen re-exposure). In the effector phase, activated lymphocytes perform their function of eliminating the antigen. Many effector functions require the participation of molecules or cells that belong to the innate immune system. For instance, some antibodies activate complement, whereas T lymphocytes release cytokines that stimulate the function of phagocytes and the inflammatory response. The immune system is able to recognize and eliminate a foreign antigen but does not normally react harmfully to self molecules (self-tolerance) . Lymphocytes display common morphologic features but differ in their origin, in how they recognize antigens, and in their functions. Lymphocytes that have not encountered antigen (nai've lymphocytes) are small (8 to 10 /lm in diameter), with a large nucleus and a small amount of cytoplasm containing mitochondria and ribosomes but no specialized organelles. After antigenic stimulation, naIve lymphocytes enter into the cell cycle and become larger 00 to 12 /lm) and have more cytoplasm and organelles (lymphoblasts). There are two main types of lymphocytes: B lymphocytes (which mature in the bone marrow) and T lymphocytes (which mature in the thymus). B lymphocytes produce antibodies and are responsible for humoral immunity. Their antigen receptors are membrane-bound antibodies that can recognize soluble antigens. Cell activation leads to a sequence of events that culminates in the generation of plasma cells, which secrete antibodies. T lymphocytes are responsible for cell-mediated immunity, The T cell receptor (TCR) recognizes cell surfaceassociated antigens, but not soluble antigens. Specifically, the TCR recognizes peptide antigens that are generated inside antigen-presenting cells (APCs) and are brought to the cell surface in association with specialized molecules of the MHC. This allows the immune system to detect intracellular pathogens. There are two functionally distinct populations of circulating T lymphocytes: helper T lymphocytes and cytotoxic T lymphocytes (CTLs). Helper T lymphocytes play the pivotal role in adaptive immunity and activate other cells, such as B lymphocytes, macrophages, and cytotoxic T cells, to perform their effector functions. CTLs lyse cells that are infected by virus or other intracellular microorganisms. A distinctive subpopulation of circulating T lymphocytes with a suppressor activity (defined as naturally occurring regulatory T cells [TregJ) has also been characterized (see later discussion).
Some antigen-stimulated Band T lymphocytes differentiate into memory cells, whose function is to mediate a rapid and enhanced response to a subsequent exposure to the specific antigen. These cells survive in a quiescent state for many years, not requiring antigen recognition for their prolonged survival in vivo. NaIve lymphocytes migrate through the peripheral (or secondary) lymphoid organs, where they recognize and are activated by antigens that are introduced through the skin and via the gastrointestinal and respiratory tracts, These effector and memory lymphocytes circulate in the blood and localize in peripheral sites of antigen entry, where they exert their effector functions. Different subsets of lymphocytes express different membrane proteins that identify their particular stage of differentiation and function. The membrane proteins are defined by means of specific monoclonal antibodies and are designated by the abbreviation CD (cluster of differentiation). Those membrane proteins mentioned in this chapter are listed in Table 3-7.
B Lrmphocytes and Their Antigen Receptors B lymphocytes develop in the bone marrow and are found mainly in secondary lymphoid organs and, in low number, in the circulation. B lymphocytes express cell surface immunoglobulins that act as antigen receptors.
Immunoglobulin Molecules Antibodies, or immunoglobulins, are the antigen-specific products of B cells. Immunoglobulins (1) serve as membrane-bound B-cell antigen receptors (see later discussion), (2) are produced and released by antigen-activated B cells, 0) bind their specific antigen, and (4) recruit other molecules or cells to destroy the target to which they are bound. The immunoglobulin variable region (antigen-binding fragment, or Fab) binds to the antigen; the constant region (crystallizable fragment, or Fc) binds to a limited number of effector molecules and cells. The immunoglobulin structure is substantially the same for all five main immunoglobulin classes or isotypes (IgM, IgD, IgG, IgA, and IgE).30 Antibodies are Y-shaped molecules composed of two identical heavy (H) chains (each 50 or 70 kD) and two identical light (L) chains (each about 25 kD). In all antibodies, there are only two types of functionally eqUivalent light chains; they are called lambda 0..) and kappa (K) chains. The light and heavy chains each have an aminoterminal sequence (variable region, or V region) that varies greatly among different antibodies; in each antibody, the variable regions of the light and heavy chains (VL and VH, respectively) form pairs to generate two identical antigen-binding sites that lie at the ends of the arms of the Y. The carboxylterminal sequences, on the other hand, are constant among immunoglobulin chains of the same heavy-chain isotype (Fig. 3-7), The heavy chains define the isotypes of immunoglobulins and are designated by the Greek letters /l, 0, 1.., E, and n. IgM and IgE heavy chains contain an extra constant-region domain. IgM molecules occur as pentamers; IgA is found in mucous secretions but not in plasma and
,-
I' •
1ABLE 3 ~ 7
Some Examples of Differentiation Antigens'
CD AntIgen Common Names
Main Cellular Expression
Known or Supposed Functions
CDl (a,b,c,d)
Thymocytes, dendritic cells
CO2
T cells, NK cells T cells, thymocytes
MHC class I-like molecule, presentation of nonpeptide antigens to some T cells Adhesion molecule (binds LFA-3) Associated with the T-cell receptor and involved in signal transduction Coreceptor for MHC class II molecules Signaling molecule Coreceptor for MHC class I molecules Subunit of LFA-l (adhesion molecule) Subunit of LFA-l (adhesion molecule) Role in B cell activation Role in B cell activation Binds to CD70, mediates costimulatory signals for T cell and B cell activation Receptor for costimulatory molecules B7-1, B7-2 Adhesion molecule (ligand for L-selectin)
CD3
CD40
Helper T cells T cells, B cell subset Cytotoxic T cells Leukocytes Leukocytes B cells B cells T cells, memory B cells, NK cells T cell subsets Precursors of hematopoietic cells, endothelial cells in high endothelial venules B cells, macrophages, DCs
CD44 CD45
Leukocytes Leukocytes
CD80
APCs
CD86
APCs
CD95 CDlS2
Multiple cell types Activated T cells
CD154
Activated CD4+ T cells
CD4 CDS CDlB CDlla CD18 CD19 CD21 CD27 CD28 CD3 4
Tyrosine phosphatase, role in activation of B cells, macrophage, and DCs induced by T cells; binds CD40L (CDl54) Mediates adhesion of leukocytes Tyrosine phosphatase, role in signal transduction Costimulator for T cell activation; ligand for CD28 and CTLA-4 Costimulator for T cell activation; ligand for CD28 and CTLA-4 Role in activation-induced apoptosis; binds FasL Inhibitory signaling to T cells; binds to CD80 and CD86 on APCs Ligand for CD40
Other Common Names
LFA-3
LFA-l a chain LFA-l ~ chain CR-2
Leukocyte common antigen B7-1 B7-2 Fas CTLA-4
CD40L
'Defined by means of monoclonal antibodies and designated by the abbreviation CD (cluster of differentiation) followed by a numbet. APCs. antigen-presenting cells; CfLA, cytotoxic lymphocyte-associated antigen; CR, complement receptor; DCs, dendritic cells; L, ligand; LFA, leukocyte functionassociated antigen; MHC, major histocompatibility complex; NK, natural killer.
• figure 3-7 Basic: Immunoglobulin structure, linear structure of atypical Immunoglobulin G molecule. The variable regions of the light and heavy chains are Indicated by the brackets, Each chain can be divided into domains on the basis of sequence similarities; the light chains comprise two domains and the heavy chain four. The Fab (fragment antigen binding) and the F~ (fragment crystallizable) regions are defined by analysis with papain and pepsin.The hinge region that links the Fc and Fab portions is flexible and allows independent movement of the two Fab arms. Membrane immunoglobulins have a very short cytoplasmic: tail that is unable to transduce the signal to the cell after reaction with their spedfic antigen. Immunoglobulins in the cell membrane are assodated with two other chains, Iga and Ig~. in a receptor complex. The cytoplasmic: tails of these two chains contains particular sequence motifs, called immunoreceptor tyrosine activation motifs (ITAMs), that transduce the intracellular signaling.
30
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
is present principally as dimers. Pentamers and dimers are formed using the joining 0) chain, a 15-kD polypeptide synthesized by the plasma cell that binds the Cterminus of heavy chains. The structural features that distinguish the heavy-chain constant regions of the various isotypes confer distinct properties that enable them to activate different effector mechanisms and to be transported across epithelia or the placenta (Table 3-8). Within the variable regions, there are hypervariable (HV) regions, also called the complementarity-determining regions, or CDRs (CDR1, CDR2, CDR3). They are localized to the surface of the molecule in such a way that when the VH and the VL regions pair in the antibody molecule, the hypervariable regions are brought together, creating a single hypervariable site that forms the binding site for antigens. Thus, final antigen specificity is particularly determined by the juxtaposition of the CDRs. The molecular region that is recognized specifically by an antibody is called the antigenic determinant, or epitope.
Membrane Immunoglobulins On B lymphocytes, immunoglobulins are bound to the membrane, where they serve as specific antigen receptors. Membrane immunoglobulins have a very short cytoplasmic tail that is unable to transduce the signal to the cell once the immunoglobulin has reacted with its specific antigen. Immunoglobulins in the cell membrane, however, are associated with two other chains, Iga and Ig~, to form a receptor complex,31 The cytoplasmic tail of these two chains contains particular sequence motifs, called immunoreceptor tyrosine activation motifs (ITAMs), which are common to other signaling molecules of the immune system, including those of the TCR complex (see Fig. 3-7 and later discussion).
Generation of Antibody Diversity The heterogeneity of antibodies allows each molecule to recognize a particular antigen. The entire collection of
( ' . TABLE 3 8
antibody specificities in a given individual (the antibody repertoire) is large enough to ensure that virtually any structure can be recognized. The problem of how this almost infinite range of specificities could be encoded by a finite number of genes was solved with the demonstration that different parts of the variable region are encoded by different sets of gene segments. 32 Indeed, each immunoglobulin chain, both light and heavy, is encoded by distinct genomic regions, one for the variable portion and one for the constant portion. For the light chain, each variable region is encoded in two different DNA segments: the variable (V) chain segment and the joining (J) chain gene segment. In the case of heavychain variable regions, in addition to the V and J segments, there is also a diversity (D) gene segment. There are multiple functional copies of the V, D, and J segments. During the development of a lymphocyte, one copy of each gene segment is joined randomly to the others by irreversible DNA recombination (somatic recombination), This process generates light- and heavychain variable regions. Several enzymes act in concert during somatic gene recombination, including the product of recombination-activating genes 1 and 2 (RAG-l and RAG-2), terminal deoxynucleotidyl transferase (TdT), and DNA-dependent protein kinase. In mature lymphocytes that respond to antigens in secondary lymphoid organs, somatic hypermutation-" introduces a very high rate of point mutations into rearranged variable region genes and generates further variability; some of the mutant immunoglobulin molecules bind antigens better than the original surface immunoglobulin, and B cells expressing them are selected to mature into antibody-secreting cells, a phenomenon called affinity maturation. Constant-region genes in the same species are polymorphic, and the allelic variants are called allozypes. The determinants of the antigen receptor that are specific for a given lymphocyte clone are called idiotopes, and the collection of all the idiotopes of a given receptor is called the idiozype. The idiotype therefore represents the col-
Immulloglobulins
Normal adult serum level (mg/mL) Molecular weight (x 103) Intravascular (%) Total circulating pool (mg/kg) Half-life (days) Rate of synthesis (mg/kg/day)
IgGI
IgG2
IgG3
IgG4
IgAl
IgA2
IgM
IgD
IgE
9 146 45 325 23 30
3 146 45 115 23
1 170 45 35 9 4
0.5 146 45 20 25
3 160 40 80 6 25
0.5 160 40 15 6
1.5 970 75 37 5 7
0.03 184 75 1 3 0.4
0,0005 188 50 0.02 3 0.02
++ ++
±
+ +++
+
+
-'
-
+++
±
±
+++ +++ +
+ +
+++ +++ +++
+ +
+
+
+
+
+
+
Biologic function Crosses placenta Fixes complement Binds cells Mononuclear cells Neutrophils Lymphocytes Mast cells Platelets
±
.
±
+ +++
'. Alternative pathway fixation; - , none; ±, weak; +, moderate; ++, intense: +++. strong. [g, immunoglobulin.
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
lection of specific determinants that distinguish each lymphocyte clone from all the others. In the antibody molecule, a given variable region can be associated with any constant region. Indeed, the constant region expressed in a B lymphocyte changes during the maturation of the cell and its subsequent proliferation in the course of the immune response, a phenomenon known as isotype switch.
BCell Development and Selection The process of gene rearrangement that gives rise to membrane immunoglobulins continually generates new populations of B cells of highly diverse specificity and provides the material on which clonal selection acts during the adaptive immune response. Nonlymphoid stromal cells of the bone marrow provide the essential microenvironment for B cell development by means of specific cell-to-cell contacts and release of growth factors. The process is characterized by sequential rearrangements and expression of the immunoglobulin genes, which regulate progression from one stage to the next.-\4 The random generation of the B cell repertoire gives rise to autoreactive immature B cells, which are eliminated during development in the bone marrow. Immature B cells express only surface IgM; in contrast to mature B cells, they undergo apoptosis and are removed from the repertoire (clonal deletion) if they bind abundant multivalent antigens such as ubiquitous cell siUrface molecules. However, a small proportion of these autoreactive immature B cells may escape this fate by replacing their autoreactive receptors with receptors that are not autoreactive. This rescue process is made possible by further gene rearrangement (receptor ectitingJ.35 Immature B cells that bind to soluble antigens are inactivated but not deleted; this state of nonreactivity is called anergy. Because anergic B cells cannot be activated by T cells, they fail to compete with other B cells in the periphery and are rapidly lost. Moreover, because B cells specific for protein antigens need T cell help in order to be activated, T cell self-tolerance by itself appears to be sufficient to prevent the production of high-affinity autoantibodies specific for self-proteins. Mature B cells leave the bone marrow and enter the peripheral lymphocyte pool as naive B cells. The influx of new mature B cells must compensate for the death of an equal number of peripheral B cells to maintain the steady state of the peripheral pool. Some naiVe B cells have a half-life of only a few days, probably because they are excluded from the lymphoid follicles. Naive B cells that successfully enter lymphoid follicles have a half-life of several weeks. After encountering the specific antigen, some B cells differentiate into effector cells that actively secrete antibodies, and some become nondividing, very-long-lived memory cells. Some B cells do not follow the developmental pathways so far described and have a distinctive receptor repertoire and t\lllctional properties. These cells are called Bl or CDS+ B cells (conventional B cells are called B2 B cells}\6 because they were first identified by the surface expression of the protein CDS (which is also present on T cells). Little is
31
known about their function. They arise early in ontogeny and, in adults, form a self-renewing population that is abundant in pleural and peritoneal cavities. They have a limited diversity in their repertoire; they produce low-affinity, polyspecific, and autoreactive antibodies (mainly IgM); and they contribute little to the adaptive immune response against protein antigens, but they do contribute to the immune response against some bacterial polysaccharides. All of these characteristics suggest that Bl B cells mediate a more primitive, less adaptive immune response than do conventional B cells. Recent studies have focused on the physiologic role of CDS in the context of regulation of antigen receptor activation, Band T cell selection, and generation and maintenance of immune toleranceY
T L,mphocytes and Their Antigen Receptors Immunoglobulins bind antigens in the blood and in the extracellular space. However, some bacteria and parasites, and all viruses, replicate inside cells, where they cannot be recognized by antibodies. These pathogens are destroyed by T cells, which are responsible for cellmediated immunity and recognize only antigens displayed on the cell surface, by molecules encoded by the genes of the MHC. Because T cells are essential in helping B cells, both cellular and humoral immune responses to protein antigens depend on antigen recognition by T lymphocytes. Infectious agents may replicate inside the cell either in the cytosol or in the vesicular system (endosomes and lysosomes). Cells infected with viruses or bacteria that live in the cytosol are killed by CTLs, which are distinguished by the presence of the cell surface molecule CD8. Pathogens or their products that are internalized by the cell in the vesicular system are detected by T cells that express the cell surface molecule CD4. CD4+ T cells, also called helper (Th) cells, do not directly kill the infected cells but activate other types of effector cells. Helper T cells comprise different cell types, the best characterized of which are Thl cells, which activate macrophages to kill intracellulose bacteria, and Th2 cells, which activate B cells to make antibodies. MHC class I molecules deliver peptides from the cytosol to the cell membrane, where the peptide-MHC complex is recognized by CD8+ T cells. MHC class II molecules deliver peptides from the vesicular system to the cell surface, where the peptide-MHC complex is recognized by CD4+ T cells. Therefore, depending on the type of MHC molecule that presents the peptide, the immune system can recognize the intracellular compartment in which the infectious agent is localized and activate different and appropriate effector mechanisms.
T Cell Receptor The antigen receptor of T cells (TCR) is a heterodimer composed of two different transmembrane glycoprotein chains (ex. and ~) bound in a structure that is very similar to the Fab fragment of the immunoglobulin molecules (Fig. 3-8).58 The organization of the gene segments encoding the TCR is very similar to that of immunoglobulins. 39 Each TCR locus consists of variable (V), joining CJ), and constant (C) region genes; the ~ locus also
32
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
contains the diversity (D) gene segments. As for immunoglobulins, the different gene segments coding for the variable regions of the TCR are present in multiple copies and undergo a process of gene rearrangement that, if successful, leads to the production of a functional TCR. During rearrangement, TCR genes are recognized by the same enzymes that recognize immunoglobulin genes. A further similarity between immunoglobulin and TCR rearrangements is the random addition of nucleotides at gene segment junctions. Because the TCR recognizes antigen but does not directly mediate the effector function, its constant chain is much simpler than that of immunoglobulins. The TCR recognizes the complex formed by the peptide and the presenting human leukocyte antigen (HLA) molecule. Moreover, T cells recognize and respond to antigens presented by an APC only if that APC expresses MHC molecules that the T cell recognizes as self-in other words, they are self-MHC restricted (see later discussion).40 Although the vast majority of T cells have the a:~ TCR, some lymphocytes have another type of TCR41 consisting of a heterodimer formed by "( and 15 polypeptide chains ("(:15 TCR). Both types are associated with a CD3 complex on the cell membrane. The function of "(:15 T cells is unknown, but they appear to represent a link between innate and adaptive immunity.42 They account for less than 5% of blood T cells; are present in epithelia of the gut, lungs, and skin; and appear to be able to recognize pathogens as well as stress-associated antigens expressed on cell surfaces. Growing evidence supports the possible relevant role of "(:15 T cells in the regulation of immune response at the tissue level.43
Membrane Coreceptors The TCR specifically recognizes the peptide-MHC complex but is independently incapable of activating the T cell. This function is carried out by a complex of pro-
teins-the CD3 complex-which is associated with the TCR on the cell surface. 44 Like the ex and ~ chains associated with membrane immunoglobulin, the cytoplasmic domains of the CD3 proteins contain sequences called ITAM that allow them, after receptor stimulation, to associate with tyrosine kinases and signal to the interior of the cell that antigen binding has occurred (see Fig. 3-8).45 Helper T cells and CTLs are characterized by the presence of the cell sUlface coreceptor proteins CD4 and CD8, respectively. CD4 binds to invariant parts of MHC class II molecules, and CD8 binds to invariant parts of MHC class I molecules. During antigen recognition, the CD molecules associate on the cell membrane with components of the TCR, participate in signal transduction, and bring about a IOO-fold increase in the sensitivity of the T cell to the antigen presented by MHC molecules (see Fig. 3-8).46
Antigen Recognition MHC class I molecules are heterodimers composed of a membrane-spanning ex chain noncovalently linked with ~2-microglobulin. The Il chain has three domains. The III and 112 domains fold together to form a cleft that represents the peptide-binding site. MHC class II molecules are heterodimers composed of two noncovalently joined transmembrane glycoproteins, Il and ~. Each chain has two domains. The III and ~I domains together form the peptide-binding cleft. Unlike antibodies that can recognize both conformational and continuous epitopes on a protein surface, T cells respond only to continuous short amino acid sequences that bind to the cleft of the outer surface of the MHC molecule.47 Because the composition of the MHC peptide-binding cleft is highly polymorphic, different allelic variants of MHC molecules bind preferentially to different peptides. Amino acids that are critical for the binding of a peptide
• Figure 3-8 Structure of the T cell receptor (TCR) complex. The a~ TCR complex of a CD4+ T cell is fonned by one a chain and one ~ chain covalently linked by a disulfide bond.The antigenbinding portion is fonned by the Va and V~ domains.TheTCR is noncovalently linked to the CD3 and ~ proteins. The cytoplasmic domains of CD3 and ~ chain contain one copy of a conserved sequence motif that is important for signaling function, called the immunoreceptor tyrosine activation motif (ITAM). The coreceptor CD4 molecule interacts with nonpolymorphic regions of class II major histocompatibility complex (MHC) molecules at the moment of antigen presentation. The major function of CD4 (and CD8) is to cooperate in signal transduction (together with the TCR) and in Tcell activation.
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
to a given MHC molecule (the peptide-binding motif) are called anchor residues. A change in anchor residues may prevent the peptide from binding; on the other hand, peptides with the same anchor residues bind the appropriate MHC molecule, regardless of the sequence of the peptides at other positions. The anchor residues binding a given MHC molecule need not be identical, but they are always related (e.g., aromatic or hydrophobic amino acid,,). Peptides that bind to MHC class I molecules are about 8 to 10 amino acids long; because of the open ends of the peptidebinding cleft of MHC class II molecules, peptides that bind MHC class II molecules are usually longer (13 amino acids or more). MHC molecules irreversibly bind peptides as an integral part of their stmcture and are unstable in the absence of peptides. This phenomenon prevents peptide exchange at cell surfaces, which would impair the efficiency of intracellular antigen recognition. MHC class I and class II molecules have different functions and are differently expressed on cells. MHC class I molecules present peptides to CTLs. Vimses can infect any nucleated cell, and MHC class I molecules are expressed with variable intensity on all cells except erythrocytes. The main function of CD4+ T cells is to activate other cells of the immune system. Therefore, MHC class II molecules are normally present on these effector cells. Class II molecules are also abundant on cells that are able to present antigen to T cells in lymphoid tissues (DCs, B lymphocytes, and macrophages). The expression of MHC molecules of both classes is enhanced by some eytokines (particularly interferons), which can induce MHC class II molecules on several cells that normally do not express them.
Complex intracytoplasmic machinery, which differs for class I and class II molecules, is responsible for the formation and loading of peptides on MHC molecules, as well as the delivery of the peptide-MHC complex to the cell surface. All proteins, including viral proteins, are formed in the cytosol. Proteins, such as the MHC molecules, that must be delivered to the cell surface are translocated into the lumen of the endoplasmic reticulum. For both MHC class I and class II molecules, loading of the specific peptide into the peptide-binding groove occurs in the vesicular system (Fig. 3_9).48,49 The MHC is not only polygenic but also highly polymorphic, because there are multiple alleles of each gene. All MHC class I and class II molecules are able to present antigens to T cells, and each molecule binds a different range of peptides. Diversity is further increased by the fact that, because of the marked polymorphism, most humans are heterozygous at these loci, and the expression of MHC genes is codominant; indeed, both alleles are expressed on the same cell, and both can present peptides to T cells.
Because there are three MHC class I genes and four potential sets of MHC class II genes, a heterozygous person expresses six different MHC class I molecules and eight different MHC class II molecules. For MHC class II molecules, the number of different products may be further increased by the combination of a and ~ chains from different chromosomes. Differences among the various HLA alleles are mainly related to variations in the residues that line the peptide-binding groove and therefore affect the peptide-binding specificity. Because T cells respond to protein-derived peptides, the expression
33
of several different HLA molecules on cell surfaces, each with different peptide-binding motifs, ensures the recognition of at least some of the peptides derived by proteolysis of the antigen. Although there is no response to the antigen in the absence of peptides available for HLA binding, HLA polymorphism extends the range of antigens to which the immune system can respond and makes antigen nonresponsiveness unlikely.<;o Allelic differences among MHC molecules also imply that the immune response against a given antigen may differ among individuals; this is a major factor in the individuality of the immune response. The polymorphism of other genes encoded in the MHC and involved in antigen presentation may also affect the immune response. For instance, the products of the two TAP alleles differ in their capacity to transport peptides; therefore, allelic variations in the TAP proteins can affect the repertoire of peptides available for MHC class I binding. The MHC contains many other genes whose function in the immune response is still not well established or is unknown. Some are induced in response to cell stress and encode the heat-shock proteins, which function as chaperones and are relevant to both adaptive and innate immunity. 51
T Cell Development and Selection T cells originate in the bone marrow and migrate at a very early stage to the thymus, where they undergo TCR gene rearrangement. In the thymus, thymocytes proliferate and mature into T cells through a series of events characterized by rearrangement of the TCR genes and expression on the cell membrane of the TCR, the coreceptors CD4 and CD8, and other surface molecules that reflect the functional maturation of the celp253 Thymocytes enter the thymic cortex and, dUring maturation, migrate from the cortex toward the medulla, coming in contact with epithelial cells, macrophages, and DCs. The medulla contains mostly mature T cells, and only mature CD4+ or CD8+ T cells exit the thymus and enter the blood. A fundamental step in thymocyte maturation (after expression of the TCR, CD4, and CD8 molecules) is the selection of cells that will make up the repertoire of mature T cells in the periphery. Mature T cells recognize only peptides bound to self-MHC molecules. On the other hand, the random generation of TCR can give rise not only to lymphocytes that do not recognize self-MHC molecules (and are therefore useless) but also to lymphocytes that recognize complexes of self-peptides/self-MHC molecules with high affinity (and are therefore potentially harmful). The dual need of selecting only cells that recognize self-MHC molecules and eliminating autoreactive cells is accomplished through processes of positive and negative selection (Fig. 3-10) that occur after the thymocytes have rearranged their a:~ TCR genes.<;4 Evidence from fetal thymic organ cultures suggests that these processes, which shape the future immunologic repertoire, are driven by the differential avidity for self-peptide/self-MHC complexes." Thymoeytes that express a TCR with low but measurable avidity for a self-peptide/self-MHC complex are allowed to
34
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
• Figure 3-9 Schematic representation of class I and class II major histocompatibility complex (MHC) pathways of antigen presentation. In the class I MHC pathway, protein antigens in the cytosol are processed (A) by proteasomes, and peptides are transported (8) into the endoplasmic reticulum (ER) by transporter proteins (transporter associated with antigen processing [TAP». In the ER, peptides bind to class I MHC molecules; the stable peptlde-class I MHC complex moves through the Goigi complex (0 and is transported to the cell surface by exocytic vesicles. In the class II MHC pathway, microbes are internalized into phagolysosomes (0) and are enzymatically degraded to generate peptides. Class II MHC molecules are synthesized in the ER (E) and transported to endosomes by exocytic vesicles. The exocytic vesicles transporting class II MHC molecules fuse with the endocytic vesicle containing the processed antigen (F). Thereafter, the MHC class II-peptide complex is transported to the cell surface.
differentiate into mature T cells, whereas immature lymphocytes that express TCRs with no avidity or with strong avidity for selfpeptide/self-MHC complexes die by apoptosis. More than 95% of developing thymocytes are thought to die in the thymus by apoptosis because they do not recognize self-MHC molecules or undergo negative selection. Cells that survive this dual selection process leave the thymus as mature na'ive T cells.
Lymphocyte Activation and Effector Functions T 4'mphocytes
Two Signals Recognition by the TCR of the specific peptide presented in the context of MHC molecules is necessary but not sufficient to activate T cells. To induce cell activation, TCR
recognition must be Simultaneously accompanied by the delivery of a second, costimulatory signal. This may occur when T cells interact with APCs. The activation of T cells on initial encounter with the specific peptide on the surface of an APC (priming) occurs in peripheral lymphoid organs. The initial interaction between naive T cells and APCs is mediated by adhesion molecules. If the naive T cell recognizes its specific peptide-MHC complex on the surface of an APC, signaling through the TCR induces conformational changes in the adhesion molecules that enhance the adhesiveness between the two cells. If no recognition takes place, the T cell dissociates from that APC and samples other APCs. The binding of the TCR and of the coreceptors (CD4 or CD8) transmits a first signal to the cell indicating that the specific antigen has
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
35
• fItIu'e 3-10 Processes of thymocyte maturation and selection in the thymus. Precursors ofT cells (CD4-CD8-TCR-) coming from the bone marrow arrive in the thymic cortex, where they mature into pre-T thymocytes (bearing a pre-T cell receptor formed by a ~ chain and a pre-T IX chain). In the ensuing maturation process, thymocytes express a complete Tcell receptor (TCR) together with both CD4 and C08 (double-positive thymocytes). During their migration from the cortex to the medulla, CD4+C08+ thymocytes go through the process of thymic selection. (1) The engagement of thymocyte TCR in a low-affinity interaction with a self-major histocompatibility complex (MHO molecule expressed on athymic epithelial cell rescues it from programmed cell death and allows the progression of thymocyte maturation as a na'ive CD4+ (or CD8+) mature T cell (positive selection). (2) If the thymocyte TCR does not engage in any interaction with peptide-MHC molecule complelles, it will die byapoptosis (lack of positive selection). (3) High-affinity or high-avidity binding between the thymocyte TCR and the peptide-MHC complex expressed by athymic antigen-presenting cell induces the death of the thymocyte by apoptosis (negative selection).
been recognized. The second signal is delivered through specialized molecules called costimulatory molecules,56,57 the best known of which are two structurally related molecules, B7-l (CD80) and B7-2 (CD86). The receptors for B7 molecules on T cells are two similar molecules, called CD28 (which is constitutively expressed on most T cells) and cytotoxic lymphocyte-associated antigen 4 (CTLA-4). Once the first signal has been provided, ligation of CD28 by B7 molecules induces lymphocyte activation. T cell activation also induces the expression on the cell surface of CTLA-4, which has a higher affinity for B7 molecules than CD28 does and delivers a negative signal to the cell, thereby limiting the proliferative response of activated T cells. The CD28/CTLA-4 and B7-1/B7-2 interactions therefore provide a regulation system that ensures that immune responses are turned on when needed and turned off when not needed (Fig. 3-11). A number of additional molecules with costimulatory function have been identified on the membrane of T lymphocytes. One of them, inducible costimulator (lCOS), presents many homologies with CD28 and its ligand is highly homologous to B7-l and B7-2. ICOS is thought to be important for stimulating the production of certain eytokines, such as IL-lO. In the same way, molecules other than CTLA-4, such as programmed death-l (PD-l), provide a negative signal for T cell activation in peripheral tissues. 5H
Approximately 100 specific peptide-MHC complexes are required on a target cell to trigger a T cell. Then, TCR aggregation initiates the events that lead to T cell activation. One of these events involves a cytoplasmic phosphatase, calcineurin, which activates nuclear activation factor of T cells (NAFT), which in turn binds to the promoter region of the IL-2 gene and is necessary to activate transcription. Because NAFT is a T cell-specific factor, blockade of the signaling pathway leading to NAFT is a way to specifically inhibit the T cell response. Both cyclosporin and FK506 inhibit calcineurin and therefore prevent the activation of NAFT. T cell activation leads to a 100-fold increase in IL-2 production, to the expression of anti-apoptotic proteins of the Bcl family, and to synthesis of the a chain of the IL-2 receptor. Resting T cells express a low-affinity IL-2 receptor composed of a ~ and a y chain. The association of the a chain with the ~ and y chains generates a receptor with a much higher affinity. Interaction of IL-2 with its receptor activates cell proliferation, which leads to the production of many T cells, all bearing an identical TCR (clonal expansion). Activation also induces the expression of CD40 ligand (CD40L) on T lymphocytes. 59 CD40L specifically binds to the CD40 molecule constitutively expressed on APe. This interaction induces and sustains the further activation of APC. This latter mechanism plays a pivotal role in the activation and differentiation of B
36
C HAP T E R
3
THE 1M M U N E
S YS T E MAN D
THE IN FL A M MAT 0 R y RES P 0 N S E
• Rgure 3-11 Mechanisms of T cell activation and the regulatory role of cytotoxic lymphocyte-associated antigen 4 (CTLA-4). A, Adhesion molecules are involved in the interactions between T cells and antigen-presenting cells (APCs). ICAM, intracellular adhesion molecule 1; LFA. leukocyte function-associated antigen. 8, Cross-linking of C028 delivers the costimulatory signal for Tcell activation and Induces the expression of C040 ligand (C) and CTLA-4 (D). See text for further explanations.
cells during the response to protein antigens (see later discussion). Once activated, CD4+ T lymphocytes can differentiate into either Thl or Th2 cells (see later discussion). Na'ive CD8+ T cells, because of their highly destructive effects, are kept under strict control and require high levels of costimulatory activity to be activated by APCs. 60 The delivery to the na'ive T cell of the first signal alone not only fails to activate the cell but also induces a state of unresponsiveness (anergy) in which the T cell is refractory to activation. The most important change induced in anergic T cells is their inability to produce IL-2. The need of the second costimulatory signal for T cell activation and the fact that this signal is provided only by APCs is important in preventing autoimmune diseases, hecause not all potentially self-reactive T cells are deleted in the thymus.
Antigen-Presenting Cells There are three main types of APCs: macrophages, DCs, and B cells. Macrophages normally express few MHC class II and no B7 molecules on their surface. Both molecules are expressed when macrophages engulf microhial constituents and are activated. As already noted, APCs have a number of receptors for common microbial constituents, which allow the immune system to distinguish antigens that are derived from infectious agents from those derived from innocuous proteins. 61 Antigens derived from infectious agents, but not those associated with innocuous proteins, induce costimulatory molecules. Indeed, innocuous proteins administered in conjunction with bacterial products (the so-called adjuvants) may elicit an immune response because bacterial prod-
ucts induce costimulatory activity in APCs that have ingested the protein. Experimental autoimmune diseases are also induced in susceptible animals by the administration of antigens mixed with bacterial adjuvants, a phenomenon that underscores the importance of costimulatory activity in the process of selflnonself discrimination. Not all infectious agents effectively induce MHC class II and costimulatory molecules on the surface of macrophages. This is particularly tme for viruses that replicate inside the cell. DCs, which constitutively express large quantities of MHC class II and costimulatory molecules, are thought to have a major role in presenting viral peptides to na'ive T cells and in priming both CD4+ and CD8+ T cells. 62 B cells also internalize soluble antigens bound to their surface immunoglobulins; peptide fragments generated from these antigens are displayed on the cell surface in association with constitutively expressed MHC class II molecules. 63 This is the pathway by which B cells can be the target of antigen-specific CD4+ helper T cells.
T Cell Receptor Degeneracy The same TCR may be activated by different peptides that share relatively little primary sequence homology (TCR degeneracy).64 Unlike the case with antibodies, there is no affinity maturation process that improves the fit of a TCR for a given peptide-MHC molecule complex during an ongoing immune response. The specificity of the TCR repertoire most likely represents a balance between two possible extremes: highly specific TCRs and highly degenerated TCRs. The former recognize partiClIlar amino acids at many positions of the MHC-bound
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
peptide; the latter recognize only certain sequence features and therefore may cross-react, although with different degrees of affinity, with many different micro1;>ial peptides, The highly specific TCR has a low probability of meeting its corresponding antigen and may never be eng'lged in the immune response; the highly degenerated TCR has a higher probability of being engaged but may also theoretically cross-react with self-peptide and induce an autoimmune response. 65 TCR engagement by different peptide ligands may lead to different functional outcomes that depend on whether the peptide acts as an agonist, partial agonist, or antagonist. Indeed, the functional consequences of T cell antigen recognition may vary depending on the nature of the peptide that contacts the TCR. It has been shown that peptides in which one or two TCR contact residues have been changed (known as altered peptide ligands, or APL<;) induce only a subset of the functional changes induced by the wild-type peptide or may even induce a state of T cell anergy that prevents response on secondary exposure to the wild-type peptide. The APLs appear to induce different intracellular pathways with respect to those induced by the usual T cell activation. These differences may depend on differences in affinity or conformational changes of the TCR. By these mechanisms, antigen receptors may translate quantitative differences in ligand binding into qualitatively different biolOgic responses. 65 T cell anergy may therefore be induced not only by antigen recognition in the absence of costimulation but also when, despite the availability of adequate costimulation, the antigen receptor signal is suboptimal-for example, when a T cell encounters an APL that does not optimally bind to the TCR. APLs have been successfully used to prevent experimental autoimmune diseases.6(,
Superantigens Some products of bacteria, mycoplasmas, and viruses induce a potent polyclonal and potentially harmful immune response. These superantigens bind to the outer surface of both the MHC class II molecules and the V~ region of the TCR, outside the peptide-binding site, and lead to T cell activation. 67 Each superantigen may activate all the T cells that express a particular set or family of Vp TCRgenes. This stimulation does not prime an adaptive specific immune response against the pathogen but may cause massive production of inflammatory cytokines such as IL-l and TNF and may lead to septic shock, the most severe cytokine-induced complication of infection by gram-negative and some gram-positive bacteria. Bacterial superantigens include the staphylococcal enterotoxins, the most common cause of food poisoning in humans, and the toxic shock syndrome toxin 1.
Effector Functions of Helper T Cells The Thl and Th2 subsets of helper T cells are distingUished on the basis of the cytokines they produce: IL-2, IFN-y, and TNF for Thl cells and IL-4, IL-5, IL-I0, and IL13 for Th2 cells. 68 ,69 It is increasingly evident, however, that many T cells cannot easily be classified as Thl or Th2
37
and that some (called ThO) have a heterogeneous pattern of cytokine secretion, Nevertheless, the differentiation of naIve CD4+ T cells into either Thl or Th2 cells appears to be a fundamental step for the subsequent evolution of the immune response, Production of Thl cells induces cellmediated immunity; production of Th2 cells leads to humoral immunity, a phenomenon that profoundly influences disease outcome, as exemplified by infections with intracellular pathogens such as leprosy. Mycobacterium leprae grows in macrophage vesicles, and effective host defense requires macrophage activation by Thl cells. In patients with tuberculoid leprosy, which is characterized by a predominantly Thl response, the infection is controlled and the patient usually survives. In lepromatous leprosy, which is characterized by a predominantly Th2 response, the main response is represented by antibody production; these antibodies cannot kill the intracellular bacteria, and the patients eventually die.
Helper T Cell Differentiation Mechanisms controlling helper T cell differentiation involve several factors, including the type of cytokines released at the time of antigen presentation, the amount and sequence of the antigenic peptide, and possibly the nature of the APC. 70 IL-12 and IL-23, the most important cytokines that stimulate the development of Thl cells, are produced by macrophages and NK cells. IFN-y, which is produced by macrophages, NK cells, and Thl cells themselves, enhances IL-12 secretion by macrophages, probably has a direct Thl-inducing effect, and inhibits the development of Th2 cells, IL-4, produced by Th2 cells, stimulates Th2 development. Both IL-4 and IL-1O inhibit the generation of Th1 cells. It has been hypothesized that activated CD4 cells produce small amounts of IL-4, which leads to Th2 differentiation, unless they are counteracted by the presence of Thl-inducing cytokines. The differing capacity of pathogens to interact with macrophages and NK cells may therefore influence the overall balance of cytokines produced early during the immune response and thus determine the preferential development of Th1 or Th2 cells. The intracellular signals that regulate Th1 or Th2 conunitment of CD4+ T cells have been the subject of intense investigations. During antigen presentation in secondary lymphoid organs, the regulatory cytokines IL-12 and IL-4 playa critical role in the orientation of the functional phenotype, through the activation of signal transducer and activator of transcription 4 CSTAT-4) or STAT-6 signaling pathways, respectively71 (Fig, 3-12). Differentiation signals regulate the expression of critical tissuespecific transcriptor factors, which further specify the Th1 or Th2 orientation through regulation of chromatin structure and accessibility of cytokine genes, The Th1-restricted transcription factor, T-bet, directly transactivates the ifng promoter and induces remodeling of the ifng locus,n Conversely, the Th2restricted transcription factor, GATA-3, is responsible for the induction of chromatin remodeling at the Th2 cytokine locus acting on several control sequences, including Il4. 73 The ability of T-bet or GATA-3 to induce remodeling at the corresponding loci suggests that modification in chromatin structure underlies the acquisition of heritable competence to transcribe the Th1 or
38
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
• Rlllre 3-12 Development of Thl and Th2 effector T cells. Cytokines produced In the innate immune response to microbes or early in adaptive immune response influence the differentiation of na'ive CD4+T cell into Th 1 and Th2 cells (see text for explanations). Cytokines produced by Thl cells inhibit the development of Th2 cells and vice versa (dashed arrows). For sake of simplicity. other cytokines produced byThI (interleukin-2 (Il-21. tumor necrosis factor-a, Iymphotoxin) and Th2 (ll-5, Il-IO, Il-13) are not shown.
Th2 cytokines. 74 DCs may function as distinct subsets able to provide T cells with selective signals leading to either Thl or Th2 responses. 12 •70 Each T cell subset produces cytokines that serve as autocrine growth factors and promote differentiation of naIve T cells to that subset. Moreover, the two subsets produce cytokines that cross-regulate each other's development and activity (see Fig. 3-12). Indeed, as previously mentioned, IFNy produced by Thl cells amplifies Thl development and inhibits proliferation of Th2 cells, whereas IL-4 and IL-IO produced by Th2 cells blocks activation of Thl cells and suppresses numerous macrophage responses. Thl and Th2 cells may be interconvertible early in their development and, in some cases, also in the late phases of the immune response. 7S Also, CD8+ T cells can respond to antigen by secreting cytokines typical of either Thl or Th2 cells. Thl and Th2 cells express different chemokine receptors 76 and therefore can be differentially attracted to the site of inflammation (see later discussion). For these reasons, chemokine receptor antagonism appears to be a promising approach for future treatment of autoimmune and allergic disorders.
Thl andTh2 Effector Functions Thl cells are important activators of macrophages; Th2 cells are the most effective activators of B cells, particularly in primary responses (Fig. 3-13). The principal Thl effector cytokine is 1FN-y, which display two main functions: it activates macrophages to destroy microbes, and it stimulates the production of 19G antibody subclasses that bind to complement and high-affinity Fey receptors and are the principal antibodies involved in microbe
opsonization and phagocytosis. An additional mechanism of macrophage activation is represented by the expression on Thl cells of CD40L, which activates the CD40 molecule on the surface of macrophages. Activated macrophages undergo several changes that augment their antimicrobial capacity and potentiate the immune response (e.g., upregulation of B7 molecules, further IL12 secretion).6H.69 Thl cells also are fundamental in recruiting macrophages to the site of infection. This occurs through the secretion by Thl cells of several additional cytokines, including (1) 1L-3 and granulocyte/ macrophage colony-stimulating factor (GM-CSF), which stimulate the production of new phagocytic cells in the bone marrow; (2) TNF-a, which induces the expression of adhesion molecules on endothelial cells; and (3) macrophage chemotactic factor (MCF) and other chemokines that direct the migration of macrophages from the endothelium to the site of infection (see later discussion). Many autoimmune disorders (e.g., rheumatoid arthritis, Crohn's disease) are characterized by infiltration and activation of Thl lymphocytes in the inflamed tissues. Th2 cells induce the production of IgM and non-complementfiXing IgG subclasses. The Th2 cytokine IL-4 induces a B-cell switch to IgE production, and IL-5 is the principal cytokine that activates eosinophils. Th2 cells typically mediate the immune response to helminths, stimulating the production of specific IgE antibodies that opsonize the parasites. Eosinophils activated by IL-S bind to IgE-coated helminths, through Fc receptors specific for e heavy chain, and release their granule contents. The same mechanism of activation via IgE is also the
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
39
• figure 3-13 Effectorfunction of hel~r T cells (Th I and Th2). Cytokines produced by Th I cells act on phagoeytes to increase phagocytosis and kllling. Interferon-y (IFN-y) also stimulates Bcells to produce immunog.lobulin G(lgG) antibodies that opsonize cells for phagocytosis. Interieukin-2 (IL-2) acts as a growth factor for Thl proliferation. LT, Iymphotoxin; TNF, tumor necrosis factor. Cytoldnes produced byTh2 cells (IL-4 and IL-l3) stimulate production of IgE antibodies, which bind to mast-cells during an allergic response. IL-4 represent also an autocrine growth factor for Th2 cells. IL-S activates eosinqphils, playing a pivotal role in the response against helminthic infections. IL-IO produced by Th2 cells (dashed alTOw) inhibits macrophage activation.
cause of mast cell degranulation in the immediate hypersensitivity immune response in allergic diseases. 68 .69
Another net result of Th2 activation is to inhibit Thlmediated inflammation through the release of antiinflammatory cytokines. IL-4 and IL-13 antagonize the macrophage-activation effect of IFN-y, IL-lO suppresses various macrophage responses, and TGF-~ (produced by some Th2 cells and by the Treg cell) inhibits leukocyte proliferation and activation. The Th2 response therefore limits the potentially dangerous consequence of uncontrolled Thl-mediated immunity. The possibility of downmodulating the Thl response through enhancement of the natut'al regulatory function of Th2 cytokines is an attractive therapeutic target in many Thl-mediated diseases. 68.69
Effector Functions of Cytotoxic T Cells CD8+ CTLs are required to eradicate intracellular microbes. After recognition of a specific class I MHC-associated antigen and subsequent activation, CTLs deliver their cytoplasmic cytotoxic granule proteins (perjorin and granzymes) to the target cell (Fig. 3-14),77 Perforin, which is also present in NK cells, has the capacity to polymerize in the lipid bilayer of the target cell membrane, thus forming an aqueous channel. Granzymes are serine proteases that enter the target cell through the perforin-induced channels and activate intracellular enzymes, called caspases, that play a pivotal role in the induction of programmed cell death (apoptosis). Activated CTL'! may also induce apoptosis through cell surface expression of the Fas ligand (FasL), which binds to and activates Fas on the cellular surface of target cells (see later discussion).
B Lymphocytes In addition to the binding of surface immunoglobulin (the antigen receptor) to its specific antigen, a second signal is required to activate B cells. The second signal is usually delivered by an already-primed CD4 cell (for thymusdependent [TD] antigens). In other cases, the second signal is provided by the pathogen itself (thymUS-independent [TI] antigens).
Thymus-Independent Activation The so-called TI antigens78 are nonprotein antigens that stimulate antibody production in athymic persons. The most important TI antigens are polymeric polysaccharides or glycolipids present in the bacterial cell wall. They induce maximal cross-linking of membrane immunoglobulins, leading to activation of B cells without the requirement of T cell help. Moreover, many polysaccharides activate the alternative pathway of complement system, generating C3d, which binds to the antigen and provides the second signal to the B cell receptor complex (Fig. 3-lSA). Antigen activates the signaling cascade by cross-linking the surface immunoglobulin molecules and therefore bringing receptor-associated tyrosine kinases (Fyn, Blk, and Lym) together with the ITAMs they phosphorylate. When phosphorylated on tyrosine, the !TAMs are recognized by cytoplasmic molecules that activate a cascade of events resulting in cell activation. Surface immunoglobulin function is also modulated by a membrane coreceptor complex, the CR2-CD19CD81 complex (see Fig. 3-1 SA). Coligation of the surface immunoglobulins with this coreceptor complex greatly increases the efficiency of cell activation. CR2 is a
40
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
• Figure 3-14 Effector functions of cytotoxic CD8+T cells (CTLs). A, After recognition of a spedfic class I MHC-associated antigen and subsequent activation, CTLs deliver their cytoplasmic cytotoxic granule proteins (perforin and granzymes) to the target infected cell. Perforin polymerizes the lipid bilayer of the target cell membrane, thus forming an aqueous channel. 8, Granzymes enter the target cell through the perforin-induced channels and activate the caspase cascade, indudng programmed cell death (apoptosis). C, Antigen-spedfic CTL is now ready to recognize and kill another adjacent infected cell.
receptor for the complement protein C3d. Therefore, as for T cell activation, the innate immune response provides signals that are essential for B cell activation. In most instances, however, the activation of B cells requires the presence of other additional signals, the most important of which is delivered by helper T cells that recognize antigen on the surface of B cells. Antibodies produced in the absence of T cell help are generally of low affinity and consist mainly of IgM because of a lack of T-mediated isotype switching processes. The antibody response to TI antigens occur mainly in the marginal zones of lymphoid follicles of the spleen. In this site, macrophages are particularly efficient at trapping polysaccharides.
Thymus-Dependent Activation Antibody responses to protein antigens require antigenspecific T cell help.79 Antigen-specific helper T cells must therefore have been primed earlier during the course of the immune response. B cells internalize the antigen bound to their surface immunoglobulin, degrade it, and present peptides on the cell surface in association with MHC class II molecules for recognition by specific helper
T cells (see Fig. 3-15B). The two cells do not need to recognize the same epitopes. What is crucial is that the epitope recognized by the T cell is part of the antigen that has been recognized and internalized by the B cell; this allows the B cell to present on the cell surface the peptide to which the T cell is specific. This linked recognition explains the immune response to the so-called haptens, small chemical groups that are unable to elicit an immune response because they are not recognized by T cells. However, haptens coupled to a protein can elicit an immune response, because T cells recognize the protein-derived peptides and provide the necessary help. This phenomenon is responsible for antibody production against substances such as penicillin that interact with host proteins and become able to induce an immune response.
B CellfT Cell Interactions The functional interaction between T and B cells take place in peripheral lymphoid organs (lymph nodes, spleen, mucosal immune system).79 Naive lymphocytes enter lymph nodes through the high endothelium venules (HEVs) (Fig. 3-16). In lymphoid organs, na'ive B
• Figure 3-15 Thymusindependent (A) and thymus-dependent (8) activation of B cells. See text for explanations. CD40L, ligand for (040; CR, complement receptor; ITAM, immunoreceptor tyrosine activation motif; TRAF,TNF receptor-associated factor.
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
41
• FI...... 3-16 Tcell and Bcell interactions in a lymph node (schematic representation).The cortex consists 01 an outer cortex (B cell zone) consisting 01 B lymphocytes organized into lymphoid follicles and some follicular dendritic cells (FOC). A deep or paracortical area (T cell zone) is composed mainly ofT lymphocytes and dendritic cells (DC).The medulla consist of strings of macrophages and plasma cells. A, Experimental models show that 1 to 2 days after antigen administration antigen is captured and processed by DCs present in the T cell zones. Na'ive T cells continuously recirculate through lymph nodes via high endothelial venules (HEY).Those Tcells whose receptors bind peptides derived from the antigen presented by DCs are trapped in the Tcell zone. Bcells recognize antigen in the primary follicle and are activated. Antigen-stimulated B cells move toward the Tcell zone. 8, At the border between T and B cell zones, antigen-specific lymphocytes proliferate, establishing a primary focus. Some proliferating Bcells differentiate into short-living, antibody-secreting plasma cells, which migrate to the spleen or to the medulla of the lymph node. Other antigen-specific Bcells migrate into primary lymphoid follicles, where they continue to proliferate (centroblasts). C, The germinal center is composed mainly of proliferating Bcells, FOCs, and a few helper Tcells. Centroblasts are closely packed in the dark zone (OZ). Later, they move into the so-called light zone (lZ), where they come in contact with a dense network of FOCs, mature into centrocytes, and express high levels of surface immunoglobulins. Germinal center B cells undergoV-region somatic hypermutation, which improves their affinity for antigen. B cells that lose their ability to bind the antigen die by apoptosis.Those with the highest affinities move to the outer edge of the LZ, where helperT cells expressing CD40 ligand are concentrated. Some germinal center cells differentiate into plasmablasts and then into plasma cells, which migrate to the bone marrow, where a subset provides a source of long-lasting, high-affinity antibody. Other germinal center cells differentiate into memory Bcells.
and T cells are anatomically segregated but are induced to migrate toward one another after activation with antigen. T lymphocytes recognize antigens presented by APC in the T cell zone; once activated, they reduce the expression of CCR7, the chemokine receptor produced in the T cell zone, and start to move. B lymphocytes recognize antigens in the follicles; once activated, they increase the expression of CCR7 and start migration to the T cell zone. The encounter between antigen-stimulated Band T cells occurs, therefore, at the interface of the follicles and the T cell zone. Recognition of the specific peptide-MHC complex on the surface of B cells activates T helper cells to express the cell surface molecule CD40L and secrete cytokines. The interaction between CD40L expressed on activated helper T cells and CD40 constitutively expressed on B cells leads to further activation of B cells. This mechanism, common also to other interactions between T cells and APCs, is particularly important for the activation and differentiation of B cells in a TD response (see Fig. 3-15B). The interaction with CD40L leads to the oligomerization of CD40 molecules. This induces the association of cytoplasmic proteins (TNF receptor-associated factors, or
TRAFs) with the cytoplasmic domains of CD40, initiating the enzymatic cascade that leads to the activation of transcription factors such as nuclear factor-KB (NF-KB) and activation protein-l (AP-l). These intracellular events play also an important role in the induction of isotype switching,80 a DNA recombination process during which the same variable region of the clonally expanded B cell is associated with different constant regions. B cells initially express IgM and IgD; later in the immune response, the same variable region may be associated with other constant portions, giving rise to IgG, 19A, or IgE. Unlike variable-gene segment recombination, switch recombination occurs only after antigen stimulation and not during B cell development. B cells undergo isotype switching to express transmembrane immunoglobulins of a different isotype before initiating the production of the secreted corresponding antibody. The second T helper-dependent mechanism of B cell activation and differentiation is related to the production of cytokines by activated T cells. Cytokines play two principal functions in antibody response: They increase the proliferation of B cells, and they promote the switch of immunoglobulin classes. Three different T-derived
42
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
cytokines (IL-2, IL-4, and IL-5) are known to enhance B cell proliferation; moreover, IL-6 produced by activated T cells and macrophages acts as an important growth factor on already differentiated and antibody-secreting B cells. IL-4 preferentially induces switching to IgE. TGF-~ induces switching to 19A, whereas IL-5 augments 19A production by cells that have already undergone switching. Although Thl cells are poor inducers of antibody production, the release of IFN-y induces switching to the IgG3 subclass that binds to high-affinity Fey receptors and complement.
and spleen migrate to the bone marrow; those originating from the mucosa-associated lymphoid tissue migrate to the lamina propria of epithelial surfaces. Plasma cells are terminally differentiated B cells that produce immunoglobulins, no longer interact with helper T cells, and are unable to change isotypes or undergo somatic hypermutation. Germinal centers last about 3 to 4 weeks after the supply of extrafollicular antigen is exhausted. Small numbers of B cells continue to proliferate in the follicles for months.
Effector Functions of Antibody-Mediated Immunity Affinity Maturation and Generation of Plasma Cells: The Germinal Center Reaction B cells that have been activated by T cells follow one of two patterns. Some migrate to the medullary cords and differentiate into short-lived plasma cells secreting IgM or IgG, thus providing an early immune response. Others migrate together with the specific helper T cells into the primary follicles, where they divide and form germinal centers (see Fig. 3-16).81 Primary follicles contain resting B cells clustered around a network of processes that extend from a specialized cell, the follicular dendritic cell, which plays a central role in the selection process during an antibody response. Although they have similar morphology, follicular dendritic cells (FDCs) are unrelated to the DCs that present antigens to the T cells. They are not derived from the hematopoietic stem cell, lack MHC class II expression, and express membrane receptors for complement and the Fc portion of immunoglobulins. FDCs play an important role in driving the maturation of the humoral immune system through their capacity to bind antigen-antibody complexes by their Fc receptors and to retain antigens on their surface for long periods. The early antibodies secreted by B cells of the medullary cord not only provide early protection but also trap antigen, in the form of immune complexes, on the surface of FDCs. Selection of B cells with surface immunoglobulins of the highest affinity for the antigen occurs at the surface of FDCs (see Fig. 3-16). B cells that lose their ability to bind the antigen die by apoptosis. On the contrary, those with the highest affinity move to the outer edge of the zone where helper T cells expressing CD40L are concentrated. Interaction with helper T cells induces B cells to become plasma cells or memory cells and prevents the selection of B cells that have acquired selfreactivity during somatic hypermutation. Other surface and soluble molecules, produced by cells other than T cells (DCs, macrophages, and monocytes), have been shown to playa fundamental role in the survival and maturation of B cells. One of these molecules is Bcel~activating factor of TNF family (BAFF). Notably, BAFF-deficient mice lack a mature B cell compartment, and BAFF overexpression has been suggested as a possible crucial mechanism for B cell hyperplasia in many autoimmune diseases. 82 Some B cells leave germinal centers as pre-plasma cells (plasmablasts) and migrate to distant sites, where they are supposed to differentiate into plasma cells with a long life span. 83.84 Those originating from lymph nodes
The first humoral response is characterized by the production of low-affinity IgM, which neutralizes microbes and circulating microbial toxins, blocking their binding to the host cellular receptors. IgG is the principal immunoglobulin in the blood and extracellular fluid, whereas 19A is the principal immunoglobulin in mucosal secretions. The different immunoglobulin isotypes have distinct properties that enable them to induce different effector mechanisms (see Table 3-8).85 Antibodies can activate a variety of effector cells that bear a receptor for their Fc portion. 86 Fc receptors are a family of related molecules with different affinities for different isotypes; the isotype determines which effector cell is preferentially activated (Table 3-9). Only Fc portions of immunoglobulins that have interacted with their antigens can activate Fc receptors. This occurs because of aggregation of immunoglobulin on the pathogen surface or because of conformational changes in the Fc portion occurring after antigen binding, or both. Antibodies coat (opsonize) the surface of microbes. The recognition of antibodies by Fc receptors on phagocytes triggers phagocytosis and killing of the opsonized microbes (see Fig. 3-2). Fc receptors may also activate mast cells, basophils, and eosinophils. Through their action, Fc receptors on NK cells and eosinophils may initiate antibody-dependent cellular cytotoxicity (ADCC) of antibody-coated infected cells and destroy them (see Fig. 3-50. Binding of antibodies to Cl is an important mode of complement activation (see Fig. 3-6).
Immunologic Memory Immunologic memory allows rapid and effective response to antigens that have been encountered previously and represents the presence of an already clonally expanded population of specific lymphocytes. These responses are called secondary, teniary, and so on, depending on the number of antigen exposures. Several hypotheses have been proposed to explain immunologic memory. It may depend on the persistence of long-lived lymphocytes in a resting state. Alternatively, lymphocytes activated by the original exposure to antigen may be restimulated repetitively by a small amount of persistent antigen (possibly at the level of FDCs), by stimulation with cross-reactive antigens, or by the release, during subsequent immune responses to other antigens, of soluble mediators that restimulate memory cells. 87-89
C HAP T E R
.:. rABI E 3
9
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
43
fl Receptors
Receptor
Structun
Cell Type
III Binding (affinity)
Function
FcyRI (CD64)
Two subunits: a chain (72 kD, CD64), Ychain (9 kD)
IgGI, IgG3 (10-" kD)
Activation of phagocytes and phagocytosis
FcyRIIA (CD32)
a chain (40 kD) with a y-Iike domain
IgGI (10-7 kD)
Phagocytosis
FcyRIIB (CD32) FcyRIIIA (CDi6) FCEIU
a chain (40 kD) with an ffiM domain Two subunits: a chain (50 kD), Ychain Three subunits: a chain (CD64), ~ chain 03 kD), y chain (9 kD) Two subunits: a chain (55 kD, CD89), Ychain (9 kD)
Macrophages Neutrophils Eosinophils Dendritic cells Macrophages Neutrophils Eosinophils Platelets B cells Mast cells Natural killer cells
IgGI, IgG3 00-7 kD)
Inhibition of activation
IgGI, IgG3 00-6 kD) IgE (10- 10 kD)
Antibody-dependent cell mediated cytotoxicity Cell activation (degranulation)
IgAI, IgA2 (10-7 kD)
Cell activation
FcaR (CD89)
Mast cells Basophils Eosinophils Macrophages Neutrophils Eosinophils
CD. cluster of differentiation: Ig, immunoglobulin: 111M, immunoreceptor tyrosine-based inhibition motif.
BCells Secondary antibody responses due to the stimulation of memory B cells differ in several aspects from primary immune responses. Memory cells not only have been clonally expanded but also have undergone somatic mutation, affinity maturation, and isotype switching. The antibody response is therefore more intense, and it is mainly high-affinity IgG. Secondary and subsequent responses lead to increasing affinity of antibody by a mechanism that is similar to that observed in a primary response and based on further somatic hypermutation and selection by antigen on FDCs in germinal centers. The first explanation offered for the maintenance of immunologic memory suggested the existence of long-lived plasma cells surviving in bone marrow. 90 However, their half-life is calculated to be at most 3 months, and it is unlikely that they are able to maintain a constant production of specific antibodies over a human lifespan. Memory B cells can survive in secondary lymphoid organs in the absence of antigen and mediate secondary immune response on restimulation. 91 Moreover, there is evidence that memory B cells also maintain serologic memory by undergoing continuous polyclonal activation. 92
TCells Long-lived effector T cells are difficult to distinguish from memory cells. The identification of memory T cells rests largely on the existence of a population of cells that have the surface characteristics of activated effector T cells but are distinct from them in that they require additional stimulation before acting on target cells. Changes in three cell surface proteins-L-selectin, CD44, and CD45-are particularly significant after exposure to antigen. L-selectin is lost on most memory cells, whereas CD44 levels increase on all memory cells and CD45 changes from a high- to a low-molecular-weight isoform (CD4SRA and CD45RO, respectively). These changes are characteristic of all activated effector T cells, yet some of the cells in which these changes occur have many char-
acteristics of resting T cells, suggesting that they are memory CD4+ T cells. The chemokine receptor CCR7 is a marker of memory T cell differentiation. According to Sallusto and coworkers,93 under physiologic conditions, memory T cells in secondary lymphOid organs are CCR7+ and express L-selectin at high levels; these cells have been defined as "central memory" T cells. 93 Conversely, memory T cells lacking CCR7 express low levels of L-selectin and high levels of integrins, a phenotype consistent with migration and tissue homing. CCR7- memory T cells are enriched for effector cells capable of producing large amounts of cytokines, as opposed to CCR7+ central memory T cells, which are poor cytokine producers. 93.94
Regulation of Lymphocyte Activation (Immune Homeostasis) During a normal immune response, after the eliciting foreign antigen has been eliminated, the immune system must terminate its activation and return to a state of rest. Moreover, because lymphocytes bearing receptors capable of recognizing self-antigens are generated constantly, the immune system must be able to prevent or abort potentially self-reactive responses. Several mechanisms, only partly understood, fulfill these control functions (Fig. 3-17),95 and their disruption may lead to autoimmune reactions. A number of the possible control mechanisms have been already described for T cells. For example, CTLA-4 appears 3 or 4 days after T cell activation and has a much higher affmity for B7-1 and B7-2, the same ligands on APCs of the T cell-activating CD28 molecule (see also Fig. 3-11). Thus, CTLA-4 is an important mechanism for the physiologic termination of T cell activation; disruption of the CTLA-4 gene in mice results in massive accumulation of activated lymphocytes in lymph nodes and spleen and inftltration of tissues by activated lymphocytes. 96,97 Cytokine counter-regulation is another important mechanism of T lymphocyte homeostasis. Th2 cytokines
44
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
• Rgure 3-17 Regulation of lymphocyte activation (immune homeostasis). See text for explanations. APC, antigen-presenting cell; IL, FasL, ligand for Fas; interleukin; MHC, major histocompatibility complex;TCR, T cell receptor; TGF, transforming growth factor.
OL-4, IL-13) inhibit macrophage activation and therefore control Th1-mediated immunity. Conversely, IFN-y, released by Th1 cells, inhibits IL-4-stimulated B-cell switching to IgE (see Fig. 3-12). Therefore, cytokines released by different T cell populations suppress different types of immune responses. Regulatory T cells (Treg) suppress or downmodulate T lymphocyte activation via the release of suppressive cytokines such as TGF-~ or IL10,98 or through direct cell-to-cell contact. Although IL-2 is an important T-cell growth factor that stimulates clonal expansion of antigen-stimulated lymphocytes, mice lacking IL-2 or the a or p chain of the IL-2 receptor develop lymphadenopathy and various manifestations of autoimmunityy9-101 This suggests that, although the function of IL-2 as a growth factor can be supplanted by other cytokines, IL-2 is part of a necessary feedback mechanism that controls lymphocyte responses, mainly through the induction of cell death for apoptosis with a pathway called activation-induced cell death. 95 ,102 Important mechanisms of regulation are also involved in humoral immune responses. Antibodies inhibit B cell activation by forming antigen-antibody complexes that bind simultaneously to Fey receptor II (FcyRIIB) and to antigen receptors of B cells. The cytoplasmic tail of FcyRIIB (see Table 3-9) contains the immunoreceptor tyrosine-based inhibition motif (ITIM) that mediates negative signals to B cells.
Apoptosls On exposure to antigen, clonal expansion of specific T and B lymphocytes increases their frequency by more than lOOO-fold within 1 week. After 1 to 3 months, the number of specific lymphocytes returns to baseline levels, leaving long-lived, functionally quiescent memory cells. This rapid decline in antigen-specific lymphocyte numbers results from apoptosis.
Apoptosis occurs by two main pathways (Fig. 3-18). Lymphocytes that are deprived of survival stimuli, such as costimulators and cytokines, lose expression of specialized anti-apoptotic proteins, mainly belonging to the Bcl familY,103 and die "by neglect." This mechanism leading to apoptosis, called passive cell death, is probably the most important mechanism that downregulates the immune response once the eliciting antigen has been eliminated and the innate immune response subsides. The stimuli that maintain quiescent and viable naive cells and memory cells are probably sufficient to maintain the expression of anti-apoptotic proteins and therefore to prevent cell death. Passive cell death differs from activation-induced cell death, in which apoptosis is actively induced, although both share the same terminal biochemical pathway. Activation-induced cell death104.105 is important in preventing uncontrolled T cell activation and as an effector mechanism of CD8+ cns and NK cells. It results from the interaction of two molecules that are coexpressed on activated cells: the surface receptor Fas (CD95) and its ligand (FasL or CD95L). Fas belongs to a family of proteins that includes TNF receptors (TNF-R) I and II and the B cell-activating molecule CD40. This family of proteins dictates signals that can induce cell survival and proliferation or apoptotic death. Activated, mature T cells express both Fas and FasL. Their interaction induces a series of intracellular events that lead to apoptosis. These events are initiated by the cytoplasmic region of Fas, which contains a sequence called the death domain and involves a proteolytic system, the central component of which is represented by a family of proteases called caspases. 106 The activation of TNFRI probably triggers a pathway of events similar to those triggered by Fas activation. The expression in the cell of
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
45
• fIIure 3-18
Pathways of apoptosis. A, Activation-induced cell death mediated by the interaction between Fas and Fas ligand. The intracellular death domains of Fas receptors bind acytosolic death dornahH.ontaining adapter called Fasassodated death domain (FADO). FADD binds and activate pro-caspase 8 (or caspase 10), which activates the effector caspase cascade, with the final activation of the caspase being activatable DNase (CAD), which is present in cytoplasm in inactive form, termed (O-CAD. Thereafter, CAD enter the nucleus and deaves DNA. 8, Intracellular mechanisms of programmed cell death. If the cells are deprived of necessary survival stimuli, the result is a rapid increase in permeability of mitochondrial membranes and release of cytochrome Co This event is allowed by the inactivation of anti-apoptotic proteins of the Sd family. Cytochrome c acts as a cofactor with activating factor-l (APAF-l), which, together with a caspase recruitment domain (CARD), activates pro-caspase 9, with the consequent further activation of the effector caspase cascade.
anti-apoptotic proteins (such as those belonging to the Bel family) protects against passive cell death but does not prevent activation-induced apoptosis. Fas appears to be a major mediator of apoptosis in CD4+ T cells; apoptosis induced by TNF-R is more relevant for CD8+ T cells. In mice, mutations in the Fas receptor Opr, lymphoproliferation phenotype) or its ligand (gld, generalized lymphoproliferative disease) are associated with massive lymphadenopathy and lupus-like autoimmunity. 107 In humans, Fas mutations cause the so-called autoimmune lymphoproliferative syndrome (ALPS),108 which is characterized by lymphadenopathy and autoimmunity. Fas-mediated apoptosis is important for eliminating cells that are specific for persistent antigens such as self-antigens. Indeed, mutations in Fas and FasL genes cause autoimmune diseases but not prolonged responses to foreign antigens. On the contrary, a prolonged immune response occurs with the constitutive expression of Bcl-2 as a transgene in B or T cells,109 a phenomenon that underlines the importance of passive cell death in the termination of responses to foreign antigens. In lymphocyte development in the thymus, apoptosis that occurs from a lack of positive selection appears to be secondary to lack of a rescue signal by the TCR, leading to passive cell death. On the contrary, apoptosis during negative selection is actively induced (activation- induced cell death) but does not appear to be secondary to Fas-FasL interactions, as occur in peripheral deletion mechanisms.
Immunologic Tolerance Immunologic tolerance is antigen-induced functional inactivation or death of specific lymphocytes that results both in the inability to respond to that antigen and in the
inhibition of lymphocyte activation during subsequent administration of the same antigen in an immunogenic form. 110 Antigens that induce an immune response are called immunogens, whereas antigens that induce tolerance are called tolerogens. Self-antigens are normally tolerogens; many foreign antigens, depending on their physicochemical form, dose, and route of administration, may act as immunogens, tolerogens, or both. For instance, protein antigens administered subcutaneously or intradermally are usually immunogenic, whereas large amounts of protein antigens administered intravenously or orally often induce unresponsiveness. The need to avoid self-aggression during the immune response to foreign antigens has been a determinant factor during the evolution of the immune system. The persistence of autoimmune diseases in the population could be explained by the fact that they more frequently affect people after they have passed the reproductive age and therefore no longer represent a serious threat to the survival of the species. Although the question concerning the balance between immunity and tolerance, including self-tolerance, has yet to be resolved, many observations are yielding a general framework. l1l- m The random generation of antigen receptors during lymphocyte maturation can give rise to lymphocytes that are specific for self-antigens. As described earlier (see Fig. 3-10), lymphocytes with high avidity for abundant self-antigens that are constitutively expressed by all cells (and therefore present in high concentration in the thymus and the bone marrow) are deleted during their development in central lymphoid organs (central tolerance). Although elonal deletion of self-reactive
46
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
lymphocytes in central lymphoid organs is an efficient process, not all self-antigens are expressed in central lymphoid organs. Lymphocytes that are specific for these antigens survive and must be eliminated or held in check by other mechanisms. These mechanisms, only partly understood, are responsible for peripheral tolerance and include not only clonal deletion but also clonal anergy, active suppression, and ignorance.
T Cell Tolerance Central Tolerance During their maturation in the primary lymphoid organs, lymphocytes encounter a number of antigens to which they respond by developing tolerance rather than activation. In fact, most of the antigens normally present in the thymus or bone marrow are self-antigens. Lymphocytes that recognize, with high affinity, self-antigen presented in association with MHC molecules by thymic APCs die by apoptosis (negative selection). This fundamental process avoids the possibility that lymphocytes with high affinity for receptors for ubiquitous self-antigens may reach maturation (see Fig. 3-10). This process affects both class I and class II MHC-restricted T cells and is therefore important for tolerance in CD8+ and CD4+ lymphocyte subpopulations. It has been shown that thymic stromal cells in both cortical and medullary epithelium also collectively express a number of tissue-specific antigens. 114 The expression of these self-antigens is regulated by a transcription factor called autoimmune regulator (AIRE). 115 Mutations in the AlRE gene cause a rare recessive inherited disorder characterized by an autoimmune polyendocrinopathy, called autoimmune polyendocrinopathy candidiasis-ectodermal dystrophy (APECED).116 Many of the genes regulated by AIRE encode tissue-specific proteins such as insulin, thyroglobulin, and zona pellucida glycoprotein. Thus, central tolerance seems to play an important role also in the mechanisms of tolerance toward tissue-specific antigens. ll ?
Peripheral Tolerance Because only a limited number of self-antigens are present in primary lymphoid organs, the mature lymphocyte repertoire contains many cells capable of recognizing other self-antigens that are exclusively expressed in the peripheral tissues. Therefore, other mechanisms of tolerance are required to avoid the response of these mature self-reacting lymphocytes. Most of the mechanisms involved in peripheral tolerance are related to the strategy of control of lymphocyte activation (see Fig. 3-17). As previously noted, activation of CD4+ T cells after recognition of antigens presented by APCs requires the simultaneous presence of a second signal given by the costimulatory molecules that are expressed only by activated and not by resting APCs. In the absence of such a costimulatory signal, T cells are able to survive but are rendered incapable of responding to the same antigen in a future encounter (clonal anergy). Thus, the continuous
presentation to mature T cells of self-antigens by resting APCs makes many self-reacting T cells anergic (Fig. 3-19). Anergy may be also induced by the inhibitory receptor CTLA-4, at the time of antigen recognition. In this situation anergy is not induced by the absence of costimulation, but by the delivery of active inhibitory signals to T cells. The possible mechanism of the preferential binding of costimulatory molecules expressed by APCs to CTLA-4 rather than to CD28 on T cells is not completely understood. However, the importance of CTLA-4 in the induction of tolerance is proven by the development of uncontrolled lymphocyte activatlon and fatal multiorgan lymphocytic infiltration suggestive of autoimmunity in knockout mice lacking CTLA-496 (see Fig. 3-19), Another important mechanism of peripheral tolerance is represented by the deletion of self-reactive T cells by activation-induced cell death. Repeated stimulation of T cells by persistent antigens induces the death of the activated cells by apoptosis. It is believed that Fas-mediated activation-induced cell death is responsible for the elimination of T cells specific for abundant peripheral antigens that are most likely responsible for the repeated activation of T cells (see Fig. 3-19). Most patients with the Fas receptor mutation associated with massive lymphadenopathy and lupus-like autoimmunity are heterozygous for the defect lOS; however, other family members, with the same heterozygous mutation and showing defective in vitro apoptosis, do not develop disease. Therefore, in addition to the Fas mutation, other genetic factors are required for disease development. In humans, a FasL mutation associated with autoimmunity has been reported, llS as well as an autoimmune Iymphoproliferative syndrome (type II) caused by caspase 10 mutations. 1l9
Studies on apoptosis have provided a possible answer for two mysteries that surround systemic autoimmune diseases: (1) why are autoantibodies directed against antigens that are normally found inside the cell, even in the nucleus? and (2) how do these antibodies contribute to disease pathogenesis? During apoptosis, nuclear autoantigens, as well as other intracellular autoantigens (such as RNA and ribosomal endoplasmic reticulum components) become localized within surface blebs. In addition, phosphatidylserine (PS), a membrane phospholipid that is normally distributed on the intracellular portion of the membrane, is redistributed to the outer membrane early in apoptosis and can bind autoantigens such as the phospholipid-binding protein ~2-glycoprotein 1. Under normal circumstances, uptake of apoptotic cells by macrophages induces the production of anti-inflammatory cytokines such as TGF-~ and the downregulation of inflammatory cytokine production. DC maturation and presentation of antigens are suppressed by the uptake of apoptotic cells, which leads to the promotion of tolerance. 120 If something goes wrong in the recognition and management of apoptotic cells, intracellular autoantigens presented at the cell surface during apoptosis could prime an autoimmune response. A further important mechanism of peripheral tolerance is related to inhibitory regulatory T cells (Treg).121.122 Distinct T cell subpopulations may presumably play these regulatory functions using different mechanisms
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
47
• fItIn 3-19 Possible mechanisms of peripheral tolerance. See text for explanations. TcR, Tcell receptor; APC, antigen-presenting cell; FasL, ligand for Fas; IL, interleukin; MHC, major histocompatibility complex; TCR, T<:ell receptor; TGF, transforming growth factor.
(see Fig. 3--19), Antigen-specific T cells (Trl) selectively producing high levels of the anti-inflammatory eytokine IL-lO are able to inhibit the development of experimental autoimmune colitis. 123 ,124 The same cytokine-mediated regulatory mechanism was also observed for cells having high production of TGF-~ and defined as Th3. 125 A third population of Treg cells is represented by resting circulating CD4+ T cells expressing high levels of the IL-2 receptor ex chain (CD2S).126 In this case, the inhibitory action requires cell-to-cell contact between CD4+CD2S+ T cells and APC or naIve T cells. Treg cells presumably arise in the thymus or in peripheral organs after self-antigen recognition. CD4+CD2S+ Treg in the thymus selectively express a particular gene (FoxP3), that produces a regulatory transcription factor, scurfin. 127 A defect of this gene in humans is associated with a severe autoimmune disease known as immunodysregulation, polyendocrinopathy, enteropathy, X-linked (IPIDO syndrome,l28 which develops in early infancy. The study of the mechanisms of peripheral tolerance offers opportunities for the elaboration of new therapeutic approaches for many autoimmune disorders. In fact, the reinstatement of tolerance toward a self-antigen relevant in disease pathogenesis may represent the most promising strategy for the cure of these disorders. The mechanisms for B cell tolerance induction were discussed earlier, in the section on B cell development and selection.
The Cryptic Self and the Immune Privilege Many self-proteins are presented too poorly to T cells to be recognized. These self-antigens are called cryptic epitopes and are collectively referred to as cryptic self. T cells that are specific for these epitopes are not tolerant, because they can respond if the antigen is presented
appropriately. They are considered ignorant, because they are not aware of the existence of the self-antigen they are able to recognize. Given the number of self-protein epitopes, an immune system from which all potential selfreactivity has been removed would probably not respond to foreign antigens. 129 It is therefore likely that a large number of potentially autoreactive T cells reside in the immune system in a resting state, without causing disease because they ignore their respective self-antigens. Ignorance may also be related to the difficulty with T cells' gaining access to some tissues, such as brain, gonads, and eye ("immunologically privileged sites"), because of the existence of blood-tissue barriers. It is now apparent that the "immune privilege" of these tissues is a complex and active phenomenon that includes local production of immunosuppressive neuropeptides and cytokines, limited MHC antigen expression, and, in the eye and testis, constitutive expression of FasL, which promotes the death of invading Fas-positive cells during viral infections. 130
Oral Tolerance Oral tolerization 131 is a phenomenon in which the oral (or, more generally, the mucosal) administration of an antigen induces a marked suppression of systemic humoral and cell-mediated immune responses to immunization with the same antigen, without affecting the response to other antigens. It is presumed that this mechanism has evolved to prevent systemic immune responses to ingested proteins and to bacteria that normally reside in the intestinal lumen. The mucosal immune system is important in the defense against infection. 132 Much of the current knowledge of mucosal immunity is based on studies of the gastrointestinal tract, but it is likely that the general features of mucosal immunity are similar in all mucosa-associated
48
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
lymphoid tissues. Immune responses to antigens delivered by the oral route differs from those to antigens encountered in other sites in two main respects: 0) the predominant production of IgA in mucosal tissues and (2) the tendency of oral immunization with protein antigens to induce systemic T-cell tolerance rather than activation, a phenomenon that is particularly appealing for the possible induction of tolerance to autoantigens in autoimmune diseases. In the intestinal mucosa, lymphocytes are scattered both among epithelial cells Cintraepithelial lymphocytes) and in the lamina propria. Ten percent of intraepithelial lymphocytes bear the y:8 TCR. Both y:8 and a.:~ intraepithelial T cells show a limited diversity of antigen receptors and are thought to have evolved to recognize commonly encountered intraluminal antigens. The small intestine contains organized mucosal lymphoid follicles (Peyer's patches). Similar follicles are found elsewhere along the gastrointestinal and respiratory tract (e.g. pharyngeal tonsils). Peyer's patches have B cell-rich areas that often contain germinal centers and CD4+ T cells, mainly in the interfollicular regions. The epithelium overlying Peyer's patches contains specialized cells, called membranous cells (M cells), which lack microvilli and engulf and transport macromolecules from the intestinal lumen into subepithelial tissues. M cells are thought to be important in delivering antigens to Peyer's patches. Lymphocyte homing to Peyer's patches and the lamina propria of . the gut has particular characteristics. Most of orally administered antigen is carried by lymphatics to the draining mesenteric lymph nodes, where an immune response is initiated. Some protein antigens are transported by M cells into Peyer's patches, where they can stimulate both T and B cells. Lymphocytes derived from both the mesenteric lymph nodes and Peyer's patches may then reach the bloodstream or the lamina propria. The reason why some protein antigens induce oral tolerance whereas others elicit effective immunity is not known. One possible explanation may be the capability of infectious organisms to activate APCs to express costimulatory molecules. Another explanation involves deletion mechanisms, the induction of a state of anergy, and active immunosuppression through the release of immunosuppressive cytokines such as TGF-~ or lL-1O, by Th3 and Tr1 regulatory T cells, respectively (see Fig. 3-19). The antigen dose influences the mechanism involved: low doses favor active suppression, and high doses favor deletion or anergy. Several studies have shown that oral administration of the specific autoantigen is effective in treating experimental autoimmune diseases, including experimental allergic encephalomyelitis and collagen-induced arthritis. 133 Induction of oral tolerance by nasal or oral administration of antigen could be an interesting therapeutic option for human autoimmune diseases in which the relevant eliciting antigen is unknown. 134 The cytokines secreted by T cells induced in the gut-associated lymphoid tissue by oral antigen administration suppress inflammation in an antigen-nonspecific way (bystander immunosuppression) in the microenvironment where the fed antigen is localized. The induction of oral tolerance to an antigen present, for instance, in the joint could theoretically result in suppression of joint inflammation when the elicited regulatory cells migrate to the joint, meet the specific antigen, and release suppressive cytokines.
THE MULTIFACTORIAL ORIGIN OF AUTOIMMUNE DISEASES Autoimmune recognition was once viewed as a distinct abnormality, as indicated by the term used by Ehrlich: "horror autotoxicus." In recent decades, it has become increasingly evident that autorecognition is a phenomenon that occurs normally in the immune response and represents the basis of formation of the normal adaptive immune response. As summarized earlier, the normal immune T cell repertoire is selected by low-affinity recognition of self-peptides presented in the context of self-MHC molecules. Moreover, in normal immune responses, a transient and limited autoimmune response may occur but is rapidly downregulated and does not lead to tissue injury. Yet, in about 5% of the population, for reasons that are still incompletely understood, a strong autoimmune response arises and leads to inflammation and tissue damage. Despite the great number of potential autoantigens, there are relatively few distinct autoimmune diseases and, in persons with a given autoimmune disease, the same autoantigens tend to be recognized. This suggests that only a small fraction of self-proteins can actually serve as autoantigens. It is also conceivable that lymphocytes that mediate autoimmune responses do not undergo clonal deletion in central lymphoid organs but rather are present in a tolerant or ignorant state in all normal persons and may be activated only in particular circumstances. The development of an autoimmune disease is presumably initiated by an aberrant, genetically regulated immune response to environmental antigens. The importance of environmental factors in eliciting disease is supported by the relatively low disease concordance in monozygotic twins and by reports that migrant populations tend to acquire the disease prevalence of the geographic area to which they move. l3S The importance of the genetic component varies among diseases; the frequency of a given disease in relatives of an index case (Le., the recurrent risk) is about 4 for rheumatoid arthritis and approaches 50 to 100 for SLE and spondyloarthropathies. Similarly, the concordance in twins is between 25% and 50% for SLE and almost lO-fold lower for rheumatoid arthritis. 136 The interaction of environmental and genetic factors at many levels determines the development and expression of an autoimmune disorder (Fig. 3_20).1 37 These considerations may explain the relatively small proportion of individuals who develop autoimmune diseases (3% to 5% of the general population), the spectrum of possible autoimmune disorders, and the heterogeneity in the clinical presentation and olltcome of many autoimmune conditions.
Genetic Predisposition The human and animal genomes contain mutations that affect the control of the immune response. That these allelic variants act in combination explains both the difficulty in mapping them and the observation that autoimmune diseases are not inherited in a simple mendelian way.
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
• FIfure 3-Z0
Pathogenic events related to development of autoimmune diseases. Genetic predisposition and environmental factors may variably act on different levels of the normal immune response leading to autoimmunity.
Almost 40 genes that predispose to autoimmunity in human and animal autoimmune diseases have been localized. There appear to be both genes that cause general defects in immune regulation, which are shared between different diseases, and genes that are unique to a given disease, which may confer susceptibility of the target organ to the autoimmune process. Some conclusions have been drawn from the analysis of murine autoimmune disease models 135 : (1) no single allele but rather a combination of genes causes the disease, and more than one combination may exist; (2) the risk of the disease depends on the number of susceptibility alleles at unlinked loci; (3) some alleles have a greater effect than others; and (4) several predisposing alleles have a weak effect and are therefore difficult to map. Only a few autoimmune syndromes have been shown to be caused by single gene mutations. These mutations usually affect genes that regulate pivotal mechanisms involved in peripheral and central tolerance. As previously mentioned, mutations of the autoimmune regulator AIRE gene leads to an autosomal recessive disease APECED,l16 whereas mutations of the FoxP3 gene (which causes complete loss of the regulatory functions of CD4+CD25+ regulatory T cells) provokes IPEX syndrome. 127 Mutations in many genes involved in regulating apoptotic pathways and IL-l production cause some monogenic autoinflammatory diseases, such as ALPS (Fas, FasL, caspase 10J, familial Mediterranean fever (MEFVJ,138 TNF-receptor-associated periodic syndrome or TRAPS (1NFRIJ,139 chronic infantile neurologic cutaneous and articular syndrome or CINCA (CIASJ),l4o and Blau syndrome (CARDl5/ NOD2J,I41 or have been associated with disease susceptibility in Crohn's disease (CARD15/NOD2J. 142 Several polymorphisms of genes related to immunologic reactivity are associated with autoimmunity. For example, polymorphisms in the gene encoding CTLA-4, the molecule involved in mechanisms of clonal anergy and immune homeostasis, are related to susceptibility to autoimmune conditions such as multiple sclerosis,I43 inflammatory bowel diseases,I44 autoimmune thyrodi-
49
tis,145 and rheumatoid arthritis. 146 An intronic polymorphism of the gene regulating the expression of another molecule involved in lymphocytic homeostasis (programmed death-1 [PDI]) has been associated with susceptibility to SLE. 147 Associations between particular MHC class I or II genes and many autoimmune diseases have been repeatedly observed. 148,149 This may be because different MHC alleles differ in their ability to present peptides to autoreactive T cells. Alternatively, some MHC alleles might enhance positive selection or decrease negative selection of autoreactive T cells in the thymus. 137 Another possible cause of autoimmunity may be related to an increased availability of autoantigens to the immune system. Mutations of genes controlling the mechanisms of clearance of apoptotic cells and nuclear material, such as Clq, predispose to a lupus-like disease. lso Finally, some genes increase susceptibility to autoimmune disease through changes in the response in the peripheral tissues. For example, some polymorphisms of the cellular receptors for the Fe portion of IgG (FcyR) are associated with susceptibility to lupus nephritis, probably by facilitating immune complex deposition in renal tissue. lSI
Environmental Fadors Infection and Autoimmunity Infection is a potentially important exogenous factor in the induction of autoimmunity. However, the mechanisms by which infections can trigger activation of autoaggressive T cells and cause tissue destruction are unknown, New insights have been provided by the observations that a single TCR may bind different peptides (so-called TCR degeneracy) and that autoreactive "ignorant" T cells are present in the circulation of normal persons. Bacterial or viral DNA or lipopolysaccharides may act as adjuvants. Coadministration of a microbial adjuvant together with the relevant antigen or peptide is necessary to induce experimental autoimmune disease. Exposure to microbial agents is important for the development of some spontaneous autoimmunity models in transgenic mice. 152 Therefore, the presence of autoreactive lymphocytes per se does not appear to be sufficient to trigger the disease; additional factors, such as infections or cyrokine release or both, seem to be needed to activate selfreactive anergic or ignorant T cells. For instance, tolerance can be broken if anergic T cells are exposed to high concentrations of IL-2 (which may theoretically occur if adjacent T cells are stimulated by another antigen) or if the inflammatory process causes efficient presentation of cryptic self-antigen on activated APCs. Epitope spreading during an autoimmune response was initially demonstrated in experimental allergic encephalomyelitis l53 induced by immunization with myelin basic protein (MBP) or its immunogenic peptides. During the course of the disease, an encephalitogenic T cell response develops not only to the original immunizing peptide but also to some cryptic epitopes within MBP. Subsequent work in other experimental models, such as nonobese diabetic (NOD) mice,I54 confirmed
50
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
these results and showed that epitope spreading can be both intramolecular and intermolecular, because determinants of other antigens at the site of inflammation, including heat-shock proteins, can also become targets of the autoimmune response. Thus, during the course of the disease, a sequence of pathogenic autoimmune responses to several cryptic epitopes develops, probably as a consequence of upregulation of processing and presentation of self-molecules in the context of inflammation. Various mechanisms, not necessarily mutually exclusive, have been hypothesized to explain how infection can trigger autoimmune diseases. Three of them (molecular mimicry, bystander activation, and superantigen activation) have been the subjects of particular attention. According to the theory of molecular mimicry, microbial antigens that share structural similarities with selfantigens can elicit a pathogenic immune response that cross-reacts with self-antigens. Many experimental autoimmune animal models are generated by hyperimmunization of genetically susceptible strains with foreign proteins that are homologous to tissue-specific selfproteins. Although in this case the triggering antigen is a homologous nonmicrobial antigen, the mechanism is equivalent to that which occurs after infection with a pathogen whose antigens, although nonhomologous, share structurally related epitopes with self-antigens. In experimental autoimmune uveitis and experimental ovarian autoimmune disease, microbial peptides induce tissue-specific autoimmunity.15s Immunization with heat-killed Mycobacterium tuberculosis in mineral oils (complete Freund's adjuvant) induces arthritis in Lewis rats. The disease can be transferred to naive, irradiated recipients with a T cell clone specific for the mycobacterial heat-shock protein 65 peptide 180-188. 156 Activation of human autoreactive T cell clones by viral and bacterial peptides has been reported. In insulin-dependent diabetes mellitus, molecular mimicry at the T cell level between glutamate decarboxylase (a pancreatic-islet autoantigen) and a coxsackievirus antigen has been demonstrated. 157 In multiple sclerosis, seven viral and a single bacterial peptide were found to stimulate at least one of seven autoimmune T cell clones specific for MBP. I 58 In patients with Lyme disease who develop chronic arthritis that is resistant to antibiotic treatment, Gross and colleagues159 demonstrated cross-reactivity between a peptide of OspA, the major surface antigen of Borrelia burgdorferi, and the self-protein LFA-l. Albani and colleagues l60 showed an abnormal immune response to a bacterial DNA) antigenic determinant carrying the "shared epitope" amino acid sequence QKRAA, which is characteristic of the HLA-DR alleles that confer susceptibility to rheumatoid arthritis. Mucosal tolerization with DNA)Pl, a peptide that induces proinflammatory T cell response CIFN-y and TNF-a) in patients with rheumatoid arthritis, was able to change T cell reactivity to production of IL-4 and IL-I0. 161 These studies demonstrated that microbial peptides with limited sequence homology are effective activators of autoreactive T cells. Moreover, they suggested the possibility that epitopes present on different microbial peptides may all be responsible for the activation of autoreactive T cells in a given autoimmune disease. In diseases in which a single pathogen has not been implicated, cross-reactive epitopes that are common to various pathogens may be important.
The bystander activation hypothesis postulates that infection-induced local inflammation-by provoking cell
death and the release of abundant quantities of self-antigens, potentiating antigen presentation and perturbing the cytokine balance-leads to bystander activation of a preexisting but controlled autoimmune response.162.163 Superantigens induce a potent polyclonal immune response and could trigger autoimmune diseases either by driving anergic autoreactive T cells out of their nonresponsive state or by facilitating activation of ignorant T cells that are specific for cryptic epitopes. 164 Superantigen-induced immune activation could also be a cause of disease exacerbation in already established autoimmune diseases.
MECHANISMS OF TISSUE INJURY IN AUTOIMMUNE-MEDIATED DISEASES The clinical and pathologic manifestations of a given autoimmune disease are determined by several factors, including the nature and location of the trigger stimulus and the type of effector mechanisms that the immune system activates to eliminate the aberrant stimulus.
General Mechanisms of Dssue Damage The effector mechanisms in autoimmunity are the same as those observed during normal immune response against microbes or other foreign antigens (Table 3-10). The majority of immune-mediated inflammatory diseases are the results of a complex combination of humoral and cellular-mediated immune responses in which multiple effector mechanisms operate.
Type I Hypersensitivity Immediate (type I) hypersensitivity is the mode of tissue damage mediated by IgE. Mast cells involved in this type of response also playa role in non-IgE-mediated autoimmune diseases. 165 However, in the vast majority of autoimmune diseases the other three types of hypersensitivity predominate.
Type " Hypersensitivity The evidence that antibodies are able to induce autoimmune disease comes from the classic experiments of induction of disease in normal animals by adoptive transfer of immunoglobulin purified from blood or tissues of affected animals. Maternal transfer of pathogenic autoantibodies to the fetus during pregnancy causes neonatal IUpUS,166 autoimmune thyroiditis,167 and autoimmune thrombocytopenia.168 Antibodies cause tissue damage by various mechanisms (Fig. 3-21). In type II hypersensitivity, antibodies opsonize the target cells and activate the complement cascade. When the opsonized cells arrive in the reticuloendothelial system of secondary lymphoid organs (especially the spleen), resident phagocytes are activated by the binding of their Fc or C3d receptors, leading to the destruction of antibody-coated cells. Antibodies can also activate the complement cascade that ultimately leads to cell lysis in the circulation. These mechanisms are responsible for
C HAP T E R
' : . TABLE 3-10
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
51
Immunopathoqeni< Mechanisms of Immune-Mediated Diseases
TYJN! of Hypersensitivity
Immune-pathologic Mechanisms
Mechanism of nssue Damage
Type 1Immediate hypersensitivity Type ll-Antibody-mediated hypersensitivity
IgE antibody
Activation of mast cells and release of vasoactive mediators Recruitment and activation of leukocytes by complement and Fc receptors Opsonization of target cells and complement activation Alteration of tissue normal function
Type III-Immune complex-mediated hypersensitivity
Tissue deposition of circulating immune complexes
Recruitment and activation of leukocytes by complement and Fc receptors
Type N-T cell-mediated hypersensitivity
Delayed hypersensitivity (CD4+ T cells) T cell-mediated cytolysis (CD8+ T cells)
Activation of tissue macrophage Direct lysis of target cells
IgM or IgG against cellular or extracellular antigens
cell destruction in autoimmune hemolytic anemia and immune thrombocytopenic purpura. Antibodies bind to tissue-specific autoantigens and recruit and activate neutrophils and macrophages through complement and Fc receptors. Tissue damage is mediated by the release of proteolytic agents from phagocytes. This occurs in acute rheumatic fever, in which antibodies directed to streptococcal cell wall antigen cross-react with myocardial antigens; Goodpasture s syndrome, in which the tissue target for autoantibodies is a noncollagenous protein in basement membranes of kidney glomeruli and lung alveoli; and pemphigus vulgaris, in which antibodies are directed against epidermal cadherin. In other circumstances, autoantibodies do not cause inflammation but interfere with cell receptor function and cause disease without eliciting tissue damage. This is the case in Graves' disease, which is caused by antibody-mediated stimulation of thyroid-stimulating hormone (TSH) receptors, and in myasthenia gravis, in which antibodies inhibits acetylcholine binding and downmodulate receptors. 169
• f1ture 3-21 Examples of antibody-mediated mechanisms of tissue or cellular damage. A, Two different modes of destruction of red blood cells (RBQ during the course of autoimmune hemolytic anemia are shown. RBC coated by specific antibodies are recognized and destroyed by maaophages of the reticular endothelial system (predominantly the spleen). Antibodies activate complement, leading to cell lysis in the drculation. B, Antibodies recruit leukocytes and activate them directly through their Fc receptors or indirectly through the complement cascade, or both.
Human or AnImal Model of Autoimmune Diseases Allergic manifestations (bronchial asthma, anaphylactic shock, urticaria) Autoimmune hemolytic anemia Autoimmune thrombocytopenic purpura Goodpasture syndrome Pemphigus vulgaris Acute rheumatic fever Antiphospholipid syndrome Neonatal lupus Myasthenia gravis Graves' disease Systemic lupus erythematosus Mixed cryoglobulinemia Rheumatoid arthritis (rheumatoid factor) Insulin-dependent diabetes Rheumatoid arthritis Multiple sclerosis Crohn's disease Autoimmune myocarditis
Type III Hypersensitivity The mechanism of tissue damage in type III hypersensitivity is similar to that described for antibodies directly bound to tissue antigens, but it is mediated by organ deposition of circulating immune complexes. The prototype of systemic immune complex-mediated disease is serum sickness, which was observed originally during passive immunization of human volunteers against diphtheria infection using serum from horses immunized with the diphtheria toxin. The preferential deposition of immune complexes in small arteries, with particular tropism to those structures in which the blood is ultrafiltered (e.g., kidney glomeruli, synovia), causes immune complex-mediated diseases including vasculitis, arthritis, SLE, and mixed cryoglobulinemia.169.170 In both type II and type III hypersensitivity, antigenspecific helper T cells are involved in the activation and maturation of B cells for the subsequent production of high-affinity autoantibodies.
52
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
Type IV Hypersensitivity In type IV (delayed-type) hypersensitivity (DTH), T cells are responsible for tissue damage. In most instances, damage is induced by the secretion (by the Thl subset of CD4+ T cells and by CD8+ T cells) of cytokines that activate tissue macrophages (lFN-y) and induce inflammation (TNF-a) (see later discussion). Occasionally, self-reactive CD8+ CTLs directly kill target cells (see Fig. 3-16). The finding of T lymphocytes and macrophages in close anatomic proximity in tissues and the predominant Thl orientation of T cells isolated from tissue lesions are considered clear evidence of the involvement of a DTHlike reaction in the determination of tissue damage in many autoimmune disorders. Diseases in which DTH plays a major role are multiple sclerosis,171 insulin-dependent diabetes mellitus, 172 Crohn's disease, 173 rheumatoid arthritis,174 and juvenile idiopathic arthritis (JIA).175.176 In the two last conditions, the dramatic hyperplasia of synovial tissue is also caused by infiltration of the lining layer with monocyte-derived macrophages recruited from the circulation. These cells, together with the resident fibroblastlike cells, play the major role in the determining cartilage and bone destruction. J77 In addition to the predominant role of DTH in inflammatory arthritis, a significant infiltration of B cells can also be found in inflamed synovial tissue, with a tendency to a discrete lymphoid organization and with formation of germinal center-like structures. 17S Moreover, a clear infiltration of immunoglobulin-producing plasma cells is consistently found in synovial tissue, even in clear proximity with macrophages and fibroblasts of the lining layer. Figure 3-22 is a schematic representation of the complex network of various cell type interactions in determining tissue damage in a classic model of inflammatory-mediated tissue damage observed in the chronic synovitis of idiopathic arthritides. There is little evidence for a direct role of CD8+ CTLs in the pathogenesis of human autoimmune diseases. This mechanism has been postulated in myocarditis associated with coxsackievirus infection, in which autoreactive CTLs have been shown to kill also myocardial cells not affected by the viral infectionY9
Recruitment of Leukocytes Into Inflamed TIssue One of the critical mechanisms leading to the initiation and maintenance of tissue inflammation is the migration of leukocytes from the circulation to the site of inflammation. This multistep process involves many soluble and surface molecules leading to the attachment of circulating cells to endothelial cells and migration through the endothelium (Fig. 3-23). 180 The capture and rolling of circulating cells by activated endothelium is followed by the activation of cells and their firm adhesion to endothelium and, finally, by the migration of cells across the endothelium (diapedesis).180.IHI A pro-inflammatOly stimulus (trauma, an infectious or other exogenous agent) leads to the production of pro-inflammatOly cytokines (TNF-ct, IL-1) by the resident cells of innate immunity. These cytokines induce the expression of a series of membrane glycoproteins (selectins) that act as adhesion molecules for circulating leukocytes 182 ,183 (Table 3-11). In particular,
endothelial (E)-selectin and platelet (P)-derived selectin are selectively expressed on the surface of cytokine-activated endothelial cells. Circulating leukocytes flowing in the bloodstream first loosely adhere to the endothelium through other constitutively expressed surface glycoproteins (Sialyl Lewis x for E-selectin, P-selectin glycoprotein ligand 1 [PSGL-IJ for P-selectin), which allows the sampling of the local environment for signs of inflammation. Similarly, leukocyte (L)-selectin is expressed on monocytes and neutrophils and naive T cells. At least three different ligands for L-selectin (glycosylationdependent cell adhesion molecule-l [GlyCAM-IJ, CD34, and mucosal addressin cell adhesion molecule-l [MadCAM-lD are expressed on endothelial cells in various organs and conditions. The engagement of PSGL-l or L-selectin with its respective endothelial ligands enhances the strength of adhesion and induces the rolling of leukocytes on endothelium. 1s4 Notably, the lack of one enzyme needed for the expression of the carbohydrate ligand CSialyl LewisX) for P- and E-selectins present on PSGL-l and on other selectin ligands results in a syndrome characterized by a significantly impaired leukocyte recruitment and increased susceptibility to infections, called type 2 leukocyte adhesion defiCiency CLAD-2).
The second step of leukocyte migration requires their firm adherence to endothelial cells. This is ensured by integrins that are constitutively expressed on circulating leukocytes. 182,183 Integrins are a large family of heterodimeric proteins composed of two noncovalently linked polypeptide chains, a and ~ (see Table 3-11). The N-termini of both chains form a globular head that interacts with specific ligands, whereas the cytoplasmic domain binds to the cell cytoskeleton. These surface proteins mediate a large number of interactions with other cells and with the extracellular matrix and are involved in many cellular functions, including cell activation, recognition, and migration. 185 After rolling, leukocytes undergo integrin affinity maturation stimulated by a series of specific cytokines (chemokines) produced by activated endothelial cells and by other cell types (epithelial cells, fibroblasts) of the inflamed tissue (see later discussion). In the meanwhile, pro-inflammatory cytokines (lL-l, TNF-a) mediate the overexpression of the ligands that are specific for highaffinity integrins. In this way, a firm adhesion between leukocytes and endothelium occurs. During inflammation, the very late antigen-4 (VLA-4, or a 4Pl) is selectively expressed on leukocytes and mediates their adhesion to activated endothelial cells expressing its ligand, vascular adhesion molecule-l (VCAM-1).186 Similarly, the leukocyte function-associated antigen-l (LFA-l, or CD11aCD18) binds to its specific ligand, the intracellular adhesion molecule (ICAM). A mutation of the gene coding for the ~ chain of LFA-l leads to a genetic disease characterized by recurrent bacterial and fungal infections and defective leukocyte accumulation at sites of infection, named type 1 leukocyte adhesion defiCiency (LAD-I). The specific inhibition of integrins involved in leukocyte recruitment is a possible therapeutic target in autoimmune diseases. ls7 The final step in leukocyte recruitment is the transmigration of cells across the endothelial lining into the site of inflammation. This process is facilitated by interaction between other integrins expressed on leukocytes and their specific ligands, which are present at the level of the adherence junction between the endothelial cells
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
53
• fIture 3-ZZ Pathogenic events leading to tissue damage during chronic synovitis. Various cell types are involved in the disease process at the level of synovial membranes. A, Effector Tcells express cytokines that activate maaophages to produce pro-inflammatory cytokines, growth factors, and other soluble products leading to tissue damage. 8, Resident fibroblast-like cells proliferate and became activated. C, Bcells may cluster in follicle-like structures, with maturation of a large number of antibody-secreting plasma cells. 0, Pro-inflammatory cytokines produced by inflammatory cells activate endothelial cells for recruitment of leukocytes from peripheral blood. E, Growth factors and proteolytic enzymes such as matrix metalloproteinases (MMPs) sustain the continuous remodeling of extracellular matrix (EM) and formation of new blood vessels. These are the two key mechanisms for the continuous growth of synovial tissue (hyperplasia), which leads to the progressive invasion and destruction of joint cartilage and bones. F, Mechanisms involved in the control of tissue inflammation ultimately lead to synovial fibrosis. HMGB-1, high mobility group box chromosomal protein 1; IFN, interferon; IL, interleukin; LTB4,Ieukotriene B4; NO, nitric oxide; PGE2, prostaglandin E2;TNF, tumor necrosis factor.
• FIglIre 3-ZJ Recruitment of leukocytes into the inflammatory site. (1) Pro-inflammatory cytokines produced by macrophages stimulate expression of adhesion molecules (selectins and integrin ligands) on the endothelial cells. (2) Chemokines produced by stromal cells, inflammatory cells, and endothelial cells attract leukocytes bearing.spedfic chemokine receptors. (3) Leukocytes bearing selectin ligands (Le., Sialyl LewisX ) weakly adhere to endothelium (rolling). (4) Leukocytes undergo integrin affinity maturation stimulated by chemokines. In this way, afirm adhesion between leukocytes and endothelium occurs (adhesion). (5) Cells transmigrate across the endothl!liallining into the site of inflammation. ECM, extracellular matrix; ICAM, intracellular adhesion molecule; IL, interleukin; JAM, junctional adhesion molecule; LFA-l, leukocyte function-assodated antigen 1; TNF, tumor necrosis factor.
54
I!:..
C HAP T E R
lABl£ 3 II
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
Adh('~ioll M()I(,(IIIl'~
Family
Name
Cell DIstribution
Ugands
Main Functions
Selectins
P-selectin (CD62P) E-selectin (CD62E) L-selectin (CD62L)
Endothelium', platelets Endothelium' Leukocytes
Sialyl Lewisx PSGL-l GlyCAM-l, CD34, MadCAM-l
Initiate leukocyte-endothelium interactions
VLA-l, -2, -3 VLA-4, -5 VLA-6 CD51CD29
Leukocytes Leukocytes Leukocytes Leukocytes
Laminin, collagens JAM-B Fibronectin, lamlnin Vitronectin, fibronectin
Cell matrix-adhesion, homing to inflamed tissues
LFA-l Mac-l
Leukocytes Leukocytes
lCAM-l, -2, -3 JAM-A
Leukocyte adhesion to endothelium Interaction between T cell and APC
Vitronectin receptor
Leukocytes, endothelium, osteoclasts
Fibronectin, fibrinogen, osteopontin, vitronectin, thrombospondin
Cell matrix-adhesion, leukocyte activation, osteoclast activation, angiogenesis
CD49CDlO4
Leukocytes
Laminin
Cell matrix-adhesion
~;av
Leukocytes, endothelium
Vitronectin
Cell matrix-adhesion, angiogenesis
~6Uy
Leukocytes
Fibronectin
Cell matrix-adhesion
~ll6
~7a4
LPAM-l
Leukocytes
VCAM-l, fibronectin
Homing to lymphoid tissues
Immunoglobulin superfamlly
ICAM-l (CD54) lCAM-2 (CDlO2) VCAM-l (CD106) PECAM (CD3l)
Endothelium' Dendritic cells Endotheliumt Leukocytes, endothelium
LFA-l, Mac-l LFA-l VLA-4 PECAM, Uy~3
Cell adhesion, ligands for integrins
Cadherins
VE-cadherin
Endothelium lateral junctions
VE-cadherin
Cell-to-cell adhesion
•Activated endothelial cells. tResting endothelial cells. APC. antigen-presenting cell; CD, cluster of differentiation; GlyCAM-I, glycosylation-dependent cell adhesion molecule-I; tCAM, intracellular adhesion molecule; JAM, junctional adhesion molecule; LFA, leukocyte function-associated antigen; Mac-I, macrophage adhesion molecule 1; MadCAM-I, mucosal addressin cell adhesion molecule-I; PECAM-I: platelet/endothelial cell adhesion molecule; PSGL-I, P-selectin glycoprotein ligand-I; VCAM-I, vascular adhesion molecule-I; VE-cadherin: vascular endothelial cadherin; VLA, very late antigen.
(platelet/endothelial cell adhesion molecule [PECAM-l] and the junctional adhesion molecule GAM] family) or in the subendothelial extracellular matrix (fibronectin, osteopontin, collagen) (see Table 3-11).181 The main stimuli for the migration of inflammatory cells through interendothelial spaces, however, are inflammatory chemokines that specifically attract adherent leukocytes to the infection site toward a chemical concentration gradient. Chemokines (chemotactic cytokines) are a large family of structurally homologous small cytokines (8 to 15 kD) that control the trafficking of leukocytes. 188.189 Chemokines are classified into four families according to the motif displayed by the first two cysteine residues (C) at the N-terminus: CC, CXC, C, and C2SC (Table 3-12). The two main subfamilies of chemokines, CXC and CC, differ from each other by the presence of one amino acid (X) between the cysteine residues. Chemokines are produced by leukocytes and by several types of tissue cells (endothelial cells, fibroblasts, epithelial cells) in both physiologic and pathologic states. Chemokines can be broadly
classified in two functional groups: inflammatory and lymphoid (or homing).189 Inflammatory chemokines are produced by leukocytes and tissues in response to a pro-inflammatory stimulus. Homing chemokines such as CCL19, CCL21, or CXC13 are constitutively expressed in the microenvironment of lymphoid tissues, skin, and mucosa. They are involved in the control of leukocyte trafficking between circulation and lymphoid structures. l90 Chemokines of both subgroups bind to heparan sulfate-containing proteoglycans on endothelial cells and are displayed in this way to circulating leukocytes expressing the specific chemokine receptors on their surfaces (see Fig. 3-23). These receptors have a characteristic structure with seven-transmembrane a-helical domains coupled to trimeric guanosine triphosphate-binding proteins. Seventeen CXC and 12 CC chemokine receptors have been identified to date (see Table 3-12), The interactions between chemokines and their respective cellular receptors induce the activation of intracellular signaling pathways that provoke cytoskeletal rearrangements by stimulating alternating polymerization and depolymerization of actin filaments, eventually leading to cell mobilization. 191 Although the
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
receptors exhibit overlapping specificity for chemokines within each subfamily, the expression of different chemokine receptors on the surface of circulating leukocytes is closely related to their specific function, state of differentiation, and degree of activation (see Table 3-12). This complex network of chernokines and cellular receptors allows the selective recruitment of circulating leukocytes to peripheral or lymphoid tissues on the basis of their actual discrete microenvironment and functional needs.
Many inflammatory chemokines and leukocytes displaying their specific surface receptors have been identified in inflamed tissues. In particular, some receptors for inflammatory chemokines (CCRl, CCR2, CCR5, and CXCR3) are regularly detected in tissues where inflammation is characterized by chronic infiltration of macrophages and predominantly Thl-oriented lympho-
.:. rABLE
3 - 12
55
cytes (Le., rheumatoid arthritis and Crohn's disease).192 Conversely, expression of CCR3 and CCR4 characterizes the leukocyte infiltration of allergic diseases, 193 and chemokine receptors for CXCLB (previously known as IL-8), CXCRl, and CXCR2 are most frequently found in acute inflammation. l88 The selective blockade of inflammatory cheniokines and their cellular receptors is a promising possible strategy for the treatment of many autoimmune disorders. 194
Cytokines and Growth Factors TNF represents the prototype of a pro-inflammatory cytokine produced by activated phagocytes. l 9S-197 TNF-a
was originally identified as a soluble molecule, present in
Chemokines and (hemokinl' Rl'( ('plots
Major Sources
Receptor
Cells Attradecl
Main Fundlons
CXCLl (Gro-a) CXCL2 (Gro-~) CXCL3 (Gro-y) CXCLS (ENA78) CXCL6 (GCP-2) CXCL7 (NAP-2) CXCLS OL-8) CXCL9 (MIG) CXCLlO OP-IO) CXCLlI 0-TAC) CXC112 (SDF-I) CXCLl3 CECA-I)
M, M, M, M, M, M, M, M, M, M, Sc Sc
CXCR2 CXCRZ CXCR2 CXCR2 exCRl CXCR2 CXCRl/2 CXCR3 CXCR3 CXCR3 CXCR4 exCRS
N, M, Tc, NK N, M, Tc, NK N, M, Tc, NK N N, M, Tc, NK N N, M, Tc, NK Tc, NK, M Tc, NK, M Tc, NK, M NaIve Tc, Bc B cells
Leukocyte recruitment Leukocyte recruitment Leukocyte recruitment Leukocyte recruitment Leukocyte recruitment Neutrophil activation, angiogenesis Neutrophil recruitment and activation Leukocyte recruitment Leukocyte recruitment, Thl response Leukocyte recruitment Lymphocyte recruitment Lymphocyte homing to lymphoid organs
Class CC chemoldnes CCLl 0-309) CCL2 (MCP-I)
M, Tc, Ec M, F
CCR8 CCR2
M, Tc M, NK, Tc, F
CCL3 (MIP la) CCL4 (MIPl~) CCL5 (RANTES)
M, Tc, Mc, F M, Mo, N, Ec Tc, Ec, P
CCRI/S CCRS CCRl/5/3
M,NK,B, Dc M, NK, Tc, Dc M, NK, Tc, B, E, Dc
CC1.7 (MCP-3) CCLS (MCP-2) CCLII (Eotaxin) CCLl3 (MCP-4) CCLl7 (TARC) CCLl9 (ELC)
M, F, P, Ec M, F, Ec Ec, M, Ep, Tc Ec, M, Ep Ec, M, Ep Sc, Ec
CCRI/2 CCR2 CCR3 CCR2/4 CCR4/8 CCR7
E, B,NK E, B E, M, Tc (Th2) E, B, Tc E, B, Tc Tc, Dc
CCL20 (MIP-3a) CCl21 (SLC)
M, Tc, DC, E, Mc Sc, Ec
CCR6 CCR7
Tc, Dc Tc, Dc
CCL22 CCL25 CC127 CCl28
Ec, M, Ep Ep K, Ec Ep
CCR4 CCR9/11 CCRIO CCR3/IO
E, B Tc Tc Tc
Leukocyte recruitment Activate macrophage and basophils, Th2 response Leukocyte recruitment, Thl response Leukocyte recruitment, HIV coreceptor Activation of basophils and Tc, chronic inflammation Leukocyte recruitment Leukocyte recruitment Allergy, Th2 response Leukocyte recruitment Tc and basophil recruitment Lymphocyte and Dc recruitment in lymphoid organs Lymphocyte and Dc recruitment Lymphocyte and Dc recruitment in lymphoid organs Tc and basophil recruitment Tc migration Tc migration to skin Tc migration to skin
XCL1 Oymphotactin)
Tc
XCRI
Dc, NK, Tm
Lymphocyte trafficking and development
Class ~C chemoldnes CX I eLl ( ractalkine)
M, Ec, Mig
C~CRI
M, Tc
Leukocyte-endothelium adhesion, brain inflammation
N. . (previous name)
Class CXC chemoldnes
(MDC) (TECK) (CTACK) (MEC)
F, Ec F, Ec F, Ec F, Ec F, Ec F, Ec Mo, F, Ec Tc, F, Ec Tc, F, Ec Tc, F, Ec
Class C chemoldnes
B, Basophils; Be, B cells; Dc, dendritic cells; E. eosinophils; Ec, endothelial cells, Ep, epithelial cells, P, fibroblasts; HIV, human immunodeficiency virus; K, keratinocytes; M, macrophages; Me, mast cells; Mig, microglial cells; Mo, monocytes/macrophages, N, neutrophils; NK. natural killer cells; P, platelets; Sc, stromal cells; Tc, T cells; Thl, type I helper T cells; Th2, type 2 helper T cells; Tm, thymocytes.
56
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
the sera of animals treated with LPS, that displayed the ability to cause tumor necrosis in ViVO. I98 Although several cell types can produce TNF-(X (T cells, NK, cells, mast cells), macrophages are the most important source of this cytokine, especially during infection with gram-negative bacteria. After cell activation, TNF-(X is synthesized as a membrane protein that is expressed as a homodimer. It is cleaved by membrane-associated metalloproteinases and released as a 17-kD polypeptide, three molecules of which polymerize to form a 51-kD TNF protein. The various biologic actions of TNF are mediated by two distinct receptors; the 55-kD TNF receptor I (TNFRI or p55 receptor) and the 75-kD TNFRII (or p75 receptor).I96 The binding of circulating TNF-(X to TNFRs leads to the recruitment of cytoplasmic proteins called TNF receptor-associated factors (TRAPs). TRAPs initiate the intracellular signaling that leads to the activation of transcription factors, such as NF-lCB and AP-1, which ultimately cause the production of inflammatory mediators and anti-apoptotic proteins (Fig. 3-24A). Notably, in the case of TNFRI, the binding with TNF-(X may lead to either inflammation or apoptosis. In the latter case, different signaling proteins (TNF receptor-associated death domain [TRADD]) are involved. Activation of this particular intracellular pathway leads to the activation of caspases, which eventually results in cell apoptosis (see Fig. 3-24A). This latter mechanism is similar to Fas-mediated apoptosis involved in activation-induced cell death (see Fig. 3-18) and represents an important strategy for self-limitation of cell activation. l99 The principal function of TNF-(X during inflammation is to stimulate the recruitment of phagocytes into the site of inflammation and promote the killing of microbes (Table 3-13). TNF-(X also induces the expression of adhesion molecules and chemokines by endothelial cells and
enhances the affinity of leukocyte integrins for their ligands. It can activate recently recruited monocytes and stimulate the pro-inflammatory activity of resident fibroblasts (see Fig. 3-22).197 When produced in large amounts, TNF-(X may enter the bloodstream and act at distant sites as an endocrine hormone. In this way, TNF-(X is able to stimulate the hypothalamus to induce fever, to act on hepatocytes for the production of acute phase reactants, and to promote metabolic changes leading to wasting of muscle and fat cells (cachexia). Very high levels of circulating TNF-(X (greater than 10-7 M) play a major role in the pathogenesis of septic shock (e.g., severe systemic hypotension, disseminated intravascular coagulation), which is induced by massive LPS-induced production of pro-inflammatory cytokines. Excessive production of TNF-(X plays a central role in macrophage activation syndrome, a complication mainly observed in patients with systemic JIA. 200 The potentially lethal effects of TNF-(X are balanced by a strategy for the downregulation of TNF activity that is related to the shedding of TNFRs from the surface of activated cells, which generates circulating soluble receptors that prevent the binding of free TNF-(X to cell-bound receptors (see Fig. 3-24A). This strategy has been successfully adopted for the therapeutic blockade of TNF activities. 196 IL-1 shares many biologic function with TNF. 201 Like TNF, its major source is activated macrophages, although neutrophils, epithelial cells, and endothelial cells can also produce IL-1. Two different isoforms of circulating IL-1 CIL-1(X and IL-1~) are known. Both are synthesized as 33-kD peptides. Biologically active IL-1(X is a 17-kD protein that is released from the cell after cleavage by an intracellular cysteine protease called caspase-l (preViously
IL·\
/
FADD
G)
TRADD ~ It
,
\\
Caspases cascade \,
Apoptosls
~ TRAF ,
AP·I
'o,~
~'j
No Signal
.-
J
Jt'l
"'"
fL.J Ra
~
,IRAK
No Sional '"
Signal
\ Gene tran.•crlplion \, Inj/ammatlon Survival signals
• Rgun 3-Z4 Mechanisms of cell activation induced by pro-inflammatory cytokines and strategies for their downregulation. Panel A: (1) The binding of circulating TNF-a to TNF receptors (TNFR) leads to the recruitment of cytoplasmic proteins, called TNF receptor-associated factors (TRAFs), that initiate the intracellular signaling. DD, death domain. (2) In the case ofTNFRl, the binding with TNF-a may lead to either inflammation or apoptosis. In the latter case, different signaling proteins (Fas-associated death domain IFADDJ) are involved. The activation of this particular intracellular pathway, due to the loss of survival signals, leads to the activation of caspases that eventually result in cell apoptosis. (3) TNFRs are shed from the surface of activated cells, and free TNF-a binding to cell receptors is prevented. Panel 8: Type I receptor for interleukin-1 (IL-1) is constitutively expressed on many cell types and mediates intracellular transmission of the signal after binding with soluble IL-1, through activation of the IL-1 receptor-associated kinase (IRAK), which eventually leads to cell activation. Type II receptor (decoy receptor), is expressed only after cell activation and lacks a cytoplasmic tail. Thus, the binding with IL-1 does not result in intracellular signal transmission. Activated macrophages also secrete a protein with a close structural homology to IL-1 that binds to the same surface receptors but is biologically inactive (IL-1 receptor antagonist ilL-RaJ).
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
known as IL-ln converting enzyme). IL-l~ may exert its biologic function either as 33-kD precursor or as a smaller cleaved product. Although the two isoforms of IL-l display a low degree of homology, they bind to the same surface receptors (lL-IR) and mediate the same biologic activities. 202 There are two different membrane receptors for IL-1. Type I receptor is constitutively expressed on many cell types and mediates the intracellular transmission of the signal after binding with soluble IL-l, through the activation of the IL-l receptor-associatedkinase (lRAK), which eventually leads to activation of NF-KB and AP-l transcription factors (see Fig. 3-24B).202 Type II receptor, which is expressed only after cell activation, lacks a cytoplasmic tail; consequently, binding with IL-l does not result in intracellular signal transmission. The major function of this receptor is to downmodulate the biologic action of IL-l; it acts as a "decoy" receptor in competition with the type I receptor (see Fig. 3-24B).203 A second strategy also downmodulates IL-l activities. Activated macrophages secrete a protein with a close structural homology to IL-l that binds to the same surface receptors but is biologically inactive (lL-l receptor antagonist, IL-Ra) (see Fig. 3-24B).204 This natural pathway of regulation of IL-l biologic activity has been adopted with the use of recombinant IL-IRa in the treatment of rheumatoid arthritis. 205 ,206 The biologic functions of IL-l largely overlap, both locally and systemically, with those described for TNF (see Table 3-13). Interleukin-6 (lL-6) is another important eytokine mainly produced by macrophages, endothelial cells, and tissue fibroblasts during acute and chronic inflammation. 207 ,20R Its biologic actions are mediated by specific binding with an IL-6 receptor that is present on cell membranes or in soluble form; this complex then binds to a signal transducing subunit, called gp130, which is also involved in signal transduction for other cytokines.209.210
11'1 • -
fABLE 3-13 Main Systl'l11i< Biologic Effects of IL-1. I NF ", 'Illd IL 6 During Inflammation
IL-l
TNF-a
IL-6
CNS Fever Production of CRH
++ +
+ +
+++ ++
Liver Acute phase reactants
+
+
++
Endothelium Chemokine expression AdheSion molecules Neoangiogenesis
++ +++ ++
++ +++ +++
+
Bone marrow Myelopoiesis Thrombocytopoiesis Inhihition erythropoiesis
++
++
++
+
++ +++ ++
CNS, ct'ntml nervous system; CRH, corticotrophin-releasing hormone; IL. interleukln; INF, tumor necrosis factor. -, none; +. moderate; ++, intense; +++, strong.
57
IL-6 has many pro-inflammatory functions (see Table 3-13). It stimulates the synthesis of acute phase reactants by the liver and the production of neutrophils from bone marrow. It induces endothelial cells activation, fibroblast proliferation, and osteoclast activation. In adaptive immunity, IL-6 stimulates the growth of B cells that have differentiated in antibody producers. 208 During the last decade, a large body of evidence has stressed the pivotal role of this cytokine in systemic JIA. In fact, many of the clinical features peculiar to this disease (chronic anemia, severe growth retardation, osteoporosis, thrombocytosis, amyloidosis) have been related to the biologic actions of IL_6. 211 - 214 Recently, the use of anti-IL-6 receptor monoclonal antibodies has been shown to be a possible promising therapeutic option for systemic JIA. 215 In recent years, a growing interest has been focused on a proinflammatory cytokine named high mobility group box chromosomal protein 1 (HMGBl).216 This nuclear protein stabilizes nucleosomes, allowing binding of DNA and facilitating gene transcription. It is passively released by necrotic cells after tissue injury. Binding of HMGBI to unactivated macrophage/ monocytes stimulates translocation of NF-KB, with the subsequent production of pro-inflammatory cytokines that, in turn, stimulate further HMGBI production, The possible crucial role of HMGBI in sepsis, lung inflammation, and arthritis has been proposed. 217 Cells of innate immunity isolated from inflamed tissues characterized by a prevalent DHT response also produce other cytokines that have pivotal roles in the cross-talk between cells of the innate and adaptive immune responses, For instance, IL12, IL-18, and IL-23 are key inducers of cell-mediated immunity that may contribute to the differentiation of recently recruited CD4+ helper T cells into IFN-y-producing Thl cells. 21 B-220 Similarly, IL-15 produced by macrophage may stimulate T cells to drive further TNF-a production by macrophages through direct cell contact (see also Fig. 3-22).221
During chronic inflammation, a large number of growth factors are also secreted by inflammatory cells. Platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), TGF-~, epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF) have been shown to play a part in the induction of angiogenesis. 222 Angiogenesis is a physiologic process that consists of the formation of new blood vessels from a preexisting microvascular bed. During chronic inflammation, this process proVides oxygen and nutrients necessary for the high metabolic requirement of resident cells and permits the migration and progressive infiltration of newly recruited inflammatory cells. In inflammatory arthritis, the growth of inflamed tissue (synovial pannus) has been compared with that of a solid tumor because its unregulated outgrowth leads to the invasion and destruction of normal tissues (e.g., joint cartilage, bone). Angiogenesis supports the extensive vascularization that occurs during the proliferation of rheumatoid pannus (see also Fig. 3-22).223 VEGF is one of the most potent endothelial cell-specific mitogens locally produced by activated monoeytes/macrophages and tissue fibroblasts on stimulation by a number of pro-inflammatory cytokines (lL-l, TNF-n, IL-6), other inflammatory mediators (e.g., PGE 2), and physical factors (e.g., hypoxia).224 The downmodulation of this growth factor has been shown to dramatically dampen the degree of inflammation in animal models of arthritis. 225
58
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
traenoic acid (5-HPETE), followed by leukotriene A4 (LTA4). LTB 4 is the most stable molecule among the leukotrienes and is the hydrolytic product of LTA4• LTB 4 is rapidly synthesized by neutrophils and macrophages on challenge with stimuli such as microbial pathogens, toxins, aggregated immunoglobulins, and pro-inflammatory cytokines. LTB 4 is one of the most powerful chemoattracrants; it induces neutrophil aggregation and degranulation and macrophage production of the pro-inflammatory cytokines, O2- and PGE/ 27
Prostaglandins and Leukotrlenes Prostaglandins and leukotrienes are acid lipids that are derived from enzymatic cleavage of arachidonic acid and are produced by most mammalian cells in response to mechanical, chemical, and immunologic stimuli. Arachidonic acid is a member of the 00-6 series of essential fatty acids contained in membrane phospholipids. Activation of the enzyme phospholipase A2 releases arachidonic acid, which is further metabolized by two main enzymatic pathways leading to the final production of a class of mediators belonging to the family of bioactive eicosanoids (Fig. 3-25), Cyclooxygenases (COX) are responsible for the production of prostaglandins both in physiologic and pathologic conditions. Two different isoforms of COX are currently known. COX-l is constitutively expressed in most tissues at a constant level throughout the cell cycle. Prostaglandins are produced in many tissues and regulate many functions, including platelet-dependent homeostasis, renal blood flow, and gastric mucosal integrity. Conversely, COX-2 is usually undetectable in normal tissues but can be rapidly induced by particular cell types (fibroblasts, monocytes, and endothelial cells) on pro-inflammatory stimulation. Activation of COX-2 is thought to playa major role in the inflammatory reactions. 226 Together with the mast cell-derived PGD 2 , the most abundant COX-2 product is PGE 2 . It can sensitize nerve endings to painful chemical and mechanical stimuli, and it also acts as a potent vasodilator. Furthermore, PGE? has a crucial role in the induction of fever after stimulation of specialized endothelial cells in hypothalamic tissue by endogenous pyrogens such as TNF and 1L-6. Leukotrienes are derived from the combined actions of 5-lipoxygenases (5-LOX) and 5-LOX-activating protein (FLAP), with initial formation of 5-hydroperoxyeicosate-
The study of the metabolism of arachidonic acid in inflammatory responses led to the discovery of lipid molecules called lipoxins (LXs), which are derived from different enzymatic cascades that are efficient endogenous mediators of inflammation resolution. 218 Lipoxins are produced after activation of two distinct intracellular pathways, depending on the cell involved and the mode of activation. As previously mentioned, the interaction between leukocytes and platelets at the site of tissue int1ammation leads to the activation in the latter cells of S-LOX, which generates LTA4• However, the activation of adherent platelets leads also to the activation of I2-LOX, which ultimately results in the production of lipoxin A4 and B/ 29 A second pathway of lipoxin production occurs in monocytes and macrophages that are exposed to anti-inflammatory cytokines such as IL-4 and IL-13. In this case, lipoxin production is initiated by a IS-LOX that leads to the generation and release of ISS-hydroxyeicosatetraenoic acid (HETE), which is rapidly taken up and converted by polymorphonuclear cells to lipoxins. 229 Therefore, in peripheral blood neutrophils, a switch in eicosanoid biosynthesis occurs, from predominantly pro-inflammatory molecules (PGE 2 and LTB4) to anti-inflammatory lipoxin, Notably, PGEs itself promotes both S-LOX and I2-LOX gene expression, thus initiating a mechanism of self-limitation of inflammation. 128
Lipoxins display a number of anti-inflammatory activities in many animal models of inflammatory diseases, mainly related to inhibition of recruitment of inflammatory cells into the site of inflammation. They have been
• Figure 3-Z5 Metabolic pathways of lipid-derived proinflammatory and anti-inflammatory mediators (eicosanoids). See text for explanation. COX, cydooxygenase; HETE, hydroxyeicosatetraenoic add.
Membrane phospholipid
I
Phospholipase A,
Arachidonic acid 5-LiPOXygena;/
L T8 4 + - -
/ Inflammation
Leukotriens (LTA 4)
!
COX-l
Prostaglandins 15-LiPOXyg7
12-LiPoxygenase
Lipoxins (LXA4, LXB4)
Control of inflammation
~OX-2
15S-H (HETE)
~poxygenase
(POE 2, P00 2)
~
Inflammation
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
59
shown to stimulate the nonphlogistic phagocytosis of apoptotic neutrophils by macrophages. 23o
THE AUTONOMIC NERVOUS AND ENDOCRINE MEDIATORS IN INFLAMMADON
Nitric Oxide and Reactive Oxygen Products
Many studies have demonstrated the role of the nervous and endocrine systems in the maintenance and control of acute and chronic inflammation. The autonomic nervous system and the hypothalamic-pituitary-adrenal (HPA) axis act cooperatively to sustain and regulate tissue inflammation. Similarly, gonadal hormones are known to actively influence the immune response, estrogens being generally implicated as immune response stimulators and androgens as natural suppressors,238.239 The noradrenergic innervation of lymphoid organs and the expression of ~2-adrenergic receptors on lymphocytes, NK cells, and monocyte/macrophages suggest that the autonomic nervous system may influence the immune response, Peripheral tissues are densely innervated by postganglionic sympathetic adrenal fibers and unmyelinated C fibers containing substance P and calcitonin gene-related peptide. 240 Peripheral afferent (sensory) neurons (PAN) are stimulated by several inflammatory mediators, including prostaglandins, bradykinin, leukotrienes, IL-1, IL-6, and neural growth factor, which act as sensitizers for the transmission of pain to the central nervous system via the nociceptive type IVC fibers that pass through the dorsal root to the spinal cord. All these sensitizers act by increasing the intraneuronal concentration of cyclic adenosine monophosphate (cAMP), which can in turn be modulated by morphine/endogenous opioids. 241 Normally, PAN do not communicate with the peripheral efferent postganglionic sympathetic neurons (PGSN) that innervate peripheral tissues. However, dUring inflammation, sensitization of PAN leads to the activation of PGSN, which may ultimately lead, through a vicious circle, to persistent inflammation and pain, mainly because of the release of neuropeptides (e.g., substance P) that promote vasodilatation, plasma extravasation, and leukocyte recruitment and activation. The sympathetic stimulation leads to a negative feedback with release of norepinephrine and other anti-inflammatory mediators (e.g., neuropeptide Y) that are able to suppress neurogenic inflammation. 241 Inflammation leads also to activation of the hypothalamic-pituitary-adrenal axis. 239 Therefore, inflammatory stress can affect immune function both via peripheral responses mediated by sympathetic innervation and via central activation of the HPA axis. The activation of the HPA axis through the overproduction of corticotropin-releasing hormone (CRH) and adrenocorticotropin (ACTH) leads to the adrenal production of anti-inflammatory glucocorticoids (GC). Glucocorticoid effects are mediated by their ability to modulate gene expression. GC act by binding to their specific receptors (GCR), which exist as a and ~ isoforms. 242 The binding of GC with GCRa leads to translocation of the GC/GCRa complex to the nucleus, where it interferes with the activation of genes inducible by AP-1 and NF-KB, such as those for IL-2, IL-6, IL-8, IL1-~, and TNF-a, and with T cell proliferation. Conversely, GCR~ is devoid of signal transmission after GC binding and can act as an inhibitor of GCRa. Enhanced GCR~ expression
Nitric oxide (NO) is produced by mammalian cells by the action of nitric oxide synthase (NOS).231 The production of NO plays an integral part in the maintenance of several physiologic functions involved in the control of cardiovascular, respiratory, and nervous systems. During inflammation, inducible NOS is activated by pro-inflammatory cytokines (IFN-y, IL-17, TNF, IL-1). This enzyme catalyzes the production of NO and L-citrulline from Larginine and molecular oxygen. The induction of NO under conditions of immune activation can be both beneficial, in the eradication of microorganisms, and harmful, by causing tissue damage. 232 Similarly, generation of reactive oxygen products is essential for the microbicidal activity of phagocytes and in directly mediated tissue damage in acute and chronic inflammatory conditions. Their generation is mediated by the activation of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, an enzyme located in the plasma membrane of phagocytes. The activation of NADPH oxidase leads to the formation of superoxide (0 2-), a univalently reduced oxygen radical. Superoxide spontaneously dismutases into hydrogen peroxide (H 20), which may react with chloride ions to form the toxic hypochlorous acid (HOC!) in a reaction catalyzed by myeloperoxidase. This latter enzyme is abundantly present in the preformed granules of phagocytes, and it is rapidly released after cell activation. 233
Proteolytic Enzymes Myeloperoxidase and a number of other proteolytic enzymes are abundantly present in the granules of professional phagocytes, such as neutrophils. 234 ,235 Among them are serine proteinases (elastases, cathepsin G), acid hydrolases (~-glucuronidases, a-mannosidases), and other peptides with bactericidal activity (lysozyme, defensins, lactoferrin, azurocidin). Monocytes and macrophages do not possess preformed granules, but they possess lysozymes that contain preformed enzymes or may actively synthesize them after activation. Matrix metalloproteinases (MMPs) comprise a large family of proteolytic enzymes produced by fibroblasts, macrophages, neutrophils, and chondrocytes on stimulation with pro-inflammatory cytokines and growth factors. 236 Their main function is the remodeling of extracellular matrix during tissue resorption. The proteolytic activity of MMPs is thought to represent a crucial component of both physiologic (embryonic development, organ morphogenesis, angiogenesis) and pathologic (chronic inflammatory diseases, tumors) conditions. 237 According to many authorities, MMPs are one of the most important classes of final mediators of tissue damage in many chronic inflammatory conditions, and their specific inhibition is considered to be a possible promising therapeutic target.
60
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
has been found on peripheral blood mononuclear cells from patients resistant to glucocorticoids. 242 ,243 The physiologic counter-regulation of GC is mediated by pro-inflammatory hormones and cytokines. Prolactin is a pro-inflammatory neuropeptide that is produced by pituitary gland but also by circulating mononuclear cells. In vitro and ex vivo studies show that prolactin antagonizes the immunosuppressive effects of GC in murine species and stimulates production of anti-DNA antibodies by normal lymphocytes. 244 Macrophage inhibitory factor (MIF) is a cytokine produced by activated macrophages in response to proinflammatory stimuli. In addition to a number of pro-inflammatory functions, such as amplification of TNF, IL-6, IL-1, and NO production by macrophages, MIF displays the unique ability to counteract the immunosuppressive effect of GC by a poorly understood mechanism. Notably, a single-nucleotide G-to-C polymorphism at position -173 of the MIF gene, which is related to an increased expreSSion of MIF by peripheral blood mononuclear cell stimulation, has been associated with systemic-onset JIA. Patients with this polymorphism required significantly longer GC treatment than did patients with the more frequent GG genotype. 245 ,246
REFERENCES 1. Abbas AK. Lichtman AH: Cellular and Molecular Immunology, 5th ed, Philadelphia. WB Saunders. 2003, 2. Janeway CA, Travers P, Walport M, Shlomchik M: Immunobiology. New York, Garland Publishing. 2001. 3, Paul WE (ed): Fundamental Immunology, 4th ed. New York. Raven Press, 1998. 4, Medzhitov R. Janeway c: Innate immunity. N EnglJ Med 343: 338-344, 2000, 5, Medzhitov R: Toll-like receptors and innate immunity. Nat Rev Immunology 1: 135-145. 2001. 6, Takeda K, Kalsho T, Akira S: Toll-like receptors. Ann Rev Immunol 21: 335--376, 2003. 7. Pasare C. Medzhitov R. Toll-like receptors: balancing host resistance with immune tolerance. Curr Opin Immuno! 15: 677-682. 2003. 8, Gordon S: Alternative activation of macrophages, Nat Rev Immunol 3: 23--35, 2003. 9, Mosser OM: The many faces of macrophage activation . .I Leukoc Bioi 73: 209-212, 2003. 10, Banchereau J. Steinman RM: Dendritic cells and the control of immunity. Nature 392: 245--252, 1998. 1I. Shortman K. Liu Y): Mouse and human dendritic cell subtypes. Nat Rev Immunol 2: 151-161, 2002. 12. Sallusto F, Lanzavecchia A: The instructive role of dendritic cells on T cell responses. Arthritis Res (SuppI 3): SI27-S132. 2002, 13, Guermonprez p. Valladeau .I, ZilVogel L, et al: Antigen presentation and T cell stimulation by dendritic cells. Ann Rev Immunol 20: 621-667, 2002. 14, Kadowaki N, Ho S, Antonenko S, et aI: Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. .I Exp Med 194: 863--869, 2001. 15, Blanco p. Palucka AK, Gill M, et al: Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Science 294: 1540-1543, 2001. 16, Ronnblom L, Alm GV: A pivotal role for the natural interferon alpha-producing cells (plasmacytoid dendritic cells) in the pathogenesis of lupus. .I Exp Med 194: 59-63, 2001, 17, Mahnke K, Knop .I, Enk AH: Induction of tolerogenic DCs: "You are what you eat." Trends Immunol 24: 646-651, 2003. 18, Colucci F, Caligiuri MA, Di Santo .II': What does it take to make a natural killer' Nat Rev Immunol 3: 413--425, 2003. 19, Moretta A, Bottino C, Vitale M, et al: Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu Rev Immunol 19: 197-223, 2001. 20, Biassoni R, Cantoni C, Marras D. et al: Human natural killer cell receptors: insights into their molecular function and structure. .I Cell Mol Med 7: 376--387. 2003. 21. Buckley CD. Pilling 0, Lord .1M, et al: Fibroblasts regulate the switch from acute resolving to chronic persistent inl1ammation, Trends Immunol 22: 199-204, 2001.
22. Molina H: Complement and immunity. Rheum Dis Clin North Am 30: 1-18, 2004. 23, Taylor PRo Carugati A. Fadok VA, et al: A hierarchical role for classical pathway complement proteins in the clearance of apoptotic cells in vivo. .I Exp Med 192: 359-366, 2000. 24, Manderson AP, Botto M, Walport MJ: The role of complement in the development of systemic lupus erythematosus. Annu Rev Immunol 22: 431-456, 2004. 25. Kemper C, Chan AC, Green .1M, et al: Activation of human CD4+ cells with CD3 and CD46 induces a T-regulatory cell 1 phenotype. Nature 421: 388-392, 2003. 26. SohnJH, Bora PS, Suk HJ, et al: Tolerance is dependent on complement C3 fmgment iC3b binding to antigen-presenting cells. Nat Med 9: 206--212, 2003 27, Kilpatrick DC: Mannan-binding lectin and its role in innate immunity. Transfus Med 12: 335--352. 2002. 28. Mantovani A, Garlanda C. Bottazzi B: Pentraxin 3, a non-redundant soluble pattern recognition receptor involved in innate immunity. Vaccine 21 (Suppl 2): S43--S47. 2003, 29. Ganz T: Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol 3: 7\0-720, 2003, 30, Frazer DT, Capra .10: Immunoglobulins: structure and function. In Paul WE (ed): Fundamental Immunology. 4th ed. New York. Raven Press, 1998. 31. Fearon DT. Carroll MC: Regulation of B lymphocyte responses to foreign and self-antigens by the CDl9/CD21 complex. Annu Rev Immunol 18: 393-422, 2000. 32. Max EE: Immunogobulins: molecular genetics. hi Paul WE (ed): Fundamental Immunology, 4th ed. New York. Raven Press, 1998. 33. Wagner SO, Neuberger MS: Somatic hypermutation of immunoglobulin genes. Annu Rev Immunol 14: 441-457, 1996. 34, Hardy RR, Hayakawa K: B cell development pathways. Annu Rev Immunol 19: 595-621, 2001. 35. Nemazee 0: Receptor selection in Band T lymphocytes. Annu Rev Immunol 18: 19-51. 2000, 36. Murakami M. Honjo T: Involvement of B-1 cells in mucosal immunity and autoimmunity. Immunol Today 16: 534-539, 1995, .~7. Raman C: CDS, an important regulator of lymphocyte selection and immune tolerance. Immunol Res 26: 255-264, 2002, 38. Ashwell .10, Klausner RD: Genetic and mutational analysis of the T-cell antigen receptor. Annu Rev Immunol 8: 139--167, 1990. 39. Muljo SA, Schlissel MS: Pre-B and pre-T-cell receptors: conservation of strategies in regulating early lytnphocyte development. Immunol Rev 175: 80-93, 2000, 40. Zinkernagel RM, Doherty PC: Restriction of in vivo T-cell mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneic system. Nature 248: 701-702, 1974. 41. Li H, Lehedeva MI, Llera AS, et al: Structure of the Vd domain of a human g:d T-cell antigen receptor. Nature 391: 502-506, 1998. 42. Hayday A: yfJ cells: a right time and a right place for a conserved third way of protection. Annu Rev Immunol18: 975-1026, 2000, 43. Hayday A, Tigelaar R: Immunoregulation in the tissues by gammadelta T celLs, Nat Rev Immunol 3: 233--242. 2003. 44. Malissen B, Malissen M: Functions of TCR and preTCR subunits: lessons from gene ablation. Curl' Opin Immunol 8: 383--393, 1996. 45. Peterson EJ. Koretzky GA: Signal transduction in T lytnphocytes. Clin Exp Rheumatol 17: 107-114, 1999. 46. Zamoyska R: C04 and CD8: modulators of T cell receptor recognition of antigen and of immune response? Curl' Opin Immunol 10: 82-87, 1998. 47. Rammensee HG: Chemistry of peptides associated with MHC class I and II molecules, Curr Opin Immunol 7: 85--96, 1995. 48, Germain RN: MHC-dependent antigen processing and peptide presentation: providing ligands for T Iytnphocyte activation, Cell 76: 287-299, 1994, 49. York lA, Rock KL: Antigen processing and presentation by the class I major histocompatibility complex. Annu Rev Immunol 14: 369-396, 1996. 50. Franco A, Ferrari C, Sette A. Chisari FV: Viral mutations, TCR antagonism and escape from the immune system. Curr Opin Immunol 7: 524-531. 1995, 51. Srivastava P: Interaction of heat shock proteins with peptides and antigen presenting cells: chaperoning of the innate and adaptive immune responses, Annu Rev Immunol 20: 395-425, 2002. 52. Anderson G. Moore NC, Owen .1.1, et a!: Cellular interactions in thymic development. Annu Rev Immunol 14: 73--99, 1996, 53, Shortman K, Wu L: Early T lymphocyte progenitors. Annu Rev Immunol 14: 29--47, 1996. 54. Starr, TK Jameson SC, Hogquist KA: Positive and negative selection of T cells, Annu Rev Immunol 21: 139-176, 200355. Ashton-Rickardt PG, Bandeira A, Delaney JR, et al: Evidence for a differential aVidity model of T-cell selection in the thymus. Cell 74: 651-663, 1994, 56. Lenschow OJ, Walunas n, Bluestone JA: CD28/B7 system of T cell cmtimulation, Annu Rev Immunol 14: 233--258. 1996, 57. Rudd CE: Upstream-downstream: CD28 cosignaling pathways and T cell function. Immunity 4: 527-534. 1996, 58. Dong C, Nurieva RI, Prasad DV: Immune regulation by novel costimulatory molecules, Immunol Res 28: 39-48. 2003. 59. Banchereau .I, Bazan F. Blanchard D. et al: The CD40 antigen and its ligand, Annu Rev Immunol 12: 881-922, 1994. 60. Lanzavecchia A: License to kill. Nature 393: 413--414, 1998. 61. Medzhitov R, Janeway CA .11': Innate immunity: the virtues of a nonc!onal system of recognition, Cell 91: 295-298, 1997
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
62. Banchereau J. Briere F, Caux C, et al: Immunobiology of dendritic cells. Annu Rev Immunol 18: 767-811, 2000. 63. Lanzavecchia A: Receptor-mediated antigen uptake and its effect on antigen presentation to class II-restricted l' lymphocytes, Annu Rev Immunol 8: 773-793, 1990. 64. Kersh GJ, Allen PM: Essential flexibility in the T-cell recognition of antigen. Nature 380: 495--498, 1996. 65. Malissen B: Translating affinity into response, Science 281: 528-529, 1998, 66. Nicholson LB, Greer ]M, Sobel RA, et al: An altered peptide ligand mediates immune deviation and prevents autoimmune encephalomyelitis. Immunity 3: 397-405, 1995. 67. Uewelyn M, CohenJ: Superantigens: microbial agents that corrupt immunity, Lancet Infect Dis 2: 156-162, 2002. 68. Abbas AK, Murphy KM, Sher A: Functional diversity of helper l' lymphocytes. Nature 383: 787-793, 1996. 69. Romagnani S: The Th1/Th2 paradigm. Immunol Today 18: 263-266, 1997. 70. Bottomly K: l' cells and dendritic cells get intimate. Science 283: 1124-1125, 1999. 71. Murphy KM, Ouyang W, Farrar ]D, et al: Signaling and transcription in l' helper development. Annu Rev Immunol 18: 451-494, 2000, 72, Szabo S], Kim ST, Costa GL, et al: A novel transcription factor, T-beta, directs Th1 lineage commitment. Cell 17: 655-669, 2000. 73. nas J, Chen CH, Yang L, et al: A critical role for NF-kappa B in GATA3 expression and TH2 differentiation in allergic airway inflammation. Nat hnmunol 2: 45-50, 2001. 74, Grogan JL, Locksley RM: l' helper cell differentiation: on again, off again. Curr Opin Immunol 14: 366-372, 2002. 75. Messi M, Giaccheno I, Nagata K, et al: Memory and flexibility of cytokine gene expression as separable properties of human T(H)1 and T(H)2 lymphocytes. Nat Immunol 4: 78-86, 2003. 76. Sallusto F, Mackay CR, Lanzavecchia A: The role of chemokine receptors in primary, effector, and memory immune responses. Annu Rev Immunol 18: <;93-620, 2000. 77. Russell]H, Ley 1']:. Lymphocyte-mediated cytotoxicity, Annu Rev Immunol 20: 323-370, 2002. 78. Fagarasan S, Honjo 1': l' independent immune response: new aspects of B cell biology. Science 290: 89-92, 2000, 79. De Franco AL: B-cell activation. Immunol Rev 176: 5-9, 2000. BO. Stavnezer J: Inununoglobulin class switching, Curr Opin Immunol 8: 199-205, 1996. 81. Maclennan ICM: Germinal centers. Annu Rev Immunol12: 117-139, 1994, 82. Mackay F, Browning]L. BAFF: a fundamental survival factor for B cells, Nat Rev Immunol 2: 465-475, 2002. 83. O'Connor BP, Gleeson MW, Noelle Rj, Erickson LD: The rise and fall of longlived humoral immunity: terminal differentiation of plasma cells in health and disease. Immunol Rev 194: 61-76, 2003, 84. Traggiai E, Puzone R, Lanzavecchia A: Antigen dependent and independent mechanisms that sustain serum antibody levels. Vaccine 21 (Suppl 2); S35-S37, 2003. 85. Liszewski MK, Fames TC, Lublin DM, et al: Control of the complement syst
61
102. Abbas AK. The control of l' cell activation vs, tolerance, Autoimmun Rev 2: 115-118, 2003. 103. Danial NN, Korsmeyer S]. Cell death: critical control points. Cell 116: 205-219, 2004. 104. Ashkenazi A, Dixit VM: Death receptors: signaling and modulation. Science 281: 1305-1308, 1998. 105, Green DR, Droin N, Pinkoski M: Activation-induced cell death in l' cells. Immunoi Rev 193: 70-81, 2003 106, Thornberry NA, Lazebnik Y: Caspases: enemies within. Science 281: 1312-1316, 1998. 107, Nagata S, Suda 1': Fas and Fas ligand: Ipr and gld mutations, Immuno! Today 16: 39-43, 1995. 108, Drappa], Vaishnaw AK, Sullivan KE, et al: Fas gene mutations in the CanaleSmith syndrome. an inherited Iymphoproliferative disorder associated with autoimmunity. N Engl] Med 335: 1643-1649, 1996. 109. Van Parijs L, Peterson DA, Abbas AK: The Fas/Fas ligand pathways and bcl2 regulate l' cell responses to model self and foreign antigens. Immunity 8: 265-274, 1998. 110. Kamradt 1', Mitchison NA: Tolerance and autoimmunity. N Engl] Med 344: 655-664, 2001. 111. Goodnow CC: Balancing immunity and tolerance: deleting and tuning lymphocyte repertoires. Proc Natl Acad Sci USA 93: 2264-2271, 1996. 112. Mondino A, Khoruts A, Jenkins MK: The anatomy of T-cell activation and tolerance. Proc Natl Acad Sci USA 93: 2245-2252, 1996. 113. Sakaguchi S: Regulatory l' cells: key controllers of immunologic self-tolerance. Cell 101: 455-458, 2000. 114. Hanahan D: Peripheral-antigen-expressing cells in thymic medulla: factors in self-tolerance and autoimmunity. Curr Opin Immuno! 10: 656-662, 1998. 115, Anderson MS, Venanzi ES, Klein L, et al: Projection of an immunological self shadow within the thymus by the aire protein. Science 298: 1395-1401, 2002. 116. Peterson P, Nagamine K, Scott A, et al: APECED: a monogenic autoimmune disease providing new clues to self-tolerance. Immunol Today 19: 384-386, 1998. 117. Mathis D, Benoist C: Back to central tolerance. Immunity 20: 509-516, 2004. 118. Wu], Wilson], He .I, et al: Fas ligand mutation in a patient with systemic lupus erythematosus and Iymphoproliferative disease. J Clin Invest 98: 1107-1113, 1996, 119. Wang], Zheng L, Lobito A, et al: Inherited human caspase 10 mutations underlie defective lymphocyte and dendritic cell apoptosis in autoimmune Iymphoproliferative syndrome type n. Cell 98: 47-58, 1999. 120. Savill], Dranstleld I, Gregory C, Haslett C: A blast from the past: clearance of apoptotic cells regulates immune responses, Nat Rev Immunol 2: 965-975, 2002, 121. Shevach EM. CD4+ CD25+ suppressor l' cells: more questions than answers. Nat Rev Immunol 2: 389-400, 2002, 122. Hori S, Takahashi 1', Sakaguchi S: Control of autoimmunity by naturally arising regulatory CD4+ l' cells, Adv Immunol 81: 331-371, 2003, 123. Groux H, O'Garra A, Bigler M, et al: A CD4+ T-cell subset inhibits antigenspecific T-cell responses and prevents colitis, Nature 389: 737-742, 1997. 124. Levings MK, Roncarolo MG: T-regulatory 1 cells: a novel subset of CD4 l' cells with immunoregulatory properties, J Allergy Clin Immunol 106: SI09-S112, 2000. 125. Prud'homme G], Piccirillo CA: The inhibitory effects of transforming growth factor-beta-l (TGF-betaO in autoinunune diseases. ] Autoimmun 14: 23-42, 2000. 126. Sakaguchi S, Sakaguchi N, Asano M, et al: Immunologic self-tolerance maintained by activated l' cells expressing IL-2 receptor alpha-chains (CD25): breakdown of a single mechanism of self-tolerance causes various autoimmune diseases.] Immunol155: 1151-1164,1995, 127. Hori S, Nomura 1', Sakaguchi S: Control of regulatory l' cell development by the transcription factor Foxp3. Science 299: 1057-1061, 2003. 128, Wildin RS, Smyk-Pearson S, Filipovich AH: Clinical and molecular features of the immunodysregulation, polyendocrinopathy. enteropathy, X linked OPEX) syndrome. ] Med Genet 39: 537--445, 2002, 129. Benoist C, Mathis D: Autoimmunity: the pathogen connection. Nature 394: 227-228, 1998. 130. Griffith TS, Ferguson TA: The role of Fas-L induced apoptosis in immune privilege, Immunol Today 18: 240-244, 1997. 131. Strobel S, Mowat AM: Immune responses to dietary antigens: oral tolerance. Immunol Today 19: 173-181, 1998. 132. MacDonald TT: The mucosal immune system, Parasite lmmuno] 25: 235-246, 2003, 133, Weiner Hi: Oral tolerance: immune mechanisms and treatment of autoimmune diseases, Immunol Today 18: 335-343, 1997, 134 Mayer L, Shao L: Therapeutic potential of oral tolerance. Nat Rev Immunol 4: 407-419, 2004. 135, Vyse 1'], Todd JA: Genetic analysis of autoimmune disease. Cell 85: 311-318, 1996. 136. Winchester R: The genetics of autoinunune-mediated rheumatic diseases: clinical and biologic implications. Rheum Dis Clin North Am 30: 213-227,2004. 137. Marrack P, Kappler ], Kotzin BL: Autoimmune disease: why and where it occurs. Nat Med 7: 899-905, 2001. 138. Sood R, Blake 1', Aksentijevich I, et al: Construction of a I-Mb restrictionmapped cosmid containing the candidate region for the familial
62
139.
140.
141. 142. 143. 144, 145. 146. 147. 148, 149. 150, 151. 152, 153. 154, 155. 156. 157. 158. 159. 160. 161. 162. 163, 164. 165. 166, 167. 168.
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
Mediterranean fever locu, CMEFV) on chromosome 16p 13.3. Genomics 42: 83-95, 1997. McDermott MF, Aksentijevich I, Galon J, et al: Germline mutations in the extracellular domains of the 55 kDa TNF receptor, TNFR1, define a family of dominantly inherited autoinflammatory syndromes. Cell 97: 133-144, 1999. Feldmann J, Prieur AM, Quartier P, et al: Chronic infantile neurological cutaneou, and articular syndrome is caused by mutations in CIAS1, a gene highly expressed in polymorphonuclear cells and chondrocyte,. Am J Hum Genet 71: 198-203, 2002. Miceli-Richard C, Lesage S, Rybojad M, et al: CAR015 mutation, in Blau syndrome. Nat Genet 29: 19-20, 2001. Hugot JP, Chamaillard M, Zouali H, et al: A~sociation of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature 41: 599--{i03, 2001. Kantarci OH, Hebrink DO, Achenbach SJ, et al: CTI.A-4 is associated with su,ceptibility to multiple sclerosis. J Neuroimmunol 134: 133-141, 2003. Xia B, Crusius JB, Wu J, et al: CTLA-4 gene polymorphisms in Dutch and Chinese patients with inflammatory bowel disease. Scand J Gastroenterol 37: 1296-1300, 2002, Kouki T, Gardine CA, Yanagawa T, et al: Relation of three polymorphi"u, of the CTI.A-4 gene in patients with Grave, di,ea,e. .l Endocrinol Invest 25: 208-213, 2002. Rodrigue, MR, Nunez-Roldan A, Aguilar F, et al: A'Sociation of the CTI.A-4 3' untranslated region polymorphism with the susceptibility to rheumatoid arthritis. Hum Immunol 63: 76-81, 2002. Prokunina L, Ca'tillejo-Lopez C, Oberg F et al: A regulatory polymorphism in PDCD1 is associated with susceptibility to 'ystemic lupus erythematosus in humans, Nat Genet 32: 666-669, 2002. Ridgway WM, Fathman CG: The a'5ociation of MHC with autoimmune di,ea'es: understanding the pathogenesis of autoimmune mabete,. Clin Immunollmmunopathol 86: 3-10. 1998. Andersson EC, Svendsen P, Svejgaard A, et al: Rev Immunogene!. A molecule ba,i, for the HLA a'5ociation in rheumatoid arthritis, Rev Immunogenet 2: 81-87, 2000. Botto M, Walport MJ: C1q, autoimmunity and apopto,i" Immunobiology 205: 395-406, 2002. Salmon .lE, Millard S, Schachter LA, et al: Fe gamma RlIA alleles are heritable risk factors for lupus nephritis in African American" Clin Inve't 97: 1348-1354, 1996, Hau,mann S, Wucherpfennig W: Activation of autoreactive T cells by peptides from human pathogens, Curr Opin Immunol 9: 831-838, 1997. Miller SO, McRae BL, Vanderlugt CL, et al: Evolution of the T-cell repertoire during the cou!'e of experimental immune-mediated demyelinating disease. Immunol Rev 144: 225-244, 1995, Kaufman DL, Clare-Salzler M, TianJ, et al: Spontaneou, 10'5 of T cell tolerance to glutamic acid decarboxylase in murine insulin-dependent diabete" Nature 366: 69-72, 1993. Wekerle H: The viral triggering of autoimmune disease. Nature Med 4: 77G-771 , 1998. Holoshitz J, Naparstek Y, Ben-Nun A, et al: Lines of T lymphocyte, induce or vaccinate again't autoimmune arthritis, Science 219: 56-58, 1983, McLaren NK, Atkinson MA: Insulin-dependent diabetes mellitus: the hypothesi, of molecular milnicry between islet cell antigens and microorganisms. Mol Med Today 2: 76-83, 1997, Wucherpfennig KW, Strominger JL: Molecular mimicry in T-cell mediated autoimmunity: viral peptides activate human T cell clone, specific for myelin ba,ic protein. Cell 80: 695-705, 1995, Gmss OM, Forsthuher T, Tary-Lehmann M, et al: Identification of LFA-1 as a candidate autoantigen in treatment-re,i'tant Lyme arthritis. Science 281: 703-706, 1998, Albani S, Key'tone EC, Oilier WER, et al: Positive selection in autoimmunity: abnormal immune responses to a bacterial dna] antigenic determinant in patients with early rheumatoid arthritis, Nat Med 1: 448-452, 1995. Prakken BJ, Samodal R, Le TO, et al: Epitope-specific immunotherapy induces immune deviation of proinflammatory T cell, in rheumatoid arthritis. Proc Nati Acad Sci USA 23: 4228-4233, 2004. Horwitz MS, Bradley LM, Harbertson J, et al: Diabetes induced by Coxsackie virus: initiation by bystander damage and not molecular mimicry. Nat Med 4: 781-785, 1998. Segal BM, Dwyer BK, Shevach EM: An interleukin (ILJ-10/IL-12 immunoregulatory circuit controls su,ceptibility to autoimmune disease. J Exp Med 187: 537-546, 1998. Behar SM, Porcelli SA: Mechanisms of autoimmune disease induction, Arthritis Rheum 38: 458-476, 1995. Robbie-Ryan M, Brown M: The role of mast cells in allergy and autoimmunity. Curr Opin Immunol 1: 728-733, 2002. Brucato A, Franceschini F, Doria A, et al: Pathogenetic a8'ociations of maternal anti-Ro/SSA antibodie,. Lupus 11: 650, 2002. Wada K, Kazukawa I, Someya T, et al: Maternal hypothyroidism in autoimmune thyroiditis and the prognosis of infants. Endocr J 47 (SuppIJ: 5133-S135, 2000, Webert KE, Mittal R, Sigouin C, et al: A retrospective 11-year ana!y'is of obstetric patients with idiopathic thrombocytopenic purpura. Blood 102: 4306-4311, 2003.
169, Martin F, Chan AC: Pathogenic roles of B cells in human autoimmunity; insights from the clinic. Immunity 20: 517-527, 2004. 170. Trende!enburg M, Schifferli JA: Cryoglobulins in chronic hepatitis C viru, infection. Clin Exp Immunol 133: 153-155, 2003, 171. La,smann H, Ransohoff RM: The CD4-Th1 model for multiple sclerosi" a crucial re-appraisal. Trends Immunol 25: 132-137, 2004. 172. Roep BO: The role of T-cells in the pathogenesis of type 1 diabetes: from cause to cure, Diabetologia 46: 305-321, 2003. 173. Romagnani P, Annunziato F, Baccari MC, Parronchi P: T cells and cytokines in Crohn's disease, Curr Opin Immunol 9: 793-799, 1997. 174. Panayi GS, Lanchbury JS, Kingsley GH: The importance of the T cell in initiating and maintaining the chronic synovitis of rheumatoid arthritis. Arthritis Rheum 35: 729-735, 1992, 175. Gattorno M, Facchetti P, Ghiotto F, et al: Synovial fluid T cell clones from oligoarticular juvenile arthritis patients di'pJay a prevalent Thl/ThO patterns of cytokine secretion irrespective to immunophenotype. Clin Exp Immunol 109: 4-11, 1997. 176. Wedderburn LR, Robinson N, Patel A, et 'II: Selective recruitment of polarized T cells expreS'ing CCR5 and CXCR3 to the inflamed joints of children with juvenile idiopathic arthriti,. Arthritis Rheum 43: 765-774, 2000, 177. Firestein GS: Evolving concepts of rheumatoid arthritis. Nature 423: 356-361, 2003. 178. Takemura S, Braun A, Crowson C, et al: Lymphoid neogenesis in rheumatoid ,ynovitis. J Immunol 167: 1072-1080, 2001. 179. Gebhard JR, Perry CM, Harkins S, et al: Cox,ackievirus B3-induced myocarditis: perforin exacerbates disease, but plays no detectable role in vims clearance. Am J Pathol 153: 417-428, 1998. 180, Alon R, Feigelson S: From rolling to arrest on blood vessels: leukocyte tap dancing on endothelial integrin ligand, and chemokines at sub-second contacts. Semin Immunol 14: 93-104, 2002, 181. Muller WA: Leukocyte-endothelial-cell interaction, in leukocyte transmigration and the inflammatOly respon,e. Trends Immuno! 24: 327-334, 2003, 182. Frenette PS, Wagner DO: Adhesion molecule,: part 1. N Engl J Med 334: 1526-1529, 1996. 183. Patel KD, Cuvelier SL, Wiehler S: Selectins: critical mediators of leukoC}1e recruitment. Semin lnununol 14: 73-81, 2002. 184. Weber C: Novel mechanistic concepts for the control of leukocyte transmigration: specialization of integrins, chemokines, and junctional molecules, Mol Med 81: 4--19, 2003, 185. Pribila JT, Quale AC, Mueller KL, Shintizu Y: Integrins and T cell-mediated immunity, Ann Rev Immunol 22: 157-180, 2004. 186, Alon R, Ka'5ner PO, Carr MW, et al: The integrin VLA-4 supports tethering and rolling in flow on VCAM-l. J Cell Bioi 128: 1243-1253, 1995. 187. von Andrian UH, Engelhardt B: Alpha4 integrins as therapeutic targets in autoimmune disease. N Engl J Med 348: 68-72, 2003, 188. Baggiolini M: Chemokines in patholo!lY and medicine. J Intern Med 250: 91-104, 2001. 189. Campbell OJ, Kim CH, Butcher EC: Chemokines in the systemic organization of immunity, Immunol Rev 195: 58-71, 2003. 190. Gunn MD, Kyuwa S, Tam C, et al: Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J Exp Med 189: 451-460, 1999. 191. Fais S, Malorni W: Leukocyte uropod formation and membranelcytoskeleton linkage in immune interactions. J Leukoc Bioi 73: 556-563, 2003. 192, Gerard C, Rollin, BJ: Chemokines and disease. Nat Immunol 2: 108--115, 2001. 193. Lloyd CM, Delaney T, Nguyen T, et al: CC chemokine receptor (CCRJ3/eotaxin is followed by CCR4/monocyte-derived chemokine in mediating pulmonary T helper lymphocyte type 2 recruitment after serial antigen challenge in vivo. J Exp Med 191: 265-274, 2000, 194. Proudfoot AE: Chemokine receptors: multifaceted therapeutic targets, Nat Rev Immunol 2: 106-115, 2002. 195. Abbas AK, Lichtman AH: Cytokines. In Abbas AK, Lichtman AH (eds): Cellular and Molecular Inuuunology, 5th ed. Philadelphia, WB Saunders, 2003, pp. 243-274, 196. Feldmann M, Maini RN: Anti-TNF alpha therapy of rheumatoid arthritis: what have we learned? Annu Rev Immunol 19: 163-196,2001. 197. O'Shea .l.l, Ma A, Lipsky P: Cytokines and autoimmunity. Nat Rev Immunol 2: 37-45, 2002. 198, Beutler BA, Milsark IW, Cerami A: Cachectin/tumor necrosis factor: production, di'tribution, and metabolic fate in vivo, J Immunol 135: 3972-3977, 1985. 199, Dempsey PW, Doyle SE, He JQ, Cheng G: The signaling adaptors and pathways activated by TNF superfamily. Cytokine Growth Factor Rev 14: 193-209, 2003. 200, Lay JD, Tsao q, Chen JY, et 'II: Upregulation of tumor necro,is factor-alpha gene by Ep'tein-Barr virus and activation of macrophages in Ep'tein-Barr virus-infected T cells in the pathogenesis of hemophagocytic syndrome. J Clin Invest 100: 1969-1979, 1997. 201. Dinarello CA: Biologic basis for interleukin-1 in disease. Blood 87: 2095-2147, 1996, 202, Greenfeder SA, Nunes P, Kwee L, et al: Molecular cloning and characterization of a second subunit of the interleukin 1 receptor complex. J Bioi Chern 270: 13757-13765, 1995.
C HAP T E R
3
THE IMMUNE SYSTEM AND THE INFLAMMATORY RESPONSE
203. Mantovani A, Locati M, Vecchi A, et al: Decoy receptors: a strategy to regulate inflarrunatory cytokines and chemokines. Trends Irrununol 22: 328-336, 2001. 204. Dayer ]M: Evidence for the biological modulation of IL-l activity: the role of IL-IRa. Clin Exp Rheumatol 20(Suppl 27): SI4-S20, 2002. 205. Horai R, Saijo S, Tanioka H, et al: Development of chronic inflammatory arthropathy resembling rheumatoid arthritis in iriterleukin 1 receptor antagonist-deficient mice. ] Exp Med 191: 313-320, 2000. 206. Dinarello CA: The role of the interleukin-l-receptor antagonist in blocking inflammation mediated by interleukin-I. N Engl] Med 343: 732-734, 2000. 207. Hirano T, Yasukawa K, Harada H, et al: Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin. Nature 324: 73-76, 1986. 208. Ishihara K, Hirano T: IL-6 in autoimmune disease and chronic inflarrunatory proliferative disease. Cytokine Growth Factor Rev 13: 357-368, 2002. 209. Taga T, Kishimoto T: Gp130 and the interleukin-6 family of cytokines. Annu Revlrrununol 15: 797-819, 1997. 210. Muller-Newen G: The cytokine receptor gp130: faithfully promiscuous. Sci STKE 201:PE40, 2003. 211. De Benedetti F, Massa M, Pignatti P, et al: Serum soluble interleukin 6 (IL-6) receptor and IL-6/soluble IL-6 receptor complex in systemic juvenile rheumatoid arthritis. ] Clin Invest 93: 2114-2119, 1994. 212. De Benedetti F, Alonzi T, Moretta A, et al: Interleukin 6 causes growth impairment in transgenic mice through a decrease in insulin-like growth factor-I: a model for stunted growth in children with chronic inflarrunation. ] Clin Invest 99: 643-650, 1997. 213. De Benedetti F, Martini A: Is systemic juvenile rheumatoid arthritis an interleukin 6 mediated disease? ] Rheumatol 25: 203-207, 1998, 214. Cazzola M, Ponchio L, De Benedetti F, et al: Defective iron supply to erythropoiesis and adequate endogenous erythropoietin production in the anemia associated with systemic-onset juvenile chronic arthritis. Blood 87: 4824--4830, 1996. 215. Yokota S: Interleukin 6 as a therapeutic target in systemic-onset juvenile idiopathic arthritis. Curr Opin Rheumatol 15: 581-586, 2003. 216. Andersson U, Wang H, Palmblad K, et al: High mobility group 1 protein (HMG:l) stimulates proinflammatory cytokine synthesis in human monocytes. ] Exp Med 192: 565-570, 2000. 217. Ulloa L, Badiwalla FM, Andersson U, et al: High mobility group box chromosomal protein 1 as a nuclear protein, cytokine, and potential therapeutic target in arthritis. Arthritis Rheum 48: 876-881, 2003. 218. Trinchieri G: Interleukin-12 and the regulation of innate resistance and adaptive immunity, Nat Rev Irrununol 3: 133-146, 2003. 219. Dinarello CA, Fantuzzi G: Interleukin-18 and host defense against infection. ] Infect Dis 187(Suppl 2); S370-S384, 2003. 220. Lankford CS, Frucht DM: A unique role for IL-23in promoting cellular immunity. Leukoc Bioi 73:49-56, 2003. 221. Liew FY: The role of innate cytokines in inflarrunatory response. Irrununol Lett 85: 131-134, 2003. 222. Koch AE: Angiogenesis. Arthritis Rheum 41: 951-962, 1998. 223. Szekanecz Z, Koch AE: Vascular endothelium and immune responses: implications for inflammation and angiogenesis. Rheum Dis Clin North Am 30: 97-114, 2004.
63
224. Berse B, Hunt ]A, Diegel R] et al: Hypoxia augments cytokine (transforming growth factor-beta [TGF-~l and IL-l)-induced vascular endothelial growth factor secretion by human synovial fibroblasts. Clin Exp Irrununol1l5: 176-182, 1999. 225. Mioda], Maciewicz R, Kendrew], et al: Treatment with soluble VEGF receptor reduces disease severity in murine collagen-induced arthritis. Lab Invest 80: 1195-1205, 2000. 226. Turini ME, DuBois RN: Cyclooxygenase-2: a therapeutic target. Annu Rev Med 53: 35-57, 2002. 227. Samuelsson B, Dahlen SE, Lindgren ]A, et al: Leukotrienes and Iipoxins: structures, biosynthesis, and biological effects. Science 237: 1171-1176, 1987. 128. Serhan CN, Chiang N: Novel endogenous small molecules as the checkpoint controllers in inflarrunation and resolution: entree for resoleomics. Rheum Dis Clin North Am 30: 69-95, 2004. . 229. Levy BD, Clish CB, Schmidt B, et al: Lipid mediator class switching during acute inflarrunation: signals in resolution. Nat Irrununol 2: 612-619, 2001. 230. Nathan C: Points of control in inflarrunation. Nature 420: 846-852, 2002. 231. Nathan C: Nitric oxide as a secretory product of mammalian cells. FASEB] 6: 3051-3064, 1992. 232. Chakravortty D, Hensel M: Inducible nitric oxide synthase and control of intracellular bacterial pathogens. Microbes Infect 5: 621-627, 2003. 233. Babior BM, Lambeth ]D, Nauseef W: The neutrophil NADPH oxidase. Arch Biochem Biophys 397: 342-344, 2002. 234. Edwards SW, Watson F: The cell biology of phagocytes. Irrununol Today 16: 508-510, 1995. 235. Mollinedo F, Borregaard N, Boxer LA: Novel trends in neutrophil structure, function and development. Immunol Today 12: 535-537, 1999. 236. Nagase H, Woessner ]F ]r: Matrix Metalloproteinases. ] Bioi Chern 274: 21491-21494, 1999. 237. Bode W: Structural basis of matrix metalloproteinase function. Biochem Soc Symp 70: 1-14, 2003. 238. Chikanza IC, Grossman AB: Reciprocal interactions between the neuroendocrine and immune systems during inflarrunation. Rheum Dis Clin North Am 26: 693-711, 2000. 239. Cutolo M, Bijlsma ]W, Lahita RG, et al: Altered neuroendocrine irrunune (NEI) networks in rheumatology. Ann N Y Acad Sci 966: xiii-xviii, 2002. 240. Besson ]M: The neurobiology of pain. Lancet 353: 1610-1615, 1999. 241. Niissalo S, Hukkanen M, Imai S, et al: Neuropeptides in experimental and degenerative arthritis. Ann N Y Acad Sci 966: 384-399, 2002. 242. Vottero A, Chrousos GP: Glucocorticoid receptor beta: view I. Trends Endocrinol Metab 10: 333-338, 1999. 243. Chikanza IC: Mechanisms of corticosteroid resistance in rheumatoid arthritis: a putative role for the corticosteroid receptor beta isoform. Ann N Y Acad Sci 966: 39-48, 2002. 244. Vera-Lastra 0, ]ara L], Espinoza LR: Prolactin and autoirrununity. Autoimmun Rev 1: 360-364, 2002. 245. Calandra T, Roger T: Macrophage migration inhibitory factor: a regulator of innate immunity, Nat Rev Irrununol 3: 791-800, 2003. 246. De Benedetti F, Meazza C, Vivarelli M, et al: Functional and prognostic relevance of the -173 polymorphism of the macrophage migration inhibitory factor gene in systemic-onset juvenile idiopathic arthritis. Arthritis Rheum 48: 1398-1407, 2003.
C H A P T E R
4
INTEGRATIVE GENOMICS Robert A. Colbert and David N. Glass
When viewed from the perspective of the pediatric rheumatology clinic, few patients with arthritis appear to have genetically determined diseases. Family histories are rarely positive for the more common diseases, including juvenile rheumatoid arthritis ORA), and diseases with a mendelian pattern of inheritance are especially uncommon. Few extended families with chronic pediatric rheumatologic illnesses suitable for traditional linkage studies have been reported. Pediatric rheumatic diseases share this scenario with autoimmune diseases in general, in which the absence of family history is the common experience. One notable exception is spondyloarthritis, particularly ankylosing spondylitis; in some families, the phenotype follows an autosomal dominant inheritance pattern linked to the human leukocyte antigen (HLA) allele HLA-B27, with penetrance as high as 20%. The paradoxical view that diseases without a family history, including the subtypes of JRA, are genetically based is considered in this context; in addition, it is commonly hypothesized that this genetic effect extends not only to a primary predisposition but also to variation in the extent and severity of disease; that is, many, if not all, aspects of the disease phenotype are also genetically determined, whether it be JRA, systemic lupus erythematosus [SLE], scleroderma, or dermatomyositis. The presence of HLA associations in most of these diseases already provides some indication of their genetic nature, although for many years HLAor HLA-linked genes were not necessarily perceived to be central to pathogenesis. It is argued herein that they are a necessary part of genetic predisposition (although only a component) and that other, non-major histocompatibility complex (MHC) genes predispose to the disease in a given individual. This does not exclude an environmental component. It is timely to note that the enormous explosion in knowledge from the Human Genome Project and its related technological advances allows questions to be asked and tested about the role of genes in all diseases, not just those traditionally viewed as having a genetic basis. Current high-throughput molecular technology not only allows screening of DNA polymorphisms but also can track newly identified genes; thus, the traditional approach, starting from a disease phenotype and then identifying the gene, can be reversed. In addition, highthroughput methodology using microarrays is also being applied to gene expression, so that virtually the entire
64
expressed genome can be evaluated at the RNA level, and perhaps eventually at the protein level. This socalled functional genomics can be combined with data from DNA polymorphisms to yield comprehensive integrative genomic approaches to human disease. This chapter reviews knowledge of the genome with respect to genes and polymorphic variability before discussing the genetic components of pediatric rheumatic diseases and the extent to which they have or do not have traditional mendelian patterns of inheritance.
THE GENOME The genome can be defined as the individual's (or cell's) total genetic information. The related science of mapping, sequencing, and analyzing genomes is known as genomics. The Human Genome Project, started in 1990, proceeded at an outstanding pace, with a physical and DNA sequence map of the human genome substantially completed well ahead of the original 2003 target. The working draft announced in spring 2000 reported assembled sequences for approximately 85% of the genome.' This map will be the basis for understanding gene function and its pathophysiology. A complete description of this project can be found at the National Human Genome Research Institute Web site, http://www.genome.gov/HGP!
Structure At the DNA level, genetic information consists of more than 3 x 109 base pairs organized into three components: paired autosomal chromosomes, two sex chromosomes, and mitochondrial DNA. A large part of this genetic material is aggregated into repetitive sequences that contribute, as either long or short interspersed sequences, to the familiar banding pattern that characterizes the morphology of chromosomes. The remainder is single copy DNA, about 10% of the whole, which is primarily organized into genes, of which only about 2% overall are protein coding genes and an additional 4% or so contain conserved sequence elements (CSEs) that may include promoter regions.' The human genome most likely encodes 30,000 to 40,000 genes. Experimentally, expressed or functional genes are documented as expressed sequence tags (ESTs), which are partially sequenced complementary DNA
C HAP T E R
(cDNA) molecules. A cDNA molecule is synthesized DNA that is complementary to RNA and reflects expressed gene products. The number of these genes expressed is probably different in different organs: A metabolically complex organ such as the liver may have 40,000 expressed genes; the synovium may well have fewer, perhaps 30,000, varying with stage of development. Genes are divided into coding and noncoding compartments (exons and introns, respectively) with additional promoter and flanking sequences that are less well understood. The latter are a major part of the regulatory process, which determines when and where specific genes are expressed. Thus, the gene is a complex unit in both its structure and its regulation.
Polymorphic Elements It is fundamental to the understanding of human diversity and of disease to recognize that, although the genome and gene structure are broadly the same for all persons (99.9% identity), variability is substantial among individuals with respect to ethnicity, and this can give rise to disease-the focus of this review. This variability may be local (Le., gene specific), or it may be part of genome-wide polymorphic elements.
Local Variability Local variability in a given gene or segment of DNA containing several genes can result from a change or mutation in a single nucleotide or from deletions, translocations, or gene conversions. The resulting functional change in even a single expressed gene product can range from deleterious (including fataD, to neutral, or even to beneficial (gain in function). Clearly, the nature and site of the change (e.g., coding regions, regulatory regions) are critical in this regard and can be reflected in changed phenotypes and disease. The use of single nucleotide polymorphisms (SNPs) is a formidable tool to evaluate both the whole genome (see later discussion) and local variability. Examples of individual gene variability of relevance to autoimmunity include complement deficiencies and the chromosome 22 deletion associated with JRA. 3
4
INTEGRATIVE GENOMICS
65
small, repetitive sequences of DNA, they are commonly called microsatellites. These polymorphisms can be distinguished by methods based on the polymerase chain reaction (PCR) (see later discussion). Their utility is enhanced by the availability of genome-wide maps and by knowledge of their polymorphic information content (PIC), which allows the selection of those polymorphisms that are likely to optimize any given data set.
Single-Nucleotide Polymorphisms SNPs are much more common than VNTRs.5 Some 4.5 million SNPs were validated by mid-2004, and there may be 10 to 20 million of them in the genome. SNPs form the basis of restriction fragment length polymorphisms (RFLPs), which are SNPs that happen to be located in an enzyme (endonuclease) recognition site. SNPs that are located in a coding region are known as cSNPs. 6 Overall, less is known about the map positions and PICs of SNPs than of VNTRs, although this situation is changing rapidly. With regard to SNPs being a tool for genomics, their number now allows a much greater frequency of markers, offsetting the fact that VNTRs are more polymorphic. Their ubiquitous presence allows their use in association studies, adding greater analytic power than is available through linkage studies, including the use of affected sibling pairs (ASPs). The ability to use SNPs will be enhanced by the new DNA technologies, which may utilize several different platforms, including gene chips, beads, and real-time PCR, although the very great number of SNPs (perhaps 200 or more in some genes) adds complexity to their analysis. 7 However, their utility in the dissection of complex genetic traits can be illustrated by two SNPs that have been associated with components of a disease phenotype. These SNPs involve a mutation that results in a gain of function in the interleukin-4 receptor gene, which is associated with severe asthma, and a mutation in a ~2-adrenergic receptor gene (ADRB2) that is associated with greater resistance to adrenergic drugs. 8,9 Genome-wide maps are being generated, and there are going to be various sources of information for constructing these maps.
Maps Genome-Wide Polymorphlsms A key aspect of research in the genetics of disease is associating sequence variations with heritable phenotypes. One of the consequences of the explosion in knOWledge relating to the genome has been the identification of polymorphic variants that occur throughout the genome in both coding and noncoding elements of DNA. There are two important broad classes of importance with respect to the science of genomics, very numerous tandem repeats (VNTRs) and SNPs.
Very Numerous Tandem Repeats VNTRs were the first genome-wide polymorphisms to be documented; more than 10,000 of them occur throughout the genome. 4 They appear to be randomly distributed in both coding and noncoding regions. Because they are
Chromosome Map Cytochemical approaches have been widely used to generate the familiar chromosome banding patterns. The addition of fluorescent in situ hybridization (FISH) has been a substantial benefit, combining cytochemical and molecular approaches. Although only broad localization is possible, several thousand genes have been mapped by this process to specific chromosomes, albeit with low resolution. Fluorescent tags on intact chromosomes cannot be resolved into separate spots unless they are 2 to 5 million base pairs apart.
Genetic Map Although a genetic map is an extension of the chromosome map described earlier, its creation is a more
66
C HAP T E R
4
INTEGRATIVE GENOMICS
detailed process by which the relationships among individual genes and their eventual assignments to specific chromosome regions and linkage groups becomes possible. Sufficient genes or polymorphic markers are mapped so as to effectively cover the whole genome. Traditionally, extended kindreds are studied for the coinheritance of a particular phenotype, whether physiologic or pathologic. Genes that cosegregate are established by this process; two genes can be shown to be closely linked and, therefore, a component of a linkage group. This sometimes involves genes of like function, the linkage group having arisen by gene duplication, but often a cluster of genes do not have similar or related functions. Examples of functional clusters are the HLA genes and the T-cell receptor genes, each of which each form close linkage groups. The statistical analyses used in such situations are logarithm of the odds Ood) scores; a score of 3.0 or more is the standard for establishing linkage between two genes. A particular linkage group may be inferred from findings in other species, a concept known as !>ynteny. Although clustering of a particular set of genes may be shared across species, the same linkage group may be found on a different chromosome in humans. For example, the MHC linkage group is located on chromosome 6 in humans and on chromosome 17 in the mouse. The recombination rate detected in families is a measure of the closeness of linkage. Recombination may be current or ancestral and translates into map distance, or centimorgans (cM). There are approximately 3500 cM in the human genome. This genetic distance, which depends on recombination frequency, may be different from the physical distance between linkage groups on the same chromosome. The HLA region itself amounts to about 3.5 cM (i.e., about 0.1% of the genome).
allow the definition of linkage disequilibrium throughout the genome and the production of the so-called Hap Map. This is an extension of the Human Genome Project in which the latest high-throughput technology is used to identify tSNPs (or tag SNPs) that identify haplotype blocks. 12 Such a map will provide tSNPs that effectively mark a particular haplotype. These specific SNPs can then be used in genome-wide screens as a surrogate for large stretches of DNA covered by linkage disequilibrium. This may allow an extremely cost-effective screening of at least 80% of the genome. It will limit the number of SNPs that must be used to ensure complete coverage of the genome for wide-scale genomic studies. The physical map will identify the genes and their locations within the total genome but will not determine whether they are functional. There are genes that cannot be expressed because of specific mutations; these genes are known as pseudogenes. Pseudogenes need to be differentiated from genes that, because of mutation, cannot function. The nonfunctioning gene is known as a null gene and is allelic with functioning variants.
Physical Map
Biotechnology and Blolnformatlcs
The advent of DNA sequencing allows definition of individual genes as well as their relationships with each other in terms of intervening DNA structure. Considerable amounts of genetic data have been generated and entered into sequence databanks, much of which is available to investigators (e.g., National Center for Biotechnology Information, http://www.ncbi.nim. nih.gov). This process allows physical mapping of individual stretches of DNA; interim physical maps are based on VNTRs and, more recently, on SNPS. IO ·11 The HLA region, for example, has on the order of 3500 kilobases (kb) of DNA, with approximately 180 genes and perhaps 80 VNTRs, but probably several thousand SNPs. Currently, a major effort is being vested in the identification of SNPs and the establishment of their map positions. Groups of alleles from closely linked genes are known as baplotypes. Some haplotypes are much more common than others, based on the concept of linkage disequilibrium. It has long been recognized that alleles of closely linked genes may not occur at random (Le., they show linkage disequilibrium). This has been most comprehensively documented in the MHC as extended or ancestral haplotypes. The generation of SNP maps will
The Human Genome Project-and with it the potential to establish the genetic basis of disease-would not have been possible without parallel technological advances. One key element is the PCR. This method allows the expansion of specific segments of DNA to make enough of them available for quantitative analysis, including sequencing. With respect to genomics, the widespread availability of polymorphic markers detectable by PCR for every part of the genome and the commercial availability of the appropriate PCR primers have created great opportunities for scientific advances. These primer pairs are organized into sets that allow a genome-wide approach. IS Although substantial work is still involved, marker sets are available that allow multiplexing of primer pairs based on product size and fluorescent tags, permitting up to 15 genotypes to be determined simultaneously on a single lane of a gel or capillary tube. In parallel, high-throughput genotyping/ sequencing machines generate more than 20,000 genotypes per week l6 The newer technologies are now capillary based rather than gel basedY This advance, combined with robotics for the PCR process, allows a laboratory with one or two technical staff members
Functional Map The functional map is based on functional genomics, the analysis of individual gene function, which is itself based on the structural or physical map.13 Through the process of identification of expressed genes (i.e., ESTs), assisted now with technological advances using gene arrays,1'1 it will be possible not only to identify all expressed genes in a given tissue but also to consider their function. Comparisons between developing and developed tissues, as well as disease and normal states, are now ongoing and should progress rapidly. This will result in a functional map.
C HAP T E R
to complete up to 1 miJIion genotypes per year. A medium-density genome screen (about every 10 cM) for a given disease may number about 150,000 genotypes. The most recent technology with SNP chips or beads may allow up to 1 miJIion genotypes to be generated per day by two or three staff members, although cost remains a significant issue. In parallel with this enormous flow of data is the development of appropriate software to assign genotypes, to validate the accuracy of the genotyping in the context of the family tree, and then to facilitate the analytic process. Automated processes are particularly important for SNPs, because they are going to be used in lO-fold or greater numbers than VNTRs. The development of software for large databases has been paralleled by conceptual advances in genetic methods. Of particUlar relevance are approaches to linkage analysis that do not depend on large kindreds. Two particularly relevant methods are allele sharing using ASPs and transmission disequilibrium testing (TDT), which uses family-based controls. I8-21 TOT is an alternative approach to establishing linkage that is valid for either simplex or multiplex families (see later discussion). SNPs wiJI promote a shift back to association studies once they are affordable in large numbers.
MONOGENIC DISEASE IN PEDIATRIC RHEUMATOLOGY The diseases inherited in a pattern recognizable as mendelian, primarily autosomal dominant and autosomal recessive, are numerous. More than 10,000 are now listed; of these, hundreds might well involve the musculoskeletal system. A few manifest directly with a form of arthritis, others with musculoskeletal features. These particular disorders make up a very small portion of the referral base for a pediatric rheumatology clinic, but they are well recognized. However, with the increasing capacity to identify particular genes and their mutations, as well as the functional consequences that lead to particUlar phenotypes, molecular analysis has become more practical and necessary-as has awareness of the phenotypes of these disorders. The familial categories of inherited disease (dominant, recessive, and X-linked) proVide patterns of inheritance that are discernible if typically penetrant. A dominant disease should be evident in every generation, shOWing vertical transmission; a recessive disease occurs in approximately one of four children in a family and is more likely to occur in the offspring of consanguineous marriages. X-linked recessive diseases become evident in male children whose mothers are the carriers, although female carriers may show some features of the disease, consistent with the Lyon hypothesis of X chromosome inactivation. In 1997, Chalom and colleagues22 categorized these diseases into three groups: arthritic diseases (e.g., Lesch-Nyhan), those resulting in contractures or stiff joints (e.g., Gaucher's disease), and those causing hypermobility (e.g., Ehlers-Oanlos syndrome) (see Chapter 40). A total of 75 disorders were identified,
4
INTEGRATIVE GENOMICS
67
probably only a portion of the whole. 23 It is recognized that not all of these disorders occur in the classic or traditional mendelian manner.
Confounding Variables In the Recognition of Mendelian Inheritance Monogenic effects may be implicated in a given instance but may not be recognizable for a variety of reasons. Transmission anomalies include inheritance of two copies of a whole chromosome from the same parent (uniparental disomy). Other variations in a monogenic disease may have more subtle bases and are summarized in Table 4-1. In addition, modifier genes may alter a specific phenotype, resulting in different levels of expression. A disease caused by a single gene could be modified by the function of other genes on a variable basis. This phenomenon might be distinguished from complex genetic traits in which a number of genes are required to generate the primary disease phenotype, although the distinction is probably relative.
COMPLEX GENETIC TRAITS A complex genetic trait may be defined as one that is dependent on multiple genes and displays nonmendelian inheritance patterns. It is likely that the diseases more commonly encountered in the pediatric rheumatology clinic are the result of complex genetic traits, although the evidence to support this statement is not yet at hand. However, as is evident from the preceding discussion, the genome-wide approaches needed to validate the concept are available, and they have already been tested in insulin-dependent (type 1) diabetes mellitus and many other autoimmune diseases. 24 •25 Examples of complex genetic traits in this category include rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, psoriatic arthritis, scleroderma, and SLE, in addition to type 1 diabetes. 26 Other potential complex genetic traits, including hypertension and osteoarthritis, have a metabolic but nonimmunologic basis. Of most relevance to this discussion is the likelihood that JRA and its various subtypes are complex genetic traits. 27 How are these recognized in the absence of genetic data? Features that suggest a complex genetic trait include a definite but limited family history but with
.~ TABlE 4-1
Variables in 'he Recogni'ion of Monogeni. Disease
Uniparental disomy New mutations Low penetrance Parental imprinting Variable phenotype Mitochondrial genes Parental reproductive choice or fitness From Ostrer H: Non-mendelian genetics in humans. In Oxford Monographs on Medical Genetics No. 35. Oxford. England. Oxford University Press. 1998.
68
C HAP T E R
4
INTEGRATIVE GENOMICS
few or no extended affected kindreds; an increased presence of other autoimmune diseases in the family28; and HLA disease associations that suggest an immunologically related complex genetic trait. Of these features, HLA associations have been extensively documented, and family histories have been reported for the disease affecting the proband, but less commonly for autoimmune diseases in general. Individual components of the phenotype are likely to be regulated separately, such that each component could be called a quantitative trait locus (QTL), with its own genetic basis. For example, vasculitis complicating SLE or uveitis complicating early-onset pauciarticular ]RA may be viewed as an individual QTL. Some patients have these features in their disease phenotype, and others do not. Studies of animal models support this QTL concept in rheumatologic disease. 29 What has also been missing and important to predict is information regarding the likelihood that other non-HLA genes are involved. It is likely that responses to therapy (or failures with therapy) are also QTLs and will prove to be genetically mediated, part of the science of pharmacogenetics.
Quantltatlon of the Family History Genomic screens are time-consuming ventures, and some quantitation of familial risk for a given disease would help to determine the probability that the disease is or is not likely to be a complex genetic trait. The standard measure of risk of affected siblings is As, calculated as the prevalence of the illness in question in sibs of the patient with the disease, divided by the prevalence of the disease in the general population. 18,30 For ]RA, one estimate of A.S is 15, similar to that for type 1 diabetes. Ankylosing spondylitis, with greater penetrance, may have a A.S of about 80.
Dlsease-Spedflc Susceptibility Genes Versus General Autoimmune Genes Mutations in genes with an immunologic function may relate to a variety of immunologic diseases. This can be the experience in both patients and animal models of autoimmunity, one such example being the las gene defects in mice and in humans that effect cell death or apoptosis. 3J.32 This gives rise to the concept that both general autoimmune-predisposing genes and diseasespecific susceptibility confer risk of autoimmunity. The number of genes involved in a particular complex trait is likely to be between 5 and 20; confirmation of this concept is being worked out as genome screens are completed in many autoimmune diseases. Analysis of 22 genome screens identified more than 13 chromosome regions or genes found in common in different autoimmune diseases 33 ; similar evidence is accruing in animal models. 34 Because different autoimmune diseases tend to have different HLA associations, it is likely that disease specificity will partly rest with the HLA region on the short arm of chromosome 6, whereas a general predisposition may be more related to the other, non-HLA susceptibility regions. This concept is a useful generalization and is being tested. There will clearly be exceptions; for
example, both general and disease-specific polymorphisms may be found within the MHC.
Disease Phenotype as a Potential Complex Genetic Trait Two strategies are commonly used in evaluating a disease as a complex trait. One is a candidate gene approach, including selected specific genes or chromosome regions that are tested for disease susceptibility; the other is a comprehensive genome screen. As the growing number of comprehensive or global screens in other autoimmune diseases identify chromosomal loci that confer disease susceptibility, a candidate gene approach for any potential autoimmune complex genetic trait becomes more feasible. The use of candidate genes has the potential to reduce the overall workload but may miss loci or decrease the resolution, whereas the genome-wide screen systematically examines every genetic region, albeit often at low resolution. A conservative approach allows both methods of ascertaining linkage or association to be tried in parallel. As the technology is enhanced, global approaches to association should become feasible.
Candidate Genes With most of the autoimmune diseases, the literature supports the involvement of individual genes or polymorphisms in susceptibility. For ]RA, a recent review 27 identified 30 such potential genes or chromosome regions outside the MHC. Although the data supporting the candidacy of such entities are conflicting at best, the probability that one or more will be involved in disease susceptibility is high, especially in those chromosome regions for which selection is based on meta-analysis of prior genome screens. 33 HLA-B27 is strongly and almost uniformly associated with spondyloarthritis. 35
Genome Screen A genome screen searches systematically throughout the genome for individual chromosome regions of susceptibility. The marker density selected for such approaches has generally been about 10 cM, which necessitates the use of more than 350 markers across the genome. This number being a reasonable compromise between identifying positive areas without excessive workload, a database of 100 ASPs (screened with all parents included) will result in 150,000 genotypes if the DNA quality is sufficient. As noted earlier, the marker sets now available with PCR primer pairs are organized so that multiple PCR products can be run on a given gel, a process known as multiplexing. Appropriately, different fluorescein tags are used to facilitate this process, leading to a high throughput of samples. One marker set has more than 800 marker pairs, substantially increasing the density of the screen.
Strategy for an Individual Genome Screen Although the general approach to genome screening is common to all potential complex genetic traits (i.e., the
C HAP T E R
use of multiple markers in all chromosome regions), the strategy selected for any given disease will vary. The level of As, discussed earlier, may be a good indication that "positive" data should result. Some autoimmune and autoinflammatory diseases (e.g., ankylosing spondylitis, SLE) have extensive multiplex kindreds. Most disorders have less extensive kindreds, but almost all autoimmune diseases appear to have ASPs as a common denominator. However, the number of available ASPs differs with each disease, and this also has a bearing on the stratagem to be adopted. Studies with 800 to 1000 ASPs are ongoing in rheumatoid arthritis, and several hundred ASPs are available for ankylosing spondylitis; in pediatric rheumatology, however, the numbers are relatively modest. On the other hand, in pediatric practice, both parents are usually available in a very high proportion of ASPs, which adds considerable analytic power that is unavailable without parental DNA. Normally, ASPs share both haplotypes in 25% of instances and one haplotype in 50%; a deviation from this expected distribution among ASPs indicates linkage. 18 If insufficient ASPs are available, other approaches are needed. An alternative method is the fiT, which requires parents and a single proband (Le., a simplex family). It is considered a test of both association and linkage. 19•2 1,36 An example of the use of familybased control genes is preferential transmission of particular genes to the proband, which is compared with the allelic distribution in genes that are not transmitted. 37 Associations generated through case-control studies have often been confounded by population stratification (founder effects), indicating the importance of familybased studies for assessment of both linkage and association. Unaffected siblings may be used if a stratagem can be devised to work out an individual's potential to contract the autoimmune disease in question. Information on parents is usually necessary, although the absence of one parent can be compensated for if genetic information in the children is sufficient. ASPs may also be used in the fiT (sib-pair fiT); this doubles the power of the test. There is considerable discussion as to the meaning of the TDT; results are not positive in the absence of linkage, although fiT results also depend on linkage disequilibrium and association.
Which Polymorphlsms? In general, microsatellite repeats (VNTRs) have been used most frequently. As noted earlier, they not only have the advantage of being mapped, but their polymorphic content is known, so VNTRs with maximum PIC content can be selected to optimize the available family materials and minimize uninformative genotyping. As an alternative, SNPs are less polymorphic and less commonly· mapped; however, their much greater number and lower cost increasingly offset the advantages of VNTRs. Because use of the fiT in simplex families requires higher-density markers (linkage disequilibrium may rarely extend more than 50 kb), the more densely distributed SNPs are likely to become the method of choice in the TDT. This is especially likely given that the technology is now available to read such polymorphisms
4
INTEGRATIVE GENOMICS
69
from DNA-based chips, which adds another automated step to the process and allows increased numbers of genotypes to be more readily processed. 38 Candidate genes can be tested in ASPs or by the TOT, the latter in either simplex or multiplex families. In this situation, association studies using a family-based pool with controls can also reduce the element of population stratification that has made case-control studies problematic.
Replication and Extension of Findings Initial findings suggestive of linkage in any genome study will need replication. For this reason, a second set of DNA on a new population would be an asset. The method of approach, whether through sib pairs or through case-control studies (see later discussion), need not be identical in design to the first study and could be focused on areas of potential interest (chromosome loci or genes) established in the initial study. Fine mapping then becomes possible with high-density markers. For example, 60 to 80 VNTRs might be available for a particular chromosome region and could be applied in multipoint linkage or association studies. In this approach, it is possible to define the point of maximum linkage or association and therefore to map more exactly the gene involved in the disease. Chromosome regions so confirmed will still have the potential to contain many genes (perhaps 20), compared with the larger number in the original region. Such an approach was applied to the mapping of the diastrophic dysplasia and hemochromatosis genes. 39--41 This method allows the gene-hunting phase to be applied with greater precision.
PEDIATRIC RHEUMATIC ILLNESSES AS COMPLEX GENETIC TRAITS To what extent are the common chronic pediatric rheumatic illnesses complex genetic traits? The subtypes of JRA, juvenile dermatomyositis, psoriatic arthropathy, ankylosing spondylitis, SLE, and scleroderma are all candidates. Supportive evidence in children is available (albeit limited) for some diseases but not at all for others; only data in adult patients are available where disease overlap occurs. For pauciarticular JRA with onset in young children, the relatively frequent occurrence of this illness has ensured that enough ASPs are available to demonstrate substantial concordance for diseaseY HLA associations are well documented, although extended multiplex families are not available for linkage studies (see Chapter 11). HLA and JRA have been shown to be linked through two approaches by the TDT in 103 HLA-typed simplex families 43 and through allele sharing in 53 ASPs. 44 Both of these studies used fewer numbers of families than has generally been the case in genomic studies in adults, suggesting that a genome-wide screen using the available number of ASPs would be successful in identifying at least the major susceptibility (if not all of the minor susceptibility) loci. An initial genome-wide screen with eight regions identified with lod scores of 2 or greater has recently been published. 45 These results do depend on
70
C HAP T E R
4
IN T E G RAT I V E
G E NOM I C S
analyses of subsets. The nonparametric, multipoint linkage for each chromosome is given in Figure 4-1. The numbers required to consider other aspects of all forms of JRA (e.g., development of chronic uveitis as a QTI)
Analysis for Chromosome 4
A
may not be available, requiring the use of simplex families and association studies to resolve some of these additional genetic issues. Both the candidate-gene approach and genome-wide screens are feasible.
Analysis for Chromosome 7
B
8
3
"
'\ \
,, , , I
2
2
\
\
.
\
I
•
I
•
I
-l.......~,.......,..,....,....-.-~~"TT"'M"T',..,..... o 00 ~ ~ ~ W
........,.....,~~"'M'"1M"T'.-.,...,.,..,O"-;:;:;~ ~ ~ ~ ~ ~ m
\
\
-1...........,..,"TT".,.....,.,.....~'"T""".,...,.....,.... o 20 80
Map poEiticn (eM)
........,..,..,.~....,.....~.,..,...,~....,-~.....,..,...,....,....-/
Map positicn (eM)
Analysis for Chromosome l2
c
.
•
1
Analysis for Chromosome 19
o
:;
III
4
o N
00
:n '"(;
VI
VI
o'" '"o
3
2
-l·1,-,-.........,rr-r",..,.,.,"'"""'..,..,...~M"T'....,........,.,."..~,..,.....TTT.,.....,....,..,..,...,.......,..,...,..."'Tခ 㓳ሀ ⅁倀
o
m
~
~
~
U
W
~
W
~
~
~
-1.......~,..,..... .........~....,. .............,........,.'T"T"'-.-,-,.,..,-........--rr-~~"'M'"1r-"""" ..........,.,..,....-/
o
~
D
~
20
Analysis for Chromosome
E
m
j Q
:3
j
8
Q
2
, ,,
... ,,
...
,
w
~
~
w m
....... , , ,,
,
I
J
J -1.......~,..,...........,-.-,-....,...,..........,..,..,~.,..... ........--rr-......,.,.,...,"'M'"1r-"""".,...".~......,..,..,.,.....y
Map poI!lition (eM)
~
Analysis for Chromosome X
F
:3
o
ro
~
Map poI!lition (eM)
Map pa!liticn (eM)
\ \
\ \
..... '
"
-1,.......,....,... . . . .,..........,~~~,............,.,~,.,....,..,....,. . . . . ....,.....'""T1~ .........;::...,.......~ o
~
~
G
~
U
W
~
~
a
~
~
~
Map pa!liticn (eM) • Figure 4-1 Nonparametric: multipoint linkage analysis results for chromosomes with peaks exceeding lod = 2.0. The results are displayed as the negative 10th logarithm of the probability (P) value for indicated subphenotypes. The X-axis indicates the chromosome distance in centimorgans (eM), and the mic:rosatellite markers are shown across the top. For each chromosome, all subphenotypes that have coincident peaks are shown.
M
C HAP T E R
The selection of candidate genes can be drawn from the literature On JRA, from the literature on other arthropathies, and from candidate loci for other autoimmune diseases in general, the last being especially useful because such data are oriented toward non-HLA genes. The occurrence of autoimmune diseases in JRA families is documented,28,46 as it is in juvenile dermatomyositis, in which, in addition to HLA associations, there is stronger evidence of an increased familial prevalence of autoimmune diseaseY In the instance of juvenile dermatomyositis, the limited occurrence of the disease (approximately 2000 patients in the United States) precludes the possibility of enough ASPs for genome screens, although such pairs do occur. In this instance, simplex families will prove to be the most reliable resource, especially if both parents are available. For the other pediatric rheumatic disorders, studies in adults-both those already reported and those underway, especially for rheumatoid arthritis, SLE, and ankylosing spondylitis-will genelflte candidate genes and loci that can be readily tested for relevance in pediatric populations.48-52
FUNCI10NAL GENONICS Understanding the Expression and Fundlon of Genes For decades, biologists have been interested in the temporal and spatial patterns of expression of individual genes and their protein products, what other gene products mey interact With, and what happens to the cell, organism, or individual when that gene product is mutated or absent. Functional genomics refers broadly to me methods and techniques aimed at answering these questions systematically for all genes encoded in the genome and complements the global approaches to the structure of the genome described earlier. Current estimates of the number of expressed genes in the human genome--those for which messenger RNAs (mRNAs) encode functional proteins-are in the range of 30,000 to 40,000. The number of genes expressed at a given time in a particular cell type depends on a variety of factors but is much smaller. Genes expressed in different cell types overlap, and they also differ considerably depending on the state of differentiation and specialized function of the cell. Gene expression at the level of individual mRNA abundance is generally referred to as the transcriptome, whereas the protein products that result from translation of these mRNAs (i.e., the protein products of the genome) are called the proteome. The ability to measure the transcriptome or proteome of a cell or tissue type has advanced dramatically over the last decade, fueled by the Human Genome Project, advances in microarray technology, and developments in protein chemistry.
Methods to Assess the Transalptome Two .different microarray-based platforms are typically used to Simultaneously assess the abundance of thousands of individual mRNA transcripts. Oligonucleotide microarrays (chips) contain hundreds of thousands of known Single-stranded oligonucleotides synthesized
4
INTEGRATIVE GENOMICS
71
in situ on a solid support. For each gene being probed, there are perhaps a dozen oligonucleotide pairs corresponding to perfect and single-base-pair mismatched sequences, proViding a measure of specificity. RNA samples of interest are reverse transcribed (i.e., DNA complementary to the RNA is synthesized in vitro), and then complementary RNA (cRNA) containing fluorescent labeled nucleotides is synthesized. cRNA is hybridized to the chip, and the resulting fluorescent signal intensity is proportional to the abundance of the particular mRNA species in the original sample. The location and intensity of the fluorescent signal on the chip is determined with the use of a high-resolution scanner and is averaged over all of the oligonucleotide pairs for the particular gene, providing a measurement of its relative expression. One commonly used type of oligonucleotide microarray, known as a GeneChip, is manufactured by Affymetrix (Santa Clara, CA). The Ul33 Plus 2.0 human chip provides virtually genome-Wide expression analysis of more than 47,000 transcripts (based on 1.3 x 106 oligonucleotide features on the chip). Most of these represent known and characterized genes (38,500), whereas others remain as ESTs corresponding to unknown or as yet not well characterized genes. The second method of assessing the transcriptome uses double-stranded cDNAs that are created by PCR and spotted onto glass microarray slides, with one probe per gene. Typically, two mRNA samples, representing different experimental conditions, are labeled with distinct fluorescent probes, then combined and hybridized to the chip. The signal intensity ratio for the two fluorescent probes is used as a measure of the relative amount of the mRNA in the two samples. A detailed comparison of the strengths and weaknesses of these two methods is beyond the scope of this review. However, one advantage of the one sample-one chip approach is that multiple cross-wise comparisons can be made. This is particularly important when comparing clinical samples in large data sets.
Data Analysis Analysis of the very large data sets that result from global microarray studies of differential gene expression is an evolving science that remains to be standardized. Even before analysis, one must consider issues of experimental design, such as whether adequate numbers and quality of samples can be obtained that will provide sufficient power to detect statistically significant and biologically meaningful differences. Sample handling is of critical importance, because exposure of cells to "stress" (e.g., heat, cold) during manipulation can dramatically change gene expression patterns. Furthermore, RNA is particularly susceptible to degradation by enzymes that are released during cell death and are present ubiquitously in the external environment. Consequently, quality control analysis must be included in the overall approach. Estimates of sample sizes for microarray experiments depend on the questions being asked, the complexity of the sample (I.e., number of cell or tissue types represented), and variability among samples. To define a relatively homogeneous disease state using peripheral
72
C HAP T E R
4
INTEGRATIVE GENOMICS
blood or tissue, sample numbers can be expected to range from 5 to 40 or more. Higher numbers allow more disease subclasses to be defined, which can be important when dealing with complex phenotypes, such as those often found in autoimmunity. Given the multiple testing bonanza inherent in analysis of data sets that may comprise 40,000 genes from one sample, conservative interpretation of probability values (and/or multiple testing corrections) is often both prudent and the only method through which manageable numbers of differentially expressed genes emerge. In attempting to define subclasses within a large group of samples, one can use supervised (with prior assumptions) or unsupervised (without prior assumptions) approaches. The best designs have a training or exploratory data set and a built-in capacity for replication of findings established through the training set. Several software packages are available for identifying overexpressed and underexpressed genes across multiple samples, recognizing patterns of expression that cluster together, identifying clusters and pathways with functional significance, and estimating overall similarities and differences between patterns of gene expreSSion in complex samples. In interpreting differential gene expression data, it is important to remember that the relative abundance of individual mRNA species does not always correlate with the abundance of the encoded protein. Many genes are regulated at the translational level, where the mRNA remains in high abundance but is translated only under certain conditions. In addition, the turnover of many individual proteins is tightly regulated by ubiquitination and subsequent degradation by intracellular proteases known as proteasomes.
Proteomlcs Proteomics refers to the systematic study of the structure
and function of all proteins in the genome. 53 Goals include the identification and quantitation of all proteins expressed in a particular cell or tissue type and determination of their modifications, subcellular locations, and interaction partners, including RNA, DNA, and other proteins. Systematic attempts to examine the proteome really began with the development of two-dimensional (20) polyacrylamide gel electrophoresis in the mid-1970s,54 predating the development of methods to sequence DNA.5S.56 This methodology was scaled up to provide enhanced resolution and detection of proteins of low abundance, making it possible to visualize as many as 2000 to 3000 proteins on one gel separation. s7 Unfortunately, the methods were cumbersome and had low throughput, and highly reproducible patterns amenable to automated recognition were difficult to obtain. Furthermore, the information obtained from 20 gels was largely descriptive, because methods to identify individual proteins (see later discussion) were not then available. Methods to assess the proteome have lagged significantly behind genomic techniques for a variety of reasons. Proteins are inherently more complex, with 20 amino acid building blocks rather than 4 nucleotides for DNA/RNA, and there are considerable post-translational modifications, such as glycosylation and phosphoryla-
tion, that affect structure and conformation. Differential mRNA splicing and proteolytic processing of translation products further increases the complexity of the proteome. In many cases a single gene locus may give rise to multiple molecular species, suggesting that the proteome is significantly more complex than the transcriptome. Because resolution of proteins based on migration in 20 gels depends primarily on two parameters, relative molecular mass (M) and isoelectric point (pO, there may be considerable overlap in a complex mixture containing thousands of cellular proteins. Methods such as mass spectroscopy (MS), which provides precise mass measurements, are highly sensitive and provide greater resolution than 20 gel separations do, but they have not been automated and can be prohibitively expensive and timeconsuming for high-throughput analyses. The problem of comigrating proteins with any of these methods can be reduced by "upstream" sample fractionation. However, the tradeoff is lower throughput and greater expense. Global detection of expressed proteins presents additional problems. Protein stains are relatively insensitive for low-abundance proteins. Isotope incorporation coupled with PhosphorImaging (Amersham Biosciences, Piscataway, NJ) can enhance detection but favors visualization of proteins with high rates of synthesis. Antibody-based methods of detection are both specific and sensitive, but they are available for only a fraction of the proteome at this time. Furthermore, most antibodies recognize either conformational or linear epitopes, but not both, and therefore are not amenable to either enzyme-linked immunosorbent assay (ELISA)-based methods or immunoblotting. As a consequence of the difficulties in performing global separation and detection of expressed proteins, new gel-independent methods, many of which are based on MS, are being developed.
Protein Identification by Mass Spectroscopy Peptide Fingerprinting One area of significant advance has been the identification of individual proteins separated from complex mixtures. Typically, a single protein spot from a 20 gel separation is removed from the gel and digested with a protease to yield specific peptide fragments, which are subjected to MS analysis. Matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) MS is most frequently used because of its sensitivity and availability, and it provides highly precise fragment masses. The masses of the peptide fragments determined experimentally (fingerprint) are matched against a database of calculated peptide fragment masses from "in silico" digested proteins, and if the protein of interest is in the database, one can obtain a match with a high degree of certainty that the proteins are identical. It is also possible to obtain peptide sequence information using tandem MS (MS/MS or MS2), in which peptide ion fragments in a complex mixture are isolated in the machine on the basis of their mass (m!z), and then fragmented in the gas phase. Because peptides fragment in a sequence-dependent fashion, in most cases an unambiguous ordering of the amino acids can be obtained from the MS/MS spectrum. This technology has
C HAP T E R
been instrumental in determining the sequences of complex mixtures of peptides derived from HLA class I and class II molecules. 58
Methods for Studying the Proteome Mlcroarray-based Methods Rapid advances in methods for microarray or chip-based analysis of differential gene expression have been possible because of several factors. Probes can be readily synthesized, and, depending on the platform chosen, this can be done in situ. The probes are highly specific, and target recognition does not depend on their secondary structure or that of the target. Targets can also be readily synthesized (copied) and made to incorporate labels that facilitate their detection without significantly changing their properties or ability to be detected. If necessary, targets can be amplified to enhance detection or to facilitate analysis of small amounts of starting material. Essentially none of these factors applies to the global analysis of expressed proteins. For example, proteins cannot be amplified, and they frequently must be in their native conformation to be properly detected. Tagging methods, which can enhance detection, run the risk of altering conformation or the structure of the region being probed. As a consequence of these limitations, there have been few Itlicroarray-based proteomic analyses identifying differentially expressed proteins in complex samples such as peripheral blood, inflammatory lesions, or synovial fluid. Protein or "antigen" arrays have been used successfully to assess autoantibody profiles from patients with various autoimmune diseases. Essentially, protein or peptide autoantigens have been attached to planar surfaces using robotic microarrayers. Up to 1152 features representing 196 distinct antigenic targets of known autoantibodies have been used to screen sera from patients with rheumatoid arthritis, SLE, and other rheumatologic diseases.59 Antibody binding is visualized with anti-human secondary antibodies conjugated to fluoroprobes, and this is followed by scanning and quantitation. The arrays are more sensitive than conventional ELISAs and offer parallel screening for multiple autoantibodies. Results so far have been encouraging, revealing high concordance with results from more conventional methods and a high degree of antigen target specificity. Studies using MS-based or 2D gel methods have also revealed differences in proteins expressed in the synovial fluid or peripheral blood of patients with rheumatoid arthritis, compared with control subjects with reactive arthritis or osteoarthritis. 6o.61
Genetk-based Methods Protein-protein interactions can be detected with the use of genetic methods known as yeast two-hybrid screens. Using molecular biologic tools, a known or "bait" protein can be expressed as a fusion product with the DNAbinding domain of a transcriptional activator. A different protein ("prey") is expressed as a fusion product with the activation domain of the transcription factor. If the bait
4
IN T E G RAT I V E
G E NOM I C S
73
and prey interact when expressed in the yeast, the result is activation of transcription of a reporter gene that can easily be detected. This method can be used to study protein-protein interactions of known gene products or to screen entire libraries to discover interaction partners. Like any method, the yeast two-hybrid system has its limitations, but it has been used successfully for a number of important applications.
Functional Genomlcs In Pediatric Rheumatic Diseases Functional and integrative genomics approaches are currently being applied to the etiology and pathogenesis of complex rheumatologic diseases, including those that primarily affect children. Studies using oligonucleotide microarrays to examine differential gene expression in peripheral blood mononuclear cells (PBMC) from patients with juvenile-onset SLE62 found prominent granulopoiesis and type I interferon (IFN)-response "signatures, " with the latter correlating with SLE Disease Activity Index (SLEDAI) scores. Furthermore, high-dose glucocorticoid treatment, which suppresses disease flares, significantly suppressed the IFN signature. These findings confirmed a role for IFN-a. in SLE pathogenesis. 63 The granulopoiesis signature was more surprising, and further analysis of cell populations led to the identification of highly granular cells in early stages of granulocyte development that were purified together with mononuclear cells during the isolation of PBMC. These were not related to corticosteroid treatment, and they were present in new, untreated patients. Similar evidence for upregulation of IFN-inducible genes in the peripheral blood of patients with adult-onset SLE was noted by Baechler and colleagues,64.65 who also found correlation between the IFN signature and disease severity, particularly with renal and hematologic involvement. These studies have reinforced the potential for targeting type I IFNs in SLE and have led to new ideas about pathogenesis. Furthermore, new biomarkers have been identified that may help investigators and eventually clinicians to monitor disease activity and, potentially, even to predict severity in SLE. Notably, in the studies of Bennett and associates,62 patients with juvenile chronic arthritis (European League Against Rheumatism [EULAR] criteria) who were used as disease controls had very different gene expression patterns from those of the SLE patients. Barnes and associates 65 recently described gene expression differences in PBMC from patients with polyarticular JRA (American College of Rheumatology [ACR] criteria) that readily distinguished them from healthy controls. Results from this pilot study revealed possible differences between the polyarticular and pauciarticular JRA subtypes, as well as juvenile-onset ankylosing spondylitis; however, more patients need to be studied for more definitive distinctions to be made. This work also revealed differential expression of CXCL chemokines, with angiogenic and angiostatic activities in peripheral blood and synovial fluid that may have relevance to disease course and the extent of joint involvement. In addition, Jarvis and coworkers66 found differentially expressed genes in the peripheral blood of patients with polyarticular JRA that
74
C HAP T E R
4
INTEGRATIVE GENOMICS
tended to normalize when the patients responded to treatment. Although functional genomics studies in the pediatric rheumatic diseases are in their infancy, these papers higWight several important points. First, it is striking that peripheral blood is such a rich source of information. Although this might have been expected in active SLE, it is perhaps more surprising in JRA, and it supports the concept that joint inflammation might represent an end result of immune dysregulation, rather than simply a site where joint antigens drive a cross-reactive local inflammatory process. Second, analysis of cell mixtures such as those present in peripheral blood and synovial fluid can provide useful information, despite the complexity of the sample. For example, differences in RNA abundance as detected by microarray analyses can reflect differences in cell populations rather than upregulation or downregulation of genes. Cell type-specific genes can indicate the presence of certain cell populations, such as the early granulocytes in patients with SLE. Third, the comprehensive nature of microarrays affords several advantages, including the ability to simultaneously measure multiple gene products in a pathway (e,g., those responsive to type I IFNs). This can be a more sensitive approach than analyzing the expression of individual genes, or even levels of the cytokines responsible for the signature. For example, it has been difficult to consistently measure increases in IFN-Cl in SLE patients, yet strong evidence for the presence of IFN-Cl (and/or IFN-~) at some point comes from the detection of an IFN signature in peripheral blood. Finally, regardless of the actual identities of the differentially represented genes, so long as there are consistent differences between the groups being compared (e.g., pauciarticular versus polyarticular JRA), it may be possible to identify biomarkers that help distinguish these individuals. In many cases, this can already be done based on clinical features, but there are also many instances in which it is impossible. For example, which patients with new-onset pauciarticular JRA are likely to develop severe erosive arthritis involving multiple joints? Which patients with arthritis, enthesitis, and HLA-B27 will develop ankylosing spondylitis? There are currently few good predictors for these outcomes, but the combination of genotyping for known susceptibility alleles, combined with differential gene expression analyses, may provide clues to some of these questions in the near future.
CONCLUSIONS It seems highly likely that the autoimmune diseases most
commonly encountered by pediatric rheumatologists are complex genetic traits that can be read from an individual's genome. Eventually, diagnosis and prognosis will be based on such traits. It is also evident that genomic polymorphisms will be used to predict responsiveness to therapy. Ultimately, therapy will be matched to an individual's genomic findings, a science known as pharmacogenetics. The methods now being developed to evaluate genomes will also allow gene therapy to be applied to chronic pediatric rheumatic illnesses.
REFERENCES 1. Collins FS, Patrinos A, ]orda E, et al: New goals for the V.S. Human Genome Project: 1998-2003. Science 282: 682-689, 1998. 2. Dermitzakis ET, Reymond A, Scamuffa N, et al: Evolutionary discrimination of mammalian conserved non-genic sequences (CNGs). Science 302: 1033-1035, 2003. 3. Sullivan KE, McDonald-McGinn OM, Driscoll DA, et al: Juvenile rheumatoiu arthritis-like polyarthritis in chromosome 22qll.2 deletion syndrome (DiGeorge anomalad/velocardiofacial syndrome/conotruncal anomaly face syndrome). Arthritis Rheum 40: 430--436, 1997. 4. Weber ]L, May PE: Abundant class of human DNA polymorphisms which can be typed using the polymerase chain reaction. Am] Hum Genet 44: 388-396, 1989. 5. Kruglyak L: The use of a genetic map of biallelic markers in linkage studies. Nat Genet 17: 21-24, 1997. 6. Beaudet AL: 1998 ASHG presidential address. Making genomic medicine a reality. Am] Hum Genet 64: 1-13, 1999. 7. Pennisi E: A closer look at SNPs suggests difficulties. Science 281: 1787-1789, 1998, 8. Martinez FD, Graves PE, Baldini M, et al: Association between genetic polymorphisms of the beta2-adrenoceptor and response to albuterol in children with and without a history of wheezing. ] Clin Invest 100: 3184-3188, 1997. 9, Hershey GK. Friedrich MF, Esswein LA, et al: The association of atopy with a gain-of-function mutation in the alpha subunit of the interleukin-4 receptor. N Engl] Med 337: 1720-1725, 1997. 10. Dib C, Faure S, Fizames C, et al: A comprehensive genetic map of the human genome based on 5,264 microsatellites. Nature 380: 152-154, 1996. 11. Cohen 0, Chumakov I, Weissenbach]: A first-generation physical map of the human genome. Nature 366: 698-701, 1993. 12, The Internationl HapMap Consortium: The International HapMap Project. Nature 426: 789-796, 2003. 13. Strachan T, Abitbol M, Davidson 0, Beckmann ]S: A new dimension for the human genome project: towards comprehensive expression maps. Nat Genet 16: 126--132, 1997. 14. Heller RA, Schena M, Chai A, et al: Discovery and analysis of inflammatory disease-related genes using cDNA microarrays. Proc Nat! Acad Sci V S A 94: 2150-2155, 1998. 15. Reed PW, Davies ]L, Copeman]B, et al: Chromosome-specific microsatellite sets for nuorescence-based, semi-automated genome mapping. Nat Genet 7: 390-395, 1994. 16. Ziegle ]S, Su Y, Corcoran KP, et al: Application of automated DNA sizing technology for genotyping microsatellite loci. Genomics 14: 1026--1031, 1992. 17. Simpson PC, Roach 0, Woolley AT, et a1: High-throughput genetic analysis using microfabricated 96-sample capillary array electrophoresis microplates. Proc Nat! Acad Sci USA 95: 2256--2261, 1998. 18. Risch N: Linkage strategies for genetically complex traits: n. The power of affected relative pairs. Am] Hum Genet 46: 229-241, 1990. 19. Spielman RS, McGinnis RE, Ewens W]: Transmission test for linkage uisequilibrium: the insulin gene region and insulin-dependent diabetes melHtus (IDDM), Am] Hum Genet 52: 506--516, 1993. 20. Spielman RS, McGinnis RE, Ewens W]: The transmission/disequilibrium test detects cosegregation and linkage. Am ] Hum Genet 54: 559-560; author reply 560-563, 1994. 21. Spielman RS, Ewens W]: A sibship test for linkage in the presence of association: the sib transmission/disequilibrium test. Am ] Hum Genet 62: 450-458, 1998. 22. Chalom EC, Ross ], Athreya BH: Syndromes and arthritis. Rheum Dis Clin North Am 23: 709-727, 1997. 23. Prahalad S, Colbert RA: Genetic diseases with rheumatic manifestations in children. Curr Opin Rheumatol 10: 488-493, 1998. 24. Lander ES, Schork N]: Genetic dissection of complex traits, Science 265: 2037-2048, 1994. 25. Risch N, Merikangas K: The future of genetic studies of complex human diseases. Science 273: 1516--1517, 1996. 26. Davies ]L, Kawaguh Y, BenneU, et al: A genome-wide search for human type 1 diabetes susceptibility genes. Nature 371: 130-136, 1994. 27. Rosen P, Thompson S, Glass 0: Non-HLA gene polymorphisms in juvenile rheumatoid artluitis. Clin Exp Rheumatol 21: 650-656, 2003. 28. Prahalad S, Shear ES, Thompson SO, et al: Increaseu prevalence of familial autoin1fnunity in simplex and multiplex families with juvenile rheumatoid arthritis, Artllfitis Rheum 46: 1851-1856, 2002. 29. Weis II. McCracken BA, Ma Y, et al: Identification of quantitative trait loci governing arthritis severity and humoral responses in the murine model of Lyme disease. ] Immunol 162: 948-956, 1999. 30. Risch N: Linkage strategies for genetically complex traits: 1. Multilocus mouels. Am] Hum Genet 46: 222-228, 1990. 31. Adachi M, Watanabe-Fukunaga R, Nagata S: AbelTant transcription caused by the insertion of an early transposable element in an intron of the Fas antigen gene of lpr mice. Proc Nat! Acad Sci V S A 90: 1756--1760, 1993. 32. Fisher GH, Rosenberg ], Straus SE, et al: Dominant interfering Fas gene mutations impair apoptosis in a human autoimmune Iymphoproliferative syndrome. Cell 81: 935-946, 1995.
C HAP T E R 33. Becker KG, Simon RM, Bailey-WilsonjE, et aI: Clustering of non-major histocompatibility complex susceptibility candidate loci in human autoimmune diSeases. Proc Nat! Acad Sci USA 95: 9979-9984, 1998. 34. Kawahito Y, Cannon GW, Gulko PS, et al: Localization of quantitative trait loci regulating adjuvant-induced arthritis in rats: evidence for genetic facto~s common to multiple autoimmune diseases. J Immunol161: 4411--4419, 1998. 35. Gonzalez-Roces S, Alvarez MY, Gonzalez S, et al: HLA-B27 polymorphism and worldwide susceptibility to ankylosing spondylitis. Tissue Antigens 49: 116--123, 1997. 36. Cleves MA, Olson JM, Jacobs KB: Exact transmission-disequilibrium tests with multiallelic markers. Genet Epidemiol 14: 337-347, 1997. 37. Thomson G: Mapping disease genes: family-based association studies. Am J Hum Genet 57: 487--498, 1995. 38. SL'Ott WK, Pericak-Vance MA, Haines ]L: Genetic analysis of complex diseases. Science 275: 1329-1330, 1997. 39. AjiPka RS, Yu P, Gnten JR, et al: Recombinations defining centromeric and telpmeric borders for the hereditary haemochromatosis locus. J Med Genet 34: 28-33, 1997. 40. Gandon G, Jouanolle AM, Chauvel B, et al: Linkage disequilibrium and extended haplotypes in the HLA-A to D6SI05 region: implications for mapping the hemochromatosis gene (HFE). Hum Genet 97: 103-113, 1996. 41. Hasthacka J, de 1a Chapelle A, Mahtani MM, et al: The diastrophic dysplasia gene encodes a novel sulfate transporter: positional cloning by fine-stntcture linkage disequilibrium mapping. Cell 78: 1073-1087, 1994. 42. Moroldo MB, Chaudhari M, Shear E, et al: Juvenile rheumatoid arthritis affected sibpairs: extent of clinical phenotype concordance. Arthritis Rheum 50: 1928-1934, 2004. 43. Moroldo MB, Donnelly P, Saunders J, et al: Transmission disequilibrium as a test of linkage and association between HLA alleles and pauciarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 41: 1620-1624, 1998. 44. Pf'~halad S, Ryan MH, Shear ES, et al: Juvenile rheumatoid arthritis: linkage to HLA demonstrated by allele sharing in affected sibpairs. Arthritis Rheum H: 2335-2338, 2000. 45. Thompson S, Moroido MB, Guyer L, et al: A genome-wide scan for juvenile rheumatoid arthritis in affected sibpair families provides evidence of linkage. Arthritis Rheum 50: 2920-2930, 2004. 46. Rossen R, Brewer EJ, Sharp RM, et al: Familial rheumatoid arthritis: linkage of HLA to disease susceptibility locus in four families where proband presented with juvenile rheumatoid arthritis. J Clin Invest 65: 629-642, 1980. 47. Ginn LR, Lin JP, Plotz PH, et al: Familial autoimmunity in pedigrees of' idiopathic inflammatory myopathy patients suggests common genetic ri~k factors for many autoimmune diseases. Arthritis Rheum 41: 400--405, 1998. 48. Jawaheer 0, Seldin MF, Amos CI, et al: Screening the genome for rheumatoid arthritis susceptibility genes: a replication study and combined analysis of 512 multicase families. Arthritis Rheum 48: 906--<)16, 2003.
4
INTEGRATIVE GENOMICS
75
49. Tsao BP, Cantor RM, Kalunian KC, et al: Evidence for linkage of a candidate chromosome 1 region to human systemic lupus erythematosus. J Clin Invest 99: 725-731, 1997. 50. Wordsworth P: Genes in the spondyloarthropathies. Rheum Dis Clin North Am 24: 843-862, 1998. 51. Moser KL, Neas BR, Salmon JE, et al: Genome scan of human systemic lupus erythematosus: evidence for linkage on chromosome 1q in African-American pedigrees. Proc Natl Acad Sci USA 95: 14869-14874, 1998. 52. Gaffney PM, Keams GM, Shark KB, et al: A genome-wide search for susceptibility genes in human systemic lupus erythematosus sib-pair families. Proc Nat! Acad Sci USA 95: 14875-14879, 1998. 53. Patterson SO, Aebersold RH: Proteomics: the first decade and beyond. Nat Genet 33 (Suppl): 311-323, 2003. 54. O'Farrell PH: High resolution two-dimensional electrophoresis of proteins. J Bioi Chem 250: 4007--4021, 1975. 55. Sanger F, Nick/en S, Coulson AR: DNA sequencing with chain-terminating inhibitors. Proc Nat! Acad Sci USA 74: 5463-5467, 1977. 56, Maxam AM, Gilbert W: A new method for sequencing DNA, Proc Natl Acad Sci USA 74: 560-564, 1977. 57, Young DA, Voris BP, Maytin EV, Colbert RA: Very-high-resolution twodimensional electrophoretic separation of proteins on giant gels. Methods Enzymo191: 190-214, 1983, 58. Hunt OF, Henderson RA, Shabanowiz J, et al: Characterization of peptides bound to the class I MHC molecule HLA-A2.1 by mass spectrometry. Science 255: 1261-1263, 1992. 59. Robinson WH, Steinman L, Utz PJ: Protein arrays for autoantibody profiling and fine-specificity mapping. Proteomics 3: 2077-2084, 2003. 60. Sinz A, Bantscheff M, Milckat S, et al: Mass spectrometric proteome analyses of synovial fluids and plasmas from patients suffering from rheumatoid arthritis and comparison to reactive arthritis or osteoarthritis. Electrophoresis 23: 3445-3456, 2002. 61. Dotzlaw H, Schulz M, Eggert M, Neeck G: A pattern of protein expression in peripheral blood mononuclear cells distinguishes rheumatoid arthritis patients from healthy individuals. Biochim Biophys Acta 1696: 121-129, 2004, 62. Bennett L, Palucka AK, Arce E, et al: Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med 197: 711-723, 2003. 63. Blanco P, Palucka AK, Gill M, et al: Induction of dendritiC cell differentiation by IFN-alpha in systemic lupus erythematosus, Science 294: 1540-1543, 2001. 64. BaecWer EC, Batliwalla FM, Karypis G, et al: Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Nat! Acad Sci USA 100: 2610-2615, 200365, Barnes M, Aronow BJ, Luyrink LK, et al: Gene expression in juvenile arthritis and spondyloarthropathy: pro-angiogenic ELR+ chemokine genes relate to COurse of arthritis, Rheumatology (Oxford) 43: 973-979, 2004. 66, Jarvis IN, Dozmorov 1, Jiang K, et al: Novel approaches to gene expression analysis of active polyarticular juvenile rheumatoid arthritis. Arthritis Res Ther 6: RI5-R32, 2004.
C H A P T E R
5
P HARMACOLOGY D RUG T HERAPY
AND
Ronald M. Laxer
The previous edition of this textbook described the tremendous progress that had been made over the preceding decade in the treatment of patients with inflammatory arthritis and other rheumatic diseases. Since that time, the development of new agents has continued, extensive clinical experience with the use of the new biologic agents has been gained, and the clinical indications for the use of these agents have been expanded. All this has resulted in better outcomes for patients with rheumatic diseases. The principal drugs used in pediatric rheumatology are those that suppress the inflammatory and immune responses. The targets of their therapeutic effects are predominantly the arachidonic acid metabolic pathways and the cells of the immune system and their products. This chapter outlines some of the most important general principles relating to use of these medications, particularly as they apply to children. The treatment of specific rheumatic disorders is detailed in the chapters dealing with each disease.
CONCEPTS IN PHARMACOLOGY Although often incomplete, knowledge of the pharmacokinetics of drugs used to treat childhood rheumatic diseases contributes to understanding of their clinical applications. This brief overview can be supplemented by referring to standard works in the field.1–3
Drug Absorption and Bioavailability Most drugs are given by the oral route and are absorbed through the mucosa of the gastrointestinal (GI) tract, particularly that of the small intestine. GI absorption may be influenced by a number of factors, including the presence or absence of food in the gastric lumen, luminal pH, gastric emptying time, and the coadministration of other drugs. Drug bioavailability, the net result of these factors, is usually determined by sequential measurement of plasma drug concentrations. Three parameters are routinely considered: peak drug concentration, the time necessary to reach peak concentration, and the area under the time-concentration curve. The area under the
76
curve after intravenous administration is considered equivalent to complete absorption after oral administration. Because the effects of many repeatedly administered drugs are cumulative, except with drugs of extremely short half-life given at infrequent intervals, bioavailability is best determined at the mean steady-state concentration of the drug—that is, the point at which drug intake is equal to drug elimination.
Volume of Distribution The volume of distribution is the volume of fluid into which a drug would need to be distributed to achieve a concentration equal to the concentration ultimately measured in plasma. If the drug stays in the plasma, its volume of distribution is smaller than if it is distributed widely in tissues. Drugs in the body are either free or bound to plasma proteins or tissue lipids. The extent and nature of binding affect the volume of distribution of the drug, the rate of renal clearance (because only free drug is filtered by the glomerulus), the drug half-life, and the amount of free drug that reaches the target tissue or receptor. Most acidic drugs are bound to plasma albumin, whereas the basic drugs are bound to lipoproteins, α1-acid glycoproteins, and globulins. In inflammatory states, plasma albumin concentration falls and α1-acid glycoproteins increase, although the extent of the decrease usually does not require any change in drug therapy. Drugs that are highly protein-bound tend to stay within the vascular compartment and have a relatively limited volume of distribution. Those that are widely bound to lipids in tissues have large volumes of distribution.
Half-Life and Clearance The half-life of a drug is the time necessary for the serum concentration to fall by 50% during the elimination phase of a time-concentration curve. Clearance is a measure of the removal of a drug from the body as a whole or from a specific part of the body, such as liver or kidney. It is expressed as the volume of body fluid from which a drug is removed per unit of time.
C H A P T E R
The first-pass effect refers to the rapid breakdown of a drug when it passes through the intestinal mucosa or enters the liver for the first time. In this case, the drug reaches the systemic circulation predominantly in the form of metabolites that may be pharmacologically inactive. Avoidance of this effect requires intravenous administration. When the rate of elimination of a drug is directly proportional to its concentration in the body, the drug is said to have first-order kinetics. Drugs that are eliminated at a constant rate, unrelated to the amount of the agent in the body, are said to follow zero-order kinetics. Some drugs obey capacity-limited kinetics: at low concentrations, first-order kinetics are observed; at higher concentrations, the enzymes used in metabolism of the drug are saturated, and zero-order kinetics is approached. Salicylate and its metabolites demonstrate this phenomenon. Time-dependent kinetics may be altered by the effect of drug metabolites. For most drugs, a steady state is not reached until the passage of five half-lives. Blood levels measured before that time may be erroneous.
Drug Biotransformation Drug biotransformation or metabolism principally occurs in the liver, kidney, skin, and GI tract. In the liver, biotransformation involves hydrolysis, oxidation, reduction, or demethylation and conjugation of the metabolite with glycine, glucuronide, sulfate, or hippurate with subsequent secretion into the bile. In the kidney, drugs may be filtered, filtered and secreted, filtered and passively reabsorbed (e.g., acetaminophen), or filtered or actively secreted and passively reabsorbed (e.g., salicylates).4 Many drugs used to treat rheumatic diseases are active in the form in which they are administered; exceptions include sulindac, salsalate, prednisone, cyclophosphamide, and azathioprine, which require biotransformation before they exert their principal effects. Because the liver and kidney play such key roles in drug metabolism, dysfunction of these organs may require alteration in drug dose. In general, however, additional toxicity caused by drugs that totally depend on the kidney for their elimination is not a danger unless renal function is diminished by more than 50%.2 In patients with significant renal or hepatic disease, monitoring of drug levels and attention to the potential of drug toxicity become more critical.
5 PHARMACOLOGY TABLE 5–1
AND
77
D RU G T H E R A P Y
Agents Used to Treat Rheumatic Diseases
Nonsteroidal anti-inflammatory drugs Disease-modifying antirheumatic drugs/slow-acting anti-inflammatory drugs Glucocorticoids Cytotoxic or immunosuppressive drugs Biologic immunomodulators
connective tissue diseases. They provide symptomatic anti-inflammatory relief and are recommended for the majority of patients with juvenile rheumatoid arthritis (JRA). In the United States, only acetylsalicylic acid (aspirin, ASA), tolmetin, naproxen, ibuprofen, and indomethacin are approved for use in young children. In Australia, only aspirin, naproxen, and ibuprofen are licensed for use in children. The range of actual NSAID use in childhood is broader in these and many other areas of the world and usually extends beyond the range of drugs approved for use in children and the manufacturer’s recommendations in terms of dosage and minimum age for use.5 In general, ASA is now used much less often because it is associated with a greater frequency of adverse effects (including Reye’s syndrome, discussed later) and is without any demonstrated superiority in
COOH OC
CH3
O
CO
ASPIRIN (acetylsalicylic acid)
CI
N CH3 CH2COOH
CH3O OH
INDOMETHACIN
COO⫺ CH2 CHOLINE MAGNESIUM COO
F
COOH
OOC
CH3
HO
OH
H
CHOLINE MAGNESIUM TRISALICYLATE
H3C
CHCH2
CH3
S
CHCOOH
CH3
H3C
O
SULINDAC IBUPROFEN
O CH3
C
H3C
CH3COONa
N
CHCOOH
CH3 O
TOLMETIN SODIUM
ANTIRHEUMATIC DRUGS
CO2Na
FENOPROFEN
The pharmacologic agents used to treat children with rheumatic disorders are grouped into five categories: the nonsteroidal anti-inflammatory drugs (NSAIDs), diseasemodifying or slow-acting antirheumatic drugs, glucocorticoids, cytotoxic or immunosuppressive agents, and biologic response modifiers (Table 5–1).
CH3 CI
CH3 NAPROXEN
MECLOFENAMATE SODIUM CH2COONa
O
KETOPROFEN
■ Figure 5–1
O
OH
C
NH
NH CI
N CI
C
The NSAIDs (Fig. 5–1) remain the mainstay of the initial treatment of the chronic arthropathies of childhood and have a role in management of some aspects of the other
CI
CH3O
CH3CHCOOH
Nonsteroidal Anti-inflammatory Drugs
NH
CHCOOH
N S
CH3
O O DICLOFENAC
PIROXICAM
Structures of nonsteroidal anti-inflammatory drugs (NSAIDs).
78
5 PHARMACOLOGY
C H A P T E R
H2N
AND
D RU G T H E R A P Y
O
Mechanism of Action
S
O N
N
CF3
H3C CELECOXIB
O O
O S H3N
O ROFECOXIB
■ Figure 5–2
Structures of cyclooxygenase 2 (COX-2) inhibitors.
terms of efficacy compared with other NSAIDs for the treatment of arthropathies; however, it continues to have a major role in the treatment of Kawasaki disease. ASA is also less convenient to use because of the need for more frequent dosing, its poor solubility in liquid form, and the necessity of monitoring serum levels. Since the last edition of this textbook, the role of Cox-2 inhibitors (Figure 5–2) has evolved tremendously. Recently, the COX-2 inhibitor rofecoxib was reported to be safe and effective in children with JRA.6 However, the role of the Cox-2 inhibitors is currently under review as recent data have suggested an increased incidence of cardiovascular events in adults with long-term use.6a-6c
NSAIDs inhibit pro-inflammatory pathways that lead to chronic inflammation. The major anti-inflammatory effect of NSAIDs is mediated by inhibition of the COX enzyme in the metabolism of arachidonic acid to prostaglandins, thromboxanes, and prostacyclins7 (Fig. 5–3) (see Chapter 3). Currently available NSAIDs (except diclofenac and indomethacin) have little effect on the lipoxygenase pathway, the other major pathway of arachidonic acid metabolism.8 The discovery of two related but unique isoforms of the COX enzyme, COX-1 and COX-2, has resulted in a greater understanding of the mechanism of action of NSAIDs.9–12 These enzymes are structurally very similar but are encoded by distinct genes and differ in their distribution and expression in tissues. The COX-1 isoenzyme has a wide tissue distribution and is constitutively expressed under basal conditions. It appears to have a “housekeeping” function and is associated with the production of prostaglandins resulting in diverse physiologic effects such as gastric cytoprotection, platelet aggregation, vascular homeostasis, and maintenance of renal blood flow (see Fig. 5–3). In contrast, COX-2 is an inducible enzyme that is upregulated at sites of inflammation by various pro-inflammatory mediators, including interleukin-1 (IL-1), tumor necrosis factor-α (TNF-α), bacterial endotoxins, and various mitogenic and growth factors. It also appears to be expressed in the central nervous system (CNS) and to have a role in the central mediation of pain and fever. After the discovery of COX-2 in the early 1990s,13 it was hypothesized that COX-1 mediated uniquely physiologic prostaglandin production whereas COX-2 mediated uniquely pathologic prostaglandin production; consequently, the anti-inflammatory effects of NSAIDs were considered to result from COX-2 and the adverse effects from COX-1 inhibition. However, this appears to have been an oversimplification.14 COX-2 may also have a
■ Figure 5–3 Synthesis of prostaglandins and leukotrienes. Sites of action of anti-inflammatory agents and cytokines are highlighted by arrows (–, downregulation; +, upregulation). COX, cyclooxygenase.
MEMBRANE PHOSPHOLIPIDS Phospholipase A2 ARACHIDONIC ACID
Cyclo-oxygenase
Lipoxygenase
⫹ LEUKOTRIENES
COX-1 Constitutive
COX-2 Inducible ⫺
⫺
• NSAIDs (reversible) • Aspirin (irreversible) May also be induced at sites of inflammation
“PHYSIOLOGICAL PROSTAGLANDINS” • gastrointestinal cytoprotection • renal blood flow • platelet aggregation • vascular homeostasis
• endotoxins • cytokines • growth factors
⫺ • COX-2 inhibitors • Corticosteroids
“PATHOLOGICAL PROSTAGLANDINS”
May also have physiological role in:
• sites of inflammation • central mediation of pain and fever
• kidney • brain • ovary and uterus • cartilage and bone • healing of gastrointestinal inflammation or ulcers
C H A P T E R
physiologic function in certain tissues; it is expressed constitutively in structures such as the ovary, uterus, brain, kidney, cartilage, and bone.12 COX-2 knockout mice have severe renal dystrophy, suggesting an important role for this isoenzyme in early renal development. Conversely, COX-1 may have nonphysiologic functions and, like COX-2, may be upregulated at sites of inflammation. Therefore, the two isoforms of COX each appear to have a role in both physiologic and pathologic prostaglandin production and to have more broad and complex functions than originally thought. Currently available NSAIDs inhibit both isoforms of COX, but most inhibit COX-1 preferentially, resulting in undesirable adverse effects such as GI toxicity while producing desirable anti-inflammatory effects through concurrent inhibition of COX-2. Existing NSAIDs are known to differ in the degree of inhibition of COX-2, compared with COX-1, that they produce. This has been found to correlate with their adverse-effect profiles: NSAIDs that are more selective for COX-2 appear to have more favorable adverse effect profiles.10 In light of the differences between the COX-1 and COX-2 isoenzymes and their binding sites for NSAIDs, there has been a profusion of research into a “safer” class of drugs that selectively bind to and inhibit COX-2 activity: the COX-2 inhibitors.9 However, the degree of COX-2 suppression needed to produce an anti-inflammatory and analgesic effects in vivo is uncertain—a threshold degree of COX-2 inhibition that must be achieved with no associated COX-1 suppression cannot be clearly defined at this time.11,15 A number of additional concerns remain about specific COX-2 inhibitors. Because COX-1 may also be induced at sites of inflammation, a specific COX-2 inhibitor may be less effective as an anti-inflammatory or analgesic agent.9,10,14 Recent data from several trials not yet published examining the Cox-2 inhibitors in the prevention of colonic disease (cancer and recurrent polyposis) have shown an increased incidence of cardiovascular events. As a result, the production of rofecoxib has been halted entirely by the manufacturer.15a Similar concerns have arisen with the use of valdecoxib, and although studies suggest that celecoxib appears to be safer, long-term use at high doses may also be associated with an increased risk of CVS events.15b Whether the events are unique to one, several or all drugs from the COX-2 class is still unclear and requires further study.15c Similarly, it is not known whether the increased cardiovascular risk is related to co-morbid cardiovascular risk factors. Until further data are available, many feel that Cox-2 inhibitors have little role to play in the treatment of children with JRA. Surprisingly, 440 mg/day of naproxen was associated with an increased cardiovascular risk in a recent study (data not yet published). The mechanism by which COX-2 inhibitors are thought to lead to increased risk relate to relatively unopposed inhibition of vasodilator and platelet inhibitory prostaglandins, without inhibiting vasoconstrictor and platelet activating prostaglandins. Currently, it is believed that COX-2 inhibitors should be administered with low-dose aspirin in patients with cardiac risk factors.16 The consequences of specific COX-
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
79
2 inhibition in tissues wherein the COX-2 isoenzyme appears to have a physiologic role, such as the renal medulla, ovary, and uterus, is also unknown.9,10 Renal toxicity may occur with COX-2 inhibitors. Three COX-2 inhibitors (celecoxib, rofecoxib, and valdecoxib) have been launched in the United States, and others are in various stages of development. The doses of existing NSAIDs that are required to reduce inflammation are generally higher than those needed to inhibit prostaglandin formation, suggesting the existence of other mechanisms by which their antiinflammatory effects are mediated (Table 5–2). In addition to inhibiting prostaglandin production, current NSAIDs inhibit specific proteinases involved in degradation of proteoglycans and collagens of cartilage17 and inhibit the generation of oxygen radicals, particularly superoxide.18–20 NSAIDs also have been shown to interfere with bradykinin release, response of lymphocytes to antigenic challenge, phagocytosis, and chemotaxis of granulocytes and monocytes.21 Individual NSAIDs may have additional specific mechanisms of action. Indomethacin blocks the action of phosphodiesterase, thus increasing intracellular cyclic adenosine monophosphate.22 This effect leads to a decrease in the generation of superoxide and hydroxyl radicals.22–24 Indomethacin also inhibits the mobility of polymorphonuclear neutrophils in inflammatory sites. Like ASA and ibuprofen, indomethacin uncouples oxidative phosphorylation and decreases the synthesis of mucopolysaccharides.25 Diclofenac and indomethacin also limit the availability of the substrate for prostaglandin and leukotriene synthesis by facilitating incorporation of arachidonic acid into triglycerides.8,26 Piroxicam at high concentrations inhibits neutrophil migration, phagocytosis, lysosomal enzyme release,27 and oxygen radical production by neutrophils.28 Meclofenamate sodium may inhibit prostaglandin binding at its receptor and inhibits phospholipase A2.29 Meclofenamate also inhibits the lipoxygenase pathway of arachidonic acid metabolism.30 Other differences in the mechanism of action of various NSAIDs have been reviewed.8,31
TABLE 5–2
Processes Influenced by NSAIDs
Prostaglandin production Leukotriene synthesis Superoxide generation Lysosomal enzyme release Neutrophil aggregation and adhesion Cell-membrane functions Enzyme activity (NADPH oxidase, phospholipase C) Transmembrane anion transport Oxidative phosphorylation Uptake of arachidonate Lymphocyte function Rheumatoid factor production Cartilage metabolism NADPH, nicotinamide adenine dinucleotide phosphate, reduced form; NSAIDs, nonsteroidal anti-inflammatory drugs. From Brooks PM, O’Day RO: Nonsteroidal antiinflammatory drugs: differences and similarities. N Engl J Med 324: 1716–1725, 1991. Copyright © 1993 Massachusetts Medical Society. All rights reserved.
80
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Pharmacology The pharmacokinetic evaluation of NSAIDs in children with JRA has been variable, ranging from extensive for the salicylates to minimal or none with the newer agents; the interested reader is referred to reviews on the subject.32,33 However, studies in adults indicate that most NSAIDs share similar pharmacokinetic properties. They are weakly acidic drugs that are rapidly absorbed after oral administration, with most of the absorption taking place in the stomach and upper small intestine. Circadian rhythms in gastric pH and intestinal motility may lead to variability in NSAID absorption, with reduced absorption of a dose given at night compared with that given in the morning.34 The majority of NSAIDs are strongly protein bound, primarily to albumin, leading to a potential for drug–disease and drug–drug interactions. Hypoalbuminemia may occur as a manifestation of disease activity in JRA, especially in patients with systemic-onset disease, and may be one of the most important factors influencing the pharmacokinetics of NSAIDs in these children. Because clinical effects are determined by unbound or free drug levels, states of hypoalbuminemia may be associated with a corresponding increase in the unbound fraction and hence with a potential for increased toxicity. However, most of the studies of NSAID pharmacokinetics in children do not report the level of disease activity.35 Protein binding may also be reduced with renal or hepatic disease. Although the strong plasma protein binding of NSAIDs also makes possible drug–drug interactions with other highly protein-bound drugs, significant clinical interactions are rare.36,37 Furthermore, children rarely need to be prescribed drugs that have a potential for interacting with NSAIDs. However, NSAIDs may potentially interact with methotrexate (MTX) through several mechanisms, including displacement from plasma protein–binding sites, competition for renal secretion, and impairment of renal function. Although the impact of NSAIDs on MTX clearance varies widely and the potential for clinically significant interactions exists in some children,38 MTX–NSAID interactions are rarely of clinical significance. The kinetics of NSAIDs at their anti-inflammatory sites of action (e.g., synovial fluid) may be more clinically relevant than their kinetics in plasma. The comparative kinetics of NSAIDs in plasma and synovial fluid are related to the half-life of the drug and to differences in protein binding at these sites. Studies in adults indicate that NSAIDs with short half-lives have less fluctuation in synovial fluid concentrations than in plasma concentrations over a given dosage interval.36 This phenomenon may partially account for the fact that the dosage interval of these drugs is longer than their plasma half-life. In addition, because synovial fluid albumin concentrations are lower than plasma concentrations, the free fraction of NSAIDs in synovial fluid can be significantly higher than in plasma and has been shown to correlate with clinical effectiveness.34 This may account for clinical effects observed with relatively low plasma drug levels. Except for naproxen and ASA, plasma concentrations correlate poorly with anti-inflammatory activity.39
NSAIDs are eliminated predominantly by hepatic metabolism; only small amounts are excreted unchanged in urine. Some NSAIDs, such as sulindac or indomethacin, are also secreted in significant amounts in bile and undergo enterohepatic recirculation.36 Most NSAIDs are metabolized by first-order or linear kinetics, whereas salicylate is metabolized by zero-order or nonlinear kinetics. For this reason, dosage adjustments are frequently required with ASA therapy, and small changes in dose may lead to large fluctuations in serum levels of ASA at the higher end of the therapeutic range.33 Naproxen may also demonstrate nonlinear pharmacokinetics at doses greater than 500 mg/day in adults because of the saturation of plasma protein–binding sites and associated increase in clearance.33,34 Interpatient differences in the metabolic clearances of individual NSAIDs may be marked, resulting in considerable variation in the extent of their accumulation at all sites throughout the body.36 In children, NSAIDs may be eliminated more rapidly than in adults; therefore, children may require more frequent doses to maintain a clinical response.35 Differences in pharmacokinetics between short- and long-term administration may be significant, but this has not been studied systematically.32 Abnormalities of hepatic function are common in children with JRA, particularly those with systemic-onset disease. Because hepatic metabolism plays a major role in NSAID elimination, it is necessary to assess hepatic function before institution of NSAID therapy in these children. NSAIDs should not be started if there is significant elevation of transaminase levels (e.g., three times normal or higher).
General Principles of NSAID Therapy The NSAIDs are generally good analgesic and antipyretic drugs and weak anti-inflammatory agents. They provide good symptomatic relief but have traditionally not been considered to influence the underlying disease process or to significantly affect long-term outcomes. The analgesic effect of NSAIDs is rapid, but the anti-inflammatory effect takes longer and requires doses up to twice as large as those needed for analgesia.40,41 NSAIDs are relatively safe for long-term use. Toxicity, although not infrequent, especially for GI side effects, is seldom serious.42–44 Given the wide variety of available NSAIDs, a few general principles can be applied in the selection of a particular NSAID for therapy in an individual patient (Table 5–3).
TABLE 5–3
General Principles of NSAID Use
A specific diagnosis should be established. The objectives of therapy (analgesia or suppression of inflammation) should be understood. The objectives of therapy must be balanced against the risks of toxicity and cost of the drug. An adequate trial (appropriate dose for at least 6–8 wk) is needed before assessing efficacy. Use of combinations of NSAIDs should be avoided. Clinical and laboratory evidence of effect and toxicity should be objectively monitored. NSAIDs, nonsteroidal anti-inflammatory drugs.
C H A P T E R
First, according to empirical evidence from clinical experience and some studies in adults, response to NSAIDs appears to have some disease specificity. Indomethacin, one of the more potent NSAIDs, may be more useful in treating manifestations of systemic JRA, such as fever and pericarditis, and in managing the spondyloarthropathies. Ibuprofen is also a very effective antipyretic agent in systemic-onset JRA. Sustained-release preparations of naproxen or, in the older child, indomethacin, given at night may be effective in reducing night pain or prolonged morning stiffness. However, studies suggest that differences in the effectiveness of NSAIDs are small but that some NSAIDs, such as ASA or indomethacin, are more toxic than others.40 Second, individual patient response to NSAIDs is variable and often relatively unpredictable; a child may fail to respond to one drug and yet respond to another. Similarly, the frequency of toxicity may vary widely. A favorable initial response occurs in more than 50% of patients on the first NSAID chosen; of those showing an inadequate response to the first NSAID, a further 50% improve when given another drug of the same class.33 Therefore, for the child whose condition has not responded to one NSAID or who is experiencing significant toxicity, a subsequent trial with another agent is usually warranted. An adequate trial of any NSAID should not be less than about 8 weeks5,45; although about 50% of children who respond favorably to an NSAID do so by 2 weeks of therapy, the mean time to therapeutic response can be 4 weeks, and up to 25% may not respond until after approximately 12 weeks of therapy.46 Third, additional factors such as availability in liquid form, frequency of dosing, cost, and tolerability of any given NSAID may influence patient preference. These factors can have a major impact on patient adherence to a particular treatment regimen and should be carefully evaluated when making therapeutic choices. A reasonable initial approach is to choose an NSAID that has a favorable toxicity–efficacy profile, can be taken on a convenient schedule (such as once or twice daily), is not too expensive, and, for young children, is available in a liquid formulation that is palatable. For these reasons, naproxen has become the initial drug of choice for children with arthritis. It is generally well tolerated and safe, is administered twice daily, and is available in a palatable liquid form.47 A recent open-label study of nabumetone in children with JRA reported an excellent safety profile with no loss of efficacy compared with previously used NSAIDs. The drug was administered at a dose of 30 mg/kg/day, in a single dose, and can be given as a liquid slurry.48 Generally, therapy should commence with the NSAID of choice at the lowest recommended dose, which can then be titrated to the patient’s clinical response. Use of multiple NSAIDs is not recommended, because this approach has no documented benefit in terms of efficacy and can be associated with a greater potential for drug interactions and organ toxicity. The dose range and schedule of administration vary with the individual NSAID (Table 5–4). Patients who are receiving long-term NSAID therapy should have their liver and renal function
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
81
monitored, and a complete blood count should be obtained at least every 6 months. Patients with active systemic disease should have more frequent monitoring of liver function, including serum albumin, particularly with any changes in dose. Once a patient has responded and is considered to be in clinical remission, withdrawal of NSAID therapy can be considered. Although there are no good studies of NSAID discontinuation in children with JRA to guide recommendations, most pediatric rheumatologists consider it reasonable to attempt gradual withdrawal of therapy after remission has been maintained for at least 6 months. Adherence to NSAID regimens may be improved by a variety of behavioral interventaions.49,50
Toxicity Serious toxicity associated with the use of NSAIDs appears to be rare in children.41 However, much of the available information is derived from case reports, case series, or retrospective cohort studies, making it difficult to derive accurate figures for the incidence and prevalence of various toxicities. Prospective studies using standardized approaches to measuring drug-related toxicities are needed to allow a better quantification of the magnitude of risk for the various adverse effects associated with NSAID therapy. In general, however, most toxicities are shared to a greater or lesser degree by all NSAIDs (Table 5–5), although individual patients may have fewer side effects with one drug than with another.23,42,43,51,52
Gastrointestinal Toxicity GI tract toxicity is common to all NSAIDs. The pathogenesis of gastroduodenal mucosal injury involves multiple mechanisms with both local and systemic effects due to NSAIDs.53 The systemic effects appear to have a predominant role and are largely the result of inhibition of prostaglandin synthesis, which leads to impairment of many cytoprotective actions such as epithelial mucus production, secretion of bicarbonate, mucosal blood flow, epithelial proliferation, and mucosal resistance to injury.53 The associated symptoms range from mild epigastric discomfort immediately after taking the medication to symptomatic or asymptomatic peptic ulceration.54 The incidence of symptomatic ulcers and potentially life-threatening ulcer complications, such as upper GI bleeding, perforation, and gastric outlet obstruction, is about 2% to 4% in adults taking NSAIDs for 1 year.55 The relative risk of upper GI bleeding and perforation associated with current NSAID use is estimated to be 4.7 overall, with variability in the magnitude of risk associated with individual NSAIDs.56 Complication-related hospitalization and death rates are estimated to be less than 1.5% per annum in adults.57 However, many of the studies on which these figures are based have limitations in ascertainment as well as attribution of adverse effects to NSAIDs, resulting in the potential for substantial underestimation or overestimation of NSAID adverse effects. Furthermore, differences in patient populations, drugs
82
C H A P T E R
TABLE 5–4
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
NSAIDs Commonly Used in Children Maximum Dose (mg/day)
Doses per day
Anti-inflammatory dose: 80–100 (<25 kg) 2500 mg/m2 (>25 kg) Antiplatelet dose: 5
4900
2–4
Kawasaki disease: high dose for initial and low dose for subsequent treatment Therapeutic serum levels (for anti-inflammatory therapy): 15–25 mg/dL (measure 5 days after initiation of therapy or alteration of dose) Nonlinear (zero-order) kinetics (see text) LFT abnormalities common (stop ASA is LFT more than three times normal) Association with Reye’s syndrome (see text) Watch for salicylism
Naproxen*
10–20
1000
2
Ibuprofen*
30–40
2400
3–4
Ketoprofen Fenoprofen
2–4 35
300 3200
3–4 4
Most frequently used initial NSAID Overall favorable toxicity/efficacy profile Pseudoporphyria in fair-skinned children (see text) May have nonlinear pharmacokinetics at higher doses Most favorable toxicity/efficacy profile Association with aseptic meningitis in patients with SLE Least favorable toxicity/efficacy profile Significant risk of nephrotoxicity
Drug
Dose (mg/kg/day)
Comments
Salicylates Acetylsalicylic acid (ASA)
Propionic Acid Group
Acetic Acid Derivatives Indomethacin*
1.5–3.0
200
3
Tolmetin
20–30
1800
Sundilac
4–6
400
2
Diclofenac
2–3
150
3
Meloxicam
0.25
15
1
Piroxicam
0.2–0.3
20
1
Nabumetone
30
2000
1
3–4
Useful in spondyloarthropathies and treatment of fever or pericarditis in systemic-onset JRA Less favorable toxicity profile Headache common at initiation and may diminish with continuation of therapy Least favorable toxicity/efficacy profile May cause false-positive result for urinary protein Absorbed as a prodrug and converted to active metabolite Significant enterohepatic recirculation May be less nephrotoxic than other NSAIDs Similar potency to indomethacin Reports of significant hepatotoxicity
Oxicams Once-daily dosing possible Relatively new agent Least favorable toxicity/efficacy profile Once-daily dosing possible—may be useful in older children or adolescents with poor medication compliance; little experience in young children Once-daily dosing possible Tablets can be mixed in water to create a slurry
JRA, juvenile rheumatoid arthritis; LFT, liver function tests; NSAIDs, nonsteroidal anti-inflammatory drugs; SLE, systemic lupus erythematosus. *Available in liquid form.
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
83
TABLE 5–5 Relative Toxicities of NSAIDs Toxicity
ASA
Ibuprofen
Fenoprofen
Naproxen
Indomethacin
Sulindac
Tolmetin
Meclofenamate
GI irritation Peptic ulcer CNS Tinnitus Hepatitis Asthma Renal function Bone marrow
+++ ++ + +++ ++ ++ + −
+ + ± + + + + +
++ + ++ +++ + + + +
++ ++ + + + + + +
++++ +++ ++++ + + + ++ +
++ + + + + + ± +
++ + + + ? + ++ +
++ + ± ± + + + +
ASA, acetylsalicylic acid; CNS, central nervous system; GI, gastrointestinal; NSAIDs, nonsteroidal anti-inflammatory drugs.
used, dosages, and periods of exposure add to the variability in estimates of prevalence. Data from a randomized, placebo-controlled study of the efficacy of a cytoprotective agent, misoprostol, in 8843 patients with rheumatoid arthritis treated with NSAIDs determined that the incidence of definite serious GI complications was 0.95% in the placebo group over the 6-month trial period.58 Possible risk factors for GI complications during NSAID therapy include advanced age, past history of GI bleeding or peptic ulcer disease, and cardiovascular disease.58 However, most patients who have a serious GI complication requiring hospitalization have not had prior GI side effects.53,54 Additional risk factors may include longer disease duration, higher NSAID dose, the use of more than one NSAID, longer duration of NSAID therapy, concomitant glucocorticoid or anticoagulant use, and serious underlying systemic disorders.53,54 Many of the studies on which these conclusions are based do not account for the interaction of multiple factors or confounding by coexisting conditions.53 It appears that infection with Helicobacter pylori increases the risk of gastroduodenal mucosal injury associated with NSAID use only minimally, if at all.59 The magnitude of this problem in children is poorly documented but has traditionally been thought to be considerably less than in adults, partly because of the absence of the associated risk factors identified in the adult population. H. pylori has not been reported to be an important pathogen in children with JRA treated with NSAIDs.60 Studies in children confirm that, although mild GI disturbances are frequently associated with NSAID therapy, the number of children who develop clinically significant gastropathy appears to be low.44 In many children who develop GI symptoms while receiving NSAIDs, alternative causes, such as the underlying disease process, psychosocial factors, and other concomitant medications, could account for their symptoms. The rigor with which these factors have been systematically evaluated and the definition of “clinically significant” gastropathy have varied considerably among studies, resulting in variability in reported rates for this complication. A retrospective study of a cohort of 702 children receiving NSAID therapy for JRA who were monitored for at least 1 year found 5 children (0.7%) with clinically sig-
nificant gastropathy defined as esophagitis, gastritis, or peptic ulcer disease.61 The retrospective nature of this study may have resulted in a substantial underestimation of the prevalence of NSAID-associated gastropathy. A prospective study of a cohort of 203 children found that, although 135 children (66.5%) had documented GI symptoms at some stage during NSAID therapy, only 9 (4.4%) had endoscopically detected ulcers or erosions; the most commonly reported GI symptoms were abdominal pain (49.7%) and appetite loss (32.0%).62 A more recent prospective study reported on 45 children who underwent routine endoscopy (in association with general anesthesia for joint injections). Nineteen patients, or 42%, had normal gastric and duodenal mucosa, and 20 had histologically mild gastritis. A clear association was seen between abdominal pain and gastroduodenal pathology, but the severity of gastric inflammation did not correlate with the duration of NSAID therapy.63 Factors frequently associated with clinically significant gastropathy in children are abdominal pain at night, melanotic stools, and a previous history of gastropathy.61 Endoscopic studies in very small numbers of highly selected groups of children with JRA receiving NSAIDs have reported a higher frequency of abnormalities.64,65 However, these endoscopic lesions, which are usually mild, have not been found to correlate well with symptoms, and their clinical significance is not clear.64,65 Further prospective studies are needed. A number of studies have shown differences in rates of serious GI complications associated with different NSAIDs. Systematic reviews have found ibuprofen to be associated with the lowest risk; indomethacin, naproxen, sulindac, and aspirin with moderate risk; and tolmetin, ketoprofen, and piroxicam with the highest risk.56,66 A recent report comparing rofecoxib (0.3 to 0.6 mg/kg/day, maximum 25 mg/day) with naproxen (7.5 mg/kg twice daily) showed equivalent efficacy but significantly fewer GI adverse events with rofecoxib.6 In adults, endoscopic studies have shown that COX-2 inhibitors induce significantly fewer ulcers than traditional NSAIDs do.67 Studies in children have also found that tolmetin may be associated with higher risk.61,62 A reasonable approach to therapy is to choose initially an NSAID with a lower risk of GI complications. GI symptoms can be further minimized by ensuring that NSAIDs are always given with food.
84
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Although some studies in healthy volunteers suggest that enteric coating may reduce acute gastric mucosal injury,68 the use of enteric-coated preparations and parenteral or rectal administration of NSAIDs aimed at preventing topical mucosal injury have not been shown to prevent gastric ulceration.53 The development of “safer” NSAIDs, such as the highly selective COX-2 inhibitors, offers considerable promise, but the clinical utility of these agents in children remains to be determined. Two large studies assessed the gastroprotective effects of celecoxib and rofecoxib. In the Celecoxib Long-term Arthritis Safety Study (CLASS),69 celecoxib was compared with diclofenac or ibuprofen in 8059 patients with rheumatoid arthritis or osteoarthritis; only at higher than clinically indicated doses did celecoxib demonstrate a lower incidence of GI-related adverse events. In the Vioxx Gastrointestinal Outcomes Research (VIGOR) study, rofecoxib (50 mg/day) was compared with naproxen (500 mg twice daily) in 8076 patients with rheumatoid arthritis.70 Significantly fewer clinically important GI adverse events occurred in the rofecoxib group. However, given the relative infrequency of clinically significant GI adverse events in patients with JRA, COX-2 inhibitors should not be the anti-inflammatory agent of first choice. Their role in the treatment of JRA remains to be determined. The utility of antacids and histamine 2 (H2)-receptor antagonists for prophylaxis against serious NSAIDinduced GI complications is controversial. Although these medications suppress symptoms, they do not prevent significant GI events such as endoscopically documented gastric ulcers. In fact, asymptomatic patients on acid-reduction therapies appear to be at greater risk for serious GI complications than are patients not taking these medications, so their routine use in asymptomatic patients taking NSAIDs cannot be recommended.53,54 Sucralfate also does not appear to offer any significant benefit in the prophylaxis of NSAID-induced gastric ulcers.71 Misoprostol, a synthetic prostaglandin E1 analogue, has been shown in adults to be effective in both prophylaxis58,72 and treatment of NSAID-induced gastroduodenal damage, allowing continuation of NSAID therapy while achieving ulcer healing.73,74 Studies of misoprostol cotherapy in children are limited but also suggest that it may be effective in the treatment of GI toxicity symptoms in children receiving NSAIDs.64,75 Misoprostol may also be associated with a protective effect on hemoglobin values in NSAID-treated patients.76,77 Omeprazole, a proton pump inhibitor, has been shown to be superior to ranitidine and misoprostol for the prevention and treatment of NSAID-related gastroduodenal ulcers in adults.77,78 Prospective studies are needed to further evaluate the role of misoprostol and omeprazole cotherapy in children. Recommendations for the treatment of established dyspeptic symptoms or active gastroduodenal ulceration, with or without continuation of NSAIDs, differ from those for the prophylaxis of gastroduodenal injury. Symptoms of active gastroduodenal ulceration after discontinuation of NSAIDs can be treated empirically with an H2-receptor antagonist or a proton pump inhibitor. If an ulcer develops,
discontinuation of the NSAID is preferred, but if NSAID therapy needs to be continued, proton pump inhibitors are recommended.53
Hepatotoxicity Hepatitis with elevation of transaminase levels can occur with any NSAID but has most commonly been reported in children with JRA receiving ASA; up to 50% of these children may have some elevation of enzyme levels,79 and up to 15% may require discontinuation of therapy for this reason.42 In one retrospective study, transaminase levels were increased in 6% of children receiving naproxen.42 One of the confounding factors is that transaminases can be elevated in untreated JRA, particularly in the systemic-onset subtype, and in systemic lupus erythematosus (SLE). Elevated transaminase levels are rarely of clinical significance and often resolve spontaneously, but they may necessitate lowering of the dose or temporary cessation of therapy. Rarely, hepatotoxicity is severe; NSAIDs have been associated with a poorly understood severe multisystem disorder, now called the macrophage activation syndrome, which consists of macrophage activation, hepatic involvement, consumptive coagulopathy, and neurologic manifestations.80,81 Therefore, liver function should be carefully monitored in children taking NSAIDs, particularly those with systemic-onset JRA.
Renal Toxicity Several types of renal complications have been associated with NSAID therapy. These include reversible renal insufficiency and acute renal failure; acute interstitial nephritis; nephrotic syndrome; papillary necrosis; and sodium, potassium, and water retention.82–85 Although these complications are more often reported in the adult population, with an estimated prevalence of 1% to 2%, a number of cases have been described in children.82,86–89 The limited data on the prevalence of these complications in children indicate that it is considerably lower than that reported in adults. A 4-year prospective study of 226 children with JRA treated with NSAIDs found the prevalence of renal and urinary abnormalities attributable to NSAID therapy to be only 0.4%90; an even lower prevalence of 0.2% was reported in another cohort of 433 children.91 Several studies demonstrated subclinical abnormalities of renal glomerular or tubular function in children with JRA receiving NSAIDs by measuring the urinary excretion of selected glomerular proteins (albumin, transferrin, immunoglobulin G [IgG]) and tubular proteins (α1- and β2-microglobulin) or enzyme markers (N-acetyl-βglucosaminidase).92,93 However, the clinical relevance of these abnormalities as potential markers for the development of more serious renal complications is not yet clear, because no long-term data are available to correlate these abnormalities with important clinical outcomes. The various renal syndromes associated with NSAID therapy differ in their pathophysiologic basis, clinical presentation, frequency, and predisposing risk factors. The most commonly reported complication is reversible
C H A P T E R
renal insufficiency, mediated through the effect of NSAIDs on prostaglandin synthesis. Inhibition of renal prostaglandin synthesis has little effect on renal function in healthy persons. However, in states of hypovolemia or salt depletion, the adrenergic and renin-angiotensin system is activated, resulting in renal vasoconstriction. Prostaglandins are needed to maintain renal perfusion under such conditions by producing local vasodilatation. Inhibition of prostaglandin synthesis with NSAID therapy may suppress this protective autoregulatory mechanism, resulting in unopposed vasoconstrictor activity and renal hypoperfusion.83,84 This type of renal insufficiency is usually reversible within 24 to 72 hours.82 Risk factors include underlying renal disease (e.g., SLE) or states of hypovolemia and salt depletion with high plasma renin activity (e.g., gastroenteritis, sepsis, congestive cardiac failure, cirrhosis, diuretic treatment) in normal persons. Indomethacin is the NSAID most often implicated with this complication.83,94 In addition to impairment of renal function, some children show signs of fluid retention, such as congestive heart failure, edema, or hypertension, when treated with NSAIDs. A modest drop in hematocrit value with ASA or other NSAIDs may result from mild degrees of sodium retention and hemodilution rather than from anemia related to GI bleeding.41 Acute interstitial nephritis with nephrotic syndrome is far less commonly reported with NSAIDs. It occurs sporadically and is thought to represent a hypersensitivitytype reaction.84 It typically has an abrupt onset with hematuria, heavy proteinuria, and flank pain. In contrast to classic drug-induced allergic nephritis, however, fever, rash, eosinophilia, and eosinophiluria usually are not present. Onset may be from 2 weeks to 18 months after initiation of NSAID therapy, and resolution may take from 1 month to almost 1 year after discontinuation of the NSAID.84 Renal failure may be severe enough to require temporary dialysis support. Glucocorticoids have been used to treat this complication, but their efficacy is unproven. Interstitial nephritis with and without nephrotic syndrome has also been described in a small number of children.41 This complication is more commonly reported with propionic acid (naproxen, fenoprofen) and acetic acid (tolmetin, indomethacin, sulindac) derivatives.84,85 Papillary necrosis is a chronic renal injury that has been most commonly associated with long-term analgesic abuse, particularly with phenacetin.84,95 It has also been reported to occur with several NSAIDs, including ibuprofen, fenoprofen, phenylbutazone, and mefenamic acid. Medullary ischemia is thought to be the initiating factor in the production of papillary necrosis.84 The syndrome is usually characterized by painless leukocyturia and hematuria, without impairment of renal function, and by demonstration of changes on the intravenous pyelogram.84,87,88,96–100 Combination NSAID therapy appears to be a risk factor in both adults and children.82,100
Central Nervous System Effects Three general categories of CNS side effects have been reported in association with NSAID therapy in adults:
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
85
aseptic meningitis, psychosis, and cognitive dysfunction.101 The NSAID most commonly reported to cause aseptic meningitis has been ibuprofen; susceptibility appears to be greater in patients with SLE. However, there does not seem to be any cross-reactivity between NSAIDs for this complication. Indomethacin and sulindac have been reported to induce psychotic symptoms, including paranoid delusions, depersonalization, and hallucinations, in a small number of patients.101 More subtle CNS effects, such as cognitive dysfunction and depression, can also occur and are probably under-recognized and under-reported. Tinnitus may occur with any NSAID, but particularly with ASA.41 A prospective study of 203 children with JRA found that CNS symptoms occurred in 55% of patients taking NSAIDs; the most common symptom was headache, which occurred in about one third of children.102 Other reported symptoms included fatigue, sleep disturbance, and hyperactivity. Seizures were noted in two patients, both of whom were taking indomethacin.
Cutaneous Toxicity A diverse group of skin reactions, including pruritus, urticaria, morbilliform rashes, erythema multiforme, and phototoxic reactions, have been described.41,103 Initially described in Australia,104 the syndrome of pseudoporphyria occurring in association with naproxen therapy in children with JRA has now been reported in several case series.104–109 It is a distinctive photodermatitis marked by erythema, vesiculation, and increased skin fragility characterized by easy scarring of sun-exposed skin (Fig. 5–4). Porphyrin metabolism is normal. All findings except scarring resolve with discontinuation of naproxen, but
■ Figure 5–4 Distant and close-up views of the face of an 8-year-old boy with pseudoporphyria who was taking naproxen. Note a blistered lesion adjacent to a superficial scar. Superficial scars are also visible on the nose.
86
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
the vesiculation may persist for several months.110 Children with fair skin and blue eyes are particularly susceptible; one study reported a relative risk of 2.96 if the child had blue-gray eyes and was taking naproxen.109 Pseudoporphyria appears to be a common side effect even in geographic areas without high sun exposure; a 6-month prospective study of children seen in a rheumatology clinic in Halifax, Nova Scotia, reported a prevalence of 12% among those treated with naproxen107; similarly, there was a 10.9% incidence among NSAIDtreated children with juvenile idiopathic arthritis (JIA) in Edinburgh.109 Although this complication has most often been reported with naproxen, many other NSAIDs have also been implicated.
Effects on Coagulation The NSAIDs decrease platelet adhesiveness by interfering with platelet prostaglandin synthesis, such as that of thromboxane B2, which promotes platelet aggregation. This inhibition is reversible in the case of all NSAIDs except ASA, which irreversibly acetylates and inactivates COX (see Fig. 5–3), an effect that persists for the life of the platelet; bleeding time returns to normal only as new platelets are released into the circulation.41 The NSAIDs also displace anticoagulants from protein-binding sites, thereby potentiating their pharmacologic effect. These effects of NSAIDs must be considered when any surgical procedure is planned.
Reye’s Syndrome Reye’s syndrome was first described in Australia in 1963 as a distinct clinicopathologic entity by Reye and associates.111 An association with salicylate therapy was later demonstrated in the early 1980s.112 Reye’s syndrome is an acute illness characterized by encephalopathy and fatty degeneration of the liver that occurs almost exclusively in children and usually in association with a viral-like prodromal illness, the most common being influenza or other respiratory illnesses, varicella, or gastroenteritis.113 The onset of the illness is characterized by profuse vomiting and varying degrees of neurologic impairment. Initially, there may be fluctuating personality changes and deterioration in consciousness, followed by extreme irritability, agitation, confusion, delirium, and coma as the encephalopathy progresses. Associated metabolic abnormalities include hyperammonemia and increased levels of hepatic transaminases, with consequences of hepatic dysfunction such as prolongation of the prothrombin time and severe hypoglycemia. Characteristic liver histopathologic abnormalities include microvesicular fat accumulation in hepatocytes on light microscopy and mitochondrial disorganization and proliferation of smooth endoplasmic reticulum on electron miscroscopy.113 The illness can be fatal, with an overall case-fatality rate of 31%; rates are significantly higher in children younger than 5 years of age and in those with a serum ammonia level greater than 45 μg/dL.113 Reye’s syndrome appears to be a heterogeneous condition; in recent years, a proportion of children presenting with Reye-like illnesses have been found to have various inborn metabolic
disorders. This subgroup is characterized by features such as younger age, recurrent episodes, family history of similar illness, frequent hypoglycemia, cardiac enlargement, muscle weakness, and lack of mitochondrial disorganization in liver tissue.113 The association between Reye’s syndrome and salicylate therapy was first reported in a small case-control study.114 Although this study initially was a source of controversy, its findings were later confirmed by larger and more rigorously designed studies.112 The association between salicylate use and Reye’s syndrome is consistent and is very strong, with odds ratios as high as 35 to 40 in the larger studies.115,116 Although demonstration of an epidemiologic association alone is not sufficient to prove causation, there is further evidence in support of an etiologic role for salicylates in Reye’s syndrome: The finding of a dose-response relationship, with higher doses of salicylate used in case subjects than in controls, and more recent epidemiologic evidence from the United States and United Kingdom indicate that there has been a dramatic decline in the incidence of Reye’s syndrome concurrent with a decline in the use of salicylates in children since public health warnings were issued in the mid1980s.113,117 In a large epidemiologic study in the United States, 14 of 361 patients with Reye’s syndrome for whom data on aspirin use were available had taken salicylate-containing medications for illnesses such as JRA and Kawasaki disease.113 Other studies have reported higher rates of Reye’s syndrome in children taking salicylates for JRA.118,119 None of the studies to date has reported any association with other NSAIDs.112 Physicians and others caring for children requiring long-term salicylate therapy should be aware of the risk of Reye’s syndrome and be able to recognize the early symptoms so that salicylate therapy can be promptly withdrawn. These children should be offered varicella vaccine and annual vaccination against influenza in accordance with the recommendations of the Advisory Committee on Immunization Practices.120
Hypersensitivity and Miscellaneous Effects The precipitation of asthma or anaphylaxis with NSAIDs has been reported in adults as a unique syndrome associated with nasal polyps; 15% to 40% of patients with nasal polyps may experience bronchospasm when given aspirin.121,122 Although this syndrome can theoretically be provoked by any NSAID, it has most commonly been reported with ASA or tolmetin; cross-reactivity between NSAIDs may occur.41 However, there appears to be a correlation with the strength of COX inhibition: More potent NSAIDs such as indomethacin induce bronchospasm at smaller doses than do less potent ones such as ibuprofen.123 Although “allergy” to ASA is often reported by patients or parents, true hypersensitivity to the drug is exceedingly rare in childhood. ASA hypersensitivity occurs in about 0.3% to 0.9% of the general population, in 20% of patients with chronic urticaria, and in 3% to 4% of patients with chronic asthma and nasal polyps.124–126 It should be noted that such patients are often hypersensitive to the other NSAIDs as well.127
C H A P T E R
Effects on the bone marrow, including aplastic anemia, agranulocytosis, leukopenia, and thrombocytopenia, have been reported but are rare.41 Mild anemia occurs in about 2% to 14% of children42 and may be partly due to hemodilution, hemolysis,128 or occult GI blood loss due to NSAID therapy.75
Salicylates The salicylates, a group of related drugs that differ by the nature of the substitutions on the carboxyl or hydroxyl groups of the molecule, include ASA (ASA), salsalate, choline salicylate, magnesium salicylate, and sodium salicylate. They are hydrolyzed in vivo to salicylic acid. Related compounds such as salicylamide and diflunisal are not hydrolyzed to salicylic acid and therefore are not considered to be salicylates.129 ASA is the oldest NSAID and has been used to treat the articular manifestations of rheumatic diseases for many years. Although newer NSAIDs have largely replaced ASA as the mainstay of anti-inflammatory drug therapy in pediatric rheumatology, ASA continues to have a role in the management of Kawasaki disease (see Table 5–4) and in the treatment of patients who are predisposed to thromboses. The general principles of NSAID mechanism of action and pharmacology, as well as the principles of therapy and the spectrum of known adverse effects, have already been addressed with reference to salicylates where relevant. However, the salicylates differ from other NSAIDs in a number of specific aspects; these are addressed in the following paragraphs.
Mechanism of Action ASA exerts its anti-inflammatory, analgesic, and antipyretic effects in part by irreversibly acetylating and inactivating COX, thus inhibiting biosynthesis and release of the prostaglandins (see Fig. 5–3; see Chapter 3).120,130,131 Other salicylates also inactivate this enzyme, but not irreversibly. A dose-dependent effect of ASA on the inhibition of prostacyclin (prostaglandin I2) production by endothelial cells and of thromboxane A2 production by platelets has also been noted and may be relevant in the management of Kawasaki disease or other types of vasculitis in children (see Chapter 24).132,133 Large doses of ASA increase urinary excretion and lower the serum concentration of urate; low doses have the opposite effect.134 This phenomenon must not be confused with hyperuricemia or with gout, which is extremely rare in children.
Pharmacology The plasma level of salicylate (ASA and salicylate ion) peaks 1 to 2 hours after a single dose, and the drug is virtually undetectable at 6 hours. ASA itself is bound very little to plasma protein, but salicylic acid binds extensively to albumin and erythrocytes. Salicylic acid is found in most body fluids (including the cerebrospinal fluid, saliva, synovial fluid, and breast milk), and it crosses the placenta. ASA is metabolized by hepatic microsomal enzymes by conjugation with glycine to form salicyluric acid and, to a lesser extent, by conjugation with the phenolic and acyl glucuronides, which are in turn excreted by the kidneys. Renal clearance of these metabolites, which have longer half-lives than the parent drug, is augmented by alkalinization of the urine.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
87
Administration ASA is quickly absorbed from the stomach and proximal small intestine.135,136 Some salicylates (methyl salicylate, salicylic acid) are well absorbed through the skin. The systemic anti-inflammatory effects of ASA are maximal, and in most cases they are achieved only if serum steady-state levels are 15 to 25 mg/dL (1.09 to 1.81 mmol/L).137,138 With levels less than 15 mg/dL, ASA does not function effectively as an anti-inflammatory agent; at greater than 30 mg/dL (2.17 mmol/L), it is likely to be toxic. The dosage necessary to reach these concentrations is usually 75 to 90 mg/kg/day, divided into four doses given with food. Lower doses suffice in the child weighing more than 25 kg. The plasma half-life of salicylate increases as the plasma concentration of the drug increases (capacity-limited kinetics). For example, at a plasma concentration of 26 mg/dL, the half-life of ASA can be as long as 16 hours, whereas at a plasma level of 4 mg/dL, it may be as short as 2.5 hours.33 Consequently, the drug need not be given as frequently once plasma concentrations are high, and toxic concentrations of the drug take much longer to decrease than would otherwise be anticipated. Therapeutic levels are not reliably attained before 2 to 5 days of administration. Serum salicylate and serum liver enzyme levels should be checked 5 days after initiation of therapy or after any dose adjustment. Although it is the author’s practice to monitor salicylate concentrations in serum taken 2 hours after the morning dose, one study indicated that the timing of blood sampling was not critical if the interval between doses was 8 hours or less and a steady state had been achieved after 5 days of therapy.139
Toxicity: Salicylism Salicylism (acute or chronic salicylate intoxication) can occur rapidly in the young child, and early signs such as drowsiness, irritability, or hyperpnea can easily be overlooked. In the very young child, metabolic acidosis and ketosis occur, whereas the older child may first experience respiratory alkalosis by direct action of ASA on the hypothalamus. Abdominal pain or vomiting may occur in some children. The child with fever and dehydration is prone to salicylism: In the child with intercurrent illness and nausea, vomiting, or diarrhea, the drug should be immediately discontinued. Symptoms of salicylism include tinnitus, deafness, nausea, and vomiting (Table 5–6). Early on, there is CNS stimulation (hyperkinetic agitation, excitement, maniacal behavior, slurred speech, disorientation, delirium, convulsions). Later, CNS depression (stupor and coma) supervenes. Because the young child, in whom there is a narrow margin between therapeutic and toxic levels,140,141 may not effectively communicate symptoms of salicylism, the family must be thoroughly schooled in the signs of overdose.
TABLE 5–6
Toxicity of Salicylates
Symptom
Frequency
Gastric irritation Tinnitus and/or diminished hearing Hepatotoxicity Reye’s syndrome Hypersensitivity (in asthma) Salicylism Mild: Lethargy, dizziness, headache, diaphoresis, nausea Moderate: Confusion, hyperpnea, metabolic acidosis, respiratory alkalosis Severe: Hyperpyrexia, convulsions, cardiovascular collapse
50% 15% 2% Rare Rare
88
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Mild salicylate toxicity requires no treatment and often only a minor decrease in salicylate dose. If the child has persisting symptoms, evidence of CNS stimulation, or depression, the drug must be discontinued. The child should be monitored for evidence of acute salicylate toxicity: fever, acute renal failure, CNS depression, pulmonary edema, bleeding, and hypoglycemia. In situations of severe chronic salicylism or acute overdose, the stomach contents should be emptied, and activated charcoal (0.5 to 1.0 g/kg) should be administered. Urine output, body temperature, serum electrolytes, and glucose should be monitored, and glucose-containing intravenous fluids should be given as required. Alkaline diuresis induced with sodium bicarbonate and furosemide increases the rate of salicylate excretion in the urine but must be carefully titrated in the presence of rapidly changing metabolic or respiratory function. Peritoneal dialysis or hemodialysis may be necessary. The reader is referred to the recommendations of Mofenson and Caraccio142 for details of the management of severe salicylate poisoning.
Contraindications ASA leads to hemolysis in children who have the enzyme defect glucose-6-phosphate dehydrogenase deficiency or pyruvate kinase deficiency. It should be avoided in children who have bleeding disorders, such as hemophilia or von Willebrand’s disease, and in patients receiving thrombolytic agents or anticoagulants. ASA should not be given during the last trimester of pregnancy because of its effects on coagulation and platelet function.
Drug Interactions ASA should be used with caution in children taking certain other drugs. Levels of MTX,38 valproic acid,143 phenytoin,144 and other NSAIDs (tolmetin, diclofenac)139,145 may be increased in children who are also receiving aspirin. ASA decreases the bioavailability of other NSAIDs by 20% to 50%142 and increases the digitalis concentration by 30%,144 although the clinical significance of this interaction is uncertain. Glucocorticoids increase the rate of excretion of ASA,146 and salicylism may occur if glucocorticoid drugs are abruptly discontinued in a child taking therapeutic amounts of ASA.
Disease-Modifying Antirheumatic Drugs A number of drugs used to treat JRA and certain other rheumatic diseases do not produce an immediate analgesic or anti-inflammatory effect but exert their beneficial effects weeks to months after initiation of therapy. These compounds, called disease-modifying antirheumatic drugs (DMARDs) or slow-acting antirheumatic drugs (SAARDs), currently include MTX, the antimalarials, sulfasalazine, leflunomide, gold compounds, and D-penicillamine. Historically, there was reluctance to start DMARD treatment until relatively late in the course of disease. Standard practice had been to use one, two, or even three NSAIDs sequentially (or occasionally even in combination) before starting DMARD therapy. Much of this delay had to do with the potentially serious toxicities of DMARDs (principally gold and D-penicillamine), the fact that JRA was not a fatal disease and therefore thought not to warrant treatment with drugs of such potentially significant toxicities, and a belief that most children would eventually “outgrow” the disease. However, there has
been a philosophical shift in therapeutic approach, based on an appreciation that irreversible damage occurs early; that active disease, leading to deleterious long-term effects, persists for many years147; and that MTX, the most frequently prescribed drug in this class, is both safe and effective.148–151 Therefore, there has been a tendency to pursue a more aggressive approach than in the past and to start DMARDs early in the course of the disease to prevent irreversible joint damage, rather than wait for significant damage to declare itself. Furthermore, experience with combination therapy in adult rheumatoid arthritis has led to use of a variety of combinations in JRA as well. MTX is the most commonly used DMARD for JRA. Gold and D-penicillamine now are rarely used for JRA. Several DMARDs are also used to treat other rheumatic diseases (e.g., hydroxychloroquine in the management of SLE and, occasionally, juvenile dermatomyositis [JDM]; sulfasalazine in treatment of the spondyloarthropathies). In the treatment of arthritis, DMARDs are usually given in addition to an NSAID. The goal of treatment is to achieve disease control with drugs from this class and then to stop other treatments (e.g., NSAIDs, prednisone).
Methotrexate Low-dose weekly MTX has emerged as one of the most useful agents in the therapeutic armamentarium for the management of rheumatic diseases in children. It is one among only a small number of agents that have been demonstrated to be efficacious in a randomized controlled trial in children; this drug is now often the firstchoice second-line agent in childhood arthritis. Although it has been most extensively studied in adult rheumatoid arthritis and in children with JRA, its use is also growing in many other chronic inflammatory disorders.152
Mechanism of Action MTX (Fig. 5–5) is a folic acid analogue and a potent competitive inhibitor of dihydrofolate reductase (DHFR) (Fig. 5–6). It may also inhibit thymidylate synthase and interfere with the metabolic transfer of single carbon units in methylation reactions, especially those involved in synthesis of thymidylate and purine deoxynucleosides, which are essential components of DNA.153 It also interferes with de novo purine biosynthesis by inhibition of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase, an enzyme in the purine biosynthetic pathway. MTX-induced inhibition of AICAR transformylase N
N
H2N
O N
N
CH2
NH2
N
C
CH3
NH CHCOOH CH2 CH2 COOH
■ Figure 5–5
Structure of methotrexate.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Intracellular Purine synthesis (guanine, adenine)
D-Ribose 5P (pentose cycle)
A
A
10 - Formyl- THF THF GAR
Guanine
FGAR
10 - FormylTHF
THF
AICAR
MTX
89
Extracellular
Cell membrane
C H A P T E R
FAICAR
IMP
AMP AMP deaminase
MTX
5’–nucleo– tidase
B
C Inosine
CD73 ⫽ ecto– 5’–nucleotidase
Adenosine
ADA (Adenosine deaminase)
Uric acid
AMP
C
D
Adenosine
Adenine GTP
Immunosuppression
A2a A2b
G␣? cAMP
G
F
G
G␥
cAMP
E A1
GTP G␣? G
G␥
■ Figure 5–6 Steps in methotrexate intracellular metabolism and possible sites of action. AICAR, 5-aminoimidazole-4-carboxamide ribonucleotide; dUMP, deoxyuridylate monophosphate; MTX, methotrexate; TMP, thymidylate monophosphate. (From Cutolo M, Sulli A, Pizzorni C, Seriolo B. Anti-inflammatory mechanisms of methotrexate in rheumatoid arthritis. Ann Rheum Dis 60: 729–735, 2001.)
and secondary inhibition of adenosine deaminase leads to accumulation and enhanced release of adenosine, a potent inhibitor of neutrophil adherence.154,155 It is believed that the effects of MTX are in fact mediated primarily through the anti-inflammatory action of adenosine acting through a variety of receptors.156 The mechanism of action of MTX in JRA or adult rheumatoid arthritis is not clearly elucidated, although MTX is known to act at a number of intracellular levels. In addition to its action as an antimetabolite, it is an antiinflammatory and immunomodulatory agent. MTX modulates the function of many of the cells involved in inflammation and affects the production of a variety of cytokines, including reducing the production of TNF-α, interferon-γ (IFN-γ), IL-1, IL-6, and IL-8, thus acting as a potent inhibitor of cell-mediated immunity.153,156 By reducing the endothelial cell production of adhesion molecules, MTX may reduce the permeability of the vascular endothelium. In addition, adenosine inhibits adherence of stimulated neutrophils to endothelial cells, thereby protecting the vascular endothelium from neutrophil-induced damage.153,157 It may also have more direct effects in inflamed joints by inhibiting the proliferation of synovial cells and synovial collagenase gene
expression.158 Although the exact mechanism of action is the subject of intense study, it is not yet possible to clearly distinguish which of the many cellular and immune effects demonstrated in vitro and in vivo are of particular relevance to MTX’s clinical effects. The reader is referred to reviews that explore this issue in greater detail.153,157 The widespread biologic effects of MTX may in fact account for its observed efficacy in a wide variety of diseases with disparate pathogeneses, such as cancer, psoriasis, and the various rheumatic and other chronic inflammatory diseases.
Pharmacology There is significant intraindividual and interindividual variability in the absorption and pharmacokinetics of MTX after oral administration.159 Although the average oral bioavailability is about 0.70 (compared with intravenous dosing), the range can be quite wide (0.25 to 1.49), with 25% of subjects in one study absorbing less than half their dose.160 Factors such as age-related differences, the influence of food, and the effects of concurrently administered medications contribute to this variability. The effect of food on the oral bioavailability
90
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Serum concentration, molar
of MTX has been controversial.161 However, a pharmacokinetic study demonstrated that factors such as age, body weight, creatinine clearance, sex, dose, and fed versus fasted state significantly influenced the disposition of MTX in adults with rheumatoid arthritis.162 The bioavailability of MTX has also been demonstrated to be greater in the fasting state in children with JRA.163 The author currently recommends that MTX be given on an empty stomach with water or clear citrus beverages. In general, oral bioavailability is about 15% less than after intramuscular administration. Bioavailability of intramuscular and subcutaneous administration is similar,164 with the latter being generally more acceptable for children who require parenteral MTX. After a single dose of MTX, the drug is present in the circulation for a relatively short period before it is redistributed to the tissues (Fig. 5–7). Peak serum levels are reached in approximately 1.5 hours (range, 0.25 to 6 hours), with elimination half-life being approximately 7 hours in subjects with normal renal function.165 Circulating levels fall rapidly as the drug is distributed into tissue and eliminated. The predominant route of elimination is renal, with more than 80% of the drug eliminated unchanged via glomerular filtration and tubular secretion within 8 to 48 hours. A smaller but significant route of elimination is the biliary tract. The pharmacokinetics of MTX are triphasic. The initial rapid phase represents tissue distribution and renal clearance; the second phase is prolonged because of slow release from tissues, tubular reabsorption, and enterohepatic
10⫺6
Cmax Toxicity likely
MTX 10⫺7
7-OH-MTX
10⫺8
10⫺9
Tmax
% excreted in urine
12
24
36
48
MTX 100 75 50 25
7-OH-MTX 12
24
36
48
recirculation; the third phase is flat between weekly doses, reflecting the gradual release of tissue MTX.165 Most MTX is delivered to cells as the parent compound and enters cells via active transport by reduced folate carriers. MTX competes with leucovorin for these carriers. Folic acid enters cells through a different set of receptors, the folate receptors. Their expression may be upregulated in inflamed synovium, allowing for more efficient transport of MTX into cells. These receptors are expressed variably, most likely under genetic control, and the interindividual variability may explain some of the differences in efficacy and toxicity. Similarly, removal of MTX from cells occurs via multidrug resistance–associated proteins, which may be enhanced in some patients and could again explain some interindividual variability in MTX response. Between 3% and 11% of the parent drug is hydroxylated in the liver to 7-hydroxymethotrexate (7-OH-MTX). A portion of intracellular MTX and 7-OH-MTX is metabolized to MTX polyglutamates by the enzyme folyl-polyglutamyl synthetase. This enzyme also polyglutamates folic acid and folinic acid, which at high levels may overwhelm this enzyme system, preventing MTX polyglutamation and allowing it to efflux from cells.153 MTX polyglutamates are long-lived derivatives that retain biochemical and biologic activity within the cell.165 These intracellular, polyglutamated MTX derivatives may in fact be the active anti-inflammatory agents,156 and levels of polyglutamates in red blood cells have been shown to correlate with efficacy in patients with rheumatoid arthritis.166 Plasma drug levels do not correlate well with clinical effects and therefore are not useful in routine monitoring of MTX therapy.159 Measurement of MTX levels in saliva also has not been helpful.165,167 The pharmacokinetics of oral MTX in JRA appear to be age dependent, with more extensive metabolism of MTX in younger children.168 This difference may account for the observation that children require higher doses of MTX than adults do to obtain similar therapeutic effects.169,170 At low doses, MTX is only moderately protein bound (11% to 57%), so the potential for interactions with other protein-bound drugs is small and usually is not clinically significant.165 Studies in adults have not shown a consistent or clinically important interaction between MTX and NSAIDs. However, several studies in children have demonstrated an interaction between MTX and NSAIDs that may be clinically significant in individual patients, particularly those with any renal dysfunction.38,171 It is thought that this interaction may be mediated through competition for protein-binding sites and through alteration of renal clearance.152 The combination of MTX and trimethoprimsulfamethoxazole should be avoided, because it may lead to hematologic toxicity through the synergistic effects of these drugs on DHFR. Although the potential for interaction between MTX and various other drugs (e.g., sulfasalazine, glucocorticoids, folate supplementation) exists, current clinical data do not support general avoidance of these combinations (see later discussion).165
Time after ingestion (h)
■ Figure 5–7 Time course of methotrexate (MTX) and 7-hydroxymethotrexate (7-OH-MTX) after an oral dose of 15 mg. (From Hillson JL, Furst DE: Pharmacology and pharmacokinetics of methotrexate in rheumatic disease: practical issues in treatment and design. Rheum Dis Clin North Am 23: 757–778, 1997.)
Efficacy The short- to medium-term efficacy of MTX in children with JRA is now well established.152,172 Benefits in initial
retrospective and uncontrolled studies were subsequently confirmed in a combined USA-USSR placebo-controlled, randomized controlled clinical trial.148 In this 6-month study of 127 children with resistant JRA (mean age, 10.1 years; mean disease duration, 5.1 years), 63% of the group treated with 10 mg/m2 of MTX improved, compared with only 32% of those treated with 5 mg/m2 and 36% of the placebo group. The assessment of efficacy was based on a composite of clinical and laboratory parameters and subjective global assessment by physician and parent but did not include any functional assessments or radiographic examinations. A Cochrane Database Systematic Review, based on only two studies of 165 patients, concluded that MTX had minimal clinically significant effects (greater than 20%) on patient-centered disability in patients with JIA.173 Various investigators have tried to determine whether MTX is more effective in some JRA subgroups than others. A randomized, controlled study from the United Kingdom concluded that MTX, at doses up to 20 mg/m2, produced significant improvement in patients with extended oligoarticular JIA but was much less effective in patients with systemic-onset JIA.174 Ravelli and associates175 studied the effects of MTX in patients with polyarticular, extended oligoarticular, and systemic-onset JIA (20, 23, and 37 subjects, respectively) and determined that the extended oligoarticular subtype was the best predictor of short-term clinical response. These patients also relapsed more quickly on discontinuation of MTX, supporting the conclusion that MTX may be most effective in patients with extended oligoarticular disease. Finally, a study from Saudi Arabia of 18 patients with systemic-onset JIA treated with MTX for a mean of 18 months found that systemic features resolved in all of the 10 patients who had such features at the start of treatment; the active joint count and functional class improved in 16 patients, and there was an overall reduction of the need for corticosteroid treatment.176 However, some children with systemiconset JRA had worsening of their disease with MTX treatment.177
Several subsequent studies suggested that MTX may also slow the radiologic progression of disease in JRA, although the available data are not conclusive.178,179 Some authors suggested that earlier treatment with MTX, possibly before the appearance of radiographic changes, may favorably influence the outcome of MTX treatment in children with systemic-onset disease.180 Similar results were found with early “aggressive” treatment in adults with recent-onset rheumatoid arthritis.181 The longer-term efficacy of MTX in JRA and its impact on function and quality of life need further study. There is also accumulating experience with the use of MTX in many other pediatric rheumatic disorders, including SLE,182 some vasculitides,183,184 sarcoidosis,185 systemic sclerosis,186 localized scleroderma,187 and uveitis.188 However, the evidence for the efficacy of MTX in these conditions is less strong and often is based on open, uncontrolled studies or extrapolated from the larger experience in adults, which is not always valid. The reader is referred to the chapters dealing with these conditions for a fuller discussion of the role of MTX in their overall management.
Dosage, Route of Administration, and Duration of Methotrexate Therapy Standard effective doses of MTX in children with JRA are in the range of 10 to 15 mg/m2/week or 0.3 to 0.6 mg/kg/week (Table 5–7). Improvement is generally
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
91
TABLE 5–7
Guidelines for Use of Methotrexate in the Treatment of Juvenile Idiopathic Arthritis Dose Initially, 10 mg/m2, once per week; give on an empty stomach with water, citrus, or carbonated beverage Increase dose as tolerated or as needed to 15 mg/m2 Administer subcutaneously once per week for doses >15 mg, intolerance, or nonadherence Administer with folic or folinic acid (see text) Clinical Monitoring Improvement should be seen by 6–12 wk Monitor every 3–6 mo, depending on course Reduce dose or discontinue and monitor for clinical or laboratory adverse events Laboratory Monitoring CBC with WBCC, differential, and platelet count; MCV; AST, ALT, albumin every 4–6 wk (see also Table 5–9) ALT, alanine aminotransferase; AST, aspartate aminotransferase; CBC, complete blood count; MCV, mean cell volume; WBCC, white blood cell count.
seen by about 6 to 8 weeks on effective doses, but may take up to 6 months. Children seem to tolerate much higher doses than adults, and some series have described using up to 20 to 25 mg/m2/week or up to 1.1 mg/kg/week in children with resistant disease with relative safety in the short term.169,170 The efficacy of the use of higher doses was not supported by a randomized controlled trial.151 Furthermore, the longer-term safety of MTX therapy at these doses is not known. Parenteral MTX administration should be considered in those children who (1) have a poor clinical response to orally administered MTX (this may be due to poor compliance or to reduced oral bioavailability for a variety of reasons); (2) need a dose in excess of about 10 to 15 mg/m2/week in order to achieve maximum clinical response (oral MTX absorption is a saturable process, whereas subcutaneous administration is not) (Fig. 5–8)189,190; or (3) develop significant GI toxicity with 24 20
AUC (M.h)
C H A P T E R
ou
16
ut bc u S
12
s
e an
8
Oral
4 0 0
10
20
30
40
50
60
Dose (mg/m2)
■ Figure 5–8 Bioavailability of oral and subcutaneously administered methotrexate. AUC, area under the curve. (Adapted from Wallace CA: New uses of methotrexate. Contemp Pediatr 11: 43–53, 1994.)
92
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
orally administered MTX (because there is some anecdotal experience that patients complain of less GI irritation when the drug is given parenterally).152,191 Studies in adult rheumatoid arthritis suggest that oral absorption of MTX is considerably reduced at doses of 15 mg or more and therefore should be administered parenterally.192,193 Furthermore, bypassing the enterohepatic circulation may also reduce hepatotoxicity. Some pediatric rheumatologists even advocate using parenteral MTX at initiation of treatment to ensure complete absorption and achievement of early disease remission.152,194 The issue of when, how, and by what criteria to attempt withdrawal of MTX therapy in JRA is currently undecided. There have been a number of studies in children treated with variable doses of MTX for variable periods in whom discontinuation of MTX was attempted after clinical “remission” of variable length was achieved.195–197 The criteria for “remission” or “relapse” have usually not been well defined or standardized among various studies, and the assessment of outcomes has been nonblinded. Given these limitations, no firm conclusions can be drawn about the optimal time and mode of MTX discontinuation in children with JRA. MTX withdrawal may result in disease flare in more than 50% of patients; this rate may be even higher in younger children.195,197 A longer period on MTX treatment after remission may not prolong the duration of improvement after stopping treatment, but the duration of clinical remission may be predicted by the degree of subclinical synovial inflammation at the time of stopping MTX.198 There are conflicting data about the ease with which remission can be reestablished when MTX is restarted after disease relapse, which may be related to the different doses of MTX used and the differing lengths of follow-up in the studies reported to date. Prospective studies with more standardized assessments of outcome and durations of follow-up are needed to address these issues.
Safety MTX is currently considered the second-line agent with the best toxicity/efficacy profile for the treatment of various rheumatic diseases in children and adults.172 Although MTX is associated with a number of potential toxicities, the documented overall frequency and severity of adverse effects in children with arthritis have been low (Table 5–8).150,152,199 Most side effects are mild and reversible and can be treated conservatively. The two areas of greatest concern and debate, especially in children, have been the potentially increased risk of hepatic cirrhosis in patients exposed to large cumulative doses of MTX and a possible increased risk of malignancies. Although the precise mechanism of all MTX-related toxicities is not yet clearly understood, at least some of its adverse effects are directly related to its folate antagonism and its cytostatic effects.200 This is especially evident in tissues with a high cell turnover rate, such as the GI tract and bone marrow, which have a high requirement for purines, thymidine, and methionine. Supplementation with folic or folinic acid may diminish these but not other types of toxicities which, along with MTX efficacy, may be mediated by different mechanisms (see later discussion).200
Gastrointestinal Toxicity Abdominal discomfort and nausea, the most frequently reported symptoms, occur in about 12% of children with JRA who receive MTX. Stomatitis or oral ulcers are reported in about 3% of children.152 Higher rates are reported in adults, with up to 60% of patients taking low-dose MTX developing GI adverse events in the form of stomatitis, anorexia, abdominal pain, indigestion, dyspepsia, nausea, vomiting, weight loss, or diarrhea.201,202 MTX-related abdominal discomfort, anorexia, nausea, or oral ulcers usually occur within 24 to 36 hours after administration of the weekly dose and can be diminished by the addition of folic acid supplementation (see later discussion), by dose reduction, or, in some troublesome cases, by conversion to subcutaneous MTX administration, although the evidence for the effectiveness of the latter strategy is only anecdotal.152 Liver Toxicity The effect of MTX on liver function and the development of hepatic fibrosis remains a major concern and has been extensively reviewed.203 MTX is associated with the potential for both acute and chronic hepatotoxicity. Mild acute toxicity, with elevations of transaminases, is common, occurring in about 9% of MTX-treated children with JRA.152 These elevations are usually transient and resolve with either MTX discontinuation or lowered dose after a brief interval off treatment.148,150 In some of these cases, concurrent administration of NSAIDs may be contributing to the elevation in transaminases, and discontinuation of NSAID treatment may allow normalization of liver function.204 The issue of greatest concern with the long-term use of low-dose MTX in children has been the potential for significant liver fibrosis or cirrhosis. However, the risk of this complication in children with JRA may differ from that in adults with rheumatoid arthritis or psoriasis treated with low-dose MTX. Long-term studies of MTX use in adult patients with rheumatoid arthritis have found a much lower incidence of liver fibrosis and cirrhosis than that initially reported in psoriatic patients; one retrospective study of 16,000 patients with rheumatoid arthritis reported a 5-year cumulative incidence of liver cirrhosis or failure of 1 in 1000 MTX-treated patients.205 Another study suggested that the incidence may be higher, with a 5-year cumulative incidence of cirrhosis of 9.4 in 1000 MTX-treated patients with rheumatoid arthritis.206 A meta-analysis of patients with rheumatoid arthritis and psoriasis found that higher cumulative dose of MTX, heavy alcohol consumption, and the presence of psoriasis were associated with higher risk of progressive liver histologic abnormalities.207 Other risk factors include preexisting liver disease, obesity, insulin-dependent diabetes mellitus, and renal insufficiency.203,208 The American College of Rheumatology has suggested guidelines for monitoring liver toxicity in patients with rheumatoid arthritis based on regular measurement of liver enzymes and performance of liver biopsy in selected cases with persistent abnormalities of liver enzymes (Table 5–9).209 These guidelines were developed by expert consensus and subsequently evaluated for their usefulness; they were found to be cost-effective
C H A P T E R
5 PHARMACOLOGY
AND
93
D RU G T H E R A P Y
TABLE 5–8 Reported Adverse Effects in Children Treated with Methotrexate* Study 1
Study 2
Study 3
Study 4
Study 5
Study 6
Study 7
Study 8
Totals
19
23
12
19
30
62
86
27
277
10.5 4–10
19.2 6–52
6 6
18.5 8–39
— 6–30
27 19–65
6 —
— 6–72
— 4–72
4–17 —
— 0.1–0.6
8–25 —
5–15 —
— 0.4–0.8
5–20 —
5 or 10 —
10–15 —
— —
2
0
1
2
6
14
8
0
33
0 1 1 0 ND 0 0 1 0 3
0 0 0 0 0 0 0 0 ND 10
ND ND 1 ND ND ND ND 0 1 1
0 1 0 0 0 0 ND 0 ND 1
0 0 2 ND ND ND ND 1 ND 3
4 0 0 0 2 ND 4 1 ND 9
0 2 1 1 0 0 0 0 0 1
1 1 0 0 0 1 ND ND ND 4
5 5 4 1 2 1 4 3 1 25
0 0 0 3
0 0 0 0
0 0 0 0
0 0 0 0
0 1 0 0
0 0 1 0
0 0 0 0
0 0 0 0
0 1 1 3
Patients (n) Treatment Duration (mo) Mean Range Methotrexate Dose mg/m2/wk mg/kg/wk
Adverse Effects (no. patients) Gastrointestinal symptoms Peptic ulcer Stomatitis Mouth ulcers Rashes Alopecia Jaundice Bacterial infections Herpes zoster Mood changes Liver function test elevations Hematuria Leukopenia Anemia Proteinuria
*Studies cited: Study 1: Truckenbrodt H, Hafner R: Methotrexate therapy in juvenile rheumatoid arthritis: a retrospective study. Arthritis Rheum 29: 801–807, 1986. Study 2: Wallace CA, Bleyer WA, Sherry DD, et al: Toxicity and serum levels of methotrexate in children with juvenile rheumatoid arthritis. Arthritis Rheum 32: 677–681, 1989. Study 3: Speckmaier M, Findeisen J, Woo P, et al: Low-dose methotrexate in systemic onset juvenile chronic arthritis. Clin Exp Rheumatol 7: 647–650, 1989. Study 4: Rose CD, Singsen BH, Eichenfield AH, et al: Safety and efficacy of methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 117: 653–659, 1990. Study 5: Halle F, Prieur AM: Evaluation of methotrexate in the treatment of juvenile chronic arthritis according to the subtype. Clin Exp Rheumatol 9: 297–302, 1991. Study 6: Graham LD, Myones BL, Rivas-Chacon RF, Pachman LM: Morbidity associated with long-term methotrexate therapy in juvenile rheumatoid arthritis. J Pediatr 120: 468–473, 1992. Study 7: Giannini EH, Brewer EJ, Kuzmina N, et al: Methotrexate in resistant juvenile rheumatoid arthritis. Results of the USA-USSR double-blind, placebo-controlled trial. The Pediatric Rheumatology Collaborative Study Group and The Cooperative Children’s Study Group. N Engl J Med 326: 1043–1049, 1992. Study 8: Huang JL: Methotrexate in the treatment of children with chronic arthritis—long-term observations of efficacy and safety. Br J Clin Pract 50: 311–314, 1996. ND, not determined. Modified from Singsen BH, Goldbach-Mansky R: Methotrexate in the treatment of juvenile rheumatoid arthritis and other pediatric rheumatic and nonrheumatic disorders. Rheum Dis Clin North Am 23: 811–841, 1997.
and clinically useful, with a sensitivity of 80% and a specificity of 82% but a low positive predictive value of 27%.208 Although a similar level of consensus does not exist for monitoring MTX-treated JRA in children, most pediatric rheumatologists tend to follow these guidelines or a variation of them.152 A study examining the relationship between hepatotoxic risk factors and liver histopathology in MTX-treated JRA found that serial biochemical abnormalities were significantly associated with Roenigk grade and the presence of liver fibrosis, suggesting that the guidelines for monitoring MTX hepatotoxicity in rheumatoid arthritis may also be applicable to patients with JRA.210 It must be remembered, however, that even close monitoring of liver function tests does not eliminate the possibility of progressive hepatic fibrosis. Erickson and colleagues208 reported one patient in a series who developed cirrhosis despite normal results on liver function tests. Furthermore, these guidelines may
not apply to patients receiving more than 25 mg/week or a cumulative dose of greater than 10 g; surveillance liver biopsies may be indicated for these patients.203 In a number of small studies in children, liver biopsies were performed after cumulative doses of up to 3000 mg had been reached; none showed any cirrhosis.150,211,212 A cross-sectional study213 reported on results of liver histology in children exposed to even higher cumulative doses of MTX (more than 3000 mg or more than 4000 mg/1.73 m2), with the mean duration of treatment being about 6 years (and some children treated for up to 10 years); no significant fibrosis or cirrhosis was found. However, 13 (93%) of 14 biopsies showed some histologic abnormality, with only 1 graded as Roenigk grade II. In addition, higher weekly doses of MTX (20 mg/m2/week or more) were not associated with significant hepatic fibrosis in 10 patients who underwent liver biopsy.214
94
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
TABLE 5–9 Recommendations for Monitoring for Hepatic Safety in Patients with Rheumatoid Arthritis Receiving Methotrexate (MTX) A. Baseline 1. Tests for all patients a. Liver blood tests (aspartate aminotransferase [AST], alanine aminotransferase [ALT], alkaline phosphatase, albumin, bilirubin), hepatitis B and C serologic studies b. Other standard tests, including complete blood cell count and serum creatinine 2. Pretreatment liver biopsy (Menghini suction-type needle) only for patients with a. Prior excessive alcohol consumption b. Persistently abnormal baseline AST values c. Chronic hepatitis B or C infection B. Monitor AST, ALT, albumin at 4- to 8-wk intervals C. Perform liver biopsy if 1. Five of nine determinations of AST within a given 12-mo interval (6 of 12 if tests are performed monthly) are abnormal (defined as an elevation above the upper limit of normal) 2. There is a decrease in serum albumin below the normal range (in the setting of well-controlled rheumatoid arthritis) D. If results of liver biopsy are 1. Roenigk grade I, II, or IIIA: resume MTX and monitor as in B, C1, and C2 above 2. Roenigk grade IIIB or IV: discontinue MTX E. Discontinue MTX in patient with persistent liver test abnormalities, as defined in C1 and C2 above, who refuses liver biopsy From Kremer JM, et al: Methotrexate for rheumatoid arthritis: suggested guidelines for monitoring liver toxicity. Arthritis Rheum 37: 316–328, Copyright © 1994 Wiley-Liss, Inc. Reprinted by permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
Although these data are encouraging, their interpretation requires some caution: First, the number of children studied is small, so the statistical power for detection of infrequent events, such as cirrhosis, is low (type II error); second, only 58% of eligible MTX-treated patients underwent biopsy, so selection bias may have occurred; and third, the clinical significance, if any, of the minor histologic abnormalities detected in most of the biopsies is not known, and there were no control biopsies to help distinguish the effects of disease (e.g., systemiconset JRA) or concomitant medications (e.g., NSAIDs) on liver histology. A follow-up study from the same group reported the results of 33 liver biopsies in 25 patients; 27 biopsies (82%) were classified as Roenigk grade I; 4 (12%) as grade II; and 2 (6%) as grade IIIA; none demonstrated significant fibrosis. The frequency of biochemical abnormalities and body mass index were the only risk factors found to significantly relate to the Roenigk grade.210 However, the same limitations as with the earlier study apply, particularly with respect to the small number of patients studied (possibility of type II error) and the unknown significance of the minor histologic abnormalities observed in most patients. Further longterm, prospective studies in larger numbers of MTXtreated children are needed to define more accurately the risk of MTX-related liver fibrosis or cirrhosis and to develop appropriate guidelines for monitoring therapy in JRA.
Infection Although MTX may potentially increase the risk for common bacterial infections, herpes zoster, and opportunistic infections, these complications are infrequently reported in treated patients. One study of 62 children with JRA treated for a mean of 161 weeks reported only 2 cases of recurrent cellulitis, 1 of osteomyelitis, and 1 of an infected Hickman catheter site; there were 8 viral infections, including 1 case of herpes zoster, 1 case of mononucleosis, and 6 cases of primary varicella. Many of the patients with varicella infection were also concurrently receiving glucocorticoids.150 The randomized controlled clinical trial of MTX treatment in JRA did not document an increased frequency of infection in children treated with MTX compared with placebo.148 Although the overall risk of infection appears to be low, it has not been precisely quantitated in adults or in children. Infections in MTX-treated patients are usually common bacterial infections (e.g., lungs, skin) or herpes zoster. The development of hypogammaglobulinemia in MTX-treated patients may predispose to infection.215 Opportunistic infections associated with MTX treatment are rare unless there is concurrent treatment with highdose glucocorticoids.216 Immunization with inactivated vaccines is not contraindicated in MTX-treated children, but immunization with live attenuated vaccines should be avoided.217 There are currently no generally accepted guidelines regarding varicella immunization in these children. However, active varicella immunization of susceptible children and family members may need to be considered before initiation of MTX therapy.205 Family members of MTX-treated patients who require polio immunization should receive inactivated vaccine. Hematologic Toxicity Hematologic toxicity includes macrocytic anemia, leukopenia, thrombocytopenia, and pancytopenia. In adult patients with rheumatoid arthritis, pancytopenia has been reported in about 1% to 2% of MTX-treated patients.218 Identified risk factors in this population include impaired renal function, advanced age, concurrent viral infection, alcohol ingestion, folate deficiency, hypoalbuminemia, and drug interactions (e.g., trimethoprim-sulfamethoxazole). MTX-associated pancytopenia has not been reported in children, and hematologic toxicity is uncommon overall; a 1997 review of published studies, including a total of 277 MTX-treated children, found only 1 case of leukopenia and 1 case of macrocytic anemia.152 Although supplemental folate treatment results in lowering of the mean corpuscular volume in MTX-treated patients, whether this will prevent pancytopenia is not yet known. Spontaneous recovery is usual within 2 weeks after withdrawal of MTX in patients with mild bone marrow suppression. Patients with moderate to severe bone marrow suppression may require folinic acid rescue and supportive therapy (e.g., colony-stimulating factors).202 MTX may have triggered the macrophage activation syndrome in one patient with systemic-onset JIA.219 Reversible asymptomatic eosinophilia has also been described.220 Malignancy The issue of whether MTX treatment is an independent risk factor for various malignancies is controversial and remains unresolved. Although in vitro
C H A P T E R
studies have shown that MTX has mutagenic and carcinogenic potential, in vivo studies in animal models (mice, rats, hamsters) have failed to demonstrate any carcinogenicity. In humans, low-dose weekly MTX therapy has not been convincingly linked to malignancy.221 However, there have been a number of case reports of an association between MTX treatment and lymphoproliferative diseases in adult patients with rheumatoid arthritis.222,223 It has not been possible to determine whether these observations were merely coincidental or were causally linked to MTX or to the underlying disease process.221 Rheumatoid arthritis is known to be associated with an increased risk of hematologic malignancy.224 There have been several cases of Epstein-Barr virus (EBV)–associated lymphoma manifesting during the course of MTX treatment for rheumatoid arthritis or dermatomyositis that regressed with discontinuation of MTX.225 Some of the reported cases demonstrated features typical of immunosuppression-induced lymphoproliferation, including extranodal location, large cell or polymorphous histology, geographic areas of necrosis, and the presence of EBV.226 The association of MTX with reversible lymphoproliferation suggests a causative role. However, MTX has not been shown to increase the risk of lymphoproliferative disorders when used in the treatment of other diseases such as psoriasis227,228 or in bone marrow transplantation.221 Several cases of both Hodgkin’s lymphoma229–231 and non-Hodgkin’s lymphoma232,233 have been reported in children with JRA treated with MTX. In two of these, EBV was implicated.231,233 Long-term prospective cohort studies, with appropriate controls, are needed to define the risk of hematologic or other malignancies in MTX-treated patients.
Pulmonary Toxicity Significant pulmonary toxicity occurring during the course of treatment with low-dose weekly MTX has been described in adult patients with rheumatoid arthritis; reported prevalence rates in published studies range from 0.3% to 18%, with a mean prevalence of 3.3%.234 However, the actual frequency of this complication is difficult to estimate, because the literature has not been clear in defining toxicity related to the drug itself rather than to secondary problems associated with MTX therapy (e.g., opportunistic lung infections). The issue is clouded further by the fact that rheumatoid arthritis itself is associated with interstitial lung disease. The mechanism of this toxicity and the risk factors for its development are poorly defined, although a number of studies implicate preexisting lung disease as an important predisposing factor in the development of MTX pneumonitis.202,234,235 Interestingly, MTX-associated pneumonitis is rarely reported in psoriatic patients. In 1998, the first pediatric case of possible MTX-induced pneumonitis was reported in an 11-year-old girl with rheumatoid factor (RF)–positive, antinuclear antibody–negative polyarticular JRA.236 However, the clinical course of lung disease in this case was not typical of that described in adults with MTX pneumonitis, clinical and laboratory information was insufficient to satisfactorily exclude alternative causes, and no biopsy was performed. Prospective studies of lung function in children with
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
95
various types of JRA have not demonstrated any significant abnormalities in pulmonary function test results in those treated with MTX.149,150,237–239
Accelerated Nodulosis MTX has been associated with a syndrome of “accelerated nodulosis,” with an estimated prevalence of 8% in adult patients with rheumatoid arthritis.201 This association was described in two teenagers with RF-positive JRA240 and in one 3-year-old girl with systemic-onset JRA.241 The nodules were similar in distribution and size to those reported in adults and developed within 6 months after the initiation of MTX treatment in these children. Although discontinuation of MTX is associated with regression of nodulosis, some patients have been successfully treated with hydroxychloroquine240 or colchicine,242 allowing stabilization of nodulosis with continued MTX treatment of the underlying disease. Although the exact mechanism of MTX-associated nodulosis is not known, it is thought to be mediated by MTX-enhanced adenosine production; therapies that inhibit adenosine production or interfere with adenosine A1 receptor function may be effective in treating MTX-associated nodulosis.201
Other Adverse Effects Central Nervous System Various CNS symptoms, including headaches, mood alterations, change in sleep patterns, irritability, fatigue, and impaired academic performance, have been reported by some pediatric rheumatologists to occur transiently in the 12 to 48 hours after the weekly dose of MTX.152 These problems may be subtle and need to be differentiated from effects of underlying disease and from various psychosocial issues that may coexist. Osteopathy Animal studies have shown that prolonged administration of low-dose MTX is associated with suppression of osteoblast activity and stimulation of osteoclast recruitment, resulting in increased bone resorption and osteopenia.243 Similar effects have been described in a small number of case reports in adults with rheumatoid arthritis or psoriasis treated with lowdose weekly MTX.244,245 Although leg pain and spontaneous fractures attributed to MTX therapy in pediatric oncology have been recognized for some time, this phenomenon has been described in only one patient with polyarticular JRA and a disease duration of 25 years recently treated with low-dose MTX.246 Teratogenicity MTX therapy is associated with spontaneous abortions.247 In 1997, a case of multiple congenital abnormalities in a baby whose mother was treated with low-dose MTX for JRA was reported.248 Although it is difficult to quantitate the risk of teratogenicity with lowdose weekly MTX treatment, women of child-bearing age should be counseled to practice effective contraception during the course of treatment. Patients should be informed that past MTX use does not predispose to congenital abnormalities and that, ideally, MTX should be discontinued before attempts at conception. There have not been any reports of azoospermia due to low-dose MTX treatment of JRA.152 MTX is excreted in breast milk
96
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
in low concentrations, but it is not known whether this affects the newborn. Women taking MTX should be advised not to breast-feed.249
N CI CHOHCH3
Folate Supplementation A number of studies have examined the issue of minimizing MTX toxicities with the use of concurrent folic or folinic acid (leucovorin) supplementation in adults with rheumatoid arthritis.250–253 These studies have evaluated the effect of folate supplementation on both the efficacy and the toxicity of low-dose weekly MTX therapy. Although the doses and regimens of folic and folinic acid used in the various trials, the doses of concurrent MTX therapy, and the assessment of “toxicity” have not been standardized, there is evidence for overall effectiveness of folate supplementation. A 1998 systematic review of all published clinical trials found that folic acid supplementation was associated with a significant reduction in mucosal and GI toxicity in MTX-treated adults with rheumatoid arthritis.254 There was no adverse effect on efficacy except with high-dose folinic acid supplementation. Currently, data are insufficient to assess the effect of long-term folate supplementation on hepatic or hematologic toxicities. It also is not clear whether folate supplementation should be commenced as prophylaxis with initiation of MTX therapy in all patients or as treatment if specific toxicities develop. There has not been any formal cost-benefit study to address this issue. Folic acid is cheaper and has a greater margin of safety in dosing compared with folinic acid. However, the two agents have not been directly compared for their clinical effectiveness and cost. Studies in children are limited. A short-term, randomized, double-blind, placebo-controlled, crossover trial of folic acid (1 mg/day) added to a stable dose of MTX (mean, 9–9.7 mg/week) in 19 children with JRA showed no effect on clinical efficacy.255 There were no observable abnormalities of liver function, but no information about other toxicities is available from this small study. Folinic acid administered 24 hours after the weekly dose of MTX at doses of approximately one-third of the MTX dose has been used effectively to treat manifestations of MTX toxicity in children with JIA,256 but at high doses this treatment was associated with disease flares.257 At present, it is not possible to make firm recommendations about routine folate supplementation in MTX-treated children. However, based on the information from adult studies and the small trial in children with JRA, it appears that low-dose (1 mg/day) folic acid supplementation does not have any detrimental effect on disease control and confers a beneficial effect in terms of GI and mucosal toxicities associated with low-dose weekly MTX treatment. Folic acid supplementation should be considered at least in symptomatic patients. High-dose folinic acid rescue should be reserved for those with severe, life-threatening toxicity (e.g., aplastic anemia).
Antimalarials Hydroxychloroquine sulfate (Fig. 5–9) is the least toxic of the 4-aminoquinolone drugs and has generally supplanted chloroquine in rheumatologic practice,258–260 although other antimalarial agents are occasionally used for recalcitrant skin disease in SLE. Hydroxychloroquine is rapidly absorbed from the intestine. Equilibrium concentrations
NH
CH(CH2)3 CH3
■ Figure 5–9
N CH2CH3
Structure of hydroxychloroquine.
are reached after 2 to 6 months of a constant daily dose, and the half-life exceeds 40 days.261 Tissue levels are much greater than plasma concentrations, and there is increased affinity of the drug for melanin as well as for the liver, pituitary, spleen, kidney, lung, and adrenals. Excretion is primarily via the kidney, although hepatic oxidative deamination accounts for part of the excretion. The antimalarial drugs inhibit the synthesis of DNA, RNA, and protein by interacting with nucleic acid.258 These drugs alter lysosomal pH, thereby interfering with ligand-receptor dissociation and antigen processing; stabilize lysosomal membranes262; inhibit antigen–antibody reactions263; suppress lymphocyte responses to mitogens; act as antioxidants262; inhibit phospholipase activity264; and inhibit neutrophil chemotaxis and phagocytosis.258 They may also antagonize the action of some of the prostaglandins.265 Antimalarials interfere with IL-1 release by monocytes263; interfere with production of TNF-α, IL6, and IFN-γ266; inhibit natural killer activity267; and induce apoptosis.268 They also have antiplatelet and antihyperlipidemic effects that are extremely important in patients with SLE.269 Antimalarials are important in many rheumatic diseases. In JRA, the therapeutic efficacy has never been established, and the only placebo-controlled study did not prove these agents to be better than placebo.270 However, recent work in adult rheumatoid arthritis has demonstrated them to be effective, especially in early disease.271 Antimalarials are also effective in combination with other DMARDs in adult rheumatoid arthritis.272 Hydroxychloroquine reduces some MTX-related toxicity, such as elevated liver enzymes and nodulosis.273,274 Studies in adult SLE have also provided proof of efficacy for hydroxychloroquine, especially in preventing flares of disease.275 Its role in SLE is primarily for treatment of dermatitis, arthritis, and serositis. It may have particular benefit as a result of its lipid-lowering effect in steroid-treated SLE patients.276–278 It has also been used in dermatomyositis, especially for skin disease, but the results are not clear.279,280
When used at recommended doses, the antimalarials are considered extremely safe. However, at least four young children have died from respiratory failure after accidental ingestion of large doses (1 to 3 g) of chloroquine,281 and it is recommended that antimalarials be used with great caution in the very young child because there is no antidote. GI intolerance occurs in 10% of adults but is probably less common in children. Antimalarials occasionally cause bleaching of the skin and hair. Rarely, neuropathy or myopathy occurs. CNS side effects are common, may be reversible with dose reduction, and may remit spontaneously. These include headache, lightheadedness, tinnitus, insomnia, and increased nervousness. Myasthenia and muscle weakness have been described.282
C H A P T E R
The major side effect of concern is retinal toxicity.283–285 Antimalarials accumulate in the pigmented cells of the retina and persist for long periods after they have been discontinued; however, the binding to melanin may not be predictive of ocular damage. Retinal toxicity, although rare, can cause blindness, even after the medication has been stopped. Retinitis is sometimes, but not always, reversible.284 Evidence in adults suggests that retinal toxicity will not occur if the dose of hydroxychloroquine is maintained at less than 6.5 mg/kg/day, even for as long as 7 years.286 Early detection of pre-maculopathy prevents visual loss if the medication is discontinued and forms the basis for routine ophthalmologic monitoring (every 6 months) with visual field testing, color vision testing, corneal examination, and visual acuity testing. A progressive loss of color vision may signify early retinopathy and is an indication for stopping the drug. Corneal deposits are not visually limiting but are also probably an indication to lower the dose. Debate continues as to the necessity of an initial ocular examination, or of biannual examinations. The author’s current policy is to continue to perform these examinations every 6 months but not to perform a baseline examination. Recommendations differ somewhat in adults.287 Each examination should include visual acuity, color vision testing, visual field corneal examination,286,288 and retinoscopy. Retinal abnormalities or interference with vision, especially with foveal recognition of red,289 is an absolute indication for discontinuing the medication. Use of hydroxychloroquine in children younger than 7 years of age may be limited by difficulty in obtaining satisfactory evaluation of color vision in this age group.
The dose of antimalarials is limited by retinal toxicity. For hydroxychloroquine, the recommended dose is less than or equal to 6.5 mg/kg/day to a maximum of 400 mg/day.290 The overall retinal toxicity seems to be related to daily dose rather than to cumulative dose. One study of children with JRA did correlate outcome with serum levels of hydroxychloroquine,291 but levels are not measured in clinical practice. Hydroxychloroquine crosses the placenta but is considered safe to use during pregnancy.292 Hydroxychloroquine does appear in breast milk, but the amount ingested per day by the breast-feeding infant would be very low.293 It seems reasonable for the mother taking hydroxychloroquine to breast feed if she had taken hydroxychloroquine during pregnancy.
Sulfasalazine Sulfasalazine is an analogue of 5-aminosalicylic acids linked by an azo bond to sulfapyridine, a sulfonamide (Fig. 5–10). Its development was based on the concept that rheumatoid arthritis might be an infectious disease and would respond to combination therapy with an anti-
COOH N HO
N
N
SO2NH
■ Figure 5–10 Structure of sulfasalazine.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
97
bacterial agent and an anti-inflammatory drug.294,295 Sulfasalazine has become a primary therapeutic choice in the treatment of mild to moderate inflammatory bowel disease, and it has been reported to be beneficial in the management of childhood arthritis,296–303 psoriatic arthritis,304 and reactive arthritis.305 Its role in ankylosing spondylitis is controversial,306–308 although it does seem to be effective for the peripheral arthritis.309 Sulfasalazine is poorly absorbed from the GI tract.310–312 Peak serum concentrations are reached after 5 days of therapy. The half-life of the drug is 10 hours. Approximately one third of the dose is absorbed in the small intestine and excreted unchanged in the bile. The remaining 70% enters the colon intact, where the azo linkage is split by bacterial enzymes to sulfapyridine, which is absorbed and excreted in the urine, and 5-aminosalicylate, which reaches high concentrations in the feces. Approximately 90% of the sulfapyridine is absorbed from the colon. Sulfapyridine is tightly protein bound and acetylated, hydroxylated, and conjugated with glucuronic acid in the liver. Both sulfasalazine and sulfapyridine reach synovial fluid in concentrations comparable to those in serum. About one third of the 5-aminosalicylic acid is absorbed, acetylated, and excreted in the urine. The rest is eliminated unchanged in the stool. The small amount of salicylate absorbed is not sufficient to reach anti-inflammatory levels in the plasma. Several mechanisms of action may explain the antiinflammatory effect of sulfasalazine. Bacterial growth is reduced by sulfasalazine and sulfapyridine, and thus the bacterial antigenic load delivered to the gut-associated lymphoid tissue may be reduced. This may be important for patients with spondyloarthropathies, in whom bacteria may gain access through inflamed gut mucosa and stimulate the immune system. Sulfasalazine interferes with a number of enzymes that are important in inflammation, in the formation of leukotrienes and prostaglandins.313 Sulfasalazine is a potent inhibitor of AICAR transformylase; as a result, there is an accumulation of extracellular adenosine with a consequent reduction in inflammation via occupancy of A2 receptors on inflammatory cells.314 Levels of matrix metalloproteinase 3 (MMP3) are decreased in patients with early rheumatoid arthritis responsive to sulfasalazine.315 Sulfasalazine reduces the release of IL-1, IL-2, TNF-α, IL-6, and IFN-γ.316–318 This effect is most likely mediated by inhibition of the degradation of I-κB (inhibitor of nuclear factor-κB [NF-κB]), which results in an inhibition of NF-κB upregulation of gene transcription,319 and by the induction of apoptosis through the activation of caspase 8.320 Sulfasalazine decreases natural killer cell activity and induces neutrophil apoptosis in vitro.321 In vitro effects on macrophages include suppression of production of IL-12, production of nitric oxide, and expression of major histocompatibility complex (MHC) class II molecules.322 Sulfasalazine may also have anti-angiogenic properties.322,323 Intolerance and toxic reactions occur in approximately 20% of sulfasalazine-treated adults with rheumatoid arthritis (range, 5% to 55%).324–348 In a placebo-controlled study of children with JRA, 29% of 35 patients developed adverse effects that led to discontinuation of the drug.299
98
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Toxicity may be more common in patients with a slow acetylator phenotype, but there is no clinical indication to document a patient’s acetylator status before starting sulfasalazine.349 Enteric-coated preparations probably cause fewer GI side effects (anorexia, nausea, vomiting, dyspepsia, diarrhea). Rashes occur in 1% to 5% of patients. A maculopapular rash occurring within 2 days after institution of therapy, especially on sun-exposed skin, is the most common dermatologic complication.324 In patients who develop hypersensitivity reactions (usually early), desensitization protocols can be carried out. Oral ulcers325 and the Stevens-Johnson syndrome326 are uncommon but important complications. Neutropenia occurs in up to 4.4% of patients treated with sulfasalazine.327 Sulfasalazine-induced thrombocytopenia has also been reported,328 and pancytopenia329 and macrocytic anemia330 may occur. It is important to note that serious hematologic toxicity can develop many months after starting treatment. Drug-induced SLE,331 Raynaud’s phenomenon,332 interstitial pneumonitis, fibrosis, alveolitis,333,334 and hepatitis (granulomatous hepatitis, elevated transaminases)335 are rare complications of sulfasalazine therapy. Hypogammaglobulinemia and IgA deficiency have been reported,350 and immunoglobulin levels should be monitored. However, serious infections have not been reported. A reversible decrease in sperm count has been observed,336 but there are no reports of increased fetal wastage or abnormalities. The drug should not be used in infants or in those with known hypersensitivity to sulfa drugs or salicylates, impaired renal or hepatic function, or specific disease contraindications (e.g., porphyria, glucose-6-phosphate dehydrogenase deficiency). Most authors also believe that sulfasalazine is contraindicated in patients with systemic-onset JRA because of an apparent increased risk of diffuse intravascular coagulation (DIC)–like reactions,298,351 as well as in patients with adult-onset Still’s disease.352 The suggested dosage in children is 30 to 50 mg/kg/day in two to three divided doses, usually taken with food or milk.296,353 Treatment is initiated at a lower dose (10 to 15 mg/kg/day) and increased weekly over 4 weeks to achieve maintenance levels. A satisfactory clinical response may occur within 4 to 8 weeks. Sulfasalazine should probably be continued for at least 1 year after disappearance of clinical disease before tapering is begun (Table 5–10). It has been reported that sulfasalazine can be used safely during pregnancy354 and that women can breast feed while taking sulfasalazine.355
Leflunomide Leflunomide (Fig. 5–11) is an immunomodulatory agent that, through its active plasma metabolite, A77-1726, inhibits de novo pyrimidine synthesis by inhibiting the enzyme dihydro-orotate dehydrogenase.356 Activated lymphocytes require de novo pyrimidine synthesis for proliferation. As a result of the inhibition, p53 in the cytoplasm translocates to the nucleus and initiates cellular arrest in the G1 phase of the cell cycle. It also inhibits tyrosine kinase,357 inhibits leukocyte-endothelial
TABLE 5–10
Guidelines for Use of Sulfasalazine
Dose Initially, 12.5 mg/kg/day (maximum, 500 mg), given in one dose; increase to maintenance dose over 4 wk Maintenance dose: 50 mg/kg/day to a maximum of 2 g/day for 1 yr or longer Clinical Monitoring After first month, then every 2–3 mo to follow disease course; discontinue if rash appears Laboratory Monitoring CBC with WBCC differential, and platelet count; hepatic enzymes; and urinalysis every week until maintenance dose is achieved, then monthly for 2 mo, then every 3 mo. Immunoglobulin levels every 6 mo. Discontinue if persistent neutropenia, thrombocytopenia, elevated hepatic enzymes, or decreased immunoglobulins CBC, complete blood count; WBCC, white blood cell count.
CF3 O N H N O
CH3
■ Figure 5–11 Structure of leflunomide.
adhesion,358 and affects cytokine production leading to immunosuppression.359 Because its actions are similar to those of MTX, it might be better classified as a DMARD. In vitro, leflunomide inhibits the production of prostaglandin E2, MMP1, and IL-6 and modulates various tyrosine kinases and growth factor receptors.360 Leflunomide is rapidly converted to A77-1726, which is highly protein bound and has a prolonged half-life of up to 18 days.361 As a result, loading doses have been recommended for the first 3 days of administration to rapidly achieve steady state. Another result of this prolonged half-life is that the metabolite remains in the circulation for prolonged periods (Table 5–11). Initial studies in adults with rheumatoid arthritis showed that, after a 3-day loading dose of 100 mg/day, daily doses of 10 and 25 mg of leflunomide were more effective than placebo361,362 as effective as sulfasalazine over 24 weeks363 and as effective as MTX at 1 year364 and at 2 years.365 After a 3-day loading dose of 100 mg/day, daily doses of 10, 20, and 25 mg have shown benefit as early as 4 weeks and continuing improvement for up to 20 weeks, after which the clinical improvements are maintained. The major side effects from leflunomide have been related to the liver, with elevation on liver function tests occurring in approximately 5% of patients with rheumatoid arthritis. However, the risk of serious hepatic disease appears to be minimal and, as with MTX, associated with preexisting liver disease, viral hepatitis, or heavy alcohol consumption. More significant hepatotoxicity was observed when MTX was combined with leflunomide.366 Side
C H A P T E R
TABLE 5–11
Guidelines for the Use of Leflunomide in the Treatment of Juvenile Idiopathic Arthritis Dose 10–20 mg/day, based on weight (usual adult dose = 20 mg/day) Clinical Monitoring Improvement should be seen by 6–12 wk Monitor every 3–6 mo, depending on course Reduce dose or discontinue and monitor for clinical or laboratory adverse events Laboratory Monitoring CBC with WBCC, differential and platelet count; AST, ALT, albumin every 4–6 wk ALT, alanine aminotransferase; AST, aspartate aminotransferase; CBC, complete blood count; WBCC, white blood cell count.
effects from leflunomide have been mild and doserelated. These include GI side effects (abdominal pain, dyspepsia, anorexia, diarrhea, gastritis), allergic rash, reversible alopecia, mild weight loss, and elevation of liver function test results.363,367 No increase in infection has been reported. Silverman et al reported on 27 patients with polyarticular course JRA who had failed treatment with or were intolerant of methotrexate and were treated with leflunomide.368 After an initial loading dose of ~100 mg/1.73 m2, patients received 10 mg/1.73 m2; the dose could be increased to 20 mg/1.73 m2 after week 8 for a poor response. After 26 weeks, 14 patients (52%) had improved according to the ACR Pedi 30 criteria; 44% and 19% met criteria for an ACR Pedi 50 and 70 respectively. Forty-four percent of patients responded as early as 4 weeks, and all who responded did so by 12 weeks. Seventeen of 27 patients entered an extension phase during which 65% of patients achieved an ACR Pedi 30, 47% an ACR Pedi 50 and 35% an ACR Pedi 70. Adverse events led to discontinuation in two. Other adverse events reported in at least 30% of patients were transient elevation of liver function tests, headache, alopecia, abdominal pain, nausea, diarrhea and dizziness. In a subsequent study, Silverman et al also compared the safety and efficacy of leflunomide with methotrexate in the treatment of patients with polyarticular course JRA in a multinational, randomized controlled trial.368a The dose of leflunomide was dependent on the weight of the child. A loading dose of 100 mg for 1, 2 or 3 days for a weight <20 kg, 20–40 kg, >40 kg, respectively, was followed by a dose of 10 mg every other day, 10 mg daily, or 20 mg daily for a weight <20 kg, 20–40 kg, >40 kg, respectively. Methotrexate was given at a dose of 0.5 mg/kg/week (maximum 25 mg per week). In addition, pharmacokinetic studies were performed at week 16. At week 16, 68% of patients receiving leflunomide showed an ACR Pedi 30, versus 89% of patients treated with methotrexate; the improvements achieved were maintained at a similar rate in a 32 week extension study. The median time to an ACR 30 did not differ between the two groups (52 days in leflunomide, 56 days in methotrexate). Body weight was a significant determinant of response, and patients weighing less than 20 kg showed the greatest discrepancy between the two groups. The incidence of treatment related adverse events was similar in both groups. The clinically active metabolite of leflunomide, M1, was lower in patients weighing less than 40 kg than those associated with clinical responses in adult rheumatoid arthritis, which may have
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
99
explained the discrepancy between responses in the patients weighing less than 40 kg.
Leflunomide is teratogenic.369 Because of the very long half-life of this drug, it has been recommended that cholestyramine be administered and then that drug levels lower than 0.02 mg/L be verified on two separate tests at least 2 weeks apart, in both men and women.369 In addition, women of child-bearing potential must have a negative pregnancy test before starting leflunomide and must practice contraception. The place of leflunomide in the treatment of patients with JRA is not yet established. At the moment, it should be considered for patients who do not tolerate MTX. A loading dose is recommended in adults with rheumatoid arthritis to rapidly achieve steady state but is not necessary and may lead to significant GI upset. The usual daily dose for adults is 20 mg, which may be reduced to 10 mg in the face of adverse effects.
Gold Compounds The use of gold and penicillamine has waned dramatically since the introduction of MTX and the newer biologic agents, and the descriptions included here are primarily for historical interest and importance. Sulfhydryl-containing organic gold compounds have been prescribed for treatment of rheumatoid arthritis since the observations of Forestier370,371 in the 1920s. In the management of JRA, they have essentially been replaced by MTX and sulfasalazine, except perhaps for patients with RF-positive disease.
Pharmacology The mechanisms of action of gold compounds are not entirely clear372 but appear to depend on the sulfhydryl bond, which is also thought to be responsible in part for the beneficial effects of D-penicillamine. Gold compounds may diminish vascular permeability, reduce the number of inflammatory cells in rheumatoid foci, and impair phagocytosis. They stabilize lysosomal membranes and suppress enzymatic activity in general. They may prevent denaturation of proteins induced by free oxygen radicals. In vitro lymphocyte responses to mitogens and specific antigens are inhibited, and monocyte interaction in cell-mediated immune function is impaired.373–375 The intramuscular preparations are 50% gold by weight (Fig. 5–12).376 The oral gold compound auranofin is 30% gold by weight and is lipid soluble. With aurothiomalate, the gold is predominantly bound to serum albumin; however, with auranofin, approximately 50% of the gold is bound to leukocytes and erythrocytes. Gold compounds are excreted in both urine and stool. They cross the placenta in substantial concentrations, apparently without harm to the fetus, and the level in breast milk is low. The gold concentration in synovial fluid is approximately half of that in blood. Gold is concentrated in organs rich in mononuclear phagocytes and in synovial type A cells. The initial half-life of intramuscularly administered gold is approximately 7 days but increases with chronic administration. After discontinuation of gold injections, plasma concentration falls to undetectable levels by 40 to 80 days, but gold is still excreted in the urine for up to 1 year.377 Auranofin is more hydrophobic than the intramuscular gold compounds and is readily absorbed orally.378,379 The tissue halflife of gold is approximately 80 days. Plasma gold concentrations in patients treated with auranofin take longer to reach
100
C H A P T E R
5 PHARMACOLOGY S
AND
D RU G T H E R A P Y
AU
TABLE 5–12
Guidelines for Use of Aurothiomalate or Aurothioglucose
HC HC
Initial Course
OH
HCOH
0.75–1.0 mg/kg/wk Test doses of 5 mg, then 10 mg, then 25 mg weekly; maximum of 50 mg/wk for at least 20 wk
HCO
Maintenance
HOCH
1 mg/kg (maximum, 50 mg) every 2 wk for 3 mo; then every 3 wk for 3 mo, then every 4 wk thereafter
CH2OH AUROTHIOGLUCOSE
Clinical Monitoring Clinical evaluation before every injection: dermatitis, pruritus, stomatitis, vasomotor (nitritoid) reaction
CH2COONa Au
S
Laboratory Monitoring
CHCOONa
Weekly before each dose: CBC, WBCC, differential, and platelet count; urinalysis. Monthly: hepatic enzymes, BUN, creatinine Discontinue drug if WBCC <3500/mm3 (<3.5 × 109/L), platelet count <100,000/mm3 (<100 × 109/L), persistent proteinuria (>0.5 g/24 hr) or hematuria
AUROTHIOMALATE
O CH2OCCH3 O CH3CO
O S
Au
O O CCH 3
P(C2H5)3
OCCH3 O AURANOFIN
■ Figure 5–12 Structure of gold compounds.
plateau levels and are only 20% of those achieved with aurothiomalate.379–381 However, no direct relationship has been observed between plasma gold concentrations and either clinical benefit or toxicity with any gold compound. The drug is principally excreted in the urine.
Intramuscular Gold Administration and Toxicity Two intramuscular gold preparations are available in North America: aurothioglucose and sodium aurothiomalate.381 Neither preparation has a clear-cut advantage over the other. Aurothioglucose is more viscous and requires a larger-bore needle for administration. Hypersensitivity reactions after aurothioglucose administration are sometimes related to the oil vehicle. Aurothiomalate may rarely induce a nitritoid reaction (flushing, tachycardia, occasionally transient hypotension). Although intramuscular gold compounds are reasonably safe (Table 5–12),382,383 a complete blood count and urinalysis should be obtained before each injection, to detect early signs of toxicity. Toxicity requiring discontinuation of the drug occurs in 25% of children,385–394 pruritic dermatitis in approximately 15%, and stomatitis in approximately 5% to 10%. Pigmentation (chrysiasis) of the skin and mucous membranes may occur. Although rashes and mouth ulcers are the most common side effects of gold therapy, the greatest difficulty encountered in maintaining children with JRA on gold may be the development of an abnormal urinary sediment,395,396 which is related to damage to the proximal tubules or, rarely, to glomerulonephritis. Because children with JRA may have minimal, transient microscopic hematuria and proteinuria
BUN, blood urea nitrogen; CBC, complete blood count; WBCC, white blood cell count.
unrelated to drug intake, a thorough renal evaluation is important before gold therapy is initiated. During gold therapy, renal function should remain normal, and microscopic hematuria should be minimal and intermittent at most and unaccompanied by significant proteinuria (300 mg/24 hours or less). If any question of toxicity arises, the drug should be discontinued and the child should be re-evaluated 1 week later before readministration of the drug is considered, perhaps at a lower dose. An immune complex–mediated membranous glomerulonephritis, heralded by proteinuria, accounts for 10% to 20% of renal toxicity and may result in the nephrotic syndrome.397 Significant proteinuria is an absolute contraindication to reinstitution of gold therapy. Hematologic abnormalities, including leukopenia, eosinophilia, and thrombocytopenia, occur in only 1% to 2% of gold-treated patients; aplastic anemia is rare but may be fatal.398 The risk of mucocutaneous, hematologic, or renal toxicity from gold compounds in adults with rheumatoid arthritis is increased in patients with the human leukocyte antigen allele HLA-DR3.399 Other toxic reactions that have been reported specifically in children with JRA include DIC,80,400 intrahepatic cholestasis and liver disease,401,402 and hypogammaglobulinemia.403 Pulmonary injury induced by gold compounds is uncommon and evidently has not been reported in children.404,405 In adults, pulmonary toxicity is characterized by cough, dyspnea, fever, pleuritic pain, eosinophilia, and skin rash. Neurologic complications of gold therapy include psychiatric syndromes and focal, central, and peripheral nerve abnormalities.406 Occasionally, glucocorticoids or chelating agents are prescribed for severe gold-induced reactions such as neutropenia or exfoliative dermatitis. 406 Reports of accidental overdose of injectable gold compounds (500 mg twice in 1 week) noted no significant ill effects,407 but other reports of acute toxicity have been published,408 including one in a child with JRA.409 One study indicated that 7 surviving infants of 10 pregnancies (there were 2 abortions and 1 stillbirth) exposed in utero to maternal gold administration had no long-term adverse effects.410 No teratogenic or embryopathic effects were encountered.
C H A P T E R
5 PHARMACOLOGY
Oral Gold Administration and Toxicity
TABLE 5–13
Triethylphosphine gold, auranofin, was effective in the treatment of adults with rheumatoid arthritis.411 Preliminary openlabel, noncontrolled studies in children with JRA indicated that almost all patients sustained some improvement after 6 months at dosages between 0.1 and 0.2 mg/kg/day.412–416 However, a subsequent double-blind trial demonstrated no significant advantage of auranofin over placebo.417 Orally administered gold appears to be better tolerated than the injectable compounds. Auranofin therapy is associated with loose stools in approximately 10% of patients, a side effect that can often be controlled by decreasing the dose. Proteinuria and hematologic toxicity appear to be less common than with intramuscularly administered gold. Stomatitis and dermatitis are about equally frequent with both the oral and the injectable compounds. D-Penicillamine D-Penicillamine
(D-β,β-dimethylcysteine) (Fig. 5–13) is now rarely used in the treatment of JRA because safer and more effective agents have replaced it. Its role in the treatment of scleroderma has been questioned.418 It has three major biochemical effects.419 It influences the formation of disulfide bonds with other sulfhydryl compounds by either an oxidation or an exchange reaction; it undergoes condensation with aldehydes and ketones to give substituted thiazolidinedione carboxylic acids, causing inhibition of collagen cross-linking and dissociation of macroglobulins; and it chelates, or forms complexes with metals. D-Penicillamine has been used in the treatment of rheumatic diseases (rheumatoid arthritis, scleroderma),420,421 in Wilson’s disease to reduce tissue concentrations of copper, in lead and mercury poisoning, and in patients with cystinuria. D-Penicillamine is absorbed orally, with peak blood levels achieved by approximately 1 to 4 hours. Absorption of the drug is significantly diminished by food, antacids, and iron. Oxidation of D-penicillamine in the presence of transition metals such as copper results in the formation of poorly absorbed disulfides. The D-penicillamine–albumin disulfide bond is quantitatively the most important during chronic therapy, because the drug tends to accumulate in this form and only slowly dissociates from albumin.422 The disulfide metabolites are excreted in urine and stool. Little unmetabolized drug can be detected in the blood 2 days after a dose. D-Penicillamine is usually started at a low dose and increased as tolerated according to a schedule such as the one presented in Table 5–13. The mechanisms whereby D-penicillamine and its metabolites influence inflammation in chronic arthritis or scleroderma are uncertain.423 D-Penicillamine–induced reduction of sulfhydryl groups on macrophages, and possibly other cells, may influence their function. Oxidation of D-penicillamine results in the production of reactive oxygen species that inhibit the reactivity of T cells. D-Penicillamine also directly inhibits myeloperoxidase, thereby reducing the production of hypochlorite, which is toxic to tissue.424 By complexing with trace metals such as copper, D-penicillamine may interfere with the action of superoxide dismutase and may thereby influence the production and scavenging of oxygen-derived free radi-
C
D RU G T H E R A P Y
CH
COOH
SH NH2
■ Figure 5–13 Structure of penicillamine.
101
Guidelines for Use of D-Penicillamine
Dose Initially 5 mg/kg/day (maximum, 250 mg/day) for 3 mo; then 10 mg/kg/day for 3 mo; then 15 mg/kg/day (maximum, 1 g/day) for 1–3 yr Clinical Monitoring Clinical evaluation every month: dermatitis, pruritus, bruising Laboratory Monitoring CBC with differential WBCC, platelet count, and urinalysis every 2 wk until dose is stable, then monthly Discontinue drug if WBCC <3500/mm3 (<3.5 × 109/L), platelet count <100,000/mm3 (<100 × 109/L), persistent proteinuria (>0.5 g/24 hr), or hematuria CBC, complete blood count; WBCC, white blood cell count. Copyright © 1998 John Wiley & Sons, Inc. Reprinted with permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
cals.425,426 It also inhibits carboxypeptidase and angiotensin II converting enzymes, which are involved in the kinin pathways,427 although the concentrations required to achieve this effect considerably exceed those achieved in vivo. It may interfere with immune complex formation428,429 and may block the replication of viral RNA.419,430 The toxicity of D-penicillamine is usually similar to that of gold,431–437 and monitoring should be carried out in the same manner as for gold toxicity. However, autoimmune diseases, including SLE, polymyositis, Goodpasture’s syndrome, and myasthenia gravis, may develop after D-penicillamine therapy.438–440 A child who has a toxic reaction to a gold compound may develop toxicity to D-penicillamine, in many cases even with the same manifestations. In adults, at least, this predisposition may be associated with HLA-DR3 (DW33) and the C4 null allele.398,441
Other Disease-Modifying Drugs Colchicine The primary use of colchicine (Fig. 5–14) in pediatrics is for treatment of familial Mediterranean fever (FMF), wherein it has been shown to reduce not only the frequency of attacks but also the development of amyloidosis. Colchicine is also occasionally used for recurrent aphthous stomatitis, Behçet’s disease, and cutaneous vasculitis. Peak plasma levels are reached 1 to 3 hours after oral administration. The drug is metabolized extensively in the liver by the cytochrome P-450 system.442 Its halflife after oral administration is 9 ± 4 hours.443 Its action is thought to depend on binding of two of its rings to
CH3O NHCOCH3
CH3O
CH3 CH3
AND
CH3O O OCH3
■ Figure 5–14 Structure of colchicine.
102
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
cellular microtubules, inhibiting the movement of intracellular granules and preventing secretion of various components to the cell exterior.444,445 Interaction with endothelial cells and neutrophils is inhibited by reducing the expression of adhesion molecules on the neutrophil membrane.446 The drug is present in granulocytes to a much greater extent than in lymphocytes or monocytes, perhaps because of reduced activity of the p-glycoprotein efflux pump in these two cell types.444 Drug interactions may occur either at the level of the intestine (reduced absorption447) or through the cytochrome P-450 system. Agents that inhibit this system may lead to colchicine toxicity; alternatively, drugs that are metabolized by this system may compete with colchicine, leading to a buildup of both agents. The effect of colchicine on microtubules has raised concern about chromosomal and gonadal aberrations. In a group of patients with FMF receiving long-term treatment with colchicine, there were no differences between patients and controls in lymphocyte mitotic rate, percentage of tetraploidy, or chromosomal breakage.448 The amount of colchicine needed to reduce sperm motility is much greater than that used with standard therapy.449 Azoospermia and oligospermia are more common in patients with Behçet’s disease than in those with FMF, which suggests that genetic makeup or the underlying disease may contribute to reduced sperm production.450 Concerns regarding growth delay in children are unfounded.451,452 The therapeutic dose of colchicine ranges from 0.5 to 2.0 mg/day as needed to prevent or significantly reduce the frequency of FMF attacks. Toxicity is extremely rare with oral administration and is generally limited to the GI tract (nausea, vomiting, abdominal pain, diarrhea). In the case of serious overdose, treatment with colchicine-specific Fab could be considered.453 However, severe toxicity can result in dehydration, multiorgan failure, and a DIC-like syndrome.444
Thalidomide Thalidomide (N-α-phthalimidoglutarimide) has acquired a well-deserved reputation as a major teratogen. However, it has been shown to be effective in a variety of immune-mediated disorders. Its immunosuppressive effects include inhibition of neutrophil chemotaxis,454 decreased monocyte phagocytosis,455 decrease in the ratio of helper T cells to suppressor T cells,456 inhibition of expression of TNF-α and IL-6 messenger RNA (mRNA),457 and inhibition of angiogenesis.458,459 Its structure includes two ring systems (Fig. 5–15). Mean peak plasma concentrations occur 4.39 ± 1.27 hours after a
O C N C O
C C
O
NH
O
■ Figure 5–15 Structure of thalidomide.
200-mg dose.460 It is metabolized primarily by spontaneous hydrolysis and has an elimination half-life of 3 to 7.3 hours. Once-daily or twice-daily administration is appropriate.461 Controlled trials have shown benefit of thalidomide compared with placebo in recurrent aphthous ulcers462 and in the recurrent oral ulceration in adult men with Behçet’s syndrome.463 There is one report of response to thalidomide in an infant with Behçet’s disease.464 In addition, several reviews465,466 have described multiple case series reporting improvement in patients with a variety of disorders, including cutaneous lupus467–469 and graft-versus-host disease.466 Single series or case reports have documented improvement in patients with palmoplantar pustulosis, sarcoidosis, rheumatoid arthritis, pyoderma gangrenosum, erythema multiforme, Weber-Christian disease, pemphigoid, Crohn’s disease (including two children),470,471 ulcerative colitis, rheumatoid arthritis,472 spondyloarthropathies,473 and adult-onset Still’s disease.474 Recently, Lehman and colleagues reported on two children with systemic-onset JRA resistant to various treatments (including etanercept) who responded well to thalidomide.475 In addition to embryopathy (which can occur with as small a dose as 100 mg given between 34 and 50 days of gestation), the major side effects of thalidomide include peripheral neuropathy and drowsiness. The neuropathy is predominantly sensory and manifests as painful paresthesias in a glove-and-stocking distribution.476 The neuropathy can progress despite discontinuation of thalidomide and may or may not be dose related; it has been reported in 1% to 70% of patients. Even after discontinuation of the drug, recovery may be delayed for several years. Baseline and routine follow-up electrophysiologic testing should be performed, and the dose should be reduced or discontinued on detection of abnormalities.477 A variety of other neurologic effects, including carpal tunnel syndrome, muscle weakness and cramps, and signs of pyramidal tract involvement, may occur. Endocrine effects include hypothyroidism, hypoglycemia, and stimulation of adrenocorticotropic hormone (ACTH) and prolactin production or secretion.478 The dose of thalidomide varies between 100 and 400 mg/day. Doses of 2.5 to 4 mg/kg/day have been suggested for children with SLE or systemic-onset JRA.475,479,480 Birth control must be practiced. Women who are prescribed thalidomide should sign a consent form indicating knowledge of potential birth defects and agreeing to adequate methods of birth control. Excellent control is maintained in a postmarketing surveillance program and a restricted distribution program, the System for Thalidomide Education and Prescribing Safety program (STEPS), monitored by Boston University, Celgene Corporation, and the U.S. Food and Drug Administration.481
Glucocorticoid Drugs Glucocorticoid drugs are the most potent anti-inflammatory agents in the treatment of rheumatic diseases.482 Reports of their use in children with rheumatic diseases, especially rheumatic fever, JRA, and SLE, began to appear
C H A P T E R
5 PHARMACOLOGY
in the 1950s and 1960s.483–486 Specific aspects of therapy are discussed in the chapters on individual diseases and in reviews.487–493 This chapter describes more general aspects, such as the pharmacology, physiology, and mechanism of action of glucocorticoids, as well as the indications and contraindications for systemic and local glucocorticoid therapy and their associated adverse effects.
CH2OH
CH2OH C
O OH
CH3
O
CH3
O
O PREDNISONE CH2OH
CH2OH
Pharmacology
Physiologic and Pharmacologic Effects Glucocorticoids are unique among the pharmacologic agents given to treat rheumatic diseases in that they are synthetic analogues of chemicals produced by the body and thus have a physiologic, as well as a pharmacologic,
C CH3
HO
C
O OH
CH3
HO CH3
CH3
CH3
O
O CH3
DEXAMETHASONE
METHYLPREDNISOLONE CH2OH C CH3
HO
O OH
CH3
O HYDROCORTISONE
■ Figure 5–16 Structure of the glucocorticoids.
role. Glucocorticoids enter cells passively and bind to two distinct cytosolic receptors, mineralocorticoid (type I) and glucocorticoid (type II) receptors (GRs). Type I receptors have highest affinity for aldosterone and are found on epithelial cells of the kidney, colon, and salivary glands, and on nonepithelial cells in the brain and heart. Activation of mineralocorticoid receptors induces activity of epithelial sodium channels, leading to sodium retention and hypertension. Type II receptors have highest affinity for dexamethasone and are present in virtually all cells, including those of the immune system. The receptors are present in the cytoplasm and consist of a
Equivalent Dose† (mg)
Relative Anti-inflammatory Potency
Relative Sodium-retaining Potency
20 6
1 4
1 1
5 5 4
4 4 5
0.8 0.8 0.5
0.75
25
0
Short-acting Hydrocortisone Deflazacort Intermediate-acting Prednisone Prednisolone Methylprednisolone Long-acting Dexamethasone
O OH
F
TABLE 5–14 Relative Doses and Equivalent Potencies of Glucocorticoids (Compared with Hydrocortisone) Glucocorticoid*
O OH
CH3
PREDNISOLONE
The glucocorticoid drugs are modeled on the principal naturally occurring glucocorticoid, hydrocortisone (cortisol). They are 21-carbon molecules that in active form have a hydroxyl group at C11. Synthetic preparations such as prednisone and cortisone must be metabolized to the active forms (prednisolone and hydrocortisone, respectively) (Fig. 5–16). Glucocorticoids that are used topically (e.g., dexamethasone) or given by intra-articular injection (e.g., triamcinolone hexacetonide) have a hydroxyl group at C11 and therefore are already in active form.494 The different relative potencies and durations of biologic action of the various synthetic analogues are outlined in Table 5–14. Orally administered glucocorticoids (prednisone, prednisolone) are rapidly absorbed. Prednisone is converted to prednisolone in the liver and reaches a peak plasma concentration within 2 hours. Hydrocortisone and prednisolone bind to the serum proteins transcortin (high affinity) and albumin (low affinity). Methylprednisolone and dexamethasone are bound primarily to albumin.494 Prednisolone has a large volume of distribution; about two thirds is taken up by muscle. After metabolism in the liver, excretion occurs principally by way of the bile.
103
D RU G T H E R A P Y
C CH3
HO
AND
*Biologic half-life: short-acting, 8–12 hr (deflazacort, ~1.5 hr); intermediate-acting, 12–36 hr; long-acting, 36–72 hr. † Oral or intravenous administration only. Adapted from Goodman LS, Gilman A (eds): Goodman and Gilman’s The Pharmacological Basis of Therapeutics, 8th ed. New York, Pergamon Press, 1990. Reproduced with permission of The McGraw-Hill Companies.
104
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
hormone-binding portion, a DNA-binding portion, and an immunogenic region. Binding of the hormone to the receptor causes translocation of the complex to the nucleus, where the DNA-binding portion binds to glucocorticoid-responsive elements (GREs) of the DNA in the promoter or enhancer region of the responsive genes. This induces mRNA transcription of specific genes that encode for proteins of importance in the inflammatory and immune responses, such as phospholipase A2 inhibitory protein. The effect is reflected indirectly in decreased prostaglandin production.495,496 Binding to negative GREs may repress gene transcription. This mechanism of glucocorticoid effect, mediated via binding to a cytosolic receptor, reflects traditional understanding of the therapeutic effects of glucocorticoids and can be categorized as “genomic” action. GRs may also interact via protein–protein interaction with other transcription factors (e.g., NF-κB), preventing activation of transcription.497 Two forms of GR have been described, GRα and GRβ. GRβ does not bind glucocorticoids and may play a role in the glucocorticoid resistance of some diseases.498 The therapeutic effects of glucocorticoids occur in a modular fashion via both genomic and nongenomic mechanisms (Table 5–15).499 This modular hypothesis postulates the following steps. ■
■
Module 1: At very low dosages of glucocorticoids, genomic effects occur (module 1). These include the classic receptor-mediated actions that result in increased transcription of certain genes (e.g., those coding for lipocortin) and decreased transcription of others (e.g., those coding for various cytokines), resulting in net anti-inflammatory and immunosuppressive effects. These genomic effects start at least 30 minutes after glucocorticoid administration and result from binding to cytosolic receptors. Module 2: As the dosage is increased up to approximately 200 to 300 mg of prednisone equivalent per day, specific nongenomic effects occur due to a greater occupation of receptors. However, it is postulated that a further increase in dosage may affect pharmacodynamics (e.g., receptor off-loading and reoccupancy), receptor synthesis, and receptor expression and may bring additional therapeutic benefit via other mechanisms. In contrast with genomic actions, these receptor-mediated actions occur within minutes after glucocorticoid administration. The clinical correlates of these effects may include the negative feedback of ACTH production, behavioral changes, cardiovascular effects, and programmed cell death (apoptosis).
TABLE 5–15 Level of Therapeutic Effect (see text) Module 1 Module 2 Module 3
■
Module 3: The assumed additional therapeutic effects of higher dosages could be obtained predominantly via nonspecific nongenomic mechanisms, mediated by membrane-bound receptors, initiated by even more rapid effects (within seconds) through physicochemical interactions with cellular membranes.499 The antianaphylactic actions of glucocorticoids may be explained by this mechanism.
This hypothesis provides a modular system of increasing therapeutic effect with increasing dosage, occurring through recruitment of a number of nongenomic actions, with increasingly more rapid onset of effect than the classic genomic actions of glucocorticoids. The actions of glucocorticoids at physiologic levels are summarized in Table 5–16. Glucocorticoids are essential for normal vascular integrity and responsiveness; they suppress leukocyte migration and immune reactions and stabilize cell membranes.500–511 Glucocorticoids influence protein, carbohydrate, fat, and purine metabolism; electrolyte and water homeostasis; cardiovascular, nervous, and renal function; and bone and muscle integrity.
Carbohydrate, Protein, and Lipid Metabolism Glucocorticoids stimulate the synthesis of glucose, diminish its peripheral use, and promote its storage as glycogen. They increase secretion of insulin by pancreatic islet cells. In the periphery, they mobilize amino acids from tissues that are then diverted in the liver to the production of glucose and glycogen. This catabolic action results in lymphoid and muscular atrophy, osteoporosis, thinning of the skin, and negative nitrogen balance. Glucocorticoids stimulate fat cell differentiation and, in high doses, redistribution of fat in a typical cushingoid distribution. They increase the production of the peroxisome proliferator-activated receptor γ2 (PPAR-γ2), a nuclear hormone receptor important in adipogenesis. Various other effects on lipids have been reported, but most have not been conclusively demonstrated to result from the direct actions of glucocorticoids themselves. There is no consistent alteration in plasma lipids in either hypercorticism or hypocorticism.512
Electrolyte and Water Balance The glucocorticoid analogues used in the treatment of rheumatic diseases have been modified to decrease their
Dose-Effect Relationships of Glucocorticoids Prednisone Equivalent (mol/L) −12
>10 >10−9 >10−4
Mechanisms
Onset of Action
Genomic actions Additional nongenomic, receptor-mediated actions Additional nongenomic, physicochemical actions
After at least 30 min Seconds to 1–2 min Within seconds
From Buttgereit F, Wehling M, Burmester GR: A new hypothesis of modular glucocorticoid actions. Steroid treatment of rheumatic diseases revisited. Arthritis Rheum 41: 761–767, 1998. Copyright © 1998. Reprinted by permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
C H A P T E R
TABLE 5–16 Actions of Glucocorticoids at Physiologic Levels Negative feedback modulation of corticotropin-releasing factor and adrenocorticotropic hormone Maintenance of blood glucose and liver glycogen levels Maintenance of cardiovascular function, blood pressure, and muscle work capacity Excretion of a water load Permissive effects on pressor, lipolytic, and gluconeogenic activities of hormones Protection against moderate stress Adapted from Munck A, Guyre PM: Glucocorticoid physiology and homeostasis in relation to anti-inflammatory actions. In Scheimer RP, Claman HN, Oronsky A (eds): Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. San Diego, Academic Press, 1989.
mineralocorticoid potency. In that manner, increased resorption of sodium ions from the distal renal tubules is moderated, as is increased urinary excretion of potassium and hydrogen ions. Patients on long-term therapy are nonetheless in positive sodium balance, have an increased extracellular fluid volume, and have a tendency toward hypokalemia and alkalosis. However, in practice, these changes are only moderate in severity, reflecting the relatively weak effect of glucocorticoids on electrolyte balance. Glucocorticoids also decrease the absorption of calcium from the intestine and increase its renal excretion, thus producing a negative calcium balance (discussed later).
Anti-Inflammatory and Immunosuppressive Actions Glucocorticoids have both anti-inflammatory and immunosuppressive effects.490–493,500–511 These effects are largely mediated by the inhibition of specific functions of leukocytes, such as the elaboration or the action of a variety of lymphokines.
Anti-inflammatory Actions Steroids inhibit both the early stages of inflammation (e.g., edema, fibrin deposition, capillary dilatation, migration of lymphocytes into inflamed areas, phagocytic activity) and the later manifestations (e.g., proliferation of capillaries and fibroblasts, deposition of collagen).512 Many of these effects are mediated by inhibition of the elaboration of a number of chemokines and cytokines, including the following: ■
■ ■ ■
Arachidonic acid and its metabolites (e.g., prostaglandins, leukotrienes): Glucocorticoids induce synthesis of lipocortins by macrophages and other cells.510,511,513 Lipocortins inhibit the binding of phospholipase A2 to its substrate and thus reduce the generation of arachidonic acid, the substrate for the COX-mediated synthesis of prostaglandins and leukotrienes Platelet-activating factor (PAF) This effect is also mediated by the induction of lipocortin.514 TNF IL-1: A number of inflammatory actions of IL-1 are inhibited, including stimulation of the production of prostaglandin E2 and collagenase, activation of T lymphocytes, stimulation of fibroblast prolifera-
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
105
tion, and enhanced hepatic synthesis of acute phase proteins. In addition, glucocorticoids can inhibit the action of humoral regulators of inflammation such as PAF and macrophage migration inhibition factor (MIF).514 The reader who is interested in the subject of the anti-inflammatory effects of glucocorticoids is referred to the extensive review by Schleimer and colleagues.515
Immunosuppressive Actions The glucocorticoid effects on the immune system are mediated principally through T lymphocytes.503 Acute administration of hydrocortisone produces a 70% decline in circulating lymphocytes. T lymphocytes are affected more than B lymphocytes, and T helper cells more than T suppressor cells. The lymphocytopenia is probably a result of sequestration of cells in the bone marrow rather than cell lysis, although drug-induced apoptotic cell death may also be involved.516 The most pronounced lymphopenia occurs 4 to 8 hours after a single dose of glucocorticoid and disappears by 24 hours. There is also a 90% decline in circulating monocytes within the initial 6 hours. Proliferative T cell responses to antigens (streptodornasestreptokinase), mitogens (concanavalin A), and cell surface antigens (as in the mixed leukocyte reaction) are reduced by glucocorticoids. IL-2 production by T cells in vitro is also reduced.517 Glucocorticoids cause an increase in the numbers of blood neutrophils by increasing the release of cells from the marginated neutrophil pool, prolonging their stay in the circulation, and reducing chemotaxis of neutrophils to sites of inflammation.518 Another effect of the decreased action of phospholipase A2 is reduction of neutrophil chemotaxis, which decreases accumulation of these cells at inflammatory sites.507 However, no consistent effect of glucocorticoids on neutrophil phagocytosis or bacterial killing has been demonstrated.518 Intravenous glucocorticoid causes a fall in circulating IgG but little discernible effect on the serum titer of specific antibodies. However, the protein catabolic effects of long-term administration may have consequences on the humoral immune system. Endothelial secretion of C3 and factor B of the complement cascade is inhibited by glucocorticoid.519
Indications for Systemic Glucocorticoid Therapy When considering the use of glucocorticoids in children with rheumatic diseases, the risk/benefit ratio must be carefully weighed, because these agents are associated with substantial toxicity when used systemically in the long term (Table 5–17). Clinicians must review the specific indications for which glucocorticoids are to be used and the outcomes that will be monitored to measure response and consequently determine the duration of therapy. The overall aim is to limit the dose and duration of steroid therapy to the lowest possible levels while achieving disease control. Administration of a single dose in the morning and use of alternate-day regimens, which have been shown to minimize the suppression of linear growth in children, should be used whenever possible.520
106
C H A P T E R
TABLE 5–17
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Adverse Effects of Glucocorticoid Drugs
Mood and behavioral disturbances Cushing’s syndrome Growth suppression Effects on bone Osteoporosis Avascular necrosis of bone Immunosuppression Lymphopenia and neutrophilia Central nervous system effects Psychosis Mood and behavioral disturbances Cardiovascular system effects Hypertension Dyslipoproteinemia Cataracts and glaucoma Metabolic effects Impaired carbohydrate tolerance Protein wasting Metabolic alkalosis Myopathy
The child who is being treated with these drugs should be under the care of a physician who is experienced in management of the disease and in minimizing the adverse effects of glucocorticoids.521–523
TABLE 5–18
Comparison of Idiopathic and Iatrogenic Cushing’s Syndromes Prominent in Idiopathic Cushing’s Syndrome Hypertension Menstrual irregularities Impotence Hirsutism Acne, striae Purpura, plethora
Equal Frequency Obesity Psychosis Edema
Prominent in Iatrogenic Cushing’s Syndrome Avascular necrosis Cataract, glaucoma Pseudotumor cerebri Pancreatitis Panniculitis
In Cushing’s syndrome, the distribution of fat is predominantly in the subcutaneous tissue of the abdomen and upper back (buffalo hump) and in the face (moon facies). Weight gain reflects both fluid retention and increased caloric intake: Children taking prednisone are often ravenously hungry. Attempts to minimize weight gain by limiting caloric and sodium intake may be useful but difficult. Skin changes, in addition to the characteristic purple striae on the lower abdomen, lower legs,
For example, in JRA the use of systemic glucocorticoids is mainly limited to treating the extra-articular features of systemic-onset disease. These include systemic “toxicity” and fevers unresponsive to NSAID therapy, severe anemia, myocarditis or pericarditis, and the macrophage activation syndrome.80,81 The presence of fever or arthritis alone in systemic-onset JRA is not sufficient indication for systemic glucocorticoid therapy. Lowdose, short-term systemic glucocorticoids may also be indicated in severe forms of polyarticular JRA with significant functional impairment and for chronic uveitis unresponsive to local therapy. High-dose systemic glucocorticoids are used in children with other inflammatory conditions such as JDM, vasculitis, and SLE.
Adverse Effects Two broad categories of adverse effects occur with the therapeutic use of systemic glucocorticoids: Those resulting from prolonged use of large doses and those resulting from withdrawal of therapy. The major manifestation of the latter is acute adrenal insufficiency with too-rapid withdrawal after prolonged therapy (see later discussion). The adverse effects of glucocorticoid excess are many (see Table 5–17). The mechanisms involved in the development of these adverse events have been recently reviewed.497
Cushing’s Syndrome Cushing’s syndrome, a term used originally to identify the effects of idiopathic hypercorticism, may also be induced by prolonged glucocorticoid administration (Table 5–18). It is characterized biochemically by high plasma glucocorticoid levels and suppression of the hypothalamic-pituitary-adrenal axis. The syndrome is characterized clinically by a number of features, including truncal obesity (Fig. 5–17), osteoporosis, thinning of the subcutaneous tissues, and hypertension.
■ Figure 5–17 Eleven-year-old girl with severe systemic-onset juvenile idiopathic arthritis requiring high-dose corticosteroid treatment. Cushingoid features demonstrated include moon facies, truncal obesity, and cutaneous striae.
C H A P T E R
upper arms, and chest, include hirsutism and acne. Hypertension is usually mild but occasionally requires treatment or reduction of the glucocorticoid dose. Osteoporosis is one of the most troublesome consequences of long-term, high-dose glucocorticoid therapy and is further discussed later in this chapter.524 Few side effects occur at the start of therapy; the major manifestations of iatrogenic Cushing’s syndrome and other toxicities are related more directly to the total dose administered than to the length of time that the patient has been taking the drug. Cushingoid effects supervene when the long-term daily dose in a child (25 kg) exceeds approximately 5 mg of prednisone. Cushingoid appearance is an important source of distortion of body image and can affect self-esteem and psychological well-being, particularly in adolescents and young adults. However, with the exception of skin striae, all of the physical features contributing to the cushingoid appearance are reversible after cessation of glucocorticoid therapy.
Growth Suppression Growth suppression is one of the most worrisome long-term adverse effects. It occurs in young children who are receiving prolonged therapy525 in dosages equivalent to 3 mg/day of prednisone and increases with higher doses.526,527 However, there may be substantial interindividual variability in the severity of growth suppression and the minimal dose required to suppress growth.528 The mechanism of glucocorticoidassociated growth suppression in children with arthritis is controversial. Glucocorticoids have been shown to inhibit production of insulin-like growth factor I (somatomedin C).529,530 In addition, the general inhibitory effect of glucocorticoids on cell growth and cell division probably contributes to growth failure.526 Although early studies showed that growth hormone did not always improve growth failure in children with glucocorticoid-induced inhibition of growth,531 more recent reports have shown not only increased height velocity in many patients532 but also catch-up growth.533 Growth suppression may in fact be a consequence of the underlying disease process, as in JRA.534 Evidence suggests that when glucocorticoids are used, growth retardation is more severe in patients with JRA than in those with SLE receiving equivalent doses.535 Furthermore, growth suppression appears to be worse in patients with systemic-onset disease compared with those with polyarticular- or oligoarticular-onset disease.535 Although alternate-day regimens have been shown to minimize this adverse effect,536–538 the usefulness of such regimens for controlling the actual disease remains unclear. Effects on Bone: Osteoporosis and Avascular Necrosis Osteoporosis is another serious consequence of long-term glucocorticoid therapy in children with rheumatic disease. There are multiple other contributing factors, including inadequate dietary intake of calcium and vitamin D, the underlying disease activity (e.g., in children with polyarticularor systemic-onset JRA),539–541 reduced physical activity,542 reduced exposure to sunlight (e.g., in children with JDM or SLE), and low body weight.543
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
107
Glucocorticoids are associated with both a reduction in bone formation and an increase in bone resorption, and it is the reduction in bone formation that appears to be the most important. Reduced bone formation is caused by a direct inhibitory effect on osteoblasts. Glucocorticoids also directly inhibit gut absorption of calcium and cause increased urinary calcium excretion, potentially resulting in secondary hyperparathyroidism and, hence, increased bone resorption544,545; however, this mechanism appears to play a minor role, at least in adults. The extent of bone loss appears to be related to both the dose and the duration of glucocorticoid therapy, although these factors do not necessarily have a consistent relationship with fracture risk. Significant trabecular bone loss occurs with doses of 7.5 mg/day or higher in most adults.546,547 Bone loss appears to occur rapidly within the first 6 to 12 months of therapy and then reaches a plateau.546 Alternate-day glucocorticoid therapy may not be protective.548 In adults, bone loss is predominantly trabecular (e.g., spine and ribs) rather than cortical, whereas in children the osteoporotic effect of glucocorticoids is more generalized. Not all patients exposed to long-term glucocorticoid therapy develop bone loss.549 However, there are no reliable biochemical markers that can be used to predict which glucocorticoid-treated children will undergo significant bone loss.550 Bone densitometry may be used to screen children who are at high risk for osteoporosis. Approaches to the prevention and treatment of glucocorticoid-associated osteoporosis are discussed later. Some of the mechanisms by which glucocorticoids result in bone loss have been explored and are depicted in Figure 5–18. They include effects on the production of local growth factors, reduction of matrix proteins and an increase in the production of enzymes that break down matrix, an increase in apoptosis of both osteoblasts and osteocytes, and an increase in osteoclastogenesis due to decreased production of osteoprotegerin and an increased production of RANK ligand.550a–550c High-dose glucocorticoids have also been associated with avascular necrosis of bone (AVN). Although the exact mechanism is not known,551 a variety of recent observations may help explain this devastating complication. Intramedullary vascular compromise may result from the increased osteocyte apoptosis induced by glucocorticoids. Absence of clearance of these apoptotic osteocytes may result in reduced blood flow and bony ischemia.552 Glucocorticoids induce adipocyte differentiation via the increased production of peroxisome proliferator-activated receptor γ2 (PPAR-γ2), which may result in increased fat in the marrow.552 Finally, glucocorticoids also increase the increased expression of endothelin-I, which may also lead to reduced intramedullary blood flow.553 Many sites can be involved, but the most common and clinically significant location for AVN is the femoral head. This complication is more frequently reported in SLE (in which the underlying disease process can be a contributing factor) and possibly also after highdose intravenous methylprednisolone therapy, although the data to support the latter association are not strong.554,555
108
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
↓ bone formation
• FLUORIDE • ANABOLIC STEROIDS
↓ collagen ↑ collagenase ↑ apoptosis
↓ growth factor
CORTICOSTEROIDS
↓ FSH induced sex hormone production
↓ sex hormones
↓ LH response to LHRH GI calcium absorption • CALCIUM • VITAMIN D
Urinary calcium excretion
↓ OPG
OSTEOPOROSIS • ESTROGENS • TESTOSTERONE
↑ RANK Ligand
• THIAZIDE ↓ calcium
↑ PTH
↑ bone resorption
• CALCITONIN • BISPHOSPHONATES
■ Figure 5–18 Approaches to the prevention and treatment of steroid-induced osteoporosis. FSH, follicle-stimulating hormone; GI, gastrointestinal; LH, luteinizing hormone; LHRH, luteinizing hormone-releasing hormone; OPG, osteoprotegerin; PTH, parathyroid hormone.
Infection and Immunity Glucocorticoids interfere with the ability to resist infection through two main mechanisms: (1) They act as immunosuppressives and unpredictably decrease the patient’s resistance to viral and bacterial infections. (2) They are also anti-inflammatory agents and hence may mask the signs and symptoms of infection. These may include, for example, important signs such as fever or abdominal pain in peritonitis. Susceptibility to infections in general is related to the dose and duration of glucocorticoid administration. However, the minimal amount of systemic steroids and the duration of administration sufficient to cause immunosuppression in an otherwise healthy child are not well defined.217 Additional factors that may affect the overall extent of immunosuppression in steroid-treated children with rheumatic diseases include the effects of the underlying disease and concurrent immunosuppressive therapies. The most profound effect of glucocorticoid administration is on cell-mediated immune reactions, including delayed hypersensitivity and allograft rejection. Patients receiving high doses of glucocorticoid for a prolonged period are prone to infections that are associated with defects of delayed hypersensitivity (e.g., tuberculosis). If possible, the Mantoux test (purified protein derivative [PPD], 5 tuberculin units) should be performed before glucocorticoids are started. If the result is positive at 72 hours, further investigations for tuberculosis should be carried out. The risk of complications of varicella infection must also be considered. The susceptible glucocorticoid-treated child who is exposed to chickenpox should receive zoster immune globulin within 96 hours (for maximum effectiveness, as soon as possible after exposure or ideally within 72 hours) or acyclovir during the infectious illness itself.217
There is little information to guide decisions about the glucocorticoid regimen in varicella-infected children who have been on chronic steroid therapy; it would seem reasonable to try to minimize doses while maintaining disease control during the course of such an infection. If bacterial infection develops in a child treated with glucocorticoids, the dose may be maintained or increased and the best available treatment for the infection vigorously administered. In those patients with Pneumocystis carinii pneumonia (PCP), early adjunctive treatment with glucocorticoids has been demonstrated to have a beneficial effect on the clinical course and outcome, at least in adult patients with acquired immunodeficiency syndrome (AIDS) and moderate to severe PCP.556 Although no controlled trials of glucocorticoids in young children have been performed, most experts would recommend glucocorticoids as part of therapy for children with moderate to severe PCP.217 The impact of glucocorticoids in immunocompromised hosts without AIDS appears to be similar to that in patients with AIDS.557
Hematologic System Glucocorticoids decrease the number of circulating lymphocytes, monocytes, basophils, and eosinophils but increase the number of circulating neutrophils.558 Excess glucocorticoid may also cause polycythemia. Central Nervous System The effect of glucocorticoids on the CNS results from changes in the concentration of plasma glucose, circulatory dynamics, and electrolyte balance. These effects are reflected by changes in mood, behavior, and electroencephalographic studies.559 Pseudotumor cerebri is rare but may occur after rapid reduction of glucocorticoid dose.560 Most glucocorticoid-induced psychoses have an acute onset, are related to high doses, and occur within 96 hours after initiation of medication.561,562 Psychosis is more common in idiopathic Cushing’s syndrome than in
C H A P T E R
iatrogenic disease. Early, there may be euphoria and mania; later in Cushing’s syndrome, depression tends to predominate. Other types of mood and behavioral disturbances, such as anxiety and insomnia, may occur. A prospective cohort study of the adverse effects of high-dose intermittent intravenous glucocorticoids in 213 children with rheumatic diseases found behavioral changes in 21 (10%).563 These abnormalities included altered mood in 14, hyperactivity in 4, sleep disturbance in 3, and psychosis in 2 children. In some cases, CNS effects may be related to the underlying disease (e.g., SLE). Abnormalities of behavior usually disappear after glucocorticoids are withdrawn. However, there are anecdotal reports of depression lasting for several weeks in some children treated with short-term steroids in the form of intraarticular triamcinolone hexacetonide or high-dose intravenous pulse methylprednisolone.
Cardiovascular System The major effect of glucocorticoids on the cardiovascular system is mediated by their influence on the regulation of renal sodium excretion, sometimes leading to hypertension. However, this complication is uncommon in children with JRA who do not have underlying renal disease. The actual mechanism of glucocorticoid-induced hypertension is not fully explained. Although sodium retention plays a role, additional factors, such as increased plasma renin activity or antidiuretic hormone, may be involved.512 Glucocorticoids also exert an important effect on capillaries, arterioles, and myocardium.512 These influences are relevant in acute relative steroid deficiency when patients treated with long-term glucocorticoids are subject to physiologic stress (see “Preventing Acute Adrenal Insufficiency [Addisonian Crisis]”). Other important possible long-term effects of chronic glucocorticoid administration include hyperlipemia and accelerated coronary atherosclerosis.564 Patients with SLE, who often are treated with large doses of glucocorticoids for prolonged periods, are at increased risk for dyslipoproteinemia and coronary artery disease (CAD) after about 10 years of disease. Although the pathogenesis of CAD in these patients is multifactorial, some studies have suggested that a long duration of glucocorticoid therapy may be an important risk factor.565 A cross-sectional study of 40 children with SLE did not find steroids to be an independent risk factor for CAD.566 These children had a median age of 15.9 years and median disease duration of 1.4 years at the time of study. In contrast to adults, children with abnormalities in coronary perfusion tended to have shorter median duration of prednisone use as well as lower cumulative dose of steroids and fewer intravenous pulsed doses of steroids. Although the study was small and these were only observed trends and not statistically significant differences, they raise the possibility that CAD may be more a manifestation of the underlying disease process than of the glucocorticoid therapy used to treat it.
Cataracts and Glaucoma Subcapsular cataracts560,567,568 are more common with glucocorticoid therapy than in idiopathic Cushing’s syndrome. The risk of cataract development becomes significant when a dose of prednisone equal to or greater than 9 mg/m2/day has been maintained for longer than 1 year. Most children who have been treated
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
109
with doses of glucocorticoid equivalent to 20 mg/day of prednisone for 4 years or longer develop cataracts.560 These cataracts often do not progress, are functionally benign, and rarely affect vision. Children on long-term glucocorticoid therapy should also be monitored for glaucoma, especially if they have a history of uveitis.
Muscle Disease The muscle wasting that results from highdose glucocorticoid administration is associated with atrophy of muscle fibers, especially type IIB fibers. Steroid myopathy can complicate the clinical assessment of a patient with SLE or JDM (see Chapters 16 and 18). Myopathy induced by glucocorticoids usually affects proximal muscles, is seldom painful, and is usually associated with normal serum levels of muscle enzymes and an electromyogram suggestive of myopathy. However, a muscle biopsy may be needed to differentiate between steroid myopathy and active myositis. Glucocorticoidinduced hypokalemia may also lead to muscular weakness and fatigue. Recovery from steroid myopathy may be slow and incomplete.569 Other Side Effects Glucose intolerance and glycosuria may occur after prolonged exposure to large doses of glucocorticoids, particularly if there is a genetic predisposition to diabetes.570 Such intolerance can usually be managed with diet or insulin; such side effects should not be important to the decision to continue glucocorticoid therapy or to initiate it in diabetic patients who need it. The role of glucocorticoids in peptic ulceration is controversial. Although some have suggested an increased risk, current evidence does not support a definitive association between peptic ulceration and glucocorticoid therapy independent of any other factors such as NSAID therapy or concomitant illness.571
Minimizing Toxicity The deleterious effects of glucocorticoids can be minimized by choosing a drug with a relatively short half-life (see Table 5–14).572 Prednisone is the drug most often given for oral therapy. Its enhanced glucocorticoid and minimal mineralocorticoid actions give it the lowest risk/benefit ratio of any of the analogues in general use.573 Adherence to the use of a single synthetic analogue simplifies communications with the patient, parents, and medical personnel and lessens the risk of an error in dose. The GR-DNA interaction induces genes that code for antiinflammatory proteins (transactivation). Genes that code for pro-inflammatory proteins are regulated by transcription factors such as NF-κB (transrepression). In this situation, the GR operates via protein interactions, inhibits the activity of the transcription factors, and thereby represses the expression of the pro-inflammatory proteins. Many glucocorticoid side effects appear to be mediated via binding to DNA elements, rather than protein elements. Therefore, if synthetic glucocorticoids could be designed to operate via the latter rather than the former pathway, the result would be maintenance of anti-inflammatory properties with reduction in toxicity. Work is in progress to develop such agents.497
Deflazacort, an oxazoline derivative of prednisone with anti-inflammatory and immunosuppressive activity,
110
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
may have a bone-sparing effect compared with prednisone.574–576 However, there are at least two case reports of children who developed symptomatic vertebral collapse while receiving deflazacort for vasculitis and a lupus-like syndrome.577 Therefore, the long-term efficacy and safety of deflazacort in children with rheumatic diseases need further study before this drug can be recommended for general use. Both the anti-inflammatory effect and the toxicity of glucocorticoids increase with larger doses and more frequent administration (Table 5–19). Four-times-daily administration is more effective than twice-daily administration of the same total dose. Daily administration is more effective than administration of the same total dose every other day.536,576,578,579 Glucocorticoid administration should be as infrequent as is consistent with achieving disease control. Short-acting glucocorticoids given in the morning (on waking) have less capacity to suppress the pituitary than do those given later in the day (which suppress the normal surge of ACTH that occurs during sleep).512 Reduction in glucocorticoid dose must be gradual and should be individualized for the child and the disease. At high doses (e.g., 60 mg/day), reductions of 10 mg are usually well tolerated; at lower doses (e.g., 10 mg/day), reductions of only 1 or 2 mg may be necessary. The dose can be tapered on every second day so that an alternate-day regimen is established as early as possible. Glucocorticoid dose tapering is often fraught with difficulty because of the adaptation of the patient’s metabolism to chronic steroid excess.580,581 Steroid pseudorheumatism may result from a rapid dose decrease in some children.582 Signs and symptoms include increased stiffness and pain in the joints, malaise, fever, and irritability. Pseudotumor cerebri may occur under similar conditions.560 It is characterized by headache, vomiting, and papilledema. It must be differentiated from other causes of increased intracranial pressure. These withdrawal effects gradually resolve over 1 or 2 weeks and are minimized if each decrement in daily prednisone is 1 mg or less per week (at the lower dose levels). A number of approaches for the prevention and treatment of corticosteroid-associated osteoporosis have been studied in adults (see Fig. 5–18), and several guidelines have addressed these issues.583–585 Vitamin D and its analogues, calcitonin, and various bisphosphonates have been used. Calcitriol (vitamin D3) or cholecalciferol (vitamin D), with or without calcitonin, was shown to prevent bone loss from the lumbar spine better than calcium alone in several randomized controlled clinical trials of adult patients starting long-term glucocorticoid therapy.586,587 A
1998 meta-analysis also concluded that treatment with calcium and vitamin D in adult patients receiving glucocorticoids effectively retards lumbar and forearm bone loss.588 The reported adverse effects included mainly constipation (calcium) and hypercalcemia and hypercalciuria (calcitriol), although these may be less frequent with physiologic doses. However, the clinical significance of these findings needs interpretation in light of the fact that none of the studies was able to demonstrate any significant decrease in fracture incidence. This is probably a result of the lack of power for detection of infrequent events in these relatively small studies. Furthermore, because none of the controlled studies included children with rheumatic diseases, the generalizability of results to this group of patients also requires caution. However, several open studies suggest that some children with rheumatic disease receiving glucocorticoids may also benefit from calcium and vitamin D supplementation.589–592 The bisphosphonates have also been studied as a potential treatment for glucocorticoid-induced osteoporosis. Etidronate, pamidronate, alendronate, and risedronate have been shown in randomized controlled trials to increase lumbar spine bone mineral density in adult patients receiving long-term glucocorticoids for a variety of diseases.593–597 However, once again, these trials did not include children and did not demonstrate any significant reduction in fracture incidence, which is the most clinically relevant outcome. Bisphosphonates have been studied in children with osteogenesis imperfecta and appear to be of benefit in reducing bone resorption, increasing bone density, and reducing the chronic bone pain associated with this condition.598–600 In addition, they have been found to be useful and safe in open-label studies of children with idiopathic juvenile osteoporosis601 and osteoporosis associated with connective tissue diseases or induced by glucorticoids.602 In summary, although much is known about the mechanisms of glucocorticoid-induced osteoporosis, knowledge about how best to prevent and treat this complication, particularly in children, is somewhat limited. Standards of practice in this area are still evolving, but include, at least, treatment with calcium and vitamin D. Prospective trials evaluating prevention and treatment of glucocorticoid-induced osteoporosis in children are also needed.
Preventing Acute Adrenal Insufficiency (Addisonian Crisis) The short-term use of glucocorticoids for days or a few weeks does not lead to adrenal insufficiency on cessation of treatment. However, prolonged therapy may lead to
TABLE 5–19 Systemic Administration of Glucocorticoid Drugs Schedule
Advantages
Disadvantages
Divided daily doses Single daily dose Alternate-day dose IV pulse therapy
Better disease control Good disease control; fewer side effects Fewer side effects Less long-term toxicity
More side effects May not control severe disease Less disease control Acute toxicities
C H A P T E R
suppression of pituitary-adrenal function that can be slow in returning to normal. This is potentially the most serious and life-threatening adverse effect associated with glucocorticoid therapy. The magnitude of the effect on adrenal function of relatively low amounts of glucocorticoid, especially if given over prolonged periods, is often underestimated (Table 5–20). However, the actual doses and duration of therapy that are associated with this suppression and the length of the recovery period after cessation of therapy are not well defined.603,604 Furthermore, some evidence suggests that a component of adrenal insufficiency may be a result of the underlying inflammatory process in some patients.605 If not recognized, suppression of the hypothalamic-pituitary-adrenal axis as a consequence of chronic glucocorticoid administration places the child at risk for vascular collapse, adrenal crisis, and death in situations that demand increased availability of cortisol.606 Under conditions of stress (serious infection, trauma, surgery), all children who may be at risk for hypothalamic-pituitary-adrenal axis suppression require additional glucocorticoids. Glucocorticoid supplementation should be prescribed before surgery in any patient who has received significant amounts of glucocorticoids at any time during the preceding 12 months (possibly as long as 36 months).606 For an elective procedure, a “steroid prep” consists of dexamethasone (0.05 to 0.15 mg/kg/24 hours) before the procedure in four divided intramuscular doses given 6 hours apart, and hydrocortisone (1.5 to 4.0 mg/kg/ 24 hours) as a continuous intravenous infusion beginning at the time of surgery and continuing for 24 hours postoperatively or until the child has recovered or is able to take prednisone again by mouth. This regimen is based on requirements for hydrocortisone during stress. Hydrocortisone (0.36 mg/kg/24 hours) is needed for physiologic maintenance. During maximal stress, 1.5 to 4.0 mg/kg/24 hours is required. Dexamethasone sodium phosphate (0.75 mg has a mineralocorticoid effect approximately equivalent to 20 mg hydrocortisone) is given preoperatively because of its long half-life; hydrocortisone is used during the procedure because of its immediate biologic availability. These recommended doses should be modified according to the magnitude of the stress and the severity and duration of suppression of the hypothalamic-pituitary-adrenal axis by exogenous steroid administration. The requirement of increased amounts of glucocorticoid during acute stress should be explained to the parents. In addition, each child should carry a card or wear a necklace or bracelet indicating that glucocorticoid medication is being taken. Such a warning can be of great
TABLE 5–20
Suppression of the Hypothalamic-PituitaryAdrenal Axis by Glucocorticoid Drugs In a 70-kg (1.73 m2) adult, prednisone in a daily dose of ≤5 mg given in the morning Does not suppress ≤5 mg given at night May suppress 7.5–15 mg for 1 mo Causes variable suppression >15 mg for 1 wk or longer Causes suppression
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
111
value to emergency medical teams if the patient is involved in an accident.
High-Dose Intravenous Glucocorticoid Therapy Intravenous glucocorticoid “pulse” therapy is sometimes used to treat the more severe, acute, systemic connective tissue diseases such as SLE, JDM, and vasculitis.607–609 It has also been used to treat the refractory systemic features of systemic-onset JRA.610–613 The rationale of this approach is to achieve an immediate, profound antiinflammatory effect and to minimize toxicity related to long-term continuous therapy in moderate to high daily doses. The main benefit appears to be rapid clinical improvement that lasts for about 3 weeks after a single bolus. This may be useful in the context of concurrent treatment with a disease-modifying agent, which may take several weeks or months to begin to exert its effect. Although oral pulse regimens have been reported,614 most publications deal with studies in which intravenous pulse therapy has been used. Methylprednisolone has been the drug of choice, given in a dose of 10 to 30 mg/kg per pulse up to a maximum of 1 g, administered according to a variety of protocols (Table 5–21): a single administration repeated as clinical circumstances warrant, a pulse each day for 3 to 5 days, or alternate-day pulses for three doses. Intravenous glucocorticoid pulse therapy, although perhaps efficacious in selected circumstances (no controlled trials have been reported in children), may be associated with potentially serious complications (Table 5–22). The most frequently reported short-term adverse effect in children is abnormal behavior in up to 10% of patients.563 These behavioral changes included altered mood, hyperactivity, psychosis, disorientation, and sleep disturbances. Nonbehavioral adverse effects included headache, abdominal complaints, pruritus, vomiting, hives, hypertension,
TABLE 5–21
Suggested Protocol for Administration of Intravenous Methylprednisone Dose Methylprednisolone up to 30 mg/kg (maximum 1 g) Preparation Prepare drug with diluent provided with package Calculated dose is added to 100 mL 5% dextrose in water and infused over 1–3 hr Monitoring Temperature, pulse rate, respiratory rate, blood pressure before beginning infusion Pulse and blood pressure every 15 min for first hour, every 30 min thereafter Slow rate or discontinue infusion, and increase frequency of monitoring, if there are significant changes in blood pressure or pulse rate Side Effects Hypertension or hypotension, tachycardia, blurring of vision, flushing, sweating, metallic taste in mouth
112
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
TABLE 5–22
Potential Acute Toxicities of Intravenous Glucocorticoid Pulse Therapy Cardiac arrhythmia secondary to potassium depletion Hypertension secondary to sodium retention Acute psychosis, convulsions Hyperglycemia with or without ketosis Anaphylaxis Infection Osteonecrosis
bone pain, dizziness, fatigue, lethargy, hypotension, tachycardia, and hyperglycemia.563 Potential longer-term effects, such as influences on bone metabolism and risk of AVN, have not been systematically studied. Although there have been a number of reports of AVN associated with intravenous-pulse methylprednisolone,615,616 a retrospective cohort study of patients with rheumatoid arthritis did not find an increased risk of AVN in those treated with pulse methylprednisolone.617
Intra-articular Steroids Injection of long-acting glucocorticoids directly into inflamed joints has emerged as a major advance in the management of various types of arthritis. Although intraarticular steroid (IAS) therapy has not been studied in the randomized controlled clinical trial setting, there is accumulating evidence for its efficacy and safety in children (i.e., a number of uncontrolled, prospective cohort studies).618–624 However, it is difficult to summarize the results of the available studies because of the variability in the populations studied and in the type, dose, and frequency of steroids used; the lack of control groups; and the variability in the assessment of outcomes, which often were not blinded and did not use standard outcome measures or lengths of follow-up. IAS therapy has been used most often in children with oligoarticular disease; indications for use have included lack of response to NSAIDs given in optimal dosage for 3 months or longer; significant NSAID toxicity; and the presence of joint deformity, growth disturbance, or muscle wasting. These drugs may also have a role as an alternative to NSAIDs in children with oligoarticular disease. In polyarticular disease, multiple IAS injections at one time can be used as a temporizing measure while awaiting response to second-line agents given systemically. IAS may also be useful as an alternative to increasing systemic therapy in children with polyarticular disease who have significant inflammation in only a few joints. Virtually all patients experience rapid resolution of symptoms and signs of joint inflammation within a few days after injection. About two thirds achieve remission for at least 12 months after a single injection.619,620 The duration of response seems to be longer in children with oligoarticular JRA than in those with polyarticular- or systemic-onset disease and in those with other forms of arthritis (e.g., spondyloarthropathies).625,626 Younger patients and those with shorter disease duration achieved longer remissions after IAS injection,618 as did patients with higher mean erythrocyte sedimentation rates in another study.627 Early use of intra-articular corticosteroid injections may result in less leg-length discrepancies in patients with asymmetric pauciarticular JRA.628 Neidel and associates629 reported a 2-year remission rate of 58% after a single injection of 1 mg/kg (maximum 40 mg) of triamcinolone hexacetonide into inflamed
hip joints; responses were better in children with pauciarticular or RF-negative polyarticular-onset disease (65%) compared to those with RF-positive polyarticular- or systemic-onset JRA.
Type of Steroid, Dosage, and Frequency of Injection A variety of preparations are available for intra-articular injection. The most frequently studied agents in children are the least soluble and hence longest-acting forms of injectable steroid: triamcinolone hexacetonide (THA)618,619,621and triamcinolone acetonide.620 These agents are completely absorbed from the site of injection over a period of 2 to 3 weeks. Because of its lower solubility, THA is absorbed more slowly than triamcinolone acetonide, thus maintaining synovial levels for a longer period and creating lower systemic glucocorticoid levels.630 THA is preferred by most pediatric rheumatologists. In a recent comparative study in patients with oligoarticular JIA, THA was found to be more effective than triamcinolone acetonide at equivalent doses.631 Intra-articular THA is superior to betamethasone632 and methylprednisolone.633 The dose of THA used in clinical studies has varied: Some data indicate that higher doses (about 1 mg/kg) may be associated with a better response.618 Although there are no hard and fast rules regarding the choice of steroid, the dosage and frequency of injection have been outlined. In general, children who weigh less than 20 kg receive 20 mg THA in large joints. Those weighing more than 20 kg receive 30 to 40 mg THA in the hips, knees, and shoulders and 10 to 20 mg in the ankles and elbows. In smaller joints such as the wrist, midtarsal, and subtalar joints, 10 mg THA is used. For injections into tendon sheaths and small joints of the hands and feet, 0.25 to 0.50 mL of a combination of methylprednisolone acetate mixed 1:1 with preservative-free 1% lidocaine (Xylocaine) is recommended. The shorter-acting steroid is associated with less risk of damage to tendon sheaths or of local soft tissue atrophy caused by leakage of steroid from the smallervolume joints. Repeated injections into the same joint are not performed more than three times per year, although there are few data on which to base this recommendation. There are also no controlled studies in children examining whether postinjection rest has a role. Although full immobilization of the injected joint is common practice in some clinics, the author’s recommendation is to limit ambulation for the first 24 hours and to avoid high-impact physical activity in the 24 to 72 hours after a joint injection. Adverse Effects Despite initial reservations about the safety of IAS therapy in children, clinical studies indicate an overall favorable adverse-effect profile. Iatrogenic septic arthritis is always a potential risk but has never been reported in children. It occurs very rarely in adults and can be avoided with aseptic precautions.633 Transient crystal synovitis occurs in a small proportion of patients.620 It is very similar to gouty arthritis and is selflimited, with resolution of symptoms within 3 to 5 days in most cases without any intervention.622 The most frequent adverse effects are atrophic skin changes at the site of injection, particularly of small joints such as wrists and ankles in young children, and asymptomatic calcifica-
C H A P T E R
tions on radiographs in joints after multiple injections.625 The frequency of these skin changes ranges from 5.6% of knees injected, to 16% of ankles, 22% of wrists, and 50% of metatarsophalangeal joints.634 The skin changes are attributed to leakage of long-acting steroids into subcutaneous tissues and can be minimized by clearing the needle track with injection of saline or local anesthetic as the needle is withdrawn from the joint. Most skin changes eventually resolve.619,634 Radiographic reviews have demonstrated joint calcifications in 6% to 50% of injected joints.635,636 These are usually asymptomatic, but in one case surgical removal was required because of the size of the calcification.637 One of the main reservations about the use of IAS in young children was based on the theoretic potential for cartilage toxicity. Cartilage damage occurred after intra-articular injection of steroids in the rabbit model but was not reproduced in higher species.633 Clinical magnetic resonance imaging studies in children to assess cartilage integrity up to 13 months after steroid injection of single joints demonstrated no toxic effects on cartilage and no detrimental effects on statural growth.638,639 Some children with multiple steroid injections (more than five per joint) who are monitored for longer periods (6 to 18 years) may have nonspecific abnormalities of cartilage on magnetic resonance imaging.640 The clinical significance of these findings is not clear.
Although the majority of adverse effects associated with IAS injections are local, they are also associated with some systemic effects. Children develop transient suppression of endogenous cortisol production lasting 10 to 30 days after IAS injection.641,642 Younger children may undergo a more prolonged period of suppression. However, the clinical significance of this finding in terms of effects on linear growth or actual risk of adrenal crisis at times of stress is not known. Although these complications have not been reported after single injections, whether there is any risk after multiple injections, particularly in younger children, needs further study. The systemic absorption of steroid may also be associated with altered salicylate kinetics, resulting in a transient fall in serum salicylate levels. Diabetic children may require a temporary increase in insulin requirements.
Cytotoxic Agents Cytotoxic drugs prevent cell division or cause cell death. They act primarily on rapidly dividing cells such as those of the immune system, particularly T lymphocytes, and are therefore immunosuppressive. Cytotoxic drugs have both immediate anti-inflammatory actions and delayed immunosuppressive effects. Pharmacologic actions are usually considered to be either specific or not specific for the cell cycle phase. The cell cycle consists of the G1 presynthetic phase, the S phase (synthesis of DNA), the G2 resting (or postsynthetic) phase, and mitosis. 6-Mercaptopurine and azathioprine inhibit biosynthesis of purine and nucleotide interconversions and act during the G1 and S phases in proliferating cells. Mycophenolate mofetil (MMF) reduces the pool of guanine nucleotide, thus interfering with purine biosynthesis; it also acts in the G1 and S phases. Alkylating agents cross-link DNA and act during all phases of the cell cycle,
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
113
whether or not a cell is replicating. These agents are maximally effective in inhibiting immunologic responses when their administration coincides with the period of proliferation of the specific immunologically competent cells. Although cytotoxic drugs have been used to treat children who are seriously ill with rheumatic diseases after other modes of therapy have proved ineffective, there have been no adequately controlled trials in such patients. In most instances, the effects of these drugs are delayed; therefore, they have proved more valuable in moderate- to long-term therapy than in an acute crisis. The potential toxicity of these drugs is substantial. Although extensive experience has been accumulating with the use of these agents, each child’s illness must be considered thoroughly before drugs from this class are recommended. These agents are not approved for unrestricted use in children with rheumatic diseases and should be used only by physicians who are familiar with their administration, toxicity, and expected benefits. Occasionally, a cytotoxic agent is used for its steroid-sparing effect.
Azathioprine Azathioprine (Fig. 5–19), a purine analogue, is inactive until it is metabolized to 6-mercaptopurine (6-MP) by the liver and erythrocytes.643 Hypoxanthine phosphoribosyl transferase metabolizes 6-MP to 6-thioinosinic acid, which suppresses the synthesis of adenine and guanine, thereby interfering with DNA synthesis. Furthermore, 6-inosinic acid inhibits phosphoribosyl pyrophosphate conversion in purine nucleotide synthesis, conversion of inosinic acid to xanthylic acid by purine nucleoside phosphorylase, and incorporation of nucleotide triphosphates into DNA. Azathioprine suppresses cell-mediated immune functions and inhibits monocyte functions.644–647 These immunosuppressive effects are related primarily to inhibition of T cell growth during the S phase of cell division. A measurable decrease in antibody synthesis occurs with long-term administration; occasionally, a decrease in serum antibody concentration occurs. Approximately 50% of the drug is absorbed after oral administration, of which one third is protein bound.643 The plasma half-life is approximately 75 minutes. The kidneys are the major route of excretion. Azathioprine crosses the placenta. However, because the fetal liver lacks the ability to convert azathioprine to its active metabolites, the fetus is generally protected from adverse effects, although rare cases of fetal bone marrow suppression have been reported.648 It is best to avoid using azathioprine during pregnancy, although this is not always possible. Azathioprine does appear in breast milk, and nursing is not recommended.649
N
S N
N N
O2N
CH3
N
N H
■ Figure 5–19 Structure of azathioprine.
114
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
Toxicity to the GI tract (oral ulcers, nausea, vomiting, diarrhea, epigastric pain) is common.650 Toxicity to the liver (mild elevation of serum concentrations of liver enzymes, cholestatic jaundice), lung (interstitial pneumonitis), pancreas (pancreatitis), or skin (maculopapular rash) is uncommonly associated with azathioprine therapy. Dose-related toxicity to the bone marrow results in leukopenia and, less commonly, thrombocytopenia and anemia. An idiosyncratic arrest of granulocyte maturation that occurs shortly after initiation of therapy has been described and results from reduced activity of the enzyme thiopurine methyltransferase (TPMT).651,652 TPMT is responsible for inactivating 6-MP. There is genetic polymorphism for TMPT, and at least eight alleles have been identified. The majority of the population has high enzymatic activity; approximately 0.03% of the population is deficient.653 These latter patients may rapidly develop severe leukopenia on exposure to azathioprine. Lower levels of enzymatic activity have been observed in AfricanAmericans compared with Caucasian-Americans.654 Testing of TMPT levels is now available and should be considered for patients who develop severe leukopenia while taking azathioprine. The bone marrow effects of azathioprine may be increased by concomitant use of trimethoprim.655 Although the risk of malignancy theoretically increases in patients treated with azathioprine, the long-term data are not conclusive. Data are insufficient with respect to childhood rheumatic diseases, but in adults with rheumatoid arthritis treated with azathioprine, the risk of malignancy did not appear to be greater than that in similar patients who did not receive azathioprine.656–658 The use of azathioprine has been reported anecdotally in many pediatric rheumatic diseases and in series of patients with JRA or SLE.659,660 Starting doses should be 1 to 1.5 mg/kg/day, increasing as needed and as tolerated to 2 to 2.5 mg/kg/day (Table 5–23).
Mycophenolate Mofetil MMF, an agent initially used with great success in patients with solid organ transplants, has been found to be effective
TABLE 5–23
Guidelines for Use of Azathioprine
Dose 0.5–2.5 mg/kg/day in a single dose (taken with food) for 1 yr or longer
in a variety of autoimmune diseases (Fig. 5–20). Mycophenolic acid (MPA), a fermentation product of Penicillium stoloniferum, is the active immunosuppressant species, and MMF was developed to increase the oral bioavailability of MPA. Its major effect is on T and B lymphocytes, for which it is relatively selective. Action of MPA is mediated through noncompetitive binding to inosine monophosphate dehydrogenase, an enzyme critical for de novo synthesis of guanine nucleotide, a pathway on which T and B lymphocytes are primarily dependent. In vitro MPA inhibits T- and B-cell mitogen proliferation,661–663 antigen-specific antibody response of memory B cells,664,665 suppression of the humoral immune response,666 and attachment of monocytes to endothelial cells.667 MPA enhances vascular cell adhesion molecules induced by TNF-α and E-selectin surface expression on endothelial cells.668 It does not inhibit IL-2 production, IL2 receptor expression, phagocytic activity, or secretion of type 1 helper T cell (Th1) cytokines. Some of the improvement seen in renal disease in MRL/lpr mice may be associated with inhibition of renal nitric oxide production.669,670 Because its effect appears to be at a late stage of T cell activation, MMF can be used safely with cyclosporine and tacrolimus. MMF is rapidly absorbed after oral administration and is hydrolyzed in the liver to the biologically active MPA. MPA is 8% albumin bound, and the activity of the drug results from the unbound MPA. Peak plasma levels occur 1 to 3 hours after a single dose, with a second peak at 6 to 12 hours as a result of enterohepatic circulation (Table 5–24). The oral bioavailability is approximately 94%. The elimination half-life is approximately 17 hours after oral administration.671 Because of its extensive binding to albumin, MMF may interact with other albumin-bound drugs. Antacids containing aluminum and magnesium decrease absorption and should not be administered simultaneously. There may be competition for renal tubular secretion with acyclovir. The effective adult dose in solid organ transplantation is 2 g/day in two divided doses. The dose used to prevent solid organ transplant rejection in children has been 46 mg/kg/day.671 Cyclosporine and, to a lesser extent, tacrolimus alter the kinetics of MMF so that higher doses are required in the transplantation setting.672 In children with autoimmune diseases, mean doses reported have included 900 mg/m2/day,673 and 22 mg/kg/day.674 Individual pharmacokinetic profiling is available and can
Clinical Monitoring Clinical evaluation at 1–2 mo and then every 3 months (or more often if disease uncontrolled)
O
OH
CH3 O
Laboratory Monitoring CBC with WBCC, differential, and platelet count weekly until stable dose is achieved, then monthly Hepatic enzymes, BUN, serum creatinine initially and then monthly Discontinue if WBCC <3500/mm3, platelet count <100,000/mm3, or elevated liver enzymes BUN, blood urea nitrogen; CBC, complete blood count; WBCC, white blood cell count.
N O
O OCH3 CH3 Mycophenolic acid portion Morpholino portion
■ Figure 5–20 Structure of mycophenolate mofetil.
O
C H A P T E R
TABLE 5–24
5 PHARMACOLOGY TABLE 5–25
Guidelines for Use of Mycophenolate Mofetil
AND
D RU G T H E R A P Y
115
Guidelines for Use of Daily Cyclophosphamide
Dose
Dose
Starting dose usually 250 mg bid, work up to 1 g bid as a maximum dose (see text for dosing guidelines) At predicted optimal dose, consider measuring trough levels and area under the curve and adjusting dose accordingly
0.5–2.0 mg/kg/day in a single oral dose 0.5–2.0 mg/kg/day as an IV pulse (with ample IV fluids for 24 hr) Encourage fluid intake to minimize risk to bladder Encourage frequent emptying of bladder
Clinical Monitoring
Clinical Monitoring
Clinical evaluation every 1–2 mo and then every 3 months (or more often if disease uncontrolled)
Clinical evaluation every month Laboratory Monitoring
Laboratory Monitoring CBC with WBCC, differential, and platelet count 4–6 wk Discontinue if WBCC <3500/mm3, platelet count <100,000/mm3, falling hemoglobin not related to disease activity CBC, complete blood count; WBCC, white blood cell count.
CBC with WBCC, differential, and platelet count and urinalysis every week until stable dose is achieved, then every month Hepatic enzymes, BUN, serum creatinine initially and then every month Discontinue drug if WBCC <1500/mm3 (1.5 × 109/L), platelet count <100,000/mm3 (100 × 109/L), hematuria BUN, blood urea nitrogen; CBC, complete blood count; WBCC, white blood cell count.
be especially helpful in determining the lowest effective dose in patients experiencing side effects.675,676 Adverse effects of MMF include toxicity on the GI tract, hematologic effects (leukopenia, anemia, thrombocytopenia, pancytopenia), and opportunistic infections. The GI side effects are usually improved by giving the dose three or four times a day instead of twice a day, or by reducing it. Hematologic toxicity usually responds to therapy cessation within 1 week. The incidence of lymphoma has been reported at 0.6%, similar to that for azathioprine.677 Although early reports of MMF use in patients with rheumatoid arthritis showed promise,678 the availability of newer agents seems to have had an effect on further study in this area. However, MMF is being used increasingly in patients with SLE,674,679–684 systemic vasculitis,673,677,685 (although flares may be common686), or inflammatory eye disease.687,688 MMF crosses the placenta and is teratogenic in rats and rabbits, and one case with severe fetal malformations has been reported.689 If immunosuppression is required, a switch to azathioprine is recommended. No human data are available regarding breast feeding at this time
Cyclophosphamide Cyclophosphamide, an alkylating agent, is a nitrogen mustard derivative (Fig. 5–21). It is well absorbed after oral administration and may also be given intravenously (Table 5–25). It is inactive until metabolized, principally in the liver, by the cytochrome P-450 mixed-function oxidase system to inactive intermediates and the active metabolite phosphoramide mustard. Phosphoramide mustard covalently binds to guanine in DNA, destroying the purine ring and thereby preventing cell replication.690,691 Cyclophosphamide potentially acts on all cells, O
CH2CH2CI P
NH
O
N
H2O
including those that are mitotically inactive (G0 interphase) at the time of administration (e.g., memory T cells).692,693 Excretion of the drug is primarily by the kidney, and the dose must be reduced in patients with renal impairment (Table 5–26). Intravenous doses should be lowered by 20% to 30% in patients with severe renal insufficiency.694 Because cyclophosphamide is dialyzable, it is important to delay hemodialysis until at least 12 hours after intravenous administration of the drug.694 Acrolein, the other principal metabolite, is thought to be therapeutically inactive but is responsible for bladder toxicity. The half-life of cyclophosphamide is approximately 7 hours. Cyclophosphamide exerts anti-inflammatory actions by its effects on mononuclear cells and cellular immunity. Alkylating agents cause B- and T-cell lymphopenia. B cells appear to be more sensitive than T cells to the effects of cyclophosphamide.695 It has been suggested that the route of administration influences the nature of the effects of this drug; daily oral low-dose therapy may more profoundly affect cell-mediated immunity, whereas intermittent high-dose intravenous therapy predominantly affects B cell immunity.696,697 In humans, IgG and IgM synthesis is depressed, and there is a measurable decrease of serum antibody concentration after chronic administration.692,693
TABLE 5–26
Recommended Reductions of Cyclophosphamide Dose in Patients with Impaired Renal Function Serum Creatinine Concentration (mmol/L) <250 250–500 >500
Oral Cyclophosphamide Dose (mg/kg/day) 2 1.75 1.5
CH2CH2CI
■ Figure 5–21 Structure of cyclophosphamide.
Adapted from Luqmani RA, Palmer RG, Bacon PA: Azathioprine, cyclophosphamide, and chlorambucil. Clin Rheumatol 4: 595, 1990.
116
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
The alkylating agents have prominent toxic effects.698–706 Short-term side effects are common; although troublesome to the child, they are seldom serious. These include anorexia, nausea and vomiting, and alopecia. Alopecia appears to be related to dose and duration of treatment and is usually reversible. Pulmonary fibrosis has been reported in a small number of patients receiving daily cyclophosphamide therapy.706a,b Leukopenia and thrombocytopenia are the most common adverse reactions, although with careful monitoring they are seldom of clinical significance. The cyclophosphamide dose should be adjusted to maintain the total granulocyte count at 1500/mm3 (1.5 × 109/L) or higher. The nadir of granulocytopenia with intravenous therapy occurs between the first and the second weeks of therapy, and the dose should be adjusted accordingly based on the complete blood count and differential white blood cell count obtained on days 7, 10, and 14 after intravenous cyclophosphamide administration. Lymphocyte counts lower than 500/mm3 (0.5 × 109/L) are also an indication to lower the dose. Glucocorticoids probably aid in protecting bone marrow from the neutropenia-inducing effects of cyclophosphamide. Cyclophosphamide is administered in one of two ways: either orally each day or by intravenous bolus every 2 to 4 weeks (Table 5–27). Intravenous pulse administration is less toxic and at least as efficacious for lupus nephritis707; in some systemic vasculitides, it is not clear whether the intravenous route is as effective as oral administration.697 Bladder toxicity (cystitis, fibrosis, transitional cell carcinoma) is a major risk of cyclophosphamide therapy and
TABLE 5–27
Suggested Protocol for the Administration of Intravenous Cyclophosphamide Dose 0.5–1.0 g/m2 cyclophosphamide Preparation Start IV with 5% dextrose in water Administer 2 L/m2 of IV fluid as 5% dextrose in water or 2/3 5% dextrose in water and 1/3 normal saline over next 24 hr; start hydration 4 hr before administration of cyclophosphamide MESNA (30% of cyclophosphamide dose) IV in 240 ml of 0.2 normal saline at 10 mL/hr over 24 hr, starting immediately after cyclophosphamide Ondansetron (0.15 mg/kg per dose) IV or PO 30 min before cyclophosphamide and every 8 hr until infusion is complete Dilute cyclophosphamide to 10 mg/mL in 5% dextrose in water and infuse prescribed dose over at least 1 hr (to minimize nausea and vomiting) Monitoring Pulse, blood pressure, and respiratory rate every 30 min during infusion, then every 4 hr for next 24 hr Urinalysis before and after infusion Monitor urinary output: Empty bladder every 2–4 hr. If urinary output falls to less than 50% of IV input over any 4-hr period, give furosemide 1 mg/kg IV. Repeat at 2–4 mg/kg in 2 hr, if necessary. MESNA, 2-mercaptoethanesulfonic acid.
results from prolonged contact of acrolein with the bladder mucosa.701–703 In rats, the toxicity appears to be mediated through nitric oxide produced by inducible nitric oxide synthase.708 To prevent cystitis, adequate hydration and frequent voiding must be emphasized for children receiving daily cyclophosphamide. Persistent nonglomerular hematuria is an indication for cystoscopy; if cystitis is observed, the dose should be reduced or the drug stopped. Intravenous pulse therapy reduces the risk of toxicity, including hemorrhagic cystitis, and confines those risks to a short period each month instead of every day. Prophylactic 2-mercaptoethanesulfonic acid (MESNA) should be considered a part of any intravenous cyclophosphamide protocol to minimize contact of acrolein with the bladder mucosa. The syndrome of inappropriate antidiuretic hormone secretion (SIADH) has been reported in patients receiving large doses of cyclophosphamide and is exacerbated by the large fluid load that must be administered.709 It does not matter which hydration solution is used.710 With the large doses of cyclophosphamide administered by intravenous bolus, children must be encouraged to empty their bladders every 2 hours and must be awakened during the night to do so; if this is not possible, furosemide should be given and catheterization considered to prevent significant contact of the bladder mucosa with acrolein. Nausea and vomiting can be a significant problem; prophylactic use of a potent antiemetic (e.g., ondansetron) is encouraged as part of intravenous protocols. An important consideration with the use of alkylating agents is their effect on fertility for both males and females.711 The stage of sexual maturity is critical in terms of inducing gonadal dysfunction; the further beyond puberty, the greater the chance of infertility with an equivalent dose of cyclophosphamide.712 In female patients with lupus nephritis, amenorrhea and oligomenorrhea occur more frequently with higher total dose and with increased age at administration.713–716 Use of intravenous bolus administration at currently recommended doses results in a much lower total cumulative dose than does daily oral administration and therefore should be the preferred route, presuming equivalent effectiveness. Ovarian destruction attributable to cyclophosphamide was reported in one child.698 Luteinizing hormone-releasing hormone (LHRH) may protect the ovary against cyclophosphamide-induced damage and may be effective in patients treated with cyclophosphamide, as was demonstrated in a study of women with lymphoma who received gonadotropin-releasing hormone agonist.717 Protocols using the gonadotropin-releasing hormone agonist analogue preserved ovarian function in young women treated with cyclophosphamide,718 as well as in adolescents with cancer.719 Oocyte or ovarian tissue cryopreservation holds potential promise for the future. In male patients, sperm cryopreservation is suggested before cyclophosphamide treatment is instituted. Additional approaches include the use of testosterone.720 Cyclophosphamide is associated with an increased risk of malignancy in adults with rheumatoid arthritis, a risk that is dose related and increases with duration of follow-up. One case-control study demonstrated a fourfold
C H A P T E R
5 PHARMACOLOGY
elevation of myeloproliferative disorders,705 and there are increased risks of bladder and skin cancer.721 The incidence of bladder cancer in adults with Wegener’s granulomatosis treated with cyclophosphamide is 5% at 10 years and 16% at 15 years; the incidence is related to both total dose and duration of treatment. Nonglomerular hematuria identified a subgroup of patients at high risk for bladder cancer.722 Short courses of very high doses of cyclophosphamide have been used either as primary therapy in myeloablative doses for SLE and aplastic anemia723 or as part of a transplantation protocol for patients with autoimmune disease. Monthly intravenous cyclophosphamide is used commonly in patients with severe lupus nephritis (World Health Organization [WHO] class IV) and other lifethreatening complications of lupus. However, because of the well-known toxicity of cyclophosphamide and the effectiveness of agents such as MMF and azathioprine in maintaining remission, it has been recommended that the duration of intravenous treatment be shortened to as little as 3 months in some patients with lupus nephritis.724 In addition, cyclophosphamide is well accepted in the treatment of severe necrotizing and granulomatous vasculitis. Cyclophosphamide crosses the placenta and is teratogenic; it is contraindicated both during pregnancy and during breast feeding.649
AND
117
D RU G T H E R A P Y
COOH
CH2CH2CI N
CH2CH2CH2
CH2CH2CI
■ Figure 5–22 Structure of chlorambucil.
for very narrow indications. It is the drug of choice for amyloidosis complicating inflammatory disease and occasionally for severe uveitis.725 However, the very high risk of malignancy, ranging up to 7.5%,726 precludes its use in all but the most severe cases. In addition to the risk of malignancy, male infertility is very common once total doses of 25 mg/kg are reached.727 Thrombocytopenia and infection are also common.725,728
Cyclosporine Cyclosporine (Fig. 5–23), a cyclic peptide of fungal origin, has been shown to have profound effects on the immune system.729 The observation that cyclosporine could virtually eliminate mitogen-induced proliferation by T cells but had little effect on other cell types730 indicated the potential of this drug in the treatment of immunologically mediated disease. It has had a major impact on the prevention of solid organ transplant rejection. Cyclosporine is inactive until complexed with its intracellular receptor, cyclophilin. In the process of T- or B-cell activation, cell receptor signaling leads to a release of intracellular calcium, which binds to calmodulin, activating the protein serine/threonine phosphatase calcineurin.731 Once activated, calcineurin stimulates the translocation of nuclear factor of activated T cells (NF-AT), a transcription factor that is an important stimulus for IL-2 gene transcription732 and cell-mediated immune responses.730,733–735 The cyclosporinecyclophilin complex binds to calcineurin, thus inhibiting
Chlorambucil Chlorambucil (Fig. 5–22), like cyclophosphamide, is an alkylating agent that preferentially reduces B cell numbers, with less effect on memory T cells and natural killer cells, and acts by cross-linking macromolecules, thereby interfering with several cellular functions. It is usually prescribed for oral administration at a dose of 0.1 to 0.2 mg/kg/day but can be given intravenously as well. Because of serious toxicity, use of chlorambucil is reserved
■ Figure 5–23 Structure of cyclosporin.
CH3
H C C
H CH3
CH3 CH
CH2
CH3
H CH3 H
CH3 CH CH3
CH2 CH3
N
C (10) CO C
(9)
N
(8)
OC
C
CH3
CH3
CO
N
CH
C (11)
H C
CH2
HO H
C
CH3
CH3 N
H
C
H CO
C (1)
N H
O
CH3 CH2 CH3 H CH2 C C N (2) (3) CO O N
N H
CH3
H
CO
(7)
H
C
N H
CH3
(6) C
C
N
O (5)
H
C
N
C
H O
CH2
CH CH3
CH3
CO
H C
H
CH3
CH CH3
CH3
(4)
CH2 CH3
CH CH3
CH3
118
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
the early phase of T cell activation and IL-2 production. Cyclosporine inhibits the production of IL-3, IL-4, IFNγ,736,737 and IL-15,738,739 and it enhances the production of transforming growth factor β1 protein.740 It is also antiangiogenic, as demonstrated by the inhibition of vascular endothelial growth factor (VEGF) expression in several systems.741,742 Cyclosporine may also result in immune suppression by inhibiting degradation of I-κB.743 In addition, it may modulate anti-inflammatory effects by inhibiting monocyte production of tissue factor, a potential stimulus of the coagulation cascade via inhibition of NF-κB.744 It may also result in apoptosis of T and B cells.745 Whether cyclosporine affects antigen presentation is uncertain. Some studies have shown that the drug has no effect on macrophage function in antigen presentation,746 whereas others have suggested that the drug interferes with antigen presentation by dendritic cells747 and Langerhans’ cells.748 Cyclosporine is incompletely and variably absorbed from the GI tract, bound principally to serum albumin and erythrocytes, metabolized by the liver, and excreted in the bile. It has a half-life of approximately 18 hours. A microemulsion formulation of cyclosporine has been developed to improve absorption and bioavailability749; this preparation has more consistent interpatient and intrapatient pharmacokinetics. Cyclosporine crosses the placenta and is present in breast milk. It is important to note the several significant drug interactions associated with cyclosporine480 and the considerable toxicity associated with the use of this drug, including impaired renal function,750,751 hypertension, hepatic toxicity,752 tremor, mucous membrane lesions, and nausea and vomiting. Hypertrichosis, paresthesias, and gingival hyperplasia have been observed. Renal toxicity may result in hypertension from interstitial fibrosis or tubular atrophy, even in adults treated with relatively low dosages (3 to 5 mg/kg/day orally). Concomitant use of NSAIDs may exacerbate this toxic effect of the drug. Cyclosporine is effective in the treatment of rheumatoid arthritis, both alone753 and in combination with MTX.754,755 Several open trials have shown improvement in children with JRA, 756–758 JDM, 759 and uveitis. 760,761 The addition of cyclosporine to MTX was associated with significant clinical improvement in 8 of 17 patients with JIA.762 However, side effects are common and often necessitate reduction or discontinuation of treatment, particularly as a result of reduced renal function or hypertension.758 Cyclosporine has been reported to have dramatic beneficial effects in patients with the macrophage activation syndrome.80,81 Patients with membranous lupus nephritis may benefit from cyclosporine treatment,763 as has been documented in pediatric patients with WHO class III or IV lupus nephritis and heavy proteinuria.764 It may be especially helpful as a steroid-sparing agent.765 Despite concern, a case-control series766 did not show any increased risk of malignancy over a 5-year follow-up period in adult patients treated with cyclosporine compared with controls. Current guidelines for cyclosporine use are outlined in Table 5–28. Factors that most commonly limit clinical use of cyclosporine are hypertension and a rise in serum creatinine of greater than 30% from baseline. Unfortunately, long-term renal damage can occur despite normal serum creatinine levels during the course of therapy.749 In the rheumatic diseases, the goal has been to achieve a whole blood trough level between 125
TABLE 5–28
Guidelines for Use of Cyclosporine
Dose 3–5 mg/kg/day orally Clinical Monitoring Blood pressure every week for first month, then monthly Laboratory Monitoring Renal function studies (BUN, creatinine, urinalysis) at start of therapy and every month Hepatic enzymes, CBC with WBCC differential, and platelet count every month Maintain 12 hr whole-blood trough drug levels between 125 and 175 ng/mL (RIA method) Reduce dose if serum creatinine increases by ≥30% BUN, blood urea nitrogen; CBC, complete blood count; WBCC, white blood cell count. ALT, alanine aminotransferase; AST, aspartate aminotransferase; CBC, complete blood count; MCV, mean cell volume; WBCC, white blood cell count.
and 175 μg/mL. It is important to note that grapefruit juice increases cyclosporine and cyclosporine metabolite levels significantly.767
Antibiotics The concept that rheumatoid arthritis was caused by microbial pathogens led to early use of antimicrobial therapy. In fact, it was for that reason that gold was first introduced as a treatment for rheumatoid arthritis. Sulfasalazine was synthesized to take advantage of its antimicrobial properties. Recently, antibiotics have again been considered in the treatment of inflammatory rheumatic disease. Penicillin plays a key prophylactic role in preventing the recurrence of acute rheumatic fever and perhaps in preventing of poststreptococcal arthritis.768 Cutaneous polyarteritis, which may be a streptococcus-related disease, is also often treated with prophylactic penicillin.769 In Wegener’s granulomatosis, treatment with trimethoprim-sulfamethoxazole may prevent disease relapses.770 Studies in rheumatoid arthritis have suggested a role for synthetic tetracycline antibiotics.771 In early disease, minocycline was more effective than placebo772 or hydroxychloroquine.773 The mechanism of action may depend more on the biochemical than on the antimicrobial effect of these agents. Antibiotics do not seem to be effective in enteric reactive arthritis, but they may have a role in urogenital reactive arthritis.774 Although a small pilot study suggested that minocycline might be effective in systemic sclerosis,775 a more recent study refuted those findings.776
Biologic Therapies Intravenous Immunoglobulin Intravenous immunoglobulin (IVIG) is prepared from pooled human plasma. Its effectiveness in the treatment of Kawasaki disease777 has encouraged its use in a num-
C H A P T E R
ber of other childhood rheumatic diseases. It is also reported to be of benefit in some cases of JDM778 and systemic-onset JRA,779 although placebo-controlled randomized trials did not show benefit in either systemic- or polyarticular-onset JRA.780,781 There are also many anecdotal reports of improvements in patients with SLE, systemic vasculitides, and autoimmune neuropathies.782 Although IVIG has a good record of safety, anaphylactoid reaction is a risk. Other potential side effects include myalgia, fever, and headache during the infusion and aseptic meningitis 24 to 48 hours afterward.783 Current preparation protocols purify the product so that it is free from contamination with human immunodeficiency virus (HIV), hepatitis C virus, and other known viruses, but there is a theoretic risk of transmission of CreutzfeldtJacob disease as well as other, as-yet unidentified pathogens. Guidelines for IVIG administration are listed in Table 5–29. The mechanisms whereby IVIG exerts its therapeutic effect are not clear and may differ in different situations. Several recent reviews have thoroughly addressed these issues.782,784,785 Potential mechanisms include (1) Fc receptor blockade (in idiopathic thrombocytopenic purpura); (2) inhibition of complement activation and function of the membrane attack complex (in dermatomyositis); (3) inhibition of cytokine production and function (in Kawasaki disease); (4) increased catabolism of IgG mediated by FcRn (IgG transport receptor) blockade (which may be responsible for the rapid reduction in autoantibody levels in autoimmune disease); and (5) generation of anti-idiotype antibodies, reducing the production of pathogenic autoantibodies (in a variety of autoimmune diseases). Its action may be mediated by specific antibodies that neutralize an as-yet unknown causative agent such as a virus. IVIG contains anti-idiotype antibodies that may bind to the idiotype of an antibody involved in the pathogenesis of the disease.786 Such anti-idiotype antibodies have been shown to suppress autoimmune disease in animal models.787
TABLE 5–29
Guidelines for Use of Intravenous Immunoglobulin (IVIG) Dose Up to 2 g/kg Preparation Start IV with normal saline Administration Give IVIG at rate of 0.5 mL/hr for 30 min, then 1.0 mL/kg/hr for 30 min, then 2.0 mL/kg/hr for the remainder Monitor Blood pressure and pulse rate every 15 min for first hour, every 30 min for second hour, every 60 min thereafter Observe Sudden fall in blood pressure (anaphylaxis) Headache, vomiting 18–36 hr after infusion (aseptic meningitis)
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
119
Antibodies to inflammatory mediators, including cytokines, may also have important therapeutic roles. The rapid defervescence that occurs after IVIG administration in Kawasaki disease suggests that interleukins, particularly IL-1 and IL-6, are removed from the circulation or neutralized, or that their production is stopped by some constituent of the IVIG. It is known that normal serum contains antibodies to IL-1α,788 IL-6,789 TNF-α,790 IFN-α2b, and IFN-γ.791 Normalization of T cell number and function has also occurred after administration of IVIG.792 IVIG can neutralize superantigens, which may be involved in Kawasaki disease.793 Another mechanism of action whereby IVIG might exert its beneficial anti-inflammatory effects is a reduction in the expression of adhesion molecules. In JDM, IVIG can decrease the activity of the membrane attack complex.794
Specific Biologic Agents Elucidation of some of the basic mechanisms involved in rheumatoid arthritis has resulted in an understanding of many of the cellular and molecular mechanisms that participate in inflammatory states. Specific biologic agents have been developed that can target one or several steps involved in the immune response. Strategies for intervention (adapted from Wallis and associates795) can be grouped as follows: ■ ■ ■ ■ ■
Tolerance induction Inhibition of MHC/antigen/T cell receptor (TCR) interaction Inhibition of cellular function and cell–cell interaction Interference with cytokines Apoptosis
Although the initial excitement regarding biologic therapy was tempered by either lack of efficacy or severe toxicity, more recent developments justify a great deal of optimism for the development of new, more focused and targeted therapies over the next few years.
Induction of Tolerance Autoreactive T cells that escape thymic deletion during ontogeny can be deleted by at least two peripheral mechanisms of tolerance.796 If the T cells interact with antigen but are not activated, T cell dormancy, or anergy, develops. If type 2 helper (Th2) T cells have the same TCR as the autoreactive clone, cytokines with an antiinflammatory profile will be released and can suppress an antigen-specific Th1 response. This is the principle behind attempts to achieve oral tolerance to an antigen. Reactive Th2 cells can leave the gut and migrate to sites in which the antigen may be localized and are stimulated to release anti-inflammatory cytokines, thereby mediating local immune suppression. Trials of oral administration of type II collagen have been attempted because of the high incidence of autoimmunity to type II collagen in adult rheumatoid arthritis (and in approximately 25% of children with JRA), as well as the success of this approach in a variety of animal models of arthritis in which type II collagen autoimmunity occurred.797 Studies in humans
120
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
have shown varying results, but results seem to be better when lower doses of collagen are administered.798 In an open trial involving 10 patients with JRA who were treated with type II chick collagen, 8 responded; no adverse effects were observed.799 With the advances that have occurred with other therapies, there have been no further studies in this area.
LJP-394 LJP-394 (abetimus sodium) is a molecule consisting of double-stranded DNA (ds-DNA) epitopes that cross-link anti–ds-DNA antibodies in solution or on cell surfaces. In rodents, LJP appears to induce B-cell tolerance by cross-linking anti–ds-DNA surface immunoglobulin receptors on B cells, leading to B-cell anergy or apoptosis. It has been shown to reduce the titers of anti–ds-DNA antibodies in patients with SLE.800 A randomized, double-blind, placebo-controlled trial was performed to examine whether LJP prevents or delays renal flares in patients with SLE and previous renal disease.801 Although the number of renal flares and the time of flares did not differ between the two groups, the time to institute “aggressive treatment” with corticosteroid or cyclophosphamide (or both) was longer, and the number of aggressive treatment courses were fewer in the LJP group. There was a significant reduction in the titer of anti–ds-DNA antibodies and a concomitant, although not significant, rise in the serum complement levels. Patients with high-affinity anti–ds-DNA antibodies had the greatest reduction in antibody titer. The treatment was well tolerated. Although it was not statistically significant, there was a trend toward more thromboembolic events in the LJP-treated group. Clinically important differences in the health-related quality of life were seen in the treatment group.802
Inhibition of MHC /Antigen/T Cell Receptor Interaction The immune response is driven by the processing and presenting of an antigenic peptide by an antigen-presenting cell to a specific TCR in the context of a specific MHC molecule. Any component of this trimolecular complex could theoretically be targeted for biologic modulation. If the initiating antigen were known, an immunization program could be developed to prevent disease, and this is the principle behind oral tolerance (see earlier discussion). TCR vaccines may be one way to prevent or reduce activity of synovial inflammation, as shown in animal models. This strategy would work only if specific variable region (Vβ subtypes could be shown to predominate in synovitis and did not change over time. Early studies in patients with rheumatoid arthritis who were vaccinated with several Vβ subtypes show promise and are continuing.803 Data in children with JRA and spondyloarthropathies suggest that this approach may have some merit.804 Another possible route of attack is to block the MHC site by anti-MHC antibodies.
Inhibition of Cellular Function and Cell–Cell Interaction T cells appear to play a central role in initiating the rheumatoid process in adult disease and in JRA, but their role in continuing to drive the inflammatory process is less clear. Results of early studies using monoclonal T cell–depleting antibodies were disappointing. Initial
clinical improvements, if they occurred at all, were short lived or were associated with profound lymphopenia, precluding further treatment.805,806 These included studies with anti-CD7 monoclonal antibodies (mAbs), CD-5 PLUS (immunotoxin composed of murine anti-CD5 mAbs conjugated to the toxin ricin),807 CAMPATH (humanized αCDw52 mAb),808,809 and cM-T412, an anti-CD4 mAb.810,811 Possible explanations for the lack of efficacy in these studies are that T cells are not necessarily critical for the perpetuation of synovitis; that the specific T cells targeted by monoclonal antibodies are not the ones involved in synovitis; that targets are too nonspecific; and that, although peripheral T cells may be affected, synovial T cells are not.812 One placebo-controlled trial in adult rheumatoid arthritis treated with nondepleting anti-CD4 mAbs showed some efficacy, but unacceptable CD4 lymphopenia and rash precluded further study.813 Generation of the immune response involves not only antigen processing and TCR/MHC interactions but also interactions between molecules expressed on the surface of T cells and on antigen-presenting cells, as well as various adhesion molecules and their companion receptors. Although preclinical studies in animals are encouraging, no studies have been reported to date in humans.814,815
CTLA-4Ig In order to activate resting T cells, two molecular signals are required: (1) the interaction of the TCR with processed peptide, presented in the appropriate MHC setting; and (2) interaction of CD28 on T cells with CD80/86 on the surface of the antigen-presenting cell. There is another high-affinity receptor, cytotoxic T lymphocyte–associated antigen-4 (CTLA-4), which can also bind to CD80/86, with a higher avidity than CD28, thereby preventing the second signal required for T cell activation. CTLA-4 immunoglobulin (CTLA-4Ig) is a soluble immunoglobulin receptor fusion protein that consists of the extracellular domain of human CTLA-4 and a fragment of the Fc domain of human IgG.816 By binding to CD28, it can prevent T cell activation. Early preclinical studies showed benefit in rodents with inflammatory synovitis, and initial studies in adults with rheumatoid arthritis showed modest improvement, both when compared with placebo alone817 and also when used together with MTX compared with MTX alone in patients who had an inadequate response to MTX.818 Anti-CD40 Ligand The interaction between CD40 (found mainly on T cells) and CD40 ligand (also known as CD154), is essential in the production of pathogenic autoantibodies and tissue injury in lupus nephritis. Antibodies to CD40 ligand have been shown to improve renal disease, even if well established, in animals with lupus nephritis. Several small studies using anti-CD40 ligand in patients with lupus nephritis have been reported. Various doses of IDEC-131 (a humanized mAb against CD154) were compared with placebo in 85 patients with mild to moderate lupus. Although the treatment was safe and well-tolerated, patients did not improve relative to placebo.819 However, subsequent studies with this agent were halted after the report of a thrombotic event in a patient with Crohn’s disease. In an open-label study
C H A P T E R
involving 28 patients with diffuse proliferative lupus nephritis, treatment with a different anti-CD154 antibody, BG9588, resulted in a significant reduction on anti–ds-DNA antibodies, a significant increase in serum C3, and improvements in proteinuria and hematuria.820 However, adverse events, including myocardial infarctions in two young patients, precluded completion of the treatment trial. Therefore, although the concept makes biologic sense, the current preparations cannot be used and require further experimentation.821 Interactions between activated endothelium and leukocytes via adhesion molecules allow leukocytes to leave the peripheral circulation to enter sites of inflammation (e.g., the synovium). The results of early studies using a murine IgG2 anti-intercellular adhesion molecule mAb to inhibit this interaction in rheumatoid arthritis were encouraging,822 but retreatment frequently resulted in allergic side effects,823 which suggested that repeated treatment would not be a useful strategy.
Rituximab Rituximab is a chimeric monoclonal mouse– human antibody that is reactive with the B cell CD20 receptor, which is present on pre-B and mature B cells but not on stem cells or plasma cells.824 It has been effective in the treatment of relapsed Hodgkin’s B cell lymphoma. The role of CD20 is not clear, but rituximab exerts its effect by removing B cells from the circulation, both by antibody-dependent and complement-dependent cellular cytotoxicity and by the induction of apoptosis of B cells. Although the antibody-producing plasma cells are not removed from the circulation, B cells that may act as antigen-presenting cells produce cytokines, and infiltrate tissues are removed for a prolonged period. Memory B cells, which are also responsible for antibody production, may be removed as well. Rituximab is theoretically beneficial in diseases in which B cell autoantibodies may be pathogenetically important. This was observed first in patients with idiopathic thrombocytopenic purpura825 and more recently in small case series or case reports of a variety of autoimmune diseases. At an intravenous dose of 375 mg/m2 administered weekly for four infusions, B cells are reduced dramatically for a period of 6 to 9 months. Although disease manifestations may return later, coincident with the reappearance of B cells, baseline medications may be tapered during this period. Side effects are rare and include flushing and itching, usually with the first dose. These manifestations are probably allergic in nature and may be alleviated with pretreatment with an antihistamine and corticosteroids.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
121
Reduced levels of immunoglobulins for prolonged periods appeared to be more common and significant in children then in adults and may require immunoglobulin replacement therapy. Patients must be observed closely for development of viral infections, because post-rituximab infections have been reported with parvovirus, varicella-zoster virus, cytomegalovirus, and enterovirus. Binstadt and colleagues826 reported on a series of four children with undiagnosed autoimmune illnesses who received rituximab treatment after failure of multiple other agents. Impressive improvement in the neurologic manifestations of these patients was observed. Nadir of IgG levels were seen at 4 to 6 months and three of the four patients required immunoglobulin replacement therapy, at least in the short term. Leandro and associates827 reported on six patients with SLE treated with a combination of two rituximab infusions of 500 mg, two infusions of cyclophosphamide at 750 mg, and high-dose oral corticosteroids, with a very good response at 6 months in five of the six. Interestingly, although hypocomplementemia in these patients returned to normal, the response of anti-DNA antibodies was quite variable. The exact role of rituximab in the treatment of autoimmune diseases is still evolving.828,829
Interference with Cytokines The biologic effects of T cell–derived and monocytederived cytokines can explain much of the clinical syndrome of synovitis as well as the systemic manifestations associated with JRA.830–832 Cytokines are critical in perpetuating and damping the immune response, and, as such, they are important targets for therapeutic manipulation. A great deal of evidence supports the role of TNF-α in the initiation and perpetuation of the rheumatoid process. Children with JRA have high levels of TNF-α in the synovial fluid and peripheral circulation.833,834 Studies in adults have shown that high levels of TNF-α in synovial fluid are associated with bone erosions. Animal studies, and early studies in adults, showed convincingly that blocking TNF improved symptoms of inflammatory arthritis. Subsequent studies showed that these agents may be dramatically beneficial, not only in reducing disease activity, but also in improving function and retarding, and perhaps reversing, structural damage. Anti-TNF agents currently in use or under study in children are etanercept, infliximab, and adalimumab.
Etanercept Etanercept is a fully human, dimeric protein containing the extracellular domain of the human p75 TNF receptor fused to the Fc region of human IgG1 (Fig. 5–24). It is produced with the use of recombinant DNA
s
s
s
■ Figure 5–24 Structure of etanercept. s s
CH3
s
s
s
s s
CH2
Fc region of human IgG1
Extracellular domain of human p75 TNF receptor
122
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
technology. By binding to TNF-α in the circulation, etanercept prevents the interaction of TNF-α with its cell surface receptor, thereby preventing cell activation and perpetuation of the inflammatory cascade. It can also modulate biologic responses that are mediated by TNF, such as expression of adhesion molecules, serum concentration of MMPs and cytokines.835 Although soluble forms of the TNF-α receptor occur naturally, they are generally inadequate to block TNF activity in systemic inflammatory disorders. The dimeric form of the TNF-α receptor is much more efficient at binding TNF, because it binds at a much greater affinity (50 to 1000 times higher) than that of the naturally occurring form. Its halflife, administered subcutaneously, is approximately 4 days. In adults with rheumatoid arthritis, steady-state concentrations are achieved in about 2 weeks.835 Etanercept has been used extensively in children with JRA since the initial report of its effectiveness in 2000.836 It is currently indicated for children who have had an inadequate response or who have not tolerated MTX and still have polyarticular disease. The initial study by Lovell and colleagues,836 as well as a large case series report by Quartier and coworkers,837 suggested that approximately 70% of children will have an initial response to etanercept. The response is usually dramatic, occurs by the third or fourth injection, and is associated with a dramatic reduction in active joint count and markers of inflammation. Although long-term data are lacking, a French study reported that only 39% of patients had a sustained improvement of 30% or more by 12 months.837 The North American follow-up study, in which 43 patients completed 2 years of treatment, showed that 81% of patients improved by at least 30%, 79% by at least 50%, and 67% by at least 70%.838 However, most of these patients were from an early responder group, and these data are less generalizable to a standard JRA population. Unfortunately, the results in children with systemic-onset JRA do not seem to be nearly as promising.839,840 Studies in adults have shown that the combination of etanercept and MTX is better than either drug alone, in terms of disease activity, functional disability as measured by the Health Assessment Questionnaire, and retardation of radiographic progression.841,842 The combination of etanercept and MTX has been used extensively in pediatric clinical practice.843,844 Although no data are available, the standard practice in treating children with JRA currently is to add etanercept to MTX for patients who have had only a partial response. Etanercept is administered subcutaneously, twice per week at a dose of 0.4 mg/kg, to a maximum of 25 mg. Higher doses do not seem to be more effective845 (Table 5–30). Administration of the dose on a once-weekly basis has been effective in adults846 and is currently being evaluated in children. It would still need to be administered in two injection sites. Given the success in treating early adult rheumatoid arthritis, research is currently underway to assess the optimum timing during the course of the patient’s arthritis. The ability to improve radiographic changes in adults suggests that etanercept may repair structural damage, in additional to improving disease activity. Therefore, it has been suggested that etanercept be administered very early in the course of disease.
TABLE 5–30
Guidelines for Use of Etanercept in the Treatment of Juvenile Idiopathic Arthritis Dose 0.4 mg/kg twice per week subcutaneously Clinical Monitoring Improvement should be seen by the third to fourth dose Monitor every 1–2 mo initially, then every 3–6 mo, depending on course Hold if suspected bacterial infection, varicella Laboratory Monitoring CBC with WBCC, differential and platelet count; AST, ALT, albumin every 4–8 weeks.
Magnetic resonance imaging studies in adults have shown that bone erosion can occur as early as 4 months after disease onset. Studies are underway in children to determine whether early administration of etanercept may lead to remission. To date, etanercept has been very well tolerated. The placebo-controlled study showed an increase in symptoms of upper respiratory tract infection as well as injection site reactions, although these were generally mild836 and may be treated with topical corticosteroids. However, postmarketing studies have reported a variety of unusual side effects; most importantly, systemic infection must be closely watched for. This includes bacterial infection, viral infection such as varicella with or without superimposed bacterial infection, and granulomatous infection. Although these are more common in elderly patients, physicians must be attuned to their development in any age group. Early postmarketing studies suggested that demyelinating syndromes, including multiple sclerosis, might be more common in patients treated with etanercept. Another TNF antagonist, lenercept, used to treat patients with multiple sclerosis; patients taking active drugs had exacerbations.847 Patients with JRA have developed demyelinating syndromes, as have adults with rheumatoid arthritis.848 Patients with previous demyelinating syndromes should not be treated with TNF antagonists, and those with a strong family history should be observed carefully for the development of symptoms that may be suggestive of demyelination. A variety of other side effects have been noted. Although rare enough to be the subject of case reports, there does seem to be an association between treatment with etanercept and the development of vasculitic skin rash849 or drug-induced lupus.850 The associations with pancytopenia and aplastic anemia are less clear.851 One case of diabetes mellitus after etanercept treatment has been reported in a child with JRA.852 Changes in mood and weight gain have also been noted.837 Other disorders in which etanercept has been studied with early promise include psoriatic arthritis,853 ankylosing spondylitis,854 uveitis,855 and Wegener’s granulomatosis.856
Infliximab Infliximab is a chimeric IgG1 anti–TNF-α antibody consisting of a mouse antibody and the constant region of the human antibody.857 It was the first anti-TNF
C H A P T E R
agent that was shown to be effective in patients with rheumatoid arthritis,858,859 and is also effective in patients with Crohn’s disease. As opposed to etanercept, it binds not only to soluble TNF-α but also to membrane-bound TNF-α, leading both antibody-dependent and complement-dependent cytotoxicity. It therefore seems to be more efficacious in granulomatous inflammatory disorders (e.g., sarcoidosis) but also seems to be somewhat more risky in the development of granulomatous opportunistic infections (see later discussion). Administration of infliximab is through the intravenous route (Table 5–31). Dose and frequency vary somewhat with the clinical response. Initial doses usually start at 3 mg/kg and are given at time 0, at 2 weeks, at 6 weeks, and then every 8 weeks depending on clinical response. Doses often require escalation, and doses up to 20 mg/kg have been used occasionally in children.860 Alternatively, the length of time between infusions can be shortened. Because of the development of anti-infliximab antibodies, administration with MTX is recommended, because these antibodies seem to correlate with infusion reactions and accelerated clearance of infliximab.857 It is not certain, however, that they actually reduce the effectiveness. Similar to etanercept, combination treatment with MTX seems to improve the response to infliximab.861 Interestingly, infliximab may improve the radiologic status without necessarily improving the clinical status of disease.862 Early studies in children with JRA showed improvement, although no placebo trials have been done.844 Improvement occurs soon after the first infusion. Gerloni et al reported the results of 24 patients with JIA (5 systemic, 5 rheumatoid factor negative polyarthritis, 1 rheumatoid factor positive polyarthritis, 10 extended oligoarthritis and 3 psoriatic arthritis patients) who were treated with infliximab. All but 5 were “young adults” at the time of treatment, and still had active disease despite treatment with weekly methotrexate at a dose of at least 10 mg/m2. All started infliximab at a dose of 3 mg/kg at times 0, 2 and 6 weeks and then were scheduled to receive infliximab again every 8 weeks, but 20% of patients who received more than 3 infusions required that the interval between infusions be shortened. Fourteen patients required an increase in their maintenance dose. Only 9 patients completed one year of therapy. Five patients dropped out during the first year (infusion reactions in 4, failure of therapy in 1). All clinical parameters improved quickly and the improvements were sustained over time. However, there was no overall significant TABLE 5–31
Guidelines for Use of Infliximab in the Treatment of Juvenile Idiopathic Arthritis Dose 3 mg/kg IV on weeks 0, 2, and 6, then every 4–8 wk thereafter, depending on course; dose may be increased up to 10 mg/kg Clinical Monitoring Improvement should be seen soon after the first dose Monitor at each visit Hold if suspected bacterial infection, varicella Laboratory Monitoring CBC with WBCC, differential and platelet count; AST, ALT, albumin every 4–8 weeks.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
123
improvement in the HAQ score, perhaps because these patients were so severely affected prior to starting infliximab treatment. Adverse events occurred in 50% of patients, including 15 infusion reactions in 7 patients (requiring discontinuation of treatment in 4 patients). Antibodies to ds-DNA developed in 2 patients by the second month of treatment, were not associated with any signs or symptoms of systemic lupus erythematosus, and disappeared despite continuing infliximab. No serious infections occurred.
Infection remains the major concern with the use of infliximab. Many more cases of tuberculosis have been reported in patients treated with infliximab than in those treated with etanercept, probably because of the destabilization of previously formed granulomata.863 Other infections that have occurred with greater than expected frequency include histoplasmosis, coccidioidomycosis, and listeriosis.857 In addition to the side effects noted with etanercept, infusion reactions ranging from mild allergic reactions to anaphylactic reactions may occur, more commonly on the second or third infusions. Therefore, infliximab must be administered under close observation. In addition to treatment of JRA, infliximab may be beneficial in other autoimmune diseases, including vasculitis,864 Kawasaki disease,865 sarcoidosis,866 uveitis,867 and Behçet’s disease.868
Adalimumab Adalimumab is a recombinant human IgG1 mAb that acts in a similar fashion to infliximab by binding to TNF both within the circulation and on the cell surface.869 Therefore, it may result in cell lysis in the presence of complement. It is administered subcutaneously with a half-life of approximately 2 weeks. Recommended dosing in adults is 40 mg every second week. As with other anti-TNF agents, the results are seen quickly but the dose may need to be given once a week for sustained improvement. Injection site reactions can be problematic. In trials in adults with rheumatoid arthritis, adalimumab was shown to be more effective than MTX, in terms of disease activity, function- and healthrelated quality of life, and ability to limit structural damage.869 Adalimumab can be added safely to MTX with increased efficacy.870 Lovell et al reported the preliminary results from a 16 week open label lead into a multicenter phase III randomized double blind placebo controlled trial of adalimumab given at a dose of 24 mg/m2 every other week to patients with polyarticular JIA with a minimum of 5 swollen and 3 tender joints despite treatment with methotrexate.870a Of the 171 patients enrolled, 155 completed the 16 week open trial; 52% remained on MTX. By week 16, 95% of the patients on MTX and 88% on monotherapy reached at least a 30% improvement by ACR criteria, which occurred in the majority of children by 2 weeks. Infection was the most common adverse event, primarily upper respiratory tract infections, although three serious events (genital herpes, pneumonia, acute gastritis) occurred. Post-marketing studies have shown significant cytopenias such as thrombocytopenia, leukopenia, and pancytopenia.
Common Issues with Anti-TNF Agents With increasing use of anti-TNF agents, a number of common concerns have arisen, one of which is the increased risk of infection, particularly tuberculosis. In general, before starting treatment with any of these agents, the following approach is
124
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
recommended. Patients should be screened for the presence of latent tuberculosis with a tuberculosis skin test. A chest radiograph is probably unnecessary unless the PPD result is positive. If the skin test is positive, thorough investigation of the patient and family for active tuberculosis must be done, and the patient must be treated accordingly. If the investigations prove negative, the patient should be given isoniazid (INH) for 9 months. Treatment with anti-TNF agents may be initiated 1 month after starting INH.871 Anti-TNF treatment should not be used in patients with active infection and should be discontinued in the case of a serious infection. Mild upper respiratory tract infections are not a reason to stop anti-TNF agents. Concern remains regarding the development of malignancy, particularly lymphoma. However, any association is difficult to sort out because of the known increased incidence of malignancy in patients with rheumatoid arthritis.851 Nevertheless, patients must be observed closely for the occurrence of malignancies. Finally, trials have been performed to block TNF in patients with congestive heart failure (CHF); neither infliximab nor etanercept was effective, and they may have even worsened the CHF.872 A recent report concluded that these agents might exacerbate, or even induce, CHF in patients with no previous risk factors.873 It does not seem that any specific laboratory monitoring is required routinely for any of these agents. Although the induction of antinuclear antibodies is common (up to approximately 15% of patients), screening for them is necessary only if suspicion of a developing autoimmune disease (e.g., drug-induced lupus) is raised.
Anakinra IL-1 plays a prominent role in rheumatoid arthritis by stimulating synoviocytes and chondrocytes to produce small inflammatory mediators (e.g., prostaglandins) and MMPs that lead to cartilage destruction and bone erosions. IL-1 also increases the expression of receptor-associated NF-κB ligand (RANK ligand), leading to osteoclast differentiation and activation and bone destruction. It exerts its effect by binding to IL-1 receptor and through cell signaling and production of these various molecules and cytokines. Interleukin-1 receptor antagonist (IL-1Ra), or anakinra, is a naturally occurring, acute phase anti-inflammatory protein, part of the IL-1 supergene family.874 IL-1Ra is the most important physiologic regulator of IL-1–induced activity. By binding to the IL-1 receptor on cell surfaces, it prevents the interaction of the receptor with IL-1 and subsequent cell signaling. An imbalance between IL-1 and IL-1Ra can lead to uncontrolled inflammation. Anakinra is a human recombinant form of IL-1Ra that is produced by recombinant technology in Escherichia coli. Its half-life is 4 to 6 hours when dosed at 1 to 2 mg/kg in adults with rheumatoid arthritis.874 It is administered subcutaneously by daily injection. Preliminary studies in animals showed improvement, which led to studies in adults with rheumatoid arthritis. Several studies showed improvement in American College of Rheumatology (ACR)-20, -50, and -70 scores versus placebo at 12 and 24 weeks.874 Although the improve-
ments have not been as dramatic as these reported with anti-TNF agents, they do offer another option for treatment. In a study by Cohen and associates875 of combination therapy with MTX, there was an increasing response to anakinra, and the response occurred early, by 2 to 4 weeks. Furthermore, the response appeared to be sustained (at least four of six patients improved) in a dosedependent fashion, and improvement appeared to continue beyond 24 weeks. Importantly, anakinra also appears to halt876 and perhaps even repair bone destruction as viewed radiologically.877 Adverse events have, in general, not been serious. The most common are injection site reactions, which tend to occur within the first 4 weeks and are rare later. They consist of rash, erythema, and pruritus and may be relieved with ice packs and application of topical corticosteroid. Rarely are they severe enough to stop treatment. Although serious infections (pneumonia, cellulitis) were more common compared with placebo, no deaths from infection have been reported, and, in contrast to anti-TNF agents, opportunistic infections have not occurred in the studies to date. It appears safe to use MTX with anakinra,875 but etanercept does not seem to add benefit to anakinra in patients with rheumatoid arthritis.878 A small series demonstrated that, in general, patients who had failed to respond to anti-TNF agents also did poorly with anakinra.879 Its role in children with JIA is unclear and is currently under study; preliminary data have indicated some promising benefit.880 Seven patients with MTX and etanercept-resistant systemiconset JRA treated with anakinra were reported from 5 separate centres.880a Most patients improved within 2 weeks and showed a sustained response, which included a reduction in the number of active joints, an improvement in the laboratory values and a reduction in the requirement for prednisone. Similarly, 4 children with MTX and etanercept-resistant systemic-onset JRA were reported from a single centre.880b All four showed improvement of at least 30% after one and two months of anakinra. Side effects were mild and did not require discontinuation from therapy.
Anti-IL6 Receptor Antibody IL-6 appears to be an important potential target, particularly in the treatment of systemic-onset JIA. There is ample evidence that IL-6 is central to the many clinical and laboratory manifestations of this disease. An imbalance between IL-6 and its soluble receptor can lead to increased IL-6 binding on cell surfaces with its receptor, binding gp130, on the cell membrane, leading to intracellular signaling and resulting cytokine production and release.881 Elegant studies have produced evidence that levels of IL-6 correlate with spikes of fever, thrombocytosis, and joint involvement,882 and, in mice transgenic for IL-6, growth retardation is observed.883 Therefore, neutralization of IL-6 would be expected to be very beneficial. Anti-IL6 receptor antibody, MRA, is a genetically engineered, humanized, mAb that is produced by grafting the complementarity-determining region of mouse anti-human IL-6 receptor to human IgG1.881
C H A P T E R
MRA competes with both the soluble and the membrane-bound IL-6 receptor to prevent cell signaling. Studies in animals showed that MRA reduces joint inflammation in collagen arthritis.884 Preliminary data show evidence for MRA in adult-onset Still’s disease,885 in Castleman’s disease,886 and in adult rheumatoid arthritis.887 The suggested dose is 5 mg/kg/week intravenously, tailored as necessary. In a very preliminary study recently presented, 11 children with systemic-onset JIA were treated with MRA in a dose-escalation trial, depending on the initial response to a dose of 2 mg/kg of MRA, to a maximum dose of 8 mg/kg. Three patients received 2 mg/kg, five received 4 mg/kg, and three received 8 mg/kg, with a 70% JIA core set response in 33.3%, 60%, and 100%, respectively. No children withdrew due to adverse events or disease flare.888 The long-term effects of anti-IL6 receptor monoclonal antibody (aIL-6R Mab) were reported in 4 children who received treatment once every two weeks for 2.5 to 3 years. Growth improved and corticosteroids were withdrawn in three and reduced in the fourth, and the aIL-6R Mab was able to be discontinued in two children after three years. Two patients had paronychiae and one developed a subcutaneous abscess.888a An international multi-centre trial is currently being planned.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
125
TABLE 5–32
Combination Therapies Reported to be Effective in the Treatment of Rheumatoid Arthritis and Juvenile Idiopathic Arthritis Sulfasalazine, methotrexate, and hydroxychloroquine272 Methotrexate and cyclosporin754,891 Chloroquine and methotrexate892 Prednisolone, methotrexate, and sulfasalazine893,894 Leflunomide and infliximab895 Biologic therapy combined with methotrexate841,842,861,870,875 IV methylprednisolone, IV cyclophosphamide, and methotrexate610,613
Apoptosis (programmed cell death) is a process whereby normal tissue growth is maintained and controlled by the expression of oncogenes. Inflammatory cytokines in rheumatoid arthritis synovial fluid upregulate the expression of apoptosis factors, but apoptosis per se appears to be defective,889 possibly because of a defect in the interaction between the oncogene Fas and its ligand. Strategies to correct defective apoptosis include monoclonal anti-Fas antibodies and administration of Fas ligand; this approach may be amenable to gene therapy (see later discussion).
fasalazine alone (COBRA Trial).893,894 There is a suggestion that the combination of leflunomide and infliximab may also be effective. 895 In addition, the improvement noted with biologic therapy can be enhanced by combining it with MTX,841,842,861,870,875 and the combination of etanercept and MTX may also retard structural damage, opening the door to potential long-term remission (TEMPO Trial).841 However, one combination of the biologic agents anakinra and etanercept was no more effective than either agent alone and was more toxic.878 In JRA, combination therapy has included intravenous methylprednisolone, intravenous cyclophosphamide, and MTX.610,613 In addition, MTX is usually used together with either etanercept or infliximab with no increase in toxicity.837,839,843,844 Combination therapy would seem particularly appropriate in cases of severe systemic-onset JRA. Important questions remaining to be answered include which patients are most at risk for long-term damage and therefore most likely to benefit from combination therapy; whether therapy should be started in combination or medication should be added only after a partial inadequate response; and whether full or reduced doses of each agent should be used.
Combination Therapies
Gene Therapy
In the past, there was a reluctance to use combinations of DMARDs for arthritis because of concerns about increased toxicity of agents with similar toxicity profiles. However, a number of factors support the use of combination therapies. In adult rheumatoid arthritis, single agents seem to lose efficacy over time890; furthermore, these agents rarely induce sustained long-term remissions. An increased appreciation of the long-term morbidity of both rheumatoid arthritis and JRA supports a more aggressive approach to medical management (Table 5–32).147 A better understanding of the mechanisms of action of these agents, as well as recent welldesigned studies of combination therapy in adults that demonstrated efficacy without a significant increase in toxicity, support the use of this approach. In adult rheumatoid arthritis, studies have demonstrated the efficacy of triple therapy with sulfasalazine, MTX, and hydroxychloroquine versus any two of these drugs alone272; MTX and cyclosporine versus MTX or placebo754,891; chloroquine and MTX versus MTX892; and prednisolone, MTX, and sulfasalazine versus sul-
The successes seen with biologic therapy have supported a potential role for gene therapy in patients with arthritis. The costs and inconvenience of ongoing therapy, and the potential ability of gene therapy to deliver sustained concentrations of therapeutic molecules directly to sites of inflammation make this an attractive option. Ideally, children with the worst prognoses would be targeted for study. However, this would most likely require systemic rather than local therapy, and systemic gene therapy has been more difficult to control. It might be ideal for oligoarticular JRA once its safety is established.896 There are a variety of mechanisms in the inflammatory process that can be targeted by gene therapy (summarized in recent reviews897,898). Proof-of-principle studies in animals have led to trials in adults with rheumatoid arthritis,899 in the form of autologous synovial cell implants containing IL-1Ra in a retroviral vector. Results of long-term outcome and safety data are awaited. There is potential for gene therapy to control gene expression (e.g., IL-10 to change a Th1 to a Th2 response; Fas ligand to correct defective apoptosis; blockade of NF-κB);
Apoptosis
126
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
their impact on systemic manifestations awaits clinical trials, which are underway.
High-Dose Immunotherapy with Transplantation Cures of adjuvant arthritis in animal models by either syngeneic900 or autologous bone marrow transplantation,901 together with “experiments in nature” in which patients with rheumatoid arthritis who underwent bone marrow transplantation for other disorders were noted to experience an improvement in their rheumatoid arthritis, have paved the way for new approaches to the treatment of autoimmune disease (i.e., autologous transplantation). The principles behind this treatment are that high-dose myeloablative therapy will destroy the autoreactive clones that initiate the autoimmune process, and the marrow can be repopulated with a “naive” population of stem cells. As the immune system redevelops after transplantation, immune cells may become “tolerized” to the putative antigens that are involved in the autoimmune process. In fact, allogeneic matched bone marrow transplantation for patients with RF-positive rheumatoid arthritis and associated aplastic anemia has led to remission of the arthritis. However, the length of the remission varied, from as short as 1 year to as long as 13 years.902,903 Allogeneic bone marrow transplantation carries a significant risk of both mortality (15% to 35%) and development of graft-versus-host disease (GVHD). The use of an autologous transplant, from either marrow or peripheral blood stem cells (autologous stem cell transplantation, or ASCT), reduces the mortality from the procedure to 1% to 5% and is not associated with GVHD. Therefore, highdose immunotherapy with ASCT has become a method that has been used in treating several autoimmune diseases. Initial studies in patients with autoimmune diseases and associated malignancies who underwent ASCT showed recurrence of disease within 5 weeks to 1 year.904 In these initial studies, the “retransplanted” stem cells were not manipulated in either a positive way (selection for CD34-positive stem cells) or a negative way (removal of T cells with potential autoreactivity). Relapses may also occur because (1) the putative autoantigens responsible for the disease were not eliminated, (2) the HLA status of the host did not change and therefore a predisposition to select arthritogenic peptides and a limited number of T cell developmental pathways persists, and (3) autoreactive T cells were not completely eliminated before transplantation. Preliminary studies of ASCT have been described in children with JRA and scleroderma, with excellent outcomes in most but not all studies.905,906 An initial mortality rate of 14% raised significant concerns despite remarkable improvement in some patients.907 Wulffraatt and associates908 reported their experience with 31 patients (25 systemic-onset, 6 polyarticular-onset) with polyarticular-course JIA treated with ASCT from eight different European pediatric transplantation centers. Bone marrow cells were transfused in 23 cases and peripheral stem cells, after harvesting with cyclophosphamide (2 g/m2) and granulocyte colony-stimulating factor, in 8. T cells were selected for CD34
stem cells by either negative or positive selection techniques. Conditioning included 5 days of antithymocyte antiglobulin (ATG) on days −9 through −6 and cyclophosphamide (50 mg/kg) on days −5 through −2; low-dose total body irradiation (TBI) was given to 21 patients on day −1. Frozen stem cells were thawed and infused on day 0. The neutrophil and platelet counts returned to normal by day 35. In vitro mitogenic T cell responses normalized within 6 to 18 months, and T cell counts were normal by 5 to 9 months. Seventeen patients had a drugfree period of 8 to 60 months. Mild relapses, which were easy to control, occurred in 7 patients. Four patients had no response to ASCT at all, and 3 patients died—2 with macrophage activation syndrome (1 induced by EBV) and 1 with disseminated toxoplasmosis. Catch-up growth was seen in younger children but not in older children or in those with long disease duration. All patients developed chills, fever, and malaise during the infusion of ATG. In addition to the two patients who died from infection-related causes, infectious complications were common (varicella-zoster in seven patients, atypical mycobacteria and Legionella pneumonia in one each). ASCT has been shown to alter laboratory abnormalities that reflect the immunologic process, including perforin expression,909 expression of myeloid-related proteins (MRP8/MRP14),910 and synovial cellularity and cytokine expression.911
Encouraging reports from the use of ASCT in patients with SLE912,913 and systemic sclerosis914 have led to the development of new studies, whose results are awaited. Although ASCT has not been curative in patients with rheumatoid arthritis, the disease seems easier to control with DMARDs after the procedure.915 Many questions remain regarding this treatment and the crucial variables in the protocols. The intensive immunotherapy required (high-dose cyclophosphamide ± irradiation ± antithymocyte globulin) may itself result in disease remission, as described in several cases of aplastic anemia and SLE.916,917 It is not clear whether irradiation is necessary, particularly because it may significantly increase the risk of malignancy; it did not seem to improve the outcome in the 31 patients described by Wulffraatt and associates.908 It is likely that manipulation of the “graft” is required before reinfusion. The number of stem cells required must be defined. Other preconditioning regimens may be more effective.918 If ASCT is ultimately proven to be effective, patient selection will be critical to its success. Patients should be chosen whose disease can be predicted to have a severe outcome but who are not yet at the stage of severe, irreversible damage. The development of prognostic markers is critical for proper selection of candidates. The ethical issues of attempting a procedure with a mortality rate of at least 5% in children with chronic diseases but much lower predicted mortality rates are monumental.919 The long-term risk of immunosuppression is significant, and safer ways to provide immunosuppression need to be developed. ACKNOWLEDGMENT The author wishes to express his gratitude to Madlen Gazarian for her participation in the development of this chapter in the previous edition.
C H A P T E R
5 PHARMACOLOGY
R E F E R E N C E S 29. 1. Gilman AG, Rall TW, Nies AS, Taylor P (eds): The Pharmacologic Basis of Therapeutics, 8th ed. New York, Pergamon Press, 1990. 2. Spino M: Pediatric dosing rules and nomograms. In MacLeod SM, Radde IC (eds): Textbook of Pediatric Clinical Pharmacology. Littleton, MA, PSG, 1985. 3. Hardman JG, Limbird LE, Molinoff PB, et al: Goodman and Gilman’s The Pharmacological Basis of Therapeutics, 9th ed. New York, McGraw-Hill, 1996. 4. Radde IC: Renal elimination of drugs during development. In MacLeod SM, Radde IC (eds): Textbook of Pediatric Clinical Pharmacology. Littleton, MA, PSG, 1985. 5. Hollingworth P: The use of non-steroidal anti-inflammatory drugs in paediatric rheumatic diseases. Br J Rheumatol 32: 73–77, 1993. 6. Kiss MH, Reiff AA, Reicin AS, et al: Rofecoxib demonstrates efficacy and tolerability in children and adolescents (ages 2–17 years) with juvenile rheumatoid arthritis (JRA) in a 12 week randomized study. Arthritis Rheum 48: S650, 2003. 6a. Mamdani M, Juurlink DN, Lee DS et al: Cyclo-oxygenase-2 inhibitors versus non-selective non-steroidal anti-inflammatory drugs and congestive heart failure outcomes in elderly patients: a population-based cohort study. Lancet 363: 1751–1756, 2004. 6b. Solomon DH, Schneeweiss S, Levin R, Avorn J: Relationship between COX2 specific inhibitors and hypertension. Hypertension 44: 140–145, 2004. 6c. Levesque LE, Brophy JM, Zhang B: The risk for myocardial infarction with cyclooxygenase-2 inhibitors: a population study of elderly adults. Ann Intern Med 2005, in press. 7. Vane JR, Botting RM: The mode of action of anti-inflammatory drugs. Postgrad Med J 66: S2, 1990. 8. Brooks PM, O’Day RO: Nonsteroidal antiinflammatory drugs: differences and similarities. N Engl J Med 324: 1716–1725, 1991. 9. Hawkey CJ: COX-2 inhibitors. Lancet 353: 307–314, 1999. 10. Donnelly MT, Hawkey CJ: Review article: COX-II inhibitors—a new generation of safer NSAIDs? Aliment Pharmacol Ther 11: 227–236, 1997. 11. Bolten WW: Scientific rationale for specific inhibition of COX-2. J Rheumatol 25 (Suppl 51): 2–7, 1998. 12. Crofford LJ: COX-1 and COX-2 tissue expression: implications and predictions. J Rheumatol 24 (Suppl 49): 15–19, 1997. 13. Needleman P, Isakson PC: The discovery and function of COX-2. J Rheumatol 24 (Suppl 49): 6–8, 1997. 14. Wallace JL, Reuter BK, McKnight W, Bak A: Selective inhibitors of cyclooxygenase-2: are they really effective, selective, and GI-safe? J Clin Gastroenterol 27 (Suppl): S28–S34, 1998. 15. Lipsky PE, Abramson SB, Crofford L, et al: The classification of cyclooxygenase inhibitors. J Rheumatol 25: 2298–2303, 1998. 15a. Juni P, Nartey L, Reichenbach S, et al: Risk of cardiovascular events and rofecoxib: cumulative meta-analysis. Lancet 364: 2021–2029, 2004. 15b. Burklow J, Ralbovsky D: NIH halts use of COX-2 inhibitor in large cancer prevention trial. Bethesda, MD: US Department of Health and Human Services, National Institutes of Health; 2004. 15c. Finckh A, Aronson MD: Cardiovascular risks of cyclooxygenase inhibitors: where we stand now. Ann Intern Med 142: 212–214, 2005. 16. Hochberg MC: COX-2: Where are we in 2003? Be strong and resolute: continue to use COX-2 selective inhibitors at recommended dosages in appropriate patients. Arthritis Res Ther 5: 28–31, 2003. 17. Lentini A, Ternai B, Ghosh P: Synthetic inhibitors of human leucocyte elastase 4: inhibition of human granulocyte elastase and cathepsin G by nonsteroidal anti-inflammatory drugs (NSAIDs). Biochem Int 15: 1069, 1987. 18. Haggag AA, Mohamed HF, Eldawy MA, Elbahrawy H: Biochemical studies on anti-inflammatory activity of salicylates as superoxide radical scavengers. IRCS Med Sci Biochem 14: 1104, 1986. 19. Minta JO, Williams MD: Some nonsteroidal antiinflammatory drugs inhibit the generation of superoxide anions by activated polymorphs by blocking ligand-receptor interactions. J Rheumatol 12: 751, 1985. 20. Biemond P, Swaak AJ, Penders JMA, et al: Superoxide production by polymorphonuclear leucocytes in rheumatoid arthritis and osteoarthritis: in vivo inhibition by the antirheumatic drug piroxicam due to interference with the activation of the NADPH-oxidase. Ann Rheum Dis 17: 170, 1986. 21. Paulus HE: Pharmacological considerations. In Roth SH (ed): Handbook of Drug Therapy in Rheumatology. Littleton, MA, PSG, 1985. 22. Weiss B, Hait WN: Selective cyclic nucleotidephosphodiesterase inhibitors as potential therapeutic agents. Annu Rev Pharmacol Toxicol 17: 441, 1977. 23. Simon LS, Mills JA: Nonsteroidal antiinflammatory drugs. N Engl J Med 302: 1179, 1980. 24. Mielke K, Otto P, Platz CM: Current Concepts of Antiinflammatory Drugs. New York, Biomedical Information, 1980. 25. Kanton TG: Ibuprofen. Ann Intern Med 91: 877, 1979. 26. Liauw HL, Moscaritola JD, Burcher J: Diclofenac sodium (Voltaren). In Lewis AJ, Furst DE (eds): Anti-Inflammatory Agents. New York, Marcel Dekker, 1987. 27. Dahl SI, Ward JR: Pharmacology, clinical efficacy and adverse effects of piroxicam, a new nonsteroidal anti-inflammatory agent. Pharmacotherapy 2: 80, 1982. 28. Montecucco C, Mazzone A, Pasotti D, et al: Effects of piroxicam therapy on granulocyte function and granulocyte elastase concentration in peripheral
30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58.
59. 60. 61. 62.
AND
D RU G T H E R A P Y
127
blood and synovial fluid of rheumatoid arthritis patients. Inflammation 13: 211, 1989. Maclean JR, Gluckman MI: On the mechanism of the pharmacologic activity of meclofenamate sodium. Arzneimittelforschung 33: 627, 1983. Boctor AM, Eickholt M, Pugsley TA: Meclofenamate sodium is an inhibitor of both the 5-lipoxygenase and cylooxygenase pathways of the arachidonic acid cascade in vitro. Prostaglandins Leukot Med 23: 229, 1986. Furst D: Are there differences among nonsteroidal antiiflammatory drugs? Arthritis Rheum 37: 1–9, 1994. Walson PD, Mortensen ME: Pharmacokinetics of common analgesics, antiinflammatories and antipyretics in children. Clinical Pharmacokinetics 17 (Suppl 1): 116–137, 1989. Skeith KJ, Jamali F: Clinical pharmacokinetics of drugs used in juvenile arthritis. Clin Pharmacokinet 21: 129–149, 1991. Davies NM, Anderson KE: Clinical pharmacokinetics of naproxen. Clin Pharmacokinet 32: 268–293, 1997. Wells TG, Mortensen ME, Dietrich A, et al: Comparison of the pharmacokinetics of naproxen tablets and suspension in children. J Clin Pharmacol 34: 30–33, 1994. Graham GG: Pharmacokinetics and metabolism of non-steroidal antiinflammatory drugs. Med J Aust 147: 597–602, 1987. Sellers EM: Plasma protein displacement reactions are rarely of clinical significance. Pharmacology 18: 225–227, 1979. Dupuis LL, Koren G, Shore A, et al: Methotrexate-nonsteroidal antiinflammatory drug interaction in children with arthritis. J Rheumatol 17: 1469–1473, 1990. American Medical Association: Drug Evaluation Annual: Antiarthritic Drugs. Chicago, American Medical Association, 1992. Giannini EH, Cawkwell GD: Drug treatment in children with juvenile rheumatoid arthritis. Pediatr Rheumatol 42: 1099–1125, 1995. Lindsley CB: Uses of nonsteroidal anti-inflammatory drugs in pediatrics. Am J Dis Child 147: 229–236, 1993. Barron KS, Person DA, Brewer EJ: The toxicity of nonsteroidal antiinflammatory drugs in juvenile arthritis. J Rheumatol 9: 149–155, 1982. O’Brien WM, Bagby GF: Rare adverse reactions to nonsteroidal antiinflammatory drugs. J Rheumatol 12: 13–20, 1985. Dowd JE, Cimaz R, Fink CW: Nonsteroidal antiinflammatory drug-induced gastroduodenal injury in children. Arthritis Rheum 38: 1225–1231, 1995. Laxer RM, Silverman ED: The pharmacological management of juvenile chronic arthritis. Bailliere’s Clin Paediatr 1: 3825–3873, 1993. Lovell DJ, Giannini EH, Brewer EJ: Time course of response to nonsteroidal antiinflammatory drugs in juvenile rheumatoid arthritis. Arthritis Rheum 27: 1433–1437, 1984. Szer IS: Chronic arthritis in children. Comp Ther 23: 124–129, 1997. Goodman S, Howard P, Haig A, et al: An open label study to establish dosing recommendations for nabumetone in juvenile rheumatoid arthritis. J Rheumatol 30: 829–831, 2003. Rapoff MA, Belmont J, Lindsley C, et al: Prevention of nonadherence to nonsteroidal anti-inflammatory medications for newly diagnosed patients with juvenile rheumatoid arthritis. Health Psychol 21: 620–623, 2002. Kroll T, Barlow JH, Shaw K: Treatment adherence in juvenile rheumatoid arthritis: a review. Scand J Rheumatol 28: 10–18, 1999. Schlegel SI, Paulus HE: Nonsteroidal antiinflammatory drugs: use in rheumatic disease, side effects and interactions. Bull Rheum Dis 36: 1, 1986. Bombardier C, Chalmers A, Jamali F: The monitoring of clinical and pharmacological effects of antiinflammatory drugs in populations [proceedings of workshops]. J Rheumatol 15: 17, 1988. Wolfe MM, Lichtenstein DR, Singh G: Gastrointestinal toxicity of nonsteroidal antiinflammatory drugs. N Engl J Med 340: 1888–1899, 1999. Singh G, Ramey DR: NSAID induced gastrointestinal complications: The ARAMIS perspective, 1997. J Rheumatol 51: 8–16, 1998. Paulus HE: FDA Arthritis Advisory Committee meeting: serious gastrointestinal toxicity of nonsteroidal antiinflammatory drugs. Arthritis Rheum 31: 1450–1451, 1988. Garcia Rodriguez LA, Jick H: Risk of upper gastrointestinal bleeding and perforation associated with individual non-steroidal anti-inflammatory drugs. Lancet 343: 769–772, 1994. Fries JF: NSAID gastropathy: the second most deadly rheumatic disease? J Rheumatol 18: 6–10, 1991. Silverstein FE, Graham DY, Senior JR, et al: Misoprostol reduces serious gastrointestinal complications in patients with rheumatoid arthritis receiving nonsteroidal anti-inflammatory drugs: a randomized, double-blind, placebo-controlled trial. Ann Intern Med 123: 241–249, 1995. Barkin J: The relation between Helicobacter pylori and nonsteroidal antiinflammatory drugs. Am J Med 105: 22S–27S, 1998. Shabib S, Laxer R, Silverman E, et al: Seroprevalence of Helicobacter pylori infection is not increased in pediatric inflammatory arthritides. J Rheumatol 21: 1548–1552, 1994. Keenan GF, Giannini EH, Athreya BH: Clinically significant gastropathy associated with nonsteroidal antiinflammatory drug use in children with juvenile rheumatoid arthritis. J Rheumatol 22: 1149–1151, 1995. Duffy CM, Gibbon M, Yang H, et al: Non-steroidal antiinflammatory druginduced gastrointestinal toxicity in a practice-based cohort of children with juvenile arthritis. J Rheumatol 27 (Suppl 58): 73, 2000.
128
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
63. Ashorn M, Verronen P, Ruuska T, Huhtala H: Upper endoscopic findings in children with active juvenile chronic arthritis. Acta Paediatr 92: 558–561, 2003 64. Mulberg AE, Linz C, Bern E, et al: Identification of nonsteroidal antiinflammatory drug-induced gastroduodenal injury in children with juvenile rheumatoid arthritis. J Pediatr 122: 647–649, 1993. 65. Hermaszewski R, Hayllar J, Woo P: Gastro-duodenal damage due to nonsteroidal anti-inflammatory drugs in children. Br J Rheumatol 32: 69–72, 1993. 66. Henry D, Lim L, Garcia Rodriguez LA, et al: Variability in risk of gastrointestinal complications with individual non-steroidal anti-inflammatory drugs: results of a collaborative meta-analysis. BMJ 312: 1563–1566, 1996. 67. Bombardier C: An evidence-based evaluation of the gastrointestinal safety of coxibs. Am J Cardiol 89(6A), 3D–9D, 2002. 68. Cole AT, Hudson N, Liew LCW, et al: Protection of human gastric mucosa against aspirin: enteric coating or dose reduction? Aliment Pharmacol Ther 13: 187–193, 1999. 69. Silverstein FE, Faich G, Goldstein JL, et al: Gastrointestinal toxicity with celecoxib vs nonsteroidal anti-inflammatory drugs for osteoarthritis and rheumatoid arthritis: the CLASS study, a randomized controlled trial. Celecoxib Long-term Arthritis Safety Study. JAMA 284: 1247–1255, 2000. 70. Bombardier C, Laine L, Reicin A, et al: Comparison of upper gastrointestinal toxicity of rofecoxib and naproxen in patients with rheumatoid arthritis. VIGOR Study Group. N Engl J Med 343: 1520–1528, 2000. 71. Agrawal NM, Roth S, Graham DY, et al: Misoprostol compared with sucralfate in the prevention of nonsteroidal anti-inflammatory drug induced gastric ulcer. Ann Intern Med 115: 195–200, 1991. 72. Agrawal NM, Aziz K: Prevention of gastrointestinal complications associated with nonsteroidal antiinflammatory drugs. J Rheumatol 51: 17–20, 1998. 73. Roth S, Agrawal N, Mahowald M, et al: Misoprostol heals gastroduodenal injury in patients with rheumatoid arthritis receiving aspirin. Arch Intern Med 149: 775–779, 1989. 74. Ballinger AB, Kumar PJ, Scott DL: Misoprostol in the prevention of gastroduodenal damage in rheumatology. Ann Rheum Dis 51: 1089–1093, 1992. 75. Gazarian M, Berkovitch M, Koren G, et al: Experience with misoprostol therapy for NSAID gastropathy in children. Ann Rheum Dis 54: 277–280, 1995. 76. Shield MJ: Diclofenac/Misoprostol: novel findings and their clinical potential. J Rheumatol 51: 31–41, 1998. 77. Hawkey CJ, Karrasch J, Szczepanski L, et al: Omeprazole compared with misoprostol for ulcers associated with nonsteroidal antiinflammatory drugs. N Engl J Med 338: 727–734, 1998. 78. Yeomans ND, Tulassay Z, Juhasz L, et al: A comparison of omeprazole with ranitidine for ulcers associated with nonsteroidal antiinflammatory drugs. N Engl J Med 338: 719–726, 1998. 79. Bernstein BH, Singsen BH, King KK, Hanson V: Aspirin-induced hepatotoxicity and its effect on juvenile rheumatoid arthritis. Am J Dis Child 131: 659–663, 1977. 80. Stephan JL, Kone-Paut I, Galambrun C, et al: Reactive haemophagocytic syndrome in children with inflammatory disorders: a retrospective study of 24 patients. Rheumatology (Oxf) 40: 1285–1292, 2001. 81. Sawhney S, Woo P, Murray KJ: Macrophage activation syndrome: a potentially fatal complication of rheumatic disorders. Arch Dis Child 85: 421–426, 2001. 82. Lindsley CB, Warady BA: Nonsteroidal antiinflammatory drugs: Renal toxicity. Review of pediatric issues. Clin Pediatr 29: 10–13, 1990. 83. Carmichael J, Shankel SW: Effects of nonsteroidal anti-inflammatory drugs on prostaglandins and renal function. Am J Med 78: 992–1000, 1985. 84. Clive DM, Stoff JS: Renal syndromes associated with nonsteroidal antiinflammatory drugs. N Engl J Med 310: 563, 1984. 85. Schrier RW, Henrich WL: Nonsteroidal anti-inflammatory drugs: Caution still indicated. JAMA 251: 1301–1302, 1984. 86. Becker-Cohen R, Frishberg Y: Severe reversible renal failure due to naproxenassociated acute interstitial nephritis. Eur J Pediatr 160: 293–295, 2001. 87. Allen RC, Petty RE, Lirenman DS, et al: Renal papillary necrosis in children with chronic arthritis. J Pediatr 97: 37, 1986. 88. Ray PE, Rigolizzo D, Wara DR, Piel CF: Naproxen nephrotoxicity in a 2 year old child. Am J Dis Child 142: 524–525, 1988. 89. Robinson J, Malleson P, Lirenman D, Carter J: Nephrotic syndrome associated with nonsteroidal anti-inflammatory drug use in two children. Pediatrics 85: 844–847, 1990. 90. Szer IS, Goldstein-Schainberg C, Kurtin PS: Paucity of renal complications associated with nonsteroidal antiinflammatory drugs in children with chronic arthritis. J Pediatr 119: 815–817, 1991. 91. Haftel HM, Mitchell JM, Adams BS: Incidence of renal toxicity from antiinflammatory medications in a pediatric rheumatology population: role of routine screening of urine. J Rheumatol 27 (Suppl 58): 73, 2000. 92. Kordonouri O, Dracou C, Papadellis F, et al: Glomerular microproteinuria in children treated with nonsteroidal anti-inflammatory drugs for juvenile chronic arthritis. Clin Exp Rheumatol 12: 567–571, 1994. 93. Malleson PN, Lockitch G, Mackinnon M, et al: Renal disease in chronic arthritis of childhood: a study of urinary N-acetyl-beta-glucosaminidase and beta2-microglobulin excretion. Arthritis Rheum 33: 1560–1566, 1990.
94. Garella S, Matarese RA: Renal effects of prostaglandins and clinical adverse effects of nonsteroidal anti-inflammatory agents. Medicine (Baltimore) 63: 165, 1984. 95. De Broe ME, Elseviers MM: Analgesic nephropathy. N Engl J Med 338: 446–452, 1998. 96. Dunn MJ, Patrono C: Renal effects of nonsteroidal antiinflammatory drugs [proceedings of a symposium]. Am J Med 81: 2B, 1986. 97. Husserl FE, Lange RK, Kantrow CM: Renal papillary necrosis and pyelonephritis accompanying fenoprofen therapy. JAMA 242: 1896, 1979. 98. Whelton A, Hamilton CW: Nonsteroidal anti-inflammatory drugs: effects on kidney function. J Clin Pharmacol 31: 588, 1991. 99. Sedor JR, Davidson EW, Dunn MJ: Renal effects of nonsteroidal antiinflammatory drugs in healthy subjects. Am J Med 81: 58, 1986. 100. Wortmann DW, Kelsch RC, Kuhns L, et al: Renal papillary necrosis in juvenile rheumatoid arthritis. J Pediatr 97: 37–40, 1980. 101. Hoppmann RA, Peden JG, Ober SK: Central nervous system side effects of nonsteroidal anti-inflammatory drugs. J Intern Med 151: 1309–1313, 1991. 102. Duffy CM, Gibbon M, Yang H, et al: Non-steroidal anti-inflammatory druginduced central nervous system toxicity in a practice-based cohort of children with juvenile arthritis. J Rheumatol 27 (Suppl 58): 73, 2000. 103. Bigby M, Stern R: Cutaneous reactions to nonsteroidal antiinflammatory drugs. J Am Acad Dermatol 12: 866–876, 1985. 104. Howard AM, Dowling J, Varigos G: Pseudoporphyria due to naproxen. Lancet 2: 819–820, 1985. 105. Allen R, Rogers M, Humphrey I: Naproxen induced pseudoporphyria in juvenile chronic arthritis. J Rheumatol 18: 893–896, 1991. 106. Creemers MCW, Chang A, Franssen MJ, et al: Pseudoporphyria due to naproxen. Scand J Rheumatol 24: 185–187, 1995. 107. Lang BA, Finlayson LA: Naproxen-induced pseudoporphyria in patients with juvenile rheumatoid arthritis. J Pediatr 124: 639–642, 1994. 108. Levy ML, Barron KS, Eichenfield A, Honig PJ: Naproxen-induced pseudoporphyria: a distinctive photodermatitis. J Pediatr 117: 660–664, 1990. 109. De Silva B, Banney L, Uttley W, et al: Pseudoporphyria and nonsteroidal antiinflammatory agents in children with juvenile idiopathic arthritis. Pediatr Dermatol 17: 480–483, 2000. 110. Mehta S, Lang B: Long-term followup of naproxen-induced pseudoporphyria in juvenile rheumatoid arthritis. Arthritis Rheum 42: 2252–2254, 1999. 111. Reye RDK, Morgan G, Baral J, et al: Encephalopathy and fatty degeneration of the viscera: a disease entity in childhood. Lancet 2: 749–752, 1963. 112. Kauffman RE: Commentary: Reye’s syndrome and salicylate use, by Karen M. Starko, MD, et al, Pediatrics 66:859–864, 1980, and National patterns of aspirin use and Reye syndrome reporting, United States, 1980–1985, by Janet B. Arrowsmith et al, Pediatrics 79:858–863, 1987. Pediatrics 102: 259–262, 1998. 113. Belay ED, Bresee JS, Holman RC, et al: Reye’s syndrome in the United States from 1981 through 1997. N Engl J Med 340: 1377–1382, 1999. 114. Starko KM, Ray CG, Dominguez LB, et al: Reye’s syndrome and salicylate use. Pediatrics 66: 859–864, 1980. 115. Forsyth BW, Horwitz RI, Acampora D, et al: New epidemiological evidence confirming that bias does not explain the aspirin/Reye’s syndrome association. JAMA 261: 2517–2524, 1989. 116. Hurwitz ES, Barrett MJ, Bregman D, et al: Public Health Service study of Reye’s syndrome and medications: report of main study. N Engl J Med 257: 1905–1911, 1987. 117. Hardie RM, Newton LH, Bruce JC, et al: The changing clinical pattern of Reye’s syndrome, 1982–1990. Arch Dis Child 74: 400–405, 1996. 118. Remington PL, Shabino CL, McGee H, et al: Reye syndrome and juvenile rheumatoid arthritis in Michigan. Am J Dis Child 139: 870–872, 1985. 119. Rennebohm RM, Heubi JE, Daugherty CC, Daniels SR: Reye syndrome in children receiving salicylate therapy for connective tissue disease. J Pediatr 107: 877–880, 1985. 120. MMWR: Prevention and control of influenza: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Morb Mortal Wkly Rep 47 (RR-6): 1–26, 1998. 121. Morassut P, Yang W, Karsh J: Aspirin intolerance. Semin Arthritis Rheum 19 (22–30), 1989. 122. Oates JA, Fitzgerald GA, Branch RA, et al: Clinical implications of prostaglandin and thromboxane A2 formation. N Engl J Med 319: 689–698, 1988. 123. Furst DE: Toxicity of antirheumatic medications in children with juvenile arthritis. J Rheumatol 33: 11–15, 1992. 124. Slepian IK, Mathews KP, McLean JA: Aspirin-sensitive asthma. Chest 87: 386, 1985. 125. Rachelefsky GS, Coulson A, Siegel SC, Stiehm ER: Aspirin intolerance in chronic childhood asthma: detected by oral challenge. Pediatrics 56: 443, 1975. 126. Fischer TJ, Guilfoile TD, Kesarwala HH, et al: Adverse pulmonary responses to aspirin and acetaminophen in chronic childhood asthma. Pediatrics 71: 313, 1983. 127. Szceklik A: Analgesics, allergy, and asthma. Drugs 32 (Suppl 4): 148–163, 1986. 128. Squires JE, Mintz PD, Clark S: Tolmetin-induced haemolysis. Transfusion 25: 410–412, 1985.
C H A P T E R 129. American Hospital Formulary Service: Drug Information 88. Bethesda, MD: American Society of Hospital Pharmacy, 1988. 130. Lasagna L, McMahon FG (eds): New perspectives on aspirin therapy [proceedings of a symposium]. Am J Med 18 (Suppl): 1983. 131. Burch JW, Stanford N, Majerus PW: Inhibition of platelet prostaglandin synthetase by oral aspirin. J Clin Invest 61: 314, 1978. 132. Preston FE, Whipps S, Jackson CA, et al: Inhibition of prostacyclin and platelet thromboxane A2 after low-dose aspirin. N Engl J Med 304: 76, 1981. 133. Pedersen AK, Fitzgerald GA: Dose-related kinetics of aspirin: presystemic acetylation of platelet cyclooxygenase. N Engl J Med 311: 1206, 1984. 134. Yu T-F, Gutman AB: Study of the paradoxical effects of salicylate in low, intermediate and high dosage on the renal mechanisms for excretion of urate in man. J Clin Invest 38: 1298, 1959. 135. Levy G: Clinical pharmacokinetics of aspirin. Pediatrics 62: 867, 1978. 136. Done AK, Yaffe SJ, Clayton JM: Aspirin dosage for infants and children. J Pediatr 95: 617, 1979. 137. Mandelli M, Tognoni G: Monitoring plasma concentration of salicylate. Clin Pharmacokinet 5: 424, 1980. 138. Kvien TK, Olsson B, Hoyeraal HM: Acetylsalicylic acid and juvenile rheumatoid arthritis: effect of dosage interval on the serum salicylic acid level. Acta Paediatr Scand 74: 755, 1985. 139. Pachman LM, Olufs R, Procknal JA, et al: Pharmacokinetic monitoring of salicylate therapy in children with juvenile rheumatoid arthritis. Arthritis Rheum 22: 826, 1979. 140. Poe TE, Mutchie KD, Saunders GH, et al: Total and free salicylate concentrations in juvenile rheumatoid arthritis. J Rheumatol 7: 717, 1980. 141. Bardara M, Cislaghi GU, Mandelli M, et al: Value of monitoring plasma salicylate levels in treating juvenile rheumatoid arthritis. Arch Dis Child 53: 381, 1978. 142. Mofenson HC, Caraccio RE: Salicylate poisoning. In Gellis SS, Kagan BM (eds): Current Pediatric Therapy. Philadelphia, WB Saunders, 1990. 143. Goulden KJ, Dooley JM, Camfield PR, Fraser AD: Clinical valproate toxicity induced by acetylsalicylic acid. Neurology 37: 1392, 1987. 144. Verbeeck RK: Pharmacokinetic drug interactions with nonsteroidal antiinflammatory drugs. Clin Pharmacokinet 19: 44, 1990. 145. Marsh CC, Schuna AA, Sundstrom WR: A review of selected investigational nonsteroidal anti-inflammatory drugs in the 1980’s. Pharmacotherapy 6: 10, 1986. 146. Needs CJ, Brooks PM: Clinical pharmacokinetics of the salicylates. Clin Pharmacokinet 10: 164, 1985. 147. Wallace CA, Levinson JE: Juvenile rheumatoid arthritis: outcome and treatment for the 1990s. Rheum Dis Clin North Am 17: 891–905, 1991. 148. Giannini E, Brewer E, Kuzmina N, et al, for the Pediatric Rheumatology Collaborative Study Group and the Cooperative Children’s Study Group: Methotrexate in resistant juvenile rheumatoid arthritis: results of the USAUSSR double-blind, placebo-controlled trial. N Engl J Med 326: 1043–1049, 1992. 149. Rose C, Singsen B, Eichenfield A, et al: Safety and efficacy of methotrexate therapy in juvenile rheumatoid arthritis. J Pediatr 117: 653, 1990. 150. Graham LD, Myones BL, Rivas-Chacon RF, Pachman LM: Morbidity associated with long-term methotrexate therapy in juvenile rheumatoid arthritis. J Pediatr 120: 468–473, 1992. 151. Ruperto N, Murray KJ, Gerloni V, et al: A randomized trial of parenteral methotrexate comparing an intermediate dose with a higher dose in children with juvenile idiopathic arthritis who failed to respond to standard doses of methotrexate. Arthritis Rheum 50: 2191–2201, 2004. 152. Singsen BH, Goldbach-Mansky R: Methotrexate in the treatment of juvenile rheumatoid arthritis and other pediatric rheumatic and nonrheumatic disorders. Rheum Dis Clin North Am 23: 811–841, 1997. 153. Kremer JM: Toward a better understanding of methotrexate. Arthritis Rheum 50: 1370–1382, 2004. 154. Baggott JE, Morgan SL, Ha T-S, et al: Antifolates in rheumatoid arthritis: a hypothetical mechanism of action. Clin Exp Rheumatol 11 (Suppl 8): S101–S105, 1993. 155. Cronstein BN, Eberle MA, Gruber HE, et al: Methotrexate inhibits neutrophil function by stimulating adenosine release from connective tissue cells. Proc Natl Acad Sci U S A 88: 241–245, 1991. 156. Cronstein BN: The mechanism of action of methotrexate. Rheum Dis Clin North Am 23: 739–751, 1997. 157. Chan ES, Cronstein BN: Molecular action of methotrexate in inflammatory diseases. Arthritis Res 4: 266–273, 2002 158. Firestein GS, Paine MM, Boyle DL: Mechanisms of methotrexate action in rheumatoid arthritis: selective decrease in synovial collagenase gene expression. Arthritis Rheum 37: 193–200, 1994. 159. Ravelli A, Di Fuccia G, Molinaro M, et al: Plasma levels after oral methotrexate in children with juvenile rheumatoid arthritis. J Rheumatol 20: 1573–1577, 1993. 160. Herman RA, Veng-Pedersen P, Hoffman J, et al: Pharmacokinetics of low dose methotrexate in rheumatoid arthritis patients. J Pharm Sci 78: 165, 1989. 161. Furst DE: Practical clinical pharmacology and drug interactions of low-dose methotrexate therapy in rheumatoid arthritis. Br J Rheumatol 34 (Suppl 2): 20–25, 1995. 162. Godfrey C, Sweeney K, Miller K, et al: The population pharmacokinetics of long-term methotrexate in rheumatoid arthritis. Br J Clin Pharmacol 46: 369–376, 1998.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
129
163. Dupuis LL, Koren G, Silverman ED, Laxer RM: Influence of food on the bioavailability of oral methotrexate in children. J Rheumatol 22: 1570–1573, 1995. 164. Jundt JW, Browne BA, Fiocco GP, et al: A comparison of low dose methotrexate bioavailability: oral solution, oral tablet, subcutaneous and intramuscular dosing. J Rheumatol 20: 1845–1849, 1993. 165. Hillson JL, Furst DE: Pharmacology and pharmacokinetics of methotrexate in rheumatic disease: practical issues in treatment and design. Rheum Dis Clin North Am 23: 757–778, 1997. 166. Angelis-Stoforidis P, Vajda FJ, Christophidis N: Methotrexate polyglutamate levels in circulating erythrocytes and polymorphs correlate with clinical efficacy in rheumatoid arthritis. Clin Exp Rheumatol 17: 313–320, 1999. 167. Press J, Berkovitch M, Laxer R, et al: Evaluation of therapeutic drug monitoring of methotrexate in saliva of children with rheumatic diseases. Ther Drug Monit 17: 247, 1995. 168. Albertioni F, Flato B, Seideman P, et al: Methotrexate in juvenile rheumatoid arthritis: evidence of age dependent pharmacokinetics. Eur J Clin Pharmacol 47: 507–511, 1995. 169. Reiff A, Shaham B, Wood BP, et al: High dose methotrexate in the treatment of refractory juvenile rheumatoid arthritis. Clin Exp Rheumatol 13: 113–118, 1995. 170. Wallace CA, Sherry DD: Preliminary report of higher dose methotrexate treatment in juvenile rheumatoid arthritis. J Rheumatol 19: 1604–1607, 1992. 171. Wallace CA, Smith AL, Sherry DD: Pilot investigation of naproxen/ methotrexate interaction in patients with juvenile rheumatoid arthritis. J Rheumatol 20: 1764–1768, 1993. 172. Giannini E, Cassidy J, Brewer E, et al: Comparative efficacy and safety of advanced drug therapy in children with juvenile rheumatoid arthritis. Semin Arthritis Rheum 23: 34, 1993. 173. Takken T, Van Der Net J, Helders PJ: Methotrexate for treating juvenile idiopathic arthritis. Cochrane Database Syst Rev 4: CD003129, 2001. 174. Woo P, Southwood TR, Prieur AM, et al: Randomized, placebo-controlled, crossover trial of low-dose oral methotrexate in children with extended oligoarticular or systemic arthritis. Arthritis Rheum 43: 1849–1857, 2000. 175. Ravelli A, Viola S, Migliavacca D, et al: The extended oligoarticular subtype is the best predictor of methotrexate efficacy in juvenile idiopathic arthritis. J Pediatr 135: 316–320, 1999. 176. al-Sewairy W, al-Mazyed A, al-Dalaan, et al: Methotrexate therapy in systemic-onset juvenile rheumatoid arthritis in Saudi Arabia: a retrospective analysis. Clin Rheumatol 17: 52–57, 1998. 177. Halle F, Prieur AM: Evaluation of methotrexate in the treatment of juvenile chronic arthritis according to the subtype. Clin Exp Rheumatol 9: 297–302, 1991. 178. Harel L, Wagner-Weiner L, Poznanski AK, et al: Effects of methotrexate on radiologic progression in juvenile rheumatoid arthritis. Arthritis Rheum 36: 1370–1374, 1993. 179. Ravelli A, Viola S, Ramenghi B, et al: Radiologic progression in patients with juvenile chronic arthritis treated with methotrexate. J Pediatr 133: 262–265,1998. 180. Ravelli A, Ramenghi B, Di Fuccia G, et al: Factors associated with response to methotrexate in systemic-onset juvenile chronic arthritis. Acta Paediatr 83: 428–432, 1994. 181. Stenger AA, Van Leeuwen MA, Houtman PM, et al: Early effective suppression of inflammation in rheumatoid arthritis reduces radiographic progression. Br J Rheumatol 37: 1157–1163, 1998. 182. Ravelli A, Ballardini G, Viola S, et al: Methotrexate therapy in refractory pediatric onset systemic lupus erythematosus. J Rheumatol 25: 572–575, 1998. 183. Shetty AK, Stopa AR, Gedalia A: Low-dose methotrexate as a steroid-sparing agent in a child with Takayasu’s arteritis. Clin Exp Rheumatol 16: 335–336,1998. 184. Langford CA, Talar-Williams C, Barron KS, Sneller MC: Use of a cyclophosphamide-induction methotrexate-maintenance regimen for the treatment of Wegener’s granulomatosis: extended follow-up and rate of relapse. Am J Med 114: 463–469, 2003. 185. Shetty AK, Zganjar BE, Ellis GS Jr, et al: Low-dose methotrexate in the treatment of severe juvenile rheumatoid arthritis and sarcoid iritis. J Pediatr Ophthalmol Strabismus 36: 125–128, 1999. 186. Lin AT, Clements PJ, Furst DE: Update on disease-modifying antirheumatic drugs in the treatment of systemic sclerosis. Rheum Dis Clin North Am 29: 409–426, 2003. 187. Uziel Y, Feldman BM, Krafchik BR, et al: Methotrexate and corticosteroid therapy for pediatric localized scleroderma. J Pediatr 136: 91–95, 2000. 188. Weiss AH, Wallace CA, Sherry DD: Methotrexate for resistant chronic uveitis in children with juvenile rheumatoid arthritis. J Pediatr 133: 266–268, 1998. 189. Balis FM, Savitch JL, Bleyer WA: Pharmacokinetics of oral methotrexate in children. Cancer Res 43: 2342–2345, 1983. 190. Balis FM, Mirro J, Reaman GH, et al: Pharmacokinetics of subcutaneous methotrexate. J Clin Oncol 6: 1882–1886, 1988. 191. Alsufyani K, Ortiz-Alvarez O, Cabral DA, et al: The role of subcutaneous administration of methotrexate in children with juvenile idiopathic arthritis who have failed oral methotrexate. J Rheumatol 31: 179–182, 2004.
130
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
192. Hamilton RA, Kremer JM: Why intramuscular methotrexate may be more efficacious than oral dosing in patients with rheumatoid arthritis. Br J Rheumatol 36: 86–90, 1997. 193. Hoekstra M, Haagsma C, Neef C, et al: Bioavailability of higher dose methotrexate comparing oral and subcutaneous administration in patients with rheumatoid arthritis. J Rheumatol 31: 645–648, 2004. 194. Wallace CA, Sherry DD: A practical approach to avoidance of methotrexate toxicity. J Rheumatol 22: 1009–1012, 1995. 195. Ravelli A, Viola S, Ramenghi B, et al: Evaluation of response to methotrexate by a functional index in juvenile chronic arthritis. Clin Rheumatol 14: 322, 1995. 196. Wallace CA, Sherry DD, Mellins ED, Aiken RP: Predicting remission in juvenile rheumatoid arthritis with methotrexate treatment. J Rheumatol 20: 118–122, 1993. 197. Gottlieb BS, Keenan GF, Lu T, Ilowite NT: Discontinuation of methotrexate treatment in juvenile rheumatoid arthritis. Pediatrics 100: 994–997, 1997. 198. Foell D, Frosch M, Schulze zur Wiesch A, et al: Methotrexate treatment in juvenile idiopathic arthritis: when is the right time to stop? Ann Rheum Dis 63: 206–208, 2004. 199. Hunt P, Rose C, Singsen B: Long-term safety and efficacy of methotrexate in juvenile rheumatoid arthritis. Arthritis Rheum 36: S61, 1993. 200. van Ede AE, Laan RFJM, Blom HJ, et al: Methotrexate in rheumatoid arthritis: an update with focus on mechanisms involved in toxicity. Semin Arthritis Rheum 27: 277–292, 1998. 201. McKendry RJR: The remarkable spectrum of methotrexate toxicities. Rheum Dis Clin North Am 23: 939–954, 1997. 202. Sandoval DM, Alarcon GS, Morgan SL: Adverse events in methotrexatetreated rheumatoid arthritis patients. Br J Rheumatol 34 (Suppl 2): 49–56, 1995. 203. West SG: Methotrexate hepatotoxicity. Rheum Dis Clin North Am 23: 883–915, 1997. 204. Wallace CA, Smith AL, Aronson H, Sherry DD: Methotrexate and naproxen kinetics in patients with juvenile rheumatoid arthritis. J Rheumatol 19 (Suppl 33): 112, 1992. 205. Walker AM, Funch D, Dreyer NA, et al: Determinants of serious liver disease among patients receiving low-dose methotrexate for rheumatoid arthritis. Arthritis Rheum 36: 329–335, 1993. 206. Chandran G, Ahern MJ, Hall PM, et al: Cirrhosis in patients with rheumatoid arthritis receiving low-dose methotrexate. Br J Rheumatol 33: 981, 1994. 207. Whiting-O’Keefe QE, Fye KH, Sack KD: Methotrexate and histological abnormalities: a meta-analysis. Am J Med 90: 711–716, 1991. 208. Erickson AR, Reddy V, Vogelgesang SA, West SG: Usefulness of the American College of Rheumatology recommendations for liver biopsy in methotrexate-treated rheumatoid arthritis patients. Arthritis Rheum 38: 1115–1119, 1995. 209. Kremer JM, Alarcon GS, Lightfoot RW, et al: Methotrexate for rheumatoid arthritis: suggested guidelines for monitoring liver toxicity. Arthritis Rheum 37: 316–328, 1994. 210. Hashkes PJ, Balistreri WF, Bove KE, et al: The relationship of hepatotoxic risk factors and liver histology in methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 134: 47–52, 1999. 211. Keim D, Ragsdale C, Heidelberger K, Sullivan D: Hepatic fibrosis with the use of methotrexate for juvenile rheumatoid arthritis. J Rheumatol 17: 846–848, 1990. 212. Kugathasan S, Newman AJ, Dahms BB, Boyle T: Liver biopsy findings in patients with juvenile rheumatoid arthritis receiving long-term, weekly methotrexate therapy. J Pediatr 128: 149–151, 1996. 213. Hashkes PJ, Balistreri WF, Bove KE, et al: The long-term effect of methotrexate therapy on the liver in patients with juvenile rheumatoid arthritis. Arthritis Rheum 40: 2226–2234, 1997. 214. Lahdenne P, Rapola J, Ylijoki H, Haapasaari J: Hepatotoxicity in patients with juvenile idiopathic arthritis receiving longterm methotrexate therapy. J Rheumatol 29: 2442–2445, 2002. 215. Rackham OJ, Sills JA, Davidson JE: Immunoglobulin levels in methotrexate treated paediatric rheumatology patients. Arch Dis Child 87: 147–148, 2002. 216. Kanik KS, Cash JM: Does methotrexate increase the risk of infection or malignancy. Rheum Dis Clin North Am 23: 955–967, 1997. 217. Peter G (ed): 1997 Redbook: report of the Committee on Infectious Diseases, 24th ed. Elk Grove Village, IL, American Academy of Pediatrics, 1997. 218. Guiterrez-Urena S, Molina JF, Garcia CO, et al: Pancytopenia secondary to methotrexate therapy in rheumatoid arthritis. Arthritis Rheum 39: 272, 1996. 219. Ravelli A, Caria MC, Buratti S, et al: Methotrexate as a possible trigger of macrophage activation syndrome in systemic juvenile idiopathic arthritis. J Rheumatol 28: 865–867, 2001. 220. Savolainen HA, Leirisalo-Repo M: Eosinophilia as a side-effect of methotrexate in patients with chronic arthritis. Clin Rheumatol 20: 432–434, 2001. 221. Bleyer WA: Methotrexate induced lymphoma? J Rheumatol 25: 404–407, 1998. 222. Shiroky JB, Frost A, Skelton JD, et al: Complications of immunosuppression associated with weekly low-dose methotrexate.J Rheumatol 18: 1172–1175, 1991. 223. Kingsmore SF, Hall BD, Allen NB, et al: Association of methotrexate, rheumatoid arthritis and lymphoma: report of two cases and literature review. J Rheumatol 19: 1462–1465, 1992.
224. Tennis P, Andrews E, Bombardier C: Record linkage to conduct an epidemiologic study on the association of rheumatoid arthritis and lymphoma in the province of Saskatchewan, Canada. J Clin Epidemiol 46: 685–695, 1993. 225. Kamel OW, van de Rijn M, Weiss LM, et al: Reversible lymphomas associated with Epstein-Barr virus occurring during methotrexate therapy for rheumatoid arthritis and dermatomyositis. N Engl J Med 328: 1317–1321, 1993. 226. Kamel OW, van de Rijn M, LeBrun DP, et al: Lymphoid neoplasms in patients with rheumatoid arthritis and dermatomyositis: frequency of Epstein-Barr virus and other features associated with immunosuppression. Hum Pathol 25: 638–643, 1994. 227. Bailin PL, Tindall JP, Roenigk HH, et al: Is methotrexate therapy for psoriasis carcinogenic? A modified retrospective prospective analysis. JAMA 232: 359–362, 1975. 228. Nysfors A, Jensen H: Frequency of malignant neoplasms in 248 long-term methotrexate-treated psoriatics: a preliminary study. Dermatologica 167: 260–261, 1983. 229. Padeh S, Sharon N, Schiby G, et al: Hodgkin’s lymphoma in systemic onset juvenile rheumatoid arthritis after treatment with low dose methotrexate. J Rheumatol 24: 2035–2037, 1997. 230. Londino AV Jr, Blatt J, Knisely AS: Hodgkin’s disease in a patient with juvenile rheumatoid arthritis taking weekly low dose methotrexate. J Rheumatol 25: 1245–1246, 1998. 231. Krugmann J, Sailer-Hock M, Muller T, et al: Epstein-Barr virus-associated Hodgkin’s lymphoma and Legionella pneumophila infection complicating treatment of juvenile rheumatoid arthritis with methotrexate and cyclosporine A. Hum Pathol 31: 253–255, 2000. 232. Cleary AG, McDowell H, Sills JA: Polyarticular juvenile idiopathic arthritis treated with methotrexate complicated by the development of nonHodgkin’s lymphoma. Arch Dis Child 86: 47–49, 2002. 233. Munro R, Porter DR, Sturrock RD: Lymphadenopathy in a patient with systemic onset juvenile chronic arthritis. Ann Rheum Dis 57: 513–517, 1998. 234. Salaffi F, Manganelli P, Carotti M, et al: Methotrexate-induced pneumonitis in patients with rheumatoid arthritis and psoriatic arthritis: report of five cases and review of the literature. Clin Rheumatol 16: 296–304, 1997. 235. Carroll GJ, Thomas R, Phatouros CC, et al: Incidence, prevalence and possible risk factors for pneumonitis in patients with rheumatoid arthritis receiving methotrexate. J Rheumatol 21: 51–54, 1994. 236. Cron RQ, Sherry DD, Wallace CA: Methotrexate-induced hypersensitivity pneumonitis in a child with juvenile rheumatoid arthritis. J Pediatr 132: 901–902, 1998. 237. Pelucchi A, Lomater C, Gerloni V, et al: Lung function and diffusing capacity for carbon monoxide in patients with juvenile chronic arthritis: effect of disease activity and low dose methotrexate therapy. Clin Exp Rheumatol 12: 675–679, 1994. 238. Camiciottoli G, Trapani S, Castellani W, et al: Effect on lung function of methotrexate and non-steroid anti-inflammatory drugs in children with juvenile rheumatoid arthritis. Rheumatol Int 18: 11–16, 1998. 239. Schmeling H, Stephan V, Burdach S, Horneff G: Pulmonary function in children with juvenile idiopathic arthritis and effects of methotrexate therapy. Z Rheumatol 61: 168–172, 2002. 240. Muzaffer MA, Schneider R, Cameron B, et al: Accelerated nodulosis during methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 128: 698–700, 1996. 241. Falcini F, Taccetti G, Ermini M, et al: Methotrexate-associated appearance and rapid progression of rheumatoid nodules in systemic-onset juvenile rheumatoid arthritis. Arthritis Rheum 40: 175–178, 1997. 242. Merrill JT, Cronstein BN, Shen C, et al: Reversal of new but not old rheumatoid nodules by colchicine: evidence from an in vitro model and case reports of 14 patients. Arthritis Rheum 39: S240, 1996. 243. May KP, West SG, McDermott MT, Huffer WE: The effect of low-dose methotrexate on bone metabolism and histomorphometry in rats. Arthritis Rheum 37: 201–206, 1994. 244. Zonneveld IM, Bakker WK, Dijkstra PF, et al: Methotrexate osteopathy in long term, low-dose methotrexate treatment for psoriasis and rheumatoid arthritis. Arch Dermatol 132: 184, 1996. 245. Preston SJ, Diamond T, Scott A, Laurent MR: Methotrexate osteopathy in rheumatic disease. Ann Rheum Dis 52: 582–585, 1993. 246. Rudler M, Pouchot J, Paycha F, et al: Low dose methotrexate osteopathy in a patient with polyarticular juvenile idiopathic arthritis. Ann Rheum Dis 62: 588–589, 2003. 247. Kozlowski RD, Steinbrunner JV, Mackenzie AH, et al: Outcome of firsttrimester exposure to low-dose methotrexate in eight patients with rheumatic disease. Am J Med 88: 589–592, 1990. 248. Buckley LM, Bullaboy CA, Leichtman L, Marquez M: Multiple congenital anomalies associated with weekly low-dose methotrexate treatment of the mother. Arthritis Rheum 40: 971–973, 1997. 249. Lloyd ME, Carr M, McElhatton P, et al: The effects of methotrexate on pregnancy, fertility and lactation. QJM 92: 551–563, 1999. 250. Morgan SL, Baggott JE, Vaughn WH, et al: The effect of folic acid supplementation on the toxicity of low-dose methotrexate in patients with rheumatoid arthritis. Arthritis Rheum 33: 9–18, 1990. 251. Morgan SL, Baggott JE, Vaughn WH, et al: Supplementation with folic acid during methotrexate therapy for rheumatoid arthritis: a double-blind, placebo-controlled trial. Ann Intern Med 121: 833–841, 1994.
C H A P T E R 252. Shiroky JB, Neville C, Esdaile JM, et al: Low-dose methotrexate with leucovorin (folinic acid) in the management of rheumatoid arthritis: results of a multicenter randomized, double-blind, placebo-controlled trial. Arthritis Rheum 36: 795–803, 1993. 253. Weinblatt ME, Maier AL, Coblyn JS: Low dose leucovorin does not interfere with the efficacy of methotrexate in rheumatoid arthritis: an 8 week randomized placebo controlled trial. J Rheumatol 20: 950–952, 1993. 254. Ortiz Z, Shea B, Suarez-Almazor ME, et al: The efficacy of folic acid and folinic acid in reducing methotrexate gastrointestinal toxicity in rheumatoid arthritis: a metaanalysis of randomized controlled trials. J Rheumatol 25: 36–43, 1998. 255. Hunt PG, Rose CD, McIlvain-Simpson G, Tejani S: The effects of daily intake of folic acid on the efficacy of methotrexate therapy in children with juvenile rheumatoid arthritis: a controlled study. J Rheumatol 24: 2230–2232, 1997. 256. Ravelli A, Migliavacca D, Viola S, et al: Efficacy of folinic acid in reducing methotrexate toxicity in juvenile idiopathic arthritis. Clin Exp Rheumatol 17: 625–627, 1999. 257. Modesto C, Castro L: Folinic acid supplementation in patients with juvenile rheumatoid arthritis treated with methotrexate. J Rheumatol 23: 403–404, 1996. 258. A reassessment of plaquenil in the treatment of rheumatoid arthritis [proceedings of a symposium]. Am J Med 75 (Suppl): 1983. 259. Maksymowych W, Russell AS: Antimalarials in rheumatology: efficacy and safety. Semin Arthritis Rheum 16: 206, 1987. 260. Hughes GRV, Rynes RI: Antimalarials in rheumatic disease: opportunity for an expanding role? Br J Clin Pract 41 (Suppl): 52, 1987. 261. Tett SE, Cutler DJ, Day RO, Brown KF: Bioavailability of hydroxychloroquine tablets in healthy volunteers. Br J Clin Pharmacol 27: 771, 1989. 262. Miyachi Y, Yoshioka A, Imamura S, et al: Antioxidant action of anti-malarials. Ann Rheum Dis 45: 244, 1986. 263. Salmeron G, Lipsky PE: Immunosuppressive potential of antimalarials. Am J Med 75: 19, 1983. 264. Matsuzawa Y, Hostetler KY: Inhibition of lysosomal phospholipase A and phospholipase C by chloroquine and 4c-bis(diethylaminoethyoxy)bdiethyldiphenylethane. J Biol Chem 255: 5190, 1980. 265. Greaves MW, McDonald-Gibson W: Effect of non-steroidal antiinflammatory drugs and antipyretic drugs on prostaglandin biosynthesis by human skin. J Invest Dermatol 61: 127, 1973. 266. van den Borne BE, Dijkmans BA, de Rooij HH, et al: Chloroquine and hydroxychloroquine equally affect tumor necrosis factor-alpha, interleukin 6, and interferon-gamma production by peripheral blood mononuclear cells. J Rheumatol 24: 55–60, 1997. 267. Wallace DJ: Antimalarial agents and lupus. Rheum Dis Clin North Am 20: 243–263, 1994. 268. Meng XW, Feller JM, Ziegler JB, et al: Induction of apoptosis in pheripheral blood lymphocytes following treatment in vitro with hydroxychloroquine. Arthritis Rheum 40: 927–935, 1997. 269. Petri M: Hydroxychloroquine use in the Baltimore Lupus Cohort: effects on lipids, glucose and thrombosis. Lupus 5 (Suppl 1): S16–S22, 1996. 270. Brewer EJ, Giannini EH, Kuzmina N, Alekseev L: Penicillamine and hydroxychloroquine in the treatment of severe juvenile rheumatoid arthritis: results of the U.S.A.-U.S.S.R. double-blind placebo-controlled trial. N Engl J Med 314: 1269–1276, 1986. 271. Suarez-Almazor ME, Belseck E, Shea B, et al: Antimalarials for treating rheumatoid arthritis. Cochrane Database Syst Rev 4: CD000959, 2000. 272. O’Dell JR, Leff R, Paulsen G, et al: Treatment of rheumatoid arthritis with methotrexate and hydroxychloroquine, methotrexate and sulfasalazine, or a combination of the three medications: results of a twoyear, randomized, double-blind, placebo-controlled trial. Arthritis Rheum 46: 1164–1170, 2002. 273. Fries JF, Singh G, Lenert L, Furst DE: Aspirin, hydroxychloroquine, and hepatic enzyme abnormalities with methotrexate in rheumatoid arthritis. Arthritis Rheum 33: 1611–1619, 1990. 274. Combe B, Guttierrez M, Anaya JM, Sany J: Possible efficacy of hydroxychloroquine on accelerated nodulosis during methotrexate therapy for rheumatoid arthritis. J Rheumatol 20: 755–756, 1993. 275. Tsakonas E, Joseph L, Esdaile JM, et al: A long-term study of hydroxychloroquine withdrawal on exacerbations in systemic lupus erythematosus. The Canadian Hydroxychloroquine Study Group. Lupus 7: 80–85, 1998. 276. Hodis HN, Quismorio FPJ, Wickham E, Blankenhorn DH: The lipid, lipoprotein, and apolipoprotein effect of hydroxychloroquine in patients with systemic lupus erythematosus. J Rheumatol 20: 661–665, 1993. 277. Rahman P, Gladman DD, Urowitz MB, et al: The cholesterol lowering effect of antimalarial drugs is enhanced in patients with lupus taking corticosteroid drugs. J Rheumatol 26: 325–330, 1999. 278. Molad Y, Gorshtein A, Wysenbeek AJ, et al: Protective effect of hydroxychloroquine in systemic lupus erythematosus: prospective long-term study of an Israeli cohort. Lupus 11: 356–361, 2002. 279. Woo TY, Callan JP, Voorhees JJ, et al: Cutaneous lesions of dermatomyositis are improved by hydroxychloroquine. J Am Acad Dermatol 10: 592–600, 1984. 280. Olson NY, Lindsley CB: Adjunctive use of hydroxychloroquine in childhood dermatomyositis. J Rheumatol 16: 1545–1547, 1989.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
131
281. Markowitz HA, McGrinley JM: Chloroquine poisoning in a child. JAMA 189: 950, 1964. 282. Dubois E: Antimalarials in the management of discoid and systemic lupus erythematosus. Semin Arthritis Rheum 8: 33, 1978. 283. Henkind P, Rothfield NF: Ocular abnormalities in patients treated with synthetic antimalarial drugs. N Engl J Med 269: 433, 1963. 284. Rynes RI, Krohel G, Falbo A, et al: Ophthalmologic safety of long-term hydroxychloroquine treatment. Arthritis Rheum 22: 832, 1979. 285. Rynes RI: Side effects of antimalarial therapy. Br J Clin Pract 41 (Suppl 52): 42, 1987. 286. Easterbrook M: Current concepts in monitoring patients on antimalarials. Aust N Z J Ophthalmol 26: 101–103, 1998. 287. Marmor MF. New American Academy of Ophthalmology recommendations on screening for hydroxychloroquine retinopathy. Arthritis Rheum 48: 1764, 2003. 288. Grierson DJ: Hydroxychloroquine and visual screening in a rheumatology outpatient clinic. Ann Rheum Dis 56: 188–190, 1997. 289. Sassaman FW, Cassidy JT, Alpern M, et al: Electroretinography in patients with connective tissue diseases treated with hydroxychloroquine. Am J Ophthalmol 70: 515, 1970. 290. Marmor MF. Hydroxychloroquine at the recommended dose (≤ 6.5 mg/kg/day) is safe for the retina in patients with rheumatoid arthritis and systemic lupus erythematosus. Clin Exp Rheumatol 22: 143–144, 2004. 291. Laaksonen A-L, Koshiadhe V, Juva K: Dosage of antimalarial drugs for children with juvenile rheumatoid arthritis and systemic lupus erythematosus: a clinical study with determination of serum concentrations of chloroquine and hydroxychloroquine. Scand J Rheumatol 3: 103–108, 1974. 292. Costedoat-Chalumeau N, Amoura Z, Duhaut P, et al: Safety of hydroxychloroquine in pregnant patients with connective tissue diseases: a study of one hundred thirty-three cases compared with a control group. Arthritis Rheum 48: 3207–3211, 2003. 293. Costedoat-Chalumeau N, Amoura Z, Aymard G, et al: Evidence of transplacental passage of hydroxychloroquine in humans. Arthritis Rheum 46: 1123–1124, 2002. 294. Svartz N: Salazopyrin, a new sulfanilamide preparation. Acta Med Scand 60: 577, 1942. 295. Svartz N: The treatment of rheumatoid arthritis with acid azo compounds. Rheumatism 4: 56, 1948. 296. Ozdogan H, Turunc M, Deringol B, et al: Sulphasalazine in the treatment of juvenile rheumatoid arthritis: a preliminary open trial. J Rheumatol 13: 124–125, 1986. 297. Huang JL, Chen LC: Sulphasalazine in the treatment of children with chronic arthritis. Clin Rheumatol 17: 359–363, 1998. 298. Ansell BM, Hall MA, Loftus JK, et al: A multicentre pilot study of sulphasalazine in juvenile chronic arthritis. Clin Exp Rheumatol 9: 201–203, 1991. 299. van Rossum MA, Fiselier TJ, Franssen MJ, et al: Sulfasalazine in the treatment of juvenile chronic arthritis: a randomized, double-blind, placebocontrolled, multicenter study. Dutch Juvenile Chronic Arthritis Study Group. Arthritis Rheum 41: 808–816, 1998. 300. Sinclair RJG, Duthie JRR: Salazopyrin in the treatment of rheumatoid arthritis. Ann Rheum Dis 8: 226, 1948. 301. Mielants H, Veys EM: HLA-B27 related arthritis and bowel inflammation. Part 1: sulfasalazine (Salazopyrin) in HLA-B27 related reactive arthritis. J Rheumatol 12: 287, 1985. 302. Brooks CD. Sulfasalazine for the management of juvenile rheumatoid arthritis. J Rheumatol 28: 845–853, 2001. 303. Burgos-Vargas R, Vazquez-Mellado J, Pacheco-Tena C, et al: A 26 week randomised, double blind, placebo controlled exploratory study of sulfasalazine in juvenile onset spondyloarthropathies. Ann Rheum Dis. 61: 941–942, 2002. 304. Clegg DO, Reda DJ, Mejias E, et al: Comparison of sulfasalazine and placebo in the treatment of psoriatic arthritis. A Department of Veterans Affairs Cooperative Study. Arthritis Rheum 39: 2013–2020, 1996. 305. Clegg DO, Reda DJ, Weisman MH, et al: Comparison of sulfasalazine and placebo in the treatment of reactive arthritis (Reiter’s syndrome). A Department of Veterans Affairs Cooperative Study. Arthritis Rheum 39: 2021–2027, 1996. 306. Clegg DO, Reda DJ, Weisman MH, et al: Comparison of sulfasalazine and placebo in the treatment of ankylosing spondylitis. A Department of Veterans Affairs Cooperative Study. Arthritis Rheum 39: 2004–2012, 1996. 307. Dougados M, van der Linden S, Seirisalo-Repo M, et al: Sulfasalazine in the treatment of spondylarthropathy: a randomized, multicenter, double-blind, placebo-controlled study. Arthritis Rheum 38: 618–627, 1995. 308. Ferraz MB, Tugwell P, Goldsmith CH, Atra E: Meta-analysis of sulfasalazine in ankylosing spondylitis. J Rheumatol 17: 1482–1486, 1990. 309. Clegg DO, Reda DJ, Abdellatif M: Comparison of sulfasalazine and placebo for the treatment of axial and peripheral articular manifestations of the seronegative spondylarthropathies. A Department of Veterans Affairs Cooperative Study. Arthritis Rheum 42: 2325–2329, 1999. 310. Schroeder H, Campbell DES: Absorption, metabolism and excretion of salicylazosulfapyridine in man. Clin Pharmacol Ther 13: 36, 1972. 311. Bird HA, Dixon JS, Pickup ME, et al: A biochemical assessment of sulfasalazine in rheumatoid arthritis. J Rheumatol 9: 36, 1982. 312. Peppercorn MA: Sulfasalazine, pharmacology, clinical use, toxicities and related new drug development. Ann Intern Med 101: 377, 1984.
132
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
313. Stenson WF, Lobos E: Sulfasalazine inhibits the synthesis of chemotactic lipids by neutrophils. J Clin Invest 69: 494, 1982. 314. Gadangi P, Longaker M, Naime D: The anti-inflammatory mechanism of sulfasalazine is related to adenosine release at inflamed sites. J Immunol 156: 1937, 1996. 315. Posthumus MD, Limburg PC, Westra J, et al: Serum matrix metalloproteinase 3 levels during treatment with sulfasalazine or combination of methotrexate and sulfasalazine in patients with early rheumatoid arthritis. J Rheumatol 29: 883–889, 2002. 316. Carlin G, Nyman AK, Gronberg A: Effects of sulfasalazine on cytokine production by mitogen-stimulated human T cells. Arthritis Rheum 37: S783, 1994. 317. Gronenberg A, Isaksson P, Smedegard G: Inhibitory effect of sulfasalazine on production of IL-1B, IL-6, and TNFa [abstract]. Arthritis Rheum 37: S383, 1994. 318. Aono H, Hasunuma T, Fujisawa K, et al: Direct suppression of human synovial cell proliferation in vitro by salazosulfapyridine and bucillamine. J Rheumatol 22: 65–70, 1995. 319. Wahl C, Liptay S, Adler G, Schmid RM: Sulfasalazine: a potent and specific inhibitor of nuclear factor kappa B. J Clin Invest 101: 1163–1174, 1998. 320. Rodenburg RJ, Ganga A, van Lent PL, et al: The antiinflammatory drug sulfasalazine inhibits tumor necrosis factor alpha expression in macrophages by inducing apoptosis. Arthritis Rheum 43: 1941–1950, 2000. 321. Akahoshi T, Namai R, Sekiyama N, et al: Rapid induction of neutrophil apoptosis by sulfasalazine: implications of reactive oxygen species in the apoptotic process. J Leukoc Biol 62: 817–826, 1997. 322. Hasko G, Szabo C, Nemeth ZH, Deitch EA: Sulphasalazine inhibits macrophage activation: inhibitory effects on inducible nitric oxide synthase expression, interleukin-12 production and major histocompatibility complex II expression. Immunology 103: 473–478 2001. 323. Volin MV, Harlow LA, Woods JM, et al: Treatment with sulfasalazine or sulfapyridine, but not 5-aminosalicyclic acid, inhibits basic fibroblast growth factor-induced endothelial cell chemotaxis. Arthritis Rheum 42: 1927–1935, 1999. 324. Taffet SL, Das KM: Sulfasalazine: adverse effects and desensitization. Dig Dis Sci 28: 833, 1983. 325. Amos RS, Pullar T, Bax DE, et al: Sulphasalazine for rheumatoid arthritis: toxicity in 774 patients monitored for one to 11 years. BMJ 293: 420, 1986. 326. Raforth R: Systemic granulomatous reaction to salicylazosulfapyridine in a patient with Crohn’s disease. Am J Dig Dis 19: 465, 1974. 327. Capell HA, Pullar T, Hunter JA: Comparison of white blood cell dyscrasias during sulfasalazine therapy of rheumatoid arthritis and inflammatory bowel disease. Drugs 32 (Suppl 1): 44, 1986. 328. Wijnands MJ, Allebes WA, Boerbooms AMT, et al: Thrombocytopenia due to aurothioglucose, sulfasalazine, and hydroxychloroquine. Ann Rheum Dis 49: 798, 1990. 329. Logan EC, Williamson LM, Ryrie DR: Sulfasalazine associated pancytopenia may be caused by acute folate deficiency. Gut 27: 868, 1986. 330. Prouse PJ, Shawe D, Gumpel JM: Macrocytic anaemia in patients treated with sulfasalazine for rheumatoid arthritis. BMJ 293: 1407, 1986. 331. Clementz GL, Dolin BJ: Sulfasalazine-induced lupus erythematosus. Am J Med 84: 535, 1988. 332. Reid J, Holt S, Housley E, Sneddon DJC: Raynaud’s phenomenon induced by sulphasalazine. Postgrad Med J 56: 106, 1980. 333. Averbuch M, Halpern Z, Hallak A, et al: Sulfasalazine pneumonitis. Am J Gastroenterol 80: 343, 1985. 334. Hamadeh MA, Atkinson J, Smith LJ: Sulfasalazine-induced pulmonary disease. Chest 101: 1033, 1992. 335. Caspi D, Fuchs D, Yaron M: Sulphasalazine induced hepatitis in juvenile rheumatoid arthritis. Ann Rheum Dis 51: 275, 1992. 336. Toth A: Reversible toxic effect of salicylazosulfapyridine on semen quality. Fertil Steril 31: 538, 1979. 337. Pullar T, Hunter JA, Capell HA: Sulfasalazine in rheumatoid arthritis: a double blind comparison of sulfasalazine with placebo and sodium aurothiomalate. BMJ 287: 1102, 1983. 338. Neumann VC, Grindulis KA, Hubball S, et al: A comparison between penicillamine and sulfasalazine in rheumatoid arthritis. BMJ 287: 1099, 1983. 339. Pullar T, Capell HA: Sulphasalazine: a “new” antirheumatic drug. Br J Rheumatol 23: 26, 1984. 340. Neumann VC, Grindulis KA: Sulphasalazine in rheumatoid arthritis: an old drug revived. J R Soc Med 77: 169, 1984. 341. Grindulis K, McConkey B: Outcome of attempts to treat rheumatoid arthritis patients with gold, penicillamine, sulfasalazine or dapsone. Ann Rheum Dis 43: 398, 1984. 342. Bax DE, Amos RS: Sulfasalazine: a safe, effective agent for prolonged control of rheumatoid arthritis. A comparison with sodium aurothiomalate. Ann Rheum Dis 44: 194, 1985. 343. Watkinson G: Sulphasalazine: a review of 40 years experience. Drugs 32: 1, 1986. 344. Kean WF: Sulfasalazine in rheumatic disease. International Rheumatology Conference, Quebec, Medicopea, 1987. 345. Proceedings: Sulfasalazine in rheumatic diseases. J Rheumatol 16, 1988.
346. Williams HJ, Ward JL, Dahl DL: A controlled trial comparing sulfasalazine, gold sodium thiomalate, and placebo in rheumatoid arthritis. Arthritis Rheum 31: 702, 1988. 347. Farr M, Scott DG, Bacon PA: Side effect profile of 200 patients with inflammatory arthritides treated with sulfasalazine. Drugs 32: 49, 1986. 348. Scott DL, Dacre JE: Adverse reactions to sulfasalazine: the British experience. J Rheumatol 15: 17, 1988. 349. Wadelius M, Stjernberg E, Wiholm BE, Rane A: Polymorphisms of NAT2 in relation to sulphasalazine-induced agranulocytosis. Pharmacogenetics 10: 35–41, 2000. 350. van Rossum MA, Fiselier TJ, Franssen MJ, et al: Effects of sulfasalazine treatment on serum immunoglobulin levels in children with juvenile chronic arthritis. Scand J Rheumatol 30: 25–30, 2001. 351. Hertzberger-ten Cate R, Cats A: Toxicity of sulfasalazine in systemic juvenile chronic arthritis. Clin Exp Rheumatol 9: 85–88, 1991. 352. Jung JH, Jun JB, Yoo DH, et al: High toxicity of sulfasalazine in adult-onset Still’s disease. Clin Exp Rheumatol 18: 245–248, 2000. 353. Suschke HJ: Sulfasalazin bei juveniler chronishcher Arthritis. Pediatr Prax 33: 681, 1986. 354. Bresnihann B. Treating early rheumatoid arthritis in the younger patient. J Rheumatol Suppl 62: 4–9, 2001. 355. Berlin CM Jr, Yaffe SJ: Disposition of salicylazosulfapyridine (Azulfidine) and metabolites in human breast milk. Dev Pharmacol Ther 1: 31–39, 1980. 356. Davis JP, Cain CA, Pitts WJ, et al: The immunosuppressive metabolite of leflunomide is a potent inhibitor of human dihydroorotate dehydrogenase. Biochemistry 35: 1270–1273, 1996. 357. Xu X, Blinder L, Shen J, et al: In vivo mechanism by which leflunomide controls lymphoproliferative and autoimmune disease in MRL/MpJ-lpr/lpr mice. J Immunol 159: 167–174, 1997. 358. Dimitrijevic M, Bartlett RR: Leflunomide, a novel immunomodulating drug, inhibits homotypic adhesion of mononuclear cells in rheumatoid arthritis. Transplant Proc 28: 3086–3087, 1996. 359. Cao WW, Kao PN, Aoki Y, et al: A novel mechanism of action of the immunomodulatory drug, leflunomide: augmentation of the immunosuppressive cytokine, TGF-beta 1, and suppression of the immunostimulatory cytokine, IL-2. Transplant Proc 28: 3079–3080, 1996. 360. Burger D, Begue-Pastor N, Benavent S, et al: The active metabolite of leflunomide, A77 1726, inhibits the production of prostaglandin E(2), matrix metalloproteinase 1 and interleukin 6 in human fibroblast-like synoviocytes. Rheumatology (Oxf) 42: 89–96, 2003. 361. Rozman B: Clinical pharmacokinetics of leflunomide. Clin Pharmacokinet 41: 421–430, 2002. 362. Mladenovic V, Domljan Z, Rozman B, et al: Safety and effectiveness of leflunomide in the treatment of patients with active rheumatoid arthritis. Arthritis Rheum 38: 1595–1603, 1995. 363. Smolen JS, Kalden JR, Scott DL, et al: Efficacy and safety of leflunomide compared with placebo and sulphasalazine in active rheumatoid arthritis: a double-blind, randomised, multicentre trial. European Leflunomide Study Group. Lancet 353: 259–266, 1999. 364. Strand V, Cohen S, Schiff M, et al: Treatment of active rheumatoid arthritis with leflunomide compared with placebo and methotrexate. Leflunomide Rheumatoid Arthritis Investigators Group. Arch Intern Med 159: 2542–2550, 1999. 365. Cohen S, Cannon GW, Schiff M, et al: Two-year, blinded, randomized, controlled trial of treatment of active rheumatoid arthritis with leflunomide compared with methotrexate. Utilization of Leflunomide in the Treatment of Rheumatoid Arthritis Trial Investigator Group. Arthritis Rheum 44: 1984–1992, 2001. 366. Cannon GW, Kremer JM: Leflunomide. Rheum Dis Clin North Am 30: 295–309, 2004. 367. Rozman B: Clinical experience with leflunomide in rheumatoid arthritis. J Rheumatol 25 (Suppl 53): 27–32, 1998. 368. Silverman ED, Spiegel L, Hawkins D, et al: Long-term open-label preliminary study of the safety and efficacy of leflunomide in patients with polyarticular course juvenile rheumatoid arthritis. Arthritis Rheum 2005; in press. 368a. Silverman ED, Mouy R, Spiegel, et al: Leflunomide versus methotrexate for the treatment of juvenile rheumatoid arthritis. New Engl J Med 2005; in press. 369. Brent RL: Teratogen update: reproductive risks of leflunomide (Arava), a pyrimidine synthesis inhibitor: counseling women taking leflunomide before or during pregnancy and men taking leflunomide who are contemplating fathering a child. Teratology 63: 106–112, 2001. 370. Forestier J: L’aurotherapie dans les rheumatismes chroniques. Bull Soc Med Hop Paris 53: 323, 1929. 371. Forestier J: Rheumatoid arthritis and its treatment with gold salts. J Lab Clin Med 20: 827, 1935. 372. Lewis AJ, Walz DT: Immunopharmacology of gold. In Ellis JP (ed): Progress in Medicinal Chemistry, vol 19. New York, Elsevier Biomedical Press, 1982. 373. Lipsky PE, Ugai K, Ziff M: Alterations in human monocyte structure and function induced by incubation with gold sodium thiomalate. J Rheumatol 6 (Suppl 5): 130, 1979. 374. Martini A, De Amici M, Visconti L, et al: Immunological evaluation in children with juvenile chronic arthritis treated with auranofin. Int J Clin Pharmacol Res 5: 149, 1985.
C H A P T E R 375. Fantini F, Corvaglia G, Bergomi P, et al: Validation of the Italian version of the Stanford Childhood Health Assessment Questionnaire for measuring functional status in children with chronic arthritis. Clin Exp Rheumatol 13: 785–791, 1995. 376. Sadler PJ: The comparative evaluation of the physical and chemical properties of gold compounds. J Rheumatol 9 (Suppl 8): 71, 1982. 377. Gottlieb NL: Comparative pharmacokinetics of parenteral and oral gold compounds. J Rheumatol 9 (Suppl 8): 99, 1982. 378. Blocka K, Furst D, Landau E, et al: Single dose pharmacokinetics of auranofin in rheumatoid arthritis. J Rheumatol 9 (Suppl 8): 110, 1982. 379. Giannini EH, Brewer EJ, Person DA: Blood gold concentrations in children with juvenile rheumatoid arthritis undergoing long-term oral gold therapy. Ann Rheum Dis 43: 228, 1984. 380. Giannini EH, Person DA, Brewer EJ Jr, et al: Blood and serum concentration of gold after a single dose of auranofin in children with juvenile rheumatoid arthritis. J Rheumatol 10: 496, 1983. 381. Nuki G, Gumpel JM: Myocrisin: 50 years experience [proceedings of an international symposium]. London, Medi-Cine Communications International, 1985. 382. Sairanen E, Laaksonen A–L: The toxicity of gold therapy in children suffering from rheumatoid arthritis. Ann Paediatr Fenn 8: 105, 1962. 383. Kean WF, Anastassiades TP: Long-term chrysotherapy: incidence of toxicity and efficacy during sequential time periods. Arthritis Rheum 22: 495, 1979. 384. Sairanen E, Laaksonen A–L: The results of gold therapy in juvenile rheumatoid arthritis. Ann Paediatr Fenn 10: 274, 1963. 385. Hicks RM, Hanson V, Kornreich HK: The use of gold in the treatment of juvenile rheumatoid arthritis. Arthritis Rheum 13: 323, 1970. 386. DeBenedetti C, Tretbar H, Corrigan JJ: Gold therapy in juvenile rheumatoid arthritis. Ariz Med 33: 373, 1976. 387. Levinson JF, Balz GP, Bondi S: Gold therapy. Arthritis Rheum 20: 531, 1977. 388. Brewer EJ Jr, Giannini EH, Barkley E: Gold therapy in the management of juvenile rheumatoid arthritis. Arthritis Rheum 23: 404, 1980. 389. Levinson JE: Gold salts in the rheumatic diseases. In Moore TD (ed): Arthritis in Childhood: Report of the Eightieth Ross Conference in Pediatric Research. Columbus, OH, Ross Laboratories, 1981, pp. 120–124. 390. Kvien TK, Hoyeraal HM, Sandstad B: Slow acting antirheumatic drugs in patients with juvenile rheumatoid arthritis—evaluated in a randomized, parallel 50-week clinical trial. J Rheumatol 12: 533, 1985. 391. Kvien TK, Hoyeraal HM, Sandstad B: Gold sodium thiomalate and D-penicillamine: A controlled, comparative study in patients with pauciarticular and polyarticular juvenile rheumatoid arthritis. Scand J Rheumatol 14: 346, 1985. 392. Manners PJ, Ansell BM: Slow-acting antirheumatic drug use in systemic onset juvenile chronic arthritis. Pediatrics 77: 99, 1986. 393. Malleson PN, Grondin C, Petty RE, et al: Outcome of gold therapy in juvenile rheumatoid arthritis. Arthritis Rheum 30: 528, 1987. 394. Grondin C, Malleson P, Petty RE: Slow-acting antirheumatic drugs in chronic arthritis of childhood. Semin Arthritis Rheum 18: 38, 1988. 395. Husserl FE, Shuler SE: Gold nephropathy in juvenile rheumatoid arthritis. Am J Dis Child 133: 50, 1979. 396. Merle LJ, Reidenberg MM, Camacho MT, et al: Renal injury in patients with rheumatoid arthritis treated with gold. Clin Pharmacol Ther 28: 216, 1980. 397. Rothermich NO: Chrysotherapy in rheumatoid arthritis. Clin Rheum Dis 5: 631, 1979. 398. Aaron S, Davis P, Percy J: Neutropenia occurring during the course of chrysotherapy: a review of 25 cases. J Rheumatol 12: 897, 1985. 399. Wooley PH, Griffin J, Panayi GS: HLA-DR antigens and toxic reaction to sodium aurothiomalate and D-penicillamine in patients with rheumatoid arthritis. N Engl J Med 303: 300, 1980. 400. Jacobs JC, Gorin LJ, Hanissian AS, et al: Consumption coagulopathy after gold therapy for JRA. J Pediatr 105: 674, 1984. 401. Favreau M, Tannenbaum H, Lough J: Hepatic toxicity associated with gold therapy. Ann Intern Med 87: 717, 1977. 402. Harats N, Shalit M, Ehrenfeld M, et al: Gold-induced granulomatous hepatitis. Isr J Med Sci 21: 753, 1985. 403. Olsen JL, Lovell DJ, Levinson JE: Hypogammaglobulinemia associated with gold therapy in a patient with juvenile rheumatoid arthritis. J Rheumatol 13: 224, 1986. 404. Winterbauer RH, Wilski KR, Wheelis RF: Diffuse pulmonary injury associated with gold treatment. N Engl J Med 294: 919, 1976. 405. Gould PW, McCormack PL, Palmer DG: Pulmonary damage associated with sodium aurothiomalate therapy. J Rheumatol 4: 252, 1977. 406. Fam AG, Gordon DA, Sarkozi J, et al: Neurologic complications associated with gold therapy for rheumatoid arthritis. J Rheumatol 11: 700, 1984. 407. Pik A, Cohen N, Yona E, et al: Should acute gold overdose be invariably treated? J Rheumatol 12: 1174, 1984. 408. Gambari P, Ostuni P, Lazzarin P, et al: Neurotoxicity following a very high dose of oral gold (auranofin). Arthritis Rheum 27: 1316, 1984. 409. Garland JS, Sheth KJ, Wortmann DW: Poor clearance of gold using peritoneal dialysis for the treatment of gold toxicity. Arthritis Rheum 29: 450, 1986. 410. Tarp U, Graudal H: A follow-up study of children exposed to gold compounds in utero. Arthritis Rheum 28: 235, 1986. 411. Paulus HE: Government Affairs: FDA Arthritis Advisory Committee meeting: Auranofin. Arthritis Rheum 28: 450, 1985.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
133
412. Giannini EH, Brewer EJ, Person DA: Auranofin in the treatment of juvenile rheumatoid arthritis. J Pediatr 102: 450, 1983. 413. Giannini EH, Brewer EJ, Person DA, et al: Longterm auranofin therapy in patients with juvenile rheumatoid arthritis. J Rheumatol 13: 768, 1986. 414. Garcia-Morteo O, Suarez-Almazor ME, Maldonado-Cocco JA, et al: Auranofin in juvenile rheumatoid arthritis: an open label, non-controlled study. Clin Rheumatol 3: 223, 1984. 415. Kvien TL, Hoyraal HM, Sandstad B, et al: Auranofin therapy in juvenile rheumatoid arthritis: a 48-week phase II study. Scand J Rheumatol 63: 79, 1986. 416. Marcolongo R, Mathieu A, Pala R, et al: The efficacy and safety of auranofin in the treatment of juvenile rheumatoid arthritis: a long-term open study. Arthritis Rheum 31: 979, 1988. 417. Giannini EH, Brewer EJ, Kuzmina N, et al: Auranofin in the treatment of juvenile rheumatoid arthritis: results of the U.S.A.-U.S.S.R. double-blind, placebo-controlled trial. Arthritis Rheum 33: 466, 1989. 418. Clements PJ, Furst DE, Wong WK, et al: High-dose versus low-dose Dpenicillamine in early diffuse systemic sclerosis: analysis of a two-year, double-blind, randomized, controlled clinical trial. Arthritis Rheum 42: 1194, 1999. 419. Howard-Lock HE, Lock CJL, Mewa A, et al: D-Penicillamine: chemistry and clinical use in rheumatic disease. Semin Arthritis Rheum 15: 261, 1978. 420. Jaffe IA: D-Penicillamine. Bull Rheum Dis 28: 948, 1978. 421. Multicenter Trial Group: Controlled trial of D-penicillamine in severe rheumatoid arthritis. Lancet 1: 275, 1980. 422. Joyce DA: D-Penicillamine pharmacokinetics and action. Agents Actions 24 (Suppl): 197–206, 1988. 423. Joyce DA: D-Penicillamine. Clin Rheumatol 4: 553, 1990. 424. Cuperus RA, Hoogland H, Wever R, Juijsers AOL: The effect of D-penicillamine on myeloperoxidase: formation of compound III and inhibition of the chlorinating activity. Biochim Biophys Acta 912: 124, 1987. 425. Theyboom RHB, Jaffe IA: Metal antagonists. Side Effects Drugs Annu 11: 211, 1987. 426. Lipsky PE: Immunosuppression by D-penicillamine in vitro: Inhibition of human T lymphocyte proliferation by copper- or ceruloplasmin-dependent generation of hydrogen peroxide and protection by monocytes. J Clin Invest 73: 53, 1984. 427. Sheikh IA, Kaplan AP: Assessment of kinases in rheumatic diseases and the effects of therapeutic agents. Arthritis Rheum 30: 138, 1987. 428. Bresnihan GP, Ansell BM: Effect of penicillamine treatment on immune complexes in two cases of seropositive juvenile rheumatoid arthritis. Ann Rheum Dis 35: 463, 1976. 429. Sim E, Dodds AW, Goldin A: Inhibition of the covalent binding reaction of complement component C4 by penicillamine, an anti-rheumatic drug. Biochem J 259: 415, 1989. 430. Chandra P, Sarin PS: Selective inhibition of the AIDS-associated virus HTLVIII/LAV by synthetic D-penicillamine. Arzneimittelforschung 36: 184, 1986. 431. Bourke B, Maini RN, Griffiths ID, et al: Fatal marrow aplasia in patient on penicillamine. Lancet 2: 515, 1976. 432. Blasberg B, Dorey JL, Stein HB, et al: Lichenoid lesions of the oral mucosa in rheumatoid arthritis patients treated with penicillamine. J Rheumatol 11: 348, 1984. 433. Dische FE, Swinson DR, Hamilton EBD, et al: Immunopathology of penicillamine-induced glomerular disease. J Rheumatol 11: 584, 1984. 434. Stein HB, Schroeder M-L, Dillon AM: Penicillamine-induced proteinuria: risk factors. Semin Arthritis Rheum 15: 282, 1986. 435. Kay A: European League Against Rheumatism Study of adverse reactions to D-penicillamine. Br J Rheumatol 25: 193, 1986. 436. Steen VD, Blair S, Medsgar A: The toxicity of D-penicillamine in systemic sclerosis. Ann Intern Med 104: 699, 1986. 437. Baum J: The use of penicillamine in the treatment of rheumatoid arthritis and scleroderma. Scand J Rheumatol 28: 65, 1979. 438. Forre O, Munthe E, Kass E: Side-effects and autoimmunogenicity of D-penicillamine treatment in rheumatic diseases. Adv Inflamm Res 6: 251, 1984. 439. Takahashi K, Ogita T, Okudaira H, et al: D-Penicillamine-induced polymyositis in patients with rheumatoid arthritis. Arthritis Rheum 29: 560, 1986. 440. Kuncl RW, Pestronk A, Drachman DB, Rechthand E: The pathophysiology of penicillamine-induced myasthenia gravis. Ann Neurol 20: 740, 1986. 441. Perrier P, Raffoux C, Thomas P, et al: HLA antigens and toxic reactions to sodium aurothiopropanol sulphonate and D-penicillamine in patients with rheumatoid arthritis. Ann Rheum Dis 44: 621, 1985. 442. Hunter AL, Klaasen CD: Biliary excretion of colchicine. J Pharmacol Exp Ther 192: 605–617, 1975. 443. Ben-Chetrit E, Scherrmann JM, Zylber-Katz E, Levy M: Colchicine disposition in patients with familial Mediterranean fever with renal impairment. J Rheumatol 21: 710, 1994. 444. Ben-Chetrit E, Levy M: Colchicine: 1998 update. Semin Arthritis Rheum 28: 48, 1998. 445. Molad T: Update on colchicine and its mechanism of action. Curr Rheumatol Rep 4: 252–256, 2002. 446. Cronstein BN, Molad Y, Reibman J, et al: Colchicine alters the quantitative and qualitative display of selectins on endothelial cells and neutrophils. J Clin Invest 96; 994–1002, 1995. 447. Venho VM, Koivuniemi A: Effect of colchicine on drug absorption from the rat small intestine in situ and in vitro. Acta Pharmacol Toxicol (Copenh) 43: 251, 1978.
134
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
448. Cohen MM, Levy M, Eliakim M: A cytogenic evaluation of long-term colchicine therapy in the treatment of familial Mediterranean fever (FMF). Am J Med Sci 274: 147, 1977. 449. Ben-Chetrit A, Ben-Chetrit E, Nitzan R, Ron M: Colchicine inhibits spermatozoal motility in vitro. Int J Fertil Menopausal Stud 38: 301–304, 1993. 450. Haimov-Kochman R, Ben-Chetrit E: The effect of colchicine on sperm production and function: a review. Hum Reprod 13: 360–362, 1998. 451. Zemer D, Livneh A, Danon YL, et al: Long-term colchicine treatment in children with familial Mediterranean fever. Arthritis Rheum 34: 973, 1991. 452. Majeed HA, Carroll JE, Khuffash FA, Hijazi Z: Long-term colchicine prophylaxis in children with familial Mediterranean fever. J Pediatr 116: 997, 1990. 453. Baud FJ, Sabouraud A, Vicaut E, et al: Brief report: treatment of severe colchicine overdose with colchicine-specific Fab fragments. N Engl J Med 332: 642–645, 1995. 454. Faure M, Thivolet J, Gaucherand M: Inhibition of PMN leukocytes chemotaxis by thalidomide. Arch Dermatol Res 269: 275–280, 1980. 455. Barnhill RL, Doll NJ, Millikan LE, Hastings RC: Studies on the anti-inflammatory properties of thalidomide: effects on polymorphonuclear leukocytes and monocytes. J Am Acad Dermatol 11: 814, 1984. 456. Gad SM, Shannon EJ, Krotoski WA, Hastings RC: Thalidomide induces imbalances in T-lymphocyte sub-populations in the circulating blood of healthy males. Lepr Rev 56: 35, 1985. 457. Rowland TL, McHugh SM, Deighton J, et al: Differential regulation by thalidomide and dexamethasone of cytokine expression in human peripheral blood mononuclear cells. Immunopharmacology 40: 11, 1998. 458. D’Amato RJ, Loughnan MS, Flynn E, Folkman J: Thalidomide is an inhibitor of angiogenesis. Proc Natl Acad Sci U S A 91: 4082, 1994. 459. Bauer KS, Dixon SC, Figg WD: Inhibition of angiogenesis by thalidomide requires metabolic activation, which is species-dependent. Biochem Pharmacol 55: 1827, 1998. 460. Chen TL, Vogelsang GB, Petty BG, et al: Plasma pharmacokinetics and urinary excretion of thalidomide after oral dosing in healthy male volunteers. Drug Metab Dispos 17: 402, 1989. 461. Jacobson JM. Thalidomide: a remarkable comeback. Expert Opin Pharmacother 1: 849–863, 2000. 462. Revuz J, Guillaume JC, Janier M, et al: Crossover study of thalidomide vs placebo in severe recurrent aphthous stomatitis. Arch Dermatol 1126: 623–627, 1990. 463. Hamuryudan V, Mat C, Saip S, et al: Thalidomide in the treatment of the mucocutaneous lesions of the Behcet syndrome: a randomized, doubleblind, placebo-controlled trial. Ann Intern Med 128: 443–450, 1998. 464. Shek LP, Lee YS, Lee BW, Lehman TJ. Thalidomide responsiveness in an infant with Behcet’s syndrome. Pediatrics 103: 1295–1297, 1999. 465. Bessmertny O, Pham T. Thalidomide use in pediatric patients. Ann Pharmacother 36: 521–525, 2002. 466. Tseng S, Pak G, Washenik K, et al: Rediscovering thalidomide: a review of its mechanism of action, side effects, and potential uses. J Am Acad Dermatol 35: 969, 1996. 467. Ordi-Ros J, Cortes F, Cucurull E, et al: Thalidomide in the treatment of cutaneous lupus refractory to conventional therapy. J Rheumatol 27: 1429–1433, 2000. 468. Duong DJ, Spigel GT, Moxley RT 3rd, Gaspari AA: American experience with low-dose thalidomide therapy for severe cutaneous lupus erythematosus. Arch Dermatol 135: 1079–1087, 1999. 469. Stevens RJ, Andujar C, Edwards CJ, et al: Thalidomide in the treatment of the cutaneous manifestations of lupus erythematosus: experience in sixteen consecutive patients. Br J Rheumatol 26: 353–359, 1997. 470. Odeka EB, Miller V: Thalidomide in oral Crohn’s disease refractory to conventional medical treatment. J Pediatr Gastroenterol Nutr 25: 250–251, 1997. 471. Weinstein TA, Sciubba JJ, Levine J: Thalidomide for the treatment of oral aphthous ulcers in Crohn’s disease. J Pediatr Gastroenterol Nutr 28: 214–216, 1999. 472. Keesal N, Wasserman MJ, Bookman A, et al: Thalidomide in the treatment of refractory rheumatoid arthritis. J Rheumatol 26: 2344–2347, 1999. 473. Wei JC, Chan TW, Lin HS, et al: Thalidomide for severe refractory ankylosing spondylitis: a 6-month open-label trial. J Rheumatol 30: 2627-2631, 2003. 474. Stambe C, Wicks IP: TNF alpha and response of treatment-resistant adultonset Still’s disease to thalidomide. Lancet 352: 544, 1998. 475. Lehman TJ, Striegel KH, Onel KB: Thalidomide therapy for recalcitrant systemic onset juvenile rheumatoid arthritis. J Pediatr 140: 125–127, 2002. 476. Ochonisky S, Verroust J, Bastuji-Garin S, et al: Thalidomide neuropathy incidence and clinico-electrophysiologic findings in 42 patients. Arch Dermatol 130: 66, 1994. 477. Lagueny A, Rommel A, Vignolly B, et al: Thalidomide neuropathy: an electrophysiologic study. Muscle Nerve 9: 837, 1986. 478. Locker D, Superstine E, Sulmar FG: The mechanism of the push and pull principle: 8. Endocrine effects of thalidomide and its analogues. Arch Int Pharmacodyn Ther 194: 39, 1971. 479. Atra E, Sato EI: Treatment of the cutaneous lesions of systemic lupus erythematosus with thalidomide. Clin Exp Rheumatol 11: 487–493, 1993. 480. American Medical Association: Drug Evaluation Annual. Chicago, American Medical Association: 1992, p 1729.
481. Zeldis JB, Williams BA, Thomas SD, Elsayed ME: S.T.E.P.S.: a comprehensive program for controlling and monitoring access to thalidomide. Clin Ther 21: 319–330, 1999. 482. Hench PS: The potential reversibility of rheumatoid arthritis. Mayo Clin Proc 24: 167, 1949. 483. Ansell BM, Bywaters EGL, Isdale IC: Comparison of cortisone and aspirin in the treatment of juvenile rheumatoid arthritis. BMJ 1: 1075, 1956. 484. Schaller JG: Corticosteroids in juvenile rheumatoid arthritis (Still’s disease). J Rheumatol 1: 137, 1974. 485. Schaller JG: Corticosteroids in juvenile rheumatoid arthritis. Arthritis Rheum 20: 537, 1977. 486. Stoeber E: Corticosteroid treatment of juvenile chronic polyarthritis over 22 years. Eur J Pediatr 121: 141, 1976. 487. Prieur A-M: The place of corticosteroid therapy in juvenile chronic arthritis in 1992. J Rheumatol 20 (Suppl 7): 32–34, 1993. 488. Kimberly RP: Steroids. Curr Opin Rheumatol 2: 510, 1990. 489. Southwood TR: Report from a symposium on corticosteroid therapy in juvenile chronic arthritis. Clin Exp Rheumatol 11: 91, 1993. 490. Thompson EB, Lippman ME: Mechanism of action of glucocorticoids. Metabolism 23: 159, 1974. 491. Fauci AS, Dale DC, Balow JE: Glucocorticosteroid therapy: mechanisms of action and clinical considerations. Ann Intern Med 84: 304, 1976. 492. Axelrod L: Glucocorticoid therapy. Medicine (Baltimore) 55: 39, 1976. 493. Baxter JD: Glucocorticoid hormone action. Pharmacol Ther B2: 605, 1976. 494. Szefler SJ: General pharmacology of glucocorticoids. In Schleimer RP, Claman HN, Oronsky A (eds): Anti-inflammatory Steroid Action: Basic and Clinical Aspects. San Diego, Academic Press, 1990. 495. Burnstein KL, Cidlowski JA: Regulation of gene expression by glucocorticoids. Annu Rev Physiol 51: 683, 1989. 496. LaPointe MC, Baxter JD: Molecular biology of glucocorticoid hormone action. In Schleimer RP, Claman HN, Oronsky A (eds): Anti-Inflammatory Steroid Action: Basic and Chemical Aspects. San Diego, Academic Press, 1989, p. 3. 497. Schacke H, Docke WD, Asadullah K: Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther 96: 23–43, 2002. 498. Leung DY, Hamid Q, Vottero A, et al: Association of glucocorticoid insensitivity with increased expression of glucocorticoid receptor beta. J Exp Med 186: 1567–1574, 1997. 499. Buttgereit F, Wehling M, Burmester G-R: A new hypothesis of modular glucocorticoid actions: steroid treatment of rheumatic diseases revisited. Arthritis Rheum 41:761–767, 1998. 500. Parillo JE, Fauci AS: Mechanisms of glucocorticoid action on immune processes. Annu Rev Pharmacol Toxicol 19: 179, 1979. 501. Claman HN: Corticosteroids and lymphoid cells. N Engl J Med 287: 388, 1972. 502. Fauci AS, Dale DC: Effect of hydrocortisone on the kinetics of normal human lymphocytes. Blood 46: 235, 1975. 503. Weston WL, Claman HN, Krueger GG: Site and action of cortisol in cellular immunity. J Immunol 110: 880, 1973. 504. Rinehart JJ, Balcerzak SP, Sagone AL, LoBuglio AF: Effects of corticosteroids on human monocyte function. J Clin Invest 54: 1337, 1974. 505. Balow JE, Rosenthal AS: Glucocorticoid suppression of macrophage migration inhibitory factor. J Exp Med 137: 1031, 1973. 506. Schreiber AD, Parsons J, McDermott P, Cooper A: Effect of corticosteroids on the human monocyte IgG and complement receptors. J Clin Invest 56: 1189, 1975. 507. MacGregor RR: Granulocyte adherence changes induced by hemodialysis, endotoxin, epinephrine, and glucocorticoids. Ann Intern Med 86: 35, 1977. 508. Granelli-Piperano A, Vassali JD, Reich E: Secretion of plasminogen activator by human polymorphonuclear leukocytes: modulation by glucocorticoids and other effectors. J Exp Med 146: 1693, 1977. 509. Priestly GC, Brown JC: Effects of corticosteroids on the proliferation of normal and abnormal human connective tissue cells. Br J Dermatol 102: 35, 1980. 510. Hirata F, Schiffmann E, Venkatasubramanian K, et al: A phospholipase A2 inhibitory protein in rabbit neutrophils induced by glucocorticoids. Proc Natl Acad Sci U S A 77: 2533, 1980. 511. Blackwell GJ, Carnuccio R, DiRosa M, et al: Macrocortin: a polypeptide causing the antiphospholipase effect of glucocorticoids. Nature 287: 147, 1980. 511a. Cock TA, Houten SM, Auwerx J: Peroxisome proliferator-activated receptorgamma: too much of a good thing causes harm. EMBO Rep. 5: 142–147, 2004. 512. Haynes RC: Adrenocorticotrophic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones. In Goodman Gilman A, Rall TW, Nies AS, et al (eds): Goodman and Gilman’s The Pharmacological Basis of Therapeutics, 8th ed. New York, Pergamon Press, 1990. 513. Flower RJ: Glucocorticoids and the inhibition of phospholipase A2. In Schleimer RP, Claman HN, Oronsky A (eds): Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. San Diego, Academic Press, 1989, p 48. 514. Parente L, Flower RJ: Hydrocortisone and “macrocortin” inhibit the zymosan-induced release of lyso-PAF from rat peritoneal leucocytes. Life Sci 36: 1225–1231, 1985.
C H A P T E R 515. Schleimer RP, Claman HN, Oronsky A (eds): Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. San Diego, Academic Press, 1989. 516. Cohen JJ: Lymphocyte death induced by glucocorticoids. In Schleimer RP, Claman HN, Oronsky A (eds): Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. San Diego, Academic Press, 1989, p 110. 517. Cupps TR: Effects of glucocorticoids on lymphocyte function. In Schleimer RP, Claman HN, Oronsky A (eds): Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. San Diego, Academic Press, 1989, p 132. 518. Butterfield JH, Gleich GJ: Anti-inflammatory effects of glucocorticoids on eosinophils and neutrophils. In Schleimer RP, Claman HN, Oronsky A (eds): Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. San Diego, Academic Press, 1989 p 151. 519. Dauchel H, Julen N, Lemercier C, et al: Expression of complement alternative path proteins by endothelial cells: different regulation by interleukin 1 and glucocorticoids. Eur J Immunol 20: 1669, 1990. 520. Hyams JS, Carey DE: Corticosteroids and growth. J Paediatr 113: 249–254, 1988. 521. Good RA, Vernier RL, Smith RT: Serious untoward reactions to therapy with cortisone and A.C.T.H. in pediatric practice. Pediatrics 19: 95, 1957. 522. Ansell BM: Problems of corticosteroid therapy in the young. Proc R Soc Med 61: 281, 1968. 523. Reimer LG, Morris HG, Ellis EF: Growth of asthmatic children during treatment with alternate-day steroids. J Allergy Clin Immunol 55: 224, 1975. 524. Hahn TJ, Hahn BJ: Osteopenia in patients with rheumatoid diseases: principles of diagnosis and therapy. Semin Arthritis Rheum 6: 165, 1976. 525. Wang SJ, Yang YH, Lin YT, et al: Attained adult height in juvenile rheumatoid arthritis with or without corticosteroid treatment. Clin Rheumatol 21: 363–368, 2002. 526. Loeb JN: Corticosteroids and growth. N Engl J Med 295: 547, 1976. 527. Brouhard B, Travis LB, Cunningham RJ, et al: Inhibition of linear growth using alternate day steroids. J Pediatr 91: 343, 1977. 528. Punthakee Z, Legault L, Polychronakos C: Prednisolone in the treatment of adrenal insufficiency: a re-evaluation of relative potency. J Pediatr 143: 402–405, 2003. 529. McCarthy TL, Centrella M, Canalis E: Cortisol inhibits the synthesis of insulin-like growth factor 1 in skeletal cells. Endocrinology 126: 1569, 1990. 530. Bennett AE, Silverman ED, Miller JJ III, Hintz RL: Insulin-like growth factors in children with systemic onset juvenile arthritis. J Rheumatol 15: 655–658, 1988. 531. Morris HG, Jorgensen JR, Elrick H, Goldsmith RE: Metabolic effects of human growth hormone in corticosteroid-treated children. J Clin Invest 47: 436, 1968. 532. Simon D, Lucidarme N, Prieur AM, et al: Effects on growth and body composition of growth hormone treatment in children with juvenile idiopathic arthritis requiring steroid therapy. J Rheumatol 30: 2402–2499, 2003. 533. Bechtold S, Ripperger P, Hafner R, et al: Growth hormone improves height in patients with juvenile idiopathic arthritis: 4-year data of a controlled study. J Pediatr 143: 512–519, 2003. 534. Ansell BM, Bywaters EGL: Growth in Still’s disease. Ann Rheum Dis 15: 295–319, 1956. 535. Bernstein BH, Stobie D, Singsen BH, et al: Growth retardation in juvenile rheumatoid arthritis (JRA). Arthritis Rheum 20 (Suppl): 212–216, 1977. 536. Ansell BM, Bywaters EGL: Alternate-day corticosteroid therapy in juvenile chronic polyarthritis. J Rheumatol 1: 176, 1974. 537. Byron MA, Jackson J, Ansell BM: Effect of different corticosteroid regimens on hypothalamic-pituitary-adrenal axis and growth in juvenile chronic arthritis. J R Soc Med 76: 452–457, 1983. 538. Kimura Y, Fieldston E, Devries-Vandervlugt B, et al: High dose, alternate day corticosteroids for systemic onset juvenile rheumatoid arthritis. J Rheumatol 27: 2018–2024, 2000. 539. Falcini F, Trapani S, Civinini R, et al: The primary role of steroids on the osteoporosis in juvenile rheumatoid arthritis patients evaluated by dual energy X-ray absorptiometry. J Endocrinol Invest 19: 165–169, 1996. 540. Bianchi ML, Bardare M, Caraceni MP, et al: Bone metabolism in juvenile rheumatoid arthritis. Bone Mineral 9: 153–162, 1990. 541. Reed AM, Haugen M, Pachman LM, Langman CB: Repair of osteopenia in children with juvenile rheumatoid arthritis. J Paediatr 122: 693–696, 1993. 542. Rabinovich CE, Wagner-Weiner L, Wuellner J, Spencer C: Osteoporosis in juvenile rheumatoid arthritis. Arthritis Rheum 40: S323, 1997. 543. Kotaniemi A, Savolainen A, Kautiainen H, Kroger H: Estimation of central osteopenia in children with chronic polyarthritis treated with glucocorticoids. Pediatrics 91: 1127–1130, 1993. 544. Adachi JD, Bensen WG, Hodsman AB: Corticosteroid-induced osteoporosis. Semin Arthritis Rheum 22: 375–384, 1993. 545. Sambrook PN: Corticosteroid induced osteoporosis. J Rheumatol 23 (Suppl 45): 19–22, 1996. 546. Adachi JD, Bensen WG, Bell MJ, et al: Corticosteroid induced osteoporosis: followup over 3 years. Third International Symposium on Osteoporosis, Copenhagen, 1990. 547. Ruegsegger P, Medici TC, Anliker M: A longitudinal study of alternate day therapy in patients with bronchial asthma using quantitative computed tomography. Eur J Clin Pharmacol 25: 615–620, 1983. 548. Gluck OS, Murphy WA, Hahn TJ, et al: Bone loss in adults receiving alternate day glucocorticoid therapy. Arthritis Rheum 24: 892–898, 1981.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
135
549. Sambrook PN, Kelly PJ, Fontana D, et al: Mechanism of rapid bone loss following cardiac transplantation. Osteoporosis Int 4: 273–276, 1994. 550. Reeve J, Loftus J, Hesp R, et al: Biochemical prediction of changes in spinal bone mass in juvenile chronic (or rheumatoid) arthritis treated with glucocorticoids. J Rheumatol 20: 1189–1195, 1993. 550a. Canalis E, Delany AM: Mechanisms of glucocorticoid action in bone. Ann N Y Acad Sci 966: 73–81, 2002. 550b. Hofbauer LC, Schoppet M: Clinical implications of the osteoprotegerin/ RANKL/RANK system for bone and vascular diseases. JAMA 292: 490–495, 2004. 550c. Hofbauer LC, Kuhne CA, Viereck V: The OPG/RANKL/RANK system in metabolic bone diseases. Musculoskelet Neuronal Interact 4: 268–275, 2004. 551. Zizic TM, Marcoux C, Hungerford DS, et al: Corticosteroid therapy associated with ischemic necrosis of bone in systemic lupus erythematosus. Am J Med 79: 596–604, 1985. 552. Weinstein RS. Glucocorticoid-induced osteoporosis. Rev Endocr Metab Disord 2: 65–73, 2001. 553. Lafage-Proust MH, Boudignon B, Thomas T. Glucocorticoid-induced osteoporosis: pathophysiological data and recent treatments. Joint Bone Spine 70: 109–118, 2003. 554. Bergstein JM, Wiens C, Fish AJ, et al: Avascular necrosis of bone in systemic lupus erythematosus. J Paediatr 85: 31–35, 1974. 555. Ansell BM: Overview of the side effects of corticosteroid therapy. Clin Exp Rheumatol 9: 19–20, 1991. 556. Bozzette SA, Sattler FR, Chiu J, et al: A controlled trial of early adjunctive treatment with corticosteroids for Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome.N Engl J Med 323: 1451–1457, 1990. 557. Fishman JA: Treatment of infection due to Pneumocystis carinii. Antimicrob Agents Chemother 42: 1309–1314, 1998. 558. Schleimer RP: Effects of glucocorticosteroids on inflammatory cells relevant to their therapeutic application in asthma. Am Rev Respir Dis 141: 559, 1990. 559. Lewis DA, Smith RE: Steroid-induced psychiatric syndromes.J Affective Disord 5: 319–332, 1983. 560. Levine SB, Leopold IH: Advances in ocular corticosteroid therapy. Med Clin North Am 57: 1167–1177, 1973. 561. Rogers MP: Psychiatric aspects. In Schur PH (ed): The Clinical Management of Systemic Lupus Erythematosus. New York, Grune & Stratton: 89, 1983. 562. Ling MH, Perry PJ, Tsuang MT: Side effects of corticosteroid therapy. Arch Gen Psych 38: 371, 1981. 563. Klein-Gitelman MS, Pachman LM: Intravenous corticosteroids: adverse reactions are more variable than expected in children. J Rheumatol 25: 1995–2002, 1998. 564. Ilowite NT, Samuel P, Ginzler E, Jacobson MS: Dyslipoproteinemia in pediatric systemic lupus erythematosus. Arthritis Rheum 31: 859, 1988. 565. Petri M, Perez-Gutthann S, Spence D, Hochberg MC: Risk factors for coronary artery disease in patients with systemic lupus erythematosus. Am J Med 93: 513–519, 1992. 566. Gazarian M, Feldman BM, Benson LN, et al: Assessment of myocardial perfusion and function in childhood systemic lupus erythematosus. J Pediatr 132: 109–116, 1998. 567. Lubkin VL: Steroid cataract: a review and conclusion. J Asthma Res 14: 55, 1997. 568. Urban RC, Cotlier E: Corticosteroid-induced cataracts. Surv Opthalmol 31: 102–110, 1986. 569. Knox AJ, Mascie-Taylor BH, Muers MF: Acute hydrocortisone myopathy in severe asthma. Thorax 41: 411–412, 1986. 570. Perlman K, Ehrlich RM: Steroid diabetes in childhood. Am J Dis Child 136: 64–68, 1982. 571. Conn HO, Poynard T: Corticosteroids and peptic ulcer: meta-analysis of adverse events during steroid therapy. J Intern Med 236: 619–632, 1994. 572. Gambertoglio JG, Amend WJ Jr, Benet LZ: Pharmacokinetics and bioavailability of prednisone and prednisolone in healthy volunteers and patients: a review. J Pharmacokinet Biopharm 8: 1, 1980. 573. Myles AB, Schiller LF, Glass D, Daly JR: Single daily dose of corticosteroid treatment. Ann Rheum Dis 35: 73, 1976. 574. Loftus J, Allen R, Hesp R, et al: Randomized, double-blind trial of deflazacort versus prednisone in juvenile chronic (or rheumatoid) arthritis: a relatively bone-sparing effect of deflazacort. Pediatrics 88: 428–436, 1991. 575. Markham A, Bryson HM: Deflazacort: a review of its pharmacological properties and therapeutic efficacy. Drugs 50: 317–333, 1995. 576. David J, Loftus J, Hesp R, et al: Spinal and somatic growth in patients with juvenile chronic arthritis treated for up to 2 years with deflazacort. Clin Exp Rheumatol 10: 621, 1992. 577. Falcini F, Trapani S, Ermini M, Bartolozzi G: Deflazacort in paediatric rheumatic diseases needs a frequent follow-up of bone densitometry [letter]. Pediatrics 95: 318, 1995. 578. MacGregor RR, Sheagren JN, Lipsett MB, Wolff SM: Alternate-day prednisone therapy: evaluation of delayed hypersensitivity responses, control of disease and steroid side effects. N Engl J Med 280: 1427, 1969. 579. Dale DC, Fauci AS, Wolff SM: Alternate day prednisone: leukocyte kinetics and susceptibility to infection. N Engl J Med 291: 1154, 1974. 580. Dixon RB, Christy NP: On the various forms of corticosteroid withdrawal syndrome. Am J Med 68: 224, 1980.
136
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
581. Amatruda TT Jr, Hollingsworth DR, D’Esopo ND, et al: A study of the mechanism of the steroid withdrawal syndrome. J Clin Endocrinol Metab 20: 339, 1960. 582. Laaksonen A–L, Sunell JE, Westeren H, Mulder J: Adrenocortical function in children with juvenile rheumatoid arthritis and other connective tissue disorders. Scand J Rheumatol 3: 137, 1974. 583. Recommendations for the prevention and treatment of glucocorticoidinduced osteoporosis: 2001 update. American College of Rheumatology Ad Hoc Committee on Glucocorticoid-Induced Osteoporosis. Arthritis Rheum 44: 1496–1503, 2001. 584. Adler RA, Hochberg MC: Suggested guidelines for evaluation and treatment of glucocorticoid-induced osteoporosis for the Department of Veterans Affairs. Arch Intern Med 163: 2619–2624, 2003. 585. Geusens PP, de Nijs RN, Lems WF, et al: Prevention of glucocorticoid osteoporosis: a consensus document of the Dutch Society for Rheumatology. Ann Rheum Dis 63: 324–325, 2004. 586. Buckley LM, Leib ES, Cartularo KS, et al: Calcium and vitamin D3 supplementation prevents bone loss in the spine secondary to low dose corticosteroids in patients with rheumatoid arthritis: a randomized, double-blind, placebo-controlled trial. Ann Intern Med 126: 961–968, 1996. 587. Sambrook P, Birmingham J, Kelly P, et al: Prevention of corticosteroid osteoporosis: a comparison of calcium, calcitriol, and calcitonin. N Engl J Med 328: 1747–1752, 1993. 588. Homik J, Suarez-Almazor ME, Shea B, et al: Calcium and vitamin D for corticosteroid-induced osteoporosis. Cochrane Database System Rev 2: CD00952, 2000. 589. Warady BD, Lindsley CB, Robinson RG, Lukert BP: Effects of nutritional supplementation on bone mineral status of children with rheumatic diseases receiving corticosteroid therapy. J Rheumatol 21: 530–535, 1994. 590. Reed A, Haugen M, Pachman LM, Langman CB: 25-Hydroxyvitamin D therapy in children with active juvenile rheumatoid arthritis: short term effects on serum osteocalcin levels and bone mineral density. J Pediatr 119: 657–660, 1991. 591. Bianchi ML: Glucorticoids and bone: some general remarks and some special observations in pediatric patients. Calcif Tissue Int 70: 384–390. 2002. 592. Cimaz R: Osteoporosis in childhood rheumatic diseases: prevention and therapy. Best Pract Res Clin Rheumatol 16: 397–409, 2002. 593. Reid IR, King AR, Alexander CJ, Ibbertson HK: Prevention of steroidinduced osteoporosis with (3-amino-1-hydroxypropylidene)-1, 1-bisphosphonate (APD). Lancet 1: 143–146, 1988. 594. Roux C, Oriente P, Laan R, et al: Randomized trial of effect of cyclical etidronate in the prevention of corticosteroid-induced bone loss. J Clin Endocrinol Metab 83: 1128–1133, 1998. 595. Adachi JD, Bensen WG, Brown J, et al: Intermittent etidronate therapy to prevent corticosteroid-induced osteoporosis. N Engl J Med 337: 382–387, 1997. 596. Saag KG, Emkey R, Schnitzer TJ, et al: Alendronate for the prevention and treatment of glucocorticoid-induced osteoporosis. N Engl J Med 339: 292–299, 1998. 597. Boling EP: Secondary osteoporosis: underlying disease and the risk for glucocorticoid-induced osteoporosis. Clin Ther 26: 1–14, 2004. 598. Brumsen C, Hamdy NAT, Papapoulos SE: Long-term effects of bisphosphonates on the growing skeleton: studies of young patients with severe osteoporosis. Medicine (Baltimore) 76: 266–283, 1997. 599. Glorieux FH, Bishop NJ, Plotkin H, et al: Cyclic administration of pamidronate in children with severe osteogenesis imperfecta. N Engl J Med 339: 947–952, 1998. 600. Falk MJ, Heeger S, Lynch KA, et al: Intravenous bisphosphonate therapy in children with osteogenesis imperfecta. Pediatrics 111: 573–578, 2003. 601. Kauffman RP, Overton TH, Shiflett M, Jennings JC: Osteoporosis in children and adolescent girls: case report of idiopathic juvenile osteoporosis and review of the literature. Obstet Gynecol Surv 56: 492–504, 2001. 602. Bianchi ML, Cimaz R, Bardare M, et al: Efficacy and safety of alendronate for the treatment of osteoporosis in diffuse connective tissue diseases in children: a prospective multicenter study. Arthritis Rheum 43: 1960-1966, 2000. 603. Christy NP: Pituitary-adrenal function during corticosteroid therapy: learning to live with uncertainty. N Engl J Med 326: 266–267, 1992. 604. Cunha Cde F, Silva IN, Finch FL: Early adrenocortical recovery after glucocorticoid therapy in children with leukemia. J Clin Endocrinol Metab 89: 2797–2802, 2004. 605. Gundbjornsson B, Skogseid B, Oberg K, et al: Intact adrenocorticotropic hormone secretion but impaired cortisol response in patients with active rheumatoid arthritis: effect of glucocorticoids. J Rheumatol 23: 596–602, 1996. 606. Graber AL, Ney RE, Nicholson WE, et al: Natural history of pituitary-adrenal recovery following long term suppression with corticosteroids. J Clin Endocrinol Metab 25: 11, 1965. 607. Cathcart ES, Idelson BA, Scheinberg MA, et al: Beneficial effects of methylprednisolone “pulse” therapy in diffuse proliferative lupus nephritis. Lancet 1: 163, 1976. 608. Miller JJ III: Prolonged use of large intravenous steroid pulses in the rheumatic diseases of children. Pediatrics 65: 989–994, 1980. 609. Laxer RM, Stein L, Petty RE: Intravenous pulse methylprednisolone treatment of juvenile dermatomyositis. Arthritis Rheum 30: 328, 1989.
610. Shaikov AV, Maximov AA, Speransky AI, et al: Repetitive use of pulse therapy with methylprednisolone and cyclophosphamide in addition to oral methotrexate in children with systemic juvenile rheumatoid arthritis: preliminary results of a long term study. J Rheumatol 19: 612–616, 1992. 611. Picco P, Gattorno M, Buoncompagni A, et al: 6-Methylprednisolone “minipulses”: a new modality of chronic glucocorticoid treatment in systemic onset juvenile chronic arthritis. Scand J Rheumatol 25: 24–27, 1996. 612. Job-Deslandre C, Menkes CJ: Administration of methylprednisolone pulse in chronic arthritis in children. Clin Exp Rheumatol 9 (Suppl 6): 15–18, 1991. 613. Wallace CA, Sherry DD: Trial of intravenous pulse cyclophosphamide and methylprednisolone in the treatment of severe systemic-onset juvenile rheumatoid arthritis. Arthritis Rheum 40: 1852–1855, 1997. 614. Hayball PJ, Cosh DG, Ahern MJ, et al: High dose oral methylprednisolone in patients with rheumatoid arthritis: pharmacokinetics and clinical response. Eur J Clin Pharmacol 42: 85, 1992. 615. Shipley ME, Bacon PA, Hazleman BL, et al: Pulsed methylprednisolone in active early rheumatoid disease: a dose-ranging study. Br J Rheumatol 27: 211–214, 1988. 616. Walters MT, Cawley MID: Combined suppressive drug treatment in severe refractory rheumatoid disease: an analysis of the relative effects of parenteral methylprednisolone, cyclophosphamide, and sodium aurothiomalate. Ann Rheum Dis 47: 924–929, 1988. 617. Williams IA, Mitchell AD, Rothman W, et al: Survey of the long term incidence of osteonecrosis of the hip and adverse medical events in rheumatoid arthritis after high dose intravenous methylprednisolone. Ann Rheum Dis 47: 930–933, 1988. 618. Allen RC, Gross KR, Laxer RM, et al: Intraarticular triamcinolone hexacetonide in the management of chronic arthritis in children. Arthritis Rheum 29: 997–1001, 1986. 619. Earley A, Cuttica RJ, McCullogh C, Ansell BM: Triamcinolone into the knee joint in juvenile chronic arthritis. Clin Exp Rheumatol 6: 153–155, 1988. 620. Hertzberger-ten Cate R, de Vries-van der Vlugt BCM, van Suijlekom-Smit LWA, Cats A: Intra-articular steroids in pauciarticular juvenile chronic arthritis, type 1. Eur J Pediatr 150: 170–172, 1991. 621. Honkanen VEA, Rautonen JK, Pelkonen PM: Intra-articular glucocorticoids in early juvenile chronic arthritis. Acta Paediatr 82: 1072–1074, 1993. 622. Padeh S, Passwell JH: Intraarticular corticosteroid injection in the management of children with chronic arthritis. Arthritis Rheum 41: 1210–1214, 1998. 623. Dent PB, Walker N: Intra-articular corticosteroids in the treatment of juvenile rheumatoid arthritis. Curr Opin Rheumatol 10: 475–480, 1998. 624. Cleary AG, Murphy HD, Davidson JE: Intra-articular corticosteroid injections in juvenile idiopathic arthritis. Arch Dis Child 88: 192–196, 2003. 625. Smith J, Scuccimari R, Gibbon M, et al: Efficacy of intra-articular triamcinolone hexacetonide in a practice-based cohort of children with juvenile arthritis. J Rheumatol 27 (Suppl 58): 95, 2000. 626. Breit W, Frosch M, Meyer U, et al: A subgroup-specific evaluation of the efficacy of intraarticular triamcinolone hexacetonide in juvenile chronic arthritis. J Rheumatol 27: 2696–2702, 2000. 627. Ravelli A, Manzoni SM, Viola S, et al: Factors affecting the efficacy of intraarticular corticosteroid injection of knees in juvenile idiopathic arthritis. J Rheumatol 28: 2100–2102, 2001. 628. Sherry DD, Stein LD, Reed AM, et al: Prevention of leg length discrepancy in young children with pauciarticular juvenile rheumatoid arthritis by treatment with intraarticular steroids. Arthritis Rheum 42: 2330–2334, 1999. 629. Neidel J, Boehnke M, Kuster RM: The efficacy and safety of intraarticular corticosteroid therapy for coxitis in juvenile rheumatoid arthritis. Arthritis Rheum 46: 1620–1628, 2002. 630. Derendorf H, Mollmann H, Gruner A, et al: Pharmacokinetics and pharmacodynamics of glucocorticoid suspensions after intra-articular administration. Clin Pharmacol Ther 39: 313–317, 1986. 631. Zulian F, Martini G, Gobber D, et al: Comparison of intra-articular triamcinolone hexacetonide and triamcinolone acetonide in oligoarticular juvenile idiopathic arthritis. Rheumatology 42: 1254–1259, 2003. 632. Balogh Z, Ruzsonyi E: Triamcinolone hexacetonide versus betamethasone: a double blind comparative study of the long-term effects of intra-articular steroids in patients with juvenile chronic arthritis. Scand J Rheumatol 67 (Suppl): 80–82, 1987. 633. Grillet B, Dequeker J: Intra-articular steroid injection: a risk-benefit assessment. Drug Safety 5: 205–211, 1990. 634. Job-Deslandre C, Menkes CJ: Complications of intra-articular injections of triamcinolone hexacetonide in chronic arthritis in children. Clin Exp Rheumatol 8: 413–416, 1990. 635. Sparling M, Malleson P, Wood B, Petty R: Radiographic followup of joints injected with triamcinolone hexacetonide for the management of childhood arthritis. Arthritis Rheum 33: 821–826, 1990. 636. Gilsanz V, Bernstein BH: Joint calcification following intraarticular corticosteroid therapy. Radiology 151: 647–649, 1984. 637. Jalavas S, Haapasaari J, Isomaki H: Peri-articular calcification after intraarticular triamcinolone hexacetonide. Scand J Rheumatol 9: 190–192, 1980. 638. Eich GF, Halle F, Hodler J, et al: Juvenile chronic arthritis: imaging of the knees and hips before and after intraarticular steroid injection. Pediatr Radiol 24: 558–563, 1994. 639. Huppertz H-I, Tschammler A, Horwitz AE, Schwab KO: Intraarticular corticosteroids for chronic arthritis in children: efficacy and effects on cartilage and growth. J Pediatr 127: 317–321, 1995.
C H A P T E R 640. Hagelberg S, Magnusson B, Jenner G, Andersson U: Do frequent corticosteroid injections in the knee cause cartilage damage in juvenile chronic arthritis? Long-term follow-up with MRI. J Rheumatol 27 (Suppl 58): 95, 2000. 641. Huppertz H–I, Pfuller H: Transient suppression of endogenous cortisol production after intraarticular steroid therapy for chronic arthritis in children. J Rheumatol 24: 1833–1837, 1997. 642. Lazarevic MB, Skosey JL, Djordjevic-Denic G, et al: Reduction of cortisol levels after single intra-articular and intramuscular steroid injection. Am J Med 99: 370–373, 1995. 643. Elion GB: Biochemistry and pharmacology of purine analogues. Fed Proc 26: 898, 1967. 644. Maibach HI, Epstein WL: Immunologic responses of healthy volunteers receiving azathioprine (Imuran). Int Arch Allergy Appl Immunol 27: 102, 1965. 645. Abdou NI, Sweiman B, Casella SR: Effects of azathioprine therapy on bone marrow-dependent and thymus-dependent cells in man. Clin Exp Immunol 13: 55, 1973. 646. Sharbaugh RJ, Ainsworth SK, Fitts CK: Lack of effect of azathioprine on phytohemagglutinin-induced lymphocyte transformation and established delayed cutaneous hypersensitivity. Int Arch Allergy Appl Immunol 51: 681, 1976. 647. Fox DA, McCune WJ: Immunologic and clinical effects of cytotoxic drugs used in the treatment of rheumatoid arthritis and systemic lupus erythematosus. Concepts Immunopathol 7: 20–78, 1989. 648. De Witte DB, Buick MK, Cyran SE, Maisels MJ: Neonatal pancytopenia and severe combined immunodeficiency associated with antenatal administration of azathioprine and prednisone. J Pediatr 105: 625, 1984. 649. Petri M: Immunosuppressive drug use in pregnancy. Autoimmunity 36: 51–56, 2003 650. Singh G, Fries JF, Williams CA, et al: Toxicity profiles of disease modifying antirheumatic drugs in rheumatoid arthritis. J Rheumatol 18: 188, 1991. 651. Leipold G, Schutz E, Haas JP, Oellerich M: Azathioprine-induced severe pancytopenia due to a homozygous two-point mutation of the thiopurine methyltransferase gene in a patient with juvenile HLA-B27-associated spondylarthritis. Arthritis Rheum 40: 1896–1898, 1997. 652. Stolk JN, Boerbooms AM, de Abreu RA, et al: Reduced thiopurine methyltransferase activity and development of side effects of azathioprine treatment in patients with rheumatoid arthritis. Arthritis Rheum 41: 1858–1866, 1998. 653. Siva C, Yokoyama WM, McLeod HL: Pharmacogenetics in rheumatology: the prospects and limitations of an emerging field. Rheumatology 41: 1273–1279, 2002. 654. McLeod HL, Siva C: The thiopurine S-methyltransferase gene locus: implications for clinical pharmacogenomics. Pharmacogenomics. 3: 89–98, 2002. 655. Bailey RR: Leukopenia due to trimethoprim-azathioprine interaction. N Z Med J 97: 739, 1984. 656. Prior P, Symmons DPM, Hawkins CF, Scott DL: Cancer morbidity in rheumatoid arthritis. Ann Rheum Dis 43: 522, 1984. 657. Silman AJ, Petrie J, Hazleman B, Evans SJW: Lymphoproliferative cancer and other malignancy in patients with rheumatoid arthritis treated with azathioprine: a 20 year follow up study. Ann Rheum Dis 47: 988, 1988. 658. Singh G, Fries JF, Spitz P, Williams CA: Toxic effects of azathioprine in rheumatoid arthritis. Arthritis Rheum 32: 837, 1989. 659. Savolainen HA, Kautiainen H, Isomaki H, et al: Azathioprine in patients with juvenile chronic arthritis: a longterm followup study. J Rheumatol 24: 2444–2450, 1997. 660. Hagelberg S, Lee Y, Bargman J, et al: Longterm followup of childhood lupus nephritis. J Rheumatol 29: 2635–2642, 2002. 661. Eugui EM, Almquist SJ, Muller CD, Allison AC: Lymphocyte-selective cytostatic and immunosuppressive effects of mycophenolic acid in vitro: role of deoxyguanosine nucleotide depletion. Scand J Immunol 33: 161–173, 1991. 662. Platz KP, Sollinger HW, Hullett DA, et al: RS-61443-a new, potent immunosuppressive agent. Transplantation 51: 27–31, 1991. 663. Grailer A, Nichols J, Hullett D, et al: Inhibition of human B cell responses in vitro by RS-61443, cyclosporine A and DAB486IL-2. Transplant Proc 23: 314–315, 1991. 664. Burlingham WJ, Grailer AP, Hullett DA, Sollinger HW: Inhibition of both MLC and in vitro IgG memory response to tetanus toxoid by RS-61443. Transplantation 51: 545–547, 1991. 665. Woo J, Zeevi A, Yao GZ, et al: Effects of FK 506, mycophenolic acid, and bredinin on OKT-3-, PMA-, and alloantigen-induced activation molecule expression on cultured CD4+ and CD8+ human lymphocytes. Transplant Proc 23: 2939–2940, 1991. 666. Smith KG, Isbel NM, Catton MG, et al: Suppression of the humoral immune response by mycophenolate mofetil. Nephrol Dial Transplant 13: 160–164, 1998. 667. Laurent AF, Dumont S, Poindron P, Muller CD: Mycophenolic acid suppresses protein N-linked glycosylation in human monocytes and their adhesion to endothelial cells and to some substrates. Exp Hematol 24: 59–67, 1996. 668. Hauser IA, Johnson DR, Thevenod F, Goppelt-Strube M: Effect of mycophenolic acid on TNF alpha-induced expression of cell adhesion molecules in human venous endothelial cells in vitro. Br J Pharmacol 122: 1315–1322, 1997.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
137
669. Yu CC, Yang CW, Wu MS, Ko YC, et al: Mycophenolate mofetil reduces renal cortical inducible nitric oxide synthase mRNA expression and diminishes glomerulosclerosis in MRL/lpr mice. J Lab Clin Med. 138: 69–77, 2001. 670. Lui SL, Tsang R, Wong D, et al: Effect of mycophenolate mofetil on severity of nephritis and nitric oxide production in lupus-prone MRL/lpr mice. Lupus 11: 411–418, 2002. 671. Sievers TM, Rossi SJ, Ghobrial RM, et al: Mycophenolate mofetil. Pharmacotherapy 17: 1178–1197, 1997. 672. Neumann I, Haidinger M, Jager H, et al: Pharmacokinetics of mycophenolate mofetil in patients with autoimmune diseases compared renal transplant recipients. J Am Soc Nephrol. 14: 721–727, 2003. 673. Filler G, Hansen M, LeBlanc C, et al: Pharmacokinetics of mycophenolate mofetil for autoimmune disease in children. Pediatr Nephrol 18: 445–449, 2003. 674. Buratti S, Szer IS, Spencer CH, et al: Mycophenolate mofetil treatment of severe renal disease in pediatric onset systemic lupus erythematosus. J Rheumatol. 28: 2103–2108, 2001. 675. Filler G, Mai I: Limited sampling strategy for mycophenolic acid area under the curve. Ther Drug Monit 22: 169–173, 2000. 676. Bennett WM: Immunosuppression with mycophenolic acid: one size does not fit all. J Am Soc Nephrol 14: 2414–2416, 2003. 677. Nowack R, Birck R, van der Woude FJ: Mycophenolate mofetil for systemic vasculitis and IgA nephropathy. Lancet 349: 774, 1997. 678. Goldblum R: Therapy of rheumatoid arthritis with mycophenolate mofetil. Clin Exp Rheumatol 11 (Suppl 8): S117–S119, 1993. 679. McMurray RW, Elbourne KB, Lagoo A, Lal S: Mycophenolate mofetil suppresses autoimmunity and mortality in the female NZB × NZW F1 mouse model of systemic lupus erythematosus. J Rheumatol 25: 2364–2370, 1998. 680. Glicklich D, Acharya A: Mycophenolate mofetil therapy for lupus nephritis refractory to intravenous cyclophosphamide. Am J Kidney Dis 32: 318–322, 1998. 681. Fu YF, Liu GL: Mycophenolate mofetil therapy for children with lupus nephritis refractory to both intravenous cyclosphosphamide and cyclosporine. Clin Nephrol 55: 318–321, 2001. 682. Chan TM, Li FK, Tang CS: Efficacy of mycophenolate mofetil in patients with diffuse proliferative lupus nephritis. Hong Kong-Guangzhou Nephrology Study Group. N Engl J Med 343: 1156–1162, 2000. 683. Kapitsinou PP, Boletis JN, Skopouli FN, et al: Lupus nephritis: treatment with mycophenolate mofetil. Rheumatology (Oxf) 43: 377–380, 2004. 684. Dooley MA, Cosio FG, Nachman PH, et al: Mycophenolate mofetil therapy in lupus nephritis: clinical observations. J Am Soc Nephrol 10: 833–839, 1999. 685. Nowack R, Gobel U, Klooker P, et al: Mycophenolate mofetil for maintenance therapy of Wegener’s granulomatosis and microscopic polyangiitis: a pilot study in 11 patients with renal involvement. J Am Soc Nephrol 10: 1965–1971, 1999. 686. Langford CA, Talar-Williams C, Sneller MC: Mycophenolate mofetil for remission maintenance in the treatment of Wegener’s granulomatosis. Arthritis Rheum 51: 278–283, 2004. 687. Larkin G, Lightman S: Mycophenolate mofetil: a useful immunosuppressive in inflammatory eye disease. Ophthalmology 106: 370–374, 1999. 688. Llinares-Tello F, Hernandez-Prats C, Munoz-Ruiz C, et al: Monitoring trough plasma concentrations of mycophenolate mofetil in patients with uveitis. J Clin Pharm Ther 29: 53–58, 2004. 689. Le Ray C, Coulomb A, Elefant E, et al: Mycophenolate mofetil in pregnancy after renal transplantation: a case of major fetal malformations. Obstet Gynecol 103: 1091–1094, 2004. 690. Fischer DS: Alkylating agents. In Fischer DS, Marsh JC (ed): Cancer Therapy. Boston, GK Hall, 1972. 691. Kovarsky J: Clinical pharmacology and toxicology of cyclophosphamide: emphasis on use in rheumatic disease. Semin Arthritis Rheum 12: 359, 1983. 692. Cupps TR, Edgar LC, Fauci AS: Suppression of human B lymphocyte function by cyclophosphamide. J Immunol 128: 2453, 1982. 693. Turk JL, Parker D: Effect of cyclophosphamide on immunological control mechanisms. Immunol Rev 65: 99, 1982. 694. Haubitz M, Bohnenstengel F, Brunkhorst R, et al: Cyclophosphamide pharmacokinetics and dose requirements in patients with renal insufficiency. Kidney Int 61: 1495–1501, 2002. 695. Stockman GD, Heim LR, South MA: Differential effects of cyclophosphamide in B and T cell compartments of adult mice. J Immunol 110: 277, 1973. 696. Hersh EM, Wong VG, Freireich EJ: Inhibition of the local inflammatory response in man by antimetabolites. Blood 27: 38, 1966. 697. Hoffman GS, Leavitt RY, Fleischer TA, et al: Treatment of Wegener’s granulomatosis with intermittent high-dose intravenous cyclophosphamide. Am J Med 89: 403–410, 1990. 698. Miller JJ 3rd, Williams GF, Leissring JC: Multiple late complications of therapy with cyclophosphamide including ovarian destruction. Am J Med 50: 530, 1971. 699. Warne GL, Fairley KF, Hobbs JB, et al: Cyclophosphamide-induced ovarian failure. N Engl J Med 289: 1159, 1973. 700. DeFronzo RA, Braine H, Colvin OM, et al: Water intoxication in man after cyclophosphamide therapy: time course and relation to drug activation. Ann Intern Med 78: 861, 1973. 701. Wall RL, Clausen KP: Carcinoma of the urinary bladder in patients receiving cyclophosphamide. N Engl J Med 293: 271, 1975.
138
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
702. Plotz PH, Klippel JH, Decker JL, et al: Bladder complications in patients receiving cyclophosphamide for systemic lupus erythematosus or rheumatoid arthritis. Ann Intern Med 91: 221, 1979. 703. Trompeter RS, Evans PR, Barratt TM: Gonadal function in boys with steroidresponsive nephrotic syndrome treated with cyclophosphamide for short periods. Lancet 1: 1177, 1981. 704. Devries CR, Freiha FS: Hemorrhagic cystitis: a review. J Urol 143: 1, 1990. 705. Baltus JAM, Boersma JW, Hartman AP, Vandenbroucke JP: The occurrence of malignancies in patients with rheumatoid arthritis treated with cyclophosphamide: a controlled retrospective follow-up. Ann Rheum Dis 42: 368, 1983. 706. Bradley JD, Brandt KD, Katz BP: Infectious complications of cyclophosphamide treatment for vasculitis. Arthritis Rheum 32: 45, 1989. 706a. Malik SW, Myers JL, DeRemee RA, Specks U: Lung toxicity associated with cyclophosphamide use. Two distinct patterns. Am J Respir Crit Care Med 154: 1851–1856, 1996. 706b. Segura A, Yuste A, Cercos A, et al: Pulmonary fibrosis induced by cyclophosphamide. Ann Pharmacother 35: 894–897, 2001. 707. Dooley MA, Falk RJ: Immunosuppressive therapy of lupus nephritis. Lupus 7: 630–634, 1998. 708. Korkmaz A, Oter S, Deveci S, et al: Involvement of nitric oxide and hyperbaric oxygen in the pathogenesis of cyclophosphamide induced hemorrhagic cystitis in rats. J Urol 170: 2498–2502, 2003. 709. Harlow PJ, DeClerck YA, Shore NA, et al: A fatal case of inappropriate ADH secretion induced by cyclophosphamide therapy. Cancer 44: 896–898, 1979. 710. Spital A, Ristow S: Cyclophosphamide induced water intoxication in a woman with Sjögren’s syndrome. J Rheumatol 24: 2473–2475, 1997. 711. Pendse S, Ginsburg E, Singh AK: Strategies for preservation of ovarian and testicular function after immunosuppression. Am J Kidney Dis 43: 772–781, 2004. 712. Rivkees SA, Crawford JD: The relationship of gonadal activity and chemotherapy-induced gonadal damage. JAMA 259: 2123–2125, 1988. 713. Mok CC, Lau CS, Wong RW: Risk factors for ovarian failure in patients with systemic lupus erythematosus receiving cyclophosphamide therapy. Arthritis Rheum 41: 831–837, 1998. 714. Boumpas DT, Austin HA 3rd, Vaughan EM, et al: Risk for sustained amenorrhea in patients with systemic lupus erythematosus receiving intermittent pulse cyclophosphamide therapy. Ann Intern Med 119: 366–369, 1993. 715. Ioannidis JP, Katsifis GE, Tzioufas AG, Moutsopoulos HM: Predictors of sustained amenorrhea from pulsed intravenous cyclophosphamide in premenopausal women with systemic lupus erythematosus. J Rheumatol 29: 2129–2135, 2002. 716. Huong du L, Amoura Z, Duhaut P, et al: Risk of ovarian failure and fertility after intravenous cyclophosphamide: a study in 84 patients. J Rheumatol 29: 2571–2576, 2002. 717. Blumenfeld Z, Avivi I, Linn S, et al: Prevention of irreversible chemotherapy-induced ovarian damage in young women with lymphoma by a gonadotropin-releasing hormone agonist in parallel to chemotherapy. Hum Reprod 11: 1620–1626, 1996. 718. Blumenfeld Z, Shapiro D, Shteinberg M, et al: Preservation of fertility and ovarian function and minimizing gonadotoxicity in young women with systemic lupus erythematosus treated by chemotherapy. Lupus 9: 401–405, 2000. 719. Pereyra Pacheco B, Mendez Ribas JM, Milone G, et al: Use of GnRH analogs for functional protection of the ovary and preservation of fertility during cancer treatment in adolescents: a preliminary report. Gynecol Oncol 81: 391–397, 2001. 720. Masala A, Faedda R, Alagna S, et al: Use of testosteron.e to prevent cyclophosphamide-induced azoospermia. Ann Intern Med 126: 292–295, 1997. 721. Radis CD, Kahl LE, Baker GL, et al: Effects of cyclophosphamide on the development of malignancy and on long-term survival of patients with rheumatoid arthritis: a 20-year followup study. Arthritis Rheum 38: 1120–1127, 1995. 722. Talar-Williams C, Hijazi YM, Walther MM, et al: Cyclophosphamide-induced cystitis and bladder cancer in patients with Wegener granulomatosis. Ann Intern Med 124: 477–484, 1996. 723. Brodsky RA, Petrie M, Smith BD, et al: Immunoablative high-dose cyclophosphamide without stem-cell rescue for refractory, severe autoimmune disease. Ann Intern Med 129: 1031–1035, 1998. 724. Contreras G, Pardo V, Leclercq B, et al: Sequential therapies for proliferative lupus nephritis. N Engl J Med 350: 971–980, 2004. 725. David J, Vouyiouka O, Ansell BM, et al: Amyloidosis in juvenile chronic arthritis: a morbidity and mortality study. Clin Exp Rheumatol 11: 85–90, 1993. 726. Buriot D, Prieue AM, Lebranchu Y, et al: Acute leukemia in 3 children with chronic juvenile arthritis treated with chlorambucil. Arch Fr Pediatr 36: 592–598, 1979. 727. Guesry P, Lenoir G, Broyer M: Gonadal effects of chlorambucil given to prepubertal and pubertal boys for nephrotic syndrome. J Pediatr 92: 299–303, 1978. 728. Savolainen HA: Chlorambucil in severe juvenile chronic arthritis: longterm followup with special reference to amyloidosis. J Rheumatol 26: 898–903, 1999. 729. Hamawy MM: Molecular actions of calcineurin inhibitors. Drug News Perspect 16: 277–282, 2003.
730. Borel J, Feurer C, Magnee C, Stahelin H: Effects of the new anti-lymphocyte cyclosporin A in animals. Immunology 32: 1017, 1977. 731. Fruman DA, Klee CB, Bierer BE, Burakoff SJ: Calcineurin phosphatase activity in T lymphocytes is inhibited by FK 506 and cyclosporin A. Proc Natl Acad Sci U S A 89: 3686–3690, 1992. 732. Schreiber SL, Crabtree JR: The mechanisms of action of cyclosporin A and FK506. Immunol Today 13: 136, 1992. 733. Bunjes D, Hardt C, Rollinghoff M, Wagner H: Cyclosporin A mediates immunosuppression of primary cytotoxic T cell responses by impairing the release of interleukin 1 and interleukin 2. Eur J Immunol 11: 657, 1981. 734. Navarro J, Touraine JL: Comparative study of cycloimmune cyclosporin A on human lymphocyte proliferation in vitro: the lack of an immunosuppressive effect by specific clonal deletion. Int J Immunopharmacol 5: 157, 1983. 735. Kronke M, Leonard WJ, Depper JM, et al: Cyclosporin A inhibits T-cell growth factor gene expression at the level of mRNA transcription. Proc Natl Acad Sci U S A 81: 5214, 1984. 736. Reem GH, Cook LA, Vilcek J: Gamma interferon synthesis by human thymocytes and T lymphocytes inhibited by cyclosporin A. Science 221: 63–65, 1983. 737. Ferraccioli G, Falleti E, De Vita S, et al: Circulating levels of interleukin 10 and other cytokines in rheumatoid arthritis treated with cyclosporin A or combination therapy. J Rheumatol 25: 1874–1879, 1998. 738. Ziolkowska M, Koc A, Luszczykiewicz G, et al: High levels of IL-17 in rheumatoid arthritis patients: IL-15 triggers in vitro IL-17 production via cyclosporin A-sensitive mechanism. J Immunol 164: 2832–2838, 2000. 739. Cho ML, Kim WU, Min SY, et al: Cyclosporine differentially regulates interleukin-10, interleukin-15, and tumor necrosis factor a production by rheumatoid synoviocytes. Arthritis Rheum 46: 42–51, 2002. 740. Ahuja SS, Shrivastav S, Danielpour D, et al: Regulation of transforming growth factor-beta 1 and its receptor by cyclosporine in human T lymphocytes. Transplantation 60: 718–723, 1995. 741. Hernandez GL, Volpert OV, Iniguez MA, et al: Selective inhibition of vascular endothelial growth factor-mediated angiogenesis by cyclosporin A: roles of the nuclear factor of activated T cells and cyclooxygenase 2. J Exp Med 193: 607–620, 2001. 742. Cho ML, Cho CS, Min SY, et al: Cyclosporine inhibition of vascular endothelial growth factor production in rheumatoid synovial fibroblasts. Arthritis Rheum 46: 1202–1209, 2002. 743. Meyer S, Kohler NG, Joly A: Cyclosporine A is an uncompetitive inhibitor of proteasome activity and prevents NF-kB activation. FEBS Lett 413: 354–358, 1997. 744. Holschermann H, Durfeld F, Maus U, et al: Cyclosporine A inhibits tissue factor expression in monocytes/macrophages. Blood 88: 3837–3845, 1996. 745. Andjelic S, Khanna A, Suthanthiran M, Nikolic-Zugic J: Intracellular Ca2+ elevation and cyclosporine A synergistically induce TGF-beta 1-mediated apoptosis in lymphocytes. J Immunol 158: 2527–2534, 1997. 746. Muller S, Adorini L, Appella E, Nagy ZA: Lack of influence of cyclosporine on antigen presentation to lysozyme-specific T cell hybridomas. Transplantation 46 (Suppl 2): 44S–48S, 1988. 747. Knight SC, Roberts M, Macatonia SE, et al: Blocking of acquisition and presentation of antigen by dendritic cells with cyclosporine. Transplantation 46 (Suppl 2): 48S–53S, 1988. 748. Furue M, Katz SI: Cyclosporine A inhibits accessory cell and antigen-presenting cell functions of epidermal Langerhans cells. Transplant Proc 20 (2 Suppl 2): 87–91, 1998. 749. Somerville MF, Scott DG: Neoral: new cyclosporin for old? Br J Rheumatol 36: 1113–1115, 1997. 750. Myers BD, Ross J, Newton L, et al: Cyclosporine-associated chronic nephropathy. N Engl J Med 311: 699–705, 1984. 751. Palestine AG, Austin HA, Balow JE, et al: Renal histopathologic alterations in patients treated with cyclosporin for uveitis. N Engl J Med 314: 1293, 1986. 752. Rodger S, Turney JH, Haynes I, et al: Normal liver function in renal allograft recipients treated with cyclosporine. Transplantation 36: 451–452, 1983. 753. Wells G, Haguenauer D, Shea B, et al: Cyclosporine for rheumatoid arthritis. Cochrane Database Syst Rev 2: CD001083, 2000. 754. Gerards AH, Landewe RB, Prins AP, et al: Cyclosporin A monotherapy versus cyclosporin A and methotrexate combination therapy in patients with early rheumatoid arthritis: a double blind randomised placebo controlled trial. Ann Rheum Dis 62: 291–296, 2003. 755. Marchesoni A, Battafarano N, Arreghini M, et al: Radiographic progression in early rheumatoid arthritis: a 12-month randomized controlled study comparing the combination of cyclosporin and methotrexate with methotrexate alone. Rheumatology (Oxf) 42: 1545–1549, 2003. 756. Pistoia V, Buoncampagni A, Scribanis R, et al: Cyclosporin A in the treatment of juvenile chronic arthritis and childhood polymyositis-dermatomyositis: results of a preliminary study. Clin Exp Rheumatol 11: 203–208, 1993. 757. Reiff A, Rawlings DJ, Shaham B, et al: Preliminary evidence for cyclosporin A as an alternative in the treatment of recalcitrant juvenile rheumatoid arthritis and juvenile dermatomyositis. J Rheumatol 24: 2436–2443, 1997. 758. Gerloni V, Cimaz R, Gattinara M, et al: Efficacy and safety profile of cyclosporin A in the treatment of juvenile chronic (idiopathic) arthritis:
C H A P T E R
759. 760. 761. 762. 763. 764. 765. 766. 767.
768. 769. 770. 771.
772. 773. 774. 775. 776. 777. 778. 779. 780.
781.
782. 783. 784. 785. 786. 787.
results of a 10-year prospective study. Rheumatology (Oxf) 40: 907–913, 2001. Zeller V, Cohen P, Prieur AM, Guillevin L: Cyclosporin A therapy in refractory juvenile dermatomyositis: experience and longterm followup of 6 cases. J Rheumatol 23: 1424–1427, 1996. Walton RC, Nussenblatt RB, Whitcup SM: Cyclosporine therapy for severe sight-threatening uveitis in children and adolescents. Ophthalmology 105: 2028–2034, 1998. Kilmartin DJ, Forrester JV, Dick AD: Cyclosporin A therapy in refractory non-infectious childhood uveitis. Br J Ophthalmol 82: 737–742, 1998. Ravelli A, Moretti C, Temporini F, et al: Combination therapy with methotrexate and cyclosporine A in juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 569–572, 2002. Hallegua D, Wallace DJ, Metzger AL, et al: Cyclosporine for lupus membranous nephritis: experience with ten patients and review of the literature. Lupus 9: 241–251, 2000. Fu LW, Yang LY, Chen WP, Lin CY: Clinical efficacy of cyclosporin A Neoral in the treatment of paediatric lupus nephritis with heavy proteinuria. Br J Rheumatol 37: 217–221, 1998. Griffiths B, Emery P: The treatment of lupus with cyclosporin A. Lupus 10: 165–170, 2001. van den Borne BE, Landewe R–B, Houkes I, et al: No increased risk of malignancies and mortality in cyclosporin A-treated patients with rheumatoid arthritis. Arthritis Rheum 41: 1930–1937, 1998. Ioannides-Demos LL, Christophidis N, Ryan P, et al: Dosing implications of a clinical interaction between grapefruit juice and cyclosporine and metabolite concentrations in patients with autoimmune diseases. J Rheumatol 24: 49–54, 1997. Birdi N, Allen U, D’Astous J: Poststreptococcal reactive arthritis mimicking acute septic arthritis: a hospital-based study. J Pediatr Orthop 15: 661–665, 1995. Fink CW: The role of the streptococcus in poststreptococcal reactive arthritis and childhood polyarteritis nodosa. J Rheumatol Suppl 29: 14–20, 1991. Stegeman CA, Tervaert JWC, de Jong PE, Kallenberg CGM: Trimethoprimsulfamethoxazole (co-trimoxazole) for the prevention of relapses of Wegener’s granulomatosis. N Engl J Med 335: 16–20, 1996. Stone M, Fortin PR, Pacheco-Tena C, Inman RD: Should tetracycline treatment be used more extensively for rheumatoid arthritis? Metaanalysis demonstrates clinical benefit with reduction in disease activity. J Rheumatol 30: 2112–2122, 2003. O’Dell JR, Haire CE, Palmer W, et al: Treatment of early rheumatoid arthritis with minocycline or placebo: results of a randomized, double-blind, placebo-controlled trial. Arthritis Rheum 40: 842–848, 1997. O’Dell JR, Blakely KW, Mallek JA, et al: Treatment of early seropositive rheumatoid arthritis: a two-year, double-blind comparison of minocycline and hydroxychloroquine. Arthritis Rheum 44: 2235–2241, 2001. Sieper J, Braun J: Treatment of reactive arthritis with antibiotics. Br J Rheumatol 37: 717–720, 1998. Le CH, Morales A, Trentham DE: Minocycline in early diffuse scleroderma. Lancet 352: 1755–1756, 1998. Mayes MD, O’Donnell D, Rothfield NF, Csuka ME: Minocycline is not effective in systemic sclerosis: results of an open-label multicenter trial. Arthritis Rheum 50: 553–557, 2004. Oates-Whitehead RM, Baumer JH, Haines L: Intravenous immunoglobulin for the treatment of Kawasaki disease in children. Cochrane Database Syst Rev 4: CD004000, 2003. Lang BA, Laxer RM, Murphy G, et al: Treatment of dermatomyositis with intravenous gammaglobulin. Am J Med 91: 169–172, 1991. Uziel Y, Laxer RM, Schneider R, Silverman ED: Intravenous immunoglobulin therapy in systemic onset juvenile rheumatoid arthritis: a followup study. J Rheumatol 23: 910–918, 1996. Silverman ED, Cawkwell GD, Lovell DJ, et al: Intravenous immunoglobulin in the treatment of systemic juvenile rheumatoid arthritis: a randomized placebo controlled trial. Pediatr Rheumatol Collaborative Study Group. J Rheumatol 21: 2353–2358, 1994. Giannini EH, Lovell DJ, Silverman ED, et al: Intravenous immunoglobulin in the treatment of polyarticular juvenile rheumatoid arthritis: a phase I/II study. Pediatr Rheumatol Collaborative Study Group. J Rheumatol 23: 919–924, 1996. Braun-Moscovici Y, Furst DE: Immunoglobulin for rheumatic diseases in the twenty-first century: take it or leave it? Curr Opin Rheumatol 15: 237–245, 2003. Duhem C, Dicato MA, Ries F: Side-effects of intravenous immune globulins. Clin Exp Immunol 97 (Suppl 1): 79–83, 1994. Yu Z, Lennon VA: Mechanism of intravenous immune globulin therapy in antibody-mediated autoimmune diseases. N Engl J Med 340: 227–228, 1999. Patel SY, Kumararatne DS: From black magic to science: understanding the rationale for the use of intravenous immunoglobulin to treat inflammatory myopathies. Clin Exp Immunol 124: 169–171, 2001. Nydegger UE, Blaser K, Hassig A: Anti-idiotypic immunosuppression and its treatment with human immunoglobulin preparations. Vox Sang 47: 92, 1984. Hahn BH, Ebling FM: Suppression of murine lupus nephritis by administration of an anti-idiotypic antibody to anti-DNA. J Immunol 132: 1887, 1984.
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
139
788. Svenson M, Hansen MB, Bendtzen K: Distribution and characterization of autoantibodies to interleukin 1 alpha in normal human sera. Scand J Immunol 32: 695, 1990. 789. Hansen M, Svenson M, Diamant M, Bendtzen K: Anti-interleukin-6 antibodies in normal human serum. Scand J Immunol 33: 777, 1991. 790. Fomsgaard A, Svenson M, Bendtzen K: Autoantibodies to tumour necrosis factor alpha in healthy humans and patients with inflammatory disease and gram-negative bacterial infections. Scand J Immunol 30: 219, 1989. 791. Ross C, Hansen MB, Schyberg T, Berg K: Autoantibodies to crude human leukocyte IFN, recombinant IFN-alpha 2b, and human IFN gamma in healthy blood donors. Clin Exp Immunol 8: 57, 1990. 792. Saulsbury FT: The effect of intravenous immunoglobulin on lymphocyte populations in children with Kawasaki syndrome. Clin Exp Rheumatol 10: 617–620, 1992. 793. Takei S, Arora YK, Walker SM: Intravenous immunoglobulin contains specific antibodies inhibitory to activation of T cells by staphylococcal toxin superantigens. J Clin Invest 91: 602–607, 1993. 794. Dalakas MC, Illa I, Dambrosia JM, et al: A controlled trial of high-dose intravenous immune globulin infusions as treatment for dermatomyositis. N Engl J Med 329: 1993–2000, 1993. 795. Wallis WJ, Furst DE, Strand V, Keystone E: Biologic agents and immunotherapy in rheumatoid arthritis: progress and perspective. Rheum Dis Clin North Am 24: 537–565, 1998. 796. Weiner HL, Friedman A, Miller A, et al: Oral tolerance: immunologic mechanisms and treatment of animal and human organ-specific autoimmune diseases by oral administration of autoantigens. Annu Rev Immunol 12: 809–837, 1994. 797. Trentham DE, Dynesius-Trentham RA, Orav EJ, et al: Effects of oral administration of type II collagen on rheumatoid arthritis. Science 261: 1727–1730, 1993. 798. Barnett ML, Kremer JM, St Clair EW, et al: Treatment of rheumatoid arthritis with oral type II collagen: results of a multicenter, double-blind, placebo-controlled trial. Arthritis Rheum 41: 290–297, 1998. 799. Barnett ML, Combitchi D, Trentham DE: A pilot trial of oral type II collagen in the treatment of juvenile rheumatoid arthritis. Arthritis Rheum 39: 623–628, 1996. 800. Furie RA, Cash JM, Cronin ME, et al: Treatment of systemic lupus erythematosus with LJP 394. J Rheumatol 28: 257–264, 2001. 801. Alarcon-Segovia D, Tumlin JA, Furie RA, et al: LJP 394 for the prevention of renal flare in patients with systemic lupus erythematosus: results from a randomized, double-blind, placebo-controlled study. Arthritis Rheum 48: 442–454, 2003. 802. Strand V, Aranow C, Cardiel MH, et al: Improvement in health-related quality of life in systemic lupus erythematosus patients enrolled in a randomized clinical trial comparing LJP 394 treatment with placebo. Lupus 12: 677–686, 2003. 803. Vandenbark AA, Morgan E, Bartholomew R, et al: TCR peptide therapy in human autoimmune diseases. Neurochem Res 26: 713–730, 2001. 804. Thompson SD, Murray KJ, Grom AA, et al: Comparative sequence analysis of the human T cell receptor β chain in juvenile rheumatoid arthritis and juvenile spondylarthropathies: evidence for antigenic selection of T cells in the synovium. Arthritis Rheum 41: 482–497, 1998. 805. Kirkham BW, Pitzalis C, Kingsley GH, et al: Monoclonal antibody treatment in rheumatoid arthritis: the clinical and immunological effects of a CD7 monoclonal antibody. Br J Rheumatol 30: 459–463, 1991. 806. Kirkham BW, Thien F, Pelton BK, et al: Chimeric CD7 monoclonal antibody therapy in rheumatoid arthritis. J Rheumatol 19: 1348–1352, 1992. 807. Olsen NJ, Brooks RH, Cush JJ, et al: A double-blind, placebo-controlled study of anti-CD5 immunoconjugate in patients with rheumatoid arthritis. The Xoma RA Investigator Group. Arthritis Rheum 39: 1102–1108, 1996. 808. Isaacs JD, Watts RA, Hazelman BL, et al: Humanised monoclonal antibody therapy for rheumatoid arthritis. Lancet 340: 748–752, 1992. 809. Isaacs JD, Manna VK, Rapson N, et al: CAMPATH-1H in rheumatoid arthritis: an intravenous dose-ranging study. Br J Rheumatol 35: 231–240, 1996. 810. Choy EH, Chicanza IC, Kingsley GH, et al: Treatment of rheumatoid arthritis with single dose or weekly pulses of chimaeric anti-CD4 monoclonal antibody. Scand J Immunol 36: 291–298, 1992. 811. Choy EH, Pitzalis C, Cauli A, et al: Percentage of anti-CD4 monoclonal antibody-coated lymphocytes in the rheumatoid joint is associated with clinical improvement: implications for the development of immunotherapeutic dosing regimens. Arthritis Rheum 39: 52–56, 1996. 812. Choy EH, Kingsley GH, Panayi GS: Monoclonal antibody therapy in rheumatoid arthritis. Br J Rheumatol 37: 484–490, 1998. 813. Choy EH, Panayi GS, Emery P, et al: Repeat-cycle study of high-dose intravenous 4162W94 anti-CD4 humanized monoclonal antibody in rheumatoid arthritis: a randomized placebo-controlled trial. Rheumatology (Oxf) 41: 1142–1148, 2002. 814. Durie FH, Fava RA, Foy TM, et al: Prevention of collagen-induced arthritis with an antibody to gp39, the ligand for CD40. Science 261: 1328–1330, 1993. 815. Webb LM, Walmsley MJ, Feldmann M: Prevention and amelioration of collagen-induced arthritis by blockade of the CD28 co-stimulatory pathway: requirement for both B7-1 and B7-2. Eur J Immunol 26: 2320–2328, 1996. 816. Kremer JM: Cytotoxic T-lymphocyte antigen 4-immunoglobulin in rheumatoid arthritis. Rheum Dis Clin North Am 30: 381–391, 2004.
140
C H A P T E R
5 PHARMACOLOGY
AND
D RU G T H E R A P Y
817. Moreland LW, Alten R, Van den Bosch F, et al: Costimulatory blockade in patients with rheumatoid arthritis: a pilot, dose-finding, double-blind, placebo-controlled clinical trial evaluating CTLA-4Ig and LEA29Y eightyfive days after the first infusion. Arthritis Rheum 46: 1470–1479, 2002. 818. Kremer JM, Westhovens R, Leon M, et al: Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA4Ig. N Engl J Med 349: 1907–1915, 2003. 819. Kalunian KC, Davis JC Jr, Merrill JT, et al: Treatment of systemic lupus erythematosus by inhibition of T cell costimulation with anti-CD154: a randomized, double-blind, placebo-controlled trial. IDEC-131 Lupus Study Group. Arthritis Rheum 46: 3251–3258, 2002. 820. Boumpas DT, Furie R, Manzi S, et al: Lessons learned from anti-CD40L treatment in systemic lupus erythematosus patients. BG9588 Lupus Nephritis Trial Group. Arthritis Rheum 48: 719–727, 2003. 821. Sidiropoulos PI, Boumpas DT: Lessons learned from anti-CD40L treatment in systemic lupus erythematosus patients, Lupus: 13: 391–397, 2004. 822. Kavanaugh AF, Davis LS, Nichols LA, et al: Treatment of refractory rheumatoid arthritis with a monoclonal antibody to intercellular adhesion molecule 1. Arthritis Rheum 37: 992–999, 1994. 823. Kavanaugh AF, Schulze-Koops S, Davis LS, Lipsky PE: Repeat treatment of rheumatoid arthritis patients with a murine anti-intercellular adhesion molecule 1 monoclonal antibody. Arthritis Rheum 40: 849–853, 1997. 824. Edwards JC, Leandro MJ, Cambridge G: B lymphocyte depletion therapy with rituximab in rheumatoid arthritis. Rheum Dis Clin North Am 30: 393–403, 2004. 825. Quartier P, Brethon B, Philippet P, et al: Treatment of childhood autoimmune haemolytic anaemia with rituximab. Lancet 358: 1511–1513, 2001. 826. Binstadt BA, Caldas AM, Turvey SE, et al: Rituximab therapy for multisystem autoimmune diseases in pediatric patients. J Pediatr 143: 598–604, 2003. 827. Leandro MJ, Edwards JC, Cambridge G, et al: An open study of B lymphocyte depletion in systemic lupus erythematosus. Arthritis Rheum 46: 2673–2677, 2002. 828. Silverman GJ, Weisman S: Rituximab therapy and autoimmune disorders: prospects for anti-B cell therapy. Arthritis Rheum 48: 1484–1492, 2003. 829. Looney RJ, Anolik J, Sanz I: B cells as therapeutic targets for rheumatic diseases. Curr Opin Rheumatol 16: 180–185, 2004. 830. De Benedetti F, Ravelli A, Martini A: Cytokines in juvenile rheumatoid arthritis. Curr Opin Rheumatol 9: 428–433, 1997. 831. Muller K, Herner EB, Stagg A, et al: Inflammatory cytokines and cytokine antagonists in whole blood cultures of patients with systemic juvenile chronic arthritis. Br J Rheumatol 37: 562–569, 1998. 832. Mangge H, Schauenstein K: Cytokines in juvenile rheumatoid arthritis (JRA). Cytokine 10: 471–480, 1998. 833. Mangge H, Kenzian H, Gallistl S, et al: Serum cytokines in juvenile rheumatoid arthritis: correlation with conventional inflammation parameters and clinical subtypes. Arthritis Rheum 38: 211–220, 1995. 834. Muzaffer MA, Dayer JM, Feldman BM, et al: Differences in the profiles of circulating levels of soluble tumor necrosis factor receptors and interleukin 1 receptor antagonist reflect the heterogeneity of the subgroups of juvenile rheumatoid arthritis. J Rheumatol 29: 1021–1080, 2002. 835. Genovese MC, Kremer JM: Treatment of rheumatoid arthritis with etanercept. Rheum Dis Clin North Am 30: 311–328, 2004. 836. Lovell DJ, Giannini EH, Reiff A, et al: Etanercept in children with polyarticular juvenile rheumatoid arthritis. Pediatric Rheumatology Collaborative Study Group. N Engl J Med 16: 763–769, 2000. 837. Quartier P, Taupin P, Bourdeaut F, et al: Efficacy of etanercept for the treatment of juvenile idiopathic arthritis according to the onset type. Arthritis Rheum 48: 1093–1101, 2003. 838. Lovell DJ, Giannini EH, Reiff A, et al: Long-term efficacy and safety of etanercept in children with polyarticular-course juvenile rheumatoid arthritis: interim results from an ongoing multicenter, open-label, extended-treatment trial. Arthritis Rheum 48: 218–226, 2003. 839. Russo RA, Katsicas MM, Zelazko M. Etanercept in systemic juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 723–726, 2002. 840. ten Cate R, van Suijlekom-Smit LW, Brinkman DM, et al: Etanercept in four children with therapy-resistant systemic juvenile idiopathic arthritis. Rheumatology (Oxf) 41: 228–229, 2002. 841. Klareskog L, van der Heijde D, de Jager JP, et al: Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial. Lancet 28: 675–681, 2004. 842. Weinblatt ME, Kremer JM, Bankhurst AD, et al: A trial of etanercept, a recombinant tumor necrosis factor receptor:Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate. N Engl J Med 240: 253–259, 1999. 843. Schmeling H, Mathony K, John V, Keysser G, et al: A combination of etanercept and methotrexate for the treatment of refractory juvenile idiopathic arthritis: a pilot study. Ann Rheum Dis 60: 410–412, 2001. 844. Lahdenne P, Vahasalo P, Honkanen V: Infliximab or etanercept in the treatment of children with refractory juvenile idiopathic arthritis: an open label study. Ann Rheum Dis 62: 245–247, 2003. 845. Takei S, Groh D, Bernstein B, et al: Safety and efficacy of high dose etanercept in treatment of juvenile rheumatoid arthritis. J Rheumatol 28: 1677–1680, 2001. 846. Keystone EC, Schiff MH, Kremer JM, et al: Once-weekly administration of 50 mg etanercept in patients with active rheumatoid arthritis: results of a multi-
847.
848. 849. 850. 851. 852. 853. 854. 855. 856.
857. 858. 859. 860. 861.
862.
862a.
863. 864. 865. 866. 867. 868. 869. 870.
870a. 871.
center, randomized, double-blind, placebo-controlled trial. Arthritis Rheum 50: 353–363, 2004. TNF neutralization in MS: results of a randomized, placebo-controlled multicenter study. The Lenercept Multiple Sclerosis Study Group and The University of British Columbia MS/MRI Analysis Group. Neurology 53: 457–465, 1999. Mohan N, Edwards ET, Cupps TR, et al: Demyelination occurring during anti-tumor necrosis factor alpha therapy for inflammatory arthritides. Arthritis Rheum 44: 2862–2869, 2001. Livermore PA, Murray KJ: Anti-tumour necrosis factor therapy associated with cutaneous vasculitis. Rheumatology (Oxf) 41: 1450–1452, 2002. Lepore L, Marchetti F, Facchini S, et al: Drug-induced systemic lupus erythematosus associated with etanercept therapy in a child with juvenile idiopathic arthritis. Clin Exp Rheumatol 21: 276–277, 2003. Cush JJ: Safety overview of new disease-modifying antirheumatic drugs. Rheum Dis Clin North Am 30: 237–255, 2004. Bloom BJ: Development of diabetes mellitus during etanercept therapy in a child with systemic-onset juvenile rheumatoid arthritis. Arthritis Rheum 43: 2606–2608, 2000. Mease PJ, Kivitz AJ, Burch FX, et al: Etanercept treatment of psoriatic arthritis: safety, efficacy, and effect on disease progression. Arthritis Rheum 50: 2264–2272, 2004. Gorman JD, Sack KE, Davis JC Jr: Treatment of ankylosing spondylitis by inhibition of tumor necrosis factor alpha. N Engl J Med 346: 1349–1356, 2002. Reiff A: Long-term outcome of etanercept therapy in children with treatment-refractory uveitis. Arthritis Rheum 48: 2079–2080, 2003. Stone JH: Limited versus severe Wegener’s granulomatosis: baseline data on patients in the Wegener’s granulomatosis etanercept trial. Wegener’s Granulomatosis Etanercept Trial Research Group. Arthritis Rheum 48: 2299–2309, 2003. Maini SR: Infliximab treatment of rheumatoid arthritis. Rheum Dis Clin North Am 30: 329–347, 2004. Elliott MJ, Maini RN, Feldmann M, et al: Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet 344: 1105–1110, 1994. Elliott MJ, Maini RN, Feldmann M, et al: Repeated therapy with monoclonal antibody to tumour necrosis factor alpha (cA2) in patients with rheumatoid arthritis. Lancet 344: 1125–1127, 1994. Kimura Y, Imundo LF, Li SC: High dose infliximab in the treatment of resistant systemic juvenile rheumatoid arthritis. Arthritis Rheum 44: S272, 2001. Maini RN, Breedveld FC, Kalden JR, et al: Sustained improvement over two years in physical function, structural damage, and signs and symptoms among patients with rheumatoid arthritis treated with infliximab and methotrexate. Arthritis Rheum 50: 1051–1065, 2004. Lipsky PE, Felson DT, Maini RN: Blocking tumor necrosis factor inhibits radiographic damage even in patients who show minimal or no clinical improvement: comment on the concise communication by Boers. Arthritis Rheum 46: 2817–2818, 2002. Gerloni V, Pontikaki I, Gattinara M, et al: Efficacy of repeated intravenous infusions of an anti-tumor necrosis factor alpha monoclonal antibody, infliximab, in persistently active, refractory juvenile idiopathic arthritis: Results of an open-label prospective study. Arthritis Rheum 52: 548–553, 2005. Wolfe F, Michaud K, Anderson J, Urbansky K: Tuberculosis infection in patients with rheumatoid arthritis and the effect of infliximab therapy. Arthritis Rheum 50: 372–379, 2004. Booth A, Harper L, Hammad T, et al: Prospective study of TNFalpha blockade with infliximab in anti-neutrophil cytoplasmic antibody-associated systemic vasculitis. J Am Soc Nephrol 15: 717–721, 2004. Weiss JE, Eberhard BA, Chowdhury D, Gottlieb BS. Infliximab as a novel therapy for refractory Kawasaki disease. J Rheumatol 31: 818–810, 2004. Pritchard C, Nadarajah K: Tumour necrosis factor alpha inhibitor treatment for sarcoidosis refractory to conventional treatments: a report of five patients. Ann Rheum Dis 63: 318–320, 2004. Murphy CC, Ayliffe WH, Booth A, et al: Tumor necrosis factor alpha blockade with infliximab for refractory uveitis and scleritis. Ophthalmology 111: 352–356, 2004. Ohno S, Nakamura S, Hori S, et al: Efficacy, safety, and pharmacokinetics of multiple administration of infliximab in Behcet’s disease with refractory uveoretinitis. J Rheumatol 31: 1362–1368, 2004. Keystone E, Haraoui B: Adalimumab therapy in rheumatoid arthritis. Rheum Dis Clin North Am 30: 349–364, 2004. Weinblatt ME, Keystone EC, Furst DE, et al: Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum 48: 35–45 2003. Lovell DJ, Ruperto N, Goodman S et al: Preliminary data from the study of adalimumab in children with juvenile idiopathic arthritis (JIA). Arthritis Rheum; 40: S436–437, 2004. Bieber J, Kavanaugh A: Consideration of the risk and treatment of tuberculosis in patients who have rheumatoid arthritis and receive biologic treatments. Rheum Dis Clin North Am 30: 257–270, 2004.
C H A P T E R 872. Khanna D, McMahon M, Furst DE: Anti-tumor necrosis factor alpha therapy and heart failure: what have we learned and where do we go from here? Arthritis Rheum 50: 1040–1050, 2004. 873. Kwon HJ, Cote TR, Cuffe MS, et al: Case reports of heart failure after therapy with a tumor necrosis factor antagonist. Ann Intern Med 138: 807–811, 2003. 874. Cohen SB: The use of anakinra, an interleukin-1 receptor antagonist, in the treatment of rheumatoid arthritis. Rheum Dis Clin North Am 30: 365–380, 2004. 875. Cohen SB, Moreland LW, Cush JJ, et al: A multicentre, double-blind, randomised, placebo controlled trial of anakinra (Kineret), a recombinant interleukin 1 receptor antagonist, in patients with rheumatoid arthritis treated with background methotrexate. Ann Rheum Dis 63: 1062–1068, 2004. 876. Bresnihan B, Cobby M: Clinical and radiological effects of anakinra in patients with rheumatoid arthritis. Rheumatology (Oxf) 42 (Suppl 2): 22–28, 2003. 877. Rau R, Sander O, Wassenberg S: Erosion healing in rheumatoid arthritis after anakinra treatment. Ann Rheum Dis 62: 671–673, 2003. 878. Genovese MC, Cohen S, Moreland L, et al; 20000223 Study Group. Combination therapy with etanercept and anakinra in the treatment of patients with rheumatoid arthritis who have been treated unsuccessfully with methotrexate. Arthritis Rheum 50: 1412–1419, 2004. 879. Buch MH, Bingham SJ, Seto Y, et al: Lack of response to anakinra in rheumatoid arthritis following failure of tumor necrosis factor alpha blockade. Arthritis Rheum 50: 725–728, 2004. 880. Reiff A, Porras O, Rudge S, et al: Preliminary data from a study of Kineret (anakinra) in children with juvenile rheumatoid arthritis. Arthritis Rheum 46: S215, 2002. 880a. Irigoyen PI, Olson J, Hom C, Ilowite NT: Treatment of systemic onset juvenile rheumatoid arthritis with anakinra. Arthritis Rheum; 40: S437–438, 2004. 880b. Henrickson M: Efficacy of anakinra in refractory systemic arthritis. Arthritis Rheum; 40: S438, 2004. 881. Choy E: Clinical experience with inhibition of interleukin-6. Rheum Dis Clin North Am 30: 405–415, 2004. 882. De Benedetti F, Martini A: Is systemic juvenile rheumatoid arthritis an interleukin 6 mediated disease? J Rheumatol 25: 203–207, 1998. 883. De Benedetti F, Alonzi T, Moretta A, et al: Interleukin 6 causes growth impairment in transgenic mice through a decrease in insulin-like growth factor-I: a model for stunted growth in children with chronic inflammation. J Clin Invest 99: 643–650, 1997. 884. Takagi N, Mihara M, Moriya Y, et al: Blockage of interleukin-6 receptor ameliorates joint disease in murine collagen-induced arthritis. Arthritis Rheum 41: 2117–2121, 1998. 885. Iwamoto M, Nara H, Hirata D, et al: Humanized monoclonal anti-interleukin-6 receptor antibody for treatment of intractable adult-onset Still’s disease. Arthritis Rheum 46: 3388–3389, 2002. 886. Nishimoto N, Sasai M, Shima Y, et al: Improvement in Castleman’s disease by humanized anti-interleukin-6 receptor antibody therapy. Blood 95: 56–61, 2000. 887. Choy EH, Isenberg DA, Garrood T, et al: Therapeutic benefit of blocking interleukin-6 activity with an anti-interleukin-6 receptor monoclonal antibody in rheumatoid arthritis: a randomized, double-blind, placebo-controlled, dose-escalation trial. Arthritis Rheum 46: 3143–3150, 2002. 888. Yokota S, Miyamae T, Imagawa T, et al: Phase II trial on anti-IL6-receptor antibody for children with systemic-onset juvenile idiopathic arthritis. Arthritis Rheum 48: S429, 2003. 888a. Yokota S, Imagawa T, Miyamae T, Mori M, Nishimoto N, Kishimoto T: Longterm therapeutic experience of humanized anti-IL-6 receptor monoclonal antibody (aIL-6R Mab) in systemic-onset juvenile idiopathic arthritis (SoJIA). Arthritis Rheum; 40: S438, 2004. 889. Nakajima T, Aono H, Hasunuma T, et al: Apoptosis and functional Fas antigen in rheumatoid arthritis synoviocytes. Arthritis Rheum 38: 485–491, 1995. 890. Felson DT, Anderson JJ, Meenan RJ: The comparative efficacy and toxicity of second-line drugs in rheumatoid arthritis: results of two metaanalyses. Arthritis Rheum 33: 1449–1461, 1990. 891. Stein CM, Pincus T, Yocum D, et al: Combination treatment of severe rheumatoid arthritis with cyclosporine and methotrexate for forty-eight weeks: an open-label extension study. The Methotrexate-Cyclosporine Combination Study Group. Arthritis Rheum 40: 1843–1851, 1997. 892. Ferraz MB, Pinheiro T, Helfenstein M, et al: Combination therapy with methotrexate and chloroquine in rheumatoid arthritis: a multicenter randomized placebo-controlled trial. Scand J Rheumatol 23: 231–236, 1994. 893. Boers M, Kostense PJ, Verhoeven AC, van der Linden S; COBRA Trial Group: Combinatietherapie Bij Reumatoide Artritis. Inflammation and damage in an individual joint predict further damage in that joint in
5 PHARMACOLOGY 894. 895. 896. 897. 898. 899. 900. 901.
902. 903. 904. 905. 906. 907. 908. 909. 910.
911.
912. 913. 914. 915. 916. 917. 918. 919.
AND
D RU G T H E R A P Y
141
patients with early rheumatoid arthritis. Arthritis Rheum 44: 2242–2246, 2001. Landewe RB, Boers M, Verhoeven AC, et al: COBRA combination therapy in patients with early rheumatoid arthritis: long-term structural benefits of a brief intervention. Arthritis Rheum 46: 347–356, 2002. Hansen KE, Cush J, Singhal A, et al: The safety and efficacy of leflunomide in combination with infliximab in rheumatoid arthritis. Arthritis Rheum 51: 228–232, 2004. Londino AV, Rothman D, Robbins PD, Evans CH: Gene therapy for juvenile rheumatoid arthritis? J Rheumatol 27 (Suppl 58): 53–55, 2000. Evans CH, Ghivizzani SC, Kang R, et al: Gene therapy for rheumatic diseases. Arthritis Rheum 42: 1–16, 1999. Muller-Ladner U, Pap T, Gay RE, Gay S: Gene transfer as a future therapy for rheumatoid arthritis. Expert Opin Biol Ther 3: 587–589, 2003. Robbins PD, Evans CH, Chernajovsky Y: Gene therapy for arthritis. Gene Ther 10: 902–911, 2003. van Bekkum DW, Bohre EP, Houben PF, Knaan-Shanzer S: Regression of adjuvant-induced arthritis in rats following bone marrow transplantation. Proc Natl Acad Sci U S A 86: 10090–10094, 1989. Knaan-Shanzer S, Houben PF, Kinwel-Bohre EP, van Bekkum DW: Remission induction of adjuvant arthritis in rats by total body irradiation and autologous bone marrow transplantation. Bone Marrow Transplant 8: 333–338, 1991. McKendry RJ, Huebsch L, Leclair B: Progression of rheumatoid arthritis following bone marrow transplantation: a case report with a 13-year followup. Arthritis Rheum 39: 1246–1253, 1996. Snowden JA, Kearney P, Kearney A, et al: Long-term outcome of autoimmune disease following allogeneic bone marrow transplantation. Arthritis Rheum 41: 453–459, 1998. Euler HH, Marmont AM, Bacigalupo A, et al: Early recurrence or persistence of autoimmune diseases after unmanipulated autologous stem cell transplantation. Blood 88: 3621–3625, 1996. Wulffraat N, van Royen A, Bierings M, et al: Autologous haemopoietic stem-cell transplantation in four patients with refractory juvenile chronic arthritis. Lancet 353: 550–553, 1999. Martini A, Maccario R, Ravelli A, et al: Marked and sustained improvement two years after autologous stem cell transplantation in a girl with systemic sclerosis. Arthritis Rheum 42: 807–811, 1999. Barron KS, Wallace C, Woolfrey CEA, et al: Autologous stem cell transplantation for pediatric rheumatic diseases. Rheumatol 28: 2337–2358, 2001. Wulffraat NM, Brinkman D, Ferster A, et al: Long-term follow-up of autologous stem cell transplantation for refractory juvenile idiopathic arthritis. Bone Marrow Transplant 32 (Suppl 1): S61–S64, 2003. Wulffraat NM, Rijkers GT, Elst E, et al: Reduced perforin expression in systemic juvenile idiopathic arthritis is restored by autologous stem-cell transplantation. Rheumatology (Oxf) 42: 375–379, 2003. Wulffraat NM, Haas PJ, Frosch M, et al: Myeloid related protein 8 and 14 secretion reflects phagocyte activation and correlates with disease activity in juvenile idiopathic arthritis treated with autologous stem cell transplantation. Ann Rheum Dis 62: 236–241, 2003. Brinkman DM, ten Cate R, Vossen JM, et al: Decrease in synovial cellularity and cytokine expression after autologous stem cell transplantation in patients with juvenile idiopathic arthritis. Arthritis Rheum 46: 1121–1123, 2002. Traynor AE, Barr WG, Rosa RM, et al: Hematopoietic stem cell transplantation for severe and refractory lupus: analysis after five years and fifteen patients. Arthritis Rheum 46: 2917–2923, 2002. Jayne D, Tyndall A: Autologous stem cell transplantation for systemic lupus erythematosus. Lupus 13: 359–365, 2004. Tyndall A, Matucci-Cerinic M: Haematopoietic stem cell transplantation for the treatment of systemic sclerosis and other autoimmune disorders. Expert Opin Biol Ther 3: 1041–1049, 2003. Snowden JA, Passweg J, Moore JJ, et al: Autologous hemopoietic stem cell transplantation in severe rheumatoid arthritis: a report from the EBMT and ABMTR. J Rheumatol 31: 482–488, 2004. Brodsky RA, Sensenbrenner LL, Jones RJ: Complete remission in severe aplastic anemia after high-dose cyclophosphamide without bone marrow transplantation. Blood 87: 491–494, 1996. Petri M, Jones RJ, Brodsky RA: High-dose cyclophosphamide without stem cell transplantation in systemic lupus erythematosus. Arthritis Rheum 48: 166–173, 2003. Kishimoto T, Hamazaki T, Yasui M, et al: Autologous hematopoietic stem cell transplantation for 3 patients with severe juvenile rheumatoid arthritis. Int J Hematol 78: 453–456, 2003. Laxer RM, Harrison C: Bioethical issues in autologous stem cell transplantation in children and adults with arthritis. J Rheumatol 28: 2147–2150, 2001.
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS Edward H. Giannini
~'rf
EVIDENCE-BASED MEDICINE AND CLINICAL INVESTIGATION Today, more than ever, the clinician is encouraged to practice "evidence-based medicine." That is, the practitioner must be able to access, summarize, and apply information from the literature to day-to-day clinical problems. 1,2 But how strong should the evidence be before a clinician modifies his or her practice because of new data in a clinical research report? The same question can be asked of any clinical or biomedical research report, whether or not it applies to clinical practice. The strength of evidence depends on many factors, including the rigor of the study design, the selection of patients and appropriate controls, and the meticulousness and appropriateness with which the data were gathered, analyzed, interpreted, and reported. In addition, the similarity between the patients described in a study and the patients seen by a particular clinician is of utmost importance in determining the relevance of a study. The reader must be able to judge the quality of the work in order to determine its overall validity and the acceptability of the conclusions. The mere fact that the work has been published (even in a reputable journal) may not be enough of a yardstick whereby one decides that the work is of superior quality. Since the last edition of this text, the Cochrane Collaboration has done much to add to the practice of evidence-based medicine, including pediatrics. 3- 7 Founded in 1993 and named after the British epidemiologist, Archie Cochrane, its chief goal is to provide systematic reviews of health care interventions and thus to promote the search for evidence in the form of clinical trials and other types of studies of interventions. At present, the Cochrane Database of Systematic Reviews, its main product, contains approximately 1700 complete reviews, with another 1300 underway. Outstanding "Users' Guides to the Medical Literature" have now been published, and the reader is encouraged to supplement the discussion in this chapter with the appropriate guide for a specific topic.H--4O
142
Any student of clinical research must now be aware of the sizable obstacles currently facing the clinical research enterprise. 41 ,42 There now appears to be a general consensus among medical scientists, public health policy makers, and the United States Congress that the remarkable advances in biotechnology that have been realized during the past 20 years have not translated, at an acceptable rate, into improved health status for the nation and the world in general. The situation is thought to be worsening as acquisition of basic science knowledge continues to accelerate. Clinical research is viewed as a continuum-beginning from basic biomedical research, progressing to clinical science and knowledge, eventuating in improved health of the public. Two "major transitional blocks" are identified that impede efforts to apply science to better human health in an expeditious fashion. The first "translational block" occurs between basic biomedical research and clinical science and knowledge, the second between clinical science and knowledge and improved health. Contributing factors to the first block include lack of willing study participants, regulatory burden, fragmented infrastmcture, incompatible databases, and a lack of qualified investigators. Contributing to the second block are career disincentives, practice limitations, high research costs, and lack of funding. These obstacles should remain foremost in the reader's mind, with the realization that the design and analysis of studies must necessarily be grounded in what is reasonable from a logistical, practical, ethical, and economic point of view. This chapter proVides readers with enough clinical, epidemiological, and biostatistical skills to critically assess the literature and to independently determine the "strength of the evidence" (Le., to become more proficient at the practice of evidence-based medicine). It also promotes basic skills that facilitate the design, undertaking, and reporting of clinical research. Although this chapter emphasizes clinical research, many of the concepts discussed here are easily translated to the realm of basic science. Whetllcr working in the laboratory or in the clinic, an investigator must understand the basic concepts of, for example, frequency distributions and statistical inferences.
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
DEFINmON OF CLINICAL RESEARCH Academicians have argued for years about exactly what is incorporated in the concept of clinical research. The Nathan Report (http://www. nih .gov/news/crp/97report/ index.htm) defines clinical research as epidemiologic and behavioral studies, outcomes research and health services research, and patient-oriented research. Research conducted with human subjects (or on material of human origin, such as tissues, specimens, or cognitive phenomena), for which an investigator (or colleague) directly interacts with human subjects, also qualifies as clinical research. This area of research includes mechanisms of human disease, therapeutic interventions, clinical trials, and development of new technologies. Excluded from this definition are in vitro studies that use human tissues but do not deal directly with patients. This definition, however, does not exclude laborato!), or translational research ("bench-to-bedside"), provided that the identity of the patients from whom the cells or tissues under study are derived is known.
GENERAL TERMINOLOGY AND BASIC CONcEPTS ASSOCIATED WITH CLINICAL STUDIES To be conversant in clinical study designs and methodology, one needs a working knowledge of the vocabula!)' and basic concepts relevant to the various approaches. Readers must attempt to generalize the usage of common terms from one type of study to the next. For example, "exposure" may mean that the subject was exposed to cigarette smoke in a retrospective casecomparison study, exposed to a drug in a clinical trial, or exposed to human leukocyte antigen (HLA)-B27 in a genetic study. All clinical investigations may be divided broadly into observational or experimental studies. In observational studies, there is no artificial manipulation of any factor that is to be assessed in the study, nor is there active manipulation of the patient. In observational studies, the subjects have received the "etiologic" agent by mechanisms other than active assignment or randomization. Examples of such mechanisms are self-imposed exposure (personal habits or nutritional patterns), prescription by a physician, atmospheric pollutants, and occupational toxins. Observational studies may be either retrospective or prospective. Retrospective implies that the data already exist and are retrieved using a systematic approach, but missing data are not retrievable. In prospective observational studies, a cohort is observed prospectively through time, and data are gathered on an ongoing basis. In this 'case, missing data may possibly be retrieved for purposes of the study. Experimental studies are those in which the experimenter artificially manipulates some study factor-subjects, therapeutic regimen, or some other parameter. In experimental studies, the subjects are observed prospectively, some artificial manipulation is conducted, and the results of this manipulation are then observed. Meta-analysis is an approach whereby the quantitative evidence from two or more studies bearing on the same
143
question is statistically combined. 43 Usually, the summa!)' statistics rather than the raw data from the various studies are combined. Meta-analysis may be considered a form of retrospective experimental study in that the data already exist and were gathered during prospective experimental studies. The Cochrane Collaboration relies heavily on meta-analytic techniques.
Hypothesis-Generating Versus Hypothesis-Testing Studies The design of a clinical investigation depends on whether the study intends to generate hypotheses to be tested in future studies or to test specific hypotheses for which the investigator has some existing evidence to support the belief that they are true or not true. Hypothesis-generating studies are considered exploratory; studies that are designed as tests of hypotheses may be called pivotal or confirmatory studies. A single study may have both confirmato!)' and explorato!)' aspects. Each type of study has distinct advantages and disadvantages. The design chosen is always deeply influenced by reality-what is economically, logistically, ethically, and scientifically possible. A common exercise used by methodologists is to design the best theoretical experiment to answer the question being posed, without regard to time, money, ethics, patient availability, or anything else that could cause a lessening in the quality of the study. (A related approach is known as the "infinite data set. "44) Then, realizing that there is no such thing as the perfect clinical study, the designer eliminates the most unrealistic "requirement." (For example, it is not likely that one can enroll 300 patients with scleroderma who will agree to the pOSSibility of being randomized to placebo for a year.) The study is compromised further and further by reality until one arrives at what can be done in consideration of all the issues. If the result is unacceptable scientifically, perhaps the question cannot (and should not) be answered. The decision to pursue or not to pursue the "compromised" study, based in reality, is one of the most difficult in the entire research process. It is worth remembering that one's rights as a clinical investigator are the exact opposite of one's constitutional rights as a citizen: the investigator (or the study) may be considered guilty (of anything and eveI)'thing) until proven innocent.
Main ObJectives, Process ObJectives, Hypotheses, and Long-Term Goals The first challenge, after identifying a clinical question to be addressed, is to clearly establish the main (or primary) objectives and the secondary objectives. Objectives are statements as to what the investigators plan to learn or accomplish by conducting the study. Process objectives follow the main objectives; they are the procedures that must be completed in order to meet the main objectives. Hypotheses are then developed. Hypotheses are formal statements that declare what the investigator will test and then either reject or fail to reject. As discussed earlier, explorato!)' (hypothesis-generating) studies may not state
144
C HAP T E R
6 0
E S I G N, MEA SUR E MEN T, AND
hypotheses a priori. Such studies frequently allow the data to drive the hypotheses to be tested in the dataexploration phase. Finally, longer-term goals are stated. Often, they are to be met only after the main objectives of the current projects are complete. They frequently give a hint as to what future direction the research may take. Clinical research proposals often misstate hypotheses or mix main objectives (specific aims) with process objectives or longer-term goals. The following example may assist the reader in formulating each of these elements of a research proposal. Suppose the question is, "Does the selective cyclooxygenase-2 inhibitor, celecoxib, produce fewer gastrointestinal (GI) adverse effects in children with juvenile rheumatoid arthritis (TRA) than does naproxen when each drug is given in anti-inflammatory doses for a period of up to 3 months?" The objectives are typically written nonjudgmentally and are more informal in their language than the hypotheses. A primary objective for this project could be, "To determine the incidence of GI adverse drug events among patients with ]RA treated for up to 3 months with celecoxib compared with naproxen." Equally acceptable (but less distinguished from the statement of hypothesis) is, "To test the hypothesis that celecoxib produces fewer GI adverse drug effects than does naproxen in children with ]RA." A secondary objective could be, "To determine the incidence of GI events requiring pharmacologic intervention in the two groups and compare the differences." The main objective would not be, for example, "To enroll 250 patients with ]RA in a clinical trial in which half are randomly assigned to celecoxib and half to naproxen." That is a process objective-one of the necessary steps that will be carried out to meet the main objective. Even large studies typically have only one or two main objectives and accompanying hypotheses. The usual null hypothesis (abbreviated Ho) is that the treatments are not different with regard to the primary outcome. Ho can be stated as, "There is no difference in the incidence of GI adverse drug effects among patients treated with naproxen compared with those treated with celecoxib." The alternative hypothesis (abbreviated H) is, "There is a difference in the incidence of GI adverse drug effects,." As stated, the Ha does not specify whether there are more or fewer GI effects with celecoxib. This is a two-tailed hypothesis. A one-tailed hypothesis could be stated as follows: "Patients with ]RA treated with celecoxib will have a lower incidence of GI adverse drug effects than those treated with naproxen." However, this hypothesis sounds as if the investigator is not interested in testing to see whether celecoxib produces more GI effects, just less. For this reason, the usual approach is to use two-tailed hypotheses, at least in early studies when the direction of the difference (if any) is not known. The choice of a one-tailed as opposed to a two-tailed hypothesis influences the statistical interpretation of the data, as discussed later. A secondary null hypothesis in this example might be, "There is no difference in the incidence of GI adverse drug events that require pharmacologic intervention among patients with ]RA treated with celecoxib compared with those treated with naproxen."
A N A LY SIS
0 FeLl N I CAL
I N YES T I GAT ION S
Finally, the longer-term goal for this example is not, "To complete the study in a 5-year time frame," but rather, "To provide safer and more effective anti-inflammatory medications to children with ]RA."
EPIDEMIOLOGIC AND BEHAVIORAL STUDIES Clinical epidemiology is a medical science that studies the distribution and determinants of disease frequency in human populations in order to understand why some people contract a disease and others do not. Epidemiology, combined with biostatistics, makes up the basic tools of the clinical investigator. Epidemiologic methods can be used to answer questions in the following categories.
Descriptive Epidemiology Studies in descriptive epidemiology typically concern themselves with patterns of disease occurrence with respect to person, place, or time. Descriptive epidemiologic studies serve as hypothesis-generating studies for studies of causation, much the same way as small exploratory clinical trials serve as preliminary studies for therapeutic confirmatory trials. The person variable is concerned with who experiences the disease. A basic tenet is that the disease does not occur at random but is more likely to develop in some people than in others. Personal factors of potential importance include age, sex, race, ethnicity, socioeconomic status, existing morbidity, health habits, and genetics. The place factor is concerned with where the disease develops. Variation in place of occurrence can be evaluated at the local, regional, or international level. The time variable is concerned with variation in the occurrence of disease in time and its seasonality or periodicity. A hypothetical example of a descriptive epidemiologic study is the investigation of a group of workers in a factory who have what is suspected of being environmentally acquired lupus. The epidemiologist would investigate the detailed characteristics of the workers to determine whether there are patterns among those who do and do not have lupus. Do all types of workers (management through hourly manufacturing employees) demonstrate the same rate of disease development? Are persons living close to the factory or its effluent also affected? Systematic investigation of the patterns of disease allows a more precise hypothesis of causation, particularly if some exposure or dose level is found to be more strongly associated with the illness.
Frequency of Disease Occurrence The frequency of disease occurrence is an important aspect of understanding a disease process. It can be measured in a number of ways. Prevalence is the number of existing cases in a defined population. Mathematically, prevalence is equal to the number of existing cases divided by the number of persons in the population. It is expressed in different ways: as a proportion (0 to 1), as a percentage (0 to 100), or by actual
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
numbers using a convenient denominator (e.g., cases per 1000 children). Point prevalence is the number of new and old cases in a defined population at a given "instant" in time. For example, hospital epidemiologists are frequently asked to estimate the point prevalence of nosocomial infections among current inpatients. The epidemiologist then counts all the confirmed and suspected cases of hospitalacquired infections within the hospital on a given day and divides this total by the census for that day to obtain the point prevalence. Period prevalence is the number of new and old cases that exist in a defined population during a given time period (e.g., 1 year). Incidence is the rate at which newly diagnosed cases develop over time in a population. Mathematically, incidence is equal to the number of new cases (numerator) divided by the number of persons at risk in the population multiplied by the time (duration) of observation (denominator). This rate is expressed in units of cases/person-time. Incidence is related to the concept of risk, defined as the proportion of unaffected individuals who will, on average, contract the disease over a specific period of time. Risk is equal to the number of new cases divided by the number of persons at risk. Risk has no units and can have values between 0 (no new occurrences) and 1(the entire population becomes affected during the risk period). Epidemiologic theory states that incidence is best estimated from prospective studies; prevalence may be calculated by prospective or retrospective approaches.
Etiology of Disease In his presidential address to the Royal Society of Medicine in January of 1965, Sir Austin Bradford Hill gave his now famous oration entitled, "The Environment and Disease: Association or Causation. "45 In it, he described what have become known as Koch's postulates Jor epidemiologists. These postulates describe what evidence must be present to establish a factor as being causally linked to a disease and include the following: • Strength of the association: How strong is the association between the factor and the outcome? For example, how significant is the probability (P) value of the association between dietary intake of calcium and bone mineral density among children with JRA? • Consistency of the association: Does the association between factor and disease persist from one study to the next, even if variations in study design and samples of patients vary substantially? • Specificity ojthe association: Is the association limited, for example, to specific alleles and types of disease, with little association between the alleles and other diseases? (As the study of causation, including genetic risk, has advanced, the issue of specificity is considered less important than it once was.) • Temporal correctness: Did the exposure to the factor occur before the disease? • Biologic gradient: Is there a dose-response relationship between the factor and the disease? For example,
145
does increasing the dose or time of exposure to cyclophosphamide result in a consequent increase in frequency of malignancy? • Biologic plausibility: Does the association make sense with what is currently understood about the disease and its pathogenesis? • Experiment: Does the association hold up under experimental conditions? For example, if one reduces the dose or time of exposure to cyclophosphamide, is there a corresponding decrease in the frequency of malignancy? No single study can prove indisputably that any potential etiologic factor causes a disease, complication, or adverse event. It is the accumulating body of knowledge concerning factor and disease, or (to generalize the terms) treatment and outcome, that finally allows the medical community to state that evidence is sufficient to prove a causal link between the two. An important concept in the study of disease etiology is that of relative risk. The relative risk (risk ratio) can be defined as the risk of developing a disease among the members of one group (e.g., boys) compared with another group (e.g., girls). Relative risk can range from zero to infinity and has no units. A relative risk of 1 means that there is no difference in risk between the groups. A relative risk greater than 1 means increased risk, and a relative risk less than 1 indicates decreased risk (protection). In pediatric rheumatology, the concept of risk is used frequently to specify increased or decreased risk of disease associated with certain genetic markers. Table 6-1 presents terms that are relevant to risk and shows how each may be calculated using a 2 x 2 contingency table. Disease state (present or absent) is considered the dependent variable (DV) and is usually placed in the columns (x-axis). The risk factor (positive or negative) is considered the independent variable and is usually placed in the rows (y-axis). Notice that the relative risk is calculated differently from the odds ratio, although in medicine the two are frequently used synonymously. More correctly, however, relative risk is calculated from incidence studies, whereas odds ratios are calculated from retrospective studies.
Epidemiologic Study Designs Aimed at the Establishment of Associations and Cause-Effect Relationships The Case-Controlled Retrospective Study One of the most common types of study designs used to establish an association or cause and effect is the observational case-controlled, retrospective study. (The term case-comparison is now frequently used because the studies are not controlled in the usual sense.) In this situation, patients who have the disease are compared with those who do not, and data documenting prior exposure to some agent are ascertained retrospectively. The most frequent statistic to come from this type of study is the odds ratio (see Table 6-1). The odds ratio is a fairly good estimate of the relative risk, provided the disease is rare.
146
C HAP T E R
11.
6
TABLE 6-1
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
Terms Assodated With Risk Factors dnd Disease
Risk Fador
Disease Present a c
Positive Negative
Disease Absent b d
The 2 x 2 table may be used to calculate associations between the risk factor and the disease:
Term
CalculaUon
Meaning
Incidence Absolute risk Attributable risk
(a + c) / (a + b + c + dl
-
Relative risk
(a / [a + bJ) -;- (c / [c + dJ)
Odds ratio
(a x d) / (b x c)
Case exposure rate
a / (a + c)
Control exposure rate
b / (b + d)
Number of new cases among those at risk Synonymous with incidence Incidence among those with the risk factor minus incidence among those without the risk factor (sometimes expressed as a percentage of the incidence rate among those with the risk factor) Incidence among those exposed divided hy incidence among those not exposed An approximation to the relative risk used in retrospective studies Among those with the disease, the proportion who had the risk factor Among those without the disease, the proportion who had the risk factor
[a / (a + b)] - [c / (c + dl]
Patients included in the study are often identified from a search of medical records or a disease registry. However, there are no standard guidelines for selection of control subjects. Control subjects may be hospitalized patients with conditions other than the disease of interest, residential neighbors, a probability sample of the general population, or a combination of these. The casecontrol classroom peers design is an appealing methodology for the selection of appropriate controls now finding uses in pediatric rheumatology.46 Originally (and still) used in studies of pediatric cancer patients, the technique identifies one classroom peer of the same sex and closest in age to the subject, and then compares the two on whatever variables are relevant to the specific study.47 Advantages of case-controlled studies include efficiency, low cost, quick results, and low risk to study subjects. The disadvantages of the case-controlled study are several, however. The temporal relationship of exposure and disease may be obscured. Historical information may be incomplete or inaccurate; a detailed study of mechanisms of disease is often not possible; and if the study is not well done, results may be biased.
The Prospective Cohort Study The observational approach that most closely resembles an experiment is the prospective cohort study. In this case, a population is defined from which the sample is drawn. Exposure to some factor is then established, and subjects are categorized as having been either "exposed" or "not exposed" to a factor thought to contribute risk of some outcome. Each of the two cohorts is monitored prospectively to observe whether the outcome develops. Relative risk is the statistic most commonly used in describing this study. The identification of exposed persons presents several problems. The first is to correctly
identify the exposed persons and measure the degree of exposure. This may be done by selecting subjects with some type of unusual occupational or environmental exposure. Another method is selecting those who are available and suitable for the needed investigation or selecting those who offer some special resource that facilitates the study, such as members of a health plan, or a combination of all these factors. The advantages of a prospective observational cohort study are that a clear temporal relationship between exposure and disease is established, and the study may yield information about the length of induction (incubation) of the disease. Relative risk can be estimated directly. The design facilitates the study of rare exposures and allows calculation of rates of disease occurrence. The disadvantages of cohort studies include the potential for loss to follow-up or alteration of behavior because of the long follow-up time that may be necessary. Cohort studies are not particularly good for rare diseases when the outcome is onset of the disease. Detailed studies of the mechanisms of the disease typically are not possible in cohort studies. A rheumatologic example of a cohort study designed to detect disease causation is the study by Inman and associates,48 who prospectively observed a cohort of persons "exposed" to Salmonella typhimurium infection to determine whether reactive arthritis developed. Prospective cohorts also find use in pediatric rheumatology when the aim is identify risk factors for development of certain complications or outcomes in a group of children who, typically, have the same disease but vary in "predictor" or "risk" variables. 49--'52
Diagnosis of Disease and Classification Criteria The diagnosis of disease, as it applies to epidemiology, refers to the performance of screening and diagnostic
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
147
tests used in populations rather than the process of differential diagnosis of individual patients. Classification criteria typically employ a set of "core variables" fashioned into an algorithm. Numerous classification criteria relevant to pediatric rheumatic disease have been developed and/or validated since the previous edition of this text. They include criteria for flare of JRA, criteria for improvement in juvenile idiopathic myopathies and lupus, and preliminary criteria for clinical remission in JRA.S3-'i6
information about several parameters: it demonstrates the tradeoff between sensitivity and specificity for different criteria or cut points; the nearer the curve follows the left border and the top border of the ROC space, the more accurate the test; the closer the curve comes to the 45degree diagonal of the ROC space, the less accurate the test is; and the area under the curve is a measure of the accuracy of the test or criteria.
Validity of a Diagnostic or Screening Test or Criteria
Prognosis refers to the possible outcomes of a disease and the frequency with which they can be expected to occur. Prognostic factors need not cause the outcome but must merely be associated with an outcome strongly enough to predict it. Prognosis is somewhat narrower in focus and more short-term in aspect than those consequences of disease and treatment that are considered in the field of outcomes research (discussed later). For example, the six most frequently measured outcomes in outcomes research are known as the 6 Ds: death, disease, disability, discomfort, dissatisfaction, and dollars. Many studies of prognosis are cohort studies that prospectively monitor large numbers of patients to determine eventual clinical events and then look for certain prognostic factors for the events. At present and in the past, the estimation of prognosis has largely concentrated on clinical variables. Studies of prognosis in JRA, for example, have included the sex of the patient, the age at onset, and a variety of clinical and laboratory variables to estimate outcome. 49 .S7-63 Genetics, genomics, and microarrays to study RNA expression (and the accompanying new technologies of informatics and computational medicine) are being investigated more intensely to delineate their ability to predict outcome. 64-71 Pharmacogenetics has entered the scene as a way to determine the probability of response or adverse reaction to certain drugs. 72 It is likely
The validity of a diagnostic or screening test or set of criteria involves a variety of parameters, as shown in Table 6-2. The table typically is constructed with the presence or absence of disease as the column labels (Le., x-axis) and the test results as the row values (Le., DV shown on y-axis). Those patients in row 1, column 1 are called true positives; those in row 1, column 2 are false positives; those in row 2, column 1 are false negatives; and those in row 2, column 2 are true negatives. Sensitivity, specificity, positive and negative predictive values, false-positive and false-negative rates, and reliability are terms used to describe the validity of a screening test. A widely used tool that allows one to compare visually the performance of a set of different criteria or different cut points for a diagnostic or screening test is known as the receiver operating characteristic (ROC) curve (Fig. 6-1). First used in industrial engineering, an ROC curve is a plot of the true-positive rate against the false-positive rate-that is, sensitivity on the y-axis and 1 minus specificity on the x-axis. An excellent example of the use of this technique is demonstrated in Brunner's paper describing the relative performances of various criteria for disease flare in JRA.S3 An ROC curve provides
,~.
fABLE 6-2
Prognosis of Disease
Estimating the Validity of a Diagnosti( Test
Test Resun
Disease Present
Disease Absent
Positive Negative
True positive (TP) False negative (FN)
False positive (FP) True negative (TN)
The 2 x 2 table may be used calculate measures of the test's validity:
Term
Calculation
Meaning
Sensitivity
TP / (TP + FN)
Specificity
TN / (TN + FP)
Positive predictive value
TP / (TP + FP)
False-positive rate
FP / (TP + FP)
Negative predictive value
TN / (TN + FN)
False-negative rate
FN / (TN + FN)
Reliability (also called reproducibility)
-
Proportion (or percentage) of persons with the disease who test positive Proportion (or percentage) of persons without the disease who test negative Proportion (or percentage) of persons who test positive who have the disease Proportion (or percentage) of persons who test positive who do not have the disease Proportion (or percentage) of persons who test negative who do not have the disease Proportion (or percentage) of persons who test negative who have the disease The ability of a test to yield the same result on retesting
148
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
1.0 - , - - - - - - - - - - - - - - - - - - - : / 1
.
0.9 +---;:D;-e.f.-:;1-'-..,.-------:::::-c:.---,-~:-,- - - 1 / /
• Worsening of 2 CRV without improvement of more than 1 of the remaining CRV by 30% or more • Worsening of 2 CRV
/
... Worsening of 3 CRV without improvement of more than 1 of the remaining CRV by 30% or more o Worsening of 3 CRV
* Worsening of 4 CRV without improvement of more than 1 CRV by 30% or more
o Worsening of 4 CRV 0.5 + - - I - - - , - ' ' - - - - - - - - - - - - - - - - j
~
Changes in CHAO, ESR and AJC only
0.4 +------.---...,--------r---r----,-------j 0.000 0.100 0.200 0.300 0.400 0.500 0.600 1 - Specificity (false positive rate)
• Figure 6-1 The receiver operating characteristic (ROO curve, a plot of the true-positive rate against false-positive rate (i.e., sensitivity on the y-axis and 1 minus specificity on the x-axis). ROes allow one to observe the tradeoff between sensitivity and specificity at various cut points of a diagnostic test. Values (or criteria for classification of patients) that fall on the upper left side of the curve are more useful (see text for further explanation). In the example shown, various criteria for describing flare in JRA are plotted. (Modified from Brunner HI, Lovell DJ, Rnck BK, Giannini EH: Preliminary definition of disease flare in juvenile rheumatoid arthritis. J Rheumatol29: 1058-1064,2002.)
that, in the future, determination of outcome will be based in large part on screening for various genetic markers that can predict whether individuals will respond to a particular therapy, whether they will experience an adverse drug effect, and whether their quality of life will be improved after a particular treatment.
Treatment of Disease For special emphasis, this chapter considers clinical trials separately from epidemiologic studies (see "Clinical Trials: Useful Guidelines").
Behavioral Studies The National Institutes of Health (NIH) considers behavioral studies as a category of clinical research. This class of studies is not considered here.
OUTCOMES AND HEALTH SERVICES RESEARCH The terms clinical effectiveness and health services research are closely related to outcomes research and are considered synonymous here. Outcomes research may be defined as the study of effectiveness, cost-effectiveness, and quality of health care. The interest in outcomes research has been generated by a more cost-conscious health care system. 73 For example, only 10% to 20% of medical procedures have been evaluated through ran-
domized controlled trials. Both established and new technologies can be ineffective. Certainly, the outcomes researcher must rely on much evidence-based medicine present in the literature as well as new studies. Outcomes researchers often use randomized controlled trial designs, prospective cohorts, retrospective cohorts, and case-control studies (described later). Chart reviews are frequently used, as are administrative data including local hospital discharge data, insurance claims, and national health survey information. In addition, meta-analysis, decision analysis, and cost-effectiveness analysis are typical study designs employed by the outcomes researcher. There are few reports of "true" outcomes research related to pediatric rheumatology. Excellent gUides are available for evaluating outcomes research. 15 .2(l-24
Decision Analysis Decision analysis may employ all of the techniques mentioned in the previous section and therefore is discussed in greater detail here. Decision analysis is an analytic formulation of a decision in which uncertainty exists. It incorporates probability of events as well as values for the consequences of action. It is decision oriented (not truth oriented), it is prescriptive (not descriptive), and it enhances, but does not replace, clinical judgment. The premises of decision analysis are that decisions must be made, the consequences of actions are uncertain, the consequences of action include both favorable and unfavorable effects, and health care resources are constrained. Decision analysis should enhance the ability to
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
make rational decisions that lead to optimal therapeutic outcomes. Often, the principles of decision-making are obscured by substitution of established protocols. "Decision trees" should not be confused with flow charts presented in clinical practice guidelines for the management of particular diseases such as JRA.74 Decision trees tend to be more complicated, more branched, and more speculative than flow charts. Elements of a decision analysis are as follows. Choices are outlined, and the method to assess the overall value of the outcome of each choice is determined. The underlying assumptions are that there is uncertainty about the future and that various possible outcomes are valued differently. A decision tree is then constructed. Decision analysis itself includes outlining the problem, laying out the options and possible outcomes in explicit detail, assessing the probabilities and values of each outcome, and selecting the "best choice." In summary, the elements of decision analysis are choices, outcomes, and utilities. The areas of outcomes research and decision analysis are in their infancy in pediatric rheumatology. Each should offer a fruitful field for investigators for many years to come, because many different "standards of care" exist for most diagnoses that are included under the topic of pediatric rheumatology. Two excellent guides for judging the quality of decision analysis studies are those by Richardson and Detsky.1B.19
PATIENT-ORIENTED RESEARCH The NIH definition of clinical research groups four categories of investigation under the major heading of patient-oriented research: mechanisms of human disease, therapeutic interventions, clinical trials, and development of new technologies. This chapter emphasizes clinical trial classification and methods. However, many of the concepts and much of the terminology presented herein can be generalized to the conduct of clinical studies in any of these four areas of clinical research.
Olnlcal Trials: Useful Guidelines For simplicity, the generic term drng is used in the follOWing discussions. It should be considered synonymous with any medicinal product, vaccine, or biologic agent. The principles discussed can also apply to interventional procedures such as surgery and radiotherapy. Clinical epidemiologists are frequently concerned with evaluating the effectiveness and safety of new therapies. Numerous useful guidance documents (as distinguished from guidelines) are now in the literature and should be consulted for a more detailed explanation of the principles outlined here. Of particular relevance to the pediatric rheumatologist is the U. S. Food and Drug Administration (FDA) document entitled, "Guidance for Industry: Clinical Development Programs for Drugs, Devices and Biological Products for the Treatment of Rheumatoid Arthritis" (available at http://wwwfda.gov/ cber/gdlnslrheumcln.pdj). This document summarizes the pOSition of the FDA as to what clinical development programs should consist of, and it provides a framework
149
for conducting studies used to obtain regulatory agency approval of therapies for rheumatoid arthritis (RA) or JRA. An extremely useful set of guidance documents also available on the FDA's Web site is the "International Conference on Harmonisation (ICH) of Technical Requirements for Registration of Pharmaceuticals for Human Use." These documents contain valuable guidance for industry and the clinical investigator as well as definitions of terms related to overall clinical development programs, considerations for individual trials, statistical methods and reporting, and a variety of other topics. The ICH guidance documents seek to establish harmonization of clinical trials in various regions throughout the world, and their acceptance is very broad. It is strongly recommended that the designer or reader of clinical trials be familiar with their content. A list of available ICH guidelines may be found at http://wwwfda.gov/ cber/guidelines.htm or http://www.ich.org. Before general and specific considerations for individual trials can be discussed, an understanding of the various systems of classification of clinical trials is essential.
Classification of Olnlcal Trials by Initiator Clinical trials may be initiated either by industry or by an individual investigator. Those that are part of a clinical development program and conducted under a sponsor's (pharmaceutical company's) investigational new drng (IND) submission are usually initiated by industry. Trials undertaken under a sponsor's IND are used frequently by the sponsor in its submission to obtain approval for a new drug, known in the United States and elsewhere as a new drng application (NDA). If the NDA is approved by the regulatory agency, the drug may be marketed and labeled for the specific indication (Le., disease or conditions) stated in the NDA. Investigator-initiated protocols are typically, but not always, conducted after the drug has been approved for market. The main objective of investigator-initiated protocols may be new dosage regimens or use in diseases other than that for which the drug has obtained an indication. Many such trials are exploratory rather than confirmatory. Funding for investigator-initiated protocols in pediatric rheumatology has come from the NIH, the FDA, manufacturers, foundations, and the European Union.
Classification of Clinical Trials by Phase Clinical drug development programs are often described as consisting of four temporal phases, numbered I through IV. This system of classification, despite its shortcomings, is perhaps the one most commonly used today by the pharmaceutical industry and by regulatory agencies75 (Fig. 6-2).
Phase I Phase I studies are human pharmacology trials whose overall aim is to establish preliminary safety and tolerability of the dose range expected to be needed for later clinical studies and to determine adverse drug effects that can be expected. Studies in this phase include both
150
C HAP T E R 6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
Therapeutic Use
•• • • c • ••
Therapeutic Confirmatory
n
Therapeutic Exploratory
•••
Human Pharmacology
0
> • ••
0
u u
0
0
u
III
PHASES OF DEVELOPMENT
0
C0 IV
Time • Most typical kind of study in this phase
o Possible information generated • Figure 6-2 Study types and the phases of development in which they are performed. Filled circles indicate the most typical kind of study in the phase shown; open circles indicate possible information generated.This graph shows that study types are not synonymous with phases of development. (From E8: General Considerations for Clinical Trials Federal Register. Vol. 62, No. 242. Wednesday, December 17, 1997/Notices p.66116.)
single- and multiple-dose administration schedules and have one or more of the following aspects,
Pharmacokinetics Pharmacokinetics (PK) studies typically determine the drug'S absorption, distribution, metabolism, and excretion pathways. PK may progress throughout a clinical development program and be assessed in separate studies or as part of larger trials to determine efficacy and safety. These studies are necessary to assess the clearance of the drug and to anticipate possible accumulation of the drug or its metabolites and the potential for drug-drug interactions, Assessing PK in subpopulations, such as those with impaired renal function or hepatic failure, is another important aspect of this phase of studies. PK data are usually expressed using the following terms 76 :
• Area under the time-concentration curve (AUC, or AUC O_24 if done over a 24-hour period) is a measure of the total amount of drug absorbed; it is frequently estimated after the drug has reached steady-state levels. • Peak concentration (Crna) is the maximum concentration reached at a particular dosage. • Time to peak concentration (Trnax) is used together with Crnax to measure the rate of absorption. • Cumulative percentage of drug recovered (Ac%) usually relates to urine data and is the cumulative amount of drug recovered over a specific time period (e.g., 24 hours) divided by the initial dose. • Elimination (or terminal) half-life (til) is a measure of how long it takes to clear a drug from the system; it can be estimated by dividing 0.693 by the absolute value of the slope of the terminal linear phase of the concentration profile plotted on a semilog scale. Special considerations for PK studies in children are available on the FDA Web site at http://wwwfda.gov/ cber/gdlns/pedphrm.pdj
Estimation of Initial Safety and Tolerability Drug safety refers to the frequency of adverse drug effects (Le., physical or laboratory toxicity that could possibly be
related to the drug) that are treatment emergent-that is, they emerge during treatment and were not present before treatment, or they become worse during treatment compared with the pretreatment state, An adverse drug effect is distinguished from an adverse event (or experience), which refers to any untoward experience that occurs while a patient is receiving the medication, whether or not it is attributable to the drug. The seriousness of an adverse event dictates how qUickly it must be reported to regulatory agencies and to others who may have ongoing experimental protocols. A serious adverse event (SAE) is defined as one that results in death, is lifethreatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability or incapacity, or is a congenital anomaly or birth defect. Investigators conducting pharmaceutical industry-sponsored studies should be aware that companies may have their own, more strict definition of SAEs. The term severity is distinguished from serious in that severity refers to the intensity of a specific event, whereas serious refers to the outcome or consequences of the event. Drug tolerability refers to how well subjects are able to tolerate overt adverse drug effects.
Assessment of Pharmacodynamics Pharmacodynamics (PD) studies assess the uptake, movement, binding, and interactions of pharmacologically active molecules at their specific sites of action. These studies also typically observe the relation of drug blood levels to clinical response or to adverse drug events, They may provide early estimates of drug activity and potential efficacy, and they help to establish the dosage and dose regimen used in later phases of drug development.
Early Assessment of Drug Activity Although early assessment of drug activity is not part of human pharmacology, phase I studies may also determine preliminary efficacy in terms of secondary variables that may be confirmed in later studies.
Phase II Phase II studies are the earliest attempt to establish efficacy in the intended patient population. Many are called therapeutic exploratory studies and will form the basis for later trials. The hypotheses may be less well defined than in later studies, and they may be data driven. The designs are flexible so that changes can be made as the data accumulate. These studies may use a variety of different types of study design, including comparisons with baseline status or concurrent controls. In these studies, the eligibility criteria are typically very narrow, leading to a homogeneous population that is carefully monitored for safety, Further studies may establish the drug's safety and efficacy in a broader population once it is determined that a drug does indeed have activity. These studies may also aim to determine more exactly the doses and regimens for later studies. Phase II studies may use dose-escalation designs to estimate dose response, which may be confirmed in later
C HAP T E R
6 DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
studies. In phase II, doses of the drug are typically, but not always, less than the highest doses used in phase I. Another important goal of phase II studies is to determine potential study end points, therapeutic regimens (including the use of concurrent medications), and subsets of the disease population (mild versus severe).
Phase III The primary objective of phase III studies is to confirm a therapeutic benefit. Therefore, the most typical kind of study is the therapeutic confirmatory study, which provides firm evidence of an agent's efficacy and safety. This type of trial always has a predefined hypothesis that is tested. These studies also estimate (with substantial precision) the size of the treatment effect attributable to the drug. Also incorporated in phase III development are further exploration of the dose-response relationship, study of the drug in a wider population and in different stages of the disease, and the effects of adding other drugs to the investigatory agent. These studies continue to add information to the accumulating safety database.
Phase IV Phase IV studies, sometimes called postmarketing surveillance studies, begin after the drug reaches the market. These studies extend the prior demonstration of the drug's safety, efficacy, and dose. The most frequent phase IV study is one of therapeutic use, which goes beyond the prior demonstration of the drug'S safety and efficacy. These studies demonstrate how the drug performs when used in the everyday setting of the clinic, by patients who may have comorbid conditions, and by patients who may be taking a host of concurrent medications.
Investigations in Special Populations Investigations in children typically take place after considerable data have been gathered in an adult population
!:
III
TABLl 6-3
with a similar disease. If clinical development includes children, it is usually appropriate to start with older children before extending the studies to younger children. The exception to the "adults first" rule is when a medication is developed to treat a condition that occurs only in childhood. The Pediatric Research Equity Act of 2003 (available at http://wwwfda.gov/opacom/lawslprea.html), referred to formerly as the Pediatric Rule, requires manufacturers to assess the safety and effectiveness of a drug or biologic product in children if the disease for which the drug was developed in adults also occurs in the pediatric population.
Classification of Olnlcal Trials by Objective The ICH believes that the classification system based on phases of development is inadequate for classifying clinical trials because one type of trial may occur in several different phases, as was shown in Figure 6-2. Table 6-3 presents an alternative classification system based on the objective of the study, using terminology described earlier.
CONSIDERATIONS FOR INDIVIDUAL CLINICAL TRIALS For special issues relevant to trials in children, the reader is referred to the document entitled, "Guidance for Industry: Ell Clinical Investigation of Medicinal Products in the Pediatric Population," which is available at http://wwwfda.govlcber/gdlnslichclinped.pdf
Objectives Clearly stated primary and secondary objectives and hypotheses form the backbone around which every clinical trial is developed. These should all be clearly stated in the protocol and study report. Table 6-3 provides a framework for developing appropriate objectives for the various types of study planned. In general, trials tend to
An Approach to Classifying Clinical Studies According to Objective
Type of Study
Objective of Study
Examples
Human pharmacology
Assess tolerance Define/describe PK and PO Explore drug metabolism and drug interactions Estimate activity Explore use for the targeted indication Estimate dosage for subsequent studies Provide basis for confirmatory study design, end points, methods Demonstrate/confirm efficacy Establish safety profile Provide an adequate basis for assessing the risk/benefit relationship to support licensing Establish dose-response relationship Refine understanding of risk/benefit relationship in general or special population Identify less common adverse reactions Refine dosing recommendation
Dose tolerance studies Single- and multiple-dose studies of PK and/or PD Drug-interaction studies
Therapeutic exploratory
Therapeutic confirmatory
Therapeutic use
PO, pharmacodynamics; PK. pharmacokinetics.
151
Earliest trials of relatively short duration in welldefined, narrow patient populations, using surrogate or pharmacology end points or clinical measures Dose-response exploration studies Randomized, parallel dose-response studies Clinical safety studies Studies of mortaliry/morbidity outcomes Large simple trials Comparative studies Comparative effectiveness studies Studies of mortality/morbidity outcomes Studies of additional end points Large simple trials
152
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
have one to three main objectives and numerous process objectives. One should be wary of trials with more than a few objectives, although exploratory trials may have objectives that are less clearly delineated than those of later trials.
Population The specific population to be studied is delineated by the inclusion/exclusion criteria. The development of these criteria is one of the more formidable tasks in the entire process. Developers of trials must attempt to reach a compromise between limiting the heterogeneity of the sample and not making the criteria so strict that recruitment of eligible subjects becomes untenable or threatens to restrict the generalizability of results. The heterogeneity of the patient population that will be allowed to enroll in the trial is influenced by the phase of development. Early, exploratory studies are often concerned with whether a drug has any effect whatsoever. In these trials, one may use a very narrow subgroup of the total patient population for which the agent may eventually be labeled. Later-phase, confirmatory trials typically relax the eligibility criteria to allow for a broader, more heterogeneous sample of the target population. Still, if the criteria for enrollment are too broad, interpretation of treatment effects becomes difficult.
Design The study's phase, objectives, ethics, and feasibility influence the specific design of a trial. New designs have appeared in recent years that reduce the time during which children receive placebo or a known inferior medication. 77 ,78 More than one type of design may be used to answer the same question. If the same results and conclusions are reached regardless of the design and analysis used, the results are said to demonstrate robustness. Although a study may be designed as being "pivotal," it is rare that any single trial establishes incontrovertible evidence of an agent's clinical worth.
Comparative and Noncomparatlve Studies Trials may be classified as either comparative or noncomparative studies. A comparative study implies that some type of comparison is made between the drug under investigation at a particular dosage level and either a placebo, another dosage level of the investigational drug, or an active comparator (an existing drug known to be effective for the specific condition). Noncomparative trials involve no such comparisons with the investigational agent. Studies that compare the agent with placebo or an active comparator are called controlled studies. For a discussion and guidance on the proper selection of a control group, the reader is referred to http://wwwfda.govlcber/gdlnslclincontr0501.htm. Studies that involve dose escalation or that compare the PK and PD for differing dosage levels of the same drug are not considered controlled in the usual sense.
Open Studies The early human pharmacology studies in the first phase of development and the postmarketing surveillance (phase IV) studies are usually open label, meaning that everyone involved with the study, including the patient and the physician, knows what the patient is receiving. Studies that occur in phases II and III may have an openlabel extension phase, during which patients who took part in the comparative phase receive the investigational drug openly for an extended period. The chief purpose is to gather longer-term safety and efficacy data. Investigator-initiated protocols may also be open if the intent is simply to gather additional information about an agent in another disease or at a dosage level other than that indicated in the label. As one would expect, the possibility of bias in interpretation of safety and efficacy information with open studies is much greater than with blinded studies. Open studies may be randomized to ensure that subject populations are similar in test and control groups.
Blinded Comparative Studies Beginning either in late phase I or early phase II, blinded, controlled (comparative) studies are performed. Blinding refers to the masking of those involved in the assessment of the patient and, in some situations, of the data analyst. The purpose of blinding is to prevent identification of the treatment until any opportunity for bias has passed. These biases include (but are not limited to) decisions about whether to enroll a patient, allocation of patients, clinical assessment of end points, and approaches to data analysis and interpretation. Designs in which both the assessor and the patient are blinded are called double-blind designs. Designs in which only the patient or only the assessor is blinded to the treatment are called single-blind designs. Studies should attempt to maintain blinding until the final patient has completed the study, although this has proved difficult in certain pediatric studies of severe diseases. Clinical studies in humans typically have a Steering Committee to provide oversight of the trial and a Data Safety and Monitoring Board (DSMB) to provide ongoing monitoring. To perform their charges, committee members may observe results of the trial categorized into groups of patients-that is, the committee members are aware of which group a patient is in but unaware of which intervention that group is receiving. The committee members are said to be group unblinded, whereas the investigators remain totally blinded.
Blind Assessor Certain studies present challenges to the maintenance of blinding because blinding is either unethical or impractical. For example, surgical versus nonsurgical interventions prevent the patient and the surgeon from being blinded because they know whether surgery was performed. In this situation, a blind assessor may be used to evaluate the patient's condition. The blind assessor may
C HAP T E R 6
153
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
be a physician, nurse, or other health professional who evaluates the patient's response to treatment but is unaware of the treatment being given.
,
II
TABLE 6-4 Example of a Randomization Schedule (Nonstratified, Blocks of 8)"
Patient Number
Double-Dummy Design to Maintain Blinding
Randomization Block 1
Another situation in which blinding of the patient is difficult is when the dosage administration regimen is different for two drugs being compared. An example in rheumatology is the comparison of methotrexate (administered once weekly) versus hydroxychloroquine (administered once daily). In this case, the double-dummy design can be a useful way to maintain the blind. In the example mentioned, patients who are to receive methotrexate take active methotrexate once per week and dummy hydroxychloroquine each day, whereas patients who are to receive hydroxychloroquine receive active hydroxychloroquine each day and dummy methotrexate once per week. Double-dummy designs are limited by ethical issues involving, for example, repeated infusions or other aggressive means of delivering the "dummy" agent.
Treatment A Treatment B
Randomization The purpose of randomization is to introduce a deliberate element of chance into patient assignment to the treatment groups. Randomization reduces (but does not eliminate) the chance of an unequal distribution of known or unknown prognostic factors among the treatment groups. It also reduces possible bias in the selection and allocation of subjects. Many randomization schemes are currently employed. The simplest form of randomization is unrestricted. Patients are assigned to one of two or more treatment groups by a sequential list of treatments. The list of treatments is known as the randomization schedule. Blocked randomization is commonly used to ensure that equal numbers of patients are placed in each treatment group (Table 6-4). Note in the table that the assignment to groups is not sequential, but when the block is full an equal number of patients will have been enrolled into each group. If the blocks are too small, there is a risk of unblinding. As an extreme example, suppose blocks were only two cells large for a study that planned to compare two drugs. If a clinical investigator knew the block size, enrolled only two patients, and became aware of what one of the patients was receiving through an adverse drug effect, then he or she would automatically know what the other patient was receiving. On the other hand, if the blocks are too large they may not be completely filled, thus increasing the likelihood of unequal assignment to the groups. In recent pediatric rheumatology studies involving two groups, block sizes of six to eight have been used. Clinical investigators are never made aware of block size during the trial. Blocks may also be stratified by some prognostic factor to ensure equal distribution of the factor among the treatment groups. In multicentered trials, randomization may be stratified by center, such that each center has its own set of blocks. This tends to produce equal numbers
1 3
2 4
6 5
8
12
13
14
9
10
11
15 16
7
Randomization Block 2 Treatment A Treatment B
'The first patient entering the study receives treatment A, the second receives treatment A, the third receives treatment B, and so on. The sequence of assignments is random. When the block is full, equal numbers of patients have been enrolled into each treatment group. After block 1 is full. the assignment moves to block 2.
of patients in each group at individual centers. In pediatric rheumatology, stratification by center is frequently not possible because only small numbers of patients are enrolled at each center. If a multicentered trial uses only one randomization schedule for all centers, whether it is unrestricted or stratified, the study is said to be randomized across all centers. Typically, a central coordinating center takes responsibility for giving all centers randomization numbers. The use of more than two stratification factors is rare. Such designs are logistically difficult and do little to achieve a balance of prognostic factors. Typically, a DMSB checks whether randomization is achieving balance of important prognostic factors while the trial is continuing. If imbalance of one (or at the most two) prognostic factors is found, the randomization scheme may be altered to achieve more balanced groups. This is known as dynamic or adaptive randomization.
Design Configurations of Comparative Studies Design configurations refer to treatment group assignments after initial randomization (Le., whether or not patients remain in the same treatment group throughout the study). All of these configurations may be open, blinded, or both. If patients remain in the same group to which they are initially assigned, the study is known as a parallel group design. In crossover designs, patients switch from one treatment to the next, often in a randomized manner, and each acts as his or her own control for purposes of analysis. The crossover design in the trial must be distinguished from the frequently used open-label extension phase (discussed earlier), in which all patients receive the active drug. Factorial designs allow for study of the interaction of two treatments that are likely to be used in combination. The simplest factorial design is a 2 x 2 design in which patients are assigned to receive drug A only, drug B only, both drug A and drug B, or neither drug A nor drug B. Factorial designs are also used to study dose response when two agents are used together. Group sequential designs are particu-
154
C HAP T E R
6 DESIGN, MEASUREMENT, AND ANALYSIS
lady well suited to interim analyses. This design implies that the various treatment groups are evaluated for safety and efficacy at periodic intervals during the trial to determine whether the trial should continue or be stopped because of safety or efficacy concerns. Other comparative designs, whose basic approaches are evident from their names, include dose-escalation and fixed-dose dose-response trials. A design recently used to study etanercept in the treatment of polyarticular JRA77 is the blinded withdrawal design. In this approach, all patients receive active medication long enough to establish whether patients respond (according to a standard definition). Patients who are not classified as "responders" after the prescribed time period are discontinued from the study and classified as therapeutic failures. Patients who respond are randomly assigned either (1) to be withdrawn blindly from active medication and given placebo or (2) to continue to receive active medication but in a blind manner. A common phenomenon in blinded-withdrawal studies is a mild flare of disease among patients who continue to receive (blinded) active medication after randomization. This is called the reverse placebo effect, because it is the reverse of the beneficial effect that often is observed in patients who are blindly randomized to placebo. The primary outcome after randomization can be time to flare or percentage who flare (according to a standard definition). A design that has gained widespread acceptance in oncology and is now being used in pediatric rheumatology is the open-randomized, actively controlled trial. Patients are randomly assigned to one of two or more treatment arms, all of which are active. The trial is open and, in general, no additional trial procedures or visits other than routine care are required. These trials have numerous advantages but also substantial disadvantages. 79 This design has gained in popularity as a method to study treatments of rare conditions in which pivotal, confirmatory trials are not possible and for which there is little economic incentive for pharmaceutical companies to develop the product or for studies of combination therapy.Ho.H! These trials tend to be more "user friendly" than studies that are part of a clinical development program. Perhaps their biggest advantage is that they are inexpensive to conduct, because third-party payers may be billed for the procedures. They are not used to seek a new label or indication for an agent, and they are considered exploratory.
N of 1 Design The N of 1 approach repeatedly and randomly crosses over individual patients from one therapy to the next. For example, the randomization scheme may be A, B, B, A, A, B. Data from numerous N of 1 trials in individuals may be combined to increase the sample size, but this is fraught with difficulties and sources of potential bias. These studies are considered exploratory and are hampered in rheumatology and other diseases by the carryover effects of the treatment, natural fluctuations of the disease state unrelated to therapy, and logistic problems. For a discussion of the usefulness
OF CLINICAL INVESTIGATIONS
of these designs in rheumatology, see the review by Giannini. H2
Intent of Comparative Studies The type of comparison that one intends to carry out must be decided on before the protocol can be synthesized. Major types of comparisons include trials to show superiority, trials to show equivalence, trials to show noninferiority, and trials to show dose-response relationship. All comparative trials must possess assay sensitivi~y, defined as the ability of a study to distinguish between active and inactive treatrnents. H3 Trials to show superiority are perhaps the most frequent type of comparative studies. They are designed to show superiority of the investigative agent compared with either placebo or an active comparator or to demonstrate a dose-response relationship. In pediatric rheumatology, placebo-controlled studies have become more difficult because some existing agents are clearly better than placebo. In such situations, the use of a placebo design is considered unethical and an active comparator is substituted. Trials to show eqUivalence do not aim to show superiority but rather equivalence (either biologic or clinical). They intend to show that the difference in response to two or more treatment approaches is clinically unimportant. For equivalence trials, the usual statistical approach is to use two-sided confidence intervals (CIs) (explained later). Equivalence is inferred if the entire CI of the true treatment difference falls within the equivalence margins. Stated another way, a statistical test of inference that results in a nonsignificant P value (i.e., no difference between the test drug and the active comparator) is not enough to conclude that the two agents are equivalent. Trials to show noninferiority are similar to equivalence trials, but the question is asked in only one direction (i.e., whether the investigative agent is no worse than the standard therapy). In this case, a one-tailed CI is used to infer noninferiority. Again, a simple one-sided test of the null hypothesis (Ho) that finds "no difference" between the treatments is insufficient to make conclusions about noninferiority. The latter two trial types can be difficult to design, and sample size requirements are often much higher than for superiority trials. This is particularly true in active control equivalence or noninferiority trials that do not use placebo or that do not use multiple doses of the new drug.H3 The lack of internal validity makes external validation necessary. Active comparators should be chosen that have been shown through convincing, confirmatory trials to be efficacious in the particular condition. Further, the same response variables should be used in the equivalence or noninferiority trials that were used in the confirmatory trials of the active comparator, and equivalence margins should be clinically sound. Trials to show dose-response relationships occur throughout the development phase of a drug and may have numerous objectives, including confirmation of efficacy, establishment of the dose-response curve, estimation
C HAP T E R 6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
of the most appropriate starting dose, strategies for dose adjustment, and estimation of the maximum dose.
Ethical Requirement for Clinical Equipoise in Comparative Studies Clinical equipoise refers to the equality regarding probability of benefit that must exist between two or more groups being compared in a study. This probability of benefit is derived from existing, scientifically valid evidenceof the effectiveness of the agents being tested, and not from anecdotal or "gut" feelings. In other words, an investigator may ethically enroll patients into a comparative trial if there is a lack of convincing evidence of superiority of one of the agents, or if there is reasonable doubt in the medical community about the clinical utility of the agents. The requirement of clinical equipoise is often mistakenly interpreted as making any placebo-controlled study unethical. Ongoing debate in the literature of the ethics of placebo-controlled trials, particularly in children, is much too extensive to discuss in detail here. 83-t85 Excellent arguments for when placebo trials are appropriate have been presented by Freedman. 86--88 According to these authors, five conditions in which a placebo control may be used are as follows: There is no standard treatment. Standard treatment is no better than placebo. Standard treatment is placebo. The net therapeutic advantage of standard treatment has been called in question by new evidence. • Effective treatment exists but is not available due to cost or short supply.
• • • •
The final arbiter is always the Human Subjects or Ethics Committee.
Conducting the Clinical Trial With the advent and widespread use of independent forprofit clinical research organizations (CROs) and site management organizations (SMOs), the quality of clinical trial conduct has risen substantially. Academic research organizations (AROs) appeared in the 1990s. Although not typically involved in the day-to-day operations of the trial, AROs provide the basic scientific and theoretical background for the trial. These functions may include biologic justification for the selection of the particular therapy, identification of biologic and clinical response variables, development of the protocol, interpretation of the results, and final report preparation. Pediatric rheumatology clinical trials, with few exceptions, are multicentered approaches. This term implies that multiple clinical investigative sites are used to enroll enough patients to meet statistical power requirements. A coordinating center (CC) is responsible for coordinating almost all trial activities. The role of the CC is determined in part by whether a CRO is used and whether the trial is part of a clinical development program or an investigator-initiated protocol. Site monitoring may be a function of the CC or the CRO. During visits to clinical sites, site monitors-known throughout industry as clinical research associates (CRAs)-verify the data on the
155
case report forms against source documentation (j.e., original reports from the laboratory and clinical records). Data collection (or capture) is moving to "paperless coordinating centers." Before the widespread use of electronic communication, almost all data were collected during the site-monitoring visit, or forms were mailed to the CC or directly to the sponsor. Now, use of e-mail and electronic entry of data via scanning of forms is increasing. The electronic age has drastically reduced the number of patients entered into trials who do not meet the eligibility criteria and the amount of inappropriate, incorrect, or missing data. Data collation refers to the distillation of data from the case report forms to summary tables and spreadsheets. This step is necessary before data analysis can be performed and facilitates the review of both safety and efficacy data by the DMSB. The DMSB is charged with, among other items, determining whether the trial should continue unchanged, be modified, or be stopped early because of safety or efficacy concerns.
Data Analysis Plan The role of the statistician begins with planning of the study and not after the data are already in hand. Plans for exploratory studies may not be as formal as those for confirmatory trials, because the former permit the use of data-driven hypotheses to be tested. That is, data exploration is encouraged in exploratory, hypothesis-generating, nonpivotal trials. A single study may have both confirmatory and exploratory aspects. At a minimum, the statistical considerations document includes the following items: 1. Whether descriptive statistics, statistical inference, or both will playa role in the analysis 2. Identification of the primary and secondary response (outcome) variables 3. Calculation of sample size, including assumptions that will be used to justify the sample size, which in turn include the a and ~ error levels, the difference that one wishes to detect as statistically significant, and how the variance estimate will be obtained 4. How the various analysis sets of patients to be used in the analysis will be formed, including the plan for handling dropouts, noncompliant patients, and other sets of patients who do not complete the protocol as written 5. Which statistical tests of inference will be used and the justification for their use 6. Plans for adjustment of P values based on the number of secondary hypotheses to be tested 7. Statement of which aspects of the analysis plan are expected to generate confirmatory data and which are exploratory, including plans for any subgroup analyses 8. Plans for handling covariates 9. The data management and statistical analysis software that will be used
Most of these items are discussed later (see "Understanding and Describing Data" and "Statistical Tests of Inference Commonly Used in Clinical Investigations"); others are selfexplanatory. Items 2 and 4 require further attention here.
156
C HAP T E R
6 0
E S I G N, MEA SUR E MEN T, AND
Primary and Secondary Response Variables Response variables are defined as those outcomes that will be used as the main evidence of the treatment effect of the investigational drug. Students of clinical research should keep in mind that the field of response variables is not static, and advances in their measurement, description, and methods of analysis will continue to change and improve, as they have done since the previous version of this text.89 Treatment effect is defined as an effect that is expected to result from a therapy. In comparative trials, the treatment effect of interest is a comparison of two or more agents. Treatment effect is distinguished from effect size, which is a measure of the responsiveness (sensitivity to change) of the outcome variable and is defined mathematically as the mean change of the variable among the patient groups divided by the standard deviation (SO) of the baseline score. A related term is the standardized response mean, which is the mean change in a variable's score from baseline to the follow-up visit, divided by the SD of this change. 90 In studies designed primarily to observe safety and tolerability of an agent, the "response" variable relates to adverse events or treatment-emergent adverse drug effects rather than to efficacy. Thus, the choice of primary response variables largely depends on the objectives of the trial and should reflect clinically relevant effects. Secondary response variables are usually (but not always) associated with the exploratory nature of the study. Surrogate endpoints are outcomes that are intended to relate to a clinically important end point but do not in themselves measure a clinical benefit. The use of biologic surrogate end points in rheumatology as primary outcomes is somewhat suspect. For example, a decrease in some targeted T cell type may not produce clinical benefit even though the desired biologic effect was achieved. A frequently encountered problem in rheumatology is that of multiplicity (Le., the use of multiple primary end points with repeated statistical testing). To avoid this problem, the use of composite variables has become popular. This strategy involves the integration or combining of multiple relevant variables into a single variable, using a predefined algorithm. Three examples of composite variables are the American College of Rheumatology 20 (ACR-20)91 and the Disease Activity Score (DAS)92 for use in trials of adults with RA and the ACR Pediatric-30 for use in trials of children with JRA. 93 Each attempts to dichotomously categorize patients as improved or not improved, using a core set of variables assembled into an algorithm. Composite variables avoid the multiplicity problem without requiring adjustment of the type I error level (described later) due to multiple hypothesis testing.
Response Variables Based on Claims Allowed The claims that the FDA allows for antirheumatic and antiinflammatory therapies for RA and JRA include reduction in signs and symptoms, major clinical response, complete clinical response, remission, prevention of disability, and prevention of structural damage (see http://wwwfda.gov/ cber/gdlns/rheumcln.pdj). The reduction in signs and symptoms claim is usually the first to be granted for marketing approval. This claim
A N A LY SIS
0 Fell N I CAL
I N V E S TI GAT ION S
is typically established in trials of at least 6 months' duration unless the product belongs to an already well-characterized pharmacologic class, in which case trials of 3 months' duration are sufficient to establish efficacy for signs and symptoms. For trials in adults, the FDA recommends that the ACR-20 criteria be used. In pediatric rheumatology, the FDA suggests that the ACR Pediatric30 be used for JRA. The major clinical response claim is awarded to agents that are able to show a response at the ACR-70 level, rather than the 20% improvement needed for a signs and symptoms claim. This claim is based on statistically significant improvement response rates by the ACR-70 definition compared with background therapy in a randomized controlled group. Trial duration should be a minimum of 7 months for an agent that is expected to have a rapid onset of action and longer for agents with less rapid effects. The complete clinical response claim is granted to a drug that produces a remission for at least 6 continuous months by the ACR-20 criteria and by radiographic arrest. Complete clinical response indicates that the patient is in remission but is still taking antirheumatic drugs. Typically, trials for a complete clinical response last a minimum of 1 year. Remission is defined as continuation of the same result after all antirheumatic drugs have been withdrawn. The remission claim is granted if remission by the ACR definition and radiographic arrest (no radiographic progression by the method of Larsen and colleagues94 or by the modified method of Sharp and associates95 ) are maintained over a continuous 6-month period while the patient is off all antirheumatic therapy. A drug need not be a cure to be awarded a remission claim. A remission claim can be granted even if the patient relapses after 6 months or more of remission. Trials aimed at a remission claim should be at least 1 year in duration. Wallace and colleagues56 developed a preliminary definition of clinical remission for use in JRA. The prevention of disability claim is granted to drugs for which the primary outcome is a functional ability measure, such as the Childhood Health Assessment Questionnaire or the Arthritis Impact Measurements Scale. In addition, the full effect of JRA on a patient is not captured without the use of a more general healthrelated measure of quality of life. For this reason, data from a validated measure such as the Medical Outcome Study Short-Form Health Survey (SF_36),96,9 7 the Childhood Health Questionnaire, or the Pediatric Quality of Life Inventory Scales (Peds QL)98,99 should also be gathered, and the patient's condition should not worsen on these measures over the duration of the trial. The prevention of structural damage claim is granted to drugs that show either a slowing of radiographic progression or the prevention of new erosions demonstrated by radiography or other measurement tools such as magnetic resonance imaging. These trials should be at least 1 year in duration. Certainly, other clinical efficacy response variables are possible, and in pediatric rheumatology the ACR Pediatric-30 is likely to be inappropriate for the study of rheumatic conditions of childhood other than JRA.
C HAP T E R
6 0
ESIGN, MEAS UREMENT, A ND AN A LYSIS OF CLINICAL INVESTIGA nONS
Whatever variables are chosen, they must possess a host of validity characteristics. These include responsiveness (sensitivity to change within the trial's duration), face (clinical sensibility), content (comprehensiveness), construct (biologic sensibility, or how the variable is hypothesized to behave as compared to how it does behave), and criterion (does it agree with the gold standard, if one exists) validity.lOo In addition, variables should be reproducible (reliability) and, if more than one variable is chosen, nonredundant with one another.
Analysis Sets (Patients) Not all patients who enter a trial complete the protocol as it is written. The analysis plan must state how subjects who drop out, are noncompliant, or in some other manner do not follow the protocol specifications will be handled. The formation of analysis sets should be aimed at minimizing bias and avoiding an increase in the possibility of an erroneous conclusion that a difference is present between groups when, in fact, it is not (type I error, described later). The per-protocol set (also called valid cases set, efficacy sample, or evaluable subjects sample) comprises those subjects who closely follow and complete the protocol. In practice, consideration of only the per-protocol set results in the loss of valuable information from patients who perhaps completed most of the study or had only one or two major protocol violations related to, for example, concurrent medication. Therefore, the full-analysis set is also used for the primary analysis. The full-analysis set refers to the intent-totreat approach and is derived from all randomized patients, including those who dropped out early or had a major protocol violation. Historically, the intent-to-treat analysis meant that all patients, whether they dropped out, were noncompliant, or otherwise deviated from the written protocol, were evaluated for outcome at the time that they would have had their last visit (because one intended to treat them until then). The concept is embodied in the brief saying, "Once randomized, analyzed." However, this approach results in the introduction of substantial bias and is problematic in rheumatology and other specialties in which patients, once off trial, are lost to follow-up and receive various other medications and procedures. It is now common to use a modified intent-to-treat analysis, the last-observation-carried-fonvard (LOCF) approach. This technique involves using the last value obtained for a response variable (no matter when in the trial it was measured) as if it were measured at the scheduled final visit. In this way, the data from noncompleters who were exposed to the drug long enough to experience treatment effects (if any) can be combined with the data from the per-protocol set. The intent-to-treat approach allows for minimal exclusion of some patients, such as those who, after randomization, never received any of the investigational medication.
UNDERSTANDING AND DESCRIBING DATA In any type of clinical investigation, the investigators collect data and want to be able to summarize, interpret, and con-
157
vey it to other parties. In order to do this, it is important to understand how measurements are made, how data can be displayed, and how the types of data determine the appropriate statistical test. The types of measurements (variables) determine which statistical test to use.
Scales of Measurement: Categorical, Ordinal, and Continuous Data The scale or level of data has important implications for how information is displayed and summarized. All data may be classified into one of the following measurement scales: categorical, ordinal, or continuous (numeric).
Categorical Variables For categorical (qualitative) variables, sometimes called nominal variables (i.e., "in name only"), each subject can be placed into one of the categories. Variables with two possible outcomes, such as Yes/No or Male/Female, are called dichotomous. Categorical variables are categorized in terms of proportions or percentages (e.g., the study population was 75% female and 25% male). The best ways to display categorical data include contingency tables and bar charts.
Ordinal Variables On an ordinal scale, the variables used have an inherent order. Subjects can be placed in "ranked" or "ordered" categories. Examples of ordinal variables include the severity scores of swelling (0 to 3+), Apgar scores, and tumor staging. Order exists among the categories, but the difference between adjacent categories is not uniform throughout the scale. Figure 6-3 shows an example of an ordinal variable, the standard method used to grade swelling by pediatric rheumatologists. The difference between swelling severity scores of 0 and 1 is not the same magnitude as the difference between scores of 2 and 3. Ordinal variables are best summarized using percentages and proportions. The entire set of data measured as an ordinal scale may be summarized by a median value.
Continuous Variables Continuous variables are observations in which the differences between numbers have meaning on a numeric scale. These are quantitative measures. Examples are age, height, weight, blood pressure, survival time, and laboratory values such as glucose, sodium, and potas-
0 - - - - - 1 + - - - - - 2+ - - - - - 3+
None No joint swelling
Mild Moderate Mild. definite Definite swelling swelling but with no with obscuring blurring of normal of skeletal landmarks skeletal outlines
Severe No discernible skeletal landmarks
• Figure 6-3 Example of an ordinal level variable. An ordinal scale is used to assess the severity of swelling in a joint.
158
C HAP T E R
6 DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
sium levels. Although continuous variables all have meaning on a numeric scale, differing degrees of precision are required for different types of studies. For instance, age in a study of adults may be estimated to the closest year; in children, age may have to be estimated to the closest month, and in neonates, to the closest day or hour. Characteristics measured on numeric scales can be displayed in tables and graphs. Means and SDs (discussed later) are used to summarize continuous variables.
When to Convert Higher Levels of Data to Lower Levels As a rule, one should work with the highest level of data possible, because of increased quantitative precision and because parametric statistics are, in general, more powerful than nonparametric equivalents (see later discussion). Yet, there are times when, for example, numeric data should be converted to ordinal or nominal data before further analyses are conducted. Situations in which "lowering" the level of data may be appropriate include the following:
• In a multicentered study in which different methods are used to generate a numeric value (e.g., the antinuclear antibody titer when different substrates are used). In this situation, one may be forced to "dichotomize" patient results, describing them as simply "normal" or "elevated," and conduct the analysis using statistics appropriate for a nominal rather than a continuous variable. • ff the experimenter suspects measurement error in the data. An example is compliance with a prescribed drug dosage or with a clinical trial or physical therapy program. It may be necessary to divide the patients dichotomously and classify each as "compliant" or "noncompliant." • If reliability (reproducibility) of the measurement tool is unknown or likely to be poor. In pediatrics, the reliability of measurement on visual analogue scales is suspected to be low in young children. Rather than reading the result in centimeters from the left side of the scale, investigators frequently place a grid that is divided into 10 equal segments over the visual analogue scale line and read the result as a whole number from 0 to 10. This effectively converts a continuous variable to an ordinal level outcome. • In clinical trials when one wishes to use a set of response variables (of any data level) to dichotomously divide patients into "responders" and "nonresponders." Examples have been presented earlier in this chapter.
Concepts Related to Measurement of Variables: Validity, Variability, and Bias In clinical research, it is obvious that not all patients treated identically will experience an identical response. This is known as the variability that is common to virtually all human experimentation. Certain important terms are associated with describing how this variability may have arisen. In some situations, its sources can be mini-
mized or eliminated altogether. Variability is sometimes called error. Error may be broadly classified into nonrandom and random error. Nonrandom error is also called bias or systematic error. It results in a lack of validity of a measure and therefore influences the accuracy of the measure. In general, validity is equated with accura9. Random error refers to imprecision. In Figure 6-4, the different types of random error and systematic error are graphically demonstrated.
Potential Sources of Variability in Measurement of Individuals Variability may arise among individuals from a number of factors, including diurnal variation; changes related to factors such as age, diet, and exercise; and environmental factors, such as season or temperature. Variability may also arise from measurement characteristics, including poor calibration, inherent lack of precision of the instrument, and misreading or misrecording of information from the instrument.
External Versus Internal Validity External validity may be equated with generalizability. It determines the population settings to which measurement and treatment variables can be generalized. Internal validity defines how valid the conclusions are within the patient sample studied; it is a basic minimum requirement. without which any study is uninterpretable. The question of external validity is meaningless without first establishing whether the study is internally valid. Obviously, both types of validity are important. However, they may be at odds, in that study design features that increase one may tend to decrease the other.
Bias An excellent, concise discussion of biases can be found
at http://www.musc.eduldclicrebmlbias.html. Space considerations prevent a thorough discussion of all types of
Accuracy and Precision
Accuracy Only
. . ::.. : . .. @ @ O ..... . ..
Precision Only
Neither Accuracy Nor Precision
.. .... -: ..
• Rgure 6-4 Combinations of accuracy and precision in describing a continuous variable. Accuracy is impaired by bias or systematic error, precision by random error.
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
bias that can enter into clinical studies. Sources of bias that may occur in clinical studies include selection, measurement, unacceptability, confounding, recall, referral, volunteer, withdrawal, attention, investigator, and verification, among others. To complicate matters further, the same type of bias may go by different names or be a subset of some other bias (see later discussion). Many are self-explanatory and need little explanation. A few of the more important types of bias are discussed here. Selection bias-the distortion of study effects resulting from the sampling of subjects-includes volunteer bias, nonresponse bias, and bias resulting from loss to followup. Another subtype of selection bias is referred to as detection bias. Measurement bias (sometimes referred to as information bias) is distortion of the study effect resulting from inaccurate determination of the study variables (either exposure or disease). Measurement bias may be divided into nondifferential and differential misciassification. Nondifferential information bias can occur if the exposure is not accurately assessed. This type of bias may occur in occupational research, for example, if job titles are used as a surrogate for exposure status. Another form of nondifferential measurement bias is unacceptableness bias, in which the exposure may be under-reported by patients if it is unacceptable behavior. This is likely to have an impact on all subjects, not just subjects with the disease of interest. Differential misclassification bias includes recall bias, in which the recall of information about exposure is influenced by whether the person has the disease (Le., cases may have more accurate memory of events leading to disease than controls who have no disease). Interoiew bias can occur if the circumstances under which different groups of subjects are interviewed are not compatible. These circumstances include time from exposure to interview, setting of the interview, person doing the interview, manner in which questions are asked (prompting), and whether the subject has knowledge of the research hypothesis. Case-control studies are particularly vulnerable to informationbias. Confounding bias is a distortion of the study effect that results from mixing of the exposure associated with the disease with the effects of one or more extraneous variables. An extraneous variable that wholly or partially accounts for the apparent effect of the exposure or that masks an underlying true association is called a confounder. Examples of confounding are (1) an apparent association between an exposure and a disease that may actually be due to another variable and (2) an apparent lack of association between exposure and disease that results from failure to control for the effect of some other factor. An example of confounding bias in pediatric rheumatology can be found in a recent report by Brunner and colleagues. 101 These investigators attempted to identify risk factors for damage in childhood-onset systemic lupus erythematosus (SLE). An association was found between damage and disease duration, indicating a possible (and logical) cause-and-effect relationship between the two. However, once the data were corrected for the confounder disease activity over time, disease duration disappeared as a predictor of damage.
159
To assess the pOSSibility of confounding, the standard technique of stratifying the data by the potential confounder may be used. One looks for an association between the exposure (as a possible causal factor) and the disease, then compares those subjects who have the confounder with those who do not to see whether an association exists. Another common method is to use Mantel-Haenszel procedures to calculate an overall relative risk in which the results from each stratum are weighted by the sample size of the stratum. 102 Only established risk factors for the disease should be investigated as potential confounders. In brief, these can be dealt with in the design of the study (i.e., by matching) or by stratification or multivariate analysis (see later discussion).
Desalblng Data and the Frequency Distribution of Continuous Variables Descriptive statistics are commonly used to graphically represent individual data points or to summarize groups of data, regardless of the data level. Many exploratory and epidemiologic studies use only descriptive statistics, rather than inferential statistics (tests of hypotheses). Graphs such as dose-response curves represent descriptive statistics. Rates and ratios are commonly used in epidemiologic studies to describe disease frequency and distribution. A rate (or proportion) implies that the numerator is part of the denominator and is usually associated with a time element (e.g., an annual case-fatality rate of 11/120 implies that the 11 deaths came from the total of 120 cases). The numerator of a ratio is not part of the denominator (e.g., the female/male ratio among patients with oligoarticular-onset JRA is 6: 1). Statisticians employ many types of distributions for describing and analyzing data. These include the binomial (BernoullO, geometric, chi-square, Poisson, t, and F distributions, among others. The frequency distribution of continuous variables is most commonly referred to in the medical literature and is the only distribution discussed in detail in this chapter. Its parameters form the basis of much of the descriptive statistics used in the reporting of data from clinical investigations. A frequency distribution of a continuous variable is simply an x-y plot of the possible values that a variable can take on (x-axis) versus the number of observations having the particular values (y-axis). If the frequency distribution is normally distributed it is called a gaussian distribution (Fig. 6-5).
Measures of Central Tendency, Skewness, and Kurtosis Every distribution of continuous variables has an arithmetic mean (average), which is calculated by adding the observations and dividing the sum by the number of observations. The median of any distribution is the centermost value. If the distribution has an even number of observations (Le., there is no center value), the median is calculated by averaging the two most center values. The median is also the 50th percentile. The mode is the
160
C HAP T E R 6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
-25.0.
-1 S.D.
o
+2 S.D.
1
1
1
I
I
1 1
1 1
1 1 I 1 1
'"
."
/
1 1
I
/1 /
I
2.1% I';
,;
1/
I
1
ONE·TAILED
/
/ / 13.6%
1 1
1
I 1 1
34.1%
34.1%
1
1 .05
I
1
"
• Figure 6-6 Effect of positive skewing on the location of the mean, median, and mode. (The mean and median are skewed to the right.) The effect of negative skewing is the mirror image of this figure.
1.64 I
TWO-TAILED
.025 1.96
•
14
1.96
•1
• Figure 6-5 The normal or gaussian distribution (bell-shaped curve), showing the approximate percentage of observations expected to be found within one and two standard deviations (SO) from the mean value. Also note the critical values for one- and two-tailed tests of hypotheses.
most frequently observed value. The mean, median, and mode are all called measures of central tendency. A distribution has only one mean and one median, but numerous modes are possible; this leads to forms such as bimodal and multimodal distributions. In a normal distribution, the mean, median, and mode are all the same value. Figure 6-6 demonstrates the effect of positive skewing on the measures of central tendency. The skewness is said to be positive because there are too many observations in the upper (shaded) tail (Le., toward the right side of the distribution). This type of skewing typically occurs in distributions that have a fixed lower boundary but no upper boundary (e.g., results of liver function tests). Negative skewing is the mirror image of Figure 6-6 and has the opposite effect on central tendency measures. An example is the age at onset of disease among patients with oligoarticular ]RA. Kurtosis is a measure of the peakedness of a distribution.
Measures of Spread or Variation The frequency distribution of continuous variables can be described by its mean and its standard deviation (SD). The SD is a measure of the spread of values. Abbreviations in common use for the mean include x for the sample mean and Il for the underlying population mean from which the sample was drawn. The SD in a formula is designated s for the sample SD and 0 for the population SD; it is the square root of the distribution's variance. Variance is calculated by subtracting the mean from each of the individual values in the distribution, squaring the differences (to eliminate the negative sign), summing the squares, and dividing the result by the
number of observations minus 1. The sample variance is abbreviated S2, and the variance of the population from which the sample was drawn is 0 2 • If a frequency distribution is normally distributed, the distance ± 1 SD from the mean includes 68.3% of the observations, ±2 SD includes 95.8% of the observations, and ±3 SD includes 99.7% of the values. Actual distributions from clinical investigations may include more or fewer of the observations at the SD cut points than these theoretical percentages indicate. To demonstrate an SD, consider the following example. The mean number of swollen joints among 551 children who were entered into trials of second-line agents was 16.1, with an SD of 11.6. Therefore, 376 (68.3%) of the children in these trials should have swollen joint counts between 28 and 45 06.1 ± 11.6). Note that if one were to consider 2 or 3 SDs from the mean, the range of values included would drop below 0, again pointing to the theoretical nature of the SD rather than actual values. The impossible values are an indication that the distribution is not perfectly normal.
Z Scores: Placing a Single Value Within a Distribution of Values Frequently, clinical investigators find it useful to locate exactly where an individual patient's value for some variable lies within a distribution of values. Because the normal distribution depends on two parameters, the mean and the SD, there is an infinite number of normal curves, based on the variable being measured. However, all tables of the normal distribution are for the distribution described by a mean of 0 and an SD of 1. Therefore, any variable with a mean not equal to 0 and an SD equal to 1 must be rescaled so that these parameters are met. The solution is to convert the variable to a standard normal variable, Z (also called a standard normal deviate). For example, in a clinical trial of calcium supplementation in postpubertal females with ]RA, eligibility criteria may include the requirement that the patient's bone mineral content (by dual-energy X-ray absorptiometry [DEXA]) must be less than 1.5 SD below the mean of the normal population. A Z score can be calculated by subtracting the population mean from the measured value in the individual and dividing the result by the SD of the
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
population: Z = (x; - /l)/a, where Xi is the individual's value. One may transform the Z scale back to the original scale by the formula Xi = /l + aZ. Thus, if the population mean is 2143 g and the population SD is 308 g, a patient whose bone mineral content is less than 1681 g (Le., 2143 - [308 x 1.5] respecting the negative direction) qualifies for the study with regard to this eligibility criterion. A Z score also can be calculated using the sample mean and SD. In addition, two means can be compared to determine whether they are statistically significant via the Z test if the sample size is large. Any continuous numeric variable can be converted to the Z scale.
161
Although the median, mode, and range (described earlier) are helpful, the mean and SD may be quite meaningless in this situation. A commonly used method is to group the ranked values in a non-normal distribution into quartiles, which are similar to percentiles, but with only four categories: Q1 = 25%, Q2 = 50%, Q3 = 75%, and Q4=100%. The spread, or dispersion, of a non-normal distribution is described in terms of the interquartile range. This is the difference between the highest value in the third quartile (Le., the 75th percentile) and the highest value in the first quartile (i.e., the 25th percentile). In other words, the formula for the interquartile range is Q3 - Q1.
Standard Error of the Mean The standard error of the mean (SEM) represents a different concept from the SD. Mathematically, it is expressed as SEM = SD/Yn, where n is the sample size. Because samples drawn from an underlying population do not each produce the same mean value (but tend to cluster around the same value), one must calculate the range of where the true (unknown) population mean lies. FrOm the formula, it can be seen that the greater the n (Le., the larger the denominator), the smaller the SEM. Thus, the SEM gives the clinical investigator an idea of how "tightly" the estimated mean from the sample represents the true, underlying mean. Investigators often ask statisticians what should be plotted when presenting the data as a mean and its accompanying measure of variability, the SD or the SEM. Because the SEM is always less than the SD, investigators tend to plot it rather than the SD. A rule of thumb is that the SD should be used when comparing values from individual subjects to a population distribution. The SEM is used when plotting mean values of two groups of subjects.
Confidence Intervals As stated earlier, the SD encompasses the variability of
individual observations and the SEM indicates the variability of means. The mean ± 1.96 SD estimates the range of values within which 95% of the observations from subjects can be expected to fall (see Fig. 6-5). Similarly, the mean ± 1.96 SEM estimates the range in which 95% of the means of repeated samples from the same population should fall. If the mean and the SEM are known, the 95% CIs can easily be estimated. These limits indicate the range of values within which the investigator is 95% sure that the true mean of the underlying population lies. One can eas!Iy calculate any CI level (e.g., 90% CI, 99% CI) by using the critical values that cut off specific areas of the curve. CIs are frequently used in addition to statistical hypothesis testing. They can be calculated for the chisquare test, the t test, regression, and a variety of other tests of statistical inference.
Describing Non-normal Distributions Not all parameters used to describe normal distributions are helpful when one is attempting to describe distributions that are nongaussian (Le., do not follow a bell-shaped curve or reasonable approximation).
STATISTICAL TESTS OF INFERENCE COMMONLY USED IN CLINICAL INVESTIGATIONS This chapter does not attempt to describe comprehensively the myriad statistical procedures that are readily available to the clinical investigator through such computer programs as Statistical Analysis System (SAS) or Statistical Package for Social Sciences (SPSS). Rather, a basic introduction to statistical concepts is provided, followed by a description of the inferential and other procedures found most commonly in the literature. Formulas are not stressed, because virtually all statistical procedures are now conducted with the use of computer programs.
Basic Concepts Relevant to Analysis Statistical approaches may be divided into frequentist methods and bayesian approaches. Frequentist methods refer to P values and CIs, which can be interpreted as the frequency of specific outcomes from the same experimental situation if it is repeated many times. That is, what are the chances of this outcome (and outcomes even more extreme) if one repeats the experiment many times? Bayesian analysis permits a calculation of the probability that, for example, a treatment is superior according to the observed data and prior knowledge. It begins with a posterior probability distribution for some parameter, which is derived from the data, and a prior probabili~y distribution for that parameter. The posterior distribution is used as the basis for statistical inference. 103 This chapter emphasizes the frequentist school, because the majority of statistics in today's literature follow frequentist rather than bayesian theory. The types of variables in the study and the number of variables studied determine the choice of the appropriate statistical approach. The first step is to determine which variables are independent (predictor or explanatory) variables and which are dependent (outcome or response) variables. An independent variable is the variable that is the explanatory factor or thought to be the cause. A dependent variable (DV) is one whose value is the outcome in the study or the response or is thought to be the effect. The second step is to determine the measurement scale of the variable: categorical (nomina!), ordinal (ranked), or continuous numeric, definitions for which were provided earlier. The third step is to deter-
162
C HAP T E R
6 DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
mine whether the study observations are independent of each other. In the design of a clinical study, one must determine whether the groups to be compared are independent or paired. Samples in which the values of one group cannot be predicted from the values of the other group are said to consist of independent groups. In other words, the patient group and the control group represent different individuals rather than the same individual measured at two different times. With paired (matched) groups, the values of one group may be predicted from values of the other. In a paired experiment, a patient may be measured before and after therapy, in which case the patient acts as her or his own control, or a patient may be paired with another individual who has been matched with respect to all of the independent variables (e.g., age, duration of disease) that may affect the DV (response). In animal studies, in which genetically identical animals are frequently used in research, paired experiments are the rule. However, in human clinical studies, it is rare that two groups can be matched for all of the independent variables that may influence the outcome variable. An investigator may wish to match the groups as closely as possible, to eliminate bias, but still treat the groups as if they were independent, thus improving the overall quality of the experimental design, as described earlier. The nature and distribution of the values of the variables also determine whether parametric or nonparametric tests can be used. The use of a parametric test is based on certain assumptions. The major assumption is that the variable of interest follows a normal distribution. It may be possible to transform variables that are not normally distributed. This technique expresses the values of observations on another scale, such as a natural log scale. This may allow the use of parametric statistical tests when the actual values obtained in the study do not follow a normal distribution. Another alternative is to use a nonparametric test. Nonparametric methods are based on weaker assumptions in that they do not assume a normal distribution or equality of variance between the different groups. There are nonparametric procedures for most statistical needs, but, because they are not based on the assumption of normality, nonparametric tests are more conservative. Unfortunately, they are also less powerful (Le., less able to reject the null hypothesis when it is false) than their parametric counterparts.
Two Types of Statistical Error and P Values Type I error is the probability of rejecting a null hypothesis when it is true (i.e., concluding that there is a difference when, in fact, there is none). It is abbreviated as ex (alpha error level). The P value is the calculated type I error level based on the data; it is defined as the likelihood that a difference at least as large as the observed difference could have occurred by chance alone. That is, it is analogous to a false-positive result in diagnostic tests (discussed earlier). Whether the P is capitalized is arbitrary and varies among publishers and journals. P values that exceed the predefined type I error level (e.g., P = .08 when ex was set at .05) are called not statistically significant; values at or below the preset type I error level
(e.g., .001) are called statistically Significant. The ex or type I error level is set; the P value is calculated. The debate about when to adjust P values to deal with the issue of multiplicity, or multiple hypothesis testing, appears far from resolved, and spirited debate continues, particularly in new technologies, such as microarray data. 104-106 When one is conducting exploratory studies not aimed at establishing definite cause-effect relationships, P need not be corrected for fear of missing a possible true association or difference. False-positive results can later be discarded in confirmatory, pivotal studies. In confirmatory studies aimed at adding pivotal evidence to a cause-effect relationship, there is no need to correct the P value for the main test of hypothesis (i.e., that on which sample size was based). However, results of secondary, exploratory hypotheses should present both uncorrected and corrected P values. An alternative to correcting the P value is to set ex lower (e.g., at .01 instead of .05) in anticipation of conducting multiple hypothesis testing. There are numerous techniques for correcting P values for multiple comparisons. By far, the most widely used (perhaps because of it'> simplicity) is the Bonferroni correction, despite its conservativeness. To adjust a P value using the Bonferroni correction, the P value obtained is multiplied by the number of statistical tests. Thus, a P value of .05 obtained in a series of 10 tests of hypotheses becomes (.05 x 10), or .5. Alternatively, when the experiment is designed, the ex error level can be divided by the number of anticipated tests (in the example, .05 .;- 10 = .005), with values of P greater than this level referred to as nonsignificant. Type II error is the probability of failing to reject a false null hypothesis in favor of the alternative hypothesis (i.e., concluding that there is no difference when, in fact, there is a difference). It is commonly abbreviated as ~ (beta error level). Table 6-5 is a summary of the types of decision errors, illustrating the concepts of the null hypothesis and ex and ~ errors. Traditionally, type I error levels are set lower than type II error levels (e.g., .05 and .2, respectively). In other words, the experimenter is more willing to make a type II error than a type I error. The conventional rationale for this approach is that type I errors are more serious, because they can result in the abandonment of an established, beneficial therapy in favor of a new therapy when no such change is warranted. Power, the ability of a statistical test to identify a true difference if in fact one eXists, is expressed mathematically as 1 minus ~. It is a consideration in the design of an experiment, because the power of the test is affected by the sample size. The distribution of the test statistic is divided into two areas: acceptance and rejection. These concepts are graphically shown in Figure 6-7. If the null II _ . TABLE
6 ')
True Situation
Oul< OUII' 01 Study Accept Ho Correct Type II error
H", null hypothesis; H" alternative hypothesis.
Type I error Correct
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
• FIgurre &-7 Theoretical visual representation of acceptance and rejection regions, alpha and beta error regions, and power.There are four possibilities when one compares the means (or other summary descriptors of a distribution of values) of two (or more) populations or samples. (1) First, one may correctly conclude that there is no difference between the two means (I.e., correctly accept, or fail to reject, the null hypothesis (HoI of no difference). In this case the mean of, for example, the treated group would fall within the distribution on the left anywhere in the acceptance region (white), but not in the rejection region (black) and also not in the stippled region (beta).This implies that the second mean arose from a population having the same underlying mean as the population that gave rise to the first mean, and the experimenter correctly recognized this situation. (2) Second, one may correctly conclude that there is a difference between the two means (I.e., correctly reject Hoin favor of the alternative hypothesis [HI or HJ that there is a difference between the two means). In this case, the lIlean of the treated group would fall within the rejection region (black) or further to the right. This implies that the second mean arose from a population with a different underlying mean from that of the population that gave rise to the first mean, and the experimenter correctly recognized this fact. (3)Third, the investigator may incorrectly conclude that there is a difference between the two means when In fact there is not a difference (I.e., incorrectly reject Hoin favor of HI when, i'n fact, it should not be rejected). In this case, the value of the second mean happened to fall into the rejection, or alpha region, even though it arose from the same population as the first mean.This is known as atype I error, and its probability of occurrence is based on the alpha error level set by the experimenter a.e., how much chance is one willing to take of concluding that there is a difference when in fact there is not?).This will determine the size or area of the rejection region. Atype I error implies that the second mean arose from a population with the same mean as the population that gave rise to the first mean, but the experimenter failed to recognize this fact because the second mean fell far out in the tail of the distribution, as shown in the graph. (4) Finally, the investigator may incorrectly conclude that there is no difference between two means when, in fact, there is a difference (I.e., incorrectly accept, or fail to reject, Howhen it is actually false). In this case, the second mean fell somewhere in the beta region, leading the experimenter to believe, incorrectly, that the second mean came from the same population that gave rise to the first mean.This is known as a type " error, and its probability of occurrence is based on where the experimenter sets the power (1 - the beta error level) of a test. Power will determine the size of the beta area on the graph and is usually heavily dependent on sample size. Atype II error implies that the second mean arose from a population with adifferent mean from that of the population that gave rise to the first mean, but that the experimenter failed to recognize this fact because the second mean fell somewhat close to the first mean (I.e., in the beta region) own.
163
some guesswork, and the resulting calculation may not always yield the correct sample size needed to answer a specific question. This problem occurs when the investigator's assumptions do not hold true for the sample that is actually enrolled in the study. Sample size should always be calculated during the development of the clinical investigative protocol. Sample size is most frequently calculated with the use of computer programs, but the investigator and the statistician must still be able to make various assumptions that they think are likely to be met during the study. For chi-square tests (discussed later), the investigator needs to know the outcome of interest (which should be specifically defined), an estimate of the magnitude of effect (Le., how much difference can one expect between, for example, a control group and a treated group), the desired type I error level (usually .05), and the type II (~) error level (i.e., 1 - power). With these pieces of information, the calculations are relatively straightforward and can be ascertained from tables or with the use of appropriate computer programs. To calculate the sample size needed for a parametric test, such as the t test, one must also estimate the variance in the variable of interest. The variance estimate may come from published data or from a pilot study that was designed to preliminarily assess the question under consideration. Because many sample size calculations result in the requirement of an unrealistic number of patients, the statistician is often asked to find ways to decrease the number of patients needed. Sample size may be decreased by increasing the acceptable type I error level, by increasing the acceptable type II error level, by increasing the size of difference reqUired to detect a statistically significant difference (which should also be clinically significant), and by choosing an outcome variable with a smaller amount of variance. The most common ways to do this include improving the precision of the measurements of the outcome variable, training investigators, using better equipment, and repeatedly measuring the outcome.
Post Hoc Power Analysis
hypothesis is rejected, one concludes that the evidence supports a significant difference between the groups. If the null hypothesis is not rejected, one concludes that there is no such difference. The lower the P value, the higher the level of significance.
In the event that an investigation yields nonsignificant differences, the concern is that the investigator has committed a type II error. The options to address this situation include calculation of the size of the difference one could detect as statistically significant with sufficient power (e.g., 80%), given the sample size and variance obtained in the study; this is termed the minimum detectable difference. Three other options are calculation of the power of the study to find the actual (observed) difference between groups statistically significant, calculation of the power of the study to find clinically meaningful differences, and calculation of the sample size that would be necessary to find the observed difference statistically significant.
Sample Size
Minimum Detectable Difference
The estimation of sample size requires considerable statistical skill as well as knowledge of the underlying basic assumptions being made by the investigator. It involves
The first option is used frequently and requires further explanation. Minimum detectable difference calculations estimate the amount of difference between groups that the
164
C HAP T E R
6 DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
investigator would be able to detect given the actual size of the sample and the variance. If the minimum detectable difference is much larger than would be considered clinically significant, the investigator may conclude that the investigation did not include a sufficient number of patients to detect a clinically meaningful difference as statistically significant. If the minimum detectable difference is smaller than the difference that is considered clinically significant, the test was adequately powered and the investigator may conclude that in fact there is no difference between the samples.
Confidence Intervals (Limits) on Statistical Tests of Inference CIs (described earlier) are frequently calculated for a statistical hypothesis test. They may be calculated for the t test, chi-square test, analysis of variance, regression, and most other tests of inference. A 95% CI is a range of values within which, the experimenter believes, there is a 95% probability that the true underlying (unknown) mean (or difference in means) lies. The most frequently reported CI is at the 95% level. The confidence limits are related to the P value. If one calculates the CI of a test that compares two means or difference in means, and zero is within in the range of the 95% CI, then the statistical test (P value) should not be significant, because there is some probability that the true difference between the two means is zero.
number of successes in N independent trials, given that the probability of success on anyone trial is y?" For example, one may ask, "What is the probability that a fair coin will produce 7 heads in 10 flips of the coin given that the probability of a "success" (heads) is 0.5 on each toss?" The binomial test yields an exact probability (P value) for the 7 successes as well as the probability of obtaining results even more extreme (Le., 8 of 10, 9 of 10, and 10 of 10 heads). The binomial test has limited applicability in describing the statistical probability that a therapy is beneficial, because the odds of success typically are not known. In pediatric rheumatology, the question may be, "What is the probability that 50 patients with ]RA treated with methotrexate will experience improvement as determined by a given index or measure?" The problem is that one is typically unsure of the exact probability of a success in a single independent trial. In some situations, the probability of success is arbitrarily given the value of .5 (Le., 50% chance), and the binomial test is done to either confirm or fail to confirm that level of probability of success.
Goodness-of-Fit Chi-Square Test
that a statistically significant difference may not necessarily indicate clinical significance. Particularly if the sample size is large, many tests may result in a statistically significant finding when in fact there is a relatively small degree of clinically significant difference between the two groups. Biologic significance, as compared with statistical significance, has come into the forefront with the testing of new immune response modifiers (biologic agents). For example, the sought-after effect of a drug (e.g., to reduce the number of CD4-positive T lymphocytes) may be significant yet produce no clinical effect. The solution to this problem is to include not only measures of surrogate markers of efficacy (e.g., laboratory values) but also outcomes that allow for interpretation of the clinical significance of such a biologic effect.
The goodness-o/fit chi-square test is related to Pearson's chi-square test (discussed later), in which observed proportions are compared with expected values. The goodness-of-fit chi-square test can be used to test the significance of a single proportion or of a theoretical model, such as the mode of inheritance of a gene. A reference population is often used to obtain the expected values. Suppose the frequency of an allele that is thought to produce risk for polyarticular ]RA is known to be 2 in 100 in the general population (ignoring population stratification for this example). However, the observed frequency of the allele in a sample of patients with polyarticular ]RA is found to be 10 in 100. Is this much deviation from the expected value significant? Briefly, one calculates how well the observed frequencies fit the model from the general population by determining the difference between each set of observed and expected values, squaring them, dividing by their respective expected values, and adding the results. In the example, the sum of (observed - expected)" + expected values = 00 - 2)2 + 2 [for those with the allele] + (90 - 98)2 + 98 [for those without the allele] = approximately 33. A chi-square table is consulted (as described in greater detail later), and the value of 33 is found to be highly statistically significant.
One-Sample Tests
One-Sample tTest
Statistical hypothesis testing may be completed on studies involving one or more groups. The most frequent approach to analYZing data from a clinical investigation that involves only one group is to compare that group with a known population or expected value.
When a statistical inference is desired on a single mean, the one-sample t test may be used. The test is similar to Student's t test for comparing two means, described later. Suppose one wishes to determine whether the mean height of 9- to 10-year-old girls with SLE is significantly less than that of the general population of 9- to lO-yearold girls. The critical ratio for t is set up as follows:
Statistical Versus Clinical Versus Biologic Significance An important concept that is frequently overlooked is
Binomial Test of Proportions Perhaps the most frequently used test for comparing one sample to a known population is the binomial test. This test asks the question, "What is the probability of x
t
=
(.~- /lo) / (s - --.In)
where x is the mean of the sample of patients with SLE, Jlo is the general population mean, s is the sample SD, and
C HAP T E R
6 DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
n is the sample size. One then consults a t table list of critical values and determines whether t is significant at some predetermined level in consideration of the degrees of freedom or independence (which here is n - 1).
Two-Sample Tests The two-sample test to be used is determined by the level of the data and by certain other assumptions, as defined later.
Chi-Square Test with One Degree of Freedom
It.
TABLE 6--6
the expected values in cells a, b, c, and dare 15, 73, 68, and 342, respectively. The next step is to find the difference between each pair of observed and expected values, square it to eliminate the negative sign, and divide by the expected value. To obtain the chi-square statistic, the results from each cell (ij) are summed:
X2
=
r [ (Ojj -
E/ / Eij
l
A short-cut formula for calculating the chi-square with 1 df is
X2
For categorical (nominal) data and ordinal data with very few ranks, the most frequently used hypothesis test is the Pearson chi-square (X2) test. This nonparametric statistical test of inference is for assessing the association between the two variables. It is most commonly performed on contingency tables such as a 2 x 2 cross-tabulation, which has one degree of freedom (l df). The numbers in the cells represent counts (frequencies), and each cell must he independent of all other cells (Le., an individual patient can contribute only once to the entire table). For example, to observe the association between HLA-DR8 and oligoarticular JRA, a 2 x 2 table can be constructed with HLA-DR8 positive or negative as the column headings and oligoarticular JRA present or absent (the latter representing control subjects) as the row headings (Table 6-6). The chi-square statistic is based on how much difference there is between "observed" and "expected" frequencies. The null hypothesis is that there is a random distribution of outcome in each "exposure" group-that is, that the columns are independent of the rows. Expected values are calculated from row and column totals. In Table 6-6,
=
(ad - bc)W.;- (a + b)(c + d)(a + c)(b + d)
where N is the total study population. The significance of the resulting chi-square statistic is determined from a table of critical values. There are several useful points to remember about the interpretation of the statistic. For chi-square with 1 df (i.e., 2 x 2 tables), the statistic becomes significant at the .05 level if the value is 3.841 or greater, and the larger the chi-square value, the more significant it is. Most tables of critical values report two-tailed probabilities; the P value is divided by 2 to find the one-tailed probabilities. Chi-square analysis with greater than 1 df (i.e., tables larger than 2 x 2) requires larger values to be significant.
Continuity Correction ofYates If the total N for a 2 x 2 chi-square table is less than about 40, the Yates continuity correction is used to compensate for deviations from the theoretical (smooth) probability distribution. The resulting chi-square value is smaller and the resulting statistical inference is more conservative. The technique involves subtracting 72 from the absolute value of each 11 E...II Mathematically, this is stated as follows:
°-
Example of the Calculation of Chi-Square with One Degree of Freedom'
As50dated Allele (HLA·DR 8) Condition (Ollgoartlcular Juvenile Rheumatoid Arthrltls)
Pnsent
Present
Cell. a
Absent (controls)
observed expected Cell c observed expected
Column Totals (c) of Obsen'ed Values
83
Absent
Row Totals (rJ of Observed Values
Cell b
= 42 = 15
observed = 46 expected = 73
88
Cell d
= 41 = 68
165
observed = 369 expected = 342 415
410
N= 498
Expected value in a cell is the row total x column total divided by the total N, or, (rt x c,)/N Expected value in cell a = (83 x 88) / 498 ;: 15 Expected value in cell b = (88 x 415) / 498;: 73 Expected value in cell c = (83 x 410) / 498 ;: 68 Expected value in cell d = (410 x 415) / 498 ;: 342 X2 = (42 - 15)2 + (46 -73)' + (41 - 68)2 + (369 - 342)' = 71 15 73 68 342 A chi-square value of 71 is significant at the .000001 level. The odds ratio (Le_, adlbc) = 8.2. Adapted from Nepom BS, Malhortrd U, Schwarz DA, et ai: HLA and T cell receptor polymorphisms in pauciarticu!ar-onset juvenile rheumatoid arthritis. Arthritis Rheum 34: 1260-1267. 1991. Copyright © 1991 Wiley-Liss, Inc. Reprinted by permission of Wiley-Liss. Inc.. a subsidiary of John Wiley & Sons, Inc.
166
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
risk, with the results from each stratum being weighted by the sample size of the stratum.
or
x = (lad 2
bcl - N12)2 + (a + b)(c + d)(a + c)(b + d)
Rsher's ExactTest Fisher's exact test is used as a replacement for the chi-
square test when the expected frequency of one or more cells is less than 5. This test is commonly used in studies in which one or more events are rare.
McNemar'sTest The chi-square test assumes independence of the cells, as noted earlier. However, experimental designs exist for observing categorical outcomes more than once in the same patient. McNemar's test (also known as the paired or matched chi-square) provides a way of testing the hypotheses in such designs. McNemar's chi-square statistic can be calculated with the following formula:
X2McNemar
=
(b -
C)2
I
(b
+ C)
or, with the continuity correction, X2CMcNemar =
(Ib - cl - 1)2 + (b + c)
An example is shown in Table 6-7. Two different concentrations of an analgesic lotion are given to 51 patients with arthritis. The null hypothesis is that the proportion of patients who receive relief from analgesic lotion 1 is the same as that from lotion 2. The results show that the null hypothesis cannot be rejected according to the McNemar test.
Mantel-Haenszel Chi-SquareTest This procedure is known as a stratified chi-square test and is frequently used to detect confounding variables. The procedure involves breaking the contingency table into various strata and then calculating an overall relative
II.
TABLE 6 7
EXdlllple 01 the MINellldr Chi Square fest
Fifty-one patients with arthritis pain in both hands are treated with two different lotions containing different concentrations of an analgesic. One lotion is placed on the left hand and the other on the right hand of each patient. Each patient rates each hand as experiencing pain relief or no relief The null hypothesis is that the same proportion of patients will receive relief from lotion 1 as from lotion 2.
lotion 1
lotion 2 Relief No Relief
Relief
No Relief
11 10
24
6
The McNemar chi-square test with the Yates correction is used: X'CMcNc,,", ( 16 - 10 I - I)' / 6 + 10 = 0.562. Interpret from regular x' table with one degree of freedom: 0.562 is less than 3.841. Therefore, do not reject the null hypothesis.
Common Errors with Chi-Square Tests Perhaps because of its frequent use, the chi-square test is often employed or interpreted inappropriately. Some of the more common mistakes include unnecessary conversion of continuous or ordinal level data to categorical data in order to use the chi-square test, nonindependence of the cells in the table (exception is when McNemar's chisquare test is being used), use of the chi-square rather than Fisher's exact test when expected cell frequencies are lower than 5, and confusion of statistical significance by chi-square values with clinical or biologic importance.
Student's tTest What the chi-square test is to categorical data, the t test is to continuous data. This test is used for comparing two sample means from either independent or matched samples. It asks the question, "Is the difference between ~ and Xc statistically significant, where ~ is the mean of the treated group and Xc is the mean of the control group?" This difference must be standardized, just as was done in the case of the standard normal variable procedure earlier, so that one set of t tables of critical values can be used. In the case of independent samples, the numerator of the t statistic is the difference in means and the denominator is the standard error of the difference. This is calculated as -.J s2 p Clint
+ lin)c
where s2 is the pooled variance, n t is the sample size of the treat~d group, and nc is the sample size of the control group. The degrees of freedom are calculated as n, + nc - 2, and a table of critical t values, for either one-tailed or two-tailed tests (depending on the hypothesis being examined), is consulted to determine whether the t statistic is significant. The matched t test is more efficient (Le., more powerful) than the independent test. The computer calculates the difference (retaining the sign) for each matched pair: d j = Xli - X 2i ' where i repres~nts each of n successive pairs. The mean difference (d) and the estimated standard error of the difference (s/-.Jn) are calculated. The null hypothesis. (0 = 0) is tested by calculating the t statistic: tpaired = d + (s/-.Jn). The degrees of freedom are equal to n - 1, where n is the number of pairs. A table of critical t values is consulted to determine whether the test statistic is significant.
Nonparametrlc t Tests The t tests described earlier are parametric tests. That is, they make assumptions about the underlying distributions, including normality and equality of variances between groups. The t test is a very robust test; it is still valid even if these assumptions are substantially violated. Modern data-analysis computer software programs provide relevant statistics and tests of underlying assumptions for application to a parametric test, and the resulting displays may warn the investigator that the results of the test
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
may not be valid. If the violations are severe, the investigator may transform the data using, for example, either natural logarithms (described earlier) or nonparametric tests. Nonparametric tests ignore the magnitude of differences between values taken on by the variables and work with ranks. No assumptions are made about the distribution of the data. In the case of the t test, either the MannWbitnry U test is used for independent data or the Wilcoxon signed-rank test is used for paired data.
K-Sample Tests Clinical investigations involving more than two samples (groups) require that modifications be made to the analysis plan to accommodate the need for multiple comparisons.
Chi-Square Test with More Than One Degree of Freedom When categorical data are analyzed, there may be more than two categories for one or both variables (i.e., the table may be larger than 2 x 2). However, if the chisquare test statistic is found to be significant in a table larger than 2 x 2, it is frequently difficult to determine which proportions were different. One must then attempt to either collapse the number of cells in the table or break the table up into several smaller tables. This may requite adjustment of the resulting P values because of multiple comparisons. The degrees of freedom of contingemcy tables larger than 2 x 2 are equal to the number of rows minus 1 plus the number of columns minus 1. For example, a 2 x 3 table has (2 - 1 + 3 - 1), or 3, degrees of freedom.
Analysis of Variance One-Way Analysis ofVariance The use of repeated t tests to detect differences among more than two means is considered unacceptable because the resulting P value does not accurately describe the chance one has taken of committing a type I error. The one-way analysis of variance (ANOVA) is used for the purpose of comparing more than two sample means. Simply stated, ANOVA divides the total variance among all subjects into two portions, the amount of variance that is a result of the difference between the groups of subjects, and the amount of variance that results from differences within each group. The ratio of the amount between and the amount within each group is known as the F ratio. The corresponding test of significance is known as the F test. If statistical significance is achieved, the investigator must go one step further. The significance may have arisen because just two means were different from one another, or perhaps all the means were different from one another. To determine exactly which means were different, tests must accommodate the fact that multiple comparisons are being made. This is determined by applying a multiple comparison. Commonly used multiple comparison tests include Tukry's honest significant difference test, the Newman-Keuls test, Scheffe's multiple contrasts test, and, if one wishes to make multiple comparisons to
167
only one group (e.g., control group, standard of care), Dunnett's test.
Multi-Way ANOVA ANOVA procedures have an additional capability that can increase the efficiency of analyses when one wishes to compare the influence of two or more independent variables on one DV simultaneously. Suppose one wished to test the effects of methotrexate and a physical therapy (PT) program on the disease status of a group of patients with polyarticular JRA. One could carry out two separate studies, conduct t tests for treatment effects on the methotrexate-treated patients and the PT-treated patients, and compare each with placebo or no PT. This approach requires substantial numbers of patients to meet samplesize requirements for each study. However, a two-way ANOVAfactorial design could make much more efficient use of the available subjects and provide information about the interaction (effect modification) between methotrexate and PT. In this situation, patients could be randomized to both treatments, yielding four groups (methotrexate alone, PT alone, both methotrexate and PT, and neither treatment). In addition to providing information about the effect of each treatment alone (j.e., the two main effects), a two-way ANOVA factorial design examines the effect of the interaction between the two treatments. ANOVA techniques can be extended to threeway, four-way, and beyond, provided the sample size is large enough.
Nonparametric ANOVA ANOVA procedures discussed to this point are parametric tests and, as such, make various assumptions about the underlying distribution. If these assumptions are substantially violated, the nonparametric equivalent of ANOVA, the Kruskal-Wallis test, can be used. This test is subject to the same sample-size limitations as the chisquare test. If the sample size in any group is less than 5, one must use Fisher's exact test and exact probabilities, as described earlier.
Correlation and Regression One of the most important measures of statistical correlation is the Pearson product-moment correlation. This statistic is appropriate for estimating the relationship between two variables, x and y, both of which are measured along a continuous scale. Correlation is a two-way model that does not require assumptions of causality. The correlation (r) can vary between -1 and +1. The magnitude of the correlation demonstrates the strength of the relationship between the two variables. The larger the absolute value of the correlation, the more strongly associated are x and y. In the extreme, where r = +1 or -1, all the data values fall perfectly on a straight line. As r approaches zero, the data demonstrate greater scatter about a best-fit line for their relationship. The sign of the correlation indicates the direction of the relationship. A positive sign means that the two variables are directly related (j.e., they tend to increase or decrease together).
168
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
A negative sign for r indicates that the two variables are inversely related (Le., the value of one tends to decrease as that of the other increases). Correlation assumes that the joint distribution of x and y is bivariate normal. That is, y is normally distributed at all values of x, and vice versa. If this assumption is violated substantially, the nonparametric Spearman rank correlation, which yields a Spearman's rho (r j, is used. Because Spearman rank correlation deals with ranks, it can be used with continuous variables that violate assumptions and with ordinal data. Regression is a one-way model in which predictor or explanatory independent (x) variables are thought to affect the dependent outcome (y) variables, but not vice versa. In simple regression models (Le., models that include only a single predictor), as well as in multiple regression models, the direction of the effects must be prespecified. The simple linear regression equation is y = a + bx, where a is the intercept and b is the coefficient. By using various values of x in the equation, the predicted value of y for a given x can be determined. Simple regression models serve as building blocks for the larger, more complex, and more realistic models, including polynomial regression models and structural equation models.
Multiple linear Regression The technique whereby a multitude of independent variables (x j' x2' x3' etc.) can be simultaneously investigated for their influence on a linear related DV (y) is known as multiple linear regression. The method models the DV as a linear function of all the (k) independent variables. That is, y
=
a + bjx j + b 2x 2 + b3x 3 + ... + bkx k
This method is particularly helpful in evaluating extraneous variables as possible confounders of the linear relationship between two continuous variables. In other words, linear regression permits the investigator to assess the separate unconfounded effects of several independent variables on a single DV The Xi terms can be continuous or categorical variables. The b i terms are the regression coefficients. Each b i is "corrected" simultaneously for the linear relationship between its associated Xi and all the other xis, as well as for the linear relationship between the other Xi'S and y, An overall flo value is calculated for the model. It represents the percentage of the total variance of y that is accounted for by the linear relationship with all the Xi'S. A common mistake is to refer to multiple linear regression as a multivariate technique; technically it is not, because it deals with multiple independent variables rather than multiple DVs.
Multiple Logistic Regression Multiple logistic regression is distinguished from multiple linear regression in that the outcome variable (DV) is dichotomous (e.g., diseased or not diseased). Its aim is the same as that of all model-building techniques: to derive the best-fitting, most parsimonious (smallest or most efficient), and biologically reasonable model to
describe the relationship between an outcome (DV) and a set of predictors (independent variables), Here, the independent variables are called covariates. (Multiple logistic regression is not technically a multivariate technique, because it deals with only one DV) Importantly, in multiple logistic regression, the predictor variables may be of any data level (categorical, ordinal, or continuous). A major use of this technique is to examine a series of predictor variables to determine those that best predict a certain outcome. A pediatric rheumatology example of the use of this technique can be found in the paper by Ruperto and associates,63 in which predictor variables that are measurable during the very early stages of JRA (e.g" number of active joints during the first 6 months of illness, erythrocyte sedimentation rate [ESR]) were tested to determine their relative predictive ability for either a favorable or a less favorable outcome (Le., a dichotomous DV) at least 5 years later. An excellent reference guide to this technique is provided by Hosmer and Lemeshow. 107
Analysis of Covariance The analysis of covariance (ANCaVA) combines the principles of ANaVA with those of regression. A chief advantage of this technique is that, unlike ANaVA, the independent variables can be of any data level. ANcaVA is often used to adjust for initial (baseline) differences between or among groups. In other words, one of its chief purposes is to eliminate systematic bias. For example, suppose two groups of patients had unequal numbers of swollen joints at baseline (even though the study may have been randomized). The initial number of swollen joints is then used as the covariate, ANcaVA adjusts the post-treatment means of the groups to what they would have been if all groups had started out equally on the covariate, The other purpose of ANCaVA is to reduce the withingroup (or error) variances, thus making the test more efficient (powerful). For example, suppose a clinical trial investigates the effect on the ESR of a biologic agent and an active comparator. Subjects are randomly assigned to receive one or the other treatment, and the change in ESR is observed. Within each treatment group, there is considerable variation in ESR, reflecting individual differences among patients in the degree of active inflammation. In other words, ESR and active inflammation are covarying (covariates). If one could statistically remove this part of the within-group variability by allowing the degree of inflammation to be the covariate in the analysis, a smaller error term would result, and the test would gain power. ANcaVA provides a method to do this. In summary, ANcaVA provides a method for adjusting for differences at baseline between groups and reduces the amount of variation within groups that is caused by covariates, thus increasing power and capability to detect differences.
Survival Analysis Survival (life table) analysis was developed primarily for the study of how long a particular cohort of subjects survives. The term survival data is now used in a broader
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
sense for data that involves time to a certain event, such as time to failure of a drug or time before remission. 55 There are two basic types of life table analysis: the fixedinterval (actuariaf) model and the Kaplan-Meier survival analysis. The latter is used much more frequently in medicine than the former. In actuarial analysis, the lengths of each interval shown on the x-axis are all equal (e.g., 1 year). This is the technique used by life insurance companies to estimate the probability of a person's surviving to a certain age. In the Kaplan-Meier approach, the end of an interval is demarcated by an event. The horizontal components of the lines are not equal, as they are in the actuarial technique. An example of the actuarial method in pediatric rheumatology can be found in a study by Giannini and colleagues lO8 of the time to occurrence of eye disease among patients with certain major histocompatibility complex alleles. An example of the KaplanMeier approach can be found in the study by Lovell and associates,77 in which time to failure in subjects given placebo was compared with time to failure in those given etanercept. Methods exist for comparing the difference of the life table graphs; the most frequently used is the generalized Wilcoxon test. Figure 6-8 is a graphic representation of a comparison between the characteristic lines of an actuarial model and a Kaplan-Meier analysis. An outstanding reference for survival analysis techniques is that of Lee and Wang. 109
169
or within raters, indicates that the values for the measure are nonreliable or nonreproducible. Of course, this has dire consequences for the interpretation of the results and for statistical interpretation. Various tests exist for expressing the degree of agreement between and within raters. The most frequently used test to express rater agreement when the outcome is dichotomous is the kappa test ratio (known also as Cohen's kappa, or K). The K statistic is given by the following formula: K = (observed agreement - agreement expected by chance) -:- (maximum possible agreement - agreement expected by chance). K scores range from -1, indicating perfect disagreement, to +1, indicating perfect agreement; a score of 0 indicates the agreement expected by chance. K scores are often expressed as percentages: less than 20% considered negligible agreement; 20% to 40%, minimal; 40% to 60%, fair; 60% to 80%, good, and greater than 80% excellent. lJo Data with three or more categories (Le., ordinal data) require the use of the more complex weighted kappa test. Kendall's W, or coefficient of concordance, can also be used. W values range from 0, which indicates poor agreement, to +1, which indicates perfect agreement among all raters. W cannot be negative, because not all the raters can disagree completely if there are more than two.
Intradass Correlation
Measures of Agreement Among and Within Raters It is often necessary to express in statistical terms how
well various raters agree with one another (inter-rater agreement) or with themselves (intra-rater agreement). These are commonly referred to as measures of reliability or reproducibility. Lack of agreement, either among
100
Cl
80
.5 .~ ;:, III
~Ql
If more than two raters are to be compared for reliability, the intraclass correlation is more appropriate than conducting repeated two-way comparisons between pairs of raters (there is a correction for correlation between raters that becomes apparent when the range of measurement is large). It evaluates the level of agreement among all raters, and the measures (scores) must be parametric in nature. The data are placed in a matrix in which the columns are the raters and the rows are the number of subjects (or the number of variables) being measured. The coefficient represents the amount of agreement: 1 is perfect agreement, and 0 is no agreement. Analysis of variance on the matrix produces an F value (described previously) and tells the investigator whether the raters are significantly different from one another. IJJ
60
Multivariate Analyses
:=
[ '0 40
Actuarial method
1:
~
Ql Q.
20
Kaplan-Meier method
o
2
3
4
5
6
Years since beginning treatment
• FItIIIIIt 6-8 Comparison of Kaplan-Meier and actuarial survival curves, showing atheoretical example of the percentage of patients surviving after 0 to 6 years of treatment. (Modified from Kramer MS: Clinical Epidemiology and Biostatistics: APrimer for Clinical Investigators and Decision-Makers. BerlinHeidelberg~NewYork, Springer-Verlag, 1988, p. 246, © 1988 Springer-Verlag.)
This section provides a basic concept of multivariate statistics and an overview of the more popular and increasingly used multivariate tests. Interested readers are referred to the text by Stevens JJ2 for in-depth discussions of the concepts and tests presented here. Multivariate statistics are appropriate when the experimental design simultaneously investigates two DVssuch as bone mineral density and bone mineral content during a trial of calcium supplementation in prepubescent girls with polyarticular JRA. Testing of these two DVs in two trial groups (calcium supplementation and no supplementation) by, for example, the t test would not be appropriate because the tests are not independent of one another; that is, the two DVs are highly correlated
170
c
HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
with each other (and if one is known, the other can be guessed). Furthermore, just as the interaction among independent variables in a factorial ANOVA provides new information, so too does the use of multivariate tests that consider more than one DV simultaneously. Multivariate techniques may use nominal, ordinal, or continuous data.
dicting group membership. Again, as in stepwise regression, computer programs can add and subtract variables from the equation (based on significance of Wilk's lambda test) until the combination that best discriminates the groups is arrived at.
Hotelllng's T2 Test
Factor analysis is used for data exploration to reveal patterns of interrelationships among variables that are not readily apparent, for confirmation of hypotheses, and for reducing the number of variables to a manageable level. In situations involving many different observations concerning the same patient or groups of patients, factor analysis can be used to determine whether it is possible that some of these observations are a result of just a few underlying factors. That is, the correlation among many DVs may be explained by some underlying factor or factors. For example, the fact that a young boy is experiencing swollen joints, a rash, intermittent fevers, and a drop in hemoglobin with an increase in white blood cell count is a result of underlying factors: he is experiencing the onset of systemic JRA, and pathologic processes are taking place in his hematopoietic system. When groups of patients are studied, the symptoms tend to "load" on the underlying factors differentially. Factor loading is expressed in a factor loading matrix, in which each row of the matrix is a variable and each column is a factor. Such a matrix examines how highly each variable correlates with, or loads on, each factor. Each variable may load onto one or more variables. Next, one must decide which factors are most important to keep and which can be discarded as not contributing enough to the explanation of the variables. This is done by calculating an eigenvalue, which is the amount of variance in the data that is explained by a particular factor. The procedure to this point is called principal component analysis. Additional steps in factor analysis include rotation of axes to determine which are general factors (most variables load significantly on them) and which are bipolarfactors (some variables load positively and some load negatively on them). Factorial complexity is determined by observing how many variables load significantly onto two or more factors.
Just as the t test is used when one wishes to compare one DV in two different groups, Hotelling's T2 test is used when there are two groups with multiple DVs. The process involves the comparisons of, for example, the vectors of the means in the treated and in the control group. A centroid for the groups is calculated, which is the point at which the mean values of the two DVs intersect for the control group when they are plotted on an x-y graph. The SD and the amount of correlation between the variables are used to calculate whether the T2 statistic is significant. Multivariate significance implies that there is a linear combination of the DVs that is significantly separating the groups. However, significance of the T2 statistic may arise from one or more of the DVs under consideration. Discriminant function analysis (described later) is used to determine which variables are contributing the most to the significant finding.
Multivariate Analysis ofVariance Just as ANOVA was used for the comparison of the means of more than two means, so too is the multivariate analysis of variance (MANOVA), rather than Hoteiling's T2, used in situations with more than two groups and multiple DVs. Its use instead of separate univariate ANOVAs is related to accounting for intercorrelations among the DVs. Vectors of means are calculated, and the centroid for each group is compared with the grand centroid (similar to the comparison of variance within groups versus variance between groups tested in univariate ANOVA). Similarly, the within-group variability must be computed for each of the DVs. There are numerous ways to calculate the significance of MANOVA, the most common of which is Wilk's lambda. As with Hoteiling's T2, significance can arise from one or several of the variables, and discriminant function analysis is needed to sort it out.
Discriminant Function Analysis In situations in which many DVs exist, statistically significant differences among the groups can arise from the effect of one, several, or all of the variables. Discriminant function analysis identifies the variables that are most important in accounting for the observed differences. In addition, discriminant function analysis allows determination of those groups that differ more in some DVs than in others. The process derives an equation (function) that best discriminates among the groups. The equation is similar to a regression equation and is the first variable multiplied by its weight, plus the second variable multiplied by its weight, and so on. In this case, rather than predicting a value of a DV, as in regression, one is pre-
Factor Analysis
Multivariate Analysis of Covariance The multivariate analysis of covariance (MANCOVA) is an extension of univariate ANCOVA in which group means at follow-up are adjusted for differences at baseline and within-group variance is reduced by removing variation caused by covariates. The objective of MANCOVA is to determine whether several groups differ on a set of DVs after the follow-up means have been adjusted for any initial differences on the covariates at baseline.
Canonical Correlation Canonical correlation is a multivariate procedure, an extension of multiple regression, that is used to examine the nature of the association or interrelation between two sets of variables. Canonical correlation breaks down the
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
complex association into additive pieces to determine the number and nature of independent relationships existing between two sets of variables.
Sample Size for Multivariate Tests A rule of thumb is that there should be at least 10 subjects for each DV investigated in the study. A study in which the subject/variable ratio is smaller is likely to be unreliable.
JUDGING THE QUALITY OF A REPORT OF A CUMICAL INVESTIGATION The most helpful and up-to-date series of guides to the reader of clinical reports was published in the journal of the American Medical Association in 1993-2000. This "Users I Guides to the Medical Literature" series8--40 provides logical checklists of questions for readers attempting to weigh the evidence from many different types of clinical studies. In brief, each Guide asks three basic questions: • Are. the results of the study valid? • What were the results? • Will the results help me in caring for my patients? Practical examples from everyday clinical situations are then used to illustrate how one determines the answer and eventually weighs the evidence from the report. Because not all epidemiologic studies deal with patients, judgin, the quality of an epidemiologic investigation requires an approach that is similar to but separate from judging clinical investigations.
Juclgl'" the Evidence from a Olnlcal Trial and the CONSORT Statement Although the Users' Guides just cited contain information for judging clinical trials, more detailed guides are available. In 1995, a group of medical journal editors, clinical epidemiologists, and statisticians developed a consensus statement about how randomized controlled trials should be reported-the Consolidated Standards of Reporting Trials (CONSORD statement. Since the previous edition of this text, the statement has been revised,113 and a study has be¢n done to determine its impact on reporting of trials,u4 The statement contains a checklist of 21 items that deal chiefly with methods, results, and discussion. It identifies key pieces of information necessary to evaluate the internal and external validity of a report. A flow diagram is also recommended; using a two-group, parallel design, randomized, controlled trial as an example, it provides a graphic display of allocation and status of patients throughout the trial. Some modification of the statement's recommendations is usually necessary because of differences in study design. Overall, however, it provides an outstanding template on which to formulate or judge the report of a clinical trial. Guidelines for determining authorship of articles that are generally accepted by editors of medical journals have now been published. ll5
171
REFERENCES 1. Evidence-Based Medicine Working Group: Evidence-based medicine: a new approach to teaching tbe practice of medicine. JAMA 268: 2420-2425, 1992. 2. Brunner HI, Giannini EH: Evidence-based medicine in pediatric rheumatology. Clin Exp Rheumatol 18: 407-414, 2000. 3. Levin A: The Cochrane Collaboration. Ann Intern Med 135: 309-312, 2001. 4. Laupacis A: The Cochrane Collaboration: how is it progressing? Stat Med 21: 2815-2822, 2002. 5. Waters E: Evidence-based paediatric practice and its relation to the Cochrane Collaboration. J Paediatr Child Health 39: 137-138, 2003. 6. Churchill R: Systematic reviews and the Cochrane Collaboration: improving the evidence-base for all psychiatric treatments. Epidemiol Psichiatr Soc 12: 137-145,2003. 7. Clarke M: The Cochrane Collaboration: providing and obtaining the best evidence about the effects of healtb care. Eval Health Prof 25: 8-11, 2002. 8. Guyatt GH, Rennie D: Users' guides to the medical literature. JAMA 270: 2096-2097, 1993. 9. Oxman AD, Sackett DL, Guyatt GH: Users' guides to the medical literature. I: How to get started. The Evidence-Based Medicine Working Group. JAMA 270: 2093-2095, 1993. 10. Guyatt GH, Sackett DL, Cook DJ: Users' guides to the medical literature, II: How to use an article about therapy or prevention. A: Are the results of the study valid? Evidence-Based Medicine Working Group. JAMA 270: 2598-2601, 1993. 11. Guyatt GH, Sackett DL, Cook DJ: Users' guides to the medical literature. II: How to use an article about therapy or prevention. B: What were the results and will they help me in caring for my patient.? Evidence-Based Medicine Working Group. JAMA 271: 59-63, 1994. 12. Jaeschke R, Guyatt G, Sackett DL: Users' guides to the medical literature. III: How to use an article about a diagnostic test. A: Are the results of the study valid? Evidence-Based Medicine Working Group. JAMA 271: 389-391, 1994. 13. Jaeschke R, Guyatt GH, Sackett DL: Users' guides to the medical literature. III: How to use an article about a diagnostic test. B: What are the results and will they help me in caring for my patient.? The Evidence-Based Medicine Working Group. JAMA 271: 703-707, 1994. 14. Levine M, Walter S, Lee H, et al: Users' guides to the medical literature. IV: How to use an article about harm. Evidence-Based Medicine Working Group. JAMA 271: 1615-1619, 1994. 15. Guyalt GH, Sackett DL, Sinclair JC, et al: Users' guides to the medical literature. IX: A method for grading health care recommendations. EvidenceBased Medicine Working Group. JAMA 274: 1800-1804, 1995. 16. Laupacis A, Wells G, Richardson WS, Tugwell P: Users' guides to the medical literature. V: How to use an article about prognosis. Evidence-Based Medicine Working Group. JAMA 272: 234-237, 1994. 17. Oxman AD, Cook DJ, Guyatt GH: Users' guides to the medical literature. VI: How to use an overview. Evidence-Based Medicine Working Group. JAMA 272: 1367-1371, 1994. 18. Richardson WS, Detsky AS: Users' guides to the medical literature. VII: How to use a clinical decision analysis. A: Are the results of the study valid? Evidence-Based Medicine Working Group. JAMA 273: 1292-1295, 1995. 19. Richardson WS, Detsky AS: Users' guides to the medical literature. VU: How to use a clinical decision analysis. B: What are the results and wiU they help me in caring for my patients? Evidence Based Medicine Working Group. JAMA 273: 1610-1613, 1995. 20. Hayward RS, Wilson MG, Tunis SR, et al: Users' guides to the medical literature. YIIl: How to use clinical prattice guidelines. A: Are the recommendations valid? The Evidence-Based Medicine Working Group. JAMA 274: 570-574, 1995, 21. Wilson MC, Hayward RS, Tunis SR, et al: Users' gUides to the medical literature. VII!: How to use clinical practice guidelines, B: What are the recommendations and will they help you in caring for your patients? The Evidence-Based Medicine Working Group. JAMA 274: 1630-1632, 1995. 22. Naylor CD, Guyalt GH: Users' guides to the medical literature. X: How to use an article reporting variations in the outcomes of health services. The Evidence-Based Medicine Working Group. JAMA 275: 554-558, 1996. 23. Naylor CD, Guyatl GH: Users' guides to the medical literature. Xl: How to use an article about a clinical utilization review, Evidence-Based Medicine Working Group. JAMA 275: 1435-1439, 1996. 24. Guyatt GH, Naylor CD, Juniper E, et al: Users' guides to the medical literature. XlI: How to use articles about health-related quality of life. EvidenceBased Medicine Working Group. JAMA 277: 1232-1237, 1997. 25. Drummond MF, Richardson WS, O'Brien BJ, et al: Users' guides to the medical literature. XlIl: How to use an article on economic analysis of clinical practice. A: Are the results of the study valid? Evidence-Based Medicine Working Group. JAMA 277: 1552-1557, 1997. 26. O'Brien BJ, Heyland D, Richardson WS, et al: Users' guides to the medical literature. XlIl: How to use an article on economic analysis of clinical practice. B: What are the results and will they help me in caring for my patients? Evidence-Based Medicine Working Group. .IAMA 277: 1802-1806, 1997. 27. Dans AL, Dans LF, Guyalt GH, Richard.on S: Users' guides to the medical literature. XlV: How to decide on the applicability of clinical trial results to your patient. Evidence-Based Medicine Working Group. JAMA 279: 545-549, 1998.
172
C HAP T E R
6
DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS
28. McAlister FA, Laupacis A, Wells GA, Sacken DL: Users' guides to the medical literature. XIX: Applying clinical trial results. B: Guidelines for determining whether a drug is exerting (more than) a class effect. Evidence-Based Medicine Working Group. JAMA 282: 1371-1377, 1999. 29. Bucher HC, Guyatt GH, Cook OJ, et al: Users' guides to the medical literature. XIX: Applying clinical trial results. A: How to use an article measuring the effect of an intervention on surrogate end points. Evidence-Based Medicine Working Group. JAMA 282: 771-778, 1999. 30. Richardson WS, Wilson MC, Guyatt GH, et al: Users' guides to the medical literature. XV: How to use an article about disease probability for differential diagnosis. EVidence-Based Medicine Working Group. JAMA 281: 1214-1219, 1999. 31. Guyatt GH, Sinclair J, Cook OJ, Glasziou P: Users' gUides to the medical literalUre. XVI: How to use a treatment recommendation. Evidence-Based Medicine Working Group and the Cochrane Applicability Methods Working Group. JAMA 281: 1836-1843, 1999. 32. Barratt A, Irwig L, Glasziou P, et al: Users' guides to the medical literature. XVU: How to use gUidelines and recommendations about screening. Evidence-Based Medicine Working Group. JAMA 281: 2029--2034, 1999. 33. Randolph AG, Haynes RB, Wyatt JC, et al: Users' guides to the medical literature. XVIII: How to use an article evaluating the clinical impact of a computer-based clinical decision support system. JAMA 282: 67-74, 1999. 34. McAlister FA, Straus SE, Guyatt GH, Haynes RB: Users' guides to the medical literature. XX: Integrating research evidence with the care of the inclividual patient. Evidence-Based Medicine Working Group, JAMA 283: 2829--2836, 2000. 35. Hunt DL, Jaescbke R, McKibbon KA: Users' guides to the meclicalliterature. XXI: Using electronic health information resources in evidence-based practice, Evidence-Based Medicine Working Group. JAMA 283:1875-1879, 2000. 36, McGinn TG, Guyal! GH, Wyer PC, et al: Users' guides to the medical literature. XXII: How to use articles about clinical decision rules. Evidence-Based Medicine Working Group. JAMA 284: 79--84, 2000. 37, Giacomini MK, Cook OJ: Users' guides to the medical literature. XXIII: Qualitative research in health care, A: Are the results of the study valid? Evidence-Based Medicine Working Group. JAMA 284: 357-362, 2000. 38. Giacomini MK, Cook OJ: Users' guides to the medical literature, XXIII: Qualitative research in health care, B: What are the results and how do they help me care for my patients? Evidence-Based Medicine Working Group. JAMA 284: 478-482, 2000. 39. Richardson WS, Wilson MC, Williams JW Jr, et al: Users' gUides to the medical literature, XXIV: How to use an article on the clinical manifestations of disease. Evidence-Based Medicine Working Group. JAMA 284: 869--875, 2000. 40. Guyatt GH, Haynes RB, Jaeschke RZ, et al: Users' guides to the medical literature. XXV: Evidence-based medicine: principles for applying the Users' Guides to patient care. Evidence-Based Medicine Working Group. JAMA 284: 1290-1296,2000. 41. Sung NS, Crowley WF Jr, Genel M, et al: Central challenges lilCing the national clinical research enterprise. JAMA 289: 1278--1287, 2003, 42. Bloom FE: Presidential address. Science as a way of life: perpleXities of a physician-scientist. Science 300: 1680-1685, 2003. 43. Petiti OM: Meta-analysis, Decision Analysis, and Cost Effectiveness Analysis. Oxford, UK, Oxford University Press, 1994. 44. Moses LE: Statistical concepts fundamental to investigations. N Engl J Med 312: 89Q-897, 1985. 45. Hill AB: The environment and disease: association or causation? Proc R Soc Med 58: 295-300, 1965. 46. Noll RB, Kozlowski K, Gerhardt C, et al: Social, emotional, and behavioral functioning of children with juvenile rheumatoid arthritis. Arthritis Rheum 43: 1387-1396, 2000. 47. Noll RB, Gartstein MA, Vannatta K, et al: Social, emotional, and behavioral functioning of children with cancer, Pediatrics 103: 71-78, 1999. 48. Inman RD, Johnston ME, Hodge M, et al: Postdysenteric reactive arthritis: a clinical and immunogenetic study follOWing an outbreak of salmonellosis. Arthritis Rheum 31: 1377-1383, 1988. 49. Arguedas 0, Fasth A, Andersson-Gare B: A prospective population based study on outcome of juvenile chronic arthritis in Costa Rica. J Rheumatol 29: 174-183, 2002, 50. Hashkes PJ, Balistreri WF, Bove KE, et al: The long-term effect of methotrexate therapy on the liver in patients with juvenile rheumatoid arthritis. Arthritis Rheum 40: 2226-2234, 1997. 51. Ravelli A, Manzoni SM, Viola S, et al: Factors affecting the efficacy of intraarticular corticosteroid injection of knees in juvenile idiopathic arthritis. J Rheumatol 28: 2100-2102, 2001. 52. Ravelli A, Martini A: Early predictors of outcome in juvenile idiopathic arthritis. Clin Exp Rheumatol 21 (5 Suppl 31): S89-S93, 2003. 53, Brunner HI, Lovell OJ, Finck BK, Giannini EH: Preliminary definition of disease flare in juvenile rheumatOid arthritis. J Rheumatol 29: 1058--1064, 2002. 54. Rider LG, Giannini EH, Harris-Love M, et al: Defining clinical improvement in adult and juvenile myositis, J Rheumatol 30: 60}-{i17, 2003. 55, Ruperto N, Ravelli A, Murray KJ, et al: Preliminary core scts of measures for disease activity and damage assessment in juvenile systemic lupus erythematosus and juvenile dermatomyositis, Rheumatology (Oxf) 42: 1452-1459, 2003. 56. Wallace CA, Ruperto N, Giannini EH; Childhood Arthritis and Rheumatology Research Alliance; Pediatric Rheumatology International Trials Organisation;
Pediatric Rheumatology Collaborative Study Group: Preliminary criteria for clinical remission for select categories of juvenile idiopathic arthritis. J Rheumatol 11: 2290-2294, 2004, 57, Gare BA, Fasth A: The natural history of juvenile chronic arthritis: a popUlation based cohort study. 1I: Outcome. J Rheumatol 22: 308--319, 1995. 58. Flato B, Lien G, Smerdel A, et al: Prognostic factors in juvenile rheumatOid arthritis: a case-control study revealing early predictors and outcome after 14.9 years, J Rheumatol 30: 386-393, 2003, 59. Oen K: Long-term outcomes and predictors of outcomes for patients with juvenile idiopathic arthritis, Best Pract Res Clin Rheumatol 16: 347-360, 2002. 60, Oen K, Malleson PN, Cabral DA, et al: Disease course and outcome of juvenile rheumatoid arthritis in a multicenter cohort. J Rheumatol 29: 1989-1999, 2002. 61. Oen K, Malleson PN, Cabral DA, et al: Early predictors of longterm outcome in patients with juvenile rheumatoid arthritis: subset-specific correlations. J Rheumatol 30: 585-593, 2003. 62. Ruperto N, Levinson JE, Ravelli A, et al: Long-term health outcomes and quality of life in American and Italian inception cohorts of patients with juvenile rheumatoid arthritis. I: Outcome status. J Rheumatol 24: 94'i-951, 1997. 63, Ruperto N, Ravelli A, Levinson JE, et al: Long-term health outcomes and quality of life in American and Italian inception cohorts of patients with juvenile rheumatoid arthritis. II: Early predictors of outcome. J Rheumatol 2-1: 952-958, 1997. 64. Murray K, Thompson SO, Glass ON: PathogeneSiS of juvenile chronic arthritis: genetic and environmental factors. Arch Dis Child 77: 530-534, 1997. 65. Glass ON, Giannini EH: Juvenile rheumatoid arthritis as a complex genetic trait. Arthritis Rheum 42: 2261-2268, 1999. 66. Jarvis IN, Dozmorov I, Jiang K, et al: Novel approaches to gene expression analysis of active polyarticular juvenile rheumatoid arthritis. Arthritis Res Ther 6: RI5-R32, 2004. 67, Prahalad S, Ryan MH, Shear ES, et al: Juvenile rheumatoid arthritis: linkage to HLA demonstrated by allele sharing in affected sibpairs. Arthritis Rheum 43: 2335-2338, 2000. 6B. Savolainen A, Saila H, Kotaniemi K, et al: Magnitude of the genetic component in juvenile idiopathic arthritis. Ann Rheum Dis 59(2): 1001, 2000. 69. Saila HM, Savolainen HA, Kotaniemi KM, et al: Juvenile idiopathic arthritis in multicase families. Clin Exp Rheumatol 19: 218--220, 2001. 70. Forre 0, Smerdel A: Genetic epidemiology of juvenile idiopathiC arthritis. Scand J Rheumatol 31: 123-128,2002. 71. Thomson W, Donn R: Juvenile idiopathic arthritis genetics: what's new? What's next? Arthritis Res 4: 302-306, 2002, 72. Rosen P, Moroldo MB, tovell OJ, et al: Discordant phenotypes for methotrexate response in juvenile rheumatoid arthritis. Arthritis Rheum 46 (Supp)): S470, 2002. 73. Katz PP: Introduction to special patient outcomes in rheumatology issue of Arthritis Care and Research, Arthritis Care Res 49 (5 Supp)): SI-SI4, 2003. 74. Hull RG: Guidelines for management of childhood arthritis. Rheumatology (OxD 40: 1309-1312, 2001. 75, Temple R: Current definitions of phases of investigation and the role of the FDA in the conduct of clinical trials, Am Heart J 139: SI33-S135, 2000, 76. Weiner DL, Yuh L: Biovavailability studies, In Buncller CR, Tsay J-Y (eds): Statistics in the Pharmaceutical Industry. New York, Marcel Dekker, 1994. pp 215-245. 77. Lovell OJ, Giannini EH, Reiff A, et al: Etanercept in children with polyarticular juvenile rheumatoid arthritis. Pediatric Rheumatolob'Y Collaborative Study Group, N Engl J Med 342: 763-769, 2000. 78. Honkanen YE, Siegel AF, Szalai JP, et al: A three-stage clinical trial design for rare disorders. Stat Med 20: 3009-3021, 2001. 79. Giannini EH: Can non-fundable trials be conducted anyway? The case for open, randomised, actively controlled trials in rheumatology, Ann Rheum Dis 57: 128--130, 1998, 80. Shaikov AV, Maximov AA, Speransky AI, et al: Repetitive use of pulse therapy with methylprednisolone and cyclophosphamide in addition to oml methotrexate in children with systemic juvenile rheumatoid arthritis: prel iminary results of a longterm study. J Rheumatol 19: 612-616, 1992. 81. Ruperto N, Murray Kj, Gerloni V, et al: A randomized trial of parente",1 methotrexate comparing an intermediate dose with a higher dose in children with juvenile idiopathic arthritis who failed to respond to standard doses of methotrexate. Althritis Rheum 50: 2191-2201. 2004. 82. Giannini EH: The N of 1 trials design in the rheumatic diseases. Arthritis Care Res 1: 109-115, 1998. 83. Temple R, Ellenberg SS: Placebo-controlled trials and active-control trials in the evaluation of new treatments. Part 1: Ethical and scientific issues. Ann Intern Med 133: 455-463, 2000, 84. Vickers AJ, de Craen A]: Why use placebos in clinical trials? A narrative review of the methodological literature, J C1in Epidemiol 53: 157-161, 2000. 85. Emanuel EJ, Miller FG: The ethics of placebo-controlled trials: a mitkUe ground. N Engl J Med 345: 915-919, 2001. 86. Freedman B, Glass KC, Weijer C: Placebo orthodoxy in clinical research. II: Ethical, legal, and regulatory myths. J Law Med Ethics 24: 252-259, 1996. 87. Freedman B: Placebo-controlled trials and the logic of clinical purpose. lim 12: 1-6, 1990. 88. Freedman B, Weijer C. Glass KC: Placebo orthodoxy in clinical research. I: Empirical and methodological myths. J Law Med Ethics 24: 243-251. 1996,
C HAP T E R 6 DESIGN, MEASUREMENT, AND ANALYSIS OF CLINICAL INVESTIGATIONS 89. Beaton DE, Bombardier C, Katz IN, Wright JG: A taxonomy for responsivene"s, J Clin Epidemiol 54: 1204--1217, 2001. 90. Fortin PR, Stucki G, Katz IN: Measuring relevant change: an emerging challenge in rheumatologic clinical trials, Arthritis Rheum 38: 1027-1030, 1995. 91. Felson DT, Anderson JJ, Boers M, et al: American College of Rheumatology. Preliminary deftnition of improvement in rheumatoid arthritis. Arthritis Rheum 38: 727-735, 1995. 92. van der Heijde OM, van 't Hof M, van Riel PL, van de Putte LB: Development of a disease activity score based on judgment in clinical practice by rheumatologists, J Rheumatol 20: 579-581, 1993. 93. Giahnini EH, Ruperta N, Ravetli A, et al: Preliminary definition of improvemerlt in juvenile arthritis. Arthritis Rheum 40: 1202-1209, 1997, 94. Larsen A, Dale K, Eek M: Radiographic evaluation of rheumatoid arthritis and related conditions by standard reference films. Acta Radial Diagn (Stockh) 18: 481-491, 1977. 95. Sharp JT, Young DY, Bluhm GB, et al: How many joints in the hands and wrists should be included in a score of radiologic abnormalities used to as!;tJss rheumatoid arthritis? ArthritL~ Rheum 28: 1326-1335, 1985. 96. Ware JE Jr, Sherbourne CD: The MOS 36-item short-form health survey (SF-36). I: Cooceptual framework and item selectioo, Med Care 30: 473-483, 199t. 97. LandgrafJM: Measuring and monitoring quality of life in children and youth: a 11I!ief commentary. Soz Praventivmed 46: 281-282, 2001. 98. VaJliji lW, Seid M, Kurtin PS: PedsQL 4.0: reliability and validity of the Pe~latric Quality of Life Inventory version 4.0 generic core scales in healthy and patient populations. Med Care 39: 80Q-812, 2001. 99. VaI1Tli lW, Seid M, Smith Knight T, et al: The PedsQL in pediatric rheumatology: reliability, validity, and responsiveness of the Pediatric Quality of Life lnv~ntory Generic Core Scales and Rheumatology Module. Arthritis Rheum 46: 714-725, 2002. 100. Hay~ RD, Anderson R, Revicki 0: Psychometric considerations in evaluating health-related quality of life measures. Qual Life Res 2: 441-449, 1993. 101. Brunner HI, Silverman ED, To T, et al: Risk factors for damage in childhood-onset systemic lupus erythematosus: cumulative disease activity
102. 103, 104, 105,
106.
107, 108.
109, 110. 111. 112. 113,
114.
115.
173
and medication use predict disease damage, Arthritis Rheum 46: 436-444; 2002. Mantel N, Haenszel W: Statistical aspects of the analysis of data from retrospective studies of disease. .I Nat! Cancer Inst 22: 719--748, 1959. Brophy JM, Joseph L: Placing trials in context using Bayesian analysis: GUSTO revisited by Reverend Bayes. JAMA 273: 871-875, 1995. Blume J, Peipert .IF:. What your statistician never told you about P-vaJues. JAm Assoc Gynecol Laparosc 10: 439-444, 2003. Pounds S, Morris SW: Estimating the occurrence of false positives and false negatives in microarray studies by approximating and partitioning the empirical distribution of p-values. Bioinformatics 19: 1236-1242, 2003. Gruvberger-Saal SK, Eden P, Ringner M, et al: Predicting continuous values of prognostic markers in breast cancer from microarray gene expression profiles. Mol Cancer Ther 3: 161-168, 2004. Hosmer OW, Lemeshow S: Applied Logistic Regression, 2nd ed. New York, John Wiley & Sons, 2000. Giannini EH, Malagon CN, Van Kerckhove C, et al: Longitudinal analysis of HLA associated risks for iridocyclitis in juvenile rheumatoid arthritis. .I Rheumatol 18: 1394--1397, 1991. Lee ET, Wang JW: Statistical Methods for Survival Data Analysis. New York, John Wiley & Sons, 2003. Jekel .IF, Elmore JG, Katz OL: Epidemiology, Biostatistics, and Preventative Medicine. Philadelphia, WB Saunders, 1996. Portney LG, Watkins MP: Foundations of Clinical Research. Applications and P,dctice, Norwalk, CT, Appleton & Lange, 1993, pp 509--516. Stevens .lP: Applied Multivariate Statistics for the Social Sciences, 4th ed. Mahwah, N.I, Lawrence Erlbaum, 2001. Moher 0, Schulz KF, Altman 0: The CONSORT statement: revised recommendations for improving the quality of reports of parallel-group I"dndomized trials. .lAMA 285: 1987-1991,2001. Moher D, .lanes A, Lepage L: Use of the CONSORT statement and quality of reports of mndomized trials: a comparative before-and-after evaluation. .lAMA 285: 1992-1995,2001. Davidoff F, DeAngelis CO, Orazen .1M, et al: Sponsorship, authorship, and accountability. N Engl.l Med 345, 825-826; discussion 826-827, 2001.
C HAP T E R
7
ASSESSMENT OF HEALTH STATUS, FUNCTION, AND QUALITY OF LIFE OUTCOMES: METHODOLOGY AND INSTRUMENTS Clar6n M. Duffy and Daniel J. Lovell
~
The rheumatic diseases of childhood influence many if not all aspects of the child's life-not only physical but also sociaJ,1 emotionaJ,2 intellectual, and economic. 3 Indeed, the impact of childhood rheumatic disease is not only on the child but extends to the entire family.4 Conversely, the family's status and functioning can significantly impact the outcome of the child's illness. 4 This chapter describes the instruments that have been developed to assess this web of influence in a quantitative fashion, and specifically focuses on the measurement of functional status and quality of life, with an emphasis on measures developed or used for juvenile rheumatoid arthritis (JRA)/juvenile idiopathic arthritis (IIA).
BACKGROUND What Do We Mean by the Terms "Fundlonal Status" and "Quality of Life"? "Functional status" can broadly be defined as the ability of an individual to perform daily activities. Such ability may be impaired for a variety of reasons and can include both physical and psychological factors. For our purpose, the assessment of functional status emphasizes impairment due to physical factors. The term "quality of life" (QoL) is much more complex. It was originally developed by sociologists to try to determine the effect of material affluence on people's lives. This sociologic approach was developed in the United States during World War II. The concept broadened so that it eventually included education, social welfare, economics, and industrial growth. s This broad societal approach was also incorporated into questionnaires that were developed to assess the status of an individual within this broad framework of concern. Many of these areas of concern or domains, although important to an individual, are well outside the influence of disease 174
and health care interventions. It is for this reason that other terminology to describe QoL was developed. Various authors have used terms such as health-related quality of life (HRQoL), life satisfaction, self-esteem, wellbeing, general health, functional status, and life adjustment to describe those aspects of QoL that relate to the overall global health status of human beings.' There remains considerable confusion about the actual terminology and definitions in this field. For the purpose of this chapter, discussion of QoL will be restricted to HRQoL. However, both functional status and HRQoL are complex concepts that contain numerous subcomponents. There are differences among the experts as to what constitutes HRQoL and, as a consequence, various instruments have been developed to measure it. Such instruments may be divided into generic and disease-specific measures. Generic HRQoL measures are those that purport to be broadly equal across different types and severity of disease, across different medical treatments or health interventions, and across different demographic or cultural subgroups. They are designed to capture aspects of health and disease that cross broad diagnostic categories and social or demographic subgroups. Disease-specific HRQoL measurements are those designed to assess specific diseases or patient populations; such instruments are usually more responsive to changes in individual subject status. The last 25 years have seen the development and validation of a variety of instruments to measure functional status, as well as generic and disease specific HRQoL. These instruments were first developed in adult rheumatology, but in the last 15 years tools specifically designed to be used in childhood-onset rheumatic diseases have been developed.
Why Measure Quality of Life? QoL measurements have been used for a wide variety of purposes. One of the earliest and currently one of the
C HAP T E R
7
ASSESSMENT OF HEALTH, FUNCTION, AND QUALITY OF LIFE OUTCOMES
strongest motivations for measurement of HRQoL is to allow comparisons of health gains or efficiency of healthrelated interventions across broad population groups, in order to be able to compare health gains achieved in treating different patient groups. This information has been used in measurement of health outcomes in a variety of health-related systems of care to assist in priority setting and allocation of limited health care resources. 5-7 In the field of rheumatology, HRQoL has gained wide popularity because it has been shown to measure outcomes that are of direct interest and importance to patients, to provide effective measurements of patient status, to be predictive of patient outcome, and to produce reliable and effective measures of treatment impact. 8-11 To further complicate this already complex field, there are a number of terms and measurement techniques that have been imported from the field of questionnaire development that are foreign and certainly not intuitively easily understood by health care personnel. The most important concepts and terms are included in Table 7-1.
HlerafChy of Outcomes Critical to understanding this area is having a clear vision of the hierarchy of outcomes and outcome measures. The first level of outcome assessment is the measurement of
.-
':1
IABLE 7-1
175
disease activity. This is the aspect of outcome assessment that has been the traditional focus of trials in rheumatology. Disease activity measures include those parameters most familiar to clinicians, such as joint counts, morning stiffness, and erythrocyte sedimentation rate. The major drawback to measures in this area is that they are not really what the patient is interested in. However, measures of disease activity are still widely used in clinical trials, because inhibition of the disease process or activity is an essential component of an effective therapeutic intervention (especially for pharmacologic interventions). At this time, measurement of disease activity is a necessary but not a sufficient approach to measuring patient outcome. Furthermore, when the performance characteristics of the traditional disease activity measures used in rheumatology were Scientifically assessed, many instruments were found to be unreliable, redundant, insensitive to change, or not correlated with long-term patient outcome. 9 The next level up the hierarchy is the measurement of functional status. The focus here is on measuring the ability of the person to perform physical activities of daily life, such as dressing, walking, climbing stairs, and self-care. Obviously, this is an area of more relevance to the patient. Several instruments have been developed and validated to quantify functional status in patients with ]RA. These are discussed in greater detail later in this chapter.
Glossary of Terms Commonly Used in Quality of Lile (Qol) Literature
Ce11ln1i effect: Situation in which the highest score on an instrument does not represent the best status a subject can have: patients with the higheSt score can still improve more.
Centellt validity: Extent to which items in the instrument comprehensively assess the domain of interest.
ConveJ1ent valid"':
Correlation of instrument scores to accepted but not gold standard parameters measuring the same domain.
Crlterlon validity: Comparison of results on an instrument as a "gold standard": such a gold standard does not exist for QoL. DlsallIIlnant Instrument: Designed to most effectively differentiate groups of people. Domatns or dimensions: Area of behavior or experience that is being measured. EvalUUve Instrument: Designed to most effectively detect change in the status of a person over time.
Face .1IdIty: Estimation of whether an instrument appears to be measuring what it is intended (seldom quantitated).
to
measure: does it look reasonable?
Floor effects: Situation in which the lowest possible score on an instrument does not represent the worst status a subject can have; patients with the worst score can still deteriorate further. ~llabll"': Extent to which an instrument can yield accurate and reliable results when used in circumstances or subjects different from those in which it was originally validated: for example, able to be used in varying socioeconomic, ethnic, and geographic disease types or disease severity.
1'ttodeI! ' A theoretical framework of what is being measured and how the instrument should perform. PItIent: preference: Instruments designed so that each individual selects those parameters on the instrument that are most important to them. Predlctlve valid"': Extent to which a score on an instrument at one point predicts patient outcome at a later time.
ReJlabll..,: Extent to which a measuring procedure yields the same results on repeated trials if all the conditions remain unchanged. R ~ (sensIt/v/tJ to change): Extent to which scores on an instrument given at different times in the same subject (or subjects) will change if there is a true change in the status of the subject. ~ or proxy reporter: Someone who answers on behalf of another and reports what they think the subject would answer for themselves (e.g., parent reporting for a child).
176
C HAP T E R
7
ASSESSMENT OF HEALTH, FUNCTION, AND QUALITY OF LIFE OUTCOMES
The next higher level in this hierarchy is the measurement of QoL. Although some authors have made distinctions between "QoL" and "health status," many others use these terms interchangeably, and they may not have separate meanings. 1l In general, QoL is the more commonly used term. Measures at this level are in accordance with the view of the World Health Organization that health is a state of physical, mental, and social well-being. 12 Disease-specific HRQoL measures, such as the Health Assessment Questionnaire (HAQ) or the Arthritis Impact Measurement Scale (AIMS) were developed to incorporate the broad World Health Organization concept of health but by addressing areas that are affected by rheumatic disease. For example, the HAQ includes questions addressing mortality, functional status, physical discomfort, psychologic discomfort, treatment side effects, and economic impact. 9 Despite concerns regarding the ability to measure QoL in children,13 major advances have been made in the development and validation of disease-specific HRQoL tools for children with rheumatic diseases. These instruments also are discussed later. The development of a core set of measures for application in clinical trials in JIA has been extremely important. 14 This core set incorporates the types of instruments already alluded to (Table 7-2).
PROCESS OF INSTRUMENT DEVELOPMENT The development and validation process for functional assessment or QoL instruments has been well established, but for most health care providers the terminology and statistical approaches are unfamiliar. Certainly, it is clear that the development of a new functional assessment or QoL instrument is labor intensive, requires sequential studies, entails input from a wide range of individuals, and needs frequent revisions of the original tool before completion. For example, more than 20 iterations were required in the development of the HAQ. The developmental process for two of the functional assessment tools validated for children with JRA each required 3 to 5 years of work. 15-18 Given the broader scope of content, QoL instruments for JRA have taken at least as long. I 9-2l The process used to develop and validate health measurement questionnaires has been described in textbooks 22 and published articles,23 and, indeed, several articles have described thoroughly the
I!:.
TABLE 7~2 (ore Set of Criteria for Evaluating Change in Treatment Trials of Children With IRA
Physician global assessment of disease activity Patient/parent assessment of overall well-being Functional ability (CHAQ) Number of joints Wilh active arthritis Number of joints with limited range of motion ESR Improvement: At least 30% improvement from baseline in 3 of any 6 variables, with no more than 1 of the remaining variables worsening by > 30% From Giannini, Ruperto, Ravelli, et al." ESR, eJYlhrocyte sedimenlalion rate.
steps used for instruments specifically focused on children with rheumatic diseases. 19.24 A key question to be addressed by anyone who is considering development of a new survey instrument or questionnaire is whether there is truly a need. At this time, there are hundreds of validated questionnaires, and review of the literature may well reveal one that could serve the purpose. Researchers tend to magnify the deficiencies of existing measures and signitlcantiy underestimate the effort required to develop an adequate new measure. 22 There are several compendia of measuring scales. 2'-27 If one or more existent scales are found, then these scales need to be evaluated. If the conclusion is that no existent questionnaire is satisfactory, then much work awaits the brave souls who choose to develop a new tool (Table 7-3). Given this background, the rest of this chapter describes the various tools available in pediatric rheumatology. For a more complete description of these instruments, the reader is referred to the instruments themselves or to published reviews. ll ,28.29
BACKGROUND ON AVAILABLE INSTRUMENTS FOR CHILDHOOD RHEUMATIC DISEASES As discussed already, there is an increasing need to incor-
porate estimates of physical, social, and mental functioning into health assessment, particularly in the assessment of chronic diseases,30-33 in an attempt to provide an all-encompassing measure of QoL. QoL not only includes both health status and functional status but should also attempt to incorporate some aspect of the patient's own perception of what particular aspects of life affect them significantly and to what extent this is influenced by the disease. 33 These facts have been incorporated into the development of measurement tools for adult rheumatic diseases·w--36; and have been demonstrated to be reliable, valid, and responsive in a variety of conditions37-42; and are now believed to be required for inclusion in clinical trials. 43 ,44 Since 1986, various groups have attempted to develop the deftnitive measure for application in JRA. The length of time involved clearly suggests that this is a complex task. The ideal instrument should be practical and easy to use; should be capable of completion by the parent and/or the child within a short time; should measure physical function; should also, as suggested by Singsen,4' measure psychological function and social function, including school, family, and behavioral issues; and should include a measurement of pain. None of the instruments developed to
1'1, 1..1
TABLE 7 -3 Relluiu~d PlOpl'rtil's of the Ideal Instrument for Juvenile Idiopathic Arthritis
Reliable Valid Responsive (sensitive to change) Discriminative ability Easy to use and score Applicable to a wide age range and to a heterogeneous population Measures physical function comprehensively Measures quality of life (including psychosocial functioning) comprehensively
C HAP T E R
7
ASSESSMENT OF HEALTH, FUNCTION, AND QUALITY OF LIFE OUTCOMES
date meets all of these criteria. However, each instrument has unique characteristics that make it distinct, and therefore each one may have different indications for use. In the following sections, each instrument is discussed with an emphasis on its development, its measurement properties, and the settings in which it might be used. Most instruments developed to date for childhood rheumatic diseases have their application in JRA (Table 7-4). However, some have been developed or have been modified for use in other childhood rheumatic diseases. Both types are discussed here.
INSTRUMENTS FOR JUVENILE RHEUMATOID
E~
177
TABLE 7-4 Instruments Developed dnd/or Used in Juwnile Rheumdtoid Arthritis
Measures of Physical Function Childhood Health Assessment Questionnaire (CHAQ) Juvenile Arthritis Assessment Scale (JAFAS) and Report (JAFAR) Juvenile Arthritis Self-Report Index (JASI)
Measures of Quality of ute Juvenile Arthritis Quality of Life Questionnaire (JAQQ) Childhood Arthritis Health Profile (CAHP) Quality of My Life Questionnaire (QoMLQ) Childhood Health Questionnaire (CHQ) Pediatric Quality of Life Inventory Scales (Peds QL)
ARTtfRmS Disease-specific measures of functional status developed for ]RA include the Childhood Arthritis Impact Measurement Scales (CHAIMS),46 the Childhood Health AsseS1lment Questionnaire (CHAQ),16 the Juvenile Arthritis Functibnal Assessment Scale (JAFAS) and Report (JAFAR),17 and the Juvenile Arthritis Self-report Index (JASI).24.47 Disease-specific measures of QoL include the Juvenile Arthritis Quality of Life Questionnaire (JAQQ)l9 and the Childhood Arthritis Health Profile (CAHP).20,21 More recently, there has been a greater focus on the use of generic instruments to assess QoL in children with JRA. Such measures include the Quality of My Life Questionnaire (QoMLQ),48 the Child Health Questionnaire (CHQ)49 and the Pediatric Quality of Life Inventory (Peds QL) (Table 7-4).50 All of these instruments are discussed briefly here, and Table 7-5 summarizes their comparative properties.
The Childhood Arthritis Impact Measurement Scales The CHAIMS was the first disease-specific measure developed for JRA. 46 This was a modification of the
1!fI • • H.BIE 7-5
Puam_
AIMS.34 However, its measurement properties were poor except for the pain dimension and, as a result, it has not had widespread use.
The Childhood Health Assessment Questionnaire The CHAQ16 was derived from the adult HAQ.9,3S It comprises two indices, Disability and Discomfort. The Disability Index assesses function in eight areas--dressing and grooming, arising, eating, walking, hygiene, reach, grip, and activities-distributed among a total of 30 items. In each functional area, there is at least one question that is relevant to children of all ages. Each question is rated on a four-point scale of difficulty in performance, scored from a to 3. The Disability Index is calculated as the mean of the eight functional areas. Discomfort is determined by the presence of pain, as measured by a 100-mm visual analogue scale (VAS). In addition, a 100-mm VAS measures patient/parent global assessment of arthritis. In the original validation study,16 mean scores for patients were 0.84 for the Disability Index and 0.82 for
Comparative Properties of Instruments Developed and/or Used for Juvenile Rheumatoid Arthritis'
Reliability Validity Responsiveness Discriminative ability Applicable to a wide range Applicable to a heterogeneous population Measures physical function Measures quality of life Measures pain Tested widely Easy to use
CHAQ
JAFAR
JASI
JAQQ
CAHP
QoMLQ
CHQ
Peels QL
Strong Strong Weak Moderate Very strong
Strong Strong Weak Moderate No
Strong Strong Moderate Strong No
NA Strong Very strong Weak Very strong
Moderate Moderate NA Moderate Moderate
Moderate Moderate NA NA Moderate
Strong Strong Moderate Moderate Strong
Strong Strong Moderate Moderate Very strong
Very strong
Strong
No
Very strong
NA
NA
Very strong
Strong
Moderate No Moderate Very strong Strong
Moderate No Moderate Moderate Strong
Very strong No No No No
Strong Strong Strong No Strong
Strong Strong No No No
No Moderate No No Strong
Moderate Strong Moderate No Moderate
Moderate Strong Moderate No Moderate
·Although most instruments have been developed using patient~ defmed as having juvenile rheumatoid arthritis (JRA) as the criteria of the American College of Rheumatoll>gy. they are probably equally applicable to patients defined by the International League of Associations for Rheumatology as juvenile idiopathic arthritis (JIA). "No. property absent; Weak, property present but weak; Moderate, property present and moderately strong; Strong. property present and strong; Vel)' strong, property present al1d vel)' strong; NA. not applicable. CHAQ. Childhood Health Assessment Questionnaire; JAFAR, Juvenile Arthritis Functional Assessment Report; JASI, Juvenile Arthritis Self-report Index; JAQQ, Juvenile Arthritis Quality of Life Questionnaire; CAHP, Childhood Arthritis Health Profile; QoMLQ, Quality of My Life Questionnaire; CHQ, Child Health Questionnaire; Peds QL. Pediatric Quality of Life Inventol)'.
178
C HAP T E R
7 Ass E SSM E N T
0 F HE A LT H, FUN C TJ 0 N, AND QUA LI T Y 0 F
the Discomfort Index. Reliability was very good. Convergent validity was also very good, with excellent correlations with Steinbrocker's functional class, active joint count, disease activity index, and degree of morning stiffness. Mean scores for parents and children were not significantly different from one another and were highly correlated, suggesting that parents can reliably report for their children. The CHAQ was completed by parents in all cases, and also by children 8 years of age and older, in a mean of 10 minutes. Responsiveness was established later,51 The CHAQ has been shown to be a useful instrument for outcome evaluation in longitudinal studies.,2-M It has also been used in a variety of settings and translated into a number of different languages while still maintaining excellent reliability, validity, and parent--child correlations. li6--75 One study did suggest that it had poor responsiveness/Ii and in two recent trials it was shown to have reasonable responsiveness. 77 •78 Several studies have demonstrated its usefulness in the evaluation of rehabilitative interventions,?9-&! Other studies suggest that it is highly predictive of the presence of significant pain. 85.86 Efforts to better understand its measurement attributes suggest that a change of 0.13 represents a minimal clinically important change score, while scores of 0.13, 0.63, and 1.75 represent mild, mild to moderate, and moderate disability, respectively.87 Although the CHAQ does not measure psychosocial function in its present form, an earlier version showed reasonable measurement properties in this domain88 ; however, this aspect has not been maintained in the version in current use. The CHAQ has excellent reliability and validity and reasonable responsiveness. It also has good discriminative properties, can be administered to children of all ages, is available in several languages, and is of great use in the clinical setting for long-term follow-up of children with JRA. Therefore, it is of value for longitudinal studies as well as clinical trials and has become the preferred measure in both settings.
The Juvenile Arthritis Functional Assessment Scale and Report The Juvenile Arthritis Functional Assessment Scale 0AFAS)89 preceded the publication of JAFAR.15 JAFAS is an observer-based scale, whereas JAFAR is a report completed by the patient or parent. Items for both instruments were derived from the AIMS, the HAQ, and the McMaster Health Index Questionnaire. 90 JAFAS requires standardized simple equipment and can be administered in about 10 minutes by a health protessional who times the child's performance on 10 physical tasks. Good reliability and convergent validity have been demonstrated. The major limitation ofJAFAS is the requirement of a trained observer and standardized equipment. JAFAR comprises one dimension and contains 23 items which assess ability to perform physical tasks in children older than 7 years of age on a three-point scale scored from 0 to 2; thus, the score range is 0 to 46, with the lower score indicating better function. Two separate versions are available, one for the child 0AFAR-C) and one for the parents 0AFAR-P). In 72 children with JRA (and their parents), mean scores for JAFAR-C and JAFAR-P were 4.39 and 4.38, respectively, and were significantly
L I FE 0
UTe 0 M E S
different from controls. Reliability was good for both versions. Construct validity was good, with predictable correlations among JAFAR-C, JAFAR-P, JAFAS, and pain. Convergent validity was also good, with moderate correlations with disease activity index and active joint counts. Similar measurement properties were found in an English study, in which both versions of JAFAR were highly correlated with one another and with active joint count, pain, Steinbrocker class, and stiffness score, but not with measures of psychological dysfunction?l A Dutch translation of the JAFAR also showed good measurement properties, although the CHAQ did slightly better than the JAFAR in this particular study.li9 Sensitivity to change or responsiveness was demonstrated in a small trial of intravenous immunoglobulin in polyarticular JRA.92 JAFAR has also proved to be a useful measure of functional ability in studies on osteopenia93.94 and sleep disturbance')' in JRA. The JAFAR has excellent reliability and validity. Data from a small controlled trial suggest that it is responsive, but further work is needed in larger trials to clearly establish this property. It cannot be administered to children younger than 7 years of age, and this prohibits its use in children with early onset of JRA. Nonetheless, it is a practical instrument that is of great use in the clinical setting and in the longitudinal follow-up of most children with JRA.
The Juvenile Arthritis Self-Report Index The JASI 24 was developed with a specific focus on physical activity in children older than 8 years of age with JRA. Its emphasis is on responsiveness, and it is aimed primarily at rehabilitation interventions. Through a detailed process, as suggested by Kirshner and Guyatt,23 an instrument with 100 items, distributed in five categories of physical function (self-care, domestic, mobility, school, and extracurricular), was developed. The score range is from 0 to 100, with higher scores indicating better function. A seven-point Likert scale of difficulty in performing tasks was included. As a secondary component, JASI Part 2, patients identify up to five tasks that are most problematic, and these tasks are evaluated on sequential follow-up. This maneuver makes this component of the JASI potentially more responsive and patient specific. In a validation study, the JASI was shown to have good measurement properties. 47 It was completed for the most part by the patients in a mean time of 49.8 minutes (including 10 minutes of instruction). The mean JASI Part 1 score for the group was 78.2 (range, 20 to 100), suggesting overall excellent function. There was reasonable spread of scores, suggesting that the JASI has discriminative ability. Reliability was demonstrated with excellent intraclass correlations. Construct validity was established by demonstration of predicted correlations with other measures used. In a subsample of patients, level of agreement was good between JASI scores and observation of performance of tasks by a therapist. JASI Part 2 was also reliable, but less so. The JASI has been developed in a meticulous fashion, resulting in excellent reliability and validity. Data for JASI Part 11 suggest that it is responsive, although further data are needed to clearly establish this property. However, it
CHAPTER
7
ASSESSMENT OF HEALTH, FUNCTION, AND QUALITY OF LIFE OUTCOMES
cannot be administered to children younger than 8 years of age, and this prohibits its use in children with early onset of JRA. Also, because it is comprehensive, it takes a long time to complete, and this may make it less attractive for clinical use. Nonetheless, the JASI is a comprehensive instrument with excellent measurement properties whose greatest value is probably as a research tool for longitudinal studies.
The 'uvenile Arthritis Quality of Ufe Questionnaire The JAQQ19 was developed by follOWing standard principles of item generation. 23 The parents of 91 patients with JRA and juvenile spondyloarthropathy were interviewed to generate items. For those 9 years of age and oldet, parents and children were interviewed separately, a pr@cess that produced a very high level of agreement between patients and parents over a wide array of perceived difficulties. 96 Additional items on psychosocial function were added by the incorporation of a previously developed psychosocial instrument,97 Generated items were subsequently reduced by application of scores assigned by patients, parents, and a panel of experts and then categorized into four dimensions (gross motor function, fine motor function, psychosocial function, and general symptoms), each with approximately 20 items. A seven-point Likert scale of frequency of difficulty with the particular item in question was then applied; scores range from 1 to 7, with higher scores indicating worse function. Respondents score all items and are also asked to identify up to five items in each dimension with which they are having difficulty; they may also volunteer their own items for each dimension. The mean score for the five highest-scoring items in each dimension is computed as the Dimension Score (range, 1 to 7); the Total JAQQ Score is computed as the mean of the four Dimension Scores (range, 1 to 7). The J~QQ was completed by 30 patients in 20 minutes initially, and then in 5 minutes on retesting 5 weeks later, showing good construct validity, with moderate correlations with measures of disease activity and excellent correlations with pain. Correlations for the psychosocial dimension were less, as predicted, being best with pain. Responsiveness was demonstrated by cQrrelations of change scores, which were moderate with sum of joint severity score and physician global assessment of change and excellent with pain. Responsiveness was also demonstrated by the ability of the JAQQ to discriminate among patients based on physician global assessment of change. After this initial study,19 the item number was reduced to 74----;gross motor function, 17 items; fine motor function, 16 items; psychosocial function, 22 items; and general symptoms, 19 items. A pain dimension, added as a supplement to the JAQQ, included a 100-mm VAS, a five-point Likert scale, and, for children younger than 10 years of age, a five-point happy/sad face modeI.98 Face and content validity of this version was confirmed in 20 pediatric rheumatologists and therapists. Construct validity and responsiveness were established. 99 Responsiveness was further established before and after commencement of new drug therapy (a mean of 8 weeks apart).100 Responsiveness of the JAQQ was also shown to be maintained over time 101 and to be a~ least as good as that of the CHAQ or CHQ.102 English and French versions are availahle, and a Dutch translation has been validated. 103
179
The JAQQ has been developed in a detailed fashion, resulting in excellent validity and responsiveness. It can be administered to children of all ages and disease onset types, in a reasonable period of time with minimal assistance, and can be scored quickly by hand; this makes it practical for use in the clinical setting. Perhaps its biggest drawback is that for each child essentially a different instrument is completed, based on its unique scoring system. This may make comparison between groups difficult, compromising the test's discriminative ability, and is the subject of ongoing study. Nonetheless, the JAQQ has excellent responsiveness and therefore is a very useful instrument for clinical trials.
The Childhood Arthritis Health Profile The CAHp 20 was developed to capture the broad range of health states in children with JRA including physical and psychosocial function. It is a parent report that is selfadministered and consists of three modules--generic health status measures, JRA-specific health status measures, and patient characteristics. The initial report focused on the development, validity, and reliability of the functional scales, both JRA specific and generic. Three functional scales--gross motor function, fine motor function, and role activities (play, family, friends).-were determined for the JRA-specific scales. Internal reliability was demonstrated by good inter-item correlations within scales and minimal item scale variation. Correlation coefficients for the JRA-specific scales with one another ranged from 0.84 to 0.97, whereas those for the generic functioning scales were 0.73, demonstrating validity of these scales and further suggesting that the JRA-specific scales provide additional information beyond that of the generic functioning scales. In a follow-up report,21 differences in the )RA-specific scales were compared with clinician-rated disease severity and activity. Significant differences were seen across disease severity and activity for gross motor function, fine motor function, usual role activities affected by JRA, and school functioning. This study confirmed the discriminative ability of the CAHP. The CAHP is a promising instrument but as yet too few data have been published to determine its usefulness. Because of its comprehensiveness and complexity, it is unlikely to be of practical use in the clinical setting. Nonetheless, because of its discriminative ability, it is likely to find a role as a research tool for longitudinal studies.
The Quality of My Ufe Questionnaire The QoMLQ was developed in an attempt to distinguish between difficulties resulting from the disease itself and those difficulties that are generic. 48 It comprises two separate 100-mm VAS, anchored with the descriptors "Worst" and "Best," that direct respondents to indicate their "quality of life," both aspects caused by the disease itself and those caused by overall difficulties not necessarily directly related to the disease. This approach demonstrated the importance of distingUishing between these factors, because clear differences were noted among respondents between the two scales. This is a short and easy to use generic instrument that has been demon-
180
C HAP T E R
7
ASSESSMENT OF HEALTH, FUNCTION, AND QUALITY OF
strated to be highly reliable and valid. 4R However, it needs further study.
The Child Health Questionnaire The CHQ49 is a generic instrument that comprises a number of different forms. The form used most commonly in children with ]RA is the Parent Form 50 (PF 50), which contains 50 items distributed in several dimensions: global health, physical activities, everyday activities, pain, behavior, well-being, self esteem, general health, and family. These sections are complemented by general questions about the child and the caregiver. Two separate scores can be computed that estimate physical and psychosocial function; both are scored from 0 to 100, with the higher score indicating better function. In a recent study of short-term outcome in 116 children with ]IA observed for less than 2.5 years, Selvaag and colleagues 104 demonstrated poorer physical status but minimal psychological impairment in]IA patients relative to controls using the CHQ. The CHQ is an instrument that has received significant attention, particularly from the Pediatric Rheumatology International Trials Organization (PRINTO), which has validated it for use in 32 languages. 73 It was used in combination with the CHAQ in a trial of methotrexate, where it was shown to be highly responsive. 7R However, one study suggested that the ]AQQ may be at least as responsive as the CHQ for studies in JIAIOZ For the reasons outlined earlier, and because of its generalizability, the CHQ has become the preferred measure of QoL for ]IA trials.
The Pediatric Quality of Life Inventory The Peds QL is a modular instrument designed to measure HRQoL in children and adolescents aged 2 to 18 years. 50 It contains a generic core integrated with a disease-specific core. The generic core has undergone various iterations, the most recent of which, the Peds QL 4.0 Generic Core Scales, contains 23 items distributed in four scales: physical, emotional, social, and school functioning. The Peds QL 3.0 Rheumatology Module contains 22 disease-specific items distributed in five scales (pain, daily activities, treatment, worry, and communication). It is completed by both the children and their parents and consists of developmentally appropriate forms for varying age groups. When it is completed separately, parentchild concordance has been demonstrated to be good. The module takes approximately 15 minutes to complete. Each item is scored on a five-point scale (0 to 4), with a higher score indicating worse function. A mean Scale Score is computed based on the number of items scored. This is then extrapolated in a reverse fashion to a scale of 0 to 100, with a higher score indicating better function. Total Scale Scores are computed as the mean across all items scored in that scale. This process is the same for the Generic Core Scale and the Rheumatology Module. This instrument was shown to have excellent reliability, validity, and responsiveness in a study of 271 children with various rheumatic diseases, 91 of whom had ]RA, and their parents. !Os Reliability varied with the age of the child, being less good for younger children. Reliability was also not as
LI FE
OUTCOM ES
good for the Rheumatology Module. Responsiveness has not been tested in a trial setting. Nonetheless this instrument represents an important addition to the pool of outcome measures available for use in]RA, and further studies are being followed with interest.
INSTRUMENTS AVAILABLE FOR USE IN RHEUMATIC DISEASES OTHER THAN JUVENILE IDIOPATHIC ARTHRITIS There has been considerable recent international effort to develop appropriate measures for use in inflammatory myopathies in both adults and children. This effort has culminated in the development of measures of disease activity and damage 106 and a core set of measures for childhood-onset and adult-onset diseases. 107 ,IOH The core set for juvenile dermatomyositis (TDM) was conducted simultaneously with a similar effort for juvenile systemic lupus erythematosus (SLE).IOR The development of specific functional status or HRQoL measures in either disease, specific to children, has not been as active as for ]RA. Through an initial survey of 267 physicians in 46 countries worldwide, followed by a nominal group technique using 40 physicians from 34 countries, 37 response variables for ]DM were examined. lOR Ultimately, a core set for JDM was arrived at and comprised muscle strength testing, muscle enzyme testing, functional ability, physician and parent/patient global assessments, and a global score tool. This instrument has recently undergone validation testing in an attempt to define improvement in the core set. 109 Such improvement is defined as a minimum of 15% improvement in the domains of muscle strength and physical function, a minimum of 20% improvement for physician and patient global assessments and overall global assessment, and a minimum of 30% improvement in muscle enzymes. In addition, however, a specific outcome measure has been developed for ]DM, the Childhood Myositis Assessment Scale (CMAS).lIO This is a therapist-administered assessment of muscle strength, endurance, and function with excellent measurement properties. It is scored on a scale of 0 to 52, based on the ability of the child to perform specific tasks scored by an observer. The CMAS has been shown to have outstanding intraand inter-observer reliability. It has been shown to have good validity and good correlations with manual muscle testing. It also has reasonable responsiveness and is undergoing further study. The CHAQ has also been validated for use in ]DM patients. It was shown to have excellent test-retest reliability, validity, and responsiveness lll - 1l4 and to be of value as a measure of outcome. liZ As a component of the initiative discussed for ]DM, a similar effort was conducted for juvenile SLE. 1OH In this component of the study, 41 response variables were tested. Ultimately, measures of disease activity included specific SLE immunologic tests and renal function measures, as well as physician and parent/patient global assessments, an overall global assessment, a measure of growth and development, and a measure of HRQoL (most likely the CHQ, but this has not been finalized). Brunner and associates pre-
C HAP T E R
7
ASSESSMENT OF HEALTH, FUNCTION, AND QUALITY OF LIFE OUTCOMES
viously validated the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) for juvenile SLE 115 and in a recent study demonstrated the responsiveness of the European Consensus Lupus Activity Measurement (ECLAM) for juvenile SLE, suggesting that it might be more sensitive than the SLEDA[ in this population. 116 Two recent studies described the extent of damage in juvenile SLE117.118 using the Systemic Lupus International Collaborating Clinics (SLICC)/ACR Damage Index. Further work is ongoing to develop new measures or test existing measures of function or HRQoL in both ]DM and]$LE.
CONCLUSION This chapter outlined the essential properties required of any instrument for application in children with rheumatic diseases and discussed the various instruments that have been developed. The focus was predominantly on outcome measures that have been developed for ]RA. The properties of these various instruments were compared (Table 7-5). It is clear that they differ significantly from one another and have been developed with different objectives in mind, so that each has unique qualities. Four measures-CHAIMS, CHAQ, ]AFAR and ]ASI-have a specific focus on physical function, whereas the others-]AQQ, CAHP, QoMLQ, CHQ, and Peds QL-attempt to measure HRQoL in addition to physical function. The ]ASI has excellent measurement properties. Because it is comprehensive and takes considerable time to cOfllplete, it is probably best applied as a research tool, rather than a tool for clinical use. The ]AFAR also has excellent measurement properties and has been tested widely; however, its use may be limited because it cannot be administered to a broad enough age range. The CHAQ has excellent measurement properties and has uhdergone the most Widespread use. It is simple to use and can be completed within a brief period. It is versatile and can be used in clinical trials and outcome studies, in a number of diseases, and is also applicable to a wide age range. It not only is of great value as a research tool but also can be used in the clinical setting. For these reasons, it has a distinct advantage and has become the preferred measure of functional status for ]RA. The ]AQQ is also comprehensive, although it can be completed quickly. It can be administered to all age groups and is highly responsive. Its most appropriate role is in clinical trials, the specific purpose for which it was designed; however, it may also have a role in longitudinal studies. The CAHP also has excellent measurementproperties, particularly in its discriminative ability. It is comprehensive but cannot be completed quickly, so its role will most likely be as a research tool for longitudinal studies. Both the QoMLQ and the Peds QL have distinct merits. The QoMLQ is simple to use but may not be comprehensive enough for most purposes; this may limit i:ts use. The Peds QL is a comprehensive instrument with excellent measurement properties that can be completed relatively qUickly. Therefore, it merits further study to ascertain whether it should be the preferred measure of HRQoL for ]IA and other pediatric rheumatic
181
diseases. That distinction is currently held by the CHQ, which also has excellent measurement properties. However, the CHQ may not be responsive enough, and the ]AQQ and Peds QL might be better. This issue needs further study. To date, although the thrust to develop instruments for application in diseases other than ]IA continues, this effort lags somewhat behind. The CMAS appears to be an excellent measure of functional status in ]DM, and the CHAQ appears to have an important role in this disease. However, a concentration of effort needs to be applied to the study of juvenile SLE in this regard. Such work is ongoing. The past several years have been a very exciting and active time of research. Work is still ongoing to improve these instruments and to clarify the role each should hold in the future, not only in ]RA but also in other pediatric rheumatic diseases.
REFERENCES 1. Ross CK. Lavigne ]V, Hayford JR, et al: Psychological factors affecting reported pain in juvenile rheumatoid arthritis. J Pediatr PS'Ychol 18: 561, 1989. 2. Miller JJ: Psychosocial factors related to rheumatic diseases in childhood. J Rheumatol 20: 1, 1993. 3. Allaire SH, DeNardo BS, Szer IS, et al: The economic impact of juvenile chronic arthritis. J Rheumatol 19: 1952. 1992. 4. Timko C, Baumgartner M. Moos RH, et al: Parenteral risk and resistance factors among children with juvenile rheumatic disease: a four-year predictive study. J Behav Med 16: 571, 1993. 5. Carr AJ, Thompson PW, Kirwan FR: Quality of life measures. Br J Rheumatol 35: 275, 1996. 6. Bell MJ, Bombardier C, Tugwell P: Measurement of functional status, quality of life, and utility in rheumatoid arthritis. Arthritis Rheum 33: 591, 1990. 7. Kaplan RM, Coons SJ, Anderson JP: Quality of life and policy analysiS in arthritis. Arthritis Care Res 5: 173, 1992. 8. Tugwell P, Bombardier C, Buchanan WW, et al: Methotrexate in rheumatoid arthritLs: impact on quality of life assessed by traditional standard-item and individualized patient preference health status questionnaires. Arch Intern Med 150: 59, 1990. 9. Fries JF: Toward an understanding of patient outcome measurement. Arthritis Rheum 26: 697, 1983. 10. Ward MM: Clinical measures in rheumatoid arthritis: which ones are most useful in assessing patients? J Rheumatol 21: 17. 1993. 11. Duffy CM, Watanabe Duffy KN: Health assessment in the rheumatic diseases of childhood. Curr Opin Rheumatol 9: 440, 1997. 12. World Health Organization: The First Ten Years of the World Health Organization. Geneva, WHO, 1958. 13. Tucker LB: Whose life is it anyway? Understanding quality of life in children with rheumatic diseases. J Rheumatol 27: 8, 2000. 14. Giannini EH, Ruperto R, Ravelli A, and the Paediatric Rheumatology International Trials Organization (PRINTO): Preliminary definition of improvement in juvenile arthritis. Arthritis Rheum 40: 1202, 1997. 15. Howe S, Levinson J, Shear E, et al: Development of a disability measurement tool for juvenile rheumatoid arthritis: the Juvenile Arthritis Functional Assessment Report for children and their parents. Arthritis Rheum 34: 873, 1991. 16. Singh G, Athreya BH, Fries JF, et al: Measurement of health status in children with juvenile rheumatoid arthritis. Arthritis Rheum 37: 1761, 1994. 17. Lovell OJ: Newer functional outcome measurements in juvenile rheumatoid arthritis: a progress report. J Rheumatol 19: 28-31 (Suppl 33), 1992. 18. Graham TB, Lovell OJ: Outcome in pediatric rheumatic disease. Curr Opin Rheum 9: 434, 1997. 19. Duffy CM, Arsenault L, Watanabe Duffy KN, et al: The Juvenile Arthritis Quality of Life Questionnaire: development of a new responsive index for juvenile rheumatoid arthritis and juvenile spondyloarthritides. J Rheumatol 24: 738, 1997. 20. Tucker LB, De Nardo BA, Abetz LN, et al: The childhood arthritis health profile (CAHP): validity and reliability of the condition specific scales labstract]' Arthritis Rheum 38: S183. 1995. 21. Tucker LB, De Nardo BA, Schaller JG: The Childhood Arthritis Health Profile: correlation of juvenile-rheumatoid arthritis specific scales With disease severity and activity [abstract]. Arthritis Rheum 39 (Supp!): S57. 1996. 22. Streiner DK, Norman GR: Health Measurement Scales: a Practical Guide to their Development and Use. Oxford, UK, Oxford University Press, 1989.
182
C HAP T E R
7
ASSESS ME NT OF HEALTH, FUNCTION, AND QUA LITY OF LIFE OUTCOMES
23. Kirshner B, Guyatt G: A methodologic framework for assessing health indices. J Chron Dis 38: 27, 1985. 24. Wright VF, Law M, Crombie V, et al: Development of a selt~report functional status index for juvenile rheumatoid arthritis. J Rheumatol 21: 536, 1994. 25. Orvaschel H, Walsh G: The assessment of adaptive functioning in children: a review of existing measures suitable for epidemiological and clinical services research. Rockville, MD, National Institutes of Mental Health, 1984. 26. McDowell I, Newell C: Measuring health a gUide to J"dting scales and questionnaires. Oxford, UK, Oxford University Press, 1987. 27. Norman GR, Streiner DL: PDQ Statistics. Toronto, BC Decker, 1986. 28. Duffy CM, Tucker L, BurgOS-Vargas R: Update on functional assessment toob. J Rheumatol 27 (Suppl 58): 11, 2000. 29. Katz P (ed): Patient outcomes in rheumatology: a review of measures. Arthritis Rheum 49 (Suppl 5); Sl, 2003. 30. Brook RH, Appel FA: Quality of care assessment: choosing a model for peer review. N Engl J Med 288: 1323, 1973. 31. Feinstein AR, Josephy BR, Wells CK: Scientific and clinical problems in indexes of functional disability. Ann Intern Med 105: 413, 1986. 32. Guyatt GH, Veldhuyzen Van lanten SJO, Feeny DH, et al: Measuring quality of life in clinical trials: a taxonomy and review. Can Med Assoc J 140: 1441, 1989. 33. Gill TM, Feinstein AR: A critical appraisal of the quality of Quality of Life measurements. JAMA 272: 619, 1994. 34. Meenan RF, Gertrnan PM, Mason JH: Measuring health status in arthritis: the arthritis impact measurement scales. Arthritis Rheum 23: 146, 1980. 35. Fries JF, Spitz P, Kraines RG, et al: Measurement of patient outcome in arthritis. Arthritis Rheum 23: 137, 1980. 36. Tugwell P, Bombardier C, Buchanan WW, et al: The MACTAR patient preference questionnaire: an individualized functional priority approach for assessing improvement in clinical trials in rheumatoid arthritis. J Rheumatol 14: 446, 1987. 37. Meenan RF, Gertman PM, MasonJH, et al: The arthritis impact measurement scales: further investigations of a health status measure. Arthritis Rheum 25: 1048, 1982. 38. Potts MK, Brandt K: Evidence of the validity of the arthritis impact measurement scales. Arthritis Rheum 30: 93, 1987. 39. Duffy CM, Watanabe Duffy KN, Gladman DD, et al: Utility of the arthritis impact measurement scales for patients with psoriatic arthritis. J Rheumatol 19: 1727, 1992. 40. Anderson JJ, Firschein HE, Meenan RF: Sensitivity of a health status measure to short-term clinical changes in arthritis. Arthritis Rheum 32: 844, 1989. 41. FriesJF, Spitz PW, Young DY: The dimensions of health outcomes: the health assessment questionnaire, disability and pain scales. J Rheumatol 9: 789, 1982. 42. Pincus T, Callaghan LF, Brooks RH, et al: Self-report questionnaire scores in rheumatoid arthritis compared with traditional physical, radiographic and labol"dtory measures. Ann Intern Med 110: 259, 1989. 43. Felson DT, Anderson JJ, Boers M, et al: The American College of Rheumatology core set of disease activity measures for rheumatoid arthritis clinical trials. Arthritis Rheum 36: 729, 1993. 44. Tugwell P, Bombardier C, Buchanan WW, et al: Methotrexate in rheumatoid arthritis patients: impact on quality of life-assessed by traditional standarditem and individualized patient preference health status questionnaires. Arch intern Med 150: 59, 1990. 45. Singsen BH: Health status (arthritis impact) in children with chronic rheumatic diseases: current measurement issues and an approach to instrument design. Arthritis Care Res 4: 87, 1991. 46. Coulton Cj, lborowsky E, Lipton J, et al: Assessment of the reliability and validity of the arthritIs impact measurement scales for children with juvenile arthritis. Arthritis Rheum 30: 819, 1987. 47. Wright VF, Longo Kimber J, Law M, et al: The Juvenile Arthritis Functional Status Index (jAS(): a validation study. J Rheumatol 23: 1066, 1996. 48. Feldman BM, Grundland B, McCullough L, Wright V: Distinction of quality of life, health-related quality of life, and health status in children referred for rheumatology care. J Rheumatol 27: 226, 2000. 49. Landgraf JM, Abetz L, Ware JE: Child Health Questinnaire (CHQ); A User's Manual. Boston, MA, The Health Institute, New England Medical Center, 1996. 50. Varni jW, Seid M, Rode CA: The Peds QL: measurement model for the Pediatric Quality of Life Inventory. Med Care 37: 126, 1999. 51. Singh G, Brown B, Athreya B: Functional status in juvenile rheumatoid arthritis: sensitivity to change of the childhood health assessment questionnaire labstractl. Arthritis Rheum 34: S81, 1991. 52. Andersson Gare B, Fasth A, Wiklund I: Measurement of functional status in juvenile chronic arthritis: evaluation of a Swedish version of the Childhood Health Assessment Questionnaire. Clin Exp Rheumatol 11: 569, 1993. 53. Andersson Gare B, Fasth A: The natural history of juvenile chronic arthritis: a population based cohort study. II: Outcome. J Rheumatol 22: 308, 1995. 54. Ruperto N, Ravelli A, LevisonJE, et al: Long term health outcomes and quality of life in American and Italian inception cohorts of patients with juvenile rheumatoid arthritis. II: Early predictors of outcome. J Rheumatol 24: 952, 1997. 55. Minden K, Kiessling U, Listing J, et al: Prognosis of patients with juvenile chronic arthritis and juvenile spondyloarthropathy. J Rheumatol 27: 2256, 2000.
56. Guillaume S, Prieur AM, Coste J, Job-Deslandre C: Long-term outcome and prognosis in oligo-articular onset juvenile idiopathic arthritis. Arthritis Rheum 43: 1858, 2000. 57. lak M, Pedersen FK: Juvenile chronic arthritis into adulthood: a long-term follow up study. Rheumatology 39: 198,2000. 58. Lomater C, Gerloni V, Gattinara M, et al: Systemic onset juvenile idiopathic arthritis: a retrospective study of 80 consecutive patients foHowed for 10 years. J Rheumatol 27: 491, 2000. 59. Spiegel LR, Schneider R, Lang BA, et al: Early predictors of poor functional outcome in systemic onset juvenile rheumatoid arthritis: a multicentre cohort study. Arthritis Rheum 43: 2402, 2000. 60. Oen K, Malleson P, Cabral 0, et al: Disease course and outcome of juvenile rheumatoid arthritis in a multicentre cohort. J Rheumatol 29: 1989, 2002. 61. Fantini F, Gerloni V, Gattinara M, et al: Remission in juvenile chronic arthritis: a cohort study of 683 consecutive cases with a mean 10 year follow up. J Rheumatol 30: 579, 2003. 62. Flato B, Lien G, Smerdel A, et al: Prognostic factors in juvenile rheumatoid arthritis: a case-control study revealing early predictors and outcome after 14.9 years. J Rheumatol 30: 386, 2003. 63. Foster HE, Marshall N, Myers A, et al: Outcome in adults with juvenile idiopathic arthritis. Arthritis Rheum 48: 767, 2003. 64. Bowyer S, Roettcher PA, Higgins GC, et al: Health status of patients with juvenile rheumatoid arthritis at 1 and 5 years after diagnosis. J Rheumatol 30: 394, 2003. 65. Oen K, Malleson P, Cabral D, et al: Early predictors of long-term outcome in patients with juvenile rheumatoid arthritis: subset-specific correlations. J Rheumatol 30: 585, 2003. 66. Doherty E, Yanni G, Conroy RM, et al: A comparison of child and parent ratings of disability and pain in juvenile chronic arthritis. J Rheumatol 20: 1563, 1993. 67. Len C, Goldenberg./, Bosi Ferraz M, et al: Crosscuitural reliahility of the Childhood Health A"essment Questionnaire. J Rheumatol 21: 2349, 1994. 68. Fantini F, Corvaglia G, Bergomi P, et al: Validation of the Italian version of the Stanford Childhood Health Assessment Questionnaire for measuring functional status for children with chronic arthritis. Clin Exp Rheumatol 13: 785, 1995. 69. Van der Net J, Prakken ABJ, Helders PJM, et al: Correlates of disablement in polyarticular juvenile chronic arthritis: a cross sectional study. Br J Rheumatol 35: 91, 1996. 70. Goycochea-Robles MV, Garduno-Espinosa .I, Vilchis-Guizar E, et al: Validation of a Spanish version of the Childhood Health A"essment Questionnaire. J Rheumatol 24: 2242, 1997. 71. Arguedas 0, Andersson-Gare B, Fasth A, et al: Development of a Costa Rican version of the Childhood Health Assessment Questionnaire. J Rheumatol 24: 2233, 1997. 72. Flato B, Soskaar 0, Vinje 0, et al: Measuring disability in early juvenile arthritis: Evaluation of a Norwegian version of the Childhood Health Assessment Questionnaire. J Rheumatol 25: 1851, 1998. 73. Ruperto N, Ravelli A, Pistorio A. et al: Cross-cultuml adaptation and psychometric evaluation of the Childhood Health Assessment Questionnaire (CHAQ> and the Child Health Questionnaire (CHQ) in 32 countries. Clin Exp Rheumatol 19 (Suppl 23): Sl-S9, 2001. 74. Pouchot J, Larbre]p, Lemelle I, et al: Validation of the French version of the Child Health Questionnaire in juvenile idiopathic arthritis. Rev Rhum 69: 468, 2002. 75. Madl SM, Al Mayouf SM, Grainger CG, Bahabri SA: The Arabic version of the Child Health Questionnaire modified for Arabic children. Saudi Med J 25: 83, 2004. 76. Ruperto N, Ravelli A, Migliavacca 0, et al: Responsiveness of clinical measures in children with oligoarticular juvenile chronic anhritis. J Rheumatol 26: 1827, 1999. 77. Lovell OJ, Giannini EH, Reiff A, et al: Etanercept in children with polyarticular juvenile rheumatoid arthritis. Pediatric Rheumatology Collaborative Study Group. N Engl J Med 342: 763-769, 2000. 78. Ruperto N, Murray K, Gerloni V, et al: A randomized trial of methotrexate in medium versus high doses in children with juvenile idiopathic arthritis who failed on standard dose. Arthritis Rheum 49 (Suppl): S232, 2002. 79. Fan JS, WesselJ, Ellsworth J: The relationship between strength and function in females with juvenile rheumatoid arthritis. J Rheumatol 25: 1399, 1998. 80. Wessel J, Kaup C, Fan J, et al: Isometric strength in children with arthritiS: reliability and relation to function. Arthritis Care Res 12: 238, 1999. 81. Miller ML, Kress AM, Berry CA: Decreased physical function in juvenile rheumatoid arthritis. Arthritis Care Res 12: 309, 1999. 82. Bekkering WP, ten Cate R, van SUijlekom-Smit LW, et al: 111e relationship between impairments in function and disabilities in independent function in children with systemic juvenile idiopathic arthritis. J Rheumatol 28: 1099, 200 1. 83. Epps H, Hurley M, Utley M: Development and evaluation of a single value score to assess global range of motion in juvenile idiopathic arthritis. Arthritis Rheum 47: 398, 2002. 84. Takken T, van der Net J, Helders PJ: Relationship between functional ahility and physical fitness in juvenile idiopathic arthritis patients. Scand .I Rheumatol 32: 174, 2003. 85. Sallfors C, Hallberg LR, Fasth A: Gender and age differences in pain, coping and health status among children with chronic arthritis. Clin Exp Rheumatol 21: 785, 2003.
C HAP T E R
7
ASSESSMENT OF HEALTH, FUNCTION, AND QUALITY OF LIFE OUTCOMES
86, Den K, Reed M, Malleson P, et al: Radiologic outcome and its relationship to functional disability in juvenile rheumatoid arthritis, ] Rheumatol 30: 832, 2003, 87, Dempster H, Porepa M, Young N, Feldman BM: The clinical meaning of functional outcome scores in children with juvenile arthritis, Arthritis Rheum 44: 1768, 2001. 88, Billings AG, Moos RF, Miller ]], et al: Psychosocial adaptation in juvenile rheumatic disease: a controlled evaluation, Health Psychol 6: 343, 1987, 89, Lovell 0], Howe S, Shear S, et al: Development of a disability measurement tool for juvenile rheumatoid arthritis, Arthritis Rheum 32: 1390, 1989. 90. Chambers LW, MacDonald LA, Tugwell P, et al: The McMaster Health Index Questionnaire as a measure of the quality of life for patients with rheumatoid
d Child Behaviour Profile. Burlington, VT, Queen City, 1983, 98. MaUl)\iIksela Fl, Olkkala KT, Korpela R: Measurement of pain in children with self-reporting and behavioural assessment. Clin Pharmacal Ther 42: 137, 1987, 99. Duffy CM, Arsenault L, Watanabe Duffy KN, et al: Validity and sensitivity to chanlle of the Juvenile Arthritis Quality of Life Questionnaire (JAQQ) [ab,tractJ. Arthritis Rheum 36 (Supp!): S144, 1993. 100. Duffy CM, Arsenault L, Watanabe Duffy KN, et al: Relative sensitivity to change of the Juvenile Arthritis Quality of Life Questionnaire following a new lteaunent [abstract). Arthritis Rheum 37 (Supp!): 5196, 1994. 101. Duffy CM, Arsenault L, Watanabe Duffy KN, et al: Relative sensitivity to chanJle of the Juvenile Arthritis Quality of Life Questionnaire on sequential follow up [abstractJ. Arthritis Rheum 38 (Supp!): S178, 1995. 102. Duffy CM, Watanabe Duffy KN, Gibbon M, et al: Accuracy of functional outcome measures in defining improvement in juvenile idiopathic arthritis [abstract). Ann Rheum Dis 59: 724. 2000. 103. Takken T, van del' Net], Kuis W, Heidel'S PJ: Aquatic fitness training for children with juvenile idiopathic arthritis. Rheumatology 42: 1408, 2003.
183
104. Selvaag AE, Flato B, Lien G, et al: Measuring health status in early juvenile idiopathic arthritis: determinants and responsiveness of the Child Health Questionnaire. J Rheumatol 30: 1602, 2003. 105. Varni lW, Seid M, Smith Knight T, et al: The Peds QL in Pediatric Rheumatology: reliability, validity and responsiveness of the Pediatric Quality of Life Inventory Generic Core Scales and Rheumatology Module. Arthritis Rheum 46: 714, 2002, 106. Isenberg DA, Allen E, Farewell V, et al: International consensus on outcome measures for patients with idiopathic inflammatory myopathies: development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology 42: 1, 2003. 107. Miller FW, Rider LG, Chung YL, et al: Proposed core set measures for disease outcome assessment in adult and juvenile idiopathic inflammatory myopathies. Rheumatology 40: 1262, 2001. 108. Ruperto N, Ravelli A, Murray K], et al: Preliminary core set of measures for disease activity and damage assessment in juvenile systemic lupus erythematosu, and juvenile dermatomyositis. Rheumatology 42: 1, 2003. 109. Rider L, Giannini EH, Harris-Love M, et al: Defining clinical improvement in adult and juvenile myositis, J Rheumatol 30: 603, 200} 110, Lovell 0], Lindsley CB, Rennenbohm RM, et aI: Development of validated disease activity and damage indices for the juvenile idiopathic inflammatory myopathies: 11. The Childhood Myositi, Assessment Scale (CMAS): a quantitiative tool for the evaluation of muscle function. The Juvenile Dermatomyositis Disease Activity Collaborative Study Group. Arthritis Rheum 42: 2213, 1999. 111. Feldman BM, Ayling-Campos A, Luy L, et al: Measuring disability in juvenile dermatomyositis: validity of the Childhood Health Assessment Questionnaire, J Rheumatol 22: 326, 1995. 112. Huber AM, Lang B, LeBlanc C, et al: Medium and long-term functional outcomes in a multi-center cohort of children with juvenile dermatomyositis. Arthritis Rheum 43: 541, 2000, 113. Huber AM, Hicks ]E, Lachenbruch PA, et al: Validation of the Childhood Health Assessment Questionnaire in the juvenile idiopathic myopathies. J Rheumatol 28: 1106, 2001. 114. Takken T, Elst E, Spermon N, et al: The physiological and physical determinants of functional ability measures in children with dermatomyositis, Rheumatology 49: 591, 2003. 115. Brunner HI, Feldman BM, Bombardier C, Silverman ED: The SLEDAI, SLAM and BILAG are sensitive to clinical change in childhood-onset sy'temic lupus erythematosus. Arthritis Rheum 42: 1354. 1999. 116, Brunner HI, Silverman ED, Bombardier C, Feldman BM: European Consensus Lupus Activity Measurement is sensitive to change in childhoodonset sy'temic onset lupus erythematosus. Arthriti, Rheum 49: 335, 2003. 117. Brunner HI, Silverman ED, To T, et al: Risk factors for damage in childhoodonset systemic lupus erythematosus: cumulative disease activity and medication u,e predict disease damage. Arthritis Rheum 46: 436, 2002. 118, Ravelli A, Duarte-Salazar C, Buratti S, et al: Assessment of damage in juvenile-onset systemic lupus erythematosus: a multi-centre cohort ,tudy. ArthritiS Rheum 49: 501, 2003.
( HAP T E R
8
A GENERAL ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN Balu H. Athreya and Carol B. Lindsley
IJif
Rheumatic diseases are chronic, multisystem diseases characterized by an unpredictable course with periods of exacerbation and remission. Care of affected children requires consultations with medical and surgical specialists. Many of these disorders cause muscle weakness and joint contractures with functional disabilities. Therefore, children with rheumatic diseases often require physical and occupational therapy, counseling, and social services (Table 8-1). Treatment of these diseases with anti-inflammatory agents, immunosuppressives, and the new biologic immune modulators is discussed in various chapters dealing with individual disease categories. This chapter discusses general principles of management of issues, other than medical, that are common to all chronic diseases. This includes discussions on compliance issues, strategies to improve self-image and coping skills, schoolrelated issues, transition to adult care, physical and occupational therapy, and one specific aspect of well-child care-immunization. There are multiple dimensions to the effects of chronic illness on children and on members of their families.! Several early studies suggested a negative impact of chronic illness on psychosocial deveiopment,I-3 family,4 school life,s.6 and family finances. 7 •H Studies focusing on children with rheumatic diseases growing up before the availability of modern drugs showed that 30% to 50% entered adult life with active disease. 9.10 Severe disability was common in this young adult population with decreased physical functions, poor health perception, and pain. ll ,12 All of the psychosocial and physical problems assume greater importance when such children reach adolescence!3 or when they grow up to be adults with continuing disease activity. For all of these reasons, a program of care for children with rheumatic diseases should plan for the whole child and the future and should be comprehensive (e.g., family-centered, community-based, coordinated, and cost-effective) (Table 8-2).14 More recent studies have not substantiated earlier pessimistic outcomes for children with rheumatic diseases. Even earlier studies that emphasized the continuing disease activity and functional problems of young
184
adults growing up with juvenile rheumatoid arthritis QRA) showed that many of these patients completed college, worked full time, and raised chiidren. ll ,I5--17 Recent well-designed studies seem to indicate that JRA is not necessarily a psychosocial stressor, and families of children with JRA are, in general, resilient.!8,19 Although these studies suggest that psychosocial intervention is not needed for every child with arthritis, they also show that some of these children do suffer disabilities, have reduced function and employment status and decreased social acceptance,21) and have overall adjustment problems and internalization of symptoms. 21 Current research efforts suggest that planning for care for these children should be based on new concepts of disablement, should be evidence-based, and should be individualized. New concepts of the "disablement" process include four distinct constructs: active pathology, impairment, functional limitation, and disability.22 Each of these stages offers potential for intervention. In addition, there may be other factors, such as coping skills, access to care, economics, and the family's psychosocial climate, that contribute to this disablement process and therefore are appropriate targets for intervention. In planning for the management of functional and psychosocial disabilities in chronic illness, one must address the issue of why some patients and families are vulnera-
I-, • 1ABLE 8 -I ,-
(olllponenb of M,lIIdgemenl of RlwllIIldli( Oisedses in (hildren
Medical and surgical management Family-centered, community-based, coordinated care (school, outreach) Psychosocial management (social services, mental health services, financial) Musculoskeletal rehabilitation (physical therapy, occupational therapy, orthopedics) Well-child care issues (growth and development, nutrition, immunization, anticipatory guidance) Continuity of care Cost-effective care
C HAP T E R 8
I'll • -
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
185
TEAM CARE
IABLE l'l-2 Steps in Implementing [,unity-Centered, \ (llIlmunity-Based (an'
Tertiary Center Recognizing the pivotal role of the child and the family in the planning of care Developing resources in the community where the child lives Recognizing parents and professionals as equals in a partnership of care Emptlwering the family with information through education and support Encouraging pediatricians to assume a greater role in casecOOirdination, become knowledgeable about available local resources, and work with community agencies Increasing communication among disciplines and from patients to hea'lth professionals Breaking barriers to development of such a system
Pediatric Rheumatologist; Rheumatology Nurse; Allied Services; Other Pediatric Specialists; Special Laboratory; Imaging; etc.
I I
Child and Parent
School .....- - Primary Physician
ble and others are not. 18,19 Based on their well-designed study, Noll and colleagues suggested that "randomly occurring, challenging life events do not alter the child's potential for inclusive fitness by denigrating their social status or emotional well-being."23 One concept to explain this qifference in vulnerability suggests that there are some risk factors that tend to push children with chronic illness and their families to dysfunction and disability, and there are resilience factors that tend to give more stability,24,25 An imbalance between these two groups of factors influences outcomes (Table 8-3). Therefore, in planning for the care of these children, emphasis should be on the child and the family, and efforts should be aimed at not only controlling the disease and managing the current problems, but also at planning for the future. Steps should be taken to improve the resilience of patients and families through better education to cope with the vicissitudes of these diseases, improved social and peer support for the children and their parents, and stress management education to help them cope better with their disease and disability. Accurate diagnosis is the first step. Rheumatic diseases may be acute and explosive in onset, or they may evolve over a period of months and years. Only one organ system may be affected, or several systems may be involved, mimicking several other inflammatory and noninflammatory diseases. It may not be possible to place an accurate diagnostic label at first, and yet life-threatening complications and functional disabilities may have to be managed. Great skill and patience are required to support the families in managing their problems in the face of uncertainties in diagnosis and prognosis. Therein reside the <-~hallenges and pleasures of rheumatology.
I:.
I ABI E H 3
Risk factors Resilience factors
• Figure 8-1
From Wallender JL. Varni VW: Adjustment in children with chronic physical disorders' programmatic research on a disability-stress coping module. hI La Greca AM, Siegel LJ. Wallender JL. Walker CE teds), Stress and Coping in Child Health. New York. Guilford, pp. 279-298, 1991.
Community Agencies
Model of team care.
Expertise should be the cornerstone of the care of children with rheumatic diseases. Comprehensive treatment centers should be based in tertiary-care academic centers. The treatment team (Fig. 8-1) should consist of a pediatric rheumatologist, a nurse specialist, physical and occupational therapists, a social worker, and a psychologist, all working with the child's primary care physician. Consultations with an orthopedic surgeon, ophthalmologist, nutritionist, and dentist should be available when required. The child and family should be the central focus of the team. The roles of the primary care physician and the rheumatologist are listed in Table 8-4. Effective communication between the primary care physician and the specialist, and the availability of one contact person at the tertiary center, are the other essential requirements for comprehensive care. 26 Specially trained nurses have fulfilled the contact function effectively in many pediatric rheumatology centers. 27
PATIENT EDUCATION Children with rheumatic diseases and their parents require education on several issues. These issues are
I! II
TABLE l'l-4
Primary physician
Risk and Resilience
Severity of disease and disability Degree of functional independence Daily hassles and struggles Family's ability to solve problems Social support Coping skills
---.,~~
Pediatric rheumatologist
Role of Physicians in the Care Managenwnt feam
Care of intercurrent illness Immunization Developmental issues Anticipatory guidance Working with the community agencies Working with the school system Overall management of the rheumatic disorder Detailed guidelines and directions for the management of a specific problem Ongoing medical explanation for the management decisions Minor counseling Physical therapy/occupational therapy Monitoring for drug toxicity Referral to other subspecialty
186
C HAP T E R
8 ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
discussed in detail in several publications (Appendix 8-1). The mode of teaching has to vary to suit the needs
and skills of parents. Language barriers, cultural backgrounds, and literacy issues must be kept in mind before such programs are organized. Education about the disease, medications, adverse effects of medications, and therapy programs should take place both individually by the pediatric rheumatologist and in groups as part of parent support groups. The belief system of the family should be explored, and all doubts and questions should be addressed. Information required by parents varies with the time that has passed since diagnosis. It also depends on the developmental needs of the child. Therefore, education must be individualized. z8 Information is easily available on the Internet. However, families need help to receive information from reliable sources, such as the Arthritis Foundation (http://www. arthritis. org), and they need assistance with interpretation of the information gathered. Parents and patients in the United States are encouraged to attend educational sessions organized by the American Juvenile Arthritis Organization (AJAO) for patients, siblings, and parents as part of annual regional and national meetings. Similar organizations also exist in other parts of the World (e.g., United Kingdom, Italy, India).
COUNSELING A trusting relationship is the most important requirement for effective counseling. To be trusted, one must be trustworthy. Open, honest communication with the child and the parents is a major component of this relationship. One must listen carefully before offering advice. Collecting information about a child's illness, behavior, and family dynamics may require many visits and observations. One must listen to what the child and family say and observe what they do. Nonverbal cues must be attended to. While treating the impact of the disease on a child and the attendant weaknesses in the family, one must also be on the lookout for their strengths. This strength may lie in the extended family, an interested schoolteacher, or a loving grandparent. It is important to know the strengths, because one can only build on the strengths while finding ways to compensate for the weaknesses. Some families grow in strength in the presence of adversity. Factors that contribute to successful coping should be supported and encouraged. 24 •29 Children with physical disabilities and deformities and children who look different because of a rash or steroid therapy are bound to feel different and selfconscious. Physical disability and fluctuation in disease activity may make it difficult for these children to participate in social and family activities. 30 Emphasis on what they can do and planning activities in which they can participate should help their sense of self-worth and morale. There should be alternative plans and backup arrangements. Involvement of siblings, friends, and classmates in helping a disabled child during activities should help create a successful experience for everyone.
'11 1:.
IABLE H -5
hI( lors ThaI Influence Adheren(('
Cognitive/emotional (e.g., limited ability to understand, depression) Behavioral (e.g., defiance, adolescent independence) Cultural (e.g., alternative concepts of disease model) Social and family issues (e.g., unstable family) Disease-related (e.g., chronicity) Medication-related (e.g., side effects) Organizational (e.g., appointments in the clinic) Economics (e.g., cost of visit) From Kroll T, Barlow]H. Shaw K: Treatment adherence in juvenile rheumatoid anhrilis: a review. Scand.J Rheumatol 28: 10-18, 1999.
ADHERENCE (COMPLIANCE) Children with chronic illness soon become tired of taking medicines day after day with no end in sight. They often ask why they have to do mindless exercises that do not appear to help them in any way. Therefore, compliance with medication and therapy programs becomes a major issue for children with chronic diseases. 31.3 2 The word "compliance" implies that the physician gives orders and the patient obeys. But the ideal situation is an informed patient who chooses to follow the treatment prescribed by a trusted physician after being convinced of the benefits and made aware of the consequences of not following the treatment. In other words, the patient chooses to adhere to the prescribed program. In pediatrics, one must consider the child, the parent, and the caretaker to make sure the treatment plan is followed. Patient and parent education and incorporation of the needs of family members in planning the care are essential to ensure that the patient adheres to the plan. Listening to the specific needs of a child and family and incorporating them into the plan, as well as designing a plan that accounts for the stresses and strengths of the family and their cultural values, increases the likelihood that the plan will be followed. Factors that affect adherence and strategies to enhance adherence are listed in Tables 8-5 and Table 8-6.
SIBLING ISSUES Stress related to living with chronic illness affects every member of the family, including siblings. The role of siblings and their impact on the developmental needs of the patient may vary with the age and cognitive level of siblings, their perception of the child with chronic illness,
-=..
TABLE 8-6
Slr.llegies 10 fadlildle dud Promole Adlwreme
Informational handouts Cues and reminders Positive feedback Discipline Steps to minimize discomfort and inconvenience Dealing effectively with complaints Monitoring disease process for changes Adapled from Rapoff M: Compliance wilh treatment regimens for pediatric rheumatic diseases. Anhritis Care Res 3: 40-47. 1989.
C HAP T E R 8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
paret}fal attitudes, and the demands placed on unaffected siblings. 33•34 Feelings may range from guilt (that siblings are responsible for the sick child in some magical way) to fear of catching the illness and embarrassment. 35 Siblings may resent the extra time and attention given to the affected child and perceived favoritism in matters of discipline. Realistically, parents may not be able to provide adequate time to satisfy the developmental needs of the siblings. This is a particular problem for single parents. Depending on the parental demands, some siblings may take an important role in the medical treatment of the affected child (such as reminding about medications and helping with therapy); others may be protected from this role by their parents. Some siblings may take a more protective role in school and outside the home. Disease severity, parental functioning, family stress, and family support systems are some of the determinants of the effects of chronic illness on siblings. In spite of ambivalent feelings, siblings of children with arthritis function well in life. 36 With the proper approach, they become a great source of extra support, both at home and at school. It is important to evaluate the needs of siblings in caring for children with chronic illness and to help support their needsY Some ideas to reduce the negative impact on siblings are given in Table 8-7.
SCHOOL ISSUES Rheumatic diseases in general do not affect the ability of the child to learn and think, except for conditions that affect the central nervous system, such as systemic lupus erythematosus (SLE). Children with arthritis can and should attend school except in special circumstances. School is a child's job. However, the school system often must institute several adaptations to the standardized schedule. 5•6 In the United States, children with chronic illness and disabilities must be educated in the "least restrictive environment." Individualized education plans ClEPs) have to be formulated at the parent's request, and there are strong "due process" requirements. 38 Depending on the medical condition and the disability, one or more of the "related school services" listed in Table 8-8 may need to be provided for children with special health care needs to ensure proper educational opportunities. 6,39 Special school services are important components of community-based services, They provide physical adap-
~~ /I TABl~
H- 7 Approd(he~ 10 Siblings of Children with ( hrolllc Illness
Include siblings in clinic visits. Talk with siblings and explore their needs. Determine the perception of the siblings on the impact of the illness on family members. Help them deal with their friends' questions. Encourage and enlist sibling participation in developing care plans. Conduct sibling group discussions at local and national support groups.
It III
1ABLE 8-8 III Children
Related School
Service~ Needed
187
by Chronl<
Administration of medications Implementation of medical procedures Emergency preparations Schedule modifications Modified physical education Transportation Building accessibility Toileting/lifting assistance: support therapies Physical therapy Occupational therapy Speech and language therapy Counseling services (school, career, personal) Data from Walker OK, Jacobs FH: Chronically ill children in schools. In Hobbs N. Perrin J (eds): The Constant Shadow: Issues in Chronic Childhood Illness in America. San Francisco, Jossey-Bass, 1984. Modified from Walker OK: Care of chronically ill children in schools. Pediatr Clin North Am 31: 221, 1984.
tations in schools for handicapped access, elevators, classes on the same floor, and a duplicate set of books. Transportation, school counseling, nutrition, adaptive physical education, and homebound instruction are some of the other services that may be needed. 5.39 Many common concerns expressed by children and parents in relation to school, as well as suggested solutions, are given in Table 8-9, The nurse can provide an individualized checklist for parents (Appendix 8-2); this list can be shared with the school nurse or teacher. School nurses are some of the best advocates for children with disabilities and special needs. Well-informed school nurses can work with teachers and physical education instructors and make appropriate modifications within the school. Therefore, communication between the tertiary center staff members (particularly the nurse) and the school nurse is essential. In addition, special educational programs for school nurses are very useful. It is important to recognize that teachers and parents tend to emphasize issues related to activities of daily living (ADL) as limiting school life, whereas children themselves rate peer acceptance and self-concept as more important. 4o Therefore, these children need more help with peer support and better coping skills. Final1y, if a child is considering vocational training or col1ege after high school, early planning is essential. This planning should start at the beginning of secondary school at the very latest. One of the author's successful "graduates" wrote a checklist in preparation for her entry into college (Table 8-10).
TRANSITION Transition to adult care is a process that should start in pediatric rheumatology centers when a child reaches adolescence. 13.41 Preparation of these children requires attention to functional vocational evaluation and training, independent liVing skills, and self-advocacy. Growing up through adolescence into young adult life is a major task for any child. This time becomes a challenge for children with chronic illness and their parents. 13 Adolescents with chronic diseases have to be encouraged
188
-
C HAP T E R
8 ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
,
II
TABLE 8 9
(ommon SdlOOI (OI11erns for Students with Rheumati( Oiseas('s
DIfficulty
Strategy
Inactivity, stiffness due to prolonged sitting
Sit at side or back of room to allow walking around without disturbing class Change position every 20 minutes Ask to be assigned jobs that require walking (e.g., collect papers) Request elevator permit Schedule classes to decrease walking and climbing Request extra time getting from classes Use wheelchair if needed Keep two sets of book: one in class, one at home Have a buddy help carry books Get a backpack or shoulder bag for books Determine cafeteria assistance plan (helper, reserved seat, wheeled cart) Request an easel-top desk or special chair Use "fat" pen/pencil, crayons Use felt-tip pen Stretch hands every 10 minutes Use tape recorder for note taking Photocopy classmate's notes Use computer for reports Request alternative to timed tests (oral test, extra time, computer) Educate teacher (messy writing may be unavoidable at times) Wear loose-fitting clothing Wear clothes with Velcro closures Get adaptive equipment from occupational therapist Modify locker or request alternative storage place Use lockers with key locks instead of dials Devise alternative signaling method
Climbing stairs or walking long distances
Carrying books or cafeteria tray
Getting up from desk Handwriting (slow, messy, painful)
Shoulder movement and dressing Reaching locker Raising hand
From Raising a Child with Arthritis: A Patient's Guide. Atlanta: Arthritis Foundation, 1998.
to take control of their disease management gradually. This requires preparation of the family and child before adolescence. Both physicians and parents have to let go of the child in a sensitive and gradual way, The parent has to trust, and the child must prove that he or she can be trusted to take care of ongoing management and needs. Adolescent support groups with profeSSional leadership may be helpful. Some of the subjects that should be
( ' . TABI E S I I)
RheulIl
Visit the college before choosing, in order to evaluate the walking distance between buildings, stairs, and elevators and for general accessibility. Know the different climate changes (e,g" if you feel better in the summer, you may want to pick a campus with a warm climate). Also, climate changes can cause flare-ups or increased stiffness, It is always good to get to know a local doctor in your college area or health service before troubles occur, so that he or she knows your past history and can help you right away if there are any problems. If you have the chance to pick your oWn schedule during your first year, make sure you give yourself enough time to get from one class to another (e.g., if you have to walk to buildings from one end of campus to the other), Also try not to overburden yourself with classes and too many credit hours-there will always be bad days, so expect them. If you are not a morning person or if you are a "night owl," schedule your classes in the afternoon, And always-no matter how "bad" the food is-eat as regularly as possible: breakfast, lunch, and dinner, It helps in the long run, even if your stomach does not think so at the time, Take extra medicine with you at the start of school and give yourself time to find the nearest pharmacy.
addressed, both in individual sessions and in group discussions, are preparing for college, sexuality, alcohol and drugs, and vocational planning.
FINANCIAL ISSUES Children with chronic illness account for a large proportion of health care expenditures in the United States,~.42 For instance, families of children with rheumatic diseases may spend hundreds to thousands of unreimbursed dollars out of pocket per year,4,7.8 not including time lost from work. In the current competitive environment, children with chronic illness and disabilities are particularly vulnerable. The high cost that goes with chronic illness, and the pressures to cut costs, may make it difficult to proVide adequate and appropriate care for children with chronic illness and disabilitiesY In the United States, families need to be educated about various types of coverage and how to work with health maintenance organizations (HMOs) and insurance companies. Families need to learn about child welfare systems and social security benefits. They also have to learn to work with their school systems. Parents must be advocates for their children and learn both their own rights and responsibilities and those of their children.3~ Both parents and physician..'i need to work through the political process to bring about changes in financing of medical care that will ensure access to appropriate services for all children with disabilities. 43 A special program developed by the AJAO teaches several skills to parents so that they can become advocates for their children. In this program, a parent and a health professional who works with the child learn to
C HAP T E R
8 ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
list items of care essential for that specific child and learn strategies to achieve those goals.
UNCONVENTIONAL REMEDIES With scientific medicine truthfully acknowledging its ignorance about the etiology of rheumatic diseases and the lack of a cure, it is easy for parents to believe those who promise miracles. This is also the age of alternative medicine. 44 There are pressures from well-meaning friends and relatives to try unproven remedies widely advertised in newspapers and on the Internet. Southwood and colleagues 45 reported that 70% of patients with juvenile arthritis had used unconventional remedies. Even if it is not discussed by patients, it can be assumed that most of them have tried or are trying one or more remedies. It is better to keep an open and noncritical relationship with patients and their family members so that they feel comfortable talking about these remedies. It is important for pediatric rheumatologists to be aware of the currently available alternative remedies, so they can provide proper guidance when patients ask about such methods. This is an opportunity to educate parents on the conduct of scientific studies and to explain to them the difference between controlled trials and testimonials. It is better to let them try some remedies that are innocuous, caution about some potentially dangerous treatments (e.g., megavitamins), and refuse to be part of certain other approaches (e.g., bee-sting therapy). It is always wise to allow room for parents to come back without losing face and feeling humiliated.
f
II
TABLE 8-1 I
189
Principles of Therapy
Relieve pain Cheat, cold, splint). Stretch individual joints. Train coordination of opposing muscles. Re-integrate physiologic movement patterns. Re-educate to improve posture and gait. Improve activities of daily living: Use of traction and/or prone position to reduce hip flexion contracture Use of resting splints for wrist support Shoe inserts and modifications From Hackett J, Johnson B, Parkin A, Southwood T: Physiotherapy and occupational therapy for juvenile chronic arthritis: custom and practice in five centres in the UK, USA and Canada. Br J Rheumatol 35: 695-699, 1996; Scull SA, Dow MB, Athreya BH: Physical and occupational therapy for children with rheumatic diseases. Pediatr Clin North Am 33: 1053---1077, 1986.
tions for referral to therapists are listed in Table 8-12. If more aggressive therapy is required, an outpatient program is indicated with two or three sessions a week. Even with this frequency, carryover into the daily routine is essential to maintain the benefits. Occasionally, children require intensive inpatient rehabilitation, which is becoming increasingly difficult to finance in the United States. Fortunately, the need for such inpatient rehabilitation is becoming less frequent thanks to better medical management. Children whose disability is serious enough to interfere with attendance at a regular school may be able to receive school-based therapy that follows an IEP.
PHYSICAL AND OCCUPATIONAL THERAPY The overall goals of rehabilitation are to maximize function, to prevent deformities, and to help a child achieve developmental milestones-physical, psychosocial, emotional, educational, and vocational. The concept is to help the child and family lead as normal a life as possible. Goals must be set in collaboration with the parents and the child. One has to account for the needs of the family, their strengths and weaknesses, their economic and human resources, their coping styles, and their cultural values. The child's developmental level and interests also must be acknowledged. 46 Physical and occupational therapy programs are vital to the management of rheumatic diseases in children. A variety of modalities and treatment approaches are used without adequate proof of their efficacy. For example, it is not known whether a resting splint to prevent dysfunctional positioning of the wrist during sleep is better than a functional splint worn during activities to prevent misalignment of the wrist joint. Consequently, there are differences of opinion and approachY General principles of therapy agreed upon by most experts are given in Table 8-11. Needs for therapy services may vary with the specific rheumatic disease, whether disabilities are acute or chronic. For minor problems and prevention of contracture, standard exercises to maintain range of motion (ROM) can be taught to parents by the pediatric rheumatologist and the nurses (Figs. 8-2 to 8-11). Usual indica-
• Figure 8-2 Neck. Tum the head toward one shoulder and then the other. Repeat two to three times on each side. (From Raising a Child with Arthritis: A Parent's Guide. Atlanta, Arthritis Foundation, 1998.)
190
I
C HAP T E R
TABU M 12
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
Inditdtions lor Refell,,1 10 1herdpish
Involvement of multiple joints Severe pain or stiffness of joints and muscles Established contracture, limitation in activities of daily living (even single area) Preoperative management Postoperative management Rapidly progressive disease Prescription for assistive devices (e.g., crutches, wheelchair) Failure to respond to standard exercise Parent-child conflict resulting in poor compliance with therapy
Essential components of treatment should include rest, management of fatigue and pain, posture and positioning, therapeutic exercises, and improving ADL,41l-50 Conditioning exercises and sports activities are discussed later.
Rest It is preferable for children with arthritis to be as active
as possible. General bed rest is rarely prescribed except for children with severe arthritis or myositis and for children with systemic disease associated with myocarditis or pericarditis. In the author's practice, children
~ A
c
• Figure 8-3 Shoulder. A, Lie on the floor with both arms at your sides. Raise one arm over the head, keeping the elbow straight, until the back of the hand reaches the floor. Return the arm slowly to the side. Repeat this exercise with the other arm. Repeat, alternating arms, two to three times. B, Shoulder abduction. Start with the arms down at the sides with palms facing out. Raise the arms out to the sides and up until palms touch, keeping elbows straight. Hold briefly, then return arms to the sides. Repeat two to three times. C, Shoulder rotation. lying down, place arms straight out from shoulder, palms toward ceiling. Bend at the elbow with the fingers pointing toward ceiling. Rolls arms forward so the hands point straight down toward feet (internal rotation). Roll arms backward so the hands point toward the head (external rotation). Repeat two to three times. (From Raising a Child with Arthritis: A Parent's Guide. Atlanta, Arthritis Foundation, 1998.)
• Figure 8-4 Elbow. A, Lie on floor with both arms at sides, palms facing the ceiling. Bring hands to shoulders by bending elbows. Return hands to the floor by straightening elbows. B, Bend elbows and hold them into the sides of the body with forearms parallel to the floor and palms down. Slowly tum forearms so palms face ceiling. Hold to the count of three, and tum arms so palms face the floor again. (From Raising a Child with Arthritis: AParent's Guide. Atlanta, Arthritis Foundation, 1998.)
A
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
.--
191
• Figure 8-5 Wrist. A, With the forearms resting firmly on atabletop and the hand hanging over the edge of the table, bend the wrist up as far as possible. Bend the wrist down as far as possible. Hold. Repeat two to three times. B, Plac.e the hand on atable or other flat surfac.e. Raise elbow toward ceiling until wrinkles appear at the wrist. C, Grasp the hand with the opposite hand. Put the palms together, with fingers around the opposite hand. Push the hand backward, stretching the wrist. Hold. Repeat two to three times. (From Raising a Child with Arthritis: AParent's Guide. Atlanta, Arthritis Foundation, 1998.)
~--~~,....---" 1"
, , I
,\\'; ."".. 11 •• - , .-If '-'
A
B
c are allowed to set their own limits based on pain and endurance, with the suggestion that, if necessary, they allow themselves a rest period during the day, usually immediately after school. Localized rest for an acutely inflamed joint is provided with a splint or a bivalved cast. If a splint or cast is applied, the limb should be taken out once or twice a day and ROM exercises or isometric exercises should be performed.
Children with functional disorders and chronic pain syndromes complain of fatigue that interferes with school attendance and participation in sports. This fatigue is often associated with nonrestorative sleep. Pharmacologic and behavioral approaches are needed to help promote sleep and thereby relieve fatigue. Fatigue out of proportion to disease activity deserves close scrutiny to differentiate between medical and psychological causes and may require cognitive and behavioral therapy.
Fatigue
Pain
Fatigue is a common problem for children with rheumatic diseases. It is a particular complaint for children with systemic diseases, anemia, and myocarditis. A defined period of rest during the middle of the day is ideal, although it is not always practical. Treatment of the systemic disease and anemia often corrects the fatigue.
Pain is a major component of many of the inflammatory musculoskeletal conditions. 51 Children with chronic arthritis often do not complain of pain because they become used to living with a certain level of constant pain. In addition to control of inflammation and use of analgesics, other modalities, such as biofeedback and transcutaneous
192
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
a I,)
• Figure 8-6 Angers. A, Place hand on atable or flat surface with fingers together. Then, separate the fingers as widely as possible and hold. Repeat two to three times. 8, Curl the fingers tightly while keeping the knuckles straight. Complete the fist by bending the knuckles, then open the hand wide. Repeat two to three times. (From Raising a Child with Arthritis; A Parent's Guide. Atlanta, Arthritis Foundation, 1998.)
B
electrical nelVe stimulation (TENS), may have to be tried, depending on the clinical diagnosis, acuteness or chronicity of pain, the child's developmental status, various effects of the pain on function, and psychosocial issues. Pain caused by an acutely inflamed joint is treated with joint aspiration, intra-articular injection of glucocorticoid (if appropriate), and use of nonsteroidal anti-inflammatory dmgs. In addition, the joint can be placed in a resting splint for a few days until the inflammation subsides. The splint should be removed two or three times a day and the joint moved through ROM actively or passively to avoid atrophy of muscles and joint stiffness. children with myositis and pain need to rest and to be allowed to move to their tolerance. An active exercise program should not be started until pain and tenderness subside. Pain out of proportion to swelling of the joint needs careful evaluation to mle out other diagnoses (e.g., leukemia). In patients with JRA or SLE, severe pain or sudden onset of pain in one joint may indicate septic arthritis. Pain after activity is not uncommon in children with rheumatic diseases. Standard treatment is rest and ice, and an occasional analgesic. Children should be encouraged to be as active as possible and to set their own limits. The author's guideline is that if the joint pain after activity
lasts more than 45 to 60 minutes, the activity level or duration should be decreased.
Chronic Pain Chronic pain is a primary manifestation of rheumatic diseases in general. A unique constellation of nociceptive, affective, cognitive, and behavioral factors interact to make the pain experience more intense for some childrenY It is also important to remember that chronic pain, whether organic in origin or unexplained, can lead to depression, and, conversely, occult depression can manifest as chronic pain. 53 Children with rheumatic diseases may also experience cycles of intense and intractable pain not amenable to a pharmacologic approach. Some of these children experience secondary fibromyalgia with such classic features as tender points and lack of sleep. This pattern affects their emotional stability and function and leads to a vicious circle. In these children, a cognitive-behavioral approach may be indicated. 54 Other techniques, such as TENS, visual imagery, relaxation, and management of time and stress, may be helpful. Approaches to pain control in regional pain, ovemse, and pain amplification syndromes are discussed in appro-
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
193
• figure 8-7 Hlp and knee. Lying on the back, bend one knee toward the chest, then lower. Repeat with the other knee. Repeat two to three times with each leg. (From Raising a Child with Arthritis: A Parent's Guide. Atlanta, Arthritis Foundation, 1998.)
• Figure 8-8 Back. Lie on the back with knees bent. Keep the back flat against the floor. Raise both bent knees toward the chest. Place hands behind thighs and pull toward the chest. Lower legs to original position. (From Raising a Child with Arthritis: AParent's Guide. Atlanta, Arthritis Foundation, 1998.)
/~
B~=m
A
• Figure 8-9 A, Thigh. Lie on the back with one leg bent at the knee and one leg straight. Raise the straight leg, keeping the knee as straight as possible. Keep the small of the back on the floor. Lower the leg and repeat with the other leg. B, Knee. Lying on the stomach, bend one knee, bringing the heel toward the buttocks.Then, lower the leg and repeat with the other knee. Repeat two to three times with each leg. (From Raising a Child with Arthritis: A Parent's Guide. Atlanta, Arthritis Foundation, 1998.)
c;;:----_-------,,' ----_..........- _~.--. "~~
_----
.....
A
c
D
• Figure 8-10 Hlp. A, Lie flat on the back with legs straight, about six inches apart. Roll the legs in and out, keeping the knees straight. Repeat two to three times. B, Lie flat on the back with the legs straight, about 6 inches apart. Slide one leg out to the side and retum. Repeat with the other leg. Repeat two to three times with each leg. C, Lying on the stomach, lift one leg. Try to keep the knee straight.Then, lower the leg and repeat with the other leg. Repeat two to three times with each leg. D, Lying on table with knees bent over the edge, bring one knee up to the chest. At the same time, keep the other flat on the table. Hold for a count of 10. Lower the leg. Repeat with the other leg. (From Raising a Child with Arthritis: AParent's Guide. Atlanta, Arthritis Foundation, 1998.)
194
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
• Figure 8-11 A, Ankle. Sit in a
chair with feet on the floor. Rrst, keeping the heels down, lift the toes up as high as possible.Then, keeping the front of the feet on the floor, lift the heels up as high as possible. Rnally, tum the soles of both feet toward each other, then tum them away from each other. Repeat each movement two to three times. 8, Lower leg (calf). Standing an arm's length away from the wall, place both hands on the wall, above the head. Place one leg straight back, keeping the foot flat on the floor and the knee straight; the forward leg should be bent at the knee. Hold until a pull is felt in the back of the straight leg and count slowly to 10. Repeat two to three times with each leg. e, Lower leg. This exercise is performed by the parent or other caregiver with the child lying on the back with legs straight. Place your hand under the child's heel. Grasp the foot and lean the lower part of your arm against the sole of the foot. Bend the foot toward the knee, but do not use too much pressure. With the other hand, grasp the leg between the ankle and knee to steady the leg. Hold for a count of 10 or 15. Repeat with the other leg. (From Raising a Child with Arthritis: AParent's Guide. Atlanta, Arthritis Foundation, 1998.)
l
priate chapters. It is important to remember that emotions, cultural factors, and family dynamics play a major part in pain perception. Therefore, exclusively medical management of pain in these syndromes is unlikely to be successful. This is particularly true in reflex neurovascular dystrophy. 51 ,55 For all of these reasons, it is important to obtain a complete developmental and psychosocial history in children with chronic pain, particularly in children for whom repeated examinations and laboratory results are negative. Children with chronic pain present a management challenge because of the psychosocial factors and the time needed to care for them. Referral to a pain clinic, psychologist, or psychiatrist is sometimes needed.
Posture/Positioning Children with arthritis assume a number of dysfunctional postures unconsciously as their bodies adapt to the pain and limitation of ROM. It is important to remind them to sit with a straight back and shoulders braced. This is particularly important for children with spondyloarthropathies. Attention should be given to the school chair to ensure that good posture is maintained. The position for reading books or work on the computer should provide for good neck and wrist positions. Children with cervical spine involvement should sleep with a single pillow or a very soft pillow. Resting knee or wrist splints may be indicated for use at night to prevent poor positioning of these joints during sleep (Fig. 8-12). Lying
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
195
Lower-extremity resting splint.This posterior shell is wom during the hours of sleep to rest the knees in extension. (From Scull SA, Dow MB, Athreya BH: Physical and occupation therapy for children with rheumatic diseases. Pedlatr Oin North Am 33:1067, 1986, with permission.) • figure .... 12
prone at least 20 minutes a day may help prevent hip flexion contractures (Fig. 8-13).
therapeutic Exerdses Therapeutic exercises serve several functions. 43-50,56 They may be aimed at improving ROM of joints, strengthening muscles, or promoting conditioning and endurance. 56 There are several types of exercises. 50,57 In active exercises, the patient is asked to move the joint. These exercises are useful to stretch and strengthen muscles (e.g., touching toes to stretch hamstrings [Fig. 8-14)). Passive exercises (active-assistive) are performed by the therapist and are useful when there is a flare of arthritis, when a joint has lost motion, and in young children. Resistive exercises are used to strengthen muscles; for example, resistance is applied as the joint moves through its range. Stretching exercises, such as those for the treatment of tight heel cords or hip flexion contracture, are done prefer:lbly by the therapist. Strengthening exercises may be isometric or isotonic. If the muscle is contracted without permitting joint motion, it is isometric. Quadriceps setting for an acutely inflamed joint is an isometric exercise. Isotonic exercises performed against gravity along with ROM and resistive exercises are useful to strengthen muscles. A child should master ROM exercises and muscle strengthening exercises before proceeding to endurance! conditioning exercises and sports activities (Fig. 8-15).
• Figure .... 14 Lower-extremity stretching exercises. Active exercises to stretch hamstrings (A); iliopsoas (B); and heel cords (C and 0). (From Scull SA, Dow MB, Athreya BH. Physical and occupation therapy for children with rheumatic diseases. Pediatr elin North Am 33:1072, 1986.)
Recreational
~ Aerobic Isotonic Isometric Range of motion and stretching
• FIpre .... 13 Prone time. Resting in the prone position on a firm sUrface
is used to stretch hip and knee flexion contractures. (From Scull SA, Dow MB, Athreya BH: Physical and occupation therapy for children with rheumatic diseases. Pediatr elin North Am 33:1058, 1986, with permission.)
Children with arthritis should begin with range-ofmotion exercises. They may progress upward on the pyramid as their disease regresses or is controlled medically. • Figure .... 15 Pyramid of graded progress in an exercise program. (From Hicks IE: Exercise in patients with inflammatory arthritis. Rheum Dis elin North Am 16: 857, 1990.)
196
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
Exercises should be done every day, either in the morning or in the evening, for at least 20 to 30 minutes. This routine may be difficult to accomplish, particularly in the morning in a family in which both parents work and there are other children. It is best for a child to take a warm bath before performing exercises. The parents and child (particularly the adolescent) should be educated on the importance and the techniques of performing home therapy exercises. The therapist should demonstrate the exercise and let the parents practice under supervision. A written description with figures (see Figs. 8-2 to 8-11) is useful. Exercises should be age-appropriate and should accommodate the family's daily schedule. Young children are easily bored with exercises, and insistence by parents on a rigid routine may lead to parent-ehild conflicts. Therefore, efforts should be made to incorporate these exercises into games, recreational activities, and play (Table 8-13).
Adlvltles of Dally Living (ADL) ADL include tasks in self-care, dressing, feeding, and hygiene. Age-appropriate independence in these activities may be difficult for children with arthritis. For children who have difficulties with actions such as turning doorknobs or opening jars, simple and more efficient techniques to perform these activities and other joint protection techniques must be taught. For more severe conditions, adaptive devices may be needed (Fig. 8-16). In certain situations, modifications to the home may be necessary, particularly for children in wheelchairs.
Sports and Exercise Children with rheumatic diseases fatigue easily and have reduced cardiopulmonary reserve. S8 Several factors limit the exercise capacity of these children, including the severity of involvement of joints and muscles, involvement of the cardiovascular or the respiratory system, and anemia. In addition, psychologic factors S9 and parental concerns may add to the lowered physical fitness of these children. 60 Involving children in age-appropriate games and play activities is an ideal way to maintain ROM of joints, increase muscle strength, improve conditioning, and boost self-esteem. Better compliance is an added bonus. Various age-appropriate activities are listed in Table 8-13. Young children can ride in their toy cars or tricycles (Fig. 8-17). By adjusting the height of the seat and making sure they do not slide up and down, ROM of the knee can be improved to gain increased flexion or extension. By reversing the handle, extension of the wrist can be increased (Fig. 8-18). School-aged children should be encouraged to participate in physical education classes to their tolerance. Adapting gym classes and allowing children to set their own limits should encourage participation. However, activities that involve weight bearing or stress on affected joints, such as headstands, somersaults, chin-ups, and handstands, should be avoided. The trampoline is usually
b_ .. lAB. [ I i 13 lor Parl'll!s
Play fxl'rdsl's by Agl' Grou,): A GUllk
Alles 6 mo-3 ,.. Playing in warm water with toys-to move all joints and help pain Toys that pull apart (e.g., pop beads)-to strengthen hand~ and arms Tickle bottoms of feet-to bend knees and hips Put plastic ring over ankle and leg and take it off Pushing and catching a balloon or soft beach ball-to strengthen arms Kicking a balloon or soft beach ball-to bend knees and strengthen legs Squeeze and roll clay-to strengthen hand and bend wrist Tricycle or toys to scoot-to strengthen leg muscles and move the joints of the legs
Ages 4-8,.. Warm bath and playing with toys in the water-to move all joints and help pain Pieces of elastic to pull into funny shapes-to strengthen arms and hands. (Theraband is a thin elastic sheet that can be drawn on and pulled into funny shapes; it is available from physical therapy or dental supply houses.) Any crafts the child likes-to strengthen and move joints of arms and hands Clay to squeeze and roll-to strengthen hand and bend wrist Tricycle or bicycle riding-to move leg in many positions Swimming classes----total body exercise
Alles 9-13,..
/
Swimming-total body exercise Crafts----to strengthen and move joints of arms and hands Bicycling-leg joint movement and strengthening of leg muscles School activities (including gym)-total body movement. (Note: Although children with juvenile rheumatoid arthritis are usually encouraged to take part in gym to their tolerance, there may be times when it is not advisable, or your physician may suggest avoiding specific gym activities. Ask your physician whether your child should be restricted from playing contact sports, doing headstands, etc.) Hitting and catching softballs or volleyballs-arm strength Ballet-total body movement
Alles 13 ,.. and Older By this age, the teenager should be helping to plan the exercise activities; parents, teen, and staff can talk together about what the teen likes to do and what joints need extra moving. Adapted from Giesecke LL, Athreya BH, Doughty RA: Home Care Guide on Juvenile Rheumatoid Althritis (for Parents). Atlantic City, N.J, Children's Seashore House, 1985: Scull SA, Dow MIl, Athreya BH: Physical and occupational therapy for children with rheumatic diseases. Pediatr Clin North Am 33: 1060, 1986.
contraindicated. Contact sports and activities that generate torsional forces on involved joints (e.g., downhill skiing) or repeated impacts should be avoided in children with severe arthritis. In general, cycling and swimming are safe and beneficial for most children with arthritis.
Therapeutic Modalities Heat, cold, splinting, bracing (orthotics), and water exercises are some of the modalities used in the treatment of rheumatic diseases.
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
197
• Rgure 8-17 Child in hot-wheels cyde. The seat and the pedals can be adjusted to increase flexion or extension at the knee.
• FIg-.e 8-16 Dressing aids: a dressing stick (A); sock aid (8); and coat hanger "hoop' (C and fJJ are used to train this child for independence. (From Scull SA, Dow MB, Athreya BH: Physical and occupation therapy for children with rheumatic diseases. Pediatr Clin North Am 33:1064, 1986.)
Heat and Cold Both intra-articular temperature and blood flow are increased in inflamed joints. Consequently, the metabolism of articular tissues is altered. 61 However, studies on the value of heat and cold in altering the intra-articular temperature and blood flow and in relief of symptoms have yielded inconclusive results. Based on an extensive review of the literature, Karen Hayes62 came to several conclusions, which are described in the following paragraphs. Both heat and cold appear to be effective in managing pain, stiffness, and limitation of ROM. Heat may be more effective for joint motion and cold more effective for pain reduction. Therefore, a patient with acute disease may benefit from cold as well as from heat; patients with chronic disease may benefit from heat. Preference of the patient may be the best determinant of the modality used. Either deep or superficial heat may be used. A warm tub bath or shower in the morning helps reduce stiff-
• Rgure 8-18
Reversed handle bars force wrist joints into extension.
198
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
ness. A heated pool (88' F to 92' F [31' C to 33' CD is ideal for therapeutic exercises,63 but one must be careful to avoid vasodilatation and hypotension. Children with moderate to severe anemia often do not tolerate warm pools. Moist packs may be useful to relieve muscle spasm. Deep heat in the form of paraffin baths may be beneficial, particularly for the small joints of the hand. Deep heat application, such as diathermy or ultrasound, is not usually recommended for children because of potential damage to the cartilage. Cold packs and ice packs are ideal for acute swelling and pain and for postexercise pain.
Splints Splints, braces, and orthotic devices are used for various purposes, including support for acutely inflamed joints (resting splints), maintenance of joints in functional positions during activity (dynamic splints) (Fig. 8-19), stretching of contracted joints (corrective splints), and provision of shock absorption (shoe inserts). Splints may be made from a variety of compounds such as plaster of Paris, polypropylene, or fiberglass. Materials to be used
depend on the purpose, need for repeated adjustments, and cost.
Resting Splints Resting splints are used to rest acutely inflamed joints. A joint is maintained in a functional position in the splint for most of the day. The splint is removed once or twice a day to exercise the muscles and move the joint through its ROM, if possible. Resting splints are often used to maintain position and prevent deformities at the wrists, knees, and ankles. The most commonly used splints are wrist splints (Fig. 8-20), to maintain the wrist in a functional position during the night, and knee splints (see Fig. 8-12), to prevent knee-flexion contractures. If a knee-flexion contracture is greater than 20 degrees, it may be necessary to use serial casting (corrective splint) or dynamic splinting. Although cervical collars may not be totally efficacious to prevent motion at the atlantoaxial joint, children with cervical spine involvement may benefit from use of a firm collar during car rides.
Functional Splints Functional splints are designed to support joints during ADL and to reduce stress. For example, a molded anklefoot orthosis (AFO) may be used to maintain the stretch of the Achilles tendon during weight bearing. A shoe insert may be used to decrease pain during weight bearing. Some splints produce a constant stretch to the muscles during walking and functioning (dynamic splints). Biomechanical orthotics are small, custom-made devices, such as shoe inserts, that are designed to correct alignment or to relieve pain.
Water Exercises Therapy in water (pool or whirlpool) is one of the best and most acceptable forms of therapy for children with
• Figure 8-19
Dynamic splint.
• Figure I-ZO Wrist cock-up splint. Aresting splint for wrist and fingers. (From Scull SA, Dow MB, Athreya BH: Physical and occupation therapy for children with rheumatic diseases. Pediatr elin North Am 33:1067, 1986.)
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
arthritis,6.>,64 The buoyancy of water facilitates weight bearing, Water also can be used to provide resistance to muscle movement. Children can walk in water up to the chest to reduce the load on their hips. A heated pool may be useful to reduce stiffness. Finally, swimming itself is a great exercise, both for ROM and for aerobic conditioning. Water therapy is ideal for children with severe morning stiffness and for postoperative rehabilitation.
Shoe Modification Citing their detailed analysis of feet in JRA, Truckenbrodt and associates65 defined eight major deformities (Table 8-14). The most common are pes planovalgus due to involvement of subtalar joints, pes cavus due to inflammation of the intertarsal joints, and pseudocavus due to inflammation of the upper ankle joint and the talonavicular joint. Soft, comfortable footwear with good arch support (e.g., sneakers), is recommended for most common problems. Cushioning with a foam insole may reduce the force of impact. For children with metatarsal pain, metatarsal bars or rocker-bottom soles may help with push-off.
III,· Il:i.i II
199
TABLE 8-15 Strategies for Managing Various Prohlems Associated with Rheumatic Diseases
Problem
Strategy
Morning stiffness
Sleep in a sleeping bag Early-morning shower Pool therapy Therapeutic exercises Active Active-assistive (passive) Resistive Aerobic Posture Splint Brace Stretch Strengthening exercises Active Faradic electrical stimulation Functional training Adaptive devices Shoe inserts Functional orthoses Heel or shoe lift
Decreased range of motion
Position
Muscle weakness
Limitation in activities of daily living Painful feet Leg-length differences Courtesy of S. Bowyer, MD.
Postsurgical Therapy
IMMUNIZATION
The therapist should be involved from the beginning if surgery is planned, Therapists are in a position to recommend the goals of surgery because they have worked with the child and the child's personality and have reviewed the disability and its current functional limitations. Children should be taught to thoroughly exercise the muscles they will be asked to use increasingly after surgery. Early mobilization is essential to prevent loss of motion. Positioning and splinting or use of a passive ROM machine may be required to maintain newly acquired motion. Postoperative therapy may include active and passive ROM, water therapy, and gradual increase in weight bearing. 5o ,66 General strategies to manage various problems encountered in patients with rheumatic diseases are given in Table 8-15.
There are two major questions related to immunization and rheumatic diseases. First, is there any relationship between immunization and onset or exacerbation of these diseases? Second, what are the recommendations for children with rheumatic diseases who are taking various medications? Although some reports suggest that immunization may precipitate an exacerbation or initiation of an arthritic or vasculitic disorder,67-71 other studies do not support this association. n ,7'> In the only available study on the relationship between influenza immunization and arthritis in children, Malleson and colleagues7.> did not find exacerbation of arthritis associated with the influenza virus vaccination. The same study documented that antibody responses were satisfactory even in children receiving glucocorticoid therapy, At present, it appears that the benefits of immunization far outweigh any risk of exacerbation of the disease in most situations,74 Children receiving chronic salicylate therapy and all children with SLE, dermatomyositis with significant muscle weakness, systemic scleroderma with cardiopulmonary or renal disease, or systemic vasculitides may benefit from yearly influenza virus (killed virus) vaccination. Studies also indicate that patients taking glucocorticoids and immunosuppressive drugs respond to influenza vaccine73,75 and pneumococcal vaccine76 with adequate antibody titers. However, such immunization should not give rise to a false sense of security, because immunity is not guaranteed. Children with SLE and splenic hypofunction should receive the pneumococcal vaccine. 76,n The author usually encourages families to keep the regular schedule of immunizations while cautioning them about the possibility of a flare-up. There are, however, a Jew speCial circumstances and precautions:
' Si • -
I ABLE 8-14 Most Frequent Deviations of the Foot in lllvt'nile Chronic Arthritis: Various Combinations Are Possible
Heelfoot (pseudocavus) Pes planovalgus Pes C;WlIS Heelfoot (pseudocavus) Hallux flexus/rigidus Hallux valgus Forefoot adduction Claw toes Hammer toes From Truckenbrodt H, Hafner R, von A1tenbockum c: Functional joint analysis of the foot in juvenile chronic arthritis, Clin Exp Rheumatol 12 (Suppl 10): 591,
1994.
200
C HAP T E R 8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
Active Disease Children with severe, active rheumatic disease should not receive any immunization.
Varicella Varicella can be a major problem for children receiving immunosuppressive therapy, glucocorticoids, methotrexate, and/or biologic agents, For all of these children, suggested management for the prevention of varicella is given in Table 8-16. Varicella vaccination is generally contraindicated in children taking glucocorticoids in doses of 2 mg/kg/day equivalent of prednisone or its equivalent, or 20 mg/day of prednisone or equivalent for longer than 14 days.78 For children receiving smaller doses, the risklbenefit ratio must be assessed. Salicylate should not be used for at least 6 weeks after varicella vaccine administration. The potential for Reye's syndrome in children treated with salicylates in association with varicella or influenza has been widely discussed. 79
Immunosuppressive Therapy Children undergoing immunosuppressive therapy or glucocorticoid therapy should not receive any live vims vaccine. Recommendations made by the Committee on Infectious Diseases of the American Academy of Pediatrics should be followed for these children. 78 The British Society of Rheumatology recommends the' use of influenza A, meningococcus C, haemophilus B, hepatitis B, and tetanus toxoid but warns that the response may be suboptimaJ.74 For adults undergoing glucocorticoid therapy, there is no consensus on what dose is low enough to be considered safe. If glucocorticoids and cytotoxic dmgs have been stopped, live vims vaccines may be given after a minimum of 3 months. It is also important to remember that the new nasal spray vaccine for influenza contains live vims and is therefore contraindicated in immunocompromised patients and their contacts. Only the inactivated parenteral form of the influenza vaccine should be used. Children receiving intravenous immunoglobulin should wait for at least 3 months after the last dose before any immunization to ensure an adequate immune response. 80
I!.
TABLE H 16 V.uicella Prevention Str,ltegy for Children Taking Glmo
Document successful vaccination in the past: measure serum antibody level. If seronegative (susceptible), immunize with varicella vaccine 3 weeks before starting therapy. Susceptible children exposed to varicella should receive varicellazoster immunoglobulin within 72 hI' after exposure, If chicken pox develops, treat with oral or parenteral acyclovir depending on severity and spread; stop Enbrel and methotrexate temporarily; but continue glucocorticoids,
REFERENCES 1. Hobbs N, Perrin 1M: Issues in the Care of Children With Chronic Illness: A Source Book on Problems, Services and Policies. San Francisco, losey-Bass, 1985. 2. Pless lB, Power C, Peckham CS: Long-term psychosocial sequelae of chronic physical disorders in childhood. Pediatrics 91: 1131-1136, 1993. 3. McAnarney ER, Pless IB, Satterwhite B, Friedman SB: Psychological problems of children with chronic juvenile arthritis, Pediatrics 53: 523-528, 1974. 4. McCormick MC, Stemmler MM, Athreya BH: The impact of childhood rheumatic diseases on the family. Arthritis Rheum 29: 872--879, 1986. 5. Lovell DA, Athreya BH, Emery HM, et al: School attendance and pallerns, special services and special needs in pediatric patients with rheumatic diseases. Arthritis Care Res 3: 196, 1990. 6. Walker DK: Care of chronically ill chlldren in schools. Pediatr Clin North Am 31: 221-233, 1984. 7. AIlaire SH, DeNardo BS, Szer IS, et al: The economic impact of juvenile rheumatoid arthritis. j Rheumatol 19: 952-955, 1992. 8. Newacheck PW, Taylor WR: Childhood chronic illness: prevalence, severity, and impact Am 1 Public Health 82: 364-371, 1992. 9, Gare BA, Fasth A: The natural history of juvenile chroniC arthritis: a population based cohort study. n: Outcome. 1 Rheumatol 22: 308-319, 1995. 10. Wallace CA, Levinson lE: luvenile rheumatoid arthritis: outcome and treatment for the 1990s. Rheum Dis Clin North Am 17: 891-905, 1991. 11. Peterson LS, Mason T, Nelson AM, et al: Psychosocial olllcomes and health status of adults who have had juvenile rheumatoid arthritis: a controlled, population-based study. Arthritis Rheum 40: 2235-2240, 1997. 12. Packham lC, Hall MA: Long-term follow-up of 246 adults with juvenile idiopathic arthritis: functional outcome. Rheumatology 41:1428-435, 2002. 13. Rettig 1', Athreya BH: Leaving home: preparing the adolescent with arthritis for coping with independence and the adult rheumatology world. In Isenberg D, Miller .1.1 (eds): Adolescent Rheumatology. London, Martin Dunitz, 1998, 14. Brewer Ellr, McPherson M, Magrab PR, Hutchins VL: Family-centered, community-based, coordinated care for children with special healthcare needs. Pediatrics 83: 1055-1060, 1989. 15. Miller 11 3rd: Psychosocial factors related to rheumatic diseases in childhood. .I Rheumatol 20 (Suppl 38): 1-11, 1993. 16. Packham lC, Hall MA: Long-term follOW-LIp of 246 adults with juvenile idiopathic arthritis: education and employment. Rheumatology 41: 1436-14.W, 2002. 17. Packham lC, Hall MA: Long-tenn follow-up of 246 adults with juvenile idiopathic arthritis: social function, relationships and sexual actiVity. Rheumatology 41: 1440-1443, 2002. 18. Routh DK: Commentary: juvenile rheumatoid arthritis as a stressor. .I Pediarr Psychol 28: 41--43, 2003. 19. Dahlquist LM: Commentary: are children with lRA and their families at risk or resilient? 1 pediatr Psychol 28: 43-46, 2003. 20. Reiter-Purtill 1, Gerhardt CA. Vannatta K, et al: A controIled longitudinal study of the social functioning of children with lRA. j Pediatr Psychol 28: 17-28, 2003. 21. LeBovidge .IS, Lavigne lV, Donenberg GR, Miller ML: Psychological adjustment of children and adolescents with chronic arthritis: a meta-analytiC review. j Pediatr Psychol 28: 29-39, 2003. 22. van der Netl, Prakken AB, Helders 1'1, et al: Correlates of disablement in juvenile chronic arthritis: a cross-sectional study. Hr j Rheumatol35: 91-100, 19%. 23. Noll RB, Kozlowski K, Gerhardt C, et al: Social, emotional, and behavioral timctioning of children with juvenile rheumatoid arthritis. Arthritis Rheum 43: 1387-1396, 2000. 24. von Weiss RT, Rapoff MA, Varni JW, et al: Daily hassles and sodal support as predictors of adjustment in children with pediatric rheumatic diseases. .I Pediatr Psycho! 27: 155-165, 2002. 25. Wallender lL. Varni VW: Adjustment in children with chronic physical disorders: programmatic research on a disability-stress coping module. In La Greca AM, Siegel Ll, Wallender jL, Walker CE (eds): Stress and Coping in Child Health. New York, Guilford, pp 279-298, 1991. 26. Mervyn FA: "They get this training but they don't know how you feel." Horsham, UK, National Fund for Research into Crippling Diseases, 1975. 27, Athreya BH: Regionalized arthritis resources. Arthritis Rheum 20 (Supp1): 604, 1977. 28. Rosenstock 1M, Strecher Vj, Becker MH: Social learning theory and the Health Belief Model. Health Educ Q 15: 1751-1783, 1988, 29. Akikusa JD, Allen RC: Reducing the impact of rheumatic diseases in childhood. Best Pract Res Clin Rheumatol 16: 333-345, 2002. 30. Sallfors C, Fasth A, Hallberg LR: Oscillating between hope and despair: a qualitative study. Child Care Health Dev 28: 495-505, 2002. 31. Rapoff MA, Lindsley CB, Christophersen ER: Parent perception of problems experienced by their children in complying with treatments for juvenile rheumatoid arthritis. Arch Phys Mecl Rehabil 66: 427--429, 1985. 32. KroIl T, Barlow lH, Shaw K: Treatment adherence in juvenile rheumatoid arthritis: a review. Scand ) Rheumatol 28: 10-18, 1999. 33. Swartz DR: Dealing with ~hronic illness in childhood. Pediatr Rev 6: 67, 1984. 34. Gallo AM, Breitmayer Bj, Knafl KA, Zoeller LH: Stigma in childhood chronic illness: a well sibling perspective. Pediatr Nurs 17: 21-25, 1991. 35. Birenbaum A: On managing a courtesy stigma. 1 Health Soc Behav 11: 196, 1970. 36. Daniels D, Miller 11 3rd, BiIllngs AG, Moos RH: Psychosocial functioning of siblings of children with rheumatic disease. j Pediatr 109: 379-383, 1986.
C HAP T E R 8 ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN 37. Williams PO, Wiliams AR, Graff ]C, et al: A community-based intervention for siblings and parents of children with chronic illness or disability: the ISEE study, .J Pediatr 143: 386-393, 2003. 38. Cassidy ]T, Lindsley CB: Legal rights of children with musculoskeletal disabilities. Bull Rheum Dis 45: 1-5, 1996. 39. Spencer CH, Fife RZ, Rabinovich CEo The school experience of children with arthritis: coping in the 1990s and transition into adulthood. Pediatr Clin North Am 42: 1285-1298, 1995. 40. Taylor ], Passo MH, Champion VL: School problems and teacher responsibilities in juvenile rheumatoid arthritis.] Sch Health 57: 186-190, 1987. 41. White PH: Success on the road to adulthood: issues and hurdles for adolescents with disabilities. Rheum Dis Clin North Am 23: 697-707, 1997. 42. NefflM, Anderson G: Protecting children with chronic illness in a competitive marketplace.]AMA 274: 1866-1869, 1995. 43. American Academy of Pediatrics, Committee on Children with Disabilities: Managed care and children with special health care needs: a subject review. Pediatrics 102: 657, 1998. 44. Jonas WB: Alternative medicine: learning from the past, examining the present, advancing to the future. ]AMA 280: 1616-1618, 1998. 45. Southwood TR, Malleson PN, Roberts-Thomson P], Mahy M: Unconventional remedies used for patients with juvenile arthritis. Pediatrics 85: 150-154, 1990. 46. Hafner R, Truckenbrodt H, Spamer M: Rehabilitation in children with juvenile chronic arthriti~. Baillieres Clin Rheumatol 12: 329-361, 1998. 47. Hackett], Johnson B. Parkin A, Southwood T: Physiotherapy and occupational therapy for juvenile chronic arthritis: custom and practice in five centres in the UK, USA and Canada. Br] Rheumatol 35: 695-699, 1996. 48. Scull SA, Dow MB, Athreya BH: Physical and occupational therapy for children with rheumatic diseases. Pediatr Clin North Am 33: 1053-1077, 1986. 49. Emery HM, Bowyer SL: Physical modalities of therapy in pediatric rheumatic dise'dses. Rheum Dis Clin North Am 17: 1001-1014, 1991. 50. Hicks JE, Nichols ]], Swezey RL: Handbook of Rehabilitative Rheumatology. Atlanta, American Rheumatism Association, 1988. 51. Varni .TW. Bernstein BH: Evaluation and management of pain in children with rheumatic diseases. Rheum Dis Clin North Am 17: 985, 1991. 52. Rapoff MA , Lindsley CB: The pain puzzle: a visual and conceptual metaphor for understanding and treating pain in pediatric rheumatic disease, ] Rheumatol 27 (Suppl 58); 29-33, 2000. 53. McGrath P], Unruh AM: Pain in children and adolescents. New York, Elsevier Amsterdam, 1987, pp 289-316. 54. Waleo GA, Varni .TW, Ilowite NT: Cognitive-behavioral pain management in children with juvenile rheumatoid arthritis. Pediatrics 89: 1075-1079, 1992. 55. Sherry DO, Weisman R: Psychologic aspects of childhood reflex neurovascular dystrophy. Pediatrics 81: 572-578, 1988. 56. Rhodes V]: Physical therapy management of patients with juvenile rheumatoid arthritis. Phys Ther 71: 910-919, 1991. 57. Basmajian]V: Therapeutic Exercises, 4th ed. Baltimore, Williams & Wilkins, 1984, pp 303-308. 58. Giannini M], Protas E]: Aerobic capacity in juvenile arthritis patients and healthy children. Arthritis Care Res 4: 131-135, 1991. 59. Malleson PN, Bennett SM, MacKinnon M, et al: Physical fitness and its relationship to other indices of health status in children with chronic arthritis. J Rheumatol 23: 1059-1065, 1996. 60. Klepper SE, Darbee], Effgen SK, Singsen BH: Physical fitness levels in children with polyarticular juvenile rheumatoid arthritis. Arthritis Care Res 5: 93-100, 1992.
201
61. Castor CW, Yaron M: Connective tissue activation. YIn: The effects of temperature studied in vitro. Arch Phys Med Rehabil 57: 5-9, 1976. 62. Hayes KW: Heat and cold in the management of rheumatoid arthritis. Arthritis Care Res 6: 156-166, 1993. 63. McNeal RL: Aquatic therapy for patients with rheumatic disease. Rheum Dis Clin North Am 16: 915-929, 1990. 64. Campion MR: Hydrotherapy in Pediatrics, 2nd ed. Oxford, ButterworthHeinemann, 1991. 65. Truckenbrodt H, Hafner R, von Altenbockum C: Functional joint analysis of the foot in juvenile chronic arthritis. Clin Exp Rheumatol 12 (Suppl 10): S91-S96, 1994. 66. Swann M: The surgery of juvenile chronic arthritis: an overview. Clin Orthop 259: 70-75, 1990. 67. Benjamin CM, Chew GC, Silman A]: Joint and limb symptoms in children after immunisation with measles, mumps, and rubella vaccine. BMJ 304: 1075-1078, 1992. 68. Institute of Medicine: Adverse effects of pertussis and rubella vaccines. In Howson CP, Howe CJ, Fineberg HV (eds): A Report of the Committee to Review the Adverse Consequences of Pertussis and Rubella Vaccines. Washington, DC, National Academy Press, 1991. 69. Tingle AJ, Allen M, Petty RE, et al: Rubella-associated arthritis: I. Comparative study of joint manifestations associated with natural rubella infection and RA-27/3 rubella immunisation. Ann Rheum Dis 45: 110-114, 1986. 70. Castresana-lsla C], Herrera-Martinez G, Vega-Molina ]: Erythema nodosum and Takayasu's arteritis after immunization with plasma derived hepatitis B vaccine.] Rheumatol 20: 1417-1418, 1993. 71. Mader R, Narendf'dn A, Lewtas], et al: Systemic vasculitis following influenza vaccination: report of 3 cases and literature review. J Rheumatol 20: 1429-1431, 1993. 72. Ray P, Black S, Shinefield H, et al: Risk of chronic arthropathy among women after rubella vaccination. Vaccine Safety Datalink Team. ]AMA 278: 551-556, 1997. 73. Malleson PN, Tekano ]L, Scheifele OW, Weber ]M: Influenza immunization in children with chronic arthritis: a prospective study. ] Rheumatol 20: 1769-1773, 1993. 74. Davies K, Woo P, British Paediatric Rheumatology Group: Immunization in rheumatic diseases of childhood: an audit of the clinical practice of British Paediatric Rheumatology Group members and a review of the evidence. Rheumatology 41: 937-941, 2002. 75. Park CL, Frank AL, Sullivan M, et al: Influenza vaccination of children during acute asthma exacerbation and concurrent prednisone therapy. Pediatrics 98: 196-200, 1996. 76. Battafarano OF, Battafarano N], Larsen L, et al: Antigen-specific antibody responses in lupus patients following immunization. Arthritis Rheum 41: 1828-1834, 1998. 77. Lipnick RN, Karsh J, Stahl Nl, et al: Pneumococcal immunization in patients with systemic lupus erythematosus treated with immunosuppressives. J Rheumatol 12: 1118-1121, 1985. 78. Report of the Committee on Infectious Diseases, 26th ed. Elk Grove Village, IL, American Academy of Pediatrics, 2003, pp 69-81. 79. Hurwitz ES, Barrett M], Bregman 0, et al: Public Health Service study of Reye's syndrome and medications: report of the main study. ]AMA 257: 1905-1911, 1987. 80. Rowley AH, Shulman ST: Current therapy for acute Kawasaki syndrome. ] Pediatr 118: 987-991, 1991.
202
C HAP T E R
8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
APPENDIX 8-1 RESOURCES FOR FURTHER INFORMATION ON RHEUMATIC DISEASES FOR PATIENTS AND PARENTS 1 1. American Juvenile Arthritis Organization Resource Catalog (l998)-Arthritis Foundation 1330 W. Peachtree Street, Atlanta, GA 30309 USA http://www.arthritis.org Check on Juvenile Authirtis
6. National Dissemination Center for Children with Disabilities P,O, Box 1492 Washington, DC 20013 USA http://www.nichcy,org
2. Lupus Foundation of America, Inc. 1300 Piccard Drive, Suite 200 Rockville, MD 20850 USA http://www,lupus.org
7. On TRAC-Taking Responsibility for Adolescent/Adult Care British Columbia's Children's Hospital Room 2, D20 4480 Oak Street Vancouver, BC V6H 3V4 Canada http://www.acsa-caah.ca/pdjlanglon_trac.pdf
3. Scleroderma Foundation 12 Kent Way, Suite 101 Byfield, MA 01922 USA http://www.scleroderma.org 4. Spondylitis Association of America 14827 Ventura Boulevard, Suite 119 PO Box 5872 Sherman Oaks, CA 91403 USA http://www,spondylitis.org 5. Family Village-A great Web site for information on all chronic diseases and rare syndromes. Gives information for resources in England also. http://wwwfamilyvillage.wisc,edu I
Verified December 2004
MONOGRAPHS/BOOKS 1. Raising a Child with Arthritis: A Parent's Guide.
Atlanta, Arthritis Foundation, 1998. 2, Tucker LB, Denardo BA, Stebulis JA, Schaller JG. Your Child with Arthritis: A Family Guide for Caregiving. Baltimore, The John's Hopkins University Press, 1996.
C HAP T E R 8
ApPROACH TO MANAGEMENT OF RHEUMATIC DISEASES IN CHILDREN
203
APPENDIX 8-2 EXAMPLE OF A SCHOOL COMMUNICATION FORM PEDIATRIC RHEUMATOLOGY PROGRAM
(Date)
(Date of Birth)
(Student's Name)
_______is followed by Rheumatology for . This chronic disease is characterized by joint . We recommend the following swelling, pain and stiffness and fatigue. In addition, this student has services be provided by the school: r:l Modified physical education to include: r:l Activities to tolerance, allow student to set own limits r:l No contact sports r:l Swimming is encouraged r:l No repeated stress or pounding to affected joints r:l No weight bearing on wrists and arms, such as handstands, cartwheels, rings r:l Adaptive physical education r:l Two (2) sets of books r:l Use of elevators if available r:l Extra time between classes r:l Provision for locker on each floor if applicable r:l Modified assignments to include use of tape recorder r:l Computer r:l Extended time lines for completion of assignments and tests r:l Use of oral tests r:l No timed tests r:l No grades for handwriting r:l Door-to-door transportation r:l Physical and Occupational Therapist evaluations to plan for accessibility and adaptation needs in school environment (as needed and on a quarterly basis) r:l We request that home instruction be implemented if more than two (2) consecutive school days are missed. r:l IEP that includes all of the above and ongoing aT and PT r:l Other: _ We are enclosing a booklet for teachers. Attached please find a list of common school problems and solutions experienced by students with arthritis. Thank you in advance for your assistance and cooperation regarding this student. Sincerely,
Attending Rheumatologist IEP, Individualized education plan; OT, occupational therapy; PT, physical therapy.
Rheumatology Nurse Specialist
C H A P T E R
9
C HRONIC A RTHRITIS
IN
C HILDHOOD
James T. Cassidy and Ross E. Petty
Chronic arthritis in childhood—characterized as juvenile rheumatoid arthritis (JRA) (Table 9–1), juvenile chronic arthritis (JCA) (Table 9–2), or juvenile idiopathic arthritis (JIA) (Table 9–3)—is one of the most common rheumatic diseases of childhood. It is also one of the more frequent chronic illnesses of children and an important cause of short- and long-term disability. Although it has been customary to refer to this disorder as a single disease, it almost certainly comprises a number of similar entities, characterized principally by arthritis of the appendicular joints, each of which has distinct modes of presentation and may have the same or different causes. It may also be considered as a closely related series of host-determined specific responses with an immunoinflammatory pathogenesis, possibly activated by contact with an external antigen or antigens. A dauntingly complex immunogenetic predisposition is often present. The relative frequencies of the three principal onset types (oligoarthritis, polyarthritis, and systemic onset) have varied considerably in published series and reflect the biases that confound estimates of incidence and prevalence (Table 9–4).1,2 There are also potential ethnic differences in onset types.3 Clinical aspects of these onset types are discussed in detail in Chapters 10, 11, and 12.
Classification Chronic arthritis in childhood is a complex area of study and investigation, not least because of inconsistencies of definition and terminology. It is genetically heterogeneous, phenotypically diverse in presentation and during the course of the disorder, and substantially without a single pathognomic diagnostic approach. The classification of childhood arthritis has been problematic for decades.4,5 In the 1970s, two sets of criteria were proposed to classify chronic arthritis in childhood: those for JRA, developed and validated by a committee of the American College of Rheumatology (ACR),6 and those for JCA, published by the European League Against Rheumatism (EULAR).7 Later, a third classification (for JIA) was proposed by the Pediatric Task Force of the International League of Associations for Rheumatology (ILAR).8 These three classifications are compared in Table
206
9–5 and Table 9–6. Problems in applying these criteria include the necessity to exclude other diseases for which there are no validated diagnostic or classification criteria for children and the fact that all three were based on populations of northern European derivation.
ACR Criteria for Classification of Juvenile Rheumatoid Arthritis The ACR criteria have been widely used, validated, and revised (see Table 9–1),6,9–11 but they are applicable primarily to white American children. They defined the age limit in children, the duration of disease necessary for a diagnosis, and the characteristics of the arthritis (Fig. 9–1). Three types of onset were recognized: oligoarticular (pauciarticular), polyarticular, and systemic. The requirement that age at onset of arthritis be less than 16 years was a criterion based more on practice patterns than on age-related biologic variation in disease. Furthermore, although persistent objective arthritis in one or more joints for 6 weeks was sufficient for diagnosis, as determined by computer-assisted statistical analysis, a duration of at least 6 months was required before the onset type could be certain (unless characteristic systemic features were present). The type of onset is defined by a constellation of clinical signs present during the first 6 months of illness. Oligoarticular onset is defined as arthritis in four or fewer joints. Polyarticular onset is defined as arthritis in five or more joints. In determination of the onset type, each joint is counted separately, except for the joints of the cervical spine, carpus, and tarsus; each of these structures is counted as one joint. Systemic-onset JRA is characterized by a daily (quotidian or intermittent) fever spiking to greater than 39˚ C for at least 2 weeks in association with arthritis of one or more joints. Most children with systemiconset disease also have a characteristic rash, and many have other evidence of extra-articular involvement, such as lymphadenopathy, hepatosplenomegaly, or pericarditis. Nine course subtypes were identified during long-term follow-up.
EULAR Criteria for Classification of Juvenile Chronic Arthritis In 1977, at the EULAR conference on the Care of Rheumatic Children in Oslo, the term juvenile chronic arthritis (JCA) was proposed for the heterogeneous group of disorders that present as chronic arthritis in childhood (see Table 9–2).7 The onset
C H A P T E R
IN
CHILDHOOD
TABLE 9–1 Criteria for the Classification of Juvenile
TABLE 9–3 Proposed Classification Criteria for Juvenile
Rheumatoid Arthritis
Idiopathic Arthritis: Durban, 1997
1. Age at onset <16 yr 2. Arthritis (swelling or effusion, or presence of two or more of the following signs: limitation of range of motion, tenderness or pain on motion, and increased heat) in one or more joints 3. Duration of disease 6 wk or longer 4. Onset type defined by type of disease in first 6 mo: a. Polyarthritis: ≥5 inflamed joints b. Oligoarthritis (pauciarticular disease): <5 inflamed joints c. Systemic-onset: arthritis with characteristic fever 5. Exclusion of other forms of juvenile arthritis Modified from Cassidy JT, Levinson JE, Bass JC, et al.: A study of classification criteria for a diagnosis of juvenile rheumatoid arthritis. Arthritis Rheum 29: 274–281, 1986.
TABLE 9–2 Criteria for a Diagnosis of Juvenile Chronic Arthritis 1. 2. 3. 4.
9 C H R O N I C A RT H R I T I S
Age at onset <16 yr Arthritis in one or more joints Duration of disease 3 mo or longer Type defined by characteristics at onset: a. Pauciarticular: <5 joints b. Polyarticular: >4 joints, rheumatoid factor negative c. Systemic: arthritis with characteristic fever d. Juvenile rheumatoid arthritis: >4 joints, rheumatoid factor positive e. Juvenile ankylosing spondylitis f. Juvenile psoriatic arthritis
From European League Against Rheumatism (EULAR) Bulletin 4. Nomenclature and Classification of Arthritis in Children. Basel, National Zeitung AG, 1977.
types of JCA and age of onset were defined according to the ACR criteria. The disorder must have been present for at least 3 months, and other rheumatic diseases must have been excluded. Included within the JCA classification, however, are juvenile ankylosing spondylitis (JAS), psoriatic arthropathy, and arthropathies associated with inflammatory bowel disease. The only substantial differences between the ACR and the EULAR criteria are (1) inclusion of JAS, psoriatic arthritis, and the arthritis of inflammatory bowel disease and (2) restriction on use of
1. Systemic 2. Oligoarthritis a. Persistent b. Extended 3. Polyarthritis (rheumatoid factor negative) 4. Polyarthritis (rheumatoid factor positive) 5. Psoriatic arthritis 6. Enthesitis-related arthritis 7. Undifferentiated arthritis a. Fits no other category b. Fits more than one category From Petty RE, Southwood TR, Baum J, et al: Revision of the proposed classification criteria for juvenile idiopathic arthritis: Durban, 1997. J Rheumatol 25: 1991–1994, 1998.
the term juvenile rheumatoid arthritis to children with arthritis and rheumatoid factor (RF) seropositivity, although no definition of RF seropositivity is provided.
ILAR Criteria for Classification of Juvenile Idiopathic Arthritis In 1993, the Pediatric Standing Committee of ILAR proposed a classification of the idiopathic arthritides of childhood (see Table 9–3). This classification8 and its subsequent revisions (Durban, Edmonton),12–14 were developed with the aim of achieving homogeneity within disease and categories. The designation undifferentiated arthritis includes conditions that, for whatever reason, either do not meet criteria for any other category or meet criteria for more than one category. These JIA criteria still require validation and consensus and will almost certainly be modified further as new evidence for pathogenesis becomes available.15–26 For the student of pediatric rheumatology, differences in nomenclature require care in interpreting the literature, because the terms JRA, JCA, and JIA are often incorrectly used as if they were interchangeable and synonymous. This dilemma has been the subject of several publications.4,16,27–29 Throughout this text, every attempt has been made to ensure that the terms JRA, JCA, and JIA accurately reflect the cited publications. If more than one set of criteria may pertain (e.g., use of the term juvenile
TABLE 9–4 Characteristics of Chronic Arthritis in Children by Type of Onset Characteristic
Polyarthritis
Oligoarthritis (Pauciarticular Disease)
Systemic Disease
Percent of cases Number of joints involved Age at onset
30 ≥5 Throughout childhood; peak at 1–3 yr 3:1 Systemic disease generally mild; possible unremitting articular involvement 5%
60 ≤4 Early childhood; peak at 1–2 yr 5:1 Systemic disease absent; major cause of morbidity is uveitis 5–15%
10 Variable Throughout childhood; no peak 1:1 Systemic disease often self-limited; arthritis chronic and destructive in half Rare
10% (increases with age) 40–50% Guarded to moderately good
Rare 75–85%* Excellent except for eyesight
Rare 10% Moderate to poor
Sex ratio (F:M) Systemic involvement Occurrence of chronic uveitis Frequency of seropositivity Rheumatoid factors Antinuclear antibodies Prognosis *In girls with uveitis.
207
208
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
TABLE 9–5 Comparison of Classifications of Chronic Arthritis in Children Juvenile Rheumatoid Arthritis (ACR)
Juvenile Chronic Arthritis (EULAR)
Juvenile Idiopathic Arthritis (ILAR)
Systemic Polyarticular Oligoarticular (pauciarticular)
Systemic Polyarticular Juvenile rheumatoid arthritis Pauciarticular Juvenile psoriatic arthritis Juvenile ankylosing spondylitis
Systemic Polyarticular RF-negative Polyarticular RF-positive Oligoarticular Persistent Extended Psoriatic arthritis Enthesitis-related arthritis Other arthritis
ACR, American College of Rheumatology; EULAR, European League Against Rheumatism; ILAR, International League of Associations for Rheumatology; RF, rheumatoid factor.
TABLE 9–6 Characteristics of the ACR, EULAR, and ILAR Classifications of Chronic Arthritis in Children Characteristic
ACR
EULAR
ILAR
Onset types Course subtypes Age at onset of arthritis Duration of arthritis Includes JAS Includes JPsA Includes inflammatory bowel disease Other diseases excluded
3 9 <16 yr ≥6 wk No No No
6 None <16 yr ≥3 mo Yes Yes Yes
6 1 <16 yr ≥6 wk Yes Yes Yes
Yes
Yes
Yes
ACR, American College of Rheumatology; EULAR, European League Against Rheumatism; ILAR, International League of Associations for Rheumatology; JAS, juvenile ankylosing spondylitis; JPsA, juvenile psoriatic arthritis.
arthritis) or a compromise is in order, the designation chronic arthritis is used.
HISTORICAL REVIEW The concept that inflammatory polyarthritis occurred in childhood was first suggested in 1864 by Cornil,30 who described a 29-year-old woman who had had chronic inflammatory arthritis since the age of 12. Diamant-Berger31 reviewed the subject in 1890 and included 35 previously published cases and 3 of his own. He commented on the acute onset of disease, the predominant involvement of large joints, a course characterized by exacerbations and remissions, frequent disturbances of normal growth, and a generally good prognosis. Still32 presented the classic description of chronic childhood arthritis in 1897, while he was a medical registrar at the Hospital for Sick Children, Great Ormond Street, London. He pointed out that the disease almost always began before the second dentition, was more frequent in girls, and was usually of insidious onset. The acute onset of disease in 12 patients who had lymphadenopathy, splenomegaly, and fever was described in detail. Serositis characterized by pleuritis and pericarditis was common, although rash was not noted. Still observed that there was often no articular pain and that children exhibited a marked tendency to early contracture and muscle atrophy. The cervical spine was affected in the majority of cases, often during the early stages of the disease. Still suggested that childhood arthritis might have a different etiology from that of rheumatoid arthritis or might include more than one disease. This classic description is an outstanding example of bedside observation.33 Today, the acute systemic onset of JRA is even now sometimes
■ Figure 9–1 The joints of the wrists and hands of a 21⁄2-year-old boy with systemic-onset disease are swollen, warm, and painful.The proximal and distal interphalangeal joints are erythematous.There are flexion contractures of the fingers. (See color insert.)
called Still’s disease. French authors have often used the term syndrome de Chauffard-Still for chronic arthritis with lymphadenopathy and splenomegaly. In 1901, Hirschsprung34 confirmed Still’s observations that a chronic articular disease was associated with lymphadenopathy and splenomegaly in young children; he also noted the occurrence of hepatomegaly. In 1939, Atkinson35 published a review of 118 cases of Still’s disease, 86 of whom were patients with severe arthritis, lymphadenopathy, and splenomegaly. In the excellent review of Edstrom,36 only 3 of 65 children with chronic arthritis had marked lymphadenopathy and splenomegaly, and Bille37 found no examples of “Still’s disease” among his 65 patients with chronic childhood arthritis. Other large series that are well documented include studies by Colver,38 Holzmuller,39 Coss and Boots,40 Pickard,41 and Lockie and Norcross.42 Monographs on the subject were published by Wissler in 194243 and by Françon in 1946.44 Coss and Boots40 did not view Still’s disease as an independent clinical entity and indicated that the term juvenile rheumatoid arthritis should be used to refer to all cases of idiopathic inflammatory arthritis. Dawson45 supported this view but stressed that children differed from adults by the severity and frequency of the systemic symptoms and by interference with normal growth and development. Many early authors stressed that JRA could begin as a type of monarthritis that most frequently affected the knee and could persist in one joint for the duration of the illness or for several years before other joints were involved.36,37,39,41
C H A P T E R
Stoeber46 summarized experience at the Rheumakinderklinik in Garmisch-Partenkirchen from 1952 to 1976 in 1654 children ranging in age from 10 months to adolescence. Until Colver published the first follow-up studies of JRA in 1937,38 the prognosis in this disease was considered to be poor.47,48 Extended periods of observation of patients experiencing onset in childhood led some early authors to conclude that severe destruction of cartilage occurred in many children and that ankylosis would supervene in most.49,50 Subsequent studies indicated that the mortality rate was in the range of 7% to 9%.41,42 However, most children recovered, often without significant disability.36,41,42 In Edstrom’s study,36 87% of patients seen early in the course of their disease experienced good functional recovery; even 65% of those reviewed after the first or second year of disease had a good prognosis. However, only 39% of children seen later than 2 years after onset returned to normal function. A long, uninterrupted period of active disease was significantly associated with a poor prognosis; only 6% of children whose disease became quiescent within the first 4 years experienced severe disability. Sury51 identified 151 patients from 1920 to 1948 who had a chronic form of arthritis with onset before the age of 15 years. Thirty-nine percent of referred patients and 19% of the children from Copenhagen were severely disabled. The peak age at onset in 100 girls was between 2 and 4 years. In 8 of 51 boys, onset was during the first year of life. Of 41 patients whose disease began with monarthritis, 23 had persistent disease in that one joint for at least the first year. At necropsy, 12 children had a verrucous endocarditis.
EPIDEMIOLOGY Chronic arthritis in children is not a rare disease, but the true frequency of its occurrence is not known (see Chapter 1).52 In historical reviews, between 2.7% to 5.2%
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
209
of all patients with “rheumatoid arthritis” (RA) had onset before 15 years of age.53 The disorder has been described in all races and geographic areas, although its incidence and prevalence vary considerably throughout the world,4,54,55 which partly reflects the ethnicity, immunogenetic susceptibility, and environment of the population under study.56,57 The possibility of interaction of environmental triggers is most clearly evident in systemic-onset disease, with studies supporting seasonal variation in incidence.58–61 Oen and Cheang54 published a meticulous review of the epidemiology of childhood arthritis. Considered as confounding variables were the diagnostic criteria employed, disease exclusions, source of the data (population, clinic, practitioner, health survey), geographic origin, race, and years of observation. No significant differences were found for diagnostic criteria or duration of the study. However, prevalence was higher for population-based studies and for data from North America, whereas clinic-based studies were more homogeneous in results. Andersson Gäre reviewed and summarized the epidemiology and outcome of children with JCA in Sweden compared with reports from other centers.62
Incidence The incidence of chronic arthritis has varied from 2 to 20 per 100,000 (Table 9–7).53,54,59,63–84 A clinic survey in Michigan from 1960 to 1970 calculated a minimal incidence of 9.2 per 100,000 children at risk per year.64 Estimates in Finland ranged from 6 to 8 per 100,000 in Laaksonen’s study53 to 18.2 per 100,000 (95% confidence interval [CI], 10.8 to 28.7) in the study of Kunnamo and colleagues67 and others.85 In Norway,80 the incidence was
TABLE 9–7 Studies of Incidence and Prevalence of Chronic Arthritis in Children Authors (ref. no.) Group I Towner et al. (65) Peterson et al. (77) Mielants et al. (70) Andersson Gäre (62) Manners & Diepeveen (74) Kaipiainen-Seppanen & Savolainen (75) Ozen et al. (79) Kiessling et al. (82) Group II Gewanter et al. (66) Kunnamo et al. (67) Prieur et al. (68) Arguedas et al. (81) Group III Laaksonen (53) Bywaters (63) Sullivan et al. (64) Rosenberg (69) Denardo et al. (72) Oen et al. (59) Malleson et al. (73) Symmonds et al. (76) Fujikowa & Okuni (78) Moe & Rygg (80)
Origin
Year
Diagnostic Criteria
Incidence (per 100,000 Children/yr)
Prevalence (per 100,000 Children)
USA USA Belgium Sweden Australia Finland
1983 1996 1993 1994 1996 1996
EULAR, ACR ACR EULAR EULAR EULAR ACR
10.8–13.9 11.7 — 10.9 — 14
83.7–113.4 86.1–94 167 86.3 400 —
Turkey Germany
1998 1998
EULAR EULAR
— 3.5
64 20
USA Finland France Costa Rica
1983 1986 1987 1998
ACR ACR EULAR EULAR
— 18.2 1.3–1.9 6.8
16–43 — 8–10 31.4
Finland UK USA Canada USA Canada Canada UK Japan Norway
1966 1968 1975 1990 1994 1995 1996 1996 1997 1998
English English ACR ACR ACR ACR ACR EULAR ACR EULAR
6–8 — 9.2 5–8 4 5 8 10 0.83 22.6
75–100 60–70 65 39.7 — 32 40 — — 148.1
ACR, American College of Rheumatology; EULAR, European League Against Rheumatism; Group I, predominantly population-based; Group II, surveys of medical practitioners; Group III, clinic-based.
210
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
22.6 per 100,000. However, 42% of these children were positive for the human leukocyte antigen B27 allele (HLA-B27). The incidence of chronic arthritis in children varied depending on the classification criteria employed (EULAR versus ILAR).86 Data from the Mayo Clinic in Olmstead County, Minnesota, reported an incidence of 13.9 per 100,000 (95% CI, 9.9 to 18.8) during the period 1960–1979.65 A more recent study from the same center indicated that the frequency of this disorder decreased from 15 per 100,000 during 1960–1969 to 7.8 per 100,000 for 1980–1993.77 Seasonal variation was also apparent in this investigation, in Sweden,62 in one Canadian study,59 and in Finland.84 This trend, however, was not confirmed in a larger study from Canada.60
Prevalence Prevalence rates for chronic arthritis in childhood have varied from 16 to 150 per 100,000 (see Table 9–7).54,83 Bywaters63 found the prevalence of “Still’s disease” in English school children to be approximately 65 per 100,000. In the Tecumseh Community Health Survey of 2000 children between 6 and 15 years of age,87 10% complained of joint pain, slightly less than 10% related a history of previous joint swelling, and 5% had morning stiffness, although only two children were found to have chronic arthritis on examination, a prevalence close to that estimated by Bywaters. The 10-year weighted cumulative prevalence per 100,000 in Michigan was 46 for girls and 19 for boys.64 The prevalence was 86.3 per 100,000 in the Swedish study62 and 83.7 to 113.4 per 100,000 (95% CI, 69.1 to 196.3) in Minnesota.65 In Norway,80 the point prevalence was 148.1 per 100,000. As a number of studies suggest, these figures undoubtedly represent minima. The survey by Manners and Diepeveen74 in Australia of 12-year-old school children reported a prevalence of 400 per 100,000 based on an examination of each child included in the survey. Clearly, rheumatic symptoms, past or present, are not uncommon in children and far exceed the estimated prevalence when objective criteria are used and an examination is performed by an experienced pediatric rheumatologist.1,88 Published data are difficult to compare because of varying referral patterns,2 the heterogeneity of the disease, its evolution over time, differences in classification criteria, dissimilarity of source populations, and variable case ascertainment.56 Substantial geographic and ethnic differences are present in regard to age at onset, relative frequencies of onset types, and immunologic markers.
Age at Onset Chronic arthritis in children was arbitrarily defined as arthritis beginning before the age of 16 years. Onset before 6 months of age is distinctly unusual; however, the age at onset is often quite early, with the highest frequency occurring between 1 and 3 years of age, although it varies considerably depending on onset type.36,40,42,51,53,64,89–93 The distribution of ages at onset of arthritis for 300 children in whom the definition of JRA conformed to that of the ACR criteria is in Figure 9–2.64 The peak age at onset was between 1 and 3 years for
■ Figure 9–2 Age at onset of JRA in 300 children: total group (-▲-), girls (-●-), and boys (-●-). For the total group and for girls, a large peak is observed at 1 to 2 years. A bimodal distribution with peaks at 2 years and at 8 to 10 years suggests heterogeneity of arthritis in boys. (From Sullivan DB, Cassidy JT, Petty RE: Pathogenic implications of age of onset in juvenile rheumatoid arthritis. Arthritis Rheum 18: 251, 1975.)
the total group and for girls, but for boys this relationship was much less impressive.
Sex Ratio Twice as many girls as boys are affected by chronic arthritis. Marked differences in this ratio are apparent in the various onset types. These observations suggest either that there are three (or more) different diseases included under the classification for arthritis affecting primarily the appendicular skeleton, or that disease expression is modified by sex chromosome–determined factors (see Table 9–4).
Geographic and Racial Distribution The incidence and prevalence data outlined previously were derived primarily from American or northern European white populations. There are few comparable data for other geographic or racial groups.94,95 Nonetheless, suggestions of racial disparity in frequency exist.54,96 Schwartz and colleagues97 concluded that the proportion of African-American children with JRA in a referral clinic population in the United States was consistent with their representation in the population served. However, there was a striking under-representation of this ethnic group in young children with oligoarticular and polyarticular onsets. Some reports suggest that chronic arthritis in children and adults is less frequent in African than in European populations; in Nigeria, however, the proportion of all patients with onset of chronic inflammatory arthritis in childhood may be somewhat higher.98,99 Lower frequencies of JRA have been reported in children of Japanese, Filipino, or Samoan origin than in whites living in Hawaii.100 The incidence of JRA in Japan was reported to be low (0.83/100,000),101 and chronic
C H A P T E R
arthritis may be less common in Americans of Chinese ancestry than in American whites,94 but there is a paucity of data describing these children. Studies from Asia are minimal, but reports from India have most often noted a higher frequency of polyarthritis, compared with oligoarthritis or systemic-onset disease.102–104 An analysis of white and Western Canadian Indian children suggests that, although chronic arthritis occurs in aboriginal children, its frequency is not higher than that for the white population, whereas HLA-B27–associated arthritis is appreciably more frequent in the aboriginal group.105 However, Oen and colleagues106 noted an impressive incidence of the disorder (23.6 per 100,000), in addition to a high frequency of seronegative spondyloarthropathies, in Inuit children of northern Canada. The data reported by Boyer and associates107 suggest that the incidence of RFseropositive polyarthritis is increased in southeast Alaskan Yupik Eskimo children. The numbers of patients in these studies are small, however, and conclusions with respect to actual incidence and prevalence of chronic arthritis in these groups are tentative. Similar studies are being conducted in American Indian children.108
ETIOLOGY AND PATHOGENESIS As indicated previously, chronic arthritis in childhood is certainly not a single disease. These children represent a heterogeneity of phenotypes with at least three primary modes of onset. Two observations are paramount in considering pathogenesis and etiology.109,110 First, it is an autoimmune disease. T-cell abnormalities and the pathologic characteristics of the chronic synovitis suggest a possible cell-mediated pathogenesis.111 Multiple autoantibodies, immune complexes, and complement activation indicate potential humoral abnormalities. Second, it is most assuredly a complex genetic trait (oligogenic or polygenic).112,113 The various forms of the disease display nonmendelian inheritance, and interactions of multiple genes are likely to be important in these diseases. Many of the putative genetic predispositions are within the major histocompatibility complex (MHC) region on chromosome 6; however, non-MHC genes undoubtedly play a role in some of the syndromes (see Chapter 4). Although there are undoubtedly genetic predispositions, as well as putative environmental triggers, any theory of pathogenesis must account for a number of factors, including the clinical heterogeneity of the disease; the higher prevalence in girls; the narrow peak ages at onset; the absence of a peak age at onset for systemic disease; and the widespread immunologic perturbations.114 There may be multiple etiologic events, or the disorder may result from a single pathogenic vector with diverse clinical patterns evolving from interactions with the host. It may be postulated that an infectious agent, infecting a child at a point of vulnerability—defined by age, intercurrent illness, prior antigenic experience, immunologic maturity, or immunogenetic predisposition—results in a permanent infection, which emerges as a clinical disorder. It is necessary to consider differing sets of conditions for development of each onset type and course subtype. Possible causes include aberrant
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
211
immunologic regulation, psychologic stress, trauma, hormonal abnormalities, and infection.
Immunopathogenic Mechanisms A number of observations contribute to the hypothesis that the immune system is intimately involved in pathogenesis. First, there is abundant evidence of altered immunity, abnormal immunoregulation, cytokine production, and polymorphisms.109,115–119 Second, there is an association between specific immunodeficiencies and rheumatic diseases, including chronic arthritis (see Chapter 33). Third, there is a close relationship between immune reactivity and inflammation, the hallmark of arthritis (see Chapter 3). Whether it is principally an immunogenetically determined disorder or an antigen-driven immunologic response is uncertain. One study alleged that breast feeding has a protective effect on the development of the disorder,120 especially in oligoarticular disease; however, a strong relationship was not confirmed in another investigation.121
The Innate Immune System: Immune Complexes Complement activation and consumption probably play a role in perpetuation of the inflammatory reaction and in determining specificity of response.122 Levels of circulating immune complexes parallel activity of the arthritis and systemic features.111,123–125 The pro-inflammatory activity of circulating immune complexes appears to be related to their size.126 Complement activation is also reflected in levels of fragment Bb of the alternative pathway and C4d, which correlate with circulating immune complexes and clinical activity of the disease.127,128 The circulating complexes consistently contain immunoglobulin M (IgM) RFs,129–131 even though most of these children are seronegative by conventional RF testing. They may be positive, however, for hidden RFs detected by acid gel filtration of their sera.132 Levels of these tightly bound RFs correlate with activity of the disease, particularly in children with many involved joints.133 The pro-inflammatory potential of such immune complexes may be enhanced because of their resistance to normal complement-mediated degradation due to this interaction with RFs. The immune complexes identified in synovial fluid also vary in size, composition, ability to activate complement, and potential for induction of cytokine secretion.111
T Lymphocytes and Cytokine Profiles Speculations on pathogenesis have centered on the possibility that there is a disordered interaction between type 1 (Th1) and type 2 (Th2) helper T cells (see Chapter 3).134,135 Th1 cells, or inflammatory CD4+ T cells, predominantly secrete interleukin-2 (IL-2), IL-3, interferon-γ (IFN-γ), granulocyte colony-stimulating factor (GM-CSF), and tumor necrosis factor-α (TNF-α) and TNFβ, and activate macrophages. Th2 cells secrete IL-3, IL-4, IL-5, IL-6, IL-10, GM-CSF, and TNF-α, which effect activation and differentiation of B cells. TNF-α and its soluble receptors and IL-6 occupy a central role in pathogenesis136 and provide a rationale for current therapeutic studies.137 Macrophage migration inhibitory factor (MIF) is increased in concentration in the vascular compartment of children with JIA. This abnormality is directly related to excess transmission of a two-point promotor haplotype (CATT(7)–MIF–173*G/C) that increases susceptibility to chronic inflammation.138 Investigations of the cytokine network in these children are often contradictory and incomplete, making interpretation of pathogenic mechanisms difficult. Differing results are undoubtedly related to the assay methods employed, stage and activity of the disease, treatment, identification of onset type and course
212
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
subtype, and whether plasma, serum, or synovial fluid was analyzed. A number of excellent reviews have been published116,119,137,139,140 (see also Chapter 3). Synovial fluid T cells appeared to be similar in type and responsiveness to those in peripheral blood in some studies,141,142 but in others CD4+ T cells were decreased in synovial fluid but increased in synovial tissue.143,144 Selective recruitment of T cells expressing CCR5 and CXCR3 has been reported.145 In most instances, T cells from the synovial fluid or membrane had increased expression of activation markers.141,142,145–148 Studies of Scola and colleagues149 indicated that cytokine expression patterns were typical of a type I bias. Murray and colleagues150 studied synovial T-cell infiltrates in 17 children and found that the level of T-cell activation (CD3+ IL-2R+) was significantly higher in oligoarthritis (especially for CD8+ cells) and that the CD4/CD8 ratio was lower. A subsequent study of the immunohistologic patterns of expression of synovial Th1 and Th2 cells found increased secretion of IL-4.151 Clonotypic restriction has been reported.152 Patterns of expression of TNF-α and -β and their receptors and other cytokines have also been studied.151,153,154 Ozen and colleagues79 observed a marked Th1 response in synovial fluid mononuclear cells in 4 of 5 children by immunofluorescent identification of intracellular cytokines through flow cytometric analysis (increased IFN-γ in addition to IL-4). The chemokine receptor CCR4 has been linked to a similar increase in the IL-4/IFN ratio.155 Coculture with heatshock protein (HSP) 60 produced only a slight increase in IFN-γ products in the one synovial fluid sample tested. A recent report indicated that synovial dendritic cells express the receptor activator for NF-κB (RANK).156 Gattorno and colleagues157 investigated the pattern of cytokine production in T-cell clones from synovial fluid mononuclear cells in five children with oligoarthritis. Large amounts of IFN-γ were produced with a predominant Th1/Th0 pattern. Razuiddin and coworkers158 found a mixed Th-cell response in stimulated peripheral blood mononuclear cells in systemic-onset disease. Studies of single nucleotide polymorphisms have confirmed different genetic influences in systemiconset disease for IL-6159,160 and MIF.161,162 Similar studies in oligoarthritis have focused on TNF haplotypes.163 Synovial cytokine levels differ in children with JRA compared to adults with RA.164 TNF-α G → A −238 and −308 polymorphisms may define outcomes in subtypes of chronic arthritis in children.165
T-cell Receptor Polymorphism Abnormalities of assembly and interactions of the trimolecular complex consisting of putative antigen and genes controlling selection of T-cell receptor (TCR) peptide chains and HLA specificities are of increasing importance in considering immunopathogenesis.166–168 Oligoclonal selection of the TCR chain has been demonstrated, a finding that is more characteristic of synovial fluid and tissues than peripheral blood.169,170 Maksymowych and associates171 confirmed a high frequency of the TCR allele TCR-Vβ6.1 among HLA-DQA*0101-positive children. A subsequent report identified this TCR null allele as a risk factor for a polyarticular course in children with early-onset oligoarticular disease positive for DQA1*0101.172 This association was not confirmed in a study from Norway,173 nor did Nepom and coworkers174 identify TCR polymorphism in their studies of oligoarticular-onset JRA. In the report of Thompson and colleagues,170 TCR-Vβ8 was clonally expanded in children with polyarticular disease and TCR-Vβ20 was increased in oligoarthritis.
Peripheral Blood Mononuclear Cells The results of studies of T-lymphocyte phenotypes in the peripheral blood have been inconsistent and are difficult to
interpret because of absence or inadequacy of control data. In general, studies of T-cell responses to mitogens such as phytohemagglutinin or concanavalin A indicate that proliferation is normal in children with inactive or oligoarticular disease but diminished in those with active disease.175–177 Multiple inhibitors may be present.178 Aberrations in suppressor T-lymphocyte function may be instrumental in immunopathogenesis and permit overproduction of autoantibodies.143,179–183 Some studies of peripheral blood lymphocytes have demonstrated normal ratios of CD4+ (helper/inducer) to CD8+ (suppressor/cytotoxic) T cells.142,184,185 Morimoto and coworkers175 found decreased CD4+ and increased CD8+ cells, an apparent result of antibody to CD4+ T cells that was present in the sera of patients with active disease. Some have also found increased CD8+ T cells in children with systemic and polyarticular onset,186,187 whereas others noted decreased CD8+ T cells, particularly in systemic-onset disease.188,189 In contrast, B-cell numbers are increased in these children.183,184 Similar discrepancies have been observed in the frequencies of markers of T-cell activation: Tac or IL-2 receptor expression was found to be normal,142,190 HLA-DR antigens normal190,.191 or increased,184 and very-late-activation antigen (VLA-1) increased only in children with active disease.190 Although many of these studies suggest a global T-cell regulatory defect, their results may have been influenced by therapy. Massa and colleagues192 found that treatment with methotrexate led to a decrease in CD4−, CD8−, and γ/δ T-cell numbers.
Autoantibodies B-lymphocyte numbers are normal to increased, depending on onset type, but their mitogen responsiveness may be impaired.184,193 Immunoglobulin levels tend to be high, at least partly reflecting the nonspecific inflammatory response. Antibody responses to new T cell–dependent antigens such as bacteriophage OX 174 may be defective.194 Some autoantibodies may more likely be epiphenomena than integral to pathogenesis, except for complications such as vasculitis, in which immune complexes participate in vascular inflammation. Autoantibodies to nuclear, immunoglobulin, and other antigens are common in sera of children with JRA.195 Among these autoimmune phenomena are antibodies to histones196–203; anti-neutrophile cytoplasmic antibodies204–206; anti-perinuclear, anti-keratin, and anti-RA33 antibodies207–209; anti-cardiolipin antibodies200,210–212; and the DEK oncoprotein (possibly associated with IFN-γ).213 Antibodies to high-mobility group (HMG) proteins214 are increased in JRA to a defined epitope on HMG-17,215,216 and in oligoarticular disease to an HMG-2 protein.217,218 Autoimmunity to types I, II, and IV collagens have been demonstrated in some studies.219–221 Immunity to cartilage link protein has been noted.222 IgA anti-gliadin antibodies have been described223 but are not predictive of celiac disease in JCA.224 Anti-cyclic citrullinated peptide antibodies have been reported, especially in RF-seropositive disease,225 but less frequently than in adults with RA.226
Hormonal Factors The often striking differences in sex ratio, as well as the characteristic preadolescent or postadolescent peaks in incidence of specific categories of childhood arthritis, suggest that reproductive hormones may play important roles in pathogenesis.227,228 In a study by Khalkhali-Ellis and colleagues,229 androgen levels in children with chronic arthritis and in aged-matched controls were similar for progesterone and dehydroepiandrosterone (DHEA). However, in prepubertal patients, 17β-estradiol was undetectable, and the concentration of the sulfated conjugate of DHEA was significantly less than in controls. Testosterone was lower in the synovial fluid than in matched serum; patients with
C H A P T E R
the lowest synovial fluid levels were those with disease of the longest duration. Therefore, low androgen levels may contribute to pathogenesis because they exert a protective effect against cartilage degradation.230 A number of studies have identified an interesting association of elevated serum prolactin levels in chronic arthritis and in systemic lupus erythematosus (SLE) (see Chapter 16). Prolactin is produced by cells of the anterior pituitary and other cells, including lymphocytes; in addition to its endocrine effects, prolactin enhances cell proliferation and survival.231 Levels were increased in children with chronic arthritis and antinuclear antibody (ANA) seropositivity.227,232,233 The prolactin concentration correlated with levels of IL-6 and with a chronic course of the disease. Both glucocorticoids and hydroxychloroquine inhibit prolactin secretion.228
Infection That infections can cause arthritis in children is not in doubt. Arthritis after viral infections is probably common, although it is usually self-limited.234 The possible role of infection in causation235 is supported by the finding that chronic arthritis is especially common among children who have impaired defense mechanisms and overt forms of immunodeficiency such as selective IgA deficiency, hypogammaglobulinemia, or deficiency of the C2 complement component (see Chapter 33). Arthritis is also a complication of infection with the human immunodeficiency virus.236 There is considerable evidence that viral infections do not just cause transient arthritis but are associated with human autoimmune disease.237 Persistent rubella virus infection has been demonstrated in children with arthritis in some studies,238,239 but was not confirmed in others.240,241 Postvaccination arthritis has been described after routine vaccinations242 and after measles-mumps-rubella (MMR) vaccine, and a chronic arthropathy was documented in one study (predominantly in females243) but not in another.241 HLA-DR associations with rubella vaccine arthritis have been demonstrated.244 Arthritis in children has also been linked to perinatal infection with the influenza virus A2H2N2.245,246 This unique epidemiologic study demonstrated the possibility of delayed expression of disease resulting from a presumed intrauterine or neonatal viral infection, not unlike the long-term effects that have been documented with intrauterine rubella infection. A somewhat tenuous link between chronic arthritis and parvovirus B19 has also been noted.247–249 IgM antibodies to parvovirus B19 were demonstrated in significantly more children with JRA, compared with those with acute self-limited arthritis or in a healthy control population.250 Remission of arthritis after viral infections has also been described.251,252 Antibody levels to specific viruses are also increased.253 Inflammatory responses may be triggered by molecular mimicry to Epstein-Barr virus proteins.254 In spite of these reports, there is no consistent evidence for an association of any specific viral infection with any form of chronic childhood arthritis. Mycoplasma, β-hemolytic streptococcus, and enteric organisms (Salmonella, Shigella, Campylobacter, and Yersinia) are all known to cause reactive arthritis (see Chapter 30). A possible role of chlamydial infection has been suggested.255 The observation that children with oligoarthritis and uveitis frequently have antibodies to bacterial peptidoglycan supports the possibility of a causative role for bacterial infection in this disease.256 The cyclic pattern of incidence of chronic arthritis documented from 1979 to 1992 in Manitoba by Oen and colleagues59 correlated with the occurrence of infections to Mycoplasma pneumoniae. RA has resulted after hepatitis B vaccination.257,258
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
213
Humoral and cellular immune responses to highly conserved bacterial HSPs are present in children with chronic arthritis.259–262 HSPs have been demonstrated in the serum and synovial fluid.262–264 Van Eden and colleagues265 postulated that reactive T-cells are part of the normal immune repertoire for TCR V-gene products; self-HSPs and bacterial HSPs may trigger this response. In 13 of 15 children with oligoarthritis, T-lymphocyte proliferative responses to HSP 60 were detected an average of 12 weeks before remission of the inflammatory disease.262,266 Spontaneous remission was characterized by CD30+ T-cells directed to HSP 60.267 Albani and associates261,268 demonstrated immune responses to the dnaJ HSP from Escherichia coli, especially in children with polyarticular disease. This protein has five amino acids that are homologous with those in the binding groove of DRB1, which in itself is increased in frequency in these children. Therefore, molecular mimicry may play a role in pathogenesis.269–271 Despite this circumstantial evidence, however, a direct link between infection and chronic childhood arthritis has remained elusive.
Psychologic Factors It is well documented that psychologic stress is particularly common in families of children with arthritis.272,273 It has not been possible, however, to be certain whether the psychosocial disturbances preceded or followed the development of arthritis. Current research indicates that psychologic factors inherent in the family and child affect their adaptation to chronic illness but are unlikely to have played a role in the causation of the disease. Some studies suggest that susceptibility to arthritis is associated with dysregulation of the autonomic nervous system that leads to lack of an appropriate response of the child’s immune system to stimuli274 and may be influenced by neuroendocrine gene polymorphisms.275
Physical Trauma Chronic arthritis has been reported by parents to follow minor physical trauma to an extremity. Such trauma may serve as a localizing factor, or it may simply call attention to an already inflamed and weakened joint. Benign hypermobility and the rare syndrome of congenital insensitivity to pain are both associated with trauma and may predispose to joint inflammation. The fact that certain joints (e.g., the knee) are frequently affected could be interpreted to suggest that trauma associated with weight bearing in the young child is a factor in initiating chronic inflammation.
Additional Studies of Pathogenesis A relatively large number of basic investigations of potential pathogenic mechanisms in chronic arthritis in children have been published recently. Each report of abnormalities in serum or synovial fluid clarifies an area of potential pathogenesis but also of future therapeutic interest. At this time, no sequential cascade of events can be identified. In synovial fluid and tissues, additional data on IFN-δ/IL-4 ratios,149,155 angiogenic factors,276 proinflammatory cytokine secretion,277 vascular endothelial growth factor,278 and Fas-induced apoptosis279,280 are of interest. Studies by DeBenedetti and colleagues118,281 reported increased expression of enzymatic generated P- and E-selectins, upregulated by Th1 cells, and additional investigations282,283 of levels of adhesion molecules and selectins in serum and synovial fluid. Decreased serum fibrinolytic activity,284 abnormalities involving matrix metalloproteinase 3 and its tissue inhibitor,285 and abnormalities of CD2R286 and CD27287 and their interactions with T cells are observed in some children. Expression of the p53 protein has been investigated.288
214
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
Animal Models of Inflammatory Joint Disease The study of animal models of human disease provides clues about etiology, but these models are, at best, approximations of human disease. Fundamental differences in etiology and pathogenesis almost certainly exist and may be difficult to recognize. In spite of these limitations, and because of the obvious difficulties involved in direct study of human rheumatic diseases, various spontaneous and induced models of disease in animals have been the focus of a number of studies289 and may be relevant to human diseases.290
Adjuvant Disease Intradermal injection of complete Freund’s adjuvant (mineral oil, detergent, and Mycobacterium butyricum) into the footpad of susceptible strains of rats results in the development of a chronic polyarthritis, sometimes accompanied by lesions of the skin and auricular cartilages, and an anterior uveitis.291,292 This disease is adoptively transferable by T lymphocytes, and Cohen and associates demonstrated that the model may be manipulated by T-cell clones capable of inducing, preventing, or ameliorating the disease.293–296 Antibody appears to have no significant role in pathogenesis. The course of adjuvant arthritis may be intermittent or chronic; if chronic, it results in tissue destruction with calcification and ankylosis of joints. The arthritogenic component of the adjuvant is a peptidoglycan dimer, muramyl dipeptide.297
Polyarthritis Induced by Type II Collagen Native type II collagen in incomplete Freund’s adjuvant induces chronic polyarthritis in certain strains of rats and mice.298–303 The disease can be passively transferred with specific antibody to type II collagen.304 Recent studies have suggested a role for cellmediated immunity in the pathogenesis of this disease.305–309 The severity of the arthritis in mice has been reduced by a synthetic collagen vaccine310 and by gene therapy with viral IL-10.311,312
Arthritis Induced by Infectious Agents Erysipelothrix rhusiopathiae and Mycoplasma are each capable of inducing arthritis in domestic pigs,313–315 and Mycoplasma species are arthritogenic in many other species, including rabbit, cow, goat, sheep, mouse, chicken, and turkey.316 Spontaneous infection with Chlamydia psittaci causes polyarthritis in lambs.317 Reovirus causes synovitis in chicks.318 Caprine arthritis is caused by infection with a retrovirus.319–321
Spontaneous Rheumatoid-Like Arthritis A disease resembling rheumatoid arthritis spontaneously occurs in dogs322 and MRL/l mice323 but rarely in primates such as monkeys.324 In the canine and mouse models, RFs are demonstrable.323,325
Transgenic Mice The NSE/hIL-6 transgenic mouse overexpresses IL-6 and is characterized by stunted growth.326 Levels of insulin-like growth factor binding protein 3 (IGFBP-3) are decreased and result in a decreased association of IGF-I in the 150-kD ternary complex secondary to increased clearance of the IGF-I. The TNF-α-transgenic mouse has been studied for the development of bone disease327 and arthritis.328
GENETIC BACKGROUND Familial Chronic Arthritis in Children Familial chronic arthritis is rare, and multigeneration cases are seldom recognized. Although disease rarely occurs in affected siblings, data on HLA segregation underscore a hereditary basis to immunopathogenesis.329,330 Within any one family, arthritis tends to have the same type of onset, and even the same complication of uveitis.331 An examination of time of onset of arthritis in families in which two or more children were affected indicated that the interval between onset of arthritis in each pair of siblings varied from 7 months to 11 years. In no instance was disease onset simultaneous, although in most cases the ages at onset were similar. An increase in the frequency of chronic arthritis developing in childhood has been reported in the parents of multiple offspring with this disorder.332 There is no association between birth order and the development of arthritis.333 The development of chronic arthritis in twins has been extensively studied.330,334–336 A concordance rate of 44% was reported in identical twins and one of 4% in dizygotic twins.337 Ansell and colleagues338 indicated that 2 of 5 pairs of identical twins were concordant for arthritis; in one pair of twin boys, however, a diagnosis of JAS was made later. Six nonidentical twins were discordant for disease. Studies by Clemens and associates339 in more than 2000 children with JCA documented a remarkable concordance between siblings for onset of disease, clinical manifestations, and disease course. Ten of 12 sibling pairs who were concordant for type of onset shared two DR antigens; the other two pairs shared one HLA-DR antigen.339 A multicenter study reconfirmed these findings in 71 sibling pairs, both of whom had chronic arthritis (and in 3 siblings in 3 families); 94% of these children were white.330 The mean interval between onset of disease in the siblings was 4.4 years (standard deviation [SD], 4.2 years). More than three quarters were concordant for onset type, and 79% were concordant for disease course. Among seven sets of twins, the interval between disease onset was shorter (3.3 months), and all were concordant for onset type (6 oligoarthritis, 1 polyarthritis) and course subtype. Uveitis was concordant in only 3 of 16 sibling pairs. There is also an increased prevalence of autoimmunity in these families with JRA.340 Chronic inflammatory rheumatic diseases are more common in parents of multiple offspring affected by chronic arthritis.332 One further association bears attention: the occurrence of JRA and adult RA in the same family. Documentation of this event is scant, and it must be concluded that JRA and RA uncommonly occur in the same family, which is consistent with their different HLA associations. However, Rossen and colleagues341 studied four families with multiple cases of JRA and RA and concluded from histocompatibility data that susceptibility to arthritis was influenced by a dominant allele with variable penetrance and expressivity. An increased frequency of autoantibodies has been documented in first-degree relatives of children with chronic arthritis.342
Human Leukocyte Antigen Relationships Although chronic arthritis has rarely been documented in siblings or in children whose first-degree relatives have another rheumatic disease, more than 50 studies of histocompatibility antigens have pointed clearly to the role
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
215
■ Figure 9–3 Proportion of the study population, by disease-onset type (A) and by sex (B), in each age-at-onset category with the HLADR4 allele.The horizontal line represents the frequency of the allele in the control group. Pauci, pauciarticular; Poly, polyarticular. (From Murray KJ, Moroldo MB, Donnelly P, et al: Age-specific effects of juvenile rheumatoid arthritis-associated HLA alleles. Arthritis Rhum 42: 1843–1853, 1999, Copyright © 1999. Wiley-Liss, Inc. Reprinted by permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.)
of certain HLA specificities in predisposing to these diseases.112,329,337,343–372 These investigations also have provided evidence of fundamental distinctions between adult RA and arthritis in children.373,374 It was hoped that HLA specificities might aid in the reclassification of children with chronic arthritis, but this remains an unfulfilled aspiration. Only recently has it been noted that risk and protective effects of these immunogenetic profiles are age related for each onset type and for a number of course subtypes. These complex interactions and age- and sex-specific windows of susceptibility and protection were examined in detail in a provocative paper by Murray and colleagues.382 As an illustration, the population distribution for HLA-DR4 in this study for children with polyarticular onset and 254 ethnically matched unrelated controls is depicted in Figure 9–3. In addition, an increased frequency of certain class III MHC genes may contribute to susceptibility in some populations375 but not in others.356,376,377
Associations with Class I Antigens An increased frequency of A2, B27, and B35 alleles has been documented.375,378–381 A2 is associated predominantly with earlyonset oligoarticular disease in girls.382 An increase in the frequency of HLA-B27 in early studies was possibly related to inclusion of children with JAS, and later investigations document inconsistent increases in the frequency of this antigen.380,383 However, HLA-B27 may confer an age-related risk to oligoarthritis in boys (50% risk at 7.3 years; 80% risk at 11.9 years).382
Associations with Class II Antigens Class II genetic associations are more numerous and complex in relation to specific onset types and course subtypes than are the few documented class I specificities.384 These associations are most obvious in early-onset oligoarthritis and in RFseropositive polyarthritis. They are more heterogeneous and inconclusive in RF-seronegative polyarthritis and systemic disease. Terminology has changed over the years for many of these specificities.385
Interactions with Non-HLA Genes A number of non-HLA genes, on chromosome 6 or on other chromosomes, may be important in a predisposition to chronic arthritis or in its pathogenesis.386 A weak association with TAP2B, a polymorphism in a member of the adenosine triphosphate–binding cassette superfamily, is present for early-onset disease.387 TAP1B may function as an additive susceptibility factor.388 Interaction with other non-HLA genes is also possible, such as IL-1A2, a variant of the IL-1α gene, in early-onset oligoarthritis.389 The gene for IL-1α, or a gene for which its polymorphism is a marker, may also contribute risk for early-onset disease and uveitis.389,390 One study suggested that homozygosity of the B allele of the LMP2 proteosome subunit may increase susceptibility to a putative subtype of chronic arthritis that is associated with B27.391 Data have also been reported for the LMP7 gene.392 Polymorphism in the osteoprotegerin gene may correlate with articular erosions and low bone mass (see Chapter 38).393
216
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
Associations with Other Autoimmune Diseases and Chromosomal Abnormalities There may also be an association of the various types of onset with genetic and chromosomal abnormalities394 and with other autoimmune diseases (see Chapters 4 and 32). Most frequently reported is insulin-dependent diabetes mellitus.395,396 In a referral diabetic clinic population of 200 children, 6 had polyarticular disease and 1 had probable early JAS.395 Autoimmune thyroiditis has also been reported.397 A syndrome that includes flexion contractures, short stature, and skin changes, the socalled Rosenbloom syndrome, may complicate diabetes mellitus and should not be confused with inflammatory joint disease (see Chapter 32).398,399 Myasthenia gravis was reported in four children with chronic arthritis.400 Autoimmune neuromyotonia has also been noted. Other non-HLA genetic associations have been suggested that might increase a predisposition to disease. Among these are selective IgA deficiency401 (see Chapter 33) and the velocardiofacial syndrome.402 One child with arthritis and deletion of the short arm of chromosome 18 was IgA deficient.403 The phenotypic profile of the chromosome translocation in the velocardiofacial syndrome—del(22q11.2) (CATCH 22)—includes chronic destructive arthritis, normal numbers of T lymphocytes, and normal or elevated serum immunoglobulins.404,405 No children had the complete DiGeorge syndrome. Sullivan and associates402 suggested that T-cell immunodeficiency in a subgroup of patients with this deletion syndrome permits polyarthritis to develop. The coexistence of IgA deficiency and arthritis in the 22q11 syndrome has been observed.406 Turner’s syndrome has also been identified in a number of patients.407 In a study of this association,408 18 of approximately 500 children with JRA were found to have Turner’s syndrome. Polyarticular disease was present in 7 who had progressive seronegative arthritis and a 45 XO genotype. It is not entirely clear whether the arthropathy of Down syndrome differs from idiopathic chronic arthritis.394 In children with Down syndrome and arthritis, the joint disease has often been indistinguishable from polyarticular disease. In some children, the arthritis is more like psoriasis, and the latter is seemingly increased in frequency in trisomy 21. Polyarthritis was reported in a child with partial trisomy 5q, monosomy 2p.394
CLINICAL MANIFESTATIONS Constitutional Signs and Symptoms Anorexia, weight loss, and growth failure occur in many children. Significant fatigue is rarely a feature of limited articular disease, but it is a common symptom in children with polyarticular or systemic disease, especially at onset and during periods of poor disease control. It may be expressed as an increased sleep requirement, lack of energy, or increased irritability. Night pain may interrupt sleep and contribute to fatigue. Sleep fragmentation may also exacerbate pain as well as fatigue in these children.409,410
Pain A child with chronic arthritis may not complain of pain at rest,411,412 but active or passive motion of a joint elicits pain, particularly at the extremes of the range of motion
(ROM). Pain is usually described as aching or stretching and is of mild-to-moderate severity. In contrast, children with pain-amplification syndromes almost always describe pain as extremely severe (see Chapter 37). Pain elicited by pressure or tenderness is usually maximal at the joint line or over hypertrophied, inflamed synovium. Bone pain or tenderness is not characteristic, and its presence should alert the examiner to the possibility of a malignancy involving bone.413 The manner in which a child communicates discomfort or dysfunction varies cognitively according to developmental age. The child may express increased irritability, or the joints may be tender on examination or painful in motion. In the young child, there may be no complaints of pain, although it is not clear that the child is not experiencing pain. Young children, however, may alter the manner in which the affected joint is used, assume a posture of guarding the joints, or entirely refuse to use a limb in order to avoid pain. Their expression of pain is thereby physical rather than vocal. In such circumstances, parents report that the child does not appear to be in pain but that he or she limps or reverts to more infantile patterns of movement. The relation of cognitive stage to expression of pain is not certain. Some studies detected no age-related differences in reporting of pain.414,415 In others, younger children with limited joint disease complained of the most severe pain,416–418 and in still others, older children reported more pain.411,412,419 This was related, it was suggested, to their concern about the significance and consequences of the disease. Pain may be an important component of functional disability. It may limit school attendance, physical activity, and social interactions.420 Debate continues about pain perception in children with arthritis.412,421 Two studies concluded that children perceived less pain than their adult counterparts did, although the evaluation instruments were not ideal in either case.422,423 One investigation reported that affected children had lower pain thresholds than healthy children did.424 In another study, Sherry and associates425 stated that only 14% of children with arthritis (mostly oligoarticular type) studied in an outpatient setting indicated that they had no pain. In a more recent report, 57 children with chronic arthritis, aged 7 to 17 years, reported the presence of pain, usually described as aching.415 No differences were found with respect to type of onset of arthritis, sex, age, or disease duration. There was often an overall lack of association between the child’s psychosocial functioning and reports of the nature or intensity of the pain.415,426 In some studies, the child’s report of pain intensity correlated with the physician’s estimate of disease activity or severity,426 but in others no correlation was found.415,423,427 In one study,428 pain as assessed by a pediatric pain questionnaire was compared with the results of thermography of affected joints. Correlations between the visual analogue scale scores determined by parents and those measured by physicians were significant (P < .01 and < .05, respectively). However, correlation between the children’s scores and thermography was not significant. The authors interpreted these findings to indicate that relationships among the multifactorial aspects of the subjective pain response in joint inflammation were far from direct. Severity of disease accounted for only a relatively small percentage of the variance among factors that influenced the subjective pain response.416 Both acute and chronic pain in children are often underappreciated and therefore undertreated.415,426 Children may not complain of pain per se because of their level of cognitive
C H A P T E R
development, and pain might better be evaluated by a child’s responses to physical examination of the joints than simply by inquiring about the presence of pain. Measurement of pain in children is facilitated by the use of instruments such as the Pediatric Pain Questionnaire.429 Using a visual analogue scale, Varni and colleagues429 confirmed that reports of pain by children as young as 5 years correlated well with reports made by parents and physicians. It therefore seems reasonable to conclude that children with arthritis experience as much pain as adults when that pain is accurately evaluated in relation to developmental stage. In a study of children with chronic arthritis,411 many reported pain as a major symptom that affected their daily activities. The pain was modestly correlated with long disease duration, older age, and variables related to demographic scales. Disease status and measurable psychologic variables accounted for a modest amount of variance in pain scores. The results suggested that factors that were contributing to pain in children were quite different from those that had been identified in adults with RA. Another study430 examined three psychologic measures in children and their families that significantly affected functioning. Greater emotional distress in the child and in the mother, as well as the level of family harmony, correlated with a higher degree of reported pain in the child.431
Characteristics of Joint Inflammation Morning stiffness and gelling after inactivity are common manifestations of inflammatory joint disease. They are infrequently vocalized by the young child but frequently reported by parents as morning slowness or stiffness. The arthritic joint exhibits the cardinal signs of inflammation: swelling, erythema, heat, pain, and loss of function (see Fig. 9−1). Swelling of a joint may result from periarticular soft tissue edema, from intra-articular effusion, or from hypertrophy of the synovial membrane. Effusion and hypertrophy of the synovial membrane are common, whereas in arthritis accompanying diseases such as Henoch-Schönlein purpura, periarticular swelling is more prominent. Involved joints are often warm but usually not erythematous. In contrast, the joint may be erythematous in septic arthritis or acute rheumatic fever and in some of the other reactive arthritides.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
The characteristic initial complaints are stiffness and pain at the back of the neck; rapid loss of extension and rotation may result. Unilateral apophyseal joint disease may result in asymmetry of neck extension. Subluxation of the atlantoaxial joints may occur early, rendering the child at risk for injury in an accident or with attempted intubation before general anesthesia. Cervical spine ankylosis carries much the same risks. It may be a major problem for the anesthetist because of difficulty in obtaining adequate neck extension and the necessity to use nasotracheal rather than orotracheal intubation or a laryngeal mask. Involvement of the thoracolumbar apophyseal joints is generally not appreciated clinically. However, scoliosis, possibly reflecting asymmetrical apophyseal joint inflammation, was up to 30 times more frequent in early studies.435,436 Scoliosis apparently is now much less frequent. Minimal reactive sclerosis of the sacroiliac joints may occur in a few children (less than 5%) and should be distinguished from the more pronounced disease observed in JAS.437,438 The sacroiliac joints may eventually fuse, particularly in children who have been bedridden. Small outpouchings of synovium are not uncommon and are particularly evident at the extensor hood of the proximal interphalangeal (PIP) joints and around the wrist or ankle. Less commonly, synovial cysts occur in the antecubital area or anterior to the shoulder.439 They may be the initial or sole manifestation of chronic arthritis and, if unilateral, may be misinterpreted as a tumor or as deep venous thrombosis (Fig. 9–4). Ultrasound imaging, magnetic resonance imaging (MRI), or arthrography aids in correct diagnosis. Large synovial cysts are an unusual complication. These may occur in the popliteal space (Baker’s cyst) (Fig. 9–5), rupture into adjacent muscles, and dissect into the calf.440 This event is characterized by sudden sharp pain and swelling in the calf, followed by crescentic ecchymoses about the malleoli. A normal child may occasionally develop a transient popliteal cyst.441
Distribution of Affected Joints Any joint may be affected, but large joints are most frequently involved. Small joints of the hands and feet may also be affected, particularly in polyarticular-onset disease. Attention must also be directed to the temporomandibular joint (TMJ) and to the cervical, thoracic, and lumbosacral spine. The sternoclavicular, acromioclavicular, and sternomanubrial joints are infrequently affected. Cricoarytenoid arthritis is unusual but may be responsible for acute airway obstruction.432 Inflammation of the synovial joints of the middle ear, the incudomalleal and incudostapedial articulations, is rarely appreciated clinically. Tympanometric studies, however, have indicated that subclinical disease may be present in almost two thirds of children with JRA.433 Disease in the apophyseal joints of the cervical spine occurs at onset in approximately 2% of children and may present as a torticollis.434 Approximately 60% eventually develop involvement of this area of the axial skeleton.
217
■ Figure 9–4 Brachial synovial cyst. A 6-year-old girl developed this dissecting cyst of her right arm as the first manifestation of chronic arthritis. Later, bilateral effusions developed in both knees. (See color insert.)
218
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD ■ Figure 9–5 A, Popliteal cyst or Baker’s cyst.The cyst was associated with pain at the back of the knee, was somewhat tender, and transilluminated. Aspiration yielded clear yellow fluid of low inflammatory activity. B, Arthrogram of a popliteal cyst, with contrast medium outlining the communication (arrow) between the synovial space and this dissecting cyst in an 18-year-old boy who had arthritis since the age of 9 years.
Tenosynovitis Tenosynovitis is more common than is usually appreciated, but it is generally not a striking or isolated clinical complaint.442 The most common sites are the extensor tendon sheaths on the dorsum of the hand, the extensor sheaths over the dorsum of the foot, and those of the posterior tibial tendon and the peroneus longus and brevis tendons around the ankle. Loss of extension of the fingers may result from a stenosing synovitis of the flexor tendon sheaths and may be responsible, in part, for a claw-hand deformity. Clinically recognized carpal tunnel syndrome is uncommon in children with involvement of the wrists. Tenosynovitis of the superior oblique tendon of the eye may cause pain on upward gaze, sometimes with diplopia, the so-called Brown’s syndrome.443,444
Extra-articular Manifestations The frequency with which extra-articular complications occur during the course of chronic arthritis emphasizes the systemic nature of this disease. It should also serve as a reminder that, on occasion, therapy induces systemic complications that may in themselves contribute significantly to morbidity. Chronic uveitis, a unique complication, is discussed in Chapter 11; episcleritis445 and keratoconjunctivitis sicca can also occur.446
Generalized Abnormalities of Growth and Development Abnormalities of growth and development are frequent complications of chronic arthritis or its treatment. Linear growth is retarded during periods of active systemic disease.447 Accelerated growth may occur with suppression of the active disease by therapy or during a remission. It is unusual, however, for a child who has suffered prolonged arrest or slowing of growth to return to the previous growth channel. Puberty and secondary sexual characteristics are often delayed.448 Levels of growth hormone and IGF-I and -II are often normal in children with chronic arthritis but may be impaired.449 IGF-I was low in the children studied by Davies and colleagues,449 and in the investigation of DeBenedetti and coworkers, it was inversely correlated with IL-6 levels in children with systemic disease.450 Many early studies commented on the general arrest of development, retardation of linear growth, asymmetry of develop-
ment, or persistence of infantile proportions.32,40 In a study of 119 children, Ansell and Bywaters451 found that long duration of active disease was associated with reduction of linear growth even in children who had not received glucocorticoid drugs. During remission, height returned to normal in 2 to 3 years if premature epiphyseal fusion had not occurred. Stunting was severe only in children with long-standing, active disease. Similar findings were reported in the extensive prognostic investigation by Laaksonen53 of 544 children. In a sequential study of height in 31 children,452 about half were below the 3rd percentile for age and sex at the 5- to 7.5-year follow-up interval. However, 3 of 9 children were below the 3rd percentile at onset of disease. Undetected disease of some duration might have accounted for this finding. In a study of 56 children between the ages of 4 and 18 years who had disease for more than 1 year, Bacon and White453 found that mean height for age was below the 35th percentile for children with polyarticular or systemic-onset disease. Mean weight for age and weight per height were significantly diminished in those with polyarticular disease. In a study of adults who had had arthritis as children, Lovell and colleagues454 documented growth retardation (height less than 5th percentile) in 50% of children with systemic onset, 11% of those with oligoarticular onset, and 16% of those with polyarticular onset. Glucocorticoid therapy could only partly explain these observations. A marked decrease in height velocity (−2 SD) was observed in 56% of children with systemic arthritis by GarciaConsuegra and associates.455 In their study, growth retardation was associated with glucocorticoid administration, nutritional status, bone mineral density, and early disease onset. Glucocorticoid medications also result in measurable growth retardation or may intensify that initiated by the disease.456 Growth retardation was evident in children treated with prednisone in a dose estimated to be equal to or greater than 5 mg/m2/day for 6 months or longer.457 Laaksonen and colleagues,458 however, found that glucocorticoids in this dose range did not produce growth retardation if used for short periods.
Localized Growth Disturbances Localized growth disturbances result from destruction of a growth center, accelerated development of ossification centers of the long bones of an inflamed joint, or premature fusion of the physis. Brachydactyly of the digits develops from premature closure of the epiphyseal growth plates. During early active disease, development of the ossification centers is accelerated, apparently related to the hyperemia of inflammation and local production of growth factors. The ultimate result may be
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
219
■ Figure 9–6 Arthritis of the second and fourth proximal interphalangeal joints on the left hand associated with brachydactyly of those fingers.
either overgrowth of the affected limb or premature fusion of the involved epiphyses, resulting in diminished length. This latter phenomenon may be widespread and symmetric, resulting in small hands and feet, or it may be isolated, as in the example of brachydactyly illustrated in Figure 9–6. Arthritis of the lower limb, especially of the knee, frequently causes accelerated growth and epiphyseal maturation. If this occurs in one knee only, a discrepancy of leg lengths results. Only rarely does premature epiphyseal fusion cause shortening of the affected leg. Although accurate clinical measurement of leg length is difficult, a good estimate can be obtained by (1) measuring the distance from the superior anterior iliac spine to the medial malleolus in the supine position, (2) estimating leg lengths with the child in a supine position by comparison of malleolar symmetry, or (3) using a set of boards of calibrated thickness to determine which is required to level the iliac crests when the child is standing. A difference of greater than 0.5 cm is probably significant, and differences of 5 cm or more occasionally occur. Inability to extend both knees makes these measurements invalid, however; under these circumstances, visual comparison of the heights of the equally flexed knees in a supine position gives some indication of the presence of a leg-length inequality. Apparent leg-length inequality may also result from pelvic rotation and scoliosis. As the child grows, inequalities of minimal-to-moderate degree may disappear, but they persist in up to two thirds of these children. Whether the long-term result is shortening or lengthening of the affected limb appears to depend considerably on the age of the child— or, more precisely, on the degree of maturation of the skeleton—at the time of the inflammatory insult. If the child is very young and the potential for growth is considerable, the affected limb tends to grow longer. If the disease occurs shortly before the physis would be expected to fuse, shortening of the affected limb is more likely. Micrognathia is a striking example of localized growth retardation (Fig. 9–7). Marked alterations of facial morphology (birdface deformity) may result. In a Swedish survey of 70 children, 56% had symptoms (crepitus, pain, difficulty opening the mouth) and 41% had radiographic evidence of TMJ pathology attributable to arthritis.459 In one patient, the disease began in a TMJ. Another study corroborated the frequency (50%) of TMJ abnormalities and alterations in mandibular growth.460 Extreme
■ Figure 9–7 Arthritis began in this young man at the age of 2 years and pursued an unremitting polyarticular course. Disease of both temporomandibular joints has caused micrognathia and retrognathia with an anterior open underbite.
micrognathia is most likely to occur if arthritis begins before 4 years of age. The mandible ossifies by intramembranous bone production. Its growth is affected by a number of factors that contribute to mandibular growth abnormalities. Pain in a TMJ may inhibit normal masseter muscle development, which in turn retards mandibular bone development, resulting in a shortened mandibular ramus and body. Destruction of the condyle of the mandible causes further diminution of overall mandibular height. In some children, overgrowth of the condyle may contribute to TMJ dysfunction.461 An association of micrognathia and arthritis of the apophyseal joints of the cervical spine per se has been asserted in a number of studies,462–464 but this observation may not be valid. Although the stereotypical abnormality is bilateral micrognathia with an anterior open overbite, unilateral TMJ disease is much more common than bilateral disease. It is characterized by mandibular asymmetry; deviation to the affected side on opening of the jaw; difficulty in palpating the affected mandibular condyle; and pain, tenderness, or crepitus of one or both TMJs. In many instances, TMJ arthritis appears to be asymptomatic, although some children report a preference to chew on the unaffected side, experience pain with maximal opening of the mouth, or are aware of clicking or other sounds accompanying movement of the joint. Little is understood about the effect of chronic arthritis on dental caries or periodontal disease.465
Osteopenia Osteopenia has emerged as a potentially major determinant of functional outcome in young adults who have had chronic arthritis as children.466–474 Children with chronic arthritis develop a diminished bone mass compared with normal children and thereby are at increased risk for fractures in adulthood and for an earlier onset of osteoporosis.475–477 An important determinant of future fracture risk is the peak bone mass achieved at the end of skeletal maturation, which is almost complete by the
220
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
late years of adolescence.478 Genetic polymorphisms of the vitamin D and calcitonin receptors479 and the osteoprotegerin gene393 may play a role in the development of low bone mass. These interrelationships are discussed in Chapter 38.
ORGAN-SPECIFIC MANIFESTATIONS OF CHRONIC ARTHRITIS Skin and Subcutaneous Tissue The classic rash of systemic-onset disease has already been mentioned and is discussed in Chapter 12. A second cutaneous change, occurring particularly in children with involvement of the hands, is a dark discoloration of the skin over the PIP joints.480 The presence of this finding may reflect disease chronicity. In children with tender joints, retention keratosis may simulate a pigmented lesion.
may improve over several years. Treatment includes the use of pressure “stockings” or “gloves.”
Vasculitis Rheumatoid vasculitis is rare and occurs most often in the older child with RF-positive polyarthritis. This type of vessel involvement must be distinguished from benign digital vasculitis (Fig. 9–8), which is more frequent, and may occasionally be associated with vascular calcification on radiographs along the course of the digital arteries. The occurrence of Raynaud’s phenomenon is generally indicative of another connective tissue disease and not of chronic arthritis per se. Pityriasis lichenoides et varioliformis acuta (PLEVA or MuchaHabermann disease) has been recorded in a few children with JRA,484,485 scleroderma,484 Takayasu’s arteritis,486 polyarteritis nodosa,485 Wegener’s granulomatosis,487 and other vasculitides.485,488 These are rare occurrences, however, and may be entirely coincidental.
Muscle Disease Nodules Nodules occur in a variety of rheumatic diseases in children. Rheumatoid nodules occur in 5% to 10% of children with chronic arthritis, almost always confined to those with polyarthritis. Nodules are most frequent below the olecranon, but they also occur at other pressure points, on the digital flexor tendon sheaths, Achilles tendons, and occiput, as well as on the bridge of the nose in a child who wears glasses.
Atrophy and weakness of muscles around inflamed joints is characteristic and is often accompanied by a shortening of the muscles and tendons that results in flexion contractures. A nonspecific myositis may account for some of the associated fatigue and muscle weakness. It does not have a characteristic distribution, and
Typical rheumatoid nodules are firm or hard, usually mobile, and nontender. The overlying skin may be erythematous. They may be solitary or multiple, may change in size over time, and may persist for months to years. They are almost always associated with RF seropositivity and in this respect are generally regarded as a poor prognostic sign. Benign rheumatoid nodules or pseudorheumatoid nodules occur as isolated abnormalities in otherwise healthy children. They are painless, may be single or multiple, and sometimes become quite large (greater than 5 cm). They occur especially over bony prominences such as the anterior tibia or scalp and are characterized by spontaneous regression and recurrence. They frequently recur after surgical excision. Subcutaneous granuloma annulare, a relatively common disease in children, may be histologically indistinguishable from either rheumatoid or pseudorheumatoid nodules. Its clinical appearance and location (shins, dorsum of the foot), umbilicated appearance, and the absence of other disease confirm the diagnosis. Other disorders, such as multicentric reticulohistiocytosis and acute rheumatic fever, are associated with subcutaneous nodules (see Chapter 31).
Lymphedema Asymmetrical lymphedema of the subcutaneous tissues of one or more extremities has been documented in several children with arthritis.481–483 The swelling is usually painless and may be somewhat pitting. The cause is unknown except for its unclarified relationship to inflammation, but does not seem to be related to local obstruction caused by joint swelling. The course is chronic but
■ Figure 9–8 Punctate erythema of the palms and finger pads was the sole manifestation of benign perivasculitis in this 5-year-old girl with chronic arthritis. The lesions were not raised or tender and disappeared after treatment with nonsteroidal anti-inflammatory drugs. (See color insert.)
C H A P T E R
histopathologic studies are sparse. Data from adults with RA suggest that it is characterized by a perivasculitis and lymphocytic infiltrates (lymphorrhages). Serum levels of muscle enzymes are not increased. A few children with widespread, prolonged, active arthritis develop progressive muscle atrophy that is most severe and persistent if it occurs before 3 years of age.489
Cardiac Disease Pericarditis The overall prevalence of pericardial involvement is estimated at 3% to 9%.490 Pericarditis tends to occur in the older child with systemic disease, but it is not related to age at onset or severity of joint disease.491 It may occasionally precede development of arthritis, or it may occur at any time during the course of the disease, usually accompanied by a systemic exacerbation. Episodes typically persist for 1 to 8 weeks. Most pericardial effusions are asymptomatic, although some children have dyspnea or precordial pain that may be referred to the back, shoulder, or neck. Examination may document diminished heart sounds, tachycardia, cardiomegaly, and a pericardial friction rub, usually at the left lower sternal border. In many cases, pericardial effusions develop insidiously, are not accompanied by obvious cardiomegaly or electrocardiographic changes, and escape recognition except by echocardiography. Tamponade is rare and is characterized by pulsus paradoxus, venous distention, hepatomegaly, and peripheral edema. Chronic constrictive pericarditis is very rare and is characterized by pulsus paradoxus, a small heart, venous distention, ascites, and peripheral edema. In general, children with pericarditis do not fare worse than others in outcome, and this complication should not necessarily be regarded as a poor prognostic sign.
Myocarditis Myocarditis is much less common than pericarditis and may result in cardiomegaly and congestive heart failure.492 In 3 children reported by Miller and French,493 failure occurred in the absence of overt pericardial effusions in children with severe active disease. At necropsy, diffuse myocardial changes typical of congestive cardiomyopathy were present in one child. Goldenberg and colleagues490 reported that 4 children had perimyocarditis and 2 had myocarditis in a retrospective study of 172 children with JRA. Children with JRA may also demonstrate abnormal diastolic relaxation, similar to that which occurs in ischemic heart disease, and therefore may have an additional risk factor for coronary artery disease.492
Endocarditis Valvular disease, seemingly unrelated to other causes, has been documented in more than 10 children: 8 had aortic insufficiency, and 2 had mitral insufficiency. Sudden deterioration in cardiac function may occur. Valve replacement was necessary in some of these patients.494
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
221
Pleuropulmonary Disease Parenchymal pulmonary disease is rare, but diffuse interstitial fibrosis occurs in a small number of children495,496 and may precede other evidence of arthritis.497 Athreya and colleagues495 noted interstitial disease in 8 of 191 children, all of whom had a systemic onset. Another report detailed pathologic findings in a child who died of pulmonary fibrosis.498 One child was described with primary pulmonary hypertension,499 and another with pulmonary hemosiderosis.500 Pulmonary function studies documented abnormalities in 10 of 16 children.501 In some children, these abnormalities may be the result of respiratory muscle weakness.502 Pleural effusions may occur with carditis, or they may be asymptomatic and detected only as incidental findings on chest radiographs. Pulmonary rheumatoid nodules as described in adult RA are rare in childhood.
Gastrointestinal Tract Except for symptoms induced by medications, gastrointestinal (GI) tract disease is rarely described in children with chronic arthritis. Intestinal pseudo-obstruction503 has been reported. Peritonitis was documented in two children.504 In children with major GI tract disease, the possibilities of inflammatory bowel disease, celiac disease, or cystic fibrosis—all of which may be associated with arthritis—should be considered. Sjögren’s syndrome is uncommonly identified in children but occasionally complicates RF-positive polyarthritis (see Chapter 21).
Lymphadenopathy and Splenomegaly Enlargement of lymph nodes and spleen may occur alone or together and are especially characteristic of systemiconset disease. Marked symmetrical lymphadenopathy is particularly common in the anterior cervical, axillary, and inguinal areas and may suggest the diagnosis of lymphoma. Mesenteric lymphadenopathy may cause abdominal pain or distention and may lead to the erroneous diagnosis of an acute surgical abdomen. Splenomegaly is usually most prominent within the first years after onset. The degree of splenomegaly may be extreme, but it is uncommonly associated with Felty’s syndrome (splenic neutropenia).505,506 Functional hyposplenia (as in SLE) has not been reported.
Hepatic Disease Hepatomegaly is less common than splenomegaly and occurs almost exclusively in children with systemic-onset disease. Moderate to marked enlargement of the liver is often associated with only mild derangement of functional studies and relatively nonspecific histopathologic changes. This type of liver disease is most evident at onset and typically diminishes with time. Chronic liver disease does not occur. Massive enlargement of the liver is usually accompanied by abdominal distention and pain. Progressive hepatomegaly is characteristic of secondary amyloidosis. Occasionally, a fatty liver is associated with glucocorticoid administration. Hepatitis (transaminasemia) related to therapy with nonsteroidal
222
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
anti-inflammatory drugs (NSAIDs) may occur (see Chapter 5). A rare, and apparently benign, transient elevation of serum alkaline phosphatase has been reported in several children.507,508 This abnormality presumably does not result from hepatotoxicity, although it may be confused with it. Reye’s syndrome had been associated with aspirin therapy509 (see Chapter 5). The macrophage activation syndrome is discussed in Chapter 12.
Neurologic Disease Involvement of the central nervous system is usually related to complicating factors such as metabolic derangement, salicylate toxicity, high fever, embolism, and other systemic diseases. In some children, central nervous system disease has been so overwhelming as to suggest a primary relationship with the arthritis or its treatment.510 Some reports may have represented unrecognized instances of Reye’s syndrome. Cerebral and cutaneous vasculitis was described in one child with RF-seropositive disease.511 Entrapment syndromes (peroneal, tibial, tarsal) are not as common in children as in adults with RA but should be considered in the differential diagnosis of tenosynovitis or vasculitis involving peripheral nerves.
Renal Disease Urinary tract abnormalities may occur as a complication of chronic arthritis or as toxic effects secondary to treatment. Intermittent hematuria or proteinuria is an occasional finding in some children, but it should be noted that low-grade proteinuria and hematuria occasionally occur in normal children. Microscopic hematuria and lowgrade proteinuria may reflect the effects of medications, including NSAIDs, gold compounds, or D-penicillamine. Abnormal urinary findings also raise the possibility that the child has vasculitis or intravascular coagulation, a disease such as SLE, or (rarely) amyloidosis. Renal papillary necrosis has been described in a number of children and is thought to be partly related to NSAID use (see Chapter 5). Hypercalciuria has been identified as an additional rare cause of hematuria in some children.512 In Anttila’s study of 165 children, transient microscopic hematuria was observed in 23%, leukocyturia in 25%, and low-grade proteinuria in 42%.513,514 Recurrent or persistent hematuria (4%), leukocyturia (6%), and proteinuria (2%) were uncommon. Hematuria and leukocyturia were more frequent during the initial observation period. Proteinuria was associated with the presence of extra-articular disease, prolonged duration of active disease, and amyloidosis (40% of the children who died). Renal biopsy was performed in 35% of the children and documented minimal glomerular changes in 22% and tubular atrophy in 13%. Chronic pyelonephritis was a common finding at necropsy. Interstitial nephritis and gold-induced nephrotic syndrome were rare. More recent studies of renal dysfunction suggest that abnormalities of tubular function are common.515 Although the frequency of proteinuria (2.3%), hemoglobinuria (3.5%), erythrocyturia (4.1%), and leukocyturia (5.3%) were low in a study of 176 children with chronic arthritis, urinary β2-microglobulin or N-acetyl-β-glucosaminidase levels were elevated in 38.5%. Increased excretion of these proteins was associated with active
polyarticular disease and was most frequent in children receiving slow-acting antirheumatic drugs or two or more NSAIDs. It is not clear whether these changes represent the effects of the disease or drug-related toxicity.
PATHOLOGY The histopathologic features of chronic arthritis in children are similar to those described in RA. There is villous hypertrophy and hyperplasia of the synovial lining layer (Fig. 9–9). The subsynovial tissues are hyperemic, edematous, and infiltrated with lymphocytes and plasma cells. Vascular endothelial hyperplasia is often prominent. There is a selective accumulation in the synovium of activated T cells, which are clustered around antigen-presenting dendritic cells. Fibrin may be layered onto the superficial surface of the synovium or incorporated within it. An exuberant synovitis and pannus formation eventually results in progressive erosion and destruction of articular cartilage and, later, of contiguous bone with pannus formation. Rheumatoid nodules and necrotizing vasculitis are rarely, if ever, present in synovial tissue from children with chronic arthritis. Onset types cannot be distinguished on the basis of synovial histopathology.516,517 In RA, the primary infiltrating cell is the T lymphocyte, which may be distributed diffusely throughout the synovium or form nodules or germinal centers. CD4+ helper-inducer T cells and CD8+ suppressor-cytotoxic T cells may be visible. In addition, macrophages and dendritic cells abound in mature synovitis. In late disease, B lymphocytes and plasma cells producing RF can be demonstrated. Multinucleated giant cells and mast cells are also present. Hypertrophy of synovial lining cells, fibroblasts, and blood vessels leads to the development of papillary fronds. Extension of the inflammatory granulation tissue or pannus that spreads from the synovium and invades the cartilage and bone results in osteolysis, which is visible radiographically as erosion and subchondral cyst formation.
■ Figure 9–9 Photomicrograph of synovial tissue from the knee of a young boy with arthritis.Villous hyperplasia and hypertrophy, edema, proliferation of new blood vessels, and infiltration by mononuclear cells are prominent.
C H A P T E R
Erosions occur preferentially in the “bare” areas of the joint, where bone is not covered by articular cartilage (see Chapter 2). They are irregular but sharply defined. Dissolution of the cartilage (chondrolysis) results from enzymatic digestion by neutral proteases, cathepsin, and collagenase from cells of the pannus. Immune complexes may also contribute to chondrolysis. Metaplasia of the granulation tissue may result in the formation of new cartilage, bone, or fibrous tissue, leading to ankylosis. End-stage disease is characterized by deformity, subluxation, and fibrous or bony ankylosis. Joint destruction usually occurs later in the disease course of childhood than in adulthood, and permanent joint damage is absent in many children even after years of chronic inflammation. (Newer studies with MRI are likely to revise the impression of the extent of joint damage in early disease). The greater thickness of juvenile cartilage may offer some protection in this regard. The hyaline cartilage of the hip is destroyed in progressive stages during the course of severe disease, and during healing it may be replaced by a fibrocartilaginous layer. Rice bodies may be present and consist primarily of amorphous fibrous material, fibrin, and small amounts of collagen. Viable cells are incorporated within this matrix and appear more normal than the synovial cells of the inflammatory foci. The majority of these cells resemble type B synovial lining cells, although a few type A cells are also visible. Residual blood vessels in some of these bodies attest to their former attachment to the synovial membrane. The rash is one of the most characteristic hallmarks of the disease (see Chapter 12). It is characterized by minimal perivascular infiltration of mononuclear cells around capillaries and venules in the subdermal tissues. A neutrophilic perivasculitis resembling that of the rash of rheumatic fever may accompany the more flagrant lesions. Subcutaneous nodules may be histopathologically typical of rheumatoid nodules, or they may have a looser connective tissue framework resembling that of the nodules of rheumatic fever (see Chapter 10). Classic rheumatoid nodules consist of three distinct zones: a central area of necrosis and granulation tissue, surrounded by a radially arranged palisade of connective tissue cells that, in turn, is enveloped by chronic inflammatory cells. In children, the central area of fibrinoid necrosis and the epithelioid palisades may be absent or less structured. The serosal lining surfaces of the pleural, pericardial, and peritoneal cavities of the body may exhibit a nonspecific fibrous serositis that is characterized clinically by effusion and pain. Enlargement of the lymph nodes is related to a nonspecific follicular hyperplasia that in rare instances may closely resemble lymphoma. Hepatic abnormalities are characterized by a nonspecific collection of periportal inflammatory cells and hyperplasia of Kupffer’s cells.
LABORATORY EXAMINATION Although the laboratory may provide support for a diagnosis of chronic arthritis, no laboratory test or combination of studies can confirm the diagnosis. The laboratory can be used to provide evidence of inflammation, to support the clinical diagnosis, to monitor toxicity of therapy, and as a research tool to understand more completely the pathogenesis of the disease.
Blood Indices Hematologic abnormalities reflect, in a general way, the extent of the inflammatory disease. Children with limited
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
223
joint disease seldom exhibit any hematologic aberrations beyond that of mild anemia. Those with moderately extensive arthritis usually have a normocytic hypochromic anemia. The anemia may be moderately severe, with a hemoglobin in the range of 7 to 10 g/dL (70 to 100 g/L) in children with severe, uncontrolled disease. Although the anemia is attributable to chronic disease (low serum iron, low total iron-binding capacity, adequate hemosiderin stores),518 iron deficiency may also play a role. Plasma iron transport and iron available for erythropoiesis are reduced in systemic disease. More severe anemia may respond to recombinant erythropoietin in combination with intravenous iron therapy.519,520 Serum ferritin is elevated in children with active arthritis and correlates closely with systemic activity521; in that sense, such elevations do not reflect iron stores but represent a response as an acute phase reactant. Erythroid aplasia has been reported522; hypoplastic crises in some instances may represent a virusassociated reactive hemophagocytic syndrome523 or the macrophage activation syndrome (MAS).524 Leukocytosis is common in children with active disease, and leukocyte counts are strikingly high, 30,000 to 50,000 cells/mm3 (30 to 50 × 109/L), in children with systemiconset disease. Polymorphonuclear leukocytes predominate. The platelet count may rise dramatically in severe involvement; in disease of long standing, thrombocytosis may signal an exacerbation. Thrombocytopenia is rare and may signal an evolution of the disease into SLE.525,526
Erythrocyte Sedimentation Rate and C-Reactive Protein The Westergren erythrocyte sedimentation rate (ESR) is a useful but not totally reliable measure of active disease at onset and during follow-up of a child with arthritis. It is occasionally helpful in monitoring the therapeutic efficacy of a medication program, although the ESR does not necessarily correlate with the articular response to medications.527 The C-reactive protein (CRP) level may be a more reliable monitor of the inflammatory response; at least it is less often increased in a child in whom no clinical inflammatory disease can be found.
Serum Immunoglobulins Increases in serum levels of the immunoglobulins are correlated with activity of the disease and reflect the acute-phase response. Extreme hypergammaglobulinemia is present in the sickest children and returns toward normal with clinical improvement. Viral antibody titers may also be increased, but levels of antibodies to rubella and rubeola viruses are usually similar to those of appropriate control groups or are associated with a polyclonal increase in immunoglobulin concentrations. In one study, 37% of 200 children with JRA had hypergammaglobulinemia, defined as a level of 1.96 SD or higher from normal, in at least one immunoglobulin class.528 In general, persistent hypergammaglobulinemia was an important hallmark of a deteriorating clinical course and poor therapeutic response. Selective IgA deficiency occurred in 4% of the children. In addition, abnormally low levels of IgA were more
224
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
frequent than in normal controls. No relation of immunoglobulin concentrations to age at onset or duration of disease was found. In contrast, another study found normal levels of immunoglobulins in 86% to 94% of children with chronic arthritis.529 Significantly increased concentrations of IgG, IgA, and complement factor C4 were present in children with active disease, whereas elevated IgM levels were characteristic of the disease itself. Studies by the same authors indicated that serum antibody levels to enteric bacteria (Escherichia coli O55 and O86, common antigen, Shigella polyvalent antigen) were normal.530 No abnormalities of immunoglobulin allotypes have been reported.531 Marked increases in low-molecular-weight IgM have been described, presumably reflecting a perturbation of intracellular assembly of subunits.532
Rheumatoid Factors Classic IgM RFs were first detected by assays relying on the agglutination of IgG-coated latex particles or human or sheep erythrocytes. Most laboratories now use a nephelometric method; an enzyme-linked immunosorbent assay (ELISA) has also been employed.133 RFs of other immunoglobulin isotypes have been reported, but their significance is uncertain. The latex fixation and sensitized sheep cell agglutination tests for the detection of classic IgM anti-IgG RFs are positive in a variable percentage of children with chronic arthritis, depending on the age of the population under study and the onset type. RFs are unusual in a child younger than 7 years of age and are seldom helpful diagnostically at onset of disease. In the U.S. Pediatric Rheumatic Disease Registry,533 20 (3%) of 686 children with polyarticular onset were RF positive. In a study by Oen and colleagues534 in Canadian First Nation children, the high frequency of RF-positive polyarticular disease (42%) was probably related to the high prevalence of HLA-DRB1 antigens bearing the RA shared epitope. The diagnostic importance of RF seropositivity in a child with possible chronic arthritis is mitigated by the frequent occurrence of abnormal titers in the other connective tissue diseases of childhood, especially in SLE. In a study of the diagnostic utility of RF serology in children,535 RF tests were as likely to be positive in children with diseases other than chronic arthritis as in that disorder (specificity, 326/332 [98%]; sensitivity, 5/105 [4.8%]). As a diagnostic aid, therefore, tests for RFs are of little utility.
Children with high titers of RFs likely represent a subgroup distinct from the larger number of children with seronegative disease and are identified in separate groups in the EULAR and ILAR classifications. RFs are common in children with later age of onset and polyarticular disease, and in those who are older, have subcutaneous rheumatoid nodules or articular erosions, or are in a poorer functional class. The percentage of children with RF seropositivity also rises progressively as the age at onset or the duration of disease of the cohort group under study increases. They are especially common in the presence of HLA-Dw4 (DRB1*0401) and Dw14 (DRB1*404) specificities. These observations suggest that RFs might be a result rather than a determining event in children who go on to experience unremitting, disabling disease during the early adult years. Anti-cyclic citrullinated peptide antibodies are also found in children with RF-positive polyarthritis.225
There is a general increase in IgM synthesis by peripheral blood B cells.536 Many studies have demonstrated other types of antiglobulins in the sera of children with chronic arthritis or have reported more sensitive and specific tests for RFs.122,130,537–551 The majority of children with seronegative disease have IgG antiIgG antibodies demonstrated by immunosorbent techniques.537,539–541,546 Pepsin agglutinators of the IgG class are found more often in children with disease than are classic RFs.538 Miller and colleagues541 employed five different immunosorbents to search for occult antiglobulins. Antiglobulins detected by binding to sepharose-linked globulin followed by acid elution were found in 8 normal children and 52 children with chronic arthritis. Only 4 children with arthritis had significantly elevated levels; antiglobulins per se were not diagnostically helpful. In the studies of Moore and associates,122,130,351,542–545,547 68% to 75% of children had “hidden RFs,” defined as IgM 19S antiglobulins detected by acid elution of IgM-containing fractions of serum from a gel filtration column. Hidden RFs were found in 59% of children who lacked classic RFs by using a complementdependent hemolytic assay, and their presence correlated well with disease activity. Not only were hidden RFs more frequent, but they were also of higher titer than in healthy children or other disease control groups. Titers correlated with activity of the disease and did not differ significantly between those with polyarticular and oligoarticular disease. Hidden RFs could be inhibited more readily by the use of human IgG than by IgG of animal origin, although the detection system continued to be a hemolytic assay employing rabbit IgG. These studies have focused on North American and European patients. In a survey of 43 children from Turkey,552 19S IgM hidden RFs were present in 56% of patients (46% with oligoarthritis, 64% with polyarthritis, 80% with systemic onset). The latex fixation test was positive in only one patient, who had polyarticular disease. Hidden RFs were present in lower percentages in India102,553–555 and Greece.556
Antinuclear Antibodies Tests for ANAs are more useful than those for RF in diagnosis and classification. Standardized serum dilution titers are usually low to moderate. Most ANAs are of the IgG class, although antibodies of the IgM and IgA classes are found. ANAs of unknown specificities are commonly demonstrable. The frequency of ANAs is highest in girls of younger age at onset, especially in those with oligoarticular disease, and lowest in older boys and in those with systemic-onset disease. ANAs reach their highest prevalence (65% to 85%) in children who have oligoarthritis and uveitis.557,558 Therefore, determination of ANA seropositivity is supportive of the diagnosis and important in identification of children most at risk for chronic uveitis. Studies have demonstrated persistent ANA seropositivity in a small but significant group of children with musculoskeletal complaints in whom no autoimmune or rheumatic disease was found.559–561 Care must therefore be exercised in interpreting the significance of ANA seropositivity in children who do not have objective evidence of arthritis. At the present, tests for ANAs are most often performed on HEp-2 cells. The frequency of ANA seropositivity is increased with the use of this substrate compared with mouse liver, as is background positivity in normal children. The most common patterns are homogeneous and speckled. The antigenic specificities of ANAs have not been identified. Evaluation of specificities by Western blot with HEp-2 cell
C H A P T E R
nuclei demonstrated extensive heterogeneity of reactivity.197 The appearance of antibody to double-stranded DNA (dsDNA) during the course of chronic arthritis should be recognized as a warning of a transition to SLE. In a study of 77 children, the frequency of antibodies to defined nuclear antigens (Sm, RNP, DNA, RNA, RAP, SS-A, and SS-B) was only 13%.562 However, the authors noted a small group of girls with polyarthritis and late onset of disease who had an increased frequency of RFs or antibodies to the RAP antigen. They suggested that these patients might be infected with the Epstein-Barr virus (which is associated with anti-RAP) and that this virus might play a role in pathogenesis. In another study, granulocyte-specific ANAs of the IgG class were found in all children with uveitis but in none of those with acute systemic-onset, and they tended to correlate with the number of affected joints.563 Half of the granulocyte-specific ANAs were capable of fixing complement and correlated with active disease.564
Serum Complement and Immune Complexes The third component of complement (C3) is often elevated in the sera of children with active disease, acting as an acute phase protein. The activated form of the molecule (C3d) may be increased as well. This observation indicates that the pathogenesis of arthritis in children may include complement-mediated tissue damage.126,565–569 Immune complexes are present in the sera of some children with systemic-onset disease and polyarthritis122 and in children who are RF seropositive.123,554 It has been suggested that children with systemic disease might have a defect in antibody-forming capacity or in macrophage function that results in decreased clearance of circulating immune complexes.
Plasma Lipids Dyslipoproteinemia occurs de novo in children with chronic arthritis, separate from the effects of glucocorti-
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
225
coids.570,571 Tselepis and colleagues572 reported that 14 patients with active arthritis had lower plasma cholesterol and high-density lipoprotein cholesterol levels and higher triglycerides than comparable children with another disease or controls. Plasma platelet-activating factor (PAF) acetylhydrolase activity was also decreased in parallel with the activity of the disease. These low levels of PAF acetylhydrolase activity may have resulted in a loss of anti-inflammatory activity, because PAF is a lipid mediator of inflammation.
Synovial Fluid Analysis Synovial fluid is usually a group II or inflammatory fluid (Table 9–8); however, the level of the leukocyte count does not always correlate with the degree of clinical activity. Very low counts, such as 600 cells/mm3 (0.6 × 109/L), have been observed in fluid from joints clinically involved by intensely active and symptomatic disease. Conversely, counts in the range of septic arthritis, such as 100,000 cells/mm3 (100 × 109/L), have been described in children with otherwise classic disease. The principal cellular constituents are polymorphonuclear neutrophils and mononuclear cells, including lymphoid dendritic cells. Synovial fluid levels of glucose may be low, as in adult RA. Synovial fluid complement levels are not as uniformly depressed as in adult disease.573 Rynes and colleagues574 found intra-articular activation of the classic complement pathway in some children, but not of the alternative pathway. Complement activation products, however, were not detected in the joint fluid of children with oligoarticular disease in the study by Miller and associates.575 Complexes of IgG, IgG RF, and complement components along with hidden RFs have been described in both synovial tissue and eluates.573,576 The concentration of glycosaminoglycans in synovial fluid (hyaluronic
TABLE 9–8 Characteristics of Synovial Fluid in the Rheumatic Diseases Group and Condition
Synovial Complement
Color and Clarity
Viscosity
Mucin Clot
WBC Count
PMN (%)
Miscellaneous Findings
Very high High
Good Fair to good
<200 <2,000
<25 <25
Debris
High
Fair to good
1,000
<25
Noninflammatory Normal Traumatic arthritis
Normal Normal
Osteoarthritis
Normal
Yellow and clear Xanthochromic and turbid Yellow and clear
Systemic lupus erythematosus
↓
Yellow and clear
Normal
Normal
5,000
10
Rheumatic fever Chronic arthritis Reactive arthritis
Normal to ↑ Normal to ↓ ↑
Yellow and cloudy Yellow and cloudy Yellow and opaque
↓ ↓ ↓
Fair Poor Poor
5,000 15,000–20,000 20,000
10–50 75 80
Tuberculous arthritis
Normal to ↑
↓
Poor
25,000
50–60
Septic arthritis
↑
Yellow-white and cloudy Serosanguineous and turbid
↓
Poor
50,000–300,000
>75
Inflammatory Lupus erythematosus cells
Pyogenic
PMN, polymorphonuclear leukocyte; WBC, white blood cell; ↓, decreased; ↑, increased.
Acid-fast bacteria Low glucose, bacteria
226
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
acid and chondroitin sulfates) is decreased compared with normal controls, accounting for the low viscosity of inflamed synovial fluid.
tion with MRI to differentiate actively inflamed synovium from synovial fluid.590 MRI may prove especially valuable in permitting an early, accurate diagnosis of many of the rare and unusual causes of monarthritis.591
RADIOLOGIC EXAMINATION
Overview
Imaging Techniques Imaging is an important tool for diagnosing arthritis and monitoring its progression. A systematic approach to the evaluation of radiographs577–581 ensures that maximal information is obtained. Technical advances in imaging techniques since about 1990 have contributed considerably to the radiologic investigation of chronic arthritis. Radiologic abnormalities have been reviewed by Reed and Wilmot582 and mimics of the disorder by Wihlborg and colleagues.583 Although plain film radiography is still the mainstay of imaging techniques, computed tomography, high-resolution ultrasonography, radionuclide imaging, and MRI have begun to contribute considerably to the knowledge of early articular changes. Radiography is the best initial investigation in most situations. The only exception to this rule is radiography of the lumbosacral spine and the sacroiliac joints: Unless the indications are very specific, the information gained may not justify the amount of radiation required. Ultrasonography584–586 is often the best way of identifying intra-articular fluid, particularly in joints such as the hip and shoulder, where fluid may be difficult to demonstrate clinically.584,587 Ultrasound studies of the wrist may help delineate joint effusion from tenosynovitis or a ganglion cyst.588 Computed tomography can demonstrate otherwise poorly defined lesions in the sacroiliac joints, TMJs, or feet. Thermography is not widely available, although it can document the extent and degree of joint inflammation by registering heat output. The role of arthrography in the investigation of arthritis in a child has been largely supplanted by MRI, but arthrography can be helpful in demonstrating popliteal cysts and loose bodies. Radionuclide imaging with technetium 99m (99mTc) is a sensitive but less specific investigative tool. The blood-flow phase illustrates vascular integrity; the blood-pool phase evaluates the homeostasis of inflow versus outflow; the bone-uptake phase demonstrates lesions characterized by a decrease or increase in retention of isotope. Unlike other imaging techniques, radionuclide scans demonstrate hemodynamics and metabolic activity in bone or joint at the time the study is being performed. Radionuclide scans therefore provide early evidence of joint inflammation with increased uptake of the isotope on both sides of the joint. They do not necessarily differentiate inflammatory arthritis from septic arthritis or other joint diseases. MRI has the potential to illuminate understanding of intra-articular pathology in a way that no other imaging technique can (see Fig. 11–8).584 It is now possible to identify abnormalities of noncalcified tissues before they evolve into lesions that result in bony changes detectable by plain radiography.589,590 Intravenous gadolinium is taken up by synovial tissue and can be used in conjunc-
Sequential radiologic changes have been examined in relation to the type of onset of the disease and will be summarized in the following paragraphs.592 Early radiographic changes reflect inflammation; they include periarticular soft tissue swelling and widening of the joint space caused by increased intra-articular fluid or synovial hypertrophy. Juxta-articular osteoporosis and growth-arrest lines are common early abnormalities.582 Generalized osteoporosis may also be significant, especially in the postpubertal girl with polyarticular disease.469 Periosteal new-bone apposition occurs most commonly in the short tubular bones of the phalanges, metacarpals, and metatarsals (Fig. 9–10) but occasionally involves the long bones as well. Widening of the midportions of the phalanges from periosteal new-bone apposition is a characteristic feature of polyarthritis. Later radiologic changes include joint-space narrowing, marginal erosions, subluxation, and ankylosis. Thinning of cartilage is difficult to assess radiographically and is often called jointspace narrowing. In fact, it is the layer of noncalcified cartilage overlying bone, not the joint space, that is narrowed. Such a change is best evaluated in joints of the lower extremity if the radiograph is taken with the patient standing. Erosions are often not generally demonstrable by plain radiographs before 2 years of active disease, even in a child with polyarthritis. Indeed, in some children with limited joint disease, erosions may not be visible even after 1 or 2 decades of constant effusion and swelling of a joint. However, in one long-term study,593
■ Figure 9–10 Severe chronic arthritis in an 8-year-old girl. Demineralization is widespread, and there is damage to the proximal epiphyses of the middle phalanges and soft tissue swelling around the proximal interphalangeal joints. Periosteal new-bone apposition (arrow) is present on the metacarpals.
C H A P T E R
erosive changes of the small joints of the hands and feet were present at 5 years in 67 of 70 children with RF-seropositive disease. Subluxation involves large as well as small joints. Of the large joints, subluxation is most common in the wrist, hip, and shoulder. Subluxation of the tibiotalar, hip, shoulder, elbow, and other joints may also occur. Bony ankylosis occurs earlier in children than in adults and is particularly pronounced in the carpal and tarsal joints and in the cervical spine. Ankylosis can be confirmed by computed tomography. Aseptic necrosis of the femoral head, humeral epiphysis, or tibial plateau is not common, even in children treated with high-dose glucocorticoids for long periods,594 although early series reported osteonecrosis more frequently.595 Some studies reported frequent fractures related to chronic disease or severe generalized osteoporosis,592,596,597 but this is an unusual event in clinical practice today. These abnormalities are particularly common in children who have had severe disease and undergone long periods of immobilization or steroid therapy. The supracondylar area of the femur is a characteristic site of a fracture that occurs after manipulation for contracture of the knee. Microfractures of the growth plates may be related to abnormal mechanical stress. The normally balanced muscle forces about joints are altered by severe joint deformities, erosion, and subluxation as well as by inflammation of the periarticular connective tissues. Abnormal compression forces on the growth plates result.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
227
Localized growth disturbances are among the most remarkable skeletal changes. Epiphyseal ossification centers are advanced in development within weeks after onset of the disease. This finding is most obvious when the disease is asymmetrical (Fig. 9–11). Radial atrophy (overtubulation) often accompanies linear overgrowth of the long bones. Even if arthritis is confined to the knee, growth of the pelvis on the same side may be stunted, and coxa valga may be present. Growth abnormalities were particularly evident in children with monarticular or oligoarticular disease. Longitudinal overgrowth was the rule, although affected bones were generally smaller in diameter than normal. Brachydactyly and micrognathia occurred predominantly in children with systemic disease and in those with early age at onset; it occurred to a lesser extent in those with polyarthritis. Accelerated epiphyseal maturation during active disease was sometimes associated with future stunting of full growth of the affected bones. Spondylitis typical of chronic arthritis was noted in all regions of the axial skeleton. Abnormalities of the upper segments of the cervical spine were the most characteristic change, and a few children had arthritis of the cervical spine at onset of disease. Atlantoaxial subluxation and disease of the thoracolumbar spine did not develop in children with monarthritis, even of long duration. Dwarfing or remodeling of the dorsolumbar vertebral bodies, probably attributable to arthritis of the apophyseal joints of that region, occurred in a few patients. Sacroiliac arthritis, both with and
■ Figure 9–11 These radiographs illustrate localized growth disturbances in children with chronic arthritis. A, Arthritis persisted for 7 years in the left wrist of this 9-year-old girl, resulting in osteoporosis and acceleration of maturation of growth centers and a small hand. B, Monarthritis of the left knee has persisted for 8 years in this 14-yearold girl, causing osteoporosis, epiphyseal advancement and enlargement, discrepancy in leg length (left leg longer than right leg), and radial atrophy of the long bones. C, Pelvis of the patient described in B. Arthritis has not been present in the hip, and the regional demineralization and miniaturization of the left side of the pelvis are secondary to the effects of inflammatory disease of the left knee.
228
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
without coincident cervical disease, was present in four children but was distinguishable from that of JAS. Early findings of soft tissue swelling or osteoporosis were present in 45% of children with polyarthritis or systemic-onset disease and in 75% of those with oligoarthritis. Periosteal newbone formation and striking metaphyseal rarefaction were found only in children with polyarthritis and systemic-onset disease. Metaphyseal bands related to focal osteoporosis and hyperemia of the zone of ossification of the epiphyseal plate were common and similar to those that accompany acute leukemia (Fig. 9–12). Periosteal new-bone apposition occurred adjacent to involved joints but not in children with monarthritis or oligoarthritis, despite a subsequent polyarticular course in some of these patients. Advanced radiologic changes were also related to the type of onset of disease (Fig. 9–13). Destruction of cartilage and bone was marked in children with polyarticular or systemiconset disease and was less frequent and usually less severe even in those with oligoarthritis who subsequently developed polyarthritis. Bony ankylosis was a late change. Large joint subluxation, especially at the hip, was particularly characteristic. Fractures of the epiphyses and vertebral compression fractures occurred only in children with severe, long-standing disease. The increased frequency of these findings in children with systemic disease was probably related to extended use of glucocorticoid drugs in this group, because fractures were rare in children in whom these agents had not been used. Fractures were not observed in children who had oligoarthritis.
Changes in Specific Joints Elbow, Wrist, and Hand Evaluation of arthritis affecting the hands is challenging because of the necessity to differentiate changes caused by arthritis from those reflecting bony maturation. Poznanski and colleagues598 proposed a scoring system to quantify the relative amounts of wrist cartilage. This measurement, the relative carpal length (RCL), compares the distance from the base of the third metacarpal to the midpoint of the distal radial growth plate with the length of the second metacarpal bone. The lower this ratio, the greater the severity of wrist and intercarpal joint cartilage loss. Mason
■ Figure 9–13 Severe symmetrical erosions of the heads of the second through fifth metatarsals.The poorly corticated lesions are prominent on the inferomedial aspects of the bones.
and colleagues599 found that one third of patients with polyarticular JRA had an RCL greater than 2 SDs below the mean for age, usually accompanied by periarticular osteopenia, joint space narrowing, or erosion at the time of diagnosis. Erosions, subluxations, and enlargement of the radial head may occur. Subluxation at the radiocarpal joint is a characteristic radiologic abnormality, particularly in the child with long-standing wrist disease. Reduced carpal length is a consistent finding.598 Bony ankylosis of the carpal bones is another characteristic change (see Figure 10–13), and similar changes may occur in the metacarpophalangeal (MCP) joints. In the wrist, there may be disproportionate shortening of the ulna, with the result that the radius compensates by becoming bowed. Pronounced ulnar subluxation of the wrist is characteristic. A recent study evaluated radiographic progression and disability in children with JIA.600 Marked bony overgrowth and enlargement of the epiphyses may also occur at the interphalangeal joints. Failure of growth of small tubular bones results in brachydactyly. This condition is occasionally but not always related to premature epiphyseal fusion, especially if selective stunting of one or two bones occurs. A common site for this finding is the fourth metacarpal bone. Asymmetrical fusion may occur if only a portion of the epiphyseal plate is affected, resulting in abnormal angulation of the joint. Radial deviation at the MCP joints is more characteristic than ulnar drift. In severe, late, uncontrolled disease, bony destruction in the hand may be extensive.
Shoulder
■ Figure 9–12 Zones of metaphyseal rarefaction in both tibias (arrow). These abnormalities are not specific for chronic arthritis and are later replaced by growth arrest lines.
The shoulder is involved in fewer than 10% of children at onset of disease, although perhaps one third eventually develop involvement of this joint.601 Children with oligoarthritis rarely have shoulder disease, but in those with polyarticular or systemic-onset disease, the frequency of involvement rises to 50% or 80%, respectively, and involve-
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
229
■ Figure 9–14 In A, the left mandibular condyle is eroded and flattened. In B, the ramus of the mandible is shortened. (A and B, courtesy of Dr. B. Blasberg.)
ment is almost always bilateral.601 Loss of ROM is most marked in internal rotation and is accompanied by marked atrophy and weakness of the rotator cuff. Superior subluxation of the shoulder and avascular necrosis may result. Disease of the acromioclavicular joint is uncommon.
Temporomandibular Joint The TMJ is usually evaluated by Panorex views and, if necessary, by tomography or MRI.602 Flattening or even complete dissolution of the mandibular condyle may occur (Fig. 9–14).592,603–606 Mandibular asymmetry with undergrowth on the affected side may be present if the disease predominantly affects one TMJ.607,608
rowing and fusion occur but are more characteristic of seronegative spondyloarthritis (tarsitis).613 Erosive disease (see Fig. 9−13) and localized growth disturbances (Fig. 9–15) are typical of severe polyarticular disease. MRI is particularly useful for identification of the extent of inflammation of the ankle joint and may confirm the presence of synovitis at a multiplicity of sites: tibial-talar, subtalar, and talonavicular joints, and peroneal and posterior tibial tendon sheaths.614
Knee Rosberg and Laine609 noted that the medial femoral condyle tended to enlarge more than the lateral condyle in children with arthritis of the knee. Overgrowth of the patella may be marked in long-standing disease. Erosion and enlargement of the intercondylar notch of the femur occur. These changes have also been reported as characteristic of hemophilia and tuberculosis. Cysts in the popliteal fossa are best demonstrated by ultrasound imaging.610 Children with monarthritis in whom the diagnosis is uncertain should undergo MRI to evaluate for other causes of persistent joint swelling.591,611
Ankle, Subtalar Joints, and Foot Loss of “joint space” is frequent in long-standing arthritis of the ankle or subtalar joints.612 Aseptic necrosis of the dome of the talus may also occur. Intertarsal joint nar-
■ Figure 9–15 Brachydactyly of the fourth metatarsal bones and juxtaarticular osteoporosis of the fourth metatarsophalangeal joint in a child with chronic arthritis affecting those joints.
230
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
Hip Articular inflammation and muscular imbalance influence growth and architecture of the hip joint.615,616 Subluxation of the hip is best evaluated on a weight-bearing anteroposterior radiograph to confirm upward and lateral displacement of the femoral head (Fig. 9–16). Ultrasonography may demonstrate increased intra-articular fluid or synovial hypertrophy, particularly between the femoral head and the medial margin of the acetabulum. Protrusio acetabuli may also develop and is sometimes rapidly progressive. Osteonecrosis of the femoral head is not as frequent as in SLE, but if suspected from the plain films, it should be evaluated by a radionuclide scan or MRI.617,618 Kobayakawa and coworkers595 reported a high frequency of necrosis of the femoral head. Of 206 children hospitalized for treatment, 36 (17.5%) had pain or restricted hip motion. Definite radiographic evidence of avascular necrosis of the femoral head (condensation, fragmentation, resorption, and flattening of the epiphysis) was found in 10 hips in 6 children, and signs suggestive of this diagnosis were found in a further 20 hips in 13 children. A sclerotic rim at the base of the femoral neck (suggesting earlier ischemia) was also observed. A more recent study demonstrated a low frequency of osteonecrosis.594
■ Figure 9–17 A, Fusion of the spinous processes of the second and third cervical vertebrae in a teenage girl with early-onset disease. B, More extensive fusion of spinous processes resulting in a severe pseudoarticulation and accompanied by destructive changes at the C5–C6 junction.There were no neurologic signs.
Spine Characteristic radiologic abnormalities of the axial skeleton occur in children with chronic arthritis, depending on onset type.437,619 The predominant changes are in the apophyseal joints of the upper cervical segments. Bony fusion is frequent and often is observed first at the C2–C3 level (Fig. 9–17). Fusion of adjacent spinous processes may also be present. A single lateral film of the cervical spine may not be sufficient to determine the presence of ankylosis in this location. The distances between spinous processes in flexion and in extension should be compared to confirm that fusion has occurred. Atlantoaxial subluxation is also common. The upper limit of the
■ Figure 9–16 Marked subluxation of the left hip of a 5-year-old boy with onset of severe, unremitting arthritis at the age of 11⁄2 years.The femoral neck is osteoporotic, and there is aseptic necrosis of the femoral head, which is subluxed laterally and superiorly out of a shallow, poorly formed acetabulum.
atlanto-odontoid distance in children is approximately 4 mm, measured at the bottom of the arch of C1 on a lateral film taken in flexion with a 40-inch tube-to-film distance. There is normally a small amount of displacement between the bodies of C2 and C3 in the young child (2 to 12 years of age); this change should not be misinterpreted as early subluxation. Instability of the atlantoaxial joint may lead to impingement on the cord and brainstem (Fig. 9–18). There may also be cephalad encroachment of the odontoid into the foramen magnum. Locke and colleagues620 studied the atlanto-odontoid distance in 200 normal children aged 3 to 15 years. If the measurement was greater than 4 mm in the neutral position, an atlantoaxial subluxation was usually confirmed. Age and sex were not significant factors in evaluating these measurements. Other signs of atlantoaxial subluxation were increased tissue density anterior to the cervical spine, flexion greater than 10 degrees between the atlas and the axis, compensatory curve of the lower cervical spine, and narrowing of the atlantovertebral foramen. Narrowing of an intervertebral disk associated with atrophy or maldevelopment of adjacent vertebral bodies probably reflects fusion of the adjacent apophyseal joint or joints, even if it is not well delineated radiologically. Vertebral bodies at areas of fusion fail to grow normally and are smaller and narrower than contiguous vertebral bodies (an altered ratio of height to width). The corresponding intervertebral spaces are reduced, and the disk is sometimes calcified. Vertebral compression fractures are sometimes apparent, especially in children who have been treated with glucocorticoids (Fig. 9–19), and commonly occur in the thoracolumbar vertebrae. Scoliosis of the thoracolumbar spine was previously more common in children with chronic arthritis than in healthy children.436 Sacroiliac arthritis is not characterized by the degree of reactive sclerosis that occurs in JAS.437,621 In long-standing disease, however, there may be subchondral sclerosis and secondary cartilaginous space narrowing. Late fusion may occur in chil-
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
231
■ Figure 9–18 A, Lateral spine of a 12-year-old girl with systemiconset disease at the age of 11 years, showing atlantoaxial subluxation (arrow) and fusion of C3–C4. B, Magnetic resonance image of the cervical spine at age 26 years. Note the impingement of the odontoid on the upper cervical cord (arrow). (B, Courtesy of Dr. J. W. McCune.)
dren with severe disease who have been in a wheelchair or at bed rest for long periods. Patient characteristics and disease variables have been evaluated for influence on the development of sacroiliac arthritis in children with chronic arthritis.23
TREATMENT Approach to Management Although chronic arthritis cannot yet be cured, spontaneous remissions fortunately occur in many children,
and, in the interim, disease control is hopefully achievable. The aims of treatment are, therefore, to control pain and preserve ROM, muscle strength, and function; to manage systemic complications; and to facilitate normal nutrition, growth, and physical and psychological development (see Chapter 8) (Table 9–9). Along with advances in therapeutics has come a raised expectation for disease control. An “adequate” response is no longer considered to be acceptable under most circumstances. The possibility of absolute control of inflammation is a goal that should be pursued within the constraints of safety and cost. Increasingly, the aim of treatment is remission rather than improvement. Although the major focus of medical therapy is on the arthritis, other extra-articular complications (e.g., uveitis, serositis, growth retardation, osteopenia) require consideration. The treatment program should be family centered, community based, and well coordinated.622,623 An ideal approach involves a multidisciplinary team that consists of a pediatric rheumatologist, nurse clinician, social worker, physical therapist, occupational therapist, and psychologist. Consultation with a physiatrist, psychiatrist, orthopedic surgeon, dentist, or nutritionist is often indi-
TABLE 9–9 Objectives of the Treatment of Chronic Arthritis in Children Immediate Relieve discomfort Preserve function Prevent deformities Control inflammation Long-term
■ Figure 9–19 Radiograph of the lumbar vertebrae of a 12-year-old girl with severe debilitating disease requiring glucocorticoid therapy, showing numerous areas of vertebral compression.
Minimize side effects of disease and treatment Promote normal growth and development Rehabilitate Educate
232
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
TABLE 9–10 Psychosocial and Educational Management of Chronic Arthritis in Children Understand the patient’s and parents’ goals Understand the psychologic effect of fluctuations in disease activity Undertake early realistic career planning Emphasize the benefit of regular school participation Facilitate normal peer group interaction
cated. Regular ophthalmologic consultation is mandatory. Because this rheumatic disorder is characterized by chronic or recurrent inflammation of the joints and varying systemic manifestations, the child and family must accept the need for long-term treatment and surveillance. Patient and family education and incorporation of the family’s needs into the management program are essential to facilitate optimal compliance and therapeutic benefit.624–626 Most children with chronic arthritis require a combination of pharmacologic, physical, and psychosocial approaches (Table 9–10). A priority in management is to foster normal psychological and social development and participation in peer-group activities. Attendance at school is strongly encouraged; only rarely is home instruction justified.627 The child should also remain in the physical education program at school (if at all possible), if not as a participant then as an involved member of the class assigned to alternative activities; otherwise, the child should be enrolled in adaptive physical education. Children should largely determine their own level of activity. Inappropriate restriction of recess time or peergroup association can be harmful both physically and psychologically. Participation in recreational activities and camps with other children with arthritis is frequently of benefit. Children who attend camps realize that they are not the only ones with arthritis, profit from group educational experiences, and have the opportunity to test their limits in a safe, supportive, and understanding environment. Vocational evaluation and advice is appropriate in the adolescent with arthritis.
Basic Medical Program The systematic approach to pharmacologic treatment is to begin with the safest, simplest, and most conservative measures.628–631 If this approach proves inadequate, other therapeutic modalities are promptly selected in an orderly progression. It is now realized that the more rapidly inflammation can be controlled, the less likely it is that there will be permanent sequelae. NSAIDs are the mainstay of initial treatment in approximately 75% of patients. Three developments, however, have fundamentally altered the current therapeutic approach beyond that point: Intra-articular glucocorticoids have proved effective in treating joint disease; low-dose methotrexate has substantially changed the choice of treatment options; and new therapeutic modalities such as etanercept and infliximab promise even further improvements in the risk/benefit ratio.632 Although combination pharmacotherapy is attractive in a child with severe disease for whom more limited regimens have failed, adequate clinical studies of effectiveness and safety are generally
lacking.633,634 Specific recommendations for therapy are presented in Chapters 10 through 12. It is not usually possible at onset of the disease to predict which children will recover and which will go on to have unremitting disease with lingering disability or enter adulthood with serious functional impairment.635–637 Therefore, the initial therapeutic approach must be vigorous in all children. Furthermore, therapeutic strategies must recognize differences in approaching the three types of onset: oligoarthritis, polyarthritis, and systemic disease. Evolution of the nine course subtypes in the ACR classification, and recognition of prognostic indicators, will lead to ongoing modifications of the program in keeping with the response of the child. Evidence of improvement is based on a review of the clinical course, repeated physical examinations, charting of the articular severity index and global responses, and laboratory estimates of inflammation.638,639 These evaluations have been systematized in the so-called core set variables.640 A clinically significant response is an improvement of at least 30% in at least three of the variables, with worsening by no more than 30% in not more than one variable. More demanding criteria require 50% or 70% improvement.641 It is undoubtedly true that a number of these evaluations are redundant.642,643 A preliminary definition of a disease flare in JRA has also been studied.644 It is almost impossible to be confident of the risk/benefit ratio for many of the therapeutic regimens for children with chronic arthritis.645 The possibility that there may be important differences in the therapeutic efficacy of antirheumatic drugs depending on ethnicity and genetic background has been largely unexplored.646 Experimental data and clinical observations confirm the efficacy of NSAIDs, gold compounds, methotrexate, glucocorticoids, and etanercept. It is not as certain that other medications, such as hydroxychloroquine, sulfasalazine, D-penicillamine, intravenous immunoglobulin (IVIG), or cyclosporine, confer a statistically and clinically significant benefit. Experience with the biologic agents is still limited, in terms of both number of children treated and duration of follow-up. Toxicity is often a foremost concern in the longterm use of steroids or immunosuppressive agents such as azathioprine or cyclophosphamide. Because of the relative lack of scientific guidance by appropriately designed studies, undertaken in adequate numbers of children with appropriate control of confounding factors such as type of onset and course of the disease, the experience and judgment of the pediatric rheumatologist often become the most important principles of guidance. Alternative or complementary therapies, although of no proven benefit, are widely used by children with chronic arthritis. Their use is often fostered by information or misinformation available through the World Wide Web. Parents should be directed to more objective sources of information, such as the Arthritis Foundation’s Guide to Alternative Therapies.
Consideration should be given to continuation of an NSAID for at least 3 to 6 months after all evidence of active disease has disappeared. Methotrexate therapy should probably be continued for 1 year or longer after a remission has been achieved. One might also consider a different mode of withdrawal, such as decreasing administration to every 2 weeks for a period of time before discontinuation.628,647 In combination therapy, a consensus has not developed on withdrawal of medica-
C H A P T E R
tions. Perhaps NSAIDs can be discontinued first along with steroids, methotrexate tapered next, and then, finally, etanercept stopped. In children, potentially toxic regimens such as prolonged use of steroid and immunosuppressive drugs should be employed only in uncontrolled or life-threatening disease.
Nonsteroidal Anti-inflammatory Drugs In most children, one of the NSAIDs is used in the initial approach to therapy (see Chapter 5). All of these drugs have antipyretic, analgesic, and anti-inflammatory actions, as well as—for those that have been approved for use in children—a record of long-term safety. Most currently used NSAIDs inhibit the activity of cyclooxygenases 1 and 2 and therefore have the potential to induce GI irritation.648,649 Although this side effect is unusual in children, ranitidine and misoprostol may ameliorate it.650 The new selective cyclooxygenase 2 (COX-2) inhibitors have not generally been evaluated in children.651 Basic immunizations should be continued, consistent with guidelines appropriate for the geographic area.652 Recent observations in clinical trials in older adults have suggested an increased risk of cardiovascular events secondary to NSAIDs. There are no comparable studies in children. Of particular concern is the allegation that this risk was increased with the administration of naproxen. The possibility exists that this toxicity (if confirmed) may be a “class effect” extending beyond the Cox-2 drugs. Therefore administration of any NSAID should likely be accompanied by an explicit dicussion of the risk/benefit ratio and the requirement of additional clinical data before a firm judgment can be made on its role in the treatment of inflammatory and rheumatic diseases. There is no convincing evidence that one NSAID is superior to another. The choice among many possibilities should be based on the documented experience with the drug in children, its tolerability, toxicity, convenience of use, and cost. Only a few of the NSAIDs have been approved in the United States for use in children (i.e., naproxen, tolmetin, and ibuprofen). Results of evaluations of several other NSAIDs indicate that they are as effective, but no more so, than salicylates, naproxen, or tolmetin. The clinical trials of NSAIDs by the Pediatric Rheumatology Collaborative Study Group (PRCSG) led to the conclusion that approximately 65% of children who were going to respond did so by 4 weeks of therapy, although some children were late responders: A 100% level of response in patients who were deemed to be responders was not obtained before 12 weeks in 72 of the 127 children who progressed to a favorable outcome.653 Clinical response to an NSAID is variable and relatively unpredictable. A child may not respond to one drug, or may eventually lose an initial response, and yet may respond to another. In such cases, it is logical to select an NSAID from a different chemical class than that first used (salicylic, propionic, indoleacetic, pyrrolealkaonic, or N-phenylanthranilic acids or oxicams; see Chapter 5). However, except for issues of toxicity, it is not often useful to try one NSAID after another in an attempt to find one to which the patient will respond; the effect of this approach may be to delay institution of more effective second-line therapy. Naproxen is effective in the management of joint inflammation in a dose of 15 to 20 mg/kg/day given with food. It needs to be given only twice daily, and it is available as a suspension
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
233
(125 mg/5 mL), a definite advantage in the treatment of young children. Naproxen is usually well tolerated, although mild epigastric discomfort is occasionally encountered. Cutaneous pseudoporphyria, a side effect of this drug and occasionally of other NSAIDs, is characterized by a bullous eruption on the face, hands, and other sun-exposed areas, which often leaves an irregular, shallow scar (see Chapter 5).654–658 Ibuprofen is a relatively mild anti-inflammatory agent and is usually well tolerated in a dosage of approximately 35 mg/kg/day, divided into 3 or 4 doses given with food. The suspension (100 mg/5 mL) consists of both S and R enantiomers and is not absorbed as well as the tablets.659 It should therefore be given in a dosage of approximately 45 mg/kg/day divided into 3 or 4 doses a day. Tolmetin, given with food in 3 divided doses totaling 25 to 30 mg/kg/day, is equally effective as an anti-inflammatory drug. Diclofenac may be useful in children who are unable to tolerate other NSAIDs because of gastric side effects. It is prescribed in 3 doses per day; a slow-acting preparation is also available. Sulindac is an inactive prodrug that is converted in vivo to the active sulfide and therefore offers little theoretical exposure to the GI mucosa. It has also been suggested that it is less nephrotoxic than other NSAIDs. Celecoxib, rofecoxib, and more recent analogues of the COX-2 inhibitors have been introduced for treatment of arthritis in adults. These drugs are reputedly less likely than traditional NSAIDs to cause gastric irritation and peptic ulcer disease.649 Other NSAIDs may have specific benefits, although their use in childhood has often not been officially approved. Indomethacin, in a dose of 1 to 3 mg/kg/day to a maximum of 125 mg/day, is useful in treating the fever and pericarditis of systemic-onset disease. In many children, intermittent fever responds only to prednisone or, alternatively, to indomethacin. Indomethacin is a potent anti-inflammatory drug. It has been of particular value in older children, but its overall use has been limited, perhaps inappropriately, by headache or epigastric pain, and occasionally by serious side effects of masked infection and sudden death.660 Piroxicam needs to be given only once a day, and it may be particularly useful in the older child or adolescent in whom compliance with taking prescribed medication is a problem.
Aspirin The role of aspirin as the drug of choice in initial management has been supplanted by the NSAIDs. The reasons for this are related more to convenience of administration and relative freedom from side effects than to superior efficacy. Aspirin probably resulted in more frequent instances of transaminasemia than the newer NSAIDs. Nonetheless, considering its long and extensive use, aspirin has a remarkable record of effectiveness in suppressing fever and other aspects of inflammation and a proven record of long-term safety. Its epitaph has not yet been carved in stone, and aspirin will probably continue to be a part of the pediatric rheumatologist’s anti-inflammatory armamentarium because of its low cost. Aspirin is started at 75 to 90 mg/kg/day in 4 doses given with food to minimize gastric irritation and to ensure therapeutic blood levels. Lower doses per kilogram may be indicated in children who weigh more than 25 kg. The therapeutic serum salicylate level is 20 to 25 mg/dL (1.45 to 1.8 mmol/L), usually measured 2 hours after the morning dose. It may be difficult to reach therapeutic levels in children with acute systemic disease, but increasing the dose beyond 130 mg/kg often results in salicylism. If high doses are required initially, they should be reduced gradually to maintenance levels as the
234
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
systemic manifestations of the disease subside. Awakening the child at night to administer aspirin is not necessary, because the serum half-life of salicylate is prolonged once therapeutic levels have been achieved. Aspirin should not be chewed, because it can cause gingival inflammation and erosions of the biting surfaces of the teeth. An adequate trial of aspirin should last at least 6 weeks. Choline magnesium trisalicylate (liquid) may offer an alternative therapeutic agent to use of the NSAIDs until questions on cardiovascular risk are addressed in children. Aspirin and other NSAIDs are associated with interstitial nephritis and renal papillary necrosis. A few children may develop analgesic nephropathy when taking aspirin alone.661,662 Azotemia and hypertension do not occur in children with NSAID nephropathy per se, but the NSAID should be discontinued or the dose reduced if renal involvement is suspected. It is estimated that this complication occurs in approximately 1% of children who receive NSAID therapy.
Analgesics Although it is not an anti-inflammatory drug, acetaminophen given 2 to 3 times a day may be useful for control of pain or fever in the systemically ill child. This drug should not be used for long-term basic management of the disease because, when used with an NSAID, it may contribute to interstitial nephritis. Ketorolac has profound analgesic effects but less prominent anti-inflammatory effects. Its principal use is in the short-term management of pain. The therapeutic role for the glucosamine compounds, if any, awaits clarification.
Methotrexate Methotrexate, because of its relatively rapid onset of action, efficacy, and acceptable toxicity, is considered the initial second-line agent for treatment for most children with chronic arthritis.663–670 The advantages of this medication are efficacy at relatively low doses, oral administration, once-a-week dosing, and apparent lack of oncogenicity or production of sterility.667,671–673 Although most patients who respond do so by 3 months, occasionally a child may require a longer period of treatment. Principal toxicities are directed at the bone marrow, liver, and, very rarely, the lung (see Chapter 5). Cirrhosis of the liver is not an expected toxic effect in children on weekly therapy.674–677 Malnutrition, viral hepatitis, diabetes mellitus, obesity, and alcohol consumption increase the risk. In this respect, the guidelines recommended by the ACR should be observed.645,678–680 Routine liver biopsies are not indicated. Methotrexate-induced pneumonitis and effects on pulmonary function have been reported rarely in children (Fig. 9–20).681–684 Accelerated nodulosis is uncommon.672,681,685 There is no consistent effect on bone mass.686 Because some NSAIDs may interfere with the protein-binding and excretion of methotrexate, their dosage should be kept constant during treatment. Folic acid, 1 mg/day (or folinic acid687,688) is given during treatment with methotrexate to reduce GI and mucosal toxicity with no diminution in therapeutic effectiveness.
■ Figure 9–20 Posteroanterior (A) and lateral (B) radiographs of the chest of a 131⁄2-year-old girl who developed polyarticular disease in the summer of 1997. In December of that year, low-dose methotrexate was started at 7.5 mg once a week.These films were obtained in May 1998.They demonstrate a diffuse, fine, fibronodular, interstitial infiltrate throughout the lower two thirds of the parenchyma. At that time, she was asymptomatic and there were no abnormalities on examination of the chest. Pulmonary function tests confirmed a mild restrictive pattern with an initial diffusing capacity for carbon monoxide 79% of predicted. High-resolution computed tomographic examination confirmed the presence of bilateral, linear, and reticular nodular interstitial infiltrates. The methotrexate was stopped and, over the course of the next year, the radiographic appearance gradually returned toward normal, as did pulmonary function.
C H A P T E R
Monitoring for toxicity includes a complete blood cell count, white blood cell differential and platelet counts, and measurement of serum levels of liver enzymes and albumin. The drug is given as a single weekly dose on an empty stomach with clear liquids 45 minutes before breakfast or, alternatively, at bed time at least 2 hours after dinner, especially on a Friday or Saturday, to avoid missing school if ill. The injectable preparation (25 mg/mL) can be used orally in children in order to prescribe the correct amount. The minimum oral starting dose is 10 mg/m2 weekly (equal to or greater than 0.3 mg/kg). If clinical response is inadequate, or if oral administration is associated with nausea, vomiting, a trial of subcutaneous administration should be attempted, or a higher dose range (0.65 to 1.0 mg/kg, with a maximum dose of approximately 30 to 50 mg/week) should be chosen for a trial of effectiveness.689–691 GI absorption of oral methotrexate in older children is 50% to 80% at less than 20 mg/week, but unreliable at 20 mg or greater per week. Determinations of plasma levels were previously recommended but are not generally necessary.692,693 Intramuscular administration, as an alternative to the subcutaneous route, is effective but not necessary.691,694 Methotrexate is discontinued if no objective response is documented or if toxicity develops despite a reduction in dose. Clinical improvement of arthritis and relatively low toxicity have been reported in a number of studies.663–666,673,689,692,695–701 Assessments included number of swollen joints, morning stiffness, systemic features of the disease, mean daily dose of glucocorticoid, ESR, and CRP concentration. Reviews by Wallace and colleagues698 and others667,695,702,703 have suggested that virtually all children respond to methotrexate and approximately one half achieve a remission. Ruperto and colleagues704 reported a 66% response rate. The poorest response is associated with systemiconset disease and the best with oligoarthritis.705 The double-blind random trial of methotrexate by the PRCSG included two treatment groups, one receiving 5 mg/m2/week and one 10 mg/m2/week, along with a placebo control group.666 Statistical improvement in articular severity was observed, with a mean of 26 for the placebo group and 63 for the 10-mg group. Recalculating these data by the newer core set of criteria for response640,641,704 (3 or more of any 6 core variables improved by at least 30%, with up to 1 variable worse) indicated that 72% of children improved with methotrexate, compared with 44% of the placebo group.697,706 The drug was discontinued in 2 of 45 patients (hematuria, persistent transaminasemia). One placebo patient developed severe GI irritation. A report of Wallace and coworkers692 included 23 children with seropositive polyarthritis who were treated with doses of methotrexate that ranged from 0.1 to 0.6 mg/kg/week. Twenty-one children improved significantly, and remission occurred in 2 children. It was possible to reduce the dose of glucocorticoids in 6 of 9 children. The average time to clinical response was approximately 3.3 months (range, 1 to 8 months). It was not possible to identify clinical involvement that would predict improvement. A study by Harel and associates707 found that carpal length as a measure of cartilage loss was significantly improved or slowed in 11 of the 17 children who responded to 2.5 years of therapy, during which all of the nonresponders had a deterioration of carpal length. Ravelli and colleagues708 provided further evidence of the effectiveness of methotrexate in that there was radiologic
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
235
improvement or a slowing of deterioration of carpal length in children who had responded during a 2-year period of administration; carpal length was significantly worse in nonresponders as a measure of joint destruction. A younger age at onset and being a male correlated with a higher risk of poorer radiologic outcome. The extended oligoarticular course subtype was the best predictor of efficacy.600,709 It is somewhat difficult to categorize methotrexate as an antiinflammatory drug or a remittent drug. Administration is associated with immunologic changes, such as a decrease of double negative (CD4−CD8−) and γ/δ T-cell levels.192 Methotrexate therapy should be continued even after a sustained clinical remission of the disease is achieved.705,710 Early withdrawal has resulted in an exacerbation of the arthritis, and reinstitution of the drug did not always result in a satisfactory clinical response. However, in children who have gone into remission, it is necessary to discontinue methotrexate at some time—the question is when.706 A study by Ravelli and coworkers710 indicated that one third of children relapsed after discontinuation of the drug. It has been reported that clinical response to the drug can be evaluated by a functional index.700 The child most at risk was the one who had an oligoarticular onset with a subsequent polyarticular course.709 A report by Gottlieb and colleagues705 strengthened the conclusion that methotrexate is a remittive agent. Approximately 50% of children in all onset groups who achieved a remission developed a relapse approximately 11 months after withdrawal of the drug. Only 50% regained control of the disease an average of 15 months after reinstitution of therapy. The most convincing evidence of the disease-modifying nature of methotrexate was the long period of time after withdrawal before relapse (range, 1.5 to 36 months). The authors identified younger age at onset (less than 5 years) as indicating a more likely chance of relapse.
Glucocorticoid Drugs Glucocorticoid medications are indicated for uncontrolled or life-threatening systemic disease, in the treatment of chronic uveitis as local ophthalmic drops or injections, and as intra-articular agents.
Systemic Glucocorticoids Systemic glucocorticoids should be instituted only with a well-considered therapeutic plan and a clear set of clinical objectives. Disease control may not be possible in some patients except at a cost of unacceptable glucocorticoid toxicity, and some inflammatory activity may have to be accepted in exchange for less drug toxicity. Systemic glucocorticoid therapy, even in high doses, probably does not limit the duration of active disease, prevent extra-articular complications, or alter the eventual outcome of the disease. Given orally or intravenously, these agents are more suited to management of systemic rather than articular disease. Although the use of glucocorticoid drugs alone to suppress joint inflammation is to be discouraged, lowdose or alternate-day prednisone is of benefit in children with severe polyarthritis unresponsive to other therapeutic programs. Low-dose prednisone711 (i.e., 0.1 to 0.2 mg/kg) can be used as a “bridging” agent in the initial treatment of the moderately to severely affected child who is started at the same time on other, slower-acting
236
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
anti-inflammatory drugs. For severe uncontrolled systemic manifestations with marked disability, prednisone is often prescribed as a single daily morning dose of 0.25 to 1.0 mg/kg/day (maximum 40 mg) or in divided doses in more severe disease. After satisfactory control of the systemic manifestations is achieved, or after it is obvious that acceptable control is not attainable, prednisone should be gradually decreased, if at all possible, as advanced therapy is introduced. Prolonged use of systemic glucocorticoids leads to iatrogenic Cushing’s syndrome, growth suppression, fractures, cataracts, and increased susceptibility to overwhelming infection (see Chapter 5). In spite of these unavoidable toxicities, systemic glucocorticoids are important and effective agents for treatment of some of the most serious manifestations of chronic arthritis.712 Growth retardation commonly develops in children who receive long-term steroids,713,714 even in doses comparable to physiologic replacement levels (i.e., prednisone 3 to 5 mg/m2/day or 0.075 to 0.125 mg/kg/day).457 This effect (among others) is related to blunting of growth hormone release, interference with collagen synthesis, delayed skeletal maturation, and negative effects on IGF reactivity. Effects of glucocorticoids on growth and bone mineralization may be ameliorated by the use of deflazacort, if it is available.715,716 It is important that the child and family be aware of the signs and symptoms of glucocorticoid deficiency and understand that the drug must not be discontinued abruptly under any circumstance because of the risk of precipitating an adrenal crisis. Vomiting or diarrhea in a child who is receiving steroids requires a parenteral glucocorticoid in an equivalent dose (see Chapter 5). Children taking systemic glucocorticoids should wear an identification bracelet or necklace indicating their name, diagnosis, and the fact that they are taking a steroid. This precaution facilitates emergency intravenous steroid administration in the presence of serious trauma or stress. Supplemental steroid is also necessary before and during surgical anesthesia. Although a number of choices exist for the specific drug to be used in these circumstances, one preference is dexamethasone for intramuscular administration before the operation and intravenous hydrocortisone during the procedure. It often becomes difficult to reduce the dose of a glucocorticoid because of a child’s adaptation to chronic steroid excess. Steroid pseudorheumatism may complicate even slow withdrawal of the drug, particularly at lower dose levels. This syndrome is characterized by increased stiffness, joint pain, fever, irritability, and malaise with each reduction of dose. It can be minimized if reductions are 1 mg or less in the lower dosage ranges and are instituted not more often than every 1 to 2 weeks.
time thereafter, with careful attention to electrolyte and fluid balance, and to the potential for cardiac arrhythmia or acute hypertension.
Intra-articular Glucocorticoids The role of intra-articular administration of depot glucocorticoids is changing, but at the present time it is clearly indicated (1) in the management of oligoarthritis that has not responded to an appropriate program of NSAIDs or (2) as an aid to physical therapy of an inflamed, contracted joint.719–721 Intra-articular glucocorticoid therapy should be considered in the management of polyarticular disease in which one or several target joints have not responded to NSAIDs and anti-inflammatory drugs. However, intra-articular injections should be given only a limited number of times (e.g., three times in a single joint during 1 year). Triamcinolone hexacetonide, if available, has been the drug of choice at a dose of 20 to 40 mg for large joints. With the use of topical anesthesia and then local infiltration with buffered lidocaine, most children older than 7 years of age can cooperate with the procedure. Although EMLA cream (lidocaine 2.5%/prilocaine 2.5%) has been recommended, Uziel and colleagues722 did not find it better than placebo. Younger children, and those in whom a hip joint or several joints are being injected, require conscious sedation or light general anesthesia. As in any such procedure, aseptic technique must be followed. The efficacy and safety of intra-articular steroid administration has been studied by a number of authors.719–721,723–728 Almost all patients have a beneficial response; in 60% the response persists for at least 6 months, and in 45% there is no evidence of inflammation in the injected joint for at least 1 year.719,721 Side effects are uncommon, although two deserve special mention. The risk of subcutaneous atrophy and cutaneous depigmentation at the site of injection (Fig. 9–21) can be avoided or minimized by carefully preventing leakage of the depot preparation around the needle track. Intra-articular or periarticular calcification occurs in approximately 15% of treated patients, as evidenced
Steroid Pulse Therapy Intravenous pulse glucocorticoid therapy offers an alternative approach to serious, unresponsive disease. The effect is immediate, and it is hoped that long-term toxicity is decreased.717,718 Methylprednisolone is the drug of choice, in a dose of 10 to 30/mg/kg per pulse. Protocols consist of single pulses spaced 1 month apart, three pulses given sequentially on 3 days each month, or three pulses administered on alternate days each month. Pulse therapy is not without potentially serious toxicities; it should always be given in a short-stay clinic with cardiovascular monitoring during the infusion and for a
■ Figure 9–21 Subcutaneous atrophy (arrow), 10 months after intra-articular injection of triamcinolone acetonide tertiary-butylacetate.
C H A P T E R
on radiographs.721 Systemic side effects are rare, although a beneficial “overflow” effect may occur in uninjected joints, or there may be transient improvement in manifestations of the disease.729 Intra-articular septa, demonstrable by ultrasonography, may impede free distribution of injected glucocorticoid within the joint and prevent a complete therapeutic response. In a retrospective series by Sherry and colleagues,730 repeated intra-articular steroid injections were compared with traditional therapy as management for unilateral knee arthritis. Fewer leg-length and thigh-circumference discrepancies were associated with the steroids. In a study from Israel by Padeh and Passwell,731 the efficacy and safety of intra-articular triamcinolone hexacetonide was examined in 70 children who had received a total of 300 injections. Excellent benefit was achieved in 246 (82%) of 300 joints for longer than 6 months. Discontinuation of other medications was possible in 61% of these patients. A complete remission or correction of joint contracture occurred in 115 (82%) of 141 children with oligoarthritis. Eleven patients with popliteal cysts and 12 patients with tenosynovitis experienced satisfactory resolution of the abnormality. Three patients developed subcutaneous atrophy that eventually cleared. Two had a transient postinjection flare of joint pain and increased swelling. None of 20 patients examined by radiography had periarticular calcifications.
Biologic Response Modifiers Tumor Necrosis Factor Inhibition Recent therapeutic approaches for children with unremitting disease include soluble TNF-α receptor (TNFR) p75 fusion protein (etanercept)732–739 and recombinant monoclonal human IgG antibody to TNF-α (infliximab and, more recently, adalimumab).739,740–743 The safety of longterm use of these agents in children was recently reviewed.744 Etanercept has become standard therapy for arthritis that has not responded adequately to methotrexate. It is administered in a minimum dose of 0.4 mg/kg subcutaneously twice a week (maximum, 25 mg per injection), with continuation of previous medications, such as methotrexate and an NSAID.745,746 Before starting therapy, base-line laboratory determinations should be reviewed (complete blood cell count, comprehensive metabolic panel, ESR, CRP, urinalysis) and a negative skin reaction to the purified protein derivative (PPD) tuberculosis skin test must be documented. Thereafter, clinical and laboratory monitoring should be continued as with methotrexate therapy (if methotrexate is not continued, laboratory monitoring is subject to reevaluation). Singledose administration each week is an option for doses of 25 mg or less in newly diagnosed patients and in older children (4 to 17 years). If the weekly dose is greater than 25 mg, two subcutaneous injection sites should be used. Higher doses (0.8 to 1.8 mg/kg) have been recommended for an inadequate initial clinical response, but efficacy was limited in one study to 2 of 8 children.747 It may be possible to evaluate the blood level of pharmacologically active etanercept to guide dosage in patients with an incomplete response.748 The drug should not be started in any child with an infection or a history of recurrent infections. It has been emphasized that even subtle signs or symptoms of infection need to be investigated promptly, especially in
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
237
younger children (6 years of age or younger). Therapy must be discontinued in the presence of infection or in the perioperative period. The risk of reactivation of tuberculosis or the development of granulomatous or fungal disease must be recognized.749 The status of prior bacillus Calmette-Guérin (BCG) vaccination as a risk factor is undetermined, but, considering the lack of published data, it is apparently not a serious consideration in starting therapy. In an extensive clinical trial,734 etanercept in a dose of 0.4 mg/kg subcutaneously twice a week was given to 69 children with an active polyarticular course whose disease had been refractory or intolerant to methotrexate (40 polyarticular, 22 systemic, and 7 oligoarticular onsets). Ages ranged from 5 to 17 years, with a median disease duration of 5.5 years. At 3 months, 51 patients (74%) met standardized clinical response criteria (30% or greater improvement in 3 of 6 core set criteria with worsening of 30% or more in not more than 1 of 6 criteria and a minimum of 2 active joints). The responders were then randomized to continued administration of TNFR Fc or placebo (in addition to baseline NSAID or low-dose prednisone) for an additional 4 months. There was a clinically and statistically impressive improvement in 19 of 25 children who had been continued on etanercept, and a disease flare in 20 (89%) of 26 children in the placebo group (P = .007). The median time to disease flare in the children taking etanercept was at least 116 days, compared with 28 days in the placebo group. Each component of the core set criteria worsened in the placebo treatment arm and remained stable or improved in children continued on etanercept. The data suggested the possibility of an increased risk of a flare among those patients with a higher baseline ESR. Among patients who demonstrated a clinical response at 90 days, those who remained on etanercept continued to improve from month 3 through month 7, whereas those who received placebo did not. The drug was well tolerated with only mild to moderate upper respiratory infections or injection-site reactions. Long-term efficacy and safety in these studies has recently been summarized.750 The percentages of patients after randomization who maintained definition of improvement were 81% for JRA 30, 79% for JRA 50, and 67% for JRA 70. Studies have not been performed to assess the effects of continued etanercept therapy in patients who do not respond within 3 months after initiation of therapy or who develop a relapse while taking the drug. Its effectiveness if used without concomitant methotrexate is problematic. There are children who are only partially responsive or not responsive to TNF-α blockade, especially those with systemic-onset disease.747,751,752 A wide spectrum of toxicities has also been encountered on long-term administration.752–754 Demyelinating disease755 and pancytopenia were described in a drug warning from the company, and a number of other severe side effects have been reported.752,756
Other biologic agents that interfere with TNF-α (i.e., infliximab, adalimumab) are being evaluated and may prove useful in selected children.739,757–762 Infliximab has not been approved for use in children; a double-blind, randomized placebo-controlled trial in polyarticular disease was completed in 2004 by the PRCSG with doses of 3 and 6 mg/kg.763 One hundred and twenty children, aged 4–17 yrs, with active polyarticular JRA despite receiving methotrexate, were enrolled in a randomized, double-blind study with placebo control for 14 weeks followed by a treatment extension of 38 weeks.
238
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
Infliximab was administered in a dose of 3 mg/kg for a 3-dose induction at weeks 0, 2, and 6. The placebo group then received a 3-dose induction of infliximab of 6 mg/kg at weeks 14, 16, and 20, and then every 8 weeks through week 44. Infliximab, administered in combination with methotrexate, resulted in a trend towards improvement (ACR-JRA 30). It was well tolerated in the 6 mg/kg group. The 3 mg/kg group was associated with more infusion reactions (35% vs. 17.5%). Antibodies to infliximab were also higher in the lower-dose children (37.7% vs. 12.2%). Adverse events were comparable among groups except for infusion reactions; one death from sepsis occurred in the placebo group at 3 weeks.
The drug in adults is given concomitantly with weekly methotrexate, as intravenous infusions of 3 mg/kg at weeks 0, 2, and 6, and thereafter every 8 weeks for maintenance. Dosage and intervals of infusion may or may not differ in children compared with adults. Data on the occurrence of occult sepsis and the risk, if any, of previous BCG vaccination or of malignancy, have not been clarified. A strict stepwise infusion protocol to minimize reactions should be followed, with no infusion time shorter than 2 hours.764 Additional precautions are the same as for etanercept, including a negative skin reaction to PPD.765
receptor antagonist (IL-1Ra). Anti-IL-6 receptor monoclonal antibody is currently in clinical trials based on previous investigations.775 Thalidomide has been recommended for treatment of systemic onset arthritis.776
Intravenous Immunoglobulin IVIG for the treatment of polyarticular or systemic-onset disease, and other rheumatic diseases, has received considerable attention, but results of clinical trials are inconsistent.777,778 A controlled randomized trial of IVIG by the PRCSG in children with systemic-onset disease indicated that the drug had little benefit compared with placebo.779 In an additional trial of IVIG in children with polyarticular disease,780 1.5 to 2.0 g/kg was given twice a month for the first 2 months and then monthly for up to 6 months thereafter, with a maximum dose of 100 g. There were no adverse effects. The authors concluded that approximately 75% of children with polyarticular disease would benefit from IVIG.
Cytotoxic and Immunosuppressive Drugs
Lahdenne and cowokers739 compared therapy with etanercept (10 patients, 0.4 mg/kg subcutaneously twice weekly) and infliximab (14 patients, 3 to 4 mg/kg intravenously at weeks 0, 2, and 6, then every 4 to 8 weeks thereafter) in children with severe polyarthritis refractory to standard therapy, including 2 with psoriatic disease (mean age, 10.2 years; range, 3.3 to 16.3 years). During the trial, 23 patients were taking methotrexate. After 12 months, 89% of the patients taking etanercept achieved an ACR 50% response, compared with 75% of those taking infliximab (P > .05). One patient was withdrawn from etanercept because of lack of compliance, and 5 patients withdrew from infliximab because of adverse events or lack of efficacy. There were no serious infections encountered during this open-label, prospective trial.
Clinical experience with cytotoxic drugs is largely anecdotal or uncontrolled. Use of the immunosuppressive and cytotoxic drugs should be reserved for children with life-threatening complications, major steroid toxicity, or severe progressive erosive arthritis.781 A number of medications have been studied for this purpose, including the purine analogues 6-mercaptopurine and azathioprine and the alkylating agents cyclophosphamide and chlorambucil. Complications of chlorambucil therapy include sterility and an increased risk of malignancies. Successful therapy for amyloidosis in children with JRA usually required the use of chlorambucil.782–786 In one study at 15 years’ follow-up,787 68% of children with chronic arthritis complicated by amyloidosis who had been treated with chlorambucil (0.08 to 0.15 mg/kg/day) were alive; the mortality rate in the untreated group was 100%. Leflunomide, an inhibitor of pyrimidine synthesis, is being tested in children788 and has had encouraging results in adults with RA.789
Other Biologic Agents
Cyclosporine
A number of immune modulators have been tried therapeutically (see Chapter 5). Transfer factor, after an initial encouraging report, did not prove effective.766 Other immunoregulatory drugs, such as levamisole, were too toxic.767 Thymopentin, the active pentapeptide of thymopoietin, had no effect on joint disease, but fever was controlled and the size of lymph nodes, liver, and spleen was decreased.768 The ratio of CD4+ to CD8+ T cells returned toward normal, and there did not appear to be any toxicity. Recombinant IFN-γ has been used experimentally.769 In one study,770 it was given to children (6 systemic onset, 3 polyarthritis, 1 oligoarthritis) for whom other therapies had failed after a long duration of disease (5 to 11 years). Eight patients had significant improvement, and 7 entered remission. Two patients with systemic onset had no response, and 4 relapsed. One child developed a severe infection. Plasmapheresis has been recommended for severe disease771 and may be particularly indicated if high levels of circulating immune complexes are demonstrated. Lymphapheresis and total lymphoid radiation have not been adequately studied in controlled series of patients. Tolerance induction by oral administration of type II collagen has been considered as a therapeutic modality.772,773 Induction of tolerance to epitopes of HSP 60 as potential therapy for childhood arthritis is reviewed in Chapter 11. Anakinra is undergoing therapeutic trials for treatment of chronic arthritis in children.774 It is a recombinant human IL-1
A number of studies of cyclosporine for the treatment of chronic arthritis in children have been reported.790–793 It is given in an oral dose of 3 to 5 mg/kg/day in two divided doses exactly 12 hours apart. Blood pressure should be determined at home for 2 weeks and then evaluated periodically along with urinalyses and estimates of renal function. There may be a small long-term risk of lymphoma. Ostensen and coworkers791 administered the drug to 14 children in a dose of 4 to 15 mg/kg/day for 6 to 20 months. Three were able to undergo glucocorticoid dose reduction; 4 had acute exacerbations of disease while taking the drug; serum creatinine concentration increased in 11, hypertrichosis occurred in 14, and hypertension developed in 1 child. Anemia occurred in 9, and the drug was discontinued in 3. The role of this drug in the treatment of arthritis in children is uncertain, but it is probably critically important in treating the MAS (reactive hemophagocytosis).524,794–797 Combined therapy with methotrexate has been recommended in selected children.798
Other Modes of Advanced Therapy The slow-acting antirheumatic drugs (SAARDs), or diseasemodifying antirheumatic drugs, consist of the antimalarials, sulfasalazine, injectable and oral gold compounds, and
C H A P T E R D-penicillamine. Much of the data regarding therapeutic efficacy of these drugs has been inferred from uncontrolled studies.799–801 The fact that their beneficial effects are not observed for several weeks to months, coupled with the unpredictable, relapsing, remitting course of arthritis, makes evaluation of this group of medications difficult. When the toxicity of many of the members of this group is considered, enthusiasm for their use must be somewhat restrained. Until recently, however, these agents were the mainstay of second-line therapy in children with polyarticular disease in whom NSAIDs had produced no response. Methotrexate has generally replaced the SAARDs in this capacity.703 On the other hand, SAARDs can be considered for children who have an incomplete response to a combination of an NSAID, methotrexate, and a TNF-α blocker.
Hydroxychloroquine Hydroxychloroquine is a useful adjunctive agent for treatment of chronic arthritis in older children.799 The therapeutic effect of hydroxychloroquine is usually subtle and is rarely evident before 2 to 3 months of therapy. If no improvement is demonstrated after treatment for 6 months, it should be discontinued. Hydroxychloroquine is never used alone but is added to an NSAID regimen in a dose of 5 to 6 mg/kg/day. The medication is available only in 200-mg tablets, and often it is not possible to achieve an exact dose per day. An alternative schedule involves calculating the weekly dose and then distributing the correct number of tablets during the week. The drug should be taken with food, because it can be a GI irritant. An ophthalmologic examination, including testing of color vision and visual fields, is usually performed before therapy is started and traditionally every 6 months thereafter.802 With current doses, ocular toxicity is uncommon and recommendations regarding the frequency of reexaminations are being reconsidered.802–804 In general, the drug has not been recommended in children younger than 4 years of age, and sometimes in those younger than 7 years of age, because of the inability of young children to discern colors adequately for testing on grids or visual fields. Although retinal toxicity is very rare, hydroxychloroquine should be discontinued at the first suspicion of retinopathy, because the effects of this drug are cumulative. Corneal deposition of drug may also occur and is usually accepted as an indication for lowering the dose of the drug rather than discontinuing it. Chloroquine is less frequently used than hydroxychloroquine and probably has greater ocular toxicity. The therapeutic effect of hydroxychloroquine has not been conclusively proven in randomized, double-blind studies. In a number of therapeutic trials involving approximately 240 children,799 clinical improvement occurred in 15% to 75% and remission in 45% of patients. Toxicity occurred in up to 60% of the children and required discontinuation of the medication in 10%. An extensive double-blind trial by the PRCSG compared hydroxychloroquine with D-penicillamine or an NSAID.805 A total of 162 children with severe, poorly controlled arthritis were observed over 12 months; 88% of the children completed 6 months of the trial, and 76% completed 12 months. The authors reported that 60% of the hydroxychloroquine group (n = 43), 46% of those taking D-penicillamine (n = 46), and 39% of those taking placebo n = 34) demonstrated clinical improvement at 12 months of therapy.806 Although no unequivocal therapeutic advantages could be attributed to hydroxychloroquine or D-penicillamine compared with placebo, it was difficult to understand why the children given placebo did so well, because failure of a therapeutic trial with an NSAID was an entry criterion. However, improvement of pain on motion was noted in the hydroxychloroquine group more often than the placebo-treated group. Late benefit was unlikely to be documented if a favorable response had not occurred by 6 months.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
239
No subgroup of children could be identified who were likely to respond to either drug. Hydroxychloroquine may also be beneficial in amelioration of the dyslipoproteinemia that occurs in chronic disease and secondary to glucocorticoid treatment.807
Sulfasalazine Sulfasalazine has been reported to have modest efficacy in some children with chronic arthritis, and it has the advantage of a more rapid onset of anti-inflammatory action than occurs with the other SAARDs. This drug should not be used in children with known hypersensitivity to sulfa drugs or salicylate, impaired renal or hepatic function, or specific contraindications such as porphyria or glucose-6-phosphate dehydrogenase deficiency. Severe side effects (fever, rash, elevation of serum levels of liver enzymes, and the MAS) have been reported in children with systemic-onset disease. Sulfasalazine is started in a dose of 500 mg/day given with food or, for small children, 12.5 mg/kg. This dose is gradually increased until a dose of 50 mg/kg/day (maximum, 2000 mg) is reached. Enteric-coated sulfasalazine may be preferable to reduce dyspepsia. Monitoring for toxicity includes measurement of hemoglobin, differential leukocyte count, platelet count, immunoglobulins, and serum levels of liver enzymes every 4 weeks initially and then every 3 to 6 months (see Chapter 5). Benefit is usually apparent within 4 to 8 weeks after initiation of therapy.
Parenteral Gold Compounds Parenteral gold compounds (sodium aurothiomalate and aurothioglucose) had been indicated in children with polyarthritis if a program of NSAIDs had failed and the disease was extensive or progressing. Before gold therapy is started, it should be determined that the child’s hematologic, renal, and hepatic functions are normal (complete blood cell count, urinalysis, blood urea nitrogen and creatinine concentrations, and serum levels of liver enzymes). A 5-mg intramuscular test dose is given initially, and weekly doses thereafter are gradually increased to a level of approximately 0.75 mg to 1 mg/kg/week (maximum, 50 mg/week). If objective, satisfactory improvement or a remission is achieved in 6 months, therapy is maintained at the same dose with injections every 2 weeks for approximately 3 months, then every 3 weeks for 3 months. If signs of improvement continue during this interval, the gold injections are decreased to every 4 weeks thereafter, with periodic adjustments based on growth and body weight. Before each administration, the child should be assessed for any associated toxicity, such as stomatitis, dermatitis, pruritus, depression of any of the cellular elements of the blood, hematuria, or proteinuria. A decrease in the leukocyte count to fewer than 3500 cells/mm3 (3.5 × 109/L), a fall in the absolute neutrophil count of 50% or more, and development of thrombocytopenia or eosinophilia, hematuria, or clinical symptoms or signs of gold toxicity are indications for at least a temporary interruption of therapy. Therapy may be cautiously resumed at a lower dose after the symptoms or signs of toxicity disappear. Contraindications to reinstitution of gold therapy are severe leukopenia or neutropenia, proteinuria, exfoliative dermatitis, significant oral ulcerations, and consumptive coagulopathy. In the authors’ experience, the most common toxicities are hematuria and dermatitis. The oral gold compound auranofin, or triethylphosphine gold, has been evaluated in a number of studies,808–811 including a 6-month, double-blind, parallel, randomized, placebo-controlled trial in more than 200 children.811 Most investigations suggested, and this controlled study conclusively demonstrated, that the effect of auranofin was modest at best and was probably not significantly better than placebo. The safety of this oral
240
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
gold compound at a starting dose of 0.1 to 0.2 mg/kg/day (maximum, 9 mg/day) was documented in the initial study.812 Toxicity was relatively mild. D-Penicillamine D-Penicillamine has rarely been used in the treatment of children with chronic arthritis since recognition of the superior effectiveness of methotrexate and publication of the PRCSG study in which neither D-penicillamine nor hydroxychloroquine was superior to placebo.805 The dose is approximately 10 mg/kg/day (maximum, 750 mg/day). This level is achieved in three increments at intervals of 8 to 12 weeks each. The drug must be taken on an empty stomach to prevent heavy metals in food from chelating with it and rendering it ineffective. A complete blood cell count and urinalysis are performed once a week until the maximum dose is achieved, after which monitoring at monthly intervals is sufficient. Of particular concern is the possible induction of a lupus-like syndrome, dermatitis, thrombocytopenia, and proteinuria. D-Penicillamine toxicity is not necessarily dose-dependent and may be related to the presence of HLA-Dw33 (HLA-DR3)813 and the C4 null allele.814
Autologous Stem Cell Transplantation Bone marrow transplantation has been initiated as experimental treatment for severe autoimmune diseases including rheumatic diseases unresponsive to conventional therapy. Autologous stem cell transplantation (ASCT) is being evaluated currently in a small number of children.637,815–826 This approach to treatment is indicated only in children in whom control of severe active disease has not been achieved by conventional therapy, including anti-TNF therapy. In one clinical protocol,819 bone marrow harvest was followed by T-cell depletion and a conditioning regimen (antithymocyte globulin, cyclophosphamide, total body irradiation). ASCT was undertaken in children with polyarticular and systemic-onset arthritis (1 and 4 patients, respectively) who had progressive, unremitting disease. At final evaluation, 3 patients were in drugfree remission and 1 had mild oligoarthritis. Deaths from posttransplant viral infections and the MAS have occurred in more than 30 children. Although follow-up has been short and patient numbers limited, the authors of this report judged the results to be encouraging. The results of ASCT in 31 children with refractory systemic JIA treated in eight European centers were summarized by Wulffraat and colleagues.827 Eighteen patients were in complete remission and taking no medication 12 to 60 months after transplantation. A partial response was observed in 6 patients (18%) and no improvement in 7 (21%). Three patients died from transplantation-related causes and 2 others from their disease. Survival and post-transplantation morbidity are likely to be better with improved techniques for harvesting and manipulating stem cells, prevention of viral infections, genetic tests to identify the potential for relapses, tailoring of medications specifically for each recipient, and experience with preventing complications such as the MAS. Serum concentrations of myeloid-related proteins 8 and 14 are markers of disease activity in children who have undergone ASCT828 and may predict flares in JIA.829
Growth Hormone The best approach to the treatment of growth failure and short stature is uncertain.830,831 Most, but not all, children with
chronic arthritis have normal baseline and stimulated blood levels of growth hormone, IGF-I, and IGF-II. Growth hormone has been used with varying success, although early studies may not have used sufficient doses. Davies and colleagues449 treated 28 children who were growth impaired with one of two regimens of growth hormone (12 or 24 IU/m2/week). All patients demonstrated adequate baseline secretion of growth hormone. Although almost all children had an increase in height during 1 year of treatment, those receiving the higher dose of growth hormone grew better. Investigations indicate that daily growth hormone in a weekly dose of 0.3 mg/kg initiates a return to normal growth rates for 1 to 2 years.832 However, glucocorticoid in a prednisone-equivalent dose of greater than 0.35 mg/kg/day interferes with growth hormone responsiveness.832,833 In the study by Touati and associates,834 recombinant growth hormone in a dose of 1.4 IU/kg/week partially counteracted the adverse effects of glucocorticoids on linear growth velocity and lean body mass. In children with rheumatic diseases, the immunostimulatory effects of growth hormone lead to the theoretical possibility of interference with immunosuppression.835 In one study,836,837 growth hormone slightly increased allograft rejection rates. Growth hormone may also increase cytochrome P-450 activity838 and thereby alter clearance of specific drugs (glucocorticoids, sex steroids, anticonvulsants, cyclosporine).
Nutrition The child’s overall nutrition, development, and growth are important aspects of long-term management. Growth retardation and impaired bone mineralization almost invariably occur during periods of active disease and are exacerbated by glucocorticoid administration, anorexia, or inanition. Nutrition and vitamin supplementation (vitamin D and folic acid) are often indicated. Assessment of nutritional status should be a component of every patient’s evaluation.839,840 Management of malnutrition is often difficult in the systemically ill, anorectic child. Nocturnal nasogastric feeding occasionally is necessary to maintain adequate nutrition in such a child.454 Specific measures to address osteopenia and osteoporosis are reviewed in Chapter 38. Marked microcytic, hypochromic anemia is usually not improved by oral iron supplementation which often adversely affects appetite. Occasionally, a child benefits from intravenous iron administration followed by recombinant erythropoetin.520,841 Henderson and Lovell842 suggested the use of a four-parameter test to screen for protein–energy malnutrition. The presence of any two of the following abnormalities indicates the need for a detailed nutritional assessment: weight below 5th percentile, weight-for-height index below 80th percentile, arm circumference below 5th percentile, and serum albumin less than 2.8 mg/dL (28 g/L).
Physical and Occupational Therapy The objectives of physical and occupational therapy are to minimize pain, maintain and restore function, and prevent deformity and disability (Table 9–11). These aspects of treatment are critically important in the child’s total management program.843 In children with active arthritis, a rest period after returning from school in the afternoon and increased time for sleep at night are advisable. Most children, however, determine their own levels of activity.
C H A P T E R
TABLE 9–11 Treatment of Chronic Arthritis: Physical Measures Relief of pain Heat/cold Maintenance of joint function Splints Range-of-motion exercises (active/passive) Posture/prone-lying exercises Adequate rest/adequate exercise Balanced physical activity Selected peer sports (tricycle, swimming)
Only rarely is it warranted to push them beyond their present capabilities or to unduly restrict daily activities. Normal play must be encouraged, and the family must realize how vital this is to the growing child in terms of peer group interactions and physical fitness, including achieving and maintaining a desirable level of exercise and musculoskeletal and cardiopulmonary conditioning.844,845 It is important, however, to avoid specific activities that cause overtiring or increased joint pain. Undesirable play is that which places excessive stress on affected joints. Tricycle or bicycle riding and swimming are almost always helpful and do not add undue loading to the joints. Swimming has the added advantage of providing active ROM and muscle strengthening without weight bearing. Although activities such as bicycling and swimming are excellent ways of maintaining muscle strength and ROM, they are not a substitute for a well-designed active and passive therapy program: The child with limited ROM tends to function within the current range rather than regaining lost ROM. Skilled physical and occupational therapists are able to assess the time and intensity of a therapy program that the child can tolerate. Daily physical therapy performed at home should seldom exceed 30 minutes twice a day (and often should be much shorter). The child’s age and temperament, the duration of morning stiffness, and the presence of systemic disease must all be considered in planning a physical therapy program. If possible, adaptive play and games are designed to achieve the desired results.845 The child and parents should agree on realistic goals. It is sometimes difficult for the parent to carry out home therapy and maintain a healthy parent–child relationship. In such circumstances, twice-weekly visits to a physical therapist may be a better plan than daily home therapy by the parent. In some children, it is necessary to institute intensive physical and occupational therapy for 1 to 3 weeks once or twice a year to prevent loss of function. During periods of active inflammation, joint pain and stiffness are reduced by heat. A hot bath in the morning and application of heat before physical therapy reduce stiffness and pain and maximize function. In children with a great deal of muscle spasm, ice may be more beneficial than heat, but this often is not preferred. Passive stretching is usually needed to regain lost ROM. Active exercise is required to rebuild muscle strength. Atrophy of the extensor muscles begins early, and active exercise must be instituted during the initial phases of the disease to maintain the strength of these muscle groups. Cock-up splints for the wrists, ring splints for the fingers, and Orthoplast posterior resting splints for the knees may be used to prevent malpositioning or for the long-term correction
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
241
of deformity. The maintenance of normal ROM or reduction of minimal contractures often requires gentle stretching and the use of resting splints. A cervical collar with a plastic insert should be worn by children with cervical spine disease during automobile travel. Malpositioning and neck discomfort may be minimized by the use of a soft collar while studying and a desk with a tilt top on which the child can do homework. At night, a small pillow under the neck, rather than under the head, is an effective way to reduce neck discomfort. Dynamic splints are helpful for certain joint contractures (elbow and knee). For severe contractures—especially contractures of the wrist, elbow, or knee—serial casting with active exercises every 2 days when the cast is removed may be more effective. If stretching or casting is too forceful or is improperly performed, subluxation of the knee or wrist or even fracture of osteoporotic bone can occur. Appropriate footwear is essential in the child with arthritis of the joints of the ankle and foot.612 Shoes should be lightweight but provide firm lateral support. In children with a leg-length discrepancy, a heel lift or an addition to the bottom of the shoe of the shorter leg should be used, depending on the depth needed. In general, the total compensation should be somewhat less than the leg-length discrepancy. One study has analyzed gait alterations in children with chronic arthritis.846 In children with pain or inflammation of the metatarsophalangeal joints or excessive pronation or supination, a custom-fitted hard insole that slips into the shoe provides considerable comfort and improvement of the gait. Hip flexion contractures can be difficult to treat. Prone lying for a significant period (at least 1 hour/day) helps to reduce the degree of contracture. Several studies416,847,848 have examined psychologic approaches to treatment for children with higher levels of associated pain. The treatment protocol included relaxation training, guided imagery, and biofeedback for the child and training in the use of behavioral techniques for management of daily physical therapy and school attendance for the mothers. The results provided modest support for the use of psychologic interventions in children whose pain significantly affects their activities of daily living.
Orthopedic Surgery At present, orthopedic surgery has a limited but important role in management of chronic arthritis in young children.849–852 In the older child, however, surgical approaches to joint contractures, dislocations, or joint replacement become important components of therapy, and the orthopedic surgeon plays an important role at this stage.852–854 Approaches to some problems, such as limb overgrowth, have not been completely clarified.855 In some reports, surgical epiphyseal arrest was judged to be necessary in approximately one third of the patients.856,857
Synovectomy The long-term outcome of children with joint disease is not altered by prophylactic synovectomy. However, synovectomy may be useful in some children for relief of mechanical impairment of joint motion related to joint pain or synovial hypertrophy.850,858–862 Care must be taken to prevent a postsurgical contracture, and the use of a continuous-motion apparatus has been recommended for this purpose, especially in the young child. Arthroscopic surgery greatly reduces the morbidity associated with synovectomy.863
242
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
Soft Tissue Surgery Soft tissue releases, posterior capsulotomy, and tendon lengthening occasionally are useful in a child with a severe contracture of the knee or hip. In other instances, balanced traction is necessary to expedite the treatment of a knee contracture, although this is often difficult to maintain in the young child. Tenosynovectomy may be indicated to reduce the risk of tendon rupture over the dorsum of the wrist or for adhesive flexor tenosynovitis and trigger finger, which sometimes occurs in children who are RF seropositive.
Reconstructive Surgery Reconstructive surgery has become important in the older patient with marked disability. Total joint prostheses, particularly for the hip or knee, have proved to be of great benefit.627,864 Usually, surgery is postponed until bone growth has ceased. Other special considerations include the status of the other lower-extremity joints, activity of the rheumatic disease in general, currently unresolved questions concerning wear and longevity of prostheses, and possible lack of adequate motivation for rehabilitation in some patients. Many children with polyarthritis are further incapacitated either before or after lower-extremity surgery because of difficulty with crutch walking due to increased use of the hands or wrists, flexion contractures of the elbows, or involvement of the shoulder joints. Preservation of muscle function in anticipation of surgical intervention requires a rehabilitation program over many years devoted to maintaining muscle strength as near normal as possible.
Counseling the Family It is of signal importance that the child and the parents be educated about the present state of knowledge of JRA, its outcome, and therapy. The family must share in the coordination of disease management, which should be community based.865,866 An optimistic attitude must be maintained. Counseling should be initiated by the physician at the time of the first visit and reinforced and continued at follow-up by the team. Educational efforts are repeated as needed during the subsequent clinical course, especially in an effort to increase compliance.867,868 In management, the first priority, concomitant with initial efforts to control the activity of the disease, is to foster normal psychologic and social development. Attendance at school is an integral part of this program. Home instruction is rarely indicated; most pediatricians sense that physically disabled students do better in regular schools. Some families have had psychologically important disruptions such as divorce, separation, death, or adoption, and they often display severe emotional disturbances. More recent studies from North America indicated that families of children with chronic illnesses are much more functional.869,870 It is important to emphasize that coordinated care of the child with a chronic disease requires a team effort.71 This concept presents problems for newly established clinical services because of the number of personnel who need to be involved in an ideal program in addition to the child, the family, the school, and the community agencies. One study found that, for most children with chronic arthritis, basic care was either divided or dupli-
cated, and many aspects of a total supportive program were neglected.871 These authors and others found that a pattern of division, duplication, and neglect was typical of the fragmented care of most children with a chronic disease.
COURSE OF THE DISEASE AND PROGNOSIS The course of chronic arthritis is especially unpredictable early in the disease. Nonetheless, certain generalizations can be made, because the type of onset is associated to some extent with the future unfolding of the disease and its manifestations (Table 9–12).10,872 After the pattern of the disease is well established, the course tends to be more predictable and repetitive. After this initial period of observation of variable duration, it is usually possible to begin to estimate prognosis and therapeutic response on which changes in the management program can be based. It is impossible to predict the eventual disease outcome in any individual child. Furthermore, “outcome” is a complex concept that can be measured in a number of ways873,874 (see Chapter 7).
Functional Disability and Psychosocial Outcome It had been estimated historically that 70% to 90% of children with chronic arthritis have a satisfactory outcome without serious disability.875–877 A small percentage (perhaps 5%) develop a recurrence of arthritis as adults878–880 Approximately 10% to 20% of children enter adulthood with moderate-to-severe functional disabilities.2,671,881 Delay in referral and initiation of an acceptable therapeutic program are associated with a poorer functional outcome. Recent data have been presented on outcome for specific onset types872,882–884 and for long-term follow-up from specific referral centers.844,877,885–893 The data of Wallace and Levinson671 documented poorer long-term results than previously recorded (Table 9–13). At 15 to 20 years, 17% of the patients were in Steinbrocher functional capacity III to IV, and 45% still had active disease (activity of disease was approximately equal for each onset type). A review of previous studies of outcome has been published.894 A study by Ruperto and colleagues636 evaluated long-term outcome in a group of 227 patients from Cincinnati and Pavia. This study examined the effect of specific demographic, clinical, and immunologic variables that were present during the first 6 months of the illness. The mean duration of disease at assessment was 15 years (range, 15.3 to 36.1 years). The best predictor of long-term disability was the initial articular severity score. Early hand involvement was also a strong predictor of future disability, pain, and impaired wellbeing. ANA seropositivity was associated with less disability. This study confirmed that quality-of-life scores are much more difficult to forecast because of the multiple domains that are functioning in this type of outcome. Extensions of this study635 indicated that long-term outcome, based on quality-of-life scales and health assessment questionnaires, was favorable in most patients 5 years or more after onset of symptoms.895,896 The Childhood Health Assessment Questionaire (CHAQ) has been evaluated for measurement of health status in early and late disease.897 Oen and colleagues898 surveyed early predictors of long-term outcome on the CHAQ in 392 patients with JRA
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
243
TABLE 9–12 Clinical Outcome by Onset Type and Course Subtypes Onset Type (N)
Course Subtype (n)
Profile
Outcome
Polyarthritis (78)
RF seropositive (16)
Female Older age Hand-wrist involvement Erosions Nodules Unremitting Female Young age Variable
Poor
ANA seropositive (38) Seronegative (24) Oligoarthritis (121)
ANA seropositive (66)
Good
Seronegative (35)
Female Young age Chronic uveitis Polyarthritis Erosions Unremitting Male Older age Variable
Excellent
Oligoarthritis (30) Polyarthritis (21)
Variable Erosions
Good Poor
RF seropositive (8) HLA-B27 positive (12)
Systemic disease (51)
Good
Excellent (except eyes) Poor Good
ANA, antinuclear antibodies; RF, rheumatoid factors. Cassidy JT, Levinson JE, Bass JC, et al: A study of classification criteria for a diagnosis of juvenile rheumatoid arthritis. Arthritis Rheum 29: 274–278, 1986.
(of 652 eligible), including 327 white patients, who were 8 years of age or older and had a minimum of 5 years of followup. The most significant early predictors were age at onset and being male. In addition to the results in Table 9–14, worse disability was observed for systemic onset in males, less disability in patients with RF-negative disease, and a shorter duration of activity in RF-positive polyarthritis. ANA seropositivity in oligoarthritis was associated with a longer duration of activity, as was a younger age at onset. The RA shared epitope was associated with less disability in systemic disease. A subsequent report examined radiologic outcome in children from the same clinics.899 More recently, investigators have attempted to avoid the confounding factors of referral-based and clinic follow-up by
TABLE 9–13 Functional Outcome of Chronic Arthritis
Author and year (ref. no.) Laaksonen, 1966 (53) Jeremy et al., 1968 (939) Ansell & Wood, 1976 (940) Hill et al., 1976 (879) Hanson et al., 1977 (941) Stoeber, 1981 (942) Wallace & Levinson, 1991 (671) Zak & Pedersen, 2000 (881) Bowyer et al., 2003 (895) Oligoarthritis Polyarthritis Systemic disease
Years of follow-up (mean) ≥16 5–20 (18) ≥15 (14.5) 5–25 (10) 10–22 (15) 15–20 26 5
Classes III and IV (% of cases) 48 24 23 33 28 41 17 11 0 12 30
Modified from Wallace CA, Levinson JE: Juvenile rheumatoid arthritis: outcome and treatment for the 1990s. Rheum Dis Clin North Am 17: 891–905, 1991.
primarily basing their outcome studies on population surveys. Data from the U.S. Pediatric Rheumatology Disease Registry895 in 703 patients observed between 1992 and 1997, before the era of biologic therapy, indicated that more than 25% of those with polyarthritis, and almost one half of those with systemic onset, had functional limitations affecting school activities. Joint space damage on radiographs was evident at 5 years in two thirds of the polyarticular and systemic-onset groups. Some investigators have also taken the approach recommended by the World Health Organization in 1980 to examine functional adaptation in relation to impairment (based on organ disease), disability (related to personal quality of life),900 and handicap (related to society’s perception of the functioning ability of the individual).901 A study by Peterson and colleagues902 from the Mayo Clinic evaluated the physical and psychosocial impacts of disease in a population-based cohort of 44 adults who had experienced onset of the disease during childhood. Controls (102)
TABLE 9–14 Functional Outcome of Juvenile Rheumatoid Arthritis by the Childhood Health Assessment Questionnaire (CHAQ)
Type of Onset (n) Systemic (40) Pauciarticular (224) Polyarticular (RF−) (80) Polyarticular (RF+) (40)
CHAQ score
Follow-up (mean yr)
Mean
Range
11.6 12.5 12.6 13.9
0.25 0 0.19 0.62
0–2.75 0–2.13 0–2.75 0–3.0
RF, rheumatoid factor. Oen K, Malleson PN, Cabral DA, et al.: Early predictors of longterm outcome in patients with juvenile rheumatoid arthritis: subset-specific correlations. J Rheumatol 30: 585–593, 2003.
244
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
were age- and sex-matched. Average follow-up was 24.7 years, and mean age was 34 years. The patients had greater disability, more bodily pain, increased fatigue, poorer health perception, and decreased physical functioning along with lower rates of employment and lower levels of exercise compared with the control group. On the other hand, educational achievement, annual income, health insurance status, and rates of pregnancy and childbirth were similar to those of the controls. Active disease was present in 66%, and 16% were under regular medical care for their arthritis. This study concluded that adults who had had chronic arthritis developed long-term physical and psychosocial impairments that have been often ignored in the evaluation of functional outcome. Many of the same outcomes and prognostic factors were identified in a study by Flato and colleagues of 268 patients with JRA who were monitored for a median of 14.9 years (range, 11.7 to 25.1 years).903 Another study from Norway904 included 53 patients with arthritis and 19 with juvenile spondyloarthropathy. Forty-three (60%) of the 72 patients were in a remission at the time of the study, and 60% reported no disability. This study and subsequent reports903,905 supported the conclusion that long-term outcome was more favorable than previously reported, perhaps related to less bias of admission to the study and the early use of more aggressive therapeutic regimens. This group of investigators had previously indicated that poor psychosocial functioning in 22% of the patients on follow-up was associated with premorbid psychosocial dysfunction, chronic family difficulties, and major life events.905 They also confirmed that approximately 19% of the patients were still suffering from chronic pain without evidence of active arthritis.904 It is also the impression of the authors, and of others,943 that a chronic pain syndrome may persist in these patients well after their arthritis has remitted. A number of investigations of adaptation to chronic illnesses in children have been completed recently.869,870,906–909 Studies of adaptation have been limited by assumptions that disease groups of chronic illnesses are homogeneous and that comprehensive adaptation models involve both risk factors and resistance factors within the child and family. In one study, 107 children with JRA,869 114 with insulin-dependent diabetes mellitus, and 88 healthy controls were evaluated every 6 months for 2 years. Differences were observed between mothers and fathers with regard to dependent variables of depression, anxiety, somatization, strain at work, days of work loss due to child’s illness, and extent to which the illness interfered with leisure time in the family. Married mothers missed more work than fathers but demonstrated less overall functional impairment. Single mothers did not differ from married mothers or fathers. The dependent variables—particularly those related to depression, anxiety, and strain at work—decreased for all parents during the 2 years of observation. Premorbid diagnostic groups were associated with maternal depression and paternal distress in passive coping. Parental depression and distress were associated with the child’s behavior in the illness, complaints of pain, and ability to deal with anger and disappointment. Anger level and anger expression styles were highly associated with depression.910 Children who turned their anger inward were most likely to report depression. The child’s functioning did not appear to be related to the clinical diagnostic group. Trajectories within the child’s and family’s adaptations could be identified independent of the diagnostic groups.870 This and other studies911 have concluded that interventions to ameliorate parental distress certainly have beneficial effects on child behavior and on parental reactions to that behavior and the chronic illness.431,912–915 The unreimbursed cost to the family of a child with a chronic rheumatic disease per year is considerable.916 Transition of the child to young-adult identification, vocational planning, and eventually a place in adult life has been
the subject of a number of studies. Excellent summaries are contained in publications by Spencer and colleagues,917 Chamberlain and Rooney,918 and White.919–921
Death In early studies of chronic arthritis in children, the overall death rate was 1% to 4%.922,923 The disease-associated death rate is now perhaps less than 1% in Europe and less than 0.3% in North America. This mortality represents, however, a 4- to 14-fold increase compared with standardized rates. In a study from England,924 the standardized mortality ratio was 3.4 for males (95% CI, 2.0 to 5.5) and 5.1 for females (95% CI, 3.2 to 7.8). The majority of the deaths in Europe were previously related to the development of amyloidosis 925; in the United States, deaths occurred predominantly in children with systemiconset disease and in many cases were related to infection. Infections were associated in the early studies with glucocorticoid therapy and are currently less often fatal. The major causes of death in 46 children observed by Bywaters at Taplow926,927 were renal failure (which in half of the children was associated with amyloidosis) and infections. At the 15-year evaluation, excess mortality was identified in girls who developed arthritis early in life and the frequency of amyloidosis had risen to 7.4%. It was rarely observed as early as 1 year after onset but could develop as late as 23 years, most commonly in children with systemic disease. Spontaneous remissions occurred. Even in Europe, amyloidosis as a complication of chronic arthritis appears to be on the decline, with a decreasing number of deaths784,928,929; however, a recent series from Turkey reported a frequency of 10%.930 Amyloidosis refers to the tissue deposition of the fibrillar protein amyloid.931 Secondary amyloidosis occurs as a complication of chronic inflammatory conditions such as arthritis or infection and in diseases such as familial Mediterranean fever. It is characterized clinically by proteinuria and the nephrotic syndrome, diarrhea, hepatosplenomegaly, or unexplained anemia in a child with profound hypergammaglobulinemia. Amyloidosis may be preceded by marked elevations of CRP, an acute phase reactant that is similar in structure to serum amyloid A (SAA) protein.932 SAA protein responds as an acute phase reactant and is increased in concentration in children with active disease. Although no HLA associations have been confirmed in children who develop amyloidosis,933 a genetic marker for the amyloid P component was identified in one study by restriction fragment length polymorphism.934 Secondary amyloidosis as a complication of chronic arthritis is exceedingly rare in North America but occurs in approximately 5 to 7% of children in certain areas of the world, particularly England, Scandinavia, Poland, and Germany.935 Diagnosis is confirmed by examination of tissue sections stained by Congo red dye. Under the polarizing microscope, amyloid deposits assume a green color that is virtually pathognomonic.936 Rectal submucosa is the most frequently recommended biopsy site; renal biopsy may be hazardous because of an increased tendency toward bleeding. Radionuclide imaging using radioiodinated autologous serum amyloid P is a noninvasive technique for diagnosis and monitoring.937,938
PERSPECTIVE In spite of new insights into causation and considerable advances in treatment, chronic arthritis remains an important cause of chronic pain and disability in child-
C H A P T E R
hood. Particularly when considering heroic pharmacologic therapy, one should remember that the disease is often self-limited, albeit after years of activity, and is seldom fatal. On the other hand, undue delay in instituting advanced treatment that may be effective can result in irretrievable damage to joints and other visceral organs and impaired skeletal maturation. The art of medicine is at least as important as its science in guiding the pediatric rheumatologist’s planning of a therapeutic approach. An important factor working in favor of the program of management is the child’s unceasing potential for growth. To a large extent, it is this intrinsic endowment for future physical and psychologic development that enables so much to be accomplished in most children. Even so, much work remains in clarifying the nature and management of these diseases and their complications.
9 C H R O N I C A RT H R I T I S 24. 25.
26. 27. 28. 29. 30. 31. 32.
R E F E R E N C E S
33. 34.
1. Petty RE: Frequency of uncommon diseases: is juvenile idiopathic arthritis underrecognized? J Rheumatol 29: 1356–1357, 2002. 2. Cuesta IA, Kerr K, Simpson P, et al: Subspecialty referrals for pauciarticular juvenile rheumatoid arthritis. Arch Pediatr Adolesc Med 154: 122–125, 2000. 3. Graham TB, Glass DN: Juvenile rheumatoid arthritis: ethnic differences in diagnostic types. J Rheumatol 24: 1677–1679, 1997. 4. Cassidy JT: What’s in a name? Nomenclature of juvenile arthritis. A North American view. J Rheumatol Suppl 40: 4–8, 1993. 5. Schneider R, Passo MH: Juvenile rheumatoid arthritis. Rheum Dis Clin North Am 28: 503–530, 2002. 6. Brewer EJ, Bass JC, Cassidy JT: Criteria for the classification of juvenile rheumatoid arthritis. Bull Rheum Dis 23: 712–719, 1972. 7. European League Against Rheumatism: EULAR Bulletin No. 4: Nomenclature and Classification of Arthritis in Children. Basel, National Zeitung AG, 1977. 8. Fink CW: Proposal for the development of classification criteria for idiopathic arthritides of childhood. J Rheumatol 22: 1566–1569, 1995. 9. Brewer EJ Jr, Bass J, Baum J, et al: Current proposed revision of JRA Criteria. JRA Criteria Subcommittee of the Diagnostic and Therapeutic Criteria Committee of the American Rheumatism Section of The Arthritis Foundation. Arthritis Rheum 20: 195–199, 1977. 10. Cassidy JT, Levinson JE, Bass JC, et al: A study of classification criteria for a diagnosis of juvenile rheumatoid arthritis. Arthritis Rheum 29: 274–281, 1986. 11. Cassidy JT, Levinson JE, Brewer EJ Jr: The development of classification criteria for children with juvenile rheumatoid arthritis. Bull Rheum Dis 38: 1–7, 1989. 12. Petty RE, Southwood TR, Baum J, et al: Revision of the proposed classification criteria for juvenile idiopathic arthritis: Durban, 1997. J Rheumatol 25: 1991–1994, 1998. 13. Petty RE: Growing pains: the ILAR classification of juvenile idiopathic arthritis. J Rheumatol 28: 927–928, 2001. 14. Petty RE, Southwood TR, Manners P, et al: International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: second revision, Edmonton, 2001. J Rheumatol 31: 390–392, 2004. 15. Fantini F: Classification of chronic arthritides of childhood (juvenile idiopathic arthritis): criticisms and suggestions to improve the efficacy of the Santiago-Durban criteria. J Rheumatol 28: 456–459, 2001. 16. Ramsey SE, Bolaria RK, Cabral DA, et al: Comparison of criteria for the classification of childhood arthritis. J Rheumatol 27: 1283–1286, 2000. 17. Foeldvari I, Bidde M: Validation of the proposed ILAR classification criteria for juvenile idiopathic arthritis. International League of Associations for Rheumatology. J Rheumatol 27: 1069–1072, 2000. 18. Berntson L, Fasth A, Andersson-Gare B, et al: Construct validity of ILAR and EULAR criteria in juvenile idiopathic arthritis: a population based incidence study from the Nordic countries. International League of Associations for Rheumatology. European League Against Rheumatism. J Rheumatol 28: 2737–2743, 2001. 19. Merino R, De Inocencio J, Garcia-Consuegra J: Evaluation of ILAR classification criteria for juvenile idiopathic arthritis in Spanish children. J Rheumatol 28: 2731–2736, 2001. 20. Krumrey-Langkammerer M, Hafner R: Evaluation of the ILAR criteria for juvenile idiopathic arthritis. J Rheumatol 28: 2544–2547, 2001. 21. Hofer MF, Mouy R, Prieur AM: Juvenile idiopathic arthritides evaluated prospectively in a single center according to the Durban criteria. J Rheumatol 28: 1083–1090, 2001. 22. Berntson L, Fasth A, Andersson-Gare B, et al: The influence of heredity for psoriasis on the ILAR classification of juvenile idiopathic arthritis. J Rheumatol 29: 2454–2458, 2002. 23. Flato B, Smerdel A, Johnston V, et al: The influence of patient characteristics, disease variables, and HLA alleles on the development of radiographi-
35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61.
IN
CHILDHOOD
245
cally evident sacroiliitis in juvenile idiopathic arthritis. Arthritis Rheum 46: 986–994, 2002. Martini A: Are the number of joints involved or the presence of psoriasis still useful tools to identify homogeneous disease entities in juvenile idiopathic arthritis? J Rheumatol 30: 1900–1903, 2003. Tsitsami E, Bozzola E, Magni-Manzoni S, et al: Positive family history of psoriasis does not affect the clinical expression and course of juvenile idiopathic arthritis patients with oligoarthritis. Arthritis Rheum 49: 488–493, 2003. Manners P, Lesslie J, Speldewinde D, et al: Classification of juvenile idiopathic arthritis: should family history be included in the criteria? J Rheumatol 30: 1857–1863, 2003. Southwood TR, Woo P: Childhood arthritis: the name game. Br J Rheumatol 32: 421–423, 1993. Holt PJ: The classification of juvenile chronic arthritis. Clin Exp Rheumatol 8: 331–333, 1990. Huppertz HI: Viewpoint: time for change in pediatric rheumatology. Rheumatol Int 13: 37–38, 1993. Cornil MV: Memoire sur les coincidences pathologiques du rhumatisme articulaire chronique. C R Mem Soc Biol (Paris) 4: 3, 1864. Diamant-Berger MS: Du Rhumatisme Noueux (Polyarthrite Deformante) Chez Les Enfants. Paris, Lecrosnier et Babe, 1891. Still GF: On a form of chronic joint disease in children. Med Chirurg Trans 80: 47, 1897. Keen JH: George Frederic Still—Registrar, Great Ormond Street Children’s Hospital. Br J Rheumatol 37: 1247, 1998. Hirschsprung H: Multipel, kronisk, infektios Ledsygdom I Barnealderen. Hospitalstid 421, 1901. Atkinson FRB: Still’s disease. Br J Child Dis 36: 100, 1939. Edstrom G: Rheumatoid arthritis in children. Acta Paediatr Scand 34: 334, 1947. Bille BSV: Kronisk polyartrit hos barn och dess guldbehandling. Nord Med 37: 307, 1948. Colver T: The prognosis in rheumatoid arthritis in childhood. Arch Dis Child 12: 253, 1937. Holzmuller G: Beitrag zur Frage der Pathogenese und Therapie der primarchronischen Gelenkerkrankungen im Kindesalter. Ztschr F Rheumaforsch 5: 57, 1942. Coss JA, Boots RH: Juvenile rheumatoid arthritis. J Pediatr 29: 143, 1946. Pickard NS: Rheumatoid arthritis in children. A clinical study. Arch Intern Med 80: 771, 1947. Lockie LM, Norcross BM: Juvenile rheumatoid arthritis. Pediatrics 2: 694, 1948. Wissler H: Der Rheumatismus im Kindesalter. Teil 2: Die chronische Polyartritis des Kindes. Der Rheumatismus Bd 24 Steinkopff, Dresden und Leipzig 152, 1942. Françon F: Conferences Cliniques De Rhumatologie Pratique. Paris, 1946, p 386. Dawson MH: Rheumatoid arthritis in children (Still’s disease). In Palmer WW (ed): Nelson Loose-Leaf Medicine. New York, T Nelson & Sons, 1946, p 626. Stoeber E: Juvenile chronic polyarthritis and Still’s syndrome. Doc Geigy 1: 1977. Spitzy H: Ueber das Vorkommen multipier chronischer deformierende Glenkentzundungen im Kindesalter. Jahrb Kinderheilk 49: 286, 1899. Chevallier P: La maladie de Chaufffard-Still et les syndromes voisins. Rev Med Paris 47: 77, 1930. Sundt H: Om arthritis deformans i Barnealderen. Med Rev 38: 145, 1921. Portis RB: Pathology of chronic arthritis of children (Still’s disease). Am J Dis Child 55: 1000, 1938. Sury B: Rheumatoid Arthritis in Children. A Clinical Study. Copenhagen, Munksgaard, 1952. Manners PJ, Bower C: Worldwide prevalence of juvenile arthritis-why does it vary so much? J Rheumatol 29: 1520–1530, 2002. Laaksonen AL: A prognostic study of juvenile rheumatoid arthritis. Analysis of 544 cases. Acta Paediatr Scand 1–163, 1966. Oen KG, Cheang M: Epidemiology of chronic arthritis in childhood. Semin Arthritis Rheum 26: 575–591, 1996. Oen K: Comparative epidemiology of the rheumatic diseases in children. Curr Opin Rheumatol 12: 410–414, 2000. Andersson GB: Juvenile arthritis—who gets it, where and when? A review of current data on incidence and prevalence. Clin Exp Rheumatol 17: 367–374, 1999. Nielsen HE, Dorup J, Herlin T, et al: Epidemiology of juvenile chronic arthritis: risk dependent on sibship, parental income, and housing. J Rheumatol 26: 1600–1605, 1999. Lindsley CB: Seasonal variation in systemic onset juvenile rheumatoid arthritis. Arthritis Rheum 30: 838–839, 1987. Oen K, Fast M, Postl B: Epidemiology of juvenile rheumatoid arthritis in Manitoba, Canada, 1975–1992: cycles in incidence. J Rheumatol 22: 745–750, 1995. Feldman BM, Birdi N, Boone JE, et al: Seasonal onset of systemic-onset juvenile rheumatoid arthritis. J Pediatr 129: 513–518, 1996. Uziel Y, Pomeranz A, Brik R, et al: Seasonal variation in systemic onset juvenile rheumatoid arthritis in Israel. J Rheumatol 26: 1187–1189, 1999.
246
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
62. Andersson GB: Juvenile Chronic Arthritis. A Population Based Study on Epidemiology, Natural History and Outcome. Göteborg, Sweden, University of Göteborg, 1994. 63. Bywaters EG: Diagnostic criteria for Still’s disease (juvenile RA). In Bennett PH, Wood PHN (eds): Population Studies of the Rheumatic Diseases. Amsterdam, Excerpta Medica, 1968, p. 235. 64. Sullivan DB, Cassidy JT, Petty RE: Pathogenic implications of age of onset in juvenile rheumatoid arthritis. Arthritis Rheum 18: 251–255, 1975. 65. Towner SR, Michet CJ Jr, O’Fallon WM, et al: The epidemiology of juvenile arthritis in Rochester, Minnesota, 1960–1979. Arthritis Rheum 26: 1208–1213, 1983. 66. Gewanter HL, Roghmann KJ, Baum J: The prevalence of juvenile arthritis. Arthritis Rheum 26: 599–603, 1983. 67. Kunnamo I, Kallio P, Pelkonen P: Incidence of arthritis in urban Finnish children. A prospective study. Arthritis Rheum 29: 1232–1238, 1986. 68. Prieur AM, Le Gall E, Karman F, et al: Epidemiologic survey of juvenile chronic arthritis in France. Comparison of data obtained from two different regions. Clin Exp Rheumatol 5: 217–223, 1987. 69. Rosenberg AM: Analysis of a pediatric rheumatology clinic population. J Rheumatol 17: 827–830, 1990. 70. Mielants H, Veys EM, Maertens M, et al: Prevalence of inflammatory rheumatic diseases in an adolescent urban student population, age 12 to 18, in Belgium. Clin Exp Rheumatol 11: 563–567, 1993. 71. Gerhardt CA, Vannatta K, McKellop JM, et al: Brief report: child-rearing practices of caregivers with and without a child with juvenile rheumatoid arthritis: perspectives of caregivers and professionals. J Pediatr Psychol 28: 275–279, 2003. 72. Denardo BA, Tucker LB, Miller LC, et al: Demography of a regional pediatric rheumatology patient population. Affiliated Children’s Arthritis Centers of New England. J Rheumatol 21: 1553–1561, 1994. 73. Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry. J Rheumatol 23: 1981–1987, 1996. 74. Manners PJ, Diepeveen DA: Prevalence of juvenile chronic arthritis in a population of 12-year-old children in urban Australia. Pediatrics 98: 84–90, 1996. 75. Kaipiainen-Seppanen O, Savolainen A: Incidence of chronic juvenile rheumatic diseases in Finland during 1980–1990. Clin Exp Rheumatol 14: 441–444, 1996. 76. Symmons DP, Jones M, Osborne J, et al: Pediatric rheumatology in the United Kingdom: data from the British Pediatric Rheumatology Group National Diagnostic Register. J Rheumatol 23: 1975–1980, 1996. 77. Peterson LS, Mason T, Nelson AM, et al: Juvenile rheumatoid arthritis in Rochester, Minnesota, 1960–1993. Is the epidemiology changing? Arthritis Rheum 39: 1385–1390, 1996. 78. Fujikawa S, Okuni M: A nationwide surveillance study of rheumatic diseases among Japanese children. Acta Paediatr Jpn 39: 242–244, 1997. 79. Ozen S, Karaaslan Y, Ozdemir O, et al: Prevalence of juvenile chronic arthritis and familial Mediterranean fever in Turkey: a field study. J Rheumatol 25: 2445–2449, 1998. 80. Moe N, Rygg M: Epidemiology of juvenile chronic arthritis in northern Norway: a ten-year retrospective study. Clin Exp Rheumatol 16: 99–101, 1998. 81. Arguedas O, Fasth A, Andersson-Gare B, et al: Juvenile chronic arthritis in urban San Jose, Costa Rica: a 2-year prospective study. J Rheumatol 25: 1844–1850, 1998. 82. Kiessling U, Doring E, Listing J, et al: Incidence and prevalence of juvenile chronic arthritis in East Berlin 1980–1988. J Rheumatol 25: 1837–1843, 1998. 83. Gare BA: Epidemiology. Baillieres Clin Rheumatol 12: 191–208, 1998. 84. Kaipiainen-Seppanen O, Savolainen A: Changes in the incidence of juvenile rheumatoid arthritis in Finland. Rheumatology (Oxf) 40: 928–932, 2001. 85. Savolainen E, Kaipiainen-Seppanen O, Kroger L, et al: Total incidence and distribution of inflammatory joint diseases in a defined population: results from the Kuopio 2000 arthritis survey. J Rheumatol 30: 2460–2468, 2003. 86. Berntson L, Andersson GB, Fasth A, et al: Incidence of juvenile idiopathic arthritis in the Nordic countries. A population based study with special reference to the validity of the ILAR and EULAR criteria. J Rheumatol 30: 2275–2282, 2003. 87. Mikkelson WM, Dodge HJ, Duff IF, et al: Clinical and serological estimates of the prevalence of rheumatoid arthritis in the population of Tecumseh, Michigan, 1959–1960. In Kellgren J (ed): The Epidemiology of Chronic Rheumatism. Philadelphia, IFA Davis, 1963, p 239. 88. Adams BS: Juvenile arthritis. More common than you think. Mich Med 100: 53–54, 2001. 89. Edstrom G: Rheumatoid arthritis and Still’s disease in children. A survey of 161 cases. Arthritis Rheum 1: 497, 1958. 90. Sairanen E: On rheumatoid arthritis in children. A clinico-roentgenological study. Acta Rheum Scand Suppl 2: 1, 1958. 91. Schesinger BE, Forsyth CC, White RH: Observations on the clinical course and treatment of one hundred cases of Still’s disease. Arch Dis Child 36: 65, 1961. 92. Grokoest AW, Snyder AL, Schlaeger R: Juvenile Rheumatoid Arthritis. Boston, Little, Brown, 1962. 93. Ansell BM: Heberden Oration, 1977. Chronic arthritis in childhood. Ann Rheum Dis 37: 107–120, 1978.
94. Hill RH: Juvenile arthritis in various racial groups in British Columbia. Arthritis Rheum 20: 162, 1977. 95. Haffejee IE, Raga J, Coovadia HM: Juvenile chronic arthritis in black and Indian South African children. S Afr Med J 65: 510–514, 1984. 96. Graham TB, Glass DN: Juvenile rheumatoid arthritis: ethnic differences in diagnostic types. J Rheumatol 24: 1677–1679, 1997. 97. Schwartz MM, Simpson P, Kerr KL, et al: Juvenile rheumatoid arthritis in African Americans. J Rheumatol 24: 1826–1829, 1997. 98. Greenwood BM: Polyarthritis in Western Nigeria. II. Still’s disease. Ann Rheum Dis 28: 617–623, 1969. 99. McGill PE, Oyoo GO: Rheumatic disorders in Sub-saharan Africa. East Afr Med J 79: 214–216, 2002. 100. Kurahara D, Tokuda A, Grandinetti A, et al: Ethnic differences in risk for pediatric rheumatic illness in a culturally diverse population. J Rheumatol 29: 379–383, 2002. 101. Fujikawa S, Okuni M: Clinical analysis of 570 cases with juvenile rheumatoid arthritis: results of a nationwide retrospective survey in Japan. Acta Paediatr Jpn 39: 245–249, 1997. 102. Aggarwal A, Misra R: Juvenile chronic arthritis in India: is it different from that seen in Western countries? Rheumatol Int 14: 53–56, 1994. 103. Chandrasekaran AN, Rajendran CP, Madhavan R: Juvenile rheumatoid arthritis—Madras experience. Indian J Pediatr 63: 501–510, 1996. 104. Seth V, Kabra SK, Semwal OP, et al: Clinico-immunological profile in juvenile rheumatoid arthritis—an Indian experience. Indian J Pediatr 63: 293–300, 1996. 105. Rosenberg AM, Petty RE, Oen KG, et al: Rheumatic diseases in Western Canadian Indian children. J Rheumatol 9: 589–592, 1982. 106. Oen K, Postl B, Chalmers IM, et al: Rheumatic diseases in an Inuit population. Arthritis Rheum 29: 65–74, 1986. 107. Boyer GS, Lanier AP, Templin DW, et al: Spondyloarthropathy and rheumatoid arthritis in Alaskan Yupik Eskimos. J Rheumatol 17: 489–496, 1990. 108. Jarvis JN, Cleland SY: Rheumatic disease in native american children: opportunities and challenge. Curr Rheumatol Rep 5: 471–476, 2003. 109. Lang BA, Shore A: A review of current concepts on the pathogenesis of juvenile rheumatoid arthritis. J Rheumatol Suppl 21: 1–15, 1990. 110. Murray K, Thompson SD, Glass DN: Pathogenesis of juvenile chronic arthritis: genetic and environmental factors. Arch Dis Child 77: 530–534, 1997. 111. Jarvis JN: Pathogenesis and mechanisms of inflammation in the childhood rheumatic diseases. Curr Opin Rheumatol 10: 459–467, 1998. 112. Glass DN, Giannini EH: Juvenile rheumatoid arthritis as a complex genetic trait. Arthritis Rheum 42: 2261–2268, 1999. 113. Thomson W, Donn R: Juvenile idiopathic arthritis genetics—what’s new? What’s next? Arthritis Res 4: 302–306, 2002. 114. Petty RE: Etiology and pathogenesis of rheumatic diseases of adolescence. Adolesc Med 9: 11–24, 1998. 115. Lipnick RN, Tsokos GC, Magilavy DB: Immune abnormalities in the pathogenesis of juvenile rheumatoid arthritis. Rheum Dis Clin North Am 17: 843–857, 1991. 116. De Benedetti F, Ravelli A, Martini A: Cytokines in juvenile rheumatoid arthritis. Curr Opin Rheumatol 9: 428–433, 1997. 117. Moore TL: Immunopathogenesis of juvenile rheumatoid arthritis. Curr Opin Rheumatol 11: 377–383, 1999. 118. De Benedetti F, Vivarelli M, Pignatti P, et al: Circulating levels of soluble E-selectin, P-selectin and intercellular adhesion molecule-1 in patients with juvenile idiopathic arthritis. J Rheumatol 27: 2246–2250, 2000. 119. Woo P: Cytokines and juvenile idiopathic arthritis. Curr Rheumatol Rep 4: 452–457, 2002. 120. Mason T, Rabinovich CE, Fredrickson DD, et al: Breast feeding and the development of juvenile rheumatoid arthritis. J Rheumatol 22: 1166–1170, 1995. 121. Rosenberg AM: Evaluation of associations between breast feeding and subsequent development of juvenile rheumatoid arthritis. J Rheumatol 23: 1080–1082, 1996. 122. Moore TL, Dorner RW: Separation and characterization of complement-fixing immune complexes in juvenile rheumatoid arthritis patients. Rheumatol Int 6: 49–52, 1986. 123. Rossen RD, Brewer EJ, Person DA, et al: Circulating immune complexes and antinuclear antibodies in juvenile rheumatoid arthritis. Arthritis Rheum 20: 1485–1490, 1977. 124. Moran H, Ansell BM, Mowbray JF, et al: Antigen-antibody complexes in the serum of patients with juvenile chronic arthritis. Arch Dis Child 54: 120–122, 1979. 125. Khalkhali-Ellis Z, Bulla GA, Schlesinger LS, et al: C1q-containing immune complexes purified from sera of juvenile rheumatoid arthritis patients mediate IL-8 production by human synoviocytes: role of C1q receptors. J Immunol 163: 4612–4620, 1999. 126. Jarvis JN, Diebold MM, Chadwell MK, et al: Composition and biological behaviour of immune complexes isolated from synovial fluid of patients with juvenile rheumatoid arthritis (JRA). Clin Exp Immunol 100: 514–518, 1995. 127. Jarvis JN, Taylor H, Iobidze M, et al: Complement activation and immune complexes in children with polyarticular juvenile rheumatoid arthritis: a longitudinal study. J Rheumatol 21: 1124–1127, 1994. 128. Aggarwal A, Bhardwaj A, Alam S, et al: Evidence for activation of the alternate complement pathway in patients with juvenile rheumatoid arthritis. Rheumatology (Oxf) 39: 189–192, 2000.
C H A P T E R 129. Moore TL, Sheridan PW, Zuckner J, et al: Separation and characterization of immune complexes containing 19S IgM rheumatoid factor-IgG in juvenile arthritis. Arthritis Rheum 26: 165–169, 1983. 130. Moore TL, Osborn TG, Dorner RW: 19S IgM rheumatoid factor-7S IgG rheumatoid factor immune complexes isolated in sera of patients with juvenile rheumatoid arthritis. Pediatr Res 20: 977–981, 1986. 131. Jarvis JN, Iobidze M, Taylor H, et al: A comparison of immunoglobulin Gcontaining high-molecular-weight complexes isolated from children with juvenile rheumatoid arthritis and congenital human immunodeficiency virus infection. Pediatr Res 34: 781–784, 1993. 132. Moore TL, Dorner RW, Osborn TG, et al: Hidden 19S IgM rheumatoid factors. Semin Arthritis Rheum 18: 72–75, 1988. 133. Jarvis JN, Pousak T, Krenz M: Detection of IgM rheumatoid factors by enzyme-linked immunosorbent assay in children with juvenile rheumatoid arthritis: correlation with articular disease and laboratory abnormalities. Pediatrics 90: 945–949, 1992. 134. Woo P: Cytokines in juvenile chronic arthritis. Baillieres Clin Rheumatol 12: 219–228, 1998. 135. Harjacek M, Diaz-Cano S, Alman BA, et al: Prominent expression of mRNA for proinflammatory cytokines in synovium in patients with juvenile rheumatoid arthritis or chronic Lyme arthritis. J Rheumatol 27: 497–503, 2000. 136. Rooney M, Varsani H, Martin K, et al: Tumour necrosis factor alpha and its soluble receptors in juvenile chronic arthritis. Rheumatology (Oxf) 39: 432–438, 2000. 137. Yokota S: Interleukin 6 as a therapeutic target in systemic-onset juvenile idiopathic arthritis. Curr Opin Rheumatol 15: 581–586, 2003. 138. Donn R, Alourfi Z, Zeggini E, et al: A functional promoter haplotype of macrophage migration inhibitory factor is linked and associated with juvenile idiopathic arthritis. Arthritis Rheum 50: 1604–1610, 2004. 139. De Benedetti F, Martini A: Is systemic juvenile rheumatoid arthritis an interleukin 6 mediated disease? J Rheumatol 25: 203–207, 1998. 140. Mangge H, Schauenstein K: Cytokines in juvenile rheumatoid arthritis (JRA). Cytokine 10: 471–480, 1998. 141. De Maria A, Malnati M, Moretta A, et al: CD3+4-8-WT31- (T cell receptor gamma+) cells and other unusual phenotypes are frequently detected among spontaneously interleukin 2-responsive T lymphocytes present in the joint fluid in juvenile rheumatoid arthritis. A clonal analysis. Eur J Immunol 17: 1815–1819, 1987. 142. Bergroth V, Konttinen YT, Pelkonen P, et al: Synovial fluid lymphocytes in different subtypes of juvenile rheumatoid arthritis. Arthritis Rheum 31: 780–783, 1988. 143. Thoen J, Forre O, Waalen K, et al: Phenotypes of T lymphocytes from peripheral blood and synovial fluid of patients with rheumatoid arthritis and juvenile rheumatoid arthritis. Evidence in favour of normal helper and suppressor functions of T lymphocytes from patients with juvenile rheumatoid arthritis. Scand J Rheumatol 16: 247–256, 1987. 144. Agosti E, Radillo O, Narchi G, et al: Immunological patterns in monoarticular juvenile rheumatoid arthritis. Clin Exp Rheumatol 6: 319–324, 1988. 145. Wedderburn LR, Robinson N, Patel A, et al: Selective recruitment of polarized T cells expressing CCR5 and CXCR3 to the inflamed joints of children with juvenile idiopathic arthritis. Arthritis Rheum 43: 765–774, 2000. 146. Konttinen YT, Bergroth V, Kunnamo I, et al: The value of biopsy in patients with monarticular juvenile rheumatoid arthritis of recent onset. Arthritis Rheum 29: 47–53, 1986. 147. Silverman ED, Isacovics B, Petsche D, et al: Synovial fluid cells in juvenile arthritis: evidence of selective T cell migration to inflamed tissue. Clin Exp Immunol 91: 90–95, 1993. 148. Kutukculer N, Caglayan S, Aydogdu F: Study of pro-inflammatory (TNFalpha, IL-1alpha, IL-6) and T-cell-derived (IL-2, IL-4) cytokines in plasma and synovial fluid of patients with juvenile chronic arthritis: correlations with clinical and laboratory parameters. Clin Rheumatol 17: 288–292, 1998. 149. Scola MP, Thompson SD, Brunner HI, et al: Interferon-gamma:interleukin-4 ratios and associated type 1 cytokine expression in juvenile rheumatoid arthritis synovial tissue. J Rheumatol 29: 369–378, 2002. 150. Murray KJ, Luyrink L, Grom AA, et al: Immunohistological characteristics of T cell infiltrates in different forms of childhood onset chronic arthritis. J Rheumatol 23: 2116–2124, 1996. 151. Murray KJ, Grom AA, Thompson SD, et al: Contrasting cytokine profiles in the synovium of different forms of juvenile rheumatoid arthritis and juvenile spondyloarthropathy: prominence of interleukin 4 in restricted disease. J Rheumatol 25: 1388–1398, 1998. 152. Chini L, Bardare M, Cancrini C, et al: Evidence of clonotypic pattern of T-cell repertoire in synovial fluid of children with juvenile rheumatoid arthritis at the onset of the disease. Scand J Immunol 56: 512–517, 2002. 153. Grom AA, Murray KJ, Luyrink L, et al: Patterns of expression of tumor necrosis factor alpha, tumor necrosis factor beta, and their receptors in synovia of patients with juvenile rheumatoid arthritis and juvenile spondylarthropathy. Arthritis Rheum 39: 1703–1710, 1996. 154. Muzaffer MA, Dayer JM, Feldman BM, et al: Differences in the profiles of circulating levels of soluble tumor necrosis factor receptors and interleukin 1 receptor antagonist reflect the heterogeneity of the subgroups of juvenile rheumatoid arthritis. J Rheumatol 29: 1071–1078, 2002. 155. Thompson SD, Luyrink LK, Graham TB, et al: Chemokine receptor CCR4 on CD4+ T cells in juvenile rheumatoid arthritis synovial fluid defines a subset
9 C H R O N I C A RT H R I T I S 156. 157.
158.
159. 160.
161. 162. 163. 164. 165. 166. 167. 168.
169. 170.
171. 172. 173. 174. 175. 176. 177. 178. 179. 180. 181.
IN
CHILDHOOD
247
of cells with increased IL-4:IFN-gamma mRNA ratios. J Immunol 166: 6899–6906, 2001. Varsani H, Patel A, van Kooyk Y, et al: Synovial dendritic cells in juvenile idiopathic arthritis (JIA) express receptor activator of NF-kappaB (RANK). Rheumatology (Oxf) 42: 583–590, 2003. Gattorno M, Facchetti P, Ghiotto F, et al: Synovial fluid T cell clones from oligoarticular juvenile arthritis patients display a prevalent Th1/Th0-type pattern of cytokine secretion irrespective of immunophenotype. Clin Exp Immunol 109: 4–11, 1997. Raziuddin S, Bahabri S, Al Dalaan A, et al: A mixed Th1/Th2 cell cytokine response predominates in systemic onset juvenile rheumatoid arthritis: immunoregulatory IL-10 function. Clin Immunol Immunopathol 86: 192–198, 1998. Oglivie EM, Fife MS, Thompson SD, et al: TDT association study confirms the IL6-174 SNP confers susceptibility to systemic JIA. Arthritis Rheum 46 (Suppl): S578, 2002. Jefferey R, Luong L, Ogilvie E, et al: The -174 single nucleotide polymorphism (SNP) of the interleukin-6 gene associated with systemic arthritis is functionally significant in the control of common haplotypes, alters transcription factor binding and repression by glucocorticoids. Arthritis Rheum 46 (Suppl): S578, 2002. Meazza C, Travaglino P, Pignatti P, et al: Macrophage migration inhibitory factor in patients with juvenile idiopathic arthritis. Arthritis Rheum 46: 232–237, 2002. De Benedetti F, Meazza C, Vivarelli M, et al: Functional and prognostic significance of the -173 polymorphism of the macrophage migration inhibitory factor (MIF) gene in systemic JIA. Arthritis Rheum 46 (Suppl): S580, 2002. Zeggini E, Thomson W, Kwiatkowski D, et al: Linkage and association studies of single-nucleotide polymorphism-tagged tumor necrosis factor haplotypes in juvenile oligoarthritis. Arthritis Rheum 46: 3304–3311, 2002. Koren S, Hanly JG: Adrenal failure in systemic lupus erythematosus. J Rheumatol 24: 1410–1412, 1997. Ozen S, Alikasifoglu M, Bakkaloglu A, et al: Tumour necrosis factor alpha G→A −238 and G→A −308 polymorphisms in juvenile idiopathic arthritis. Rheumatology (Oxf) 41: 223–227, 2002. Grom AA, Giannini EH, Glass DN: Juvenile rheumatoid arthritis and the trimolecular complex (HLA, T cell receptor, and antigen). Differences from rheumatoid arthritis. Arthritis Rheum 37: 601–607, 1994. Grom AA, Hirsch R: T-cell and T-cell receptor abnormalities in the immunopathogenesis of juvenile rheumatoid arthritis. Curr Opin Rheumatol 12: 420–424, 2000. Wedderburn LR, Patel A, Varsani H, et al: Divergence in the degree of clonal expansions in inflammatory T cell subpopulations mirrors HLA-associated risk alleles in genetically and clinically distinct subtypes of childhood arthritis. Int Immunol 13: 1541–1550, 2001. Sioud M, Kjeldsen-Kragh J, Suleyman S, et al: Limited heterogeneity of T cell receptor variable region gene usage in juvenile rheumatoid arthritis synovial T cells. Eur J Immunol 22: 2413–2418, 1992. Thompson SD, Murray KJ, Grom AA, et al: Comparative sequence analysis of the human T cell receptor beta chain in juvenile rheumatoid arthritis and juvenile spondylarthropathies: evidence for antigenic selection of T cells in the synovium. Arthritis Rheum 41: 482–497, 1998. Maksymowych WP, Gabriel CA, Luyrink L, et al: Polymorphism in a T-cell receptor variable gene is associated with susceptibility to a juvenile rheumatoid arthritis subset. Immunogenetics 35: 257–262, 1992. Grom AA, Von Knorre C, Murray KJ, et al: T-cell receptor BV6S1 null alleles and HLA-DR1 haplotypes in polyarticular outcome juvenile rheumatoid arthritis. Hum Immunol 45: 152–156, 1996. Ploski R, Hansen T, Forre O: Lack of association with T-cell receptor TCRBV6S1*2 allele in HLA-DQA1*0101–positive Norwegian juvenile chronic arthritis patients. Immunogenetics 38: 444–445, 1993. Nepom BS, Malhotra U, Schwarz DA, et al: HLA and T cell receptor polymorphisms in pauciarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 34: 1260–1267, 1991. Morimoto C, Reinherz EL, Borel Y, et al: Autoantibody to an immunoregulatory inducer population in patients with juvenile rheumatoid arthritis. J Clin Invest 67: 753–761, 1981. Astrup LB, Morling N, Ryder LP, et al: Concanavalin-A-activated suppressor cells in patients with juvenile rheumatoid arthritis. Scand J Immunol 16: 361–367, 1982. Ellsworth JE, Stein LD, Thebert PJ, et al: Abnormalities of lymphokine generation in children with juvenile rheumatoid arthritis. Pediatr Res 16: 221A, 1982. De Benedetti F, Marconi M, Ravelli A, et al: Multiple inhibitors of mitogeninduced proliferation of normal lymphocytes in juvenile chronic arthritis sera. Clin Exp Rheumatol 8: 505–511, 1990. Barron KS, Lewis DE, Brewer EJ, et al: Cytotoxic anti-T cell antibodies in children with juvenile rheumatoid arthritis. Arthritis Rheum 27: 1272–1280, 1984. Oen K, Wilkins JA, Krzekotowska D: OKT4:OKT8 ratios of circulating T cells and in vitro suppressor cell function of patients with juvenile rheumatoid arthritis (JRA). J Rheumatol 12: 321–327, 1985. Murata H, Yata J: Defect of suppressor cell induction in patients with juvenile rheumatoid arthritis. Asian Pac J Allergy Immunol 4: 95–99, 1986.
248
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
182. Alpert SD, Turek PJ, Foung SK, et al: Human monoclonal anti-T cell antibody from a patient with juvenile rheumatoid arthritis. J Immunol 138: 104–108, 1987. 183. Tsokos GC, Mavridis A, Inghirami G, et al: Cellular immunity in patients with systemic juvenile rheumatoid arthritis. Clin Immunol Immunopathol 42: 86–92, 1987. 184. Tsokos GC, Inghirami G, Pillemer SR, et al: Immunoregulatory aberrations in patients with polyarticular juvenile rheumatoid arthritis. Clin Immunol Immunopathol 47: 62–74, 1988. 185. Wouters CH, Ceuppens JL, Stevens EA: Different circulating lymphocyte profiles in patients with different subtypes of juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 239–248, 2002. 186. Alarcon-Riquelme ME, Vazquez-Mellado J, Gomez-Cordillo M, et al: Immunoregulatory defects in juvenile rheumatoid arthritis. Comparison between patients with the systemic or polyarticular forms. J Rheumatol 15: 1547–1550, 1988. 187. Wedderburn LR, Maini MK, Patel A, et al: Molecular fingerprinting reveals non-overlapping T cell oligoclonality between an inflamed site and peripheral blood. Int Immunol 11: 535–543, 1999. 188. Dobloug JH, Chattopadhyay C, Forre O, et al: Con A-induced suppressor cell activity and T-lymphocyte subpopulations in peripheral blood lymphocytes of patients with rheumatoid arthritis and juvenile rheumatoid arthritis. Scand J Immunol 13: 367–373, 1981. 189. Oen K: Defects in pokeweed mitogen (PWM) induced immunoglobulin (Ig) synthesis by lymphocytes of patients with juvenile rheumatoid arthritis. J Rheumatol 12: 728–734, 1985. 190. Odum N, Morling N, Platz P, et al: Increased prevalence of late stage T cell activation antigen (VLA-1) in active juvenile chronic arthritis. Ann Rheum Dis 46: 846–852, 1987. 191. Forre O, Thoen J, Dobloug JH, et al: Detection of T-lymphocyte subpopulation in the peripheral blood and the synovium of patients with rheumatoid arthritis and juvenile rheumatoid arthritis using monoclonal antibodies. Scand J Immunol 15: 221–226, 1982. 192. Massa M, De Benedetti F, Robbioni P, et al: Association of methotrexate treatment with a decrease of double negative (CD4-CD8-) and gamma/delta T cell levels in patients with juvenile rheumatoid arthritis. J Rheumatol 20: 1944–1948, 1993. 193. Oen K, Krzekotowska D: In vitro immunoglobulin production by lymphocytes of patients with juvenile rheumatoid arthritis: effects of Staphylococcus aureus Cowan I stimulation and monocyte depletion. J Rheumatol 15: 1539–1546, 1988. 194. Ilowite NT, Wedgwood RJ, Rose LM, et al: Impaired in vivo and in vitro antibody responses to bacteriophage phi X 174 in juvenile rheumatoid arthritis [published erratum appears in J Rheumatol 15: 386, 1988]. J Rheumatol 14: 957–963, 1987. 195. Leak AM: Autoantibody profile in juvenile chronic arthritis. Ann Rheum Dis 47: 178–182, 1988. 196. Pauls JD, Silverman E, Laxer RM, et al: Antibodies to histones H1 and H5 in sera of patients with juvenile rheumatoid arthritis. Arthritis Rheum 32: 877–883, 1989. 197. Malleson P, Petty RE, Fung M, et al: Reactivity of antinuclear antibodies with histones and other antigens in juvenile rheumatoid arthritis. Arthritis Rheum 32: 919–923, 1989. 198. Monestier M, Losman JA, Fasy TM, et al: Antihistone antibodies in antinuclear antibody-positive juvenile arthritis. Arthritis Rheum 33: 1836–1841, 1990. 199. Leak AM, Woo P: Juvenile chronic arthritis, chronic iridocyclitis, and reactivity to histones. Ann Rheum Dis 50: 653–657, 1991. 200. Malleson PN, Fung MY, Petty RE, et al: Autoantibodies in chronic arthritis of childhood: relations with each other and with histocompatibility antigens. Ann Rheum Dis 51: 1301–1306, 1992. 201. Leak AM, Tuaillon N, Muller S, et al: Study of antibodies to histones and histone synthetic peptides in pauciarticular juvenile chronic arthritis. Br J Rheumatol 32: 426–431, 1993. 202. Stemmer C, Tuaillon N, Prieur AM, et al: Mapping of B-cell epitopes recognized by antibodies to histones in subsets of juvenile chronic arthritis. Clin Immunol Immunopathol 76: 82–89, 1995. 203. Kageura H: Activation of dorsal development by contact between the cortical dorsal determinant and the equatorial core cytoplasm in eggs of Xenopus laevis. Development 124: 1543–1551, 1997. 204. Speckmaier M, Rother E, Terreri T, et al: Prevalence of anti-neutrophil cytoplasmic antibodies (ANCA) in juvenile chronic arthritis. Clin Exp Rheumatol 14: 211–216, 1996. 205. Mulder L, Van Rossum M, Horst G, et al: Antineutrophil cytoplasmic antibodies in juvenile chronic arthritis. J Rheumatol 24: 568–575, 1997. 206. Bakkaloglu A, Ozen S, Saatci U, et al: Antineutrophil cytoplasmic antibodies in juvenile chronic arthritis. Clin Rheumatol 18: 304–307, 1999. 207. Gabay C, Prieur AM, Meyer O: Occurrence of antiperinuclear, antikeratin, and anti-RA 33 antibodies in juvenile chronic arthritis. Ann Rheum Dis 52: 785–789, 1993. 208. Serra CR, Rodrigues SH, Sztajnbok FR, et al: Antiperinuclear factor and antibodies to the stratum corneum of rat esophagus in juvenile idiopathic arthritis. J Pediatr 134: 507–509, 1999. 209. Hromadnikova I, Vavrincova P, Stechova K, et al: Anti-keratin antibodies in patients with juvenile idiopathic arthritis. Clin Exp Rheumatol 19: 470–473, 2001.
210. Caporali R, Ravelli A, De Gennaro F, et al: Prevalence of anticardiolipin antibodies in juvenile chronic arthritis. Ann Rheum Dis 50: 599–601, 1991. 211. Gattorno M, Buoncompagni A, Molinari AC, et al: Antiphospholipid antibodies in paediatric systemic lupus erythematosus, juvenile chronic arthritis and overlap syndromes: SLE patients with both lupus anticoagulant and high-titre anticardiolipin antibodies are at risk for clinical manifestations related to the antiphospholipid syndrome. Br J Rheumatol 34: 873–881, 1995. 212. Serra CR, Rodrigues SH, Silva NP, et al: Clinical significance of anticardiolipin antibodies in juvenile idiopathic arthritis. Clin Exp Rheumatol 17: 375–380, 1999. 213. Dong X, Wang J, Kabir FN, et al: Autoantibodies to DEK oncoprotein in human inflammatory disease. Arthritis Rheum 43: 85–93, 2000. 214. Wittemann B, Neuer G, Michels H, et al: Autoantibodies to nonhistone chromosomal proteins HMG-1 and HMG-2 in sera of patients with juvenile rheumatoid arthritis. Arthritis Rheum 33: 1378–1383, 1990. 215. Neuer G, Bustin M, Michels H, et al: Autoantibodies to the chromosomal protein HMG-17 in juvenile rheumatoid arthritis. Arthritis Rheum 35: 472–475, 1992. 216. Neuer G, Bautz FA, Bustin M, et al: Sera from JRA patients contain antibodies against a defined epitope in chromosomal protein HMG-17. Autoimmunity 17: 23–30, 1994. 217. Jung F, Neuer G, Bautz FA: Antibodies against a peptide sequence located in the linker region of the HMG-1/2 box domains in sera from patients with juvenile rheumatoid arthritis. Arthritis Rheum 40: 1803–1809, 1997. 218. Rosenberg AM, Cordeiro DM: Relationship between sex and antibodies to high mobility group proteins 1 and 2 in juvenile idiopathic arthritis. J Rheumatol 27: 2489–2493, 2000. 219. Rosenberg AM, Hunt DW, Petty RE: Antibodies to native type I collagen in childhood rheumatic diseases. J Rheumatol 11: 421–424, 1984. 220. Petty RE, Hunt DW, Rosenberg AM: Antibodies to type IV collagen in rheumatic diseases. J Rheumatol 13: 246–253, 1986. 221. Myers LK, Higgins GC, Finkel TH, et al: Juvenile arthritis and autoimmunity to type II collagen. Arthritis Rheum 44: 1775–1781, 2001. 222. Guerassimov A, Duffy C, Zhang Y, et al: Immunity to cartilage link protein in patients with juvenile rheumatoid arthritis. J Rheumatol 24: 959–964, 1997. 223. Pellegrini G, Scotta MS, Soardo S, et al: Elevated IgA anti-gliadin antibodies in juvenile chronic arthritis. Clin Exp Rheumatol 9: 653–656, 1991. 224. Lepore L, Pennesi M, Ventura A, et al: Anti-alpha-gliadin antibodies are not predictive of celiac disease in juvenile chronic arthritis. Acta Paediatr 82: 569–573, 1993. 225. Van Rossum M, van Soesbergen R, de Kort S, et al: Anti-cyclic citrullinated peptide (anti-CCP) antibodies in children with juvenile idiopathic arthritis. J Rheumatol 30: 825–828, 2003. 226. Avcin T, Cimaz R, Falcini F, et al: Prevalence and clinical significance of anticyclic citrullinated peptide antibodies in juvenile idiopathic arthritis. Ann Rheum Dis 61: 608–611, 2002. 227. Neeck G, Michels H: Endocrine aspects of paediatric rheumatic diseases. Baillieres Clin Rheumatol 10: 349–363, 1996. 228. Chikanza IC, Kuis W, Heijnen CJ: The influence of the hormonal system on pediatric rheumatic diseases. Rheum Dis Clin North Am 26: 911–925, 2000. 229. Khalkhali-Ellis Z, Moore TL, Hendrix MJ: Reduced levels of testosterone and dehydroepiandrosterone sulphate in the serum and synovial fluid of juvenile rheumatoid arthritis patients correlates with disease severity. Clin Exp Rheumatol 16: 753–756, 1998. 230. Da Silva JA, Larbre JP, Spector TD, et al: Protective effect of androgens against inflammation induced cartilage degradation in male rodents. Ann Rheum Dis 52: 285–291, 1993. 231. Chikanza IC: Prolactin and neuroimmunomodulation: in vitro and in vivo observations. Ann N Y Acad Sci 876: 119–130, 1999. 232. McMurray RW, Allen SH, Pepmueller PH, et al: Elevated serum prolactin levels in children with juvenile rheumatoid arthritis and antinuclear antibody seropositivity. J Rheumatol 22: 1577–1580, 1995. 233. Picco P, Gattorno M, Buoncompagni A, et al: Interactions between prolactin and the proinflammatory cytokine network in juvenile chronic arthritis. Ann N Y Acad Sci 876: 262–265, 1999. 234. Petty RE, Tingle AJ: Arthritis and viral infection. J Pediatr 113: 948–949, 1988. 235. Pugh MT, Southwood TR, Gaston JS: The role of infection in juvenile chronic arthritis. Br J Rheumatol 32: 838–844, 1993. 236. Berman A, Cahn P, Perez H, et al: Human immunodeficiency virus infection associated arthritis: clinical characteristics. J Rheumatol 26: 1158–1162, 1999. 237. Vaughan JH: Viruses and autoimmune disease. J Rheumatol 23: 1831–1833, 1996. 238. Chantler JK, Tingle AJ, Petty RE: Persistent rubella virus infection associated with chronic arthritis in children. N Engl J Med 313: 1117–1123, 1985. 239. Smith CA, Petty RE, Tingle AJ: Rubella virus and arthritis. Rheum Dis Clin North Am 13: 265–274, 1987. 240. Phillips PE, Dougherty RM: Detection of rubella virus using polymerase chain reaction after vaccination and in arthritis. Arthritis Rheum Suppl 33: S134, 1990. 241. Frenkel LM, Nielsen K, Garakian A, et al: A search for persistent rubella virus infection in persons with chronic symptoms after rubella and rubella immunization and in patients with juvenile rheumatoid arthritis. Clin Infect Dis 22: 287–294, 1996. 242. Maillefert JF, Tonolli-Serabian I, Cherasse A, et al: Arthritis following combined vaccine against diphtheria, polyomyelitis, and tetanus toxoid. Clin Exp Rheumatol 18: 255–256, 2000.
C H A P T E R 243. Weibel RE, Benor DE: Chronic arthropathy and musculoskeletal symptoms associated with rubella vaccines. A review of 124 claims submitted to the National Vaccine Injury Compensation Program [published erratum appears in Arthritis Rheum 39: 1930, 1996]. Arthritis Rheum 39: 1529–1534, 1996. 244. Mitchell LA, Tingle AJ, MacWilliam L, et al: HLA-DR class II associations with rubella vaccine-induced joint manifestations. J Infect Dis 177: 5–12, 1998. 245. Pritchard MH, Matthews N, Munro J: Antibodies to influenza A in a cluster of children with juvenile chronic arthritis. Br J Rheumatol 27: 176–180, 1988. 246. Pritchard MH, Munro J: Successful treatment of juvenile chronic arthritis with a specific antiviral agent. Br J Rheumatol 28: 521–524, 1989. 247. Schwarz TF, Roggendorf M, Suschke H, et al: Human parvovirus B19 infection and juvenile chronic polyarthritis. Infection 15: 264–265, 1987. 248. Nocton JJ, Miller LC, Tucker LB, et al: Human parvovirus B19-associated arthritis in children. J Pediatr 122: 186–190, 1993. 249. Soderlund M, Von Essen R, Haapasaari J, et al: Persistence of parvovirus B19 DNA in synovial membranes of young patients with and without chronic arthropathy. Lancet 349: 1063–1065, 1997. 250. Oguz F, Akdeniz C, Unuvar E, et al: Parvovirus B19 in the acute arthropathies and juvenile rheumatoid arthritis. J Paediatr Child Health 38: 358–362, 2002. 251. Saulsbury FT: Remission of juvenile rheumatoid arthritis with varicella infection. J Rheumatol 26: 1606–1608, 1999. 252. Bateman HE, Kirou KA, Paget SA, et al: Remission of juvenile rheumatoid arthritis after infection with parvovirus B19. J Rheumatol 26: 2482–2484, 1999. 253. Cassidy JT, Shillis JL, Brandon FB, et al: Viral antibody titers to rubella and rubeola in juvenile rheumatoid arthritis. Pediatrics 54: 239–244, 1974. 254. Massa M, Mazzoli F, Pignatti P, et al: Proinflammatory responses to self HLA epitopes are triggered by molecular mimicry to Epstein-Barr virus proteins in oligoarticular juvenile idiopathic arthritis. Arthritis Rheum 46: 2721–2729, 2002. 255. Maximov AA, Shaikov AV, Lovell DJ, et al: Chlamydial associated syndrome of arthritis and eye involvement in young children. J Rheumatol 19: 1794–1797, 1992. 256. Burgos-Vargas R, Howard A, Ansell BM: Antibodies to peptidoglycan in juvenile onset ankylosing spondylitis and pauciarticular onset juvenile arthritis associated with chronic iridocyclitis. J Rheumatol 13: 760–762, 1986. 257. Pope JE, Stevens A, Howson W, et al: The development of rheumatoid arthritis after recombinant hepatitis B vaccination. J Rheumatol 25: 1687–1693, 1998. 258. Maillefert JF, Sibilia J, Toussirot E, et al: Rheumatic disorders developed after hepatitis B vaccination. Rheumatology (Oxf) 38: 978–983, 1999. 259. Graeff-Meeder ER, Van Eden W, Rijkers GT, et al: Heat-shock proteins and juvenile chronic arthritis. Clin Exp Rheumatol 11 (Suppl 9): S25–S28, 1993. 260. Life P, Hassell A, Williams K, et al: Responses to gram negative enteric bacterial antigens by synovial T cells from patients with juvenile chronic arthritis: recognition of heat shock protein HSP60. J Rheumatol 20: 1388–1396, 1993. 261. Albani S, Ravelli A, Massa M, et al: Immune responses to the Escherichia coli dnaJ heat shock protein in juvenile rheumatoid arthritis and their correlation with disease activity. J Pediatr 124: 561–565, 1994. 262. Prakken AB, Van Hoeij MJ, Kuis W, et al: T-cell reactivity to human HSP60 in oligo-articular juvenile chronic arthritis is associated with a favorable prognosis and the generation of regulatory cytokines in the inflamed joint. Immunol Lett 57: 139–142, 1997. 263. Conroy SE, Tucker L, Latchman DS, et al: Incidence of anti Hsp 90 and 70 antibodies in children with SLE, juvenile dermatomyositis and juvenile chronic arthritis. Clin Exp Rheumatol 14: 99–104, 1996. 264. Prakken B, Kuis W, Van Eden W, et al: Heat shock proteins in juvenile idiopathic arthritis: keys for understanding remitting arthritis and candidate antigens for immune therapy. Curr Rheumatol Rep 4: 466–473, 2002. 265. Van Eden W, Van der Zee R, Taams LS, et al: Heat-shock protein T-cell epitopes trigger a spreading regulatory control in a diversified arthritogenic Tcell response. Immunol Rev 164: 169–174, 1998. 266. Prakken AB, Van Eden W, Rijkers GT, et al: Autoreactivity to human heatshock protein 60 predicts disease remission in oligoarticular juvenile rheumatoid arthritis. Arthritis Rheum 39: 1826–1832, 1996. 267. De Kleer IM, Kamphuis SM, Rijkers GT, et al: The spontaneous remission of juvenile idiopathic arthritis is characterized by CD30+ T cells directed to human heat-shock protein 60 capable of producing the regulatory cytokine interleukin-10. Arthritis Rheum 48: 2001–2010, 2003. 268. Albani S, Keystone EC, Nelson JL, et al: Positive selection in autoimmunity: abnormal immune responses to a bacterial dnaJ antigenic determinant in patients with early rheumatoid arthritis. Nat Med 1: 448–452, 1995. 269. Danieli MG, Markovits D, Gabrielli A, et al: Juvenile rheumatoid arthritis patients manifest immune reactivity to the mycobacterial 65-kDa heat shock protein, to its 180–188 peptide, and to a partially homologous peptide of the proteoglycan link protein. Clin Immunol Immunopathol 64: 121–128, 1992. 270. Nuallain EM, Monaghan H, Reen DJ: Antibody response of restricted isotype to heat shock proteins in juvenile chronic arthritis. Scand J Immunol 38: 83–88, 1993. 271. Albani S: Infection and molecular mimicry in autoimmune diseases of childhood. Clin Exp Rheumatol 12 (Suppl 10): S35–S41, 1994. 272. Henoch MJ, Batson JW, Baum J: Psychosocial factors in juvenile rheumatoid arthritis. Arthritis Rheum 21: 229–233, 1978.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
249
273. Vandvik IH, Hoyeraal HM, Fagertun H: Chronic family difficulties and stressful life events in recent onset juvenile arthritis. J Rheumatol 16: 1088–1092, 1989. 274. Kuis W, Kavelaars A, Prakken BJ, et al: Dialogue between the brain and the immune system in juvenile chronic arthritis. Rev Rhum Engl Ed 64: 146S–148S, 1997. 275. Donn RP, Farhan A, Stevans A, et al: Neuroendocrine gene polymorphisms and susceptibility to juvenile idiopathic arthritis. Rheumatology (Oxf) 41: 930–936, 2002. 276. Scola MP, Imagawa T, Boivin GP, et al: Expression of angiogenic factors in juvenile rheumatoid arthritis: correlation with revascularization of human synovium engrafted into SCID mice. Arthritis Rheum 44: 794–801, 2001. 277. Jarvis JN, Wang W, Moore HT, et al: In vitro induction of proinflammatory cytokine secretion by juvenile rheumatoid arthritis synovial fluid immune complexes. Arthritis Rheum 40: 2039–2046, 1997. 278. Vignola S, Picco P, Falcini F, et al: Serum and synovial fluid concentration of vascular endothelial growth factor in juvenile idiopathic arthritides. Rheumatology (Oxf) 41: 691–696, 2002. 279. Pignatti P, Massa M, Travaglino P, et al: Activation-induced cell death and Fas-induced apoptosis in patients with systemic or pauciarticular juvenile idiopathic arthritis. Clin Exp Rheumatol 19: 339–344, 2001. 280. Harjacek M, Diaz-Cano S, De Miguel M, et al: Expression of galectins-1 and -3 correlates with defective mononuclear cell apoptosis in patients with juvenile idiopathic arthritis. J Rheumatol 28: 1914–1922, 2001. 281. De Benedetti F, Pignatti P, Biffi M, et al: Increased expression of alpha(1,3)fucosyltransferase-VII and P-selectin binding of synovial fluid T cells in juvenile idiopathic arthritis. J Rheumatol 30: 1611–1615, 2003. 282. Bardare M, Corona F, Di Francesco L, et al: Levels of circulating intercellular adhesion molecule 1 and E-selectin in serum and synovial fluid of juvenile chronic arthritis patients. Acta Univ Carol Med Praha 40: 81–85, 1994. 283. Bloom BJ, Nelson SM, Alario AJ, et al: Synovial fluid levels of E-selectin and intercellular adhesion molecule-1: relationship to joint inflammation in children with chronic arthritis. Rheumatol Int 22: 175–177, 2002. 284. Mussoni L, Pintucci G, Romano G, et al: Decreased fibrinolytic activity in juvenile chronic arthritis. Ann Rheum Dis 49: 973–975, 1990. 285. Gattorno M, Vignola S, Falcini F, et al: Serum and synovial fluid concentrations of matrix metalloproteinases 3 and its tissue inhibitor 1 in juvenile idiopathic arthritides. J Rheumatol 29: 826–831, 2002. 286. Potocnik AJ, Menninger H, Yang SY, et al: Expression of the CD2 activation epitope T11-3 (CD2R) on T cells in rheumatoid arthritis, juvenile rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, and Lyme disease: phenotypic and functional analysis. Scand J Immunol 34: 351–358, 1991. 287. Gattorno M, Prigione I, Vignola S, et al: Levels of soluble CD27 in sera and synovial fluid and its expression on memory T cells in patients with juvenile idiopathic arthritides. Clin Exp Rheumatol 20: 863–866, 2002. 288. Taubert H, Thamm B, Meye A, et al: The p53 status in juvenile chronic arthritis and rheumatoid arthritis. Clin Exp Immunol 122: 264–269, 2000. 289. Greenwald RA, Diamond HS: CRC Handbook of Animal Models for the Rheumatic Diseases. Boca Raton, FL, CRC Press, 1988. 290. Sokoloff L: Animal models of rheumatoid arthritis. Int Rev Exp Pathol 26: 107–145, 1984. 291. Pearson CM: Development of arthritis, periarthritis and periostitis in rats given adjuvants. Proc Soc Exp Biol Med 91: 95–101, 1956. 292. Waksman BH, Bullington SJ: Studies of arthritis and other lesions induced in rats by injection of mycobacterial adjuvant. III. Lesions of the eye. Arch Ophthalmol 64: 751–762, 1960. 293. Holoshitz J, Naparstek Y, Ben Nun A, et al: Lines of T lymphocytes induce or vaccinate against autoimmune arthritis. Science 219: 56–58, 1983. 294. Van Eden W, Holoshitz J, Nevo Z, et al: Arthritis induced by a T-lymphocyte clone that responds to Mycobacterium tuberculosis and to cartilage proteoglycans. Proc Natl Acad Sci U S A 82: 5117–5120, 1985. 295. Lider O, Karin N, Shinitzky M, et al: Therapeutic vaccination against adjuvant arthritis using autoimmune T cells treated with hydrostatic pressure. Proc Natl Acad Sci U S A 84: 4577–4580, 1987. 296. Stanescu R, Lider O, Van Eden W, et al: Histopathology of arthritis induced in rats by active immunization to mycobacterial antigens or by systemic transfer of T lymphocyte lines. A light and electron microscopic study of the articular surface using cationized ferritin. Arthritis Rheum 30: 779–792, 1987. 297. Zidek Z, Masek K, Jiricka Z: Arthritogenic activity of a synthetic immunoadjuvant, muramyl dipeptide. Infect Immun 35: 674–679, 1982. 298. Moder KG, Nabozny GH, Luthra HS, et al: Immunogenetics of collagen induced arthritis in mice: a model for human polyarthritis. Reg Immunol 4: 305–313, 1992. 299. Staines NA, Wooley PH: Collagen arthritis—what can it teach us? Br J Rheumatol 33: 798–807, 1994. 300. Nabozny GH, David CS: The immunogenetic basis of collagen induced arthritis in mice: an experimental model for the rational design of immunomodulatory treatments of rheumatoid arthritis. Adv Exp Med Biol 347: 55–63, 1994. 301. Cremer MA, Rosloniec EF, Kang AH: The cartilage collagens: a review of their structure, organization, and role in the pathogenesis of experimental arthritis in animals and in human rheumatic disease. J Mol Med 76: 275–288, 1998.
250
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
302. Myers LK, Rosloniec EF, Cremer MA, et al: Collagen-induced arthritis, an animal model of autoimmunity. Life Sci 61: 1861–1878, 1997. 303. Anthony DD, Haqqi TM: Collagen-induced arthritis in mice: an animal model to study the pathogenesis of rheumatoid arthritis. Clin Exp Rheumatol 17: 240–244, 1999. 304. Stuart JM, Townes AS, Kang AH: Collagen autoimmune arthritis. Annu Rev Immunol 2: 199–218, 1984. 305. Kakimoto K, Katsuki M, Hirofuji T, et al: Isolation of T cell line capable of protecting mice against collagen-induced arthritis. J Immunol 140: 78–83, 1988. 306. Holmdahl R, Andersson M, Goldschmidt TJ, et al: Type II collagen autoimmunity in animals and provocations leading to arthritis. Immunol Rev 118: 193–232, 1990. 307. Holmdahl R, Malmstrom V, Vuorio E: Autoimmune recognition of cartilage collagens. Ann Med 25: 251–264, 1993. 308. Durie FH, Fava RA, Noelle RJ: Collagen-induced arthritis as a model of rheumatoid arthritis. Clin Immunol Immunopathol 73: 11–18, 1994. 309. Staines NA, Harper N, Ward FJ, et al: Arthritis: animal models of oral tolerance. Ann N Y Acad Sci 778: 297–305, 1996. 310. Moore AR: Collagen-induced arthritis. Methods Mol Biol 225: 175–179, 2003. 311. Ma Y, Thornton S, Duwel LE, et al: Inhibition of collagen-induced arthritis in mice by viral IL-10 gene transfer. J Immunol 161: 1516–1524, 1998. 312. Kim KN, Watanabe S, Ma Y, et al: Viral IL-10 and soluble TNF receptor act synergistically to inhibit collagen-induced arthritis following adenovirusmediated gene transfer. J Immunol 164: 1576–1581, 2000. 313. Sikes D: A rheumatoidlike arthritis in swine. Lab Invest 8: 1406–1415, 1959. 314. Drew RA: Erysipelothrix arthritis in pigs as a comparative model for rheumatoid arthritis. Proc R Soc Med 65: 994–998, 1972. 315. Goudswaard J, Hartman EG, Janmaat A, et al: Erysipelothrix rhusiopathiae strain 7: a causative agent of endocarditis and arthritis in the dog. Tijdschr Diergeneeskd 98: 416–423, 1973. 316. Cole BC: Mycoplasma-induced arthritis in animals: relevance to understanding the etiologies of the human rheumatic diseases. Rev Rhum Engl Ed 66: 45S–49S, 1999. 317. Storz J: Chlamydia and chlamydia-induced diseases. Springfield, IL, Charles C Thomas, 1971. 318. Walker ER, Friedman MH, Olson NO, et al: Ultrastructural study of avian synovium infected with an arthrotropic reovirus. Arthritis Rheum 20: 1269–1277, 1977. 319. Crawford TB, Adams DS, Sande RD, et al: The connective tissue component of the caprine arthritis-encephalitis syndrome. Am J Pathol 100: 443–454, 1980. 320. Wilkerson MJ, Davis WC, Baszler TV, et al: Immunopathology of chronic lentivirus-induced arthritis. Am J Pathol 146: 1433–1443, 1995. 321. Hanson J, Hydbring E, Olsson K: A long term study of goats naturally infected with caprine arthritis-encephalitis virus. Acta Vet Scand 37: 31–39, 1996. 322. Pedersen NC, Castles JJ, Weisner K: Noninfectious canine arthritis: rheumatoid arthritis. J Am Vet Med Assoc 169: 295–303, 1976. 323. Hang L, Theofilopoulos AN, Dixon FJ: A spontaneous rheumatoid arthritislike disease in MRL/l mice. J Exp Med 155: 1690–1701, 1982. 324. Bywaters EG: Observations on chronic polyarthritis in monkeys. J R Soc Med 74: 794–799, 1981. 325. Schumacher HR, Newton C, Halliwell RE: Synovial pathologic changes in spontaneous canine rheumatoid-like arthritis. Arthritis Rheum 23: 412–423, 1980. 326. De Benedetti F, Meazza C, Oliveri M, et al: Effect of IL-6 on IGF binding protein-3: a study in IL-6 transgenic mice and in patients with systemic juvenile idiopathic arthritis. Endocrinology 142: 4818–4826, 2001. 327. Li P, Schwarz EM, O’Keefe RJ, et al: Systemic tumor necrosis factor alpha mediates an increase in peripheral CD11bhigh osteoclast precursors in tumor necrosis factor alpha-transgenic mice. Arthritis Rheum 50: 265–276, 2004. 328. Zwerina J, Hayer S, Tohidast-Akrad M, et al: Single and combined inhibition of tumor necrosis factor, interleukin-1, and RANKL pathways in tumor necrosis factor-induced arthritis: effects on synovial inflammation, bone erosion, and cartilage destruction. Arthritis Rheum 50: 277–290, 2004. 329. Howard JF, Sigsbee A, Glass DN: HLA genetics and inherited predisposition to JRA. J Rheumatol 12: 7–12, 1985. 330. Moroldo MB, Tague BL, Shear ES, et al: Juvenile rheumatoid arthritis in affected sibpairs. Arthritis Rheum 40: 1962–1966, 1997. 331. Rosenberg AM, Petty RE: Similar patterns of juvenile rheumatoid arthritis within families. Arthritis Rheum 23: 951–953, 1980. 332. Saila H, Savolainen A, Kauppi M, et al: Occurrence of chronic inflammatory rheumatic diseases among parents of multiple offspring affected by juvenile idiopathic arthritis. Clin Exp Rheumatol 21: 263–265, 2003. 333. Prahalad S, Fraser AM, O’Brien E, et al: Lack of association between birth order and juvenile idiopathic arthritis. Arthritis Rheum 48: 2989–2990, 2003. 334. Husby G, Williams RC Jr, Tung KS, et al: Immunologic studies in identical twins concordant for juvenile rheumatoid arthritis but discordant for monoclonal gammopathy and amyloidosis. J Lab Clin Med 111: 307–314, 1988. 335. Prahalad S, Ryan MH, Shear ES, et al: Twins concordant for juvenile rheumatoid arthritis. Arthritis Rheum 43: 2611–2612, 2000.
336. Prahalad S, Ryan MH, Shear ES, et al: Juvenile rheumatoid arthritis: linkage to HLA demonstrated by allele sharing in affected sibpairs. Arthritis Rheum 43: 2335–2338, 2000. 337. Nepom B: The immunogenetics of juvenile rheumatoid arthritis. Rheum Dis Clin North Am 17: 825–842, 1991. 338. Ansell BM, Bywaters EG, Lawrence JS: Familial aggregation and twin studies in Still’s disease. Juvenile chronic polyarthritis. Rheumatology 2: 37–61, 1969. 339. Clemens LE, Albert E, Ansell BM: Sibling pairs affected by chronic arthritis of childhood: evidence for a genetic predisposition. J Rheumatol 12: 108–113, 1985. 340. Prahalad S, Shear ES, Thompson SD, et al: Increased prevalence of familial autoimmunity in simplex and multiplex families with juvenile rheumatoid arthritis. Arthritis Rheum 46: 1851–1856, 2002. 341. Rossen RD, Brewer EJ, Sharp RM, et al: Familial rheumatoid arthritis: linkage of HLA to disease susceptibility locus in four families where proband presented with juvenile rheumatoid arthritis. J Clin Invest 65: 629–642, 1980. 342. Southwood TR, Roberts-Thomson PJ, Ahern MJ, et al: Autoantibodies in patients with juvenile chronic arthritis and their immediate family relatives. Ann Rheum Dis 49: 968–972, 1990. 343. Hall A, Ansell BM, James DCO, et al: HL-A antigens in juvenile chronic polyarthritis (Still’s disease). Ann Rheum Dis Suppl 34: 36, 1975. 344. Gershwin ME, Opelz G, Terasaki PI, et al: Frequency of HLA-Dw3 in juvenile rheumatoid arthritis. Tissue Antigens 10: 330–336, 1977. 345. Stastny P, Fink CW: Different HLA-D associations in adult and juvenile rheumatoid arthritis. J Clin Invest 63: 124–130, 1979. 346. Glass D, Litvin D, Wallace K, et al: Early-onset pauciarticular juvenile rheumatoid arthritis associated with human leukocyte antigen-DRw5, iritis, and antinuclear antibody. J Clin Invest 66: 426–429, 1980. 347. Suciu-Foca N, Godfrey M, Jacobs J, et al: Increased frequency of DRw5 in pauciarticular juvenile rheumatoid arthritis. In Terasaki P (ed): Histocompatibility Testing 1980. Los Angeles, University of California Press, 1980, p 953. 348. Glass DN, Litvin DA: Heterogeneity of HLA associations in systemic onset juvenile rheumatoid arthritis. Arthritis Rheum 23: 796–799, 1980. 349. Forre O, Dobloug JH, Hoyeraal HM, et al: HLA antigens in juvenile arthritis. Genetic basis for the different subtypes. Arthritis Rheum 26: 35–38, 1983. 350. Ansell BM, Albert ED: Juvenile chronic arthritis, pauciarticular type. In Albert ED, Baur MR, Mayr WR (eds): Histocompatibility Testing 1984. New York, Springer-Verlag, 1984, p 368. 351. Moore TL, Oldfather JW, Osborn TG, et al: HLA antigens in black and white patients with juvenile arthritis: associations with rheumatoid factor, hidden rheumatoid factor, antinuclear antibodies, and immune complex levels. J Rheumatol 11: 188–196, 1984. 352. Nepom BS, Nepom GT, Mickelson E, et al: Specific HLA-DR4-associated histocompatibility molecules characterize patients with seropositive juvenile rheumatoid arthritis. J Clin Invest 74: 287–291, 1984. 353. Morling N, Friis J, Heilmann C, et al: HLA antigen frequencies in juvenile chronic arthritis. Scand J Rheumatol 14: 209–216, 1985. 354. Sher MR, Schultz JS, Ragsdale CG, et al: HLA-DR and MT associations with the clinical and serologic manifestations of pauciarticular onset juvenile rheumatoid arthritis. J Rheumatol 12: 114–118, 1985. 355. Miller ML, Aaron S, Jackson J, et al: HLA gene frequencies in children and adults with systemic onset juvenile rheumatoid arthritis. Arthritis Rheum 28: 146–150, 1985. 356. Brautbar C, Mukamel M, Yaron M, et al: Immunogenetics of juvenile chronic arthritis in Israel. J Rheumatol 13: 1072–1075, 1986. 357. Manners PJ, Dawkins RL, McCluskey J, et al: An immunogenetic study of juvenile chronic arthritis. Aust Paediatr J 22: 317–321, 1986. 358. Hoffman RW, Shaw S, Francis LC, et al: HLA-DP antigens in patients with pauciarticular juvenile rheumatoid arthritis. Arthritis Rheum 29: 1057–1062, 1986. 359. Nepom BS, Palmer J, Kim SJ, et al: Specific genomic markers for the HLADQ subregion discriminate between DR4+ insulin-dependent diabetes mellitus and DR4+ seropositive juvenile rheumatoid arthritis. J Exp Med 164: 345–350, 1986. 360. Stastny P: HLA and the role of T cells in the predisposition to disease. Rheum Dis Clin North Am 13: 1–6, 1987. 361. Fernandez-Vina MA, Fink CW, Stastny P: HLA antigens in juvenile arthritis. Pauciarticular and polyarticular juvenile arthritis are immunogenetically distinct. Arthritis Rheum 33: 1787–1794, 1990. 362. De Inocencio J, Giannini EH, Glass DN: Can genetic markers contribute to the classification of juvenile rheumatoid arthritis? J Rheumatol Suppl 40: 12–18, 1993. 363. Haas JP, Nevinny-Stickel C, Schoenwald U, et al: Susceptible and protective major histocompatibility complex class II alleles in early-onset pauciarticular juvenile chronic arthritis. Hum Immunol 41: 225–233, 1994. 364. Fink CW, Fernandez-Vina M, Stastny P: Clinical and genetic evidence that juvenile arthritis is not a single disease. Pediatr Clin North Am 42: 1155–1169, 1995. 365. Ploski R: Immunogenetic polymorphism and disease mechanisms in juvenile chronic arthritis. Rev Rhum Engl Ed 64: 127S–130S, 1997. 366. Pratsidou-Gertsi P, Kanakoudi-Tsakalidou F, Spyropoulou M, et al: Nationwide collaborative study of HLA class II associations with distinct types of juvenile chronic arthritis (JCA) in Greece. Eur J Immunogenet 26: 299–310, 1999.
C H A P T E R 367. Sanjeevi CB, Miller EN, Dabadghao P, et al: Polymorphism at NRAMP1 and D2S1471 loci associated with juvenile rheumatoid arthritis. Arthritis Rheum 43: 1397–1404, 2000. 368. Thomas E, Barrett JH, Donn RP, et al: Subtyping of juvenile idiopathic arthritis using latent class analysis. British Paediatric Rheumatology Group. Arthritis Rheum 43: 1496–1503, 2000. 369. Zeggini E, Donn RP, Ollier WE, et al: Evidence for linkage of HLA loci in juvenile idiopathic oligoarthritis: independent effects of HLA-A and HLADRB1. Arthritis Rheum 46: 2716–2720, 2002. 370. Smerdel A, Ploski R, Flato B, et al: Juvenile idiopathic arthritis (JIA) is primarily associated with HLA-DR8 but not DQ4 on the DR8-DQ4 haplotype. Ann Rheum Dis 61: 354–357, 2002. 371. Alsaeid K, Haider MZ, Kamal H, et al: Prevalence of human leukocyte antigen (HLA) DRB1 alleles in Kuwaiti children with juvenile rheumatoid arthritis. Eur J Immunogenet 29: 1–5, 2002. 372. Alsaeid KM, Haider MZ, al Awadhi AM, et al: Role of human leukocyte antigen DRB1*0307 and DRB1*0308 in susceptibility to juvenile rheumatoid arthritis. Clin Exp Rheumatol 21: 399–402, 2003. 373. Jacobs JC, Berdon WE, Johnston AD: HLA-B27-associated spondyloarthritis and enthesopathy in childhood: clinical, pathologic, and radiographic observations in 58 patients. J Pediatr 100: 521–528, 1982. 374. Sheerin KA, Giannini EH, Brewer EJ Jr, et al: HLA-B27-associated arthropathy in childhood: long-term clinical and diagnostic outcome. Arthritis Rheum 31: 1165–1170, 1988. 375. Hall PJ, Burman SJ, Laurent MR, et al: Genetic susceptibility to early onset pauciarticular juvenile chronic arthritis: a study of HLA and complement markers in 158 British patients. Ann Rheum Dis 45: 464–474, 1986. 376. Arnaiz-Villena A, Gomez-Reino JJ, Gamir ML, et al: DR, C4, and Bf allotypes in juvenile rheumatoid arthritis. Arthritis Rheum 27: 1281–1285, 1984. 377. Vicario JL, Martinez-Laso J, Gomez-Reino JJ, et al: Both HLA class II and class III DNA polymorphisms are linked to juvenile rheumatoid arthritis susceptibility. Clin Immunol Immunopathol 56: 22–28, 1990. 378. Petty RE, Hunt DW, Rollins DF, et al: Immunity to soluble retinal antigen in patients with uveitis accompanying juvenile rheumatoid arthritis. Arthritis Rheum 30: 287–293, 1987. 379. Paul C, Schoenwald U, Truckenbrodt H, et al: HLA-DP/DR interaction in early onset pauciarticular juvenile chronic arthritis. Immunogenetics 37: 442–448, 1993. 380. Moroldo MB, Donnelly P, Saunders J, et al: Transmission disequilibrium as a test of linkage and association between HLA alleles and pauciarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 41: 1620–1624, 1998. 381. Smerdel A, Lie BA, Ploski R, et al: A gene in the telomeric HLA complex distinct from HLA-A is involved in predisposition to juvenile idiopathic arthritis. Arthritis Rheum 46: 1614–1619, 2002. 382. Murray KJ, Moroldo MB, Donnelly P, et al: Age-specific effects of juvenile rheumatoid arthritis-associated HLA alleles. Arthritis Rheum 42: 1843–1853, 1999. 383. Wordsworth P: Genes in the spondyloarthropathies. Rheum Dis Clin North Am 24: 845–863, 1998. 384. Thomson W, Barrett JH, Donn R, et al: Juvenile idiopathic arthritis classified by the ILAR criteria: HLA associations in UK patients. Rheumatology (Oxf) 41: 1183–1189, 2002. 385. Nepom BS, Glass DN: Juvenile rheumatoid arthritis and HLA: report of the Park City III workshop. J Rheumatol Suppl 33: 70–74, 1992. 386. Rosen P, Thompson S, Glass D: Non-HLA gene polymorphisms in juvenile rheumatoid arthritis. Clin Exp Rheumatol 21: 650–656, 2003. 387. Donn RP, Davies EJ, Holt PL, et al: Increased frequency of TAP2B in early onset pauciarticular juvenile chronic arthritis. Ann Rheum Dis 53: 261–264, 1994. 388. Ploski R, Undlien DE, Vinje O, et al: Polymorphism of human major histocompatibility complex-encoded transporter associated with antigen processing (TAP) genes and susceptibility to juvenile rheumatoid arthritis. Hum Immunol 39: 54–60, 1994. 389. Ploski R, McDowell TL, Symons JA, et al: Interaction between HLA-DR and HLA-DP, and between HLA and interleukin 1 alpha in juvenile rheumatoid arthritis indicates heterogeneity of pathogenic mechanisms of the disease. Hum Immunol 42: 343–347, 1995. 390. McDowell TL, Symons JA, Ploski R, et al: A genetic association between juvenile rheumatoid arthritis and a novel interleukin-1 alpha polymorphism. Arthritis Rheum 38: 221–228, 1995. 391. Pryhuber KG, Murray KJ, Donnelly P, et al: Polymorphism in the LMP2 gene influences disease susceptibility and severity in HLA-B27 associated juvenile rheumatoid arthritis. J Rheumatol 23: 747–752, 1996. 392. Prahalad S, Kingsbury DJ, Griffin TA, et al: Polymorphism in the MHCencoded LMP7 gene: association with JRA without functional significance for immunoproteasome assembly. J Rheumatol 28: 2320–2325, 2001. 393. Falcini F, Simonini G, Cimaz R, et al: Osteoprotegerin serum levels and osteoprotegerin polymorphism in juvenile idiopathic arthritis: correlations with articular erosions and low bone mass. Arthritis Rheum 46 (Suppl): S579, 2002. 394. Ihnat DH, McIlvain-Simpson G, Conard K, et al: Inflammatory arthropathies in children with chromosomal abnormalities. J Rheumatol 20: 742–746, 1993. 395. Rudolf MC, Genel M, Tamborlane WV Jr, et al: Juvenile rheumatoid arthritis in children with diabetes mellitus. J Pediatr 99: 519–524, 1981. 396. Agrawal S, Desai MP: Simultaneous occurrence of type I diabetes mellitus and juvenile rheumatoid arthritis. Indian Pediatr 40: 568–571, 2003.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
251
397. Mihailova D, Grigorova R, Vassileva B, et al: Autoimmune thyroid disorders in juvenile chronic arthritis and systemic lupus erythematosus. Adv Exp Med Biol 455: 55–60, 1999. 398. Rosenbloom AL, Silverstein JH, Lezotte DC, et al: Limited joint mobility in childhood diabetes mellitus indicates increased risk for microvascular disease. N Engl J Med 305: 191–194, 1981. 399. Sherry DD, Rothstein RR, Petty RE: Joint contractures preceding insulindependent diabetes mellitus. Arthritis Rheum 25: 1362–1364, 1982. 400. Glass JB, Sher PK, Lennon VA, et al: The association of pauciarticular juvenile arthritis and myasthenia gravis. Am J Dis Child 145: 1176–1180, 1991. 401. Cassidy JT, Petty RE, Sullivan DB: Occurence of selective IgA deficiency in children with juvenile rheumatoid arthritis. Arthritis Rheum 20: 181–183, 1977. 402. Sullivan KE, McDonald-McGinn DM, Driscoll DA, et al: Juvenile rheumatoid arthritis-like polyarthritis in chromosome 22q11.2 deletion syndrome (DiGeorge anomalad/velocardiofacial syndrome/conotruncal anomaly face syndrome). Arthritis Rheum 40: 430–436, 1997. 403. Petty RE, Malleson P, Kalousek DK: Chronic arthritis in two children with partial deletion of chromosome 18. J Rheumatol 14: 586–587, 1987. 404. Verloes A, Curry C, Jamar M, et al: Juvenile rheumatoid arthritis and del(22q11) syndrome: a non-random association. J Med Genet 35: 943–947, 1998. 405. Zori RT, Boyar FZ, Williams WN, et al: Prevalence of 22q11 region deletions in patients with velopharyngeal insufficiency. Am J Med Genet 77: 8–11, 1998. 406. Davies K, Stiehm ER, Woo P, et al: Juvenile idiopathic polyarticular arthritis and IgA deficiency in the 22q11 deletion syndrome. J Rheumatol 28: 2326–2334, 2001. 407. Wihlborg CE, Babyn PS, Schneider R: The association between Turner’s syndrome and juvenile rheumatoid arthritis. Pediatr Radiol 29: 676–681, 1999. 408. Zulian F, Schumacher HR, Calore A, et al: Juvenile arthritis in Turner’s syndrome: a multicenter study. Clin Exp Rheumatol 16: 489–494, 1998. 409. Zamir G, Press J, Tal A, et al: Sleep fragmentation in children with juvenile rheumatoid arthritis. J Rheumatol 25: 1191–1197, 1998. 410. Bloom BJ, Owens JA, McGuinn M, et al: Sleep and its relationship to pain, dysfunction, and disease activity in juvenile rheumatoid arthritis. J Rheumatol 29: 169–173, 2002. 411. Hagglund KJ, Schopp LM, Alberts KR, et al: Predicting pain among children with juvenile rheumatoid arthritis. Arthritis Care Res 8: 36–42, 1995. 412. Kuis W, Heijnen CJ, Hogeweg JA, et al: How painful is juvenile chronic arthritis? Arch Dis Child 77: 451–453, 1997. 413. Cabral DA, Tucker LB: Malignancies in children who initially present with rheumatic complaints. J Pediatr 134: 53–57, 1999. 414. Ross DM, Ross SA: Childhood pain: The school-aged child’s viewpoint. Pain 20: 179–191, 1984. 415. Vandvik IH, Eckblad G: Relationship between pain, disease severity and psychosocial function in patients with juvenile chronic arthritis (JCA). Scand J Rheumatol 19: 295–302, 1990. 416. Walco GA, Varni JW, Ilowite NT: Cognitive-behavioral pain management in children with juvenile rheumatoid arthritis. Pediatrics 89: 1075–1079, 1992. 417. Sallfors C, Hallberg LR, Fasth A: Gender and age differences in pain, coping and health status among children with chronic arthritis. Clin Exp Rheumatol 21: 785–793, 2003. 418. Malleson PN, Oen K, Cabral DA, et al: Predictors of pain in children with established juvenile rheumatoid arthritis. Arthritis Rheum 51: 222–227, 2004. 419. Beales JG, Keen JH, Holt PJ: The child’s perception of the disease and the experience of pain in juvenile chronic arthritis. J Rheumatol 10: 61–65, 1983. 420. Lovell DJ, Athreya B, Emery HM, et al: School attendance and pattern: Special services and special needs in pediatric patients with rheumatic diseases. Arthritis Care Res 3: 196, 1990. 421. Kuis W, Heijnen CJ, Sinnema G, et al: Pain in childhood rheumatic arthritis. Baillieres Clin Rheumatol 12: 229–244, 1998. 422. Laaksonen AL, Laine V: A comparative study of joint pain in adult and juvenile rheumatoid arthritis. Ann Rheum Dis 20: 386, 1961. 423. Scott PJ, Ansell BM, Huskisson EC: Measurement of pain in juvenile chronic polyarthritis. Ann Rheum Dis 36: 186–187, 1977. 424. Thastum M, Zachariae R, Scholer M, et al: Cold pressor pain: comparing responses of juvenile arthritis patients and their parents. Scand J Rheumatol 26: 272–279, 1997. 425. Sherry DD, Bohnsack J, Salmonson K, et al: Painless juvenile rheumatoid arthritis. J Pediatr 116: 921–923, 1990. 426. Thompson KL, Varni JW, Hanson V: Comprehensive assessment of pain in juvenile rheumatoid arthritis: an empirical model. J Pediatr Psychol 12: 241–255, 1987. 427. McGhee JL, Burks FN, Sheckels JL, et al: Identifying children with chronic arthritis based on chief complaints: absence of predictive value for musculoskeletal pain as an indicator of rheumatic disease in children. Pediatrics 110: 354–359, 2002. 428. Ilowite NT, Walco GA, Pochaczevsky R: Assessment of pain in patients with juvenile rheumatoid arthritis: relation between pain intensity and degree of joint inflammation. Ann Rheum Dis 51: 343–346, 1992. 429. Varni JW, Thompson KL, Hanson V: The Varni/Thompson Pediatric Pain Questionnaire. I. Chronic musculoskeletal pain in juvenile rheumatoid arthritis. Pain 28: 27–38, 1987.
252
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
430. Ross CK, Lavigne JV, Hayford JR, et al: Psychological factors affecting reported pain in juvenile rheumatoid arthritis. J Pediatr Psychol 18: 561–573, 1993. 431. Power TG, Dahlquist LM, Thompson SM, et al: Interactions between children with juvenile rheumatoid arthritis and their mothers. J Pediatr Psychol 28: 213–221, 2003. 432. Malleson P, Riding K, Petty R: Stridor due to cricoarytenoid arthritis in pauciarticular onset juvenile rheumatoid arthritis. J Rheumatol 13: 952–953, 1986. 433. Siamopoulou-Mavridou A, Asimakopoulos D, Mavridis A, et al: Middle ear function in patients with juvenile chronic arthritis. Ann Rheum Dis 49: 620–623, 1990. 434. Uziel Y, Rathaus V, Pomeranz A, et al: Torticollis as the sole initial presenting sign of systemic onset juvenile rheumatoid arthritis. J Rheumatol 25: 166–168, 1998. 435. Rombouts JJ, Rombouts-Lindemans C: Scoliosis in juvenile rheumatoid arthritis. J Bone Joint Surg Br 56: 478–483, 1974. 436. Ross AC, Edgar MA, Swann M, et al: Scoliosis in juvenile chronic arthritis. J Bone Joint Surg Br 69: 175–178, 1987. 437. Martel W, Holt JF, Cassidy JT: Roentgenologic manifestations of juvenile rheumatoid arthritis. AJR Am J Roentgenol 88: 400, 1962. 438. Ladd JR, Cassidy JT, Martel W: Juvenile ankylosing spondylitis. Arthritis Rheum 14: 579–590, 1971. 439. Bloom BJ, Tucker LB, Miller LC, et al: Bicipital synovial cysts in juvenile rheumatoid arthritis: clinical description and sonographic correlation. J Rheumatol 22: 1953–1955, 1995. 440. Rennebohm RM, Towbin RB, Crowe WE, et al: Popliteal cysts in juvenile rheumatoid arthritis. AJR Am J Roentgenol 140: 123–125, 1983. 441. Dinham JM: Popliteal cysts in children. The case against surgery. J Bone Joint Surg Br 57: 69–71, 1975. 442. Sherry DD, Sapp LR: Enthesalgia in childhood: site-specific tenderness in healthy subjects and in patients with seronegative enthesopathic arthropathy. J Rheumatol 30: 1335–1340, 2003. 443. Wang FM, Wertenbaker C, Behrens MM, et al: Acquired Brown’s syndrome in children with juvenile rheumatoid arthritis. Ophthalmology 91: 23–26, 1984. 444. Moore AT, Morin JD: Bilateral acquired inflammatory Brown’s syndrome. J Pediatr Ophthalmol Strabismus 22: 26–30, 1985. 445. Read RW, Weiss AH, Sherry DD: Episcleritis in childhood. Ophthalmology 106: 2377–2379, 1999. 446. Jain V, Singh S, Sharma A: Keratonconjunctivitis sicca is not uncommon in children with juvenile rheumatoid arthritis. Rheumatol Int 20: 159–162, 2001. 447. Saha MT, Verronen P, Laippala P, et al: Growth of prepubertal children with juvenile chronic arthritis. Acta Paediatr 88: 724–728, 1999. 448. Rusconi R, Corona F, Grassi A, et al: Age at menarche in juvenile rheumatoid arthritis. J Pediatr Endocrinol Metab 16 Suppl 2: 285–288, 2003. 449. Davies UM, Jones J, Reeve J, et al: Juvenile rheumatoid arthritis. Effects of disease activity and recombinant human growth hormone on insulin-like growth factor 1, insulin-like growth factor binding proteins 1 and 3, and osteocalcin. Arthritis Rheum 40: 332–340, 1997. 450. De Benedetti F, Alonzi T, Moretta A, et al: Interleukin 6 causes growth impairment in transgenic mice through a decrease in insulin-like growth factor-I. A model for stunted growth in children with chronic inflammation. J Clin Invest 99: 643–650, 1997. 451. Ansell BM, Bywaters EGL: Growth in Still’s disease. Ann Rheum Dis 15: 295, 1956. 452. Bernstein BH, Stobie D, Singsen BH, et al: Growth retardation in juvenile rheumatoid arthritis (JRA). Arthritis Rheum 20: 212–216, 1977. 453. Bacon MC, White P: A new approach to the assessment of growth in JRA. Arthritis Rheum 30: S132, 1987. 454. Lovell DJ, White PH: Growth and nutrition in juvenile rheumatoid arthritis. In Woo P, White PH, Ansell BM (eds): Paediatric Rheumatology Update. Oxford, Oxford University Press, 1990. 455. Garcia-Consuegra MJ, Merino MR, Lama MR, et al: Growth in children with juvenile idiopathic arthritis. Ann Pediatr (Barc) 58: 529–537, 2003. 456. Simon D, Fernando C, Czernichow P, et al: Linear growth and final height in patients with systemic juvenile idiopathic arthritis treated with longterm glucocorticoids. J Rheumatol 29: 1296–1300, 2002. 457. Allen DB, Julius JR, Breen TJ, et al: Treatment of glucocorticoid-induced growth suppression with growth hormone. National Cooperative Growth Study. J Clin Endocrinol Metab 83: 2824–2829, 1998. 458. Laaksonen AL, Sunell JE, Westeren H, et al: Adrenocortical function in children with juvenile rheumatoid arthritis and other connective tissue disorders. Scand J Rheumatol 3: 137–142, 1974. 459. Olson L, Eckerdal O, Hallonsten AL, et al: Craniomandibular function in juvenile chronic arthritis. A clinical and radiographic study. Swed Dent J 15: 71–83, 1991. 460. Ronchezel MV, Hilario MO, Goldenberg J, et al: Temporomandibular joint and mandibular growth alterations in patients with juvenile rheumatoid arthritis. J Rheumatol 22: 1956–1961, 1995. 461. Avrahami E, Segal R, Solomon A, et al: Direct coronal high resolution computed tomography of the temporomandibular joints in patients with rheumatoid arthritis. J Rheumatol 16: 298–301, 1989. 462. Bjork A, Skieller V: Contrasting mandibular growth and facial development in long face syndrome, juvenile rheumatoid polyarthritis, and mandibulofacial dysostosis. J Craniofac Genet Dev Biol Suppl 1: 127–138, 1985.
463. Ganik R, Williams FA: Diagnosis and management of juvenile rheumatoid arthritis with TMJ involvement. Cranio 4: 254–262, 1986. 464. Stabrun AE, Larheim TA, Rosler M, et al: Impaired mandibular function and its possible effect on mandibular growth in juvenile rheumatoid arthritis. Eur J Orthod 9: 43–50, 1987. 465. Walton AG, Welbury RR, Foster HE, et al: Juvenile chronic arthritis: a dental review. Oral Dis 5: 68–75, 1999. 466. Hillman L, Cassidy JT, Johnson L, et al: Vitamin D metabolism and bone mineralization in children with juvenile rheumatoid arthritis. J Pediatr 124: 910–916, 1994. 467. Woo PM: Growth retardation and osteoporosis in juvenile chronic arthritis. Clin Exp Rheumatol 12 Suppl 10: S87–S90, 1994. 468. Cassidy JT, Langman CB, Allen SH, et al: Bone mineral metabolism in children with juvenile rheumatoid arthritis. Pediatr Clin North Am 42: 1017–1033, 1995. 469. Cassidy JT, Hillman LS: Abnormalities in skeletal growth in children with juvenile rheumatoid arthritis. Rheum Dis Clin North Am 23: 499–522, 1997. 470. Henderson CJ, Cawkwell GD, Specker BL, et al: Predictors of total body bone mineral density in non-corticosteroid-treated prepubertal children with juvenile rheumatoid arthritis. Arthritis Rheum 40: 1967–1975, 1997. 471. Kotaniemi A, Savolainen A, Kroger H, et al: Development of bone mineral density at the lumbar spine and femoral neck in juvenile chronic arthritis— a prospective one year followup study. J Rheumatol 25: 2450–2455, 1998. 472. Falcini F, Ermini M, Bagnoli F: Bone turnover is reduced in children with juvenile rheumatoid arthritis. J Endocrinol Invest 21: 31–36, 1998. 473. Cimaz R, Biggioggero M: Osteoporosis. Curr Rheumatol Rep 3: 365–370, 2001. 474. French AR, Mason T, Nelson AM, et al: Osteopenia in adults with a history of juvenile rheumatoid arthritis. A population based study. J Rheumatol 29: 1065–1070, 2002. 475. Zak M, Hassager C, Lovell DJ, et al: Assessment of bone mineral density in adults with a history of juvenile chronic arthritis: a cross-sectional long-term follow-up study. Arthritis Rheum 42: 790–798, 1999. 476. Henderson CJ, Specker BL, Sierra RI, et al: Total-body bone mineral content in non-corticosteroid-treated postpubertal females with juvenile rheumatoid arthritis: frequency of osteopenia and contributing factors. Arthritis Rheum 43: 531–540, 2000. 477. Haugen M, Lien G, Flato B, et al: Young adults with juvenile arthritis in remission attain normal peak bone mass at the lumbar spine and forearm. Arthritis Rheum 43: 1504–1510, 2000. 478. Cassidy JT: Osteopenia and osteoporosis in children. Clin Exp Rheumatol 17: 245–250, 1999. 479. Masi L, Cimaz R, Simonini G, et al: Association of low bone mass with vitamin D receptor gene and calcitonin receptor gene polymorphisms in juvenile idiopathic arthritis. J Rheumatol 29: 2225–2231, 2002. 480. Gedalia A, Gewanter H, Baum J: Dark skin discoloration of finger joints in juvenile arthritis. J Rheumatol 16: 797–799, 1989. 481. Athreya BH, Ostrov BE, Eichenfield AH, et al: Lymphedema associated with juvenile rheumatoid arthritis. J Rheumatol 16: 1338–1340, 1989. 482. Bardare M, Falcini F, Hertzberger-ten Cate R, et al: Idiopathic limb edema in children with chronic arthritis: a multicenter report of 12 cases. J Rheumatol 24: 384–388, 1997. 483. Schmit P, Prieur AM, Brunelle F: Juvenile rheumatoid arthritis and lymphoedema: lymphangiographic aspects. Pediatr Radiol 29: 364–366, 1999. 484. Ellsworth JE, Cassidy JT, Ragsdale CG, et al: Mucha-Habermann disease in children—the association with rheumatic diseases. J Rheumatol 9: 319–324, 1982. 485. Person DA, He XH, Brewer EJ: Vasculitis in children with juvenile rheumatoid arthritis. J Rheumatol 13: 219, 1986. 486. Hall S, Nelson AM: Takayasu’s arteritis and juvenile rheumatoid arthritis. J Rheumatol 13: 431–433, 1986. 487. Wedderburn LR, Kwan JT, Thompson PW, et al: Juvenile chronic arthritis and Wegener’s granulomatosis. Br J Rheumatol 31: 121–123, 1992. 488. Cullen B, Mongey AB, Molony J: Juvenile chronic arthritis associated with systemic vasculitis. Ir Med J 79: 72–74, 1986. 489. Vostrejs M, Hollister JR: Muscle atrophy and leg length discrepancies in pauciarticular juvenile rheumatoid arthritis. Am J Dis Child 142: 343–345, 1988. 490. Goldenberg J, Ferraz MB, Pessoa AP, et al: Symptomatic cardiac involvement in juvenile rheumatoid arthritis. Int J Cardiol 34: 57–62, 1992. 491. Lietman PS, Bywaters EGL: Pericarditis in juvenile rheumatoid arthritis. Pediatrics 32: 855, 1963. 492. Oguz D, Ocal B, Ertan U, et al: Left ventricular diastolic functions in juvenile rheumatoid arthritis. Pediatr Cardiol 21: 374–377, 2000. 493. Miller JJ III, French JW: Myocarditis in juvenile rheumatoid arthritis. Am J Dis Child 131: 205–209, 1977. 494. Bultink IE, Lems WF, Dijkmans BA, et al: Severe aortic regurgitation in RF positive polyarticular JIA. Ann Rheum Dis 61: 282–283, 2002. 495. Athreya BH, Doughty RA, Bookspan M, et al: Pulmonary manifestations of juvenile rheumatoid arthritis. A report of eight cases and review. Clin Chest Med 1: 361–374, 1980. 496. Lovell D, Lindsley C, Langston C: Lymphoid interstitial pneumonia in juvenile rheumatoid arthritis. J Pediatr 105: 947–950, 1984. 497. Uziel Y, Hen B, Cordoba M, et al: Lymphocytic interstitial pneumonitis preceding polyarticular juvenile rheumatoid arthritis. Clin Exp Rheumatol 16: 617–619, 1998.
C H A P T E R 498. Brinkman GL, Chaikof L: Rheumatoid lung disease. Am Rev Respir Dis 80: 732, 1959. 499. Padeh S, Laxer RM, Silver MM, et al: Primary pulmonary hypertension in a patient with systemic-onset juvenile arthritis. Arthritis Rheum 34: 1575–1579, 1991. 500. Chu SH, Shyur SD, Peng YH, et al: Juvenile idiopathic arthritis with pulmonary hemosiderosis: a case report. J Microbiol Immunol Infect 35: 133–135, 2002. 501. Wagener JS, Taussig LM, DeBenedetti C, et al: Pulmonary function in juvenile rheumatoid arthritis. J Pediatr 99: 108–110, 1981. 502. Knook LM, De Kleer IM, Van der Ent CK, et al: Lung function abnormalities and respiratory muscle weakness in children with juvenile chronic arthritis. Eur Respir J 14: 529–533, 1999. 503. Snape WJ Jr: Pseudo-obstruction and other obstructive disorders. Clin Gastroenterol 11: 593–608, 1982. 504. Bhettay E, Thomson AJ: Peritonitis in juvenile chronic arthritis. A report of 2 cases. S Afr Med J 68: 605–606, 1985. 505. Rosenberg AM, Mitchell DM, Card RT: Felty’s syndrome in a child. J Rheumatol 11: 835–837, 1984. 506. Toomey K, Hepburn B: Felty syndrome in juvenile arthritis. J Pediatr 106: 254–255, 1985. 507. Lockitch G, Pudek MR, Halstead AC: Isolated elevation of serum alkaline phosphatase. J Pediatr 105: 773–775, 1984. 508. Jacobs JC: JRA and hyperphosphataemia. J Pediatr 107: 828–829, 1985. 509. Rennebohm RM, Heubi JE, Daugherty CC, et al: Reye syndrome in children receiving salicylate therapy for connective tissue disease. J Pediatr 107: 877–880, 1985. 510. Falcini F, Taccetti G, Montanelli F, et al: Benign intracranial hypertension during treatment with acetylsalicylic acid: presentation of 2 cases in children. Pediatr Med Chir 11: 319–321, 1989. 511. Gururaj AK, Chand RP, Chuah SP: Cerebral infarction in juvenile rheumatoid arthritis. Clin Neurol Neurosurg 90: 261–263, 1988. 512. Gedalia A, Mendez EA, Craver R, et al: Renal involvement in juvenile rheumatoid arthritis: report of two cases. Clin Rheumatol 20: 153–156, 2001. 513. Anttila R, Laaksonen AL: Renal disease in juvenile rheumatoid arthritis. Acta Rheumatol Scand 15: 99–111, 1969. 514. Anttila R: Renal involvement in juvenile rheumatoid arthritis. A clinical and histopathological study. Acta Paediatr Scand Suppl 227: 3–73, 1972. 515. Malleson PN, Lockitch G, Mackinnon M, et al: Renal disease in chronic arthritis of childhood. A study of urinary N-acetyl-beta-glucosaminidase and beta 2-microglobulin excretion. Arthritis Rheum 33: 1560–1566, 1990. 516. Cassidy JT, Brody GL, Martel W: Monarticular juvenile rheumatoid arthritis. J Pediatr 70: 867–875, 1967. 517. Fletcher MR, Scott JT: Chronic monarticular synovitis. Diagnostic and prognostic features. Ann Rheum Dis 34: 171–176, 1975. 518. Roy CN, Weinstein DA, Andrews NC: 2002 E. Mead Johnson Award for Research in Pediatrics Lecture: the molecular biology of the anemia of chronic disease: a hypothesis. Pediatr Res 53: 507–512, 2003. 519. Fantini F, Gattinara M, Gerloni V, et al: Severe anemia associated with active systemic-onset juvenile rheumatoid arthritis successfully treated with recombinant human erythropoietin: a pilot study. Arthritis Rheum 35: 724–726, 1992. 520. Cazzola M, Ponchio L, De Benedetti F, et al: Defective iron supply for erythropoiesis and adequate endogenous erythropoietin production in the anemia associated with systemic-onset juvenile chronic arthritis. Blood 87: 4824–4830, 1996. 521. Pelkonen P, Swanljung K, Siimes MA: Ferritinemia as an indicator of systemic disease activity in children with systemic juvenile rheumatoid arthritis. Acta Paediatr Scand 75: 64–68, 1986. 522. Ewer AK, Darbyshire PJ, Southwood TR: Systemic-onset juvenile chronic arthritis and bone marrow hypoplasia. Br J Rheumatol 32: 78–80, 1993. 523. Prieur AM, Fischer A, Griscelli C: Still’s disease and haemophagocytic syndrome. Ann Rheum Dis 44: 806, 1985. 524. Grom AA, Passo M: Macrophage activation syndrome in systemic juvenile rheumatoid arthritis. J Pediatr 129: 630–632, 1996. 525. Sherry DD, Kredich DW: Transient thrombocytopenia in systemic onset juvenile rheumatoid arthritis. Pediatrics 76: 600–603, 1985. 526. Ragsdale CG, Petty RE, Cassidy JT, et al: The clinical progression of apparent juvenile rheumatoid arthritis to systemic lupus erythematosus. J Rheumatol 7: 50–55, 1980. 527. Giannini EH, Brewer EJ: Poor correlation between the erythrocyte sedimentation rate and clinical activity in juvenile rheumatoid arthritis. Clin Rheumatol 6: 197–201, 1987. 528. Cassidy JT, Petty RE, Sullivan DB: Abnormalities in the distribution of serum immunoglobulin concentrations in juvenile rheumatoid arthritis. J Clin Invest 52: 1931–1936, 1973. 529. Gutowska-Grzegorczyk G, Baum J: Serum immunoglobulin and complement interrelationships in juvenile rheumatoid arthritis. J Rheumatol 4: 179–185, 1977. 530. Gutowska-Grzegorczyk G, Baum J: Antibody levels to enteric bacteria in juvenile rheumatoid arthritis. Arthritis Rheum 20: 779–784, 1977. 531. Hall PJ, De Lange GG, Ansell BM: Immunoglobulin allotypes in families with pauciarticular-onset juvenile chronic arthritis. Tissue Antigens 25: 212–215, 1985. 532. Roberts-Thomson PJ, Shepherd K, Southwood TR, et al: Low molecular weight IgM in juvenile chronic arthritis. Arch Dis Child 63: 1453–1456, 1988.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
253
533. Bowyer S, Roettcher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group. J Rheumatol 23: 1968–1974, 1996. 534. Oen K, Schroeder M, Jacobson K, et al: Juvenile rheumatoid arthritis in a Canadian First Nations (aboriginal) population: onset subtypes and HLA associations. J Rheumatol 25: 783–790, 1998. 535. Eichenfield AH, Athreya BH, Doughty RA, et al: Utility of rheumatoid factor in the diagnosis of juvenile rheumatoid arthritis. Pediatrics 78: 480–484, 1986. 536. Massa M, De Amici M, Ravelli A, et al: Changes in the production of IgM in response to pokeweed mitogen in juvenile rheumatoid arthritis. Correlations with disease activity. Minerva Pediatr 42: 273–276, 1990. 537. Torrigiani G, Ansell BM, Chown EE, et al: Raised IgG antiglobulin factors in Still’s disease. Ann Rheum Dis 28: 424–427, 1969. 538. Munthe E: Anti-IgG and antinuclear antibodies in juvenile rheumatoid arthritis. Scand J Rheumatol 1: 161–170, 1972. 539. Florin-Christensen A, Arana RM, Morteo OG, et al: IgG, IgA, IgM, and IgD antiglobulins in juvenile rheumatoid arthritis. Ann Rheum Dis 33: 32–34, 1974. 540. Schur PH, Bianco NE, Panush RS: Antigammaglobulins in normal individuals and in patients with adult and juvenile rheumatoid arthritis and osteoarthritis. Rheumatology 6: 156–166, 1975. 541. Miller JJ III, Olds-Arroyo L, Akasaka T: Antiglobulins in juvenile rheumatoid arthritis. Arthritis Rheum 20: 729–735, 1977. 542. Moore T, Dorner RW, Zuckner J: Hidden rheumatoid factor in seronegative juvenile rheumatoid arthritis. Ann Rheum Dis 33: 255–257, 1974. 543. Moore TL, Dorner RW: 19S IgM Forssman-type heterophile antibodies in juvenile rheumatoid arthritis. Arthritis Rheum 23: 1262–1267, 1980. 544. Emancipator K, Moore TL, Dorner RW, et al: Hidden and classical 19S IgM rheumatoid factor in a juvenile rheumatoid arthritis patient. J Rheumatol 12: 372–375, 1985. 545. Speiser JC, Moore TL, Weiss TD, et al: Hidden 19S IgM rheumatoid factor in adults with juvenile rheumatoid arthritis onset. Ann Rheum Dis 44: 294–298, 1985. 546. Schlump U, Howard A, Ansell BM: IgG-anti-IgG antibodies in juvenile chronic arthritis. Scand J Rheumatol 14: 65–68, 1985. 547. Moore TL, El Najdawi E, Dorner RW: IgM rheumatoid factor plaque-forming cells in juvenile rheumatoid arthritis. Arthritis Rheum 30: 335–338, 1987. 548. Magsaam J, Ferjencik P, Tempels M, et al: A new method for the detection of hidden IgM rheumatoid factor in patients with juvenile rheumatoid arthritis. J Rheumatol 14: 964–967, 1987. 549. Ilowite NT, Wedgwood JF, Ramakrishnan T, et al: The rheumatoid factor cross-reactive idiotype in juvenile rheumatoid arthritis: role of the CD5-positive B cell. Clin Immunol Immunopathol 67: S74–S82, 1993. 550. Andersson GB, Fasth A: Serum concentration of hyaluronan, IgM and IgA rheumatoid factors in a population based study of juvenile chronic arthritis. Scand J Rheumatol 23: 183–190, 1994. 551. Bharadwaj A, Aggarwal A, Misra R: Clinical relevance of IgA rheumatoid factor (RF) in children with juvenile rheumatoid arthritis. Rheumatol Int 19: 47–49, 1999. 552. Ince A, Akhter IM, Moore TL, et al: Hidden 19S IgM rheumatoid factors in Turkish patients with juvenile rheumatoid arthritis. J Rheumatol 25: 190–192, 1998. 553. Danda D, Naveed M, Misra R: Detection of IgM hidden rheumatoid factor in juvenile rheumatoid arthritis: a novel approach using high performance liquid chromatography. J Indian Rheum Assoc 2 (Suppl): 80–84, 1994. 554. Bhardwaj A, Aggarwal A, Agarwal V, et al: Role of IgM & IgA rheumatoid factors in complement activation in patients with juvenile rheumatoid arthritis. Indian J Med Res 111: 103–109, 2000. 555. Agarwal V, Misra R, Aggarwal A: Immune complexes contain immunoglobulin A rheumatoid factor in serum and synovial fluid of patients with polyarticular juvenile rheumatoid arthritis. Rheumatology (Oxf) 41: 466–467, 2002. 556. Siamopoulou-Mavridou A, Mavridis AK, Terzoglou C, et al: Autoantibodies in Greek juvenile chronic arthritis patients. Clin Exp Rheumatol 9: 647–652, 1991. 557. Petty RE, Cassidy JT, Sullivan DB: Clinical correlates of antinuclear antibodies in juvenile rheumatoid arthritis. J Pediatr 83: 386–389, 1973. 558. Schaller JG, Johnson GD, Holborow EJ, et al: The association of antinuclear antibodies with the chronic iridocyclitis of juvenile rheumatoid arthritis (Still’s disease). Arthritis Rheum 17: 409–416, 1974. 559. Allen RC, Dewez P, Stuart L, et al: Antinuclear antibodies using HEp-2 cells in normal children and in children with common infections. J Paediatr Child Health 27: 39–42, 1991. 560. Cabral DA, Petty RE, Fung M, et al: Persistent antinuclear antibodies in children without identifiable inflammatory rheumatic or autoimmune disease. Pediatrics 89: 441–444, 1992. 561. Deane PM, Liard G, Siegel DM, et al: The outcome of children referred to a pediatric rheumatology clinic with a positive antinuclear antibody test but without an autoimmune disease. Pediatrics 95: 892–895, 1995. 562. Alspaugh MA, Miller JJ: A study of specificities of antinuclear antibodies in juvenile rheumatoid arthritis. J Pediatr 90: 391–395, 1977. 563. Permin H, Horbov S, Wiik A, et al: Antinuclear antibodies in juvenile chronic arthritis. Acta Paediatr Scand 67: 181–185, 1978. 564. Hoyeraal HM: Granulocyte reactive antinuclear factors in juvenile rheumatoid arthritis. Scand J Rheumatol 5: 84–90, 1976. 565. Jarvis JN, Pousak T, Krenz M, et al: Complement activation and immune complexes in juvenile rheumatoid arthritis. J Rheumatol 20: 114–117, 1993.
254
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
566. Jarvis JN, Lockman JC, Levine RP: IgM rheumatoid factor and the inhibition of covalent binding of C4b to IgG in immune complexes. Clin Exp Rheumatol 11: 135–141, 1993. 567. Jarvis JN, Taylor H, Iobidze M, et al: Complement activation and immune complexes in children with polyarticular juvenile rheumatoid arthritis: a longitudinal study. J Rheumatol 21: 1124–1127, 1994. 568. Jarvis JN, Xu C, Wang W, et al: Immune complex size and complement regulate cytokine production by peripheral blood mononuclear cells. Clin Immunol 93: 274–282, 1999. 569. Jiang K, Chen Y, Xu CS, et al: T cell activation by soluble C1q-bearing immune complexes: implications for the pathogenesis of rheumatoid arthritis. Clin Exp Immunol 131: 61–67, 2003. 570. Iiowite NT, Samuel P, Beseler L, et al: Dyslipoproteinemia in juvenile rheumatoid arthritis. J Pediatr 114: 823–826, 1989. 571. Bakkaloglu A, Kirel B, Ozen S, et al: Plasma lipids and lipoproteins in juvenile chronic arthritis. Clin Rheumatol 15: 341–345, 1996. 572. Tselepis AD, Elisaf M, Besis S, et al: Association of the inflammatory state in active juvenile rheumatoid arthritis with hypo-high-density lipoproteinemia and reduced lipoprotein-associated platelet-activating factor acetylhydrolase activity. Arthritis Rheum 42: 373–383, 1999. 573. Mollnes TE, Paus A: Complement activation in synovial fluid and tissue from patients with juvenile rheumatoid arthritis. Arthritis Rheum 29: 1359–1364, 1986. 574. Rynes RI, Ruddy S, Spragg J, et al: Intraarticular activation of the complement system in patients with juvenile rheumatoid arthritis. Arthritis Rheum 19: 161–168, 1976. 575. Miller JJ III, Olds LC, Huene DB: Complement activation products and factors influencing phagocyte migration in synovial fluids from children with chronic arthritis. Clin Exp Rheumatol 4: 53–56, 1986. 576. Martin CL, Pachman LM: Synovial fluid in seronegative juvenile rheumatoid arthritis: studies of immunoglobulins, complements, and alpha 2-macroglobulin. Arthritis Rheum 23: 1256–1261, 1980. 577. Pettersson H, Rydholm U: Radiologic classification of knee joint destruction in juvenile chronic arthritis. Pediatr Radiol 14: 419–421, 1984. 578. Pettersson H, Rydholm U: Radiologic classification of joint destruction in juvenile chronic arthritis. Acta Radiol [Diagn] (Stockh) 26: 719–722, 1985. 579. Poznanski AK, Conway JJ, Shkolnik A, et al: Radiological approaches in the evaluation of joint disease in children. Rheum Dis Clin North Am 13: 57–73, 1987. 580. Van Rossum MA, Zwinderman AH, Boers M, et al: Radiologic features in juvenile idiopathic arthritis: a first step in the development of a standardized assessment method. Arthritis Rheum 48: 507–515, 2003. 581. Azouz EM: Arthritis in children: conventional and advanced imaging. Semin Musculoskelet Radiol 7: 95–102, 2003. 582. Reed MH, Wilmot DM: The radiology of juvenile rheumatoid arthritis. A review of the English language literature. J Rheumatol Suppl 31: 2–22, 1991. 583. Wihlborg C, Babyn P, Ranson M, et al: Radiologic mimics of juvenile rheumatoid arthritis. Pediatr Radiol 31: 315–326, 2001. 584. Lamer S, Sebag GH: MRI and ultrasound in children with juvenile chronic arthritis. Eur J Radiol 33: 85–93, 2000. 585. El Miedany YM, Housny IH, Mansour HM, et al: Ultrasound versus MRI in the evaluation of juvenile idiopathic arthritis of the knee. Joint Bone Spine 68: 222–230, 2001. 586. Friedman S, Gruber MA: Ultrasonography of the hip in the evaluation of children with seronegative juvenile rheumatoid arthritis. J Rheumatol 29: 629–632, 2002. 587. Frosch M, Foell D, Ganser G, et al: Arthrosonography of hip and knee joints in the follow up of juvenile rheumatoid arthritis. Ann Rheum Dis 62: 242–244, 2003. 588. Goldenstein C, McCauley R, Troy M, et al: Ultrasonography in the evaluation of wrist swelling in children. J Rheumatol 16: 1079–1087, 1989. 589. Ruhoy MK, Tucker L, McCauley RG: Hypertrophic bursopathy of the subacromial-subdeltoid bursa in juvenile rheumatoid arthritis: sonographic appearance. Pediatr Radiol 26: 353–355, 1996. 590. Murray JG, Ridley NT, Mitchell N, et al: Juvenile chronic arthritis of the hip: value of contrast-enhanced MR imaging. Clin Radiol 51: 99–102, 1996. 591. Ramsey SE, Cairns RA, Cabral DA, et al: Knee magnetic resonance imaging in childhood chronic monarthritis. J Rheumatol 26: 2238–2243, 1999. 592. Cassidy JT, Martel W: Juvenile rheumatoid arthritis: clinicoradiologic correlations. Arthritis Rheum 20: 207–211, 1977. 593. Williams RA, Ansell BM: Radiological findings in seropositive juvenile chronic arthritis (juvenile rheumatoid arthritis) with particular reference to progression. Ann Rheum Dis 44: 685–693, 1985. 594. Lang BA, Schneider R, Reilly BJ, et al: Radiologic features of systemic onset juvenile rheumatoid arthritis. J Rheumatol 22: 168–173, 1995. 595. Kobayakawa M, Rydholm U, Wingstrand H, et al: Femoral head necrosis in juvenile chronic arthritis. Acta Orthop Scand 60: 164–169, 1989. 596. Elsasser U, Wilkins B, Hesp R, et al: Bone rarefaction and crush fractures in juvenile chronic arthritis. Arch Dis Child 57: 377–380, 1982. 597. Varonos S, Ansell BM, Reeve J: Vertebral collapse in juvenile chronic arthritis: its relationship with glucocorticoid therapy. Calcif Tissue Int 41: 75–78, 1987. 598. Poznanski AK, Hernandez RJ, Guire KE, et al: Carpal length in children—a useful measurement in the diagnosis of rheumatoid arthritis and some congenital malformation syndromes. Radiology 129: 661–668, 1978.
599. Mason T, Reed AM, Nelson AM, et al: Frequency of abnormal hand and wrist radiographs at time of diagnosis of polyarticular juvenile rheumatoid arthritis. J Rheumatol 29: 2214–2218, 2002. 600. Magni-Manzoni S, Rossi F, Pistorio A, et al: Prognostic factors for radiographic progression, radiographic damage, and disability in juvenile idiopathic arthritis. Arthritis Rheum 48: 3509–3517, 2003. 601. Libby AK, Sherry DD, Dudgeon BJ: Shoulder limitation in juvenile rheumatoid arthritis. Arch Phys Med Rehabil 72: 382–384, 1991. 602. Kuseler A, Pedersen TK, Herlin T, et al: Contrast enhanced magnetic resonance imaging as a method to diagnose early inflammatory changes in the temporomandibular joint in children with juvenile chronic arthritis. J Rheumatol 25: 1406–1412, 1998. 603. Sfondrini G, Schiavi A, Lalli A, et al: [TMJ in subjects with juvenile rheumatoid arthritis: functional and cephalometric analysis.] Mondo Ortod 16: 287–297, 1991. 604. Marini I, Vecchiet F, Spiazzi L, Capurso U: Stomatognathic function in juvenile rheumatoid arthritis and in developmental open-bite subjects. ASDC J Dent Child 66: 30–5,12, 1999. 605. Martini G, Bacciliero U, Tregnaghi A, et al: Isolated temporomandibular synovitis as unique presentation of juvenile idiopathic arthritis. J Rheumatol 28: 1689–1692, 2001. 606. Jank S, Schroder D, Haase S, et al: Temporomandibular disorders in juvenile patients with rheumatic diseases. Mund Kiefer Gesichtschir 7: 214–219, 2003. 607. Blasberg B, Lowe AA, Petty RE, et al: Temporomandibular joint disease in children with juvenile rheumatoid arthritis. Arthritis Rheum 30: S27, 1987. 608. Pedersen TK, Herlin T, Melsen B: Non-surgical treatment of the unilateral affection of the temporomandibular joint in children with juvenile chronic arthritis. Clin Exp Rheumatol 11 (Suppl 9): S79, 1993. 609. Rosberg G, Laine V: Natural history of radiological changes of knee joint in juvenile rheumatoid arthritis. Acta Paediatr Scand 56: 671–675, 1967. 610. Cellerini M, Salti S, Trapani S, et al: Correlation between clinical and ultrasound assessment of the knee in children with mono-articular or pauci-articular juvenile rheumatoid arthritis. Pediatr Radiol 29: 117–123, 1999. 611. Gylys-Morin VM, Graham TB, Blebea JS, et al: Knee in early juvenile rheumatoid arthritis: MR imaging findings. Radiology 220: 696–706, 2001. 612. Truckenbrodt H, Hafner R, von Altenbockum C: Functional joint analysis of the foot in juvenile chronic arthritis. Clin Exp Rheumatol 12 (Suppl 10): S91–S96, 1994. 613. Garcia-Morteo O, Gusis SE, Somma LF, et al: Tarsal ankylosis in juvenile and adult onset rheumatoid arthritis. J Rheumatol 15: 298–300, 1988. 614. Remedios D, Martin K, Kaplan G, et al: Juvenile chronic arthritis: diagnosis and management of tibio-talar and sub-talar disease. Br J Rheumatol 36: 1214–1217, 1997. 615. Hafner R: Influence of inflammation and muscular imbalance on growth and form of the hip in juvenile chronic arthritis. Rev Rhum Engl Ed 64: 169S–172S, 1997. 616. Spencer CH, Bernstein BH: Hip disease in juvenile rheumatoid arthritis. Curr Opin Rheumatol 14: 536–541, 2002. 617. Gabriel H, Fitzgerald SW, Myers MT, et al: MR imaging of hip disorders. Radiographics 14: 763–781, 1994. 618. Fantini F, Corradi A, Gerloni V, et al: The natural history of hip involvement in juvenile rheumatoid arthritis: a radiological and magnetic resonance imaging follow-up study. Rev Rhum Engl Ed 64: 173S–178S, 1997. 619. Hensinger RN, DeVito PD, Ragsdale CG: Changes in the cervical spine in juvenile rheumatoid arthritis. J Bone Joint Surg Am 68: 189–198, 1986. 620. Locke GR, Gardner JI, Van Epps EF: Atlas-dens interval (ADI) in children: a survey based on 200 normal cervical spines. Am J Roentgenol Radium Ther Nucl Med 97: 135–140, 1966. 621. Hall MA, Burgos VR, Ansell BM: Sacroiliitis in juvenile chronic arthritis. A 10-year follow-up. Clin Exp Rheumatol 5 (Suppl 1): S65–S67, 1987. 622. Hobbs N, Perrin JM: Issues in the Care of Children with Chronic Illness. A Source Book on Problems, Services, and Policies. San Francisco, JosseyBass, 1985. 623. Hobbs N, Perrin JM, Ireys HT: Chronically Ill Children and Their Families. Problems, Prospects, and Proposals from the Vanderbilt Study. San Francisco, Jossey-Bass, 1985. 624. Rapoff MA, Purviance MR, Lindsley CB: Educational and behavioral strategies for improving medication compliance in juvenile rheumatoid arthritis. Arch Phys Med Rehabil 69: 439–441, 1988. 625. Andre M, Hedengren E, Hagelberg S, et al: Perceived ability to manage juvenile chronic arthritis among adolescents and parents: development of a questionnaire to assess medical issues, exercise, pain, and social support. Arthritis Care Res 12: 229–237, 1999. 626. Rapoff MA, Belmont J, Lindsley C, et al: Prevention of nonadherence to nonsteroidal anti-inflammatory medications for newly diagnosed patients with juvenile rheumatoid arthritis. Health Psychol 21: 620–623, 2002. 627. Chmell MJ, Scott RD, Thomas WH, et al: Total hip arthroplasty with cement for juvenile rheumatoid arthritis. Results at a minimum of ten years in patients less than thirty years old. J Bone Joint Surg Am 79: 44–52, 1997. 628. Cassidy JT: Medical management of children with juvenile rheumatoid arthritis. Drugs 58: 831–850, 1999. 629. Cron RQ, Sharma S, Sherry DD: Current treatment by United States and Canadian pediatric rheumatologists. J Rheumatol 26: 2036–2038, 1999. 630. Ilowite NT: Current treatment of juvenile rheumatoid arthritis. Pediatrics 109: 109–115, 2002.
C H A P T E R 631. Cron RQ: Current treatment for chronic arthritis in childhood. Curr Opin Pediatr 14: 684–687, 2002. 632. Arkachaisri T, Lehman TJ: Use of biologics in the treatment of childhood rheumatic diseases. Curr Rheumatol Rep 2: 330–336, 2000. 633. Wallace CA, Sherry DD: Trial of intravenous pulse cyclophosphamide and methylprednisolone in the treatment of severe systemic-onset juvenile rheumatoid arthritis. Arthritis Rheum 40: 1852–1855, 1997. 634. Wallace CA: On beyond methotrexate treatment of severe juvenile rheumatoid arthritis. Clin Exp Rheumatol 17: 499–504, 1999. 635. Ruperto N, Levinson JE, Ravelli A, et al: Long-term health outcomes and quality of life in American and Italian inception cohorts of patients with juvenile rheumatoid arthritis. I. Outcome status. J Rheumatol 24: 945–951, 1997. 636. Ruperto N, Ravelli A, Levinson JE, et al: Long-term health outcomes and quality of life in American and Italian inception cohorts of patients with juvenile rheumatoid arthritis. II. Early predictors of outcome. J Rheumatol 24: 952–958, 1997. 637. Petty RE: Prognosis in children with rheumatic diseases: justification for consideration of new therapies. Rheumatology (Oxf) 38: 739–742, 1999. 638. Ruperto N, Ravelli A, Migliavacca D, et al: Responsiveness of clinical measures in children with oligoarticular juvenile chronic arthritis. J Rheumatol 26: 1827–1830, 1999. 639. Feldman BM, Grundland B, McCullough L, et al: Distinction of quality of life, health related quality of life, and health status in children referred for rheumatologic care. J Rheumatol 27: 226–233, 2000. 640. Giannini EH, Ruperto N, Ravelli A, et al: Preliminary definition of improvement in juvenile arthritis. Arthritis Rheum 40: 1202–1209, 1997. 641. Ruperto N, Ravelli A, Falcini F, et al: Responsiveness of outcome measures in juvenile chronic arthritis. Italian Pediatric Rheumatology Study Group. Rheumatology (Oxf) 38: 176–180, 1999. 642. Cassidy JT, Johnson JC, Hewett JE, et al: The clinical utility of measures of disease severity in children with juvenile rheumatoid arthritis (JRA). Clin Exp Rheumatol 12: S122, 1994. 643. Ravelli A, Viola S, Ruperto N, et al: Correlation between conventional disease activity measures in juvenile chronic arthritis. Ann Rheum Dis 56: 197–200, 1997. 644. Brunner HI, Lovell DJ, Finck BK, et al: Preliminary definition of disease flare in juvenile rheumatoid arthritis. J Rheumatol 29: 1058–1064, 2002. 645. Yazici Y, Erkan D, Paget SA: Monitoring by rheumatologists for methotrexate-, etanercept-, infliximab-, and anakinra-associated adverse events. Arthritis Rheum 48: 2769–2772, 2003. 646. Helliwell PS, Ibrahim G: Ethnic differences in responses to disease modifying drugs. Rheumatology (Oxf) 42: 1197–1201, 2003. 647. Cimaz R, Corona F, Scarazatti M, et al: Methotrexate treatment every other week in patients with juvenile chronic arthritis. Br J Rheumatol 35: 1030–1031, 1996. 648. Keenan GF, Giannini EH, Athreya BH: Clinically significant gastropathy associated with nonsteroidal antiinflammatory drug use in children with juvenile rheumatoid arthritis. J Rheumatol 22: 1149–1151, 1995. 649. Dowd JE, Cimaz R, Fink CW: Nonsteroidal antiinflammatory drug-induced gastroduodenal injury in children. Arthritis Rheum 38: 1225–1231, 1995. 650. Gazarian M, Berkovitch M, Koren G, et al: Experience with misoprostol therapy for NSAID gastropathy in children. Ann Rheum Dis 54: 277–280, 1995. 651. Kiss MH, Reiff AA, Reicin AS, et al: Rofecoxib demonstrates efficacy and tolerability in children and adolescents (age 2–17 years) with juvenile rheumatoid arthritis (JRA) in a 12-week randomized study. Arthritis Rheum 48 (Suppl 9): S650, 2003. 652. Davies K, Woo P: Immunization in rheumatic diseases of childhood: an audit of the clinical practice of British Paediatric Rheumatology Group members and a review of the evidence. Rheumatology (Oxf) 41: 937–941, 2002. 653. Lovell DJ, Giannini EH, Brewer EJ Jr: Time course of response to nonsteroidal antiinflammatory drugs in juvenile rheumatoid arthritis. Arthritis Rheum 27: 1433–1437, 1984. 654. Levy ML, Barron KS, Eichenfield A, et al: Naproxen-induced pseudoporphyria: a distinctive photodermatitis. J Pediatr 117: 660–664, 1990. 655. Allen R, Rogers M, Humphrey I: Naproxen induced pseudoporphyria in juvenile chronic arthritis. J Rheumatol 18: 893–896, 1991. 656. Lang BA, Finlayson LA: Naproxen-induced pseudoporphyria in patients with juvenile rheumatoid arthritis. J Pediatr 124: 639–642, 1994. 657. Wallace CA, Farrow D, Sherry DD: Increased risk of facial scars in children taking nonsteroidal antiinflammatory drugs. J Pediatr 125: 819–822, 1994. 658. Girschick HJ, Hamm H, Ganser G, et al: Naproxen-induced pseudoporphyria: appearance of new skin lesions after discontinuation of treatment. Scand J Rheumatol 24: 108–111, 1995. 659. Makela AL, Lempiainen M, Ylijoki H: Ibuprofen levels in serum and synovial fluid. Scand J Rheumatol Suppl 39: 15–17, 1981. 660. Jacobs JS: Sudden death in arthritic children receiving large doses of indomethacin. JAMA 199: 932–934, 1967. 661. Wortmann DW, Kelsch RC, Kuhns L, et al: Renal papillary necrosis in juvenile rheumatoid arthritis. J Pediatr 97: 37–40, 1980. 662. Allen RC, Petty RE, Lirenman DS, et al: Renal papillary necrosis in children with chronic arthritis. Am J Dis Child 140: 20–22, 1986. 663. Truckenbrodt H, Hafner R: Methotrexate therapy in juvenile rheumatoid arthritis: a retrospective study. Arthritis Rheum 29: 801–807, 1986.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
255
664. Speckmaier M, Findeisen J, Woo P, et al: Low-dose methotrexate in systemic onset juvenile chronic arthritis. Clin Exp Rheumatol 7: 647–650, 1989. 665. Halle F, Prieur AM: Evaluation of methotrexate in the treatment of juvenile chronic arthritis according to the subtype. Clin Exp Rheumatol 9: 297–302, 1991. 666. Giannini EH, Brewer EJ, Kuzmina N, et al: Methotrexate in resistant juvenile rheumatoid arthritis. Results of the U.S.A.-U.S.S.R. double-blind, placebocontrolled trial. The Pediatric Rheumatology Collaborative Study Group and The Cooperative Children’s Study Group. N Engl J Med 326: 1043–1049, 1992. 667. Lovell DJ: Ten years of experience with methotrexate. Past, present and future. Rev Rhum Engl Ed 64: 186S–188S, 1997. 668. Murray KJ, Lovell DJ: Advanced therapy for juvenile arthritis. Best Pract Res Clin Rheumatol 16: 361–378, 2002. 669. Giannini EH: Methotrexate: vital new information about a middle aged drug. J Rheumatol 29: 2031–2033, 2002. 670. Ramanan AV, Whitworth P, Baildam EM: Use of methotrexate in juvenile idiopathic arthritis. Arch Dis Child 88: 197–200, 2003. 671. Wallace CA, Levinson JE: Juvenile rheumatoid arthritis: outcome and treatment for the 1990s. Rheum Dis Clin North Am 17: 891–905, 1991. 672. Giannini EH, Cassidy JT, Brewer EJ, et al: Comparative efficacy and safety of advanced drug therapy in children with juvenile rheumatoid arthritis. Semin Arthritis Rheum 23: 34–46, 1993. 673. Wallace CA: The use of methotrexate in childhood rheumatic diseases. Arthritis Rheum 41: 381–391, 1998. 674. Martini A, Ravelli A, Viola S, et al: Methotrexate hepatotoxic effects in children with juvenile rheumatoid arthritis. J Pediatr 119: 333–334, 1991. 675. Kugathasan S, Newman AJ, Dahms BB, et al: Liver biopsy findings in patients with juvenile rheumatoid arthritis receiving long-term, weekly methotrexate therapy. J Pediatr 128: 149–151, 1996. 676. Hashkes PJ, Balistreri WF, Bove KE, et al: The long-term effect of methotrexate therapy on the liver in patients with juvenile rheumatoid arthritis. Arthritis Rheum 40: 2226–2234, 1997. 677. Hashkes PJ, Balistreri WF, Bove KE, et al: The relationship of hepatotoxic risk factors and liver histology in methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 134: 47–52, 1999. 678. Kremer JM, Alarcon GS, Lightfoot RW Jr, et al: Methotrexate for rheumatoid arthritis. Suggested guidelines for monitoring liver toxicity. American College of Rheumatology. Arthritis Rheum 37: 316–328, 1994. 679. Kremer JM: Not yet time to change the guidelines for monitoring methotrexate liver toxicity: they have served us well. J Rheumatol 29: 1590–1592, 2002. 680. Pincus T: Guidelines for monitoring of methotrexate therapy: “evidence-based medicine” outside of clinical trials. Arthritis Rheum 48: 2706–2709, 2003. 681. Falcini F, Taccetti G, Ermini M, et al: Methotrexate-associated appearance and rapid progression of rheumatoid nodules in systemic-onset juvenile rheumatoid arthritis. Arthritis Rheum 40: 175–178, 1997. 682. Cron RQ, Sherry DD, Wallace CA: Methotrexate-induced hypersensitivity pneumonitis in a child with juvenile rheumatoid arthritis. J Pediatr 132: 901–902, 1998. 683. Camiciottoli G, Trapani S, Castellani W, et al: Effect on lung function of methotrexate and non-steroid anti-inflammatory drugs in children with juvenile rheumatoid arthritis. Rheumatol Int 18: 11–16, 1998. 684. Schmeling H, Stephan V, Burdach S, et al: Pulmonary function in children with juvenile idiopathic arthritis and effects of methotrexate therapy. Z Rheumatol 61: 168–172, 2002. 685. Muzaffer MA, Schneider R, Cameron BJ, et al: Accelerated nodulosis during methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 128: 698–700, 1996. 686. Bianchi ML, Cimaz R, Galbiati E, et al: Bone mass change during methotrexate treatment in patients with juvenile rheumatoid arthritis. Osteoporos Int 10: 20–25, 1999. 687. Ravelli A, Migliavacca D, Viola S, et al: Efficacy of folinic acid in reducing methotrexate toxicity in juvenile idiopathic arthritis. Clin Exp Rheumatol 17: 625–627, 1999. 688. Endresen GK, Husby G: Folate supplementation during methotrexate treatment of patients with rheumatoid arthritis. An update and proposals for guidelines. Scand J Rheumatol 30: 129–134, 2001. 689. Wallace CA, Sherry DD: Preliminary report of higher dose methotrexate treatment in juvenile rheumatoid arthritis. J Rheumatol 19: 1604–1607, 1992. 690. Albertioni F, Flato B, Seideman P, et al: Methotrexate in juvenile rheumatoid arthritis. Evidence of age dependent pharmacokinetics. Eur J Clin Pharmacol 47: 507–511, 1995. 691. Ravelli A, Gerloni V, Corona F, et al: Oral versus intramuscular methotrexate in juvenile chronic arthritis. Italian Pediatric Rheumatology Study Group. Clin Exp Rheumatol 16: 181–183, 1998. 692. Wallace CA, Bleyer WA, Sherry DD, et al: Toxicity and serum levels of methotrexate in children with juvenile rheumatoid arthritis. Arthritis Rheum 32: 677–681, 1989. 693. Ravelli A, Di Fuccia G, Molinaro M, et al: Plasma levels after oral methotrexate in children with juvenile rheumatoid arthritis. J Rheumatol 20: 1573–1577, 1993. 694. Alsufyani K, Ortiz-Alvarez O, Cabral DA, et al: The role of subcutaneous administration of methotrexate in children with juvenile idiopathic arthritis who have failed oral methotrexate. J Rheumatol 31: 179–182, 2004.
256
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
695. Rose CD, Singsen BH, Eichenfield AH, et al: Safety and efficacy of methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 117: 653–659, 1990. 696. Graham LD, Myones BL, Rivas-Chacon RF, et al: Morbidity associated with long-term methotrexate therapy in juvenile rheumatoid arthritis. J Pediatr 120: 468–473, 1992. 697. Moroldo MB, Giannini EH: Estimates of the discriminant ability of definitions of improvement for juvenile rheumatoid arthritis. J Rheumatol 25: 986–989, 1998. 698. Wallace CA, Sherry DD, Mellins ED, et al: Predicting remission in juvenile rheumatoid arthritis with methotrexate treatment. J Rheumatol 20: 118–122, 1993. 699. Ravelli A, Ramenghi B, Di Fuccia G, et al: Factors associated with response to methotrexate in systemic-onset juvenile chronic arthritis. Acta Paediatr 83: 428–432, 1994. 700. Ravelli A, Viola S, Ramenghi B, et al: Evaluation of response to methotrexate by a functional index in juvenile chronic arthritis. Clin Rheumatol 14: 322–326, 1995. 701. Huang JL: Methotrexate in the treatment of children with chronic arthritis— long-term observations of efficacy and safety. Br J Clin Pract 50: 311–314, 1996. 702. Giannini EH, Cassidy JT: Methotrexate in juvenile rheumatoid arthritis. Do the benefits outweigh the risks? Drug Saf 9: 325–339, 1993. 703. Giannini EH, Cawkwell GD: Drug treatment in children with juvenile rheumatoid arthritis. Past, present, and future. Pediatr Clin North Am 42: 1099–1125, 1995. 704. Ruperto N, Ravelli A, Falcini F, et al: Performance of the preliminary definition of improvement in juvenile chronic arthritis patients treated with methotrexate. Italian Pediatric Rheumatology Study Group. Ann Rheum Dis 57: 38–41, 1998. 705. Gottlieb BS, Keenan GF, Lu T, et al: Discontinuation of methotrexate treatment in juvenile rheumatoid arthritis. Pediatrics 100: 994–997, 1997. 706. Cassidy JT: Outcomes research in the therapeutic use of methotrexate in children with chronic peripheral arthritis. J Pediatr 133: 179–180, 1998. 707. Harel L, Wagner-Weiner L, Poznanski AK, et al: Effects of methotrexate on radiologic progression in juvenile rheumatoid arthritis. Arthritis Rheum 36: 1370–1374, 1993. 708. Ravelli A, Viola S, Ramenghi B, et al: Radiologic progression in patients with juvenile chronic arthritis treated with methotrexate. J Pediatr 133: 262–265, 1998. 709. Ravelli A, Viola S, Migliavacca D, et al: The extended oligoarticular subtype is the best predictor of methotrexate efficacy in juvenile idiopathic arthritis. J Pediatr 135: 316–320, 1999. 710. Ravelli A, Viola S, Ramenghi B, et al: Frequency of relapse after discontinuation of methotrexate therapy for clinical remission in juvenile rheumatoid arthritis. J Rheumatol 22: 1574–1576, 1995. 711. Michels H: What is low-dose corticosteroid therapy in juvenile idiopathic arthritis? A worldwide, questionnaire-based survey. Z Rheumatol 59 Suppl 2: II/127-II/130, 2000. 712. Michels H: Is there a renaissance of corticosteroid therapy? Rev Rhum Engl Ed 64: 183S–185S, 1997. 713. Falcini F, Taccetti G, Trapani S, et al: Growth retardation in juvenile chronic arthritis patients treated with steroids. Clin Exp Rheumatol 9 Suppl 6: 37–40, 1991. 714. Wang SJ, Yang YH, Lin YT, et al: Attained adult height in juvenile rheumatoid arthritis with or without corticosteroid treatment. Clin Rheumatol 21: 363–368, 2002. 715. Loftus J, Allen R, Hesp R, et al: Randomized, double-blind trial of deflazacort versus prednisone in juvenile chronic (or rheumatoid) arthritis: a relatively bone-sparing effect of deflazacort. Pediatrics 88: 428–436, 1991. 716. Falcini F, Trapani S, Ermini M, et al: Deflazacort in pediatric rheumatic diseases needs a frequent follow-up of bone densitometry. Pediatrics 95: 318, 1995. 717. Picco P, Gattorno M, Buoncompagni A, et al: 6-methylprednisolone “minipulses”: a new modality of glucocorticoid treatment in systemic onset juvenile chronic arthritis. Scand J Rheumatol 25: 24–27, 1996. 718. Adebajo AO, Hall MA: The use of intravenous pulsed methylprednisolone in the treatment of systemic-onset juvenile chronic arthritis. Br J Rheumatol 37: 1240–1242, 1998. 719. Allen RC, Gross KR, Laxer RM, et al: Intraarticular triamcinolone hexacetonide in the management of chronic arthritis in children. Arthritis Rheum 29: 997–1001, 1986. 720. Earley A, Cuttica RJ, McCullough C, et al: Triamcinolone into the knee joint in juvenile chronic arthritis. Clin Exp Rheumatol 6: 153–155, 1988. 721. Sparling M, Malleson P, Wood B, et al: Radiographic followup of joints injected with triamcinolone hexacetonide for the management of childhood arthritis. Arthritis Rheum 33: 821–826, 1990. 722. Uziel Y, Berkovitch M, Gazarian M, et al: Evaluation of eutectic lidocaine/prilocaine cream (EMLA) for steroid joint injection in children with juvenile rheumatoid arthritis: a double blind, randomized, placebo controlled trial. J Rheumatol 30: 594–596, 2003. 723. Job-Deslandre C, Menkes CJ: Complications of intra-articular injections of triamcinolone hexacetonide in chronic arthritis in children. Clin Exp Rheumatol 8: 413–416, 1990. 724. Hertzberger-ten Cate R, Cats A: Toxicity of sulfasalazine in systemic juvenile chronic arthritis. Clin Exp Rheumatol 9: 85–88, 1991.
725. Huppertz HI, Tschammler A, Horwitz AE, et al: Intraarticular corticosteroids for chronic arthritis in children: efficacy and effects on cartilage and growth. J Pediatr 127: 317–321, 1995. 726. Breit W, Frosch M, Meyer U, et al: A subgroup-specific evaluation of the efficacy of intraarticular triamcinolone hexacetonide in juvenile chronic arthritis. J Rheumatol 27: 2696–2702, 2000. 727. Ravelli A, Manzoni SM, Viola S, et al: Factors affecting the efficacy of intraarticular corticosteroid injection of knees in juvenile idiopathic arthritis. J Rheumatol 28: 2100–2102, 2001. 728. Zulian F, Martini G, Gobber D, et al: Comparison of intra-articular triamcinolone hexacetonide and triamcinolone acetonide in oligoarticular juvenile idiopathic arthritis. Rheumatology (Oxf) 42: 1254–1259, 2003. 729. Magni MS, Viola S, Beduschi L, et al: Transient remission of systemic manifestations following intraarticular triamcinolone hexacetonide injection in a boy with systemic juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 735, 2002. 730. Sherry DD, Stein LD, Reed AM, et al: Prevention of leg length discrepancy in young children with pauciarticular juvenile rheumatoid arthritis by treatment with intraarticular steroids. Arthritis Rheum 42: 2330–2334, 1999. 731. Padeh S, Passwell JH: Intraarticular corticosteroid injection in the management of children with chronic arthritis. Arthritis Rheum 41: 1210–1214, 1998. 732. Weinblatt ME, Kremer JM, Bankhurst AD, et al: A trial of etanercept, a recombinant tumor necrosis factor receptor: Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate. N Engl J Med 340: 253–259, 1999. 733. Horneff G, Forster J, Seyberth HW, et al: Recommendations by the Pediatric and Adolescent Rheumatology Study Committee on therapy with etanercept (p75 TNF-alpha receptor immunoglobulin fusion protein). Pharmacotherapy Committee. Z Rheumatol 59: 365–369, 2000. 734. Lovell DJ, Giannini EH, Reiff A, et al: Etanercept in children with polyarticular juvenile rheumatoid arthritis. Pediatric Rheumatology Collaborative Study Group. N Engl J Med 342: 763–769, 2000. 735. Kietz DA, Pepmueller PH, Moore TL: Clinical response to etanercept in polyarticular course juvenile rheumatoid arthritis. J Rheumatol 28: 360–362, 2001. 736. Martini A: Etanercept improves active polyarticular juvenile rheumatoid arthritis. Clin Exp Rheumatol 19: 122–124, 2001. 737. Culy CR, Keating GM: Etanercept: an updated review of its use in rheumatoid arthritis, psoriatic arthritis and juvenile rheumatoid arthritis. Drugs 62: 2493–2537, 2002. 738. Gerloni V, Pontikaki I, Gattinara M, et al: Treatment of juvenile idiopathic arthritis (JIA) with etanercept: short and medium term results. Arthritis Rheum 46 (Suppl): S482, 2002. 739. Lahdenne P, Vahasalo P, Honkanen V: Infliximab or etanercept in the treatment of children with refractory juvenile idiopathic arthritis: an open label study. Ann Rheum Dis 62: 245–247, 2003. 740. Elliott MJ, Maini RN, Feldmann M, et al: Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid arthritis. Lancet 344: 1105–1110, 1994. 741. Elliott MJ, Maini RN, Feldmann M, et al: Repeated therapy with monoclonal antibody to tumour necrosis factor alpha (cA2) in patients with rheumatoid arthritis. Lancet 344: 1125–1127, 1994. 742. Pisetsky DS: Tumor necrosis factor blockers in rheumatoid arthritis. N Engl J Med 342: 810–811, 2000. 743. Honkanen VEA, Tynjälä P, Vâhäsalo P: Infliximab in juvenile arthritis: 1-year follow-up. Arthritis Rheum 46 (Suppl): S480, 2002. 744. Dekker L, Armbrust W, Rademaker CM, et al: Safety of anti-TNFalpha therapy in children with juvenile idiopathic arthritis. Clin Exp Rheumatol 22: 252–258, 2004. 745. Schmeling H, Mathony K, John V, et al: A combination of etanercept and methotrexate for the treatment of refractory juvenile idiopathic arthritis: a pilot study. Ann Rheum Dis 60: 410–412, 2001. 746. Haapasaari J, Kautiainen H, Hannula S, et al: Good results from combining etanercept to prevailing DMARD therapy in refractory juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 867–870, 2002. 747. Takei S, Groh D, Bernstein B, et al: Safety and efficacy of high dose etanercept in treatment of juvenile rheumatoid arthritis. J Rheumatol 28: 1677–1680, 2001. 748. Gudbrandsdottir S, Larsen R, Sorensen LK, et al: TNF and LT binding capacities in the plasma of arthritis patients: effect of etanercept treatment in juvenile idiopathic arthritis. Clin Exp Rheumatol 22: 118–124, 2004. 749. Elwood RL, Pelszynski MM, Corman LI: Multifocal septic arthritis and osteomyelitis caused by group A streptococcus in a patient receiving immunomodulating therapy with etanercept. Pediatr Infect Dis J 22: 286–288, 2003. 750. Lovell DJ, Giannini EH, Reiff A, et al: Long-term efficacy and safety of etanercept in children with polyarticular-course juvenile rheumatoid arthritis: interim results from an ongoing multicenter, open-label, extended-treatment trial. Arthritis Rheum 48: 218–226, 2003. 751. Russo RA, Katsicas MM, Zelazko M: Etanercept in systemic juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 723–726, 2002. 752. Quartier P, Taupin P, Bourdeaut F, et al: Efficacy of etanercept for the treatment of juvenile idiopathic arthritis according to the onset type. Arthritis Rheum 48: 1093–1101, 2003. 753. Skytta E, Pohjankoski H, Savolainen A: Etanercept and urticaria in patients with juvenile idiopathic arthritis. Clin Exp Rheumatol 18: 533–534, 2000.
C H A P T E R 754. Livermore PA, Murray KJ: Anti-tumour necrosis factor therapy associated with cutaneous vasculitis. Rheumatology (Oxf) 41: 1450–1452, 2002. 755. Mohan N, Edwards ET, Cupps TR, et al: Demyelination occurring during anti-tumor necrosis factor alpha therapy for inflammatory arthritides. Arthritis Rheum 44: 2862–2869, 2001. 756. Lepore L, Marchetti F, Facchini S, et al: Drug-induced systemic lupus erythematosus associated with etanercept therapy in a child with juvenile idiopathic arthritis. Clin Exp Rheumatol 21: 276–277, 2003. 757. Gerloni V, Pontikaki I, Desiati F, et al: Evaluation of disease activity, disability and quality of life in patients with persistently active refractory juvenile chronic arthritis after one year treatment with monoclonal anti-tumor necrosis factor-α, antibody (infliximab) [abstract]. American College of Rheumatology 65th Annual Scientific Meeting, San Francisco, CA, November 11–16: 2001. 758. Vouyiouka O, Rizou S, Grafakou O: Infliximab and etanercept in the treatment of juvenile idiopathic arthritis. The Athens experience [abstract]. Ann Rheum Dis 60: ii17–ii52, 2001. 759. Scarazatti M, Beltramelli M, Dell’Era L, et al: Infliximab in juvenile idiopathic arthritis [abstract]. Ann Rheum Dis 60: ii17–ii52, 2001. 760. Vinje O, Obiora E, Forre O: Juvenile chronic polyarthritits treated with infliximab [abstract]. Ann Rheum Dis 59: 713–749, 2000. 761. Chaturvedi VP: Pilot study of chimeric monoclonal anti-tumor necrosis factor alpha anti-body (infliximab) with methotrexate in polyarticular juvenile idiopathic arthritis [abstract]. American College of Rheumatology 65th Annual Scientific Meeting, New Orleans, LA, October 24–25: 2002. 762. Honkanen VEA, Tynjälä P, Vâhäsalo P, et al: Infliximab in juvenile arthritis: 1-year follow-up [abstract]. American College of Rheumatology 66th Annual Scientific Meeting, New Oreans, LA, October 24–25: 2002. 763. Lovell DJ, Ruperto N, Cuttica R, et al: Randomized trial of infliximab (IFX) plus methotrexate (MTX) for the treatment of polyarticular juvenile rheumatoid arthritis (JRA). Arthritis Rheum 50: L8, 2004. 764. Crandall WV, Mackner LM: Infusion reactions to infliximab in children and adolescents: frequency, outcome and a predictive model. Aliment Pharmacol Ther 17: 75–84, 2003. 765. Myers A, Clark J, Foster H: Tuberculosis and treatment with infliximab. N Engl J Med 346: 623–626, 2002. 766. Hoyeraal HM, Froland SS, Salvesen CF, et al: No effect of transfer factor in juvenile rheumatoid arthritis by double-blind trial. Ann Rheum Dis 37: 175–179, 1978. 767. Prieur AM, Buriot D, Lefur JM, et al: Possible toxicity of levamisole in children with rheumatoid arthritis. J Pediatr 93: 304–305, 1978. 768. Bardare M, Corona F, Ogliari MT, et al: Thymopentin in the treatment of juvenile chronic arthritis. Clin Exp Rheumatol 8: 89–93, 1990. 769. Pernice W, Schuchmann L, Dippell J, et al: Therapy for systemic juvenile rheumatoid arthritis with gamma-interferon: a pilot study of nine patients. Arthritis Rheum 32: 643–646, 1989. 770. Coto C, Varela G, Hernandez V, et al: Use of recombinant interferon gamma in pediatric patients with advanced juvenile chronic arthritis. Biotherapy 11: 15–20, 1998. 771. Lepore L, Agosti E, Pitacco F, et al: Therapy with plasmapheresis and lymphoplasmapheresis combined with immunosuppressive agents in 2 cases of intractable juvenile rheumatoid arthritis. Pediatr Med Chir 9: 321–324, 1987. 772. Barnett ML, Combitchi D, Trentham DE: A pilot trial of oral type II collagen in the treatment of juvenile rheumatoid arthritis. Arthritis Rheum 39: 623–628, 1996. 773. McKown KM, Carbone LD, Kaplan SB, et al: Lack of efficacy of oral bovine type II collagen added to existing therapy in rheumatoid arthritis. Arthritis Rheum 42: 1204–1208, 1999. 774. Reiff A, Porras O, Rudge S, et al: Preliminary data from a study of Kineret™ (anakinra) in children with juvenile rheumatoid arthritis. Arthritis Rheum 46 (Suppl): S215, 2002. 775. De Benedetti F, Massa M, Pignatti P, et al: Serum soluble interleukin 6 (IL-6) receptor and IL-6/soluble IL-6 receptor complex in systemic juvenile rheumatoid arthritis. J Clin Invest 93: 2114–2119, 1994. 776. Lehman TJ, Striegel KH, Onel KB: Thalidomide therapy for recalcitrant systemic onset juvenile rheumatoid arthritis. J Pediatr 140: 125–127, 2002. 777. Stein LD, Roifman CM: Intravenous immunoglobulin therapy. Bull Rheum Dis 44: 3–5, 1995. 778. Prieur AM: Intravenous immunoglobulins in Still’s disease: still controversial, still unproven. J Rheumatol 23: 797–800, 1996. 779. Silverman ED, Cawkwell GD, Lovell DJ, et al: Intravenous immunoglobulin in the treatment of systemic juvenile rheumatoid arthritis: a randomized placebo controlled trial. Pediatric Rheumatology Collaborative Study Group. J Rheumatol 21: 2353–2358, 1994. 780. Giannini EH, Lovell DJ, Silverman ED, et al: Intravenous immunoglobulin in the treatment of polyarticular juvenile rheumatoid arthritis: a phase I/II study. Pediatric Rheumatology Collaborative Study Group. J Rheumatol 23: 919–924, 1996. 781. Laxer RM: Long-term toxicity of immune suppression in juvenile rheumatic diseases. Rheumatology (Oxf) 38: 743–746, 1999. 782. Ansell BM, Eghtedari A, Bywaters EG: Chlorambucil in the management of juvenile chronic polyarthritis complicated by amyloidosis. Ann Rheum Dis 30: 331, 1971. 783. Deschenes G, Prieur AM, Hayem F, et al: Renal amyloidosis in juvenile chronic arthritis: evolution after chlorambucil treatment. Pediatr Nephrol 4: 463–469, 1990.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
257
784. Savolainen HA, Isomaki HA: Decrease in the number of deaths from secondary amyloidosis in patients with juvenile rheumatoid arthritis. J Rheumatol 20: 1201–1203, 1993. 785. Rostropowicz-Denisiewicz K: Some remarks on management of juvenile chronic arthritis complicated by amyloidosis. Acta Univ Carol [Med] (Praha) 40: 91–94, 1994. 786. Savolainen HA: Chlorambucil in severe juvenile chronic arthritis: longterm followup with special reference to amyloidosis. J Rheumatol 26: 898–903, 1999. 787. David J: Amyloidosis in juvenile chronic arthritis. Clin Exp Rheumatol 9: 73–78, 1991. 788. Silverman E, Spiegel IR, Hawkins D, et al: Long-term open-label preliminary study of the safety and efficacy of leflunomide in patients with polyarticular-course juvenile rheumatoid arthritis. Arthritis Rheum 52: 554–562, 2005. 789. Weinblatt ME, Kremer JM, Coblyn JS, et al: Pharmacokinetics, safety, and efficacy of combination treatment with methotrexate and leflunomide in patients with active rheumatoid arthritis. Arthritis Rheum 42: 1322–1328, 1999. 790. Bjerkhoel F, Forre O: Cyclosporin treatment of a patient with severe systemic juvenile rheumatoid arthritis. Scand J Rheumatol 17: 483–486, 1988. 791. Ostensen M, Hoyeraal HM, Kass E: Tolerance of cyclosporine A in children with refractory juvenile rheumatoid arthritis. J Rheumatol 15: 1536–1538, 1988. 792. Reiff A, Rawlings DJ, Shaham B, et al: Preliminary evidence for cyclosporin A as an alternative in the treatment of recalcitrant juvenile rheumatoid arthritis and juvenile dermatomyositis. J Rheumatol 24: 2436–2443, 1997. 793. Gerloni V, Cimaz R, Gattinara M, et al: Efficacy and safety profile of cyclosporin A in the treatment of juvenile chronic (idiopathic) arthritis. Results of a 10-year prospective study. Rheumatology (Oxf) 40: 907–913, 2001. 794. Mouy R, Stephan JL, Pillet P, et al: Efficacy of cyclosporine A in the treatment of macrophage activation syndrome in juvenile arthritis: report of five cases. J Pediatr 129: 750–754, 1996. 795. Ravelli A, De Benedetti F, Viola S, et al: Macrophage activation syndrome in systemic juvenile rheumatoid arthritis successfully treated with cyclosporine. J Pediatr 128: 275–278, 1996. 796. Ravelli A, Viola S, De Benedetti F, et al: Dramatic efficacy of cyclosporine A in macrophage activation syndrome. Clin Exp Rheumatol 19: 108, 2001. 797. Grom AA: Natural killer cell dysfunction. A common pathway in systemiconset juvenile rheumatoid arthritis, macrophage activation syndrome, and hemophagocytic lymphohistiocytosis? Arthritis Rheum 50: 689–698, 2004. 798. Ravelli A, Moretti C, Temporini F, et al: Combination therapy with methotrexate and cyclosporine A in juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 569–572, 2002. 799. Grondin C, Malleson P, Petty RE: Slow-acting antirheumatic drugs in chronic arthritis of childhood. Semin Arthritis Rheum 18: 38–47, 1988. 800. Rosenberg AM: Treatment of juvenile rheumatoid arthritis: approach to patients who fail standard therapy. J Rheumatol 23: 1652–1656, 1996. 801. Fleischmann R: Safety and efficacy of disease-modifying antirheumatic agents in rheumatoid arthritis and juvenile rheumatoid arthritis. Expert Opin Drug Saf 2: 347–365, 2003. 802. Rynes RI: Antimalarial drugs in the treatment of rheumatological diseases. Br J Rheumatol 36: 799–805, 1997. 803. Marmor MF, Carr RE, Easterbrook M, et al: Recommendations on screening for chloroquine and hydroxychloroquine retinopathy: a report by the American Academy of Ophthalmology. Ophthalmology 109: 1377–1382, 2002. 804. Mavrikakis I, Sfikakis PP, Mavrikakis E, et al: The incidence of irreversible retinal toxicity in patients treated with hydroxychloroquine: a reappraisal. Ophthalmology 110: 1321–1326, 2003. 805. Brewer EJ, Giannini EH, Kuzmina N, et al: Penicillamine and hydroxychloroquine in the treatment of severe juvenile rheumatoid arthritis. Results of the U.S.A.-U.S.S.R. double-blind placebo-controlled trial. N Engl J Med 314: 1269–1276, 1986. 806. Van Kerckhove C, Giannini EH, Lovell DJ: Temporal patterns of response to D-penicillamine, hydroxychloroquine, and placebo in juvenile rheumatoid arthritis patients. Arthritis Rheum 31: 1252–1258, 1988. 807. Wallace DJ, Metzger AL, Stecher VJ, et al: Cholesterol-lowering effect of hydroxychloroquine in patients with rheumatic disease: reversal of deleterious effects of steroids on lipids. Am J Med 89: 322–326, 1990. 808. Giannini EH, Brewer EJ Jr, Person DA: Auranofin in the treatment of juvenile rheumatoid arthritis. J Pediatr 102: 138–141, 1983. 809. Giannini EH, Brewer EJ, Person DA, et al: Longterm auranofin therapy in patients with juvenile rheumatoid arthritis. J Rheumatol 13: 768–770, 1986. 810. Kvien TK, Hoyeraal HM, Sandstad B, et al: Auranofin therapy in juvenile rheumatoid arthritis: a 48-week phase II study. Scand J Rheumatol Suppl 63: 79–83, 1986. 811. Giannini EH, Brewer EJ Jr, Kuzmina N, et al: Auranofin in the treatment of juvenile rheumatoid arthritis. Results of the USA-USSR double-blind, placebo-controlled trial. The USA Pediatric Rheumatology Collaborative Study Group. The USSR Cooperative Children’s Study Group. Arthritis Rheum 33: 466–476, 1990. 812. Brewer EJ Jr, Giannini EH, Person DA: Early experiences with auranofin in juvenile rheumatoid arthritis. Am J Med 75: 152–156, 1983. 813. Wooley PH, Griffin J, Panayi GS, et al: HLA-DR antigens and toxic reaction to sodium aurothiomalate and D-penicillamine in patients with rheumatoid arthritis. N Engl J Med 303: 300–302, 1980.
258
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
814. Clarkson RW, Sanders PA, Grennan DM: Complement C4 null alleles as a marker of gold or D-penicillamine toxicity in the treatment of rheumatoid arthritis. Br J Rheumatol 31: 53–54, 1992. 815. Wulffraat NM, Vlieger A, Brinkman JP, et al: Autologus stem cell transplantation (ASCT) in retractory polyarticular and systemic JCA. Arthritis Rheum 41: S129, 1998. 816. Quartier P, Prieur AM, Fischer A: Haemopoietic stem-cell transplantation for juvenile chronic arthritis. Lancet 353: 1885–1886, 1999. 817. Kuis W, Wulffraat NM, Petty RE: Autologous stem cell transplantation: an alternative for refractory juvenile chronic arthritis. Rheumatology (Oxf) 38: 737–738, 1999. 818. Wulffraat NM, Kuis W: Autologous stem cell transplantation: a possible treatment for refractory juvenile chronic arthritis? Rheumatology (Oxf) 38: 764–766, 1999. 819. Wulffraat NM, Kuis W, Petty R: Addendum: Proposed guidelines for autologous stem cell transplantation in juvenile chronic arthritis. Paediatric Rheumatology Workshop. Rheumatology (Oxf) 38: 777–778, 1999. 820. Vossen JM, Brinkman DM, Bakker B, et al: Rationale for high-dose cyclophosphamide and medium-dose total body irradiation in the conditioning of children with progressive systemic and polyarticular juvenile chronic arthritis before autologous stem cell transplantation. Rheumatology (Oxf) 38: 762–763, 1999. 821. Tyndall A, Millikan S: Bone marrow transplantation. Baillieres Best Pract Res Clin Rheumatol 13: 719–735, 1999. 822. Lepore L, Kiren V: Autologous bone marrow transplantation versus alternative drugs in pediatric rheumatic diseases. Haematologica 85: 89–92, 2000. 823. Frosch M, Strey A, Vogl T, et al: Myeloid-related proteins 8 and 14 are specifically secreted during interaction of phagocytes and activated endothelium and are useful markers for monitoring disease activity in pauciarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 43: 628–637, 2000. 824. Wulffraat M, de K, I, Brinkman D, et al: Autologous stem cell transplantation for refractory juvenile idiopathic artrhitis: current results and perspectives. Transplant Proc 34: 2925–2926, 2002. 825. Wedderburn LR, Abinun M, Palmer P, et al: Autologous haematopoietic stem cell transplantation in juvenile idiopathic arthritis. Arch Dis Child 88: 201–205, 2003. 826. DeKleer IM, Brinkman DM, Ferster A, et al: Autologous stem cell transplantation for refractory Juvenile idiopathic arthritis: analysis of clinical effects, mortality, and transplant related morbidity. Ann Rheum Dis 63: 1318–1326, 2004. 827. Wulffraat NM, Brinkman D, Ferster A, et al: Long-term follow-up of autologous stem cell transplantation for refractory juvenile idiopathic arthritis. Bone Marrow Transplant 32 Suppl 1: S61–S64, 2003. 828 Wulffraat NM, Haas PJ, Frosch M, et al: Myeloid related protein 8 and 14 secretion reflects phagocyte activation and correlates with disease activity in juvenile idiopathic arthritis treated with autologous stem cell transplantation. Ann Rheum Dis 62: 236–241, 2003. 829. Schulze zur Wiesch A, Foell D, Frosch M, et al: Myeloid related proteins MRP8/MRP14 may predict disease flares in juvenile idiopathic arthritis. Clin Exp Rheumatol 22: 368–373, 2004. 830. Hopp RJ, Degan J, Corley K, et al: Evaluation of growth hormone secretion in children with juvenile rheumatoid arthritis and short stature. Nebr Med J 80: 52–57, 1995. 831. Bechtold S, Ripperger P, Hafner R, et al: Growth hormone improves height in patients with juvenile idiopathic arthritis: 4-year data of a controlled study. J Pediatr 143: 512–519, 2003. 832. Allen DB, Goldberg BD: Stimulation of collagen synthesis and linear growth by growth hormone in glucocorticoid-treated children. Pediatrics 89: 416–421, 1992. 833. Simon D, Touati G, Prieur AM, et al: Growth hormone treatment of short stature and metabolic dysfunction in juvenile chronic arthritis. Acta Paediatr Suppl 88: 100–105, 1999. 834. Touati G, Prieur AM, Ruiz JC, et al: Beneficial effects of one-year growth hormone administration to children with juvenile chronic arthritis on chronic steroid therapy. I. Effects on growth velocity and body composition [published erratum appears in J Clin Endocrinol Metab 83: 1547, 1998]. J Clin Endocrinol Metab 83: 403–409, 1998. 835. Manfredi R, Tumietto F, Azzaroli L, et al: Growth hormone (GH) and the immune system: impaired phagocytic function in children with idiopathic GH deficiency is corrected by treatment with biosynthetic GH. J Pediatr Endocrinol 7: 245–251, 1994. 836. Broyer M, Guest G, Crosnier H, et al: Recombinant growth hormone in children after renal transplantation. Societe Francaise de Nephrologie Pediatrique. Lancet 343: 539–540, 1994. 837. Crosnier H, Guest G, Souberbielle JC, et al: Treatment with recombinant human growth hormone (rhGH) in children with chronic kidney failure or renal transplantation. Arch Pediatr 1: 716–722, 1994. 838. Cheung NW, Liddle C, Coverdale S, et al: Growth hormone treatment increases cytochrome P450-mediated antipyrine clearance in man. J Clin Endocrinol Metab 81: 1999–2001, 1996. 839. Henderson CJ, Lovell DJ, Specker BL, et al: Physical activity in children with juvenile rheumatoid arthritis: quantification and evaluation. Arthritis Care Res 8: 114–119, 1995.
840. Knops N, Wulffraat N, Lodder S, et al: Resting energy expenditure and nutritional status in children with juvenile rheumatoid arthritis. J Rheumatol 26: 2039–2043, 1999. 841. Martini A, Ravelli A, Di Fuccia G, et al: Intravenous iron therapy for severe anaemia in systemic-onset juvenile chronic arthritis. Lancet 344: 1052–1054, 1994. 842. Henderson CJ, Lovell DJ: Nutritional aspects of juvenile rheumatoid arthritis. Rheum Dis Clin North Am 17: 403–413, 1991. 843. Hafner R, Truckenbrodt H, Spamer M: Rehabilitation in children with juvenile chronic arthritis. Baillieres Clin Rheumatol 12: 329–361, 1998. 844. Takken T, van der NJ, Kuis W, et al: Physical activity and health related physical fitness in children with juvenile idiopathic arthritis. Ann Rheum Dis 62: 885–889, 2003. 845. Klepper SE: Exercise and fitness in children with arthritis: evidence of benefits for exercise and physical activity. Arthritis Rheum 49: 435–443, 2003. 846. Frigo C, Bardare M, Corona F, et al: Gait alterations in patients with juvenile chronic arthritis: a computerized analysis. J Orthop Rheumatol 9: 82–90, 1996. 847. Lovell DJ, Walco GA: Pain associated with juvenile rheumatoid arthritis. Pediatr Clin North Am 36: 1015–1027, 1989. 848. Schanberg LE, Sandstrom MJ: Causes of pain in children with arthritis. Rheum Dis Clin North Am 25: 31–53, 1999. 849. Arden GP, Ansell BM: Surgical Management of Juvenile Chronic Polyarthritis. London, Academic Press, 1978. 850. Arden GP: Surgical treatment of juvenile rheumatoid arthritis. Ann Chir Gynaecol Suppl 198: 103–109, 1985. 851. Greene WB: Surgical alternatives for adolescents with severe arthritis. Bull Rheum Dis 43: 3–5, 1994. 852. Pahle JA: Orthopaedic management of juvenile chronic arthritis (JCA). Z Rheumatol 55: 376–387, 1996. 853. Swann M: The surgery of juvenile chronic arthritis. An overview. Clin Orthop 70–75, 1990. 854. Hamalainen M: Surgical treatment of juvenile rheumatoid arthritis. Clin Exp Rheumatol 12 (Suppl 10): S107–S112, 1994. 855. Price CT: Are we there yet? Management of limb-length inequality. J Pediatr Orthop 16: 141–143, 1996. 856. Simon S, Whiffen J, Shapiro F: Leg-length discrepancies in monoarticular and pauciarticular juvenile rheumatoid arthritis. J Bone Joint Surg Am 63: 209–215, 1981. 857. Rydholm U, Brattstrom H, Bylander B, et al: Stapling of the knee in juvenile chronic arthritis. J Pediatr Orthop 7: 63–68, 1987. 858. Granberry WM: Synovectomy in juvenile rheumatoid arthritis. Arthritis Rheum 20: 561–564, 1977. 859. Jacobsen ST, Levinson JE, Crawford AH: Late results of synovectomy in juvenile rheumatoid arthritis. J Bone Joint Surg Am 67: 8–15, 1985. 860. Rydholm U, Elborgh R, Ranstam J, et al: Synovectomy of the knee in juvenile chronic arthritis. A retrospective, consecutive follow-up study. J Bone Joint Surg Br 68: 223–228, 1986. 861. Swann M, Ansell BM: Soft-tissue release of the hips in children with juvenile chronic arthritis. J Bone Joint Surg Br 68: 404–408, 1986. 862. Kvien TK, Pahle JA, Hoyeraal HM, et al: Comparison of synovectomy and no synovectomy in patients with juvenile rheumatoid arthritis. A 24-month controlled study. Scand J Rheumatol 16: 81–91, 1987. 863. Hafner R, Pieper M: Arthroscopic synovectomy of the knee joint in chronic juvenile arthritis. Z Rheumatol 54: 165–170, 1995. 864. Parvizi J, Lajam CM, Trousdale RT, et al: Total knee arthroplasty in young patients with juvenile rheumatoid arthritis. J Bone Joint Surg Am 85: 1090–1094, 2003. 865. Surgeon General’s Report: Children with Special Health Care Needs. Commitment to Family-Centered, Community-Based, Coordinated Care. Bethesda, MD: U.S. Department of Health and Human Services, 1987. 866. Athreya BH, McCormick MC: Impact of chronic illness on families. Rheum Dis Clin North Am 13: 123–131, 1987. 867. Rapoff MA, Lindsley CB, Christophersen ER: Parent perceptions of problems experienced by their children in complying with treatments for juvenile rheumatoid arthritis. Arch Phys Med Rehabil 66: 427–429, 1985. 868. Kroll T, Barlow JH, Shaw K: Treatment adherence in juvenile rheumatoid arthritis—a review. Scand J Rheumatol 28: 10–18, 1999. 869. Frank RG, Hagglund KJ, Schopp LH, et al: Disease and family contributors to adaptation in juvenile rheumatoid arthritis and juvenile diabetes. Arthritis Care Res 11: 166–176, 1998. 870. Frank RG, Thayer JF, Hagglund KJ, et al: Trajectories of adaptation in pediatric chronic illness: the importance of the individual. J Consult Clin Psychol 66: 521–532, 1998. 871. Pless IB, Satterwhite B, Van Vechten D: Division, duplication and neglect: patterns of care for children with chronic disorders. Child Care Health Dev 4: 9–19, 1978. 872. Oen K, Malleson PN, Cabral DA, et al: Early predictors of longterm outcome in patients with juvenile rheumatoid arthritis: subset-specific correlations. J Rheumatol 30: 585–593, 2003. 873. Dempster H, Porepa M, Young N, et al: The clinical meaning of functional outcome scores in children with juvenile arthritis. Arthritis Rheum 44: 1768–1774, 2001. 874. Michels H: The chronically ill child with rheumatoid arthritis. Z Arztl Fortbild Qualitatssich 91: 219–226, 1997.
C H A P T E R 875. Michels H, Hafner R, Morhart R, et al: Five year follow-up of a prospective cohort of juvenile chronic arthritis with recent onset. Clin Rheumatol 6 Suppl 2: 87–92, 1987. 876. Billings AG, Moos RH, Miller JJ III, et al: Psychosocial adaptation in juvenile rheumatic disease: a controlled evaluation. Health Psychol 6: 343–359, 1987. 877. Fantini F, Gerloni V, Gattinara M, et al: Remission in juvenile chronic arthritis: a cohort study of 683 consecutive cases with a mean 10 year followup. J Rheumatol 30: 579–584, 2003. 878. Aptekar RG, Decker JL, Bujak JS, et al: Adult onset juvenile rheumatoid arthritis. Arthritis Rheum 16: 715–718, 1973. 879. Hill RH, Herstein A, Walters K: Juvenile rheumatoid arthritis: follow-up into adulthood—medical, sexual and social status. Can Med Assoc J 114: 790–796, 1976. 880. FitzGerald O, Bresnihan B: Juvenile chronic arthritis: spectrum of disease in an adult rheumatology department. Ir J Med Sci 155: 266–271, 1986. 881. Zak M, Pedersen FK: Juvenile chronic arthritis into adulthood: a long-term follow-up study. Rheumatology (Oxf) 39: 198–204, 2000. 882. Guillaume S, Prieur AM, Coste J, et al: Long-term outcome and prognosis in oligoarticular-onset juvenile idiopathic arthritis. Arthritis Rheum 43: 1858–1865, 2000. 883. Lomater C, Gerloni V, Gattinara M, et al: Systemic onset juvenile idiopathic arthritis: a retrospective study of 80 consecutive patients followed for 10 years. J Rheumatol 27: 491–496, 2000. 884. Spiegel LR, Schneider R, Lang BA, et al: Early predictors of poor functional outcome in systemic-onset juvenile rheumatoid arthritis: a multicenter cohort study. Arthritis Rheum 43: 2402–2409, 2000. 885. Hafner R, Truckenbrodt H: Course and prognosis of systemic juvenile chronic arthritis—retrospective study of 187 patients. Klin Padiatr 198: 401–407, 1986. 886. Schuchmann L, Michels H, Morhart R, et al: Prospective observation study on the clinical course of chronic juvenile arthritis (JCA). Monatsschr Kinderheilkd 134: 164–167, 1986. 887. Packham JC, Hall MA: Long-term follow-up of 246 adults with juvenile idiopathic arthritis: functional outcome. Rheumatology (Oxf) 41: 1428–1435, 2002. 888. Packham JC, Hall MA, Pimm TJ: Long-term follow-up of 246 adults with juvenile idiopathic arthritis: predictive factors for mood and pain. Rheumatology (Oxf) 41: 1444–1449, 2002. 889. Packham JC, Hall MA: Long-term follow-up of 246 adults with juvenile idiopathic arthritis: social function, relationships and sexual activity. Rheumatology (Oxf) 41: 1440–1443, 2002. 890. Packham JC, Hall MA: Long-term follow-up of 246 adults with juvenile idiopathic arthritis: education and employment. Rheumatology (Oxf) 41: 1436–1439, 2002. 891. Minden K, Niewerth M, Listing J, et al: Long-term outcome in patients with juvenile idiopathic arthritis. Arthritis Rheum 46: 2392–2401, 2002. 892. Oen K: Long-term outcomes and predictors of outcomes for patients with juvenile idiopathic arthritis. Best Pract Res Clin Rheumatol 16: 347–360, 2002. 893. Foster HE, Marshall N, Myers A, et al: Outcome in adults with juvenile idiopathic arthritis: a quality of life study. Arthritis Rheum 48: 767–775, 2003. 894. Ravelli A: Toward an understanding of the long-term outcome of juvenile idiopathic arthritis. Clin Exp Rheumatol 22: 271–275, 2004. 895. Bowyer SL, Roettcher PA, Higgins GC, et al: Health status of patients with juvenile rheumatoid arthritis at 1 and 5 years after diagnosis. J Rheumatol 30: 394–400, 2003. 896. Brunner HI, Giannini EH: Health-related quality of life in children with rheumatic diseases. Curr Opin Rheumatol 15: 602–612, 2003. 897. Selvaag AM, Flato B, Lien G, et al: Measuring health status in early juvenile idiopathic arthritis: determinants and responsiveness of the child health questionnaire. J Rheumatol 30: 1602–1610, 2003. 898. Oen K, Malleson PN, Cabral DA, et al: Disease course and outcome of juvenile rheumatoid arthritis in a multicenter cohort. J Rheumatol 29: 1989–1999, 2002. 899. Oen K, Reed M, Malleson PN, et al: Radiologic outcome and its relationship to functional disability in juvenile rheumatoid arthritis. J Rheumatol 30: 832–840, 2003. 900. LeBovidge JS, Lavigne JV, Donenberg GR, et al: Psychological adjustment of children and adolescents with chronic arthritis: a meta-analytic review. J Pediatr Psychol 28: 29–39, 2003. 901. Hutchison T: The classification of disability. Arch Dis Child 73: 91–93, 1995. 902. Peterson LS, Mason T, Nelson AM, et al: Psychosocial outcomes and health status of adults who have had juvenile rheumatoid arthritis: a controlled, population-based study. Arthritis Rheum 40: 2235–2240, 1997. 903. Flato B, Lien G, Smerdel A, et al: Prognostic factors in juvenile rheumatoid arthritis: a case-control study revealing early predictors and outcome after 14.9 years. J Rheumatol 30: 386–393, 2003. 904. Flato B, Aasland A, Vinje O, et al: Outcome and predictive factors in juvenile rheumatoid arthritis and juvenile spondyloarthropathy. J Rheumatol 25: 366–375, 1998. 905. Aasland A, Flato B, Vandvik IH: Psychosocial outcome in juvenile chronic arthritis: a nine-year follow-up. Clin Exp Rheumatol 15: 561–568, 1997.
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
259
906. Carter BD, Kronenberger WG, Edwards JF, et al: Psychological symptoms in chronic fatigue and juvenile rheumatoid arthritis. Pediatrics 103: 975–979, 1999. 907. Huygen AC, Kuis W, Sinnema G: Psychological, behavioural, and social adjustment in children and adolescents with juvenile chronic arthritis. Ann Rheum Dis 59: 276–282, 2000. 908. Noll RB, Kozlowski K, Gerhardt C, et al: Social, emotional, and behavioral functioning of children with juvenile rheumatoid arthritis. Arthritis Rheum 43: 1387–1396, 2000. 909. von Weiss RT, Rapoff MA, Varni JW, et al: Daily hassles and social support as predictors of adjustment in children with pediatric rheumatic disease. J Pediatr Psychol 27: 155–165, 2002. 910. Hagglund KJ, Clay DL, Frank RG, et al: Assessing anger expression in children and adolescents. J Pediatr Psychol 19: 291–304, 1994. 911. Cassidy JT, Johnson JC, Hewett JE, et al: Parental distress in families of children with juvenile rheumatoid arthritis. Arthritis Rheum 37: S405, 1994. 912. Press J, Neumann L, Uziel Y, et al: Assessment of quality of life of parents of children with juvenile chronic arthritis. Clin Rheumatol 21: 280–283, 2002. 913. Wagner JL, Chaney JM, Hommel KA, et al: The influence of parental distress on child depressive symptoms in juvenile rheumatic diseases: the moderating effect of illness intrusiveness. J Pediatr Psychol 28: 453–462, 2003. 914. Gerhardt CA, Vannatta K, McKellop JM, et al: Comparing parental distress, family functioning, and the role of social support for caregivers with and without a child with juvenile rheumatoid arthritis. J Pediatr Psychol 28: 5–15, 2003. 915. Reiter-Purtill J, Gerhardt CA, Vannatta K, et al: A controlled longitudinal study of the social functioning of children with juvenile rheumatoid arthritis. J Pediatr Psychol 28: 17–28, 2003. 916. Allaire SH, DeNardo BS, Szer IS, et al: The economic impacts of juvenile rheumatoid arthritis. J Rheumatol 19: 952–955, 1992. 917. Spencer CH, Fife RZ, Rabinovich CE: The school experience of children with arthritis. Coping in the 1990s and transition into adulthood. Pediatr Clin North Am 42: 1285–1298, 1995. 918. Chamberlain MA, Rooney CM: Young adults with arthritis: meeting their transitional needs. Br J Rheumatol 35: 84–90, 1996. 919. White PH: Future expectations: adolescents with rheumatic diseases and their transition into adulthood. Br J Rheumatol 35: 80–83, 1996. 920. White PH: Resilience in children with disabilities—transition to adulthood. J Rheumatol 23: 960–962, 1996. 921. White PH: Success on the road to adulthood. Issues and hurdles for adolescents with disabilities. Rheum Dis Clin North Am 23: 697–707, 1997. 922. Baum J, Gutowska G: Death in juvenile rheumatoid arthritis. Arthritis Rheum Suppl 20: 253, 1977. 923. Bernstein B: Death in juvenile rheumatoid arthritis. Arthritis Rheum Suppl 20: 256, 1977. 924. Thomas E, Symmons DP, Brewster DH, et al: National study of cause-specific mortality in rheumatoid arthritis, juvenile chronic arthritis, and other rheumatic conditions: a 20 year followup study. J Rheumatol 30: 958–965, 2003. 925. Schuchmann L, Michels H, Renaud M, et al: Amyloidosis—a dreaded complication of juvenile chronic arthritis (JCA) (author’s transl). Klin Padiatr 193: 67–72, 1981. 926. Bywaters EGL: Deaths in juvenile chronic polyarthritis. Arthritis Rheum Suppl 20: 256, 1977. 927. Arden GP: Sepsis in juvenile chronic polyarthritis. In Arden GP, Ansell BM (eds): Surgical Management of Juvenile Chronic Polyarthritis. London, Academic Press, 1978, p 225. 928. David J, Vouyiouka O, Ansell BM, et al: Amyloidosis in juvenile chronic arthritis: a morbidity and mortality study. Clin Exp Rheumatol 11: 85–90, 1993. 929. Ansell BM: Chlorambucil therapy in juvenile chronic arthritis (juvenile idiopathic arthritis). J Rheumatol 26: 765–766, 1999. 930. Ozdogan H, Kasapcopur O, Dede H, et al: Juvenile chronic arthritis in a Turkish population. Clin Exp Rheumatol 9: 431–435, 1991. 931. Woo P: Amyloidosis in pediatric rheumatic diseases. J Rheumatol Suppl 35: 10–16, 1992. 932. Gwyther M, Schwarz H, Howard A, et al: C-reactive protein in juvenile chronic arthritis: an indicator of disease activity and possibly amyloidosis. Ann Rheum Dis 41: 259–262, 1982. 933. Burman SJ, Hall PJ, Bedford PA, et al: HLA antigen frequencies among patients with juvenile chronic arthritis and amyloidosis: a brief report. Clin Exp Rheumatol 4: 261–263, 1986. 934. Woo P, O’Brien J, Robson M, et al: A genetic marker for systemic amyloidosis in juvenile arthritis. Lancet 2: 767–769, 1987. 935. Michels H, Linke RP: Clinical benefits of diagnosing incipient AA amyloidosis in pediatric rheumatic diseases as estimated from a retrospective study. Amyloid 5: 200–207, 1998. 936. Linke RP, Gartner HV, Michels H: High-sensitivity diagnosis of AA amyloidosis using Congo red and immunohistochemistry detects missed amyloid deposits. J Histochem Cytochem 43: 863–869, 1995. 937. Hawkins PN, Myers MJ, Lavender JP, et al: Diagnostic radionuclide imaging of amyloid: biological targeting by circulating human serum amyloid P component. Lancet 1: 1413–1418, 1988.
260
C H A P T E R
9 C H R O N I C A RT H R I T I S
IN
CHILDHOOD
938. Hawkins PN, Richardson S, Vigushin DM, et al: Serum amyloid P component scintigraphy and turnover studies for diagnosis and quantitative monitoring of AA amyloidosis in juvenile rheumatoid arthritis. Arthritis Rheum 36: 842–851, 1993. 939. Jeremy R, Schaller J, Arkless R, et al: Juvenile rheumatoid arthritis persisting into adulthood. Am J Med 45: 419–434, 1968. 940. Ansell BM, Wood PHN: Prognosis in juvenile chronic polyarthritis. Clin Rheum Dis 2: 397–412, 1976.
941. Hanson V, Kornreich H, Bernstein B, et al: Prognosis of juvenile rheumatoid arthritis. Arthritis Rheum 20: 279–284, 1977. 942. Stoeber E: Prognosis in juvenile chronic arthritis. Follow-up of 433 chronic rheumatic children. Eur J Pediatr 135: 225–228, 1981. 943. Rapoff MA, Lindsley CB: The pain puzzle: a visual and conceptual metaphor for understanding and treating pain in pediatric rheumatic disease. J. Rheuatol 27 9Suppl 58); 29–33, 2000.
10
( HAP T E R
POLYARTHRITIS Ross E. Petty and James T. Cassidy
~
DEFINmONS In all systems of classification of childhood arthritis, arthritis that affects more than four joints during the first 6 months of disease is defined as polyarthritis. 1-3 In the classification of the International League of Associations for Rheumatology (ILAR),3 polyarthritis is further defined as rheumatoid factor (RF)-negative if tests for RF are negative, and RF-positive if RF is detected on two occasions at least 3 months apart. The lLAR classification requires the application of exclusion criteria (Table 10-1). This chapter considers both RF-negative and RF-positive polyarthritis.
a later peak between the ages of 6 and 12 years. HIt is likely that the older group represents disease that is similar to, if not identical to, adult rheumatoid arthritis.
Sex Ratio Polyarthritis is predominantly a disease of girls, and the reported ratio of girls to boys using the ACR criteria was 3.5 to 1,9 In a study using the EULAR criteria,6 this ratio was 4.5:1.
Geographic and Racial Distribution EPIDEMIOLOGY Polyarthritis accounts for approximately 30% of patients with juvenile idiopathic arthritis OIA). A small proportion of these patients (perhaps 3% to 5%)4 are RF positive, although this proportion is as high as 42% in Native Canadian children. s
Incidence and Prevalence There are no large studies of the incidence and prevalence of polyarthritis, and estimates derived from reported investigations vary widely. In one study, Moe and Rygg6 estimated the incidence of polyarthritis (RFpositive or -negative) in northern Norway at approximately, 8.9/100,000 children per year and the point prevalence at 54.2/100,000 (European League Against Rheumatism [EULAR] criteria). In a trans-Canadian registry of patients seen in pediatric rheumatology clinics,7 polyarticular juvenile rheumatoid arthritis ORA) had an incidence of 0.49/100,000 children per year (American College of Rheumatology [ACR] criteria). These great discrepancies are not readily explicable, although genetic differences may be important. Both studies were registry based; the number of patients in the Norwegian study was only 43, whereas 154 children with polyarthritis were included in the Canadian study.
Age at Onset Using the ACR criteria for polyarticular JRA, the distribution of ages at onset appears to be biphasic (Fig. 10-1), with an early peak between the ages of 1 and 4 years and
Boyer and colleagues lO suggested that the incidence of RF-positive polyarthritis (ACR criteria) is increased in Southeast Alaska. In an extensive review of the epidemiology of childhood arthritis, Oen and Cheang ll analyzed the reported proportion of children with polyarthritis in different racial groups. Polyarthritis accounted for a much higher proportion of East Indian (61%), and North American Indian (64%) children with chronic arthritis, compared with white children (27%).
ETIOLOGY AND PATHOGENESIS The etiology of polyarthritis is unknown. The immunoinflammatory pathogenesis is multifactorial and is discussed in Chapter 9. No environmental agent has been consistently associated with polyarthritis, but it is likely that genetic polymorphisms play an important role, particularly in RF-positive polyarthritis. Interleukins, cytokines, and other products of inflammatory cells, such as macrophages and T lymphocytes, are undoubtedly involved in disease pathogenesis (see Chapter 3) A pro-inflammatory cytokine profile predominates. Most studies of cytokines in children with polyarthritis have not distinguished between children with and without RF. In general, the soluble interleukin-2 receptor (SIL-2R) is moderately increased in the blood J2-16 and synovial fluid, J6 and its level correlates with the activity of the arthritis. There is increased IL-1a16 in the blood, and IL1[3,16 IL-6, IL-1a, and tumor necrosis factor-a (TNF-a)l7 in the synovial fluid. The soluble TNF-a receptor p55 is increased in both,12.IH but the ratio of sTNF to TNF-a may be low. 19 Serum levels of IL-6 are higher in polyarthritis
261
262
C HAP T E R
• TABLE 10 1
10
POLYARTHRITIS
ILAR Crilerid for C1dssificatioll of Polyarthritis
RF-Negatlve Polyarthrftls Arthritis affecting five or more joints during the first 6 mo of disease; a test for RF is negative Exclusions: • Psoriasis or a history of psoriasis in the patient or first-degree relative • Arthritis in an HLA-B27-positive male beginning after the sixth birthday • Ankylosing spondylitis, enthesitis-related arthritis, sacroiliitis with inflammatory bowel disease, Reiter's syndrome, or acute anterior uveitis or a history of one of these disorders in a firstdegree relative • IgM RF on at least two occasions at least 3 mo apart • The presence of systemic ]IA in the patient
RF-Posltlve Polyarthrftls Arthritis affecting five or more joints during the first 6 mo of disease; two or more tests for RF at least 3 mo apart during the first 6 mo of disease are positive Exclusions: • Psoriasis or a history of psoriasis in the patient or first-degree relative • Arthritis in an HLA-B27-positive male beginning after the sixth birthday • Ankylosing spondylitis, enthesitis-related arthritis, sacroiliitis with inflammatory bowel disease, Reiter's syndrome, or acute anterior uveitis or a history of one of these disorders in a firstdegree relative • The presence of systemic ]IA in the patient IgM, immunoglobulin M; lLAR, International League of Associations for Rheumatology; ]lA, juvenile idiopathic artbritis; RF, rheumatoid factor,
B cells are not necessary for the development of inflammatory joint disease such as polyarthritis, and children with agammaglobulinemia may develop inflammatory joint disease. Immune complexes were detected in 64% of children with polyarticular JRA, especially those with active disease. 23 In 24%, immunoglobulin A (IgA)-containing immune complexes were present, and in synovial fluid immune complexes containing IgA, but not IgM, were detected.
GENETIC BACKGROUND Polyarthritis is seldom familial; if two siblings are affected with ]IA, concordance for polyarticular onset type is 42%.9 The human leukocyte antigen alleles HLA-Dw4, -DR1, and -DR4 are risk factors for polyarthritis. 24- 29 DRI (DQA1*OlOl) is also a risk factor for polyarthritis in older children and is associated with TCR-~6S1 null alleles. 24 The strong association between RF-seropositive polyarthritis and DR4-related susceptibility alleles and their shared epitope parallels that in RF-seropositive rheumatoid arthritis (RA) in adults. White children with RFseropositive polyarthritis have a high frequency of two copies of alleles of DR4, particularly for the combinations of Dw4 (DRB1*0401) and Dw14 (DRB1*0404)Y·29 Data from a study of Native Canadian children indicated that DRB1*0901 may be a risk factor in that population, whereas DRBl*08 alleles were possibly protective. 50 Inconsistent HLA typing has been found in RF-negative polyarthritis. Dw4 is only slightly increased in frequency,31 but there are increases in the frequencies of DRS, DR8, and DP3Y-35 These data support the understanding that children with RF have a disease that is different from that of children without RF, but they also suggest that RF-negative polyarthritis may, in itself, be quite heterogeneous. Genes outside the major histocompatibility locus also play a part in predisposition to ]IA, although most studies have examined these relationships in systemic arthritis or oligoarthritis. 36
CLINICAL MANIFESTATIONS
2
In children with polyarthritis, articular disease predominates, although systemic features occur in some patients. Children with RF-positive polyarthritis tend to have more severe disease than those who are RF negative, and they are subject to the development of rheumatoid nodules and, rarely, vasculitis.
Joint Disease (particularly in those with active disease) than in controls, but not as high as in those with systemic ]IA.20 Levels of IL12 were also associated with active disease in this study.20 Serum levels of adhesion molecules sE-selectin, sPselectin, and soluble intercellular adhesion molecule-l (sICAM-l) were not increased in children with active polyarticular ]IA. 21 RF may have a pathogenic role as a component of immune complexes, in fixing complement, and in causing damage to blood vessels in synovium,22 but it has been demonstrated that
Children with arthritis in five or more joints during the first 6 months of disease (Fig. 10-2), are classified as having arthritis of polyarticular onset by the ACR criteria!; as having either polyarticular onset (if RF negative) or ]RA (if RF positive) by the EULAR criteria 2; and as haVing RFnegative or RF-positive polyarthritis by the lLAR classitlcation (see Table 10-1).3 Onset may be acute, but it is more often insidious, with progressive involvement of additional joints. Morning stiffness or gelling after inactivity is an important indicator of active arthritis and may persist for hours, or occasionally all day. The arthritis
C HAP T E R
10 POLYARTHRITIS
263
Systemic Manifestations Systemic manifestations in children with polyarthritis include low-grade fever, anorexia, growth failure, and, occasionally, weight loss. Fatigue may be a prominent symptom, particularly in children with RF-positive polyarthritis. There may be slight-to-moderate hepatosplenomegaly or lymphadenopathy, but this is less prominent than in children with systemic JIA. Chronic asymptomatic uveitis develops less frequently 00%) than in early-onset oligoarticular disease and is more common in children with RF-negative compared with RF-positive polyarthritis (see Chapter 11). • figure 10-2 Symmetric polyarthritis affecting the metacarpophalangeal, proximal interphalangeal, and radiocarpal joints.
may be remittent or indolent, tends to be symmetric, and usually involves the large joints of the knees, wrists, elbows, and ankles. The cervical spine and temporomandibular joints are often involved in polyarticular JRA. Small-joint disease of the hands or feet may occur early or late in the course of the disease. 37 The interphalangeal joint of the thumb, the second and third metacarpophalangeal (MCP) joints, and the proximal interphalangeal (PIP) joints are most commonly involved; the other MCP and PIP joints are less frequently involved. The distal interphalangeal (DIP) joints are affected in 10% to 45% of children in association with involvement of the other small joints. The joints are swollen and warm but usually are not erythematous. Often, the digits exhibit soft tissue swelling between the joints as much as around them. Boutonniere deformities (PIP joint flexion and DIP hyperextension) and flexion contractures are more common than swan-neck deformities (PIP joint hyperextension and DIP flexion).38 Children with RF-positive polyarticular JIA have onset late in childhood or adolescence. These patients often develop a pattern of symmetric small and large joint involvement that resembles that of adult RA, with rheumatoid nodules, early onset of erosive synovitis, and a chronic course persisting well into adulthood. In children with RF-seronegative polyarthritis, there is less tendency for the disease to encompass a large number of joints, to be symmetric, to involve the small joints of the hands or feet, or to be associated with rheumatoid nodules. AnselP" found that the knees, wrists, and ankles were most commonly affected in children with polyarticular juvenile chronic arthritis OCA; EULAR criteria) and that the arthritis was usually symmetric. Approximately 20% of children had MCP joint disease, and a similar number, but not necessarily the same children, had PIP joint involvement. Approximately one fourth had involvement of the DIP joints. Joints of the second and third fingers were most commonly affected. Tenosynovitis of the dorsal flexor tendon sheaths of the hands occurred in one sixth of the patients, and in one or more of the other flexor tendons in one fifth. Involvement of a PIP joint or flexor tendon sheath was particularly common in children younger than 5 years of age. Arthritis of the metatarsophalangeal or PIP joints of the feet was also common.
Liem and Rosenberg40 studied growth in children with JRA and reported that in those with polyarthritis height-for-age Z scores (an expression of the number of standard deviations from the normal mean) fell slightly during the first 3 to 4 years of followup but returned to normal by 8 years after diagnosis. However, in those children with RF, the negative deviation was much more marked and prolonged, although it was statistically significantly different only at year 5. There was no demonstrable influence of corticosteroid use in this study. Evaluation of the Z scores for levels of alkaline phosphatase indicated that those children with RF polyarthritis had significantly higher scores by 4 years after diagnosis, an unexplained finding.
Nodules Rheumatoid nodules occur in 5% to 10% of children with JRA, almost always in those with RF-positive polyarthritis (Fig. 10-3).41-43 Nodules most frequently occur below the olecranon, but they also occur at other pressure points and on the digital flexor tendon sheaths, Achilles tendons, and occiput, as well as on the bridge of the nose in a child who wears glasses.
• AlIIIre 10-3 Multiple rheumatoid nodules over the metacarpophalangeal (MCP) and proximal interphalangeal joints were a constant feature of polyarticular JIA in this girl.They appeared over joints, pressure points, and tendon sheaths. Note also the radial deviation of the second and third MCP joints, the ulnar deviation of the radiocarpal joint, and the loss of extension of the interphalangeal joint of the thumb.
264
C HAP T E R
10 POLYARTHRITIS
Typical rheumatoid nodules are Hrm or hard, usually mobile, and nontender. The overlying skin may be erythematous. They may be solitary or multiple, may change in size over time, and they may persist for months to years. They are almost always associated with RF seropositivity, and in this respect they are generally regarded as a poor prognostic sign. Rheumatoid nodules must be distinguished from those of rheumatic fever and the so-called benign rheumatoid nodules that are not associated with objective arthritis (Table 10-2). Benign rheumatoid nodules or pseudorheumatoid nodules occur as isolated abnormalities in otherwise healthy children (Fig. 10-4).44-49 They are painless, may be single or multiple, and sometimes become quite large (greater than 5 cm). They occur especially over bony prominences such as the anterior tibia or scalp and are characterized by spontaneous regression and recurrence. They frequently recur after surgical excision. In a very few patients, arthritis may develop at an older age. 46 RF and antinuclear antibody (ANA) are absent. Histologically, they resemble the nodules of adult RA. Subcutaneous granuloma annulare, a relatively common disease in children, may be histologically indistinguishable from either rheumatoid or pseudorheumatoid nodules. 50 Its clinical appearance, location (shins, dorsum of the foot), umbilicated appearance, and the absence of other disease confirm the diagnosis. Other diseases, such as multicentric reticulohistiocytosis, are associated with subcutaneous nodules. 51
.' III TABLE 10 2 Nodules in Children with Rheumatic Diseases
Nodule Type
AssocIated Disease
Rheumatoid nodules Benign rheumatoid nodules Deep granuloma annulare Rheumatic fever nodules Extensor sheath nodules Calcinotic nodules
Polyarticular JRA No associated disease No associated disease Acute rheumatic fever Scleroderma Scleroderma Dermatomyositis Postinfectious Polyarteritis nodosa
Erythema nodosum Superficial aneurysms
Vasculitis Rheumatoid vasculitis is rare and occurs most often in the older child with RF-positive polyarthritis (Table 10-3; Fig. 10-5). This devastating, widespread complication involves small to medium-sized vessels and must be distinguished from the more frequent benign digital vasculitis (see Figure 9-8) and may occasionally be associated with vascular calcification that is apparent on radiographs along the course of the digital arteries. 52 •53 A second skin change has been described in children with polyarticular JRA.54 This consists of a dark coloration of the skin over the PIP joints of the fingers. The presence of this finding may reflect disease chronicity. In children with tender joints, retention keratosis may simulate a pigmented lesion.
Felty Syndrome Felty's syndrome, the presence of splenomegaly and neutropenia in a patient with RF-positive polyarthritis, is very rare in children and youth. 55
Cardiac Disease Small pericardiaI effusions may be detected on echocardiographs, but clinically evident pericarditis or pleuritiS is infrequent. Cardiac valve disease is an uncommon but important complication. Leak and associates 56 reported the occurrence of aortic regurgitation in four children with
I!' III
TABLE 10-3
Clinical Features of Rheumatoid Vaslllhlis
Fever Peripheral neuropathy Cutaneous ulcers Digital arteritis Raynaud's phenomenon Gastrointestinal hemorrhage Mesenteric thrombosis Myocardial infarction Nephritis
• Figure 10-4 A, Alarge benign rheumatoid nodule overlies the tibial tuberde in this young boy. 8,The lesion of granuloma annulare is histologically indistinguishable from the benign rheumatoid nodule shown in A. (8, Courtesy of Dr. J. Prendiville.)
C HAP T E R
• FIgure 10-5 Systemic. necrotizing vasculitis in a 6-year-old boy with systemic. JIA. He developed widespread cutaneous and visceral vasculitis that led to his death 1 year after onset of disease.
RF-positive polyarthritis. Several other instances of aortic or mitral valve regurgitation have also been reported. 57.5S
Pulmonary Disease Pulmonary disease is rare, but interstitial pneumonitis59 and bronchiolitis obliterans6o have been reported. Rarely, pulmonary disease precedes the onset of arthritis. 60 A familial form of deforming RF-positive polyarthritis with pulmonary fibrosis and renal and cutaneous vasculitis has been reported,61 but it is unlikely that this represents simple JIA.
DIAGNOSIS The differential diagnosis of polyarthritis is considerably different from that of oligoarthritis (Table 10-4). Septic polyarthritis is unusual (see Chapter 28), although arthritis caused by Neisseria gonorrhoeae may have an early migrato!)' polyarticular phase, and infectious arthritis in children may involve more than one joint. Lyme disease may be polyarticular, but it can usually be differentiated
II III .-
TABU 10-4 Poly,nthrilis
Differential Diagnosis of Inflammatory
RF-Negative
RF-Posltlve
]IA polyarthritis RF-negative Enthesitis-related arthritis Inflammatory bowel disease Systemic ]IA Psoriatic ]IA Polyarthritis related to infection Lyme disease Reactive arthritis Sarcoidosis
]TA polyarthritis RF-positive Systemic lupus erythematosus
.rIA. juvenile idiopathic arthritis; RF. rheumatoid factor.
10
POLYARTHRITIS
265
from polyarticular JIA by its intermittent pattern of activity and the accompanying cutaneous, neurologic, and cardiac abnormalities (see Chapter 29). The onset of polyarthritis in a preadolescent or adolescent girl should suggest the possible diagnosis of systemic lupus erythematosus (SLE) (see Chapter 16), The arthritis of SLE may mimic that of JIA, and without serologic studies the correct diagnosis may be difficult or impossible to make until the later occurrence of one of the more characteristic clinical hallmarks of SLE: butterfly rash, alopecia, nephritis, central nervous system disease, Raynaud's phenomenon, leukopenia, or hemolytic anemia. Any of these clinical findings, or the presence of an active urinary sediment in a child with polyarthritis, strongly suggests the diagnosis of SLE. This diagnosis would be supported by the presence of anti-dsDNA antibodies or hypocomplementemia. Ragsdale and colleagues62 presented the course of 10 children who developed SLE after an initial diagnosis of JRA. In all cases, anti-dsDNA antibodies were detectable before the development of clinical disease characteristic of SLE. Raynaud's phenomenon in the child with polyarthritis should always include a differential diagnosis such as scleroderma, SLE, or mixed connective tissue disease; in the authors' experience, Raynaud's phenomenon is very rarely associated with JRA. Although anemia is common in severe polyarthritis, it is not Coombs' test positive, and the presence of hemolytic anemia would support the diagnosis of another connective tissue disease, usually SLE. The differential diagnosis of polyarthritis also includes enthesitis-related arthritis (ERA) (or juvenile ankylosing spondylitis), in which large-joint oligoarthropathy of the lower extremities usually predominates at onset rather than disease of the axial skeleton (see Chapter 13). Transient pain in the lower back or buttock may be an early sign. The diagnosis of ERA should be considered particularly in a boy older than 10 years of age who has enthesitis or a family history of ankylosing spondylitis. RF and ANA are absent in children with ERA; on the other hand, the HLA-B27 antigen is present in more than 92% of children with juvenile ankylosing spondylitis, compared with 6% to 8% in the general North American white population. Occult inflammatory bowel disease may manifest as, or be complicated by, polyarthritis that is usually more transient than that of JRA. Whipple's disease can cause peripheral arthritis in children but is very rare (see Chapter 15). Dermatomyositis or scleroderma occasionally manifest with arthritis. The associated features of these illnesses generally lead quickly to a correct diagnosis. Dermatomyositis is almost invariably associated with a characteristic skin rash, and isolated polymyositis is rare. Scleroderma, on the other hand, may begin insidiously, and subtle subcutaneous calcifications may be misinterpreted as rheumatoid nodules. Malignant infiltration of bone or synovium can mimic polyarthritis, although in most instances the lesion is in juxta-articular bone rather than in the joint (see Chapter 39). Sometimes, however, joint effusions occur in children with malignancies. Nonarticular bone pain or tenderness, back pain with rest or activity, and severe constitutional symptoms may be warning clues. In addition
266
C HAP T E R
10
POLYARTHRITIS
to the systemic manifestations of malignancy, many children with hematologic malignancies have moderateto-severe anemia or elevation of the erythrocyte sedimentation rate (ESR) that is out of keeping with other features of their disease. The leukocyte count or platelet count may be low or normal, whereas in children with JIA, these laboratory measurements usually are increased. The serum level of lactic dehydrogenase is often very high. Radiographs of affected joints may suggest one of these disorders; however, the correct diagnosis in some children may be delayed for 3 to 6 months. Examination of the bone marrow is usually diagnostic (e.g., leukemia, neuroblastoma). Arthritis as a manifestation of malignancy becomes increasingly uncommon during late adolescence as red marrow ceases to occupy the metaphyses of the long bones. Sickle cell anemia in the very young child causes a dactylitis (hand-foot syndrome) that may mimic true arthritis, and in other children it causes microinfarcts that give rise to periostitis and periarthritis. It is reported that joint effusions adjacent to infarcted bone occur in 20% of acute sickle cell crises,63 and the coexistence of polyarthritis and sickle cell disease has been reported.64 Occasionally, the hypermobility syndrome or one of the Ehlers-Danlos syndromes presents as polyarthralgia. Swelling, pain, and temperature and color changes associated with reflex sympathetic dystrophy are usually unilateral and are readily differentiated from signs of JIA. Rarely, a child with a mucopolysaccharidosis, particularly Morquio's or Scheie's syndrome, has joint stiffness or bony enlargement suggesting polyarthritis. Children with immunodeficiencies, especially selective IgA deficiency and hypogammaglobulinemia, may have a polyarthritis that mimics JIA (see Chapter 33). Children with the velocardialfacial syndrome (22q11 deletion) develop chronic polyarthritis that may be clinically indistinguishable from JIA. 65 Jacobs and associates 66 and Athreya and Schumacher!>7 described children with familial hypertrophic synovitis who developed characteristic flexion contractures of the finger joints during the first few months of life. A bent thumb, the first diagnostic sign of this disorder, was often noted soon after birth. Symmetrical effusions affected the large joints. Progressive but minimal limitation of motion of the joints developed with age. There was usually no pain or systemic manifestation of fever or inflammation. Autosomal dominant inheritance was suggested in some families. Hypertrophic villi with giant cells but no other inflammatory infiltrates were present on histologic examination of the synovium. Synovial lining cells were hyperplastic, but vascular endothelial proliferation was not prominent. Radiographs demonstrated flattening of the proximal ossification centers of the femurs. Familial arthritis and camptodactyly has also been described6B and in some cases was associated with pericarditis. 69.7o The authors have seen two children with an isolated joint contracture resembling JIA in whom the eventual diagnosis was myositis ossificans progressiva (see Chapter 20). This diagnosis was not evident in either child until later in the course, when typical ossifications in muscle were observed radiographically. Familial osteochondritis dissecans also can mimic polyarticular JIA.7 1 The generalized soft tissue puffiness of the dorsa of the hands and feet in disorders such as lymphedema praecox, Noonan's syndrome, and Turner's syndrome
may mimic swelling caused by a combination of small joint effusions and tenosynovitis in children with polyarticular JIA.72 The absence of other evidence of inflammation in these patients should readily differentiate such conditions from polyarticular JIA. Other rare causes of diagnostic confusion include Poncet's disease (see Chapter 31)73 and the Torg osteolysis74 or related syndromes. 75
PATHOLOGY The histologic appearance of the synovium is nonspecific and does not materially differ from that seen in other chronic inflammatory arthritides. The synovial lining layer is hyperplastic, and there is villous hypertrophy. The subsynovial tissue is edematous. The vascular endothelium is hyperplastic and infiltrated by lymphocytes and plasma cells (see Figure 9-9). In adult RA, the primary infiltrating cell is the T lymphocyte, which may be distributed diffusely throughout the synovium or form nodules or germinal centers. 76 In addition, macrophages and dendritic cells abound, and, in late disease, B lymphocytes and RF-producing plasma cells are evident. Hypertrophy of synovial lining cells, fibroblasts, and blood vessels leads to the development of papillary fronds that may reach 2.5 x 0.2 cm in size. Extension of the inflammatory granulation tissue or pannus that spreads from the synovium and invades the cartilage and bone results in osteolysis, which is visible radiographically as erosion and subchondral cyst formation. Erosions occur preferentially in the "bare" areas of the joint, where bone is not covered by articular cartilage (see Chapter 2). They are irregular but sharply defined. Dissolution of the cartilage (chondrolysis) results from enzymatic digestion by neutral proteases, cathepsin, and collagenase from cells of the pannus. Immune complexes may also contribute to chondrolysis. 77 Metaplasia of the granulation tissue may result in formation of new cartilage, bone, or fibrous tissue, resulting in ankylosis. 76 End-stage disease is characterized by def01mity, subluxation, and fibrous or bony ankylosis. Joint destruction usually occurs much later in the course of JIA than in adult RA, and permanent joint damage is absent in many children with JIA even after years of chronic inflammation. (Newer studies with magnetic resonance imaging are likely to change this impression of the extent of joint damage in early disease.) The greater thickness of juvenile cartilage may offer some protection in this regard. The hyaline cartilage of the hip is destroyed in progressive stages during the course of severe]lA, and during healing it may be replaced by a fibrocartilaginous layer. 7R Rice bodies consist primarily of amorphous fibrous material, fibrin, and small amounts of collagen (Fig. 10-6).79--81 Viable cells are incorporated within this matrix and appear more normal than the synovial cells of the inflammatory foci. The majority of these cells resemble type B synovial lining cells, although a few type A cells are also visible. Residual blood vessels in some of these bodies attest to their former attachment to the synovial membrane. Subcutaneous nodules may be histopathologically typical of rheumatoid nodules (Fig. 10-7), or they may have a looser connective tissue framework resembling that of the nodules of rheumatic fever. 4 1,82 Classic rheumatoid nodules consist of three distinct zones: a central area of necrosis and granulation tissue, surrounded by a radially
C HAP T E R
10
POLYARTHRITIS
267
white blood cell counts and platelet counts and have a normocytic hypochromic anemia characteristic of chronic inflammation. These changes are usually more severe than those seen in children with oligoarthritis but less severe than in children with systemic arthritis (see also Chapter 9).
Autoantibodies Rheumatoid Factors
• figure 10-& Rice bodies from the knee of a 15-year-old boy with persistent arthritis of knees and ankle.
• figure 10-7 Photomicrograph of a rheumatoid nodule shows a central area of fibrinoid necrosis surrounded by a palisade of epithelioid cells and peripheral fibroblastic proliferation.
arranged palisade of connective tissue cells, which in turn is enveloped by chronic inflammatory cells. In ]IA, the central area of fibrinoid necrosis and the epithelioid palisades may be absent or less structured.
LABORATORY EXAMINAnON As is the case in other types of idiopathic arthritis, the
laboratory provides evidence of inflammation, is useful in excluding other diagnoses, and is important in classification, prognostication, and guiding therapy. There is no single diagnostic test, however.
Indicators of Inflammation Children with polyarthritis, particularly those with RFpositive polyarthritis, almost always have significant elevation of indices of inflammation such as ESR, C-reactive protein (CRP), and immunoglobulins. Many have elevated
The latex fixation test for the detection of classic IgM antiIgG RFs is positive in a variable percentage of children with polyarthritis. Classic RFs occur more frequently in children with polyarthritis than in any other category of ]IA. In the u.s. Pediatric Rheumatic Disease Registry,83 20 (3%) of 686 children with a polyarticular onset of]RA were RF positive. In a study by Oen and colleaguesll4 in Native Canadian children, the high frequency of RF-positive polyarticular disease (42%) was undoubtedly related to the high prevalence of HLA-DRB1 antigens bearing the RA shared epitope. RFs are unusual in a child younger than 7 years of age and are seldom helpful diagnostically at onset of disease. The diagnostic importance of RF seropositivity in a child with possible JRA is mitigated by the frequent occurrence of abnormal titers in the other connective tissue diseases of childhood, especially in SLE. In a study of the diagnostic utility of RF serology in children,8s RF tests were as likely to be positive in children with diseases other than ]RA as in those with JRA (Table 10-5). As a diagnostic aid, therefore, tests for RFs are of little utility. However, children with high titers of RFs most likely represent a subgroup distinct from the larger number of children with seronegative polyarthritis. The evidence for this hypothesis is not unequivocal, because studies often identify the seropositive group in retrospect. RFs are more common in children with later age at onset of arthritis and in those who are older, have subcutaneous rheumatoid nodules or articular erosions, or are in a poor functional class (Table 10_6).86 The percentage of children with RF seropositivity also rises progressively as the age at onset or duration of disease of the cohort group under study increases. These observations suggest that RFs might be a result rather than a determining event in children who go on to develop unremitting, disabling disease during the early adult years. Many studies have demonstrated other types of antiglobulins in the sera of children with ]RA or have reported more sensitive and specific tests for RFs. The
I=. ~
TABLE 10-5
Utility of Rheumatoid Factor as a Diagnosti( Test'
Juvenile Rheumatoid Arthritis Rheumatoid Fador Present Absent
Present
Absent
5
6 326
100
'Sensitivity = 5/105 (4.8%); specificity = 326/332 (98%). Modified from Eichenfield AH, Athteya BH, Doughty RA, Cebui RO: Utility of rheumatoid factor in the diagnosis of juvenile rheumatoid arthritis. Pediatrics 78: 480, 1986.
268
I!.
C HAP T E R
10
POLYART H R I TIS
TABLE 10-6 Clinital and Laboratory Correlations with RheumatoId Fador Positivity
Late age at disease onset Long duration of disease Poor functional capacity Rheumatoid nodules Rheumatoid vasculitis Erosions
majority of children with IgM RF-negative JRA can be shown to have IgG anti-IgG antibodies by immunosorbent techniques. A7 Miller and colleaguesHH employed five different immunosorbents to search for occult antiglobulins. Antiglobulins detected by binding to sepharose-linked globulin followed by acid elution were found in 8 normal children and in 52 children with JRA; only 4 children with JRA had significantly elevated levels. They concluded that the presence of these antiglobulins per se was not diagnostically helpful in distinguishing children with JRA. In the review by Lawrence and associates,H9 up to 75% of children with ]RA were shown to have "hidden RFs," defined as IgM 195 antiglobulins detected by acid elution of IgM-containing fractions of serum from a gel filtration column. Titers correlated with activity of the disease and did not differ significantly between polyarticular and oligoarticular disease. Most studies have focused on North American and European patients. In a survey of 43 children from Turkey,90 195 IgM hidden RFs were found in 56% of the patients (46% with oligoarthritis, 64% with polyarthritis, and 80% with systemic onset). The latex fixation test was positive in only one patient who had polyarticular disease. Hidden RFs were present in lower percentages in India91 .92 and in GreeceY3 RFs of IgA and IgE isotypes are reportedY4-96
Oen and colleagues99 reported the findings in a group of children with RF-negative and RF-positive polyarthritis. In early radiographs (taken within 2 years after disease onset), joint space narrowing (including decreased joint space, ankylosis, and carpal collapse) was demonstrated in 12% of 39 children in the RF-negative group and 31% of 21 children in the RF-positive group. In late films (taken at last follow-up, a median of 6.5 years after onset in RF-negative polyarthritis, and 8.7 years in RFpositive polyarthritis), the frequency of joint space narrowing had increased to 42% and 80%, respectively. Erosions and growth abnormalities likewise increased with time (Table 10-7). Details of the location of radiographic abnormalities are shown in Table 10--8 and Table 10-9. Pedersen and colleagues lOO studied the temporomandibular joint (TM]) using orthopantomograms in 169 consecutive patients with leA. Radiographic abnormalities of the TM] were more severe and more frequent in those with polyarthritis (65.8%) than in those with other onset types. More severe disease also correlated with early age at onset of arthritis and with long duration of disease. The presence of RF in young children was associated with severe TM] disease.
TREATMENT Polyarthritis, perhaps to a greater extent than in any other category of JIA, requires both intensive pharmacotherapy and the application of physical modalities such as physical therapy. Recognition of these diseases, particularly in the presence of RF positivity, should prompt early aggressive therapy to minimize joint damage and functional loss. The probability of a benign
Antinuclear Antibodies ANAs of unknown specificities are commonly present in children with polyarthritis. They are present in low to medium titers (up to 1:640, occasionally higher) in a proportion of these children. Low and colleagues97 found that 75% of children with RF-positive polyarthritis had antibodies to cyclic citrullinated peptides (CCP), although there was considerable variation depending on the substrate used in the assays, and these antibodies were also demonstrable at lower frequency in children with RF-negative polyarthritis, oligoarthritis, or systemic arthritis. Another study of 100 children with JRA (ACR criteria)9A showed that anti-CCP was present in half of those with RF-positive polyarthritis but in none of those with polyarticular RF-negative polyarthritis, oligoarthritis, or systemic arthritis. Standardization of the techniques used to measure anti-CCP antibodies is critical, and the clinical applicability of this laboratory investigation is not certain at present.
RADIOLOGIC EXAMINATION Radiologic changes often occur early and, particularly in children with RF, may be severe. Examples of typical changes are seen in Figures 10--8 through 10-10 (also see Figures 9-10, 9-14, and 9-17).
• Figure 10-8 Extensive ankylosis of the carpal and metacarpal joints in a 12-year-old girl 2 years after onset of polyarticular JlA.The entire carpus is ankylosed, and there are fusions between the carpals and the second and third metacarpals as well. There is also undergrowth of the fourth right metacarpal and disruption of the radiocarpal relationship.
C HAP T E R
11[·. TABLE to-7 -
10 POLYARTHRITIS
269
Radiographic Abnormalities in Polyarthritis
("'" of Patients)
Polyarthritis RF·Negatlve Abnonnallty Joint space nan'owing Erosions Growth abnormalities
PoIyartbrltls RF-PosItIve
Early
Late
Early
Late
(n=39)
(n = 54)
(n= 21)
(n = 28)
12
42
31
80
17
40
55
75
16
42
45
31
RF. rheumatoid factor, From Oen KG. Reed M, Malleson PN, et al: Radiologic outcome and its relationship to functional disability in iuvenile rheumatoid arthritis, .J Rheum 30: 832-840. 2003.
Medical Management
• fItpn 10-' Radiograph of the hand of a 15-year-old girl taken shortly before her death. Severe polyarthritis was present, with marked bony overgrowth and enlargement of the epiphyses. Erosions and destruction of the small joints were evident, with invagination of the abutting bones.The distal interphalangeal joints were also involved, and partial ankylosis of the carpus was present.
course of short duration is very low, and a prolonged disease course with the threat of significant joint damage should be anticipated. The effects of systemic manifestations of the disease must also be monitored carefully. The principles outlined in Chapter 8 should be applied.
• figure 10-10 Protrusio acetabuli of the left hip in a girl with longstanding JlA. Pain in the hip was accompanied by marked reduction in range of motion, although at the time the overall disease activity was minimal.
Initial treatment with nonsteroidal anti-inflammatory drugs (NSAlDs) is appropriate. Naproxen or ibuprofen is most commonly used in North America, but indomethacin is favored by some pediatric rheumatologists in Europe and elsewhere. Some physicians combine initial NSAID therapy with a disease-remittive agent, usually methotrexate, particularly in children with RF-positive polyarthritis, In any event, failure of NSAlDs to control the disease within 6 to 8 weeks should prompt the addition of methotrexate. This drug is usually given by mouth initially, in doses of 0.35 to 0.65 mg/kg/week, but in the absence of an adequate response, once an oral dose of 15 mg/week is reached, administration should be changed to the subcutaneous route. The response to methotrexate is usually excellent. IOt-103 Although methotrexate has become the drug of choice, leflunomide may have a place, although this is not yet established. 104 .105 In a study of 40 children from China with active polyarticular JRA,I04 leflunomide plus methotrexate was reported to be superior to methotrexate alone. In a North American multicenter trial, Silverman and colleagues lO5 reported that leflunomide alone was an effective agent in patients with polyarthritis for whom methotrexate had failed. The anti-TNF agents have joined the ranks of effective agents in children with polyarthritis,106-11O and, as experience with these and other biologic agents increases, it is likely that they will find an important place in the management of patients with methotrexate-resistant disease. In the double-blind, randomized, controlled trial conducted by the Pediatric Rheumatology Collaborative Study Group,106 etanercept in a dose of 0.4 mg/kg was given subcutaneously twice a week to all patients. Seventy-four percent of patients demonstrated a response by the core criteria and were entered into the double-blind study. During this phase of the study, patients receiving etanercept had many fewer disease flares, and, when flares did occur, they occurred much later than in the group receiving placebo. Follow-up studies indicated that etanercept offered sustained clinical improvement for longer than 2 years and was well tolerated. 107 ,108 The combination of methotrexate and etanercept was effective in a small group of children with
270
C HAP T E R
I!.
10
POLY ART H R IT I S
TABlE 10 R Pnlydrlhlllts
Lo(alion alld I rl'lllU'lI( Y (II II) of Rddioljr
Joint Space Narrowing location Cervical spine Shoulders Elbows Wrists Hands Hips Knees Ankles Feet
Early
Erosions
Late
Early
0 0 0
7 0 8
5
9 O·
6 6 5 6
3 4 0 <1 0
12 1 3 0
Nl'lj
Growth Abnormalities
Late
3 0 0 0 0
In
0 6 5 6 12 11 2 1 11
Early 0 0 0 4 4 0 1 0 2
Late 0 0 3
9 20 8 3 2
9
RF, rheumatoid factor. From Oen KG, Reed M, Malleson PN, et al: Radiologic outcome and its relationship to functional disability in juvenile rheumatoid arthritis. J Rheum 30: 832-840, 2003.
polyarthritis resistant to methotrexate and sulfasalazine or cyclosporine.109 A comparison of the efficacy and toxicity of etanercept and infliximab in children with active polyarthritis was reported by Lahdenne and colleagues yo Both agents were found to be very rapidly effective, and at 12 months Core Set 75 was achieved by 67% of patients. Five patients in the infliximab group withdrew because of lack of efficacy, as did one patient in the etanercept group. The patients were not randomized in this study, so it is uncertain whether one agent has greater benefit or toxicity than the other. Glucocorticoids are important as intra-articular therapy. Breit and colleagues 111 reported a longer median duration of response in children with ]CA who were RF negative (lOS weeks), than in those who were RF positive (63 weeks). Glucocorticoids have a limited role as systemic agents, particularly acting as a "bridge" until disease-modifying agents begin to have their effect. Drugs such as the gold compounds and penicillamine are seldom used, but hydroxychloroquine probably has a role as an adjunctive agent, used in combination with methotrexate (see Chapter 5). Autologous stem cell transplantation has not found a significant role in the management of polyarthritis,
I! III
TABLE 10 9 Pnlydrlhrilb
Cervical spine Shoulders Elbows Wrists Hands Hips Knees Ankles Feet
Physical and Occupational Therapy Children with polyarthritis, like those with other types of chronic arthritis, should be encouraged to participate in school and recreational activities with other children. Additional rest may be needed by some children. In general, these patients are quite capable of judging their own physical limitations. At the same time, joint protection should be promoted. Undesirable play is that which places undue stress on affected joints. Tricycle and bicycle riding and swimming are almost always helpful and do not add undue loading to the joints. Swimming has the added advantage of providing active range of motion and muscle strengthening without weight-bearing. Notwithstanding the advantages of active exercise, it is important to have a carefully designed passive therapy program: Children tend to function within the range of motion they have, not the range they should be trying to achieve. Physical therapy should be instituted as soon as the degree of inflammation subsides sufficiently to facilitate
I O(
Joint Space Narrowing Location
except in children with systemic ]IA and a polyarticular course (see Chapter 10).
Erosions
Growth Abnormalities
Early
Late
Early
Late
Early
Late
5 0 0 4 2 0 0 1 6
8 25 11 16 10 11 0 6 17
0 6 2 11 5 0 0 0 5
0 18 2 14 8 5 1 1 14
0 0 0 5 3 0 0 2 3
0 13 0 3 3 0 0 4 2
RF. rheumatoid factor. From Oen KG, Reed M, Malleson PN, et al: Radiologic outcome and its relationship to functional disability in juvenile rheumatoid arthritis. J Rheum 30: 832--840, 2003,
C HAP T E R
the child's cooperation. Physical therapy aimed at restoration of normal range of motion can be facilitated by pretreatment with an analgesic such as acetaminophen and the application of heat or ice. Major contractures are often more amenable to therapy after intra-articular injection of triamcinolone hexacetonide. A cast applied at the time of joint injection can make use of the conscious sedation used to inject the joint, and may be changed in 24 to 48 hours (serial casting). Arthritis of the small joints of the hands is a particularly important problem in children with polyarthritis and requires early and ongoing management. Loss of range of motion at the MCP and PIP joints is frequent, causing diminished tuck and grasp strength. Subluxation of the MCP joints may occur, particularly in children with RFpositive polyarthritis.
Surgery Surgical management of polyarthritis is less common than in the past, thanks to the effectiveness of methotrexate and, potentially, the biologic agents. Nonetheless, some children with resistant or untreated disease will require replacement of hips, knees or, occasionally, small joints of the hands. Such procedures are seldom needed in childhood, but in adults with long-standing disease of juvenile onset they add greatly to function and quality of life.
COURSE OF THE DISEASE AND PROGNOSIS The child most at risk for an unsatisfactory outcome is the one who has been referred late after onset or the one who has a relatively late age at onset and long duration of disease, early involvement of the small joints of the hands and feet, rapid appearance of erosions, unremitting inflammatory activity, RF seropositivity, and subcutaneous nodules (see Table 10-6).112 These children have the greatest number of joints involved, often 20 or more, and eventual disability is largely related to the extent of
,~.
1l\BLE 10-10
Prot....ls Remission' Before age 16 yr After age 16 yr CHAQ score 0 >0 but <0.5 O.S-l.S >1.5
POLYARTHRITIS
271
articular involvement. Widespread symmetrical involvement of the PIP and MCP joints of the hands or feet is characteristically associated in polyarthritis with a more guarded outlook than for disease that is confined to the large joints. This is the so-called adult pattern of JIA, and it is related to development of RF seropositivity. Hip disease occurs in approximately one half of the children and almost always is accompanied by persistent inflammatory diseasey,115 It often leads inexorably to destruction or abnormal development of the femoral heads and acetabula. This type of severe hip disease is a major cause of disability in JIA, and hip involvement is justifiably interpreted as a poor prognostic sign. Limitation of range of articular motion often develops early and is related to synovial proliferation, effusion, or muscle spasm. Later on, it may result from contractures of soft tissues, joint destruction, or ankylosis. Remission is unlikely if arthritis has persisted longer than 7 years. Onset of puberty has no relation to activity of the disease or likelihood of a remission. In the Cincinnati series, 45% of the patients still had active arthritis 10 years after onset. 1l6 Fantini and colleagues ll7 noted that 15.7% of their patients with polyarticular JCA were in remission at last visit, 8.3% had a temporary remission, but 75.9% had never had a remission. Oen and colleagues lB described the outcome of children with JRA in a multicenter cohort that included 80 children with RF-negative polyarticular JRA and 40 with RF-positive polyarticular JRA (Table 10-10). All had been diagnosed between 1977 and 1994 and had been monitored for a minimum of 5 years. Although the ACR criteria were used for classification, children with psoriatic arthritis, ERA, seronegative enthesitis arthritis syndrome, juvenile ankylosing spondylitis, or arthritis with inflammatory bowel disease were excluded. Oen's group concluded that for children with RF the disease was unremitting; only 25% of children with negative tests for RF had gone into remission by age 16 years. Furthermore, children who had not gone into remission by this age were likely to have ongoing active arthritis into their late 20s or early 30s. These quite recent data suggest that polyarthritis continues to be associated with significant morbidity and functional disability.
REFERENCES
Prognosis 01 Polyarthritis
RF-Negatlve Polyarticular IRA (n= SO)
10
RF-PosIUve Polyarticular IRA (n= 40)
19 (24%) 4 (5%) 1509%)
0 0 0
36% 31% 25% 8%
8% 34% 40% 18%
'Remission - absence of active arthritis while off medication for at least 2 yr (no. and "", of patients), CHAQ, Childhood Health Assessment Questionnaire; JRA, juvenile rheumatoid arthritis; RF, rheumatoid factor. Data modified from Oen K, Malleson PN, Cabral DA. et al: Early predictors of longterm outcome in patients with juvenile rheumatoid arthritis: subset-specifiC correlations. J Rheumatol 30: 585-593. 2003.
1. Brewer EJ. Bass J. Baum J, et al: Current proposed revision of JRA criteria. Arthritis Rheum 20 (Supp\); 195-199, 1977. 2. European League Against Rheumatism: EULAR Bulletin No.4: Nomenclature and Classification of Arthritis in Children, Basel. National Zeitung AG, 1977, 3. Petty RE, Southwood TR, Manners P, et al: International League of Associations for Rheumatology classification of juvenile idiopathic anhritis: Second Revision, Edmonton 2001. J Rheumatol 31: 390-392, 2004. 4. Bowyer S, Roettcher P: Pediatric rheumatology clinic popuiations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group, J Rheumatol 234: 1968--1974, 1996. 5, Oen K, Schroeder M, Jacobson K. et al: Juvenile rheumatoid arthritis in a Canadian First Nations (aborigina\) population: onset subtypes and HLA associations. J Rheumatol 25: 783-790, 1998. 6. Moe N, Rygg M: Epidemiology of juvenile chronic anhritis in nonhern Norway: a ten year retrospective srudy. Clin Exp Rheumatol 16: 99-101, 1998. 7, Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry. J Rheumatol 23: 1981-1987, 1996. 8. Sullivan DB, Cassidy JT. Petty RE: Pathogenic implications of age of onset in juvenile rheumatoid arthritis, Arthritis Rheum 18: 251-255, 1975. 9. Moroldo MB, Chaudhari M. Shear E, et al: Juvenile rheumatoid arthritis affected sibpairs: extent of clinical phenotype concordance. Arthritis Rheum 50: 1928--1934.2004.
272
C HAP T E R
10 POLYARTHRITIS
10. Boyer GS, Lanier AP, Templin OW, et al: Spondyloarrhropathy and rheumatoid althritis in Alaskan Yupik Eskimos. .I Rheumatol 17: 48~96, 1990. 11. Oen KG, Cheang M: Epidemiology of chronic arrhritis in childhood. Semin Arrhritis Rheum 26: 575-591, 1996. 12. Mangge H, Kenzian H, Gallistl S, et al: Serum cytokines in juvenile rheumatoid arrhritis: correlation with conventional inflammation parameters and clinical subtypes. Arrhritis Rheum 38: 211-220, 1995. 13. De Benedetti F, Massa M, Pignatti P, et al: Serum soluble imerleukin 6 OL-6) receptor and IL-6/soluble IL-6 receptor complex in systemic juvenile rheumatoid arthritis. J Coo Invest 93: 2114-2119, 1994. 14. De Benedetti F, Alonzi 1', Moretla A, et al: Interleukin 6 causes growth impairment in transgenic mice through a decrease in insulin-like growth factor-I: a model for stunted growth in children with chronic inflammation. .I Clin Invest 99: 643--650, 1997. 15. Lipnick RN, Sfikakis PP, Klipple GL, et al: Elevated soluble CD8 antigen and soluble interleukin-2 receptors in the Sera of patients with juvenile rheumatoid arthritis. Clin Immunol Immunopathol 68: 64--67, 1993. 16. Madson KL, Moore n, Lawrence .1M 1Il, et al: Cytokine levels in sentm and synovial fluid of patients with juvenile rheumatoid arthritis. J Rheumatol 21: 2359-2363, 1994. 17. Kutukculer N, Caglayan S, Aydogdu F: Study of pro-inflammatory (TNFalpha, IL-1 alpha, IL-6) and l' cell-derived ClL-2, IL-4) cytokines in plasma and synovial fluid of patients with juvenile chronic arthritis: correlations with clinical and laboratory parameters. Clin Rheumatol 17: 288-292, 1998. 18. Gattorno M, Picco P, Buoncompagni A, et al: Sentm p55 and p75 tumour necI'Dsis factor receptors as markers of disease activity in juvenile chronic arrhritis. Ann Rheum Dis 55: 243-247, 1996. 19. Rooney M, Varsani H, Marrin K, et al: Tumour necrosis factor alpha and its soluble receptors in juvenile chronic arthritis. Rheumatology (Oxf) 39: 432-438, 2000. 20. Yuilmaz M, Kendirli SG, Altintas 0, et al: Cytokine levels in sentm of patient swith juvenile rheumatoid arrhritis. Clin Rheumatol 20: 30--35, 2001. 21. DeBenedetti F, Vivarelli M, Pignatti P, et al: Circulating levels of soluble Eselectin, P-selecting and intercellular adhesion molecule-l in patients with juvenile idiopathic arrhritis. .I Rheumatol 27: 2246-2250, 2000. 22. Jarvis IN: Pathogenesis and mechanisms of inflammation in childhood rheumatic disease. Curr Opin Rheumatol 10: 45~67, 1998. 23. Agarwal V, Misra R, Aggarwal A: Immune complexes contain immunoglobulin A rheumatoid factor in serum and synovial t1uid of patients with polyarticular juvenile rheumatoid arrhritis. Rheumatology (Oxf) 41: 466-467, 2002. 24. Grom AA, von Knorre C, Murray KJ, et al: T-cell receptor BV6s1 null alleles and HLA-DR1 haplotypes in polyarricular outcome juvenile rheumatoid arthritis. Hum Immunol 45: 152-156, 1996. 25. Clemens LE, Albert E, Ansell BM: HLA studies in IgM rheumatoid-factor-positive arrhritis of childhood. Ann Rheum Dis 42: 431-434, 1983. 26. Forre 0, Dobloug JH, Hoyeraal HM, et al: HLA antigens in juvenile arthritis: genetic basis for the different subtypes. Arthritis Rheum 26: 35-.~8, 198327. Nepom BS, Nepom GT, Mickelson E, et al: Specific HLA-DR4-associated histocompatibility molecules characterize patients with seropositive juvenile rheumatoid arrhritis. J Clin Invest 74: 287-291, 1984. 28. Murray KJ, Moroido MB, Donnelly P, et al: Age-specific effects of juvenile rheumatoid arthritis-associated HLA alleles. Arrhritis Rheum 42: 1843-1853, 1999. 29. Vehe RK, Begovich AB, Nepom BS: HLA susceptibility genes in rheumatoid factor positive juvenile rheumatoid arrhritis. .I Rheumatol Suppl 26: 11-15, 1990. 30. Oen K, EI Gabalawy HS, Canvin .1M, et al: HLA associations of seropositive rheumatoid arrhritis in a Cree and Ojibway population. J Rheumatol 25: 2319-2323, 1998. 31. Morling N, Hellesen C, Jakobsen BK, et al: HLA-A, B, C, 0, DR antigens and primed lymphocyte typing (PLT) defined DP-antigens in juvenile chronic arrhritis. Tissue Antigens 17: 433-441, 1981. 32. Morling N, Friis J, Heilmann C, et al: HLA antigen frequencies in juvenile chronic arrhritis. ScandJ Rheumatol 14: 209-216, 1985. 33. Fernandez-Vina MA, Fink CW, Stastny P: HLA antigens in juvenile arthritis: pauciarticuiar and polyarricular juvenile arthritis are immunogenetically distinct. Arthritis Rheum 33: 1787-1794, 1990. 34. Arnaiz-Villena A, Gomez-Reino JJ, Gamir ML, et al: DR, C4, and Bf allotypes in juvenile rheumatoid arthritis. Al'lhritis Rheum 27: 1281-1285, 1984. 35. Gao X, Fernandez-Vina M, Olsen NJ, et al: HLA-DPBl'0301 is a major risk factor for rheumatoid factor-negative adult rheumatoid arrhritis. Al'lhritis Rheum 34: 1310--1312, 1991. 36. F0rre 0, Smerdel A: Genetic epidemiology of juvenile idiopathic arthritis. ScandJ Rheumatol 31:123-128, 2002. 37. Naidu SH, Ostrov BE, Pellegrini VD Jr: Isolated digital swelling as the initial presentation of juvenile rheumatoid arthritis. J Hand Surg Am 22: 653--657, 1997. 38. Chaplin 0, Pulkki 1', Saarimaa A, et al: Wrist and finger deformities in juvenile rheumatoid arthritis. Acta Rheumato! Scand 15: 206-223, 1969. 39. Ansell BM: Joint manifestations in children with juvenile chronic polyarthritis. Arthritis Rheum 20: 204-206, 1977. 40. LiemJJ, Rosenberg AM: Growth patterns in juvenile rheumatoid arthritis. Clin Exp Rheumatoi 21: 663--668, 2003. 41. Bywaters EGL, Glynn LE, Zeldis A: Subcutaneous nodules of Still's disease. Ann Rheum Dis 17: 278, 1958.
42. Bywaters EG, Cardoe N: Multiple nodules in juvenile chronic polyarrhritis. Ann Rheum Dis 31: 421, 1972. 43. Kaye BR, Kaye RL, Bobrove A: Rheumatoid nodules: review of the spectntm of associated conditions and proposal of a new classification, with a reporr of four seronegative cases. Am J Med 76: 279-292, 1984. 44. Altman RS, Caffrey PR: Isolated subcutaneous rheumatic nodules. Pediatrics 34: 869, 1964. 45. Burrington JD: "Pseudorheumatoid" nodules in children: report of 10 cases. Pediatrics 45: 473-478, 1970. 46. Simons FE, Schaller JG: Benign rheumatoid nodules. Pediatrics 56: 29--.l~, 1975. 47. Schaller JG: Benign rheumatoid nodules. Arthritis Rheum Suppl 20: 277, 1977. 48. Mesara BW, Brody GL, Oberman HA: "Pseudorheumatoid" subcutaneous nodules. Am J Clin Pathol45: 684--691, 1966. 49. Kabukcuoglu S, Tel N, Pasaoglu 0, Ilhan H: Benign rheumatoid nodules in chIldhood.Turk J Pediatr 41: 365-368, 1999. 50. Mersara BW, Brody GL, Oberman HA: "Pseudorheumatoid" subcutaneous nodules. Am J Clin Pathol 45: 684--691, 1966. 51. Havill S, Duffill M, Rademaker M: Multicentric reticulohistiocytosis in a child. Australas J Dermatol 40: 44-46, 1999. 52. Forsyth CC: Calcification of the digital arteries in a child with rheumatoid arthritis. Arch Dis Child 35: 296, 1960. 53. Reid MM, Fannin TF: Extensive vascular calcification in association with juvenile rheumatoid arrhritis and amyloidosis. Arch Dis Child 43: 607-610. 1968. 54. Gedalia A, Gewanter H, Baum J: Dark skin discoloration of finger joints in juvenile arthritis. J Rheumatol 16: 797-799, 1989. 55. Bloom BJ, Smith P, Alario AJ: Felty syndrome complicating juvenile rheumatoid arrhritis. J Pediatr Hematol Oncol 20: 511-513. 1998. 56. Leak AM, Millar-Craig MW, Ansell BM: Aortic regurgitation in seropositive juvenile arthritis. Ann Rheum Dis 40: 229-234, 1981. 57. Ozer S, Alehan 0, Ozme S, et al: Mitral and aortic insufficiency in polyarticular juvenile rheumatoid arthritis. Pediatr Cardiol15: 151-153, 1994. 58. Delgado EA, Petty RE, Malleson PN, et al: Aortic valve insuf!1ciency and coronary arrery narrowing in a child with polyarticular juvenile rheumatoid arthritis. J Rheumatol 15: 144-147, 1988. 59. Uziel Y, Hen B, Cordoba M, Wolach B: Lymphocytic interstitial pneumonitis preceding polyarticular juvenile rheumatoid arthritis. Clin Exp Rheumatol 16: 617-619, 1998. 60. DiKensoy 0, Bayram N, Bingol A, Filiz A: Bronchiolitis obliterans in a case of juvenile rheumatoid arthritis presented with pneumomediastinum. Respiration 69: 100--102, 2002. 61. Rohayem J, Leupold W, Paul K-D, Gahr M: Pulmonary fibrosis and other clinical manifestations of small vessel vasculitis in a family with seropositive juvenile rheumatoid arthritis. Pediatr Pulmonol 33: 65-70, 2002. 62. Ragsdale CG, Petty RE, Cassidy .IT, Sullivan DB: The clinical progression of apparent juvenile rheumatoid arrhritis to systemic lupus erythematosus. J Rheumatol 7: 50--55, 1980. 63. De Cuelaer K, Forbes M, Roper 0, Serjeant GR: Non-gouty arthritis in sickle cell disease: reporr of 37 consecutive cases. Ann Rheum Dis 43: 599-603, 1984. 64. Nistala K, Murray KJ: Co-existent sickle cell disease and juvenile rheumatoid arthritis: two cases with delayed diagnosis and severe destntctive arrhropathy. J Rheumatol 28: 2125-2128, 2001. 65. Davies K, Stiehm ER, Woo P, Murray K.f: Juvenile idiopathic polyarricular arrhritis and 19A deficiency in the 22qll deletion syndrome. J Rheumatol 28: 2326-2334, 2001. 66. Jacobs JC, Downey JA: Juvenile rheumatoid arthritis. In Downey, .lA, Low NL (eds); The Child with Disabling mness. Philadelphia, WB Saunders, 1974, p 5. 67. Athreya BH, Schumacher HR: Pathologic features of a familial arrhropathy associated with congenital flexion contractures of fingers. Arrhritis Rheum 21: 429-437, 1978. 68. Malleson P, Schaller JG, Dega F, et al: Familial arthritis and camptodactyly. Arthritis Rheum 24: 1199-1204, 1981. 69. Bulutlar G, Yazici H, Ozdogan H, et al: A familial syndrome of pericarditis. arrhritis, camptodactyly, and coxa vara. Arthritis Rheum 29: 436-438, 1986. 70. Laxer RM, Cameron BJ, Chaisson 0, et al: The camptodactyly-arrhropathypericarditis syndrome: case reporr and literature review. Arrhritis Rheum 29: 43~44, 1986. 71. Robinson RP, Franck WA, Carey EJ, et al: Familial polyarticular osteochondritis dissecans masquerading as juvenile rheumatoid arthritis. J Rheumatol 5: 190--194, 1978. 72. Zulian F, Schumacher HR, Calore A, et al: Juvenile arthritis in Turner's syndrome: a multicenter study. Clin Exp Rheumatol 16: 489-494, 1998. 73. Pugh MT, Southwood TR: Tuberculous rheumatism, Poncet disease: a sterile controversy? Rev Rhum Ed Fr 60: 855-860, 1993. 74. Eisenstein OM, Poznanski AK, Pachman LM: Torg osteolysis syndrome. Am J Med Genet 80: 207-212, 1998. 75. Singh JA, Williams CB, McAlister WH: Talo-patello-scaphoid osteolysis, synovitis, and shorr fourth metacarpals in sisters: a new syndrome' Am J Med Genet 121A: 118-125, 2003. 76. Harris ED Jr: Rheumatoid Al'lhritis. Philadelphia, WE Saunders 1987. 77. Cooke TDV: The interaction and local disease manifestations of immune complexes in arricular collagenous tissue. In Maroudas A. Holborow EJ (eds): Studies in Joint Disease. London, Pittman, 1980, p 158.
CHAPTER 78. Bernstein B, Forrester 0, Singsen B, et 011: Hip joint restoration in juvenile meumatoid arthritis. Anhritis Rheum 20: 1099--1104, 1977. 79. Berg E, Wainwright R, Barton B, et 011: On the nature of rheumatoid rice bodies: an immunologic, histochemical, and electron microscope study. Arthritis Rheum 20: 1343-1349, 1977. 80. Wynne-Roberts CR, Cassidy JT: Juvenile rheumatoid arthritis with rice bodies: light and electron microscopic studies. Ann Rheum Dis 38: 8-13, 1979. 81. Chung C, Coley BD, Martin LC: Rice bodies in juvenile rheumatoid arthritis. AJR Am J Roentgenol 170: 698-700, 1998. 82. Bennett GA, Zeller .TW, Bauer W: Subcutaneous nodules of rheumatoid arthritis and rheumatic fever: a pathologic study. Arch Pathol 30: 70, 1970. 83. Bowyer S, Roetlcher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group. J Rheumatol 23: 1968-1974, 1996. 84. Oen K, Schroeder M, Jacobson K, et 011: Juvenile rheumatoid arthritis in a Camldian First Nations (aboriginal) population: onset subtypes and HLA associations. J Rheumatol 25: 783-790, 1998. 85. Eichentield AH, Athreya BH, Doughty RA, Cebui RD: Utility of rheumatoid factor in the diagnosis of juvenile rheumatoid arthritis. Pediatrics 78: 48Q....484, 1986. 86. Cassidy./T, Valkenburg HA: A five year prospective study of rheumatoid factor tests in juvenile rheumatoid arthritis. Arthritis Rheum 10: 83-90, 1967. 87. Schlump lJ, Howard A, Ansell BM: IgG-anti-igG antibodies in juvenile chronic arthritis. Scand J Rheumatol 14: 65--{58, 1985. 88. Miller JJ 111, Olds-Arroyo L, Akasaka T: Antiglobulins in juvenile rheumatoid arthritis. Arthritis Rheum 20: 729-735, 1977. 89. Lawrence JM 3rd, Moore TL, Osborn TG, et 011: Autoantibody studies in juvenile rheumatoid arthritis. Semin Ar
10 POLYARTHRITIS
273
100. Pedersen TK, Jensen JJ, Melsen B, Herlin T: Resorption of the temporomandibular condylar bone according to subtypes of juvenile chronic arthritis. J Rheumatol 28: 2109--2115, 2001. 101. Giannini EH, Brewer EJ, Kuzmina N, et 011: Methotrexate in resistant juvenile rheumatoid arthritis: results of the U.S.A.-U.S.S.R. double-blind, placebo-controlled trial. The Pediatric Rheumatology Collaborative Study Group and the Cooperative Children's Study Group. N Engl J Med 326: 1043-1049, 1992. 102. Ruperto N, Murray KJ, Gerloni V, et 011: A randomized trial of parenteral methotrexate comparing an intermediate dose with a higher dose in children with juvenile idiopathic arthritis who failed to respond to standard doses of methotrexate. Arthritis Rheum 50: 2191-2201, 2004. 103. Alsufyani K, Ortiz-Alvarez 0, Cabral DA, et 011: The role of subcutaneous administration of methotrexate in children with juvenile idiopathic arthritis who have failed oral methotrexate. J Rheumatol 31: 179--182,2004. 104. Gao JS, Wu H, Tian J: Treatment of patients with juvenile rheumatoid arthritis with combination of leflunomide and methotrexate. Zhonghua Er Ke Za Zhi 41: 435-438, 2003. 105. Silverman E, Spiegel L, Hawkins 0, et 011: Long-term open-label preliminary study of the safety and efficacy of leftunomide in patients with polyarticular-course juvenile rheumatoid arthritis. Anhritis Rheum 52: 554-562, 2005. 106. Lovell OJ, Giannini EH, Reiff A, et 011: Etanercept in children with polyarticular juvenile rheumatoid arthritis. Pediatric Rheumatology Collaborative Study Group. N Engl J Med 342: 81D-811, 2000. 107. Lovell OJ, Giannini EH, Reiff A, et 011: Long-term efficacy and safety of etanercept in children with polyarticular-course juvenile rheumatoid arthritis: interim results from an ongoing multicenter, open-label, extended-treatment trial. Anhritis Rheum 48: 218-226, 2003. 108. Kietz DA, Pepmueller PH, Moore TL: Therapeutic use of etanercept in polyarticular course juvenile idiopathic arthritis over a two year period. Ann Rheum Dis 61: 171-173, 2002. 109. Schmeling H, Mathony K, John V, et 011: A combination of etanercept and methotrexate for the treatment of refractory juvenile idiopathic arthritis: a pilot study. Ann Rheum Dis 60: 410-412, 2001. 110. Lahdenne P, Vahasalo P, Honkanen V: Infliximab or etanercept in the treatment of children with refractory juvenile idiopathic arthritis: an open label study. Ann Rheum Dis 62: 245-247, 2003. 111. Breit W, Frosch M, Meyer U, et 011: A subgroup-specific evaluation of the efficacy of intraarticular triamcinolone hexacetonide in juvenile chronic arthritis. J Rheumato! 27: 2696--2702, 2000. 112. Oen K, Malleson PN, Cabral DA, et 011: Disease course and outcome of juvenile rheumatoid arthritis in a multicenter cohort. J Rheumatol 29: 1989--1999, 2002. 113. Oen K, Malleson PN, Cabral DA, et 011: Early predictors of longterm outcome in patients with juvenile rheumatoid arthritis: subset-specific correlations. J Rheumatol 30: 585-593, 2003. 114. Isdale IC: Hip disease in juvenile rheumatoid arthritis. Ann Rheum Dis 29: 603-608, 1970. 115. Blane CE, Ragsdale CG, Hensinger RN: Late effects of JRA on the hip. J Pediatr Orthop 7: 677-680, 1987. 116. Wallace CA, Levinson JE: Juvenile rheumatoid arthritis: outcome and treatment for the 1990s. Rheum Dis Clin North Am 17: 891-905, 1991. 117. Fantini F, Gerloni V, Gattinara M, et 011: Remission in juvenile chronic arthritis: a cohort study of 683 consecutive cases with a mean 10 year followup. J Rheumatol 30: 579-584, 2003.
C HAP T E R
II
OLIGOARTHRITIS Ross E. Petty and James T. Cassidy
iJtf DEFINITION Arthritis that affects four or fewer joints during the first 6 months of disease is called pauciarticular juvenile rheumatoid arthritis (IRA) by the American College of Rheumatology (ACR) criteria 1; pauciarticular juvenile chronic arthritis (ICA) by the European League Against Rheumatism (EULAR) criteria 2; or oligoarthritis by the International League of Associations for Rheumatology CILAR) criteria. 3 It is a chronic inflammatory arthritis of unknown origin that begins before the 16th birthday and lasts for at least 6 weeks. In the ILAR classification of juvenile idiopathic arthritis (IIA), oligoarthritis is further characterized as either persistent (if no more than four joints are affected during the disease course) or extended Cif, after the initial 6-month period, the total number of affected joints exceeds four). The ILAR classification also requires that patients who otherwise fulfill these criteria be excluded from the category if they have psoriasis, if there is a family history of psoriasis or a disease associated with the human leukocyte antigen allele HLA B27 in a first-degree relative, if the disease began in a male older than 6 years of age, or if two positive tests for rheumatoid factor (RF) were obtained at least 3 months apart. Such exclusions do not apply to the EULAR or ACR criteria (see Chapter 9) (Table 11-1). Oligoarthritis is a distinctly, if not uniquely, pediatric disease, and it is the most commonly diagnosed category of chronic arthritis among children in North America and Europe.
EPIDEMIOLOGY Oligoarthritis accounts for 50% to 60% of all cases of chronic arthritis in children, at least in North American and European white populations. Oen and Cheang 4 noted that the proportion of all children with chronic arthritis who had oligoarthritis (ACR or EULAR) was higher in North American and European patients (58%) than in East Indian (25%), North American Indian (26%), or other racial groups 01%). Several reviews of incidence and prevalence studies have been published.4-7
Incidence Reports of the incidence of oligoarthritis are difficult to interpret because of the variation in criteria used to
274
classify the patients. Using information from hospitals and community physicians, Andersson-Gare and colleaguesH determined an annual incidence of oligoarthritis (EULAR) of 7/100,000 children younger than 16 years of age in Sweden. Using the same criteria, a Norwegian study~ reported a somewhat higher incidence of 11.2/100,000/ year. It should be noted that 42% of these children were HLA B27-positive, strongly suggesting that children with enthesitis-related arthritis (ERA) or juvenile ankylosing spondylitis (IAS) were included in the group. In studies that used the ACR criteria, estimates of the incidence of pauciarticular ]RA have ranged from less than 1I100,000/year in]apan lO to more than I8/100,000/year in Finland. ll
Prevalence The prevalence of oligoarthritis in reported studies varies greatly depending on the diagnostic criteria used; whether the study was hospital, clinic, or community based6 ; and the geographic location of the study.7 Using information from hospitals and community physicians, Andersson-Gare and associates 8 found 146 children with oligoarthritis (EULAR) in a population of 400,600 children younger than 16 years of age, a prevalence of 36/100,000. In the study by Manners and Diepeveen,12 oligoarthritis fulfilling the EULAR criteria for ]CA was found in 9 of 2241 twelve-year old school children who were examined by the authors. This prevalence (4/1000) is markedly higher than that reported in other studies but may be closest to reality, because it was community based and verified by physical examination by a pediatric rheumatologist. It is possible that this high prevalence is not representative of the disease worldwide, however.
Age at Onset Oligoarthritis has a striking age at onset distribution, with a peak incidence between 2 and 4 years of age (Fig. 11-1).13 A small proportion of children with oligoarthritis have disease onset after this time, but when this occurs it raises the possibility of alternative diagnoses, such as ERA or ]AS in boys or developing polyarthritis in girls.
C HAP T E R
,-:. I <\BLE 11-1
Classification of Oligo,uthritis (ILAR Criteria)
Arthritis in four or fewer joints during the first 6 mo of disease Persistent oligoarthritis: Never more than four joints affected Extended oligoarthritis: More than four joints affected after the first 6 mo of disease Exclusions: • Psoriasis or a history of psoriasis in the patient or a first-degree relative • Arthritis in an HLA-B27 positive male beginning after the sixth birthday • Ankylosing spondylitis, enthesitis-related arthritis, sacroiliitis with inflammatory bowel clisease, Reiter's syndrome, or acute anterior uveitis or a history of one of these disorders in a firstdegree relative. • Presence of IgM RF on at least two occasions at least 3 mo apart • Presence of systemic arthritis IgM, immunoglobulin M; lLAR, International League of Associations for Rheumatology: RF, rheumatoid factor.
Sex Ratio In North America and Europe, oligoarthritis is predominantly a disease of girls, with a female/male ratio of approximately 3:1. In children with uveitis, the ratio of girls to boys is even higher: 5:1 to 6,6:1.13--16 In Asia, however, oligoarthrltis occurs predominantly in boys, and uveitis is reported to be rare. 17
GENmCS Early-onset oligoarthritis, particularly if it is complicated by uveitis, appears to be very uncommon in populations of non-European origin. Oligoarthritis is seldom familial. When sibling pairs both have ]RA, however, three quarters are concordant for onset type (most commonly pauciarthritis),18 It is likely that oligoarthritis, like other types of childhood arthritis, is a multigenic disease.
26 24
•
22 20 18
j
16
~ 14
c
8 6
2 Years • FIguN 11-1 Age at onset of oligoarticular JRA: total group (-.1-), girls (-e-), boys (-0-). (From Sullivan DB, Cassidy IT, Petty RE: Pathogenic implications of age of onset in juvenile rheumatoid arthritis. Arthritis Rheum 18: 251, 1975.)
II 0
LI GOA R T H R IT I S
275
HLA Genes There are quite characteristic associations of oligoarthritis, or subsets of oligoarthritis, with some HLA genes. The unusual disease association of an A locus antigen, AZ, has been reported and confirmed in children with ]RA (ACR criteria) in general and in early-onset oligoarticular ]RA in girls in particular. 19-21 An increase in the HLA-BZ7 allele in early studies was possibly related to inclusion of children with ]AS (the recently proposed ERA).22,23 Later investigations documented inconsistent increases in the frequency of this antigen in subgroups of children with ]RA. 24,25 However, BZ7 may confer an age-related risk for oligoarticular ]RA in boys (50% risk at 7.3 years; 80% risk at 11.9 years).19 Among the class II alleles, the most consistent association has been with the DRBl*08-DQAl·04-DQBl·04 haplotype encoding DR8 and DQ4. These two alleles are in strong linkage disequilibrium, but a recent report showed that it was DR8, not DQ4, that was associated with ]IA. 26 DR8, DRS, DR6, DPBl*OZOI, and certain DQ alleles are also reportedly more frequent in children with early-onset oligoarthritis I9 ,24,27,28 and in oligoarticular ]RA, with relative risks in the range of Z to 13. 24 ,27,29-34 An association with DR6 has been reported in a few studies. 19 Linkage disequilibrium probably accounts for some of the reported associations. The transmission disequilibrium test was used by Moroldo and colleagues24 to examine linkage and association in 101 white families who had a child with oligoarthritis. DR8 and DRS (as well as AZ, B27, and B3S) had significantly higher frequencies of transmission to the affected child; DR4 and DR7 were found less often. These data suggest that these numerous HLA associations partly reflect linkage between the HLA genetic region in children with ]RA and a population stratification effect. However, age and sex influenced these effects. Prahalad and colleagues35 used sibling pairs with arthritis to confirm the linkage of pauciarticular ]RA with the HLA-DR region, especially DR8 and DRIl. Using restriction fragment length polymorphisms, Morling and colleagues33 found that children with oligoarticular ]RA had increased frequencies of DRBl·08; DRB3·01l0Z/03 (DRwSZ); DQAl·0401 and 0501; DQBl·0301; DPAl·0Z01; and DPBl*OZ, compared with healthy controls. Both sub-types of DRS (DRIl and DRIZ) and DR8 (DRBl·080l) contributed to susceptibility to early-onset oligoarticular disease. 36 DRIl, DRIZ, and DR8 haplotypes share similar DQAl alleles: DQAl·0401, ·0501, and ·060J.37,38 These three DQ alleles have a common motif in exon Z at the 4Z to 53 positions that was present in 86% of children with ]CA but in only 36% of controls.37,38 Haas and colleagues38 demonstrated that distinct differences in the DQAl promoter are strongly associated with susceptibility to early-onset disease. Nepom and associates 39 identified a 13-nucleotide region of sequence identity in the first hypervariable region of DRS, DR6, and DR8 alleles that is a possible "shared epitope" that could be important in antigen recognition, DRI and DR4 are present in lower frequency in young girls with persistent oligoarticular ]RA and antinuclear antibody (ANA) seropositivity, compared with the normal population. 19 DRI is a risk factor for extended oligoarthritis as well as for polyarthritis in older children. 40 It is in linkage
276
C HAP T E R
II
OLIGOARTHRITIS
disequilibrium with DQA"O101, which was associated in one study with progressive erosive disease in children with early-onset oligoarticular JRA and was negatively associated with the presence of uveitis. 4l This DQA gene, although not present in all children with the disease, may be critically important in the development of this onset type. 42 DQA*0101 43 and A2*0101 44 are also binding sites for the 45-kD DEK protooncogene. Anti-DEK antibodies are characteristic of oligoarticular-onset disease (78% positive), especially in children who are ANA seropositive and have a history of uveitis, and may negate the regulatory function of the gene,45 or they may simply be a retlection of autoimmunity.46 One study associated ANA seropositivity in early-onset disease with DQB1*0603,47 Oligoarticular JRA is also associated with DP2 (DPB1*020l),48-50 which is present in 64% of patients but only 25% of controls. It has been suggested that this DP allele increases the risk conferred by DR alleles but is not sufficient in itself to increase susceptibility to oligoarticular JRA.51 A number of studies have discussed the role of interactions among alleles at different loci in producing susceptibility to disease. 33.52 Interactions between class I and class II genes led to the hypothesis that at least two genetic loci are involved in the predisposition to oligoarthritis; they have been namedjRA 1 and]RA 2. 24 ,40,41.53-56 A third locus, ]RA 3, has been postulated for a DP gene. In linkage studies, Zeggini and coworkers 21 demonstrated linkage to HLA-A, -B, and -DRBl in girls with oligoarthritis, with persistent and extended types. They suggested that linkage appeared to be attributable to preferable maternal transmission of these alleles.
Non-HLA Genes A number of genes involved in antigen presentation or cytokine expression may be important in either a predis-
I! II
TABLE I I 2
position to JRA or its pathogenesis.57 A weak association with the transporter associated protein gene TAP2B, a polymorphism in a member of the adenosine triphosphatebinding cassette superfamily, is present in patients with early-onset disease. 58 TAP1B may function as an additive susceptibility factor,59 One study suggested that homozygosity of the B allele of the LMP2 proteosome subunit may increase susceptibility to a putative subtype ofJRA that is associated with B27. 60 Interaction with other non-HLA genes is also possible; IL-1A2, a variant of the IL-1~ gene, is associated with early-onset oligoarthritis. 61 Children with extended oligoarthritis (ILAR criteria) were shown to have a high frequency of the interleukin-1 receptor antagonist gene ILlRN*2; this was also observed, to a lesser extent, in children with ERA. 62 The gene for the cytokine IL-1~, or a gene for which its polymorphism is a marker, may contribute risk for early-onset disease and uveitis. 6163 A significant increase in ILlRN*2 in Czech patients with JIA, compared with controls, was reported, especially in those with oligoarthritis, ERA, and other categories. 62 Crawley and colleagues64 found a decrease in the IL-10 phenotype associated with low IL-lO production in children with arthritis affecting fewer than five joints, compared with those with more than four affected joints. The frequency of the tumor necrosis factor TNFa2 microsatellite allele was significantly increased in Latvian children with oligoarticular JCA, and the frequency of TNFa9 was significantly decreased in this population,65 The TNFa2 allele is associated with high TNF-a production. 66 Zeggini and colleagues reported an increased frequency of the intronic +851 TNF SNP in persistent oligoarticular JIA (odds ratio, 3.86; 95% confidence interval, 1.6 to 9.2).67 Genetic associations are summarized in Table 11-2.
HlIllIdn I ellkofyte Antigen A.,.,odalions in Oligoarthrilis
HLA Gene
CrIteria
Associations
A2 H27 DR6 DRB1'08
ACR ACR ACR ACR EULAR ACR ACR ACR ACR ACR EULAR ACR ACR ACR ACR EULAR EULAR ACR ACR ACR EULAR
Young age, female sex Erosion when present with DR8 Oligoarthritis Oligoarthritis Early onset Persistent disease Oligoarthritis Progressive erosive disease Decreased in lIveitis Oligoarthritis Oligoarthritis Oligoarthritis Extended oligoarthritis Decreased in persistent oligoarthritis Decreased in oligoarthritis Early onset Early onset Oligoarthritis Oligoarthritis o ligoarthritis Early onset, ANA positive
DRB3'01/2/3 DPA'01Ol DPA1'020l DPBl'020l DR 1 DR 4 DRll (DRS) DR12 (DRS) DQA1'040l DQA1'OSOl DQB1'0301 DQB1'0603
Reference No. 19,21 232 19 24 36 232 24 41 41 41 48 50 40 40 19 36 36 24 24 24 47
ACR. American College of Rheumatology; ANA, antinuclear antibody; EULAR. European League Against RheumatLsm; HLA. human leukocyte antigen.
C HAP T E R
ETIOLOGY AND PATHOGENESIS The etiology of oligoarthritis is unknown. The narrow age-at-onset profile suggests the possibility of exposure to a ubiquitous environmental agent, possibly a virus, but none has been consistently identified. In a study of six twin pairs with oligoarticular ]RA, there was an average of approximately 3 months (range, 0 to 12 months) separating disease onset in each twin, again raising the question of an environmental agent as an initiating event.68 One study alleged that breast feeding has a protective effect on the development of ]RA,69 especially oligoarticular disease; however, a strong relationship was not confirmed in another investigation.70 Studies of T lymphocytes have yielded conflicting results. Maksymowych and associates71 reported a high frequency of the T cell receptor allele TCR-V~6.1 among HLA-DQA*0101-positive children. A subsequent report identified this TCR null allele as a risk factor for a polyarticular course in children with early-onset oligoarticular disease positive for DQAl*0101. 72 This association was not contlrmed in a Norwegian study,73 nor did Nepom and coworkers74 identify TCR polymorphism in their studies of oligoarticular-onset ]RA. In the report of Thompson and colleagues,75 TCR-V~8 was clonally expanded in children with polyarticular disease, and TCR-V~20 was increased in those with oligoarthritis. Murray and colleagues76 studied synovial T cell infiltrates in 17 children (12 with polyarticular and 5 oligoarticular disease) and found that the level of T cell activation (CD3+ IL-2W) was significantly higher in oligoarthritis (especially for CD8+ cells) and that the CD4/CD8 ratio was lower. In a subsequent report,n these investigators found that IL-4 messenger RNA (mRNA) was more frequently identified in the synovium of children with oligoarticular ]RA than in those with polyarticular onset or course. They suggested that the presence of IL-4 mRNA might be important in restricting the disease to an oligoarticular pattern. The high frequency of autoantibodies to nuclear antigens indicates a break in immunologic tolerance, but there is no evidence that autoantibodies as such participate in disease pathogenesis. The identity of the specificities of the antigens to which ANAs react in children with oligoarthritis are still largely unknown. Antibodies to an epitope on the high-mobility group HMG-17 protein are increased in ]RA, and antibodies to an HMG-2 protein are increased in oligoarticular disease. 78.79 A number of studies have identified an interesting association between elevated serum prolactin levels and]RA as well as SLE (see Chapter 18). Levels were increased in children with ]RA and were associated with ANA seropositivity.80 Modest hyperprolactinemia was also identified in prepubertal girls who were ANA seropositive and had oligoarthritis. 81 .82 The prolactin concentration correlated with levels of IL-6 and with a chronic course of the disease. Humoral and cellular immune responses to highly conserved bacterial heat shock proteins (HSPs) are present in children with ]RA. 83-
II 0
LI GOA RT H R I TI S
277
tive T cells are part of the normal immune repertoire for TCR V-gene products; self-HSPs and bacterial HSPs may trigger this response. 93-95 In 13 of 15 children with oligoarthritis, T lymphocyte proliferative responses to Hsp-60 were detected an average of 12 weeks before remission of the inflammatory disease. 89.9o The investigators hypothesized that induction of tolerance to specific T cell epitopes of Hsp-60 by nasal administration may be a promising route of immunotherapy for childhood arthritis. 96
CLINICAL MANIFESTATIONS The first 6 months of disease is characterized by inflammation in four or fewer joints. These children are not systemically ill, and, except for chronic uveitis, extraarticular manifestations are distinctly unusual. Oligoarthritis in a child is predominantly a disease of the lower extremities. In a study97 of 64 children with oligoarticular ]RA (ACR), one or both knees were most commonly affected at disease onset (89%), followed by the ankles (36%). Arthritis affecting small joint of the fingers and toes occurred in only 6% of the children, and arthritis in elbows, hips, wrists, or temporomandibular joints in 3%. Although it is the authors' impression that wrists and small joints of the hands or feet are seldom affected in oligoarticular ]RA at onset, others disagree. 98 In at least half of the reported cases, only a single joint is affected (monarticular onset), usually the knee.99-101 Uveitis may be present at onset of the disease; it eventually affects up to 20% of children and is usually asymptomatic.
DIAGNOSIS The differential diagnosis depends on a number of factors, including the onset type and pattern of joint involvement, the duration of disease at the time the child is evaluated, and the sex and age of the child. In some instances, oligoarticular ]IA is a diagnosis of exclusion.102.103 In a child with monarthritis of recent onset (Le., within 72 hours), the differential diagnosis must include septic arthritis, trauma (including nonaccidental trauma resulting in a hemarthrosis), and hematologic disease (including hemophilia, leukemia, and malignancy) (Table 11-3).104 If the monarthritis is long-standing, sepsis (except for tuberculosis), trauma, and malignancy are very unlikely. A painful joint effusion of short duration may be caused by trauma, or, rarely in children, it may be associated with an internal structural abnormality such as a discoid meniscus 105 or osteochondritis dissecans (see Chapter 36). The monarthritis of hemophilia results from bleeding into a major joint, which is often initiated by even minor trauma. Recurrent involvement has been described in occult celiac disease. 106.107 Rare causes of chronic monarthritis, such as tuberculosis, sarcoidosis, and villonodular synovitis, should also be considered. A migrating monarthritis, sometimes associated with fever and a rash, has been described in children of Assyrian ancestry.108 The various forms of idiopathic osteolysis may mimic arthritis of a limited number of joints (e.g., wrists) at onset (see Chapter 37). Rarely, arthritis
278
C HAP T E R
I! II TABLE
tI 3
II
OLIGOARTHRITIS
Ditll'rential Oiaynosis of Monarthritis
Acute Monarthritis Early rheumatic disease Oligoarthritis Enthesitis-related arthritis Psoriatic arthritis Arthritis related to infection Septic arthritis Reactive arthritis Malignancy Leukemia Neuroblastoma Hemophilia Trauma
Chronic Monarthritis Juvenile idiopathic arthritis Oligoarthritis Enthesitis-related arthritis Juvenile psoriatic arthritis Villonodular synovitis Sarcoidosis, Blau's syndrome Tuberculosis Hemophilia "Pseudoarthritis" (e.g., hemangioma, synovial chondromatosis, lipoma arborescens) Episodic fever syndromes (mevalonate kinase deficiency, familial Mediterranean fever, chronic infantile neurocutaneous and articular syndrome)
has resulted from administration of a drug such as isotretinoin l09 ,1l0 or antithyroid medication,lll,m Oligoarticular JIA is the most common cause of chronic oligoarthritis, especially in girls younger than 6 years of age, Psoriatic arthritis, which affects large and small joints in an asymmetrical pattern, is also a possibility at this age (see Chapter 14). JAS is a more likely cause of a monarthritis or oligoarthritis of the lower extremities with onset in the older child or adolescent. 113 A diagnosis of oligoarticular JRA or psoriatic arthritis may be substantiated by the demonstration of asymptomatic anterior uveitis by slit-lamp examination, although sarcoidosis also rarely manifests in this way. ANA seropositivity supports the diagnosis of JRA or psoriatic arthritis but may also be found in some healthy children 1l4 and in some children with noninflammatory musculoskeletal pain. ll5 Lyme disease may present as an oligoarthritis (see Chapter 29). In the child with oligoarticular JIA, the affected joint is swollen and often warm but usually not very painful or tender, and almost never red. The child is not systemically ilL If a joint is acutely painful and erythematous or if the child is febrile, septic arthritis is more likely the correct diagnosis. 116,ll7 Immediate joint aspiration is always indicated in such a patient to exclude septic arthritis or osteomyelitis, Needle or arthroscopic synovial biopsy is useful in children with monarthritis in whom granulomatous disease is suspected. 99,1l8,1l9 Culture and microscopic examination of synovial tissue may be more rewarding in the case of tuberculosis than culture of the fluid only. A negative purified protein derivative (PPD) skin test virtually excludes the diagnosis of active tuberculosis.
Biopsy should not be performed simply to confirm a diagnosis of JRA. Arthritis of the hip joint is rare at the onset or during the course of oligoarticular JRA. In the authors' experience of 145 children with oligoarthritis seen early in their course, only 1 girl had initial involvement of the hip. Onset of apparent arthritis in the hip in a very young child should be considered first to be a septic process or a congenital dislocation. 120 In the older child, osteonecrosis of the femoral head (Legg-Calve-Perthes disease) is a diagnostic consideration. In the adolescent age group, a slipped capital femoral epiphysis may initially mimic JRA. In older boys, JAS may manifest as unilateral or bilateral hip disease (see Chapter 13), In children with transient synovitis of the hip,121,122 pain may be severe, but the process is self-limited, lasting no more than one to a few weeks; the results of all laboratory and radiologic studies are normal (see Chapter 13).
COMPLICATIONS: UVEITIS One of the most devastating complications of JRA is chronic nongranulomatous uveitis. It is predominantly a chronic, anterior, nongranulomatous inflammation affecting the iris and ciliary body-an iridocyclitis (Fig. 11-2). The posterior uveal tract, the choroid, is rarely affected clinically (Fig. 11-3). Papillitis has been reported in three patients, 14,123.124 and macular edema and degeneration have been described in one or two others. 125 Less common ocular complications include scleritis, episcleritis, and keratoconjunctivitis sicca. In Kanski's study,126 only 3 of 131 children with chronic uveitis developed keratoconjunctivitis sicca. Ohm 127 first described chronic uveitis and band keratopathy in a child with arthritis in 1910. The association of ocular disease and JRA was confirmed by several authors. Il8-1l0 In Sury's patients, m chronic uveitis was found in 15% of the total, and two thirds of these patients had band keratopathy. The major-ity of his patients had an insidious onset of uveitis with little or no early disturbance of vision; diagnosis was often delayed until slit-lamp examination.
Conju netiva \
\
Anterior chamber, Lens_
--
"
Pupil--Optic nerve Cornea ....--
Ciliary m. " .... "
/
I
~~~ / /
/
/
Choroid
• figure ll-Z Schematic view of a sagittal section of the eye. Chronic anterior uveitis (iridocyclitis) involves the iris and dliary muscle primarily, but secondary effects occur in the cornea, anterior chamber. lens, vitreous, and (rarely) retina.
C HAP T E R
II 0
LI GOA RT H R I TIS
279
Etiology and Pathogenesis The pathogenesis of chronic uveitis and the basis of its association with JRA are not known. Children with uveitis and JRA have a higher frequency of immunity to soluble retinal antigen (S antigen)148-150 and to a low-molecularweight iris antigen l51 ,152 than do children with arthritis alone. It is not clear whether immunity to these ocular antigens is pathogenic or merely reflects the inflammatory process.
Genetic Background
• figure 11-3 Retinal edema and hyperemia in the left eye, representing posterior uveitis in a 6-year-old girl with a I-year history of juvenile rheumatoid arthritis. She had minimal blurring of vision. Anterior uveitis and synechiae were also present.
Epidemiology Chronic uveitis is especially likely in young girls who have oligoarthritis with an early age at onset who are ANA seropositive (Table 11-4).14,34,123,132-141 On average, uveitis is found in 15% to 20% of children with oligoarticular JRA99,IOO,142 and in about 5% of those with polyarticular-onset disease. Uveitis is very rare in children with systemic-onset disease. 125 . 132 ,133,143,144 The frequency of uveitis has varied considerably in reported series of patients with JRA or JCA, from 4% in Minnesota145 to 16% in Finland. 1I It appears to be particularly rare in Asian and African populations. JRA with uveitis has been reported in whites, African Americans, Hispanic Americans,146 black and Indian South African children,80 and Polynesians from New Zealand. 147 The authors have observed this complication in North American aboriginal children.
'--
'1> ..
I ABLE ]] -4
Characteristics of Children with Chronic Uveitis
Charac:terlstlc
Female/Male ratio Mean age at onset of arthritis (yr) Arthritis category (%) Oligoarthritis Polyarthritis Systemic Serology (%) Rheumatoid factor positive Antinuclear antibody positive
Overall Avenge Reported Range 4.4:1 4
82 18 <1 <1
80
3:1 to 7.5:1 2-5 63-95 7-37 0-6 0-5 60-95
From Petty RE: Current knowledge of the etiology and pathogenesis of chronic uveitis accompanying juvenile rheumatoid arthritis, Rheum Dis Clin North Am 13: 19-36. 1987.
The HLA alleles most frequently associated with earlyonset oligoarthritis, ANA seropositivity, and uveitis are A2, DRS (DRB1*1104), DR1 (DRB1*OIQl), DP2 (DPB1*020l), DQ1 (DQA1*050l), DR6, and DRw52. 34,153,154 Malagon and coworkers 34,154 compared 72 children with early-onset oligoarticular arthritis and uveitis with 77 children with early-onset oligoarticular arthritis but no uveitis. They found that DRS correlated with the presence of uveitis and DR1 correlated with its absence. The presence of DR8 had no association with uveitis but was increased in all children with oligoarthritis compared with controls. In the study by Melin-Aldana and colleagues,155 DRS (DRBl*1104) was significantly associated with an increased risk of uveitis along with DQA1*0501 and DQBl*0301, with which it was in linkage disequilibrium. A2 was Significantly associated with anti-DEK antibodies, which occurred in 97% of children with uveitisY HLA-DR11 was present in 83.7% of Italian children with uveitis, most of whom had an oligoarticular onset. 156
Clinical Manifestations The onset of chronic uveitis is usually insidious and often entirely asymptomatic, although up to one half of the children have some symptoms attributable to the uveitis (pain, redness, headache, photophobia, change in vision) later in the course of their disease (Table 11-5). Uveitis is detected in fewer than 10% of patients before the onset of arthritis,126 usually in the course of a routine ophthalmic examination. In almost half of all patients with uveitis, it occurs just before arthritis is diagnosed, at
[
.. TABLE] ]-5 Oculal Signs and Symptoms in Children with Chronic Uveitis and Arthritis
Percentage Affected Charaderlstlc Bilateral uveitis Symptoms Ocular pain and or redness Change in vision Photophobia Headache None
Mean
Range
64
25-89
65
0-25 0-20 0-8 0-6 51-97
From Petty RE: Current knowledge of the etiology and pathogenesis of chronic uveitis accompanying juvenile rheumatoid arthritis, Rheum Dis Clin North Am 13: 19-36. 1987.
280
C HAP T E R
II
OLIGOARTHRITIS
the time of diagnosis, or shortly thereafter. 14 ,126,140 Most children develop uveitis within 5 to 7 years after onset of arthritis, although the risk is never entirely absent I4 ,16,134,157 (Fig. 11-4). The disease is bilateral in approximately 70%
12 11 10
~Ql
~
0.
0
~
E ;:]
Z
9 8
7 6 5 4 3 2 1 0 1
2
3
4
5
6
7
A
8
9 10 11 12 13 14 15 >15
Age(y)
12 11 10
.l!l c: Ql
.~
0.
0 Qi
.c E ;:] Z
9 8
7 6 5 4 3 2
I I
1 0 1
2
3
4
5
6
7
B
8
I
I
9 10 11 12 1314 15 >15
Age (y)
12 11 10
9
~Ql
8
0.
7 6
~
0
...
il
5
E ;:]
4
Z
3
Pathology
2 1 0 1
C
to 80% of children. 14 ,139,158 Patients with unilateral disease are unlikely to develop bilateral involvement after the first year of disease; however, there are exceptions, and unilateral uveitis may persist for many years in a few children before the other eye is involved. The early detection of chronic uveitis requires slitlamp biomicroscopy, which should be performed at the time of diagnosis in every child with ]IA and repeated at prescribed intervals during the first few years of the disease. The frequency of ophthalmic examinations is influenced by the level of risk of uveitis (Table 11-6). The authors recommend that slit-lamp examinations be performed every 3 months for the first 2 years in children in the high-risk group (early age at onset, oligoarthritis, female sex, ANA seropositivity) and every 4 to 6 months thereafter for a period of 7 years at a minimum. In children with polyarticular disease, slit-lamp examinations should be done initially at 4- to 6-month intervals. In children with a systemic onset, examinations once a year after the first year are probably sufficient. Any child who has had uveitis should be considered to be at high risk, even if it has remitted, and continued surveillance is mandatory. The earliest signs of uveitis on slit-lamp examination are the presence of inflammatory cells and increased protein concentration ("flare") in the aqueous humor of the anterior chamber of the eye (Fig. 11-5). Deposition of inflammatory cells on the inner surface of the cornea (keratopunctate deposits or keratic precipitates) may develop later. Posterior synechiae between the iris and the anterior surface of the lens result in an irregular or poorly reactive pupil (Table 11-7; Fig. 11-6). This abnormality may be the first obvious clue to the presence of uveitis on ophthalmoscopic examination, but it is often a sign of disease of considerable duration or severity. Band keratopathy occurs late (Fig. 11-7). Secondary cataracts and glaucoma are also quite common, but phthisis bulbi is a rare late manifestation of uveitis. Band keratopathy and cataracts occurred in 42% to 58% of patients in reported series, and glaucoma occurred in 19% to 22%.144.159,160 These complications are still occasionally encountered in some children with chronic uveitis in spite of vigorous and carefully monitored ophthalmic treatment, although their frequency may be diminishing
2
3
4
5
6
7
8
9 10 11 12 13 14 15 >15
Time of onset (y)
• Figure 11-4 Graphs showing the temporal relationships between arthritis and uveitis in children with juvenile rheumatoid arthritis. A, The distribution of age at onset of arthritis in aseries of 38 children who developed uveitis, B, The distribution of age at onset of uveitis in the same children. Note that in four patients, uveitis began after their 15th birthday: at 15{1I2}, 18,31, and 39 years of age. C, Interval between onset of arthritis and diagnosis of uveitis in these patients. Note that for 1 patient the interval was 29 years, and for another, 34 years.
Reports of the histopathology of uveitis are few. Descriptions of extremely severe, long-standing disease that led to blindness do not necessarily illuminate the pathogenic process. 161 Reported changes include increased iris vascularity162 with scanty lymphocyte and plasma-cell infiltrates. 162.163 Plasma cells containing immunoglobulin M (IgM) were described in one patient. 164 Patients in whom granulomatous changes were present may have had sarcoidosis rather than ]RA.165.166 Immunoglobulin levels were increased in the aqueous humor of children with ]RA and uveitis.167.168 Studies of the vitreous showed an increased IgG concentration,
C HAP T E R
,--
II ..
TABLE I 1-6
RIsk*
Examination
Type of
Antinuclear
Frequency (mo)
Disease
AntIbodies
6
Moderate
3-4
High
OLIGOARTHR1TIS
281
Frequenty of Uveitis Monitoring
12
Low
II
Systemic arthritis Polyarthritis
Age at Onset (yr)
+ or-
Disease DuratIon (yr)
>7
Polyarthritis Oligoarthritis Oligoarthritis
+ or-
~6
~6 ~6
9
+
Oligoarthritis
+
~6
9
>7
'Risk decreases by one level for age at onset >6 yr.
• FI. . . 11-5 A slit-lamp examination shows "flare" in the fluid of the anterior chamber (caused by increased protein content) and keratic predpitates on the posterior surface of the cornea, representing small collections of inflammatory cells. (Courtesy of Dr. H. J. Kaplan.) (See color insert.)
.~ III
I ABLE 11-7 Frequency of Complications of Chronic Uveitis ... Reported Cases
Complication
Mean Frequency (%)
Synechiae Band keratopathy Cataract Glaucoma Phthisis bulbi
62 37 40 19
9
Range (%)
37-75 11-56 6-75 8-25 0-14
From Petty RE: Current knowledge of the etiology and pathogenesis of chronic uveitis accompanying juvenile rheumatoid arthritis. Rheum Dis Clin North Am 13: 19-36, 1987.
activated complement C3c, and increased C1q binding, suggesting the presence of immune complexes. t69 Kaplan and associates 170 found that 90% of vitreous lymphocytes in one adult with uveitis and "juvenile" RA were B lymphocytes,
Differential Diagnosis Uveitis and arthritis occur together in a number of diseases with high frequency.144,171 Uveitis also occurs in
• Figure 11-6 Left eye of a 7-year-old boy shows an irregular pupil that resulted from adhesions of the iris to the anterior surface of the lens. (See color insert.)
children without evidence of joint involvement as an isolated disorder. 172 Inflammation of the anterior uveal tract is found in Kawasaki disease,173 complicates ERA QAS),174 and is observed in psoriatic arthritis,138,17'; arthropathy of inflammatory bowel disease,176-178 and reactive arthritis with urethritis and conjunctivitis,179,1HO Uveitis also occurs in chronic infantile neurologic cutaneous and articular syndrome (CINCA), 181-184 sarcoidosis, 165,166,185,186 Blau's syndrome,187,188 Vogt-Koyanagi-Harada syndrome, and Beh~et's disease. 144 Posterior uveitis rarely complicates rheumatic diseases in children, and its presence suggests the diagnosis of sarcoidosis rather than JIA.
Laboratory Examination The most characteristic laboratory abnormality found in children with arthritis and uveitis is the presence of ANAs, usually in low titer (less than 1:640). The specificities of these antibodies are unknown,126,189-191 and the pattern of immunofluorescence on HEp-2 cells is speckled or homogeneous. Occasionally ANAs react with nucleoli, but antibodies to double-stranded DNA (dsDNA) and the extractable nuclear antigens (ENAs) are absent. Antibodies that are specific for granulocyte nuclei
282
C HAP T E R
II
OLIGOARTHRITIS
• Rgure 11-7 A, Early band keratopathy is noted as a semilunar band just inside the limbus medially and laterally. It does not extend across the pupil. 8,The semiopaque band extends across the midplane of the comea in this example of more advanced band keratopathy. It is fenestrated and does not extend to the limbus. (8, See color insert.)
have been reported to occur in children with uveitis. 192 These antibodies were complement fixing and correlated with disease activity. 193 When tissue sections were used as substrate, ANAs were found with significantly higher frequency (65% to 88%) in children with oligoarticular JRA and uveitis than in those with oligoarticular JRA alone. 189,190 Neuteboom and coworkers l94 found ANAs in 55% of children with uveitis and chronic arthritis with HEp-2 cells substrate, but in only 32% when rat liver was used. They detected antibody to dsDNA in 3 of 22 children and antiSS-A in 1 of 22 children with uveitis. Using HeLa cells as a source of nuclear material, they also demonstrated reactivity of sera from children with arthritis and uveitis to a 15-kD antigen by Western blotting. Reactivity to histones has been associated with oligoarticular disease and uveitis. 195 ,196 Massa and colleagues l97 reported that ANA-seropositive patients had significantly elevated IgG antibodies to nucleosomes H3 and H4, to DNA-free subparticles, and to H3H4-DNA, hut that these antibodies were generally not associated with uveitis.
Management The treatment of chronic uveitis should be supervised by an ophthalmologist who is experienced in management of this complication. 198 The initial approach consists of glucocorticoid eye drops (dexamethasone or methylprednisolone), with or without a mydriatic agent to dilate the pupil and help prevent posterior synechiae. A shortacting mydriatic drug is preferred, given if possible once a day in the evening, so that pupillary dilatation does not interfere with school work and reading. Some data suggest that nonsteroidal anti-inflammatory drugs (NSAIDs) may be of some benefit. 199-201 Although this effect is not a major one, it should be considered when NSAID treatment of arthritis is altered. In unresponsive disease, glucocorticoid drops may be given hourly during waking hours, with glucocorticoid ointment placed in the conjunctival sac at bedtime. Chylack lS reported that 36% of children had no response to topical glucocorticoids after 6 months of intensive therapy and therefore required systemic administration of the drug. In a few instances, subtenon injections of glucocorticoid may be required. It may be advisable in severe or resistant disease to employ supplemental oral prednisone in low dosage (e.g., 2 to 4 mg/day). Some
children require larger amounts of oral glucocorticoid; single daily or alternate-day therapy may be useful in these patients. 126 Occasionally, high-dose intravenous methylprednisolone (30 mg/kg) is of benefit. Although the results of slit-lamp examination may return to normal soon after treatment is initiated, it is not advisable to discontinue steroids at that time, because signs of inflammation frequently reappear. Long-term ophthalmic glucocorticoid administration may lead to the development of Cushing's syndrome. Methotrexate may be an effective mode of therapy in children with severe uveitis in whom all other forms of treatment have failed. 202 ,203 Chlorambucil,204,20s cyclosporine,206 and plasmapheresis 207 may be useful in some cases of idiopathic uveitis, but their role in the uveitis of JIA is not clarified. Cyclosporine was given to 14 children with chronic uveitis refractory to glucocorticoids for a mean duration of 21 months. 20B Visual acuity was improved or did not deteriorate further in 92% of eyes, and results of the ophthalmic examination were improved in 76%. In a follow-up study, the same investigators studied the effect of mycophenolate mofetil in nine eyes that had not responded to cyclosporine. 209 Eight eyes improved in visual acuity; by ophthalmoscopy score, 5 were unchanged and 2 eyes worsened. Refractory uveitis has also been treated with intravenous immunoglobulin (IVIG).209 Smiley 131 noted that neither adrenocorticotrophic hormone nor azathioprine had proved efficacious in controlling ocular inflammation, although some ophthalmologists believe that azathioprine may be useful. The efficacy of anti-TNF biologic agents in the treatment of uveitis is uncertain. Reifplo reported effectiveness of etanercept in an uncontrolled study of seven children with active uveitis and JRA that had not responded satisfactorily to corticosteroids and methotrexate or cyclosporine. Follow-up of these patients indicated that at least four had a sustained response. 21l Smith and coworkers 212 conducted a randomized, blinded, placebo-controlled trial in 12 children with JRA and uveitis in whom the methotrexate response was insufficient and found no difference between etanercept and placebo at 6 months. Infliximab has been anecdotally reported to be of benefit, but there are also reports of new onset or worsening of uveitis during treatment with anti-TNF agents. 2B
C HAP T E R
Band j<eratopathy has been treated with topical chelation and by lasers. Cataracts seldom interfere significantly with vision in childhood, but they may require surgical removal. The managem~nt of complicated uveitis and glaucoma remains unsatisfactory, but results of lensectomy or vitrectomy for complicated catara~t are improved. 158 The subject of cataract surgery in children with JRA and uveitis was thoroughly reviewed by Hooper and cbworkers. 214 It was their recommendation that cataract surget'y should be performed only in the absence of vitreous opacit~es, hypotony, or cyditic membrane formation; that the anteri?r chamber should be free of inflammatory cells; and that combining anterior segment surgery with a pars plana vitrectomy IS the safest approach. Perioperative glucocorticoids are recor~mended. The timing of cataract extraction challenges the judgment of the ophthalmologist in weighing the danger of opera~ion on an inflamed eye against the risk of amblyopia. Oper~tive complications are minimized with microsurgery and cryoe*traction.
Cou~ and Prognosis of Uveitis Srniley 134 emphasized that the course of chronic uveitis is rarel~ less than 2 years and often as long as 17 or 18 years: The authors' experience and that of others,132 however, indicates that some children have much shorter courses. The activity of the uveitis does not parallel that of the arthritis,14,133,1 34,160,215 and it may occur for the first time after the arthritis is in remission. Visual loss may occult because of complications of the uveitis or as a result of amblyopia related to suppression of visual images from a cataract. Although prognosis for sight in chronic uveitis has been improving, visual outcomes still remain far less than satisfactory, with estimates of blindness (less than 20/400 AU) in both eyes as high as 15% to 30%.14,134,139 It has been suggested that the frequency and severity of uveitis may actually be decreasing. 13~,140.J41,216,217 Whether this represents intrinsically less severe disease (a "benign" form of uveitis), earlier treatment, or more aggressive therapy is not certain. The prognosis for uveitis is worse in children in whom the onset of uveitis occurs before diagnosis of arthritis or shortly thereafter. 158,137,140 It is also worse in those with an initial severe inflammatory response,137 chronicity of inflarnmation,34 or ANA seronegativity.l40 Kanski's review l58 of the Taplow experience confirmed that visual prognosis was good in 25% and fair in 50% of studied patients. The remaining 25% developed visual impairment from cataract or ghiucoma, In the early Ann Arbor series, 3 children (8%) completely recovered from their uveitis, and 55% retained normal vision, 14 However, 16% of the children were blind in one or both eyes. During the course of the disease, even minimal symptoms were associated with moderate-to-advanced ocular disease, Twentyfive children had protracted or recurrent ocular inflammation. A series of patients from the same center 10 years later137 described an improved prognosis. Visual outcome was worse if uveiti$ was present at the time of diagnosis of arthritis and in children with persistently active disease. In the Taplow series, however, Kanski noted that 8 of 26 eyes with continuously active uveitis for more than 10 years remained unaffected by seconpary complications and retained normal visual acuity. 126 Cabral and colleagues,138 in a study of 49 patients (82 affected eyes), reported that only 15 affected eyes had corrected visual acuity of 20/40 or worse, at an average of 9.4 years after onset
II OLIGOARTHRITIS
283
of uveitis, and 8 had corrected visual acuity of 20/200 or worse. This study confirmed the correlations between a poor visual prognosis and the presence of uveitis at diagnosis of arthritis, the presence of complications (e.g" synechiae), and persistent disease activity. Complications of uveitis were frequent in the recent series reported by Paroli and associates. 156 Of 42 children with ]RA-associated uveitis who were observed for at least 1 year, cataract was present in 64%, band keratopathy occurred in 59%, and glaucoma occurred in 25%. Nonetheless, visual acuity was normal or good in almost two thirds of these children.
LABORATORY EVALUATION Laboratory indicators of inflammation may be normal in children with oligoarthritis, although mild to moderate elevation of the erythrocyte sedimentation rate (ESR) and elevation of C-reactive protein levels may occur. Hemoglobin levels and white blood cell and platelet counts are usually normal, and the presence of marked abnormalities in these parameters should suggest a diagnosis other than oligoarthritis. Tests for RF are almost always negative, although, occasionally, children with a single affected joint (often the wrist) have RF. In contrast, tests for ANAs are positive in low titer (1:640) in 65% to 85% of children with oligoarthritis, particularly in girls and in those with uveitis. 36 ,189.190,218 Antibodies that react with citrullinated peptides have rarely been demonstrated in children with oligoarthritis, their frequency depending to some extent on the antigen used. 21 9-221 Anticardiolipin antibodies were reported in 18% of children with oligoarthritis (compared with 4% in a healthy population).222 They do not appear to be associated with intravascular thrombosis, however. Elevated concentrations of activated C3 (C3c, C3d) were demonstrated in about one third of children with active oligoarthritis (a lower frequency than in children with systemic arthritis or polyarthritis).223 Circulating immune complexes are not characteristic of oligoarthritis. Routine synovial fluid analysis does not distinguish one type of JIA from another. The fluid is usually moderately inflammatory, with a cell count of 5 to 20,000 cells/mm3, mostly polymorphs.
RADIOGRAPHIC EVALUATION The radiographic changes in oligoarthritis are similar to those seen on other kinds of arthritis, although often less severe. In a recent follow-up study224 of 97 children with pauciarticular ]RA, joint space narrowing was present in only 5% of children early in the disease course, increasing to approximately 15% at a median of 6.2 years after disease onset. Erosions were seen in 10% of children with early disease, and in approximately 25% of children 6 years later. Bone overgrowth was more common; it occurred in more than 20% of children early in the disease and slightly more frequently later in the disease course. Not surprisingly, overgrowth was most common at the knee (Figs. 11-8 and 11-9).
284
C HAP T E R
II
OLIGOARTHRITIS
• Figure 11-8 A-e, Anteroposterior and lateral radiographic films of the knees of a 3-year-old girl who developed monarthritis of the left knee at the age of 2 years with an initial flexion contracture of 32 degrees.There is marked joint space narrowing with regional osteoporosis of the left knee and epiphyseal enlargement. D, Post-gadolinium magnetic resonance imaging sagittal studies of the left knee.There is a large joint effusion with marked inflammatory synovial hypertrophy, demonstrated by enhancement of the pannus throughout all compartments of the joint.There is also thinning and irregularity of the articular cartilage involving the femur, tibia, and patella.There is almost bone-on-bone apposition of the femorotibial articulation. Asymmetrical enlargements of the epiphyses of the left knee are visible, with relative hypoplasia of the menisci.
Magnetic resonance imaging with gadolinium confirms the presence of synovitis, and increased intra-articular fluid, and, occasionally, bone marrow edema. Its main utility, however, is in differentiating other causes of joint swelling, particularly in the child with monoarthritis. 225
MANAGEMENT Prompt and accurate diagnosis is essential to the optimal outcome of oligoarthritis. Because of the subtlety of the signs and symptoms, medical attention may not be
sought early in the disease course. The aim of therapy should be to achieve total remission of signs and symptoms of joint inflammation. Initial management of oligoarthritis should include careful clinical general and musculoskeletal assessment. In the child with monarthritis, other possible causes of inflammation in a single joint should be excluded. Evaluation by a physical therapist and occupational therapist to assess joint range, muscle strength, and function should be obtained. A slit-lamp examination by an experienced ophthalmologist is essential to exclude the pOSSibility of uveitis, as soon as possible after the diagnosis of
C HAP T E R
II
OUGOARTHRITIS
285
• FIgure 11-9 These radiographs illustrate the 5-year progression of osteoporosis, joint space narrowing, and degenerative changes in the !
oligoarthritis. A recommended program for monitoring of uveitis is shown in Table 11-6. Initial pharmacotherapy usually consists of an NSAID. Napr<j)xen in a dose of 15 to 20 mg/kg/day is often the drug of choice because of its twice-daily dosing and the availability of a suspension. The parents and patient should be cautioned to take the medication with food to minimize the risk of gastric upset and reminded of the risk of naproxen-induced pseudoporphyria. A trial of at least 6 weeks is recommended. During this time, the physiFal therapist may recommend passive and active stretching for joints with limited motion. If there has been insufficient response after 6 to 8 weeks, consideration should be given to administration of intra-articular corticosteroids, preferably triamcinolone hexacetonide (20 to 40 mg per large joint), or as a second choice, triamcinolone acetonide. It may be necessary to serially cast joints with restricted range of motion. This can be done at the same time as joint injection using conscious sedation with a drug such as midazolam (see Chapter 5). The response to NSAIDs and intra-articular corticosterotds is usually very good, and most patients with oligoarthritis respond to this approach. Joint injections can be repeated two or more times, but children who are resistant to such therapy require the addition of a second-line agent. A different NSAID may also be of benefit. The child who has an extended oligoarticular course is known to have a guarded outcome, and such children should be given the benefit of methotrexate earlier rather than later in the disease course. After a course of at least 6 months of NSAIDs and intra-articular corticosteroids on one or two occasions, oral methotrexate (0.30 to 0.5 mg/~g/week) should be started, with the dose increased as tolerated to 0.65 mg/kg/week (see Chapter 5).
Attention to the possible development of a leg-length inequality (in the child with a single affected knee) or contractures of affected joints requires ongoing assessment and physical therapy. Major leg-length inequalities are unusual in the child who is treated early and effectively. Sherry and colleagues226 demonstrated the effectiveness of intra-articular corticosteroids in preventing this complication. Should a leg-length inequality of greater than 1 cm develop, a lift to partially compensate for the difference should be applied to the sole of the shoe of the shorter leg. Flexion contractures around the knee or ankle are usually much more responsive to physical therapy after corticosteroid injection of the affected joint. Serial casting may be initiated at the time of joint injection and is an effective way of regaining lost range of motion in the knee in particular. An active physical therapy program should be undertaken under the guidance of the therapist. Surgical management of oligoarthritis is rarely necessary. Occasionally, soft tissue releases are required in the child who has not received early treatment and has developed a flexion contracture that is unresponsive to intra-articular corticosteroids and physical therapy.
DISEASE COURSE AND PROGNOSIS The disease course and prognosis in children with oligoarthritis have improved considerably in the last 10 to 15 years. In particular, the frequency of significant joint contractures or leg-length inequalities has diminished, probably as the result of prompt institution of pharmacologic and physical therapy, in particular the use of intra-articular corticosteroids. 226 Bowyer and colleagues227
286
C HAP T E R
II
OLIGOARTHRITIS
reported on the health status of 232 children with pauciarticular JRA who were monitored for 1 to 5 years. By 1 year after disease onset, half no longer required medications, and 98% were in Steinbrocker functional class I or II. No patient with pauciarticular JRA (ACR) was in Steinbrocker class III or IV 5 years after diagnosis. Childhood Health Assessment Questionnaire (CHAQ) scores were zero in the majority of patients, but approximately 25% had scores of between 0 and 0.5, and 12% had higher scores. Measures of psychosocial outcome were also evaluated in this study. Educational achievement and employment were comparable to the national norms. Bowyer and colleagues also found little negative impact of pauciarthritis on school participation: only 6% of the children were unable to participate in a full school program 5 years after diagnosis. 227 Sequelae from uveitis remain an important issue, although here, too, some centers report decreased frequency and severity of this complication. Oen and associates 228 studied the outcome of children with pauciarticular JRA (ACR criteria) who had been monitored for at least 5 years from disease onset and who were at least 8 years of age. Using stringent criteria for remission (absence of active arthritis, off medications for at least 2 years), these authors found that 47% of children with pauciarticular JRA were in remission 10 years after disease onset. Ninety-four percent of the remissions occurred before the age of 16 years. Although some patients had a monocyclic course, 25% relapsed approximately 5 years after the end of the first episode. An extended pauciarticular course was observed in 20% of children with pauciarticular onset, at a median of 3.9 years after disease onset. Children with pauciarticular JRA were mostly (85%) in Steinbrocker functional class I; 14.5% were in class II, and 0.5% in class III or IV. Radiographic outcome was evaluated in the same population group.224 Although joint erosion is unusual in pauciarticular JRA, it was demonstrable by plain radiographs in 10% of patients early in the disease course, and in 25% of patients at some time during the disease course, most commonly in knees, elbows, and shoulders. Joint space narrowing was even less frequent, but growth abnormalities occurred in approximately one quarter of the patients, most commonly in the knees and elbows. These results were confirmed by Bowyer and coworkers. 227 AI-Matar and colleagues 229 studied the patterns of joint involvement that would predict progression of disease in 205 children with oligoarthritis (ACR) who were monitored for 5 to 26 years. They concluded that the early presence of ankle or wrist disease, symmetrical joint involvement, and an elevated ESR predicted the progression of disease to affect more than four joints. ANA test positivity (titer of 1:40 or greater on HEp-2 cell substrate or 1:20 or greater on mouse liver substrate) and younger age at onset of disease correlated with longer active disease duration in the study by Oen and colleagues. 23o ANA positivity also correlated with worse outcome in this group of patients.
Fantini and colleagues 231 reported the follow-up of 420 children with pauciarticular-onset JCA (EULAR). Mean follow-up for these patients was 8.1 years, with a range from less than 1 year to more than 30 years. At last follow-up, 36% of these patients were in remission, which was defined as absence of clinical or laboratory evidence of active arthritis for a period of at least 6 months in the absence of antirheumatic therapy. Fiftythree percent of children with oligoarticular JCA had never gone into remission, and 13% had remitted but relapsed. Twenty-four percent of these patients had polyarticular course (equivalent to extended oligoarthritis of the ILAR criteria). Flato and associates 232 compared physical and psychosocial status in patients witl1 pauciarticular JRA (ACR), who were monitored for a median of 14.9 years, with those of the healthy population. They found that early age at onset and an elevated ESR on first admission to hospital predicted persistent disease. In addition to these factors, DRB1·01 was found to predict joint erosions in these patients. Elevated ESR at onset, early age at onset, and the presence of hip disease in the first 6 months were predictors of disability. Early age at onset has not always been found to be a predictor of bad outcome, however. 233 ,234 Patients with persistent pauciarticular disease fared better than those with extended pauciarticular disease with respect to development of erosions, Health Assessment Questionnaire (HAQ) scores, and remission rates ,233.234 The course of oligoarthritis is variable. Some children pursue an oligoarticular course and never have more than four affected joints (persistent oligoarthritis). In such children, the diseases often goes into remission, although flares of disease may occur many years later. In a second group, there is a progressive increase in the number of affected joints after the first 6 months of disease (extended oligoarthritis), so that by 1 or 2 years after onset, they have polyarthritis, although the number of affected joints is often much lower than in children with polyarthritis at onset. In this group, the prognosis is somewhat guarded, and fewer children enter remission. Because of the limited extent of the joint involvement, serious functional disability is uncommon. Fixed flexion contractures may persist, however, or osteoarthritis of a weight-bearing joint may eventually develop late in the course of the disease after clinical remission (see Fig. 11-9). Mortality is extremely rare.
PERSPECTIVE Oligoarthritis is deceptively complex. Current understanding recognizes two subcategories (persistent and extended), and further studies may reveal other groups within this category. Its early recognition is essential to optimal management, which usually leads to a good functional outcome. Nonetheless, the disease is chronic and is complicated by chronic anterior uveitis, which may further compromise function.
C HAP T E R
REFERENCES 1. Brewer Ej, Bass j, Baum j, et al: Current proposed revision of jRA criteria. Artbritis Rheum 20 (Supp!); 195-199, 1977. 2. European League Against Rheumatism: EULAR Bulletin No.4: Nomenclature and Classification of Arthritis in Children. Basel, National Zeitung AG, 1977. 3. Pelly RE, Southwood TR, Manners P, et al: International League of As1lociations for Rheumatology classification of juvenile idiopathic arthritis: Second Revision, Edmonton 2001. j Rheumatol 31: 390-392, 2004. 4. Oe" KG, Cheang M: Epidemiology of chronic arthritis in childhood. Semin ArthritiS Rheum 26: 575-591, 1996. 5. Andersson-Gare B: juvenile arthritis-who gets it, where and when? A review of current data on incidence and prevalence. Clin Exp Rheumatol 17: 367-374, 1999. 6. Manners ~r, Bower C: Worldwide prevalence of juvenile arthritis-why does it vary so much? j Rheumatol 29: 1520-30, 2002. 7. Oen K: Comparative epidemiology of the rheumatic diseases in children. CUrt Opin Rheumatol 12: 410-414, 2000. 8. Andersson-Gare B, Fasth A, Andersson j, et al: Incidence and prevalence of juvenile chronic arthritis: a population survey. Ann Rheum Dis 46: 277-281, 19117. 9. Me N, Rygg M: Epidemiology of juvenile chronic arthrits in northern Norway: a ten-year retrospective study. Clin Exp Rheumatol 16: 99--101, 19p8. 10. Fujikawa S, Okuni M: A nationwide surveillance study of rheumatic diseases a~ong japanese children. Acta Paediatr jpn 39: 242-244, 1997. 11. Kunnamo I, Kallio P, Pelkonen P: Incidence of arthritis in urban Finnish children: a prospective study. Arthritis Rheum 29: 1232-1238, 1986. 12. M~nners Pj, Diepeveen DA: Prevalence of juvenile chronic arthritis in a population of 12-year-old children in urban Australia. Pediatrics 98:84--90, 1996. 13. Sullivan DB, Cassidy]T, Petty RE: Pathogenic implications of age of onset in juyenile rheumatoid arthritis. Arthritis Rheum 18: 251-255, 1975. 14. C~ssidy]T, Sullivan DB, Petty RE: Clinical patterns of chronic iridocyclitis in cljildren with juvenile rheumatoid arthritis. Arthritis Rheum 20 (Suppll: 224--227, 1977. 15. Cl11ylack LT jr: The ocular manifestations of juvenile rheumatoid arthritis. Arthritis Rheum 20 (Supp!): 217-223, 1977. 16. Sdhaller j, Kupfer C, Wedgwood Rj: Iridocyclitis in juvenile rheumatoid arthritis. Pediatrics 44: 92-100, 1969. 17. Stte Y, Kidon M, Koh E: More males and less uveitis in Asian children with juvenile idiopathic arthritis in Singapore. Clin Exp Rheumatol 21: 536, 2003. 18. Moroldo MB, Chaudhari M, Shear E, et al: juvenile rheumatoid arthritis affected sibpairs: Extent of clinical phenotype concordance. Arthritis Rheum 5IP: 1928-1934, 2004. 19. Murray Kj, Moroldo MB, Donnelly P, et al: Age-specific effects of juvenile rheumatoid arthritis-associated HLA genes. Arthritis Rheum 42: 1843-1853, 1'P99. 20. Oen K, Petty RE, Schroeder ML: An association between HLA-A2 and juvenile rheumatoid arthritis in girls. j Rheumatol 9: 916-920, 1982. 21. Zrggini E, Donn RP, Oilier WER, Thomson W, and the British Paediatric Rjleumatology Study Group: Evidence of linkage of HLA loci in juvenile icliopathic oligoarthritis: independent effects of HLA-A and HLA-DRB1. Arthritis Rheum 46: 2716-2720, 2002. 22. Sthaller jG, Ocbs H, Thomas ED. et al: Histocornpatibility antigens in childhood-onset arthritis. j Pediatr 88: 926-930, 1976. 23. Prieur AM: HLA B27 associated chronic arthritis in children: review of 65 Cases. Scand j Rheumatol Suppl66: 51-56, 1987. 24. Moroldo MB. Donnelly P, Saunders j, et al: Transmission disequilibrium as a ttfst of linkage and association between HLA alleles and pauciarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 41: 1620-1624, 1998. 25. Wordsworth P: Genes in the spondyloarthropathies. Rheum Dis Clin North Aim 24: 84~63, 1998. 26. Smerdel A , Ploski R, Flato B, et al: juvenile idiopathic arthritis (jIA) is primarily associated with HLA-DR8, but not DQ4 on the DR8-DQ4 haplotypes. Ainn Rheum Dis 61: 354--357, 2002. 27. !fall Pj, Burman Sj, Laurent MR, et al: Genetic susceptibility to early onset pauciarticular juvenile chronic arthritis: a study of HLA and complement markers in 158 British patients. Ann Rheum Dis 45: 464--474, 1986. 28. Raul C, Yao Z, Nevinny-Stickel C, et al: Immunogenetics of juvenile chronic alrthritis. I: HLA interaction between A2,DR5/8-DR/DQ and DPBl'0201 is a !leneral feature of all subsets of early onset pauciarticular juvenile chronic arthritis. II: DPB18 polymorphism plays a role in systemic juvenile chronic arthritis. Tissue Antigens 45: 280-283, 1995. 29. Stastny P, Fink CWo Different HLA-D associations in adult and juvenile rheumatoid arthritis. j Clin Invest 63: 124--130, 1979. 30. Glass 0, !irvin 0, Wallace K, et al: Early onset pauciarticular juvenile rheumatoid arthritis associated with human leukocyte antigen-DRw5, iritis 'lnd antinuclear antibody. j Clin Invest 66: 426-429, 1980. 31. Suciu-Foca, Godfrey M, Jacobs j, et al: Increased frequency of DRw5 in Jilauciarticular juvenile rheumatoid arthritis. In Terasaki P (ed): Histocompatibility Testing 1980. Los Angeles, University of California Press, 1980, p 953.
II
OLIGOARTHRITIS
287
32. Sher MR, Schultz jS, Ragsdale CG, et al: HLA-DR and MT associations with the clinical and serologic manifestations of pauciarticular onset juvenile rheumatoid arthritis. j Rheumatol 12: 1141-118, 1985. 33. Morling N, Friis j, Fugger L, et al: DNA polymorphoisms of HLA class II genes in pauciarticular juvenile rheumatoid arthritis. Tissue Antigens 38:16-23, 1991. 34. Malagon C, Van Kerckhove C, Giannini EH, et aI: The iridocyclitis of early onset pauciarticular juvenile rheumatoid arthritis: outcome in immunogenetically characterized patients. j Rheumatol 19: 160-163, 1992. 35. Prahalad S, Ryan MH, Shear ES, et al: juvenile rheumatoid arthritis: linkage to HLA demonstrated by allele sharing in affected sibpairs. Arthritis Rheum 43: 2335-2338, 2004. 36. Dracou C, Constantinidou N, Constantopoulos A: juvenile chronic arthritis profile in Greek children. Acta Paediar jpn 40: 558--563, 1998. 37. Haas jP, Andreas A, Rutkowski B, et al: A model for the role of HLA-DQ molecules in the pathogenesis of juvenile chronic arthritis. Rheumatol Int 11:191-197, 1991. 38. Haas jP, Kimura A, Truckenbrodt H, et al: Early-onset pauciarticular juvenile chronic arthritis is associated with a mutation in the Y-box of the HLA-DQAl promoter. Tissue Antigens 45: 317-321, 1995. 39. Nepom BS, Milhotra U, Schwarz DA, et al: HLA and T cell receptor polymorphisms in pauciarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 34: 1260-1267, 1991. 40. Glass ON, Giannini EH: jRA as a complex genetic trait. Arthritis Rheum 42: 2261-2268, 1999. 41. Van Kerckhove C, Luyrink L, Taylor j, et al: HLA-DQAl'OI01 haplotypes and disease outcome in early onset pauciarticular juvenile rheumatoid arthritis. j Rheumatol 18: 874--879, 1991. 42. Murray Kj, Szer W, Grom AA, et al: Antibodies to the 45 kDa DEK nuclear antigen in pauciarticular onset juvenile rheumatoid arthritis and iridocyclitis: selective association with MHC gene. j Rheumatol 24: 560-567, 1997. 43. Adams BS, Tan HMO: Allele-specific DQAl promotor binding by DEK: A putative autoantigen in juvenile rheumatoid arthritis. Arthritis Rheum 41: SI88, 1998. 44. Forero L, Zwirner NW, Fink CW, et al: juvenile arthritis, HLA-A2 and binding of DEK oncogene-peptides. Hum Immunol 59: 443-450, 1998. 45. Szer IS, Sierakowska H, Szer W: A novel autoantibody to the putative oncoprotein DEK in pauciarticular onset juvenile rheumatoid arthritis. j Rheumatol 21: 2136-2142, 1994. 46. Dong X, Wang j, Kabir FN, et al: Autoantibodies to DEK oncoprotein in human inflammatory disease. Arthritis Rheum 43: 85-93, 2000. 47. Donn RP, Thomson W, Pepper L, et al: Antinuclear antibodies in early onset pauciarticular juvenile chronic arthritis (jCA) are associated with HLADQBl'0603: a possible JCA-associated human leucocyte antigen haplotype. Br j Rheumatol 34: 461-465, 1995. 48. Morling N, Hellesen C, Jakobsen BK, et al: HLA-A, B, C, 0, DR antigens and primed lymphocyte typing (PLTl defined DP-antigens in juvenile chronic arthritis. Tissue Antigens 17: 433-441, 1981. 49. Hoffman RW, Shaw S, Francis LC, et al: HLA-DP antigens in patients with pauciarticular juvenile rheumatoid arthritis. Arthritis Rheum 29: 1057-1062, 1986. 50. Begovich AB, Bugawan TL, Nepom BS, et al: A specific HLA-DP beta allele is associated with pauciarticular juvenile rheumatoid arthritis but not adult rheumatoid arthritis. Proc Nat! Acad Sci USA 86: 9489-9493, 1989. 51. Paul C, Schoenwald U, Truckenbrodt H, et al: HLA-DP/DR interaction in early onset pauciarticular juvenile chronic arthritis. Immunogenetics 37: 442-448, 1993. 52. Scholz S, Albert ED: Immunogenetic aspects of juvenile chronic arthritis. Clin Exp Rheumatol 11 (Supp!) 9: S37-S41, 1993. 53. Thompson SO, Moroldo MB, Guyer L, et aI: A genome-Wide scan for juvemle rheumatoid arthritis in affected sibpair families provides evidence of linkage. Arthritis Rheum 50: 2920-2930, 2004. 54. Forre 0, Dobloug jH, Hoyeraal HM, et al: HLA antigens in juvenile arthritis: genetic basis for the different subtypes. Arthritis Rheum 26: 35-38, 1983. 55. Albert ED, Scholz S: juvenile arthritis: genetic update. Baillieres cUn Rheumatol 12: 209--218, 1998. 56. Feichtlbauer P, Gomolka M, Brunnler G, et al: HLA region rnicrosatellite polymorphisms in juvenile arthritis. Tissue Antigens 52: 220-229, 1998. 57. Ploski R, Forre 0: Non-HLA genes and susceptibility to juvenile chronic arthritis. CUn Exp Rheumatol 12 (Suppl) 10: SI5-S17, 1994. 58. Donn RP, Davies Ej, Holt PL, et al: Increased frequency of TAP2B in early onset pauciarticular juvenile chronic arthritis. Ann Rheum Dis 53: 261-264, 1994. 59. Ploski R, Undlien DE, Vinje 0, et aI: Polymorphism of human major histocompatibility complex-encoded transporter associated with antigen processing (TAP) genes and susceptibility to juvenile rheumatoid arthritis. Hum Immunol 39: 54-60, 1994. 60. pryhuber KG, Murray Kj, Donnelly P, et al: Polymorphism in the LMP2 gene influences disease susceptibility and severity in HLA-B27 associated juvenile rheumatoid arthritis. j Rheumatol 23: 747-752, 1996.
288
C HAP T E R
II 0
LIGOARTHRITIS
61. Ploski R, McDowell n, Symons .lA, et al: Interaction between HLA-DR and HLA-DP, and between HLA and interleukin I alpha in juvenile rheumatoid arthritis indicates heterogeneity of pathogenic mechanisms of the disease. Hum Immunol 42: 343-347, 1995. 62. Vencovsky .I, Jarosova K, Ruzickova S, et al: Higher frequency of allele 2 of the interleukin-I receptor antagonist gene in patients with juvenile idiopathic arthritis, Arthritis Rheum 44: 2387-2391, 2001. 63. McDowell n, Symons .lA, Ploski R, et al: A genetic association between juvenile rheumatoid arthritis and a novel interleukin-I alpha polymorphism. Arthritis Rheum 38: 221-228, 1995. 64. Crawley E, Kay R, Sillibourne .I, et al: Polymorphic haplotypes of the interleukin-IO 5' flanking region determine variable interleukin-IO transcription and are associated with particular phenotypes of juvenile rheumatoid arthritis. Arthritis Rheum 42: 1101-1108, 1999. 65. Nikitina Zake L, Cimdina I, Rumba I, et al: Major histocompability complex class I chain related (MIC) A gene, TNFa microsatellite alleles and lliFH alleles in juvenile idiopathic arthritis patients from Latvia, Hum Immunol 63: 418-423, 2002. 66. Pociot F, Briant L, Jongeneel CV, et al: Association of tumor necrosis factor (TNF) and class 11 major histocompatibility complex alleles with the secretion of TNF-alp and TNF-beta by human mononuclear cells: a possible link to insulin-dependent diabetes mellitus. EurJ Immunol 23: 224--231, 1993. 67. Zeggini E, Thompson W, Kwiatkowski 0, et al: Linkage and association studies of single-nucleotide polymorphism-tagged tumor necrosis factor haplotypes in juvenile oligoarthritis. Arthritis Rheum 46: 3304--3311, 2002. 68. Moroldo MB, Tague BL, Shear ES, et al: Juvenile rheumatoid arthritis in affected sibpairs. Arthritis Rheum 40: 1962-1966, 1997. 69. Mason T, Rabinovich CE, Fredrickson DO, et al: Breast feeding and the development of juvenile rheumatoid arthritis. .I Rheumatol 22: 1166-1170, 1995. 70. Rosenberg AM: Evaluation of associations between breast feeding and subsequent development of juvenile rheumatoid arthritis. .I Rheumatol 23: 1080-1082, 1996. 7J. Maksymowych WP, Gabriel CA, Luyrink L, et al: Polymorphism in a T-cell receptor variable gene is associated with susceptibility to a juvenile rheumatoid arthritis subset. Immunogenetics 35: 257-262, 1992. 72. Grom AA, von Knorre C, Murray KJ, et al: T-cell receptor BV6S1 null alleles and HLA-DRI haplotypes in polyarticular outcome juvenile rheumatoid arthritL•. Hum Immunol 45: 152-156, 19%. 73 Ploski R, Hansen T, Forre 0: Lack of association with T-cell receptor TCRBV6s1'2 allele in HLA-DQAl'OI01-positive Norwegian juvenile chronic arthritis patients, Immunogenetics 38: 444--445, 1993. 74. Nepom BS, Malhotra U, Schwarz DA, et al: HLA and T cell receptor polymorphisms in pauciarticular-onset juvenile rheumatoid arthritis. Arthritis Rheum 34: 1260-1267, 1991. 75. Thompson SO, Murray KJ, Grom AA, et al: Comparative sequence analysis of the human T cell receptor beta chain in juvenile rheumatoid arthritis and juvenile spondylarthropathies: evidence for antigenic selection of T cells in the synovium. Arthritis Rheum 41: 482-497, 1998. 76. Murray KJ, Luyrink L, Grom AA, et al: Immunohistological characteristics of T cell infiltrates in different forms of childhood onset chronic arthritis. .I Rheumatol 23: 2116-2124, 1996. 77. Murray K), Grom AA, Thompson SO, et al: Contrasting eytokine profiles in the synovium of different forms of juvenile rheumatoid arthritis and juvenile spondyloarthropathy: prominence of interleukin 4 in restricted disease. ) Rheumatol 25: 1388--1398, 1998. 78. )ung F, Neuer G, Bautz FA: Antibodies against a peptide sequence located in the linker region of the HMG-1/2 box domains in sera from patients with juvenile rheumatoid arthritiS. Arthritis Rheum 40: 1803-1809, 1997. 79. Rosenberg AM, Cordeiro OM: Relationship between sex and antibodies to high mobility group proteins 1 and 2 in juvenile idiopathic arthritis. ) Rheumatol 27: 2489-2493, 2000. 80. McMurray RW, Allen SH, Pepmueller PH, et al: Elevated serum prolactin levels in children with juvenile rheumatoid arthritis and antinuclear antibody seropositivity. .I Rheumatol 22: 1577-1580, 1995. 81. Picco P, Gattorno M, Buoncompagni A, et al: Prolactin and interleukin 6 in prepubertal girls with juvenile chronic arthritis. ) Rheumatol 25: 347-351, 1998. 82. Picco P, Gattorno M, Buoncompagni A, et al: Interactions between prolactin and the proinflammatory eytokine network in juvenile chronic arthritis. Ann N Y Acad Sci 876: 262-265, 1999, 83. de Graeff-Meeder ER, van der Zee R, Rijkers GT, et al: Recognition of human 60 kD heat shock protein by mononuclear cells from patients with juvenile chronic arthritis. Lancet 337: 1368--1372, 1991. 84. Boog CJ, de Graeff-Meeder ER, Lucassen MA, et al: Two monoclonal antibodies generated against human hsp60 show reactivity with synovial membranes of patients with juvenile chronic arthritis.) Exp Med 175: 1805-1810, 1992. 85. de Graeff-Meeder ER, Rijkers GT, Voorhorst-Ogink MM, et al: Antibodies to human HSP60 in patients with juvenile chronic arthritis, diabetes mellitus, and cystic fibrosis. Pediatr Res 34: 424--428, 1993. 86. de Graeff-Meeder ER, van Eden W, Rijkers GT, et al: Heat-shock proteins and juvenile chronic arthritis, Clin Exp Rheumatol 11 (Supp!) 9: S25-S28, 1993.
87. Life P, Hassell A, Williams K, et al: Responses to gram negative emeric bacterial antigens by synovial T cells from patients with juvenile chronic arthritis: recognition of heat shock protein HSP60, ) Rheumatol 20: 1388--1396, 1993. 88. Albani S, Ravelli A, Massa M, et al: Immune responses to the Escherichia coli dna) heat shock protein in juvenile rheumatoid arthritis and their correlation with disease actiVity. ) Pediatr 124: 561-565, 1994. 89. Prakken AB, van Hoeij M), Kuis W, et al: T-cell reactivity to human HSp60 in oligo-articular juvenile chronic arthritis is associated with a favorable prognosis and the generation of regulatory cytokines in the inflamed joint. Immunol Lett 57: 139-142, 1997. 90. Prakken AB, van Eden W, Rijkers GT, et al: Autoreactivity to human heatshock protein 60 predicts disease remission in oligoarticular juvenile rheumatoid arthritis. Arthritis Rheum 39: 1826-1832, 1996. 91. Conroy SE, Tucker L, Latchman OS, et al: Incidence of anti Hsp 90 and 70 antibodies in children with SLE, juvenile dermatomyositis and juvenile chronic arthritis. Clin Exp Rheumatol 14: 99-104, 1996. 92. van Eden W, Anderton SM, van der Zee R, et al: (Altered) self peptides and the regulation of self reactivity in the peripheral T cell pool. Immunol Rev 149: 55-73, 1996. 93. van der Zee R, Anderton SM, Prakken AB, et al: T cell responses to conserved bacterial heat-shock-protein epitopes induce resistance in experimental autoimmunity. Semin Immunol 10: 35-41, 1998. 94. van Eden W, van der Zee R, Paul AG, et al: Do heat shock proteins control the balance of T-cell regulation in inflammatory diseases? Immunol Today 19: 303-307, 1998. 95. van Eden W, van der Zee R, Taams LS, et al: Heat-shock protein T-cell epitopes trigger a spreading regulatory control in a diversified arthritogenic Tcell response. Immunol Rev 164: 169-174, 1998. 96, Prakken B, Wauben M, van Kooten P, et aI: Nasal administration of arthritisrelated T cell epitopes of heat shock protein 60 as a promising way for immunotherapy in chronic arthritis. Biotherapy 10: 205-211, 1998. 97. Huemer C, Malleson PN, Cabral DA, et al: Patterns of joint involvement at onset differentiate oligoarticular juvenile psoriatic arthritis from pauciarticular juvenile rheumatoid arthritis. ) Rheumatol 29: 1531-1535, 2002. 98. Sharma 5, Sherry DO: Joint distribution at presentation in children with pauciarticular arthritis. ) Pediatr 134: 642-643, 1999. 99. Cassidy)T, Brody GL, Martel W: Monarticular juvenile rheumatoid arthritis. ) Pediatr 70: 867-875, 1967. 100. Bywaters EGL, Ansell BM: Monoarticular arthritis in children. Ann Rheum Dis 24: 116, 1965. 101. Schaller .I, Wedgwood R): Pauciarticular juvenile rheumatoid arthritis. Arthritis Rheum 12: 330, 1969. 102. Brewer E) )r: Pitfalls in the diagnosis of juvenile rheumatoid arthritis. Pediatr Clin North Am 33: 1015-1032, 1986. 103. Cassidy./T: Miscellaneous conditions associated with arthritis in children. Pediatr Clin North Anl 33: 1033-1052, 1986. 104. Cabral DA, Tucker LB: Malignancies in children who initially present with rheumatic complaints. ) Pediatr 134: 53-57, 1999. 105. Rush P), Shore A, Wilmot 0, et al: Discoid meniscus presenting as juvenile rheumatoid arthritis.) Rheumatol 13: 1173-1177, 1986, 106, Lepore L, Martelossi S, Pennesi M, et al: Prevalence of celiac disease in patients with juvenile chronic arthritis, .I Pediatr 129: 311-313, 1996. 107. Falcini F, Ferrari R, Simonini G, et al: Recurrent monoarthritis in an ll-yearold boy with occult coeliac disease: successful and stable remission after gluten-free diet. Clin Exp Rheumatol 17: 509-511, 1999. 108. Miller JJ III, Emery HM: Migrating monopredominant arthritis in children of Assyrian ancestry. J Rheumatol 23: 178--180, 1996. 109. Dubourg G, Koeger AC, Huchet B, et al: Acute monoarthritis in a patient under isotretinoin. Rev Rhum Engl Ed 63: 228--229, 1996. 110, De Francesco V, Stinco G, Campanella M: Acute arthritiS during isotretinoin treatment for acne conglobata. Dermatology 194: 195, 1997. 111. Bajaj S, Bell M), Shumak S, et al: Antithyroid arthritis syndrome. J Rheumatol 25: 1235-1239, 1998. 112. Mathieu E, Fain 0, Sitbon M, et al: Systemic adverse effect of antithyroid dnlgs. Clin Rheumatol 18: 66-68, 1999. 113. Burgos-Vargas R, Vazquez-Mellado): The early clinical recognition of juvenile-onset ankylosing spondylitis and its differentiation from juvenile rheumatoid arthritis. Arthritis Rheum 38: 835-844, 1995. 114. Allen Re, Dewez p. Stuart L, et al: Antinuclear antibodies using HEp-2 cells in normal children and in children with common infections, ) Paediatr Child Health 27: 39-42, 1991. 115. Cabral DA, Petty RE, Fung M, et al: Persistent antinuclear antibodies in children without identifiable inflammatory rheumatic or autoimmune disease. Pediatrics 89: 441-444, 1992. 116. Fink CW, Dich VQ, Howard) )r, et al: Infections of bones and joints in children. Arthritis Rheum 20: 578--583, 1977. 117. Kornreich HK, Bernstein RH, Key KK, et al: The rheumatic presentation of osteomyelitis. Arthritis Rheum 22: 631, 1979. 118. Konttinen YT, Bergroth V, Kunnamo I, et al: The value of biopsy in patients with monarticular juvenile rheumatoid arthritis of recent onset. Arthritis Rheum 29: 47-53, 1986. 119. Jacobs )C, Phillips PE, Johnston AD: Needle biopsy of the synoviuI1l of children. Pediatrics 57: 696-701, 1976.
C HAP T E R 120. CO(.perman DR, Emery H, Keller C: Factors relating to hip joint arthritis following three childhood diseases-juvenile rheumatoid arthritis, Perthes disea~e, and postreduction avascular necrosis in congenital hip dislocation. ] Pediatr Orthop 6: 706--712, 1986. 121. Hellstrom B: The diagnosis and course of rheumatoid arthritis and benign astlptic arthritis in children. Acta Paediatr Scand 50: 529, 1961. 122. Jambs BW: Synovitis of the hip in children and its significance. Pediatrics 47: 558--566, 1971. 123. Chylack LT ]r: The ocular manifestations of juvenile rheumatoid arthritis. Arthritis Rheum 20 (Supp!): 217-223, 1977. 124. Chadwick A], Rosen ES: Papillitis and Still's disease. Am] Ophthalmol 65: 784-787, 1968. 125. ]o~e DG, Good RA: Iridocyclitis and pauciarticular juvenile rheumatoid arthritis.] Pediatr 78: 910--911, 1971. 126. Kanski ]]: Anterior uveitis in juvenile rheumatoid arthritis. Arch Ophthalmol 95: 1794-1797, 1977. 127. Ohm]: Bandformige Hornhauttrubung bei einem neunjhrigen Madchen und ih~e Behandlung mit subkonjunktivalen ]odkalium-einspritzungen. Kiin Monatsbl Augenheilkd 48: 243, 1910. 128. FrIedlander A: 2 Tilfaelde af kronisk septisk PolyartritL_ i Barnealderen med OJ,,nkomplikationer. Ugeskr Laeger 95: 1190, 1933. 129. H<;>lm E: Iridocyclitis and ribbon-like keratitis in cases of infantile polyarthritis (Still's disease). Trans Ophthalmol Soc U K 55: 478, 1935. 130. Bh"gvad 0: Iridocyclitis and disease of the joints in children. Acta Ophthalmol (Copenh) 19: 219, 1941. 131. Silry B: Rheumatoid Arthritis in Children. A Clinical Study. Copenhagen, Mlmksgaard, 1952. 132. K<;>taniemi K, Kaipiainen-Seppanen 0, Savolainen A, et al: A populationbased study on uveitis in juvenile rheumatoid arthritis. Clin Exp Rheumatol 11: 119-122, 1999. 133. Chylack LT ]r, Bienfang DC, Bellows AR, et aI: Ocular manifestations of juvenile rheumatoid arthritis. Am] Ophthalmol 79: 1026-1033, 1975. 134. Smiley WK: The eye in juvenile chronic polyarthritis. Clin Rheum Dis 2: 413, 1976. 135. S
II
OLIGOARTHRITIS
289
155. Melin-Aldana H, Giannini EH, Taylor ], et al: Human leukocyte antigenDRBl'1104 in the chronic iridocyclitis of pauciarticular juvenile rheumatoid arthritis.] Pediatr 121: 56-60, 1992. 156. Paroli MP, Speranza S. Marino M, et al: Prognosis of juvenile rheumatoid arthritis-associated lIveitis. Eur] Ophthalmol 13: 616-621, 2003. 157. Akduman L, Kaplan H], Tychsen L: Prevalence of uveitis in an outpatient juvenile arthritis clinic: onset of uveitis more than a decade after onset of arthritis.] Ophthalmic Nurs Technol 16: 177-182, 1997. 158. Kanski ]]: Juvenile arthritis and uveitis. Surv Ophthalmol 34: 253-267, 1990. 159. Petty RE: Current knowledge of the etiology and pathogenesL_ of chronic uveitis accompanying juvenile rheumatoid arthritis. Rheum Dis Clin North Am 13: 19-36, 1987. 160. Rosenberg AM, Oen KG: The relationship between ocular and articular disease activity in children with juvenile rheumatoid arthritis and associated uveitis. Arthritis Rheum 29: 797--800, 1986. 161. Sabates R, Smith T, Apple D: Ocular histopathology in juvenile rheumatoid arthritis. Ann Ophthalmol 11: 733-737, 1979. 162. Chylack LT ]r, Dueker DK, Pihlaja 0]: Ocular manifestations of juvenile rheumatoid arthritis: pathology, fluorescein iris angiography, and patient care patterns. In Miller ]] III (ed): Juvenile Rheumatoid Arthritis. Littleton, MA, PSG, 1978, P 149. 163. Merriam ]C, Chylack LT ]r, Albert DM: Early-onset pauciarticular juvenile rheumatoid arthritis: s histopathologic study. Arch Ophthalmol 101: 1085-1092, 1983. 164. Godfrey WA, Lindsley CB, Cuppage FE: Localization of IgM in plasma cells in the iris of a patient with iridocyclitis and juvenile rheumatoid arthritis. Arthritis Rheum 24: 1195-1198, 1981. 165. Hollwich F, Oamaske E: Eye symptoms in Still's disease. Med Monatsschr 22: 109-114, 1968. 166. Hinzpeter EN, Naumann G, Bartelheimer HK: Ocular histopathology in Still's disease. Ophthalmol Res 2: 16, 1971. 167. Rahi AH, Kanski ]], Fielder A: Immunoglobulins and antinuclear antibodies in aqueous humour from patients with juvenile "rheumatoid" arthritis (StHl's disease). Trans Ophthalmol Soc U K 97: 217-222, 1977. 168. Kanski 1/: Clinical and immunological study of anterior uveitis in juvenile chronic polyarthritis. Trans Ophthalmol Soc U K 96: 123-130, 1976. 169. Person DA, Leatherwood CM, Brewer E], et al: Immunology of the vitreous in juvenile rheumatoid arthritis. Arthritis Rheum 24: 591, 1981. 170. Kaplan H], Aaberg TM, Keller RH: Recurrent clinical uveitis: cell surface markers on vitreous lymphocytes. Arch Ophthalmol 100: 585-587, 1982. 171. Kanski J]: Care of children with anterior uveitis. Trans Ophthalmol Soc U K 101 (Pt 3): 387-390, 1981. 172. Perkins ES: Pattern of uveitis in children. Br] Ophthalmol50: 169-185, 1966. 173. Ohno S, Miyajima T, Higuchi M, et al: Ocular manifestations of Kawasaki'S disease (mucocutaneous lymph node syndrome). Am ] Ophthalmol 93: 713-717, 1982. 174. Ladd ]R, Cassidy ]T, Martel W: Juvenile ankylosing spondylitis. Arthritis Rheum 14: 579-590, 1971. 175. Shore A, Ansell BM: ]uvenile psoriatic arthritis-an analysis of 60 cases.] Pediatr 100: 529-535, 1982. 176. Lindsley CB, Schaller ]G: Arthritis associated with inflammatory bowel disease in children.] Pediatr 84: 16, 1974. 177. Passo M, Brandt K, Fitzgerald J: Arthritis associated with inflammatory bowel disease in children: relationship between arthritis and activity of the inflammatory bowel disease. Arthritis Rheum 22: 645, 1979. 178. Rankin GB, Watts HD, Melnyk CS, et al: National Cooperative Crohn's Disease Study: extraintestinal manifestations and perianal complications. Gastroenterology 77: 914-920, 1979. 179. Davies NE, Haverty JR, Boatwright M: Reiter's disease associated with shigellosis. South Med] 62: 1011-1014, 1969. 180. Iveson ]M, Nanda BS, Hancock ]A, et al: Reiter's disease in three hoys. Ann Rheum Dis 34: 364-368, 1975. 181. Ansell MB, Bywaters EG, Elderkin FM: Familial arthropathy with rash, uveitis and mental retardation. Proc R Soc Med 68: 584-585, 1975. 182. Prieur AM, Griscelli C: Arthropathy with rash, chronic meningitis, eye lesions, and mental retardation. ] Pediatr 99: 79-83, 1981. 183. Kaufman RA, Lovell DJ: Infantile-onset multisystem inflammatory disease: radiologic fmdings. Radiology 160: 741-746, 1986. 184. Yarom A, Rennebohm RM, Levinson ]E: Infantile multisystem inflammatory disease: a specific syndrome? ] Pediatr 106: 390--396, 1985. 185. Lindsley CB, Godfrey WA: Childhood sarcoidosis manifesting as juvenile rheumatoid arthritis. Pediatrics 76: 765-768, 1985. 186. Hoover DL, Khan ]A, Giangiacomo J: Pediatric ocular sarcoidosis. Surv Ophthalmol 30: 215-228, 1986. 187. Blau EB: Familial granulomatous arthritis, iritis, and rash. ] Pediatr 107: 689-693, 1985. 188. Pastores GM, Michels W, Stickler GB, et al: Autosomal dominant granulomatous arthritis, uveitis, skin rash, and synovial cysts. ] Pediatr 117: 403-408, 1990. 189. Petty RE, Cassidy]T, Sullivan DB: Clinical correlates of antinuclear antibodies in juvenile rheumatoid arthritis.] Pediatr 83: 386-389, 1973. 190. Schaller ]G, Johnson GD, Holborow E], et al: The association of antinuclear antibodies with the chronic iridocyclitis of juvenile rheumatoid arthritis (Still's disease). Arthritis Rheum 17: 409-416, 1974. 191. Rosenberg AM, Romanchuk KG: Antinuclear antibodies in arthritic and nonarthritic children with uveitis. ] Rheumatol 17: 60--61, 1990.
290
C HAP T E R
II
OLIGOARTHRITIS
192. Permin H, Horbov S, Wiik A, et al: Antinuclear antibodies in juvenile chronic arthritis. Acta Paediatr Scand 67: 181-185, 1978. 193. Hoyeraal HM: Granulocyte reactive antinuclear factors in juvenile rheumatoid arthritis. Scand 1 Rheumatol 5: 84-90, 1976. 194. Neuteboom GH, Hertzberger-ten Cate R, de long 1, et al: Antibodies to a 15 kD nuclear antigen in patients with juvenile chronic arthritis and uveitis. Invest Ophthalmol Vis Sci 33: 1657-1660, 1992. 195. Leak AM, Woo P: luvenile chronic arthritis, chronic iridocyclitis, and reactivity to histones. Ann Rheum Dis 50: 653-657, 1991. 196. Leak AM, Tuaillon N, Muller S, et al: Study of antibodies to histones and histone synthetic peptides in pauciarticular juvenile chronic arthritis. Br 1 Rheumatol 32: 426-431, 1993. 197. Massa M, De Benedetti F, Pignatti P, et al: Lack of temporal association of iridocyclitis with IgG reactivities to core histones and nucleosome subpartides in pauciarticular juvenile chronic arthritis. Br 1 Rheumatol 34: 507-511, 1995. 198. Nguyen QD, Foster CS: Saving the vision of children with juvenile rheumatoid arthritis-associated uveitis. lAMA 280: 1133-1134, 1998. 199. Olson NY, Lindsley CB, Godfrey WA: Treatment of chronic childhood iridocyclitis with nonsteroidal anti-inflammatory dmgs. 1 Allerg Clin Immunol 79: 220, 1981. 200. March WF, Coniglione TC: Ibuprofen in the treatment of uveitis. Ann Ophthalmol 17: 103-104, 1985. 201. Dunne lA, lacobs N, Morrison A, et al: Efficacy in anterior uveitis of two known steroids and topical tolmetin. Br 1 OphthaJmol 69: 120-125, 1985. 202. Weiss AH, Wallace CA, Sherry DO: Methotrexate for resistant chronic uveitis in children with juvenile rheumatoid arthritis. 1 Pediatr 133: 266-268, 1998. 203. Shetty AK, Zganjar BE, Ellis GS lr, et al: Low-dose methotrexate in the treatment of severe juvenile rheumatoid arthritis and sarcoid iritis. 1 Pediatr Ophthalmol Strabismus 36: 125-128, 1999. 204. Mehra R, Moore n, Catalano 0, et aJ: Chlorambucil in the treatment of iridocyclitis in juvenile rheumatoid arthritis. 1 Rheumatol8: 141-144, 198]. 205. Palmer RG, Kanski 11, Ansell BM: Chlorambucil in the treatment of intractable uveitis associated with juvenile chronic arthritis. 1 Rheumatol ]2: 967-970, 1985, 206. Nussenblatt RB, Palestine AG, Chan CC: Cyclosporin A therapy in the treatment of intraocular int1ammatory disease resistant to systemic corticosteroids and cytotoxic agents. Am 1 Ophthalmol 96: 275-282, 1983. 207. Wizemann Al, Wizemann V: Therapeutic effects of short-term plasma exchange in endogenous uveitis, Anl1 Ophthalmol 97: 565-572, 1984. 208. Kilmartin 01, Forrester]V, Dick AD: Cyclosporin A therapy in refractory noninfectious childhood uveitis. Br 1 Ophthalmol 82: 737-742, 1998. 209. Rosenbaum lT, George RK, Gordon C: The treatment of refractory uveitis with intravenous immunoglobulin. Am 1 Ophthalmol 127: 545-549, 1999. 210. Reiff A, Takei S, Sadeghi S, et al: Etanercept therapy in children with treatment-resistant uveitis. Arthritis Rheum 44: 1411-1415, 2001. 2] 1. Reiff A: Long-term outcome of etanercept therapy in children with treatmentrefractory uveitis, Arthritis Rheum 48: 2079-2080, 2003. 212. Smith .lA, Smith S, Whitcup SM, et al: The treatment of lRA-associated uveitis with etanercept. Arthritis Rheum 46: S482, 2002. 213. Smith JR, Levinson RD, Holland GN, et al: Differential efficacy of tumor necrosis factor iMibition in the management of inflammatory eye disease and associated rheumatic disease. Arthritis Rheum 45: 252-257, 2001. 214, Hooper PL, Rao NA, Smith RE: Cataract extraction in uveitis patients, Surv Ophthalmol 35: ]20-144, 1990.
2I 5. Cimaz RG, Fink CW: The articular prognosis of pauciarticular onset juvenile arthritiS is not influenced by the presence of uveitis. 1 Rheumatol 23: 357-359, 1996. 216. Sherry DO, Mellins ED, Wedgwood R]: Decreasing severity of chronic uveitis in children with pauciarticular arthritiS. Am} Dis Child 145: 1026-1028. 1991. 217. Gori S, Broglia AM, Ravelli A, et al: Frequency and complications of chronic iridocyclitis in ANA-positive pauciarticular juvenile chronic arthritis. Int Ophthalmol 18: 225-228, 1994. 218, Leak AM, Ansell BM, Burman S}: Antinuclear antibody studies in juvenile chronic arthritis, Arch Dis Child 61: 168-172, 1986. 219, Hassfeld W, Vinje 0, Flato B, et al:Anti-citrulline autoantibodies in juvenile rheumatoid arthritis (IRA). Arthritis Rheum 48: S100, 2003. 220. Low 1M, Chauhan AK, Kietz DA, et al: Determination of anti-cyclinc citrulJinated peptide antibodies in the sera of patients with juvenile idiopathic arthritis. Arthritis Rheum 48 (Supp!): S99, 2003. 221. Van Rossum M, Van Soesbergen R, De Kort S, et al: Anti-cyclic citmlJinated peptide (anti-CCP) antibodies in children with juvenile idiopathic arthritis. J Rheumatol 30: 825-828, 2003, 222. Serra CRB, Rodrigues SH, Silva NP, et al. Clinical significance of anticardiolipin antibodies in juvenile idiopathic 31thritis. Clin Exp Rheumatol 17: 375-380, 1999. 223. Miller IT Ill, Hsu YP, Moss R, et al: The immunologic and clinical associations of the split products of C3 in plasma in juvenile rheumatoid arthritis. Arthritis Rheum 22: 502-507, 1979. 224. Oen K, Reed M, Malleson PN, et al: Radiologic outcome and its relationship to functional disability in juvenile rheumatoid arthritis. 1 Rheumatol 30: 832-840, 2003. 225. Ramsey SE, Cairns RA, Cabral DA, et al: Knee magnetic resonance imaging in chlldhood chronic monarthritis. .I Rheumatol 26: 2238-2243, 1999. 226. Sherry DO, Stein LD, Reed AM, et al: Prevention of leg length discrepancy in young children with pauciarticular juvenile rheumatoid arthritis by treatment with intraarticular steroids. Arthritis Rheum 42: 2330-2334, 1999. 227. Bowyer SL, Roettcher PA, Higgins GC, et al: Health status of patients with juvenile rheumatoid arthritis at 1 and 5 years after diagnosis. 1 Rheumatol 30: 394--400, 2003. 228. Oen K, Malleson PN, Cabral DA, et al: Disease course and outcome of JRA in a multicenter cohort, J Rheumatol 29: 1989-1999, 2002, 229. AI-Matar MJ, Petty RE, Tucker LB, et al: The early pattern of joint involvement predicts disease progression in children with oligoarticular (pauciarticular) juvenile rheumatoid arthritis, Arthritis Rheum 46: 2708-271 S, 2002. 230. Oen K, Malleson PN, Cabral DA, et al: Early predictors of longterm outcome in patients with juvenile rheumatoid arthritis: subset-specific correlations. J Rheumatol 30: 585-593, 2003. 231. Fantini F, Gerloni V, Gattinara M, et al: Remission in juvenile chronic arthritis: a cohort study of 683 consecutive cases with a mean 10 year fol!owup. J Rheumatol 30: 579-584, 2003. 232. Flato B, Lien G, Smerdel A, et al: Prognostic factors in juvenile rheumatoid arthritis: a case-control study revealing early predictors and outcome after 14.9 years. J Rheumatol 30: 386-393, 2003, 233. Ruperto N, Ravelli A, Levinson lE, et al: Longterm health outcomes and quality of life in American and Italian inception cohorts of patients with juvenile rheumatoid arthritis, lJ: Early predictors of outcome. J Rheumatol 24: 952-958, 1997. 234, Minden K, Kiessling U, Listing J, et al: Prognosis of patients with juvenile chronic arthritiS and juvenile spondyJoarthopathy. J Rheumarol 27: 2256-2263, 2000.
12
C HAP T E R
SYSTEMIC ARTHRITIS Ross E. Petty and James T. Cassidy
~iIf
Systemic arthritis is one of the most perplexing diseases of childhood. The onset can be quite nonspecific and may suggest bacterial or viral infection, malignancy, or another rheumatic disease. The evolution of the disease eventually makes the diagnosis, which is entirely clinical, evident. For almost a century, this disorder bore the name "Still's disease," in recognition of its early description by George Frederic Still. I
DEFINITION AND CLASSIFICATION This disease-known as systemic arthritis by the International League of Associations for Rheumatology (ILAR) classification of juvenile idiopathic arthritis (J1A),2 systemic-onset juvenile rheumatoid arthritis (JRA) by the American College of Rheumatology (ACR) classification,3 or systemic-onset juvenile chronic arthritis (JCA) by the European League Against Rheumatism (EULAR) classificatiol14-is unique among the chronic arthritides of childhood in several ways. In particular, the range and severity of characteristic extra-articular features marks this disease as a systemic illness with joint inflammation that ranges from mild to severe. In some ways it resembles a fever of unknown origin. Diagnosis of systemic arthritis by the ILAR criteria requires the presence of arthritis and a documented quotidiah fever of at least 2 weeks' duration plus one of the following: typical rash, generalized lymphadenopathy, enlargement of liver or spleen, or serositis. Criteria and exchlsions are shown in Table 12-1.
EPIDEMIOLOGY Systemic arthritis is the least common type of chronic arthritis in childhood. It accounts for 10% to 15% of children with ]IA seen in rheumatology clinics in North America and Europe. Moe and Rygg5 estimated the annual incidence of systemic JCA (EULAR criteria) at 0.8/100,000 in Norway. Incidence data for systemic arthritis from elsewhere in the world are not available. Most studies indicate no definite peak age at onset of systemic arthritis.6-9 However, in a recent large multicenter study, there was a broad peak of onset between 1 and 5 years. lO It occasionally manifests before 1 year of age, and it also occurs in adolescence and adulthood.
Children of both sexes are affected with approximately equal frequency.6-9 This characteristic further differentiates systemic arthritis from other categories of JIA, in which females predominate. Systemic JIA is found worldwide and may account for a greater proportion of all childhood arthritis in Asia than in North America or Europe. At least three studies have addressed the questions of the seasonal occurrence of systemic arthritis In the earliest study, Lindsley' documented a marked seasonal variation in 28 children over a 10 year period, with no patients having disease onset in the winter months. A Canadian study of 221 children8 demonstrated no Significant seasonal variation overall, although there was some regional disparity, and in the prairie region onset of systemic arthritis was less common in the winter months. No seasonal variation was observed in 59 patients from IsraelY
ETIOLOGY AND PATHOGENESIS Clinically, systemic arthritis resembles an infectious disease, but it has not been consistently associated with any pathogen. Its low frequency (rarity) in siblings or other relatives and the absence of consistently identifiable seasonality of onset indicate that, if an infectious agent is involved in its etiology, it is only one of several factors that are needed to cause the disease.
Cytokine Profiles Abnormal expression of three of the most important proinflammatory cytokines-interleukin-6 (lL-6), IL-l, and tumor necrosis factor-a (TNF-a)-is characteristic of systemic JIA. De Benedetti and Martini l l suggested that systemic JIA is an IL-6-mediated disease. Evidence to support that hypothesis is strong. IL-6 is markedly elevated in the blood l2- 16 and synovial fluid. 12-14 The IL-6 level increases just before each fever spike and correlates with the systemic activity of the disease, arthritis, and increase in acute phase reactants.12.14.15 The abnormalities in regulation of IL-6 are also probably responsible for the limitation of growth, thrombocytosis. and microcytic anemia seen in this disease.12-14.17.18 Complexes of IL-6 with its soluble receptor, sIL-6R, prolong the activity of the cytokine and thereby potentiate its effects. 14 ,19 However, recent studies 20 have demonstrated an unidentified factor
291
292
I!.,
C HAP T E R
12
SYSTEMIC ARTHRITIS
TABt E 12 I Systemic JlIVI'l1ile Ic!lopdthi< Arlhritis IL AR Did<jl1osli( ("I i1l'rid
Arthritis in any number of joints together with a fever of at least 2 weeks' duration that is documented to be daily (quotidian) for at least 3 days and is accompanied by one or more of the following: • Evanescent rash • Generalized lymphadenopathy • Enlargement of liver or spleen • Serositis Exclusions: • Psoriasis or a history of psoriasis in the patient or a first-degree relative • Arthritis in an HLA B27 positive male beginning after the sixth birthday • Ankylosing spondylitis, enthesitis-related arthritis, sacroiliitis with inflammatory bowel disease, Reiter's syndrome, or acute anterior uveitis or a history of one of these disorders in a firstdegree relative • The presence of IgM RF on at least two occasions at least 3 mo apart HLA, human leukocyte antigen; IgM, immunoglobulin M; [LAR, International League of Associations for Rheumatology; RF, rheumatoid factor, Cassidy]T, Levinson ]E, Bass ]C, Baum], Brewer E], Fink CW, et al: A study of classification criteria for a diagnosis of juvenile rheumatoid arthritis, Arthritis Rheum 29(2): 274-281, 1986, Copyright © 1986]ohn Wiley & Sons, Inc, Reprinted with permission of Wiley-Liss, Inc"~ a subsidiary of John Wiley & Sons, Inc,
in sera from children with systemic JIA that inhibits the bioactivity of IL-6 by preventing binding of the IL-6 sIL6R complex to gp130. It has been suggested that polymorphisms of the IL-6 gene may affect IL-6 expression and contribute to cytokine dysregulation. 18 ,21 Two studies have confirmed a high frequency of the -174G allele of the IL-6 gene in systemic arthritis,lO,18 particularly in children with disease onset after 5 years of age. In vitro studies 22 have documented increased production of IL-6 by peripheral blood mononuclear cells from patients with systemic JIA. Furthermore, the inhibitory effect of IL-lO on IL-6 production was blunted in these patients. The presence of the -174 G/C allele did not correlate with either finding. IL-6 most likely induces an increase in the concentration of the interleukin-1 receptor antagonist, ILlRa, which is also coincident with the febrile peak,14-J6,23 A further imbalance in IL-6 homeostasis is suggested by the observations that the sIL-6R concentrations are significantly increased in children with systemic Jlk. 24 TNF-a levels are increased in systemic JIA, but, in contrast to IL-6, do not correlate with elevations of temperature,14 Polymorphisms of the TNF-a gene that are associated with high production of TNF-a are associated with systemic ]RA in Japanese children.25 There are marked increases in the soluble TNF-a receptor protein p55 (sTNFaRp55) in the blood, and lesser elevations of sTNFaRp75 in blood and synovial fluid. 16,26 Soluble IL-2R is increased in concentration; its levels correlate with the activity of the disease. 13 ,27-29 Serum IL-7 levels30 and IL-8 levels 31 ,32 are also increased.
Lymphocytes and Autoantibodies Studies of the phenotype and function of T lymphocytes have been contradictory. In some studies the frequency
of CD8+ T cells is increased,33,34 whereas in others it was decreased. 35 ,36 B lymphocyte numbers are increased:".3B Autoantibodies and immune complexes are characteristically not detected in children with systemic arthritis, and it seems unlikely that they play any major role in pathogenesis.
GENETIC BACKGROUND Systemic JIA is rarely familial. As described earlier, there is evidence that polymorphisms of cytokine genes may be important in determining the predisposition to systemic arthritis. Studies of major histocompatibility class II associations with systemic-onset JRA have yielded inconsistent results, and to date no strong, consistent association with any human leukocyte antigen (HLA) gene has been identified. DR5, DR8, Dw7, and possibly DR4,'w as well as DPB1 polymorphisms,40 have been reported to be more frequent in certain populations, but this is not consistently confirmed. In one study there was no association between DR4 and severe arthritis in children with systemic-onset JRA. 41 The frequencies of DRB1*0405 and DQB1·0401 were increased in Japanese children with systemic JRA, but not in those with other types of arthritis. 25 The presence of these HLA genes together with the high-production TNF-a gene was significantly associated with systemic disease (odds ratiO, 3.84).
CLINICAL MANIFESTATIONS Children with systemic arthritis are usually very ill. They are fatigued, anemic, febrile, and in pain. These features of the disease predominate early in the course and may overshadow or precede the presence of arthritis. The high frequency of extra-articular manifestations emphasizes the systemic nature of this disease (Table 12-2). Ten percent of children have onset of JIA with severe systemic involvement that may precede development of overt arthritis by weeks, months, or, rarely, years. The longest interval between onset of systemic signs and appearance of arthritis in the authors' patients has been approximately 10 years. The presence of arthritis must be confirmed, however, before a diagnosis of JRA can be considered definite.
~ -
II
TABLE 12 2 Eslill1dted Freqll('/lcy of Exlrd d1tiClIldl Ma/lifestations of Systemic JlIvelllle IdIopathic Arthl ills
Feature Fever Typical rash Hepatosplenomegaly Lymphadenopathy Pericarditis Pleuritis Abdominal pain
Frequency (%) 100
95 85 70
35 20 10
C HAP T E R
Mu~loskeletal
DATE
Disease
Arthritis Any number of joints can be affected at onset or during the dIsease course. Characteristically, the onset is oligoarticular, but the course is polyarticular. The knees, wrists, and ankles are most commonly involved, but cervical spine and hip disease, as well as inflammation of the small joints of the hands and the temporomandibular joint, occur in more than half of the patients. At onset of the systemic features, the joint disease may be minimal, but usually it increases in severity over weeks or months. In some children, the joint disease is a severe polyarthritis that is very resistant to treatment and can eventually result in significant disability. In others, it is less severe and eventually goes into clinical remission. Schneider and colleagues42 reported the development of severe destructive joint disease in 12 (32%) of 38 patients at a mean follow-up of 5 years.
Tenosynovitis The most common sites of tenosynovitis, which occurs in approximately 10% of children with systemic ]IA, are the extehsor tendon sheaths on the dorsum of the hand, the extensor sheaths over the dorsum of the foot, and those of the posterior tibial tendon and the peroneus longus and brevis tendons around the ankle. Occasionally, stenosing tenosynovitis results in trigger finger or loss of interphalangeal joint extension. Tenosynovitis of the superior oblique tendon may cause Brown's syndrome.
HOUR
·c
6/27
12 6/28
SYSTEMIC ARTHRITIS 6/29
6/30
7/1
712
293 7/3
48124$12481248124812481248124812481248124812481248124812 All PN All PII AN PII All PII All PII All PII All PI
·F
40.8 105 40.0 104 ~ 39.4 103 :;) 38.8 102 38.3 101 ~ 37.8 100 ~ 31.2 98 36.1 98 38.0 91
tc f5
150 130
110 90 10 50 30 " - -_ _- -.........._ - - ~ "'--~_ 10 u.............u.....................I...l..~.J...L..Jl..L..L ..............1...L.~ ............L...L.I.. u..LI • Figure lZ-l Intermittent fever of systemic-onset JIA in a 3-year-old girt The fever spikes usually occurred daily in the late evening to early morning (quotidian pattem), retumed to normal or below normal, and were accompanied by severe malaise, tachycardia, and rash.
Systemic Disease Fever The temperature rises to 39° C or higher on a daily or twice-daily basis, with a rapid return to baseline or below the baseline (Fig. 12-1). This quotidian pattern is highly suggestive of the diagnosis of ]RA-although very early in the course of the disease the classic quotidian fever may not be apparent, and the pattern may be indistinguishable from that of sepsis. The fever may occur at any time of the day but is characteristically present in the late afternoon to evening in conjunction with the rash. The temperature may be subnormal in the morning. Chills are frequent at the time of the fever, but rigor is rare. These children are often quite ill while febrile but may be surprisingly well during the rest of the day. Hyperpyrexia, a temperature higher than 40.5° C, is a rare but serious complication of an acute systemic onset. The fever must be present for at least 2 weeks to fulfill the diagnostic criteria, but it usually lasts for several months, recurs with flares of disease, and occasionally persists for years.
Rash The intermittent fever is almost always accompanied by a c1~ssic rash that consists of discrete, erythematous macules 2 to 5 mm in size (Fig. 12_2).43.44 This rash is usually described as being salmon pink, but very early in the
• Figure lZ-Z Typical rash of systemic-onset disease in a 3-year-old boy.The rash is salmon-colored, macular, and nonpruritic. Individual lesions are transient, occur in crops over the trunk and extremities, and may occur in a linear distribution (Koebner's phenomenon) after minor trauma such as ascratch. (See color insert.)
294
C HAP T E R
12
SYSTEMIC ARTHRITIS
disease it may be more erythematous, although never purpuric. It most commonly occurs on the trunk and proximal extremities but may develop on the face, palms, or soles. The macules are often surrounded by a zone of pallor, and larger lesions develop central clearing. The rash tends to be migratory and is strikingly evanescent in anyone area: Individual lesions disappear within a few hours and leave no residua. The rash may be much more persistent in children who are systemically very ill, and it may reappear with each systemic exacerbation. Individual lesions may be elicited by rubbing or scratching the skin (the Koebner phenomenon or isomorphic response) or by a hot bath or psychological stress. The rash is sometimes pruritic,45 particularly in older patients. A series of adolescents with a linear urticarial rash and a systemic inflammatory disease resembling systemic JIA, or possibly adult-onset Still's disease, was recently reported. 46 These interesting observations raise questions about the specificity of the systemic arthritis rash and the phenotype of systemic JIA in adolescents.
Cardiac Disease
Pericarditis The overall prevalence of pericardial involvement in JRA is estimated at 3% to 9%.47,48 Pericarditis and pericardial effusions occur almost exclusively in systemic-onset disease (see Figure 10-9).48--54 Pericarditis tends to occur in the older child, but it is not related to sex, age at onset, or severity of joint disease. 49 It may precede development of arthritis or occur at any time during the course of the disease, usually accompanied by a systemic exacerbation. Episodes typically persist for 1 to 8 weeks. Most pericardial effusions are asymptomatic, although some children have dyspnea or precordial pain that may be referred to the back, shoulder, or neck. Examination may document diminished heart sounds, tachycardia, cardiomegaly, and a pericardial friction rub, usually at the left lower sternal border (Table 12-3).52 In many cases, pericardial effusions develop insidiously, are not accompanied by obvious cardiomegaly or electrocardiographic changes, and escape recognition except by echocardiography.5o,53 In a study from Los Angeles,53 an effusion or pericardial thickening was present in 36% of patients, and 81% of children who had active systemic manifestations at the time of the study had abnormal echocardiographic findings (low voltage, ST-segment elevation, diffuse T-wave inversion). In more than half of these patients, pericarditis would not have been diagnosed without echocardiography. Tamponade is rare 47 ,51,S4-S7 and is characterized by venous distention, hepatomegaly, and peripheral edema. Pulsus paradoxus (wherein the pulse volume and systolic blood pressure decrease during deep inspiration) can be demonstrated. An enlarged cardiac silhouette is present on chest radiography, and pericardial fluid is confirmed by echocardiography. Chronic constrictive pericarditis is very rare SJ ,S5,S6,S8 and is characterized by pulsus paradoxus, a small heart, venous distention, ascites, and peripheral edema. In general, children with pericarditis do not fare worse than others in outcome. This complication should
I!.
TABLE 12 3
Mdnifeslalions of Pericdrdilis
Manifestation
Frequency (%)
Symptoms Precordial pain Dyspnea
38 20
Signs Tachycardia Friction rub Tachypnea
83 67 60
InvestlgaUons Radiography: cardiomegaly Electrocardiography: ST-segment elevation Electrocardiography: T-wave abnormality Echocardiography: effusion
71 31 71 >90
Adapted from Brewer E Jr: Juvenile rheumatoid arthritis: cardiac involvement. Arthritis Rheum 20 (SupplJ 231, 1977.
not necessarily be regarded as a poor prognostic sign, although pericarditis is frequently present in children with JRA who die. 49 ,S2
Myocarditis Myocarditis is much less common than pericarditis and may result in cardiomegaly and congestive heart failure. S9 ,60 In three cases reported by Miller and French,s9 failure occurred in the absence of overt pericardial effusions in children with severe active systemic disease, At necropsy, diffuse myocardial changes typical of congestive cardiomyopathy were present in one child. Goldenberg and colleagues48 reported four children with systemic JRA complicated by perimyocarditis and two with myocarditis in a retrospective study of 172 children withJRA.
Endocarditis Valvular disease, seemingly unrelated to other causes, has been documented in only two individuals with systemic JRA.61-62 Although these patients had onset of systemic arthritis in early childhood, aortic insufficiency was not diagnosed until years later. A good outcome was reported after placement of a prosthetic valve in one patient.
Pleuropulmonary Disease Parenchymal pulmonary disease is rare, but diffuse interstitial fibrosis occurs in a small number of children63-66 and may precede other evidence of JRA.67-69 Athreya and colleagues65 noted interstitial disease in 8 of 191 children with JRA, all of whom had a systemic onset. Another report detailed pathologic findings in a child who died of pulmonary fibrosis. 63 Pulmonary function studies in 16 children with JRA documented abnormalities in 10 of them. 67 In some children, such abnormalities may be the result of respiratory muscle weakness. 7o ,71 Pleural effusions
C HAP T E R
may occur with carditis, or they may be asymptomatic and be detected only as incidental findings on chest radiqgraphs. One child observed by the authors had idiopathic pulmonary hemosiderosis as the first sign of JRA; another had primary pulmonary hypertension. 72 Pulmonary rheumatoid nodules, as described in adult RA, are rare in childhood. The exceptional occurrence of pulmonUry interstitial and intra-alveolar cholesterol granulomas was reported in one patient.73
Lymphadenopathy and Splenomegaly Enlargements of lymph nodes and spleen may occur alone or together and are characteristic of systemic-onset JRA. Marked symmetrical lymphadenopathy is particularly common in the anterior cervical, axillary, and ing~inal areas and may suggest the diagnosis of lymphoma. Mesenteric lymphadenopathy can cause abdominal pain or distention and may lead to the erroneous diagnosis of an acute surgical abdomen. Splenomegaly usually is most prominent within the first years after onset. The degree of splenomegaly may
12
SYSTEMIC ARTHRITIS
295
be extreme (Fig. 12-3), but it has not been associated with Felty'S syndrome (splenic neutropenia) in children with systemic JIA. Functional hyposplenia, as seen in systemic lupus erythematosus, has not been reported in systemic JIA.
Macrophage Activation Syndrome The most devastating complication of systemic JIA is a rare syndrome, termed the macrophage activation syndrome (MAS), which bears close resemblance to reactive hematophagocytic lymphohistiocytosis (HLH) and is associated with serious morbidity and sometimes death (Table 12-4).74 MAS may be somewhat more common in boys, and it is most strongly associated with systemic arthritis (7%)75 although it has been reported in some patients with polyarthritis76 or adult-onset Still's disease,77 and the authors have observed it in a patient with polyarticular psoriatic arthritis. The initial reports described a syndrome of diffuse intravascular coagulation associated with hepatosplenomegaly, elevated serum levels of liver enzymes, and hematocytopenia in children with active systemic disease. 7s-BO The association of these abnormalities with HLH was noted by Hadchouel and colleagues. B1 Subsequently, a more complete description of the clinical syndrome has emerged. MAS is characterized clinically by the rapid development of fever, hepatosplenomegaly, lymphadenopathy, hepatic failure with encephalopathy, purpura, bruising, and mucosal bleeding,74.7B,Bl Laboratory studies indicate the presence of hematocytopenias, especially thrombocytopenia, elevated liver enzyme concentrations, mild hypoalbuminemia, and elevated triglycerides, and ferritin. The prothrombin time (PT) and the partial thromboplastin time (PTT) are prolonged, and blood levels of the vitamin K-dependent clotting factors are decreased. Fibrin degradation products are present in the plasma. The erythrocyte sedimentation rate (ESR) is paradoxically low in association with hypofibrinogenemia induced by consumptive coagulopathy and disseminated intravascular coagulation. Interferon-a (IFN-a), TNF-a, and other cytokines are increased. I I The demonstration of macrophages phagocytosing other hematopoietic cells in the bone marrow is diagnostic (Fig. 12-4),
I!,:. TABLE 12-4
Macrophage Activation Syndrome
Acute onset of Bmising, purpura, and mucosal bleeding Enlarged lymph nodes, liver, spleen Elevation of AST, ALT, PT, PTT, and fibrin degradation products Elevation of ferritin and triglycerides Fall in WBCC and platelet counts Fall in ESR Fall in fibrinogen, clotting factors Bone marrow examination shows active phagocytosis by macrophages and histiocytes Treatment: IV glucocorticoid, cyclosporine
• fIpre lZ-J This 8-year-old boy had onset of systemic JIA at the age of 4 years. There is symmetrical large and small joint arthritis. Note the axillary lymphadenopathy and marked hepatosplenomegaly.
ALT, alanine aminotransferase; AST, aspanate aminotransferase: ESR, erythrocyte sedimentation rate; PT, prothrombin time; PIT, panial thromboplastin time; WHCC, white blood cell count.
296
C HAP T E R
12 SYSTEMIC ARTHRITIS perforin expression in children with systemic JIA. They reported reduced perforin expression on natural killer (NK) cells and some cytotoxic CDs+ T cells in patients who had active systemic JIA, compared with normal controls or children with other types of arthritis. Grom and coworkers86 reported that patient~ with MAS as a complication of systemic JIA had marked reduction of NK cell activity during the acute stage and after resolution of MAS. Those patients with low perforin expression had a history of previous episodes of MAS.86 Absolute decrease in NK cell numbers97 and function98 also characterized systemic JIA. It is suggested that these abnormalities might predispose the patient to the development of MAS.74
• Figure lZ-4 Bone marrow aspiration demonstrating phagocytosis of erythrocytes and platelets by a histiocyte in a child with macrophage activation syndrome.
Similar laboratory abnormalities have been reported in children with systemic JIA even in the absence of MAS. Subclinical coagulation abnormalities characterized by prolonged PT and PTT and increased fibrin degradation products, fibrinopeptide A, and factor VIII-related antigen, are common in systemic-onset JRA but not in polyarticular disease. 79.82 Active systemic disease is also associated with increased levels of fibrin o-dimey83 and soluble adhesion molecules. 84 These markers of fibrin degradation correlated with severe disease, fever, and leukocytosis, and serial levels fell with continuing clinical response to diseasemodifying agents. Acquired factor VIII inhibitor has been described in some children. 8s Likewise, increased serum levels of hepatic enzymes have been well recognized in children with systemic JIA. The etiology of this disorder is unknown, but it often follows an infection, particularly by members of the herpesvirus family, including Epstein-Barr virus.7 s.86 Early reports noted the onset of MAS after changes in medications, particularly the institution of treatment with a gold compound, nonsteroidal anti-inflammatory drug (NSAID), hydroxychloquine, or D-penicillamine,87-89 More recently, it was suggested that methotrexate 90 .91 and etanercept 92 might trigger the syndrome. Considering the wide array of pharmacologic agents that have been reported to be associated with MAS, it seems likely that these events were coincidental, occurring in a child who was susceptible to MAS and who required additional therapy for uncontrolled systemic JIA. The histopathologic picture of Kikuchi's disease has been reported occasionally in patients with MAS,93 Smith and coworkers94described the presence of microthrombi and endothelial proliferation in skin biopsy specimens from 10 patients with the disorder. They postulated that continuing damage to endothelial cells and the resulting vasculitis induced disseminated intravascular coagulation. Prieur and Stephan9:1 studied 44 published cases and suggested that activated macrophages release proteinases that trigger the plasminogen-plasmin cascade. MAS has also been described in children with rheumatic diseases who were recipients of autologous stem cell transplantation and subsequently experienced overwhelming viremia. Because of the similarities between MAS and familial HLH, in which perforin expression is decreased, Wulffraat and associates96 studied
MAS must be treated vigorously and rapidly because of the extreme morbidity and high fatality rate. 99 •1OO One approach has been to use intravenous methylprednisolone pulse therapy,74.78.81 although some patients have been glucocorticoid resistant and require other treatment. 99 ,lOO Mouy and associates lOl described the use of cyclosporine in five children with this disorder, all of whom had rapid resolution of their illness over the course of a few days. Ravelli and colleagues described the successful use of cyclosporine in a child with glucocorticoid-resistant MAS beginning 11 months after institution of methotrexate for severe systemic JIA.102
Hepatic Disease Hepatomegaly is less common than splenomegaly. Moderate to marked enlargement of the liver is often associated with only mild derangement of functional studies and relatively nonspecific histopathologic changes. 103 This type of liver disease is most evident at onset and usually diminishes with time. Chronic liver disease does not occur. Massive enlargement of the liver is usually accompanied by abdominal distention and pain. Progressive hepatomegaly is characteristic of secondary amyloidosis. Unexplained acute yellow atrophy has been recorded. 104 Occasionally, a fatty liver is associated with glucocorticoid administration. Hepatitis (transarninasemia) related to NSAID therapy may occur (see Chapter 5). A rare, and apparently benign, transient elevation of serum alkaline phosphatase has been observed in several children with JRA. lO s. 106 This abnormality presumably does not result from hepatotoxicity, although it may be confused with it. Reye's syndrome occasionally occurred in children treated with aspirin. 107 Intercurrent infection with varicella or influenza appears to precipitate this serious complication (see Chapter 5).
Amyloidosis Secondary amyloidosis occurs as a complication of chronic inflammatory conditions such as arthritis or infection and in diseases such as familial Mediterranean fever. 108.1119 Although no HLA associations have been confirmed in children who develop amyloidosis, llO a genetic marker for amyloid P component was identified. 111 Secondary amyloidosis as a complication of JRA is exceedingly rare in North America but has occurred in 5% to 7% of children with chronic arthritis in Europe. 1l2 · 113 Rectal submucosa is the most frequently recommended biopsy site; renal biopsy can be hazardous because of an
C HAP T E R
increased tendency toward bleeding. Radionuclide imaging u$ing radioiodinated autologous serum amyloid P is a nonirlvasive technique for diagnosis and monitoring of amyI6idosis. 114 ,115 Th~ major causes of death in 46 children with arthritis mpnitored by Bywaters at Taplow116.117 were renal failure (which in half of the children was associated with amyl~idosis) and infections. At the 15-year evaluation, excess mortality was identified in girls who had onset of arthrids early in life, After 15 years of arthritis, the frequency of amyloidosis had risen to 7.4% among children with arthritis in the United Kingdom. 113 It was rarely obsewed as early as 1 year after onset of the arthritis but coule! develop as late as 23 years after onset, most commonly in children with systemic disease. Spontaneous remissions occurred. Even in Europe, amyloidosis as a complication of chronic arthritis appears to be on the declipe, with a decreasing number of deaths. 118-12l However, a recent series from Turkey reported a 10% frequency .122
DIA$NOSIS AND DIFFERENTIAL DIAGNOSIS The diagnosis of systemic arthritis is clinical, and it is a diagnosis of exclusion. In its fully developed state, the cliniqal characteristics are quite diagnostic. In particular, the presence of a classic rash and quotidian fever indicate :the likelihood of this diagnosis. The ILAR criteria, emphasizing the importance of the fever to the diagnosis, ~equire that it be present for 2 weeks and documented to be quotidian for 72 hours.
Differential Diagnosis The differential diagnosis of suspected systemic JIA may be difficult, especially at onset or early in the course of the disease, when the child may have a high spiking fever and evidence of systemic inflammation but no arthdtis and no specific sign or symptom that allows a definitive diagnosis. 122 In such children, the possibilities of Olalignancy, inflammatory bowel disease, vasculitis such as polyarteritis nodosa, or other connective tissue dise4J.ses such as systemic lupus erythematosus should be conSidered (Table 12-5). Children with systemic-onset disease may be thought to have an acute infectious disease or septicemia. Infectious mononucleosis and other viral: illnesses may mimic the disease, but for the most part :the arthropathies that occur secondary to viral infection$ are transient. Documenting the presence of arthritis or a typical rash helps to establish the diagnosis. Laboratory tests are of little value in diagnosis. In many instances, the diagnosis is one of exclusion until a full co~plement of characteristic abnormalities has been observed, a process that can take several months or possibly years. Many rare entities, such as chronic inflammatory neurologic and articular syndrome (CINCA) and the episodic fever syndromes, need to be excluded (see Chapters 34 and 35). Fever in children with infectious diseases is of the septic type. It is more hectic, spikes less predictably, and usu~lly does not repetitively return to baseline each day,
I!.
12
SYSTEMIC ARTHRITIS
297
fABLE 12-5 Differential Diagnosis of Systemic JuvenIle Idiopathic Arthritis
Infection Bacterial endocarditis Acute rheumatic fever Cat scratch disease (Bartonella) Lyme disease (Borrelia burgdorferi) Brucellosis Many others Inflammatory bowel disease Malignancy Connective tissue diseases Systemic lupus erythematosus Dermatomyositis Vasculitis Polyarteritis Kawasaki disease Castleman's disease Familial Mediterranean fever Episodic fever syndromes Mevalonate kinase deficiency (hyperimmunoglobulin D syndrome) Periodic fever, aphthous stomatitis, pharyngitis, and adenitis (PFAPA) TNF receptor-associated periodic syndrome (TRAPS) Muckle-Wells syndrome Familial cold autoinflammatory syndrome Chronic infantile neurologic cutaneous and articular syndrome (CINCA)
as does the fever of JIA, and the child remains ill even during a relatively afebrile interval. A sustained or remittent fever is characteristic of acute rheumatic fever and should respond dramatically to salicylates. Although many children with systemic JIA have isolated pericarditis, a pericardial effusion with evidence of endocarditis such as a diastolic murmur suggests a diagnosis of rheumatic fever or bacterial endocarditis. Onset of rheumatic fever in the developed countries of the world typically occurs in children between the ages of 5 and 15 years (see Chapter 31). The arthritis is characteristically acute and painful, migratory, and asymmetrical, involving the peripheral joints without sequelae. The initial episode usually lasts no longer than 6 weeks but may persist for as long as 3 months. Evidence of a prior infection with /3-hemolytic group A streptococci is present; however, the antistreptolysin 0 titer may be chronically increased to a moderate degree in one third of children with JIA as a manifestation of inflammation rather than evidence of recent streptococcal infection. 123 In acute rheumatic fever, a two-tube rise or fall in antistreptolysin 0 titer should be documented.
Adult-Onset Stili's Disease Adult-onset Still's disease was first reported in 1942. 124 It may manifest in either sex as fever of unknown origin. Hallmarks of adult-onset Still's disease are similar to those of childhood-onset disease. 12'>-128 Bywaters l25 described 14 young women with characteristic features of Still's disease, including fever, rash, polyarthritis, and an elevated ESR. In a National Institutes of Health study, all of the patients were men, and half were actually experiencing an exacerbation of arthritis after a long remission of systemic
298
C HAP T E R
12
SYSTEMIC ARTHRITIS
disease that began in childhood. 126 If investigations for infectious, hematologic, or neoplastic causes are unrewarding, diagnostic suspicion often focuses on a connective tissue disease. Characteristic radiographic changes (e.g., pericapitate involvement) have been described. 129
II If W
'
TABLE 12--6 Radiographic Changes in Systemic Juvenil.· Idiopathic Arthritis'
Joint Space Narrowing location
PATHOLOGY The rash of ]RA is one of the most characteristic clinical hallmarks of the disease. It is characterized by minimal perivascular infiltration of mononuclear cells around capillaries and venules in the subdermal tissues. 43 A neutrophilic perivasculitis resembling that of the rash of rheumatic fever may accompany the more flagrant lesions. The serosal lining surfaces of the pleural, pericardial, and peritoneal cavities of the body may exhibit nonspecitk fibrous serositis that is characterized clinically by effusion and pain. Enlargement of the lymph nodes is related to a nonspecific follicular hyperplasia that in rare instances may closely resemble lymphoma. Hepatic abnormalities are characterized by a nonspecific collection of periportal inflammatory cells and hyperplasia of Kupffer's cells.
LABORATORY EXAMINATION Indicators of inflammation are usually striking in active systemic ]IA. The white blood cell count is often higher than 30,000 to 50,000 cells/mm3 (30 to 50 x 109/0, with a predominance of polymorphonuclear leukocytes. The platelet count is usually elevated, sometimes to more than 1,000,000/mm3 (1000 x 109/L), although rarely thrombocytopenia occurs early in the disease course. l30 Hemoglobin in the range of 7 to 10 g/dL (70 to 100 gIL) is usual, and occasionally it is even lower. Erythrocytes are characteristically hypochromic and normocytic. Erythroid aplasia has been reported 131 .132 The ESR is usually very high, except in patients with MAS, in whom it may be normal. C-reactive protein is elevated, and ferritin levels are often very high, reflecting the extent of inflammatory disease. Polyclonal elevation of immunoglobulin levels characterizes the immune response, but autoantibodies such as rheumatoid factors and antinuclear antibodies are uncommon. Complement levels are usually increased as part of the acute-phase response, and increased levels of complement activation products have been reported. 133 Synovial fluid analysis confirms the presence of an inflammatory arthritis, with cells in the range of 10,000 to 40,000/mm3 , predominantly polymorphonuclear leukocytes.
RADIOLOGIC EXAMINATION Radiographs demonstrate bone and soft tissue changes in a high proportion of children with systemic ]IA (Table 12-6). In addition, growth abnormalities may be marked, and generalized delay in bone age is a frequent observation. Juxta-articular osteoporosis indicates the effect of active arthritis. In a recent study of 30 children with
Overall Cervical Shoulders Elbows Wrists Hands Hips Knees Ankles Feet
Early
Late
30
39 16 6 2 3 3 8 3 1 0
6 3 7 1 1 2
Growth Abnormalities
Erosions
Early
Late
35
63
20 2 5 3 3 0 3 4
Early
Late
10
25
4
-
-
6 8 5 2 21 1 1 7
-
-
6 1 1
-
-
1 0 2 1 1 2
"Frequencies (%) of abnonnalities at each joint, given as a percentage of the number of radiographs of each joint at early (within first 2 years) or late radiographic examination. Joint space narrowing included decrease in joint space. ankylosis, and carpal collapse. From Oen K, Reed M, Malleson PN et al. Radiologic outcome and its relationship to functional disability in juvenile rheumatoid arthritis. .I Rheumatol 30: 832-840, 2003,
systemic ]RA, Oen and colleagues 134 found that radiographs performed within the first 2 years of disease revealed joint space narrowing in 30%, erosions in 35%, and growth abnormalities in 10%. In later radiographs (a median of 6.4 years after onset), joint space narrowing was demonstrated in 39%, erosions in 63%, and growth abnormalities in 25%. Changes in the cervical spine and hips were most common in late radiographs. Subluxation of any joint was very uncommon.
TREATMENT Approach to Management The child with systemic ]IA is often acutely ill and may require hospitalization for initial management. In addition to medications to minimize joint inflammation, attention must be directed to diagnosis and management of extra-articular manifestations of this disease. Careful assessment of pulmonary and cardiac status, the severity of anemia, and the possibility of MAS (reactive histiocytosis) are all part of the initial evaluation. The general principles of management outlined in Chapter 8 should be applied to the management of systemic ]IA. It is appropriate to use an NSAID as initial therapy, both to aid in control of the systemic inflammatory features (e.g., fever) and to modulate joint pain and inflammation. In general, therapy should be considered in two parts: therapy directed at controlling the systemic disease and that directed at control of arthritis.
Pharmacologic Therapy for Systemic Disease The systemic features seldom respond in a satisfactory manner to NSAIDs alone, and if the diagnosis is secure,
C HAP T E R
the early use of glucocorticoids is indicated. Intravenous methylprednisolone (30 mg/kg/day to a maximum of 1 g/ day on 1 to 3 consecutive days) is effective in controlling systemic aspects of the disease, but the effect may be short-lived, and oral prednisone (l to 2 mg/kg/day to a maximum of 60 mg/day in one or more doses) may be necessary.13S Systemic disease may require the use of cyclosporine or intravenous immunoglobulin (IVIG). Cyclosporine is important in treating the MAS that accompanies systemic disease.101.136 In several small, uncontrolled trials, IVIG was found to be useful for the systemic but not the articular aspects of the disease. 137- 140 A controlled, randomized trial of IVIG by the Pediatric Rheumatology Collaborative Study Group in children with systemic-onset diseases indicated that the drug had little benefit compared with placebo. 141 However, the results were considered inconclusive because of the small sample size. The place of IVIG therefore remains unclear, but it is appropriate to consider its use in the child with active systemic manifestations of the disease that have not responded to NSAIDs and glucocorticoids with or without cyclosporine. The usual dose is 2 g/kg/day on 1 to 3 consecutive days, followed by 2 g/kg given once a month for a period of at least 6 months.
Pharmacologic Therapy for Arthritis If arthritis does not respond to NSAIDs alone, intraarticular glucocorticoids (preferably triamcinolone hexacetonide), or the introduction of methotrexate (0.35 to 0.65 mg/kg/week), or both, is indicated. Oral methotrexate is usually prescribed initially, but at doses greater than 15 mg/week; or, if the response has been inadequate, the drug should be administered in the same dose subcutaneously. Patients with no response to oral methotrexate may have an excellent response when the drug is given subcutaneously.142 Although methotrexate is an effective agent in many children, the response rate is not as high as it is for oligoarthritis or polyarthritis. In a placebo-controlled study, there was no significant difference in systemic feature score, ESR, or C-reactive protein between those children who received oral methotrexate and those who received placebo. 143 If the number of joints with resistant inflammation is small, intra-articular triamcinolone hexacetonide is very effective. Even in those children with active arthritis in many joints, this approach should be considered.
Cydosporlne In an open, prospective trial, Gerloni and colleagues 144 evaluated the efficacy of cyclosporine in 34 children with systemic JIA. The most dramatic response was rapid disappearance of fever in 52% of the patients. The effects on joint inflammation were less impressive, although a reduction of at least 50% in the number of actively int1amed joints was observed in 11 of 29 patients after 3 months of therapy. A rise in hemoglobin level, a fall in ESR, and a statistically significant decrease in prednisone requirement were also observed. In this study, toxicity to cyclosporine resulted in its discontinuation in one quarter of those
12
SYSTEMIC ARTHRITIS
299
treated. A further 50% of patients discontinued the drug because of disease flare or inefficacy. The role of cyclosporine in the management of systemic JIA is still not entirely clear, particularly with regard to the joint disease.
Biologic Agents Intravenous Immunoglobulin IVIG is used to treat systemic]lA, particularly the systemic features of the disease, although the evidence supporting its use is limited. Silverman and associates l38 noted good short-term effects on the systemic manifestations and less consistent improvement in the joint disease. In a follow-up study,139 27 children with systemic JIA were treated with monthly IVIG for 4 to 54 months. By 6 months, 20 patients had a significant decrease in the number of days with fever or a marked decrease (greater than 50%) in the number of actively inflamed joints. At a mean of 37.6 months' followup, 11 of the initial responders were in remission, 3 had improved but had chronically active disease, and 6 had no response. Prieur and colleagues 140 reported that, among 10 patients with systemic JIA treated with IVIG, rash improved in 4, fever in 6, and lymphadenopathy and splenomegaly in 3. Joint disease was not consistently affected in the short or long term, and the progression of systemic features (pericarditis, polyarthritis) or side effects of the IVIG (purpura, urticaria) required further treatment with prednisone. Eleven patients received at least 17 and as many as 52 monthly infusions. The role of IVIG appears to be restricted predominantly to management of systemic aspects of the disease.
Anti-TNF Agents In a multicenter clinical trial of the TNF-a receptor p75 fusion protein (etanercept), there were 22 patients with systemic arthritis who were refractory to or intolerant of methotrexate. 14S In the double-blind phase of this trial, 7 of 8 patients (88%) receiving placebo flared compared with 4 of 9 (44%) receiving etanercept. However, the frequency of flare was much lower in children with polyarticular JRA (21%). In the 2-year follow-up study,146 30% improvement in the core set criteria occurred in 59%, 50% improvement in 53%, and 70% improvement in 47%. Each component of the JRA core set criteria worsened in the arm that received placebo and remained stable or improved in the arm that continued on etanercept. A review of the data concerning etanercept and systemic JIA was published by Horneff and colleagues. 147 Studies of the efficacy of monoclonal antibody to TNF-a Cinfliximab) in systemic JIA have not been reported. Anecdotally, however, infliximab may be effective in some children with systemic JIA, even if they have failed etanercept. There is a clinical impression, however, that neither etanercept nor infliximab is as effective in children with systemic JIA as in those with polyarthritis.
Antl-IL6 Receptor Experience with anti IL-6 receptor in systemic JIA is limited to two studies.148.149 Yokota and coworkers 148 treated
300
C HAP T E R
12
SYSTEMIC ARTHRITIS
11 children with methotrexate-resistant systemic JIA with the anti-IL-6 receptor antibody (MRA) in doses of 2 to 8 mglkg over several months. There was an abrupt reduction in disease activity in 10 of the 11 patients. The core set 70 response with 33% with 2 mglkg, 60% with 4 mglkg, and 100% with 8 mglkg. Wilkinson and collegues J49 reported the results of a single dose of MRA in nine patients with systemic ]lA. They, too, noted an abrupt response in clinical and laboratory parameters in seven patients.
Thalidomide Two recent reports support the use of thalidomide in children with treatment resistant systemic JIA. After an initial report 150 of favorable response in two children with treatment-resistant systemic JIA, a multicenter retrospective review151 of experience in 13 such children provided further evidence that this drug may have a role. All received thalidomide in a dose of 3 to 5 mglkg/day and were evaluated at 6, 12, and 24 months by active joint count, dose of prednisone required, and laboratory values (white blood cell count, hemoglobin, and ESR). Improvement in active joint count, reqUired prednisone dose, and inflammatory indices were seen by 6 months, and in the 5 patients who were monitored for 24 months this appeared to be sustained.
Autologous Stem Cell Transplantation Most children with rheumatic diseases who have received autologous stem cell transplantation (ASCT) have had systemic JIA. Kuis, Wulffraat, and their colleagues in Utrecht have had the greatest experience with this approach. 152 The Utrecht protocol uses bone marrow-derived stem cells prepared by CD3 depletion and enrichment for CD34+ stem cells. The patient is conditioned with antithymocyte globulin (5 mg/kglday on days -9 to -6), cyclophosphamide (50 mglkglday on days -5 to -2), and, in the earlier version of this protocol, lowdose (4 Gy) total body irradiation. The results of such therapy are promising but are not without significant mortality, and the recommended protocol has been modified. Two of 25 patients 152 had fatal MAS, and a third patient has died of this complication. 153 Because of MAS, ASCT is now considered to be contraindicated in patients in whom the fever cannot be controlled by corticosteroids. Total body irradiation has not been used in the most recently transplanted patients, with no evident loss of efficacy. Overall, 17 of 25 patients were in medicationfree remission 8 to 60 months after transplantation. Many of the children were treated before the availability of anti-TNF agents, and it is currently recommended that ASCT be considered only in those children who have failed or have toxicity to methotrexate and to etanercept or infliximab.
COURSE OF THE DISEASE AND PROGNOSIS The acute manifestations of the systemic JIA are variable in duration and last from weeks to months. Systemic features such as fever, rash, and pericarditis tend to subside
during the initial months to years of the disease (2 to 5 years) but may recur in conjunction with exacerbations of the arthritis. About half of the children with systemic JIA eventually recover almost completely, often after a pattern of oligoarticular disease for a variable period. The other half continue to show progressive involvement of more and more joints and moderate-to-severe functional disability in some studies l54 ,155 but not in others. 156 The eventual functional outcome in these children depends more on the number of joints involved and on continuing activity than on the nature of the systemic disease. 157 The persistence of systemic symptoms without arthritis is unusual and is seldom a cause of permanent disability. However, in the Cincinnati series, 48% of children with systemic JRA still had active arthritis 10 years later. 158 In a more recent study, Oen and colleagues l59 evaluated outcome in 48 children with systemic JRA. The probability of remission 10 years after onset was only 37%. Male sex correlated with worse disability in children with systemic disease. 160 There remains a great need for more effective therapy in children with this disease. In early studies of JRA, the death rate was 2% to 4%.161,162 The disease-associated death rate is now perhaps less than 1% in Europe and less than 0.5% in North America. The majority of the deaths associated with JRA in Europe were related to amyloidosis; in the United States, deaths occurred in children with systemic arthritis and, in many cases, were related to infections associated with glucocorticoid therapy. MAS remains a serious and potentially fatal threat.
REFERENCES 1. Still GF: On a form of chronic joint disease in children. Med ChiI' Trans 80: 47, 1897. 2. Petty RE, Southwood TR. Manners P, et al: International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: Second Revision, Edmonton 2001. j Rheumatol 31: 390-392, 2004, 3. Cassidy jT, Levinson j, Bass jC, et al: A study of classification criteria for a diagnosis of juvenile rheumatoid arthritis. Arthritis Rheum 29: 274-281, 1986. 4. European League Against Rheumatism: Bulletin No.4: Nomenclature and Classification of Arthritis in Children. Basel, National Zeitung AG, 1977. 5. Moe N, Rygg M: Epidemiology of juvenile chronic arthritis in northern Norway: a ten-year retrospective study. Clin Exp Rheumatol 16: 99-101. 1998. 6, Sullivan DB, Cassidy jT, Pelty RE: Pathogenic implications of age of onset in juvenile rheumatoid arthritis. Arthritis Rheum 18: 251, 1975. 7, Lindsley CB: Seasonal variation in systemic onset juvenile rheumatoid arthritis. Arthritis Rheum 30: 838-839, 1987. 8. Feldman B, Birdi N, Boone jE. et al: Seasonal onset of systemic-onset juvenile rheumatoid arthritis, j Pediatr 129: 513-519, 1996. 9. Uziel Y, Pomeranz A, Brik R, et al: Seasonal variation in systemic onset juvenile rheumatoid arthritis in Israel. j Rheumatol 26: 1187-1189, 1999. 10. Ogilvie EM, Fife MS, Thompson SO, et al: The -174 allele of the interleukin6 gene confers susceptibility to systemic arthritis in children. Arthritis Rheum 48: 3202-3206. 2003. 11. De Benedetti F. Martini A: Is systemic juvenile rheumatoid arthritis an interleukin 6 mediated disease? j Rheumatol 25: 203-207, 1998. 12. De Benedetti F, Massa M, Robbioni P, et al: Correlation of serum interleukin6 levels with joint involvement and thrombocyrosis in systemic juvenile rheumatoid arthritis. Arthritis Rheum 34: 1158--1163. 1991. 13. Lepore L, Pennesi M, Saletta S, et al: Study of IL-2, 1L-6, TNF alpha, IFN garruna and beta in the serum and synoviai fluid of patients with juvenile chronic arthritis. Clin Exp Rheumatol 12: 561-565, 1994. 14. Rooney M, David j, Symons j, et al: Inflammatory cytokine responses in juvenile chronic arthritis. Br j Rheumatol 34: 454-460, 1995. 15. Prieur AM, Kaufmann MT, Griscelli C, et al: Specific interleukin-l inhihitor in serum and urine of children with systemic juvenile chronic arthritis. Lancet 2: 1240-1242, 1987.
C HAP T E R 16. Mangge H, Kenzian H, Gallistl S, et al: Serum cytokines in juvenile rheumatoid arthritis: correlation with conventional inflammation parameters and clinical subtypes. Arthritis Rheum 38: 211-220, 1995. 17. De Benedetti F, Alonzi T, Moretta A, et al: Interleukin 6 causes growth impairment in transgenic mice through a decrease in insulin-like growth factor-I: a model for stunted growth in children with chronic inflammation. ] Clin Invest 99: 643-650, 1997. 18. Fishman 0, Faulds G, Jeffery R, et al: The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. ] Clin Invest 102: 1369-1376, 1998. 19. De Benedeni F, Massa M, Pignani P, et al: Serum soluble interleukin 6 (IL-6) receptor and IL-6/soluble IL-6 receptor complex in systemic juvenile rheumatoid arthritis. ] Clin Invest 93: 2114-2119, 1994. 20. Pignatti P, Ciapponi L, Galle P, et al: High circulating levels of biologically inactive IL-6/SIL-6 receptor complexes in systemic juvenile idiopathic arthritis: evidence for serum factors interfering with the binding to gp130. Clin Exp lmmunol 131: 355-363, 2003. 21. De Benedetti F, Ravelli A, Martini A: Cytokines in juvenile rheumatoid arthritis. Curr Opin Rheumatol 9: 428--433, 1997. 22. Pignatti P, Vivarelli M Meazza, et al: Abnormal regulation of interleukin 6 in systemic juvenile idiopathic arthritis. ] Rheumatol 28: 1670-1676, 2001. 23. De Benedetti F, Pignani P, Massa M, et al: Circulating levels of interleukin 1 beta and of interleukin 1 receptor antagonist in systemic juvenile chronic arthritis. Clin Exp Rheumatol 13: 779-784, 1995. 24. Keul R, Heinrich PC, Muller-Newen G, et al: A possible role for soluble IL-6 receptor in the pathogenesis of systemic onset juvenile chronic arthritis. Cytokine 10: 729-734, 1998. 25. DatI" Y, Seki N, Kamizono S, et al: Identification of a genetic risk factor for systemic juvenile rheumatoid arthritis in the 5'-flanking region of the TNF alpha gene and HLA genes. Arthritis Rheum 42: 2577-2582, 1999. 26. Muzaffer MA, Dayer ]-M. Feldman BM, et al: Differences in the profiles of circulating levels of soluble tumor necrosis factor receptors and interleukin 1 receptor antagonist reflect the heterogeneity of the subgroups of juvenile rheumatoid arthritis.] Rheumatol 29: 1071-1078, 2002. 27. Silverman ED, Laxer RM, Nelson DL, et al: Soluble interleukin-2 receptor in juvenile rheumatoid arthritis. ] Rheumatol 18: 139&-1402, 1991. 28. tipnick RN, Sfikakis PP, Klipple GL, et al: Elevated soluble CDS antigen and soluble interleukin-2 receptors in the sera of patients with juvenile rheumatoid arthritis. Clin Immunol hnmunopathol 68: 64-67, 1993. 29. Madson KL, Moore TL, Lawrence ]M III, et al: Cytokine levels in serum and ~ynovial fluid of patients with juvenile rheumatoid arthritis. ] Rheumatol 21: 2359-2363, 1994. 30. De Benedetti F. Massa M, Pignani P, et al: Elevated circulating interleukin-7 levels in patients with systemic juvenile rheumatoid arthritis.] Rheumatol 22: 1581-1585, 1995. 31. De Benedetti F, Pignatti P, Bernasconi S, et al: Interleukin 8 and monocyte chemoanractant protein-l in patients with juvenile rheumatoid arthritis: relation to onset types, disease activity, and synovial fluid leuko~ytes. ] Rheumatol 26: 425-431, 1999. 32. Yilmaz M, Kenderli SG, Altintas 0, et al: Cytokine levels in semm of patients with juvenile rheumatoid arthritis. Clin Rheumatol 20: 30-35, 2001. 33. Alarcon-Riquelme ME, Vazquez-Mellado ], Gomez-Cordillo M, et al: Immunoregulatory defects in juvenile rheumatoid arthritis: comparison between patients with the systemic or polyarticular forms. ] Rheumatol 15: 1547-1550, 1988. 34. Wedderburn LR, Maini MK, Patel A, et al: Molecular fmgerprinting reveals non-overlapping T cell oligodonality between an inflamed site and peripheral blood. Int Immunol 11: 535-543, 1999. 35. Oen K: Defects in pokeweed mitogen (PWM) induced immunoglobulin (Ig) synthesis by lymphocytes of patients with juvenile rheumatoid arthritis. ] Rheumatol 12: 72&-734, 1985. 36. Doublog ]H, Chanoppadhyay C, Forre 0, et al: Con A-induced suppressor cell activity and T-Iymphocyte subpopulations in peripheral blood lymphocytes of patients with rheumatoid arthritis and juvenile rheumatoid arthritis. SC'dnd] [mmunol 13: 367-373, 1981. 37. Tsokos GC, Mavridis A, Inghirami G, et al: Cellular immunity in patients with systemic juvenile rheumatoid arthritis. Clin Immunol lnununopathol 42: 86-92, 1987. 38. Tsokos GC, Inghirami G, Pillemer SR, et al: Immunoregulatory aberrations in patients with polyarticular juvenile rheumatoid arthritis. Clin Immunol Immunopathol 47: 62-74, 1988. 39. Nepom B: The immunogenetics of juvenile rheumatoid arthritis. Rheum Dis Clin North Am 17: 825-B42, 1991. 40, Paul C, Yao Z, Nevinny-Stickel C, et al: Immunogenetics of juvenile chronic arthritis. I: HLA interaction between A2, DR5/8-DR/DQ, and DPBl"0201 is a general feature of all subsets of early onset pauciarticular juvenile chronic arthritis. II: DPBl polymorphism plays a role In systemic juvenile chronic arthritis. Tissue Antigens 45: 280-283, 1995, 41. Desaymard C, Kaplan C, Fournier C, et al: Major histocompatibility complex markers and disease heterogeneity in one hundred eight patients with systemic onset juvenile chronic arthritis. Rev Rhum Engl Ed 63: 9-16. 1996.
12 SYSTEMIC ARTHRITIS
301
42. Schneider R, Lang BA, Reilly B] et al. Prognostic indicators of joint destruction in systemic-onset juvenile rheumatoid arthritis. ] Pediatr 120: 200-205, 1992. 43. Isdale IC, Bywaters EGL: The rash of rheumatoid arthritis and Still's disease. Q] Med 25: 377, 1956. 44. Ansell BA, Rudge S, Schaller ]G: A Colour Atlas of Paediatric Rheumatology. Aylesbury, UK, Wolfe. 1991. 45. Schaller J, Wedgwood RJ: Pmritus associated with the rash of juvenile rheumatoid arthritis. Pediatrics 45: 296-298, 1970. 46. Prendiville ]S, Tucker LB, Cabral DA, Crawford R1: A pruritic linear urticarial rash, fever and systemic inflammatory disase in five adolescents: adult-onset Still disease or systemic juvenile idiopathic arthritis sine arthritis? Pediatr Dermatol 21: 580-588, 2004. 47. Alukal MK, Costello PB, Green FA: Cardiac tamponade in systemic juvenile rheumatoid arthritis requiring emergency pericardiectomy. ] Rheumatol 11: 222-225, 1984. 48. Goldenberg], Ferraz MB, Pessoa AP, et al: Symptomatic cardiac involvement in juvenile rheumatoid arthritis. Int] Cardiol 34: 57-62, 1992. 49. Lietman PS, Bywaters EGL: Pericarditis in juvenile rheumatoid arthritis. Pediatrics 32: 855, 1963. 50. Bernstein B, Takahashi M, Hanson V: Cardiac involvement in juvenile rheumatoid arthritis. J Pediatr 85: 313-317, 1974. 51. Scharf], Levy], Benderly A, et al: Pericardial tamponade in juvenile rheumatoid arthritis. Arthritis Rheum 19: 760-762, 1976. 52. Brewer E ]r: ]uvenile rheumatoid arthritis-cardiac involvement. Arthritis Rheum 20: 231-236, 1977. 53. Bernstein B: Pericarditis in juvenile rheumatoid arthritis. Arthritis Rheum Suppl 20: 241, 1977. 54. Svantesson H, Bjorkhem G, Elborgh R: Cardiac involvement in juvenile rheumatoid arthritis: a follow-up study. Acta Paediatr Scand 72: 345-350, 1983. 55. Majeed HA, Kvasnicka]: Juvenile rheumatoid arthritis with cardiac tamponade. Ann Rheum Dis 37: 273-276, 1978. 56. Yancey CL, Doughty RA, Cohlan BA, et al: Pericarditis and cardiac tamponade in juvenile rheumatoid arthritis. Pediatrics 68: 369-373, 1981. 57. Goldenberg], Pessoa AP, Roizenblan S, et al: Cardiac tamponade in juvenile chronic arthritis: report of two cases and review of publications. Ann Rheum Dis 49: 549-553, 1990. 58. Bauer-Vinassac D, Chapsal], Duboc 0, et al: (Aortic insufficiency and rapidly developing constrictive pericarditis in rheumatoid polyarthritis of juvenile onset. Apropos of a case and review of the literature]. Ann Med Interne (Paris) 138: 141-142, 1987. 59. Miller]] III, French JW: Myocarditis in juvenile rheumatoid arthritis. Am] Dis Child 131: 205-209, 1977. 60. Oguz 0, Ocal B, Ertan U, et al: Left ventricular diastolic functions in juvenile rheumatoid arthritis. Pediatr Cardiol 21: 374-377, 2000. 61. Kramer PH, Imboden]B ]r, Waldman FM, et al: Severe aortic insufficiency in juvenile chronic arthritis. Am] Med 74: 10Bs-1091 , 1983. 62. Heyd], Glaser]: Early occurrence of aortic valve regurgitation in a youth with systemic-onset juvenile rheumatoid arthritis. Am] Med 89: 123-124, 1990. 63. Brinkman GL, Chaikof L: Rheumatoid lung disease. Am Rev Respir Dis 80: 732, 1959. 64. Jordan ]0, Snyder CH: Rheumatoid disease of the lung and cor pulmonale: observations in a child. Am] Dis Child 108: 174, 1964. 65. Athreya BH, Doughty RA, Bookspan M, et al: Pulmonary manifestations of juvenile rheumatoid arthritis: a report of eight cases and review. Clin Chest Med 1: 361-374, 1980. 66. Lovell 0, Lindsley C, Langston C: Lymphoid interstitial pneumonia in juvenile rheumatoid arthritiS. ] Pediatr 105: 947-950, 1984. 67. Wagener JS, Taussig LM, DeBenedeni C, et al: Pulmonary function in juvenile rheumatoid arthritis. ] Pediatr 99: 10&-110, 1981. 68, Uzlei Y, Hen B, Cordoba M, et al: Lymphocytic interstitial pneumonitis preceding polyarticular juvenile rheumatoid arthritis. Clin Exp Rheumatol 16: 617-619, 1998. 69. Noyes BE, Albers GM, deMello DE, et al: Early onset of pulmonary parenchymal disease associated with juvenile rheumatoid arthritis. Pediatr Pulmonol 24: 444-446, 1997. 70. Braidy]F, Poulson]M: Diaphragmatic weakness and myositis associated with systemic juvenile rheumatoid arthritis. Can Med Assoc] 130: 47--49, \984, 71. Knook LM, De Kleer 1M, Van der Ent CK, et al: Lung function abnormalities and respiratory muscle weakness in children with juvenile chronic arthritis. Eur Respir ] 14: 529-533, 1999. 72. Padeh S, Laxer RM, Silver MM, et al: Primary pulmonary hypertension in a patient with systemic-onset juvenile arthritis. Arthritis Rheum 34: 1575-1579, 1991. 73. Schultz R, Mattila], Gappa M, Verronen P: Development of progressive pulmonary interstitial and intra-alvaolar cholesterol granulomas (PICG) associated with therapy resistant chronic systemic juvenile arthritis (ClAl. Pediatr Pulmonol 32: 397--402, 2001. 74. Grom AA: Natural killer cell dysfunction: a common pathway in systemiconset juvenile rheumatoid arthritis, macrophage activation syndrome, and hemophagocytic Iymphohistiocytosis? Arthritis Rheum 50: 689-69R, 2004. 75. Sawhney S, Woo P, Murray K]: Macrophage activation syndrome: a potentially fatal complication of rheumatic disorders. Arch Dis Child 85: 421-426, 2001.
302
C HAP T E R
12
SYSTEMIC ARTHRITIS
76. Stephan JL, Kone-Paut I, Galambrun C, et al: Reac1ive haemophagocytic syndrome in children with inflammatory disorders: a retrospective srudy of 24 patients. Rheumatology (Oxford) 40: 1285-1292, 2001 77. Aellen P, Raccaud 0, Waldburger M, et al: Still's disease in adults with disseminated intravascular coagulation. Schweiz Rundsch Med Prax 80: 376-378, 1991. 78. Silverman ED, MiIler JJ III, Bernstein B, et al: Consumption coagulopathy associated with systemic juvenile rheumatoid arthritis. J Pediatr 103: 872-876, 1983. 79. Scott JP, Gerber P, Maryjowski MC, et al: Evidence for intravascular coagulation in systemic onset, but not polyarticular, juvenile rheumatoid arthritis. Arthritis Rheum 28: 256-261, 1985. 80. De Vere-Tyndall A, Macauley D, Ansell BM: Disseminated intravascular coagulation complicating systemic juvenile chronic arthritis ("StiIl's disease"). Clin Rheumatol 2: 415-418, 1983, 81. Hadchouel M, Prieur AM, Griscelli C: Acute hemorrhagic, hepatic, and neurologic manifestations in juvenile rheumatoid arthritis: possible relationship to drugs or infection. ) Pediatr 106: 561-566, 1985. 82. Gallistl S, Mangge H, Neuwirth G, et al: Activation of the haemostatic system in children with juvenile rheumatoid arthritis correlates with disease activity. Thromb Res 92: 267-272, 1998. 83. Bloom BJ, Tucker LB, Miller LC, et al: Fibrin D-dimer as a marker of disease activity in systemic onset juvenile rheumatoid arthritis. J Rheumatol 25: 1620-1625, 1998. 84. Bloom BJ, Miller LC, Tucker LB, et al: Soluble adhesion molecules in juvenile rheumatoid arthritis. J Rheumatol 26: 2044-2048, 1999. 85. De Inocencio), Lovell D), Gabriel CA: AcqUired factor YIIJ inhibitor in juvenile rheumatoid arthritis. Pediatrics 94: 550-553, 1994. 86. Grom AA, Villanueva), Lee S, et al: Natural kiIler cell dysfunction in patients with systemic-onset juvenile rheumatoid arthritis and macrophage activation syndrome . .1 Pediatr 142: 292-296, 2003. 87, Jacobs )C, Gorin L), Hanissian AS, et al: Consumption coagulopathy after gold therapy for JRA. J Pediatr 105: 674-675, 1984. 88. Manners P), Ansell BM: Slow-acting antirheumatic drug use in systemic onset juvenile chronic arthritis. Pediatrics 77: 99-103, 1986. 89. Barash J, Cooper M, Tauber Z: Hepatic, cutaneous and hematologic manifestations in juvenile chronic arthritis. Clin Exp Rheumatol 9: 541-543, 1991. 90. Havelli A, Caria MC, Buratti S, et al: Methotrexate as a possible trigger of macrophage activation syndrome in systemic juvenile idiopathic arthritis, J Rheumatol 28: 865-867, 2001. 91. Eraso R, Gedalia A, Espinoza LR: Methotrexate as a possible trigger of macrophage activation syndrome in systemic juvenile idiopathic arthritis. J Rheumatol 29: 1104-1105, 2002. 92. Ramanan AV, Schneider R: Macrophage activation syndrome follOWing initiation of etanercept in a child with systemic onset juvenile rheumatoid arthritis. ) Rheumatol 30: 401-403, 2003. 93. Ramanan AV, Wynn RF, Kelsey A, Baildam EM: Systemic juvenile idiopathic al1hritis, Kikuchi's disease and haemophagocytic Iymphohistiocytosis: is there a link? Case report and literature review. Rheumatol (Ow 42: 596-598. 2003. 94. Smith K), Skelton HG, Yeager J, et al: Cutaneous histopathologic, immunohistochemical, and clinical manifestations in patients with hemophagocytic syndrome. Military Medical Consortium for Applied Retroviral Research (MMCARR). Arch Dermatol 128: 193-200, 1992. 95. Prieur AM, Stephan .IL: Macrophage activation syndrome in rheumatic diseases in children. Rev Rhum Ed Fr 61: 447-451, 1994. 96. Wulffraat NM, Rijkers GT, Elst E, et al: Reduced perforin expression in systemic juvenile idiopathic arthritis is restored by autologous stem-cell transplantation. Rheumatology (OxO 42: 375-379, 2003. 97. Wouters CH, Ceuppens ]L, Stevens ERA: Different circulating lymphocyte profiles in patients with different substypes of juvenile idiopathic arthritis. Clin Exp Rheumatol 20: 239-248. 2002. 98. ViIlaneuva )L, Yang L, Graham BT, et al: The extent of NK dysfunction in patients with systemic onset juvenile rheumatoid arthritis and macrophage activation syndrome [abstract), Clin Exp Rheumatol 21: 546, 2003. 99. Quesnel B, Catteau B, Axnar V, et al: Successful treatment of juvenile rheumatoid almritis associated haemophagocytic syndrome by cyclosporin A with transient exacerbation by conventional-dose G-CSF. Br .I Haematol 97: 508--510, 1997. 100. Henter )1, Arico M, Egeler RM, et al: HLH-94: a treatment protocol for hemophagocytic Iymphohistiocytosis. HLH study Group of the Histiocyte Society. Med Pediatr Oncol 28: 342-347, 1997. 101. Mouy R, StephanJL, PiIlet P, et al: Efficacy of cyclosporine A in the treatment of macrophage activation syndrome in juvenile arthritis: report of five cases, .I Pediatr 129: 750-754, 1996. 102. Ravelli A, De Benederti F, Viola S, Martini A: Macrophage activation syndrome in systemic juvenile rheumatoid arthritis successfully treated with cyclosporine. J Pediatr 128: 275-278, 1996. 103, Schaller ), Beckwith B, Wedgwood R): Hepatic involvement in juvenile rheumatoid arthritis, ) Pediatr 77: 203-210, 1970, 104. Boone JE: Hepatic disease and mortality in juvenile rheumatoid arthritis. Arthritis Rheum Suppl 20: 257, 1977. 105. Jacobs JC:)RA and hyperphosphatemia.) Pediatr 107: 828-829, 1985. 106. Lockitch G, Pudek MR, Halstead AC: Isolated elevation of serum alkaline phosphatase. J Pediatr 105: 773-775, 1984.
107. Rennebohm RM, Heubi )E, Daugherty CC, et al: Reye syndrome in children receiving salicylate therapy for connective tissue disease. ) Pediatr 107: 877-880, 1985. 108. Woo P: Amyloidosis in pediatric rheumatic diseases. ,r Rheumatol Suppl 35: 10-16, 1992. 109. Gwyther M, Schwarz H, Howard A, et al: C-reactive protein in juvenile chronic arthritis: an indicator of disease activity and possibly amyloidosis. Ann Rheum Dis 41: 259-262, 1982. 110. Burman S1, Hall P), Bedford PA, et al: HLA antigen frequencies among patients with juvenile chronic arthritis and amyloidosis: a brief report. Clin Exp Rheumatol 4: 261-263, 1986. 111. Woo P, O'Brien J, Robson M, et al: A genetic marker for systemic amyloidosis in juvenile arthritis. Lancet 2: 767-769, 1987. 112. Calabro )): Amyloidosis and juvenile "heumatoid arthritis. J Pediatr 75: 521, 1969 113. Schnitzer TJ, Ansell BM: Amyloidosis in juvenile chronic polyarthritis. Arthritis Rheum 20: 245-252, 1977. 114. Hawkins PN, Myers MJ, Lavender JP, et al: Diagnostic radionuclide imaging of amyloid: biological targeting by circulating human serum amyloid P component. Lancet 1: 1413-1418, 1988. 115. Hawkins PN, Richardson S, Vigushin DM, et al: Serum amyloid P component scintigraphy and turnover studies for diagnosis and quantitative moniloring of AA amyloidosis in juvenile rheumatoid arthritis. Arthritis Rheum .36: 842-851, 1993. 116. Bywaters EGL: Deaths in juvenile chronic polyarthritis, Arthritis Rheum Suppl 20: 256, 1977. 117. Arden GP: Sepsis in juvenile chronic polyarthritis. In Arden GP, Ansell BM (eds): Surgical Management of Juvenile Chronic Polyarthritis. London, Academic Press, 1978, p 225. 118. Savolainen HA, Isomaki HA: Decrease in the number of deaths from secondary amyloidosis in patients with juvenile rheumatoid arthritis. J Rheumatol 20: 1201-1203, 1993. 119. Ansell BM: Chlorambucil therapy in juvenile chronic arthritis (juvenile idiopathic arthritis).) Rheumatol 26: 765-766, 1999. 120. David J, Vouyiouka 0, Ansell BM, et al: Amyloidosis in juvenile chronic arthritis: a morbidity and mortality study. Clin Exp Rheumatol 11: 85-90,1993. 121. Ozdogan H, Kasapcopur 0, Dede H, et al: Juvenile chronic arthritis in a Turkish population. Clin Exp Rheumatol 9: 431-435, 1991. 122. Miller LC, Sisson BA, Tucker LB, et al: Prolonged fevers of unknown origin in children: patterns of presentation and outcome. ) Pediatr 129: 419-423, 1996. 123, Sievers K, Ahvonen P: Serological patterns in juvenile rheumatoid arthritis. Rheumatism 19: 88, 1963. 124. Pindborg S: Et tilfaelde af Still's sygdom. Ugeskr Laeger 104: 1417, 1942. 125. Bywaters EG: Still's disease in the adult. Ann Rheum Dis 30: 121-133, 1971. 126. Aptekar RG, Decker )L, Bujak .IS, et al: Adult onset juvenile rheumatoid arthritis. Arthritis Rheum 16: 715-718, 1973, 127. Cush J), Medsger TA Jr, Christy WC, el al: Adult-onset Still's disease: clinical course and outcome. Arthritis Rheum 30: 186-194, 1987. 128. Roberts-Thomson PJ, Southwoocl TR, Moore BW, et al: Adult onset Still's disease or coxsackie polyarthritis? Aust N Z) Med 16: 509-511, 1986. 129. Bjorkengren AG, Pathria MN, Sartoris D), et al: Carpal alterations in adullonset Still disease, juvenile chronic arthritis, and adult-onset rheumatoid arthritis: comparative srudy. Radiology 165: 545-548, 1987. 130. Sherry DD, Kredich OW: Transient thrombocytopenia in systemic onset juvenile rheumatoid arthritis. Pediatrics 76: 600-603, 1985. 131. Rubin RN, Walker BK, Ballas SK, et al: Erythroid aplasia in juvenile rheumatoid arthritis. Am J Dis Child 132: 760-762, 1978. 132. Ewer AK, Darbyshire PJ, Southwood TR: Systemic-onset juvenile chronic arthritis and bone marrow hypoplasia. Br J Rheumatol 32: 78-80, 1993. 133. Miller II Ill, Hsu YP, Moss R, et al: The immunologic and clinical associations of the split products of C3 in plasma in juvenile rheumatoid arthritis, Arthritis Rheum 22: 502-507, 1979. 134. Oen K, Reed M, Malleson PN, et al: Radiologic outcome and its relationship to functional disbility in juvenile rheumatoid arthritis. 1 Rheumatol 30: 832-840, 2003. 135. Adebajo AO, Hall MA: The use of intravenous pulsed methylprednisolone in the treatment of systemic-onset juvenile chronic arthritis. Br J Rheumatol 37: 1240-1242, 1998. 136. Grom AA, Passo M: Macrophage activation syndrome in systemic juvenile rheumatoid arthritis. ) Pediatr 129: 630-632, 1996. 137. Groothoff ,rW, van Leeuwen EF: High dose intravenous gammaglobulin in chronic systemic juvenile arthritis. Br Med) (Clin Res Ed) 296: 1362-1363, 1988. 138. Silverman ED, Laxer RM, Greenwald M, et al: Intravenous gamnul globulin therapy in systemic juvenile rheumatoid arthritis. Arthritis Rheum 33: 1015-1022, 1990. 139. Llziel Y, Laxer RM, Schneider R, et al: Intravenous immunoglobulin therapy in systemic onset juvenile rheumatoid arthritis: a followup study.) Rheumatol 23: 910-918, 1996. 140. Prieur AM, AcUeff A, Debre M, et al: High dose immunoglobulin therapy in severe juvenile chronic arthritis: long-term follow-up in 16 patients. Clin Exp Rheumatol 8: 603-608, 1990. 141. Silverman ED, Cawkwell GD, Lovell DJ, et ai: Intravenous immunoglobulin in the treatment of systemic juvenile rheumatoid arthritis: a randomized placebo controlled trial. Pediatric Rheumatology Collaborative Study Group. ) Rheumatol 21: 2353-2358, 1994.
C HAP T E R 142. Alsufyani K, Ortiz-Alvarez 0, Cabral 0, et al: The role of subcutaneous administration of methotrexate in children with juvenile idiopathic arthritis who have failed oral methotrexate. J Rheumatol 31: 179-182, 2004. 143. Woo P, Southwood TR, Prieur AM, et al: Randomizerd, placebo-controlled, crossover trial of low-dose oml methotrexate in children with extended oligoarticular or systemic arthritis. Arthritis Rheum 43: 1849-1857, 2000. 144. Gerloni V, Cimaz R, Gattinam M et al. Efficacy and safety profile of cyclosporin A in the treatment of juvenile chronic (idiopathic) arthritis: results of a 10-year propsective srudy. Rheumatology 40: 907-913, 2001. 145. Quartier P, Taupin P, Bourdeaut F, et al: Efficacy of etanercept for the treatment of juvenile idiopathic athritis according to the onset type. Arthritis Rheum 48: 1093-1101, 2003. 146. Lovell OJ, Giannini EH Reiff A, et al: Long-term efficacy and safety of etanercept in children with polyarticular-course juvenile rheumatoid athritis, Arthritis Rheum 48: 218-226, 2003, 147. Homeff G, Schmeling H, Biedermann T, et al: The German etanercept registry for treatment of juvenile idiopathic arthritis. Ann Rheum Dis 63: 1638-1644, 2004, 148. Yokota 5, Miyamae T, [magawa T, et al: Phase II trial of anti-lL-6 receptor antihody (MRA) for children with systemic-onset juvenile idiopathic arthritis. Arthritis Rheum 47: 5429, 2003. 149. Wilkinson NMR, Livermore P, Wythe H, Woo P: Phase II safety study of antiinterleukin-6 receptor antibody in systemic juvenile idiopathic arthritis, Presented at Paediatric Rheumatology European Society meeting. GarmischPartenkircher. Sept. 2003. 150. Lehman lJA, Striegel KH, Onel KB: Thalidomide thempy for recalcitrant systemic onset juvenile rheumatoid arthritis. J Pediatr 140: 125-127, 2002. 151. Schechter SJ, Sundel R, Oliveira 5, et al: Thalidomide for severe systemic onset .I1A. Arthritis Rheum 48: 592, 2003.
12
SYSTEMIC ARTHRITIS
303
152. Wulffmat NM, Brinkman 0, Ferster A, et al: Long-term follow-up of autologous stem cell transplantation for refractory juvenile idiopathic arthritis, Bone Marrow Transplantation 342: 561-564, 2003153. Quartier P, Prieur AM, Fischer A: Haemopoietic stem-cell transplantation for juvenile chronic althritis. Lancet 353: 1885-1886, 1999. 154. Schneider R, Lang BA, Reilly BJ, et al: Prognostic indicators of joint destruction in systemic-onset juvenile rheumatoid arthritis. J Pediatr 120: 20D-205, 1992. 155. Lin SJ, Huang JL, Chao HC, et al: A follow-up study of systemic-onset juvenile rheumatoid arthritis in children. Taiwan Erh Ko I Hsueh Hui Tsa Chih 40: 176-181, 1999. 156. van der Net J, Kuis W, Prakken AB, et al: Correlates of disablement in systemic onset juvenile chronic arthritis: a cross sectional study. Scand J Rheumatol 26: 188-196, 1997. 157. Lomater C, Gerloni V, G'ittinara M. et al: Systemic onset juvenile idiopathic arthritis: a retrospective study of 80 consecutive patients followed for 10 years, J Rheumatol 27: 491-496, 2000. 158. Wallace CA, Levinson .IE: Juvenile rheumatoid arthritis: outcome and treatment for the 1990s. Rheum Dis Clin Nonh Am 17: 891-905, 1991. 159. Oen K, Malleson PN, Cabral DA, et al: Disease course and outcome of juvenile rheumatoid arthritiS in a multicenter cohort. J Rheumatol 29: 1989-1999, 2002. 160. Oen K, Malleson PN, Cahral DA. et al: Early predictors of longerm outcome in patients with JRA: subset-specific correlations. .I Rheumatol 30: 585-593, 2003. 161. BaumJ, Gutowska G: Death in juvenile rheumatoid arthritis. Arthritis Rheum Suppl 20: 253-255, 1977. 162. Bernstein B: Death in juvenile rheumatoid arthritis. Arthritis Rheum Suppl 20: 256, 1977.
CHAPTEF
13
JUVENILE ANKYLOSING SPONDYLITIS James T. Cassidy and Ross E. Petty
;~~
DEFINmON AND CLASSIFICATION Juvenile ankylosing spondylitis (JAS) is a chronic inflammatOl)' arthritis of the axial and peripheral skeletons, frequently accompanied by enthesitis, characterized by rheumatoid factor (RF) and antinuclear antibody (ANA) seronegativity, and having a firm genetic basis. Radiologic evidence of bilateral inflammation of the sacroiliac joints is required for a definitive diagnosis. Ankylosing spondylitis (AS) in adults is defined by sets of criteria that are based on clinical, laboratory, and radiographic abnormalities. Only a few children or adolescents meet these criteria, principally because of the low frequency of spinal or sacroiliac signs or symptoms in the young patient. In addition, criteria for the diagnosis of AS in adults (Tables 13-1 through 13-3) are not applicable to the younger age group for a number of reasons: Data for some of the physical measurements have not been published for children, or, if reported (back range),l they have not yet been validated. In addition, limitations of spine and chest motion may reflect disease duration and are therefore of little aid in facilitating early diagnosis. 2 The fact that peripheral joint disease precedes clinical axial involvement by years in many children precludes an early diagnosis by criteria in which abnormalities of axial motion or radiologic changes are essential diagnostic features. Reasons for these differences between adults and children, whether they have immunologic, genetic, biochemical, or structural bases, are not understood. 3,4
Historical Review of the Spondyloarthropathy Concept The term spondyloarthropathy refers to a group of rheumatic diseases that predominantly affect the joints of the axial skeleton (as well as appendicular joints) and that differ from peripheral chronic arthritis in many ways.',6 Wright and MoW introduced the term spondarthrilis to include AS, psoriatic arthritis, ulcerative colitis, Crohn's disease, juvenile chronic arthritis, Whipple's disease, Beh\;et's syndrome, reactive arthritis, and acute anterior uveitis. They observed that these patients were RF seronegative, lacked subcutaneous nodules, and had inflammatory peripheral arthritis; many had radiologic sacroiliac
304
arthritis. They also observed a tendency toward familial aggregation. It is the familiality of these disorders, based on transmission of the histocompatibility antigen HLA-B27, that currently unites the somewhat smaller group included under the title seronegative spondyloarthritides: AS, the arthritides of inflammatory bowel disease (IBD) , reactive arthritis, and psoriatic arthritis. There are several reasons for grouping these disorders together under the heading of spondyloarthropathy: 1. Inflammation of the joints of the axial skeleton and of enthe-
ses is the most important clinical feature exhibited by members of this group of diseases that is less often observed in the other chronic arthritides. 2. Relatives of children with JAS commonly have AS, psoriatic arthritis, IBD, or, less commonly, reactive arthritis, related to the high frequency of B27 in these families. 3. A number of extra-articular features are shared by several diseases in this group, Iritis, usually acute, occurs in all members of the group. The cutaneous manifestations of psoriasis and reactive arthritis may be indistinguishable. 4. RFs are absent, and other autoantibodies are infrequent. Therefore, although individual members of the spondyloarthropathy group differ from each other, they share characteristics that distinguish them from the disease complex of chronic peripheral inflammatory arthritis and the other connective tissue diseases (Table 13-4), However, this classification does not recognize the heterogeneity within each of the four principal categories, particularly psoriatic arthritis and the arthritis of IBD, Only some of the patients with psoriasis have involvement of axial joints, and most children with arthritis related to IBD have peripheral arthritis rather than spinal involvement. Arthritis that is predominantly accompanied by inflammation of the entheses is recognized to be different from other types of inflammatory arthritis (although enthesitis occurs in a number of the chronic arthropathies of children), Its recognition in children and adolescents has been hampered by the lack of pediatric criteria resulting from the rarity of early axial spinal involvement. As a consequence, other approaches to the definition of these types of arthropathy have emerged, Recognition of the seronegative enthesitis and arthritis (SEA) syndromeR permitted the identification of children who, although they did not satisfy adult criteria for AS, were different from other children with chronic inflammatory joint disease in that they had inflammation of the entheses and peripheral arthritis, but usually not arthritis affecting the axial skeleton early after onset, Follow-up of these children9 indicated that many (although not all) developed
C HAP T E R
- : . TABLE 13 1
13
JUVENILE ANKYLOSING SPONDYLITIS
305
New York Criteria for a Diagnosis of Ankylosing Spondylitis (AS)
Olnlcal CrIteria 1. Limitation of lumbar spine motion in all three planes 2. Pain or history of pain at the dorsolumbar junction or lumbar spine 3. Limitation of chest expansion to 2.5 cm or less at the level of the fourth intercostal space
DefInite AS Grade 3-4 bilateral sacroiliac arthritis on radiography with at least one clinical criterion, or Grade 3-4 unilateral or grade 2 bilateral sacroiliac arthritis on radiography with clinical criterion 1 or clinical criteria 2 and 3
Probable AS Grade 3-4 bilateral sacroiliac arthritis on radiography without clinical criteria From Bennett PH, Wood PHN: Population Studies of the Rheumatic Diseases. New York, Excerpta Medica, 1968, p 456.
spine or sacroiliac joint involvement that permitted the classification of their disease as ]AS. The absence or rarity of axial spinal involvement in childhood led, also, to the definition of enthesitis-related arthritis (ERA) in the International League of Associations for Rheumatology OLAR) classification. 1Q-12 The suggested relationship among these three definitions is illustrated in Figure 13-1. Children with the SEA syndrome have some of the characteristics of ]AS but lack the sacroiliac joint involvement needed to confirm that diagnosis. 13 These children are seronegative
IS • -
TABLE 13-2
Amor Criteria for Classification of Spondyloarthropathies
Points
CrIteria Olnlcal Lumbar or thoracic night pain or stiffness Asymmetrical oligoarthritis Buttock pain alternating in site Buttock pain triggered by pelvic movement Sausage digit Enthesopathy Iritis Nonspecific urethritis or cervicitis within 1 mo before onset Diarrhea within 1 mo before onset Psoriasis, balanitis, or chronic enterocolitis
1 2 1 2 2 2 2 1 1 2
~I
TABLE 13-3 Classification Criteria of the European Spondyloarthropathy Study Group (ESSG)
Inflammatory spinal pain
RadIoIogk Sacroiliitis
(lack RF and ANA), have enthesitis (usually around the heel or knee), and have arthritis of a few joints, particularly the large and small joints of the lower extremities. The SEA syndrome probably represents, for the most part, children with very early ]AS or ERA, rather than a separate disease. It will take decades of observation and evaluation to determine whether ]AS, ERA, and the SEA syndrome are simply earlier or later, milder or more severe, versions of the same disease, although it seems highly likely that this will be the case. The disorder remits in some children, and they will presumably never develop a diagnosable seronegative spondyloarthropathy; others progress to juvenile rheumatoid arthritis (JRA) or systemic lupus erythematosus (see Fig. 13-1). In 1982, a group of 39 such children, representing approximately 20% of a pediatric rheumatic disease clinic population, were described as having the SEA syndrome.s Of these 39 children, 8 had bilateral sacroiliac arthritis consistent with a diagnosis of ]AS; 2 each had IBD and reactive arthritis; and 1 had reactive arthritis. s The remaining 26 children had no rheumatic disease that could be identified. The striking similarities of this group of patients and those with ]AS with regard to age at onset, mean number of affected joints, and family history of arthritis and back symptoms suggested that the SEA syndrome might be an early or mild form of ]AS. The high frequency of
or (~
stage 2 if bilateral;
~
stage 3 if unilateral)
plus
2
One of the follOWing: Positive family history Psoriasis Inflammatory bowel disease Urethritis, cervicitis, or diarrhea within 1 mo before arthritis Buttock pain alternating between right and left gluteal areas Enthesopathy Sacroiliac arthritis
GeneUc Presence of HLA-B27, family history of pelvospondylitis, reactive arthritis, psoriasis, uveitis, or chronic enterocolitis
1berlIpeutIc Amelioration of pain within 48 hr of treatment with nonsteroidal anti-inflammatory drug
Synovitis (asymmetrical or predominantly in lower limbs)
3
2
A definite diagnosis of spondyloarthropathy is confirmed if six or more points are present (sensitivity. 91.9%; specificity, 97.9%). From Amor B, Dougados M, Mijiyawa M: Criteres de classification des spondylarthropathies. Rev Rhum Mal Osteoartic 57: 85--89. 1990.
Modified from Dougados M, van der Linden 5, Juhlin R. el al: The European Spondyloarthropathy Study Group preliminary criteria for the classification of spondyloarthropathy. Arthritis Rheum 34: 1218-1227. 1991. Copyright © 1991 John Wiley & Sons. Inc. Reprinted with permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
306
C HAP T E R
I!.
13
TABLE 11 4
JUVENILE ANKYLOSING SPONDYLITIS
Overlapping
Charaderlstlc Enthesitis Axial arthritis Peripheral arthritis HLA-B27 positive ANA positive RF positive Systemic disease Eyes Skin Mucous membranes Gastrointestinal tract
lhdrdCterbti(~ 01
the
Spolldyl()"rthropdthie~
Juvenile Ankyloslng Spondylitis
Juvenile Psoriatic Arthritis
+++ +++ +++ +++
+ ++ +++
+
Inflammatory Bowel Disease
Reactive
Arthritis
+ ++ +++ +++
++ + +++ +++
+ + +
+
++
+
+
ANA, antinuclear antibodies; RF, rheumatoid factor. Frequency of characteristics: -, absent; +, <25%; ++, 25-50%;
+++
++++
+++,
HLA-B27 further supported this possibility. A follow-up study of these children a mean of 11 years after onset9 indicated that 12 (52%) of 23 who did not have a definite spondyloarthropathy at the time of the original report now had definite or possible spondyloarthropathy. The presence of HLA-B27 (62%, P < .004) and arthritis (as opposed to arthralgia, P < .05) and onset of disease after the age of 5 years (p < ,01) correlated with the evolution of the SEA syndrome to an identifiable spondyloarthropathy. Overall, 64% had definite or possible spondyloarthropathy, 10% JRA, 13% noninflammatory diseases, and 5% continuing SEA syndrome. Burgos-Vargas and Clark 14 described a group of Mexican children with the SEA syndrome who developed an inexorable course of progressive axial disease and sacroiliac arthritis. Within 5 years after onset, 75% had definite JAS. This was in contrast with the aforementioned report,9 in which clinical progression was varied and remissions more frequent. A definite diagnosis in these latter children was often achieved only after a follow-up of approximately 10 years. Jacobs and associates 15 studied 58 patients selected on the basis of the presence of HLA-B27 who had been monitored for a mean of 5 years. Two thirds were boys, and most had onset
• Figure 13-1 Diagnostic considerations in children with the seronegative enthesitis and arthritis syndrome (SEA), AS, ankylosing spondylitis; IBD, inflammatory bowel disease; PsA, psoriatic arthritis; RcA, reactive arthritis.
50-750/0;
++++,
+ + +++
75% or more.
of symptoms after 9 years of age; none had RF, and only 7 had ANA (8.6%). Altogether, 51 of 58 had disease that satisfied the American College of Rheumatology (ACR) criteria for a diagnosis of JRA, 16 1 had reactive arthritis, and 6 had episodic arthritis and enthesitis. In all, 75% of the children with HLA-B27 had enthesitis, and many had other features of one of the spondyloarthropathies, although definitive diagnostic outcomes were not presented. Sheerin and associates 17 monitored 36 of 85 children with arthropathy and HLA-B27 positivity for a mean of approXimately 9 years. Although only 2 of these children met New York criteria for AS, 27 (75%) of the 36 fulfilled the investigators' criteria for JAS. Five patients (24%) had enthesitis at onset, and at follow-up 22 patients (61%) had enthesitis or a history compatible with enthesitis. A group of 26 children with atypical spondyloarthropathy who fulfilled diagnostic criteria for the SEA syndrome or one of the other HLA-B27-associated syndromes were monitored by Hussein and colleagues 18 in an effort to define common characteristics among these various diseases. No diagnostic outcomes at follow-up were described in this study, although the proposed criteria tested favorably. A number of attempts to develop criteria for the classification or diagnosis of AS have been published. 1o.1J.J3.19 The New York criteria define adult AS (see Table 13-1),13.20 whereas the more inclusive criteria of Amor and coworkers 21 (see Table 13-2) and those of the European Spondylarthropathy Study Group (ESSG)19 (see Table 13-3), which are often preferred in clinical studies, describe spondyloarthropathy, a much broader category. In the system of classification of juvenile idiopathic arthritides proposed by the ILAR,10--12 the dilemma of categorization of these diseases is dealt with by recognizing ERA, juvenile psoriatic arthritis, and arthritis related to IBD as separate categories. In this classification, the category ERA best describes what has been called JAS. The ILAR criteria for a diagnosis of ERA are presented in Table 13-5. The unifying, if not entirely unique, feature of these disorders is the presence of inflammation of the enthesis-enthesitis. ERA can be viewed as an umbrella term that includes children withJAS (who fulfill the New York criteria for AS) and most patients with SEA syndrome (especially those who are HLA-B27 positive). These criteria have not yet been formally compared with the definition used in this chapter or with the New York criteria for AS. There remains a clear need for development and standardization of diagnostic criteria for JAS. In one study,22 2958 consecutive children with various forms of childhood arthritis (including 324 definite spondyloarthropathies and 334 possible
C HAP T E R
If!. II
13
JUVENILE ANKYLOSING SPONDYLITIS
307
Enthesitis-Related Arthritis (ILAR Classification)
TABLE 13-5
DefInition Arthritis and enthesitis
or Arthritis or enthesitis with at least two of the following: Sacroiliac joint tenderness and/or inflammatory spinal pain Presence of HLA-B27 Family history in at least one first- or second-degree relative of medically confirmed HLA-B27-associated disease Anterior uveitis that is usually associated with pain, redness, or photophobia Onset of arthritis in a boy after 8 yr of age
Exduslons Psoriasis confirmed by a dermatologist in at least one first- or second-degree relative Presence of systemic arthritis (LAR, International League of Associations for Rheumatology. From Petty RE, Southwood TR, Baum J, et al: Revision of the proposed classification criteria for juvenile idiopathic arthritis: Durban, 1997. J Rheumatol 25: 1991-1994, 1998.
spondyloarthropathies) and 2300 control subjects were evaluated with the adult AS criteria of Amor and coworkers 21 and those of the ESSG (see Tables 13-2 and 13-3).19 Children with JAS were identified by the Amor criteria with a sensitivity of 73.5% and specificity of 97.6%, and by the ESSG criteria with a sensitivity of 78.7% and specificity of 92.2%. Inflexible application of any system of classification risks putting patients into categories in which they do not belong. This was a major problem with the spondyloarthropathy concept, but it could equally be problematic with the ILAR classification. Using the spondyloarthropathy concept, many patients with psoriatic arthritis did not have enthesitis or disease of the axial skeleton, although they were categorized as having AS. On the other hand, the occasional child with psoriatic arthritis and sacroiliac arthritis would probably best be categorized as having ERA but is excluded from that category because of the presence of psoriasis. Thus many classification problems still await clinical and laboratory evidence for their scientific resolution!
EPIDEMIOLOGY Inddence and Prevalence The relative frequencies of JAS derived from data accumulated in the national pediatric rheumatic disease reg-
I!. II
istries of the United States,23 Canada,24 and the United Kingdom25 and from studies in Sweden 26 and Finland27 ,28 are summarized in Table 13-6. JAS accounted for 1% to 7% of children in these studies. Ladd and colleagues29 reported on 15 patients with onset of JAS (New York criteria) at or before age 16 years who were monitored over a 10-year period; during that time, 208 children with JRA were registered in the same clinic, for a ratio of approximately 1: 14. Increasing awareness of the possibility of the occurrence of JAS-like diseases in childhood and their clinical and laboratory differentiation from other chronic arthritides of childhood will probably result in an increase in the proportion of children with inflammatory arthritis in this category. It is not certain that JAS and AS are the same diseases, although they are undoubtedly closely related. The proportion of adults with AS who developed an onset of the disease in childhood ranges from 8.6%19 to 11%.30 Frequently cited data in adults for prevalence are 0.5% (0 1.9%.31,32 Estimates of the frequency of AS range widely, however, among ethnic and racial groups. Using modified New York criteria,20 including radiographic evidence of sacroiliac arthritis, Carter and coworkers 33 determined a prevalence of 129/100,000 (0.13%) in an American population of Northern European extraction. A follow-up study of the same population confirmed that the incidence of AS was not changing over time. 34 On the basis of the prevalence of 827 antigen
TABLE 13-6 Relative Frequencies of Juvenile Rheumatoid Arthritis (JRA). Juvenile Ankylosing Spondylitis (JAS). and the Spondyloarthropathies (SpA)
23
United States United Kingdom 25 Canada" Sweden26 Finland 27
JASt
JRA"
location and Ref. No.
SpA
No.
%
No.
%
No.
%
2071 1483 521 216
71 79 61 96 99
75 37 65 2 1
3 2 7 1 1
757 348 274
26 19 32 3
114
"EqUivalent to the American College of Rheumatology definition. tIncludes 15 children with definite and 50 with probable JAS.
7
308
CHAPTER
13
JUVENILE ANKYLOSING SPONDYLITIS
and the frequency of sacroiliac arthritis in the B27-positive population, the prevalence of AS was estimated to be 0.86% to 1%,31,35 and was highest in B27-positive persons,35 Although this estimate includes asymptomatic persons, it also excludes the 8% of the AS population who do not have B27, and it may be a more accurate reflection of the prevalence of the entire spectrum of AS, whereas the estimate of Carter and colleagues33 more accurately represents patients with clinically and radiologically evident disease.
Age at Onset JAS usually has its onset in late childhood or adolescence, although instances of onset in younger children have been reported.-'6,-'7 The age distribution appears to be homogeneous and presumably is continuous with that described in adult populations, suggesting that, at least on this basis, the disease as seen in adults is the same or very similar to that in children. 29.-'8,39
Sex Ratio JAS has a much higher frequency in boys than in girls: of 247 children with this disorder, 216 were boys, for a male/female ratio of 7:1. 29 ,38-44 This disproportionate representation of boys may not accurately represent the actual occurrence of the disease in girls, The strong correlation between JAS and HLA-B27 and the equal distribution of this antigen in males and females suggest that JAS could be as common in girls as in boys. Furthermore, in radiographic surveys of B27-positive adult blood donors, sacroiliac arthritis was as common in women as in men,45 In a questionnaire survey of members of the National Ankylosing Spondylitis Society in the United Kingdom, the male/female ratio was 2.7:1.-'0 However, manifestations in women may be less severe,46 and they may have more peripheral and less axial disease. 47 It is possible that these observations contribute to the relative infrequency of the diagnosis in women as well as diagnositic confusion with ileitis condensans ilei.
Geographic and Radal Distribution Few data are specifically related to geographic and racial differences in the frequency of JAS. 48 The low incidence of AS in African Americans49 and in Japanese 50 and the high frequency in the Haida Indians of Pacific Canada 51 reflect, in part, the frequency of B27 in these populations. Other factors may be significant, however, because this antigen occurs in only 50% of African Americans with AS52 and in only 65% to 90% of Japanese with the disease. 49
The strong familial association with B27 suggests that a genetically determined mechanism is central to pathogenesisY B27 itself may be involved in pathogenesis, or it may merely serve as a marker gene. Molecular mimicry, in which there is an immune response to an amino acid sequence that is shared by certain B27 molecules and Klebsiella species, has been suggested as another pathogenic mechanism. S6 ,57 Reports of an association between B27 and gastrointestinal (GI) isolation of Klebsiella species in adults with AS suggest a role for these organisms, but these studies remain largely unconfirmecl.'H,w The observations of Mielants and associates60 ,61 support a pathogenic relationship between spondylitis and inflammation of the GI tract. Clinical or occult GI inflammation62 may be related in AS to the association with HLA antigens 63 as well as cellular immunity to cartilage proteoglycans. 64 The reactivity of synovial fluid lymphocytes to microbial antigens correlates with the specificity of extra-articular bacterial isolates in adult patients with chronic arthritis. 65 A modulatory effect of the surface expression of the B27 molecule on the invasive capability of arthritogenic bacteria, such as Yersinia enterocolitica and Salmonella enteritidis, has been demonstrated in murine cells66 ,67 but not in human fibroblasts or lymphocytes. 6H ,69 Some investigators have found that, although bacterial invasion of B27-expressing cells is normal, killing of the organisms in such cells is impaired, with the result that infective organisms persist within the host. 7t) An inflammatory infiltrate that includes CD8+ and CD14+ cells develops at sites of enthesitis in subchondral bone with bone absorption and new bone formation.7 1 Tumor necrosis factor-a. (TNF-a.) messenger RNA is increased in affected bone. 72 Expression of inflammatory cytokines and their receptors has been studied in synovial tissues and CD2R on T cells. 73 A type 1 helper T-cell (Thl) response has been suggested, with lymphocytic and mononuclear cellular infiltrates. Cells of the synovial membrane express TNF-a., TNF-~, and TNF receptors similar to those of children with JRA. 74 The B27 transgenic rat is an important animal model for studying these relationships.7s-77 This animal develops inflammatory disease that bears a striking resemblance to human spondyloarthropathy. One Lewis strain and another Fisher line express B27 and human ~2-microglobulin; they spontaneously develop chronic inflammatory disease affecting both axial and peripheral joints, the GI tract, skin, and nails. The lamina propria of the gut is infiltrated by B27-positive T lymphocytes. It has been postulated that the high density of the B27 molecule on mononuclear cells of the affected rat strains facilitates the presentation of a B27 peptide to a reactive T-lymphoeyte population, triggering the inflammatory process. An alternative hypothesis attributes susceptibility to rnisfolding of B27 heavy chains in the endoplasmic reticulum. 7H Maintaining the B27 transgenic rat in a germ-free environment abrogates development of the spondyloarthropathy. Spondylitis in turkeys has been associated with infection with Pasteurella anatipestifer. 79
GENETIC BACKGROUND ETIOLOGY AND PATHOGENESIS There is no known cause of JAS. The clinical, genetic and epidemiologic similarities of JAS and diseases such as reactive arthritis, in which enteric or genitourinary tract infections playa triggering role, suggest an infectious etiology, although none has been proven. 53 Although no organisms can be isolated from the joints, a local inflammatory response to antigen is supported by antibody and cellular immune studies. 54 ,55
There is often a striking familial occurrence of spondylitis and related diseases in adults and children. 80 Ansell and colleagues81 noted that 6 of 12 monozygotic twin pairs concordant for arthritis were B27 positive, which made the diagnosis of JAS likely (if not certain) in these children. Family studies have indicated that AS is inherited as an autosomal dominant trait with penetrance of about 20%.82 Although the risk of development of AS in a B27-positive person is not precisely known (approximately 1% to 3%), epidemiologic studies suggest that AS
C HAP T E R
occurs 10 to 20 times more frequently in relatives of patients with AS (20%) and 50 to 80 times more frequently in their siblings. 83-85 Thus, B27-positive persons with a family history of AS have a lO-fold greater risk of AS than that of B27-positive persons with no family history of AS. 84 The general risk that a B27-heterozygous parent with AS will have a male child with the disease is approximately 5% to 10% (20% if the child is also B27 positive; close to zero if the child is B27 negative).84 The risk of having a female child with ]AS is lower. Risk mayor may not be increased in first-born children. 86 .87 Familial disease may not be concordant for phenotypes in each member. 88 The studies of Brewerton83 and Schlosstein89 and their colleagues (and subsequently many others) indicated that B27 was strongly associated with AS in adults (Table 13-7). The association between ]AS and the B27 antigen is as strong in children as in adults. Of 247 children with ]AS, B27 was present in 91%.29.38-44 One study demonstrated restriction fragment length polymorphisms of B27 that had striking associations with AS. 90 There are now 25 known subtypes of B27;91,92 all except one are associated with AS. No specific associations with these subtypes have been identified by oligonucleotide probes B*2701-2706. 93 In the Gambian population of Africa, a unique subtype, B2703, is not associated with AS.94 The possibility that homozygosity for B27 was responsible for the juvenile onset of AS was not supported by data in one small study,95 nor was the idea that disease severity was genetically determined. 96 •97 Although B27 has the strongest genetic association with AS and contributes the greatest share (15% to 60%) of the attributable genetic risk,82.98 other genetic factors undoubtedly playa role. There is also an increase in HLA-A2, an association that is shared with other chronic arthritides of childhood. 99 Woodrow 1oo used meta-analysis to calculate a relative risk of spondylitis of 1. 72 in A2-positive patients. Reports of an increased frequency of HLA-A28 in B27-positive patients with AS suggest that this antigen also may contribute to disease susceptibility.lOl.102 HLA-B60 is increased in adults with AS, independent of the presence of B27.103.104 The reported increased frequency of Cwl and CW2 105.106 probably reflects a linkage disequilibrium with B27. One study alleged that a -308.1 polymorphL~m in the promoter region of the TNF gene was associated with AS and independent of B27. 107 A polymorphism in the interleukin-l receptor antagonist OL-l Ra) gene is also associated with ankylosing spondylitis. 108 There are few known class II associations with ]AS. A higher frequency of HLA-DRBP08 (44.9%) was reported in a Mexican population with ]AS than in a control population (25.4%).109
t.
TABLE 13 7
13
JUVENILE ANKYLOSING SPONDYLITIS
Maksymowych and colleagues llO reported that the LMP2A allele frequency in patients with adult- and juvenile-onset AS with uveitis was twice that in those without this complication (odds ratio, 2.51). Ploski and colleagues lll reported an increase of B·4001, DRBP08, and DPB1·0301 and the LMP2 bib phenotype in patients with ]AS compared with B27-positive controls or adults with AS.
CLINICAL MANIFESTATIONS The onset of ]AS may be insidious and characterized by intermittent musculoskeletal pain and stiffness or objective inflammation of peripheral joints, particularly those of the lower extremities, together with enthesitis at one or more sites around the knee or foot. In other patients, oligoarthritis may have an abrupt onset. Important additional diagnostic information may indicate the presence of a complication such as iritis or cardiovascular disease. Furthermore, cutaneous, mucous membrane, GI, or genitourinary abnormalities may lead to consideration of an alternative diagnosis from among the other members of the spondyloarthropathy group. 112 Systemic signs are often minimal, but fatigue, sleep disturbances, and lowgrade fever may be present. Symptoms related to the back are usually absent at onset but become increasingly evident during the disease course in adolescents. Differing modes of presentation and course may characterize specific population groupS.113.114
Enthesltls Entheses-the sites of attachment of ligament, tendon, fascia, or capsule to bone-are characteristic sites of inflammation in the spondyloarthropathies. Although the presence of exquisite, well-localized tenderness at characteristic entheses strongly suggests spondyloarthropathy, it must be noted that enthesitis occurs occasionally in]RAl15 and even in systemic lupus erythematosus; such findings could also be confused with other noninflammatory conditions such as Osgood-Schlatter disease and Sever's disease. The presence of enthesitis is, however, the most helpful feature in differentiating]AS from]RA, 116 although it is not rare in normal children. 115 Enthesitis is a characteristic early manifestation of ]AS and occurs with greater frequency in ]AS than in adult-onset AS. It frequently produces severe pain and resultant disability, which may be
Genetic Assodations with Ankylosing Spondylitis (AS) or Juvenile Ankylosing Spondylitis (JAS)
Gene
AssocI.don
Ref. Nos.
HLA-B27 HLA-A2 HLA-A28 HLA-B60 HLA-DRBl'08 LMP-2A LMP-2 bib TNF polymorphism
t t
29, 39, 41-44, 83, 89 99, 100 101, 102 103, 104 109
IL-1Ra polymorphism
t t t t t t t
309
in AS, lAS in AS, lAS in B27-positive AS in AS in Mexican children with lAS in Mexicans with AS, lAS, and iritis in lAS in AS in AS
110 111
107 108
HLA. human leukocyte antigen; IL-lRa, interleukin-l receptor antagonist; LMP, _ _; TNF, tumor necrosis factor.
310
C HAP T E R
13
JUVENILE ANKYLOSING SPONDYLITIS
the child's most important complaints. JJ7 Deposits of acid mucopolysaccharides in the extensor tendons of the adult foot, resembling tendonitis, have been described as "tamale foot. "118
Arthritis The presenting joint symptoms recorded in the largest reported series of JAS are summarized in Table 13-8. Initial musculoskeletal symptoms are often difficult for the child to localize and include pain in the buttocks, groin, thighs, heels, or around the shoulders. The vague quality and localization of this pain and its frequent spontaneous disappearance early in the disease are recurring sources of delay and confusion in diagnosis. In distinction to AS, JAS seldom causes symptoms of involvement of the axial skeleton at onset; only 24% of children with JAS are reported to have pain, stiffness or limitation of motion of the lumbosacral spine or sacroiliac joints at presentation. In contrast, peripheral joint symptoms occur at onset in 82%, whereas only 16% have involvement of the upper extremities (see Table 13-8). With the exception of one report in which hip disease was frequent at onset,119 distal joints are affected more commonly than proximal joints. In most instances, the number of joints involved is limited (four or fewer), although approximately 25% of children have a polyarticular onset. Shoulders are not uncommonly affected, and even the temporomandibular joint may be involved. The least commonly affected joints are the small joints of the hands. Pain at costosternal and sternoclavicular joints and the sternomanubrium, often in conjunction with tenderness over the proximal clavicle, may be associated with significant
I!:.
TABLE 13 H Mus(uloskele1al Signs and SYIllI)loll1s in Juvenile Ankylosing Spondylitis (lAS)'
Clinical Evidence of Joint Involvement at Onset
Percentage
Arthritis, Painful Umltatlon of Range of Motion Proximal limb joints Distal limb joints Upper limb joints Lower limb joints Axial skeleton joints
35 44 16 82 24
Joint Involvement During Course No peripheral joints affected 1 to 4 peripheral joints affected More than 4 peripheral joints affected Sacroiliac involvementt Lumbosacral spine affected! Cervical spine involvement
3 43 54 95 90
-
Enthesltls Around the kneet Around the ankle and foot t
80 90
Myopathy
Pain/Wastingt
50
'Clinical or radiographic evidence of involvement of the sacroiliac joints, and particularly the lumbosacral spine. may not be evident until adulthood. Data are from published studies. 'Estimate.
impairment of chest expansion. In the series of Schaller and associates,38 5 of 7 patients had decreased chest expansion. Aside from the number and distribution of the affected joints, there is nothing clinically to distinguish the peripheral joint disease of JAS from that of JRA. Burgos-Vargas and colleagues 120 have identified a subgroup of children with typical adult-type onset of disease. Whether this presentation represents a distinct entity or merely an extreme end of the clinical spectrum is not certain. 121
Iritis The iritis is characterized by an acutely red, painful, photophobic eye. It is usually unilateral, is frequently recurrent, and usually, but not always, leaves no ocular residua. It rarely precedes the onset of musculoskeletal complaints. 122 Acute iritis occurs in 20% of adults with AS, particularly in those with peripheral joint involvement, but may be less common in JAS. 123 In the series of Ladd and colleagues,29 only 1 of 15 patients developed an acute iritis; in the series of Schaller and associates,43 2 of 20 patients had this complication. However, Hafner4 4 indicated that 14% of 71 patients had a history of an acute iritis, and AnselJ37 observed this complication in 21 of 77 patients. These higher figures may reflect a longer follow-up time in the latter two studies or other confounding factors.
Cardiopulmonary Disease Although cardiovascular disease is uncommon, it can occasionally be severe,124 and marked aortic insufficiency, sometimes with dilatation, has been reported in at least 7 patients with JAS,125-131 in 1 patient with sacroiliac arthritis and regional enteritis,37 and in 1 patient with reactive arthritis. 132 The apparently low frequency of such complications in children may reflect the fact that follow-up is generally of shorter duration than in adults in whom cardiac disease (aortic insufficiency, heart block) develops in approximately 5% of patients an average of 15 years after onset. 133 Rarely, cardiac involvement may precede development of sacroiliac disease. 134 None of 36 consecutive patients with ]AS who were monitored for a mean of 4.3 years had symptoms related to the cardiovascular system, and only one developed the murmur of aortic regurgitation. Echocardiographs documented no structural cardiac abnormalities, and electrocardiography no conduction defects, but color Doppler assessment confirmed mild mitral regurgitation in 2 patients and mild aortic regurgitation in 3; systolic ventricular function was impaired in 1. Transesophageal echocardiography demonstrated aortic root abnormalities and valvular disease in 82% of adults as compared with 27% of controls. 136 Valve thickening was demonstrable as nodularity of the aortic cusps and basal thickening of the anterior mitral valve leaflet, creating the characteristic subaortic bump. Aortic valve regurgitation was present in almost half of the patients. Few data relating to pleuropulmonary disease are available. In a study of 18 children aged 8 to 17 years who fulfilled the Amor criteria. 21 abnormalities of pulmonary function were present in 33%.137 All patients had nonnal chest radiographs at baseline
I"
C HAP T E R
13
JUVENILE ANKYLOSING SPONDYLITIS
311
and on follow-up at 2 years. No patient had symptoms attributable to the respiratory system, and all had normal chest expansion. Nonetheless, 6 patients (33%) had abnormal pulmonary function tests. The most common abnormality was reduction in the forced vital capacity (22%); occasionally, increased functional residual capacity (11%) and residual volume (5%) were observed. Restrictive patterns were more common than diffusion defects, and diffusing capacity of the lungs for carbon monoxide was reduced in only 11%. Small airways disease was not present. In adults, although diminished chest expansion and resultant decreased vital capacity are not infrequent, clinical parenchymal pulmonary disease is rare. In the review by Rosenow and associates,138 1.3% of 2,080 adults with AS had radiographic evidence of pleuropulmonary disease (apical pleural thickening). High-resolution computed tomography (Cn in 26 adults with AS demonstrated a much higher frequency of pulmonary parenchymal abnormalities (69%) than was evident on plain radiography (15%).139 These findings included interstitial lung disease in 4 patients, bronchiectasis in 6, paraseptal emphysema in 3, and tracheal dilatation and apical fibrosis in 2 each. Cor pulmonale can develop secondary to kyphoscoliosis and decreased chest wall movement, characteristic of advanced spondylitis, but it has not been reported in children or adolescents.
tion. The earliest lesion in the sacroiliac joints is subchondral inflammation, rather than synovitis, with formation of granulation tissue with few inflammatory cells. The surfaces of the sacroiliac joints are little affected and pannus is not present. 147 Enchondral ossification on the iliac side of the joint accounts for the radiographic appearance of erosions. Ball commented,l48 "As a rule, it seems that in any synovial joint in ankylosing spondylitis tlle outcome represents a balance of erosive synovitis and capsular and/or ligamentous ossification. In joints of low mobility the ossific process tends to be the dominant feature." Enthesitis l49 is characterized by nonspecifiC inflammation. Granulation tissue, infiltrated with lymphocytes and plasma cells and causing a localized osteitis, undermines the bony and cartilaginous attachment of the ligament or tendon. Healing of this lesion gives rise to a bony spur, such as a calcaneal spur at the insertion of the plantar fascia into the calcaneus or a syndesmophyte at the attachment of the outer fibers of the annulus fibrosus to the anterolateral aspects of the rim of the vertebral body.
Nervous System
At onset, ]AS may mimic other inflammatory arthropathies, mechanical causes of back or lower extremity pain, or, very occasionally, infection or malignancy. It is the sequential development of the combination of arthritis, enthesitis, and axial skeletal involvement that points to a spondyloarthropathy; the absence of cutaneous, GI,150 or genitourinary tract disease (in which spondylitis occurs-see Chapters 15 and 30) further narrows the diagnosis to ]AS. In most instances, however, children lack sacroiliac and back symptoms at onset, and differentiation of ]AS from ]RA and other disorders of the entheses, back, and sacroiliac joints may be difficult. Arthritis of the cervical spine is infrequent and, when present, mimics that in children with polyarticular ]RA. Thoracolumbar pain may reflect Scheuermann's disease. Lumbar and lumbosacral pain have a myriad of causes, including spondylolysis, spondylolisthesis, osteoid osteoma, osteomyelitis, diskitis, and (rarely) lumbar disk herniation. Trauma may cause chronic pain in the sacrum and coccyx. Sacroiliac tenderness and pain occurs in many patients with ]AS, but septic sacroiliac disease, osteomyelitis, Ewing's sarcoma of the ilium, and familial Mediterranean fever l51 also produce pain in and around these joints. Pain that mimics enthesitis may result from a number of causes, including excessive running or jogging. Usually, the pain of traumatic enthesopathy is less severe and more diffuse than that caused by inflammation. Osteochondrosis of the tibial tuberosity (OsgoodSchlatter disease), of the inferior pole of the patella (Sinding-Larsen-]ohansson syndrome), or of the apophysis of the calcaneus (Sever's disease) may mimic inflammatory enthesitis at those sites. The coexistence of enthesitis at multiple sites usually eliminates these disorders from consideration. The absence of B27 positivity also assists in differentiating these disorders from the inflammatory enthesitis of ]AS. 152 Pressure over bony
Central nervous system disease is rare. Atlantoaxial subluxation leading to severe cervico-occipital pain was reported in 1 boy with ]AS I40 and in 2 boys with the SEA syndrome. 141 The cauda equina syndrome, caused by bony impingement on the cauda equina and characterized by weakness of bowel and bladder sphincters, saddle anesthesia, and leg weakness, occurs in adults l42 but has not been reported in children.
Renal Disease Renal abnormalities are rare. Papillary necrosis, perhaps secondary to nonsteroidal anti-inflammatory drugs (NSAIDs), has been reported. 143 Immunoglobulin A (IgA) nephropathy, occasionally with iritis,144 was observed in 115 adults with spondyloarthropathies. 145 Most of these patients had elevated serum IgA concentrations; some had impaired renal function and hypertension. AnselJ37 documented amyloidosis in 3.8% of 77 patients with ]AS seen before 1980; she noted its association with severe peripheral arthropathy and a persistently elevated erythrocyte sedimentation rate (ESR).
PATHOLOGY The pathology of]AS has not been studied, but it is probable that abnormalities are similar to those of AS. However, the synovitis is in general much milder, and the degree of cartilage erosion in peripheral joints much less, in AS compared with adult rheumatoid arthritis (RA). 146 The synovitis itself is otherwise virtually indistinguishable from that of RA, although there may be relatively more polymorphonuclear leukocytes present. The characteristic pathologic changes in the apophyseal and sacroiliac joints are enchondral and capsular ossifica-
DIFFERENTIAL DIAGNOSIS
312
C HAP T E R
13
JUVENILE ANKYLOSING SPONDYLITIS
prominences, including entheses, may produce pain in children with leukemia or bone tumors. In most instances, however, the pain resulting from such infiltrative diseases is less discrete and more severe than that of inflammatory enthesitis and frequently awakens the child from sleep.
ities. Symptoms of pain or tenderness at the origin of the adductor longus near the symphysis pubis,153 at the costochondral junctions, and over spinous processes, although infrequent, support a diagnosis. Observation of stance and gait may indicate that the child stands or walks so as to avoid pressure on inflamed entheses.
Musculoskeletal Examination
Peripheral Joints
The musculoskeletal examination can be divided into three parts: (1) the entheses, (2) the peripheral joints, and (3) the axial skeleton including joints of the pelvis, spine, and chest (see Table 13-8).
The peripheral arthropathy of JAS may be indistinguishable from that of JRA or the other spondyloarthropathies, although the number and distribution of affected joints provide helpful diagnostic clues to the differentiation of these diseases. The arthritis is often asymmetric and involves the lower extremities. Isolated hip disease may be the presenting feature,154 but this would be highly unlikely in a child with JRA. Although involvement of one or both knees is characteristic of both oligoarticular JRA and JAS, the child's age at onset, especially if a boy, is a useful distinguishing feature. Small joints of the toes are commonly involved. For example, the presence of arthritis in the metatarsophalangeal joint of the first toe, the ankle, or the knee strongly suggests the diagnosis; in contrast, symmetric disease of the small joints of the hands or polyarticular disease, particularly in a girl, is more likely to be JRA (Table 13-9), A highly characteristic intertarsal joint inflammation occurs in many children (Fig. 13-3). This tarsitis is accompanied by pain, tenderness, and restriction of
Entheses A careful history and a thorough but gentle palpation of entheses may document evidence of past or present inflammation. Enthesitis is often remarkably discrete and painful. A diagnosis of JAS is strongly supported by marked tenderness on the patella at the 2, 6, and 10 o'clock positions, at the tibial tuberosity, at the attachment of the Achilles tendon or plantar fascia to the calcaneus, at the attachment of the plantar fascia to the base of the fifth metatarsal, and at the heads of the metatarsals (Fig. 13-2). Tenderness is less commonly demonstrable at the greater trochanters of the femurs, superior anterior iliac spines and iliac crests, pubic symphysis, and ischial tuberosities, but seldom at entheses of the upper extrem-
• Figure 13-2 A, Atrows indicate the most common sites of tenderness associated with enthesitis at the insertions of the quadriceps muscles into the patella and the attachments of the patellar ligament to the patella and tibial tuberosity. 8, Atrow indicates the site of tenderness at the insertion of the Achilles tendon into the calcaneus. C, Arrows Indicate the most common sites of tenderness associated with enthesitis at the insertion of the plantar fascia into the calcaneus, base of the fifth metatarsal, and heads of the first through fifth metatarsals. Swelling in this area is best visualized by having the child lie prone on the examining table with the feet over the edge. (8 and C, From Petty RE, Malleson P: Spondyloarthropathies of childhood. Pediatr elin North Am 33: 1079-1096,1986.)
C HAP T E R
1:..
13
JUVENILE ANKYLOSING SPONDYLITIS
313
-jJ
TABLE 13-9
Comparison of JRA, JAS, and the SEA Syndrome
Charadelfstlc Male/female ratio Average age at onset (yr) Average number of joints involved Family history of arthritis (%) Back signs (%) ANA positive (%) RF positive (%) HLA-B27 positive (°;6)
JRA
JAS
1:4
7:1
5
>10
9 (may be many) 30 2
30-80 15 15
6 (rarely many)
SEA 9:1 10
5 (r
>65
65
>100
45
90
72
o o
o o
.IRA, juvenile rheumatoid arthritis: .lAS, juvenile ankylosing spondylitis: SEA, seronegative enthesitis and arthritis syndrome; ANA, antinuclear antibodies: HLA, human leukocyte antigen; RF, rheumatoid factor.
movement in the midfoot that, in the presence of disease of the first metatarsophalangeal joint, results in a characteristic deformity of the foot. Burgos-Vargas and associatesl>s concluded that a diagnosis of ]AS could be confirmed or strongly suspected shortly after onset of disease in children who displayed enthesopathy, midtarsal foot involvement, sparing of the hands, and progressive onset of lumbosacral disease,
Axial Skeleton Involvement of the joints of the axial skeleton is central to the diagnosis. In children with sacroiliac inflammation, pain may be elicited by direct pressure over one or both sacroiliac joints, compression of the pelvis, or distraction of the sacroiliac joints by Patrick's test. Examination of the back should be directed at detecting asymmetry in
• figure 13-3 Tl-magnetk resonance images of bony ankylosis (top) and T2-hyperintensive signals from the synovial sheath and bursae (bottom). (From Burgos-Vargas R, Granados-Arriola J: Ankylosing spondylitis and related diseases in the Mexican mestizo. Spine 44: 665, 1990.)
the standing position. Abnormalities in contour, such as loss of the normal lumbar lordosis or thoracic kyphosis, or occiput-to-wall apposition, are best observed while the child is standing upright. The contour of the back on full forward flexion may demonstrate loss of the normal smooth curve in the lower part of the thoracolumbar spine (Fig. 13-4), or there may be restriction of hyperextension, signifying early axial disease. The rigid spine of long-standing AS is rare in children. Cervical spine involvement is also a late development. 156 Although observations of abnormalities of the contour of the back are often more informative than actual numeric measurements, sequential measurement of thoracolumbar mobility is useful in documenting progression of the disease.
• Rgure 11-4 A 15-year-old boy shown in the position of maximal forward flexion. Note the flattened back (arrow). Radiographs demonstrated bilateral sacroiliac arthritis but no abnormality of the lumbosacral spine.
314
C HAP T E R
13
JUVENILE ANKYLOSING SPONDYLITIS
The modified Schober test 1'\7,lSll provides one index of abnormality (Fig, 13-5), With the child standing with the feet together, a line joining the dimples of Venus is used as a landmark for the lumbosacral junction. A mark is made 5 cm below (point A) and 10 cm above (point B) the lumbosacral junction. With the patient in maximal forward flexion with the knees straight, the increase in distance between points A and B is used as an indicator of lumbosacral spine mobility. Normal values plus or minus 1 standard deviation are indicated in Figure 13-6.' In general, a modified Schober measurement of less than 6 cm (e.g., an increase from 15 cm to less than 21 cm) should be regarded as abnormal. However, care should be exercised in interpreting this measurement, because there are large normal variations at each age and the data have not been adequately validated in children with musculoskeletal disease, Measurement of the distance from t1ngertips to floor on maximal forward flexion is often used to quantitate spinal motion but is poorly reproducible and does not correlate with the Schober measurement. Furthermore, finger-tofloor distance reflects hip as well as back flexion.
Thoracic disease may be reflected in limitation of chest expansion. Normal thoracic excursion varies a great deal, and normal age- and sex-adjusted ranges have not been established. However, in a specific child, sequential
10
Boy.
Girls
9 B
e em
~ "------',
==;v-"
,,------
,.---
~
~
4
2
10
II
12
13
14
15
12
13
14
15
Age Iyrs)
• Figure 13-6 Normal values for the modified Schober test: mean (dashed line) ± 1 standard deviation (solid lines). (Adapted from Moran HM, Hall MA, Barr A, et al: Spinal mobility in the adolescent. Br J Rheumatol Rehab 18: 181-185, 1979.)
measurement of thoracic motion may be useful in documenting progressive loss of range. In the adolescent, any thoracic excursion of less than 5 em (maximum expiration to maximum inspiration, measured at the fourth intercostal space) should be regarded as probably abnormal. Even in the absence of symptoms, chest expansion in children with ]AS may be restricted to 1 or 2 em, Pain and tenderness at the costosternal and costovertebral joints may be elicited by firm palpation. Sternomanubrial tenderness sometimes occurs, but sternoclavicular pain is more common,
LABORATORY EXAMINATION
• Figure 13-5 Schober test. A, Measurement 10 cm above and 5 cm below the lumbosacral junction (the dimples ofVenus) in the upright position. 8, Measurement of the distance between the upper and the lower marks when the child is bending forward.
There are few distinguishing laboratory features. Anemia is usually mild and characteristic of the anemia of chronic disease. White blood cell counts are usually normal or moderately elevated with normal differential counts. Indices of inflammation are frequently abnormal. The platelet count and the ESR are often elevated and may remain so for years. Very high values for the ESR (>100 mm) occasionally occur but should also suggest the possibility of occult IBD. Conversely, a normal ESR may accompany clinically active disease. Elevated immunoglobulin levels reflect inflammation, and selective IgA deficiency has been reported. 1'\9,160 High levels of IgA and C4 161 ,162 and of circulating immune complexes l63 in adults with AS suggest an immunoreactive state. Characteristically, RFs are absent. ANAs do not occur in children with ]AS more commonly than in a healthy population. Antiphospholipid antibodies have been demonstrated in 290/0 of adults with AS,I64 but children with ]AS have not been studied. Although there are no repOlts of systematic studies of other autoantibodies in ]AS, experience suggests that they are not common or expected. HLA-B27 is present in 90% of children with ]AS ancl in approximately 8% of the overall white population; it does not constitute a diagnostic test but rather an indicator of risk, The diagnosis of ]AS rests on clinical characteristics, and the use of HLA typing for diagnosis may lead to
C HAP T E R
misdiagnosis, although typing is important as a criterion to classify patients for study (see Geographic and Racial Distribution). There are no specific studies of synovial fluid, but the changes are probably similar to those in adults with AS, in which the differential white blood cell count includes more neutrophils and fewer lymphocytes than in RA. I 65 It has been reported that the predominant large mononuclear cell in synovial fluid in RA is lymphocyte derived, whereas that in AS is of macrophage origin. l60 Macrophages containing degenerated neutrophils are more common in synovial fluid of patients with AS and related diseases such as reactive arthritis than in those with RA. 167 Descriptions of the synovial fluid are otherwise similar to those for RA, except that the complement level is usually normaP6R or increasecJ.37
RADIOLOGIC EXAMINATION Saaolllac Joints Radiographic demonstration of bilateral sacroiliac arthritis is necessary to establish an unequivocal diagnosis of inflammatory spondylitis using the New York criteria, 13 although the classic radiographic changes of bilateral sacroiliac disease may not be identified for several years after onset in children and are not required for a diagnosis of ERA. 1O· 11 The use of radiographic criteria poses a dilemma, because interpretation of sacroiliac radiographs in children and adolescents is difficult, even for the experienced pediatric radiologist. Nonetheless, characteristic radiographic changes in the sacroiliac joints, peripheral joints, and entheses aid in diagnosis and confirm the diagnostic impression (Table 13_1O).169-17I Ansell 37 observed that the mean interval from onset of symptoms to radiographic evidence of sacroiliac arthritis was 6.5
13
JUVENILE ANKYLOSING SPONDYLITIS
315
years (range, 1 to 15 years). However, in the report by Ladd and colleagues,29 sacroiliac joint arthritis was consistently the first radiographic abnormality of the axial skeleton, and diagnostic findings were confirmed at the onset of low back symptoms in the majority of children when technically adequate views of the sacroiliac joints were obtained. Radiographic techniques used to evaluate the spine and sacroiliac joints vary from institution to institution. 172 A single anteroposterior view of the pelvis may provide a screening examination and minimizes radiation exposure, although posteroanterior, stereoscopic, angulated (3D-degree), or oblique views are preferred by some pediatric radiologists for definitive evaluation. A systematic search for widening or narrowing of the sacroiliac joints, sclerosis on the iliac or sacral sides of the joints, and fusion of the sacroiliac joints is required to maximize the yield from this investigation. The apparent widening of the sacroiliac joint is the result of erosions of the subchondral border occurring particularly in the inferior synovial portion of the joint (Fig. 13-7). The lesion may appear initially as haziness of the cortical margins, followed by dissolution of the subchondral plate that results in a punchedout appearance. Although these changes may be uniiaterJ.1 initially, they eventually become bilateral and symmetric. An osteoblastic reaction occurring on both sides of the joint results in increased density or reactive sclerosis (Fig. 13-8). Late changes may include fusion of the sacroiliac joints and regional osteoporosis (Fig. 13-9). The sacroiliac joint has some unique anatomic characteristics, an understanding of which assists in the interpretation of certain radiologic features. The sacral side of the joint is covered by hyaline cartilage, whereas the iliac side is protected by a thin layer of fibrocartilage. These differences may account for the higher frequency of abnormalities on the iliac side. Only the lower one third to one half of each joint is diarthrodial and enclosed in a synovial membrane; the upper portion is a fibrous synostosis. 173 For purposes of documenting abnormalities of the sacroiliac joints in children and adolescents with suspected JAS, a plain anteroposterior radiograph of the pelvis is usually sufficient.
• • • TABI f 13-10 Radiologic Characteristics of Juvenile '
Saaolilae Disease (BIlateral) Diffuse osteoporosis of the pelvis Blurring of subchondral margins Erosions (iliac side first) Reactive sclerosis Joint-space narrowing Fusion (late)
Vertebral Column Vertebral epiphysitis with anterior vertebral squaring Anterior ligament calcification "Bamboo spine" EnthesItIs (e.g., at Calcaneus, nblal Tuberosity) Soft tissue swelling Erosions or spur formation at insertions
PerIpheral Joints Soft tissue swelling Accelerated ossification and epiphyseal overgrowth Periostitis Joint-space narrowing, erosions Ankylosis
• Figure 13-7 Juvenile ankylosing spondylitis. Early radiographic changes (angulated view). There is moderate reactive sclerosis of both sacroiliac joints with widening, irregularity, and haziness of the subchondral bone margins in this ll-year-old boy at onset of disease. (From ladd JR, Cassidy IT, Martel W: Juvenile ankylosing spondylitis. Arthritis Rheum 14: 579-590, 1971.)
316
C HAP T E R
13
JUVENILE ANK YLOSING SPONDYLITIS
• Figure 13-8 Juvenile ankylosing spondylitis. Moderately advanced radiographic changes. A, The widening, erosions, and reactive sderosis are marked on this anteroposterior view of the pelvis. B, Oblique view of right sacroiliac joint demonstrates the same changes in more detail.
Unless erosions are clearly demonstrable, a CT examination is then obtained. If uncertainty remains, magnetic resonance imaging (MRl) is the most appropriate investigation unless the changes are clearly unilateral, in which case a scintigraphic study may be of help. From the viewpoint of classification, only plain radiographs are used; the application of newer techniques to diagnosis and classification requires study. CT is a considerable advance over plain radiography for demonstrating abnormalities in the sacroiliac joints. In a study of 40 adults with sacroiliac disease, conventional radiography yielded abnormal results in 10, but CT documented abnormalities in 30 patients. CT was particularly useful in the early stages of the disease. 174 Tomograms at three levels through the synovial portion of the joint were sufficient to demonstrate all but the smallest erosions (Fig. 13-10). A CT technique described by Oudjhane and colieagues J75 may even be more efficient at demonstrating erosions and sclerosis of the sacroiliac joints.
• Figure 13-9 Juvenile ankylosing spondylitis. Advanced radiographic changes (angulated view). This film was obtained 14 years after onset of dinical disease in the patient shown in Rgure 13-7.The sacroiliac joints are virtually fused, especially on the left, but marked bilateral reactive sclerosis and erosions are still present on the right. (From Ladd JR, Cassidy JT, Martel W: Juvenile ankylosing spondylitis. Arthritis Rheum 14: 579-590, 1971.)
MRI identifies early changes in both the sacroiliac joints and the spine and may be the most sensitive indicator of inflammation. 176 It is critical in establishing an early diagnosis in specific patients and in stratifying therapy. In one large study,177 dynamic MRI was used to evaluate 100 children younger than 6 years of age with probable spondyloarthropathies (ESSG criteria) and 30 control children. Early sacroiliac joint inflammation was confirmed in many patients for whom other studies were unrevealing, particularly in children with acute onset of disease. The investigators pointed out that this technique should not be used in every child with back pain because of cost; nevertheless, demonstration of involvement of the sacroiliac joints could influence the choice of anti-inflammatory therapy. Scintigraphic study of the sacroiliac joint is of limited value in the growing child or adolescent, unless there are distinct unilateral abnormalities. Sufficient experience in the interpretation of radionuclide scans in this age group is required before interpretation of bone scans can be relied on, and even then the yield is limited. Increased uptake in one sacroiliac joint can result not only from inflammation or infection but from asymmetric weight-bearing caused by arthritis in a lower-extremity joint or from enthesitis around the foot.
• Figure 13-10 Coronal computed tomography of the sacroiliac joints. Bilateral sclerosis and erosions on the iliac sides of the joints are present, worse on the right than on the left.
C HAP T E R
13
JUVENILE ANKYLOSING SPONDYLITIS
317
Spine Radiologic changes in the lumbosacral spine are less frequent and occur much later than abnormalities in the sacroiliac joints. 29 ,17B Periostitis with deposition of new bone along the anterior margin of the vertebral border results first in the shining corner and then in flattening of the normally concave anterior margin of the vertebral body (Fig. 13-11). Syndesmophyte formation (Fig. 13-12), the hallmark of advanced disease in adults, is rare in children and adolescents but develops during the adult years in some patients with juvenile-onset disease. Periostitis at the iliac crests or the inferior pubic rami and erosion at the symphysis pubis are uncommon. Arthritis affecting the cervical spine is less commonly symptomatic than that of the lumbosacral spine but can cause severe damage,179 This is documented by the MRI study shown in Figure 13-13A-G.
Entheses Radiographic evaluation of entheses around the calcaneus, and rarely the patella, may demonstrate subtle changes in soft tissue denSity. Loss of the distinct margins at the insertion of the Achilles tendon, together with effacement of the triangular fat shadow, may be an early sign of inflammation. Erosion of bone at the insertion of the Achilles tendon or spur formation at that site is readily evaluated by a lateral radiograph of the calcaneus (Fig. 13-14), Azouz and Duffy170 have described changes
• Figure 13-12 Radiographs of the lumbar spine. A, Syndesmophytes (arrows). B, Calcification of the anulus fibrosus (arrows) and anterior squaring. The bamboo-like appearance of the spine indicates long-standing juvenile ankylosing spondylitis and is a rare finding in childhood. These films were obtained from a 36-year-old man with onset of disease at the age of 14 years. (A and B, From Ladd JR, Cassidy JT, Martel W: Juvenile ankylosing spondylitis. Arthritis Rheum 14: 579-590, 1971.)
in the bone marrow subjacent to an inflamed enthesis in children with ]AS.
Peripheral Joints Radiographic changes of peripheral joints are characterized by juxta-articular osteoporosis and joint-space narrowing, especially at the hips, knees, and ankle joints. Erosions such as those that develop in children with]RA are exceptional. A characteristic ring osteophyte may be evident in the femur (Fig. 13-15). Ankylosis may occur but is not common in childhood, except at the intertarsal joints. Tarsitis is characteristic of ]AS. IBO
TREATMENT General Management
• Figure 13-11 Marked straightening of the lumbar spine on a lateral film in early juvenile ankylosing spondylitis.
Children with ]AS have frequently had undiagnosed symptoms for months or years or have been misdiagnosed as having]RA or other disorders. A team of health professionals helps to provide necessary education and care. Explanation of the correct diagnosis, its chronicity, possible complications, and the need for long-term medical treatment, physical therapy, and follow-up facilitates compliance with the therapeutic recommendations. In the
318
C HAP T E R
13
JUVENI LE ANKYLOSING SPONDYLITIS
• Figure 13-13 Radiographs of the upper cervical spine in a child with juvenile ankyloslng spondylitis. A, Extension view identifies the upper limit of the odontoid process of Q (upper arrow) and the posteroinferior margin of the anterior arch of C1 (lower arrow). 8, Flexion view documents a widened atlantoaxial space (arrow), confirming the presence of subluxation. C, T2-weighted magnetic resonance image delineates an inflammatory mass In the space between the C1 anterior arch and the odontoid process (lower arrow), as well as indentation of the odontoid on the lower medulla/upper cervical cord (upper arrow).
older adolescent, realistic career goals must be discussed along with current means of minimizing work-related stress to the low back and joints of the lower extremities. The patient should be encouraged to participate as fully as possible in age-appropriate social and recre-
• Figure 13-14 Lateral radiographs of the calcaneus with erosions at the site of insertion of the plantar fasda into the calcaneus (A, arrowheads) and spur formation at the insertion of the plantar fasda into the calcaneus (8, arrowhead). (A, From Petty RE, Malleson P: Spondyloarthropathies of childhood. Pediatr elin North Am 33: 1079-1096, 1986.)
ational activities. The relatively good long-term prognosis should be emphasized, ,particularly in the adolescent, who may regard the diagnosis as marking the end of recreational, social, educational, and career goals. Management should be individualized according to the patient's specific problems. Current treatments is most successful in controlling the signs and symptoms of the disease; none has been demonstrated to alter the progression of ankylosis. If widespread, severe joint inflammation is the overwhelming problem, systemic anti-inflammatory medications (NSAIDs, sulfasalazine, glucocorticoids) are appropriate. If the joint disease is localized, it may be more useful to use NSAIDs and intra-articular glucocorticoid in joints that are particularly problematic. Enthesitis, particularly around the foot, is unlikely to respond to systemic anti-inflammatory dmgs alone; the use of custom-made orthotics often provides relief. In all patients, exercise and maintenance of good posture to minimize loss of range of motion (ROM) in the spine and its articulations are recommended. A firm mattress and thin pillow are important adjuncts to the program. Smoking should be discouraged because of its demonstrated adverse effects on pulmonary function. lSI
• Figure 13-15 Radiograph of the hips of an adolescent boy with ankylosing spondylitis demonstrates bilateral rim osteophytes at the junction of the femoral head and neck.
C HAP T E R
13
JUVENILE ANKYLOSING SPONDYLITIS
319
Anti-Inflammatory Medications
Physical and Occupational Therapy
NSAIDs form the basis of the pharmacologic management. Although there are no reported trials of NSAIDs in ]AS, it is probable that these children respond best to naproxen, indomethacin, or tolmetin. Because of lower toxicity, the use of naproxen OS to 20 mg/kg/day) is recommended before indomethacin 0 to 2 mg/kg/day). Although indomethacin is often effective, toxicity is common, and the drug must be monitored carefully, beginning with low doses. Headache, epigastric pain, and inability to pay attention in school occur in 20% to 30% of children taking this drug and frequently necessitate cessation of its use (see Chapter 5 for warning on a potential risk of cardiovascular events). Sulfasalazine has widespread currency in treatment, 182 based largely on experience in adults,183-189 although no prospective, randomized, placebo-controlled trials have been reported. In an open-label, multicenter study, children with B27-positive late-onset oligoarthritis responded well to sulfasalazine. 190 This group of patients were those whose peripheral disease responded best in other small trials as well.191.192 The response rate to sulfasalazine (40 to 50 mg/kg/day: upper limit 2000 mg) was as high in patients with the spondyloarthropathies as in those with ]RA, but the duration of response was shorter. 193 Beneficial effects are usually not evident for several weeks after initiation of treatment. Toxicity to bone marrow and liver must be monitored closely. Glucocorticoids have a role only in short-term therapy in the severely ill patient,194 as topical agents in the management of acute iritis, and for intra-articular administration in children with limited but severe joint disease.195.196 The use of CT to guide injection of 40 mg of triamcinolone into the sacroiliac joints was reported in 30 adults with AS. 197 The injections were considered effective as judged both clinically and by dynamic MRI. Local injections of glucocorticoids at sites of enthesitis may occasionally be useful, particularly at the plantar fascia--ealcaneal junction, taking care to avoid direct injection into tendon or extravasation into subcutaneous tissue. There is little reported experience with other secondline agents. In general, gold compounds and o-penicillamine are not recommended. Hydroxychloroquine and methotrexate are sometimes used, but there are no reports of their efficacy in children. In a study of adults who had not responded to NSAIDs and sulfasalazine, modest benefit was demonstrated with the use of methotrexate in a dose of 7.5 to 15 mg/week. 198, 199 There are no reports of the use of immunosuppressive drugs in children or adults. In a single patient treated with azathioprine, severe pancytopenia developed because of an accompanying and presumably coincidental mutation of the thiopurine methyltransferase gene. 2oa Etanercept has been advocated for refractory disease.201.202 In a controlled trial in AS, it significantly improved signs and symptoms of the disease. 203 Infliximab in another trial produced a 50% improvement in 53% of the patients at week 12. 204 Pamidronate (60 mg IV/mo in adults) impressively reduced bone turnover but did not alter inflammatory indices. 205
Physical therapy should be directed at preventing loss of ROM and poor functional positioning in the spine and chest, as well as stabilizing or regaining lost ROM in peripheral joints. Attention to posture and daily active ROM exercises for the back and deep-breathing exercises for the chest help to preserve range. Some young patients with ]AS breathe predominantly with the diaphragm and have to be retaught to use the intercostal muscles. Strengthening of abdominal and back muscles should be undertaken cautiously. Swimming is an ideal form of physical activity that can be encouraged to augment these specific exercises. Painful enthesitis in the feet may be relieved by the use of custom-made orthotics, fitted to support the fat cushion under the heel and to take pressure off the plantar aspects of the heel and metatarsophalangeal joints. If the Achilles enthesis alone is involved, the use of a slightly higher heel may help to reduce stress at this site. Therapeutic ultrasonography and transcutaneous nerve stimulation are sometimes useful in the management of pain caused by enthesitis around the foot. Enthesitis can be quite resistant to therapy and may be the most functionally limiting aspect of the disease.
Surgery Orthopedic surgery has a limited role in management. Later in life, joint reconstruction and replacement are invaluable contributions to function and quality of life in the patient with severely damaged joints. The outcome of total hip replacement in young adults who were monitored for up to 30 years indicates that the probability that both components of low-friction arthroplasty will survive 10 years is 91%; this figure was 70% at 20 years.206.207 Bony ankylosis as a result of exuberant overgrowth of bone around the prosthesis has been reported after hip joint replacement as an almost unique complication related to this disease. This complication may be amenable to prophylactic use of medicationY
COURSE OF THE DISEASE AND PROGNOSIS The early course of ]AS is often remitting and may be mild. Often, it is only in retrospect that the initial musculoskeletal complaints are recognized as harbingers of this disorder. Almost half of these children have 4 or fewer joints affected dUring the entire course of the disease; even in those in whom this number is exceeded, it is uncommon cumulatively to have more than 6 or 7 inflamed joints. With few exceptions, the children eventually develop peripheral joint disease, if they do not have it at onset. Lower-extremity predominance remains the rule throughout the course, with hips, knees, ankles, and feet more commonly affected than upper-extremity joints. Enthesitis may increase in frequency of involved sites during the course of the disease. 208 Subtle losses in ROM of the thorax or back should be documented as early as possible. Anse1l 37 noted that
320
C HAP T E R
13
JU V E NIL E
AN K Y LOS I N G
SP 0
limitation of ROM of the spine was not detected until 11 to 33 years after onset of symptoms. A similar delay was also reported in the series of Ladd and colleagues. 29 However, from more recent experience, it is an impression that limitation of range of back motion may occur much earlier. In the study of Burgos-Vargas and colleagues,14 all patients had decreased back mobility by 5 years. Accurate data on long-term prognosis are very limited. 209-2I1 Outcome measures for adults with AS have been evaluated. 212- 215 A number of indices (Ankylosing Spondylitis Scales, Assessment in Ankylosing Spondylitis 34) have been developed as measures of outcome. Life expectancy in AS is reduced compared to the US population. Cardiopulmonary and cerebrovascular disease are the leading causes of death. At least during the years of childhood and adolescence, functional outcome probably remains good. 44 ,216 In one study,217 however, outcome in ]AS was worse than in AS. Peripheral joint disease may be more common in children than in adults, and persistent hip disease, in particular, is associated with a poor functional outcome. 208 ,218 Acute iritis seldom leaves significant residua, even if recurrent, but uncommonly can be severe. Aortitis is rare but, if present, contributes to late morbidity and mortality. Although amyloidosis may develop in adults with AS, little information is available in children. 37
REFERENCES 1. Moran HM. Hall MA, Barr A, et al: Spinal mobility in the adolescent. Rheumatol Rehabil 18: 181-185, 1979. 2. Goie The HS, Steven MM, van der Linden SM, Cats A: Evaluation of diagnostic criteria for ankylosing spondylitis: a comparison of the Rome, New York and modified New York criteria in patients with a positive clinical history screening test for ankylosing spondylitis. Br J Rheumatol 24: 242-249, 1985. 3. Petty RE, Malleson P: Spondyloarthropathies of childhood. Pediatr Clin North Am 33: 1079-1096, 1986. 4. Burgos-Vargas R, Petty RE: Juvenile ankylosing spondylitis. Rheum Dis Clin North Am 18: 123-142, 1992. 5. Rosenberg AM: Juvenile onset spondyloarthropathies. Curr Opin Rheumatol 12: 425-429, 2000. 6. Pepmueller PH, Moore TL: Juvenile spondyloarthropathies. Curr Opin Rheumatol 12: 269-273, 2000. 7. Wright V, Moll JM: Seronegative Polyarthritis. Amsterdam, The Netherlands, North Holland, 1976. 8. Rosenberg AM, Petty RE: A syndrome of seronegative enthesopathy and arthropathy in children. Arthritis Rheum 25: 1041-1047, 1982. 9. Cabral DA, Oen KG, Petty RE: SEA syndrome revisited: a longterm followup of children witb a syndrome of seronegative enthesopathy and arthropathy. J Rheumatol 19: 1282-1285, 1992. 10. 'Fink CW: Proposal for the development of classification criteria for idiopathic arthritides of childhood. J Rheumatol 22: 1566--1569. 1995. II. Perry RE, Southwood TR, Baum J, et al: Revision of the proposed classification criteria for juvenile idiopathic arthritis: Durban, 1997. J Rheumatol 25: 1991-1994, 1998. 12. Petty RE, Southwood TR, Manners P, et al: International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: Second Revision, Edmonton 2001. J Rheumatol 31: 390--392, 2004. 13, Bennett PH, Wood PHN: Population Studies of the Rheumatic Diseases. New York, Excerpta Medica, 1968. 14, BurgOS-Vargas R, Clark P: Axial involvement in the seronegative enthesopathy and arthropathy syndrome and its progression to ankylosing spondylitis. J Rheumatol 16: 192-197, 1989. 15. Jacobs JC, Berdon WE, Johnston AD: HLA-B27-associated spoodyloarthritis and enthesopathy in childhood: clinical, pathologic, and radiographic observations in 58 patients. J Pediatr 100: 521-528, 1982. 16. Cassidy JT, Levinson JE, Bass JC, et al: A study of classification criteria for a diagnosis of juvenile rheumatoid arthritis, Arthritis Rheum 29: 274-281,1986. 17. Sheerin KA, Giannini EH, Brewer EJ Jr, et al: HLA-B27-associated arthropathy in childhood: long-term clinical and diagnostic outcome, Arthritis Rheum 31: 1165-1170, 1988.
N 0 Y LI TI S 18. Hussein A, Abdul-Khaliq H, von der HH: Atypical spondyloarthritis in children: proposed diagnostic criteria. Eur J Pediatr 148: 513-517, 1989. 19. Dougados M, van der LS, Juhlin R, et al: The European Spondylarthropathy Study Group preliminary criteria for the classification of spondylarthropathy. Arthritis Rheum 34: 1218-1227, 1991. 20. van der LS, Valkenburg HA, Cats A: Evaluation of diagnostic criteria for ankylosing spondylitis. A proposal for modification of the New York criteria. Arthritis Rheum 27: 361-368, 1984. 21. Amor B, Dougados M, Mijiyawa M: Criteria of the classification of spondylarthropathies. Rev Rhum Mal Osteoartic 57: 85-89, 1990. 22. Prieur AM, Listrat V, Dougados M, et al: Criteria for classification of spondylarthropathies in children. Arch Fr Pediatr 50: 379-385, 1993. 23. Bowyer S, Roettcher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group, J Rheumatol 23: 1968-1974, 1996. 24. Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry. J Rheumatol 23: 1981-1987, 1996. 25. Symmons DP, Jones M, Osborne J, et al: Pediatric rheumatology in the United Kingdom: data from the British Pediatric Rheumatology Group National Diagnostic Register. J Rheumatol 23: 1975-1980, 1996 26. Andersson GB, Fasth A, Andersson.l, et al: Incidence and prevalence of juvenile chronic arthritis: a population survey. Ann Rheum Dis 46: 277-281, 1987. 27, Kaipiainen-Seppanen 0, Savolainen A: Incidence of chronic juvenile rheumatic diseases in Finland during 1980--1990. Clin Exp Rheumatol 14: 441--444, 1996. 28. DoU!'ado OM, Favero S, Baranauskas V, et al: Effects of the Ga-As laser irradiation on myonecrosis caused by Botbrops moojenl snake venom. La.,ers Surg Med 33: 352-357, 2003. 29, Ladd JR, Cassidy JT, Martel W: Juvenile ankylosing spondylitis. Arthritis Rheum 14: 579-590, 1971. 30. Gomez KS, Raza K, Jones SO, et al: Juvenile onset ankylosing spondylitismore girls than we thought? J Rheumatol 24: 73S--737, 1997. 31. Braun J, Bollow M, Remlinger G, et al: Prevalence of spondylarthl'Opathies in HLA-B27 positive and negative blood donors. Arthritis Rheum 41: 58-67, 1998. 32. Khan MA: Update on spondyloarthropathies. Ann Intern Med 136: 896--907. 2002. 33. Carter ET, McKenna CH, Brian DO, et al: Epidemiology of ankylosing spondylitis in Rochester, Minnesota, 1935-1973- Arthritis Rheum 22: 365-370, 1979. 34. Carbone LD, Cooper C, Michet C.f, et al: Ankylosing spondylitis in Rochester. Minnesota, 1935-1989. Is the epidemiology changing? Arthritis Rheum 35: 1476--1482, 1992. 35. Calin A: The epidemiology of ankylosing spondylitiS: a clinician's point of view. In Lawrence RC, Shulman LE (eds): Epidemiology of the Rheumatic Diseases. New York, Gower, 1984, p 51. 36. Edstrom G, Thune S, Wittbom-Cigen G: Juvenile ankyloslng spondylitis. Acta RheumalOl Scand 6: 161-173, 1960, 37. Ansell BM: Juvenile spondylitis and related disorders. In Moll JMH (ed): Ankyloslng Spondylitis. Edinburgh, Churchill Livingstone, 1980, p 120. 38. Schaller J, Bitnum S, Wedgwood l\J: Ankylosing spondylitis with childhood onset..1 Pediatr 74: 505-516, 1969. 39. Bywaters EGL: Ankylosing spondylitis of childhood onset. Clin Rheum Dis 2: 387, 1976. 40. Sturrock RD, Dick HM, Henderson N: Association of HLA 27 in juvenile rheumatoid arthritis and ankylosing spondylitis. J Rheumatol 1: 269, 1974. 41. Edmonds J, Morris RI, Metzger AL, et al: Follow-up study of juvenile chronic polyarthritis With particular reference to histocompatibility antigen W. 27. Ann Rheum Dis 33: 289-292, 1974. 42. Veys EM, Colgne E, Mielants H, et al: HLA and juvenile chronic polyarthritis. Tissue Antigens 8: 61--65, 1976. 43. Schaller JG: Ankylosing spondylitis of childhood onset. Arthritis Rheum 20, 398--401, 1977. 44. Hafner R: Juvenile spondarthl'itls. Retrospective study of 71 patients. Monatsschr Kinderheilkd 135: 41--46, 1987. 45. Calin A, Fries JF: Striking prevalence of ankylosing spondylitis in "healthy" w27 positive males and females. N Engl J Med 293: 835-839, 1975. 46. Masi AT: HLA B27 and other host interactions in spondyloarthropathy syndromes. J Rheumatol 5: 359-362, 1978. 47, Resnick 0, Dwosh IL, Goergen TG, et al: Clinical and radiographic abnormalities in ankylosing spondylitis: a comparison of men and women. Radiology 119: 293-297, 1976. 48. Zeng QY: Ankylosing spondylitis in Shantou, China: 15 years' clinical experience. J Rheumatol 30: 1816--1821, 2003. 49. Baum J, Ziff M: The rarity of ankylosing spondylitis in the black race. Arthritis Rheum 14: 12-18, 1971. 50. Sonozaki H, Sekl H, Chang S, et at Human lymphocyte antigen, HL-AD, in Japanese patients with ankylosing spondylitis. Tissue Antigens 5: 131-136, 1975. 51. Gofton JP, Robinson HS, Trueman GE: Ankylosing spondylitis In a Canadian Indian population. Ann Rheum Dis 25: 525-527, 1966. 52. Good AE, Kawanishi H, Schultz .IS: HLA B27 in blacks with ankylosing spondylitis or Reiter's disease [letter]. N Engl J Med 294: 166--167, 1976.
C HAP T E R 53. Masi AT, Walsh EG: Ankylosing spondylitis: integrated clinical and physiological perspectives. Clin Exp Rheumatol 21: 1--8, 2003, 54. Keat A: Infections and the immunopathogenesis of seronegative spondyloarthropathies. Curr Opin Rheumatol 4: 494-499, 1992, 55. Pacheco-Tena C, Alvarado DLB, Lopez-Vidal Y, et al: Bacterial DNA in synovial l1uid cells of patients with juvenile onset spondyloarthropathies. Rheumatology (OxD 40: 920-927, 2001. 56. Dominguez-Lopez ML, Cancino-Diaz ME, Jimenez-Zamudio L, et al: Cellular immune response to Klebsiella pneumonlae antigens in patients with HLAB27+ ankylosing spondylitis. J Rheumatol 27: 1453-1460, 2000. 57, Tiwana H, Natt RS, Benitez-Brito R, et al: Correlation between the immune responses to collagens type I, III, IV and V and KlebSiella pneumonlae in patients witb Crohn's disease and ankylosing spondylitis. Rheumatology (Oxf) 40: 15-23, 2001. 58. Geczy AF, Seager K, Bashir HV, et al: The role of KlebSiella in the pathogenesis of ankylosing spondylitis. II. EVidence for a specific B27-associated marker on the lymphocytes of patients with ankylosing spondylitis. J Clin Lab Immunol 3: 23-28, 1980. 59. Cameron FH, Russell PJ, Easter JF, et al: Failure of Klebsiella pneumonlae antibodies to cross-react with peripheral blood mononuclear cells Ii-om patients with ankylosing spondylitis, Arthritis Rheum 30: 300-305, 1987, 60. Mielants H, Veys EM, Goemaere S, et al: A prospective study of patients with spondyloarthropathy with special reference to HLA-B27 and to gut histology. J Rheumatol 20: 1353-1358, 1993. 61, Mielanr., H, Veys EM, Cuvelier C, et al: The evolution of spondyloanhropathies in relation to gut histology. III. Relation between gut and joint. J Rheumatol 22: 2279-2284, 1995. 62. El Maghraoui A, Dougados M, Freneaux E, et al: Concordance between abdominal scintigraphy using technetium-99m hexamethylpropylene amine oxime-labelled leucocytes and ileocolonoscopy in patients with spondyloarthropathies and without clinical eVidence of inl1ammatory bowel disease. Rheumatology (OxD 38: 543-546, 1999. 63. Mielants H, Veys EM, Joos R, et al: HLA antigens in seronegative spondylarthropathies: reactive arthritis and arthritis in ankylosing spondylitis. Relation to gut inl1ammation. J Rheumatol 14: 466-471, 1987. 64. Jobanputra P, Choy EH, Kingsley GH, et al: Cellular immunity to cartilage proteoglycans: relevance to the pathogenesis of ankylosing spondylitis, Ann Rheum m, 51: 959-%2, 1992. 65. Ford DK, da Roza D, Schulzer M: Lymphocytes from the site of disease but not blood lymphocytes indicate the cause of arthritis. Ann Rheum Dis 44: 701-710, 1985. 66. Kapasi K, Inman RD: HLA-B27 expression modulates gram-negative bacterial invasion into transfected L cells, J Immunol 148: 3554-3559, 1992. 67, Kapasi K, Inman RD: MEl epitope of HLA-B27 confers class I-mediated modulation of gram-negative bacterial invasion, J Immunol 153: 833--840, 1994. 68. Ortiz-Alvarez 0, Yu DT, Petty RE, et al: HLA-B27 does not affect invasion of arthritogenic bacteria into human cells. J Rheumatol 25: 1765-1771, 1998, 69. Huppertz HI, Heesemann J: Invasion and persistence of Salmonella in human fibroblasts positive or negative for endogenous HLA B27, Ann Rheum Dis 56: 671--676, 1997, 70, Granfors K: Host-microbe interaction in HLA-B27-associated diseases, Ann Med 29: 153-157, 1997. 71. McGonagle D, Conaghan PG, Emery P: Psoriatic arthritis: a unified concept twenty years on. Arthritis Rheum 42: 108D-I086, 1999, 72. Braun J, Bollow M, Neure L, et al: Use of immunohistologic and in situ hybridization techniques in the examination of sacroiliac joint biopsy specimens from patients with ankylosing spondylitis, Arthritis Rheum 38: 499-505, 1995. 73. Potocnik AJ, Menninger H, Yang SY, et al: Expression of the CD2 activation epitope TlI-3 (CD2R) on T cells in rheumatoid arthritis, juvenile rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, and Lyme disease: phenotypic and functional analYSiS, Scand J Immunol 34: 351-358, 1991. 74, Grom AA, Murray KJ, Luyrink L, et al: Patterns of expression of Ulmor necrosis factor alpha, tumor necrosis factor beta, and their receptors in synovia of patients with juvenile rheumatoid arthritis and juvenile spondylarthropathy, Arthritis Rheum 39: 1703-1710, 1996. 75. Hammer RE, Maika SD, Richardson JA, et al: Spontaneous inl1anunatOly disease in transgenic rats expressing HLA-B27 and human beta 2m: an animal model of HLA-B27-associated human disorders. Cell 63: 1099-1112, 1990. 76. Taurog JD: Immunology, genetics, and animal models of the spondyloarthropathies. Curr Opin Rheumatol 2: 586-591, 1990. 77. Breban M, Hammer RE, Richardson JA, et al: Transfer of the inl1ammatory dL,ease of HLA- B27 transgenic rats by bone marrow engraftment. J Exp Med 178: 1607-1616. 1993. 78. Mear JP, Schreiber KL, Munz C, et al: Misfolding of HLA-B27 as a result of its B pocket suggest, a novel mechanism for its role in susceptibility to spondyloarthropathies. J Immunol 163: 6665--6670, 1999. 79, Cooper GL, Charlton BR: Spondylitis in turkeys associated with experimental Pasteurella anatlpest!fer infection. Avian Dis 36: 290-295, 1992. 80. Brophy S, Calin A: Ankylosing spondylitis: interaction between genes, joints, age at onset, and disease expression. J Rheumatol 28: 2283-2288, 2001. 81, Ansell BM, Bywaters EG, LawrenceJS: Familial aggregation and twin studies in Still's disease. Juvenile chronic polyarthritis. Rheumatology 2: 37--61, 1969.
13
JUVENILE ANKYLOSING SPONDYLITIS
321
82. Ruhin LA, Amos CI, Wade JA, et al: Investigating the genetic basis for ankylosing spondylitis. Linkage studies with the major histocompatibility complex region, Arthritis Rheum 37: 1212-1220, 1994. 83. Brewelton DA, Hart PD, Nicholls A, et al: Ankylosing spondylitis and HL-A 27. Lancet 1: 904-907, 1973. 84. van der Linden SM, Valkenburg HA, de Jongh BM, et al: The risk of developing ankylosing spondylitis in HLA-B27 positive individuals. A comparison of relatives of spondylitis patients with the general population. Arthritis Rheum 27: 241-249, 1984. 85, Liu Y, Li .J, Chen B, et al: Familial aggregation of ankylosing spondylitis in Southern China. J Rheumarol 28: 550-553, 2001, 86. Baudoin P, van der Horst-Bruinsma IE, Dekker-Saeys AJ, et al: Increased risk of developing ankylosing spondylitis among first-born children. Arthritis Rheum 43: 2818-2822, 2000. 87, van Santen M, Bolwijn P, Versrappen F, et al: A randomized clinical trial comparing fitness and biofeedback training versus basic treatment in patient~ with fibromyalgia, J Rheumatol 29: 575-581, 2002. 88, Isogai Z, Tsuji T, Kurita K, et al: HLA-B27-positive familial case of a daughter with juvenile pustular psoriasis and her father with ankylosing spondylitis. Pediatr Dermatol 16: 491-493, 1999. 89. Schlosstein L, Terasaki PI, Bluestone R, et al: High association of an HL-A antigen, W27, with ankylosing spondylitis, N EnglJ Med 288: 704-706,1973. 90. McDaniel DO, Barger BO, Reveille]D, et al: Analysis of restriction fragment length polymorphisms in rheumatic diseases. Rheum Dis Clin North Am 13: 353-367, 1987. 91. Khan MA: Spondyloarthropathies. Curr Opin Rheumatol 10: 279-281, 1998. 92. Mougin B, Garnem P, Borel 0, et al: A routine assay for the direct analysis of HLA-DR-related shared epitope and B27 alleles in chronic inl1ammatory arthritis. J Immunol Methods 256: 47-53, 2001. 93. Maclean L: HLA-B27 subtypes: implications for the spondyloarthropathies. Ann Rheum Dis 51: 929-931, 1992. 94. Hill AV, Allsopp CE, Kwiatkowski D, et al: HLA dass I typing by PCR: HLAB27 and an African B27 subtype, Lancet 337: 640--642, 1991. 95. Kvien TK, Moller P, Dale K: Juvenile ankylosing spondylitis and HLA B27 homozygosity. Scand J Rheumatol 14: 47-50, 1985. 96. Hamersma J, Cardon LR, Bradbury L, et al: Is disease severity in ankylosing spondylitis genetically determined? Arthritis Rheum 44: 1396-1400, 2001. 97. Haywood KL, Garratt AM, Dziedzic K, et al: Generic measures of healthrelated quality of life in ankylosing spondylitis: reliability, validity and responsiveness. Rheumatology (OxD 41: 1380-1387, 2002. 98. Brown MA, Kennedy LG, MacGregor AJ, et al: Susceptibility to ankylosing spondylitis in twins: the role of genes, HLA, and the environment, Arthritis Rheum 40: 1823-1828, 1997. 99, Oen K, Petty RE, Schroeder ML: An association between HLA-A2 and juvenile rheumatoid arthritis in girls. J Rheumatol 9: 916-920, 1982. 100. Woodrow JC: Genetics. In Moll JMH (ed): Ankylosing Spondylitis. Edinburgh, Churchill Livingstone, 1980, p 26. 101, Arnett FH Jr, Schacter BZ, Hochberg MC, et aI: HLA-A28 in patients with B27associated rheumatic diseases. Arthritis Rheum 20: 106, 1977, 102. Calin A, Porta J, Payne R: HLA-A28 in B27 positive controls and patients with ankylosing spondylitis. Arthritis Rheum 20: 1428, 1977. 103, Robinson WP, van der Linden SM, Khan MA, et al: HLA-Bw60 increases susceptibility to ankylosing spondylitis in HLA-B27+ patients. Arthritis Rheum 32: 1135-1141, 1989. 104. Brown MA, Pile KD, Kennedy LG, et al: HLA dass I association.' of ankylosing spondylitis in the white population in the United Kingdom. Ann Rheum Dis 55: 268-270, 1996. 105. Truog P, Steiger U, Contu I, et al: Ankylosing spondylitis (AS): a population and family study using HL-A serology and MLR. In Kissmeyer-Nielsen F (ed): Histocompatibility Testing, Copenhagen, Munksgaard, 1975, p. 788, 106. van den Berg-Loonen EM, Dekker-Saeys BJ, Meuwissen SG, et al: Histocompatibility antigens and other genetic markers in ankylosing spondylitis and inl1ammatory bowel diseases, J Immunogenet 4: 167-175, 1977. 107, McGarry F, Walker R, Sturrock R, et al: The -308.1 polymorphism in the promoter region of the tumor necrosis factor gene is associated with ankylosing spondylitis independent of HLA-B27, J Rheumatol 26: 1110-1116, 1999. 108. McGarry F, Neilly J, Anderson N, et al: A polymorphism within the interleukin 1 receptor antagonist (IL-IRa) gene is associated with ankylosing spondylitis. Rheumatology (Oxf) 40: 1359-1364, 2001. 109. Maksymowych WP, Gorodezky C, Olivo A, et al: HLA-DRBl'08 influences the development of disease in Mexican Mestizo With spondyloarthropathy. J Rheumarol 24: 904-907, 1997. 110. Maksymowych WP, Jhangri GS, Gorodezky C, et al: The LMP2 polymorphism is associated with susceptibility to acute anterior uveitis in HLA-B27 positive juvenile and adult Mexican subjects with ankylosing spondylitis. Ann Rheum Dis 56: 488-492, 1997. 111. Ploski R, Flaro B, Vinje 0, et al: Association to HLA-DRB1'08, HLADPBl'0301 and homozygosity for an HLA-linked proteasome gene in juvenile ankylosing spondylitis, Hum Immunol 44: 88-96, 1995. 112. Turner PG, Green ]H, Galasko CS: Back pain in childhood. Spine 14: 812--814, 1989. 113. Boyer GS, Templin DW, Bowler A, et al: Spondyloarthropathy in the community: clinical syndromes and disease manifestations in Alaskan Eskimo populations, J Rheumarol 26: 1537-1544, 1999.
322
CHAPTER
13
JUVENILE ANKYLOSING SPONDYLITIS
114, Baek HJ, Shin KC, Lee YJ, et al: Juvenile onset ankylosing spondylitis (lAS) has less severe spinal disease course than adult onset ankylosing spondylitis (AAS): clinical comparison between JAS and AAS in Korea, J Rheumatol 29: 1780-1785, 2002, 115, Sherry DO, Sapp LR: Enthesalgia in childhood: site-specific tenderness in healthy subjects and in patients with seronegative enthesopathic arthropathy, J Rheumatol 30: 1335-1340, 2003, 116, Niepel GA, Sit'aj S: Enthesopathy, Clin Rheum Dis 5: 857, 1979, 117, Burgos-Vargas R, Pacheco-Tena C, Vazquez-Mellado J: A short-term followup of enthesitis and arthritis in the active phase of juvenile onset spondyloarthropathies, Clin Exp Rheumatol 20: 727-731, 2002, 118, JimeneZ-Balderas FJ, Fernandez-DiezJ, Fraga A: Tamale foot: deposit of acid mucopolysaccharides in the synovial sheaths of extensor tendons of the foot, resembling tendinitis, in a patient with juvenile ankylosing spondylitis, J Rheumatol 27: 1788-1791, 2000, 119, Kleinman P, Rivelis M, Schneider R, et al: Juvenile ankylosing spondylitis, Radiology 125: 775-780, 1977, 120, Burgos-Vargas R, Vazquez-Mellado J, Cassis N, et al: Genuine ankylosing spondylitis in children: a case-control study of patients with early definite disease according to adult onset criteria, J Rheumatol 23: 2140-2147, 1996, 121. Petty RE: Is ankylosing spondylitis in childhood a distinct entity? J Rheumatol 23: 2013-2014, 1996, 122, Monnet 0, Breban M, Hudry C, et al: Ophthalmic findings and frequency of extraocular manifestations in patients with HLA-B27 uveitis: a study of 175 cases, Ophthalmology 111: 802-809, 2004, 123, Liao CH, Huang JL: Juvenile ankylosing spondylitis with uveitis, Asian Pac J Allergy 1mmunol 21: 69-71, 2003, 124, Huppertz H, Voigt [, Muller-Scholden J, et al: Cardiac manifestations in patients with HLA B27-associated juvenile arthritis, Pediatr Cardiol 21: 141-147, 2000, 125, Stewart SR, Robbins DL, Castles JJ: Acute fulminant aortic and mitral insufficiency in ankylosing spondylitis, N Engl J Med 299: 1448-1449, 1978. 126, Reid GO, Patterson MW, Patterson AC, et al: Aortic insufficiency in association with juvenile ankylosing spondylitis, J Pediatr 95: 78-80, 1979, 127, Kean WF, Anastassiades TP, Ford PM: Aortic incompetence in HLA B27-positive juvenile arthritis, Ann Rheum Dis 39: 294-295, 1980, 128, Gore JE, Vizcarrondo FE, Rieffel CN: Juvenile ankylosing spondylitis and aortic regurgitation: a case presentation, Pediatrics 68: 423-426, 1981. 129, Pelkonen P, Byring R, Pesonen E, et al: Rapidly progressive aortic incompetence in juvenile ankylosing spondylitis: a case repcllt, Arthritis Rheum 27: 698-700, 1984. 130, Kin1 TH, Jung SS, Sohn SJ, et al: Aneurysmal dilatation of ascending aorta and aonic insufficiency in juvenile spondyloarthropathy, Scand J Rheumatol 26: 218-221, 1997, 131. Simpson J, Borzy MS, Silberbach GM: Aortic regurgitation at diagnosis of HLA-B27 associated spondyloarthropathy, J Rheumatol 22: 332-334, 1995, 132, Hubscher 0, Susini J: Aortic insufficiency in Reiter's syndrome of juvenile onset. J Rheumatol 11: 94-95, 1984, 133, Johnson WL, Toone E: The clinical and pathological cardiac manifestations of rheumatoid spondylitis, Va Med Mon (918) 95: 132-140, 1968, 134, Lee SJ, [m HY, Schueller WC: HLA-B27 positive juvenile arthritis with cardiac involvement preceding sacroiliac joint changes, Heart 86: E19, 2001. 135, Stamato T, Laxer RM, de Freitas C, et al: Prevalence of cardiac manifestations of juvenile ankylosing spondylitis, Am J Cardio1 75: 744-746, 1995, 136, Roldan CA, Chavez J, Wiest PW, et al: Aortic root disease and valve disease associated with ankylosing spondylitis, J Am Coil Cardiol 32: 1397-1404, 1998. 137, Camiciottoli G, Trapani S, Ermini M, et al: Pulmonary function in children affected by juvenile spondyloarthropathy, J Rheumatol 26: 1382-1386, 1999, 158. Rosenow E, Strimlan CV, Muhm JR, et al: Pleuropulmonary manifestations of ankylosing spondylltis, Mayo Clin Proc 52: 641-649, 1977, 139, Fenlon HM. Casserly I, Sant SM, et al: Plain radiographs and thoracic highresolution CT in patients with ankylosing spondylitis, AJR Am J Roentgenol 168: 1067-1072, 1997, 140, Reid GO, Hill RH: Atlantoaxial subluxation in juvenile ankylosing spondylitis, J Pediatr 93: 551-532, 1978. 141. Foster HE, Cairns RA, Burnell RH, et al: Atlantoaxial subluxation in children with seronegative enthesopathy and arthropathy syndrome: 2 case reports and a review of the literature, J RheumalOl 22: 548-551, 1995, 142, BanlesonJD, Cohen MD, Harrington TM, et al: Cauda equina syndrome secondary to long-standing ankylosing spondylitis, Ann Neurol 14: 662-669, 1983, 143, Allen RC, Petty RE, Lirenman OS, et al: Renal papillary necrosis in children with chronic arthritis, Am J Dis Child 140: 20-22, 1986, 144, Mustonen J: IgA glomerulonephritis and associated diseases, Ann Clin Res 16: 161-166, 1984, 145, Bruneau C, Villiaumey J, Avouac B, et al: Seronegative spondyloarthropathies and IgA glomemlonephritis: a report of four cases and a review of the literature, Semin Arthritis Rheum 15: 179-184, 1986, 146, .Iulkunen H: Synovial inflammatory cell reaction in chronic althritis, Acta Rheumatol Scand 12: 188-196, 1966, 147, Shichikawa K, Tsujimoto M, Nishioka J, et al: Histopathology of early sacroiliitis and enthesitis in ankylosing spondylitis, Adv [nflammat Res 9: 15, 1985, 148, Ball J: Pathology and pathogenesis, In Moll JMB (ed): Ankylosing Spondylitis, Edinburgh, Churchill Livingstone, 1980, p 96,
149, Ball .I: Enthesopathy of rheumatoid and ankylosing spondylitis, Ann Rheum Dis 30: 213-223. 1971. 150, Palm 0, Moum B, Ongre A, et al: Prevalence of ankylosing spondylitis and other spondyloanhropathies among patients with inflanunatory bowel disease: a population study (the IBSEN study), J Rheumatol 29: 511-515,2002, 151. Lehman TJ, Hanson V, Kornreich H, et al: HLA-B27-negative sacroiliitis: a manifestation of familial Mediterranean fever in childhood, Pediatrics 61: 423-426, 1978, 152, Shenry DO, Petty RE, Tredwell S, et al: Histocompatibility antigens in Osgood-Schlatter disease, J Pediatr Orthop 5: 302-305, 1985, 153, Olivieri I, Barbieri P, Gemignani G, et al: Isolated juvenile onset HLA-B27 associated peripheral enthesitis, J Rheumatol 17: 567-568, 1990, 154, Bowyer S: Hip contracture as the presenting sign in children with HLA-B27 arthritis, J Rheumatol 22: 165-167, 1995, 155, Burgos-Vargas R, Naranjo A, Castillo J, et al: Ankylosing spondylitis in the Mexican mestizo: patterns of disease according to age at onset. J Rheumarol 16: 186-191, 1989, 156, Maghraoui AE, Bensabbah R, Bahiri R, et al: Cervical spine involvement in ankylosing spondylitis, Clin Rheumatol 22: 94-98, 2003, 157, Macrae IF, Wright V: Measurement of back movement. Ann Rheum Di' 28: 584-589, 1969, 158. Moll JM, Wright V: Normal range of spinal mobility, An objective clinical study, Ann Rheum Dis 30: 381-386, 1971. 159, Barkley DO, Hohermuth HJ, Howard A, et al: [gA deficiency in juvenile chronic polyarthritis,.1 Rheumatol 6: 219-224, 1979. 160, Cassidy .IT: Selective IgA deficiency and chronic arthritis in children, In Moore TO (ed): Arthritis in Childhood, Report of the Eightieth Ross Conference in Pediatric Research, Columbus, OH, Ross Laboratories, 1981. p 82, 161. Kinsella TO, Espinoza L, Vasey FB: Serum complement and immunoglobulin levels in sporadic and familial ankylosing spondylitis, J Rheumatol 2', 308-313, 1975, 162, Cowling P, Ebringer R, Ebringer A: Association of inflammation with raised semm IgA in ankylosing spondylitis, Ann Rheum Dis 39: 545-549, 1980, 163, Corrigall V, Panayi GS, Unger A, et al: Detection of inunune complexes in semm of patients with ankylosing spondylitis, Ann Rheum Db 37: 159-163, 1978. 164, Juanola X, Mateo L, Domenech 1', et al: Prevalence of antiphospholipid antibodies in patients with ankylosing spondylitis, J Rheumatol 22: 1891-1895, 1995, 165, Kendall MJ, Farr M, Meynell MJ, et al: Synovial fluid in ankylosing spondylitis, Ann Rheum Dis 32: 487--492, 1973, 166, Traycoff RB, Pascual E, Schumacher HR Jr: Mononuclear cells in human synovial fluid, Identification of Iymphoblasts in rheumatoid althritis, Arthritis Rheum 19: 743-748, 1976, 167, Spriggs AI, Boddington MM, Mowat AG: Joint fluid cytology in Reiter's disease, Ann Rheum Dis 37: 557-560, 1978, 168, Bunch TW, Hunder GG, McDuft1e FC, et al: SynovilJl fluid complement determination as a diagnostic ;lid in intlammatory joint clisease, Mayo Clin Proc 49: 715-720, 1974, 169, Riley MJ, Ansell BM, Bywaters EG: Radiological manifestations of ankylosing spondylitis according to age at onset. Ann Rheum Dis 30: 138-148, 1971. 170, Azouz EM, Duffy CM: Juvenile spondyloarthropathies: clinical manifestations and medical imaging, Skeletal Radiol 24: 399--408, 1995, 171. Flato B, Smerdel A, Johnston V, et al: The intluence of patient characteristics, disease variables, and HLA alleles on the development of radiographically evident sacroiliitis in juvenile idiopathic arthritis, Arthritis Rheum 46: 986-994, 2002, 172, Luong AA, Salonen DC: Imaging of the seronegative spondyloanhropathies. Curr Rheumatol Rep 2: 288-296, 2000, 173, Bellamy N, Park W, Rooney PJ: What do we know about the sacroiliac joint' Semin Arthritis Rheum 12: 282-313, 1983, 174, Geijer M, Sihlbom H, Gothlin JR, et al: The role of CT in the diagnosis of sacro-iliitis, Acta Radiol ,'\9: 265-268, 1998, 175, Oudjhane K, Azouz EM, Hughes S, et al: Computed tomography of the sacroiliac joints in children, Can Assoc Radiol J 44: 313-314, 1993, 176, Kurugoglu S, Kanberoglu K, Kanberoglu A. et al: MRI appearances of inflammatory vertebral osteitis in early ankylosing spondylitis, Pediarr Radiol 52: 191-194. 2002, 177, Bollow M, Biedermann T, KannenbergJ, et al: Use of dynamic magnetic resonance imaging to detect sacroiliitis in HLA-B27 positive and negative children with juvenile althritides, J Rheumatol 25: ';56-564, 1998, 178, Brophy S, Mackay K, Al Saidi A, et al: The natural history of ankylosing spondylitis as defined by radiological progression, J Rheumatol 29: 1236-1243, 2002, 179, Lee HS, Kim TH, Yun HR, et al: Radiologic changes of cervical spine in ankylosing spondylitis. Clin Rheumatol 20: 262-266, 2001. 180, Pacheco-Tena C, Londono JD, Cazarin-Barrientos J, et al: Development of a radiographic index to assess the tarsal involvement in patients with spondyloarthropathies, Ann Rheum Dis 61: 330-354, 2002, 181. Averns HL, Oxtoby J, Taylor HG, et al: Smoking and outcome in ankylosing spondylitis, Scand J Rheumatol 25: 158-142, 1996, 182, Burgos-Vargas R, Vazquez-Mellado J, Pacheco-Tena C, et al: A 26 week randontised, double blind, placebo controlled exploratory study of sulfasalazine in juvenile onset spondyloarthropathies, Ann Rheum Dis 61: 941-942, 2002,
C HAP T E R 183, Dougados M, Boumier P, Amor B: Sulphasalazine in ankylosing spondylitis: a dou hIe blind controlled study in 60 patients, Br Med] (Clin Res Ed) 293: 911-914, 1986, 184, Dougados M, Maetzel A, Mijiyawa M, et al: Evaluation of sulphasalazine in the treatment of spondyloarthropathies, Ann Rheum Dis 51: 955-958, 1992 185, Fe!telius N, Hallgren R: Sulphasalazine in anky10sing spondylitis, Ann Rheum Dis 45: 396-399, 1986, 186, Nissila M. Lehtinen K, Leirisalo-Repo M, et al: Sulfasalazine in the treatment of ankylosing spondylitis, A twenty-six-week. placeho-controlled clinical trial. Arthritis Rheum 31: 1111-1116,1988. 187. Davis MJ. Dawes PT, Beswick E, et al: Sulphasalazine therapy in ankylosing spondylitis: its effect on disease activity. immunoglobulin A and the complex immunoglobulin A-alpha-1-antitrypsin. Br J Rheumatol 28: 410-413, 1989, 188. Fraser SM, Sturrock RD: Evaluation of sulphasalazine in ankylosing spondylitis-an interventional study, Br J Rheumatol 29: 37-39, 1990, 189. Corkill MM, Jobanputra P, Gibson T, et al: A controlled trial of sulphasalazine treatment of chronic ankylosing spondylitis: failure to demonstrate a clinical effect. Br I Rheumatol 29: 41-45, 1990. 190. Ansell BM, Hall MA, Loftus ]K, et al: A multicentre pilot study of sulphasalazine in juvenile chronic arthritis. Clin Exp Rheumatol 9: 201-203, 1991 191. Suschke HJ: Therapy of juvenile chronic arthritis with sulfasalazine. Z Rheumalol 46: 83-85, 1987, 192. Joos R. Veys EM, Mielants H, et al: Sulfasalazine treatment in juvenile chronic arthritis: an open study,] Rheumatol 18: 880-884, 1991, 193. HuangJL, Chen LC: Sulphasalazine in the treatment of children with chronic arthritis. Clin Rheumatol 17: 359--363, 1998. 194. MilicI' J.I. Ill: Prolonged use of large intravenous steroid pulses in the rheumati(' diseases of children. Pediatrics 65: 989--994. 1980, 195. Allen RC, Gross KR, Laxer RM. et al: 1ntraarticular triamcinolone hexacelonide in the management of chronic arthritis in children. Arthritis Rheum 29: 997-1001, 1986. 196. Huppertz HI, Tschammler A, Horwitz AE, et al: 1ntraarticular corticosteroids for ('hronic arthritis in children: efficacy and effects on cartilage and growth. J Pediatr 127: 317-321, 1995. 197. Braun J, Bollow M, Seyrekbasan F, et al: Computed tomography guided corticosteroid injection of the sacroiliac joint in patients with spondyloarthropathy with sacroiliitis: clinical outcome and followup by dynamic magnetic resonance imaging, .I Rheumatol 23: 659--664, 1996. 198. Creemers MC, Franssen M.I, van de Putte LB, et al: Methotrexate in severe ankylosing spondylitis: an open study. J Rheumatol 22: 1104-1107, 1995. 199. Roychowdhury B, Bindey-Bagot S, Bulgen DY, et al: Is methotrexate effective in ankylosing spondylitis? Rheumatology (Oill 41: 1330-1332, 2002. 200. Leipold G, Schutz E, Haas JP, et al: Azathioprine-induced severe pancytopenia due to a homozygous two-point mutation of the thiopurine methyltransferase gene in a patient with juvenile HLA-B27-associated spondylarthritis, Arthritis Rheum 40: 1896-1898, 1997, 201. Homeff G, Burgos-Vargas R: TNF-alpha antagonists for the treatment of juvenile-onset spondyloarthritides. Clin Exp Rheumatol 20: S137-S142, 2002.
13
JUVENILE ANKYLOSING SPONDY LITIS
323
202, Davis JC, Jr., van del' HD, Braun J, et al: Recombinant human tumor necrosis factor receptor (etanercept) for treating ankylosing spondylitis: a randomized, controlled trial. Arthritis Rheum 48: 3230-3236, 2003. 203. Gorman .ID, Sack KE, Davis .IC ]1': Treatment of ankylosing spondylitis by inhibition of tumor necrosis factor alpha. N Engl J Med 346: 1349-1356, 2002, 204. Braun], Brandt.l, Listing], et al: Treatment of active ankylosing spondylitis with infliximab: a randomised controlled multicentre trial. Lancet 359: 1187-1193, 2002, 205. Maksymowych WP, Jhangri GS, Fitzgerald AA, et al: A six-month randomized, controlled, double-blind, dose-response comparison of intravenous pamidronate (60 mg versus 10 mg) in the treatment of nonsteroidal antiinflammatory drug-refractory ankylosing spondylitis. Arthritis Rheum 46: 766-773, 2002. 206, Sochart DH, Porter ML: Long-term results of total hip replacement in young patients who had ankylosing spondylitiS: eighteen to thirty-year results with survivorship analysis. .I Bone Joint Surg Am 79: 1181-1189, 1997, 207, Lehtimaki MY, Lehto MU, Kautiainen H, et aI: Charnley total hip arthroplasty in ankylosing spondylitis: survivorship analysis of 76 patients followed for 8-28 years. Acta Orthop Scand 72: 233-236. 2001. 208. Burgos-Vargas R, Vazquez-Mellado.1: Cohort study comparing juvenile ankylosing spondylitis (jAS) and juveniie rheumatoid arthritis (jRAl, J Rheumatol 19 (Suppl 33): 118, 1992. 209. Kumar A, Bansal M, Srivastava DN, et al: Long-term outcome of undifferentiated spondylarthropathy. Rheumatol Inl 20: 221-224, 2001. 210. Haywood KL, Garratt M, Jordan K, et al: Disease-specific, patient-assessed measures of health outcome in ankylosing spondylitis: reliability, validity and responsiveness. Rheumatology (OxfJ 41: 1295-1302, 2002. 211. Doran MF, Brophy S, Mackay K, et al: Prediclors of [ongterm outcome in ankylosing spondylitis. J Rheumatol 30: 316-320. 2003, 212. Khan MA, Khan MK, Kushner 1: Survival among patients with ankylosing spondylitis: a life-table analysis. J Rheumatol 8: 86-90, 1981. 213. van del' HD, Bellamy N, Calin A, et al: Preliminary core sets for endpoints in ankylosing spondylitis. A'5essments in Ankylosing Spondylitis Working Group.] Rheumatol 24: 2225-2229, 1997. 214. Guillemin F, Challier B, Urlacher F, et aI: Quality of life in ankylosing spondylitis: validation of the ankylosing spondylitis Arthritis Impact Measurement Scales 2, a modified Arthritis Impact Measurement Scales Questionnaire. Arthritis Care Res 12: 157-162, 1999, 215. Spoorenberg A, van del' HD, de Klerk E. et al: A comparative study of the usefulness of the Bath Ankylosing Spondylitis Functional Index and the Dougados Functional Index in the assessment of ankylosing spondylitis. J Rheumatol 26: 961-965, 1999. 216. Calin A, Elswood ]: The natural history of juvenile-onset ankylosing spondylitis: a 24-year retrospective case-control study. Br J Rheumatol 27: 91-93, 1988. 217. Garcia-Morteo 0, Maldonado-Cocc'o JA, Suarez-Almazor ME, et aI: Ankylosing spondylitis of juvenile onset: comparison with adult onset disease. Scand J Rheumatol 12: 246-248, 1983. 218. Marks SH, Barnett M, Calin A: A case-control study of juveniie- and adult· onset ankylosing spondylitis. J Rheumatol 9: 739--741, 1982.
(
HAP T E R
14
PSORIATIC ARTHRITIS Ross E. Petty and Taunton R. Southwood
~wr
DEFINITION AND CLASSIFICATION The definition and diagnosis of juvenile psoriatic arthritis 0PsA) has changed considerably in the last three decades. Initially it was recognized as the occurrence of inflammatory arthritis before the age of 16 years, associated with psoriasis, which either preceded the onset of arthritis or followed it within the subsequent 15 years. 1 More recent attempts to formulate criteria for the diagnosis and classification of JPsA include the "Vancouver criteria"2 and an international consensus proposal of classification criteria for juvenile idiopathic arthritis 3 (Table 14-1). An important feature of both sets of criteria is the recognition of a diagnosis of JPsA in a child with arthritis who has yet to develop the rash of psoriasis. This entity, also known as psoriatic arthritis sine psoriasis, has also been recognized in adults. 4 A follow-up study of children with probable psoriatic arthritis defined by the Vancouver criteria found that definite psoriatic arthritis had occurred in approximately half of the children after a mean of 2.1 years.'; Key clinical elements of the classification criteria for JPsA include arthritis, psoriasis, a family history of psoriasis, dactylitis, and nail pitting. Arthritis is defined as swelling within a joint or limitation in range of joint movement with joint pain or tenderness that persists for more than 6 weeks, is observed by a physician, and is not the result of primarily mechanical disorders. Dactylitis can be distinguished from arthritis if swelling of one or more digits extends beyond the joint margins. It represents the combined effects of arthritis and tenosynovitis. Psoriasis, or a positive family histOly of psoriasis in a first-degree relative (parent or sibling), requires that the skin rash be diagnosed with certainty as psoriasis. Such strict definitions may be difficult to use in clinical situations, but they help improve communication and attempts at understanding the etiology and pathogenesis of the disease. 6 Traditionally, psoriatic arthritis in adults and children has been included under the term "spondyloarthropathies," although there may be important differences between psoriatic arthritis in children and adults. 7- 11 JPsA shares few of the characteristics of the spondyloarthropathies, such as sacroiliac disease, enthesitis, acute uveitis, or an association with the human leukocyte antigen (HLA) 827 allele. At onset, JPsA is often similar to the asymmetrical oligoarticular disease described by Moll and Wright ll (Table 14-2). 324
EPIDEMIOLOGY Incidence and Prevalence Psoriasis affects 1% to 3% of the general population, Li and as many as 20% to 30% of patients are reported to have an associated arthritis. 14-16 In the pediatric population, the frequency of psoriasis is lower (about 0.5% of the population younger than 16 years of age J7), and the proportion with arthritis may also be lower. These figures must be viewed with caution, however, because at least half of the children with psoriatic arthritis develop joint inflammation before the onset of psoriasis. Published studies suggest that JPsA accounts for 21Yo to 150/0 of all children with chronic arthritis. I 8--20 Several recent surveys of arthritis in children that used similar diagnostic criteria have confirmed that JPsA accounts for approximately 7% of all cases of chronic childhood arthritis (Table 14-3).18--21 Estimates of the incidence have suggested that 2.3 to 3 per 100,000 children develop psoriatic arthritis every year, with a prevalence of 10 to 15 per 100,000. 2.21 The ethnic associations of psoriatic arthritis have only rarely been studied, but a multicenter survey in the United States found that more than 90% of patients with definite psoriatic arthritis were white, 5% were Hispanic, and 2.5% were African American. 1H Psoriatic arthritis has also been reported in Chinese, Indians, and Malaysians living in SingaporeY
Age at Onset and Sex Ratio In the pediatric population, the age at onset of psoriatic arthritis appears to be bimodally distributed. 2 A first peak occurs during the preschool years (mainly in girls), and a second occurs during middle to late childhood, centering on 10 years of age. Unlike adult psoriatic arthritis, psoriasis begins after the arthritis in the majority of children, and simultaneous onset is found in fewer than 10% of the patients (Table 14-4).l.2·23--2';.116 This may explain the earlier age at diagnosis of probable psoriatic arthritis (7.4 ± 4.5 years) compared with definite psoriatic arthritis 00 ± 4.9 years) reported in an American survey.1H JPsA is very uncommon before the age of 1 year. It is somewhat more frequent in girls than in boys (see Table 14-4), and in one survey girls accounted for 60% of 128 cases of JPsA. 20 The
C HAP T E R
" II ,-
14 P S 0
R I ATI CAR T H R I TI S
325
a predisposition to psoriasis might lead to arthritis and even to the peculiarly asymmetrical large and small joint involvement in the disease.
I ABI E 14-] Criteria for Diagnosis and Classification 01 Juvenile Psoriatic Arthritis
ILAR CriterIa·
Environmental Triggers
Arthritis and psoriasis Or Arthritis and at least two of the following: Dactylitis Nail pitting or onycholysis Family history of psoriasis in a first-degree relative Exclusions: Presence of rheumatoid factor Presence of systemic juvenile idiopathic arthritis Presence of a family history of HLA-B27-assodated disease Onset of arthritis in a male with HLA-B27 after the age of 6 yr
There have been a few reports of psoriatic arthritis precipitated by physical trauma. 27 .28 Other environmental triggers, such as bacterial infections, have also been implicated. The onset of psoriasis in children may be preceded by streptococcal upper respiratory tract infection or, less commonly, skin infection. 29 .3o Some studies in children with psoriatic arthritis have demonstrated synovial fluid T cell responses to streptococci,3l,32 However, in other studies,33 the response of synovial membrane mononuclear cells from patients with psoriatic arthritis was seen only to superantigens of streptococcus, and not to typical streptococcal antigens. The pathogenic relevance of immune responses to streptococcal antigens remains unclear. Viral infections have also been implicated in the disease, and JPsA may occur after a viral infection such as chickenpox. l However, a wider study of the epidemiology of childhood arthritis found no correlations between the onset of JPsA and coincident infections with mycoplasma, respiratory syncytial virus, adenovirus, influenza A and B, parainfluenza, rubella, cytomegalovirus, or herpes simplex..34
VIIICOIMII' CrIterIat Definite juvenile psoriatic arthritis: Arthritis with typical psoriatic rash Or Arthritis with three of the following minor criteria: Nail pining or onycholysis Family history of psoriasis (first- or second-degree relative) Psoriasis-like rash Dactylitis Probable juvenile psoriatic arthritis: Arthritis with two of the four minor criteria 'Petty RE. Southwood TR, Manners P, et al: International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: Second Revision, Edmonton 2001. J Rheumatol 31: 390-392, 2001. tSouthwood TR, Petty RE, Malleson PN, et al: Psoriatic arthritis in children. Arthritis Rheum 32: 1007-1013, 1989.
Pathogenesis incidence of arthritis in adults who have psoriasis is 3.5 per 100,000 men and 3.4 per 100,000 women. 26
There is increasing evidence that psoriatic arthritis is mediated by activated COW T cells. These cells invade the epidermis in psoriatic skin lesions induced by the Koebner response, and long-lived C08+ clones have been recovered from psoriatic skin biopsies. 3,;.36 Synovial fluid C08+ T cell numbers are higher in patients with psoriatic arthritis than in those with other forms of arthritis,3 7 although in the synovium itself the ratio of C04+ to C08+ T cells is variable. 38 Patients with psoriatic arthritis
ETIOLOGY AND PATHOGENESIS The cause of JPsA is unknown. Satisfactory theories of the etiology and pathogenesis of JPsA should explain not only the link between psoriasis and arthritis but also how
' . TABLE 14-2
Classification of Psoriatic Arthritis
Percentage of Cases
Onset Type
Charaderlstlcs
Monarticular or asymmetrical oligoarthritis Symmetrical polyarthritis Predominant distal interphalangeal joint Spondylitis Arthritis mutilans
Dactylitis, often becomes polyarticular Polyarticular, large and small joints Accompanying nail disease Peripheral arthritis with sacroiliac joint disease Severely deforming, often with sacroiliac joint disease and ankylosis
I!:.
TABLE 14-3
70 15 5 5 5
Epidemiology of Juvenile Psoriatic Arthritis (JPsA)
Author and Year (ref. no.) Gare & Fasth, 1992 (21) Bowyer et aI., 1996 (18) Malleson et aI., 1996 (19) Symmons et aI., 1996 (20)
Total no. of Patients Surveyed
JPsA
Age at Onset (yr)
Sex
Inddence (per
(0/0)
(F/M)
100.000)
213 1568 861 1831
2.8 5.5 7.0 7.0
11.5 8.7 10.1 10.1
1.0 1.3 1.6 1.6
0.3 0.23
326
,,'",II
C HAP T E R
TABLE. 14-4
14
PSORIATIC ARTHRITIS
Juvenile Psoriatic Arthritis Summary of Reported Seri('s
Charaderlstlc Patients (n) Male/female ratio Age at onset (yr) roint disease Skin disease Disease sequence Psoriasis first (%) Arthritis first (%) Simultaneous (%) Oligo-onset (~4 joints) Poly-onset (2:5 joints) DIP joints affected (%) Sacroiliac arthritis (%) Nail changes (%) Uveitis (%)
Lambert et al.23
CaiabroZ4
43 11:32
12 5:7
9.3 10.4 40 53 7 55 45 21 28 70 9
NA
12 67 33 0 42 58 50 17 92
a
SIII s1l6
Shore & Anselll
Wesolowskr 5
et al. z
Total
24 7:17
60 35:25
21 13:8
35 24:11
195 0.95:1
10 11
11
NA NA
6.7 12.6
6.7-11.0 8.8-12.6
33 58 9 58 42 62 29 83 13
42 43 15 73 27 42 4r
33 62 5 86 14 10 100' 86 14
43 48 10 94 6 29
33-67 33-62 5-15 42-94 6--58 10-62
11
11-100
51 17
51-92 0-17
8.8
77
8
Southwood
DIP, distal interphalangeal: NA, not available. 'Only selected patients had pelvic radiographs.
who are infected with human immunodetlciency virus (HIV) often undergo a flare in the arthritis activity when CD4+ T cell numbers are low. 39 An important role of non-major histocompatibility complex (MHC)-restricted cytotoxicity was suggested by the studies of Spadaro and colleagues,40 which demonstrated low levels of peripheral blood and synovial fluid natural killer cells. Cytokines are important in the pathogenesis of psoriatic arthritis. 41 ,42 Tumor necrosis factor-a (TNF-a) activates nuclear factor-KB (NF-KB) , which leads to proliferation of synovial cells, increases leukocyte infiltration, and promotes production of other pro-inflammatory cytokines and osteoclastogenesis mediated by the receptor-associated NF-KB ligand (RANKL),43.44 Angiogenesis, influenced by cytokines including TNF-a, appears to have an important role in the pathogenesis of psoriatic arthritis. 4s In one study, interleukin-2 OL-2) was demonstrated in the synovium of patients with psoriatic arthritis but not in those with rheumatoid arthritis. 46 The pro-inflammatory cytokines TNF-a, IL-I~, and IL-8, as well as TNF-a and IL2 receptors, have been detected in synovial fluid from patients with psoriatic arthritisY Almost all of the studies of cytokines in psoriatic arthritis have been performed in adults, and it is not certain that the same observations would be found in children with JPsA. To date there have been no unique associations of cytokine profiles in JPsA. Psoriasis can be induced in scid/scid mice by minor histocompatibility mismatched naive CD4+ T cells. 4B Expression of a2, as, or ~1 integrin subunits by murine suprabasal keratinocytes is also associated with histopathologic changes characteristic of human psoriasis. 49 In a mouse model of psoriasis in which bone morphogenic protein-6 is overexpressed, overproduction of bone pro-osteoblastic cytokines leads to the periostitis and enthesitis typical of psoriatic arthritis. so
GENETIC BACKGROUND There is convincing clinical evidence of a strong genetic contribution to the susceptibility to psoriasis and psoriatic arthritis. 51 A concordance of 55% to 70% is documented
for psoriasis and psoriatic althritis in monozygotic twins'"z However, differing genetic susceptibilities may underlie psoriasis and psoriatic arthritis. Genomic DNA extracted from 395 probands with psoriasis provided strong evidence for a susceptibility gene on chromosome 6p, close to the HLA region, which was inherited paternally.53 However, in probands with psoriatic arthritis, this linkage was less evident and there was no paternal effect. There appear to be ethnic differences in the occurrence of psoriatic arthritis in adults. In a study of patients of Asian origin, psoriatic arthritis was significantly more common in patients of Indian extraction, than in Chinese or Malaysians,zz Given the probable importance of synovial CD8+ T cells in the pathology of psoriatic arthritiS, it might be expected that MHC class I molecules are important in this disease, and in adults with psoriatic arthritis associations with HLA-BI3, -BI7, -BI9, -B39, and -Cw6 have been reported. 54 It has been suggested that various HLA markers are associated with peripheral joint disease progression in psoriatic arthritis, The marked clinical heterogeneity of this disorder suggests the possibility that several distinct genetic associations might be important. Indeed, some subset specific associations are described. In one study, B22 appeared to be "protective," whereas B39 and DQw3 were associated with progressive disease."" The presence of HLA-B27 in patients with psoriatic arthritis is associated with spinal inflammation and sacroiliitis.'i6 None of these associations has been convincingly demonstrated in JPsA, although the number of children investigated has been comparatively small. Childhood disease appears to be linked to the MHC class II molecules DRI and DR6," and it has been suggested that the haplotype HLADRBI·OI, -DQAI·OIOl, -DQBP05 predisposes to the disease. 57 DRBI·04, DQAI·03, and DQBP03 appear to be "protective" alleles, which are found in significantly lower frequency in patients than in the normal popUlation. JPsA differs from other forms of childhood arthritis in that it does not appear to be associated with A2 or DR8. s
C HAP T E R
It is important to note that genes other than those cod-
ing the HLA system have been implicated in the pathogenesis of psoriatic arthritis. Immunologic heavy-chain gene polymorphisms and T cell receptor polymorphisms have been associated with psoriatic arthritis. 58,59 A mutation of the pro-inflammatory cytokine TNF-aa promotor was present in 32% of patients with psoriatic arthritis, compared with only 7% of controls. 6o The functional implications of this mutation have yet to be determined.
CLINICAL MANIFESTATIONS
Olnlcal Patterns JPsA usually begins with inflammation of only a few joints during the first 6 months of the disease, initially making its differentiation from oligoarticular juvenile idiopathic arthritis QIA) difficult. Somewhat less commonly, children with JPsA have symmetrical polyarthritis at disease onset. It has been suggested that psoriatic arthritis in adults is less painful than other forms of arthritis,61 and the uncomplaining younger child often presents with an apparently painless limp or is unaware of the presence of dactylitis of one or more toes. Most older children, however, have symptoms of joint stiffness and pain, particularly on awakening. Huemer and colleagues62 compared the patterns of joint involvement at onset in children with oligoarticular JPsA and pauciarticular JRA. Children with JPsA were much more likely to have small joint disease than those with pauciarticular JRA. They suggested that a criterion consisting of small joint disease and/or wrist disease and/or dactylitis alone may increase the ability to differentiate oligoarticular JPsA from oligoarticular juvenile idiopathic arthritis. The course of the disease is usually characterized by an increase in the number of affected joints. Occasionally, the reverse pattern is observed-that is, an initial polyarthritis resolves with the exception of one or two persistently inflamed joints. There appear to be several clinical patterns of JPsA that are similar to those of adult psoriatic arthritis (see Table 14-2). A scattered asymmetrical polyarthritis is the most common long-term manifestation of psoriatic arthritis in both children and adults. Longitudinal follow-up studies have suggested that the most clinically useful subgrouping is (1) patients who develop axial disease, who are likely to be HLA-B27 positive, and (2) those with peripheral joint disease, who are likely to be HLA-B27 negative. 56
14
nl@1
1Ii.~ TABLE 14-5
Joints Affected in Juvenile Psoriatic Arthritis (%)
Shore lr Anselr
Joint Knee Finger Toe Ankle Wrist Elbow Hindfoot MCP DIP finger MTP Cervical spine Lower spine Sacroiliac joints Hip Sternoclavicular Temporomandibular Shoulder
327
PSORIATIC ARTHRITIS
Southwood et al.t
Initial
Cumulative
Initial
Cumulative
53 28 25 21
77 40 45 63 62 43 38 53 42 33 32
57 17 20 14 0 6
89 60 46 63 23 20 23
3 0 0
11 11
11
23
0
34 9
11 10
8 8 8 7 7 5 0
38 15
11
0
17
DIP, distal interphalangeal; Mep. metacarpophalangeal; MTP, metatarsophalangeal. 'Shore A, Ansell BM: Juvenile psoriatic arthritis: an analysis of 60 cases. J Pediatr 100: 529-535. 1982. 'Southwood TR. Petty RE. Malleson PN, et a1: Psoriatic arthritis in children. Arthritis Rheum 32: 1007-1013, 19R9.
in other forms of chronic oligoarthritis. Distal interphalangeal (DIP) joint involvement occurred in 29% of children in one series,2 and dactylitis (defined as swelling of a digital joint and periarticular tissues extending beyond the joint margin, often giving a typical "sausage digit" appearance) occurred in 49% (see Fig. 14-1).2 The presence of dactylitis implies underlying tendinitis, and tendon nodules have been described in 14% of children. The limb girdle joints (glenohumeral and hip) and the cervical spine are relatively spared in JPsA compared with other juvenile arthritides. Sacroiliitis occurs in a minority of patients. 5
Arthritis A predominantly asymmetrical large and small joint arthritis is typical. The most commonly affected joint is the knee, but JPsA also has a particular predilection for the small joints of the hands and feet (Table 14-5).2,5 Swelling of a single small joint, or the presence of dactylitis, especially in a toe (Fig. 14-1), is higWy suggestive of psoriatic arthritis; small joint disease is relatively uncommon
• Figure 14-1 Dactylitis in a child with psoriatic arthritis. The second and fifth toes are diffusely swollen. (From Petty RE, Malleson P: Spondyloarthropathies of childhood. Pediatr elin North Am 33: 1079, 1986.)
328
C HAP T E R
14
PSORIATIC ARTHRITIS
Enthesltls Enthesitis-inflammation of the entheses (the sites of insertion into bone of tendon, ligament, or joint capsule)-has been proposed as an important unifying clinicopathologic feature in adult psoriatic arthritis. I I Patients have been described in whom isolated enthesitis occurred in association with psoriasis. 63 In JPsA, however, clinical evidence of enthesitis is unusual unless the patient is HLA-B27 positive, and it correlates more closely with enthesitis-related arthritis (see Chapter 13).
Extra-Articular Manifestations Skin Disease The typical rash of psoriasis in children is characterized by well-demarcated, erythematous, scaly lesions occurring over the extensor surfaces of the elbows and forearms, knees, and interphalangeal joints. This form of psoriatic skin lesion, psoriasis vulgaris, occurs in more than 80% of children who have the skin rash of psoriatic arthritis. Less common presentations include guttate psoriasis, inverse (flexural) psoriasis, pustular psoriasis, and the generalized forms (von Zumbusch and erythrodermic generalized psoriasis). In small children, the rash may not be obvious; a careful search of the hairline behind the ears, navel, and groin may be revealing (Fig. 14-2). Auspitz's sign, a small pinpoint area of bleeding found on removal of a psoriatic skin scale, perhaps reflects the underlying predisposition to angiogenesis. 64 A number of disorders enter into the differential diagnoses, including atopic eczema and contact dermatitis, drug eruptions, tinea corporis, and, less commonly in children, pityriasis rosea, lichen planus, discoid lupus, and mycosis fungoides. 65 The onset of the skin manifestations of the disease is rarely coincident with the onset of joint disease. In most adult studies, the rash of psoriasis appears to precede the onset of arthritis, but in children the ratio is evenly divided (see Table 14--4). Approximately
• Fllllre 14-Z This scaly pink rash over the sacrum and in the gluteal crease was initially treated as "diaper" dermatitis.The isolated patch on the left buttock (arrow) suggested the diagnosis of psoriasis.
25% of children with jPsA sine psoriasis develop a typical
psoriatic rash within 2 years. 5
Nail Changes There are several forms of nail involvement in psoriasis. The most common is nail pitting (found in about one third of patients), and the least common is complete nail dystrophy or onycholysis. Nail pits are typically small (0.5 to 1 mm diameter), round, shallow, and dimple-like, most easily seen on the fingernails as a disruption to the normal reflection of light from the nail surface (Fig. 14-3). Horizontal, but not longitudinal, nail ridging is also associated with psoriasis. The association of nail dystrophy and DIP joint arthritis is not as common in children as in adults. 66 Typical nail pitting may occur in normal persons, in children with fungal infections of the nail, in those with eczema, and in those who bite their nails. There is a close anatomic relationship between the DIP joints and the nail beds, and it is possible that subclinical enthesitis may explain the presence of nail dystrophy.l1 An alternative explanation is that abnormal angiogenesis leads to altered nail bed blood supply. Although nail fold capillaries usually appear normal on magnified inspection,67 nail fold video capillaroscopy has demonstrated morphologic abnormalities. 68
Uveitis Asymptomatic anterior uveitis, clinically indistinguishable from that seen in children with oligoarthritis, occurs in 15% to 20% of children with JPsA and is associated with the presence of antinuclear antibodies (ANA),l Rarely, older patients may complain of reduced visual acuity and photophobia. Young children may appear to squint in bright light. The acute symptomatic anterior uveitis observed in 5% to 10% of adult patients69 is rare in childhood disease and is likely to be associated with axial joint involvement in children with HLA-B27. All children with JPsA should undergo regular slit-lamp examination of the
• Figure 14-3 The nail of the index finger has multiple pits (arrowhead) characteristic of psoriasis.The digit is also swollen, suggesting dactYlitis. (From Petty RE, Malleson P: Spondyloarthropathies of childhood. Pediatr C1in North Am 33: 1079, 1986.)
C HAP T E R
anterior chamber by an experienced ophthalmologist at least every 6 months. Signs of anterior uveitis apparent on slit-lamp biomicroscopy include protein flare and increased cells in the anterior chamber, similar to the signs observed in anterior uveitis accompanying oligoarticular JRA. Patients with long-standing uveitis may have an irregular pupil with posterior synechiae, band keratopathy, cataract formation, and visual impairment, including blindness. There is a suggestion70 that the chronic anterior uveitis associated with psoriatic arthritis is relatively resistant to treatment with topical glucocorticoids, although this has not been tested formally.
Other Systemic Manifestations It is uncommon for children with psoriatic arthritis to have
fever, but patients with significant arthritis may have all of the constitutional features of a chronic inflammatory disease, such as anorexia, anemia, and poor growth. Amyloidosis has been reported.! Rarely, features of colitis, mucositis, and urethritis occur in adults with psoriatic arthritis. The SAPHO syndrome (synovitis, acne, pustulosis, hyperostosis, and osteitis) and CRMO (chronic recurrent multifocal osteomyelitis) have been postulated to be part of the same spectrum of disease as psoriatic arthritis. 71 Reported cardiac manifestations include aortic incompetence and mitral valve prolapse. 72 Upper limb lymphedema is a rare complication of psoriatic arthritis in adults.73
PATHOLOGY Little is known about specific histopathologic changes in children with JPsA. The inflammatory synovial infiltrate is indistinguishable by light microscopy from that of other forms of chronic arthritis. 74 Synovial histology of 14 children with long-standing JPsA demonstrated hyperemia and an inflammatory round cell infiltrate. l There are no other systematic studies of the histopathology of the synovium in JPsA; most information is gleaned from the study of adults with the disease. The histopathology of inflamed synovium and lesional skin is typified by marked angiogenesis. 75 Distinctive capillary hyperemia and neovascularity, with tortuous and bushy vessels, were demonstrated by knee arthroscopy in patients with early psoriatic arthritis. 64 .75 At the cellular level, these changes are reflected by endothelial cell hypertrophy, dilated rough endoplasmic reticulum, thickened arteriolar basement membranes, and increased deposition of perivascular collagen. 76 Endothelial cell expression of adhesion molecules such as CD54, which interact with CDlla/CD18 expressed on the surface of T cells, has been demonstrated in affected skin and synovium. 77 The vascular changes are likely to contribute to the inflammatory process by attracting T cells to the synovial compartment. Cutaneous lymphocyte antigen and its receptor E-selectin are expressed in both skin and joint. 78.79 T cells expressing cutaneous antigen, however, appear to migrate preferentially to the skin only, not the joint. 80 In three patients with psoriatic arthritis, T cells expressing the chemokine receptor CCR5 were present in greater numbers in synovial fluid than in peripheral
14
PSORIATIC ARTHRITIS
329
blood, although this finding did appear to discriminate among the various forms of inflammatory arthritis. 8! Evidence of selective T cell traffic between the affected skin and inflamed joint is equivocal.
LABORATORY EXAMINATION No laboratory tests are pathognomonic for JPsA. Most patients have elevated acute-phase reactants (erythrocyte sedimentation rate, C-reactive protein), the anemia of chronic disease, and thrombocytosis, as do adults with the disease. 82 However, approximately one third of patients have no laboratory evidence of an acute phase response, and this occasionally misleads the clinician into excluding the diagnosis of arthritis. ANAs are found in 30% to 60% of all children with JPsA. Although the antigenic specificities of the ANAs are unknown, antihistone antibodies have been reported. 2 Rheumatoid factor is not present.
RADIOLOGIC EXAMINATION Plain radiographic features of JPsA8:Hl5 generally follow a sequence of changes similar to those of other forms of childhood arthritis. In early arthritis, particularly within the first few weeks of the disease, soft tissue swelling around the joint (with or without joint effusion) is the only abnormality. Periarticular osteoporosis may occur within a few months after the onset of joint swelling, and periosteal new bone formation is common in digits affected by dactylitis (Fig. 14-4). The presence of periostitis due to enthesitis may contribute to the "pencil in cup" appearance of the DIP joint found in adults with the disease. 86 Such an appearance cannot be explained by synovitis alone, because the joint itself contains only a vestigial amount of synovium. l1 Instead, there may be a central erosion surrounded by joint capsular calcification at the sites of the flexor and extensor digitorum entheses. Joint-space narrowing, indicating significant cartilage loss, and erosive disease of bone are usually late features of JPsA (Fig. 14-5). Bone remodeling may eventually occur, secondary to persistent periostitis, altered epiphyseal growth, and osteoporosis. Sacroiliac erosions are uncommon, and the osteolysis typical of adult psoriatic arthritis is rare in children. Other imaging techniques (including magnetic resonance imaging, nuclear scans with technetium 99m-Iabeled immunoglobulin, and ultrasonography) have been used to demonstrate the presence of enthesitis. 87,88
TREATMENT There have been no controlled studies of the efficacy and safety of antirheumatic medications in children with JPsA. As a result, the approach to treatment is based on studies of other types of chronic arthritis in children and of psoriatic arthritis in adults. Many experienced clinicians use a combination of nonsteroidal anti-inflammatory drugs (NSAIDs) and long-acting intra-articular glucocorti-
330
C HAP T E R
14 P S 0
R I A TIC ART H R I TIS
• Allllre 14-4 .4, Radiograph shows periostitis of the fifth metatarsal and proximal phalanx of a 14-year-old boy with psoriatic arthritis of recent onset (arrowheads). 8, Radiograph of the foot of a 12-year-old girt with psoriatic arthritis of several years' duration.There is marked erosion of the head of the fifth metatarsal and the base of the proximal phalanx with dislocation of the joint.The other joints are relatively normal. (.4, from Petty RE, Malleson P: Spondyloarthropathies of childhood. Pediatr Oin North Am 33: 1079, 1986.)
coids (e.g., triamcinolone hexacetonide) for the initial treatment of oligoarthritis, adding methotrexate in the event of an inadequate response or at the first evidence of progression to polyarticular disease. Naproxen 05 to 20 mg/kg/day in two divided doses) or ibuprofen (30 to 40 mg/kg/day in three or four doses per day) is most commonly used, especially in younger children. Other NSAIDs, such as piroxicam (0.5 mg/kg/day in a single dose) or indomethacin (up to 2.5 mg/kg/day), have been used successfully in the symptomatic treatment of psoriatic arthritis in children, although these dmgs have not been formally evaluated in JPsA. All NSAIDs are taken with or just after food; gastric irritation can be further relieved with misoprostol or a histamine 2 blocker such as ranitidine. A selective cyclooxygenase-2 inhibitor such as celecoxib or rofecoxib could be considered, although there have been no trials of these agents in JPsA. Patients with JPsA do not appear to have more dmg side effects than children with other forms of arthritis. Intra-articular triamcinolone hexacetonide (1 mg/kg per large joint, 0.5 mg/kg for small joints) is effective in treating the child with oligoarthritic JPsA, but its efficacy in polyarticular disease is less clear. Intra-articular glucocorticoids do not appear to be effective for treating dactylitis or tenosynovitis, particularly after the underlying bone has become thickened by periosteal reaction. There is some anecdotal evidence that the response to repeated joint injections diminishes over time, perhaps as the degree of joint destmction becomes more severe. It is important, therefore, to use intra-articular corticosteroids earlier, rather than later, in the disease course. Methotrexate is the slow-acting antirheumatic dmg most frequently used to treat psoriatic arthritis in children
• Allllre 14-5 A, Psoriatic arthropathy affecting the distal interphalangeal joints of the third to fifth fingers (amJwheads). 8, Magnified view of the distal interphalangeal joint of the third finger.
C HAP T E R
and adults89-98; occasionally it is used in combination with oral glucocorticoids. 89 Methotrexate treatment is usually initiated at a dose of 0.35-0.5 mg/kg by mouth once a week given at least an hour before eating. If the dose is tolerated but the drug is ineffective, the dose may be gradually increased to 1 mg/kg/week (maximum of 25 mg/week). Common side effects include nausea, abdominal pain, and mood change. Changing from oral to subcutaneous administration of methotrexate in the same dose, particularly after a weekly dose of 15 to 20 mg has been reached, may improve efficacy and reduce side effects. It is possible that the early use of methotrexate may reduce the rate of progression of the disease and even the risk of polyarthritic evolution. Methotrexate may also help to control the rash of psoriasis. In several studies, including a 12-month prospective, randomized, controlled trial of psoriatic arthritis treatment in adults, cyclosporine appeared to be as effective as methotrexate for the control of the disease.99-103 However, long-term administration of cyclosporine has been associated with hypertension, and a life-table analysis suggested that methotrexate is likely to be better tolerated. 103 Hypertension is rare in most children treated with cyclosporine for arthritis, although they may develop raised creatinine levels. Although anti-TNF biologic agents are used to treat psoriasis, there are only anecdotal reports of their use in the treatment of JPsA. 104 A wide variety of conventional and unconventional drugs have been used to treat psoriatic arthritis in adults, including sulfasalazine,lO\ gold,I06,107 azathioprine,108 cWoroquine,109 etretinate,1l0 fish oil, III and vitamin D3 112 A few reports of surgical approaches to psoriatic arthritis have been published.l13,114 Children with JPsA have successfully undergone total hip replacement. The role of disease education is important in managing psoriatic arthritis in young people.1l\ The skin rash of psoriasis usually responds to a combination of moisturizing emollients to prevent fissuring, keratolytics for descaling the skin (e.g., 5% salicylic acid emollient), coal tar preparations, antihistamines to prevent pmritus, and topical glucocorticoid creams. 6'; The management of a child with psoriatic arthritis and significant skin or nail disease should always involve the expertise of a pediatric dermatologist.
COURSE OF THE DISEASE AND PROGNOSIS JPsA has a relatively poor outlook compared with other oligoarticular forms of chronic arthritis in childhood. In a study of 63 children with JPsA who had been monitored for longer than 5 years, 70% continued to have active arthritis, and one third of these had limited vocational and avocational activities, 5 Ten percent of patients reported by Shore and Ansell 1 were severely incapacitated by their disease, A study of the prognosis of adult psoriatic arthritis suggested that patients who were HLAB27 positive and had axial disease were more likely to develop erosions than those who had peripheral joint involvement only.56 Deaths in ]psA secondary to amyloidosis have been reported but are rare. 1 The influence of methotrexate and the biologics on the prognosis of JPsA has yet to be assessed.
14
PSORIATIC ARTHRITIS
331
REFERENCES 1. Shore A. Ansell BM: Juvenile psoriatic arthritis: an analysis of 60 cases. J Pediatr 100: 529-535, 1982. 2. Southwood TR, Petty RE, Malleson PN. et al: Psoriatic arthritis in children. Arthritis Rheum 32: 1007-1013, 1989. 3. Petty RE, Southwood TR, Manners 1', et al: International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: Second Revision, Edmonton 2001. .I Rheumatol 31: 390-392, 20()J. 4. Barth WF: Psoriatic arthritb sine psoriasis. III Klippel JH, Dieppe P (eds): Rheumatology. London, Mosby, 1994, pp 34.1-34.35. Roberton OM, Cabml DA, Malleson PN, Perry RE: Juvenlle psoriatic arthritis: follow-up and evaluation of diagnostic criteria. .I Rheumatol 23: 1. 166-170, 1996. 6. Petty RE, Southwood TR: Classification of childhood arthritis: divide and conquer. .I Rheumatol 25: 1869-1870, 1998. 7. Scarpa R: Juvenile psoriatic arthritis: a new clinical entity? .I Rheumatol 24: 408-409, 1997. 8. Moll JMH, Wright v: Psoriatic arthritis. Semin Arthritis Rheum 3: 55-78, 19739. Dougados M, van der Linden S, Juhlin R, et al: The European Spondylarthropathy Study Group preliminary criteria for the classification of spondylarthropathy. Arthritis Rheum 34: 1218-1227, 1991. 10. Thomson GTD, Inman RD: Diagnostic conundra in the spondyloarthropathies: towards a base for revised nosology. J Rheumatol 17: 426-429, 1990. 11. McGonagle 0, Conaghan PG, Emery 1': Psoriatic arthritis. Arthritis Rheum 42: 1080-1086, 1999. 12. Moll .1M, Wright v: Psoriatic arthritis. Semin Arthritis Rheum 3: 55-78, 1973. 13. Christophers E, Mrowietz U: Psoriasis. III Freedberg 1M, Eisen AZ, Wolff K, et al (eds): Fitzpatrick's Dermatology in General Medicine. 5th ed. New York. McGraw-Hill, 1999, pp 495-521. 14. Baker H: Epidemiological aspeClS of psoriasis. BrJ Dermatol 78: 249-261. 1966. 15. Barii-Druko V, Dobri I, Pai A, et al: Frequency of psoriatic arthritis in geneml population and among psoriatics in department of dermatology. Acta Dermatol Venereol 74 (Suppl 186): 107, 1994. 16. Green L, Meyers OL. Gordon W, Briggs B: Arthritis in psoriasis. Ann Rheum Dis 40: 366-369, 1981. 17. Church R: The prospect of psoriasis. Br J Dermatol 70: 139-145, 1958. 18. Bowyer S, Roeltcher 1', members of the Pediatric Rheumatology Database Research Group: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. .I Rheumatol 23: 1968-1974, 1996. 19. Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry, .I Rheumatol 23: 1981-1987, 1996. 20. Symmons DPM, Jones M, Osborne .I, et al: Pediatric rheumatology in the United Kingdom: data from the British Paediatric Rheumatology Group National Diagnostic Register. .I Rheumatol 23: 1975-1980, 1996. 21. Gare BA, Fasth A: Epidemiology of juvenile chronic arthritis in southwestern Sweden: a 5-year prospective population srudy. Pediatrics 90: 950-958, 1992. 22. Thumboo .I, Tham SN, Tay YK, et al: Patterns of psoriatic arthritis in Orientals. .I Rheumatol 24: 1949-1953, 1997. 23. Lambert JR, Ansell BM, Stephenson E, et al: Psoriatic arthritis in childhood. Clin Rheum Dis 2: 339-352, 1976. 24. Calabro JJ: Psoriatic arthritis in children. Arthritis Rheum 20 (Supp!): 415-416, 1977. 25. Wesolowska H: Clinical course of psoriatic arthropathy in children. Mater Med Pol 55: 185-187, 1985. 26. Harrison BJ, Silman AJ, Barren EM, et al: Presence of psoriasis does not influence the presentation or short-term outcome of patients with early inflammatory polyarthritis. J Rheumatol 24: 1744-1749, 1997. 27. Langevitz 1', Buskila 0, Gladman DO: Psoriatic arthritis precipitated by physicaltrauma. .I Rheumatol 17: 695-697, 1990. 28. Punzi L. Pianon M, Bertazzolo N, et al: Clinical laboratory and immunogenetic aspects of post-traumatic psoriatic arthritis: a study of 25 patients. Clin Exp Rheumatol 16: 277-281, 1998. 29. Telfer NR, Chalmers RJ, Whale K, Colman G: The role of streptococcal infection in the initiation of guttate psoriasis. Arch Dermatol 128: 39-42, 1992. 30. Vasey FB, Deitz C, Fenske NA, et al: Possible involvement of group A streptococci in the pathogenesis of psoriatic arthritis. J Rheumatol 9: 719-722, 1982, 31. Rantakokko K, RimpiHiinen M, UksilaJ, et al: Antibodies to streptococcal cell wall in psoriatic arthritis and cutaneous psoriasis. Clin Exp Rheumatol 15: 399-404. 1997. 32. Grininton FM, Skinner MA, Birchall NM, Tan I'll: Gamma delta + T cells from patients with psoriatic and rheumatoid arthritis respond to streptococcal antigen. .I Rheumatol 20: 982-987, 1993. 33. Thomssen H, Hoffmann B, Schank M, et al: TI,ere is no disease-specific role for streptococci-responsive synovial T lymphocytes in the pathogenesis of psoriatic arthritis, Med Micribiol Immunol (Berlin) 188: 203--207, 2000. 34. Oen K, Fast M, Postl B: Epidemiology of juvenile rheumatoid arthritis in Manitoba, Canada. 1975-1992: cycles in incidence. J Rheumatol 22: 745-750. 1995. 35. Paukkonnen K, Naukkarinen A, Horstmanheinlo M: The development of manifest psoriatic lesions is linked with the invasion of CD8+ T cell~ and CD11c+ macrophages into the epidermis. Arch Dermatol Res 284: 375-379, 1992.
332
C HAP T E R
14
PSORIATIC ARTHRITIS
36. Chang ]C, Smith LR, Froning K], et 'II: Persistence of T cell clones in psoriatic lesions. Arch Dermatol 133: 703-708, 1997. 37. Costello 1', Bresnihan B, O'Farrelly C, Fitzgerald 0: Predominance of CD8+ T lymphocytes in psoriatic arthritis.] Rheumatol 26: 1117-1124, 1999. 38. Konig A, Krenn V, Gillitzer R, et al: Inflammatory infiltrate and interleukin-8 expression in the synovium of psoriatic arthritis-an immunohistochemical and mRNA analysis. Rheumatol Int 17: 159-168, 1997. .~9. Vasey FB, Seleznick M], Fenske NA, Espinoza LR: New signposts on the road to understanding psoriatic arthritis. ] Rheumatol 16: 1405-1407, 1989. 40. Spadaro A, Scrivo R, Moretti T, et 'II: Natural killer cells and gamma/delta T cells in synovial fluid and peripheral blood of patients with psoriatic arthritis. Clin Exp Rheumatol 22: 389-394, 2004. 41. Ritchlin C, Hass-Smith SA, Hicks D, et al: Patterns of cytokine production in psoriatic synovium. ] Rheumatol 25: 1544--1552, 1998. 42. Gottlieb SL, Gilleaudeau 1', Johnson R, et 'II: Response of psoriasis to a lymphocyte-selective toxin (DAB389IL-2) suggests a primary immune but not keratinocyte, pathogenic basis. Nat Med I: 442-447, 1995. 43. Danning CL, Illei G, Hitchon C, et 'II: Macrophage-derived cytokine and nuclear factor leB p65 expression in synovial membrane and skin of patients with psoriatic arthritis. Arthritis Rheum 43: 1244--1256, 2000. 44. Ritchlin CT, Haas-Smith SA, Li 1', et 'II.: Mechanisms of TNF-alpha- ancl RANKL-mediated osteoclastogenesis and bone resorption in psoriatic arthritis.] Clin Invest 111: 821-831, 2003. 45. Canete ]D, Pablos ]L, Sanmarti R, et 'II: Antiangiogenic effects of anti-tumor necrosis factor a therapy with infliximab in psoriatic arthritis. Althritis Rheum 50: 1636--1641, 2004. 46. Wong WM, Howell WM, Coy SD, et al: Interleukin-2 is found in the synovium of psoriatic arthritis and spondyloarthritis, not in rheumatoid arthritis. Scand] JUleumatol 25: 239-245, 1996. 47. Partsch G, Wagner E, Leeb BF, et al: Upregulation of cytokine receptors sTNF-R55, sTNF-R75, and sIL-2R in psoriatic arthritis synovial tluid. ] Rheumatoi 25: 105-110, 1998. 48. Carroll ]M, Romero MR, Watt FM: Suprabasal integrin expression in the epidermis of transgenic mice results in developmental defects and a phenotype resembling psoriasis. Cell 83: 957-968, 1995. 49. Schon MP, Detmar M, Parker CM: Murine psoriasis-like disorder induced by naive CD4+ T cells. Nat Med 3: 183-188, 1997. 50. Blessing M, Schlrmacher 1', Kaiser S: Overexpression of bone morphogenic protein-6 (BMP-6) in the epidermis of transgenic mice: inhibition or stImulation of proliferation depending on the pattern of transgene expression and formation of psoriatic lesions. ] Cell Bioi 135: 227-239, 1996. 51. Tomfohrde], Silverman A, Barnes R, et 'II: Gene for familial psoriasis susceptibility mapped to the distal end of human chromosome 17q. Science 264: 1141-1145, 1994. 52. Eldar ]T, Henseler T, Christophers E, et al: Of genes and antigens: the genetics of psoriasis.] Invest Dermatol 103 (Supp!): 150S, 1994. 53. Burden AD, Javed S, Bailey M, et 'II: Genetics of psoriasis: paternal inheritance and a locus on chromosome 6p,] Invest Dermatol 110: 958--960, 1998. 54. Gladman DD, Anhorn KA, Schachter RK, Mervart H: HLA antigens in psoriatic arthritis. ] Rheumatol 13: 586--592, 1986. 55. Gladman DD, Farewell VT, Kopciuk A, Cook RJ: HLA markers ancl progression in psoriatic arthritis.] JUleumatol 25: 730-733, 1998. 56. MarS'll S, Armadens-Gil L, Martinez M, et 'II: Clinical radiographic and HLA associations as markers for different patterns of psoriatic arthritis. Rheumatology .~8: 332-337, 1999. 57. Thomson W. Barrett .TH, Donn RP, et aJ: ]uvenile idiopathic arthritis classified hy the lLAR criteria: HLA associations in UK patients. Rheumatology 41: 1183-1189, 2002. 58. Sakkas LI, Marchesoni A, Kerr LA, et al: Immunoglobulin heavy chain gene polymorphism in Italian patients with psoriasis and psoriatic arthritis. Br ] Rheumatol 30: 449-450, 1991. 59. Sakkas LI' Loqueman N, Bird H, et 'II: HLA class 1I and T cell receptor gene polymorphisms in psoriatic arthritis and psoriasis. ] Rheumatol 17: 1487-1490, 1990. 60. Hohler R, Kruger A, Schneider PM, et 'II: A TNF-alpha promoter polymorphism is associated with juvenile onset psoriasis and psoriatic arthritis. ] Invest Dermatol 109: 562-565, 1997. 61. Buskila D, Langevitz 1', Gladman DO, et 'II: Patients with rheumatoid arthritis are more tender than those with psoriatic arthritis. J Rheumatol 19: 1115-1119,1992. 62. Huemer C, Malleson PN, Cabral DA, et 'II: Pattern of joint involvement at onset differentiates oligoarticular juvenile psoriatic arthritis from pauciarticular juvenile rheumatoid arthritis. ] Rheumatol 29: 1531-1535, 2002. 63. Salvarani C, Cantini F, Olivieri I, et al: Isolated peripheral enthesitis and/or dactylitis: a subset of psoriatic arthritis . .1 Rheumatol 24: 1106-1110, 1997. 64. Reece R], Canete JD, Parsons W], et al: Distinct vascular patterns of early synovitis in psoriatic, reactive and rheumatoid arthritis. Arthritis Rheum 42, 1481-1484, 1999. 65. Griffiths C, Kirby B: Psoriasis management within primary care. Prescriber 10: 47, 1999. 66. Eastmond C], Wright V: The nail dystrophy of psoriatic althritis. Ann Rheum Dis 38: 226-228, 1979.
67. Hafner R, Michels H: Psoriatic arthritis in children. Curr Opin Rheumatol 8: 467-472, 1996. 68. Bhushan M. Moore T, Herrick AI., Griffiths CEo Nailfold capillary microscopy in psoriasis. BrJ Dennatol 142: 1171-1176, 2000. 69. Paiva ES, Macaluso DC, Edwarcls A, Rosenbaum ]T: Characterization of uveitis in patients with psoriatic arthritis. Ann Rheum Dis 59: 67-70, 2000. 70. Cabral DA, Petty RE, Malleson PN, et al: Visual prognosis in children with chronic anterior uveitis and arthritis. .1 Rheumatol 21: 2370-2375, 1994. 71. Laxer RM, Shore AD, Manson D, et 'II: Chronic recurrent multifocal osteomyelitis and psoriasis: a report of a new association and review of related disorders. Semin Arthritis Rheum 17: 260-270, 1988. 72. Pines A, Ehrenfeld M, Fisman EZ, et 'II: Mitral valve prolapse in psuriatic althritis. Arch Intern Med 146: 1371-1373, 1986. 73. Mulherin DM, FitZGerald 0, Bresnihan B: Lympheclema of the upper Iimh in patients with psoriatic arthritis. Semin Arthritis Rheum 22: 350-356, 1993. 74. Murray KJ, Luyrink L, Grom AA, et 'II: Immunohistological characteristics of T cell infiltrates in different forms of childhood onset chronic arthritis. ] Rheumatol 23: 2116--2124, 1996, 75. Canete ]D, Rodriguez ]R, Salvador G, et al: Diagnostic usefulness of synovial vascular morphology in chronic arthritis: a systematic survey of 100 cases. Semin Arthritis Rheum 32: 378--387, 2003. 76. Espinoza LR, Vasey FB, Espinoza CG, et 'II: Vascular changes in psoriatic synovium: a light and electron microscope study. Althritis Rheum 25: 677-{)84, 1982. 77. Dunky A, Neumuller .1, Menzel J: Interactions of Iymphocyles from patients with psoriatic arthritis or healthy controls and cultured endothelial cells. Clin Immunol Immunopathol85: 297-314, 1997. 78. Jones SM, Dixey], Hall ND, McHugh NJ: Expression of the cutaneous lymphocyte antigen and its counter-receptor E-selectin in the skin and joints of patients with pSOliatic arthritis. Br J RheumalOl 36: 748--757, 1997. 79. Veale D, Yanni G, Rogers S, et 'II: Reduced synovial membrane macrophage numbers, ELAM-I expression, and lining layer hyperplasia In psoriatic arthritis as compared with rheumatoid arthritis. Arthritis Rheum 36: 893-9()O. 1993. 80. Pitzalis C, Cauli A, Pipitone N, et al: Cutaneous lymphocyte antigen-positive T lymphocytes preferentially migrate to the skin but not to the joint in psoriatic arthritis. Arthritis Rheum 39: 137-145, 1996. 81. Mack M, Bruhl H, Gruber R, et al: Predominance of mononuclear cells expressing the chemokine receptor CCR5 in synovial effusions of patients with different forms of arthritis. Arthritis Rheum 42: 981-988, 1999. 82. Khan MA, Kammer GM: Laboratory findings and pathology in psoriatic arthritis. In Gerber LH, Espinoza LR (eds): Psoriatic Arthritis. Orlando, FL, Grune & Stratton, 1985, p 109. 83. Gladman OD, Stafford-Brady P, Chang CH, et 'II: Longitudinal study of clinical and radiological progression in psoriatic arthritis. .1 JUleumatol 17: 809-812, 1990. 84. Macchioni 1', Boiardi L, Cremonesi T, et 'II: The relationship between semmsoluble interleukin-2 receptor and radiological evolution in psoriatic althritis patients. Rheumatol Int 18: 1. 27-33, 1998. 85. Jenkinson T, Armas .1, Evison G, et 'II: The cervical spine of psoriatic arthritis: a clinical and radiological study. BrJ Rheumatol 33: 255-259, 1994. 86. Fournie B, GraneJ], Bonnet M, et al: Incidence of signs explaining psoriatic rheumatism in radiological involvement of the fingers and toes. Rev Rhum 59: 177-180, 1992. 87. Lehtinen A, Taavitsainen M, Leirisalo-Repo M: Sonographic analysis of enthesopathy in the lower extremities of patients with spondyloarthropathy. Clin Exp Rheumatol 12: 143-148, 1994. 88. Stoeger A, Mur E, Penz-Schneeweiss D. et al: Technetium-99m human immunoglobulin scintigraphy in psoriatic arthropathy: first results. Eur] Nucl Med 21: 342-344, 1994. 89. Bjorksten B, Back OL: Methotrexate and prednisolone treatment of a child with psoriatic arthritis. Acta Paediatr Scand 64: 664--666, 1975. 90. Abu-Shakra M, Gladman DD, Thorne JC, et 'II: Long-term methotrexate therapy in psoriatic arthritis: clinical and radiological outcome. ] JUleumatol 22: 241-245, 1995. 91. Black RL, O'Brien WM, Van Scott E], et al: Methotrexate therapy in psoriatic arthritis: douhle blind study on 21 patients. ]AMA 189: 743-747, 1964. 92. Cuellar ML, Espinoza LR: Methotrexate use in psoriasis and psoriatic arthritis. Rheum Dis Clin Nolth Am 23: 797-809, 1997. 93. Espinoza LR, Zakraoui L, Espinoza CG, et al: Psoriatic arthritis clinical response and side effects of methotrexate therapy. .1 Rheumatol 19: 872-877, 1992. 94. Falk ES, Vandbakk 0: Prevalence of psoriatic arthritis clinical response and side effects of methotrexate therapy. Acta Dermatol Venereol 73 (Supp(): 6--9, 1993. 95. Roenigk HH, Fowler-Bergfeld W, Curtis GH: Methotrexate for psoriasis in weekly oral doses. Arch Dermatol 99: 86-93, 1969. 96. Spadaro A, Taccari E, Sensl F, et 'II: Soluble interleukin-2 receptor and interleukin-6 levels: evaluation during cyclosporin A and methotrexate treatment in psoriatic arthritis. Clin Rheumatol 17: 8j-85, 1998. 97. WiIlkens RF, Williams H], WardJR, et al: Randomized, double-blind, placebo controlled triai of low-dose pulse methotrexate in psoriatic arthritis. Althritis Rheum 27: 376-381, 1984.
C H A I' T E R 98. Zacharias H, Zacharias E: Methotrexate treatment of psoriatic arthritis. Acta Dermatol Venereal 67: 270--273, 1987. 99. Ellis CN, Fradin MS, Messana ]M, et al: Cyclosporine for plaque-type psoriasis: results of a multidose, double-blind trial. N Engl] Med 324: 277-284, 1991. 100. Gupta AK, Matteson EI, Ellis CN, et al: cyclosporine in the treatment of psoriatic arthritis. Arch Dennatol 125: 507-510, 1989. 101. Macchioni P, Boiardi L, Meliconi R, et al: Serum chemokines in patients with psoriatic arthritis treated with cyclosporin A. ] Rheumatol 25: 320-325, 1998. 102. Olivieri I, Salvarani C, Cantini F, et al: Therapy with cyclosporine in psoriatic arthritis. Semin Arthritis Rheum 27: 36-43, 1997. 103. Spadaro A, Taccari E, Mohtadi B, et al: Life-table analysis of cydosporin A treatment in psoriatic arthritis: comparison with other disease-modifying antirheumatic drugs. Clin Exp Rheumatol 15: 609-614, 1997. 104. HorneiT G, Burgos-Vargas R: TNF-alpha antagonisl~ for the treatment of juvenileonset spondyloarthritides. Clin Exp Rheumatol 20 (Suppl 28) S137-S142, 2002. 105. Fraser SM, Hopkins R, Hunter ]A, et al: Sulphasalazine in the management of psoriatic arthritis. Br] Rheumatol 32: 923-925, 1993. 106. Mader R, Gladman DO, Long], et al: Injectable gold for the treatment of psoriatic arthritis: long term follow-up. Clin Invest Med 18: 139--143, 1995. 107. Palit.l, Hill ], Capell HA, et al: A multicentre double-blind comparison of auranofin, intramuscular gold thiomalate and placebo in patients with psoriatic arthritis. Br] Rheumatol 29: 28()..-283, 1990.
14
PSORIATIC ARTHRITIS
333
108. Levy ]], Paulus HE, Barnett EV, et al: A double blind controlled evaluation of azathioprine treatment in rheumatoid arthritis and psoriatic arthritis. Arthritis Rheum 15: 116-117, 1972. 109. Gladman DO, Blake R, Brubacher B, Farewell VT: Chloroquine therapy in psoriatic arthritis.] Rheumatol 19: 1724-1726, 1992. 110. K1inkhoff AV, Gertner E, Chalmers A, et al: Pilot study of etretinate in psoriatic arthritis. ] Rheumatol 16: 789--791, 1989. 111. Gupta AK, Ellis CN, Tellner DC, et al: Double blind, placebo controlled study to evaluate the efficacy of fish oil and low dose UVB in the treatment of psoriasis. Br] Dennatol 120: 801--807, 1989. 112. Huckins 0, Felson DT, Holick M: Treatment of psoriatic arthritis with oral 1,25-dihydroxyvitamin 03: a pilot study. Arthritis Rheum 33: 1723-1727, 1990. 113. Hicken G], Kitaoka HB, Valente RM: Foot and ankle surgery in patienl~ with psoriasis. Clin Orthop Rei Res 300: 201-206, 1994. 114. Zangger P, Gladman DO, Bogoch ER: Musculoskeletal surgery in psoriatic arthritis.] Rheumatol 25: 725-729, 1998. 115. Lubrano E, Helliwell P, Parsons W, et al: Patient education in psoriatic arthritis: a cross sectional study on knowledge by a validated self-administered questionnaire. ] Rheumatol 25: 1560--1565, 1998. 116. sills EL: Psoriatic arthritis in childhood. Johns Hopkins Med J 146: 49, 1980.
( HAP T E R
15
ARTHROPATHIES OF INFLAMMATORY BOWEL DISEASE Carol B. Lindsley and Ronald M. Laxer
I-~
DEFINITION AND CLASSIFICATION The arthropathies of inflammatory bowel disease (IBD) may be defined as any noninfectious arthritis occurring before or during the course of either Crohn's disease (CD; regional enteritis) or ulcerative colitis CUC). Arthritis is probably the most common extraintestinal complication of these disorders. There are two patterns of joint inflammation: peripheral polyarthritis and, less commonly, involvement of the sacroiliac (S1) joints and axial skeleton. Arthritis associated with IBD is not included in the American College of Rheumatology (ACR) classification of childhood arthritis but is included in both the European League Against Rheumatism (EULAR) and International League of Associations for Rheumatology (ILAR) criteria (see Chapter 9).
EPIDEMIOLOGY Incidence and Prevalence Arthropathy has been reported in 7% to 21% of children with IBDI-5 (Table 15-1). Passo and colleagues' found arthritis in 9% of 44 children with DC and in 15.5% of 58 children with CD. Arthralgia was much more common, occurring in 32% of those with DC and 22% of those with CO.' Differentiation of DC from CD is not always easy, and differences in the reported frequencies of arthritis in each may reflect the accuracy of diagnosis in these types of IBD.!..1 Other children have myalgia, skeletal pain associated with glucocorticoid-induced osteopenia, or secondary hypertrophic osteoarthropathy without objective arthritis. Although there has been concern about an increasing incidence of IBD worldwide, a recent Swedish study reported a stable and unchanged rate of DC and CO during the past 30 years. 6
Age at Onset and Sex Ratio In a study of 136 patients with onset of IBO before the age of 20 years,4 age at onset did not differ in patients with and without arthritis. The ratios of boys to girls in
334
those with and without peripheral arthritis were almost identical, although the five children who developed spondylitis were boys.
ETIOLOGY AND PATHOGENESIS The causes of both IBD and the accompanying arthritis are obscure. The possible roles of gastrointestinal (GI) infections or allergic reactions to foods absorbed across an inflamed mucosa remain speculative. The SI arthritis probably shares its etiology with that of ankylosing spondylitis (AS), and studies of associated enteric species and immunity to them may be relevant. 7 Peripheral arthropathy may involve entirely different immunoinflammatory mechanisms (immune complexes), however, and it is clinically more closely related to the activity of the intestinal disease. Picco and colleaguesB found increased gut permeability in all subtypes of juvenile arthritis using the lactulose/mannitol test, but IBO patients with spondyloarthropathy had the highest levels. Reciprocally, subclinical gut inflammation in the majority of patients with seronegative spondyloarthropathy has been describedY
GENETIC BACKGROUND There is a pronounced tendency for familial, racial, and ethnic clustering of DC and CO. Hamilton ancl associates! reported that approximately 15% of children with DC and 8% of those with CD had first-degree relatives with IBO. Both diseases are more common in children of Jewish descent, who composed 21% of the IBD population but only 2% of the general population in one study.! Published reports support the view that genes of the major histocompatibility complex are important in determining susceptibility to DC in particular, \tl but inherited predispositions are undoubtedly polygenic. In Japanese!! and Jewish patients,12 but not in other ethnic groups, the human leukocyte antigen (HLA) DRBl*1502 (OR2) allele is increased in frequency. It is estimated that SI arthritis is at least 30 times more common in patients with IBO than in the general population,13 a fact that reflects the
C HAP T E R
• TABLE 15-1
IS
ARTHROPATHIES OF INFLAMMATORY BOWEL DISEASE
335
Arthritis in Inflammatory Bowel Disease in Children
Author and Year (ref. no.) Farmer & Michener, 1979 (1) Hamilton et aI., 1979 (2) Burbige et aI., 1975 (3) Lindsley & SchaUer, 1974 (4) Passo et a!., 1986 (5) Lindsley & SchaUer, 1974 (4) Hamilton et aI., 1979 (2) Passo et aI., 1986 (5)
Disease
No. Patients
No. with Arthritis
% with Arthritis
CD CD CD CD CD DC DC DC
522 58 58 50 58 86 87 44
39 11 6 5 9 18 8 4
7 19 10 10 15 21 9 9
CD. Crohn's disease (regional enteritis); lIC, ulcerative colitis.
high frequency of HLA-B27 in such patients. The peripheral polyarthritis accompanying IBD has no known HLA association. Recent studies have identified NOD2/Card 15 variants that are associated with CD in both children and adults, particularly in those with ileal disease and lower weight at time of diagnosis. 14 However, no association has been reported with articular involvement nor AS.IS
CLINICAL MANIFESTATIONS Arthritis and Enthesltls Two distinct patterns of joint disease occur. The more common one in patients with IBD is inflammation affecting peripheral joints. Lower-extremity joints, especially ankles and knees, are most frequently affected,3-5 although upper-extremity joints, occasionally also including small joints of the hand and the temporomandibular joints, may be involved. Lindsley and Schaller4 reported that four or fewer joints were affected at onset or during the course of the illness in 11 of 18 children; in 5 children, S to 9 joints were affected; in only 2 children were more than 10 joints affected, including small joints of the hand. Episodes of acute peripheral arthritis are usually brief, lasting 1 or 2 weeks (occasionally longer), and tend to recur." In some children, arthritis may last for several months, particularly if the GI disease is active. Rarely, joint inflammation persists for months, although permanent functional loss or joint damage is unusual. However, erosive disease has been described in young adults with juvenile-onset disease. 16.17 Whereas the SI arthritis bears little relation to the activity of the gut disease, the peripheral arthritis reflects the activity and course of the GI inflammation. A clinical flare-up in a child's arthritis is suggestive of poor control of the underlying IBD. In adult patients, additional clinical phenotypes have been described in the peripheral arthropathy group (non-HLA-B27 associated): type I, which is similar to the previously described disease and is frequently associated with uveitis and erythema nodosum (EN), and type II, which is a symmetrical polyarthritis that is independent of IBD activity, of longer duration, and rarely associated with EN. 18 SI arthritis, which may be asymptomatic but often is characterized by pain and stiffness in the lower back,
buttocks, or thighs, is a much less common complication of IBD than is polyarthritis. It is sometimes accompanied by enthesitis identical to that occurring in other forms of spondyloarthritis. SI arthritis may also be associated with chronic symmetrical oligoarthritis predominantly affecting the joints of the lower limbs: 5% to 10% of established cases of AS in adults are associated with chronic IBD.19 Also, an additional category of asymptomatic SI disease 08% ) occurs in patients with IBD, most often in those with greater disease duration. 20 Hypertrophic osteoarthropathy is a relatively rare, very painful musculoskeletal complication of IBD. 21 The pain occurs symmetrically in the limbs (rather than the joints) and may be accompanied by increased sweating and purple discoloration of the affected limbs. Osteoporosis can be a significant component of articular disease or, rarely, a presenting manifestation when associated with fractures. 22 Patients treated with steroids are especially at high risk, and in addition they may develop avascular necrosis, most commonly involving the femoral head. Chronic recurrent multifocal osteomyelitis has been associated with IBD in some patients. 23
Gastrointestinal Disease and Extra-articular Manifestations Gastrointestinal Disease Cramping abdominal pain, often with localized or generalized tenderness, anorexia, and diarrhea, sometimes occurring at night, is characteristic of IBD. Differentiation of DC and CD on the basis of GI symptoms alone is unreliable, although bloody diarrhea is highly suggestive of DC, whereas perianal skin tags and fistulae are typical of CD (Table 15-2). GI symptoms usually precede joint disease by months or years, although occasionally both systems are affected simultaneously or joint symptoms precede intestinal disease. In the latter case, the arthritis resembles that of juvenile rheumatoid arthritis, juvenile AS, or the seronegative enthesopathy and arthropathy (SEA) syndrome, with a course punctuated by intermittent abdominal pain that may be incorrectly ascribed to the effects of anti-inflammatory drugs. Low-grade diarrhea, anemia, unexplained fever, weight loss, growth retardation out of proportion to the extent and activity of the joint disease, or a family history of IBD should alert the physician to the possibility of
336 ~~
C HAP T E R
IS
ARTHROPATHIES OF INFLAMMATORY BOWEL DISEASE
TARLE 15 2 Gastrointestilldl and Otlll'r Systemi< Diseasp in Childrpn with Inflallllllatory Bowel Diseases
Symptom or Sign Diarrhea Hematochezia Abdominal pain Weight loss Fever Vomiting Perianal disease Finger clubbing Erythema nodosum Oral lesions Uveitis Pyoderma gangrenosum
Ulcerative Colitis
Crohn's Disease
++++ ++ ++ ++
++ + +++ ++++ +++ ++ +++ ++
+ + +
+ +
+ (+) (+)
Frequency: (+). rare; +, <25%; ++.25-50%; +++.50-75%; ++++,
+ + (+) (+) ~75%.
occult IBD. Mucocutaneous lesions (EN, aphthous stomatitis, pyoderma gangrenosum) seem to be more common in children who have arthritis (especially peripheral arthritis) as a complication of IBD, although this association is not supported by some clinical studies. s Although there are no clearcut correlations between the extent of GI inflammation and arthritis, most reports support the view that there is a higher frequency of arthritis in children with extensive, as opposed to segmental, bowel disease,1,2,5 Patients with arthritis usually have active gut disease, although the onset of arthritis is not necessarily related to obvious flare-ups in GI tract inflammation. The occurrence of first-time joint symptoms after proctocolectomy for DC has been associated with the development of "pouchitis. "24
Cutaneous Disease Erythema Nodosum The lesions of EN (nodular panniculitis) occur most commonly in the subcutaneous fat of the pretibial region (Fig. 15-1) as erythematous, painful, slightly elevated lesions, 1 to 2 cm in diameter, that erupt in groups and reappear sequentially in new areas after several days. The nodules tend to persist for several weeks and recur in crops for several months. As they heal, they frequently leave pigmented areas that persist for many months, Articular pain and synovitis accompany each exacerbation in approximately two thirds of instances. Erythema nodosum is more likely associated with peripheral arthritis that is of short duration and involving few joints, lH Although EN may also occur as a distinct, isolated clinical syndrome, it is commonly associated with systemic i1lness of diverse causes, including IBD.2s,26
Pyoderma Gangrenosum The lesions of pyoderma gangrenosum may occur alone or in concert with IBD (Fig. 15-2). They often arise after minor trauma, may be single or multiple, and usually begin as a pustule that breaks down and rapidly enlarges to form a chronic, painful, deep, undermined ulcer with a red, raised border. They have rarely been reported in children but may in fact be the initial clinical manifestation. In adults, the lesions occur with IBD, rheumatoid arthritis, or other systemic diseases.D A single report, not confirmed, of pyoderma gangrenosum in a 2-year-old boy with joint effusions but without IBD was associated with enhanced leukocyte mobiJity.2H
• Figure 15-1 Erythema nodosum. A,This young girl had tender, drcumscribed purple-red nodules on the shins. 8, lesions on the forearm of a child.
C HAP T E R
IS
ARTHROPATHIES OF INFLAMMATORY BOWEL DISEASE
337
PATHOLOGY The histopathology of the synovitis of IBD is nonspecific with proliferation of lining cells and inflltration of the synovium with lymphocytes, plasma cells, and histiocytes. 40 Granulomatous synovitis occasionally occurs. 4J For a discussion of the full spectrum of the histopathology of lBO, the reader is referred to current textbooks of gastroenterology.
DIAGNOSIS AND LABORATORY EXAMINATION
• FIgure 15-2 Pyodenna gangrenosum on the upper back of a child.These lesions begin as nodules but progress to ulcers with considerable loss of subcutaneous tissue.
Vasculitis Vasculitis of several types has been reported in patients with IBO and arthritis. Involvement of large vessels was found in at least two studies. 29.3o Takayasu's arteritis in patients with CO was first described in 197031 and has been reported in several other adults and in a 15-yearold boy,32 as well as in a young adult with UC and juvenile AS.33 It seems unlikely that coincidence could account for the simultaneous occurrence of these rare diseases, but the data are insufficient to allow for certainty. A syndrome of cutaneous vasculitis, glomerulonephritis, and circulating immune complexes was reported in two adults with IBO and spondyloarthritis (one with juvenile-onset colitis).34 Immunoglobulin A nephropathy has been described in AS and in at least three patients with IBO.35.36 Cutaneous vasculitis was reported as the presenting feature in a 14-year-old girl with COY
Uveitis Lyons and Rosenbaum 38 compared the characteristics of uveitis in 17 adults with IBO and 89 patients with spondyloarthritis. Twelve of the 15 patients with uveitis and IBO had CD, and 82% were female. Uveitis accompanying IBO was usually bilateral, posterior, and of insidious onset and chronic duration. The frequency of HLA-B27 was half that in the spondyloarthritis group. Episcleritis, scleritis, and glaucoma were more common among patients with IBO. At least in adults, the uveitis associated with IBO was frequently complicated by cataract (35%), glaucoma (24%), cystoid macular edema (24%), or posterior synechiae (29%).38 There are no reported studies of uveitis in children with IBD and arthritis. However, it is known that children with IBO may develop asymptomatic uveitis. 39
Making the diagnosis of arthritis associated with IBD rests on recognition of the significance of this association and on a high level of clinical suspicion. A diagnosis of IBD should be suspected in any child with arthritis accompanied by lower abdominal pain, hematochezia, unexplained weight loss, anemia, fever, or poor growth. Occult GI blood loss can be verified by repeated stool guaiac examinations. This suspicion would be supported by laboratory evidence of inflammation (high erythrocyte sedimentation rate and other acute phase reactants, low serum albumin), and negative results for rheumatoid factor and antinuclear antibody tests. Antibodies to neutrophil cytoplasmic antigens (pANCA-a perinuclear pattern on immunofluorescence) and anti-Saccharomyces cerevisiae antibody (ASCA) are frequently present in the sera of children with IBO. 42 Tests for antineutrophil cytoplasmic antibody (ANCA) were positive in 73% of children with UC and 14% of those with CO.43 In spite of the known association of this autoantibody with systemic vasculitis, vasculitis does not appear to be more frequent in ANCA-positive patients with IBO.44 Synovial fluid analyses of children with IBD have not been reported, although in adults counts of synovial fluid white blood cells have ranged from 5,000 to 15,000/mm3 (5 to 15 x 109/L), with a predominance of neutrophils. Synovial fluid protein, glucose, and hemolytic complement levels have been norma1. 45
RADIOLOGIC EXAMINATION Radiographs of peripheral joints document only soft tissue thickening and joint effusions. SI arthritis, when it occurs, is not clearly distinguishable from that associated with juvenile AS. Spur formation, the result of enthesitis, is sometimes identified at the insertion of the plantar fascia into the calcaneus. Periostitis may be demonstrable by radiography or by radionuclide scanning (Fig. 15-3). Burbige and coworkers3 noted erosive lesions secondary to granulomatous synovitis in one child. Although radionucleotide scanning or magnetic resonance imaging is optimal for documenting early changes in SI joints, highresolution computed tomography (HRCT) is more reliable in detecting erosions and documenting calcifications.46
TREATMENT Successful management of the peripheral arthritis generally depends on effective treatment of the GI disease: Control of the primary disease usually results in remission of the
338
C HAP T E R IS
ARTHROPATHIES OF INFLAMMATORY BOWEL DISEASE
Methotrexate and anti-TNF agents may also be helpful in patients who have no response to initial therapy. A physical therapy program with range-of-motion exercises to maintain back and chest motion (as for juvenile AS) may help to prevent or slow the effects of the disease. Custom-made orthotics may be useful to minimize pain secondary to enthesitis around the foot. Vasculitis accompanying lED should be treated with systemic glucocorticoids. Topical glucocorticoids are used to treat uveitis.
COURSE OF THE DISEASE AND PROGNOSIS The outcome of the GI disease is the most important determinant of overall prognosis in the child with lED and arthritis. I Prognosis of the peripheral joint disease is usually excellent, although axial disease may progress independent of the course of the GI inflammation, Permanent changes in the spine and hips are frequent in this group of children. Poor nutrition and accompanying growth retardation may be major problems in poorly controlled disease, • Rgure 15-3 Bone scan documents increased uptake in the sacroiliac joints and along the femoral shafts, representing sacroiliac joint arthritis and periostitis in a 14-year-old girl with ulcerative colitis.
peripheral arthritis. Colectomy in DC may be followed by a striking remission in peripheral joint symptoms, although colectomy for the control of peripheral joint arthritis alone is certainly not indicated. Peripheral arthritis may be managed with nonsteroidal anti-inflammatory agents, but there is increasing evidence that these dmgs can exacerbate IBDY Selective cyclooxygenase-2 inhibitors may be preferred in this situation. Early use of sulfasalazine or glucocorticoids may provide the best management of the arthropathy of IBD directly by way of a beneficial effect on the GI inflammation, although no therapeutic trials have been published. For persistent arthritis in one or two joints, intra-articular glucocorticoid should be considered. In CD, the use of budesonide, predominantly a topically acting steroid, resulted in the remission of joint symptoms in 74% of affected patients. 4H Methotrexate results in improvement of both GI symptoms and arthritis in CD. 49 Anti-TNF therapy, particularly infliximab, produced prompt, substantial improvement in CD of GI symptoms as well as arthritis,SO As these potent anti-inflammatory drugs are used earlier and more aggressively in the treatment of IBD, it is possible that the associated arthropathies will decrease in frequency or severity. The HLA-B27-associated spondylitis of lED is much more likely than the peripheral arthritis to persist and progress without remission, independent of the activity of the GI disease and unaffected by procedures such as colectomy. It is therefore much more difficult to manage in the long term. Sulfasalazine is the initial dmg choice, in a dose of 30-50 mg/kg/day to a maximum of 2.5 g/day.
REFERENCES I, Farmer RG, Michener WM: Prognosis of CrollO's uisease with onset in childhoou or auolescence, Dig Dis Sci 24: 7~2. 1979. 2. Hamilton JR, Bruce MD, Abuourhaman M, et al: Inllammatory bowel uiseast' in children and adolescents. Auv Pediatr 26: 311-41, 1979. 3. Burbige EJ. Shi-Shung H. Bayless TM: Clinical manifestalions of Crohn's disease in children and adolescents. Pediatrics 5~: 866-71. 1975. 4. Lindsley C, Schaller JG: Arthritis associaled with inllammalory bowel disease in children. .I Pedialr 84: 16-20. 1974. ~. Passo MH, Fit1.geralu JF, Brandt KD: Althritis associated with inflammatory bowel uisease in chiluren: relationship of joint uisease to aClivity and severity of bowel lesion. Dig Dis Sci 31: 492-497, 1986. 6. Bensen BS, Moum B, Ekbom A: Incidence of Lnllammalory bowel disease in children in southeastern Norway. Scand.l Gastroenterol 37: 54ll--54~, 2002. 7. Keat A: Infections and Ihe immunopalhogenesis of sernnegalive spondyloarthropathies. Curl' Opin Rheumalol 4: 494-499, 1992. 8, Picco P, Gattorno M, Marchese N, el al: Increased gut permeability in juvenile chronic arthritiues: a multivariate analysis of the uiagnostic parameters. Clin Exp Rheumatol 18: 773-778, 2000. 9. Devos M, Cuvelier C, Mielants H. et al: lleocolonoscopy in seronegative spondyloat1hropathy. Gastroenterology 96: 339-344, 1989. 10. Satsangi.l, Jewell DP, Bell .11: The genetics of inllammatory bowel uisease. Gut 40: 572-~74, 1997. 11. Futami S, Aoyama N. Honsako Y, et al: HLA-DRBI'I ~02 alleles. subtype of DR15 is associated with susceptibility to ulcerative colitis and its progression. Dig Dis Sci 40: 814--818, 1995. 12. Toyoua H, Wang S-}. Yang H, et al: Distinct association of HlA class 11 gent·s with inllammatory bowel disease. Gastroenterology 104: 741-748, 1995. 13. Brewet10n DA, James DCO: The histocompatibility antigen HLA-27 anu disease. Semin Arthritis Rheum 4: 191-207, 1975. 14. Tomer G, Ceballos C. Concepcion E, Benkov l\J: NOD2/CAR[)I~ variants are associated with lower weight at diagnosis in children with Crohn's disease. Am.l Gastroenterol 98: 2479-2484. 2003. I~. Ferreiros-Vldal 1, Amarelo .1, Barros F, et al: Lack of association of ankylosing spondylitis with tile most cnmmon NOD2 susceptibility alleles in Crolm's disease. J Rheumatol 30: 102-104, 2003. 16. e1 Magh~aoui A, Aouragh A. Hachlm M, et 'II: Erosive arthritis in juvenile onset Crohn's disease. Clin Exp Rheumato; 18: 541, 2000. 17. Benbouazza K, Bahiri R. Krami HE, et al: Erosive polyarthritis in Cmhn's disease: report of a case. Rev Rlulln 66: 743-746, 1999. 18. Orchard TR. Woruswol1h BP. Jewell DP: Peripheral arthropathies in intLtmmatory bowe! disease: their articular distribution and natural history. Gut 42: 387-391. 1998. 19. Wordsworth P: Arthritis and inllammalory bowel disease. Curl' Rheumatol Rep 2: 87--81>, 2000. 20. de Vlam K, Mielants H. Cuvelier C, et al: Sponuyloal1hropathy is underestimated in inllammatory bowel disease: prevalence and HLA association. .I Rheum 27: 21>60-2865, 2000. 21. Neale G. Kelsall AR, Doyte FH: Cmhn's disease and diffuse symmetrical periostitis. Gut 9 383-387. 19(,8.
C HAP T E R
IS
ARTHROPATHIES OF INFLAMMATORY BOWEL DISEASE
22. Thearle M, Horlick M, Bilezikian]p, et al: Osteoporosis: an unusual presentation of childhood Crohn's disease. Endocrinol Metab 85: 2122-2126, 2000. 23. Huher AM, Lam PY, Durry CM. et al: Chronic recurrent multifocal osteomyelitis: clinical outcomes after more than five years of follow-up. J Pediatr 141: 198--203, 2002. 24. Balhir-Gurman A, Schapira 0, Nahir M: Arthritis related to ileal pouchitis following total proctocolectomy for ulcerative colitis. Semin Arthritis Rheum 30: 242-248, 2001. 25. Lorher.l: TI1e changing etiology of erythema nodosum in children. Arch Dis Child 33: 137-141, 1958. 26. Winkelmann RK, Forstrom L: New observations in the histopathology of erythema nodosum. J Invest Dermatol 65: 441-446, 1975. 27. Hurwitz S: Clinical Pediatric Dermatology. 2nd ed. Philadelphia, WB Saunders, 1993, p 684. 28. .Iacohs .IC, Goetzl EJ: "Streaking leukocyte factor," arthritis and pyoderma gangrenosum. Pediatrics 56: 570-578, 1975. 29. Yassinger S, Adelman R, Cantor 0: Association of inflammatory bowel diseaSe and large vascular lesions. Gastroenterology 71: 844-846, 1976. 30. Gorrnally S, Bourke W. Kierse B, et al: Isolated cerebral thrombo-ernbolism and Crohn disease. Eur .I Pediatr 154: 815-818, 1995. 31. Soloway M, Moir TW, Linton OW: Takayasu's arteritis: report of a case with unusual findings. Am.l Cardiol 25: 258--263, 1970. 32. Hilario MO, Terreri MT, Prismich G, et al: Association of ankylosing spondylitis, Crohn's disease and Takayasu's arteritis in a child. Clin Exp Rheumatol 16: 92-94, 1998. 33. Aoyagi S, Akashi H, Kawara T, et al: Aortic root replacement for Takayasu arteritis associated with ulcerative colitis and ankylosing spondylitis-report of a case. Jpn CircJ 62: 64-68, 1998. 34. Peeters A.I, van den Wall BAW, Daha MR, Breeveld FC: Intlammatory bowel disease and ankylosing spondylitis was associated with cutaneous vasculitis, glomerulonephritis and circulating 19A immune complexes. Ann Rheum Dis 49: 638--640, 1990. 35. Dard S, Kenollch S, Mery JP, et al: A new association: ankylosing spondylitis (AS) and Berger disease (B])). Kidney Int 24: 129, 1983. 36. McCallum 0, Smith L, Harley F, et al: IgA nephropathy and thin basement membrane disease in association with Crohn disease. Pediatr Nephrol II: 637-<">40, 1997.
339
37. Kay MH, Wyllie R: Cutaneous vasculitis as the initial manifestation of Crohn's disease in a pediatric patient. Am .I Gastroenterol 93: 1014, 1998. 38. Lyons JL, Rosenbaum JT: Uveitis associated with inflammatory howel disease compared with uveitis associated with spondyloarthropathy. Arch Ophthalmol 115: 61-64, 1997. 39 Hofley P, Roarty.l, McGinnity G. et al: Asymptomatic uveitis in children with chronic inflammatory howel disease. J Pediatric Gastroenterol Nutri 17, 397-400, 1993. 40. Ansell BM, Wigley RAD: Arthritis manifestations in regional enteritis. Ann Rheum Dis 23: 64-72, 1964. 41. Lindstrom H, Wramsby H, Ostherg G: Granulomatous arthritis in Crohn's disease. Gut 13 257-259, 1972. 42. Khan K, Schwarzenberg SJ, Sharp H, et al: Role of serology and routine laboratory tests in childhood intlammatory bowel disease. Intlamm Bowel Dis 8: 325-329, 2002. 43. Olives JP, Breton A. Hugot JP, et al: Antineutrophil cytoplasmic antihodies in children with int1ammatory bowel disease: prevalence and diagnostic value. .I Pediatr Gastrol Nutr 25: 142-148, 1997. 44. Rosa C, Esposito C, Caglioti A, et al: Does the presence of ANCA in patients with ulcerative colitis necessarily imply renal involvement! Nephrol Dial Transplant 11: 2426-2429. 1996. 45. Bunch TW, Hunder GG, McDuffie FC, et al: Synovial tluid complement determination as a diagnostic aid in int1ammatory joint disease. Mayo Clin Proc 49: 715-720, 1974. 46. Mester AR, Makb EK, Karlinger K, el al: Enteropathic arthritis in the sacroiliac joint: imaging and different diagnosis. Elir J Radiol 35: 199-208, 2000. 47. Evans JM, McMahon AD, Murray FE, et al: Non-steroidal anti-inflammatory drugs are associated with emergency admission to hospital for colitis due to inflammatory bowel disease. Gut 40: 619-622, 1997. 48. Florin THJ, Graffner H, Nilsson LG, Persson T: Treatment of joint pain in Crohn's patients with budesonide controlled ileal release. Clin Exp Pharmacol Physiol 27: 295-298, 2000. 49. Mack DR, Young R, Kaufmann 5S, et al: Methotrexate in patients with Crohn's disease after 6-mercaptopurine. .1 Rheumatol 132: 83Q-835 , 1998. 50. Van den Bosch F, Kruithof E, De Vos M, et al: Crohn's disease associated with spondyloarthropathy: effect of TNF-alpha hlockade with infliximab on articular symptoms. Lancet 356: 1821-1822, 2000.
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
Ross E. Petty and Ronald M. Laxer
I]'
Systemic lupus erythematosus (SLE) is an episodic, multisystem, autoimmune disease characterized by widespread inflammation of blood vessels and connective tissues and by the presence of antinuclear antibodies (ANAs), especially antibodies to native (double-stranded) DNA (dsDNA). Its clinical manifestations are extremely variable, and its natural history is unpredictable. Untreated, SLE is often progressive and has a significant fatality rate.
1. SLE is an episodic disease. A history of intermittent symptoms (e.g., arthritis, pleuritis, dermatitis) may precede the diagnosis by months or years. 2. SLE is a multisystem disease, and children usually present with symptoms and signs of disease affecting more than one organ system. 3. SLE is characterized by the presence of antinuclear antibodies (especially those directed to dsDNA) and of other autoantibodies.
HISTORICAL REVIEW The word lupus, derived from the Latin word for wolf, was originally used in medicine from the 13th to the 19th centuries to describe a dermatitis characterized by recurrent, florid facial ulcerations.! The acute and chronic types of the skin disease were first clarified by Kaposi in 1872. 2 In 1895, Osler recognized the systemic nature of this disease, its characteristic exacerbations and remissions, and suggested that "erythema exudativum" was a form of vasculitis:' Cardiac involvement was described in detail by Libman and Sacks in 1924,4 and by Gross in 1940.; The clinical features of SLE as recognized today, however, were first delineated by Baehr, Klemperer, and Schifrin in 1935. 6 They emphasized that characteristic visceral involvement could occur in the absence of the typical cutaneous lesions. In 1948, description of the lupus erythematosus (LE) cell by Hargraves, Richmond, and Morton7 permitted recognition of a hroader spectrum of patients with SLE. The demonstration of ANAs by the more sensitive technique of indirect immunofluorescence microscopy allowed more accurate diagnosis in patients with less typical presentations. Identification of an antibody to dsDNA in the serum and in the pathologic lesions of patients with the disease led to the concept of immune complex disease as an explanation for the pathogenesis of many of the features of SLE. The role of inflammation evoked by soluble immune complexes by way of complement activation was soon demonstrated. Later, altered cell-mediated immunity in active SLE and, more recently, the importance of immunogenetic predispositions were documented. The introduction of glucocorticoid dntgs for the treatment of connective tissue diseases represented a major advance in management. It became possible not only to prolong the lives of severely affected patients and control acute manifestations of the disease, but also to follow the unfolding course of the illness in successfully treated patients.
DEFINITION AND CLASSIFICATION The diagnosis of SLE is a clinical one and is supported by specific laboratory abnormalities. Three prominent features are characteristic of the diagnosis:
342
Criteria for the Classification of Patients The most widely used criteria for the classification of SLE are those of the American College of Rheumatology (ACR), which were revised in 19828 and modified in 1997 (Table 16-1).9 Although designed as classification criteria, they are widely used for diagnosis. In adults, the presence of four criteria has a sensitivity and specificity of 96%. Evaluations of the 1982 criteria in childhood SLEw concluded that they had a sensitivity of 96% and a specificity of 100% in childhood lupus, compared with a rheumatic disease control group. Of 103 children with SLE in this study, 99 fulfilled four or more criteria; none of the 101 control subjects did so, and only three of the control subjects fulfilled three criteria. A number of studies have validated the 1982 criteria in adults. ll ,!2 The 1997 modification has not yet been validated, and concern about the nonspecificity of the anticardiolipin assay has been raised.!3 An apparent redundancy that has not been addressed is the fact that a positive test for ANA is one criterion, whereas antibodies to dsDNA or to Sm antigen are components of another criterion. Although almost all patients with SLE have ANA, its presence in low or moderate titer has very low specificity for the disease; and, because it is almost certain that patients with antibodies to dsDNA or Sm also have ANA, it seems evident that ANA should be removed as a criterion. Some patients with what the experienced clinician would call SLE do not fulfill the ACR classification criteria. A study from Sweden reported that more than half of patients with "incomplete" SLE fulfilled four or more of the ACR criteria after a median time of 5.3 years, and that the presence of a malar rash and anticardioJipin antibodies predicted this evolution. I. Further proposed modifications of the ACR criteria l ;.!!> may be more sensitive (if less specific) but have not been evaluated in children or adolescents.
C HAP T E R
I!. III
TI\BI [16-1 Criteria for the Cldssificdtion of Svsh'mi( lupus Erythemdtosus
ACR 1982 Crlterla*
ACR 1997 CrIteriat
Malar (butterfly) rash Discoid-lupus rash Photosensitivity Oral or nasal mucocutaneous ulcerations Nonerosive arthritis Nephritis! Proteinuria >0.5 g/day Cellular casts Encephalopathy! Seizures Psychosis Pleuritis or pericarditis Cytopenia Positive immunoseroiogyl Antibodies to dsDNA Antibodies to Sm nuclear antigen Positive LE-cell preparation
Malar (butterfly) rash Discoid-lupus rash Photosensitivity Oral or nasal mucocutaneous ulcerations
Biologic false-positive test for syphilis
Positive antinuclear antibody test
Nonerosive arthritis Nephritis! Proteinuria >0.5 g/day Cellular casts Encephalopathy! Seizures Psychosis Pleuritis or pericarditis Cytopenia Positive immunoserologyl Antibodies to dsDNA Antibodies to Sm nuclear antigen Positive finding of antiphospholipid antibodies based on: 1. IgG or IgM anticardiolipin antibodies, or 2. Lupus anticoagulant, or 3. False positive serologic test for syphilL~ for at least 6 mo, confirmed by Treponema pa/lidum immobilization or fluorescent treponemal antibody absorption test Positive antinuclear antibody test
ACR, American College of Rheumatology; IgG, immunoglobulin G: IgM, immunoglobulin M: LE, lupus erythematosus. 'Adapted ffOm Tan EM, Cohen AS, Fries JF, et al: The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 25: 1271-1277. 1982. tAdapted from Ferraz MH, Goldenberg J, Hilario MO, et al: Evaluation of the 1982 ARA lupus criteria data set in pediatric patients. Clin Exp Rheumatol 12: 83--137, 1994. 'Anyone item satisfies that criterion.
EPIDEMIOLOGY Inddence and Prevalence Several studies in adults 17-21 have estimated the incidence of SLE at 2,0 to 7.6 per 100,000 per year, and the prevalence at 12 to 50 per 100,000 individuals. Data in children are few. Early studies from the United States estimated incidence at O.5Yo and 0,60 21 per 100,000 per year. From national registries, it is estimated that the mean annual incidence is 0.36 per 100,000 (confidence interval [CI], 0.23 to 0.61) in Canadian children 22 and 0.37 23 to 0.9 per 100,000 per year (CI, 0.10 to 0.94)24 in Finnish children, Using a questionnaire survey of pediatric departments of 1290 hospitals, the frequency of SLE in children in Japan was estimated at 0.47 per 100,000 per year. 2'; The estimate in the Canadian study of Malleson and colleagues22 may be the most accurate, because all patients submitted to the registry were evaluated by pediatric rheumatologists.
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
343
Data from pediatric rheumatic disease registries confirm that SLE accounts for fewer than 1% of patients in pediatric rheumatology clinics in the United Kingdom,26 1.5% to 3% of patients in Canada,22 and 4.5% of patients in the United States. 27 These differences probably reflect many factors, including referral patterns and ethnic diversity. Although there are no accurate prevalence data, it has been inferred that there are between 5,000 and 10,000 children with SLE in the United States. 2H It has been suggested that the overall incidence of SLE may be increasing. 29 It is difficult to be certain whether this reflects the effects of better ascertainment rather than a true increase in incidence. There are no data addressing this question in children and adolescents,
Age at Onset The proportion of all patients with onset of SLE in childhood has been estimated at 15% to 17%.30,31 This information dates from the 1950s and 1960s, however, and it is likely that the proportion now is higher, Onset of SLE is rare before 5 years of age and uncommon before adolescence, when it is almost as frequent as in any subsequent decade.
Sex Ratio Women outnumber men in ratios ranging from 5: 1~2 to 10: 1~3 in large series of adults with SLE. In childhood, girls are affected 4.5 times more frequently than boys, although the overall ratio varies with age at onset. 34-37 In one study,34 the ratio of girls to boys with SLE in the 0- to 9-year-old age range was 4:3; in the 10- to 14-yearold age range, it was 4:1, and in the 15- to 19-year-old range, it was 5: 1. Three of the youngest patients with SLE were boys, including the youngest child, who had disease onset at the age of 3 years. A similar trend was noted in a study by King and associates, ~8 in which girls outnumbered boys by 3: 1 in the group with onset before 12 years of age and by almost 10:1 in the group with onset after 12 years of age. This difference in sex distribution is not present in all series, however, and a large study of SLE in children found no difference in the sex ratio of children who were younger or older than 10 years of age at onset of disease. 28
Influence of Geography and Race SLE is recognized worldwide. In studies of 234 children with SLE in the United States in whom racial origin was noted,34,3H--40 123 were white, 49 Hispanic American, 52 African American, and 10 Asian. Although these data suggest a disproportionate representation by children of African American, Asian, or Hispanic origin, population studies necessary to support this impression have not been reported. 41 -45 Nonetheless, surveys suggest a predisposition for SLE among Puerto Rican and nonwhite residents of New York City that is higher than that for non-Hispanic whites. 17 An increased incidence of SLE
344
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
among Sioux, Crow, Arapaho,46 and southeast Alaskan Indian adults 47 has been noted, but no data relating to children in these groups are available. In the pediatric rheumatology clinic in Vancouver, British Columbia, the proportion of children with SLE whose parents are aboriginal Canadians or from India greatly exceeds their representation in the overall population. 48 Higher frequency and greater disease severity have been associated with non-white race,49 including Native Americans,50 African Americans and Afro-Caribbeans. 51 Among adults in the United Kingdom, Johnson and colleagues52 observed a prevalence of SLE in Asians that was three times that in whites, and the prevalence in Afro-Caribbeans was almost six times higher than in whites.
GENETIC BACKGROUND It is generally believed that susceptibility to SLE is multifactorial and that multiple genetic factors play an important role. A "threshold liability model" has been used to describe the occurrence of the disease. 53 ,54 This model asserts that disease will occur if the disease liability, determined by the presence of genetic susceptibility alleles interacting with environmental events or circumstances, exceeds a certain threshold, In linkage studies, a region of chromosome 1 from 1q31 to 1q42 was identified as a potential location of a disease susceptibility locus in diverse ethnic groups. 55 The identity of the gene or genes is not yet known. However, Tsa0 54 proposed that the region containing the major histocompatibility (MHC) locus on chromosome 6 is not the most prominent susceptibility locus, suggesting that there are at least six other loci on chromosomes 1, 2, 4, and 16 that contribute to the genetic predisposition to SLE and many others that may possibly contribute.
Family and Twin Studies Family studies have found a higher than expected prevalence of SLE in relatives of patients with the disease. In a case-control study, 10% of patients with SLE had a firstdegree relative with SLE, compared with I % in control families. 56 The degree of disease clustering can be expressed as the ratio of the prevalence in families in which there is an affected member to the prevalence of the disease within the population as a whole. The risk ratio of affected sibling pairs to population prevalence quantifies sibling-pair familial clustering; this ratio is estimated at 20 in SLE, similar to that in diabetes mellitus. 56,57 Twin studies have confirmed concordance for SLE in 24% of 4S monozygous twin pairs but only 2% of 62 dizygous twin pairs. 58 A recent report described identical triplet girls who developed SLE between 8 and 11 years of age. 59 A connective tissue disease other than SLE occurs in approXimately 1 of 10 families of patients with SLE. 6G-64 The influence of the sex of the affected parent on inheritance of SLE (imprinting) is a subject of some interest. The transmission of the disease from father to son may mark a specific subset of SLE. 65
Histocompatibility Antigens Using the family-based transmission disequilibrium test, the MHC contribution to susceptibility to SLE has been localized to the segment of chromosome 6pll-21 that contains the class II gene locus. 66 The susceptibility haplotypes identified were ORB1·1S01 (OR2)/OQBl* 0602; ORB1·0801 (OR8)/OQB1*0402; and ORBl*0301 (OR3)/OQB1*0201. This study confirmed the earlier report that HLA-OR2 and HLA-DR3 independently increase the relative risk for development of SLE by twofold to threefold in whites. 67 Among African Americans, in whom OR3 is infrequent, OR2 and OR7 are associated with SLE. These associations are attributable largely to linkage disequilibrium between DRB1·IS01, ORBS·0101 and DQB1·0602. 6H Class III MHC genes (tumor necrosis factor-a [TNF-al, C4) are probably more important contributors to susceptibility. Mason and Isenberg69 stated that in white subjects, the haplotype A1;B8;OR3 conferred a lO-fold increase in the risk of developing SLE; the presence of two C4A null alleles increases the risk I7-fold. Although the contribution of specific HLA haplotypes to susceptibility to SLE is modest, there are, nonetheless, strong associations between some HLA genes and specific autoantibodies. These are depicted schematically in Figure 16-1. Studies of associations with antigens of the MHC and the presence of anti-dsONA, the serologic hallmark of SLE, have been somewhat conflicting. Griffing and colleagues70 reported an association between antidsDNA and HLA-OR3. Others found associations with OR271 or OR7. 72 In one study, 96% of patients with hightiter antibody to dsDNA had OQB1·0201 Clinked to OR3 and OR7), OQB1·0602 Clinked to OR2 and DR6), or DQB1·0302 Clinked to some DR4 allotypes),73 The most important HLA and other genetic associations are summarized in Table 16-2. In a recent study of HLA class II alleles and lupus nephritis in 244 Italian patients OQA1·0101 was associated with renal disease, an effect that was greatly enhanced by the presence of ORB·1S01; 80% of ORB1*1SOl-positive patients had class IV nephritis, and the remainder had class III nephritis. 74
Complement Component and Receptor Genes Deficiencies of C1q, Clr, CIs, C4, and C2 are important but rare genetic associations with lupus-like syndromes. 67 ,75 Among patients with homozygous deficiency of C1q in one study, more than 90% (30 patients) had SLE54; 80% of these were children at disease onset. 76--7H Absence of CIq, C1r, and CIs is associated with SLE, nephritis, and antibodies to dsDNA, whereas complete absence of C2, C3, and C4 is associated with milder lupuslike syndromes. 76 ,79 C4A and possibly C4B null alleles (C4A*QO and C4B·QO respectively), resulting in partial deficiencies, have been associated with lupus in African American and Asian populations,75 Homozygous C4A deficiency is said to occur in lO% to lS% of white patients with lupus, although rarely in healthy individuals; partial C4A deficiency occurs in 50% to 80% of SLE
C HAP T E R
16
• figure 16-1 Representation of the inter-related human leukocyte antigen (HLA) assodations with autoantibody subsets in systemic lupus erythematosus. (From Tan FK, Arnett FC: The genetics of lupus. Curr Opin Rheumatol 10: 399, 1998.)
SYSTEMIC
Lupus
ERYTHEMATOSUS
345
~::::::::::::::;? DQ6(DQ81*0602)-DR2(*1501/1503)
DQ8(DQ81 *0302)-DR4 DQ7(DQA1*0501 )-DR5, DR4 DQ4(DQA1*0401 )-DR8, DR3 DQ2.1 (DQA1*0501 )-DR3(*0301)
l!!::;;:=-----=:" DQ6(DQA1*0602)-DR2, DR13 DQ8(DQ81 *0302)-DR4
DQ2.2-DR7-DR53 DQ7(DQ81 *0301 )-DR5,DR4-DR53
4~~S~~t=~DQ8(DQ81*0302)-DR4-DR53
--,,"'.
patients, compared with 10% to 20% of control subjects. 69 Mutations of the complement-regulatory mannose-binding protein were reported to be increased in patients with SLE. 80 The authors suggested that there may be an additive effect of defective mannose-binding protein and the c4 null allele. Complement deficiencies probably predispose to SLE because of a deficiency of the processing and clearance of immune complexes and apoptotic cells.
Cytoklne Polymorphlsms The frequency of the TNF-a. promoter 308 polymorphism is increased in African Americans,8! white Europeans,82-85 and in some Chinese patients with lupus. H6 Whether the TNF-308 polymorphism is responsible for the higher levels of TNF-a. production in HLADR3-positive individuals is not certain. 85 It has been suggested that individuals with SLE who have low levels
~~.
DQ4(DQ81*0402)-DR8, DR3(*0302) DQ6(DQ81 *0604, *0605)-DR6(*1302) DQ5(DQ81 *05), DR1, DR10
of TNF-a. production may be protected from the development of glomerulonephritis: TNF-a. levels are lower in DR2-positive individuals and higher in DR3- and DR4positive individuals, who are more prone to lupus nephritis. 87 Different levels of TNF-a. may also explain the discrepant incidences of SLE in West Africans and African Americans, most of whom are of West African descent. An uncommon TNF-a.2 allele is common in West Africa and directs high levels of TNF production; absence of this gene in the African American population results in lower TNF-a. production and increased susceptibility to SLE. 88 Associations with polymorphisms of IL-10 have been sought but not consistently found. In whites and African Americans, a polymorphism of the interleukin OL)-Ia. gene (-889 CIC) was associated with a high risk of developing SLE. 89
Fc Receptor and T Cell Receptor Polymorphlsms 1ABLE 16- 2 Genetic Associations with Systemic Lupus l rylhemalosus
Majar Histocompatibility Complex Class II DRBI*ISOI DRBP080I DRBI*0301 DQAI*OIOI DR2 DR7
White race. renal disease White race White race Renal disease African Americans African Americans
Majer Histocompatibility Complex Class III C4C*QO C4B*QO C1q deficiency Clr deficiency CIs detlciency C2 deficiency Mannose-binding protein mutation TNFa 308 promoter polymorphism TNFa 2 deletion ILIa (-889 CIC) FcyR IIa-RI3I PO 1 polymorphism
PAR? (poly (ADP-ribose) polymerase) (?) ACE (angiotensin-converting enzyme) (?)
The genes that encode the Fc receptor of immunoglobulin G (IgG) are located on chromosome 1q23. Polymorphisms in the receptor for IgG2 (FcyRiIa-RI3l) result in reduced binding and delayed elimination of IgG2containing immune complexes90 and might therefore be important in susceptibility to immune complex-mediated manifestations of SLE. There have been conflicting reports of associations of SLE and the isoforms of FcyRII, and FcyRIII. An increased frequency of homozygosity for the low-affinity FcyRII receptor that binds IgG2 to leukocytes and platelets was found in one white population,9! but not in another. 92 The latter study, however, found an increased frequency of the low-affinity phenotype in African Americans with SLE. The conferred risk appears to be small, however. 93 A single base pair mutation of the FcyRIII gene confers a small added risk of developing SLE. 94 .95
Other Genetic Fadors Early studies suggested that genetically determined differences in the T cell receptor were associated with SLE, although a recent study discounts this association.% Polymorphisms of the programmed cell death gene PD-1,
346
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
a member of the immunoglobulin superfamily located on chromosome 2q35-37, were associated with SLE in European and Mexican populations. 97 PD-1 is important in the regulation of autoreactive CD8+ T and B cells, and alteration of its function could contribute to T cellmediated autoimmune disease. 98 Studies of polymorphisms in PARP (poly (ADP-ribose) polymerase), encoded on chromosome lq41-42,99 and angiotensinconverting enzyme (ACE) 100 have yielded conflicting results. Other associations with unidentified genes are suggested by linkage studies in SLE.IOl
ETIOLOGY AND PATHOGENESIS Except for drug-induced lupus, the etiology of SLE is unknown. It is evident that SLE results from the interaction of many factors-some genetic, some acquired, some, possibly, environmental. The end result is grossly disordered immunity with the presence of persistent autoreactive Band T lymphocytes that create a multitude of clinical manifestations, many of which are antibody or immune complex mediated. In some aspects of the disease, immune complexes play the most important role; in others, antibodies or hormones are important. Different pathogenic mechanisms may predominate in different aspects of the disease.
Immune Dysregulatlon The immunopathogenetic factors that contribute to the manifestations of SLE are multiple, complex, and interactive. Many genes contribute to the regulation of these mechanisms. Studies in murine models of lupus have contributed substantially to the understanding of these interactions but do not completely reflect the heterogeneity of the human disease.
B Lymphocyte Function There has been a resurgence of interest in the role of B lymphocyte abnormalities in the pathogenesis of SLE. 102 Antibody-mediated mechanisms are operative in many of the clinical manifestations, such as acute hemolytic anemia, thrombocytopenia, leukopenia, and the antiphospholipid syndrome (APLS). In lupus nephritis and vasculitis, antigen-antibody complexes are directly related to the pathologic manifestations of the disease. B cell numbers are increased in patients with active lupus and produce increased levels of antibodies and hypergammaglobulinemia. 103 The frequency of IgGsecreting B cells in the peripheral blood correlates with disease activity.IM.IOS This may reflect polyclonal B cell activation by an exogenous antigen, antigen-stimulated B cell proliferation, or intrinsic abnormalities of the B cell and its control. Intrinsic B cell abnormalities are also present, including hyper-responsiveness to IL-6 106 and epitope spreading. 107 High-affinity IgG anti-dsDNA antibodies are characteristic. There is evidence that such antibodies arise by somatic hypermutation 102 during polyclonal B cell
activation induced by environmental agents such as viruses or bacteria, or they may develop by exposure of the immune system to self-DNA-protein complexes in nucleosomes or chromatin. During apoptosis, nuclear and cytoplasmic antigens appear in blebs on the cell membrane. Complexes such as the Ro (SS-A) antigen, small nuclear ribonucleic acid (snRNA), and nucleosomes are presented to the immune system by this mechanism. Not only do B cells produce autoantibodies, but they also influence antigen presentation and the differentiation of helper T cell Thl and Th2 responses. J08 Control of established autoantibody production is impaired because of defective CDW and natural killer cell function 109 and inefficiency of the idiotype-anti-idiotype network. 110 Persistence of antigen and antibody in the form of immune complexes is aided by suboptimal clearance of immune complexes by the reticuloendothelial system. This represents the effects of a number of factors, including exceeding the capacity of the reticuloendothelial system to remove the high level of complexes generated by autoantibody-antigen interactions, the low level of CRI complement receptors on cell surfaces,l11 and the presence of low-affinity receptors for IgG2 (FCyRID.91 B cells produce high levels of antibodies to autoantigens; control of the production of these antibodies is defective, and mechanisms that normally eliminate the immune complexes formed by such antibodies are also deficient. These circumstances set the stage for antibody- and immune complex-mediated tissue damage.
Immune Complex-Mediated Disease Evidence that immune complexes are important in pathogenesis is summarized in Table 16-3. Lupus nephritiS is the prototypical immune complex-mediated disease. Antibodies to dsDNA and ribonucleoprotein have been eluted from the renal glomeruli l12 and have been demonstrated by fluorescence microscopy in the kidney, skin, and central nervous system (CNS). Some immune complexes are formed in the circulation and deposited in tissues; others develop in situ-a mechanism that is favored by the high affinity of the antigen (e.g., dsDNA) for the target tissue (e.g., type IV collagen in the basement membrane).113 In situ localization of antibodies to dsDNA and the formation of immune complexes may be of central importance in membranous glomerulonephritis. C1q can bind directly to dsDNA, and therefore complement activation and its consequences might result without participation of autoantibody. Demonstration of properdin in some biopsy specimens is evidence of involvement of the alternative pathway.lJ.1 These mechanisms are further described in Chapter 5. Immune complexes induce an inflammatory lesion by activation of the complement cascade. Consequently, "'1 I! II
TABLE I b 3 [vidence of the Involvement of Immune Complexes in Pathogenesis 01 Systemic Lupus Lrythellldto~u~
Immune complexes are present in serum and affected tissues (renal glomerulus, dermal-epidermal junction, choroid plexus) Immune complexes activate complement, resulting in hypocomplementemia during active disease and the presence of complement activation products
C HAP T E R
chemotactic factors are generated (C3a, C5a), granulocytes and macrophages are attracted, and inflammation results. Low serum levels of C3 and C4, along with the detection of complement components in immune complexes in the renal glomerulus, indicate that activation of both the classic and alternative pathways is involved in the production of the renal lesions. ll5 A number of factors contribute to immune complex deposition in SLE, including the availability of large amounts of antigen (resulting, at least in part, from enhanced apoptosis), the exaggerated autoantibody response, and the diminished clearance of immune complexes by an overburdened or inefficient reticuloendothelial system. Genetic contributions to the failure to eliminate immune complexes effectively include polymorphisms of the FcyRs and the decrease in the complement receptor CR1 on erythrocytes. In experimental models of glomerulonephritis, the deposition of immune complexes is influenced by a variety of factors, including filtration pressure and release of vasoactive amines by IgEsensitized basophils, in addition to immune complex size, complement fixation, and antibody affinity.116-119 The glomerular mesangium functions normally to clear immune complexes from the vascular compartment and thereby occupies a central pathogenic role in certain types of experimental chronic nephritis.
Deposition of immune complexes in the subendothelial spaces of the choroid plexus may contribute to the clinical manifestations of lupus encephalopathy by interfering with normal cerebrospinal fluid circulation. Immune complexes were demonstrated in the choroid plexus in one lO-year-old girl who died of CNS lupus. 12o
T Lymphocyte Function Patients with active SLE have a T lymphocytopenia involving, particularly, the CD4+/CD45RA+ subset of T cells that activates CD8+ T cells to suppress hyperactive B cells. There is a shift from the ThO to the Th2 cell cytokine phenotype in SLE.98 As a result, the cytokine profile tends to support B cell activation by IL-lO, IL-4, IL-5, and IL-6. (The interactions of cytokines and cells are discussed in detail in Chapter 5.) T cell abnormalities are summarized in Table 16-4.
Dendritic Cell Function
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
347
constitute a heterogeneous population of cells with a central role in antigen presentation to na'ive T cells. In a recent review of the role of DCs in pathogenesis of SLE, Pascual and colleagues 121 ,122 postulated that interferon-a (IFN-a) is critical in the pathogenesis of SLE because of its central role in regulating the maturation of monocytes into DCs. Serum from patients with SLE induces monocytes to become DCs that can present antigens to T cells. 123 The serum factor has been identified as IFN-a produced by plasmacytoid DCS. 123
Neutrophil Abnormalities Patients with SLE may be neutropenic, but neutrophil function is usually normal. However, a lupus-like syndrome has been described, in some mothers and maternal grandmothers of boys with chronic granulomatous disease, in which bacterial killing by neutrophils is defective. 124 In addition, there are reports of children with both X-linked and autosomal recessive chronic granulomatous disease who also have a disease resembling SLE (see Chapter 33).125,126
Apoptosis Many of the characteristic autoantibodies found in SLE are directed to antigens that become concentrated in surface blebs of apoptotic cells. 127,128 Abnormalities in the control of apoptosis are therefore potentially of importance in the pathogenesis of SLE. Emlen and colleagues 129 described increased apoptosis of a mixed population of lymphocytes from patients with SLE in vitro. Not only is apoptosis increased in SLE, but apoptotic cells persist because of defective clearance. Low levels of C1q prevent efficient uptake of apoptotic cells by macrophages. 13o Increased Bcl-2 expression on T cells from patients with SLE13I correlates with disease activity132 and interferes with apoptosis. The expression of Fas protein is increased on CD8+ T cells in patients with SLE,1:\3 and this results in increased apoptosis of these cells and lymphocytopenia. 133 Patients with mutations of the Fas gene (Canale Smith syndrome, autosomal recessive lymphoproliferative syndrome)134 only occasionally develop a lupus-like syndrome. Such mutations appear to be rare in patients with SLE.135 A recent study136 demonstrated that cyclooxygenase-2 inhibition augmented Fas signaling and resulted in apoptosis of autoreactive T cells.
Dysregulation of dendritic cell (DC) interactions may play a central role in the pathogenesis of SLE. DCs
I:.
Hormones rABLE 16 4 T Lymphocyte Abnormalities in Systemic Lupus t rythematosus (SLE)
CD4+ T cells Decreased by glucocorticoids Decreased in active SLE Decreased in presence of antilymphocyte antibodies CD4+ subset decreased: CD45 RA+ CD4+ subset increased: CD45 Ro CDS+ T cells Normal or increased in active SLE CD3+ double negative (CD4-, CDS-) T cells
Steroid hormones have many effects on both the innate and adaptive immune systems and are therefore of importance in disease pathogenesis. m ,138 Although sex hormones do not cause SLE, they appear to play a role in disease predisposition and, possibly, disease severity or activity. Lupus occurs primarily in women between menarche and menopause; in childhood and after menopause, the ratio of females to males with the disease is much lower. However, a recent population-based case-control study by Cooper and associates,139 involVing 240 adult women with SLE and 321 controls, showed that
348
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
menarche occurred later and menopause occurred earlier in patients who subsequently developed SLE, implying that women with longer exposure to estrogen were not at greater risk of developing SLE. A relative deficiency of androgenic hormones and an increase in estrogenic hormones is characteristic of the patient with SLE, regardless of sex. Some studies in children 140 have reported that estrogen levels in children with SLE were normal but testosterone levels were lower in postpubertal boys and girls with SLE, compared with normal children or with children with juvenile rheumatoid arthritis (JRA). Children with SLE also had significantly elevated follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin. In women with SLE, levels of 16a-hydroxyestrone and estriol are increased. 141 Early studies noted an association between increased disease activity and the use of oral contraceptives containing high levels of estrogen, but more recent reports, in which the estrogen dose was much lower, failed to confirm such a relationship.142 Increased frequency of flares of SLE during the third trimester of pregnancy or in the postpartum period have been reported in some studies 143 but not in others. 144 In men with SLE, levels of estrogenic hormones are increased. 141,145 Relative deficiency of androgenic hormones may also play a role. Plasma androgens are decreased in women and men with SLE. 146 Males with Klinefelter's syndrome have low levels of testosterone, increased LH, and an increased frequency of SLE.147 A study148 of 35 males with SLE found no significant differences from controls with respect to mean serum testosterone or estradiol levels, although testosterone tended to be lower in those with active disease. Gonadotropins (FSH, LH) were higher in men with SLE. Elevated prolactin levels have been reported in pregnant women 149 and in men with active SLE.145,148 Bromocriptine, which inhibits the secretion of prolactin by the anterior pituitary gland, has been used to suppress activity in these patients and in a few children with SLE. 150
Environmental Fadors Ultraviolet Irradiation Sunlight is well known to exacerbate disease activity, and ultraviolet B (UVB) irradiation has multiple effects that are important in pathogenesis, including induction of apoptosis of keratinocytes. l5l In the process of apoptosis, DNA is degraded to nucleosomes, and during apoptosis, blebs containing small nuclear ribonucleoproteins (snRNPs) such as the 52-kD Ro/SS-A appear on the cell surface, rendering them available as immunogens. 127 ,128 Expression by keratinocytes of the intercellular adhesion molecule ICAM-l is enhanced by UVB152 but suppressed by ultraviolet A,153 UVB increases production of IL-8 and TNF-a in the epidermis,154 an effect that could increase leukocyte-mediated inflammation. There is some evidence that ultraviolet Al may actually be therapeutic in patients with SLE,155
Viral Infection There are no compelling data linking any virus with SLE. Elevated titers of antibodies to Epstein-Barr virus (EBV),156 cytomegalovirus, and herpes simplex virus 1 (HSVl),157 for example, probably reflect polyclonal B cell activation rather than specific viral infection. The frequency of EBV detected by polymerase chain reaction studies of peripheral blood leukocytes from 20 children with SLE was no different from that in children with .IRA or normal control subjects. 158 Parvovirus infection can mimic SLE, but whether it is pathogenically related is unclear. 159
Drugs and Chemicals Certain drugs are associated with the induction of a lupus diathesis that differs from idiopathic lupus in being dependent on the continuing presence of the drug (Table 16-5). Discontinuation of the offending agent is usually associated with disappearance of the serologic and clinical manifestations of lupus. The etiology and pathogenesis of drug-induced lupus or lupus-like disorders were recently reviewed. 160 There is no evidence, however, that pharmacologic agents are involved in the etiology or pathogenesis of idiopathic SLE, L-Canavanine, a chemical that is found in alfalfa sprouts and seeds, has been associated with the induction or exacerbation of SLE in a few patients. 161 Furthermore, a diet consisting almost exclusively of alfalfa sprouts and seeds induced reversible SLE in cynomolgus macaque monkeys with high levels of antibody to dsDNA, hypocomplementemia, and diffuse glomerulonephritis. 162 No strong relationship between use of hair dye or cigarette smoking and the incidence of lupus was found. 163 Other evidence of the role of environmental factors includes the presence of immune complexes in skin biopsy specimens ("lupus band") and lymphocytotoxic antibodies, ANAs, and positive LE-cell preparations among laboratory technicians working with SLE sera. 164
Animal Models of Systemic Lupus Erythematosus Murine models of lupus-like disease have been extensively studied and have yielded important information related to human disease. 165 There are both spontaneous and induced
E:..
TABL E 16 5
Drugs Implicated in lupus like Syndronw\
DefInitely AssocIated
Possibly AssocIated
a-Methyldopa Chlorpromazine Ethosuximide Hydralazine Isoniazid Minocycline Phenytoin Procainamide Trimethadione
Captopril Carbamazepine L-Canavanine Metoprolol Penicillamine Penicillin Propylthiouracil Quinidine Sulfonamides Zafirlukast
C HAP T E R
models of lupus in mice. In 1948, pairs of mice in a colony at the University of Otago Medical School in New Zealand were selected solely on the basis of coat color for inbreeding. From these lines, the New Zealand Black (NZB) and New Zealand White (NZW) strains were developed. NZB mice develop hemolytic anemia and thymic lesions characterized by the presence of germinal follicles; they are deficient in C5. The first-generation offspring (FI) of the cross between NZB and NZW animals, the NZBIWF1, develop a severe lupus-like disease characterized by the production of autoantibodies, glomerulonephritis, lymphoid hyperplasia, hemolytic anemia, hypocomplementemia, and other clinical and laboratory abnormalities. BXSB mice are derived from a cross between a C57BU6J female and an SB/Le male (which has a pigment abnormality and giant lysosomal granules similar to those in ChediakHigashi syndrome). The gene associated with the autoimmune features of this strain is carried on the Y chromosome of the BXSB mouse. MRL/I mice are derived from a cross between the leukemia-prone AKR/J mouse and several nonautoimmune strains. These mice develop polyarthritis, glomerulonephritis, lymphoid hyperplasia, and vasculitis. l66 All strains have hypergammaglobulinemia, ANAs (anti-dsDNA and anti-ssDNA), immune complexes, hypocomplementemia, and cryoglobulinemia. Anti-Sm antibodies, which are highly characteristic of human lupus, are also present in MRL/I mice. 167 Spontaneous and induced models are summarized in Table 16--6.
CLINICAL MANIFESTATIONS
Olnlcal Presentation SLE ranges from an insidious, chronic illness with a long history of intermittent signs and symptoms to an acute, rapidly fatal disease. Constitutional symptoms are common at onset and during exacerbations (Table 16-7).
filii
IABLE 16-6
Murine Models of Lupus
Model
Charader\sUcs
e---------'
Spontaneous
(NZB x NZW)FI, NZM2410, (SWR x NZB)Fl MRVlpr. MRVgld
BXSB/yaa
Develops ANAs and glomerulonephritis Single-gene mutation (Fas or Fas ligand) that leads to autoimmunity when expressed in MRL background Contains Y-Iinked autoimmune accelerator gene that causes more severe disease in males
GenetIcally Engineered Mutations Fas, FasL, Bel2 Sap, Clq, C4, DNAase Ctla-4. p2l, PD-I, Lyn, Fyn BLyS, PD-I, Lyn, Fyn, FcyRIIB FcyIll, ICAM-I
Dysregulation of apoptosis Dysregulation of antigen clearance Activation and dysregulation of T cells Activation and dysregulation of B cells Pro-int1ammatory mechanisms
ANAs. antinuclear antibodies. Modified from Nguyen C, Limaye N, Wakeland EK: Susceptibility genes in the pathogenesis of murine lupus. Arthritis Res 4 (Suppl 3): S255-S263, 2002. For references to each model. please refer to this publication.
16
r__. "
SYSTEMIC
Lupus
ERYTHEMATOSUS
349
TABLE 16-7 Clinical Features of Systemic Lupus Erythematosus
Constitutional Cutaneous Musculoskeletal Vascular Cardiac Pulmonary Gastrointestinal Liver, spleen, nodes Neurologic
Ocular Renal
Fever, malaise, weight loss Buttert1y rash, discoid lupus, periungual erythema, photosensitivity, alopecia, mucosal ulcerations Polyarthralgia and arthritis, tenosynovitis, myopathy, aseptic necrosis Raynaud's phenomenon, livedo reticularis, thrombosis, erythromelalgia, lupus profundus Pericarditis and effusion, myocarditis, Libman-Sacks endocarditis Pleuritis, basilar pneumonitis, atelectasis, hemorrhage Peritonitis, esophageal dysfunction, colitis Hepatomegaly, splenomegaly, lymphadenopathy Organic brain syndrome, seizures, psychosis, chorea, cerebrovascular accident, polyneuritis and peripheral neuropathy, cranial nerve palsies, pseudotumor cerebri Exudates, papilledema, retinopathy Glomerulonephritis, nephrotic syndrome, uremia, hypertension
Intermittent or sustained fever, fatigue, weight loss, and anorexia are manifestations of active disease in most children and may occur as the presenting symptoms. A single system may be affected at onset, although multisystem disease is more characteristic (Table 16-8). As awareness of the occurrence of SLE in children has increased, early diagnosis has become the rule rather than the exception.
Cutaneous Involvement Rashes are common at onset and during active disease and are varied in character and distribution. t68 The classic "butterfly" rash 169 occurs in one third to one half of children at onset of disease170; however, although it is highly suggestive of SLE, it is not pathognomonic (Fig. 16-2). It is characteristically symmetrical, occurring on both malar eminences, over the bridge of the nose, and sometimes on the forehead, but sparing the nasolabial folds. It is usually well demarcated and may be slightly raised. Follicular plugging is present, but the lesions do not usually result in scarring. This photosensitive rash may be precipitated by exposure to sunlight. A similar distribution of erythema occurs in juvenile dermatomyositis, but the lesion is usually less well demarcated. A rare syndrome of telangiectasia in a butterfly-like distribution and membranoproliferative glomerulonephritis may be confused with SLE.170 As pointed out by Sherwood and colleagues,171 Bloom's syndrome (OMIM 210900),172 Cockayne's syndrome (OMIM 216400),173 and Rothmund-Thomson syndrome (OMIM 268400)174 all have a dermatologic lesion in the butterfly distribution, thereby raising the question of a possible diagnosis of SLE. A number of other cutaneous manifestations of SLE may occur at onset or during the course of disease. The skin changes in 57 Thai children with SLE are summarized in Table 16-9. 170 Periungual erythema and livedo reticularis may have been
350
1:..
C HAP T E R
TABLE 16-M
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
Frequency of C1ini
Manife~tdtion~ of Sy~temi< lupu~ [rylhemato~u~
Manifestation
Present at Presentation (0/0)
Present at Any nme (%)
Olnlcal Malar rash Althritis Fatigue Renal disease Other rashes Fever Weight loss Nasal or oral ulcers Alopecia Anorexia Pleuritis/pericarditis CNS disease Photosensitivity Raynaud's' phenomenon Lymphadenopathy Digital ulceration Myocarditis Myositis Thrombocytopenic purpura
68 61 59 51 45 46 34 29 27 21 15 15 15 15 14 5 3 3 1
72 66 60 59 48 46 36 36 31 22 24 21 19 18 15 6 3 3 2
Laboratory Low C3 LowC4 Low C3 and low C4 Elevated creatinine Lymphopenia Hemolytic anemia Anti-dsDNA (Crithidia) Anti-Sm Anti-RNP Anticardiolipin Anti-Ro Rheumatoid factor Lupus anticoagulant Anti-La
70 52 48 9 33 20 80 43 34 26 19 12 8 7
-
-
• Figure 16-Z Malar erythema of acute systemic lupus erythematosus.
35 21 88 54 40 39 26 15
11 9
From Hiraki L, Schneider R, Hebert D et al. Clinical and laboratory manifestations of systemic lupus erythematosus CSLE) in pediatric patients. Anhritis Rheum 46: S315. 2002.
The classic butterfly rash has erupted over both cheeks and spread over the bridge of the nose. It may be punctate and follicular or an erythematous blush. The rash does not leave a scar. (See color insert.)
Periungual erythema reflects dilatation and tortuosity of the nail fold capillaries, similar to that observed in dermatomyositis and scleroderma. Nail fold infarcts may also occur (Fig. 16-6). Livedo reticularis occurs, particularly in the lower extremities; it reflects active disease and is
I! ..
TABLl 16-9 Cutaneous Mdnife~t,llions of Systemi< lupus Erythematosus ill Children
Manifestation
under-reported in this study, because these abnormalities are much more readily observed in fair-skinned individuals of European ancestry. Maculopapular rashes as manifestations of vasculitis or perivasculitis may occur anywhere on the body, but particularly on sun-exposed areas such as the face and upper anterior chest. These lesions are occasionally tender, and angiitic papules on the soles and palms may resemble Osler nodes and Janeway spots of bacterial endocarditis. Most heal without scarring or pigmentation (Fig. 16-3). Petechiae and purpura may represent perivasculitis or result from thrombocytopenia (Fig. 16-4). Sharply demarcated chronic leg ulcerations may develop around the malleoli (Fig. 16-5), and gangrene may result from severe cutaneous vasculitis or arterial thrombosis associated with the APLS. The lesions of subacute cutaneous lupus begin as papules that evolve into annular lesions with raised edges. They are often widespread on trunk, limbs, and face. These lesions may become crusted, hyperpigmented, and atrophic. 175
Skin lesions (all types) Malar "butterfly" rash Vasculitis (petechiae, palpable purpura) Raynaud's phenomenon Periungual erythema Periungual gangrene Nail involvement Alopecia Subacute lupus erythematosus Bullous lupus erythematosus Discoid lupus erythematosus Photosensitivity Urticarial lellkocytoclastic vasculitis Livedo reticlliaris
Number (N = 57)
Percentage
44 42 22
77 74 38
4 5 2
7 9 4
2 18 2
4 32 4
2 11
19
23
40 2
1
2
Data modified from Wananukul S, Watana D. Pongprasit P: Cutaneous manifestations of childhood systemic lupus erythematosus. Pediatr Dermatol 15: 342-346. 1998.
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
351
• Figure 16-6 Nail fold infarct in a 22-year-old man with systemic lupus erythematosus for 14 years. Ischemic necrosis of the fingers developed after 2 years of remission from a clinical course punctuated with cardiopulmonary onset, polyarthritis, diffuse proliferative nephritis, and, more recently, central nervous system lupus.The digital lesions healed completely in 5 months with prednisone and prazosin therapy.
• figure 1&-3 Vasculitis seen as punctate erythema of the fingertips of a child with systemic lupus erythematosus.
associated with the presence of antiphospholipid antibodies (aPL).176 Discoid lesions were observed in 19% of the patients with SLE reported from Thailand l70 but may be less common in children in North America and Europe. Isolated discoid lesions are even less frequent in children.177,178 They most commonly occur on the scalp or limbs in an asymmetrical distribution, are sharply demarcated papulosquamous lesions that are often photosensitive, and heal with atrophy, scarring, and alteration of pigmentation (Fig. 16-7). When they occur on the scalp, they may be associated with localized alopecia. Discoid lupus
• figure 16-4 Vasculitic purpura in ateenage girt with an acute exacerbation of systemic lupus erythematosus. (See color insert.)
• figure Ui-5 Chronic and well-demarcated ulcers of the skin in a child with systemic lupus erythematosus.
• Figure 16-7 Discoid lesions and mucocutaneous disease are present in this teenage boy who had anti-Ro antibody. (See color insert)
352
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
appears to be much more common in black children than in other racial groups. Alopecia, usually characterized by diffuse thinning of the hair, is associated with active disease. Frontal hair is usually affected initially and becomes brittle and kinky. The initial complaint of the child or parent is that there is excessive hairfall on the pillow, in the hair brush, or after shampooing. Patchy alopecia is less common; total alopecia is rare. Nail changes, including loss of the nails, occur in up to 15% of adults with long-standing disease 179 but are rare in children. Other infrequent cutaneous complications of SLE include bullae180-182 and papular mucinosis. 183 Telangiectases and urticarial lesions occur occasionally. Nodules, which resemble either erythema nodosum or rheumatoid nodules, occur in approximately 5% of patients and may suggest the possible presence of an overlap syndrome. Lupus profundus l84 has rarely been described in childhood. 184 Chilblain lupus is a chronic, unremitting form of SLE that occurs predominantly in women, although the authors have observed it in several children. It results from cold exposure and possibly hyperviscosity.l86 Persistent warts are common in children with SLE, and their appearance appears to be related both to the disease l87 and to the effects of immunosuppressive treatment.
Mucosal Involvement The oral mucosa is the site of ulceration in some children with SLE. The classic lesion is a painless, shallow, ragged ulcer on the hard palate. Although this involvement is uncommon, erythema of the hard palate is quite common and suggestive of the diagnosis (Fig. 16-8). Occasionally. ulceration or perforation of the nasal septum is observed. Aphthous stomatitis is perhaps a more frequent but less specific finding, because such changes occur in the otherwise healthy population.
Musculoskeletal Disease Arthralgia and arthritis affect most children with SLE. Arthritis commonly involves the small joints of the hands, wrists, elbows, shoulders, knees, and ankles. The arthritis is characteristically short in duration, lasting several days, although it is sometimes persistent and can be migratory. Joint pain is often severe even though objective findings are minimal. In some children, the arthritis is persistent and is characterized by swelling, tenderness, and loss of range of motion. Although the synovitis of SLE may be minimally proliferative, it is only occasionally erosive 11lH and usually does not result in permanent deformity. A Jaccoud type of arthritis (reversible subluxation related to tenosynovitis) may be associated with the presence of antibodies to Ul ribonucleoprotein (Ul RNP).189 Tenosynovitis on the dorsum of the hand and wrist commonly accompanies arthritis of the small joints. Myalgia or muscle weakness is characteristic of the acutely ill patient 190 and is most prominent proximally. The occurrence of myositis is often related to the pres-
• Rill" 16-8 Mucocutaneous ulc:erations of acute systemic lupus erythematosus. Ashallow, painless. erythematous ulc:eration with an irregular margin is seen on the hard palate.
ence of systemic vasculitis and involvement of viscera. Steroid myopathy, in which serum levels of muscle enzymes are normal, must be distinguished later in the disease from a recurrence of myositis, in which enzyme levels are usually elevated. Ischemic necrosis of bone is a significant risk in young patients,191 especially in those treated for long periods with glucocorticoid drugs. '92 It can occur anywhere but is most common in weightbearing bones, such as the femoral heads and tibial plateaus (Fig. 16-9).
Lupus Nephritis Lupus nephritis is probably present to some degree in all children with SLE and is a major determinant of longterm outcome. Clinically evident nephritis occurs in at least 75% of children 193 and may be more frequent and of greater severity in children than in adults. The frequency of its occurrence at disease onset is uncertain, because lupus nephritis is usually initially asymptomatic, although a few children develop gross hematuria or edema associated with the nephrotic syndrome. Using data from his own and other reported series, Cameron 193 estimated that the most common initial manifestation of nephritis is microscopic hematuria (79%), followed by proteinuria, including nephrotic syndrome (55%), Decreased glomerular filtration (50%) and hypertension (40%) were also common, but acute renal failure as a presenting manifestation of renal lupus was rare 0.4%). Hypertension may be initial sign of glomerulonephritis. Correlations between clinical disease and renal biopsy abnormalities are summarized in Table 16-10.
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
353
Central Nervous System Disease
• FIgue 1~' Aseptic necrosis of the femoral head in ayoung woman with systemic lupus erythematosus who had been treated with prednisone for 11 years. She developed anterior groin pain on weight-bearing. The joint space is preserved, but there is mottled increased density and deformity of the femoral head. Asemilunar area of separation of the bone is present, indicating osteonecrosis.The patient's symptoms and disability resolved only partially with reduction of prednisone and atrial of no weight-bearing.
Significant renal disease usually develops within 2 years after onset but occasionally appears more than a decade later. Histologic evidence of renal disease may precede the development of changes in the urinary sediment by months, and routine monitoring for proteinuria and hematuria, as well as assessment of renal function by measurement of creatinine clearance or radionuclide glomerular filtration rate, are important components of management in all children from the onset of disease, The expertise of a nephrologist as a member of the team that is providing ongoing care is essential. Interstitial cystitis occurred early in the course of disease in an 8-year-old girl with SLE and glomerulonephritis. She had not received cyclophosphamide, and the disease responded to cyclosporin A. 194 This rare complication was the subject of a recent review. 195
,1: • r -
ABLE 16-1 () Clinicopathologic Correlation in Lupus Nephritis
1;pe of Renal Disease
Glomerular Mesangial Focal proliferative Diffuse proliferative Membranous Extraglomerular Interstitial Necrotizing arteritis End-stage
Nephrotic Renal Syndrome Failure Remission
±
±
++ +++
++ ++
+
+ + +++
±
++
+ ++ ±
+ ±
+
-. absent; ±. minimal; +. moderate; ++. marked; +++. very marked.
Uremic Death
±
+++ + ± ±
+++
CNS disease is a major cause of morbidity and mortality, occurring in 20% to 95% of affected children. I96-199 In approximately one half of those who develop CNS disease it is present at onset; it begins during the first year of disease in 40%, or may not appear until many years later. The ACR has delineated a number of different neurologic syndromes associated with SLE in adults. 200 ,201 Using these definitions, Sibbit and colleagues l99 studied the occurrence in 55 patients with pediatric-onset SLE. Overall, some neuropsychiatric abnormality occurred in 95%. The frequencies of individual manifestations are shown in Table 16-11. Diagnostic tests used in the evaluation of CNS lupus are listed in Table 16-12. The choice of tests is guided by the clinical manifestations of CNS disease. Collectively, neuropsychiatric manifestations are most common and include depression, difficulty concentrating and remembering, and psychosis (including hallucinations and paranoia).201-203 Depression is particularly common and may be difficult to differentiate from concern about the effects of the disease or side effects of therapy. The change in physical appearance caused by glucocorticoids is especially difficult for adolescent patients to accept and alone may lead to major depression and noncompliance with treatment. Emotional lability is common in young patients with SLE, often in the absence of other evidence of CNS disease.
II • -
TABLE 16-11 Prevalence of Neuropsychiatric Manifestations in Childhood-Onset Systemic Lupus Erythematosus
Symptom Headache Recurrent headache Migraine Mood disorder Depressive features Mixed features Major depressive episode Manic features Cognitive disorder Seizure disorder Isolated seizures Epilepsy Acute confusional state Anxiety disorder Peripheral nervous system disorder Dysesthesia/paraesthesia Cranial nerve disorder Cerebrovascular disease Cerebral infarction Transient ischemic attack Chronic multifocal disease Hemorrhage Sinus thrombosis Psychosis Chorea Demyelinating syndrome Myelopathy Aseptic meningitis Autonomic disorder
Ufetlme Prevalence (0/0) 71 36
23 2 32 2 55 47 15 35 21 14 1 8 12 1 1 0 12 7 4 1 1 ()
Modified from Sibbitt WL Jr, BrandlJR, Johnson CR. et al: The incidence and prevalence of neuropsychiatric syndromes in pediatric onset systemic lupus erythemalosus. J Rheumatol 29: 1538, 2002.
354
II.
C HAP T E R
16 S Y S T E M I C L u PUS E R Y THE MAT 0
TABLE 16 12 £VahMlioll of (1.'1111,,1 Nervoll' Sy'lem LlIpu, Erylhl'mdlo""
Examination
Purpose
Cerebrospinal fluid examination Neurocognitive testing
Exclude infection, subarachnoid hemorrhage Detection of subtle changes in cognition Provide evidence of diffuse or local abnormality Document focal lesion Characterize focal lesion
Electroencephalography Computed tomography Magnetic resonance imaging Magnetic resonance angiography Positron emission tomography
Characterize focal lesion
SUS
girl with a retinal vein thrombosis and SLE; it may have been related to an aPL syndrome. Other, less common neurological manifestations have been noted in large series of children and adolescents,197,198 including visual loss, vertigo, aseptic meningitis, papilledema, and pseudotumor cerebri; cranial nerve palsy including facial palsy is rare. 2lB Pseudotumor cerebri can develop as a direct complication of active SLE, or it may be related to inappropriately rapid tapering of the glucocorticoid dose. 219 Narcolepsy, a disorder that is characterized by extreme sleepiness and cataplexy and is strongly associated with HLA-DR2, DQw1, was reported in an 18-year-old girl. 220 Myasthenia gravis has occurred in a small number of patients, although the significance of the association is uncertain. 221
Document abnormal function
Cardiac Involvement Cognitive impairment has been noted in approximately one half of the patients, and it is not uncommon for a child with acute SLE to have increasing difficulty with school and falling academic grades. 203 Silber and coileagues 202 reported significant psychometric abnormalities in 21 of 32 children. Cognitive impairment was present in 52%, and an additional 29% were considered borderline abnormal. A study that compared children with SLE and those with JRA indicated that the former had greater neuropsychologic impairment. Furthermore, a specific deficit in complex problem-solving ability was observed. Long disease duration was found to correlate with lower intelligence quotient score. 204 Headache, a frequent and annoying complaint, has many causes, and determining the relationship of headache to CNS lupus is often difficult. In most reports, vascular migraine-like headaches are more frequent in patients with SLE than in control subjects,202 although whether the frequency of headache in patients with SLE is higher than that in the general population is in doubt. 20S In three girls with cerebral vein thrombosis, unremitting headaches were a prominent complaint. 206 Seizures are occasionally the initial manifestation of lupus. They are usually generalized tonic-clonic in type, although focal seizures sometimes occur. Cerebrovascular accidents may occur independently or with venous sinus or cerebral vein thrombosis,198.206 and they may be associated with hypertension, aPL-related thrombosis, or intracranial hemorrhage secondary to thrombocytopenia. Chorea is an uncommon complication, occurring in 4% to 10% of children with SLE. 1%.I99.207-213 It may precede other manifestations of disease, leading to an incorrect diagnosis of acute rheumatic fever, or it may occur months or years after onset. It is usually unilateral. A rare manifestation of CNS lupus that resembles Parkinson's disease may be difficult to differentiate from psychosis. 216 One child in a report by Sharar and colleagues 216 presented with rigidity, irritability, and mutism leading to an initial diagnosis of psychosis. A second child had progressive bradykinesia and was thought to be depressed. Technetium 99m hexamethyl propylenamine oxime (99mTc-HMPAO) singlephoton emission computed tomography (SPECT) scanning documented impaired blood flow to the basal ganglia. Both children responded to dopamine-agonist drugs. Myelopathy has been reported in only 12 children with SLE.217 The authors observed transient myelopathy after lumbar puncture in one
Pericarditis Pericarditis is the most common cardiac manifestation, occurring in up to 30% of children with acute SLE. It may be clinically silent, or it may be represented by precordial pain that is exacerbated by lying down or deep breathing and relieved by sitting up and leaning forward. It is only rarely accompanied by a friction rub or obvious cardiomegaly. Occasionally, lower anterior midline chest pain that is thought to be pleuropericardial in origin persists for many months or years. Pericardial inflammation leading to constriction222 or tamponade 22}-227 is rare but can be a presenting feature of SLE.224 22S 0
Myocarditis Myocarditis occurs in approximately 15% of children. 22H It is characterized clinically by congestive heart failure, cardiomegaly, arrhythmias, and a narrow pulse pressure. The presence of tachycardia in the absence of fever suggests the presence of myocarditis and should prompt electrocardiographic investigation.
Coronary Artery Disease It is well established that the adult patient with SLE has a high risk of atherosclerotic heart disease229 ; approximately 5% of these patients have nonfatal myocardial infarctions, and an equal number have fatal outcomes. There have been a few instances of myocardial infarction in childhood SLE, usually in older patients with longstanding disease.230-242 However, the extent of coronary artery disease is probably considerably underestimated and multifactorial in pathogenesis. Gazarian and colleagues243 studied children and adolescents with SLE by thallium myocardial perfusion scans, radionuclide angiography with multiple gated acquisition, and resting M-mode and two-dimensional echocardiography. None of the patients had a history of ischemic heart disease or electrocardiographic abnormalities. Four children had reversible abnormalities of myocardial perfusion scans, and one child had a fixed myocardial perfusion defect. Anticardiolipin antibodies and elevated plasma lipids were present in many of the children with and without perfuSion defects. In this study, duration of
C HAP T E R
glucocOrticoid therapy was shorter in the group with perfusion: defects. In contrast, in studies with adults, those with most severe coronary artery disease had highest cumuilitive dose and longest duration of therapy with prednisone. 244 Arteries other than the coronary vessels are also affected. One study245 found that carotid media-intima thic~ss as determined by ultrasonography was greater in 26 ipatients with juvenile-onset SLE than in a control population and correlated with the presence of nephrotic-range proteinuria. A number of factors contribute to the risk of atheromata in lupus,246 including long-term administration of glucocorticoids,247 disease-related elevation of atherogenic plasma lipids,248,249 oxidative stress secondary to the effect of autoantibodies on high-density lipoproteins,2'o apolipoprotein A-I,m and elevated homocysteine Ilevels. 2S2 In a study of 19 children with SLE, Ilowite and colleagues 248 found dyslipoproteinemia in the majority. The dyslipoproteinemia of active SLE was characterized by elevations of the very-low-density lipoprotein (VLDL) cholesterol and triglyceride levels, and decreases of highdenSity lipoprotein (HDL) cholesterol and apoprotein A-I levels. Glucocorticoid-induced changes included increased total cholesterol, VLDL cholesterol, and triglyceride levels. Elevated levels of homocysteine may also contribute to the atherogenic process. In a large study of adults with SLE,Petri and coworkers 252 reported that 15% had elevated homocysteine levels that were associated with incre:lsed risks for stroke and arterial thrombotic events (odd:" ratios of 2.24 and 3.74, respectively). Folic acid supplementation reversed the elevations of homocysteineconcentration.
ti)e
Valwllitls The classic cardiac lesion of SLE, Libman-Sacks endocarditis, is probably less common in children than in adults and is often subclinical. It is characterized by the presence of 1- to 4-mm nodules of fibrinoid necrosis of the supporting collagenous tissues of the valve. 4 The mitral valve is most commonly affected; the aortic, pulmonic, and tricuspid valves may be involved, in descending order of frequency, A clinically significant or changing murmur mayor may not be present. Superimposed infectious endocarditis can also occur. TraQsesophageal echocardiography in 69 adults with SLE disclosed valvular abnormalities in 61%. 253 The most common findings were valvular thickening (51%), vegetations (43%), regurgitation (250/0), and stenosis (4%). In a study of children and adolescents with SLE who had no clinically significant cardiac disease, echocardiographic evidence of mild mitral valve prolapse was found in 3 of 40 patients, Doppler evaluation confirmed regurgitation of the mitral valve in 4, tricuspid valve in 3, aortic valve in 2, and pulmonary valve in 1 of the 40. 254 There are reports of valve disease in children with SLE that required surgical replacement. 255 ,256 The importance of aPL to the development of cardiac valve disease is controversial, although a strong association has been noted in some
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
355
studies. 257 ,258 Similarly, antibodies to Ro (SS-A) and La (SS-B) have been reported to be associated with a higher frequency of valvular disease in children with SLE. 228
Pleuropulmonary Disease Clinical or subclinical pleuropulmonary disease is a frequent manifestation of childhood SLE (Table 16-13). In a study of 22 children,48 17 (770/0) had respiratory symptoms (cough, chest pain, dyspnea, orthopnea), and radiographic abnormalities were demonstrated in one half. Pleuropulmonary disease may be particularly common in North American Indian children. 48 The most frequently described pleuropulmonary manifestations are pleural effusions and pleuritis,259,260 acute and chronic pneumonitis,261,262 and pulmonary hemorrhage. 26 3--26' Pleural effusions are common; they may result from inflammatory pleuritis or may occasionally be secondary to the nephrotic syndrome. Noninflammatory pleural effusions are usually asymptomatic, Inflammatory pleuritis usually causes unilateral or bilateral pleuritic chest pain that is exacerbated by deep inspiration. At times pleuritic pain is the initial manifestation of the disease, The effusions are usually of small volume and seldom cause respiratory embarrassment, but occasionally very large bilateral pleural effusions occur and may require urgent paracentesis. Acute lupus pneumonitis, consisting of pulmonary infiltrates and atelectasis, occurs in 10% to 15% of children. 261 ,266 Patients usually have symptoms similar to pulmonary hemorrhage, except that anemia is not a feature and hemoptysis is absent or minimal. Bibasilar rales may be present, and radiographs reveal basilar infiltrates. Chronic interstitial lung disease characterized by chronic nonproductive cough, recurrent pleuritic chest pain, and dyspnea on exertion has been reported in adults with SLE,267 but not in children. Pulmonary hemorrhage occurs in 5% to 6% of children with SLE48 and is rarely the presenting manifestation. 2GB It is associated with a high mortality. Intra-alveolar hemorrhage can mimic
Li
TABLE 16-13 Comparison of Pulmonary Manilestdtions in Childhood-Onset and Adult-Onset Systemit lupus EI ythematosus
Abnonnallty
Childhood Onset (0/0)
AduR Onset (0/0)
Subclinical (abnormal pulmonary function tests only) Pleural effusions Shrinking lungs (diaphragmatic dysfunction) Pulmonary infiltrates/ atelectasis Acute lupus pneumonitis Pneumothorax Pleuropulmonary infections Pulmonary hemorrhage Diffuse interstitial disease Pulmonary hypertension
60
>50
27 13
>50 >25
13
>50
9
>10
9 Common
>50
6
Occasional
Occasional
<2 <5
Rare
Occasional
Modified from Delgado EA, Malleson PN, Pirie GE, Petty RE: The pulmonary manife,tations of childhood onset 'ystemic lupus erythematosus. Semin Arthritis Rheum 19: 285, 1990.
356
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
• Figure 16-10 Radiograph (A) and thin-section computed tomogram (8) of the chest of a 14-year-old girl with acute onset of pulmonary hemorrhage and thrombocytopenia as the initial manifestation of systemic lupus erythematosus, showing widespread airspace and interstitial disease.The abnormalities completely resolved after therapy with high-dose prednisone.
responded to cotrimoxazole; one died, presumably from the effects of Pneumocystis and other concomitant infections. Infection with other opportunistic organisms, such as Nocardia, has also been reported. 275.276 The relationship between aPL and pulmonary disease has recently been reviewed. 277 The pulmonary disorders that may be associated with aPL include pulmonary embolism, pulmonary hypertension, and pulmonary artery thrombosis.
congestive heart failure and pneumonitis, or it can present with sudden pallor and tachycardia with hemoptysis, although hemoptysis is not always present. A rapidly falling hematocrit with shifting pulmonary infiltrates in a patient with lupus should suggest the diagnosis of pulmonary hemorrhage even in the absence of hemoptysis. Chest radiography, including computed tomography, demonstrates characteristic changes (Fig. 16-10). Pulmonary hypertension may be present in such patients. Shrinking lung, the loss of lung volume resulting from diaphragmatic dysfunction,269.27o is less commonly identified, although it may be observed after long-term follow-up as progressive elevation of the level of the diaphragm. Pneumothorax and cavitary nodules have been reported occasionally.48 A syndrome of reversible hypoxemia has been observed in severely ill adults with SLE without evidence of parenchymal lung involvement. 271 Changes in the quality of the voice (dysphonia or hoarseness) have been reported occasionally in adult patients,272.273 and the authors have observed two adolescents with persistent hoarseness, which was one of the presenting complaints in one patient. Pneumocystis carinii pneumonia has been documented in three children, all of whom were receiving glucocorticoids and azathioprine therapy and were mildly lymphopenic. 274 The clinical presentations included cough and dyspnea. Two
I!;. TABLE 16-14
Tests of pulmonary function often document a moderate to marked functional impairment despite a normal radiologic appearance of the lungs (Table 16-14).278.279 Restrictive lung disease and diffusion defects are the most commonly observed abnormalities. Pulmonary function test abnormalities may be present in children with no clinical or radiographic evidence of lung disease. In a study of 15 such children, 40% had reduced forced vital capacity and 26% had reduced carbon dioxide diffusing capacity in the lung (DLCO). These abnormalities increased over time. 280 Unexplained dyspnea and cyanosis are present in some patients. If these signs are transient or episodic, subclinical thromboembolic phenomena should be considered.
Pulmolldly flllldioll ill Childhood Ollsd Systemic Lupus ErytlH'mdtosus
0/0 With Pulmonary Function Test Abnormality
Study (Ref. no.) De ]onste et al. (259) Singsen & Platzker (260) Weiss et al. (276) Delgado et al. (45)
Subject (n) 3 20 28 13
Olnlcal Dlsun (%) 87
Radiographic Changes (%) 87
-,
_I
? 85
? 54
Any 87 35 64 62
Restrictive 50 35 64 62
'Diffusion measured in 6 children was diminished for surface area but normal or increased for remaining lung volume. 'Patients wilh clinical or radiographic evidence of lung disease were excluded from this study. '20 patients studied. 'Diffusion was abnormal at one time or another in 6 of 9 patient'. (Diffusion was appropriate for lung size in 4 of the 6.>
Obstructive
Diffusion
a
lOa'
55
25
3
35*
8
671
C HAP T E R
Antlphosphollpld Syndrome The APLS is characterized by the presence of antibodies to phospholipids such as cardiolipin. It is called "primary" if there is no associated disease and "secondary" if it is associated with SLE or other autoimmune diseases. An international consensus statement proposed classification criteria for APLS 281 that have been validated in adults. l82 (Table 16-15). Definite APLS requires the presence of one clinical criterion (vascular thrombosis or pregnancy loss) plus one laboratory criterion (presence of ~2-g1ycoprotein I-dependent anticardiolipin antibodies of medium or high titer or lupus anticoagulant). Lupus anticoagulant is detected by a functional assay of clotting, usually the dilute Russell viper venom time (DRVVT) or a prolonged activated partial thromboplastin time (aPTT) that is not correctable by the addition of phospholipid. In addition to these classification criteria, there are other characteristic clinical and laboratory features of the APLS (Table 16-16). Primary APLS has been described in more than 50 children, most frequently as deep venous thrombosis of the lower extremities, pulmonary embolism, and thrombotic events in the brain. 2~3-28S Primary APLS may be a precursor to secondary APLS in some children. 286 The frequency of thromboembolic events was studied by Levy and associates 287 in 149 patients with pediatric lupus. Thirteen patients (8%) had one or more events; in 3 patients they occurred at disease onset. Venous thrombosis occurred in 11 episodes (cerebral venous thrombosis in 9, deep vein thrombosis in 4), pulmonary embolism in 2 episodes, and arterial occlusion in 3 episodes. All patients had lupus anticoagulants. The overall risk of a thromboembolic event in a patient with a lupus anticoagulant was 54%.
': II
1I\B1 E 16-15 Sllllltrome
Criteria for Classification of Antiphospholipid
Olnlca!' CrIterIa Vascular thrombosis Pregnancy morbidity One or more unexplained deaths of a morphologically normal fetus at or beyond the 10th week of gestation, or One or more premature births (before 34 wk) of a mOrphologically normal neonate, because of severe preeclampsia or eclampsia or placental insufficiency, or Three or more consecutive unexplained spontaneous abortions before the 10th week of gestation; maternal anatomic or horlnonal abnormalities and paternal and maternal chromosomal abnormalities excluded
Laboratory CrIteria Medium to high titer of IgM or IgG anticardiolipin antibody on two or more occasions at least 6 wk apart measured by a standard ~z-glycoprotein I-dependent anticardiolipin ELISA, or Lupus anticoagulant on two or more occasions at least 6 wk apart ELISA, enzyme-linked immunosorbent assay; IgG, immunoglobulin G; IgM. immunogl<1bulin M. Modified ft
16
I!. III
SYSTEMIC
Lupus
ERYTHEMATOSUS
357
TABLE 16 -16 Features of Antiphospholipid Syndrome Not Included in the Classification Criteria
Qlnlcal
Laboratory
Livedo reticularis Cardiac valvulopathy Multiple-sclerosis-like syndrome Chorea Seizures Transient cerebral ischemia
Thrombocytopenia Hemolytic anemia Low positive anticardiolipin antibody IgA anticardiolipin Anti-~z-glycoprotein
I Antibodies to prothrombin, annexin, or neutral phospholipids Biologic false-positive test for syphilis
IgA, immunoglobulin A. Adapted from Wilson WA. Gharavi AE, Koike T, et al: International consensus statement on preliminary classification criteria for definite antiphospholipid syndrome. Arthritis Rheum 42: 1309-1311, 1999.
Other manifestations of the APLS include chorea, 199 spontaneous recurrent abortions, hemolytic anemia, thrombocytopenia, livedo reticularis, and cardiovascular crises such as myocardial infarction and valvular lesions,288,289 Menorrhagia in adolescent girls may result from this circulating anticoagulant. Spontaneous bleeding is uncommon, however, and thrombosis is more characteristic. APLS has occasionally been observed in neonates of mothers with aPL, although, for the most part, liveborn infants of aPL-positive mothers are healthy.290 APL are not restricted to patients with SLE; they can occur in a wide variety of neoplastic, infectious, inflammatory, and autoimmune diseases. The Euro-Phospholipid Project Group has evaluated 1000 patients with primary or secondary APLS.291 Fifteen percent of these patients were younger than 15 years of age at the time of diagnosis. Primary APLS was present in 53.1%; secondary APL" was seen in patients with SLE (36.2%), a lupus-like syndrome (5%), primary Sjogren's syndrome (2,2%), rheumatoid arthritis 0.8%), systemic sclerosis (0.7%), systemic vasculitis (0.7%), or dennatomyositis (0.5%), Deep vein thrombosis, thrombocytopenia, and livedo reticularis were the most common initial clinical features, but during the disease course neurologic manifestations, especially migraine and stroke, occurred in approximately 20%.
CatastrophiC antiphospholipid syndrome (CAPLS) is a term applied to an aPL-mediated disorder in which there are multiple thrombi of small vessels affecting viscera over a relatively short period. As the name implies, the outcome can be disastrous. The clinical and laboratory features in 80 patients (including 10 younger than 20 years of age) have been reviewed in detail. 292 Almost half of the patients had previous evidence of APLS. CAPLS was precipitated by infection in 35%, traumatic and invasive procedures in 13%, withdrawal of anticoagulants or low international normalized ratio (INR) in 8%, and malignancy in 8%. The clinical presentation was characterized by multisystem disease involving the cardiopulmonary systems (adult respiratory distress syndrome) in 25%, the CNS (arterial thrombosis, seizures) in 22%, abdominal pain in 22%, and renal failure in 13%. Other organs were affected less commonly. Thrombocytopenia and hemolytic anemia were frequent, and there was laboratory evidence of disseminated intravascular coagulation in 19%. Anticardiolipin antibodies
358
C HAP T E R
16 S Y S T E M I C L u PUS E RY THE MAT 0
SUS
were present in almost all patients, and a lupus anticoagulant was detected in two thirds. Mortality in reported patients was 48%. A number of abnormalities in addition to aPL can contribute to intravascular thrombosis in patients with SLE. Elevated homocysteine concentrations are associated with arterial thrombotic events in lupus. 252 In adults, acquired protein S deficiency has been identified as an additional risk factor for thrombosis, often in association with aPL. 293.294 Much less commonly, epistaxis, easy bruising, gingival bleeding, or menorrhagia may occur as a result of acquired hypoprothrombinemia (factor II deficiency).295 Such patients also have a lupus anticoagulant, and antibodies to factor II have been demonstrated. 296 The disorder usually responds to glucocorticoids with or without vitamin K and freshfrozen plasma. Antibodies to lamin Bl, a nuclear polypeptide located between chromatin and the nuclear membrane, have been associated in adults with the presence of a lupus anticoagulant (uncorrectable significant prolongation of the apm, but not with antibodies to cardiolipin or ~2-glycoprotein,297 although they may actually protect against thrombosis. 298
Other Hematologic Abnormalities Coombs' test-positive hemolytic anemia only occasionally causes clinically significant anemia. The combination of idiopathic thrombocytopenic purpura and acute hemolytic anemia (Evans' syndrome) occurs in isolation and in patients SLE. 299.3oo Thrombotic thrombocytopenic purpura (TTP) has an acute onset and is often fatal. Its presence should always suggest the possibility of SLE. In addition to anemia and purpura, these patients have fever, changing CNS signs, and nephritis. TTP may occur as a hypersensitivity reaction to drugs or recent immunization, or it may complicate malignancy, inflammatory arthritis, or pregnancy. It has been associated with a wide variety of infections. TIP occurs primarily in young women but is similar in many ways to the hemolytic-uremic syndrome in children. The primary laboratory abnormalities include leukocytosis, thrombocytopenia, and microangiopathic hemolytic anemia characterized by the presence of bizarre red blood cells, especially helmet cells. The Coombs test is usually negative. Histologically, there is widespread occlusion of arterioles and capillaries by fibrincontaining hyaline thrombi. Punch biopsy of skin can often document these abnormalities. Although outcomes are improving,300 treatment is often unsuccessful and the course may be brief and fulminant. Glucocorticoids and. anticoagulants have been used successfully in some persons, and splenectomy is occasionally performed. Plasmapheresis and blood exchange have met with some success.301-303
Thrombocytopenia was observed in 50% of 106 children with SLE, often in association with leucopenia. 304 The frequency of thromboembolic events was higher (19%) in thrombocytopenic patients than in those with normal platelet counts (6%). Thrombocytopenia occasionally results from myelofibrosis. 305 A syndrome of auto-erythrocyte sensitization, consisting of large inflammatory lesions in the skin in response to minor trauma, was first described by Gardner and Diamond. 306 Subcutaneous injection of autologous erythrocyte membranes reproduces the lesions (Fig. 16-11). The syndrome is most often encountered in adolescent girls and has been associated with psychiatric disturbances. It has been described in one boy.307 There is no known relationship of this syndrome to SLE, but its clinical manifestations may mimic SLE.
• Figure 16-11 Volar surfaces of the foreanns of a lO-year-old boy diagnosed as having the Gardner-Diamond syndrome. A large area of inflammatory change and ecchymosis was produced in the right foreann by the subcutaneous injection of "ghosts" prepared from his red blood cell membranes. Except for subcutaneous bleeding, no inflammatory changes were produced in the left ann by injection of a comparable amount of buffer.
Vasculitis The vasculitis of SLE affects small blood vessels, arterioles, and venules, in contrast to polyarteritis, in which there is preferential involvement of the medium-sized arteries. 308 Lupus crisis is the sudden development of overwhelming, often fatal, systemic disease resulting from widespread acute vasculitis. Raynaud's phenomenon, a frequent finding in children with systemic connective tissue diseases, is characterized by sequential color changes in the distal extremities. The initial event is blanching of the distal phalanges, usually related to exposure to cold or emotional upset. This initial phase of digital ischemia is followed by cyanosis caused by anoxia and desaturation. A reactive phase of hyperemia supervenes, accompanied by ischemic pain. To make the diagnosis, blanching and at least one other color phase should be observed. Raynaud's phenomenon in SLE is in part vasospastic but is also indicative of structural vascular disease. The skin of the fingers may eventually become atrophic and shiny (sclerodactyly). Vascular necrosis, digital ulceration, and gangrene follow in a minority of affected children (Fig. 16-12). Significant Raynaud's phenomenon should suggest the possibility of an overlap syndrome (see Chapter 21). Edema of the lower extremities is usually related to congestive heart failure or nephrotic syndrome rather than to localized vasculitis.
Ocular Disease Cotton-wool spots (cytoid bodies) are evidence of retinal vasculitis (Fig. 16-13). They occur singly or in small numbers in a para-arteriolar location in the posterior pole of the retina. They are highly suggestive of the diagnosis of SLE in the absence of other systemic diseases (e.g., hypertension, diabetes, severe anemia) that can cause similar exudative lesions in the nerve cell layer. They have been occasionally reported in children with dermatomyositis, polyarteritis, or scleroderma. Other ocular manifestations
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
359
• Figure 16-13 An oval white cotton-wool spot (CNS) in the posterior retinal pole. ACNS is invariably in a para-arteriolar position and often partially obliterates the adjacent arteriole. CNSs arise from segmental vasculitis within and adjacent to these vessels.The surrounding retina generally shows an edematous sheen. In systemic lupus erythematosus, usually only afew CNSs are present. Identical lesions may rarely be seen in other connective tissue diseases. In hypertension, diabetes, or septicemia, numerous CNSs may be present. (See color insert.)
• ~ 16-12 Gangrene affecting the distal phalanx of the third finger of a 12-year-old boy with systemic lupus erythematosus.The distal digit eventually autoamputated.
include subretinal edema or hemorrhage, occlusion of the central retinal vein, and episcleritis. Severe vaso-occlusive retinopathy is associated with aPL, eNS disease, and a high rate of visual loss.309 Keratoconjunctivitis sicca may be part of secondary Sjogren's syndrome.
Sjiipen's Syndrome Sjogren's syndrome is an uncommon complication of childhood SLE, although it may be under-recognized. Until 1988, only 11 children had been reported with Sjogren's syndrome accompanying SLE or mixed connective tissue disease (secondary Sjogren's syndrome).310 It is characterized clinically by a gritty feeling in the eyes, conjunctival injection, and photophobia. Schirmer's test indicates deficient tear flow (less than 5 mm wetting of a filter paper strip in 15 minutes), and Rose-Bengal or fluorescein staining of the cornea may demonstrate superficial corneal erosions. Patients with keratoconjunctivitis sicca usually also have inflammation of the salivary glands with a deficiency of saliva resulting in painful or painless unilateral or bilateral parotid gland swelling, dif· ficulty chewing and swallowing, abnormalities of taste, severe dental caries, and halitosis. Demonstration of ecta-
sia of the parotid ducts by sialography31l or decreased uptake of 99mTc-pertechnetate, or abnormalities on ultrasonography, or magnetic resonance images312 support the diagnosis, which is confirmed, if necessary, by biopsy of a minor salivary gland from the lower lip demonstrating periductal lymphocytic infiltrates. 313 Dryness of other mucosal surfaces, including nose, pharynx, and vagina, may occur. Sjogren's syndrome is strongly associated with the presence of antibodies to the extractable nuclear antigens Ro/SS-A and La/SS-B (Table 16-17).
Gastrointestinal Manifestations Disorders of the gastrointestinal tract directly related to SLE are uncommon, although side effects of medications not uncommonly cause abdominal pain. An association between pancreatitis and SLE has been recorded in a number of children. 31 4-317 Symptoms include diffuse abdominal pain, nausea, and vomiting. Elevated serum
I!. II
TABLE 16-17
Charaderistics of Sjogren's Syndrome
Definition: Keratoconjunctivitis sicca (dry eyes secondary to decreased tear production by lacrimal glands) and xerostomia (dry mouth secondary to decreased saliva production by salivary glands) Primary Sjogren :s- syndrome: Not associated with any other disease; rare in childhood Secondary Sjogren's syndrome: Associated with a connective tissue disease, most often SLE or mixed connective tissue disease; disease uncommon in childhood Autoantibodies: Anti-Ro/SS-A (95%), ami-La/SS-B (85%)
360
C HAP T E R
16 S Y S T E M I C L u pus E R Y THE MAT 0
lipase and amylase levels suggest the diagnosis. There is usually a response to glucocorticoids, but this complication is occasionally fataJ.316 Drugs, including glucocorticoids and immunosuppressives, have been implicated in the etiology of pancreatitis, but the evidence for this causality is scant. 316 Protein-losing enteropathy has been reported in 3 children and more than 20 adults with SLE.318 Rare reports of children with SLE and celiac disease,319 ulcerative colitis,3 20 or Crohn's disease 321 probably represent coincidental occurrence of both diseases rather than specific associations.
Hepatic Disease Hepatomegaly occurs in up to two thirds of children with SLE but is usually of mild degree. In some cases, it may also be a manifestation of fatty infiltration of the liver related in part to glucocorticoid therapy.322 Rare hepatic complications include granulomatous hepatitis, nodular regenerative hyperplasia, and lupus anticoagulant-associated Budd-Chiari syndrome. 323 The relationship of so-called lupoid hepatitis, a term first used to describe a disease occurring in young women with hepatitis and systemic, serologic, and occasionally familial evidence of SLE, is controversial,324,325 and it is now generally agreed that there does not appear to be a true association with SLE.32S-327 This syndrome actually comprises a subset of patients with chronic active hepatitis who have hypergammaglobulinemia and autoantibodies, including ANAs, antimitochondrial antibodies, antibodies to the liver-kidney microsomal antigens and, in 70%, antibodies to smooth muscle, but no antibodies to dsDNA. It is rare in children and must be differentiated from the more common hepatic involvement of SLE.325 Some cases begin with an episode of acute viral hepatitis. Clinically, these patients develop prolonged jaundice, intermittent fever, and hepatosplenomegaly. Areas of fibrosis, nodular degeneration, piecemeal necrosis, and a striking infiltration of lymphocytes and plasma cells are present in liver biopsy specimens. 325
Lymphatic TIssue Involvement Splenomegaly of moderate degree is a feature of active disease. Splenic infarcts or perisplenitis may cause recurrent left upper quadrant pain. Functional asplenia, defined as failure of the spleen to accumulate radiocolloid, is a rare but potentially fatal complication. 328 ,329 It is not associated with clinical symptoms, except in the case of overwhelming pneumococcal sepsis. It can be suspected, however, if Howell-Jolly bodies are seen on examination of the peripheral blood smear by interference phase-contrast microscopy, and confirmed by demonstration of failure of splenic uptake of colloid such as 99mTc-labeled phytate. The authors have seen two such children, one of whom died of overwhelming pneumococcal sepsis. Spleen scans in 11 other children with active SLE failed to detect any abnormalities. In some instances, the functional defect is reversible. 329
SUS
About half of the children with SLE have localized or generalized lymphadenopathy. Occasionally, this may be so extreme as to initially suggest a diagnosis of lymphoma, a malignancy that rarely has been reported in association with childhood SLE.330-332 Kikuchi's syndrome (histiocytic necrotizing lymphadenitis) has been reported,333
Malignancy In a study of 724 adult patients with SLE who were monitored prospectively for 24 years, the overall risk for cancer was not increased over that of the general population. However, the risk of non-Hodgkin's lymphoma was increased fourfold. 3.31 An increased frequency of lymphoma was also demonstrated in a Danish study, in which increases in lung, liver, and vaginal cancers were also documented. 332 Whether these increased risks are conferred by the disease or by treatment with drugs such as cyclophosphamide is uncertain. In a short-term study of 75 patients with SLE who had received intravenous cyclophosphamide, neoplasia developed in 4. 334
Autoimmune Endoalnopathles Autoimmune thyroid disease occurs with increased frequency in children with SLE, and Hashimoto's thyroiditis may occasionally herald the onset of the disease. In one study, 4 of 35 children with SLE had clinically evident hypothyroidism, and a further 2 had an elevated serum thyroxine (T4) concentration but normal levels of thyroidstimulating hormone. Antithyroglobulin and antimicrosomal antibodies occurred in two thirds of patients with clinical or laboratory evidence of thyroid disease, but antibodies to thyroglobulin and microsomes were present, respectively, in 30% and 60% of euthyroid children with SLE.335 The high frequency of antithyroid autoantibodies in children with SLE is similar to that reported in adults with the disease. 336 No association of SLE with Graves' disease has been reported. There have been case reports of SLE and hypoparathyroidism,337 primary hyperparathyroidism,338 juvenile-onset diabetes mellitus,339 and Addison's disease..340,.34i Endocrine gland dysfunction secondary to arterial thrombosis as part of the APLS is rare and has not been reported in children.
PATHOLOGY The basic pathologic lesions are an immune complexmediated vasculitis and fibrinoid necrosis, inflammatory cellular infiltrate, and sclerosis of collagen. 342 The characteristic histopathologic lesions of SLE include hematoxylin bodies and so-called onion-skin lesions. Hematoxylin bodies, which can be found in any organ, are formed by the interaction of antibody to deoxynucleoprotein and degraded nuclear material. They are basophilic structures that, when engulfed by a phagocytic cell, result in the LE cell. In the absence of necrosis, hematoxylin bodies in tissue sections are pathognomonic for SLE. Onion-skin
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
361
lamination resulting from sclerosis of collagen reflects the effec~ of arteritis. It is typically seen in the penicilliary arteries of the spleen (Fig. 16-14) and in other organs in associ~tion with foci of fibrinoid necrosis. Endothelial thickening of capillaries, venules, and arterioles is characteristic of SLEj larger vessels are usually spared. Secondary changes include vascular obstruction and thrombosis, which are uncommon, except in the eNS and iI1 mesenteric vessels. In areas of severe inflammation, there may be almost total destruction of the connective tissues combined with cellular necrosis and the deposition of basophilic nuclear debris. These changes are most commonly seen in lymph nodes, heart, and glomeruli and are attribijtable to immune complex deposition. In one of the most complete histologic studies of necropsy findings in children with 5LE, Cook and associates 30 reported characteristic microscopic changes in the arterioles, consisting of fibrinoid necrosis of the media, proliferation and swelling of the intima, and varying degrees of periarteriolar inflammation (Fig. 16-15). The fibrinoid material is deposited within the interfibrillary ground substance of the connective tissues;
• Figure 16-15 Immune complex vasculitis in a patient with systemic lupus erythematosus. A small artery is the focus of the necrotizing arteritis, with periarterial round cell infiltration and involvement of the adjacent vein.
nearby connective tissue fibers are irregularly thickened. This pathologic change is most often identified in glomeruli and arterioles of the skin, heart, and other parenchymal organs but may be present in necrotic lymph nodes and synovium. It is not distinguishable microscopically from fibrinoid changes of other connective tissue diseases.
Renal Lesions A wide spectrum of abnormalities, including glomerulonephritis, interstitial nephritis, and necrotizing angiitis, characterize the renal pathology in SLE. The predominant abnormality, however, is usually one of the types of glomerulonephritis categorized in the World Health Organization (WHO) classification (Table 16-18).343 Indices of severity and activity can also be applied. The distribution of histologic patterns in 60 patients in the study of Sorof and coworkers344 is shown in Table 16-19. • FJg-. 16-14 Onion-skin lamination of the penidlliary arteries of the spleen, illustrating a typical pattern of sderosis of collagen in systemic lupus erythematosus (SLE).This section was from a young patient who had idiopathic thrombocytopenic purpura and who required asplenectomy.Three years later, she developed the typical manifestations of SLE.
,:. III rABLE
16-18
Glomerulonephritis The WHO classification of lupus nephritis (see Table 16-18) recognizes six categories that include at least two
World Health Organization Classification of Lupus Nephritis
Dass
'JYpe of Glomerulonephritis
Desalptlon
I IIA IIB
Normal Minimal change Mesangial glomerulitis
III
Focal and segmental proliferation
IV
Diffuse proliferative glomerulonephritis
V
Membranous glomerulonephritis
VI
Glomerular sclerosis
No detectable disease Normal LM; mesangial immunoglobulin and complement by IFM; mesangial deposits by EM Class IIA features plus mesangial hypercellularity (>3 cells per mesangial area or increased mesangial matrix); minimal tubular or interstitial disease Focal areas of intracapillary and extracapillary cellular proliferation, necrosis, karyorrhexis, and leukocyte infiltration in <50% of the glomeruli; subendothelial or mesangial deposits on IFM or EM; focal tubular and interstitial disease Class III changes involving more glomerular surface area and >50% of the glomeruli; IFM and EM show abundant subendothelial deposits; marked interstitial involvement, membranoproliferative has prominent mesangial cell proliferation and capillary wall thickening No mesangial, endothelial, or epithelial cellular proliferation; diffusely and uniformly thickened capillary walls; IFM and EM show mesangial and subepithelial deposits; minimal interstitial involvement Segmental or extensive sclerosis of glomeruli; fibrous crescents are common
EM. electron microscopy; IFM. immunofluorescence microscopy; LM, light microscopy.
362
.~ II
C HAP T E R
I
IV V
SYSTEMIC
Lupus
ERYTHEMATOSUS
TABLE 16 1.9 .Chdnge.in Rendl Histology Over . Tillle Dlstrlbullon of Hlstologl( P"II"rns 111 60 Pdht>nls
WHOaalS II III
16
Initial
Second
BIopsy (%)
Biopsy (%)
0 23 37 10
28
0 11
41 26 22
third or More Biopsy (%)
2 10 40 10
40
WHO, World Health Organization. Data from Sorof .1M, Perez MD, Brewer ED, et ai: Increasing incidence of childhood class V lupus nephritis. .I RheumatoI 25: 1413-1418, 1998.
different pathogenic mechanisms. Light, immunofluorescence, and electron microscopic findings are all used to define these categories. WHO class I denotes normal renal histology. Mesangial, focal, and diffuse proliferative glomerulonephritis result from the deposition of immune complexes from the circulation. Membranous glomerulonephritis results, at least in part, from in situ formation of immune complexes. In addition, mixed histologic patterns are common, and changes from one category to another over time are frequent. The relative frequency of each histologic type varies from series to series and reflects, at least in part, the indications used to perform the renal biopsy. However, mesangial and proliferative lesions (classes II, III, and IV) constitute the findings in at least 70% of biopsied patients. 344 Mesangial nephritis (WHO class II) is the most minimal glomerular lesion of lupus nephritis. By light microscopy, glomeruli appear normal (class lIA) or, at most, show a minimal increase in the number of mesangial cells and matrix (class IIB) (Fig. 16-16). The diagnosis depends on the demonstration of mesangial deposition of IgG and C3 by immunofluorescence microscopy (Fig. 16-17). Byelectron microscopy, electron-dense deposits are noted in the mesangium and along the paramesangial capillary basement membranes. Mesangial disease may progress to focal or diffuse proliferative nephritis. 345 Although children with class II nephritis may have low-grade protein-
• Figure 16-16 Mesangiallupus nephritis. Except for afew lobular areas of hypercellularity, these glomeruli from a renal biopsy of a 12-year-old black girl with acute systemic lupus erythematosus appear normal.
• FlllUre 16-17 Mesangiallupus nephritis. Immunofluorescent section from the patient in Rgure 16-16 stained for immunoglobulin G(IgG). Massive deposits of immunoglobulin are identified in the mesangium, in contrast to the paucity of abnormalities identified by light microscopy.
uria and hematuria, most do not develop renal insufficiency, particularly if the active systemic disease is adequately treated initially, with normalization of serologic abnormalities (Table 16-20). Focal segmental proliferative glomerulitis (WHO class III) is characterized by mesangial changes and areas of hypercellularity in fewer than half of the glomeruli in the biOpsy specimen (Figs. 16-18 and 16-19). The lesions may be proliferative, necrotizing, sclerosing, or a combination of these changes. In addition to mesangial changes, the lesions within each glomerulus tend to be located in the peripheral capillary loops, in contrast to the predominantly centrilobular distribution in class II lesions or poststrepto-
I'
"j
I~_.
TABLE 16 20
Mesdngidl Lupus Nel>hrilis
May be the initial immune complex lesion Usually no clinical features of renal disease, or there may be minimal proteinuria or hematuria Remission or progression of the nephritis may occur with transition to diffuse or membranous disease
• FlllUre 16-18 Focal proliferative lupus nephritis. A lobular area of hypercellularity and necrosis is seen in an otherwise normal glomerulus. The majority of the glomeruli appeared normal on light microscopy.
C HAP T E R
• r:...- II-I'
Focal proliferative glomerulonephritis demonstrated on direct immunofluorescence microscopy staining for Clq. Some areas of the glomerulus show intense staining, whereas others are entirely normal. (Courtesy of Dr. David Lirenman.)
coccal glomerulonephritis. Segmental proliferation of the glomerular tufts, which is usually accompanied by mesangial proliferation, is also present. Granular or "lumpybumpy" deposits of immunoglobulins and complement components are seen by immunofluorescence microscopy along the basement membranes. By ultrastructural studies, these are subendothelial electron-dense deposits of immune complexes along the capillary basement membrane with accompanying proliferative changes. Less often, a few subepithelial and intramembranous deposits may be found. Focal segmental proliferative nephritis may progress to diffuse disease in from 70/0346 to 35% of patients. 347 This transition is most likely to occur with prolonged, severe, active disease (Table 16-21). Diffuseproliferative glomerulonephritis (WHO class IV) is characterized by the uniform hypercellularity of more than 50% of the glomeruli in the biopsy specimen (Figs. 16-20, 16-21, and 16-22). The severity of the individual glomerular changes is usually more pronounced than in focal, disease. Fluorescence microscopy demonstrates the characteristic "lumpy-bumpy" deposition of immunoglobulin and complement along the peripheral capillary walls, as well as in the mesangium. Interstitial infiltrates and extraglomerular immune complex deposition may also be noted along the tubular basement membranes, within the walls of the peritubular capillaries, or in the interstitium. By electron microscopy, extensive immune deposits can be seen in the mesangium and subendothelial spaces, and, to a lesser extent, in the subepithelial areas and intramembranously. Ultrastructurally, these electrondense deposits may exhibit a fingerprint or microtubular . : . I ARL E 16 21
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
363
• Figure l1-Z0 Diffuse proliferative lupus glomerulonephritis. All glomeruli in the biopsy core were uniformly involved. Marked hypercellularity, hyaline thrombi, hematoxylin bodies, and areas of necrosis were present. Ared blood cell cast (atrow) is seen in an adjacent tubule.
appearance, a result of crystallization within the immune complexes (Table 16-22). Membranous glomerulonephritis (WHO class V) has been considered to be quite uncommon as an isolated abnormality. The frequency of this lesion may be increasing; it was found in 28% of initial biopsy samples of children with SLE in one center. 344 The frequency of this histologic pattern at the same center increased from 17% before 1995 to 64% after 1995. The characteristic lesion is deposition of immune complexes along the subepithelial surface of the glomerular basement membrane with obliteration of the foot processes but little cellular proliferation (Figs. 16-23 and 16-24). The basement membranes assume a rigid, enlarged, glassy appearance. There may be slight increases in the number of mesangial cells and in the density of the mesangial matrix. Silver or periodic acid-Schiff stains define a
Focal Proliferalive I upus Glomerulonephritis
Minimal proteinuria Microscopic hematuria Nephrotic syndrome or renal insufficiency in 20% Responsive to glucocorticoid therapy Does not usually progress to renal failure
• Figure 11-21 Fluorescence microscopy image of a renal biopsy section from ayoung girl who died with central nervous system lupus and nephrotic syndrome. Deposits of immunoglobulin Gare shown in a"lumpy-bumpy' pattern (subendothelial) and afiner granular pattern (epimembranous).
364
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
• Figure 16-22 Electron microscopy study of a biopsy specimen from a patient with diffuse proliferative glomerulonephritis showing subendothelial and paramesangial immune deposits (atrow).
I!. Ii
TABLE 16-22
Diffuse Proliferative Lupus Glomerulonephritis
Proteinuria and hematuria Nephrotic syndrome and renal insufficiency in 60% Limited remission in 50% Renal failure in some patients, 5-10 yr after onset
• Figure 16-24 Electron microscopic section of membranous nephritis showing an epimembranous complex (atrow).
spike pattern along the basement membrane related to immune complex deposition on the capillary loops. Fluorescence microscopy shows a granular deposition of IgG and complement along the basement membrane that is usually finer and smaller than the deposits found in diffuse glomerulonephritis. In addition, there may be intra-basement membrane deposits, suggesting that in certain situations immune complexes can traverse the basement membrane from the subendothelial to the subepithelial surfaces (Table 16-23). Glomerular sclerosis (class VI) is actually not a category of the WHO classification but is customarily appended to it to accommodate the classification of patients in whom the predominant histologic change is focal segmental glomerular sclerosis. Although extensive sclerosis indicates a poor prognosis, minimal to mild sclerosis may be present in patients who have a stable disease course..~46 Correlations between glomerular histopathologic changes and clinical features of renal disease are summarized in Table 16-10. Children with mesangial lupus nephritis seldom have clinical evidence of renal disease, although there may be minimal proteinuria and microscopic hematuria. In focal segmental proliferative glomerulitis, there may be proteinuria and mild hematuria, but renal insufficiency is either absent or minimal, and the nephrotic syndrome is distinctly uncommon. Children with diffuse proliferative glomerulonephritis have proteinuria and hematuria in most cases, and, in 60%, nephrotic syndrome or renal insufficiency. Children with membranous lupus glomerulonephritis have persistent nephrotic
1_-
IlIi .. TABLE 16-23
• Figure 16-23 Membranous lupus nephritis.This patient was a 17-yearold white girl with nephrotic syndrome and congestive heart failure. She died of arrhythmia. libman-Sacks endocarditis was found at necropsy (see Rg. 18-27). There was uniform thickening of endothelial walls with a"glassy" refractivity on hematoxylin-eosin staining. Minimal hypercellularity was observed, and deposits of inflammatory cells were present in the tubular interstitial tissues.
Membr'\I1ous Lupus Glomerulonephntis
Persistent nephrotic syndrome Hypertension in 30% Remissions in 30% Eventual renal insufficiency in majority
C HAP T E R
syndrome, and one third have hypertension. Severe glomerular sclerosis represents an end-stage lesion in which the nephrotic syndrome, renal failure, and hypertension are common. In addition to histologic type, an estimate of the level of in:t1ammatory activity can be made on the basis of the renal biopsy by comparing the number of active lesions with those of chronic disease. 348 Active disease is characterized histologically by the presence of intracapillllry cellular proliferation; polymorphonuclear infiltrates; karyorrhexis; epithelial crescents; subendothelial fibrinoid change (wire loops); hyaline, fibrin, or platelet thrombi; interstitial inflammation; and necrotizing vasculitis. Chronicity is marked by segmental, global, mesangial, or vascular sclerosis, glomerular obsolescence, thickening of capillary basement membrane, fibrous adhesions or crescents, and tubulointerstitial scarring (Table 16-24).349
Inter$tltlal Nephritis Approximately half of the children with significant involvement of the glomerulus also have evidence of interstitial nephritis,350 and the severity of interstitial disease usually correlates with the severity of the glomerular changes. Rarely, severe interstitial nephritis is observed in the absence of glomerular abnormalities. Focal and diffuse infiltration with inflammatory cells, tubular necrosis, and interstitial fibrosis may be observed. Immunoglobulins and complement are deposited along the peritubular capillaries or the tubular basement membrane in a granular pattern. In some patients, tubular acidosis is present.351 Other patients have interstitial nephritis and isosthenuria as part of Sjogren's syndrome. Analgesic nephropathy as a cause of interstitial inflammation and tubular damage should always be considered in patients with these changes. The importance of renal tubular disease in determining outcome was evaluated by Daniel and colleagues. 352 The more severe the tubular lesion, the greater the likelihood of end-stage renal failure. Tubule cell expression of ICAM-I and CD40 also predicted progression of disease, even in the absence of limited tubular atrophy.
Necrotizing Angiitis Some children with lupus nephritiS may also demonstrate an lmmune complex arteriolitis in renal tissues that may be 'indistinguishable from other forms of necrotizing arteritis. Fibrinoid necrosis, thrombosis, and arteriolar
I,:
I( IABU 16-24 lupus
Assessment of Activity and Chronicity in Renal
~cators of ActIYe Disease
Indicators of Chronicity
C,Uular proliferation N~crosis, karyorrhexis Cellular crescents Wire loops, hyaline thrombi Leukocytic glomerular infiltration Interstitial infiltration
Glomerular sclerosis Interstitial fibrosis Fibrous crescents Tubular atrophy
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
365
inflammation are usually present. This type of vasculitis may be associated clinically with fulminant renal failure and malignant hypertension or renal venous thromboses. The role of the renal biopsy in the evaluation of lupus nephritis is controversia1,353 It is seldom needed to make the diagnosis of SLE, but it is indicated in at least three circumstances: (1) in the child with nephrotiC syndrome in whom differentiation of diffuse proliferative from membranous glomerulonephritis is important; (2) in one in whom, despite high-dose glucocorticoids, renal function is deteriorating, or there is persistent hematuria or proteinuria or both, to determine whether the renal disease is likely to be responsive to cytotoxic agents (Le., if there is evidence of activity or chronicity); and (3) as a prerequisite to entry into clinical therapeutic trials. 353 Extensive glomerular abnormalities have been identified on biopsy of patients who have no clinical evidence of renal disease. 354-356 Diffuse nephritis without clinically apparent disease may not always portend as poor a prognosis as that for clinically obvious disease. 357- 359 Histologic abnormalities change over time in many patients,344 and sequential biopsy findings have provided some insight into the progression of the renal lesions. However, these studies must be interpreted with caution, because indications for biopsy and the influence of therapy undoubtedly affect the results.348.360 In general, however, sequential biopsies in children have demonstrated progression of mesangial or focal proliferative glomerulonephritis to diffuse proliferative glomerulonephritis in months to years.347.361.362 Occasionally, progression from diffuse proliferative to membranous disease or, less commonly, from focal proliferative to membranous disease has been documented. 363 .364 In some studies, improvement was noted in 15% to 20% of patients in whom the initial biopsy had shown diffuse proliferative or focal proliferative glomerulonephritis, but membranous disease tended to be worse on subsequent examination.
Skin The typical cutaneous lesion of acute SLE is liquefaction of the epidermal basilar layer with disruption of the dermal-epidermal junction, edema of the dermis, infiltration of T lymphocytes throughout the epidermis and around blood vessels, and fibrinoid degeneration of the connective tissues (Fig. 16-25). In more chronic lesions, there is epidermal atrophy, hyperkeratosis, follicular plugging, and proliferation of elastic tissues. IgG and C3 can be identified by fluorescence microscopy along the epidermal basement membranes in uninvolved and non-sunexposed skin as well as in lesional biopsy specimens (Fig. 16-26).365-367 This finding of the "lupus band" on dermal punch biopsy specimens is not pathognomonic for SLE but may be a useful diagnostic test in difficult cases.
Lungs The pulmonary histology in acute pulmonary involvement in SLE is nonspecific. A basilar interstitial pneumonitis may lead to atelectasis and basophilic mucinous
366
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
fibrosis, especially in steroid-treated patients,247 The basic lesion of the endocardium consists of nodule formation along the atrial or ventricular surfaces of the valve leaflets, or on the endocardium of the chambers (Fig. 16-27). The Libman-Sacks lesion has been described as having three zones: an outer exudative zone of fibrin containing nuclear debris, including hematoxylin bodies; a middle zone of proliferating fibroblasts; and the inner zone characterized by neovascularization. 369 The mitral valve is most commonly involved.
CenkalNe"ousSydem
All layers of the heart may be involved by the pathologic processes of SLE. Pericarditis consists of focal or diffuse fibrinous depOSits or fibrosis. Pericardial fluid contains high numbers of leukocytes, almost all of which are neutrophils. 368 Myocarditis is typified by plasma cell and lymphocyte infiltration of the myocardium, sometimes with
There are no pathognomonic pathologic findings in CNS lupus, and anatomically identifiable lesions do not always account for the neurologic manifestations of the disease. 37o True vasculitis is rare; perivasculitis is more common and may lead to infarction, encephalomalacia, or other focal lesions (Figs. 16-28 and 16-29). Microinfarcts and nonspecific destructive and proliferative arteriolar and capillary lesions occur. 371 Thrombosis as part of the APLS may affect small arterioles or venules, although the direct evidence that this is responsible for many of the CNS lesions is unclear. In a study of the brains of patients with chorea (many of whom were children), infarcts, but not arteritis, were found in the basal ganglia. 212 Most patients with chorea and SLE have aPL. 372 Retinal cotton-wool spots (cytoid bodies) represent areas of inflammatory edema and degeneration of the ganglion cells arising from a periarteriolitis within the nerve fiber layer, presumably related to immune complex deposition. In an immunofluorescence microscopy study, immunoglobulin was demonstrated in the vascular layer of the capillaries of the choroid and around the basement membranes of the ciliary processes and bulbar conjunctivae. 373
• Rgare 16-26 Direct immunofluorescence photomicrograph showing granular deposits of immunoglobulin Galong the basement membrane zone ("lupus band") in a biopsy sample of nonaffected skin in a patient with systemic lupus erythematosus. (Courtesy of Dr. Richard Crawford.)
• figure 16-27 libman-Sacks endocarditis.This patient was a 17-year-old girl with an acute exacerbation of systemic lupus erythematosus who had cardiac and pulmonary failure.The heart was minimally enlarged. A loud systolic murmur and afaint diastolic murmur were present at the apex. Necropsy examination of the mitral valve leaflets showed many nonbacterial verrucae at the margins.
• Rgare 16-25 Skin biopsy of systemic lupus erythematosus rash (hematoxylin and eosin stain, magnification x 33) demonstrating follicular plugging, squamatization, and vacuolation of the basal keratinocytes; thickening of the basement membrane zone; and telangiectasia. There is a small dermal perivascular lymphocytic infiltrate. (Courtesy of Dr. Richard Crawford.)
edema of the alveoli with hyaline membrane formation. There is interstitial infiltration of lymphocytes and other inflammatory cells, alveolar hemorrhage, formation of hyaline membranes, and focal necrosis. Fibrinoid necrosis of arterioles and capillary thrombi may occur, and hematoxylin bodies may be seen.
Heart
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
367
The walls of the gastrointestinal tract are also sites of focal or diffuse vasculitis. Acute hemorrhage or infarction results from thrombosis of the mesenteric vessels. Mesenteric vasculitis is a rare but serious occurrence in the child with SLE. Hepatic biopsies in 33 adults with SLE and elevated liver enzymes demonstrated steatosis (12 patients), cirrhosis (4), chronic active hepatitis (3), chronic granulomatous hepatitis (3), centrilobular necrosis (3), chronic persistent hepatitis (2), and microabscess (2).374 More recent studies have reported the presence of multiple fibrin microthrombi, liver infarction, hepatic veno-occlusive disease, and Budd-Chiari syndrome (hepatic vein occlusion with cirrhosis and ascites) related to the presence of aPL,375 Hyperplasia of plasma cells, vasculitis, and the occurrence of hematoxylin bodies characterize lesions of the lymph nodes.
LABORATORY EXAMINATION
• fItI-e 16-28 Magnetic resonance image of the brain of a 12-year-old girl wlttl systemic lupus erythematosus who suffered widespread seizures due to central nervous system vasculitis. Pale areas of the deep cortex were observed on the initial study.
Laboratory investigations include indicators of inflammation, tests for the presence of autoantibodies (particularly those directed to nuclear antigens), tests that evaluate end-organ involvement, and tests that monitor the effects of therapy, including drug-related toxicity. A useful overall approach to use of the laboratory in diagnosis of SLE has recently been described. 376
Indicators of Inflammation
• ,.... 16-29 The abnormalities seen in Rgure 18-28 were no longer demonstrated on a second study 1 month later.
Most acute phase indices of inflammation are increased in children in proportion to the activity of the systemic disease. These include an increased erythrocyte sedimentation rate (ESR), polyclonal hypergammaglobulinemia, and increased serum levels of a2-globulins. 376 C-reactive protein (CRP), an important acute phase protein that is elevated in most inflammatory conditions, is often normal.376-379 However, it is increased in patients with SLE and systemic infection380 and in those with serositis377 or arthritis. 38 1.382 Serum ferritin levels are high in adult patients with active SLE and correlate with elevated anti-dsDNA and low complement. 383 In one study, factor VIII-related antigen (von Willebrand's factor) was elevated in approximately half of the children. 384 The discrepancies among the acute phase responses may reflect the intricate eytokine abnormalities in SLE and their influence on the synthesis of specific acute phase proteins. Patients with SLE have low levels of IL-l, the cytokine responsible for the induction of CRPj high ferritin levels may reflect the high IL-6 or TNF-a. levels seen in this disease. In some studies, however, correlations between eytokine levels and acute phase protein levels were inconsistent.385
Other Organs The synovitis in children with SLE is nonspecific and is usually mild in degree. Changes include proliferation of syrtovial lining cells and microvascular abnormalities, including vasculitis and inflammatory cell infiltration. The myopathy is not specific and includes necrotizing arteritis, mild interstitial myositis, and vacuolization of muscle fibers.
Hematologic Abnormalities Anemia Mild or moderate anemia occurs in approximately one half of children with SLE and is usually typical of chronic disease (normocytic, hypochromic), with decreased
368
I!'.
c
HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
TABLE 16-25 Hematologic Abnormalities in Systemi( lupus Erythematosus
Abnormality Anemia (hematocrit <30%) Acute hemolytic anemia Leukopenia <4500 WBClmm; «4.5 x 109/L) <2000 WBC/mm; «2 x 109/L) Thrombocytopenia <150,000 plts/mm3 «150 x 109fL) <100,000 pltsfmm' «100 x 109fL)
Frequency (0/0) 50
5 40 10 30
5
pits, platelets; WBC, white blood cells.
serum iron and normal or slightly reduced iron-binding capacity (Table 16-25), In other patients, it reflects autoimmune hemolysis caused by IgG complementfixing antibodies to erythrocytes that are detected by an antiglobulin test (Le" Coombs' test), Occasionally, lupus presents with severe hemolytic anemia, but ordinarily the level of hemoglobin is only moderately depressed. Anemia associated with cold agglutinins directed against the I antigen of erythrocyte membranes is also found in some children. Hemolytic anemia is seldom severe and is rarely fatal. Pernicious anemia has been rarely reported in adults with SLE.386 Hypersplenism, drug sensitivity, and, rarely, microangiopathy may also contribute to anemia. Drug-induced gastrointestinal tract ulceration occasionally contributes to anemia. The presence of Howell-Jolly bodies indicates the splenic dysfunction and should be sought regularly (Fig. 16-30).
Leukocytes and Platelets Although leukocytosis may occur, lymphocytopenia (fewer than 1500 cells/mm3) is common in patients with active disease,387 and neutropenia, often in association with thrombocytopenia, occurs in up to one half of these patients,304 Severe neutropenia (fewer than 1000 neutrophils/mml [1 x 109/L)) is uncommon, Some children have clinically important immune thrombocytopenic
purpura, usually in association with splenomegaly. Bone marrow examination in these cases confirms an increased number of megakaryocytes with poor platelet budding, Patients with thrombocytopenic purpura and hemolytic anemia (Evans' syndrome) may progress to SLE or eventually develop the features of TfP, Myelofibrosis has been reported in fewer than 10 patients, including adolescents. 305
Coagulation Abnormalities Lupus Anticoagulants The lupus anticoagulant is defined as prolongation of the aPTT and prothrombin time or the presence of an increased DRWT, Addition of fresh plasma incompletely corrects the defect. This phenomenon results from the effect of antibodies that bind ~2-glycoprotein I and prothrombin, thereby interfering with the interaction of the prothrombin-activator complex (factors Xa and V, calcium, and phospholipids) and preventing the conversion of prothrombin to thrombin by thromboplastin,385 Abnormalities of at least one of nine different tests were demonstrated in 41% of patients with SLE, but no individual test did better than the DRWT, which detected only 78% of patients with these abnormalities,390 It is likely, therefore, that a combination of assays is necessary to effectively screen a lupus population for lupus anticoagulant, and in this study, a combination of the DRWT, aPTT, and dilute thromboplastin tissue assay identified all patients with any abnormal test result. A combination of DRWT and aPTT detected 91% of those with any abnormal test result. 390 In this study, there were no correlations between abnormal clotting tests and the presence of high titers of anticardiolipin antibodies,
Antiphospholipid Antibodies Most patients with lupus anticoagulant have antibodies to cardiolipin that cross-react with a range of negatively charged phospholipids (Table 16-26). The reported frequencies of anticardiolipin antibodies in childhood SLE have ranged from 37%3R7 to 100%.48 A correlation with the presence of CNS disease was observed in one study.410 A similar antibody species directed against phospholipid is responsible for the false-positive serologic reaction for syphilis seen in this disease. aPLs are not restricted to patients with SLE. They can be detected in a wide variety of neoplastic, infectious, inflammatory, and autoimmune diseases, In a study of fI • TABLE 16-26 Antigenic specificities Mechanism of action
Clinical correlations
• Figure 16-30 Photomicrograph of a smear of peripheral blood stained with Wright's stain, showing erythrocytes containing small, dark Howell-Jolly bodies, (Courtesy of Dr. L Wadsworth.)
Anticardiolipm Antibodies Phospholipids (diphosphatidyl glycerol) Inhibition of prothrombin activator complex (Xa, V, Ca'+, lipid), platelet aggregation, .!- prostaglandin I, release Recurrent thromboses, livedo reticularis, chorea, recurrent fetal loss, cerebrovascular accidents, hypertension
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
369
other aPL in childhood SLE, a lupus anticoagulant was found in 55% and a false-positive serologic test for syphilis in 38%.388
Antlnudear Antibodies ANAs are present in the sera of almost all children with active SLE (Table 16-27). In fact, the absence of ANAs, particularly in the presence of symptomatic disease, all but excludes the diagnosis of SLE. The titers of ANAs as demonstrated by immunofluorescence microscopy on HEp-2 cells range from low (1:80) in inactive disease to extremely high (greater than 1:5120) in active disease. There is considerable interpatient variability, and determination of an ANA titer alone is not sufficient to diagnosis SLE or to monitor the disease course. The pattern of nuclear immunofluorescence is usually peripheral or homogeneous and suggests the presence of antibodies to dsDNA (Fig. 16-31).376 The first evidence for the presence of ANAs in SLE was the demdnstration of the LE cell (Fig. 16-32).7 The antibody responsible for this phenomenon is directed to deoxyribonucleoprotein (anti-DNP) or anti-DNA histone. Although the LE cell test is quite specific for SLE, the cell can be present in other disorders, and its low sensitivity makes it a poor screening test. For these' reasons, and because it is labor intensive, it has been supplanted by the more sensitive, if less specific, test for ANAs.
,~.
I ABI E 16-27 Characteristic Autoantibodies in Systemic I upus Erythematosus
Antl.uclear AntIbodies
Other Autoantibodies
Anti-dsDNA antibodies Antj·DNP antibodies Anti-Ro (SSIA) antibodies Anti-La (SS/B) antibodies Anti-Sm antibodies Anti-histone antibodies
Anti-erythrocyte antibodies Anti-Iymphocytotoxic antibodies Anti-tissue specific antibodies Antiphospholipid antibodies Rheumatoid factors
• Figure 16-3Z lupus erythematosus cell preparation. The lupus erythematosus cell, a polymorphonuclear leukocyte containing a homogeneous nuclear indusion, is identified by the arrow.
Antibodies to DNA Antibodies to dsDNA are virtually pathognomonic of SLE, occur in almost all children with active disease, and are present at particularly high titers in children with active nephritis (Table 16-28).391-393 They are rarely, if ever, associated with other rheumatic diseases. Antibodies to single-stranded DNA (anti-ssDNA) occur in approximately 50% of children but are also present in those with a variety of other rheumatic disorders and infections and are, therefore, of little diagnostic significance. 394 Antibody to dsDNA is measured by a number of methods,376 including a radioimmunoassay that uses radiolabeled dsDNA, a fluorescence microscopy assay that uses the protozoan Crithidia luciliae, or an enzymelinked immunosorbent assay (ELISA). The radioimmunoassay is sensitive and provides semiquantitative data, but, because the substrate may contain contaminating ssDNA, it may lack specificity. The kinetoplast of C. luciliae consists only of dsDNA; although it is less sensitive than radioimmunoassay, assays using this substrate are highly specific for antibodies to dsDNA (Fig. 16-33). Enzyme-linked immunosorbent assays (ELISAs) provide semiquantitative information, but, like the radioimmunoassay, they may have a higher level of false-positive results. For diagnostic purposes, the Crithidia assay is often preferred; for monitoring levels of antibody to dsDNA during the course of therapy, the
II II
TABLE 16-28
Antigenic specificity Mechanism of action
• FIgwe 16-31 Demonstration of antinuclear antibodies (ANA) on HEp-2 cells by indirect immunofluorescence microscopy.The homogeneous pattern of staining is characteristic of systemic lupus erythematosus. (Courtesy of Kallestad laboratories, Minneapolis, MN.)
Clinical correlations
Anti-dsDNA Antibodies
Native, double-stranded DNA Forms complement-activating immune complexes that deposit in tissues Highly specific for systemic lupus erythematosus Active glomerulonephritis
370
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
I!1_-..
1ABI E 16 29
Antigenic specificity Mechanism of action Clinical correlations
Anli Ro (SS A) Antibodies
52- and 6o-kD peptides complexed to YI, Y3, Y4, and Y5 RNAs Interferes with RNA translation or transport Neonatal lupus syndromes, especially complete heart block Subacute cutaneous lupus Sjogren's syndrome Renal and pulmonary disease
Anti-Ro/SS-A
• figure 16-33 Demonstration of antibody to double-stranded DNA (dsDNA) using Crithidia ludliae. The brightly staining kinetoplast at the base of the flagellum indicates the presence of antibodies to dsDNA, as revealed by Indirect immunofluorescence. (Courtesy of Kallestad Laboratories, Minneapolis, MN.)
radioimmunoassay or ELISA is superior. Increased or increasing levels of antibody to dsDNA are often a clue to the development of active renal disease, particularly if accompanied by decreased or decreasing levels of complement. There is considerable heterogeneity among antidsDNA antibodies, and only some are capable of inducing immune complex disease. Factors that may affect pathogenicity include the antibody isotype, the ability to penetrate cells and activate complement, and the avidity of the antibody for dsDNA and other antigens (i.e., crossreactivity).395.3% The relationship between serum level of anti-dsDNA and disease activity is controversial.395 In some studies, high levels of antibody were correlated with active disease; in others there was no such correlation. Some studies have demonstrated that an increase in antidsDNA levels precedes a flare of disease; others have shown a preCipitous fall in anti-dsDNA levels at the time of a flare.
The Ro/SS-A antigen consists of two polypeptides of 52 kD and 60 kD that are complexed to cytoplasmic RNAs Y1, Y3, Y4, and YS. Most reactive sera recognize the 6O-kD moiety. Anti-Ro is most strongly associated with neonatal lupus erythematosus, subacute cutaneous lupus, homozygous C2 and C4 deficiency, interstitial lung disease, skin photosensitivity,398.399 and Sjogren's syndrome. 4°O These antibodies are also present in about one fourth of patients with typical SLE,'particularly in association with active renal disease. 401 Up to 15% of normal individuals have low levels of anti-Ro activity402 (Table 16-29). Patients who have only anti-Ro antibody occasionally have negative findings on tests for ANA by immunofluorescence microscopy on tissue substrates (ANA-negative lupus),403,404 although the majority of such patients have a positive result on HEp-2 cells. The authors have seen one such patient, a teenaged boy with anti-Ro antibodies, who presented with classic discoid cutaneous lesions, developed CNS disease, and died from cerebromalacia (see Fig. 16-7). Another developed a butterfly rash with anti-Ro antibodies after griseofulVin administration for a presumed cutaneous fungal infection.
Anti-La/SS-B The La/SS-B antigen is a 48-kD nuclear phosphoprotein that is complexed to the Y RNAs, as well as to other immature transcripts of RNA polymerase III. Patients with anti-La almost always also have anti-Ro, although the converse is not true. Anti-La is associated with Sjogren's syndrome399 and with neonatal lupus. 405 Approximately 5% of the normal population have low levels of anti-La detected by ELISA (Table 16-30).
Antibodies to Extractable Nuclear Antigens
Anti-Sm
Antibodies to the extractable nuclear antigens (Sm, Ro/SS-A, La/SS-B) are strongly associated with SLE, neonatal lupus erythematosus, and Sjogren's syndrome. High titers of anti-RNP are associated with SLE and mixed connective tissue disease. The presence of anti-Sm constitutes one of the criteria for classification of SLE and is highly specific for this disease. The frequencies of antibodies to the extractable nuclear antigens are well documented in adults with SLE. In a long-term follow-up of 100 adults with SLE, 39 had anti-Ro, 13 anti-La, and 7 anti-Sm. 397 In a study of 22 children with SLE, anti-Ro was present in 29%, anti-La in 10%, anti-Sm in 14%, and antiRNP in 19%.48
The Sm antigen is composed of five small uridine-rich RNAs (Ul, U2, U4, U5, and U6) and is associated with a
.~
1_ _
.. I ABlE 16 30
Antigenic specificity Mechanism of action Clinical correlations
Anti I d (55 B) Antibodies
48-kD nuclear phosphoprotein that is complexed to the Y RNAs Interferes with action of RNA polymerase III Neonatal lupus syndromes Sjogren's syndrome Almost always found in association with anti-Ro
C HAP T E R
'='11
(ABLE 16-31
Anti-SmAntibodies
Vridine-rich RNAs VI, V2, V4, V5, and V6 associated with the core Sm peptides BIB', D, E, F, and G Involved with RNA processing, messenger RNA synthesis and splicing Specific for systemic lupus erythematosus Related to central nervous system disease
Antigenic specificities
Mechanism of action Clinical correlations
number of polypeptides with which anti-Sm antibodies react,406 Core Sm proteins B (29 kD), B' (28 kD), D (16 kD), E (12 kD), F (11 kD), and G (9 kD) have been identified. The polypeptides most often recognized by anti-Sm are B/B'.406 B' is found only in human tissues. Antibodies to the Sm antigen complex that are specific for SLE usually occur together with anti-U1 RNP antibodies in CNS disease (Table 16-31),407,408 although more recent studies have failed to demonstrate strong associations between anti-Sm and disease characteristics. 409 Anti-Sm antibodies are found in higher frequency in African Americans than in whites. 410
Antl·U1 RNP Antibodies to a portion of a polypeptide associated with Ul RNA (anti-RNP) are found in low titers in children with SLE, but in high titers they are associated with mu:ed connective tissue disease. 41l They are characterize~ by a speckled pattern of nuclear fluorescence on tests for ANAs and are confirmed by counterimmunoeld<;:trophoresis or ELISA (Table 16-32).
Antlhistone Antibodies Antibodies to histones are present in many children with SU:, but they were first detected in patients with druginduced SLE by Fritzler and Tan. 412 Specificities differ markedly between spontaneous SLE and the druginduced disease (Table 16-33).4l.3-415 In procainamideinduced lupus, antibodies react with H2A, H2B, and the HZA-H2B complex. In hydralazine-induced disease, they react primarily with H3 and H4. The finding of antihistone antibodies in the absence of high titers of antidsDNA antibodies is highly suggestive of drug-induced SLE. Antibodies to histones are most commonly demonstrated by an ELISA.
_I
'I •
fABLE 16-32
Antigenic specificity Mechanism of action Clinical correlations
RNP. ribonucleoprotein.
Anti-UI RNP Antibodies
Polypeptide associated with VI RNA Interferes with synthesis of RNA Low titer: systemic lupus erythematosus High titer: mixed connective tissue disease
16
e III
SYSTEMIC
Lupus
TABLE 16 -33
Anti-Histone Antibodies
Antigenic specificities Mechanism of action Clinical correlations
ERYTHEMATOSUS
371
Histone complex (H2A-H2B) HI > H2B > H2A > H3 > H4 Interferes with nucleosome packaging of DNA Drug-induced lupus; systemic lupus erythematosus
Antiglobulins (Rheumatoid Fadors) Rheumatoid factors are present in approximately 10% to 30% of children with SLE, but high titers in the presence of a lupus-like disease may suggest the presence of an overlap syndrome. Antibodies to 19A are common in 19Adeficient patients with lupus. 416
Immune Complexes Although immune complexes are fundamental to the pathogenesis of SLE, their measurement in the peripheral blood adds little to the diagnosis or management of the disease. In some studies,417 detection of serum immune complexes correlated better with disease activity than did the anti-dsDNA or C3 levels, but this has rarely been the authors' experience. Intertest agreement of immune complex assays is often poor, and correlations with disease subsets or disease activity is variable. For these reasons, immune complex assays are of very limited utility in the diagnosis or management of SLE. 418 Cryoglobulins containing ANA and DNA are sometimes present in active SLE and may be responsible for certain expressions of the disease by intravascular activation of complement or precipitation of the hyperviscosity syndrome (Raynaud's phenomenon, acrocyanosis, purpura, and abdominal pain).
Complement Determination of the serum complement level is one of the most important laboratory measures of active SLE.376 Specific components of the complement sequence (e.g., C3, C4) may be assayed, or the total hemolytic complement (CH 50) may be titrated by red cell lysis. The CH 50 reflects the integrity of the total complement cascade and can be measured for either the classic or the alternative pathway of complement activation. It is abnormally low in approximately 90% of children with active nephritis. The C3 concentration is depressed less often than CH50 or C4. A low C4 concentration is a consistent and reliable indicator of active nephritis in SLE if the baseline level is known in a particular patient and the child does not have a genetically determined deficiency of C4. A more precise method of determining the state of complement activation is to measure its activation products. Elevations of complement factors Bb and C4d more accurately reflected disease activity than did measurements of C3, C4, or CH 50 in one study.419 Normal or moderately reduced levels of C3 and C4 in a patient with undetectable complement activity as measured by the hemolytic assay suggest a genetically determined deficiency of one of the complement components.
372
I! II
C HAP T E R
lABL E 16 34
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
Evaluation of Lupus Nephritis
Urinalysis Chemical and microscopic; culture if white blood cells present Measurement of glomerular function Plasma creatinine, urea nitrogen Creatinine clearance; 24-hr protein excretion Radionuclide glomerular filtration rate Evaluation of disease activity Serum anti-dsDNA antibody level Serum complement assay Renal ultrasonography and biopsy Light, immunofluorescence, and electron microscopy
Urinalysis and Evaluation of Renal Involvement Most children with active lupus nephritis have abnormalities of the urinary sediment that are characteristic of the specific type of renal involvement, or they have telescoped urine (a urinary sediment that shows, at one time, the sequential features characteristic of the course of glomerulonephritis). Proteinuria is probably the most common abnormality, but hematuria and red blood cell casts are more important hallmarks of active glomerulitis. The latter are also useful clinical markers in follOWing the course of treatment. Proteinuria is indicative of both glomerular and tubular abnormalities and, like the creatinine clearance, may not be a satisfactory short-term indicator of therapeutic response. In severe renal disease, the sediment also contains increased numbers of cellular and fatty casts. With the onset of the nephrotic syndrome, doubly refractile oval
,,:~ TABLE \6 35 Part I Part II Part III Part IV
Part V
Analysis of Inflammatory Fluids The synovial fluid in SLE is usually inflammatory with a low white blood cell count (fewer than 2000 cells/mm' [2 x 109/L]). The protein content varies from transudative to exudative levels. Synovial fluid complement levels
Lupus Adivity Index (LAI)
Physician's global assessment on a VAS Assessment of four symptoms (fatigue, rash, joint involvement, serositis) on a VAS Assessment of severity of involvement of four organ systems (CNS, renal, pulmonary, hematologic) on a VAS Assign points for medication used: Prednisone 0-15 mg/day 16-39 mg/day >40 mg/day Any immunosuppressive agent Not used Used Assign points for three laboratory values: Proteinuria 1+
Interpretation
fat bodies appear and assume the form of Maltese crosses on polarizing microscopy. Profuse proteinuria and a ftxed speciftc gravity (1.010) are characteristic of the chronic phase of lupus nephritis. Broad casts of renal failure are seen at that time, and there may be few cellular elements in the sediment. Other abnormalities may include renal tubular acidosis. Evaluation of the extent and activity of renal involvement in SLE requires the integration of several laboratory features (Table 16-34). The combination of high levels of anti-dsDNA and low levels of complement, especially C4, in the presence of an abnormal urinalysis makes active lupus nephritis a virtual certainty.420 Rarely, a child with seemingly active SLE has a near-normal complement level. (Because serum complement components are increased by inflammatory disease, a previously elevated level may be simply lowered to normal levels by immune complex fixation, rather than being reduced to subnormal levels.) Occasionally, a child with SLE has hemolytic complement values in the subnormal range for long periods with no other indication of active renal disease. This observation may reflect the catabolic events associated with glucocorticoid therapy, general illness, or uremia that impairs the syntheses of complement components, particularly C3, rather than active disease per se. 421
2+ to 3+ 4+ Anti-dsDNA C3, C4, or CH so LAI score is arithmetic mean of the follOWing: Part I Part II Part III Part IV Part V
o to 5 points o to 3 points o to 3 points 1 point 2 points 3 points
o points 3 points 1 point 2 points 3 points 0-3 points depending on degree of abnormality 0-3 points depending on degree of abnormality
Number of points Mean of 4 point values Maximum of 4 point values Mean of 2 point values Mean of 3 point values
CH,,,, total hemolytic complement; eNS, central nervous system: VAS. visual analogue scale. Adapted from Petri M. Genovese M, Engle E, Hochberg M: Definition, incidence, and clinical description of flare in systemic lupus erythematosus. A prospective whort study. Arthritis Rheum 34: 937, © 1991 Wiley-Liss, Inc. Reprinted by pennission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
C HAP T E R
are low, reflecting, in part, the low levels in the blood. The :pleural fluid contains increased protein (greater than 3 g/dO, increased white blood cells (2500 to 5000/mm3 with mononuclear cells predominating), glucose levels near those of serum, and decreased C3 and C4. LE cells may be present in the smear.
GENERAL ASSESSMENT The challenge of assessment of disease activity has resulted in the development of a number of disease activity scores. Those most commonly used are the Lupus Activity Index (LAI) (Table 16-35),422 the SLE Disease ActiVity Index (SLEDAI) (Table 16-36),423 the Systemic Lupus Activity Measure (SLAM) (Table 16-37),424 and the British Isles Lupus Assessment Group (BlLAG) Activity Index (Table 16-38).425 In a recent comparison, the LA! was found to be most reliable, although all four had high
~~ I
I ABL E 16-36
16 S Y S T E M I C L u PUS E RY THE MAT 0
SUS
373
validity.426 SLEDAI has also been evaluated by Hawker and coworkers. 427 In one small study of 11 children and adolescents with SLE, the LA! and SLEDAI compared closely to the physician's global assessment of disease activity.428 Brunner and colleagues429 found SLEDAI, BlLAG, and SLAM to be equally sensitive to clinical change in 35 children with SLE. A "flare" of disease activity was defined by Sibley and colleagues430 as the exacerbation or development of new signs and symptoms that in the opinion of the attending physician required a change in therapy. Petri and coworkers 422 defined a flare as a change in physician's global assessment of 1 or more on a 3-point scale. The Systemic Lupus International Collaborating Clinics (SLICC) Damage Index for SLE provides a measure of accumulated organ damage, whether it results from the disease or its treatment, and has been used in children431.432 (Table 16-39). Instruments to measure the
Systemi< lupus Erythematosus Activity Index (SlEDAI)
Desalptor
Definition
8 8
Seizure Psychosis
8
Organic brain syndrome
8
Visual disturbance
8 8
Cranial nerve disorder Lupus headache
8 8
CVA Vasculitis
4
Arthritis
4
Myositis
4 4 4 2 2 2 2 2
Urinary casts Hematuria Proteinuria Pyuria New rash Alopecia Mucosal ulcers Pleurisy Pericarditis
2 2 1 1 1
Low complement Increased DNA binding Fever Thrombocytopenia Leukopenia
Recent onset. Exclude metabolic, infectious, or drug causes. Altered ability to function in normal activity due to severe disturbance in the perception of reality. Include hallucinations; incoherence; marked loose associations; impoverished thought content; marked illogical thinking; bizarre, disorganized, or catatonic behavior. Exclude uremic and drug causes. Altered mental function with impaired orientation, memory, or other intellectual function, with rapid onset and fluctuating clinical feaUlres. Include clouding of consciousness with reduced capacity to focus, and inabiliry to sustain attention to environment, plus at least two of the following: perceptual disturbance, incoherent speech, insomnia or daytime drowsiness, or increased or decreased psychomotor activity. Exclude metabolic, infectiOUS, or drug causes. Retinal changes of SLE. Include cytoid bodies, retinal hemorrhages, serous exudate or hemorrhages in the choroid, or optic neuritis. Exclude hypertensive, infectious, or drug causes. New onset of sensory or motor neuropathy involving cranial nerves. Severe, persistent headache; may be migrainous, but must be nonresponsive to narcotic analgesia. New onset of one or more CVAs. Exclude arteriosclerosis. Ulceration, gangrene, tender finger nodules, periungual infarction, splinter hemorrhages, or biopsy or angiographic proof of vasculitis. More than two joints with pain and signs of inflammation (e.g., tenderness. swelling, effusion). Proximal muscle aching/weakness, associated with elevated creatine kinase/aldolase or electromyographic changes or a biopsy showing myositis. Heme-granular or RBC casts. >5 RBCs/hpf. Exclude stone, infection, or other cause. >0.5 g/24 hr. New onset or recent increase of more than 0.5 g/24 hr. >5 WBCs/hpf. Exclude infection. New onset or recurrence of inflammatory-type rash. New onset or recurrence of abnormal, patchy, or diffuse loss of hair. New onset or recurrence of oral or nasal ulcerations. Pleuritic chest pain with pleural rub or effusion, or pleural thickening. Pericardial pain with at least one of the follOWing: rub, effusion, or electrocardiographic or echocardiographic confirmation. Decrease in CH so ' C3, or C4 below the lower limit of normal for testing laboratory. >25% binding by Farr assay or above normal range for testing laboratory. >38'C. Exclude infectious cause. <100,000 plts/mm3 . <3000 WBCs/mm3• Exclude drug causes.
Score
4
CH". total hemolytic complement; CVA. cerehrovascular accident; hpf, high-power field; pits. platelets; RBC, red blood cell; SLE. systemic lupus erythematosus; WEC. white blood cell. Scor~ if descriptor has been present in preceding 10 days or at time of evaluation. From Bombardier C. Gladman DD, Urowitz M. et al: Derivation of the SLEDAI. A disease activity index for lupus patients. Arthritis Rheum 35: 630. Copyright © 1992 Johlj Wiley & Sons, Inc. Reprinted with permission of Wiley-Uss, Inc.• a subsidiary of John Wiley & Sons, Inc.
374
I!: i
C HAP T E R
TABLE 16 17
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
Systemic lupus Adivity Medsure (SLAM)'
Category Constitutional Weight loss Fatigue Fever Mucocutaneous Malar rash or photosensitive rash, or periungual erythema or nail fold infarct Alopecia Discoid, bullous, or lupus profundus Cutaneous vasculitis Ocular Cytoid bodies Hemorrhages or episcleritis Papillitis or pseudotumor cerebri Visceral Diffuse lymphadenopathy Large liver or spleen Pleural effusion Pneumonitis Raynaud's phenomenon Hypertension Carditis Abdominal pain Neuromuscular Stroke Seizure Cortical dysfunction Headache Myalgia/myositis Synovitis/tenosynovitis Laboratory Anemia Leukopenia Lymphopenia Thrombocytopenia Elevated erythrocyte sedimentation rate Elevated serum creatinine Abnormal urine sediment
Mild
Moderate
Severe 3 3 3
2 2
2
,~.
TABLE 16 38
Systems Nonspecific Mucocutaneous Central nervous system Renal Musculoskeletal Cardiovascular and respiratory Vasculitis Hematologic
Grading System
3 3
A (9 points) B (4 points)
3 3
3 1
1 1 1 1 1 1 1
2 2 2 2
3 3
2 2 2
3 3 3
2 2
3 3 3 3 3
2 2 2 2 2 2
2 2
2 2 2
British Isles lupus Assessmenl Group (BILAG)
3
3 3 3 3
3 3 3
'Maximum score is 86. Score if abnormality was present anytime in the previoUS month. For definitions of grade of severity refer to Liang et al. Modified from Liang MH, Sacher SA, Larson MG, Schur PH: Reliability and validity of six systems for the clinical assessment of disease actiVity in systemic lupus erythematosus. Arthritis Rheum 32: 1107, © 1989 Wiley-Liss, Inc. Reprinted by pennission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
quality of life in patients with SLE have also been developed and evaluated. 433 These indices have not yet been thoroughly evaluated in children and adolescents.
TREATMENT SLE is a chronic disease, characterized by remissions and relapses, and it is associated with considerable morbidity and significant mortality. Treatment is in itself associated with morbidity, the effects of which may be permanent. The patient and family must cope with a chronic, unpredictable disease that ranges widely in severity. For the physician and other health care proViders, the challenge is not only to proVide the most appropriate pharmacologic care but also to ensure the
C (l point) D (0 points)
Intention to treat disease process (not just symptoms) Potential problem, mild, or reversible symptoms Stable disease System unaffected
ScorIng A or B in any system denotes active disease; possible scores range from 0 to 72 Score based on patient's status in previous month. Adapted from Symmons DPM. Coppock JS, Bacon PA, et al: Development and assessment of a computerized index of clinical disease actiVity in systemic lupus erythematosus. Q J Med 69: 927, 1988. By permission of Oxford University Press.
health of the child in the broadest sense and to help the child and family cope with the effects of the disease and its treatment. A brief summary of management is presented in Table 16-40.
General Measures The importance of general supportive care cannot be underestimated. Incorporation of the child and parents into the planning of the overall treatment program is best accomplished by informing them in broad terms of the character and treatment of the disease. This is best achieved by a team of health care providers who are experienced in the management of this multisystem disease in children and adolescents. The team should include a pediatric rheumatologist, nurse, social worker, and psychologist. It is the authors' practice to seek the consultation of a nephrologist early in the disease course, to provide optimal surveillance of the renal complications of the disease. Dermatologists, nutritionists, and other subspecialists may become involved as needed. Most children benefit from the continuity proVided by care by the same team over the long course of their disease. Planning for transition to an adult care environment should begin in the teenage years. Although SLE is a serious problem, the steady improvement in long-term outcome justifies a reasonably optimistic approach to the patient and family and energetic treatment of each exacerbation or complication. Careful attention to all of the details of managing SLE has contributed as much to the improved prognosis as any single therapeutic program or drug. As with other illnesses in childhood, rigorous restraints on general activity are usually unnecessary and undesir-
C HAP T E R
11'
_I
(ABLE 16- 39 Ddnldge Indl'x
Systemic LUllOS International Clinics (SL ICC)
Item
Score
Ocular Any cataract ever RetiM,al change or optic atrophy
1 1
Neuiopsychlatrl, Cognitive impairment SeizUres requiring therapy for 6 mo Cerebrovascular accident (CVA) Cranial or peripheral neuropathy (exclude optic) Transverse myelitis
1 1 1 (score 2 if >1 CVA) 1 1
R..a Glomerular filtration rate <50% Proleinuria >3.5 g/24 hr Or End-stage renal failure regardless of ,dialysis or transplantation
1 1
1
ra~lographic)
Shrinking lung (radiographic) Plevral fibrosis (radiographic) Pulmonary infarction (radiographic)
"~.
SYSTEMIC
Lupus
ERYTHEMATOSUS
375
TABLE: 16-39 Systl'mic lupus (nlernati()fldl Clinics (SIICC) Damage Index---col1t'd
Skin Scarring chronic alopecia Extensive scaring or panniculum other than scalp and pulp space Skin ulceration (excluding thrombosis) for >6 mo Premature gonadal failure Diabetes (regardless of treatment) Malignancy (excluding dysplasia)
1 1
1
1 (score 2 if >1 site)
P2, pulmonic second sound; RV, right ventricular. Damage: (non-reversible change, not related to active inflammation) occuring since onset of SLE, ascertained by clinical assessment and present for 6 months unless othetwise stated. Repeat episodes must occur at least 6 months apart to score 2. From Gladman DO, Ginzler E, Goldsmith C, et al: The development and initial validation of the Systemic Lupus International Collaborating Clinics/American College of Rheumatology Damage Index for systemic lupus erythematosus. ArthritLq Rheum 39: 363-369, 1996.
3
Pul"""''' Puhnonary hypertension (RV prfdominance or loud P2) Pulmonary fibrosis (physical and
16
1 1 1
able, not to mention often impossible. Except during periods of severe active disease, regular school attendance should be expected, and communication with and education of concerned teachers and physical education instructors helps ensure the child's optimal participation in school activities. The child should be encouraged to participate in compatible extracurricular activities as the disease permits.
~Iar
Angina or coronary artery bypass MY(lCardial infarction (M!) ever Cardiomyopathy (ventricular dysfunction) Valvular disease (murmur >3/6) Pericarditis for 6 mo or pericardiectomy
1 1 (score 2 if >1 MI) 1 1 1
PerIpheral VlllCUIar disease Cla~dication for 6 mo Minor tissue loss (pulp space) SigjJ:ificant tissue loss ever (loss of digit or limb) Venous thrombosis with swel1ing, ulcer, or stasis
1 1 1 (score 2 if >1 site) 1
Continued next column.
General Counseling, education, team approach Adequate rest, appropriate nutrition Use of sunscreen Immunizations, especial1y antipneumococcal vaccine Prompt management of infection
Nonsteroidal Anti-Inflammatory Drugs
Anticoagulation 1 (score 2 if >1 site) 1 1 1
M~I
Ml.1scle atrophy or weakness Deforming or erosive arthritis induding reducible deformities Osteoporosis with fracture or vertebral col1apse Avascular necrosis Osteomyelitis
TABLE 16-40 Approach to Malldgl'nll'nt 01 Systl'lllic lupus Erythematosus
For musculoskeletal signs and symptoms
~nal
Infarction or resection of bowel below duodenum, spleen, liver, or g"Ubladder ever for any cause Mesenteric insufficiency Chtonic peritonitis Stricture or upper gastrointestinal trllct surgery ever
I!.
1
If anticardiolipin antibodies are present in significant titers: Low-dose aspirin unless thrombosis has occurred Heparin, followed by warfarin if thrombosis has occurred
Hydroxychloroqulne For cutaneous disease and as an adjunct to glucocorticoids for systemic disease
Glucocortlcolds
1
Oral prednisone 1-2 mg/kg/day IV methylprednisolone initially, and at monthly intervals for maintenance therapy in severe disease
1 1 (score 2 if >1 site) 1
Immunosuppresslves Azathioprine 1-2 mg/kg/day (PO) Cyclophosphamide 1-2 mg/kg/day (PO), or 500-1000 mg/m'/mo (IV) in severe disease
376
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
Although there are no foods that have been demonstrated to be either helpful or detrimental to the patient with SLE, a well-balanced diet with appropriate caloric intake is important. Because of the probability of unwanted weight gain in the child with SLE who is being treated with glucocorticoids, early nutritional counseling should be provided. Avoidance of "junk food" and foods high in sodium helps minimize excessive weight gain. Education of patient, family, and teachers about the effects of treatment on body image assists the child and adolescent adapt to such changes. Hypertension must be carefully controlled. Certain general aspects of treatment are of primary importance. The dangers of exposure to excessive sunlight should be stressed. Sunscreens with sun protection factors (SPF) greater than 15 that protect against UVB light169 should be applied to all exposed skin whenever the child is outside, whether or not it is sunny. (Many experts recommend an SPF of at least 30.) Even exposure to fluorescent lights that emit DYB may be associated with symptoms. 434 A variety of suitable sunscreens is available; some have a water base and are most appropriate for use on the face, and others contain alcohol and may be used on other exposed skin. Some are water resistant, but reapplication after bathing or swimming is adVisable. The risk of infection is increased in SLE, and the sequelae of infection may be catastrophic. Functional asplenia328.329 makes the child with SLE extremely susceptible to severe, sometimes fatal pneumococcal sepsis. Prevention of infection by immunization is particularly important in these children, and the recommendations in this regard outlined in Chapter 8 should be followed. Other factors that predispose to infection include neutropenia and lymphopenia, immunosuppressive treatment, hypocomplementemia, and the nephrotiC syndrome. A high index of suspicion for infections facilitates prompt diagnosis and appropriate treatment. Antibiotics should not be used prophylactically and should not be given before appropriate cultures have been obtained. Fever, which may accompany active SLE, should always be considered first to be caused by infection, and cultures of blood, pharynx, and urine should be taken. Basilar pneumonitis should always be initially regarded as bacterial, and blood cultures as well as cultures and a Gram stain of sputum should be obtained. Acute CNS disease should also be regarded initially as an effect of infection, and cerebrospinal fluid examinations should be performed. Exposure to varicella in the immunosuppressed child requires administration of zoster immune globulin within the first 96 hours after exposure. Varicella should be treated with acyclovir in the immunosuppressed child or adolescent. Laboratory markers of infection may be somewhat blunted by the primary disease. In a child with SLE, elevation of a previously low white blood cell count into the normal range may represent the maximal response to infection. Marked elevations of serum levels of CRP are strongly suggestive of infection in a patient with SLE. The many complications of tuberculosis should be considered in the differential diagnosis.
CNS infection with organisms such as Cryptococcus may occur.
General Aspects of Pharmacologic Therapy SpeCific treatment should be individualized and based on the extent and severity of the disease (see Table 16-40). Pharmacologic management of SLE in children was recently reviewed by Carreno and associates. 435
Nonsteroidal Anti-inflammatory Drugs The primary role of nonsteroidal anti-inflammatory drugs (NSAIDs) in the management of pediatric SLE is to treat musculoskeletal complaints.436 Myalgia, arthralgia, or arthritis may respond well to anti-inflammatory doses. Low-dose aspirin is indicated in the child with significant titers of aPL. Occasional reports have linked the use of ibuprofen436-438 or naproxen438 to the occurrence of aseptic meningitis or other hypersensitivity reactions in patients with SLE. There are no studies of the use of selective cyclooxygenase-2 inhibitors in SLE.
Hydroxychloroquine Hydroxychloroquine is most often given as an adjunct to therapy with glucocorticoids, rather than as the mainstay of treatment. 439 The role of this medication has been changing since the studies of the Canadian Hydroxychloroquine Study Group 440.441 reported that this drug made an important contribution to control of the disease. In a randomized, double-blind, placebocontrolled study of the effect of withdrawing hydroxychloroquine therapy in adults with clinically stable SLE, the frequency and severity of flares of SLE activity were much higher in patients who received the placebo than in those maintained on hydroxychloroquine. In addition, hydroxychloroquine may also have a role in reversing glucocorticoid-induced changes in plasma lipids. 442--444 For these reasons, it is the authors' practice to treat children and adolescents with hydroxychloroquine (up to 6 mg/kg/day) at the initiation of steroid therapy and for up to 2 years or longer. 445 Because of the possibility that the risk of retinal toxicity from antimalarials may be increased in the presence of impaired renal function, children with SLE should be carefully monitored for retinal toxicity related to the drug (see Chapter 5). Children with SLE may be more prone than those with ]IA to develop retinal damage.
Glucocorticoids Without doubt, glucocorticoids constitute the mainstay of pharmacologic therapy, and almost all children with SLE require oral prednisone or prednisolone or intravenous methylprednisolone at some stage of the disease. These agents have wide application in the treatment of many of the manifestations of SLE. 446 The choice between prednisone and prednisolone is one of personal experience and preference; prednisone is most frequently used in North America, and prednisolone is
C HAP T E R
often favored in Europe. High-dose intravenous methylprednisolone (30 mg/kg/day to a maximum of 1 g/day on 1 to 3 consecutive days) is often indicated in children with severe acute disease such as acute hemolytic anemia, CNS disease, or overwhelming systemic disease ("lupus crisis"), or as part of the long-term management of l\lPUS nephritis.
16
I! II
SYSTEMIC
Lupus
TABL E 16-42
Approa(h to Therapy for Lupus Nephritis'
II
'--
IABLE 16-41 GllI(ocorti(oid Therapy for Children with Systeuu( Lupus Erythematosus
In'1tallon of Thet'lllpy (lint 4-6 wk)
Orj1I prednisone
IV methylprednisolone
1'Hi0 mg/day (0.5-2 mg/kg/day) in at least two divided doses (depending on severity and type of organ involvement) Indicated for severe disease (active lupus nephritis, hematologic crisis, CNS disease)30 mg/kg/day on 1 to 5 consecutive days
hperlng the Prednisone Dose If the dose is 20-60 mg/day If the dose is 10-20 mg/day If nhe dose is <10 mg/day
eNS, central nervous system.
Decrease by 2.5-5.0 mg/wk Decrease by 1-2.5 mg every 2-4wk Decrease by 0.5-1 mg every 2-4 wk
377
Type of NephriUs
Thet'lllpy
Mesangial nephritis Focal nephritis
Symptomatic management Prednisone 0.5 mg/kg/day for 2-4 mo, then taper to maintenance level Prednisone 1 mg/kg/day for 3-6 mo, then taper to maintenance level Cyclophosphamide 1-2 mg/kg/day, or IV pulse every month for 6 mo, possibly longer Prednisone 0.5-1 mg/kg/day, with possible addition of cyclophosphamide or azathioprine
Diffuse nephritis
Initial Glucocorticoid Therapy Prednisone or prednisolone should be given in a dose sufficient to achieve disease control, often 1 to 2 mg/kg/day (Table 16-41). At least at the initiation of therapy, oral glucocorticoids should be given in two or three divided doses to maximize the anti-inflammatory and immunosuppressive effects. The initial dose of glucocorticoid and the frequency of its administration depend on the severity of the disease and the organ systems affected. The potential benefits of sustained glucocorticoid administration must be bal~nced against the serious complications that result from this treatment (see Chapter 5), Low-dose therapy (less than 0.5 mg/kg/day of prednisone), administered in divided doses by mouth, is usually sufficient to control fever, dermatitis, arthritis, and serositis and to increase the sense of well-being and energy within hours to days after starting treatment. Although low-dose glucocorticoids may be sufficient to control hemolytic anemia or thrombocytopenia and reverse abnormalities of the ESR, anti-dsDNA antibodies, and complement levels, achievement of these effects may require several weeks to a few months. High-dose oral prednisone (1 to 2 mg/kg/day in divided doses) is employed for treatment of acute hemolytic anemia, CNS disease, parenchymal pulmonary disease, and the more severe types of lupus nephritis (Table 16-42). In addition, intravenous methylprednisolone (30 mg/kg/day on one or more consecutive days) may be necessary to manage these aspects of the disease, particularly if a rapid response is needed.
ERYTHEMATOSUS
Membranous nephritis
'In all patients, manage hypertension and nephrotic syndrome.
Hypertension, uremia, and preexisting psychosis are relative contraindications to high steroid doses.
Tapering the Glucocorticoid Dose After the acute manifestations of the disease are controlled, glucocorticoids should be reduced to the lowest possible level that will maintain the well-being of the child. Tapering from high dose to minimum dose is one of the most challenging aspects of management of SLE in children. In a justified determination to minimize glucocorticoid toxicity, the physician may be tempted to make an injudiciously rapid dose reduction; too often, the result is a flare of the disease, with a subsequent requirement to increase the dosage by 25% to 50% or more. A smooth, gradual tapering of drug dose, with frequent monitoring to make certain that clinical and laboratory measures of disease activity are suppressed, is much more satisfactory. In Table 16-41, average dosage schedules are suggested as guidelines, but these must be individualized. Ordinarily, the initial glucocorticoid dose is maintained for a period of 3 to 4 weeks. Thereafter, a weekly or biweekly decrease in the daily prednisone dose of approximately 5 mg is appropriate when the child is taking 20 to 60 mg/day. The average reduction every 2 to 4 weeks at daily doses of 10 to 20 mg should be approximately 2.5 mg/day. At doses lower than 10 mg/day, a biweekly or monthly reduction of the daily dose of not more than 1 mg is appropriate. Consolidation of prednisone administration into a single daily dose, preferably given in the morning, may be successful at levels of 10 mg/day or lower. Alternate-day therapy may risk inadequate disease suppression: despite its obvious advantages with respect to side effects, it has a limited role in the management of acute SLE, although it may be used in children who require low doses (less than 10 mg/day) to maintain disease control. No studies in children have been performed comparing these treatment guidelines, and the recommendations in the preceding paragraph are based on clinical experience. It is generally agreed that most children with SLE are withdrawn completely from glucocorticoid drugs only with great difficulty during the initial years after diagnosis. Even long-term, very low dose maintenance
378
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
therapy can minimize the tendency toward exacerbations during the protracted course of this disease.
are frequently such, however, that institution of another agent is indicated, even if the disease appears to be well controlled.
Maintenance Glucocorticoid Therapy Clinical response, the white blood cell and platelet counts, hemoglobin concentration, serum complement, and levels of anti-dsDNA antibodies, together with urinalysis and indices of renal function, are periodically assessed to determine the adequacy of treatment. As long as these parameters are improving, treatment should be maintained. It is the authors' practice not to alter the regimen of glucocorticoid administration, if at all possible, until the serum C3, C4, or CH50 are close to normal, there is no clinically significant hematocytopenia, and the levels of anti-dsDNA have fallen substantially (Fig. 16-34). In many children with proliferative nephritis, treatment with adequate doses of glucocorticoid leads to a return of the microscopic urinalysis findings toward normal, although proteinuria and a depressed creatinine clearance are less responsive to therapy. Failure of highdose prednisone (2 mg/kg/day in divided doses) to result in substantial improvement in clinical, serologic, and urinary abnormalities within 8 weeks should prompt consideration of additional therapies. The decision to initiate therapy with a cytotoxic drug such as azathioprine or cyclophosphamide is usually influenced by a number of factors, including steroid responsiveness, steroid dependency, and steroid toxicity. In a patient who is steroid responsive and dependent at an acceptably low prednisone dose, the disease may be best managed without cytotoxic drugs. Side effects of glucocorticoids
:~
Immunosuppressive Agents Immunosuppressive agents are often required to control SLE and permit an acceptable quality of life. A metaanalysis of eight studies in adults with SLE that compared the effect of prednisone alone with that of prednisone plus azathioprine, or prednisone plus cyclophosphamide, demonstrated that an immunosuppressive agent given with prednisone was superior to prednisone alone. 447 Depending to a large extent on the severity of the disease and specific organ involvement, a choice of immunosuppressive agent is made among azathioprine, cyclophosphamide, methotrexate, cyclosporine, and mycophenolate mofetil (Table 16-43).
Azathioprine Azathioprine has probably been used longer to treat childhood lupus than any other second-line agent. However, its role is still being defined. 448 It has traditionally been used as the initial second-line agent, largely because of its perceived safety. For children with SLE not controlled by acceptable doses of prednisone, addition of azathioprine in a dose of 1.5-2.5 mg/kg/day may be indicated to permit a reduction in steroid dose. Experience in 66 pediatric patients with SLE, 19 of whom were treated with azathioprine, indicated that early treatment with prednisone and azathioprine was effective in controlling disease and preserving renal function. 449 It is probable that the appropriate role of azathioprine in childhood SLE is in the management of steroid-resistant or steroid-dependent disease with or without class III or IV nephritiS. Some authorities recommend the use of azathioprine together with corticosteroids in the first month after diagnosis of lupus nephritis. 450 .451 Most pediatric rheumatologists would agree that azathioprine has a role in the management of nonrenal SLE; controversy surrounds its place in the management of lupus complicated by moderate or severe nephritis. 452
E -------- ---I 1--I
80
I
I-J
1_, I
I!:..
I I
40
I I I
4
6
8
10
12
2
4
6
8
10
12
2
• Figure 16-34 The course of a young girl with acute systemic lupus erythematosus and diffuse proliferative glomerulonephritis is charted in relation to urinary protein values measured in g/24 hours (.4); serum hemolytic complement units (8); creatinine dearanc.e in Iiters/24 hours (C); and anti-DNA antibody expressed as DNA binding in percent (D)The doses of cydophosphamide (E) and prednisone (F) are Indicated in mg/day. Gradual resolution of systemic disease was accompanied by a fall in the DNA binding level to a normal value of less than 20 percent and a rise in the serum hemolytic complement level to a normal value above 110 CH so units. Resolution of the acute phase of renal disease was accompanied initially by an increase in urinary protein excretion and adecrease in creatinine dearance. Afall In daily protein excretion and a gradual rise of the creatinine dearance to relatively normal values followed. Initial deterioration in the creatinine dearance rate with glucocorticoid therapy is occasionally observed.
TABLE 16-/13 Approadl to the Use of Immunosuppressive Drugs in Systellli( lupus Erythematosus
Drug and Dosage Azathioprine 1-2 mg/kg/day (PO)
Cyclophosphamide 1-2 mg/kg/day (PO) 500-1000 mg/m1/mo (IV)
eNS. central nervous system.
Indication For patients who are unresponsive to glucocorticoids and hydroxychloroquine or who develop unacceptable toxicity to these drugs For patients with diffuse proliferative glomerulonephritis or significant CNS disease For patients with life-threatening manifestations or resistant CNS or renal disease
C HAP T E R
Cycl()phosphamide
16 S Y S T E M I C Lu PUS E RY THE MAT 0
SUS
379
that, although there was initial improvement, methotrexate did not make a major contribution to steroid requirement or disease control. Silverman449 suggested that methotrexate may have a role in the management of resistant arthritis and skin disease in childhood lupus. The use of intrathecal dexamethasone and methotrexate has been suggested in the treatment of CNS lupus,467 and intravenous methotrexate combined with intravenous cyclophosphamide effectively treated refractory lupus nephritis in children after cyclophosphamide alone had failed to control the disease. 468
Cyclophosphamide is an important drug for management of the most severe aspects of SLE, particularly severe lupus nephritis and CNS disease. 453 Because of its potential toxicity, however, cyclophosphamide is not indicated for minor manifestations of disease and should be used only by experienced clinicians who are familiar with the anticipated benefits and toxicity of this drug. In conjunction with glucocorticoids, it has beeh demonstrated to be superior to prednisone alone, especially in severe nephritis. 454 There have been no controlled trials in children, but in extensive comparative studies455 ,456 of oral prednisone, or prednisone plus either azathioprine, oral cyclophosphamide, intravenous cyclophosphamide, or intravenous cyclophosphamide plus azathioprine, renal function was better in the group receiving oral or intravenous cyclophosphamide than in those receiving azathioprine or prednisone alone. These differences in outcome did not become apparent until 5 to 10 years after treatment was started. Mortality, however, was not impressively differentamong the treatment groupS.457 Intravenous pulse cyclophosphamide, used in combination with oral prednisone, is an effective regimen for the prevention of disease flares and the preservation of renal function in severe disease, and it is less toxic than daily oral cyclophosphamide. A regimen of seven monthly pulses of cyclophosphamide, followed by a pulse of cyclophosphamide every 3 months thereafter for a period of 2 years, was found to be optimal.458 This regimen is also recommended for management of lupus in children and adolescents. 459 Lehman and Onel 460 cited a favorable response to 36 months of intravenous cyclophosphamide therapy and noted minimal side effects. The combination of oral prednisone and intravenous pulse cyclophosphamide for the treatment of severe neuropsychiatric lupus in adults and older adolescents was dramatically effective in the majority of those refractory to other therapy, including glucocorticoids and azathioprine. 461 Cyclophosphamide is probably the drug of choice among the second-line agents for severe nephritis or CNS disease. Its short- and long-term toxicity limit its prolonged use, however. For patients who require treatment for longer than 3 years, an alternative should be sought because of the risk of ovarian failure and malignancy. Gonadotropin-releasing hormone (GnRH) and GnRH agonists are currently being investigated for use in preservIng gonadal function in patients who require cyclophosphamide therapy.462
Experience with the use of mycophenolate mofetil (MMF) in childhood SLE is very limited, but recent studies in adults are very promising. 47.'H76 Ginzler and colleagues 475 compared intravenous cyclophosphamide (given according to the National Institutes of Health protocol) with oral MMF (in a dose of 1 g/day increasing to 3 g/day as tolerated) as induction therapy in 140 patients with SLE and active class IV nephritis, or class III or V nephritis with creatinine greater than 1.3 or proteinuria greater than 2 g/24 hours. After 6 months, the outcome in the MMF-treated group was significantly better with respect to remissions, partial remissions, serious infections, and mortality. This short-term study suggested that MMF may have a major role in the management of SLE, but studies in children are, at present, anecdotal. The toxicity of this agent, at least in the short term, appears to be less than that of cyclophosphamide.
Methotrexate
Biological Modulation
Experience with methotrexate to treat childhood SLE is limiued,46.'H65 although it is quite extensive in adults. 466 In a study of 10 children, the addition of 2.5 to 10 mg of methotrexate per week reduced the requirement for glucocorticoid and permitted cessation of cyclophosphamide in the majority of patients. 464 In contrast, Ravelli andcolleagues465 treated 11 children with methotrexate 025-17.0 mg/m2/week) and prednisone and concluded
Intravenous immunoglobulin (IVIG) has been used to a very limited extent in adults with refractory SLE,4n-482 including those with thrombocytopenia, in whom it evoked a mild but transient improvement in platelet counts. 479 .480 In one small study, IVIG resulted in modest temporary improvement in a measure of disease activity and a decline in the titer of anti-dsDNA antibodies. 483 Lafferty and colleagues483 reported that IVIG was suc-
Cyclosporine An early study showed that cyclosporine in an average daily dose of 5 mg/kg permitted reduction in prednisone dose and better disease control in 8 of 13 children with severe glucocorticoid-dependent or glucocorticoidresistant SLE in childhood. 469 In an open study of 40 children with class III or IV lupus nephritis and heavy proteinuria, cyclosporine (5 mg/kg/day) was compared with prednisone (2 mg/kg/day) plus cyclophosphamide (2 mg/kg/day). Cyclosporine was as effective as prednisone plus cyclophosphamide in reducing proteinuria, allowed significantly greater growth, and was well tolerated. 470 The researchers suggested that cyclosporine could have an important steroid-sparing effect, as reported in adult lupus patients. 471 Because cyclosporine can be nephrotoxic, it may be difficult to evaluate in the patient with nephritis. Its use in treatment of lupus was summarized by Griffiths and Emery. 472
Mycophenolate Mofetil
380
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
cessful in treatment of the acquired factor VIII inhibitor that had resulted in bleeding. Experience in the use of IVIG to treat manifestations of childhood lupus is largely unreported, and its role should be considered to be very limited at present. Plasmapheresis may occasionally be of benefit in the child with a severe acute complication such as TTP,302 but it has not been shown to be beneficial in the treatment of lupus nephritis. From a review of more than 100 case reports or open trials, Wallace 484 concluded that plasmapheresis was most effective if the patient had high levels of circulating immune complexes and was receiving cyclophosphamide, and that it was ineffective if the patient was taking corticosteroids alone. Whether synchronized plasmapheresis and cyclophosphamide therapy has any significant role in disease management is uncertain. 484,485 Monoclonal antibodies have been used in SLE. In an open trial of murine antibody to IL-lO, given daily for 21 days with a 6-month follow-up period, disease control was achieved in five of six patients, and prednisone dose was significantly reduced. 486 Anti-CD20 (rituximab) has been reported to be of benefit in a few patients. 487--490 The potential use of monoclonal antibodies in SLE was reviewed by Strand. 491
Other Approaches Experimental therapies include thalidomide 492 ,493 and extracorporeal photochemotherapy.494 None has been evaluated in children. Autologous bone marrow transplantation (ABMT) has been performed in several adolescents with SLE with good short-term effect, but the long-term safety and effectiveness of such therapy is not yet known. 49 5--497 In the European ABMT data base, 14 of 23 patients with SLE were described as having improved. 498 A selective B cell modulator, LJP 394, significantly reduced the levels of anti-dsDNA antibodies in patients with SLE, but its effect on disease course is not known. 499
MANAGEMENT OF SPECIFIC ASPECTS OF THE DISEASE Acute Hemolytic Anemia Acute hemolytic anemia can be a medical emergency requiring the use of high-dose intravenous methylprednisolone (30 mg/kg/day on one or more consecutive days). Some children have no response even to high-dose glucocorticoid therapy. Splenectomy may be necessary in such children, although this procedure may be less efficacious than in idiopathic thrombocytopenia. For the usual reasons, blood transfusions should be avoided in these patients if not absolutely necessary; in addition, they may be at added risk because of the high frequency of anti-erythrocyte and anti-leukocyte antibodies, and because of the occurrence of anti-IgA antibodies if SLE is associated with selective IgA deficiency.
Antiphosphollpld Syndrome The optimal management of patients with the APLS is controversial and depends on the presence of laboratory abnormalities that demonstrate aPL or their effects on coagulation and the history of venous or arterial thrombotic events. 500 In patients who have an aPL and a platelet count greater than 70,000/mm3, C7 x 109/0, lowdose aspirin (3 mg/kg/day) is recommended to reduce the risk of thrombosis. In patients who have high levels of antibody to cardiolipin, or in whom venous or arterial thrombosis has already occurred, anticoagulation with heparin followed by warfarin is recommended. SOl Studies in adults showed that an INR greater than 3.0 was effective in decreasing the likelihood of thrombosis without increasing the risk of serious bleeding. 502 Patients who are receiving long-term anticoagulation should be under the care of a hematologist.
Central Nervous System Disease CNS manifestations of SLE usually require high-dose oral or intravenous glucocorticoids once the possibility of infection has been excluded. Thirty-one adults and adolescents with progressing neuropsychiatric SLE as demonstrated by serial imaging studies were treated with intravenous cyclophosphamide (500 mg/m 2). All had received glucocorticoids, and one third had received azathioprine or chlorambucil. Improvement was noted in 61%, and a further 29% stabilized after intravenous cyclophosphamide was given. 461 The use of both glucocorticoids and cyclophosphamide in treating CNS lupus in children was supported by another, smaller study. so.> Initial therapy in this group of seven children with neuropsychiatric lupus consisted of intravenous methylprednisolone and intravenous cyclophosphamide, followed by intravenous cyclophosphamide every month for at least 3 months, and oral prednisone in a dose of 1 to 2 mg/kg/day. Significant improvement was noted within 1 week after institution of therapy, and all patients recovered except for a residual neurologic deficit in one patient.
Lupus Nephritis Lupus nephritis almost always requires the prolonged use of oral corticosteroid, The dose, route of administration, and duration of treatment are controversial. Mesangial or focal nephritis may respond to low to moderate doses of oral prednisone (0.5 to 1.0 mg/kg/day) given over a period of several months, The efficacy of therapy is judged by a decrease in hematuria, improvement in renal function, and normalization of C3, C4, and CH so ' with a fall in the level of anti-dsDNA antibodies. Proteinuria may persist for a lengthy period and, if it is the only abnormality, should not prevent tapering the prednisone dose. Diffuse proliferative glomerulonephritis usually requires high-dose prednisone (l to 2 mg/kg/day) and, in many cases, the addition of azathioprine (2 mg/kg/day) or cyclophosphamide (2 mg/kg/day). The choice between these drugs is controversial, but, in the authors' experience, cyclophos-
C HAP T E R
phamide is the drug of choice in this situation (unless the renal abnormalities are mild, in which case azathioprine may be effective). Management of hypertension with a drug such as nifedipine or amlodipine is essential. If there is not a prompt response to oral cyclophosphamide and prednisone, the addition of intravenous pulse cyclophosphamide and intravenous methylprednisolone is appropriate. Membranous glomerulonephritis is less likely to be responsive to glucocorticoids than is proliferative disease. 504 The majority of children with SLE do not develop renal insufficiency if they are treated early and maintained in serologic and clinical remission. Some children appear to enter permanent remissions; late in the disease course, gradual deterioration of renal function may again be observed (end-stage renal disease) in the absence of serologic deterioration (Table 16-44). Some children require dialysis and transplantation for management of end-stage renal disease. Gipson and coworkers 505 reviewed the outcome of renal transplantation in 254 children with lupus registered in the Network for Organ Sharing. The median age was 19 years, and 80% were female. In this study, patients with SLE were almost twice as likely to die as transplant recipients with other diseases. A somewhat smaller registry study showed no such difference, however. 506 Patient survival at 3 years was 95%, and graft survival was 71%. Recurrences of the nephritis in the allograft were rare. 507
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
381
The disease may flare at any time, either spontaneously or as a reaction to infection or some other identifiable event. Spontaneous remissions occur. Fever and prominent constitutional symptoms are present at the onset of disease and during exacerbations. With the exception of eNS disease, which can occur at any time, there is a tendency for flares of disease to mimic earlier patterns of organ involvement. Overall outcome has improved as a result of the increased availability of specialized multidisciplinary diagnostic and treatment resources, more complete understanding of the short- and long-term complications to which the patient with lupus is subject, and attention to the details of day-to-day management. 508
Morbidity
SLE is characterized by a prolonged course over many years that is punctuated by exacerbations and remissions.
With the increasing life expectancy of children with SLE, a number of factors that affect their long-term quality of life are assuming increased importance (Table 16-45). A comparison of two cohorts of Taiwanese children with SLE509 noted significantly higher rates of infection, skeletal disease, and end-stage renal disease in the cohort diagnosed between 1980 and 1990, compared with the cohort diagnosed between 1991 and 2001. Recurrent infections contribute significantly to the morbidity of children with SLE.51O In a review of 32 patients with onset of SLE before 16 years of age who were monitored for 10 years 0979-1988),511 infection occurred in almost two thirds, and disseminated infection with Aspergillus, Candida, Nocardia, Clostridium perfringens, or Pseudomonas contributed to all five deaths. It is not certain whether these infections were primarily related to the longer-term course of the disease in surviving patients or to the therapy, but infections were more common in those patients who had received prolonged high-dose glucocorticoid therapy. In contrast to this experience, infection has only occasionally been a serious problem in the authors' clinics. The development of functional asplenia in some children is a contributing factor to infection with Streptococcus pneumoniae. P carinii pneumonitis occurred in one of our patients, and Pseudomonas osteomyelitis and septic arthritis in another. Osteonecrosis occurs in 10% to 12% of patients and probably reflects the effects of disease and prolonged use of corticosteroids. Although it is not associated with higher mortality, avascular necrosis contributes to a diminished function and quality of life. 512
I' _I
I! II
Dysnipoprotelnemla Insufficient attention has been paid to management of the lipid abnormalities of SLE and prevention of their long-term sequelae. Intermittent monitoring of total cholesterol, VLDL cholesterol, and triglycerides should alert the physician to the advisability of dietary, lifestyle, or pharmacologic intervention. The lipid-lowering effects of hydroxychloroquine should be exploited in achieving this aim. Trials of lipid-lowering agents such as the statins are underway.
COURSE OF THE DISEASE AND PROGNOSIS
I ABLE t6 -41j
End-Stage Lupus Nephritis
OIanIderlstks Cliniqllly inactive Normal serum complement, anti-dsDNA Hypertension Intermittent sepsis
Manafement Minimize glucocorticoids Antihypertensives Hem(jldialysis Renal transplantation
TABLE 16 -45
Renal Central nervous system Cardiovascular Immune Musculoskeletal Ocular Endocrine
Morbidity in Childhood Lupus
Hypertension, dialysis, transplantation Organic brain syndrome, seizures, psychosis, neurocognitive dysfunction Atherosclerosis, myocardial infarction, cardiomyopathy, valvular disease Recurrent infection, functional asplenia, malignancy Osteopenia, compression fractures, osteonecrosis Cataracts, glaucoma Diabetes, obesity, growth failure, infertility, fetal wastage
382
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
Atherosclerotic complications of SLE and its treatment are emerging as an important residual problem of the illness, becoming clinically apparent only after a decade or more of active disease. Whether these developments are related primarily to the lupus per se, to prolonged hyperlipoproteinemia, or the long-term complications of glucocorticoid therapy is not clear. Undoubtedly, all of these factors contribute to the final prognostic profile.
Mortality There has been a remarkable improvement in the outcome for patients with SLE during the past three decades (Table 16--46).511-519 Although SLE is a serious, life-threatening disease, an optinlistic approach to the care of these children is now justified by current data. The prognosis for an individual child with SLE, however, is relatively unpredictable. Generalizations about prognosis are especially unreliable during the first 24 months after diagnosis. Estimates can be related to the degree of activity and severity of the systemic disease, the type and progression of nephritis, clini-
IJ:~
TABLE 16-46 Survivdl ill (hildrl'1I wilh Syslemil Lupus Erylhemdlosus % Survival
Study and year (ref. no.)
Patients (n)
5yr
42
42/72-
10 yr
15 yr
Meislin & Rothfield,
1968 (39) Walravens & Chase,
1976 (40)
50 49 108 49
Garin et al., 1976 (362) King et al., 1977 (38) Fish et al., 1977 (513) Abeles et al.,
1980 (514)
60-70
72 78 86
67 70
89/10090
85
55
92
85
71 167* 56\
91
Platt et al., 1982 (511) Glidden et al.,
1983 (516)
77
McCurdy et al.,
1992 (532) Yang, 1994 (519) Baqi et al., 1998 (518)
28\ 29
DRUG-INDUCED LUPUS
'% with renal dL.ease/% with no renal disease.
Serologic or clinical manifestations of lupus develop in some patients taking certain drugs (see Table 16-5), The presence of ANAs is much more common than the pres-
IUfe table analysis. 'All 167 patients had nephritis. 163% had World Health Organization class lIJ or IV renal histology.
I"'~
TABLE Ib- f l7
cally apparent vasculitis, and the multisystenlic character of the disease. The prognosis is poorest in patients with diffuse proliferative nephritis or persistent CNS disease, and it is best in those with mesangial disease or focal nephritis. 363,52o,521 Encephalopathy, especially organic brain syndrome and cerebrovascular accident, have become major factors in survival in some studies. Sepsis has replaced renal failure as the most common cause of death. 509 This trend is illustrated in Table 16--47. Malignant hypertension, gastrointestinal bleeding and perforation, acute pancreatitis, and pulmonary hemorrhage are other terminal events. Infection may occur with common pathogens or with opportunistic organisms such as fungi or protozoa. Death from lupus crisis is rare today, although in a report of 31 children seen in the 1990s in India 522 the most common cause of death was uncontrolled disease activity. In this case series, mortality was 32%. The researchers suggested that late referral was the most important underlying factor. Reports on prognosis before 1968 were based on early experience with glucocorticoids and in general were restricted to children with classic SLE and severe disease. Platt and colleagues511 reviewed 70 children who were observed from 1958 to 1981. Survival was 77% at 15 years (see Table 16-46). Age at onset and the presence of CNS disease did not affect mortality. Prognosis for life and renal function were poorest after 7 years of follow-up in those with diffuse nephritis. African Americans, especially young children in this racial group, and Hispanic Americans have a poorer prognosis, compared with white children (Table 16-48).518523 Geographic variation in lupus mortality in the United States524 probably reflects differences in socioeconomic status (access to health care) and ethnic differences. In the Taiwanese study,'09 survival was significantly better in patients diagnosed after 1991 than in those diagnosed before 1991. The two factors that were poor prognostic factors were female sex and the development of end-stage renal disease. Infection was the leading cause of death.
(duses of Ol'dlh in (hildrl'n wilh Syslemi( lupus Erylhemdlosm
Study and Year (ref. no.) Meislin & Rothfield, 1968 (39) Walravens & Chase, 1976 (40) Garin et aI., 1976 (362) King et al., 1977 (38) Cassidy et al., 1977 (34) Fish et al., 1977 (513) Abeles et al., 1980 (514) Platt et al., 1982 (511) Glidden et al., 1983 (516)
No. DeadITotal 18/42 12/50 6/49 28/108 111587/49 10/67 11/70 9/55
eNS, central nervous system.
'In some cases, death was attributed to more than one cause.
Infection
Renal
CNS
Cardiac
Other
7 1 2 8 8 6 1 9 3
6 4 2 12 7 0 3 2 3
2 0 2 0 1 0 0 0 3
0 5 0 0 3 0 0 1 0
:I
3 0 0 1 0 6 2
1
C HAP T E R
CII
rABLE
16-48
Mortality in Childhood Systemic Lupus [rythematosus (deaths! I 0" person-years)
Age (yr) 5-9 10--14 15-19
White Male
White Female
Black Male
Black Female
0.1 0.3 0.9
0.4 1.5 3.8
0.2 0.8 1.8
0.6 4.4 12.9
Adapted from Kaslow RA, Masi AT: Age, sex, and race effects on mortality from .wstemic lupus erythematosus in the United States. Arthritis Rheum 21: 473, © 197t; Wiley-Liss, Inc. Reprinted by permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
383
early and judicious use of glucocorticoids, hydroxychloroquine, and second-line agents such as azathioprine and cyclophosphamide. New agents such as MMF and the biologics offer new hope that morbidity and mortality can be further reduced. Nonetheless, the disease often remains active throughout adolescence and into adulthood. The transition of the medical care of these young adults from the pediatric to the medical setting is an important part of their ongoing management. A great deal is yet to be learned from the long-term follow-up of patients with childhood onset of SLE.
REFERENCES ence of any clinical abnormality related to SLE in patients who take medications implicated in autoimmunity. Cutaneous and pleuropericardial disease are the most prominent clinical manifestations; CNS disease and lupus nephritis are distinctly uncommon. ANAs usually react with histones,4I3,)25 and anti-dsDNA antibody is not usually present. Semm complement levels remain normal. The anticonvulsant drugs are the most common cause of dmg-induced SLE in childhood. 526 .527 Diphenylhydantoin, mephenytoin, trimethadione, and ethosuximide have been most often incriminated. Singsen and associates 527 described five children who received ethosuximide and subsequently developed SLE with fever, malar rash, arthritis, and ANA seropositivity. Pleuropericardial effusions were less commonly observed, Clinical renal disease occurred in seven children and resulted in the deaths of three. Serum ANA titers did not differ significantly from results in 101 asymptomatic patients with seizures. Even after the drug was discontinued, 70% of these children remained ANA positive. It was concluded that asymptomatic children who develop ANA positivity while receiving anticonvulsant drugs should be carefully monitored but need not have their medications discontinued. Patients who develop an SLE-like illness while receiving hydralazine have usually been treated with moderate to high doses of the drug. 52H During drug administration, antihydralazine antibodies were present in some patients, and antibodies to dsDNA were observed with this type of drug hypersensitivity. In some children, the SLE-like disease may not abate after the drug is discontinued.
Minocydine529 and zafirlukast530 have occasionally been implicated in a lupus-like syndrome in children. 53! Five adolescent girls treated with minocycline for acne developed fever, fatigue, rashes, arthralgia, arthritis, and hepatitis 6 weeks to 2 years after initial dmg therapy. All had ANAs and anti-histone antibodies but no antidsDNA. In all, the symptoms disappeared between 2 and 4 months after discontinuation of the antibiotic. 529
PERSPECIlVE SLE is a challenging disease to diagnose and manage. Lupus can assume many guises and should be suspected particularly in older girls or adolescents with systemic illness that includes dermatitis, arthritis, and a myriad of other signs. The course is inevitably prolonged, but the outcome has improved dramatically with the availability of appropriately trained health care providers and the
1. Smith CD, Cyr M: The hislOry of lupus erythematosus: from Hippocrates to
Osler. Rheum Dis Clin North Am 14: 1-14, 1988. 2. Kaposi M: Neue Beitrage zur Kenntnisse des Lupus erythematodus. New reports on knowledge of lupus erythematosus. Arch Dermatol Syph 4: 36-78. 1872, 3. Osler W: On the visceral complications of erythema exudativum multiforme. Am J Med 110: 629-646, 1895. 4. Libman E, Sacks B: A hitherto undescribed form of valvular and mural endocarditis. Arch Intern Med 33: 701-737, 1924. 5. Gross L: Cardiac lesions in Libman-Sacks disease with a consideration of its relationship to acute diffuse lupus erythematosus. Am J Pathoi 16: 375. 1940. 6. Baehr G, Klemperer P, Schifrin A: A diffuse disease of the peripheral circulation usually associated with lupus erythematosus and endocarditis. Trans Assoc Am Phys 50: 139, 1935. 7. Hargraves MM, Richmond H, Morton R: Presentation of two hone marrow elements: the "Tart" cell and the "L.E." cell. Mayo Clin Proc 23: 25-28, 1948. 8. Tan EM, Cohen AS, Fries JF, et al: The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 25: 1271-1277. 1982. 9. Hochberg MC: Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 40: 1725. 1997. 10. Ferraz MB, Goldenherg J, Hilario MO, et al: Evaluation of the 1982 ARA lupus criteria data set in pediatric patients. Clin Exp Rheumatol 12: 83-87, 1994. I I. Trimble RB. Townes AS, Robinson H, et al: Preliminary criteria for the classification of systemic lupus erythematosus: evaluation in early diagnosed SLE and rheumatoid arthritis. Arthritis Rheum 17: 184-188, 1974. 12. Passas CM. Wong RL, Peterson M, et al: A comparison of the specificity of the 1971 and 1982 American Rheumatism Association criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 28: 620-623, 1985. 13. Piette J-C: Updating the American College of Rheumatology criteria for systemic lupus erythematosus: comments on the letter by Hochberg. Arthritis Rheum 41: 751, 1988. 14. Hallengren CS, Nived 0, Surfeit G: Outcome of incomplete systemic lupus erythematosus after 10 years. Lupus 13: 85-88, 2004. 15. Costenbader KH, Mandl LA, Liang MH: Improving the utility of ACR criteria for clinical trials. Lupus 10: S117, 2001. 16. Sanchez ML, Alarcon GS, McGwin G, et al: Can the weighted criteria improve our abiliry to capture a larger number of lupus patients into observational and interventional studies? A comparison with the American College of Rheumatology criteria. Lupus 12: 46~70, 2003. 17. Nobrega FT, Ferguson RH. Kurland LT, et al: Lupus erythematosus in Rochester, Minnesota, 195(}-196S: a preliminary study. In Bennen PH. Wood PHN (eds): Population Studies of the Rheumatic Diseases. International Congress Series, No. 148, Excepta Medica Foundation, 1968. 18. Fessel WJ: Systemic lupus erythematosus in the community: incidence, prevalence, outcome, and first symptoms-the high prevalence in black women. Arch Intern Med 134: 1027-1035, 1974. 19. Fessel WJ: Epidemiology of systemic lupus erythematosus. Rheum Dis Clin North Am 14: 2(}-23 , 1988. 20. Hochberg M: The incidence of systemic lupus erythematosus in Baltimore, Maryland, 197(}-1977. Arthritis Rheum 28: 80-B6. 1985. 21. Siegel M, Lee ML: The epidemiology of systemic lupus erythematosus. Semin Arthritis Rheum 3: I-54. 1973. 22. Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: result from the Canadian Pediatric Rheumatology Association Disease Registry. J Rheumatol 23: 1981-1987. 1996. 23. Pelkonen PM, Jalanko H]. Lantto RR, et al: Incidence of systemic connective tissue diseases in children: a nationwide prospective study in Finland. J Rheumatol 21: 2143-2146. 1994. 24. Kaipiainen-Sappanen 0, Savo!ainen A: Incidence of chronic juvenile rheumatic diseases in Finland during 198(}-1990. Clin Exp Rheumatol 14: 441-444, 1996.
384
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
25, Fujikawa S, Okuni M: A nationwide surveillance study of rheumatic diseases among Japanese children, Acta Paediatrica Japonica 39: 242-244, 1997, 26, Symmons DPM, Jones M, Osborne J, et al: Pediatric rheumatology in the UK: data from the British Pediatric Rheumatology Group National Diagnostic Register, J Rheumatol 23: 1975-1980, 1996, 27, Bowyer S, Roettcher P, and the members of the Pediatric Rheumatology Database Research Group: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey, J Rheumatol 23: 1968-1974, 1996, 28, Lehman TJA, McCurdy DK, Bernstein BH, et al: Systemic lupus erythematosus in the first decade of life, Pediatrics 83: 235-239, 1989, 29, Uramoto KM, Michet CJJ, Steinsson K: Trends in the incidence and mortality of systemic lupus erythematosus, 1950-1992, Arthritis Rheum 42: 46--50, 1999, 30, Cook CD, Wedgwood RJP, Craig JM, et al: Systemic lupus erythematosus: description of 37 cases in children and a discussion of endocrine therapy in 32 of the cases, Pediatrics 26: 570-585, 1960, 31. Harvey AM, Shulman LE, Tumulty PH, et al: Systemic lupus erythematosus: a review of the literature and clinical analysis of 138 cases, Medicine (Baltimore) 33: 291-437, 1954, 32, Lee P, Vrowitz MB, Bookman AAM, et al: Systemic lupus erythematosus: a review of 110 cases with reference to nephritis, the nervous system, infections, aseptic necrosis and prognosis, Q J Med 46: 1-32, 1977, 33, Estes D, Christian CL: The natural history of systemic lupus erythematosus by prospective analysis, Medicine (Baltimore) 50: 85-95, 1971. 34, Cassidy ]T, Sullivan DB, Petty RE, Ragsdale C: Lupus nephritis and encephalopathy: prognosis in 58 children, Arthritis Rheum 20 (Supp!): 315-322, 1977, 35, Norris DG, Colon AR, Stickler GB: Systemic lupus erythematosus in children: the complex problems of diagnosis and treatment encountered in 101 such patients at the Mayo Clinic, Clin Pediatr (Phila) 16: 774--778, 1977, 36, Hagge WW, Burke EC, Stickler GB: Treatment of systemic lupus erythematosus complicated by nephritis in children, Pediatrics 40: 822-827, 1967, 37, Celerrnajer DS, Thorner PS, Baumal R, Arbus GS: Sex differences in childhood lupus nephritis, Am J Dis Child 138: 586--588, 1984, 38, King KK, Kornreich HK, Bernstein BH, et al: The clinical spectrum of systemic lupus erythematosus in childhood, Arthritis Rheum 20 (Supp!): 287-294, 1977, 39, Meislin AG, Rothfield NF: Systemic lupus erythematosus in childhood: analysis of 42 cases with comparative data on 200 adult cases followed concurrently, Pediatrics 42: 37-49, 1968, 40, Walravens PA, Chase HP: The prognosis of childhood systemic lupus erythematosus, Am J Dis Child 130: 929--933, 1976, 41. Kaslow RA: High rate of death caused by systemic lupus erythematosus among V,S, residents of Asian descent. Arthritis Rheum 25: 414-418, 1982, 42, Feng PH, Boey ML: Systemic lupus erythematosus in Chinese: the Singapore experience, Rheumatollnt 2: 151-154, 1982, 43, Lee BW, Yap HK, Yip WC, et al: A 10 year review of systemic lupus erythematosus in Singapore children. Aust Paediatr J 23: 163-165, 1987. 44, Chen]H, Lin CY, Chen WP, et al: Systemic lupus erythematosus in children. Chinese' Microbiol Immunol 20: 23-28, 1987. 45. Tejani A: Nicastri AD, Chen CK, et al: Lupus nephritis in Black and Hispanic children. Am J Dis Child 137: 481-483, 1983. 46. Moreton RO, Gershwin ME, Brady C, Steinberg AD: The incidence of systemic lupus erythematosus in North American Indians. J Rheumatol 3: 186-190, 1976, 47, Boyer GS, Templin DW, Lanier AP: Rheumatic diseases in Alaskan Indians of the southeast coast: high prevalence of rheumatoid arthritis and systemic lupus erythematosus. J Rheumatol18: 1477-1484. 1991. 48. Delgado EA, Malleson PN, Pirie GE, Petty RE: The pulmonary manifestations of childhood onset systemic lupus erythematosus. Semin Arthritis Rheum 19: 285-293, 1990, 49. Sutcliffe N, Clarke AE, Gordon C, et al: The association of socioeconomic status, race, and psychosocial factors and outcomes in patients with systemic lupus erythematosus. Rheumatology 38: 1130-1137, 1999. 50, Calco-Alen J, Reveille JD,Rodrlquez-Valverde V, et al: Clinical, immunogenetic and outcome featues of Hispanic systemic lupus erythematosus patients of different ethnic ancestty. Lupus 12: 377-385, 2003, 51. Reveille JD, Bartolucci A, Alarcon GS: Prognosis in systemic lupus erythematosus: negative impact of increasing age at onset, black race, and thrombocytopenia as well as causes of death. Arthritis Rheum 33: 37-48, 1990. 52, Johnson AE, Gordon C, Palmer RG, Bacon PA: The prevalence and incidence of systemic lupus erythematosus in Birmingham, England: relationship to ethnicity and country of birth. Arthritis Rheum 38: 551-558, 1995. 53. Wanstrat A, Wakeland E: The genetics of complex autoimmune diseases: non-MHC susceptibility genes, Nat Immunol 2: 802-809, 2001. 54. Tsao BP: TIle genetics of human systemic lupus erythematosus, Trends Immunol 24: 595-602, 2003. 55. Tsao BP, Cantor RM, Kalunian KC, et al: Evidence for linkage of a candidate chromosome I region to human systemic lupus erythematosus. J Clin Invest 99: 725-731, 1997, 56, Hochberg MC: The application of genetic epidemiology to systemic lupus erythematosus. J Rheumatol 14: 867-869, 1987.
57. Vyse TJ, ToddJA: Genetic analysis of autoimmune disease, Cell 85: 311-3IH, 1996. 58. Deapen D, Escalante A, Weinrib L, et al: A revised estimate of twin concordance in systemic lupus erythematosus, Arthritis Rheum 35: 311-318, 1992, 59. Kamat SS, Pepmueller PH, Moore n: Triplets with systemic lupus erythematosus. Arthritis Rheum 48: 3176-3180, 2003. 60. Leonhardt ET: Family studies in systemic lupus erythematosus. Clin Exp Immunol 2 (Suppl): 743-759, 1967. 61. Larsen RA: Family studies in systemic lupus erythematosus. J Chronic Dis 25: 187-190, 197262, DeHoratius RJ, Pillarisetty RP, Messner RP, Talal N: Antinucleic acid antibodies in systemic lupus erythematosus patients and their families: incidence and correlation with lymphocytotoxic antibodies. J Clin Invest 56: 1149-1154, 1975, 63, Mitchell AJ, Rusin LHJ, Diaz LA: Circumscribed scleroderma with immunologic evidence of systemic lupus erythematosus, Arch Dermatol 116: 69---73, 1980. 64, Lippman SM, Arnett FC, Conley CL, et al: Genetic factors predisposing to autoimmune disease: autoimmune hemolytic anemia, chronic thrombocytopenic purpura, and systemic lupus erythematosus. Am J Med 73: 827-840, 1982. 65, Labita RG, Chiorazzi N, Gibofsky A, et al: Familial systemic lupus erythematosus in males. Arthritis Rheum 26: 39-42, 1983, 66, Graham RR, Ortmann WA, Langefeld CD, et a1. Visualizing human leukocyte antigen class risk haplotypes in human systemic lupus erythematosus. Am J Hum Genet 71: 543-553, 2002. 67. Tan FK, Arnett FC: The genetics of lupus. Curl' Opin Rheumatol 10: 399-408, 1998, 68. Vyse 1], Kotzin BL: Genetic susceptibility to systemic lupus erythematosus. Ann Rev Immunol 16: 261-292, 1998, 69. Mason q, Isenberg DA: Immunopathogenesis of SLE. Baillieres Clin Rheumatol 12: 385-403, 1998, 70. Griffing WL, Moore SB, Luthra HS, et al: Associations of antibodies to native DNA with HLA-DRw3: a possible major histocompatibility-linked human immune response gene. J Exp Med 152: 319---325, 1980. 71, Ahearn JM, Provost IT, Dorsch CA, et al: Interrelationships of HLA-DR, MB and MT phenotypes, autoantibody expression and clinical features in systemic lupus erythematosus. Arthritis Rheum 25: 1031-1040, 1982. 72. Schur PH, Meyer I, Garovoy M, Carpenter CD: Associations between systemic lupus erythematosus and the major histocompatibility complex: clinical and immunologic considerations, Clin Immunol Immunopathol 24: 263-375, 1982. 73. Khanduja S, Arnett FC, Reveille JD: HLA-DQ beta genes encode an epitope for lupus specific DNA antibodies [abstract], Clin Res 38: 975A, 1995, 74. Marchini M, Antonioli R, LIeD A, et al: HLA class II antigens associated with lupus nephritis in Italian SLE patients. Hum Immunol 64: 462-468, 2003, 75, Pickering MC, Walport MJ: Links hetween complement abnormalities and systemic lupus erythematosus. Rheumatology (Oxf) 39: 133-141, 2000. 76. Bowness P, Davies KA, Norsworthy PJ, et al: Hereditary Clq deficiency and systemic lupus erythematosus. Q J Med 87: 455-464, 1994. 77. Slingsby JH, Norsworthy P, Pearce G, et al: Homozygous hereditary Clq deficiency and systemic lupus erythematosus, Arthritis Rheum 39: 663-670, 19%. 78. Berkel Al, Petry F, Sanal 0, et al: Development of systemic lupus erythematosus in a patient with selective complete Clq deficiency. Eur J Pediatr 156: 113-115, 1997, 79, Walport MJ, Lachmann PJ: Complement deficiencies and abnormalities of the complement system in systemic lupus erythematosus and related disorders. Curl' Opin Rheumatol 2: 661-663, 1990. 80. Davies EJ, Teh LS, Ordi-Ros], et al: A dysfunctional allele of mannose-binding protein associated with systemic lupus erythematosus in a Spanish population, J Rheumatol 24: 485-488, 1997. 81. Sullivan KE, Wooten C, Schmeckpeper BJ, et al: A promoter polymorphism of tumor necrosis factor associated with systemic lupus erythematosus in African Americans. Arthritis Rheum 40: 2207-2211, 1997. 82, Sturfelt G, Hellmer G, Truedsson L: TNF microsatellites in systemic lupus erythematosus: a high frequency of the TNFdabc 2-3-1 haplotype in mlllticase SLE families. Lupus 5: 618-622, 1996. 83, Wilson AG, Gordon C, di Giovine, FS, et al: A genetic association between systemic lupus erythematosus and tumor necrosis factor alpha. Eur J Immunol 24: 191-195, 1994. 84. Rood MJ, van Krugten MY, van del' Linden MW, et al: TNF-30HA and HLADR3 alleles contribute independently to susceptibility to systemic lupus erythematosus. Arthritis Rheum 43: 129--134, 2000, 85. van del' Linden MW, van del' Slik AR, Zanelli E, et a1: Six microsatellite markers on the short arm of chromosome 6 in relation to HLA-DR3 and TNF-30HA in systemic lupus erythematosus. Genes Immunity 2: 373-380, 2001, 86. Lll LY, Ding WZ, Fici D, et al: Molecular analysis of major histocompatibiliry complex allelic associations with systemic lupus erythematosus. Arthritis Rheum 40: 1138-1145, 1997. 87, Jacob CO, Fronek Z, Lewis GD, et al: Heritable major histocompatibility complex class II-associated differences in production of tumour necrosis factor-IX: relevance to genetic predisposition to systemic lupus erythematosus. Proc Natl Acad Sci V S A 87: 1233-1237, 1990, 88, Wilson AG, Duff GW: Genetics of tumour necrosis factor in systemic lupus erythematosus. Lupus 5: 87-88, 1996,
C HAP T E R 89. Parks CG, Cooper GS, Dooly MA, et al: Systemic lupus erythematosus and Ilenetic variation in the interleukin 1 gene cluster: a population based study in the southeastern United Sates. Ann Rheum Dis 63: 91-94, 2004. 90. Salmon JE, Pricop L: Human receptors for inununoglobulin G: key elements in the pathogenesis of rheumatic disease. Arthritis Rheum 44: 739-750, 2001. 91. Ouits AJ, Bootsma H, Derksen RHWM, et al: Skewed distribution of IgG Fe receptor I1a (CD32) polymorphism is associated with renal disease in systemic lupus erythematosus patients. Arthritis Rheum 39: 1832-1836, 1995. 92. Salmon JE, Millard S, Schachter LA, et al: FcyRllA alleles are heritable risk for lupus nephritis in African Americans. J Clin Invest 97: 1348-1354, 1995. 93. IKarassa FB, Trikalinos TA, Ioaniddis JP: The Fcy receptor I1a polymorphism in susceptibility to systemic lupus erythematosus and lupus nephritis: a meta-analysis. Arthritis Rheum 46: 1563-1571, 2002. 94. \VU J, Edberg JC, Redecha PB, et al: A novel polymorphism of FeyIIIa (CD16) alters receptor function and predisposes to autoimmune disease. J Clin Invest 100: 1059-1070, 1997. 95. Karassa FB, Trikalinos TA, Ioaniddis JP: The Fey RlIlA-FI58 allele is a risk factor for the development of lupus nephritis: a meta-analysis. Kidney Int 63: 1475-1482, 2003. 96. Huang 0, Morris D, Dunckley H, et al: Lack of linkage between antinuclear *ntibody or clinical features of systemic lupus erythematosus and TCRNVb lind in families of subjects with SLE. Lupus 6: 527-532, 1997. 97. Prokunia L, Castmejo-Lopez C, Oberg F, et al: A regulatory polymorphism in POVDl is associated with susceptibility to systemic lupus'erythematosus in humans. Nat Genet 32: 666-669, 2002. 98. Nishimura H, Honjo T: I'D-I: an inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol 22: 265-268, 2001. 99. Tsao BP: Lupus susceptibility genes on human chromosome 1. Int Rev Immunol 19: 319-334, 2000. 100. Parsa A, Peden E, Lum RF, et al: Association of antiotensin-converting ,mzyme polymorphisms with systemic lupus erythematosus and nephritis: analysis of 644 SLE families. Genes Immun 3 (Suppl 1) S42-S46, 2002. 101. Nath SK, Quntero-Del-Rlo AJ, Kilpatrick J, et al: Linkage at 12q24 with sys!emic lupus erythematosus (SLE) is established and confmned in Hispanic ~nd European American familes. Am J Hum Genet 74: 73-82, 2004. 102. Lipsky PE: Systemic lupus erythematosus: an autoimmune disease of B cell hyperactivity. Nature Immunology 2: 764-766, 2001. 103. lOinman DM: B-cell abnormalities characteristic of systemic lupus erythematosus. In Wallace D, Hahn BH (eds); Dubois' Lupus Erythematosus, 5th ed. Baltimore, Williams & Wilkins, 1997, I' 195. 104. Becker TM, Lizzio EF, Merchant LP, et al: Increased multiclonal antibodyforming cell activation in the peripheral blood of patients with SLE. Int Arch All Appl Immunol 66: 293-303, 1982. 105. Balow JE, Tsokos GC: T and B lymphocyte function in patients with lupus nephritis: correlation with renal pathology. Clin Nephrol 21: 93-97, 1984. 106. l.inker-Israeli M, Deans RJ, Wallace DJ, et al: Elevated levels of endogenous U.-6 in systemic lupus erythematosus: a putative role in pathogenesis. .I Immunol 147: 117-123, 1991. 107. Monneaux F, Muller S: Epitope spreading in systemic lupus erythematosus: identification of triggering peptide sequences. Arthritis Rheum 46: 1430-1438,2002. 108. Harris I'D, Haynes L, Sayles PC, et al: Reciprocal regulation of polarized (ytokine production by effector Band T cells. Nature Immunol 1: 475-481, 2000. 109. Unker-Israeli M, Quismorio FP Jr, Horwitz DA: CD8+ lymphocytes from patients with systemic lupus erythematosus sustain, rather than suppress, ~pontaneous polyclonal IgG production and synergize with CD4+ cells to llpport antibody synthesis. Arthritis Rheum 33: 1216-1225, 1990. 110. Williams RC Jr, Malone CC, Huffman GR, et al: Active systemic lupus erythematosus is associated with depletion of the nanlral generic anti-idiotype (anti-F(ab'12) system. J Rheumatol 22: 1075-1085, 1995. 111. j(rych M, Atkinson JP, Holers VM: Complement receptors. Curl' Opin Immunol 4: 8-13, 1992. 112. Koffler D, Schur PH, Kunkel HG: Immunological studies concerning the nephritis of systemic lupus erythematosus. J Exp Med 126: 607-624, 1967. 113. lzui S, Lambert PH, Miescher PA: [n vitro demonstration of a particular affmlty of glomerular basement membrane and collagen for DNA: a possible basis for a local formation of DNA-anti-DNA complexes in systemic lupus erythematosus. J Exp Med 144: 428-443, 1976. 114. Rothfield N, Ross HA, Minta JO, Lepow [H, et al: Glomerular and dermal deposition of properdin in systemic lupus erythematosus. N Engl J Med 287: 681-685, 1972. 115. Gewurz H, Pickering R.I, Mergenhagen SE, Good RA: The complement proIIIe in acute glomerulonephritis, systemic lupus erythematosus and hypocomplementemic chronic glomerulonephritis. Int Arch Allergy Appl [mmunol 34: 556- 570, 1968. 116. Steward MW, Katz FE, West N]: The role of low affinity antibody in immune wmplex disease: the quantity of anti-DNA antibodies in NZBIW Fl hybrid t,lice. Clin Exp Immunol 21: 121-130, 1975. 117. Winfield JB, Faiferman I, Koffler D: Avidity of anti-DNA antibodies in serum and IgG glomerular eluates from patients With systemic lupus erythematosus: association of high avidity antinative DNA antibody with glomerulonephritis. J Clin Invest 59: 90-96, 1977. 118. \.eon SA, Green A, Ehrlich GE, et a1: Avidity of antibodies in SLE: relation to severity of renal involvement. Arthritis Rheum 20: 23-29, 1977.
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
385
119. Hale GM, Highton J, Kalmakoff J, Palmer DG: Changes in anti-DNA antibody affinity during exacerbations of systemic lupus erythematosus. Scand J Rheumatol 15: 243-250, 1986. 120. Sher JH, Pertschuk LP: Immunoglobulin G deposits in the choroid plexus of a child with systemic lupus erythematosus. J Pediatr 85: 385-387, 1974. 121. Palucka AK, Banchereau.l, Blanco 1', Pascual V: The interplay of dendritic cell subsets in systemic lupus erythematosus. Immunol Cell Bioi 80: 484-488, 2002. 122. Pascual V, Banchereau J, Palucka AK: The central role of dendritic cells and interferon-alpha in SLE. Curl' Opin Rheumatol 15: 548-556, 2003. 123. Blanco 1', Palucka AK, Gill M, et a1: Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Science 294: 1540-1543,2001. 124. Rupee RA, Petropoulou T, Belohradsky M, et al: Lupus erythematosus tumidus and chronic discoid lupus erythematois in carriers of X-linked chronic granulomatous disease. Eur J Dermatol 10: 184-189, 2000. 125. Manzi S, Urbach AH, McCune AB, et aI: Systemic lupus erythematosus in a boy with chronic granulomatous disease: case report and review of the literature. Arthritis Rheum 34: 101-105, 1991. 126. Badolato R, Notarangelo LD, Plebani A, Roos D: Development of systemic lupus erythematosus in a young child affected with chronic granulomatous disease follOWing withdrawal of treatment with interferon-gamma. Rheumatology (Ox£) 42: 804-805, 2002. 127. Miranda-Carus ME, Anhalt G, Rosen A: Autoantigens targeted in systemic lupus erythematosus are clustered in two populations of surface structures on apoptotic keratinocytes. J Exp Med 179: 1317-1330, 1994. 128. White S, Rosen A: Apoptosis in systemic lupus erythematosus. Curr Opin Rheumatol 15: 557-562, 2003. 129. Emlen W, Niebur J, Kadera R: Accelerated apoptasis of lymphocytes from patients with systemic lupus erythematosus. J Immunol 152: 3685-3692, 1994. 130. Taylor PR, Carugati A, Fadok VA, et al: A hierarchical role for classical pathway complement proteins in the clearance of apoptotic cells in vivo. J Exp Med 192: 359-366, 2000. 131. Aringer M, Wintersberger W, Steiner CW, et al: High levels of bcl-2 protein in circulating T lymphocytes, but not B cells, of patients with systemic lupus erythematosus. Arthritis Rheum 37: 1423-1430, 1994. 132. Rose LM, Latchman DS, Isenberg DA: Apoptosis in peripheral lymphocytes in systemic lupus erythematosus: a review. Br J Rheumatol 36: 158-163, 1997. 133. Mysler E, Bini P, Drappa J, et al: The apoptosis-l/fas protein in human SLE. J Clin Invest 93: 1029-1034, 1994. 134. Vaishnaw AK, Toubi E, Ohsako S, et al: The spectrum of apoptotic defects and clinical manifestations, including systemic lupus erythematosus, in humans with CD95 (Fass/APO 1) mutations. Arthritis Rheum 42: 1833-1842, 1999. 135. Elkon KB: Apoptosis in SLE: Too little or too much? Clin Exp Rheumatol 12: 553, 1994. 136. Xu L, Zhang L, Yui Y, et al: Human lupus T cells resist inactivation and escape death by upregulating COX-2. Nat Med 10: 411-415, 2004. 137. Beagley KW, Gockel CM: Regulation of innate and adaptive immunity by the female sex hormones oestradiol and progesterone. FEMS Immunol Med Microbial 38: 13-22, 2003. 138. Wilder RL: Neuroimmunoendocrinology of the rheumatic diseases. Ann N Y Acad Sci 96: 13-19,2002. 139. Cooper GS, Dooley MA, Treadwell EL, et al: Hormonal and reproductive risk factors for development of systemic lupus erythematosus. Arthritis Rheum 46: 1830-1839, 2002. 140. Athreya BH, Rafferty JH, Sehgal GS, Lahita RG: Adenohypophyseal and sex hormones in pediatric rheumatic diseases. J Rheumatol 20: 725-730, 1993. 141. Lahita RG, Bradlow HL, Kunkel HG, Fishman J: Increased 16 alpha hydroxylation of estradiol in systemic lupus erythematosus. J Clin Endocrinol Metab 53: 174-178, 1981. 142. Juikunen HI: Oral contraceptives in systemic lupus erythematosus: side effects and influence on the activity of SLE. Scand J Rheurnatol 20: 427-433, 1991. 143. Petri M, Howard D, Repke]: Frequency of lupus flare in pregnancy: the Hopkins Lupus Pregnancy Center experience. Arthritis Rheum 34; 1538-1545, 1991. 144. Lockshin MD: Pregnancy does not cause systemic lupus erythematosus to worsen. Arthritis Rheum 32: 665-670, 1989. 145. Lavalle C, Loyo E, Paniagua R, et al: Correlation study between prolactin and androgens in male patients with systemic lupus erythematosus. J Rheumatol 14: 268-272, 1987. 146. Jungers 1', Dougados M, Pelissier C, et al: Influence of oral contraceptive therapy on the activity of systemic lupus erythematosus. Anhritis Rheum 25: 618-623, 1982. 147. Gilliland WR, Stashower ME: Klinefelter's syndrome and systemic lupus erythematosus. Clin Exp Rheumatol 18: 107-109, 2000. 148. Mok CC, Lau CS: Profile of sex hormones in male patients with systemic lupus erythematosus. Lupus 9: 252-257, 2000. 149. Jara-Quezada L, Graef A, Lavalle C: Prolactin and gonadal hormones during pregnancy in systemic lupus erythematosus. J Rheumatol 18: 349-353, 1991. 150. McMurray RW, Weidensaul 0, Allan SH, Walker SE: Efficacy of bromocriptive in an open label therapeutic trial for SLE. J Rheumatol 22: 2084-2091, 1995. 151. Casciola-Rosen L, Rosen A: Ultraviolet light-induced keratinocyte apoptosis: a potential mechanism for the induction of skin lesions and autoantibody production in LE. Lupus 6: 175-180, 1997. 152. Funk JO, Holler E, Kohlhuber F, et al: Protein kinase C activation is involved in ultraviolet B irradiation-induced endothelial cell ICAM-I up-regulation
386
153. 154.
155. 156.
157. 15H.
159.
160. 161. 162. 163.
164.
165. 166.
167. i6H.
169. 170. 171.
172. 173. 174. 175. 176.
177. 178. 179.
180.
181. 182. 1H3. IH4.
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
and lymphocyte-endothelium interaction in vitro. Scand J Immunol 44: 54-60, 1996. Treina G, Scaletta C, Fourtanier A, et al: Expression of intercellular adhesion molecule-I in lNA-irradiated human skin cells in vitro and in vivo. Br r Dermatol 135: 241-247, 1996. . Strickland I, Rhodes LE, Flanagan BF, Friedmann PS: TNF-alpha and IL-8 are upregulated in the epidermis of normal human skin after LNB exposure: correlation with neutrophil accumulation and E-selectin. J Invest Dermatol lOH: 763-768, 1997. Cohen MR, Isenberg DA: Ultraviolet imtdiation in systemic lupus erythematosus: friend or foe? Br J Rheumatol 35: 1002-1007, 1996. Incaprera M, Rindi L, Bazzichi A, Garzelli C: Potential role of the EpsteinBarr virus in systemic lupus erythematosus autoimmunity. Clin Exp Rht'umatol 16: 289-294, 1998. Rider JR, allier WER, Lock RJ, et al: Human cytomegalovirus infection and systemic lupus erythematosus. Clin Exp Ri,eumatol IS: 405-409, 1997. Tsai YT, Chiang BL, Kao YE, Hsieh KH: Detection of Epstein-Barr virus and cytomegalovirus genome in white blood cells from patients with juvenile rheumatoid atthritis and childhood systemic lupus erythematosus. Int Arch Allergy Immunol 106: 235-240, 1995. Severin MC, Levy Y, Shoenfeld Y: Systemic lupus erythematosus and parvovilus B-19: casual coincidence or causative culprit? Clin Rev Allergy Immunol 25: 41-48, 2003. Rubin RL: Etiology and mechanisms of drug-induced lupus. Curr Opin Rheumatol 11: 357-363, 1999. Prete P: The mechanisms of action of L-canavanine in inducing autoimmune phenomena. Atthritis Rheum 28: 1198--1200, 1985. Bardana EJ Jr, Manilow MR, Houghton DC, et al: Diet-induced systemic lupus erythematosus in primates. Am J Kidney Dis I: 345-352, 1982. Sanchez-Guerrero J, Karlson EW, Colditz GA, et al: Hair dye use and the risk of developing systemic lupus erythematosus: a cohott study. Atthritis Rheum 39: 657-662, 1996. Carr RI, Hoffmann AA, Harbeck RJ: Comparison of DNA binding in normal population, general hospital laboratory personnel, and personnel from laboratories studying SLE. J Rheumatol 2: 178--183, 1975. Nguyen C, Limaye N, Wakeland EK: Susceptibility genes in the pathogenesis of murine lupus. Atthritis Res 4 (Suppl 3): S255-S263, 2002. Alexander EL, Moyer C, Travlos GS, et al: Two histopathologic types of inflammatoty vascular disease in MRVMp autoimmune mice: model for human vasculitis in connective ri"ue disease. Atthritis Rheum 28: 1146-1155, 1985. Eisenberg RA, Tan EM, Dixon FJ: Presence of anti-Sm reactivity in autoimmune mouse strains. J Exp Med 147: 592-587, 1978. Sontheimer RD: The lexicon of cutaneous lupus erythematosus: a review and personal perspective on the nomenclature and classification of the Clltaneous manifestations of lupus erythematosus. Lupus 6: 84-95, 1997. Millard TP, Hawk JLM, McGregor JM. Photosensitivity in lupus. Lupus 9: 3-10, 2000. Wananukul S, Watana D, Pongprasit P: Cutaneous manifestations of childhood systemic lupus erythematosus. Pediatr Dermatol 15: 342-346, 1998. ShelWood MC, Pincott JR, Goodwin FJ, Dillon MJ: Dominantly inherited glomenilonephritis and an unusual skin disease. Arch Dis Child 62: 1278-1280, 1997. German J: Bloom's syndrome. Derm Clin 13: 7-18, 1995. Jung EG: The red face: Photogenodermatoses. Clin Dermatol 11: 275-281, 1993. Oates RK, Lewis MB, Walker-Smith JA: The Rothmund-Thomson syndrome: case repott of an unusual syndrome. Aust Pediatr J 7: 103-107, 1971. Sontheimer RD, Provost IT: Cutaneous manifestations of lupus erythematosus. In Wallace DJ, Hahn DH (eds): Dubois' Lupus Erythematosus, 4th ed. Philadelphia, Lea & Febiger, 1997. Abrcon-Segovia D, DeJeze M, aria CV, et al: Antiphospholipid antibodies and the antiphospholipid syndrome in systemic lupus erythematosus prospective analysis of 500 consecutive patients. Medicine
185. Koransky .IS, Esterly NB: Lupus panniculitis (profundus). J Pediatr 98: 241-244, 1981. 186. Millard LG, Rowell NR: Chilblain lupus erythematosus (Hutchinson): a clinical and laboratory study of 17 patients. Br J Dermato] 98: 497-506, 1978. 187. Yell JA, Burge SM: Watts and lupus erythematosus. Lupus 2: 21-23, 1993. 188. Ragsdale CG, Petty RE, Cassidy JT, Sullivan DB: The clinical progression of apparent juvenile rheumatoid atthritis to systemic lupus elythematosus. J Rheumatol 7: 50-55, 1980. 189. Reilly PA, Evison G, McHugh NJ, Maddison PJ: Atthropathy of hands and feet in systemic lupus erythematosus. J Rheumatol 17: 777-784, 1990. 190. Foote RA. Kimbrough SM, Stevens JC: Lupus myositis. Muscle Nerve S: 65-68, 1982. 191. Smith FE, Sweet DE, Bnmner CM, Davis .IS IVth: Avascular necrosis in SLE: an apparent predilection for young patients. Ann Rheum Dis 35: 227-232. 1976. 192. Assouline-Dayan Y, Chang C, Greenspan A. et al: Pathogenesis and natural history of osteonecrosis. Semin Atthritis Rheum 32: 94-124, 2002. 193. Cameron JS: Lupus nephritis in childhood and adolescence. Pediatr Nephrol 8: 230-249, 1994. 194. L~Manna A, Polito C, Papale MR, et al: Chronic interstitial cystitis and systemic lupus erythematosus in an 8-year old girl. Pediatr Nephrol 12: 139-140, 1998. 195. MinJK, Byun)Y, Lee SH, et al: Urinary bladder involvement in patients with systemic lupus erythematosus: with review of the literature. Korean J Intern Med 15: 42-50, 2000. 196. Yancey CI, Doughty RA, Athreya BH: Central nervous system involvement in childhood systemic lupus erythematosus. Atthritis Rheum 24: 1389-1395, 1981. 197. Parikh S, Swaiman KF, Kim Y: Neurologic characteristics of childhood lupus erythematosus. Pediatr Neurol 13: 198-201, 1995. 198. Steinlin MI, Blaser SI, Gilday DL, et al: Neurologic manifestations of pediatric systemic lupus erythematosus. Pediatr Neul'Ol 13: 191-197, 1995. 199. Sibbitt WL Jr, Brandt JR, Johnson CR, et al: The incidence and prevalence of neuropsychiatric syndromes in pediatric onset systemic lupus erythematosus. J Rheumatol 29: 1536-1542, 2002. 200. ACR Ad Hoc Committee on Neuropsychiatric Lupus Nomenclature: The American College of Rheumatology nomenclature and case definitions for neuropsychiatric lupus syndromes. Althritis Rheum 42: 599-608, 1999. 20 \. Nived 0, Sturfelt G, Liang MH, De Pablo P: The ACR nomenclature for CNS lupus revisited. Lupus 12: 872-876, 2003. 202. Silber TJ, Chatoor I, White PH: Psychiatric manifestations of systemic lupus erythematosus in children and adolescents: a review. Clin Pediatr 23: 331-335, 1984. 203. Wyckotl' PM, Miller LC, Tucker LB, Schaller JG: Neuropsychological assessment of children and adolescents with systemic lupus erythematosus. Lupus 4: 217-220, 1995. 204. Papero PH, Bluestein HG, White P, Lipnick RN: Neuropsychologic deficits and antineuronal antibodies in pediatric systemic lupus erythematosus. Clin Exp Rheumatol 8: 417-424, 1990. 205. Cuadrado MJ, Sanna G: Headache and systemic lupus erythematosus. Lupus 12: 943-946, 2003. 206. Uziel Y, Laxer RM, Blaser S, et al: Cerebral vein thrombosis in childhood systemic lupus erythematosus. .J Pediatr 126: 722-727. 1995. 207. Herd JK, Medhi M, llzendoski DM, et al: Chorea associated with systemic lupus erythematosus: report of two cases and review of the literature. Pediatrics 61: 308-315, 1978. 208. Kukla LF, Reddy C, Silkalna G, et al: Systemic lupus erythematosus presenting as chorea. Arcb Dis Child 53: 345-347, 1978. 209. Arisaka 0, Obinata K, Sasaki H, et al: Chorea as an initial manifestation of systemic lupus erythematosus: a case repott of a 10-year old girl. Clin Pediatr 23: 298--300, 1984. 210. Bmyn GW, Padberg G: Chorea and systemic lupus erythematosus: a critical review. Eur Neurol 23: 435-448, 1984. 211. Asherson RA, Derksen RH, Harris EN, et al: Chorea in systemic lupus e,ythematosus and "lupus-like" disease: association with antiphospholipid antibodies. Semin Atthritis Rheum 16: 253-259, 1987. 2J2. Groothuis JR, Groothuis DR, Mukhopadhyay D, et al: Lupus-associated chorea in childhood. Am J Dis Child 131: 1131-1134, 1977. 213. Zetterstrom R, Berglund G: Systemic lupus etythematosus in childhood: a clinical study. Acta Paediatl' Scand 45: 189-204, 1956. 214, Olfat MO, AI-Mayouf SM, Muzaffer MA. Pattern of neuropsychiatric manifestations and outcome in juvenile systemic lupus erythematosus. Clin Rheumatol 23: 395-399, 2004. 215. Watanabe T, Onda H. Hemichorea with antiphospholipid antibodies in a patient with lupus nephritis. Pediatr Nephrol 19: 451-453, 2004. 216. Shahar E, Goshen E, Tauber Z, Lahat E: Parkinsonian syndrome complicating systemic lupus erythematosus. Pediatric Neurol 18: 456-458, 1998. 2J7. AI-Mayouf, SM, Bahabri S: Spinal cord involvement in pediatric systemic lupus erythematosus: case report and literature review. Clin Exp Rheumatol 17: 505-508, 1999. 218. Blaustein DA, Blaustein SA: Antinuclear antibody negative systemic lupus erythematosus presenting as bilateral facial paralyses. J Rheumatol 2';: 798--800, 1998. 219. DelGiudice GC, Scher CA. Athreya BH, et al: I'seudotumor cerebri and childhood systemic lupus erythematosus. J Rheumatol 13: 748--752, 1986.
C HAP T E R 220. f'ablos JL, del Rincon E. Francisco F. Mateo I: Narcolepsy in systemic lupus erythematosus. J Rheumatol 20: 375-376. 1993. 221. Ciaccio M, Parodi A, Rebora A: Myasthenia gravis and lupus erythematosus. rnt J Dermatol 28: 317-320, 1989. 222. JlCobson EJ, Reza MJ: Constrictive pericarditis in systemic lupus elYthematosus: demonstration of immunoglobulins in the pericardium. Arthritis Rheum 21: 972-974, 1978. 223. Kahl LE: The spectrum of pericardial tamponade in systemic lupus erythematosus. Arthritis Rheum 35: 1343-1349. 1992 224. Ehrenfeld M, Asman A, Shpilberg 0, Samra Y: Cardiac tamponade as the pre$enting manifestation of systemic lupus erythematosus. Am J Med 86: (126-627, 1989. 225. Lerer RJ: Cardiac tamponade as an initial finding in systemic lupus erythe,natosus. Am J Dis Child 124: 436-437. 1972. 226. Malcic I. Senecic I, Dasovic A, Radonic M: Incipient pericardial tamponade as the first symptom of systemic lupus erythematosus in 2 children. Reumatizam 42: 19-22, 1995. 227. Tamura EJ: Cardiac tamponade in a child with systemic lupus erythematosus and IgG2 and IgG4 subclass deficiencies. Eur J Pediatr 157: 475-478, 1998. 228. Oshiro AC. Derbes SJ, Stopa AR, Gedalia A: Anti-Ro/SS-A and anti-La/SS-B antibodies associated with cardiac involvement in childhood systemic lupus erythematosus. Ann Rheum Dis 56: 272-274. 1997. 229. Utowitz MB. Gladman DO: Accelerated atheroma in lupus: background. Lupus 9: 161-165. 2000. 230. Homcy CJ. Liberthson RR, Fallon JT, et al: Ischemic heart disease in systemic lupus erythematosus in the young patient: report of six cases. Am J Cardiol 49: 478-484, 1982. 231. Spiera H, Rothenberg RR: Myocardial infarction in four young patients with SLE. J Ri,eumatol 10: 464-466, 1983. 232. Maider YS, Robert~ We: Coronary arterial disease in systemic lupus erythematosus: quantification of degrees of narrowing in 22 necropsy patients (21 women) aged 16 to 37 years. Am J Med 70: 775-781. 1981. 233. Englund JA. Lucas RV Jr: Cardiac complications in children with systemic lupus erythematosus. Pediatrics 72: 724-730. 1983. 234. Hadui E. Garcia-Rubi D. Robles E, et al: Cardiovascular manifestations in systemic lupus erythematosus: prospective study of 100 patients. Angiology 36: 431-441, 1985. 235. Mandell BF: Cardiovascular involvement in systemic lupus erythematosus. Semin Arthritis Rheum 17: 12~141. 1987. 236. Bonfiglio TA. Botti RE, Hagstrom JWC: Coronary arteritis. occlusion. and myocardial infarction due to lupus erythematosus. Am Heart J 83: 153-158. 1972. 237. nor I: Myocardial infarction and ischemic heart disease in infants and chilqren: analysis of 29 cases and review of the literature. Arch Dis Child 44: ~68-281. 1969. 239. Friedman OM, Lazarus HM, Fierman AH: Acute myocardial infarction in fl'Idiatric systemic lupus erythematosus. J Pediatr 117: 263-266, 1990. 240. &;haller JG. Gilliland BG. Ochs HD, et al: Severe systemic lupus erythematosus with nephritis in a boy with deficiency of the fourth component of complement. Arthritis Rheum 20: 1519-1525. 1977. 241. Fearon WF, Cooke JP: Acute myocardial infarction in a young woman with systemic lupus erythematosus. Vasc Med 1: 19-23. 1996. 242. MJlIer OJ, Maisch SA, Perez MD, et al: Fatal myocardial infarction in an 8-yearold girl with systemic lupus erythematosus, Raynaud's phenomenon, and secondary antiphospholipid antibody syndrome. J Rheumatol 22: 768-773, 1995. 243. Gazarian M, Feldman BM, Benson LN. et al: Assessment of myocardial perf\lSion and function in childhood systemic lupus erythematosus. J Pediatr 132: 109-116. 1998. 244. Petri M. Perez-Gunhann S, Spence D, Hochberg MC: Risk factors for coronary artery disease in patient~ with systemic lupus erythematosus. Am .l Med 9:'\: 513-519, 1992. 245. Falaschi F, Ravelli A, Martignoni A. et al: Nephrotic-range proteinuria, the major risk factor for early atherosclerosis in juvenile-onset systemic lupus erythematosus. Arthritis Rheum 43: 1405-1409. 2000. 246. Alves JD, Ames PRe Atherosclerosis, oxidative stress and auto-antibodies in systemic lupus erythematosus and primaly antiphospholipid syndrome. Immunobiology 207: 23-28. 2003. 247. Bulkley BH, Roberts WC: TI,e heart in systemic lupus erythematosus and the changes induced in it by corticosteroid therapy: a study of 36 necropsy patients. Am J Med 58: 243-264, 1975. 248. Uowite NT, Samuel P, Ginzler E, et aI: Dyslipoproteinemia in pediatric systemic lupus erythematosus. Arthritis Rheum 31: 859-863, 1988. 249. Smith GW, Hannan SF, Scott PJ, et al: Immune complex-like activity associated with abnormal serum lipoproteins in systemic erythematosus. Clin Exp Immunol 48: 8-16, 1982. 250. Alves JD, Ames PRJ, Donohue S, et al: Antibodies to high-density lipoprot
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
387
253. Roldan CA. Shively BK, Crawford MH: An echocardiographic study of vaivular heart disease associated with systemic lupus erythematosus. N Engl] Med 335: 1424-1430, 1996. 254. AI-Abbad A.l. Cabral DA, Sanatani S, et al: Echocardiography and pulmonary function testing in childhood onset systemkic lupus erythematosus. Lupus 10: 32-37, 2001. 255. Chauvaud SM, Kalangos A, Berrebi A.l, et al: Systemic lupus erythematosus valvulitis: mitral valve replacement with a homograft. Ann Thorac Surg 60: 1803-1805, 1995. 256. Morin AM, Boyer AS, Nataf P, Gandjbakhch I: Mitral valve insufficiency caused by systemic lupus erythematosus requiring valve replacement: three case reports and a review of the Iiteralure. Thorac Cardiovasc Surg 44: 314-316, 1996. 257. Kaplan SO, Chartash EK, Pizzarello RA, Furie RA: Cardiac manifestations of the antiphospholipid syndrome. Am Heart J 124: 1331-1338. 1992. 258. Balada E, Ordi-Ros J, Paredes F, et al: Antiphospholtidylserine antihodies contribute to the diagnosis of antiphospholipid syndrome in patients with systemic lupus erythematosus. ScandJ Rheumato! 30: 235-241, 2001. 259. Caeiro F, Michielson FM, Bernstein R, et al: Systemic lupus erythematosus in childhood. Ann Rheum Dis 40: 325-331, 1981. 260. Schaller J: Lupus in childhood. Clin Rheum Dis 8: 219-228, 1982. 261. De Jongste .lC, Neijens HJ, Duivennan E.l, et al: Respiratory tmct disease in systemic lupus erythematosus. Arch Dis Child 61: 478-483, 1986. 262. Singsen BH. Platzker CG: Pulmonary involvement in the rheumatic disorders of children. In Kendig EL, Chernick V (eds): Disorders of the Respimtory Tract in Children, 4th ed. Philadelphia. WB Saunders, 198j, p 846. 263. Rajani KB, Ashbacher LV, Kinney TR: Pulmonary hemorrhage and systemic lupus erythematosus. J Pediatr 93: 810-812. 197H. 264. Ramirez RE, Glasier C, Kirks 0, et al: Pulmonary hemorrhage associated with systemic lupus erythematosus in children. Radiology 152: 409-412, 1984. 265. Miller RW, Salcedo .lR, Fink R.l. et al: Pulmonary hemorrhage in pediatric patients with systemic lupus erythematosus. J Pediatr 108: 57~579, 1986. 266. C;:ift,i E, Yal,inkaya F, Ince E, et al: Pulmonary involvement in childhoodonset systemic lupus erythematosus: a report of five cases. Rheumatology 43: 487-491, 2004. 267. Weinrib L, Sharma OP, Quismorio FP Jr: A long term study of interstitial lung disease in systemic lupus erythematosus. Semin Arthritis Rheum 16: 48-56, 1990. 268. Uziel Y, Laxer RM, Silverman ED: Persistent pulmonary hemorrhage as the sole initial clinical manifestation of pediatric systemic lupus erythematosus. Clin Exp Rheumatol 15: 697-700, 1997. 269. Nadorra RL, Landing BH: Pulmonary lesions in childhood onset systemic lupus erythematosus. Pediatr Pathol 7: 1-18. 1987. 270. Warrington KJ, Moder KG, Brutinel W, Mark W: The shrinking lungs syndrome in systemic lupus erythematosus. Mayo Clin Proc 75: 467-472, 2000. 271. Abramson SG, Dobro J, Eberle MA, et al: Acute reversible hypoxemia in systemic lupus erythematosus. Ann Intern Med 114: 941-947, 1991. 272. Kraus A. Guerra-Bautista G: Laryngeal involvement as a presenting symptom of systemic lupus erythematosus. Ann Rheum Dis 49: 421. 1990. 273. Teitel AD, MacKenzie CR, Stern R, Paget SA: Laryngeal involvement in systemic lupus erythematosus. Semin Arthritis Rheum 22: 203-214, 1992. 274. Foster HE, Malleson PN, Petty RE, et al: Pneumo
388
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
287. Levy DM, Massicotte MP, Harvey E, et al: Thromboembolism in paediatric lupus patients. Lupus 12: 741-746, 2003. 288. Lubbe WF, Asherson RA: Intracardiac thrombus in systemic lupus erythematosus associated with lupus anticoagulant. Arthritis Rheum 31: 1453-1454, 1988. 289. Brown JH, Doherty CC, Allen DC, et al: Fatal cardiac failure due to myocardial microthrombi in systemic lupus erythematosus. Br MedJ 296: 1505, 1988. 290. Tincani A, Lojacono A, Taglietti M, et al.: Pregnancy and neonatal outcome in primary antiphospholipid syndrome. Lupus 11: 649, 2002. 291. Cervera R, Piette J-C, Font .I, et al: Antiphosphospholipid syndrome: clinical and immunologic manifestations and patterns of disease expression in a cohort of 1000 patients. Arthritis Rheum 46: 1019-1027, 2002. 292. Asherson RA, Cervera R, Piete J-C, et al: Catastrophic antiphospholipid syndrome: clues to the pathogenesis from a series of 80 patients. Medicine (Baltimore) 80: 355-377, 2001. 293. Ginsberg .IS, Demers C, Brill-Edwards P, et al: AcqUired protein S deficiency is associated with antiphosphollpid antibodies and increased thrombin generation in patients with systemic lupus erythematosus. Am .I Med 98: 379-383, 1995. 294. Tomas .IF, A1berca I, Tabernero MD, et al: Natural anticoagulant protein and antiphospholipid antibodies in systemic lupus erythematosus. .I RheumalOl 25: 57-62, 1998. 295. Eberhard A, Sparling C, Sudbury S, et al: Hypoprothrombinemia in childhood systemic lupus erythematosus. Semin Arthritis Rheum 24: 12-18, 1994. 296. Fleck RA, Rapaport SI, Roa LVM: Anti-prothrombin antibodies and the lupus anticoagulant. Blood 72: 512-519, 1988. 297. SenecalJL, Rauch .I, Grodizicky T, et al: Strong association of autoantibodies to human nuclear !amin B1 with lupus antiagulant antibodies in systemic lupus erythematosus. Arthritis Rheum 42: 1347-1353, 1999. 298. Dieude M, SenecalJL, RauchJ, et al: Association of autoantibodies to nuclear lamin B1 with thromboprotection in systemic lupus erythematosus. Arthritis Rheum 46: 2695-2707, 2002. 299. Gatenby PA, Smith H, Kirwan P, et al: Systemic lupus erythematosus and thrombotic thrombocytopenic purpura: a case report and review of relationship. .I Rheumatol 8: 504-508, 1981. 300. Bell WR, Braine HG, Ness PM, Kickier TS: Improved survival in thrombocytopenic purpura: hemolytic uremic syndrome--clinical experience in 108 patients. N EnglJ Med 325: 398-403, 1991. 301. Breckenridge RL Jr, Solberg LA, Pineda AA, et al: Treatment of thrombotic thrombocytopenic purpura with plasma exchange, antiplatelet agents, corticosteroid and plasma infusion: Mayo Clinic experience. .I Clin Apheresis 1: 6--13, 1996. 302. Bmnner HI. Freedman M, Silverman ED: Close relationship between systemic lupus erythematosus and thrombotic thrombocytopenic purpura in childhood. Anhritis Rheum 42: 2346-2355, 1999. 303. Rock GA, Shamak KH, Buskard NA, et al: Comparison of plasma exchange with plasma infusion in the treatment of thromboc'ytopenic purpura. N Engl .I Med 325: 393-397, 1991. 304. Schmugge M, Revel-Vilk S, Hiraki L, et al: Thrombocytopenia and thromboembolism in pediatric systemic lupus erythematosus. .I Pediatr 143: 666-669, 2003. 305. Ramakrishna R, Kyle PW, Day PJ, Manoharan A: Evans' syndrome, myelofibrosis and systemic lupus erythematosus: role of procollagens in myelofibrosis. Pathology 27: 255-259, 1995. 306. Gardner FH, Diamond LK: Autoerythrocyte sensitization: a form of purpura producing painful bmising following autosensitization to red blood cells in certain women. Blood 10: 675, 1955. 307. Kurczynski EM, Cassidy JT, Heyn RM: Autoerythrocyte sensitization in a yOllllg boy. Lancet 1: 424-425, 1973. 308. Ansari A, Larson PH, Bates HD: Vascular manifestations of systemic lupus erythematosus. Angiology 37: 423-432, 1986. 309. Au A, O'Day J: Review of severe vaso-occlusive retinopathy in systemic lupus el'ythematosus and the antiphospholipid syndrome: associations, visual outcomes, complications and treatment. Clin Exp Ophthalmol 32: 87-100, 2004. 310. Deprettere AJ, Van Acker KJ, De Clerck LS, et al: Diagnosis of Sjogren's syndrome in children. Am J Dis Child 142: 1185-1187, 1988. 311. Bloch KJ, Buchanan WW, Wohl MJ, BunimJJ: Sjogren's syndrome: a clinical, pathological, and serological study of sixty-two cases. Medicine (Baltimore) 44: 187-231, 1965. 312. Spath M, Kmger K, Dresel S, et al: Magnetic resonance imaging of the parotid gland in patients with Sjogren's syndrome. J Rheumatol 18: 1372-1378, 1991. 313. Daniel TE: Labial salivary gland biopsy in Sjogren's syndrome. Arthritis Rheum 27: 147-156, 1984. 314. Garcia-Consuegra J, Merino R, Alonso A, Goded F: Systemic lupus erythematmus: a case report with unusual manifestations and favourable outcome after plasmapheresis. Eur J Pediatr 151: 581-582, 1992. 315. Saab S, Corr MP, Weisman MH: Corticosteroids and systemic lupus erythematosus pancreatitis: a case series. J Rheumatoi 25: 801-806, 1998. 316. Kolk A, Horneff G, Wllgenbus KK, et al: Acute lethal necrotising pancreatitis in childhood systemic lupus erythematosus: possible toxicity of immunosuppressive therapy. Clin Exp Rheumatol 13: 399-403, 1995. 317. HuangJ-L, Huang C-C, Chen C-Y, Hung 1-]: Acute pancreatitis: an early manifestation of systemic lupus erythematosus. Pediatr Emerg Care 10: 291-293, 1994.
318. Molina]F, Brown RF, Gedalia A, Espinoza LR: Protein losing enteropathy as the initial manifestation of childhood systemic lupus erythematosus. .I Rheurnatol 23: 269-271, 1996. 319. Mukamel M, Rosenbach Y, Mimount M, Dinari G: Celiac disease associated with systemic lupus erythematosus. Israel J Med Sci 30: 656-658, 1994. 320. Stevens HP, Ostlere LS, Rustin MH: Systemic lupus erythematosus in association with ulcerative colitis: related autoimmune diseases. Br J Dermatol 130: 385-389, 1994. 321. Nagata M, Ogawa Y, Hisano S, Ueda K: Crohn's disease in systemic lupus erythematosus: a case report. Eur J Pediatr 148: 525-526, 1989. 322. Gibson T, Myers AR: Subclinical liver disease in systemic lupus erythematosus. J Rheumatol 8: 752-759, 1981. 323. Ravelli A, Caria MC, Malattia C, et aI: Uncommon causes of liver disease in juvenile systemic lupus erythematosus. Clin Exp Rheumatol 19: 474, 2001. 324. Hall S, Czaja AJ, Kaufman OK, et al.: How lupoid is lupoid hepatitis? J Rheum 13: 95-98, 1986. 325. Miele-Verganl G, Vergani 0: Autoimmune liver disease in chidren. Ann Acad Med Singapore 32: 239-243, 2003. 326. Maddrey WC: Chronic hepatitis. In Zakim 0, Boyer 11) (eds): Hepatology: A Textbook of Liver Disease, 2nd ed. Philadelphia, WB Saunders, 1990, p 1025. 327. Miller MH, Urowitz MB, Gladman DO: The liver in systemic lupus erythematosus. QJ Med 211: 401-409, 1984. 328. Dillon AM, Stein HB, English RA: Splenic atrophy in systemic lupus erythematosus. Ann Intern Med 96: 40-43, 1982. 329. Malleson P, Petty RE, Nadel H, et al: Functional asplenia in childhood onset systemic lupus erythematosus. J Rheumatol 15: 1648-1652, 1988. 330, Efremidis A, Eiser A, Grishman E, Rosenberg V: Hodgkin's lymphoma in an adolescent with SLE. Cancer 53: 142-146, 1984. 331. Abu-Shakra M, Gladman DO, Urowitz MB: Malignancy in systemic lupus erythematosus. Arthritis Rheum 39: 1050--1054, 1996. 332. Mellemkjaer L, Andersen V, Linet MS, et al: Non-Hodgkin's lymphoma and other cancers among a cohort of patients with systemic lupus erythematosus. Arthritis Rheum 40: 761-768, 1997. 333. Chen YH, Lan JL: Kikuchi disease in systemic lupus erythematosus: clinical features and literature review. J Microbiol Immunol Infect 31: 187-192, 1998. 334. Martin F, Lauwerys B, Lefebvre C, et aI: Side-effects of intravenous cyclophosphamide pulse therapy. Lupus 6: 254-257, 1997. 335. Eberhard BA, Laxer RM, Eddy AA, Silverman ED: Presence of thyroid abnormalities in children with systemic lupus erythematosus. J Pediatr 119: 277-279, 1991. 336. Miller FW, Moore GF, Weintraub BD, Steinberg AD: Prevalence of thyroid disease and abnormal thyroid function test results in patients with systemic lupus erythematosus. Arthritis Rheum 30: 1124-1131, 1987. 337. Gazarian M, Laxer RM, Kooh S, Silverman ED: Hypoparathyroidism associated with systemic lupus erythematosus. .I Rheumatol 22: 2156--2158, 1995, 338. BenekH M, Savas MC, Erdem Y, et al: Primary hyperparathyroidism in a patient with systemic lupus erythematosus-antiphospholipid syndrome. Nephron 79: 215-216, 1998. 339. Fmman LS: Diabetes mellitus, islet-cell antibodies and HLA-B8 in a patient with systemic lupus erythematosus. Am J Dis Child 131: 1252-1254, 1977. 340. Ortiz-Alvarez 0, Metzger DL, Malleson PN, et al: Addison's disease in a child with systemic lupus erythematosus. Pediatr Rheumatol Online Journal 2: 134-138, 2004. 341. Koren S, Hanly JG: Adrenal failure in systemic lupus erythematosus. J Rheumatol 24: 1410--1412, 1997. 342. Gardner DL: Systemic lupus erythematosus. In Gardner OJ (ed): Pathology of the Connective Tissue Diseases, 2nd ed. Baltimore, Williams & Wilkins, 1965, p 144. 343. Donadio]V Jr: Renal involvement in SLE: the argument for aggressive treatment. In Bacon PA, Hadler NM (eds): The Kidney and Rheumatic Disease. Boston, Butterworth Scientific: 45, 1982, 344. Sorof JM, Perez MD, Brewer ED, et al: Increasing incidence of childhood class V lupus nephritis. J Rheumatol 25: 1413-1418, 1998. 345. Ginzler EM, Nicastri AD, Chen CK et al: Progression of mesangial and focal to diffuse lupus nephritis. N Engl J Med 291: 693-696, 1974. 346. Morel-Maroger 1: The course of lupus nephritis: contribution of serial renal biopsies. Adv Nephrol 6: 79-118, 1976. 347. Zinamerman SW, Jenkins PG, Shelf WO, et al: Progression from minimal or focal to diffuse proliferative lupus nephritis. Lab Invest 32: 665-672, 1975. 348. Balow .IE, Austin HA 3rd, Muenz LR, et al: Effects of treatment on the evolution of renal abnormalities in lupus nephritis. N Engl J Med 311: 491-495. 1984. 349. Grande JP, Balow JE: Renal biopsy in lupus nephritis. Lupus 7: 611-617, 1998, 350. Brentjens FR, Sepulveda M, Baliah T, et al: Interstitial inamune complex nephritis in patients with systemic lupus erythematosus. Kidney lnt 7: 342-350, 1975, 351. Fortenberry JD, Kenney RO: Distal renal tubular acidosis as the initial manifestation of systemic lupus erythematosus in an adolescent. J Adolesc Health 12: 148-151, 1992. 352. Daniel L, Sichez GH, Giorgi R, et al: Tubular lesions and tubular cell adhesion molecules for the prognosis of lupus nephritis. Kidney lnt 60, 2215-2221, 2001, 353. Malleson PN: The role of the renal biopsy in childhood onset systemic lupus erythematosus: a viewpoint. Clin Exp RheumalOl 7: 563-566, 1989.
C HAP T E R 354. Woolf A, Croker B, Osofsky SG, Kredich DW: Nephritis in children and young adults with systemic lupus erythematosus and normal urinary sediment. Pediatrics 64: 678-685, 1979. 355. Weis LS, Pachman LM, Potter EV, et al: Occult lupus nephropathy: a correlated light, electron and immunofluorescent microscopic study. Histopathology 1: 401-419, 1977. 356. Mahajan SK, Ordonez NG, Feitelson Pi, et al: Lupus nephropathy without clinical renal involvement. Medicine (Baltimore) 56: 493-501, 1977. 357. Leehey D], Katz AI, Azaran AH, et al: Silent diffuse lupus nephritis: long-term follow-up. Am] Kidney Dis 2: 188-196, 1982. 358. Bennett WM, Bardana E], Norman D], et al: Natural history of "silent" lupus ~ephritis. Am] Kidney Dis 1: 359-363, 1982. 359. Magil AB, Bailon HS, Chan V, et al: Diffuse proliferative lupus glomerulonephritis: determination of prognostic significance of clinical, laboratory and pathologic factors. Medicine (Baltimore) 63: 210-220, 1984. 360. Stamenkovic I, Favre H, Donath A, et al: Renal biopsy in SLE irrespective of clinical findings: longterm follow up. Clin Nephrol 26: 109-115, 1986. 361. Baldwin DS, Gluck MC, Lowenstein ], GaUD GR: Lupus nephritis: clinical course as related to morphologic forms and their transitions. Am] Med 62: 12-30, 1977. 362. Garin EH, Donnelly WH, Fennell RS III, Richard GA: Nephritis in systemic lupus erythematosus in children. ] Pediatr 89: 366-371, 1976. 363. Rush Pi, Baumel R, Shore A, et al: Correlation of renal histology with outcome in children with lupus nephritis. Kidney Int 29: 1066-1071, 1986. 364. Ty A, Fine B: Membranous nephritis in infantile systemic lupus erythematosus associated with chromosomal abnormalities. Clin Nephrol 12: 137-141, 1979. 365. Grossman], Schwartz RH, Callerame ML, Condemi]J: Systemic lupus erythematosus in a 1-year-old child. Am] Dis Child 129: 123-125, 1975. 366. Wertheimer D, Barland P: Clinical significance of immune deposits in the skin in SLE. Arthritis Rheum 19: 1249-1255, ]976. 367. Davis BM, Gilliam .IN: Prognostic significance of subepidermal immune deposits in uninvolved skin of patients with systemic lupus erythematosus: u lO-year longitudinal study. ] Invest Dermatol 83: 242-247, 1984. 368. Mandell BF: Pericardial effusions in patients with systemic lupus erythematosus. Arthritis Rheum 36: 1029-1030, 1993. 369. Shapiro RF, Gamble CN. Wiesner KB, et al: Immunopathogenesis of LibmanSacks endocarditis: assessment by light and immunofluorescent microscopy in two cases. Ann Rheum Dis 36: 508-516, 1977. 370. Johnson RT, Richardson EP: The neurological manifestations of systemic Itlpus erythematosus: a clinical-pathological study of 24 cases and review of the literature. Medicine (Baltimore) 47: 337-369, 1968. 371. Hanly]G, Walsh NMG, Sangalang V: Brain pathology in systemic lupus erythematosus.] Rheumatol 19: 732-741, 1992. 372. I!'hamashta MA, Gil A, Anciones B, et al: Chorea in systemic lupus erythelilatosus: association with antiphospholipid antibodies. Ann Rheum Dis 47: 681--683, 1988. 373. Aronson AJ, Ordonez NG, Diddie KR, Ernest ]T: Immune-complex depositIon in the eye in systemic lupus erythematosus. Arch Intern Med 139: 1312-1313, 1979. 374. Runyon BA, LaBrecque DR, Anuras S: The spectrum of liver disease in systemic lupus erythematosus: report of 33 histologically-proved cases and review of the literature. Am] Med 69: 187-194, 1980. 375. Asherson RA. Khamashta MA, Hughes GRV: The hepatic complications of the antiphospholipid antibodies. Clin Exp Rheumatol 9: 341-344, 1991. 376. Eigner W: The use of laboratory tests in the diagnosis of SLE. ] Clin Pathol 53: 424-432, 2000. 377. Zein N, Ganuza C, Kushner I: Significance of serum C-reactive protein elevation in patients with systemic lupus erythematosus. Arthritis Rheum 22: 7-12, 1979. 378. Maury CP, Helve T, Sjoblom C: Serum beta 2-microglobulin, sialic acid, and C-reactive protein in systemic lupus erythematosus. Rheumatol Int 2: 145-149, 1982. 379. Ter Borg E, Horst G, Limburg p. et al: C-reactive protein levels during diselise exacerbations and infections in systemic lupus erythematosus: a prospective longitudinal study.] Rheumatol 17: 1642-1648, 1990. 380. Morley]], Kushner [: Serum C-reactive protein levels in disease. Ann N Y Acad Sci 389: 406-418, 1982. 381. Moutsopoulos HM, Mavridis AK, Acritidis NC, Avgerinos PC: High C-reactive protein response in lupus polyarthritis. Clin Exp Rheumatol I·. 53-55, 1<)83 382. Spronk PE, ter Borg ~, Kallenberg CG: Patients with systemic lupus erythem,llosus and ]accoud's arthropathy: a clinical subset with an increased C-reactive protein response? Ann Rheum Dis 51: 358-361, 1997. 383. Nishiya K, Hashimoto K: Elevation of serum ferritin levels as a marker for atit.ive systemic lupus erythematosus. Clin Exp RheumatoJ 15: 39-44, 1997. 384. B(lwyer SL, Ragsdale CG, Sullivan DB: Factor VIII related antigen and childhood rheumatic diseases.] Rheumatol 16: 1093-1097, 1989. 385. Lacki ]K, Leszczynski P, Kelemen], et al: Cytokine concentration in serum of lupus erythematosus patients: the effect on acute phase response. ] Med 2H: 99-107, 1997. 386. C,>stello C, Abdelaal M, Coomes EN: Pernicious anemia and systemic lupus erythematosus in a young woman.] Rheumatol 12: 798-799, 1985. 387. RiVero S], Diaz-]ouanen E, Alarcon-SegOVia D: Lymphocytopenia in systemic lupus erythematosus. Arthritis Rheum 21: 295-305, 1978.
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
389
388. Bajaj SP, Rapaport SI, Fierer DS, et al: A mechanism for the hypoprothrombinemia of the acqUired hypoprothrombinemia-lupus anticoagulant syndrome. Blood 61: 684--{)92, 1983. 389. Arnout J: Antiphospholipid syndrome: diagnostic aspe':ts of lupus anticoagulants. Thromb Haemostasis 86: 83-91, 2002. 390. Johns AS, Charnley L, Ockelford PA, et al: Comparison of tests for the lupus anticoagulant and antiphospholipid antibodies in systemic lupus erythematosus. Clin Exp Rheumatol 12: 523-526, 1994. 391. Lehman 1], Hanson V, Singsen BH, et al: The role of antibodies directed against double-stranded DNA in the manifestations of systemic lupus erythematosus in childhood.] Pediatr 96: 657--Q61, 1980. 392. Morimoto C, Sano H, Abe T, et al: Correlation between clinical activity of systemic lupus erythematosus and the amounts of DNA in DNA/anti-DNA antibody immune complexes.] Immunol 129: 1960-1965, 1982. 393. Miller]] III, Hsu YP, Osborne CL, et al: Comparison of three assays for antiDNA with three assays for the measurement of the role of complement in systemic lupus erythematosus in adolescents.] Rheumatol 7: 665-670, 1980. 394. Hahn BH: Mechanisms of disease: antibodies to DNA. N Engl ] Med 338: 1359-1368, 1998. 395. Schiffer LE, Hussain N, Wang X, et al: Lowering anti-dsDNA antibodieswhat's new? Lupus 11: 885-894, 2002. 396. lang YJ, Stollar BD: Anti-DNA antibodies: aspects of structure and pathogeneicity. Cell Mol Life Sci. 60: 309-320, 2003. 397. Worrall ]G, Snaith ML, Batchelor JR, Isenberg DA: SLE: A rheumatological view: analysis of the clinical features, serology and immunogenetics of 100 SLE patients during long-term follow-up. Q] Med 74; 319-330, 1990. 398. Reichlin M: Antibodies to ribonuclear proteins in systemic lupus erythematosus. Rheum Dis Clin North Am 20: 29-43, 1994. 399. Wahten M, Tengner P, Gunnaisson I, et al: RO/SS-A and La/S5-B antibody level variation in patients with Sjogren's and systemic lupus erythematosus. ] Aut.oimmun 11: 29-38, 1998. 400. Wasicek GA, Reichlin M: Clinical and serological differences between systemic lupus erythematosus patients with antibodies to Ro versus patients with antibodies to Ro and La. ] Clin Invest 69: 835-843, 1982. 401. Wechsler HL, Stavrides A: Systemic lupus erythematosus with anti-Ro antibodies: clinical, histologic and immunologic findings. ] Am Acad Dermatol 6: 73-83, 1982. 402. Harley]B, Gaither KK: Autoantibodies. Rheum Dis Clin North Am 14: 43-56, 1988. 403. Maddison Pi, Provost IT, Reichlin M: Serological fmdings in patients with "ANAnegative" systemic lupus erythematosus. Meditine (Baltimore) 60: 87-94, 1981. 404. Gillespie ]P, Lindsley CB, Linshaw MA, Richardson WP: Childhood systemic lupus erythematosus with negative antinuclear antibody test. ] Pediatr 98: 578-581, 1981. 405. Taylor PV, Scott is, Gerlis LM, et al: Maternal autoantibodies against fetal cardiac antigens in congenital complete heart block. N Engl ] Med 315: 667-672, 1986. 406. Williams DG, Sharpe NG, Wallace G, Latchman DS: A repeated proline-rich sequence in B/B and N is a dominant epitope recognized by human and murine autoantibodies.] Autoimmun 3: 715-725, 1990. 407. Barada FA ]r, Andrews BS, David]S IV, Taylor RP: Antibodies to Sm in patients with systemic lupus erythematosus: correlations of Sm antibody titers with disease activity and other laboratory parameters. Arthritis Rheum 24: 1236-1244, 1981. 408. Beaufils M, Kouki F, Mignon F, et al: Clinical significance of anti-Sm antibodies in systemic lupus erythematosus. Am] Med 74: 201-205, 1983. 409. Gulko PS, Reveille ]D, Koopman W], et al: Survival impact of autoantibodies in systemic lupus erythematosus.] Rheumatol 21: 224-228, 1994. 410. Arnett FC, Hamilton RG, Roebber MG, et al: Increased frequencies of Sm and nRNP autoantibodies in American blacks compared to whites with systemic lupus erythematosus.] Rheumatol 15; 1773-1776, 1988. 411. Sharp GC, Irvin WS, Tan EM, et al: Mixed connective tissue disease: an apparently distinct rheumatic disease syndrome associated with a specific antibody to an extractable nuclear antigen. Am.r Med 52: 148-159, 1972. 412. Fritzler I\ij, Tan EM: Antibodies to histone in drug-induced and idiopathic lupus erythematosus. ] Clin Invest 62: 560-567, 1978. 413. Fritzler M], Ryan ]P, Kinsella TO: Clinical features of SLE patients with antihistone antibodies. ] Rheumatol 9: 46-51, 1982. 414. Portanova ]P, Arndt RE, Tan EM, Kotzin BL: Anti-histone antibodies in idiopathic and drug-induced lupus recognize distinct intrahistone regions. ] Immunol 138: 446-451, 1987. 415. Rubin RL,]oslin GF, Tan EM: Specificity of antihistone antibodies in systemic lupus erythematosus. Arthritis Rheum 25: 779-782, 1982. 416. Petty RE, Palmer NR, Cassidy]T, et al: The association of autoimmune diseases and anti-lgA antibodies in patients with selective 19A deficiency. Clin Exp Immunol 37: 83-88, 1979. 417. Abrass CK, Nies KM, Louie is, et al: Correlation and predictive accuracy of circulating immune complexes with disease activiry in patients with systemic lupus erythematosus. Arthritis Rheum 23: 273-282, 1980. 418. Gauthier YJ, Emlen W: Immune complexes in systemic lupus erythematosus. In Hahn BH, Wallace D] (eds); Dubois' Lupus Erythematosus, 5th ed. Baltimore: Williams & Wilkins, 1997, p 207. 419. Buyon ]P, Tamerius ], Belmont HM, Abramson SB: Assessment of disease activity and impending flare in patients with systemic lupus erythematosus. Arthritis Rheum 35: 1028-1037, 1992.
390
C HAP T E R
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
420. Garin EH, Donnelly WH, Shulman ST, et al: The significance of serial measurements of serum complement C3 and C4 components and DNA ninding capacity in patients with lupus nephritis. Clin Nephrol 12: 148-155, 1979. 421. Sliwinski AJ, Zvaifler NJ: Decreased synthesLs of the third component of complement in hypocomplementemic systemic lupus erythematosus. Clin Exp Immunol 11: 21-29, 1972. 422. Petri M, Genovese M, Engle E, Hochberg MC: Definition, incidence, and clinical description of flare in systemic lupus erythematosus: a prospective cohort study. Arthritis Rheum 34: 937-944, 1991. 423. Bombardier C, Gladman DO, Urowitz M, et al: Derivation of the SLEDAJ: a disease activity index for lupus patients. Arthritis Rheum 35: 630--640, 1992. 424. Liang MH, Socher SA, Larson MG, SChur PH: Reliability and validity of six systems for the clinical assessment of disease activity in systemic lupus eryt hematosus. Artluitis Rheum 32: 1107-1118, 1989. 425. Symmons DPM, Coppock JS, Bacon PA, et al: Development and assessment of a computerized index of clinical disease activity in systemic lupus erythematosus. Q J Med 68: 927-937, 1988. 426. Petri M, Hellmann 0, Hochberg M: Validity and reliability of lupus activity measures in the routine clinic setting. J Rheumatol 19: 53-59, 1992. 427. Hawker G, Gabriel S, Bombardier C, et al: A reliability study of SLEDAI: a dist'ase activity index for systemic lupus erythematosus. J Rheumatol 20: 657~60, 1993. 428. Merino R, Mendez S, Garcia-Consuegra, Madero R: Evaluation of two disease activity indices of systemic lupus erythematosus in pediatric patients. Clin Exp Rheumatol 13: 680--681, 1995. 429. Brunner HI, Feldman BM, Bombardier C, Silverman ED: Sensitivity of the Systemic Lupus Erythematosus Disease ActiVity Index, British Isles Lupus A"essment Group Index and Systemic Lupus Activity measure in the evaluation of clinical change in childhood-onset systemic lupus erythematosus. Arthritis Rheum 42: 1354-1360, 1999. 430. Sibley JT, Olszynski WP, Decoteau WE, Sundaram MB: The incidence and prognosis of central nervous system disease in systemic lupus erythematosus. J Rheumatol 19: 47-52, 1992. 431. Gladman DO, Ginzler E, Goldsmith C, et al: The development and initial validation of the Systemic LUpus International Collaborating Clinics/American College of Rheumatology Damage Index for systemic lupus erythematosus. Arthritis Rheum 39: 363-369, 1996. 432. Brunner HI, Silverman ED, Bombardier C, et al: Disease activity and steroids are risk factors for damage, while second line agents are protective in childhood-onset systemic lupus erythematosus. Arthritis Rheum 43 (SupplJ S171, 2000. 43.~. Stoll T, Gordon C, Seifert B, et al: Consistency and validity of patient administered assessment of quality of life by the MaS SF-36: its association with disease activity and damage in patients with systemic lupus erythematosus. J Rheumatol 24: 1608-1614, 1997. 434. Rihner M, McGrath J Jr: Fluorescent light photosensitivity in patients with systemic lupus erythematosus. Arthritis Rheum 35: 949-952, 1992. 435. Carreno L, Lopez-Longo FJ, Gonzalez CM, Monteagudo I: Treatment options for juvenile-onset sy'temic lupus erythematosus. Pediatr Drugs 4: 241-256, 2002. 436. Ostensen M, Villiger PM: Non-steroidal anti-inflammatory drugs in systemic lupus erythematosus. Lupus 10: 135-139, 2001. 437. Mandell BF, Raps ED: Severe systemic hypersensitiVity reaction of ibuprofen occurring after prolonged therapy. Am J Med 82: 817-820, 1987. 438. Weksler BB, Lehany AM: Naproxen-induced recurrent aseptic meningitis. DICP 25: 1183-1184, 1991. 439. D'Cruz 0: Antimalarial thaerapy: a panacea for mild lupus? Lupus 10: 148-151,2001. 440. The Canadian Hydroxychloroquine Study Group: A randomized study of the effect of Withdrawing hydroxychloroquine sulfate in systemic lupus erythematosus. N Engl J Med 324: 150-154, 1991. 441. Tsakonas E, Joseph L, Esdaile JM, et al: A long-term study of hydroxychloroquine withdrawal on exacerbations of systemic lupus erythematosus. Lupus 7: 80-85, 1998. 442. Wallace DJ, Metzger AL, Stecher VJ, et al: Cholesterol-lowering effect of hydroxychloroquine in patients with rheumatic disease: reversal of deleterious effects of steroids on lipids. Am J Med 89: 322-326, 1990. 443. Hodis HN. Quismorio FP Jr, Wickham E, Blankenhorn DH: The lipid, lipoprotein, and apolipoprotein effects of hydroxychloroquine in patients with systemic lupus erythematosus. J Rheumatol 20: 661~65, 1993. 444. Petri M: Hydroxychloroquine use in the Baltimore lUpus cohort: effects on lipids, glucose and thrombosis. Lupus 5 (Suppl 1): Sl6-S22, 1996. 445. Laaksonen A-L, Koskiahde V, Juva K: Dosage of antimalarial drugs for children with juvenile rheumatOid arthritis and systemic lUpus erythematosus: a clinical study with determination of serum concentration of chloroqUine and hydroxychloroquine. Scand J Rheumatol 3: 103-108, 1974. 446. Chatham WW, Kimberly RP: Treatment of lupus with corticosteroids. Lupus 10: 140-147, 2002. 447. Felson DT, Anderson J: Evidence for the superiority of immunosuppressive drugs and prednisone over prednisone alone in lupus nephritis: results of a pooled analysis. N Engl J Med 311: 1528-1533, 1984. 448. Abu-Shakra M, Shoenfeld Y: Azathioprine therapy for patients with systemic lupus erythematosus. Lupus 10: 152-153, 2001. 449. Silverman E: What's new in the treatment of pediatric SLE. J Rheumatol 2.~: 16';7-1660, 1996.
450. Silverman ED, Eddy AA: Systemic lupus erythematosus in children. In Maddison PJ, Isenberg DA, Woo P, Glass ON (eds): Oxford Textbook of Rheumatology, 2nd ed. Oxford, Oxford Medical, 1998, p 1190. 451. Nossent HC, Koldingsnes W; Long-term efficacy of azathioprine treatment for proliferative lupus nephritis. Rheumatology 39: 969-974, 2000. 452. Tucker LB: Controversies and advances in the management of ,ystemic lUpus erythematosus in children and adolescents. Best Pract Res Clin Rheumatol 16: 4712-480, 2002. 453. Takada K, mei GG, Boumpas DT: Cyclophosphamide for the treatment of systemic lupus erythematosus. Lupus 10: 154-161, 2001. 454. Donadio]V Jr. Holley KE, Ferguson RH, llstrup OM: Treatment of diffuse proliferative lupus nephritis with prednisone and combined prednisone and cyclophosphamide. N Engl J Med 299: 1151-1155, 1978. 455. Steinberg AD, Decker JL: A double-blind controlled trial comparing cyclophosphamide, azathioprine and placebo in the treatment of lupus glomerulonephritis. Arthritis Rheum 17: 923-937, 1974. 456. Steinberg AD, Steinberg SC: Long-tenn preservation of renal function in patient' with lupus nepluitis receiving treatment that includes cyclophosphamide versus those treated with prednisone only. Arthritis Rheum 34: 945-950, 1991. 457. Balow JE, Austin HA, Tsokos ac, et al: NIH Conference: Lupus nephritis. Ann Intern Med 106: 79-94, 1987. 458. Boumpas DT, Austin HA, Vaughn EM, et al: Controlled trial of pulse methylprednisolone versus two regimens of pulse cyclophosphamide in severe lupus nephritis. Lancet 340: 741-745, 1992. 459. Lehman 1]: A practical gUide to systemic lupus erythematosus. Pediatr Clin North Am 42: 1223--1238, 1995. 460. Lehman TJA, Onel K: Intermittent intravenous cyclophosphamide arrests progression of the renal chronicity Index in childhood systemic lupus erythematosus. J Pediatr 136: 243-247, 2000. 461. Neuwelt MC, Lacks S, Kaye BR, et al: Role of intravenous cyclophosphamide In the treatment of severe neuropsychiatric systemic lupus erythematosus. Am J Med 98: 32--41, 1995. 462. Slater CA, Liang MH. McCune JW, et al: Preserving ovarian funciion in patient' receiving cyclophosphamide. Lupus 8: 3-10, 1999. 463. Rothenberg RJ, Graziano RM, Grandone]T. et al: The use of methotrexate in steroid-resistant systemic lupus erythematosus. Arthritis Rheum 31: 612~i';, 1988. 464. Abud-Mendoza C, Sturbaum AK, Vazquez-Compean R, Gonzalez-Amaro R: Methotrexate therapy In childhood systemic lupus erythematosus. J Rheumatol 20: 731-733, 1993. 465. Ravelli A, Ballardini G, Viola S, et al: Methotrexate therapy in refractory pediatric onset systemic lupus erythematosus. J Rheumatol 25: 572-575, 1998. 466. Sato EI: Methotrexate therapy in systemic lupus erythematosus. Lupus 10: 162-164, 2001. 467. Valesini G. Priori R, Francia A, et al: Central nervous system involvement In systemic lupus erythematosus: a new therapeutic approach with intrathecal dexamethasone and methotrexate. Springer Semin Immunopathol 16: 313-321 1994. 468. Lehman TJ, Edelheit BS, Onel KB: Comnined intravenous methotrexate and cyclcophosphamide for refractory childhood lupus nephrtitls. Ann Rheum Dis 63: 321-323, 2004. 469. Feutren G, Querin S, Noel LH, et al: Effects of cyclosporine in severe systemic lupus erythematosus. J Pediatr 111: 1063-1068, 1987. 470. Fu LW, Yang LY, Chen WP, Lin CY: Clinical etficacy of cyclosporin A (NeOl'alJ in the treatment of paediatric lUpus nephritis with heavy proteinuria. Br J Rheumatol 37: 217-221, 1998. 471. Caccavo 0, Lagana B. Millerhofer AP, et al: Long-term treatment of systemic lupus erythematosus with cyclosporin A. Arthritis Rheum 40: 27-35, 1997. 472. Griffiths B, Emery P: the treatment of lupus with cyclosporine A. Lupus 10: 165-170, 2001. 473. Chan TM, Li FK, Tang CS, et al: Efficacy of mycophenolate mofetil in patients with diffuse proliferative lupus nephritis. N EnglJ Med 343: 1156-1162.2000. 474. Austin HA, Balow ]E: Treatment of lupus nephritis. Semin Nephrol 20: 265-276, 2000. 475. Ginzler EM, Aranow C, Buyon J, et al. A multicenter sUldy of myocophenolate mofetil (MMF) vs intravenous cyclophosphamide (IVC) as induction therapy for severe lupus nephritis (LN): preliminary results. Arthritis Rheum 48 (Suppl) s647 , 2003. 476. Adu 0, Cross J, Jayne DRW: Treatment of ,ystemic lupus erythematosus with myocphenolate mofetil. Lupus 10: 203-208, 2001. 477. Akashl K, Nagasawa K, Mayumi T, et al: Successful treatment of refractOly systemic lupus erythematosus with intravenous immunoglobulins. J Rheumatol 17: 375-379, 1990. 478. Winder A, Molacl Y, Ostfeld I. et al: Treatment of systemic lupus erythematosus by prolonged administration of high dose intravenous immunoglobulin: report of 2 cases. J Rheumatol 20: 495-498, 1993. 479. Cohen MG. Li EK: Limited effects of intravenous IgG in u'eating systemic lupus erythematosus-aSSOCiated thrombocytopenia. Arthritis Rheum 34: 787-788. 1991. 480. ter Borg EJ, Kallenberg CGM: Treatment of severe thrombocytopenia in systemic lupus erythematosus with intravenous gammaglobulin. Ann Rheum Dis 51: 1149-1151, 1992. 481. Schroeder JO, Zeuner RA, Euler HH, Loffler H: High dose intravenous immunoglobulins in systemic lupus erythematosus: clinical and serological results of a pilot study. J Rheumatol 23: 71-75, 1996.
C HAP T E R 482. Rauova L, Lukac), Levy Y, et al: High-dose intravenous immunoglobulins for lupus nephritis-a salvage immunomodulation. Lupus 10: 209-213, 2001. 483. l.afferty TE, Smith )B, Schuster S), DeHoratius R]: Treatment of acquired factor VIII inhibitor using intravenous immunoglobulin in two patients with systemic lupus erythematosus. Arthritis Rheum 40: 775-778, 1997. 484. Wallace D): Apheresis for lupus erythematosus: state of the art. Lupus 10: 193-196, 2001. 485. Hanly)G, Hong C, Zayed E, et al: Immunomodulating effects of synchronised plasmapheresis and intravenous bolus cyclophospha mide in systemic lupus erythematosus. Lupus 4: 457-463, 1995. 486. Llorente L. Richaud-Patin Y, Garcia-Padilla C, et al. Clinical and biologic effects of anti-interleukin-10 monoclonal antibody administration in systemic lupus erythematosus. Arthritis Rheum 43: 1790-1800, 2000. 487. Saito K, Nawata M, Nakayamada S, et al: Successful treatment with anti-CD10 monoclonal antibody (rituximab.l of life-threatening refractory systemic lupus erythematosus with renal and central nervOus system involvement. I.upus 12: 798-800, 2003. 488. Weide R. Heymanns), Pandorf A, Koppler H: Successful long-term treatment of systemic lupus erythematosus with ritxuimab maintenance therapy. Lupus 12: 779--782, 2003. 489. Eisenberg R: SLE-Rituximab in lupus. Arthritis Res Ther 5: 157-159, 2003. 490. Anolik), Sanz I, Looney R]: B cell depletion therapy in systemic lupus erythematosus. Curr Rheumato! Rep 5: 350-3456, 2003. 491. Strand V: Monoclonal antibodies and other biologic therapies. Lupus 10: 21&-221, 2001. 492. (:alabrese L, Fleischer AB: Thalidomide: current and potential clinical appli'·ations. Am) Med 108: 487-495, 2000. 493. Karim MY, Ruiz-lrastorza G, Khamashta MA, Hughes GRV: Update on therapy-thalidomide in the treatment of lupus. Lupus 10: 188-192, 2001. 494, Jl;pobler RM, Graninger W, et al: Extracorporeal photochemotherapy for the treatment of systemic lupus erythematosus, Arthritis Rheum 35: 319--324, 1992. 495. I~un RK, Traynor AE, Pope R, et al: Treatment of autoimmune disease by intense immunosuppressive conditioning and autologous hematopoietic stem cell transplantation. Blood 92: 3505-3514, 1998. 496, Trysberg E, Lindgren I, Tarkowski A: Autologous stem cell transplantation in ~ case of treatment-resistant central nervous system lupus. Ann Rheum Dis 59: 23&-238, 2000. 497, Wulfraat NM, Sanders EAM, Kamphuis SSM, et al: Prolonged remission with<>ut treatment after autologous stem cell transplantation for refratory childhood systemic lupus erythematosus. Arthritis Rheum 44: 728-731. 2001. 498, Tyndall A: Immunoablation and haemopoietic stem cell transplantation fOr sclvere autoimmune disease with special reference to systemic lupus erythematosus. Lupus 10: 214-215, 2001. 499, Furie RA, Cash )M, Cronin ME, et al: Treatment of systemic lupus erythematosus with L)P 394. ) Rheumatol 28: 257-265, 2001. 500, Ruiz-Irastorza G, Khamashta MA, Hughes GRV: Antiaggregant and anticoagulant therapy in systemic lupus erythematosus and Hughes' syndrome. IJupus 10: 241-245, 2001. 501. ~'dW DG, Bounameaux H, de Moerloose P, Sarasin FP: Prophylactic :/ntithrombotic therapy for patients with systemic lupus erythematosus with or without antiphospholipid antibodies, Arch Intern Med 160: 2042-2048, 2000. 502, Ruiz-Irastorza G, Cuadrado M) Mujic F, Taub NA, Hunt B), Hughes GRV: The n1anagement of thrombosis in the antiphospholipid-antibody syndrome, iii Engl) Med 332: 993-997, 1995, 503. Baca v, Lavalle C, Garcia R, et al: Favorable response to intravenous methylprenisolone and cyclophosphamide in children with severe neuropsychiatric lupus. ) Rheumatol 26: 432-439, 1999. 504. KClein M, Radhakrishnan ], Appel G: Cyclosporine treatment of glomerular diseases. Annu Rev Med 50: 1-15, 1999. 505, Gipson DS, Ferris ME, Dooley MA, et al: Renal transplantation in children with lupus nephritis. Anl] Kidney Dis 41: 455-463, 2003. 506. Bartosh SM, Fine RN, Sullivan EK: Outcome after transplantation of young patients with systemic lupus erythematosus: a report of the North American pediatric renal transplant cooperative study. Transplantation 72: 973-978, 2001.
16
SYSTEMIC
Lupus
ERYTHEMATOSUS
391
507. Baqi N, Tejani A. Recurrence of the original disease in pediatric renal transplantation.) Nephrol 10: 85-92, 1997. 508. Urowitz MB, Kagal A, Rahman P, Gladman DD: Role of specialty care in the management of patients with systemic lupus erythematosus. ) Rheumatol 29: 1207-1210, 2002. 509. Wang LC, Yang YH, Lu MY, Chiang BL: Retrospective analysis of mortality and morbidity of pediatric systemic lupus erythematosus in the past two decades. ) Microbiol Immunol Infect 36: 203-208, 2003. 510. Lacks S, White P: Morbidity associated with childhood systemic lupus erythematosus.) Rheumatol 17: 941-945, 1990. 511. Platt )L, Burke BA, Fish A), et al: Systemic lupus erythematosus in the first two decades of life. Am) Kidney Dis 2: 212-222, 1982. 512. Gladman DD, Chaudhry-Ahluwalia V, Ibanez 0, et al: Outcomes of symptomatic osteonecrosis in 905 patients with systemic lupus erythematosus. ) Rheumatol 28: 222&-2229, 2001. 513. Fish A], Blau EB, Westberg NG, et al: Systemic lupus erythematosus within the first two decades of life. Am) Med 62: 99--117, 1977. 514. Abeles M, Urman )D, Weinstein A, et al: SLE in the younger patient: survival studies.) Rheumatol 7: 515-522, 1980. 515. Wallace D), Podell T, Weiner ), et al: Systemic lupus erythematosus: survival patterns-experience with 609 patients. )AMA 245: 934-938, 1981. 516. Glidden RS, Mantzouranis EC, Borel Y: Systemic lupus erythematosus in childhood: clinical manifestations and improved survival in fifty-five patients. Clin Immunol Immunopathol 29: 19&-210, 1983. 517. Dumas R: Lupus nephritis: collaborative study by the French Society of Paediatric Nephrology. Arch Dis Child 60: 126-128, 1985. 518. Baqi N, Moazami S, Singh A, et al: Lupus nephritis in children: a longitudinal study of prognostic factors and therapy. ) Am Soc Nephrol 7: 924-929, 1998. 519. Yang LY: Lupus nephritis in children: a review of 167 patients. Pediatrics 94: 335-340, 1994. 520. Harisdangkul V, Nilganuwonge S, Rockhold L: Cause of death in systemic lupus erythematosus: a pattern based on age at onset. South Med) 80: 1249-1253, 1987. 521. Austin HA III, Muenz LR, Joyce KM, et al: Prognostic factors in lupus nephritis: contribution of renal histologic data. Am ) Med 75: 382-391, 1983. 522. Singh S, Devidaya L, Kumar L, )opshi K: Mortality patterns in childhood lupus-10 years' experience in a developing country. Clin Rheumatol 21: 462-465, 2002. 523. Tejani A, Nicastri AD, Chen CK, et al: Lupus nephritiS in black and Hispanic children. Am) Dis Child 137: 481-483, 1983. 524. Walsh S), DeChello LM: Geographical variation in monality from systemic lupus erythematosus in the United States. Lupus 10: 637-646, 2001. 525. Grossman L, Barland P: Histone reactivity of drug-induced antinuclear antibodies: a comparison of symptomatic and asymptomatic patients. Arthritis Rheum 24: 927-931, 1981. 526. Beernink DH, Miller )) 1II: Anticonvulsant-induced antinuclear antibodies and lupus-like disease in children.) Pediatr 82: 113-117, 1973527. Singsen BH, Fishman L, Hanson V: Antinuclear antibodies and lupus-like syndromes in children receiving anticonvulsants. Pediatrics 57: 529--534, 1976. 528. Irias )): HydralaZine-induced lupus erythematosus-like syndrome. Am) Dis Child 129: 862-864, 1975. 529. Akin E, Miller LC, Tucker LB: Mino(ycline-induced lupus in adolescents. Pediatrics 101: 926, 1998. 530. Finkel TH, Hunter D], Paisley)E, et al: Dmg-induced lupus in a child after treatment with zafirlukast (Accolate). ) Allergy Clin Immunol 103: 533-534, 1999. 531. Akachaisri T, Lehman 1JA: Systemic lupus erythematosus and related disorders of childhood. Cure Opin Rheumatol 11: 384-392, 1999. 532. McCurdy DK, Lehman T)A, Bernstein B, et al: Lupus nephritis: prognostic factors in children. Pediatrics 89: 240, 1992. 533. Kaslow RA, Masi AT: Age, sex, and race effects on mortality from systemic lupus erythematosus in the United States. Arthritis Rheum 21: 473, 1978.
( HAP T E R
17
NEONATAL
Lupus
ERYTHEMATOSUS
Earl D. Silverman, Dawn Spence, and Robert M. Hamilton
~ Neonatal lupus erythematosus (NLE) is a disease of the developing fetus and neonate defined by characteristic clinical features in the presence of specific maternally derived autoantibodies. The transplacental passage of these autoantibodies is necessary but not sufficient to cause the disease. The autoantibodies associated with NLE are directed against a group of small cytoplasmic and nuclear ribonucleoproteins and their associated RNAs, collectively referred to as RoRNP or Ro particle. The most common clinical manifestations of NLE are cardiac, dermatologic, and hepatic.
ETIOLOGY AND PATHOGENESIS Maternal autoantibodies directed against RoRNP or occasionally against other autoantigens are required for the development of NLE. However, many mothers with these autoantibodies do not deliver children with NLE. Other factors, including viral infection, maternal estrogens, apoptosis, and ultraviolet light, may have a role in pathogenesis. 1.2
Autoantlgens Anti-Ro and anti-La autoantibodies are directed against the RoRNP.3.4 There are at least two Ro proteins, a 6o-kD and a 52-kD polypeptide, and a single La protein (48 kD).3,S,6 The Ro and La proteins are closely associated, and both are usually complexed to small RNAs. The Ro and La proteins, rather than their associated RNAs, are the major antigenic targets of the autoantibodies.
La Protein The La protein is found in association with RNA polymerase III transcripts, which include small viral RNAs, UIRNA, and precursors of tRNA and 55 ribosomal RNA (55 rRNA).4,7-1O The La protein has an important role in the production of polymerase III RNA transcripts, can modulate 5' processing of pre-tRNAs, and therefore is important in controlling transcriptional and post-transcriptional events during the synthesis, maturation, and nuclear export of polymerase III transcripts, including the RNAs to which it binds. 1J- 14 La exists in at least two forms; one is associated with the Ro protein, and the other is
392
independent of Ro. 1s A structural basis for the association of Ro and La proteins has been demonstrated. 16 Proteins homologous to La exist in many species, including fruit flies (Drosophila melanogaster), frogs (Xenopus laevis), mosquitoes, and yeast (Saccharomyces cerevisiae), demonstrating the importance of La in normal cell function. 17- 19 There are species-specific inserts that may be of pathogenic significance in the development of anti-La antibodies and in the pathogenesis of NLE. 20 La is present in numerous nuclear and cytoplasmic complexes, where it acts as a molecular chaperone for polymerase III transcripts, functions as an RNA-folding protein, is important in the very early steps of U6snRNP assembly, and is important during the cell cycle by increasing histone protein production,21-24 La binds to, enhances translation of, and alters gene expression of several viral RNAs, including human parainfluenza virus type 3, poliovirus, and human immunodeficiency virus,25-2H and it alters the regulation of interferon-inducible protein kinase after viral infection. 29•3o Infection with poliovirus or adenovirus induces a relocation of La from the nucleus to the cytoplasm and cell surface. 31 ,3 2 The interaction of La with viruses and the subsequent alteration of the cellular distribution of La may permit maternally-derived autoantibodies to bind directly to their autoantigen target on the cell surface. Alternatively, La may be expressed on the cell surface as a result of apoptosis during normal fetal development. The La protein can shuttle between the nucleus and the cytoplasm, but it is mainly located in the nucleus. 32 During early development in X. laevis, there are extremely high levels of La mRNA that progressively and significantly decrease during gestation,18 More recently it has been demonstrated that cardiac mRNA levels of La are highest during the second trimester of gestation and then decline to adult levels near term. These observations may help explain why the developing rather than the adult heart is the target of autoantibodies in NLE. There are three functional La mRNA forms, which are upregulated and downregulated in paralle[,33,3-i In vivo, mRNA for exons 1 and l' have been identified in endothelial cells and liver cells but not in smooth muscle cells. Exon 1 resides mostly in the nucleus, whereas exon l' La mRNA is mainly in the cytoplasm. 35 Any of the La isoforms may be the target of the immune response in NLE.
C HAP T E R
17
NEONATAL
Lupus
ERYTHEMATOSUS
393
Ro Proteins
Autoantibodies
The function of the Ro proteins had been much more difficult to determine. The 60-kD Ro protein is important in maintaining production of functional 5S rRNA.36 This function of 60-kD Ro is in association with La, and the two proteins are either complexed together or bind in close proximity to each other. 37 The 52-kD Ro protein is mainly located in the eytoplasm,38 whereas 60-kD Ro is predominantly found in the nucleus, suggesting that the two Ro antigens likely have different cellular functions. Cell stress in vivo can alter the antigenic configuration and cell surface expression of 52-kD Ro, which wOl\ld then allow for direct tissue damage by anti-Ro antibodies.39 The 52-kD Ro protein has an alternatively spliced form referred to as 52~, whereas the more common protein in adult life is the 52a protein. The 52~ form is maximally expressed between 14 and 16 weeks' gestation, a time when the level of 52a expression is at its lowest. In fetal hearts by 22 to 25 weeks' gestation and in adult hearts, the 52~ transcript is markedly diminished, and 52a is the dominant form. These data suggest that the differential exptession of the 52-kD Ro isoforms may be important in the development of heart block because the maximal expression of 52~ occurs at the time of cardiac ontogeny, when maternal antibodies begin to cross the placental bartier. 40
In the mid-1980s, it was recognized that NLE was associated with maternal anti-Ro antibodies. 6B ,69 After the discovery that there were two major Ro proteins and one La protein, it became clear that the greatest risk for the development of congenital atrioventricular (AY) block (CAVB) was associated with maternal anti-52-kD Ro and anti-La antibodies. 7G--76 The risk of NLE was associated with higher mean maternal antibody titers to all three proteins. 71 ·77- 79 Cutaneous NLE (C-NLE) was associated with higher titers of maternal autoantibodies to these antigens than occur with CAVB. 7G--76.79,80 One study suggested that anti-La antibody production might be important in the development of CNLE, whereas anti-Ro antibody production might be more important in the development of CAVB. 81 Examination of the fine specificity of the repertoire of maternal anti-La antibodies allowed identification of antibodies directed against a small La polypeptide, named DD, that was found only in the sera of mothers of children with NLE and not in sera from mothers of unaffected children. 71 Although this finding was specific for NLE, it had only a 30% sensitivity because many mothers without anti-DD antibodies delivered children with NLE, and anti-DD antibodies were present in only 30% of children with NLE. Similarly, the fine specificity of the anti-Ro antibody response in mothers of children with NLE differs from the anti-Ro response of other women with rheumatic disorders. 82 Anti-52-kD Ro antibodies are more specific for the development of NLE than anti-60-kD Ro antibodies. The anti-Ro antibodies from mothers who delivered a child with CAVB preferentially recognized peptides of different amino acids on the 52-kD Ro protein (amino acids 200 to 239), including peptides containing the zinc finger (amino acids 1 to 135), than mothers with anti-Ro who delivered healthy children (preferentially recognized amino acids 176 to 196).83 Differences in the fine specificity of the immune response to the RoRNP may be important in fetal outcome. Further studies are required to determine the "at-risk" pregnancies and should examine the response to isoforms of the Ro and La proteins, the relative amount of which can vary during gestation. 8.33 .40,84 Although there still is controversy about whether calreticulin is a part of the RoRNP, autoantibodies directed against calreticulin are present in the sera from patients with autoimmune diseases. 41 .44,85 Although elevated anticalreticulin antibody levels have been reported in mothers of children with NLE compared with healthy normal subjects, when the anti-calreticulin response was compared with that of healthy pregnant women, no differences in the mean titer of these autoantibodies could be demonstrated. However, one report cited differences in anti-calreticulin antibody titers when compared with normal, healthy control subjects,86 The presence of other antigens on the fetal heart or placenta has been suggested to be important in the development, or protection from the development, of CAVB. There is cross-reactivity between a subset of
Caireticulin and RoRNP The most controversial issue regarding RoRNP is the relationship of calreticulin to the RoRNP. Some investigators have posited that calreticulin is part of RoRNP associated with the hY RNAs, whereas others disagree. 41 ,42 Calreticulin was initially described as a calcium-binding protein present in the endoplasmic reticulum. It is present in many animal species, including Leishmania,43 the filarial nematode Onchocerca volvulus (specifically the RAL1 protein),41.44 Caenorhabditis elegans,45 D. melanogaster,46 and x. laevisY Calreticulin has three structural domains, each with a different function. It is present on the cell membrane of neutrophils and may be shed during cytotoxic T cell activat/on, which permits calreticulin to be present extracellularly.4B.49 Although a major function of calreticulin is as a resident calcium-binding protein in the endoplasmic reticulum, it is also found outside of the endoplasmic reticulum in the nuclear envelope, the nucleus, and cytotoxic granules in T cells, and in vesicles of sperm cells. 50,51 Similar to other autoantigens, stress can increase gene expression of calreticulin. 52 ,53 In addition to calcium binding, calreticulin may have a role in regulating T cell activation,54 in controlling gene transcription (including the glucocorticoid receptor),55-{iO in modulating integrinmediated functions,61-
394
C HAP T E R
17 N EON ATAL L u PUS E R Y THE MAT 0
anti-La, but not anti-Ro, antibodies and laminin. 87,88 Sera from mothers of infants with CAVB may also contain antiendogenous retrovirus-3 (ERV-3) antibodies. Anti-La, anticalreticulin, and anti-ERV-3 antibodies can bind to the placenta or placental trophoblast or both.88-90 Binding of these autoantibodies to placental tissue may alter the quantity and repertoire of these autoantibodies in the fetal circulation and therefore affect binding to the target fetal tissue,89--91 Anti-Iaminin autoantibodies bind to fetal but not adult heart and cardiac tissue, and ERV-3 and laminin are maximally expressed between U and 17 weeks' gestation. 89,91 Cardiac laminin, calreticulin or ERV-3 may be a target for maternal autoantibodies, and direct binding to these fetal cardiac proteins may initiate or potentiate inflammation in the fetal heart. 88 ,89,91 Antibodies directed against a p57 recombinant protein and a-fodrin have been detected in the sera of mothers of infants with NLE, Anti-p57 antibodies were present in approximately one third of the sera from mothers of children with NLE and were almost always associated with anti-Ro antibodies,92 Anti-a-fodrin antibodies were initially reported to be present in sera from patients with Sjogren's syndrome but not in the sera from patients with rheumatoid arthritis or systemic lupus erythematosus (SLE),93 Preliminary data in NLE suggest that maternal anti-120-kD a-fodrin antibodies may be an additional serologic marker for the risk of development of NLE.94
GENETIC BACKGROUND Specific HLA-DR and DQ genes are important in the production of anti-Ro and anti-La antibodies, In patients with autoimmune diseases, DR3 was associated with anti-52kD Ro and anti-La autoantibodies but not anti-60 kD Ro antibodies lO1 ,102 This association is present in most but not all ethnic backgrounds and is independent of the presence of an autoimmune disease. I03-111 High-titer anti-Ro antibodies are associated with the DQw1/DQw2 heterozygote. ll2 ,m In mothers of children with NLE, the HLA antigen profile more closely resembles that present in patients with primary Sjogren's syndrome than that associated with patients with SLE.77,114 It is likely that extended haplotypes are more important than single loci in determining production of anti-Ro and anti-La antibodies. In patients of most ethnic backgrounds, high levels of anti-Ro and anti-La antibodies are associated with all or at least most of DRBl*0301, DQA1*0501, and DQB1*0201 extended haplotypes, whereas the DQA1*0501 allele is present in the other patients.104.lOS.115 Glutamine at position 34 of DQA1 and leucine at position 26 of DQB1 are associated with antiRo and anti-La antibodies, although the extended haplotype (in linkage disequilibrium) may have the strongest correlation with this autoantibody response. 1l6,l17 The genetic control of anti-RoRNP antibodies is further complicated by the demonstration that response to peptides of the 60-kD Ro protein may be under different genetic control and distinct from the HLA associations of the response to the complete 60-kD protein,u8
SUS
In Japanese mothers of children with NLE, the production of anti-Ro and anti-La antibodies was associated with the extended haplotypes DRB1*U01-DQA1*0501DQB1*0301 and DRB1*08032-DQA1*0103-DQB1*0601, as well as the individual alleles DRB1*U01, DRB1*08032, and DQB1*0301. l19 All of the anti-Ro and anti-La antibody-positive mothers had DRB1 alleles that shared the same amino acid residues at positions 14 to 31 and 71 of DRB1 and were homozygous or heterozygous at DQ6 and DQ3 alleles that shared the same amino acid residues at positions 27 to 36 and 71 to 77 of the hypervariable regions of DQB1. l1 O Individual manifestations of NLE may also be influenced by maternal HLA, as the maternal DRS haplotype DRB1*1101-DQAl*0501DQB1*0301 and individual class II alleles making up this haplotype, including DQA1 alleles with glutamine at position 34 of the first domain, were significantly associated with C-NLE but not CAVE. DQB1*0602 carried on DR2 haplotypes was associated with CAVB but not C_NLE. 120 ,121 Although not conclusive, a few studies have suggested that maternal MHC class I haplotypes may predispose to the delivery of child with CAVB. 1l 4,122,123 Despite the strong association of maternal HLA antigens with NLE, there generally has not been any association of HLA genes in the offspring with the development of NLE, although there have been exceptions. 120 ,121,124-l27 One study demonstrated that children with NLE tended to have DRB, DQA, and DQB genes identical to those of their mothers. 128 One report suggested that DR3 in the fetus might protect against in utero death, whereas another postulated that DR2 in the infant may be protective (although this was associated with maternal DR2).69,126 Some data suggest children with NLE are more likely to have a tumor necrosis factor-a (TNF-a) polymorphism, which is associated with high TNF-a production, and TGF-P gene polymorphism, which is associated with increased TGF-P production and may lead to fibrosis. 129 This work suggests that there may be factors in the developing fetus that influence the development of NLE. Multiple genes outside the MHC locus, including T cell receptor genes (in mother or child, or both) can influence the onset and progression of autoimmune diseases. 128 ,130,131 The same may be true in NLE,
CLINICAL MANIFESTAnONS OF NEONATAL LUPUS ERYTHEMATOSUS Cardiac Neonatal Lupus Erythematosus The most clinically significant manifestations of NLE are cardiac, specifically CAVB. In most cases, the CAVB is isolated, but it may be associated with other cardiac lesions, including ventricular septal defect or patent ductus arteriosus. The first reported case of CAVB associated with maternal autoimmune disease (i.e., Mikulicz's syndrome or Sjogren's syndrome) was published in 1901. 132 However, it was not until the 1950s that it was generally recognized that autoantibodies in the mother were
C HAP T E R
17 N EON ATAL L u PUS E RY THE MAT 0
SUS
395
associated with NLE. It was another 20 to 30 years until the association with anti-Ro and anti-La antibodies was reported. 7G-72, 133--137
Epidemiology It is estimated that CAVB occurs in 1 in 14,000 live births, and at least 90% of the cases of CAVB are the result of transplacental passage of maternal autoantibodies. This is likely an underestimate of the true incidence of abnormalities of fetal cardiac conduction, because when severe, CAVB can result in intrauterine death. 13H-143 In most cases, these deaths occurred in fetuses of mothers wit~out a diagnosed autoimmune disease, and the first demonstration of autoantibodies in these mothers occurred during the pregnancy or after delivery of a child with NLE. Our experience suggests that 25% to 50% of pregnancies complicated by fetal CAVB result in intrauterine death and most mothers do not have an autoimmune disease. It is likely that CAVB occurs in more than 1 of 14,000 live births and may be a factor resulting in death of the fetus in 1 of 7000 to 8000 pregnancies carried only to the late second trimester,
Pathology Although necropsy studies are few, the characteristic pathologic findings in CAVB are an absence or a degeneration of the atrioventricular (AV) node with replacement by fibrosis, calcification, or fatty tissue (Fig. 17-1). The distal conducting system may be normal. The few reports of cases of CAVB not associated with maternal aut~)antibodies have demonstrated a normal AV node but there were other conduction defects. Abnormal histology is present in other areas of the heart, and therefore, the inflammation and scarring may be a more generalized process associated with ventricular endomyocardial fibroelastosis (EFE) with complement deposition and an inflammatory cell infiltrate. 134 ,144 These latter abnormalities suggest the possibility that AV node conduction defects may be the result of the susceptibility of the conduction system to inflammatory damage rather than a specific localization of the autoantibodies to this area of the developing heart. It likely that maternally derived autoantibodies lead to fetal myocarditis, which can result in EFE and disruption of the conducting system. 14S The presence of myocardial inflammation has been substantiated by the demonstration of immunoglobulin (IgG), complement, and fibrin deposition on the myocardium.134.144.146-1S0 Because the fetus is unable to produce this immunoglobulin, the demonstration of IgG deposits on the myocardium implicates maternal immunoglobulin as the likely source of the pathology. In addition to maternally derived IgG, maternal cells have been found in fetal hearts from children with CAVB.Is I However, there is likely a role for the fetal immune system in the inflammatory response and myocardial damage, because we have demonstrated IgM and a T cell infiltrate in the myocardium of fetuses who died of CAVB and EFE. 144 In vitro experiments have demonstrated that apoptosis of cardiomyocytes leads to expression of Ro and La on the cell surface and that these apoptotic cells
• Figure 17-1 Section of ventricular septum from the fetus of a mother with systemic lupus erythematosus.The baby died at birth from nonimmune hydrops secondary to complete congenital heart block. A, dystrophic calcification in the region of an atrioventricular node; B, dense fibrosis; C, valve leaflet; 0, ventricular septum. (Courtesy of Dr. J. Dimmick.)
can activate production of pro-inflammatory cytokines, which may lead to scarring. 1S2- 1S4 Although we could not demonstrate evidence of increased apoptosis in the hearts of fetuses who died of CAVB and EFE, work by others suggests that increased apoptosis may nevertheless be important in the development of CAVB and EFE.IS O Anti-Ro and anti-La antibodies only rarely cause conduction abnormalities in the mothers of children with NLE, although conduction abnormalities and myocarditis are more common in SLE patients with antiRo and anti-La antibodies, than in SLE patients without these antibodies. 3,lss,ls6
Treatment The detection of fetal bradycardia (heart rate of less than 120 beats per minute) requires that the mother be referred for immediate fetal cardiac echocardiography to confirm the rate and the possible presence of AV block. If isolated AV block is present, the maternal anti-Ro and anti-La antibody status should be determined. Most fetuses tolerate CAVB if the ventricular rate is greater than
396
c
HAP T E R
17
NEONATAL
Lupus
ERYTHEMATOSUS
60 beats per minute. Slower rates and signs of congestive heart failure are associated with poor fetal outcome. However, fetal ventricular pacing has not resulted in a live birth, although pacemakers have been successfully implanted into the fetus. 157,158 In view of the problems with direct intrauterine pacing, it is recommended that if the fetal heart persists at a rate of less than 55 beats per minute or if there is any sign of cardiac decompensation, maternal therapy should be initiated. This therapy consists of oral or intravenous sympathomimetics that can be administered safely and effectively to increase the fetal heart rate. 159-162 Maternal administration of dexamethasone has also been used with some success in reversing hydrops fetalis, and in particular, if it occurs in the late second to early third trimester. 163 The use of inotropic therapy may improve the fetal outcome. However, if hydrops fetalis persists, immediate delivery should follow. Initially, the mother should be given fluorinated glucocorticoids and thyroid releasing hormone to increase fetal lung development, which is followed 48 hours later by delivery by cesarean section. The delivery is controlled, and intubation, ventilation, and pacemaker insertion or sympathomimetic therapy are available. 164 If there is good ventricular function, no pleural or pericardial effusions, and a satisfactory cardiac output, direct fetal therapy is not warranted. The treatment of choice is maternal administration of a fluorinated glucocorticoid such as dexamethasone at a dose of 4 mg/day. This recommendation is based on the successful treatment of CAVB and carditis with dexamethasone. A large series of cases reported that one fetus with third-degree heart block temporarily reverted to second-degree heart block, and one fetus with second-degree heart block reverted to first-degree block. In these cases, the heart rate reverted to the initial rate, but all fetuses were successfully delivered. The three pregnancies with fetuses with hydrops fetalis resulted in permanent reversal of the hydrops after therapy and resulted in live births. 165 Betamethasone can be used instead of dexamethasone. 166 In the past, it had been suggested that mothers with anti-Ro and anti-La antibodies and a history of delivering a child with CAVB should receive glucocorticoid therapy and undergo plasmapheresis. 167 ,168 This aggressive therapy was replaced by the suggestion that glucocorticoid therapy with or without intravenous immunoglobulin should be used prophylactically; there have been case reports of the successful use of this therapy.169.170 The risk of recurrence of CAVB is low in subsequent pregnancies, however, and therefore any prophylactic therapy is likely to be "successful." The current recommendation is to monitor pregnancies associated with maternal anti-Ro and anti-La antibodies with serial fetal echocardiograms. Any therapies should be reserved for pregnancies complicated by fetal heart block (of any degree).
Long-Term Cardiac Outcome The long-term outcome of children with CAVB is guarded. Many children require early pacemaker insertion. Most large series suggest a lO-year survival rate of
about 80%, with the highest rate of mortality within the first year of life. 171-173 Most cases of CAVB leading to postnatal death are associated with EFE. It had been initially suggested that EFE was the result of poor or late pacemaker placement, but later studies have demonstrated that despite adequate pacing, EFE can lead to a cardiomyopathy which frequently leads to death or the need for cardiac transplantation. 173-175 The decision to insert a permanent pacemaker early after delivery is determined by the ability of the neonate to tolerate its intrinsic heart rate. The slower the rate, the more likely that early pacemaker insertion will be necessary. Current guidelines suggest that all children should have pacemaker insertion before the end of adolescence. 176-178
Cutaneous Neonatal Lupus Erythematosus The rash of C-NLE was first reported in 1954 in a child born to a mother with an autoimmune disease. 179 It was not until 1981, however, that the association of C-NLE and maternal anti-Ro antibodies was described. 180 Similar to mothers of children with CAVB, mothers of children with C-NLE are usually clinically well despite the presence of circulating anti-Ro or anti-La antibodies or both. It is likely that a rash is present in 15% to 250/0 of children with NLE, although it is difficult to determine the tme percentage of children born of these mothers who will develop C-NLE, because the rash can be easily missed and spontaneously resolves. 181 There is a female predominance in infants with C-NLE, with a female-to-male ratio of 2: 1 to 3: 1.182,183 The reason for the increased incidence in females may be related to the observation that estrogens enhance surface expression of Ro and La proteins on keratinocyte cells (discussed later). It is not clear whether there is a similarly increased risk of CAVB in female offspring. The photosensitive nature of the cutaneous lesions has led investigators to examine the effect of ultraviolet irradiation on keratinocyte cell surface expression of the components of RoRNP (these in vivo and in vitro experiments are discussed later), Clinically, the dermatitis more closely resembles the lesions of subacute cutaneous lupus erythematosus (SCLE) than the malar rash of SLE (Figs. 17-2 to 17-4), Most patients with SCLE have anti-Ro or anti-La antibodies or both, and these antibodies are directed against similar proteins on RoRNP, as are the antibodies present in mothers of children with NLE. 82 ,184,185 The rash of C-NLE is rarely in a malar distribution, and the lesions are not indurated, whereas follicular plugging or dermal atrophy, typical of discoid lupus erythematosus, is rare; the rash may mimic Langerhans cell histiocytosis.176.181.186 The face and scalp are the most commonly involved areas, but the rash may occur at any site, including the palms and soles. Commonly, it develops around the eyes in a raccoon-like distribution, The rash tends to consist of discrete, round, or elliptical plaques with a fine scale that has central clearing, and it tends to be papulosquamous or characterized by annular erythema. An infant may have one or both of these typical rashes. In North America, papulosquamous lesions are most commonly described,
C HAP T E R
17 N EON ATAL L u PUS E RY THE MAT 0
SUS
397
~ 17-2 Neonatal lupus syndrome.This erythematous rash appeared a day after birth, accompanied by thrombocytopenia and leukopenia. (Courtesy of Drs. D.C. Rada andT. Kestenbaum.) (See color insert.)
•
A child with neonatal lupus syndrome.The rash on this baby is more discoid in character. (Courtesy of Dr. D. Kredich.)
• figure 17-3
whereas in Japan, annular erythema is more commonly seen. 187 Bullous lesions may be seen especially on the soles of the feet, and the rash may resemble cutis marmorata telangiectasia, congenita bullous impetigo, prim~ry herpes simplex infection, and erythema multiforme. The dermatitis of C-NLE may be present at birth but more commonly develops within the first few weeks of life.181.182.186 The most common age of appearance is of the rash is 6 weeks, but it may not be recognized until as late as 12 weeks. l86 New lesions may appear for several months, but they rarely develop beyond 6 months of postnatal life, consistent with the disappearance of maternal antibodies from the infant's circulation. The lesions may be induced or exacerbated by sun exposure, but instances of C-NLE that are present at birth, along with the observations that involvement of the soles of the feet and diaper area occur, illustrate that sun exposure is not required for development of the rash. Rash appearing after phototherapy for neonatal jaundice has been reported, but it is uncommon. 16 The mean duration of the rash has been reported to be 17 weeks. l86 The lesions of C-NLE are transient and usually resolve without scarring, although some mild epidermal atrophy may result. Cutaneous telangiectasias, beginning at age 6 ~o 12 months, occur in approximately 10% of affected infants. 1S8,161,162 The telangiectasias may occur in areas that were not initially involved with C-NLE and therefore are not just the result of healing of the initial inflammatory
rash. 161 The most common area for telangiectasia is at the temples near the hairline, an area not usually affected by the acute lesions. Telangiectasias tend to be bilateral. This lesion may be the presenting feature of C-NLE, although it is not clear in these instances whether the initial rash was subtle and missed or the telangiectasias occurred de novo without the characteristic earlier rash. l88 There have been reports of the telangiectasias with atrophy persisting into adolescence. 189
Pathology Biopsies of the lesions of C-NLE demonstrate the typical abnormal histopathology of SCLE, which includes epidermal basal cell damage, a mild mononuclear cell dermal infiltrate, vacuolation of the basal layer, and epidermal colloid bodies. IgG, IgM, and complement are deposited usually at the dermoepidermal junction. 190 ,191 Most mothers of infants with C-NLE are anti-Ro antibody positive, often in combination with anti-La antibodies. The percentage of mothers with elevated anti-Ro antibody levels depends on the assay used. A few cases of C-NLE have been reported in association with antibodies to U1RNP in the absence of anti-Ro or anti-La antibody and have included one report of twins discordant
398
C HAP T E R
17 N EON AT ALL u pus E RY THE MAT 0
SUS
the child, because the natural history of the skin lesions is spontaneous resolution without scarring; therefore, aggressive treatment is not indicated. However, topical application of a mild glucocorticoid cream may hasten the resolution of the lesions and be used for cosmetic reasons, although it is possible that steroid use increases the risk of developing telangiectasias. Telangiectasias can be treated with pulse dye laser therapy, although they may also spontaneously improve. IRR
Liver Disease
• Figure 17-4 Neonatal lupus syndrome. Four-month-old girl with the erythematous rash of neonatal lupus syndrome across the bridge of the nose, lower eyelids and superior forehead.This baby was born at 33 weeks of gestation with complete congenital heart block. Her mother had high titers of anti-Ro antibody.
for NLE. 192-194 The cutaneous lesions in some of these infants were considered to be atypical in morphologic appearance. 194 We examined one infant with C-NLE who had anti-U1RNP but not anti-Ro or anti-La antibodies when tested by enzyme-linked immunosorbent assay.19S However, anti-La antibodies were identified by immunoblot. In some cases of C-NLE, the antibodies may he directed to epitopes that cannot be detected by enzyme-linked immunosorbent assay.
Differential Diagnosis The differential diagnosis of isolated C-NLE includes the other causes of annular and polycyclic lesions as follows. 196-202 When the rash occurs concomitant with CAVB, the diagnosis is much easier: For a polycyclic lesion: urticaria, erythema marginatum, tinea corporis, seborrheic dermatitis, and ichthyosiform genodermatosis For annular erythema: erythema annulare centrifugum, familial annular erythema, erythema multiforme, infantile epidermodysplastic erythema, infection with Pityrospornm, annular erythema of infancy, and erythema gyratum atrophicans
Treatment The usual approach to management of C-NLE is reassurance offered to the parents and continued observation of
Hepatic dysfunction in NLE is characterized by abnormal levels of liver enzymes and hepatomegaly that were initially ascribed to congestive heart failure, intrauterine hydrops fetalis, disseminated intravascular coagulation, or total parenteral nutrition. It was subsequently suggested that hepatic involvement occurs in approximately 15% of infants with NLE. However, a large, unselected series demonstrated that liver involvement occurred in approximately 25% of all infants with NLE. IRI Liver disease can present as an isolated disorder or in association with C-NLE, CAVB, or any other manifestation of NLE. Usually, patients have mild hepatomegaly, with or without splenomegaly, and cholestasis with mildly to moderately elevated transaminases. 203 ,204 Hepatitis may be the only manifestation of NLE in a mother with anti-Ro and anti-La autoantibodies. 179. IRI ,IR6 Although a liver biopsy is usually not clinically indicated, histologic abnormalities are similar to idiopathic neonatal giant cell hepatitis with mild bile duct obstruction, occasional giant cell transformation and mild portal fibrosis.1 77,2tJs It is possible in this regard that idiopathic neonatal hepatitis may be another manifestation of NLE. Liver biopsies should be reserved for infants with clinical evidence of severe dysfunction or with persistent, moderate dysfunction. Abnormalities of liver function usually resolve, although deaths secondary to hepatic failure before age 6 months have been reported. 1R1 ,20J-2tJ7 The only report of a repeat biopsy in an infant with hepatic abnormalities demonstrated persistence of mild fibrosis; the child had a good long-term outcome,2tJ3 There have not been any instances of late failure or cirrhosis; however, it has been only since ahout 1980 that hepatic disease has been identified as part of the syndrome.
Hematologic Disease Thrombocytopenia is the most common hematologic manifestation, whereas anemia and neutropenia are less frequently present. Usually, hematologic involvement develops in conjunction with other stigmata, but thrombocytopenia, anemia, neutropenia, and pancytopenia have been reported as isolated manifestations of NLE (NLE should therefore be considered in the differential diagnosis of neonatal cytopenias).181,20K-21I Antiplatelet antibodies have been rarely detected, suggesting that other factors may be responsible for the neonatal thrombocytopenia. The thrombocytopenia and neutropenia may be secondary to anti-Ro or anti-La antibodies,
C HAP T E R
beqmse thrombocytopenia in SLE is associated with these autoantibodies and anti-Ro antibodies have been demonstrated to bind to neutrophils. 212-215 Children with chronic immune thrombocytopenic purpura may have anti-Ro antibodies. 204 ,212-214 There have been isolated repprts of aplastic anemia and neonatal thrombosis associated with the transplacental passage of maternal autoantibodies. m ,215-217 The thrombocytopenia and other hematologic manifestations tend to resolve over several weeks and, unless there is bleeding, do not require treatment. However, if the condition is severe or life threatening, high-dose glucocorticoids or intravenous immunoglobulin may be necessary!l~
17
NEONATAL
Lupus
ERYTHEMATOSUS
399
The ultrasound abnormalities included subependymal cysts, increased echogenicity of the white matter and echogenic lenticulostriate vessels. The only clinical abnormality in any of these children was one case of macrocephaly. On follow-up, all were developmentally normal. 225 We have examined four infants with hydrocephalus and NLE, one of whom required a ventricular shunt. There have been multiple case reports of chondrodysplasia punctata associated with NLE, including one child at our institution with C-NLE and radiographic changes in the hip and ankle consistent with this diagnosis. U6,227 Individual case reports have included congenital nephrotic syndrome in an infant with NLE 228 and a child with C-NLE and Turner's syndrome. 229
OIlier Manifestations There have been numerous reports of different diseases of the newborn associated with the transplacental passage of maternal anti-Ro, anti-La, or other autoantibodies, as well as unusual clinical manifestations in neonates with classic NLE. When there is a single case report, it is not clear whether the illness in the neonate is related to the maternal autoantibodies or is a disease coincidentally occurring in offspring of mothers with autoantibodies. Similarly, when unusual features are identified in children with definite NLE, it is not clear whether the other illness is coincidental or the result of maternal autoantibodies. Multiple neurologic manifestations have been described. A myelopathy with a gait abnormality and spastic paraparesis has been reported in two cases of infants with C-NLE.219 The neurologic disease became apparent in one child at age 1 year and in the other at age 16 months. Although it is possible that there was undiagnosed neonatal thrombocytopenia or vasculitis that resulted in intracerebral hemorrhage or infarction without any other clinical disease, there was no obvious cause other than the presence of anti-Ro antibodies. 192 Other neurologic diseases have included an infant with vasculopathy, a nonspecific marker of an insult to the developing blood vessels of the brain; hydrocephalus in two female siblings; and a case of transient neonatal myasthenia gravis.uo-m Three children with CAVB have been identified with cerebral ultrasonography and color Doppler flow imaging studies that demonstrated evidence of a vasculopathy in the area of the thalamus. A short-term follow-up study of these children revealed no signs of progression or neurologic impairment. The in"1estigators suggested that when infants are seen with clinical signs and symptoms consistent with a vasculopathy in the gangliothalamic region, they should be examined for other manifestations of NLE, and the mothers should be tested for anti-Ro and anti-La antibodies!2] There has been one case report of an adolescent with CAVE who developed moyamoya disease!24 A study of 10 consecutive children with other clinical features of NLB demonstrated abnormal computed tomographic (CT) scans or ultrasound findings, or both, in nine. The CT abnormalities included decreased attenuation of the cerebral white matter, basal ganglia calcifications, ventriculomegaly, and benign macrocephaly of infancy.
COURSE OF THE DISEASE AND PROGNOSIS Children NLE is a disease caused by the transplacental passage of maternal autoantibodies and not by the production of these autoantibodies by the fetus or neonate. Skin, liver, and hematologic complications generally resolve with minimal sequelae, whereas CAVB is permanent; the longterm cardiac outcome of these children is described in the section on CAVB. It was initially reported that children with NLE might be at a high risk for developing SLE in later life!30,231 In our experience, none of more than 100 children with NLE has developed a connective tissue disease; however, most of these children have been followed for less than 20 years. A report from Japan suggested a less optimistic outcome; 8% of children had persistent or recurrently positive autoantibodies, and one met the American College of Rheumatology classification criteria for SLE. 125 In a second study, 12% of children with NLE developed an autoimmune disease (Le., juvenile rheumatoid arthritis, Hashimoto's thyroiditis, psoriasis, diabetes mellitus, and nephrotic syndrome), although only a minority had autoantibodies, and none had anti-Ro antibodies or SLE.232 The increased risk of autoimmune disease in these offspring may reflect the genetic predisposition of a child born to a mother with an autoimmune disease, as opposed to a direct delayed consequence of having had NLE. The risk of development of an autoimmune disease in the child with NLE is probably not greater than the risk for developing SLE in any other offspring of anti-Ro antibody-positive women with SLE and is likely related to the linkage of HLA class II genes to the production of anti-Ro and anti-La antibodies. 107 ,233-237 Parents of these infants should be counseled that the risk of their offspring developing autoimmune diseases is similar to the risk in children of women with SLE.
Mothers Initially, NLE was the name of the disorder given to infants born with the characteristic skin or cardiac findings in the presence of maternal connective tissue
400
C HAP T E R
17
NEONATAL
Lupus
ERYTHEMATOSUS
disease, particularly SLE and Sjogren's syndrome. All mothers of infants with NLE therefore had an autoimmune disease. However, it rapidly became apparent that CAVB and the characteristic rash of C-NLE could be seen in the offspring of mothers who did not have any signs or symptoms of a connective tissue disorder. It was initially surmised that the mothers of children with CAVB would develop a connective tissue disease, but we and others found that only a minority of mothers had a connective tissue disease at the time of delivery of their children with CAVB, and at long-term follow-up the majority remained healthy.76,97-99.238,239 We have demonstrated that mothers of children with C-NLE were more likely to have had a diagnosed autoimmune disorder at the time of delivery of the child with NLE and at longterm follow-up than were mothers of children with CAVB,240 Because almost all children with NLE are born to mothers with anti-Ro or anti-La antibodies, or both, it is important to estimate the risk of delivering a child with NLE from mothers with a known connective tissue disease (particularly SLE) or from mothers who have previously delivered a child with NLE. Large series of pregnancies in women with SLE who had anti-Ro or antiLa antibodies, or both, have suggested that the risk of delivering a child with NLE varied between 1% and 10%.78,211,241,242 Later large, prospective studies have suggested that the risk of delivering a child with NLE in mothers with anti-Ro or anti-La antibodies, or both, was 1% to 2%.243,244 Retrospective studies have suggested a recurrence rate for CAVB of between 8% and 250/0 in subsequent pregnancies. 126,13l,208 Our experience and that of others in pregnancies after the delivery of a child with CAVB or C-NLE suggests that the risk is approximately 15% to 20%,24.>,244
ANIMAL MODELS OF NEONATAL LUPUS ERYTHEMATOSUS The major targets of NLE are the heart, skin, and liver. To better determine the pathogenesis of NLE, in vivo and in vitro models have been established. The in vivo models have used the infusion of human autoantibody containing sera or have generated autoantibodies by immunization of mice to generate autoantibodies. The in vitro models have used isolated cardiac myocytes or whole organ culture.
In Vitro Experiments Culture of isolated neonatal, but not adult, rabbit cardiac myocytes with sera from anti-Ro or anti-La antibodypositive women led to changes in the repolarization of these cells. 245 ,246 Binding of the maternal sera to rabbit tissue was likely the result of the presence of RoRNP antigen on the surface of fetal hearts. 247 ,248 However, unlike that which occurs with keratinocytes, the cellular localization of 48-kD La, 52-kD Ro, or 60-kD Ro in fetal cardiac myocytes is not altered by culture in the presence of 17~-estradiol or progesterone (discussed later). 249
Calcium channels are important in maintaining cardiac rhythm, and antibodies against these ions therefore are important in CAVB. Experiments using cultured human fetal cardiomyocytes have demonstrated that anti-Ro antibodies (particularly anti-52-kD Ro antibodies) alter calcium L-type and T-type channels. 250 ,251 Affinity-purified antibodies, which recognize both 52-kD Ro forms, also recognize the human 5-HT4 receptor and antagonized the serotonin-induced L-type calcium channel activation on isolated human atrial cells. 252 However, a study using sera from the Research Registry for Neonatal Lupus failed to show an association of anti-5HT4 receptor antibodies and the development of CAVB.253 Autoantibodies from mothers of children CAVB have been shown to bind to and modify the response of muscarinic acetylcholine receptor activation of neonatal but not adult rat atria. 254-256 Taken together, it appears that antibodies directed against calcium channels and receptors important in generation of cardiac rhythm, which are present on cardiomyocytes, may playa role in the pathogenesis of CAVB.
Skin Irradiation of keratinocytes enhances the expression of Ro, U1RNP, and Sm antigens.257-259 Estradiol treatment of keratinocytes can induce a marked increase in mRNA and expression of Ro, U1RNP, and Sm autoantigens. 248,257,258,260 Other factors, including TNF-a and exposure to viruses, upregulate the surface expression of 52-kD Ro and La on keratinocytes. 261 The direct binding of antiRoRNP antibodies to skin has been demonstrated. 248 ,262 The difference in disease expression in offspring born to the same mother may be at least partially explained by the demonstration that sera from children with C-NLE can be cytotoxic to keratinocytes from patients with NLE but not to cells obtained from normal individuals. This cytotoxicity was enhanced by ultraviolet B irradiation. These data suggest that keratinocytes from children with C-NLE may have abnormal surface expression of the Ro and La antigens and that ultraviolet irradiation can further increase that expression. 263 These results are consistent with the demonstration that the rash of C-NLE may be present at birth or later and that it does not occur in all infants born to mothers with autoantibodies directed against RoRNP.
Langendorff Experiments Initial ex vivo experiments examined the effect of antiRo- and anti-La-containing sera on conduction in isolated rabbit hearts. The perfusion of isolated Langendorff preparations of adult rabbit hearts with purified IgG from sera containing anti-Ro and anti-La antibodies induced heart block and altered the peak slow inward current.264 However, sera from women with SLE or Sjogren's syndrome without a history of delivering a child with NLE also resulted in heart block in the isolated, whole rabbit heart, although the heart block occurred only with perfusion with affinity-purified anti-52-kD antibodies. 265 These observations are not unique to anti-Ro antibody-containing sera, because similar alterations of cardiac conduction have been observed with sera from
C HAP T E R
patients with conduction defects associated with Chagas' disease. 266 Perfusion of Langendorff preparations of human fetal hearts with affinity-purified anti-52-kD Ro derived from mothers of children with CAVB also resulted in the development of complete AV block. At a wholecell and single-channel level, perfusion experiments with the human heart demonstrated an inhibition of L-type calcium currents. 267 Similarly, when isolated rat hearts were used, a 2: 1 AV block followed by complete inhibition of AV nodal action potential was demonstrated, and calcium channels were inhibited in isoll.lted cellular preparations. 268 These results suggested that rodents may be an appropriate species for monitoring the fetal effects of maternal anti-Ro and anti-La antibodies.
In VIvo Experiments Immunization of female BALB/c mice with recombinant RoRNP proteins generates high-titer antibodies that cross the placenta during pregnancy and are associated with various degrees of AV conduction abnormalities in the pUpS.267 However, conduction abnormalities were seen in only a low percentage of the offspring born to these mice, and advanced conduction abnormalities rarely developed. 269 Apoptosis has been proposed as a mechanism for the tissue damage. 270- m In human fetal cardiac myocytes, apoptosis results in surface translocation of RoRNP.273 In vivo murine experiments have supported this hypothesis because the passive transfer of human IgG containing anti-52-kD Ro, anti-60-kD Ro, and anti-La autoantibodies led to the formation of human IgG-apoptotic cell complexes in organs targeted in NLE (Le., heart, skin, liver, and bone) but not in thymus, lung, brain, or gut. Experiments with affinity-purified antibodies demonstrated that anti-La, but not anti-Ro, antibodies formed these complexes.274 It is possible that apoptosis, a normal event during cardiac development, may result in the binding of maternal anti-RoRNP to the apoptotic cells that cause an inflammatory reaction. The neighboring cells may be damaged as bystanders. Initial binding may be by maternalanti-52-kD Ro antibodies or antibodies to isoforms of the La protein, which are maximally expressed early in gestation, with levels decreasing with gestational age until 25 weeks, when adult levels are achieved. 33,40 This hYIJothesis allows for the selective damage to fetal but not to maternal conducting tissue.
REFERENCES 1. Smeenk RJ: Immunological aspects of congenital atrioventricular block. Pacing Clin Electrophysiol 20: 2093-2097, 1977. 2. Zhu J: Cytomegalovirus infection induces expression of 60 kD/Ro antigen on human keratinocytes. Lupus 4: 396-406. 1995. 3. Wolin S, Stietz]: The Ro small cytoplasmic ribonucleoproteins: identification of the antigenic protein and its binding site on the Ro RNAs. Proc Nat! Acad Sci USA 81: 1996--2000, 1984. 4. Lerner MB, Boyle ]A, Hardin JA, Steitz JA: Two novel classes of small ribonucleoproteins detected by antibodies associated with lupus erythematosus. Science 21 L 400-402. 1981.
17
NEONATAL
Lupus
ERYTHEMATOSUS
401
5. Ben-Chetrit E, Chan EKL, Sullivan KF, Tan EM: A 52-kD protein is a novel component of the SS-AlRo antigenic particle. J Exp Med 167: 1560-1571, 1988. 6. Harmon CE, Deng J-5. Peebles CL, Tan EM: The importance of tissue substrate in the SS-AlRo antigen-antibody system. Arthritis Rheum 27: 166-173, 1984. 7. Hendrick JP, Wolin SL, Rinke J, et al: Ro small cytoplasmic ribonucleoproteins are a subclass of La ribonucleoproteins: further characterization of the Ro and La small ribonucleoproteins from uninfected mammalian cells. Mol Cell Bioi I: 1138-1149, 1981. 8. Rosa MD, Gottlieb E, Lerner MR, Steitz ]A: Striking similarities are exhibited by two small Epstein-Barr virus-encoded ribonucleic acids and the adenovirus-associated ribonucleic acids VAl and VAIl. Mol Cell Bioi 1: 785-796, 1981. 9. Rinke J, Steitz JA: Precursor molecules of both human 5S ribosomal RNA and transfer RNAs are bound by a cellular protein reactive with anti-La lupus antibodies. Cell 29: 149-159, 1982. 10. Madore SJ, Wieben ED, Pederson T: Eukaryotic small ribonucleoproteins: anti-La human autoantibodies react with Ul RNA-protein complexes. J BioI Chern 259: 1929-1933, 1984. 11. Maraia R], Kenan 0], Keene JD: Eukaryotic transcription termination factor La mediates transcript release and facilitates reinitiation by RNA polymerase lII. Mol Cell Bioi 14: 2147-2158, 1994. 12. Fan H, Goodier ]L, Chamberlain ]R, et al: 5' Processing of tRNA precursors can be modulated by the human La antigen phosphoprotein. Mol Cell Bioi 18: 3201-3211, 1998. 13. Gottlieb E, Steitz ]A: The RNA binding protein La influences both the accuracy and the efficiency of RNA polymerase III transcription in vitro. EMBO] 8: 841-850, 1989. 14. Gottlieb E, Steitz ]A: Function of the mammalian La protein: evidence for its action in transcription termination by RNA polymerase. EMBO] 8: 851-861, 1989. 15. Deng j-S, Sontheimer RD, Giliam IN: Molecular characteristics of SS-B/La and SS-AlRo cellular antigens.] Invest Dermatol 84: 86-90, 19B5. 16. Dickey WO, van Egmond JE, Hardgrave KL, et al: Presence of anti-LaCS5-B) is associated with binding to the 13-kD carboxyl terminus of 6o-kD RoCSS-A) in systemic lupus erythematosus.] Invest Dermatol 100: 412-416, 1993. 17. Pardigon N, Strauss ]H: Mosquito homolog of the La autoantigen binds to Sindbis virus RNA. J Virol 70: 1173-1181, 1996. 18. Scherly 0, Stutz F, Lin-Marq N, Clarkson SG: La proteins from Xenopus laevis. cDNA cloning and developmental expression. ] Mol BioI 231: 196--204, 1993. 19. Yoo C), Wolin SL: La proteins from Drosophila melanogaster and Saccharomyces cerevisiae: a yeast homolog of the La autoantigen is dispensable for growth. Mol Cell Biol 14: 5412-5424, 1994. 20. Semsei I, Troster H, Bartsch H, et al: Isolation of rat cDNA clones coding for the autoantigen SS-B/La: detection of species-specific variations. Gene 126: 265-268, 1993. 21. Pannone BK, Xue 0, Wolin SL: A role for the yeast La protein in u6 snRNP assembly: evidence that the La protein is a molecular chaperone for RNA polymerase 1JJ transcripts. EMBO] 17: 7442-7453, 1998. 22. Rosenblum JS. Pemberton LF. Bonifaci N, Blobel G: Nuclear import and the evolution of a multifunctional RNA-binding protein. J Cell Bioi 143: 887-899. 1998. 23. Goodier ]L, Fan H, Maraia IV: A carboxy-terminal basic region controls RNA polymerase III transcription factor activity of human La protein. Mol Cell BioI 17: 5823-5832, 1997. 24. McLaren RS, Caruccio N, Ross J: Human La protein: a stabilizer of histone mRNA. Mol Cell Bioi 17: 3028-3036, 1997. 25. De BP, Gupta S, Zhao H, et ai: Specific interaction in vitro and in vivo of glyceraldehyde-3-phosphate dehydrogenase and LA protein with cis-acting RNAs of human parainfluenza virus type 3. J BioI Chern 271: 24728-24735, 1996. 26. Craig AW, Svitkin YV, Lee HS, et aI: The La aUloantigen contains a dimerization domain that is essential for enhancing translation. Mol Cell Bioi 17: 163-169, 1997. 27. Chang YN, Kenan OJ, Keene JD, et al: Direct interolctions between autoantigen La and human immunodeficiency virus leader RNA. J Virol 68: 7008-7020, 1994. 28. Meerovitch K, Svitkin YV, Lee HS, et al: La autoantigen enhances and corrects aberrant translation of poliovirus RNA in reticulocyte lysate. .I Virol 67: 3798-3807, 1993. 29. Huhn P, Pruijn GJ, van Venrooij WJ, Bachmann M: Characterization of the autoantigen La CSS-B) as a dsRNA unwinding enzyme. Nucleic Acids Res 25: 410-416. 1997. 30. Xiao Q, Sharp TV, Jeffrey IW, et al: The La antigen inhibits the activation of the interferon-inducible protein kinase PKR by sequestering and unwinding double-stranded RNA. Nucleic Acids Res 22: 2512-2518, 1994. 31. Peek R, van Venrooij WJ, Simons F, Pruijn G: The SS-AISS-B autoantigenic complex: localization and assembly. Clin Exp Rheumatol 12: SI5-SIB, 1994. 32. Shiroki K, Isoyama T, Kuge S, et al: Intracellular redistribution of truncated La protein produced by poliovirus 3Cpro-mediated cleavage. .I Virol 73: 2193-2200, 1999. 33. Grolz 0, Bachmann M: The nuclear autoantigen LalSS-associated antigen B: one gene, three functional mRNAs. Biochem] 323: 151-158, 1997.
402
C HAP T E R
17 NEONATAL Lupus ERYTHEMATOSUS
34. Hilker M, Troster H, Grolz 0, et al: The autoantigen La/SS-B: analysis of the expression of alternatively spliced La rnRNA isoforms. Cell Tissue Res 284: 383-389, 1996. 35. Grolz 0, Bachmann M: An altered intracellular distribution of the autoantigen La/SS-B when translated from a La mRNA isoform. Exp Cell Res 234: 329-335, 1997. 36. O'Brien CA, Wolin SL: A possible role for the 60-kD Ro autoantigen in a discard pathway for defective 5S rRNA precursors. Genes Dev 8: 2891-2903, 1994. 37. Shi H, O'Brien CA, Van Horn 01, Wolin SL: A misfolded fonn of 5S rRNA is complexed with the Ro and La autoantigens. RNA 2: 769-784, 1996. 38. Pourmand N, Blange I, Ringertz N, Pettersson I: Intracellular localisation of the Ro 52kD auto-antigen in HeLa cells visualised with green fluorescent protein chimeras. Autoimmunity 28: 225-233, 1998. 39. Igarashi T, Itoh Y, Fukunaga Y, Yamamoto M: Stress-induced cell surface expression and antigenic alteration of the Ro/SSA autoantigen. Autoimmunity 22: 33-42, 1995. 40. Buyon lP, Tseng CE, DiDonato F, et al: Cardiac expression of 52~, an alternative transcript of the congenital heart block~..ssociated 52-kd SS-AfRo autoantigen, is maximal during fetal development. Arthritis Rheum 40: 655-660, 1997. 41. Meilof lF, Van der Lelij A, Rokeach LA, et al: Autoimmunity and filariasis: autoantibodies against cytoplasmic cellular proteins in sera of patients with onchocerciasis. 1 Immunol lSI: 5800--5809, 1993. 42. Lieu TS, Sontheimer RD: A subpopulation of WIL-2 cell calreticulin molecules is associated with RO/SS-A ribonucleoprotein particles. Lupus 6: 40--47, 1997. 43. loshi M, Pogue GP, Duncan RC, et al: Isolation and characterization of Leishmania donovani calreticulin gene and its conservation of the RNA binding activity. Mol Biochem Parasitol 81: 53-64, 1996. 44. Rokeach LA, Haselby lA, Meilof lF, et al: Characterization of the autoantigen calreticulin. J Immunol 147: 3031-3039, 1991. 45. Smith Ml: A C. (>jegan.~ gene encodes a protein homologous to mammalian calreticulin. DNA Seq 2: 235-240, 1992. 46. Smith MJ: Nucleotide sequence of a Drosophila melanogaster gene encoding a calreticulin homologue. DNA Seq 3: 247-250, 1992, 47. Treves S, Zorzato F, Pozzan T: Identification of calreticulin isoforms in the central nervous system. Biochem J 287: 579-581, 1992. 48. Dupuis M, Schaerer E, Krause KH, Tschopp 1: The calcium-binding protein calreticulin is a major constituent of lytic granules in cytolytic T lymphocytes. J Exp Med 177: 1-7, 1993. 49. Eggleton P, Lieu TS, Zappi EG, et al: Calreticulin is released from activated neutrophils and binds to Clq and mannan-binding protein. Clin Immunol Immunopathol 72: 405-409, 1994. 50. Nash PO, Opas M, Michalak M: Calreticulin: not just another calcium-binding protein. Mol Cell Blochem 135: 71-78, 1994. 51. Baksh S, Spamer C, Heilmann C, Michalak M: Identification of the Zn'+ binding region in calreticulin. FEBS Lett 376: 53-57, 1995, 52. Nguyen TO, Capra 10, Sontheimer RD: Calreticulin is transcriptionally upregulated by heat shock, calcium and heavy metals. Mol Immunol 33: 379-386, 1996. 53. Waser M, Mesaeli N, Spencer C, Michalak M: Regulation of calreticulin gene expression by calcium. 1 Cell BioI 138: 547-557. 1997. 54. Burns K, Helgason CD, Bleackley RC, Michalak M: Calreticulin in T-lymphocytes. Identification of calreticulin in T-lymphocytes and demonstration that activation of T cells correlates with increased levels of calreticulin mRNA and protein. 1 BioI Chern 267: 19039-19042, 1992. 55. Michalak M, Burns K, Andrin C, et al: Endoplasmic reticulum form of calreticulin modulates glucocorticoid-sensitive gene expression. J Bioi Chern 271: 29436-29445, 1996. 56. Winrow CJ, Miyata KS, Marcus SL, et al: Calreticulin modulates the in vitro DNA binding but not the in vivo transcriptional activation by peroxisome proliferator-activated receptor/retinoid X receptor heterodimers. Mol Cell Endocrinol 111: 175-179, 1995, 57. Burns K, Duggan B, Atkinson EA, et al: Modulation of gene expression by calreticulin binding to the glucocorticoid receptor. Nature 367: 476-480, 1994, 58, Crossin KL, Tai MH, Krushel LA, et al: Glucocorticoid receptor pathways are involved in the inhibition of astrocyte proliferation. Proc Natl Acad Sci USA 94: 2687-2692, 1997. 59. Dedhar S, Rennie PS, Shago M, et al: Inhibition of nuclear hormone receptor activity by calreticulin, Nature 367: 480--483, 1994, 60. St-Arnaud R, Prud'homme 1, Leung Hagesteijn C, Dedhar S: Constitutive expression of calreticulin in osteoblasts inhibits mineralization, 1 Cell BioI 131: 1351-1359, 1995, 61. Zhu Q, Zelinka 1', White T, Tanzer ML: Calreticulin-integrin bidirectional signaling complex. Biochem Biophys Res Commun 232: 354-358, 1997, 62. Rojiani MY, Finlay BB, Gray V, Dedhar S: In vitro interaction of a polypeptide homologous to human Ro/SS-A antigen (calreticulin) with a highly conserved amino acid sequence in the cytoplasmic domain of integrin alpha subunits. BiochemL,try 30: 9859-9866, 1991. 63. Coppolino MG, Woodside MJ, Demaurex N, et al: Calreticulin is essential for integrin-mediated calcium signalling and cell adhesion, Nature 386: 843-847, 1997.
64. Van Leeuwen JEM, Kearse KP: The related molecular chaperones calnexin and calreticulin differentially associate with nascent T cell antigen receptor proteins within the endoplasmic reticulum, 1 Bioi Chern 271: 25345-25349, 1996, 65, Spee 1', Neefjes]: TAP-translocated peptides specifically bind proteins in the endoplasmic reticulum, including gp96, protein disulfide isomerJse and calreticulin. Eur.r Immunol 27: 2441-2449, 1997, 66, Verreck FA, Elferink 0, Vermeulen cr, et al: DR4Dw4/DR53 molecules contain a peptide from the autoantigen c~lreticulin. Tissue Antigens 45: 270--275, 1995. 67, Max H, Halder T, Kalbus M, et al: A 16mer peptide of the human autoantigen calreticulin is a most prominent HLA-DR4Dw4-associated self-peptide. Hum lmmunol 41: 39-45, 1994, 68. Taylor PV, Scott JS, Gerlis LM, et al: Maternal antibodies against fetal cardiac antigens in congenital complete heart block. N Engl .1 Med 315: 667-672, 1986. 69, Watson RM, Lane AT, Barnett NK, et al: Neonatal lupus erythematosus, A clinical, serological and immunogenetic study with review of the literature. Medicine (Baltimore) 63: 362-378, 1984. 70. Buyon JP, Ben-Chetrit E, Karp S, et al: AcqUired congenital heart block: pattern of maternal antibody response to biochemically defined antigens of the SSAfRo-SSB/La system in neonatal lupus. J Clin Invest 84: 627-634, 1989. 71. Silverman ED, BuyonJ, Laxer RM, et al: Autoantibody response to the Ro/La particle may predict outcome in neonatal lupus erythematosus. Clin Exp Immunol 100: 499-505, 1995. 72. Silverman E, Mamula M, Hardin lA, Laxer R: Importance of the immune response to the Ro/La particle in the development of congenital heart block and neonatal lupus erythematosus, 1 Rheumatol 18: 120--124, 1991. 73. McCreadie M, Celermajer 1, Sholler G, et al: A case control study of con~ genital heart block: association with maternal antibodies to Ro(SS~A) and La(SS-B). Br J Rheumatol 29: 10-14, 1990. 74, Taylor PV, Taylor KF, Norman A, et al: Prevalence of maternal Ro (SS~A) and La (SS-B) autoantibodies in relation to congenital heart block. Br .1 Rheumatol 27: 128-132, 1988. 75. Buyon Jp, Winchester Rl, Slade SG, et al: Identification of mothers at risk for congenital heart block and other neonatal lupus syndromes in their children. Comparison of enzyme-linked immunosorbent assay and immunoblot for measurement of anti-SS-AfRo and anti-SS-B/La antibodies. Arthritis Rheum 36: 1263-1273, 1993. 76. Julkunen H, Kurki 1', Kaaja R, et al: Isolated congenital heart block. Longterm outcome of mothers and characterization of the immune response to SS-AfRo and to SS-B/La. Arthritis Rheum 36: 1588-1598, 1993, 77, Julkunen H, Siren MK, Kaaja R, et al: Maternal HLA antigens and antibodies to SS-AfRo and SS~B/La. Comparison with systemic lupus erythematosus and primary Sjogren's syndrome. Br 1 Rheumatol 34: 901-907, 1995. 78. Ramsey-Goldman R, Hom D, Deng J-S, et al: Anti-SSA antibodies and fetal outcome in maternal systemic lupus erythematosus. Arthritis Rheum 29: 1269-1273, 1986. 79, Julkunen H, Kaaja R, Siren MK, et al: Immune-mediated congenital heart block (CHB): identifying and counseling patients at risk for having children with CHB. Semin Arthritis Rheum 28: 97-106, 1998. 80, Miyagawa S, Fukumoto T, Hashimoto K, et al: Maternal autoimmune response to recombinant Ro/SSA and La/SSB proteins in Japanese neonatal lupus erythematosus, Autoimmunity 21: 277-282, 1995, 81. Yukiko N: Immune responses to SS-A 52-kDa and 60-kDa proteins and to SS-B 50-kDa protein in mothers of infants with neonatal lupus erythematosus. Br 1 Dermatol 142: 908-912, 2000, 82. Lee LA, Alvarez K, Gmss T, et al: The recognition of human 60-kDa Ro ribonucleoprotein particles by antibodies associated with cutaneous lupus and neonatal lupus. J Invest Dermatol 107: 225-228, 1996. 83. Salomonsson S, Dorner T, Theander E, et al: A serologic marker for fetal risk of congenital heart block. Althritis Rheum 46: 1233-1241, 2002, 84, Grolz 0, Troster H, Semsei I, Bachmann M: Analysis of expression of the gene encoding for the nuclear autoantigen La/SS-B using reporter gene constructs. Biochim Biophys Acta 1396: 278-293, 1998. 85. Rokeach LA, Zimmerman PA, Unnasch TR: Epitopes of the Onchocerca volvulus RALI antigen, a member of the calreticulin family of proteins, recognized by sera from patients with onchocerciasis, Infect Immun 62: 3696-3704, 1994. 86. Orth T, Dorner T, Meyer Zum Buschenfelde KH, Mayet WJ: Complete congenital heart block is associated with increased autoantibody titers against calreticulin, Eur 1 Clin Invest 26: 205-215, 1996. 87. Chang SH, Huh MS, Kim HR, et al: Cross-reactivity of antibodies immllnoadsorbed to laminin with recombinant human La CSS-B) protein, J Autoimmun 11: 163-167, 1998. 88. Li JM, Horsfali AC, Maini RN: Anti-La (SS-BJ but not anti-R052 (SS~A) antibodies cross-react with laminin: a role in the pathogenesis of congenital heart block? C1in Exp Immunol 99: 316-324, 1995, 89, Horsfall AC, Li JM, Maini RN: Placental and fetal cardiac laminin are targets for cross-reacting autoantibodies from mothers of children with congenital heart block. 1 Autoimmun 9: 561-568, 1996, 90, Houen G, Koch C: Human placental calreticulin: purification, characterization and association with other proteins. Acta Chern Scand 48: 905-911, 1994.
C HAP T E R 91. U ]M, Fan WS, Horsfall AC, et al: The expression of human endogenous ~trovirus-3 in fetal cardiac tissue and antibodies in congenital heart block. Clin Exp Immunol 104: 388--393, 1996. 92. Maddison P], Lee L, Reichlin M, et al: Anti-p57: a novel association with neonatal lupus. Clin Exp Immunol 99: 42-48, 1995. 93. Haneji N, Nakamura T, Takio K, et al: Identification of alpha-fodrin as a candidate autoantigen in primary Sjogren's syndrome. Science 276: 604-607, 1997. 94. Miyagawa S, Yanagi K, Yoshioka A, et al: Neonatal lupus erythematosus: maternal IgG antibodies bind to a recombinant NH2-terminal fusion protein encoded by human alpha-fodrin cDNA.] Invest Dennatol 111: 1189-1192, 1998. 95. Derksen RH, Meilof ]F: Anti-Ro/SS-A and anti-La/SS-B autoantibody levels in relation to systemic lupus erythematosus disease activity and congenital hean block. A longitudinal study comprising two consecutive pregnancies in j patient with systemic lupus erythematosus. AnIlritis Rheum 35: 953--959, 1992. 96. Franceschini F, Brucato A, Quinzanini M, et al: Anti-Ro/SSA autoantibodies over time in motihers of children with congenital complete hean block. Clin Exp Rheumatol 17: 634-635, 1999. 97. McCune AB, Weston WL, Lee LA: Maternal and fetal outcome in neonatal lupus erythematosus. Ann Intern Med 106: 518-523, 1987. 98. Waltuck], Buyon ]P: Autoantibody-associated congenital hean block: out('Orne in mothers and children. Ann Intern Med 120: 544-551, 1994. 99. Press./, Uziel Y, Laxer RM, et al: Long-term outcome of mothers of children with complete congenital heart block. Am] Med 100: 328-332, 1996. 100. Julkunen H, Miettinen A, Walle TK, et al: Autoimmune response in mothers of children witih congenital and postnatally diagnosed isolated heart block: a population based study. ] Rheumatol 31: 183--189, 2004 101. Bell DA, Maddison P./: Serologic subsets in systemic lupus erythematosus: an examination of autoantibodies in relationship to clinical features of disease and HLA antigens. Arthritis Rheum 23: 1268-1273, 1980. 102, Ehrfeld H, Hartung K, Renz M, et al: MHC associations of autoantibodies 'Igainst recombinant Ro and La proteins in systemic lupus erythematosus. Results of a multicenter study. SLE Study Group. Rheumatollnt 12: 169-173, 1992. 103. Fei HM, Kang H, Scharf S, et al: Specific HLA-DQA and HLA-DRB1 alleles confer susceptibility to Sjogren's syndrome and autoantibody production. I Clin Lab Anal 5: 382-391, 1991. 104. ·I.ulli P, Sebastiani GO, Trabace S, et al: HLA antigens in Italian patients with systemic lupus erythematosus: evidence for the association of DQw2 with the autoantibody response to extractable nuclear antigens. Clin Exp \lheumatol 9: 475-479, 1991. 105. Kerttuia TO, Collin P, Polvi A, et al: Distinct immunologic features of Finnish Sjogren's syndrome patients with HLA alleles DRBl'0301, DQA l'0501, and DQBl'0201. Alterations in circulating T cell receptor gamma/delta subsets. Arthritis Rheum 39: 1733--1739, 1996. 106. Martin-Villa ./M, Maninez-Laso./, Moreno-Pelayo MA, et al: Differential contribution of HLA-DR, DQ, and TAP2 alleles to systemic lupus erytihematosus susceptibility in Spanish patients: role of TAP2'Ol alleles in Ro autoantibody production. Ann Rheum Dis 57: 214-219, 1998. 107. Miyagawa S, Shinohara K, Nakajima M, et al: Polymorphisms of HLA class II genes and autoimmune responses to Ro/SS-A-La/SS-B among ./apanese subjeL1s. Anhritis Rheum 41: 927-934, 1998. lOB. Smolen ./S, Klippel ./H, Penner E, et al: HLA-DR antigens in systemic lupus erytihematosus: association with specificity of autoantibody responses to nuclear antigens. Ann Rheum Dis 46: 457-462, 1987. 109. Wilson WA, Scopelitis E, Michalski ]P: Association of HLA-DR7 with both antibody to SSMRo) and disease susceptibility in blacks with systemic lupus erythematosus. ./ Rheumatol 11: 653--657, 19R4. 110. Miyagawa S, Dohi K, Shima H, Shirai T: HLA antigens in anti-RoCSS-A)positive patients with recurrent annular erytihema. ./ Am Acad Dennatol 28: IR'5-188, 1993, 111, Miyagawa S, Dohi K, Shima H, Shirai T: Absence of HLA-B8 and HLA-DR3 In ./apanese patients with Sjogren's syndrome positive for anti-SSACRo). ./ Rheumatol 19: 1922-1924, 1992. 112. Hamilton RG, Harley ]B, Bias WE, et al: Two Ro (SS-A) autoantibody responses in systemic lupus erythematosus. Correlation of HLA-DRlDQ specit1cities with quantitative expression of Ro (SS-A) autoantibody. Arthritis Rheum 31: 496-505, 1988. 113. Fujisaku A, Frank MB, Neas B, et al: HLA-DQ gene complementation and other histocompatibility relationships in man with the anti-Ro/SSA autoantibody response of systemic lupus erythematosus. ] Clin Invest R6: 606-611. 1990 114. Colombo G, Brucato A, Coluccio E, et al: DNA typing of maternal HLA in congenital complete heart block: comparison with systemic lupus erythematosus and primary Sjogren's syndrome. Arthritis Rheum 42: 1757-1764, 1999. 115. Rischmueller M, Lester S, Chen Z, et al: HLA class II phenotype controls diversification of the autoantibody response in primary Sjogren's syndrome (pSS). Clin Exp Immunol 111: 365-371, 1998. 116. Reveille .10, Macleod MJ, Whittington K, Arnett FC: Specific amino acid residues in the second hypervariable region of HLA-DQA1 and DQB1 chain genes promote tihe Ro (SS-A)/La (SS-B) autoantibody responses. .I Immunol 146: 3871-3876, 1991.
17
NEONATAL
Lupus
ERYTHEMATOSUS
403
117. Skarsvag S, Hansen KE, Moen T, Eggen BM: Distributions of HLA class II alleles in autoantibody subsets among Norwegian patients with systemic lupus erythematosus. Scand./ Immunol 42: 564-571, 1995. 118. Scofield RH, Dickey WD, Hardgrave KL, et al: Immunogenetics of epitopes of the carboxyl terminus of the human 60-kD Ro autoantigen. Clin Exp Immunol 99: 256-261, 1995. 119. Miyagawa S, Shinohara K, Kidoguchi K, et al: Neonatal lupus erythematosus: HLA-DR and -DQ distributions are different among the groups of antiRo/SSA-positive mothers with different neonatal outcomes. ./ Invest Dermatol 108: 881-885, 1997. 120. Miyagawa S, Fukumoto T, Hashimoto K, et al: Neonatal lupus erythematosus: haplotypic analysis of HLA class II alleles in child/mother pairs, Arthritis Rheum 40: 982-983, 1997. 121. Miyagawa S, Shinohara K, Kidoguchi K, et al: Neonatal lupus erythematosus: studies on HLA class II genes and autoantibody profiles in Japanese mothers. Autoimmunity 26: 95-101, 1997. 122. Miyagawa S, Kidoguchi K, Kaneshige T, Shirai T: Neonatal lupus erytihematosus: analysis of HLA class 1 genes in Japanese child/motiher pairs. Lupus 8: 751-754, 1999. 123. Siren MK, ./ulkunen H, Kaaja R, et al: Role of HLA in congenital heart block: susceptibility alleles in mothers. Lupus 8: 52-59, 1999. 124. Brucato A, Gasparini M, Vignati G, et al: Isolated congenital complete heart block: long-term outcome of children and immunogenetic study. .I Rheumatol 22: 541-543, 1995. 125. Kaneko F, Tanji 0, Hasegawa T, et al: Neonatal lupus erythematosus in Japan. ] Am Acad Dermatol 26: 397-403, 1992. 126. Arnaiz-Villena A, Vazquez-Rodriguez .1./, Vicario ./L, et al: Congenital heart block immunogenetics. Evidence of an additional role of HLA class III antigens and independence of Ro autoantibodies, Anhritis Rheum 32: 1421-1426, 1989, 127. Siren MK, ./ulkunen H, Kaaja R, Koskimies S: Congenital hean block: HLA differences between affected children and healthy siblings in four Finnish families. APMlS 105: 463--468, 1997. 128. Siren MK, ]ulkunen H, Kaaja R, et al: Role of HLA in congenital heart block: susceptibility alleles in children. I.upus R: 60-67, 1999. 129. Clancy RM, Buyon'/p: Clearance of apoptotic cells: TGF-beta in the balance between inflammation and fibrosis . ./ Leukoc BioI 74: 959-960, 2003. 130. Garchon HJ, Bach ./F: The contribution of non-MHC genes to susceptibility to autoimmune dL'eases. Hum Immunol 32: 1-;0, 1991. 131. Frank MB, McArthur R, Harley ]B, Fujisaku A: Anti-Ro(SSA) autoantibodies are associated with T cell receptor beta genes in systemic lupus erytihematosus patients . ./ Clin Invest 85: 33--39, 1990. 132. Morquio L: Sur une maladie infantile et familiale canK1erisee et la aort subite. Arch Med Enfants 4: 467-475, 1901. 133. East WR, Lumpkin LR: Systemic lupus erythematosus in the newhorn. Minn Med 52: 477-478, 1969. 134. Hogg G: Congenital acute lupus erythematosus associated with subendocardial fibroelastosis. Am./ Clin Pathol 28: 648-654, 1957. 135. Taylor PV, Scott .IS, Gerlis LM, et al: Connective-tissue disease, antibodies to ribonucleoprotein, and congenital hean block. N Engl ./ Med 309: 209-212, 1983. 136. Harley ./B, Kaine .fl., Fox OF, et al: Ro (S5-A) antibody and antigen in a patient with congenital complete hean block. Arthritis Rheum 28: 1321-1325, 1985. 137. Taylor PV, Taylor KF, Norman A, et al: Prevalence of maternal Ro (SS-A) and La (SS-B) autoantibodies in relation to congenital heart block. Br .I Rheumatol 27: 128-132, 1988. D8. ]ulkunen H, Kaaja R, Wallgren E, Teramo K: Isolated congenital heart block: fetal and infant outcome and familial incidence of hean block. Obstet Gynecol 82: 11-16, 1993. 139. Schmidt KG, Ulmer HE, Silverman NH, et al: Perinatal outcome of fetal complete atrioventricular block: a multicenter experience. .I Am Coll Cardiol 17: 1360-1366, 1991. 140. Cecconi M, Renzi R, Bettuzzi MG, et al: Congenital isolated complete atrioventricular block: long-term experience with 38 patients. G Ital Cardiol 23: 39-53, 1993. 141. Friedman RA, Moak JP, Garson A./: Clinical course of idiopathic dilated cardiomyopathy in children. .I Am Coll Cardiol 18: 152-156, 1991. 142. Groves AMM, Allan ill, Rosenthal E: Outcome of isolated congenital complete heart block diagnosed in utero. Br Hean./ 75: 190-194, 1996. 143. Buyon ]P, Hiebert R, Copel], et al: Autoimmune-associated congenital hean block: demographics, mortality, morbidity and recurrence rates obtained from a national neonatal lupus registry. .I Am Coli Cardiol ;1: 1658-1666, 1998. 144. Nield LE, Silverman ED, Taylor GP, et al: Maternal anti-Ro and anti-La antibody-associated endocardial fibroelastosis. Circulation 105: 843--848, 2002. 145. Herreman G, Sauvaget F, Genereau T, Galezowski N: Congenital atrioventricular block and maternal autoimmune diseases. Ann Med Interne (Paris) 141: 234-238, 1990. 146. Carter JB, Blieden I.C, Edwards .IE: Congenital heart block. Arch Pathol 97: 51-57, 1974. 147. Hackel DB: Patihology of primary congenital complete heart block. Mod Pathol I: 114-128, 1988.
404
C HAP T E R
17
NEONATAL
Lupus
ERYTHEMATOSUS
148. Ho SY, Fagg N, Anderson RH, et al: Disposition of the atrioventricular conduction tissues in the heart with isomerism of the atrial appendages: its relation to congenital complete heart block. J Am Coli Cardiol 20: 904-910, 1992. 149. Morales B, Hernandez I, Farru 0, Chuaqui B: Heart cartilage and disruption of the bundle of His in a case with congenital AV-block and corrected transposition. Pathologe 12: 254-258, 1991. 150. Clancy RM, Kapur RP, Molad Y, et al: Immunohistologic evidence supports apoptosis, 19G deposition, and novel macrophage/fibroblast crosstalk in the pathologic cascade leading to congenital heart block. Arthritis Rheum 50: 173-182, 2004. 151. Stevens AM, Hermes HM, Rutledge JC, et al: Myocardial-tissue-specific phenotype of maternal microchimerism in neonatal lupus congenital heart hlock. Lancet 362: 1617-1623, 2003. 152. Miranda-Carus ME, Boutjdir M, Tseng CE, et al: Induction of antibodies reactive with SSA/Ro-SSB/La and development of congenital heart block in a murine model. J Immunol 161: 5886-5892, 1998. 153. Miranda-Carus ME, Askanase AD, Clancy RM, et al: Anti-SSA/Ro and antiSSB/La autoantibodies bind the surface of apoptotic fetal cardiocytes and promote secretion of TNF-alpha by macrophages. J Immunol 165: 5345-5351, 2000. 154. Clancy RM, Askanase AD, Kapur RP, et al: Transdifferentiation of cardiac fibroblasts, a fetal factor in anti-SSA/Ro-SSB/La antibody-mediated congenital heart block. J Immunol 169: 2156--2163, 2002. 155. Logar D, Kveder T. Rozman B, Dobovisek J: Possible association between anti-Ro antibodies and myocarditis or cardiac conduction defects in adults with systemic lupus erythematosus. Ann Rheum Dis 49: 627-629, 1990. 156. Gordon PA, Rosenthal E, Khamashta MA, Hughes GR: Absence of conduction defects in the electrocardiograms [correction of echocardiogramsJ of mothers with children with congenital compiete heart block. J Rheumatol 28: 366--369, 2001. 157. Carpenter RJ Jr, Strasburger JF, Garson A Jr, et al: Fetal ventricular pacing for hydrops secondary to complete atrioventricular block. J Am Call Cardiol 8: 1434-1436, 1986. 158. Walkinshaw SA, Welch CR, McCormack J, Walsh K: In utero pacing for fetal congenital heart block. Fetal Diagn Ther 9: 183-185, 1994. 159. Groves AM, Allan LD, Rosenthal E: Therapeutic trial of sympathomimetics in three cases of complete heart block in the fetus. Circulation 92: 3394-3396, 1995. 160. Yoshida H, Iwamoto M, Sakakibara H, et al: Treatment of fetal congenital complete heart block with maternal administration of beta-sympathomimetics (terbutaline): a case report. Gynecol Obstet Invest 52: 142-144, 2001. 161. Minassian VA, Jazayeri A: Favorable outcome in a pregnancy with complete fetal heart block and severe bradycardia. Obstet Gynecol 100: 1087-1089, 2002. 162. Matsushita H, Higashino M, Sekizuka N, et al: Successful prenatal treatment of congenital heart block with ritodrine administered transplacentally. Arch Gynecol Obstet 267:51-53, 2002. 163. Kiuttu J, Hartikainen AL, Makitalo R, Ruuska P: Congenital heart block with hydrops fetalis treated with high-dose dexamethasone: a case report. Eur J Ohstet Gynecol Reprod Bioi 42: 155-158, 1991. 164. Deloof E, Devlieger H, Van Hoestenberghe R, et al: Management with a staged approach of the premature hydropic fetus due to complete congenital heart block. Eur J Pediatr 156: 521-523, 1997. 165. Copel JA, Buyon JP, Kleinman CS: Successful in utero therapy of fetal heart block. Am J Obstet Gynecol 173: 1384-1390, 1995. 166. Bierman FZ, Baxi L, Jaffe I, Driscoll J: Fetal hydrops and congenital complete heart block: response to maternal steroid. J Pediatr 1988: 646-648, 1988. 167. Buyon JP, Swersky SH, Fox HE, et al: Intrauterine therapy for presumptive fetal myocarditis with acquired heart block due to systemic lupus erythematosus: experience in a mother with a predominance of SS-BiLa) antibodies. Arthritis Rheum 30: 44-49, 1987. 168. Buyon J, Roubey R, Swersky S, et al: Complete congenital heart block: risk of occurrence and therapeutic approach to prevention. J Rheumatol 15: 1104-1108, 1988. 169. Kaaja R, Julkunen H, Ammala P, et al: Congenital heart block: successful prophylactic treatment with intravenous gamma globulin and corticosteroid therapy. Am J Obstet Gynecol 165: 1333-1334, 1991. 170. Wong JP, Kwek KY, Tan JY, Yeo GS: Fetal congenital complete heart block: prophylaXis with intravenous gammaglobuJin and treatment with dexamethasone. Aust N Z J Obstet Gynaecol 41: 339-341, 2001. 171. Eronen M, Siren MK, Ekblad H. et al: Short- and long-term outcome of children with congenital complete heart block diagnosed in utero or as a newborn. Pediatrics 106: 86--89, 2000. 172. Balmer C, Fasnacht M, Rahn M, et al: Long-term follow up of children with congenital complete atrioventricular block and the impact of pacemaker therapy. Europace 4:345-349, 2002. 173. Jaeggi ET, Hamilton RM, Silverman ED, et al: Outcome of children with fetal, neonatal or childhood diagnosis of isolated congenital atrioventricular block. A single institution's experience of 30 years. J Am Coil Cardiol 39: 130-137, 2002. 174. Ercmen M, Heikkila P, Teramo K: Congenital complete heart block in the fetus: hemodynamic features, antenatal treatment, and outcome in six cases. Pediatr Cardiol 22: 385-392, 2001. 175. Moak JP, Barron KS, Hougen TJ, et al: Congenital heart block: deveiopment of late-onset cardiomyopathy, a previously underappreciated sequela. J Am Coil Cardiol 37: 238-242, 2001.
176. Michaelsson M, Riesenfeld T, Jonzon A: Natural history of congenital complete atrioventricular block. Pacing Clin Electrophysiol 20: 2098-2101. 1997. 177. Michaelsson M, Jonzon A, Riesenfeld T: Isolated congenital complete atrioventricular block in adult life: a prospective study. Circulation 92: 442-449, 1995. 178. Friedman RA: Congenital AV block. Pace me now or pace me later? Circulation 92: 283-285, 1995. 179. McCuistion CH, Schoch EP: Possible discoid lupus erythematosus in a newborn infant. Arch Dermatol 70: 782-785, 1954. 180. Franco HL, Weston WL, Peebles C, et al: Autoantibodies directed against sicca syndrome antigens in the neonatal lupus syndrome. J Am Acad Dermatol 4: 67-72, 1981. 181. Cimaz R, Spence DL, Hornberger L, Silverman ED: Incidence and spectrum of neonatal lupus erythematosus: a prospective study of infants born to mothers with anti-Ro autoantibodies. J Pediatr 142: 678-683, 2003. 182. Weston WL, Morelli JG, Lee LA: The clinical spectnun of anti-Ro-positive cutaneous neonatal lupus erythematosus. J Am Acad Dermatol 40: 675-681. 1999. 183. Ng PP, Tay YK, Giam YC: Neonatal lupus erythematosus: our local expelience. Ann Acad Med Singapore 29: 114-118, 2000. 184. Deng J-S, Sontheimer RD, Gilliam ]N: Relationships berween antinuclear antibodies and anti-Ro/SSA antibodies in subacute cutaneous lupus erythematosus. JAm Acad Dermatol 11: 494-499, 1984. 185. Vazquez-Botet M, Rodriguez R, Sanchez JL: Neonatal lupus erythematosus. P R Health Sci J 16: 162-166, 1997. 186. Neiman AR, Lee LA, Weston WL, Buyon JP: Cutaneous manifestations of neonatal lupus without heart block: characteristics of mothers and children enrolled in a national registry. J Pediatr 137: 674-68, 2000. 187. Scheker LE, Kasteler JS, Callen JP: Neonatal lupus erythematosus mimicking Langerhans cell histiocytosis. Pediatr Dermatol 20: 164-166, 2003. 187. Miyagawa S, Kitamura W, YoshiokaJ, Sakamoto K: Placental transfer of anticytoplasmic antibodies in annular erythema of newborns. Arch Dermatol 117: 569-572, 1981. 188. Thornton CM, Eichenfield LF, Shinall EA, et al: Cutaneous telangiectases in neonatal lupus erythematosus. J Am Acad Dermatol 33: 19-25, 1995. 189. High WA, Costner MI: Persistent scarring, atrophy, and dyspigmentation in a preteen girl with neonatal lupus erythematosus. J Am Acad Dermatol 2003; 48: 626-628, 2003. 190. Lee LA: Neonatal lupus erythematosus. J Invest Dermatol 100: 95-13S, 1993. 191. Bielsa I, Herrero C, Collado A, et aJ: Histopathologic findings in cutaneous lupus erythematosus. Arch Dermatol 130: 54-58, 1994. 192, Solomon BA, Laude TA, ShaJita AR: Neonatal lupus erythematosus: discordant disease expression of UlRNP-positive antibodies in fraternal twins-is this a subset of neonatal lupus erythematosus or a new distinct syndrome' JAm Acad Dermatol 32: 858-862, 1995. 193. Dugan EM, Tunnessen WW, Honig PJ, Watson RM: U1RNP antibody-positive neonatal lupus. A report of rwo cases with immunogenetic studies. Arch Dermatol 128: 1490-1494, 1992. 194. Provost TT, Watson R, Gammon WR, et al: The neonatal lupus syndrome associated with UlRNP (nRNP) antibodies. N Engl J Med 316: 1135-1138, 1987. 195. Sheth AP, Esterly NB, Ratoosh SL, et al; U1RNP positive neonatal lupus erythematosus: association with anti-La antibodies? Br J Dermatol 132: 520-526. 1995. 196. Bouderlique C, Debillon T, Mesnard B, et al: Neonatal lupus presenting as telangiectatic and atrophic lesions. Pediatrie 45: 251-254, 1990. 197. Carrascosa JM, Ribera M, Bielsa I, et al: Cutis marmorata telangiectatica congenita or neonatal lupus? Pediatr Dermatol 13: 230-232, 1996. 198. Giam YC: Cutaneous neonatal lupus erythematosus in Chinese neonates. J Singapore Paediatr Soc 34: 39-43, 1992. 199. Kettler AH, Stone MS, Bruce S, TschenJA: Annular eruptions of infancy and neonatal lupus erythematosus. Arch Dermatol 123: 298-299, 1987. 200. Lee LA, Weston WL: Cutaneous lupus erythematosus during the neonatal and childhood periods. Lupus 6: 132-138, 1997. 201. Puig L, Moreno A, Alomar A, de Moragas JM: Erythema gyratum atrophicans transiens neonatale: a variant of cutaneous neonatal lupus erythematosus. Pediatr Dermatol 5: 112-116, 1988. 202. Vaughn RY, GuiJI MA, Cook J: Atrophic plaques in a neonate. Neonatal lupus erythematosus iNLE). Arch Dermatol 128: 683, 686, 1992. 203. Laxer RM, Roberts EA, Gross KR, et al: Liver disease in neonatal lupus erythematosus. J Pediatr 116: 238-242, 1990. 204. Lee LA, Reichlin M, Ruyle SZ, Weston WL: Neonatal lupus liver disease. Lupus 2: 333-338, 1993. 205. Lee LA, Sokol RJ, Buyon JP: Hepatobiliary disease in neonatal lupus: prevalence and clinical characteristics in cases enrolled in a national registry. Pediatrics 109: Ell, 2002. 206. Schoenlebe J, Buyon JP, Zitelli BJ, et al: Neonatal hemochromatosis associated with maternal autoantibodies against Ro/SS-A and La/SS-B ribonucleoproteins. Am J Dis Child 147: 1072-1075, 1993. 207. RoshJR, Silverman ED, Groisman G, et al: Intrahepatic cholestasis in neonatal lupus erythematosus. J Pediatr Gastroenterol Nutr 17: 310-312, 1993. 208. Fonseca E. Contreras F, Garcia-Frias E, Carrascosa MC: Neonatal lupus erythematosus with multisystem organ involvement preceding cutaneous lesions. Lupus 1: 49-50, 1991.
C HAP T E R 209, Watson R, Kang IE, May M, et al: Thrombocytopenia in the neonatal lupus ~yndrome, Arch Dermatol 124: 560-563, 1988. 210, Kleinman D, Katz VL, Kuller JA: Perinatal outcomes in women with systemic lupus erythemalOsus, I Perinatol 18: 178-182, 1998. 211, Lockshin MD, Bonfa E, Elkon K, Druzin ML: Neonatal lupus risk to newborns of mothers with systemic lupus erythemalOsus, Arthritis Rheum 31: (,97-701, 1988, 212, Kurata Y, Miyagawa S, Kosugi S, et al: High-liter antinuclear antibodies, antiSSAIRo antibodies and anti-nuclear RNP antibodies in patients with idiopathic thrombocytopenic purpura, Thromb Haemost 71: 184-187, 1994, 213, Alexander EL, Arnett FC, Provost TT, Stevens MB: Sjogren's syndrome: assodation of anti-Ro(SS-A) antibodies with vasculitis, hematologic abnormalities, and serologic hyperreactivity, Ann Intern Med 98: 155-159, 1983, 214, Watson RM, Braunstein BL, Watson AI, et al: Fetal wastage in women with 3nti-Ro (SSA) antibody, I Rheumatol 13: 90-94, 1986. 215. K)lnagasegar S, Cimaz R, Kurien BT, et al: Neonatal lupus manifests as isolated neutropenia and mildly abnormal liver functions. I Rheumatol 29: 187-191, 2002. 216. Wolach B. Choc L, Pomeranz A, et al: Aplastic anemia in neonatal lupus erythematosus, Am I Dis Child 147: 941-944, 1993, 217. Selander B, Cedergren S, Domanski H: A case of severe neonatal lupus erythematosus without cardiac or cutaneous involvement. Acta Paediatr 87: 10S-107, 1998, 218, Fonseca E. Contreras F, Garcia-Frias E, Carrascosa MC: Neonatal lupus erythematosus with multisystem organ involvement preceding cutaneous lesions. Lupus 1: 49--50, 1991. 219. Kaye EM. Butler IJ, Conley S: Myelopathy in neonatal and infantile lupus erythematosus, I Neurol Neurosurg Psychiatry 50: 923-926, 1987. 220. Wang HS, Kuo MF, Chang TC: Sonographic lenticulostriate vasculopathy in infants: some associations and a hypothesis, AINR Am I Neuroradiol 16: 97-102, 1995. 221. Cabanas F, Pellicer A, Valverde E, et al: Central nervous system vasculopathy in neonatal lupus erythematosus, Pediatr Neurol 15: 124-126, 1996, 222, Nakayama FF, Takigawa M, Iwatsuki K, et al: Hydrocephalus in two female siblings with neonatal lupus erythemalOsus, Arch Dermatol 130: 1210-1212, 1994. 223. IUder LG, Sherry DD, Glass ST: Neonatal lupus erythematosus simulating transient myasthenia gravis at presentation. I Pediatr 118: 417-419, 1991. 224. Inoue K, Fukushige I, Ohno T, et al: Central nervous system vasculopathy associated with neonatal lupus. Pediatr Neurol 26: 68-70, 2002, 225. l'rendiville IS, Cabral DA, Poskitt KI, et al: Central nervous system involvement in neonatal lupus erythematosus, Pediatr Dermatol 20: 60-67, 2003. 226. "lcioglu N, Hall CM: Maternal systemic lupus erythematosus and chondrodysplasia punctata in two sibs: phenocopy or coincidence? I Med Genet .~5: 690-694, 1998. 227. Austin-Ward E, Castillo S, Cuchacovich M, et al: Neonatal lupus syndrome: a case with chondrodysplasia punctata and other unusual manifestations. I Med Genet 35: 695-697, 1998. 228, Westenend PJ: Congenital nephrotic syndrome in neonatal lupus syndrome. I Pediatr 126: 851, 1995. 229. Ruas E, Moreno A, Tellechea 0, et al: Neonatal lupus erythematosus in an infant with Turner syndrome. Pediatr Dermatol 13: 298-302, 1996. 230. Jackson R, Gulliver M: Neonatal lupus erythematosus progressing into systemic lupus erythematosus. Br I Dermatol 101: 81-86, 1979. 231. Fox RI, McCuistion CH, Schoch EP: Systemic lupus erythematosus association with previous neonatal lupus erythemalOsus. Arch Dermatol 115: 340, )979. 232. Martin V, Lee LA, !\skanase AD, et al: Long-term followup of children with neonatal lupus and their unaffected siblings, Arthritis Rheum 46: 2377-2383, 2002. 233. Reichlin M, Friday K, Harley IB: Complete congenital heart block followed by anti-Ro/SSA in adult life. Am I Med 84: 339--344, 1988. 234. McCue CM, Manatakas ME, Tingelstad IB, Ruddy S: Congenital heart block in newborns of mothers with connective tissue disease. Circulation 56: !l2-90, 1977. 235. Waterworth RF: Systemic lupus erythematosus occurring with congenital romplete heart blork. N Z Med I 92: 311-312, 1980. 236. Esscher E, Scott IS: Congenital heart block and maternal systemic lupus erythematosus. Br Med I 1: 1235-1238, 1979. 237. lanham IG, Walport MI, Hughes GR: Congenital heart block and familial connective tissue disease. I Rheumatol 10: 823-825, 1983. 238, Hrucato A, Franceschini F, Gasparini M, et al: Isolated congenital complete ileart block: long-term outcome of mothers, maternal antibody specificity tlhd immunogenetic background, I Rheumatol 22: 533-540, 1995. 239. Julkunen H, Eronen M: Long-term outcome of mothers of children with isolated heart block in Finiand. Arthritis Rheum 44: 647-<552, 2001. 240. lawrence S, Luy L, laxer R, et al: The health of mothers of children with cutaneous neonatal lupus erythematosus differs from that of mothers of children with congenital heart block. Am I Med 108: 705-709, 2000. 241. Fu L~, Hwang B, Lee BH: Newborns of Chinese mother with systemic lupus I'rythematosus (SLE). Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi 33: ;i41-349, 1992, 242. Leu LY, Lan JL: The influence on pregnancy of anti-SSAIRo antibodies in systemic lupus erythematosus. Zhonghua Min Guo Wei Sheng Wu Ii Mian Yi XUI' Za Zhi 25: 12-20, 1992.
17
NEONATAL
Lupus
ERYTHEMATOSUS
405
243. Brucato A, Frassi M, Franceschini F, et al: Risk of congenital complete heart block in newborns of mothers with ami-Ro/SSA antibodies detected by counterimmunoelectrophoresis: a prospective study of 100 women. Arthritis Rheum 44: 1832-1835, 2001. 244, Gladman G, Silverman ED, Yuk L, et al: Fetal echocardiographic screening of pregnancies of mothers with anti-Ro andlor anti-La antibodies. Am I Perinatol 19: 73-80, 2002. 245. Alexander EL, Buyon IP, Lane I, et al: Anti-SS-AiRo SS-B/La antibodies bind to neonatal rabbit cardiac cells and preferentially inhibit in vitro cardiac repolarization. I Autoimmun 2: 463-469, 1989. 246, Alexander E, Buyon IP, Provost TT, Guarnieri T: Anti-Ro/SS-A antibodies in the pathophysiology of congenital heart block in neonatal lupus syndrome, an experimental model. In vitro electrophysiologic and immunocytochemical srudies. Arthritis Rheum 35: 176-189, 1992. 247. Deng j-S, Bair LW}, Shen-Schwarz S, et al: Localization of Ro(SS-A) antigen in the cardiac conduction system. Arthritis Rheum 30: 1232-1238, 1987. 248, Lee LA, Harmon CE, Huff IC, et al: The demonstration of SS-AiRo antigen in human fetal tissues and in neonatal and adult skin. J Invest Dermatol 85: 143-146, 1985. 249. Tseng CE, Miranda E, Di Donato F, et al: mRNA and protein expression of SSAIRo and SSBILa in human fetal cardiac myocytes cultured using a novel application of the Langendorff procedure. Pediatr Res 45: 260-269, 1999. 250. Qu Y, Xiao GQ, Chen L, Boutjdir M: Autoantibodies from mothers of children with congenital heart block downregulate cardiac L-type Ca channels, J Mol Cell Cardiol 33: 1153-1163, 2001. 251. Xiao GQ, Hu K, Boutjdir M: Direct inhibition of expressed cardiac L- and T-rype calcium channels by IgG from mothers whose children have congenital heart block. Circulation 103: 1599--1604, 2001. 252. Eftekhari P, Roegel IC, Lezoualc'h F, et al: Induction of neonatal lupus in pups of mice immunized with synthetic peptides derived from amino acid sequences of the serotoninergic 5-HT4 receptor, Eur I Immunol 31: 573-579, 2001. 253. Buyon JP, Clancy R, Di Donato F, et al: Cardiac 5-HH4) serotoninergic receptors, 52kD SSAIRo and autoimmune-associated congenital heart block. J Autoimmun 19: 79-86, 2002. 254, Eftekhari P, Roegel IC, Lezoualc'h F, et al: Induction of neonatal lupus in pups of mice immunized with synthetic peptides derived from amino acid sequences of the serotoninergic 5-HT4 receptor. Eur J Immunol 31: 573-579, 2001. 255. Borda E, Leiros CP, Bacman S, et al: Sjogren autoantibodies modify neonatal cardiac function via Ml muscarinic acetylcholine receptor activation. Int I Cardiol 70: 23-32, 1999, 256. Borda E, Sterin-Borda L: Autoantibodies against neonatal heart Ml muscarinic acetylcholine receptor in children with congenital heart block. J Autoimmun 16: 143-150, 2001. 257. Furukawa F, Lyons MB, Lee LA, et al: Estradiol binding to cultured human keratinocytes of antibodies specific for S5-AlRo and 5S-B/La. I Immunol141: 1480-1488, 1988. 258. Furukawa F, Imamura S, Norris DA: Stimulation of anti-RNP antibody binding to cultured keratinocytes by estradiol. Arch Dermatol Res 283: 258-261, 1991. 259. LeFeber WP, Norris DA, Ryan SR, et al: Ultraviolet light induces binding of antibodies to selected nuclear antigens on cultured keratinocytes. I Clin Invest 74: 1545-1551, 1984. 260. Wang D, Chan EK: 17-Beta-estradiol increases expression of 52-kDa and 60-kDa SS-AiRo autoantigens in human keratinocytes and breast cancer cell line MCF-7, I Invest Dermatol 107: 610-614, 1996. 261. Domer T, Chaoui R, Feist E, et al: Significantly increased maternal and fetal IgG autoantibody levels to 52 kD Ro (S5-A) and La(SS-B) in complete congenital heart block. J Autoimmun 8: 675-684, 1995. 262, Lee LA, Weston WL, Krueger GG, et al: An animal model of antibody binding in cutaneous lupus. Arthritis Rheum 29: 782-788, 1986. 263. Yu HS, Chiang Le, Chang CH, et al: The cytotoxic effect of neonatal lupus erythematosus and maternal sera on keratinocyte cultures is complementdependent and can be augmented by ultraviolet irradiation. Br J Dermatol 135: 297-301, 1996. 264. Garcia S, Nascimento IH, Bonfa E, et al: Cellular mechanism of the conduction abnormalities induced by serum from anti-Ro/SSA-positive patients in rabbit hearts, J Clin Invest 93: 718-724, 1994. 265. Viana VS, Garcia 5, Nascimento IH, et al: Induction of in vitro heart block is not restricted to affinity purified anti-52 kDa RolSSA antibody from mothers of children with neonatal lupus, Lupus 7: 141-147, 1998. 266. de Oliveira SF, Pedrosa RC, Nascimento IH, et al: Sera from chronic chagasic patients with complex cardiac arrhythmias depress electrogenesis and conduction in isolated rabbit hearts. Circulation 96: 2031-2037, 1997. 267. Boutjdir M, Chen L, Zhang ZH, et al: Arrhythmogenicity of IgG and anti-52kD SSAIRo affinity-pUrified antibodies from mothers of children with congenital heart block. Circ Res 80: 354-362, 1997. 268. Boutjdir M, Chen L, Zhang ZH, et al: Serum and immunoglobulin G from the mother of a child with congenital heart block induce conduction abnormalities and inhibit L-type calcium channels in a rat heart model. Pediatr Res 44: 11-19, 1998.
406
C HAP T E R
17 NEONATAL Lupus ERYTHEMATOSUS
269. Miranda-Carus ME, Boutjdir M, Tseng CE, et al: Induction of antibodies reactive with SSA/Ro-SSB/La and development of congenital heart block in a murine model. J Immunol 161: 5886--5892, 1998. 270. Clancy RM, Askanase AD, Kapur RP, et al: Transdifferentiation of cardiac fibroblasts, a fetal factor in anti-SSA/Ro-SSB/La antibody-mediated congenital heart block. J lmmunol 169: 2156--2163, 2002. 271. Tran HB, Macardle PJ, Hiscock,/, et al: Anti-La/SSB antibodies transported across the placenta bind apoptotic cells in fetal organs targeted in neonatal lupus. Arthritis Rheum 46: 1572-1579, 2002.
272. Clancy RM, Buyon ,/P: Clearance of apoptotic cells: TGF-beta in the balance between inflammation and fibrosis. J Leukoc BioI 74: 959-960, 2003273. Miranda ME, Tseng CE, Rashbaum W, et al: Accessibility of SSA/Ro and SSB/La antigens to maternal autoantibodies in apoptotic human fetal cardiac myocytes. J Immuno! 161: 5061-5069, 1998. 274. Tran HB, Macardle P,/, Hiscock,/, et al: Anti-La/SSB antibodies transportetl across the placenta bind apoptotic cells in fetal organs targeted in neonatal lupus. Arthritis Rheum 46: 1572-1579,2002.
18
C H A P T E R
J UVENILE D ERMATOMYOSITIS James T. Cassidy and Carol B. Lindsley
Juvenile dermatomyositis (JDM) is a multisystem disease of uncertain origin that results in nonsuppurative inflammation of striated muscle and skin. It is characterized early in its course by an immune complex vasculitis of varying severity and later by development of calcinosis.
HISTORICAL REVIEW The clinical presentation of JDM was described by four different investigators in 1887.1–4 Unverricht4 clarified the cutaneous and muscular manifestations of the disease, introducing the term dermatomyositis. He recognized that the childhood form of the disease was not always fatal, although subsequent reports emphasized the poor prognosis. The clinically distinctive features of JDM were not detailed until some time later.5–14 Experience with this disease in the era before steroids was reported by Karelitz and Welt (22 children),5 Scheuermann (47 children),15 Hecht (5 children),16 and Selander (22 children).17 The first postmortem study that described the classic histopathologic features of the disease in a child was by Batten in 1912.18 Pearson19 recognized the uniqueness of this disorder in children and distinguished it from that in adults in his classification in 1966. In 1953, Wedgwood and colleagues6 reviewed data on 26 children treated with glucocorticoids, and in 1963, Cook and colleagues20 reported 15 deaths among 50 children and emphasized the therapeutic role of these agents for treatment. Sullivan and colleagues11,13 in 1972 and 1977 and Rose12 in 1974 stressed the importance of an adequately high initial dose and sufficiently long therapeutic course to reverse the dismal prognosis of juvenile dermatomyositis.
Definition and Classification In childhood, chronic idiopathic inflammatory myositis is a relatively heterogeneous disorder, although almost all affected children have the characteristic skin and muscle abnormalities of JDM.21–26 The five criteria in Table 18–1 are applicable to its diagnosis, although their sensitivity and specificity have not been validated in children but are probably 45% to 90% and 90%, respectively. A diagnosis of JDM requires the presence of the pathognomonic rash and two of the other criteria.27–29 In general, the first two criteria (i.e., proximal muscle weakness and classic rash) are almost always present; criterion 3 (i.e., elevated serum levels of muscle enzymes), 4 (i.e., electromyographic changes), and 5 (i.e., histopathologic
changes) provide additional laboratory support for the diagnosis. A diagnosis of JDM is not necessarily excluded by failure to meet one or more of these criteria, except that related to the dermatitis. JDM differs in a number of respects from inflammatory myositis in adults (Fig. 18–1). Other types of inflammatory myositis, including polymyositis (i.e., muscle inflammation without cutaneous disease)30,31 and dermatomyositis with other systemic diseases such as malignancy,32–35 are rare in childhood. Myositis may occur in other connective tissue diseases,36–38 such as scleroderma (see Chapter 19) and the overlap syndromes (see Chapter 21). Other types of myositis, rare or not occurring in children, include inclusion body myositis,31,39 focal myositis,40 and eosinophilic myositis.41
EPIDEMIOLOGY Incidence Estimates of the incidence of JDM are given in Table 18–2.42–47 Rose and Walton48 calculated the incidence of dermatomyositis and polymyositis in Great Britain in 1966 at 0.4 per 100,000 persons in the general population. This is similar to the frequency in the United States of 0.542 and 0.55 per 100,00043 and to the rate of 0.44 per 100,000 in studies from Israel,49 in which the incidence for Jewish children was 0.7 per 100,000 for those 0 to 9 years old and 1.78 per 100,000 for those 10 to 19 years old. Symmons and colleagues44 calculated a rate of 0.19 per 100,000 for children younger than 16 years in the United Kingdom and Ireland. Mendez and colleagues47 calculated rates of 0.25 to 0.41 for children 2 to 17 years of age from 1995 to 1998 in the United States from two national registries. In general, 16% to 20% of patients with dermatomyositis have onset in childhood.42,50 There are no reliable data on prevalence.
Age at Onset and Sex Ratio The data of Medsger and coworkers42 suggested a bimodal distribution of ages at onset with a peak in the 5- to 14-year-old range, and a second, much larger peak
407
408
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S divergent geographic areas are illustrated in Figure 18–2. In these pooled data, the female-to-male ratio was 1.7:1. This ratio was 2.7:1 or higher among children with onset at 10 years of age.30,38,44,45,51–53 It was reversed, however, in a review of 25 Arab children.54 In the Israel study,49 the ratio was 3:1 in the youngest cohort, and 2:3 in the older one. The average age at onset was 7 years.11,13,55 Onset was especially common from the 4th to the 10th year, with two peak ages for girls at 6 years and another at 11 years; for boys the most common age at onset is 6 years. All the boys were younger than 10 years at onset of the disease.44
TABLE 18–1 Criteria for a Diagnosis of Juvenile Dermatomyositis 1. Symmetric weakness of the proximal musculature 2. Characteristic cutaneous changes consisting of heliotrope discoloration of the eyelids with periorbital edema and an erythematous, scaly rash over the dorsal aspects of the metacarpophalangeal and proximal interphalangeal joints (i.e., Gottron’s papules) 3. Elevation of the serum level of one or more of the skeletal muscle enzymes: creatine kinase, aspartate aminotransferase, lactate dehydrogenase, and aldolase 4. Electromyographic demonstration of the characteristics of myopathy and denervation 5. Muscle biopsy documenting histologic evidence of necrosis and inflammation
Geographic and Racial Distribution Adapted from Bohan A, Peter JB: Polymyositis and dermatomyositis. N Engl J Med 292: 344, 403, 1975.
Dermatomyositis is widely distributed throughout the world.56 Striking racial differences in incidence have been described in adults in the United States, with the frequency of the disease among 55- to 64-year-old black women 10 times that of white women in the same age group.42 Such differences are less marked in children, and in the data of Mendez and colleagues in the United States,47 the average 4-year annual rate was comparable for whites and blacks and somewhat lower for Hispanic patients.
in the 45- to 64-year-old range (Fig. 18–2).42,49 Such a distinctly bimodal distribution underscores the clinical heterogeneity of this disorder and the uniqueness of the childhood form. Data from nine series derived from
ETIOLOGY AND PATHOGENESIS Most studies suggest that JDM is an autoimmune angiopathy (Table 18–3).57–59 Cell-mediated immunity to muscle antigens (i.e., activated T cells) and immune complex disease may participate in pathogenesis.60 Response of patients’ lymphocytes in vitro to allogeneic or autologous muscle extracts has been described in some reports,61–63 but not all.64 Some reports have described a ■ Figure 18–1 Incidence of inflammatory myositis. TABLE 18–2 Incidence of Juvenile Dermatomyositis Population United States, 1970 0–4 yr 5–9 yr 10–14 yr United States, 1982 PM (n = 17) JDM (n = 26) United States, 1990 (Pennsylvania) White female (n = 14) White male (n = 5) Black female (n = 1) Black male (n = 1) United Kingdom and Ireland, 1995 PM (n = 3) JDM (n = 48) Finland, 1996 JDM (n = 4) Japan, 1997 PM/JDM United States, 1995–1998 White Black Hispanic JDM, juvenile dermatomyositis; PM, polymyositis.
Study
Incidence/100,000 (range)
Female/Male
42
Medsger et al
Hanissian et al30
0.06 0.37 0.43 0.32
3:1 1:1.3 4.7:1 4.7:1
Oddis et al43
0.8 1.1 0.4 0.7 0.9
(0.01–1.59) (0.01–2.19) (0–1.8) (0–1.4) (0.01–1.75)
Symmons et al44
2.5:1 2.8:1 1:1 5:1
0.19 (0.14–0.26) Kaipiainen et al45 0.5 Fujikawa et al46 Mendez et al47
0.16 0.32 (0.29–0.34) 0.34 0.33 0.27
2.3:1
C H A P T E R
■ Figure 18–2 Age at onset for girls (dashed line) and boys (solid line).
cytotoxic effect on muscle monolayers in culture by lymphocytes,65–70 although the effect has not been reported uniformly.71 In one study, peripheral blood lymphocytes from 4 of 5 children with active disease produced a lymphotoxin that caused necrosis or impaired protein synthesis in monolayers of human fetal muscle in the presence of homogenates of autologous muscle.66 Normal muscle cells do not express major histocompatibility class (MHC) I antigens, but in dermatomyositis, these antigens are strongly expressed.72–75 One report suggests that dysregulation of apoptosis in myofibrils, as suggested by the observation of overexpression of BCL2 protein in muscle specimens from children with this disease, are operative in pathogenesis.76 An immune complex–mediated vasculitis may be an important initiating or perpetuating event.77–81 Complement activation and immune complex deposition have been demonstrated.81,82 Whitaker and Engel77 identified
18 J U V E N I L E D E R M AT O M Y O S I T I S
409
immunoglobulin and complement in vessel walls of skeletal muscle in juvenile and adult dermatomyositis, although the frequency and intensity of deposition were more pronounced in children. Immunoglobulin (Ig) G, IgM, and the third component of complement (C3) were deposited alone or in combination in this study and others.80 Elevated plasma levels of factor VIII–related antigen,83 fibrinopeptide A, and C3 provide additional evidence of endothelial cell injury that may be induced by immune complexes, although antibodies that directly stimulate or are toxic to endothelial cells may also be important.84 Some data suggest that increased circulating concentrations of thrombospondin-1, a potent anti-angiogenic factor, may play a role in susceptibility to vascular thrombosis in JDM.85 Reports have suggested a role for soluble adhesion molecules in pathogenesis.86,87 Interleukin-1 (IL-1) expression is increased in capillary endothelial cells.74 Polymorphisms in the IL-1 receptor antagonist gene may be additional risk factors.88 Plasma levels of cytokines or their receptors (i.e., IL-1Ra, sIL-2R, IL-10, and soluble tumor necrosis factor receptors [sTNFR] p55 and p75) were significantly elevated in active disease.89 A number of chemokines are overexpressed in muscle by at least 15-fold.90 Maternal cell chimerism is present in peripheral blood mononuclear cells and in muscle tissue in 80% to 90% of children with JDM.91–93 It was identified in 25% of siblings and 15% of controls in these reports. Studies by Artlett and colleagues91,94 on microchimerism suggested that maternal cells persisting in the peripheral blood of children (19 of 26 with JDM, 2 of 21 healthy controls) may be involved in pathogenesis. Human leukocyte antigen (HLA) alleles may control the frequency and potential pathogenic mechanisms of this phenomenon. Reed and colleagues93 assessed immunologic activity in chimeric cells in 60 of 72 children with JDM, in 11 of 48 unaffected siblings, and in 5 of 29 healthy controls. In all groups, chimerism was associated with the maternal HLADQA1*0501 allele. Chimeric cells were reactive against the
TABLE 18–3 Etiology and Pathogenesis of Juvenile Dermatomyositis Potential Pathogenic Mechanism
Evidence
Abnormalities of cell-mediated immunity
Patient’s lymphocytes proliferate in vitro in presence of muscle cells Patient’s lymphocytes are cytotoxic to human fetal or animal myocytes in vitro Patient’s lymphocyte supernatants (lymphokines) are cytotoxic to myocytes in culture
Immune complex disease
Presence of circulating immune complexes in patient’s sera Immunoglobulin and complement deposited in vascular endothelium
Association with immunodeficiency
Occurrence of disease in children with hypogammaglobulinemia, selective IgA deficiency, or C2 complement component deficiency
Relation to infection
Myxovirus-like tubuloreticular structures in endothelial cells in muscle biopsy specimens Occurrence of limited myositis in association with influenza Increased coxsackievirus B titers in children with JDM (?) Increased toxoplasmosis titers in children with JDM (?)
Genetic predisposition
HLA–DRB1*0301 and DQA1*0501 Single nucleotide polymorphisms in tumor necrosis factor-α promoter region
410
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
host’s cells with an inflammatory memory response that reflected previous anti-host cell reactivity. The HLA maternal genotype influenced the fate of the transferred cells and activation of the chimeric cells. Reports of geographic and seasonal clustering of cases support the possibility of an environmental factor in causation.44,54,95 Evidence for the participation of infectious agents is for the most part indirect.96 Onset has been preceded by upper respiratory infections (e.g., coxsackievirus, influenza, streptococcus), occurring an average of 3 months before onset, and with a number of other organisms (e.g., toxoplasmosis, parvovirus, hepatitis B, Borrelia, leishmania).22,38 However, only 24 of 139 children in a multicenter study had preceding infections.38 Although acute muscle inflammation may result from viral illness, there are no unequivocal data supporting a viral cause for JDM.97,98 Serologic evidence of coxsackievirus B infection was reported in 83% of children with early JDM, compared with 25% of control subjects,99,100 but a search for the presence of viral genome in cells from patients with JDM using the polymerase chain reaction was unsuccessful.101 Acute transitory myositis may occur after influenza infection.102–105 Children with JDM have been reported to express IgG antibodies against viral non–syncytium-inducing protein, perhaps induced by persistent parvovirus B19 infection.106 Enteroviral titers are similar to control groups.54 Dermatomyositis has also followed immunizations (e.g., rubella, bacillus Calmette-Guérin),107–111 Relapse with β-hemolytic streptococcal disease may be related to molecular mimicry between the M5 protein and skeletal muscle myosin and immune responses to homologous peptide regions.38,112–114 Toxoplasma gondii was demonstrated in muscle in one patient with JDM.115 Elevated antibody titers to toxoplasma have been reported in some studies,116,117 but appropriate environmental controls were lacking. A dermatomyositis-like disease has been described in a few children with agammaglobulinemia in association with echovirus infection84,118 and occasionally in patients with selective immunoglobulin A (IgA) deficiency119,120 or deficiency of the second component of complement (C2), all children in whom an inordinate susceptibility to infection might be anticipated121 (see Chapter 35). Electron microscopic examination of muscle has demonstrated tubuloreticular structures within endothelial cells that resembled the myxovirus-like particles identified in patients with systemic lupus erythematosus (SLE).122 This finding, however, may be artifactual and reflect degenerative or regenerative alterations in cytoplasmic constituents of endothelial cells (Fig. 18–3).123,124 A study of muscle and peripheral blood mononuclear cells from children with JDM demonstrated the presence of tubuloreticular structures and cylindrical confronting cisternae in all six patients studied,125 probably representing the effect of interferon-α, which could be induced by viral infections. Other reports have confirmed these paramyxovirus-like particles in affected muscle126,127 and indicated the possible presence of picornavirus128 or coxsackievirus.129 Experimental myositis can be induced by the injection of muscle homogenates in Freund’s adjuvant in guinea pigs,130 rabbits,131 and rats.132 Although this disorder was different from JDM, cell-mediated immunity to muscle was detected, and the disease could be transferred by lymphocytes.133–135 It has not been possible, however, to reproduce in animals the characteristic vasculopathy of JDM. Experimental induction of a polymyositis-like syndrome in neonatal animals has been reported after inoculation with Semliki Forest virus136 and Ross River virus137 and in mice with coxsackievirus138–140 and encephalomyocarditis virus.141 A familial dermatomyositislike disease has been described in household cats and dogs.142,143
■ Figure 18–3 A, Heliotrope discoloration and violaceous suffusion with edema of the upper eyelids in an 11-year-old with acute dermatomyositis. B, Erythematous, scaly rash in a malar distribution. (See color insert.)
GENETIC BACKGROUND Familial Dermatomyositis There are several reports of the rare occurrence of familial JDM.6,144–151 In all instances but one, the disease has been typical dermatomyositis. In one daughter-father pair,146 the daughter had JDM, and the father had polymyositis with positive lupus erythematosus cell preparations. The twin girls reported by Harati and associates147 developed JDM within 2 weeks of each other after upper respiratory tract infections. A brother-sister pair was reported.152 There is an increased frequency of other autoimmune diseases in families of children with JDM.149
Human Leukocyte Antigen Relationships Despite the infrequency of familial dermatomyositis, there may be an immunogenetic predisposition to this disease marked by the presence of HLA-B8 and HLADR3.153–155 Friedman and colleagues154 studied HLA antigens in 65 children with JDM: B8 was present in 43% (21% of control subjects) and DR3 (DRB1*0301) in 57% (30% of control subjects). The prevalence of B8 is com-
C H A P T E R
monly increased among patients with other connective tissue or autoimmune diseases such as SLE, and its increased frequency in JDM is therefore not unexpected. MHC class I overexpression on muscle cells has occurred in the absence of histologic abnormalities.75 An association with DQA1*0501 in white children41,96,59,156 and other ethnic groups (i.e., African Americans and Hispanics) has been published.157 A study of Czech children, however, did not demonstrate an HLA association.158 The frequency of MHC class II–associated DM molecules (DMA*0103 and DMB*0102) also is increased.159 A number of distinct HLA associations occur with myositis-specific antibodies.160,161 Data on mannose-binding lectin (MBL) polymorphisms confirm an association of decreased MBL and dermatomyositis in adults, suggesting a pathogenic role for this protein.162 DQA1*0501 is associated with JDM in white children.96 It is also present in increased frequency in other necrotizing myopathies such as Duchenne muscular dystrophy. Increased expression of interferon (IFN)-αβ–inducible genes 6 to 16, myxovirus resistance protein p78, latent cytosolic transcription factor, LMP2, and TAP196 support a model of antigen (viral) induction of autoimmune disease in genetically susceptible children. The TNF-α-308A allele (A for G polymorphism in the promoter region) is also overrepresented in white children with JDM (18 of 37 patients, 5 of 29 controls).163,164 It is associated with a prolonged disease course of occlusive capillaropathy and calcinosis requiring immunosuppressive therapy (76% versus 13%).165 In these patients, there is also increased production of TNF-α by peripheral blood mononuclear cells in vitro and by regenerating JDM muscle fibers in vivo.166,167
CLINICAL MANIFESTATIONS Classic JDM presents with an insidious progression of malaise, easy fatigue, muscle weakness, fever, and rash that may predate diagnosis by 3 to 6 months (Table 18–4).10–13,158,168–175 There is, however, great variation in the rapidity of evolution of the clinical manifestations. Onset is usually insidious with development of progressive muscle weakness and pain; a more acute onset occurs in approximately one third of children. Children with myositis-specific and myositis-associated antibody seropositivity characteristically present with distinct phenotypic disease.
TABLE 18–4 Frequency of Manifestations of Juvenile Dermatomyositis at Onset Manifestation Easy fatigue Progressive proximal muscle weakness Classic rash Fever Muscle pain or tenderness Lymphadenopathy Arthritis Hepatomegaly Splenomegaly Nonspecific rash Dyspnea Dysphagia
Frequency (%) 80–100 16–96 35–85 50–80 30–80 50–75 10–65 10–20 10–15 10–15 5–15 5–9
18 J U V E N I L E D E R M AT O M Y O S I T I S
411
Constitutional Signs and Symptoms The onset of JDM is usually characterized by fever in the range of 38º C to 40º C. An affected child often complains of ease of fatigue, which probably represents muscle weakness. Malaise, anorexia, and weight loss follow. Parents may report that the child had become irritable, with alterations in gross motor function, or regression of motor milestones.
Musculoskeletal Disease Muscle weakness at onset is predominantly proximal, and complaints related to weakness of the limb girdle musculature of the lower extremities are most common. Weakness of the anterior neck flexors and back muscles leads to inability to hold the head upright or maintain a sitting posture. The child may stop walking, be unable to dress or climb stairs, or wash the face or comb hair. The affected child may also complain of moderate muscle pain or stiffness. Physical examination demonstrates symmetric weakness that is maximal in the proximal muscles of the shoulders and hips, in the neck flexors, and in abdominal musculature. Affected muscles are occasionally edematous and indurated and may be tender. Functional muscle examination may demonstrate that the child is unable to arise from a supine position without rolling over, move from sitting to standing, get out of bed without assistance, or is unable to squat or rise from a squatting position without help. Gowers’ sign is often present. The child with weakness of the pelvic girdle musculature has difficulty climbing or descending stairs. The Trendelenburg sign indicates weakness of the hip abductors. Later in the disease or in children with an especially severe course, the distal muscles of the extremities may become involved. Occasionally, the disease is so severe that the child is unable to move at all. Although not as clinically dramatic, the distal muscles of the extremities are involved in many patients. Although muscle weakness may be impressive, the deep tendon reflexes are usually well preserved. In severely affected children (approximately 10%), pharyngeal, hypopharyngeal, and palatal muscles are affected. Difficulty swallowing may be related to this involvement or to esophageal hypomotility.176 Dysphonia, weakness of the voice, nasal speech, or regurgitation of liquids through the nose may be signs of impending difficulties. The threat of aspiration is increased in these children. Subtle or even asymptomatic dysfunctional swallowing can be demonstrated on barium swallow in up to 67%.177 Sequential musculoskeletal examinations by an experienced physician or physical therapist should be recorded using a standard scale.178,179 The importance of this examination becomes even more critical later during the course of the disease when serum levels of muscle enzymes may be less dependable indicators of disease activity. Selected muscle groups that should always be evaluated are the neck flexors and extensors; shoulder abductors; elbow flexors and extensors; hip flexors, extensors, adductors, and abductors; and knee flexors and extensors. Assessment of function can also be documented with the Childhood Myositis Assessment Scale (CMAS), the Childhood
412
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
Health Assessment Questionnaire, or the Disease Activity Scale (includes skin assessment).180–183
Some children with JDM have arthralgia or subtle arthritis that is transient and nondeforming, sometimes accompanied by tenosynovitis or flexor nodules. Early development of flexion contractures, particularly at the knees, hips, and shoulders, is common and usually represents the effects of muscle inflammation rather than synovitis. The presence of significant, persistent arthritis in a child with myositis and the skin changes of dermatomyositis suggests the possibility of an overlap syndrome such as mixed connective tissue disease. Reports document, however, the presence of arthritis in 35% of 79 children with JDM.184 Another study identified oligoarthritis in 67% and polyarthritis in 33% of 80 children,185 frequencies that are much higher than was previously appreciated.
Mucocutaneous Disease In three fourths of the children, the cutaneous abnormalities are pathognomonic of the disease; a less characteristic rash occurs in the remainder. Occasionally, dermatitis is the first manifestation of the disorder, although asymptomatic (undiagnosed) muscle weakness often occurs in such children. More often, the cutaneous abnormalities become evident in the first few weeks after the onset of muscle symptoms. The three most typical cutaneous manifestations are heliotrope discoloration of the upper eyelids, Gottron’s papules, and periungual erythema and capillary loop abnormalities. The classic heliotrope dermatitis occurs over the upper eyelids as a violaceous, reddish purple suffusion that often is associated with a malar rash that resembles that of SLE in its distribution but is less well demarcated (Fig. 18–3). Edema of the eyelids and face often accompanies this dermatitis and may be extensive. The symmetric changes over the extensor surfaces of joints (i.e., Gottron’s papules) tend to be associated with shiny, erythematous, atrophic, scaly plaques (Fig. 18–4). These atrophic areas of skin have a bright pink-red appearance. Occasionally, the lesions appear to be thickened and pale early in disease (hence the name collodion patches). Gottron’s papules are especially common over the proximal interphalangeal joints of the hands and less so over the metacarpophalangeal and distal interphalangeal joints. The skin over the toes is rarely, if ever, affected. The extensor surfaces of the elbows and knees and, less frequently, the malleoli may also be involved. Similar rashes may cover the entire extensor surface of the limbs and the trunk. Characteristic abnormalities of the periungual skin and capillary bed are present in 50% to 100% of children. The periungual skin is often intensely erythematous, and careful examination with the naked eye or the 40+ lens of an ophthalmoscope documents the presence of telangiectases. Dilatation of isolated loops, thrombosis and hemorrhage (often visible), dropout of surrounding vessels,184 and arborized clusters of giant capillary loops are distinctive, if not pathognomonic, for JDM
■ Figure 18–4 A, Gottron’s papules. B, Symmetrical, scaly, erythematous papules over the metacarpophalangeal and proximal interphalangeal joints of the hand of an 8-year-old girl. (See color insert.)
(Figs. 18–5 to 18–7). There is often associated marked cuticular overgrowth. Similar changes occur in other connective tissue diseases,186 especially systemic scleroderma, but are seldom as dramatic as those seen in the child with JDM. These abnormalities, like the noninflammatory vasculopathy, correlate with a more severe chronic disease course, cutaneous ulceration, or the development of calcinosis.187–190 The acute nail fold abnormalities may abate with remission. Cutaneous involvement can vary from the slightest erythematous tinge over the knuckles or eyelids to a generalized pruritic, scaly rash. At onset, edema and induration of skin, periorbital regions, and subcutaneous tissues are common; less frequently, the extremities and trunk are affected.191 Extensive rashes often
■ Figure 18–5 Normal nail fold capillary pattern (magnification × 100). (Courtesy of Dr. Jay Kenik.)
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
413
Dermatomyositis sine myositis or amyopathic dermatomyositis is rare in children,198–202 although the classic rash of JDM may occur in the absence of clinical muscle involvement,203 or muscle disease may not be initially documented.204 Do most of these children eventually develop overt myositis if followed for long enough? In the review by Plamondon and Dent,204 none of 27 patients developed clinical myopathy at a mean follow-up of 32.8 months. Magnetic resonance imaging (MRI) may detect subtle muscle abnormalities in these patients, but current reports are inadequate to evaluate this possibility.
Calcinosis
■ Figure 18–6 Abnormal nail fold capillary pattern with the classic changes of dermatomyositis.The capillaries are thickened and tortuous, with the typical peripheral bushy pattern. (Courtesy of Dr. Jay Kenik.)
exhibit an infiltrative character with hyperemic borders in addition to the acute intradermal and subcutaneous edema. Photosensitivity occurs in up to one third of patients.172,192 Sun exposure has been associated with onset of the disease and exacerbations. Gingival and buccal ulcerations develop in 10% to 46% of children and are associated with pain on swallowing.193,194 Oral ulcers may precede or accompany the onset of the disease, and components of the sicca syndrome may be present. Cutaneous ulceration occurred relatively frequently in the series of Crowe and coworkers55 and was associated with severe and prolonged disease in 11 of 42 patients. Vasculitic ulcers at the corners of the eyes, in the axillae, over the elbows or pressure points, and over stretch marks may become a serious problem of management (Figs. 18–8 and 18–9). Children with a generalized rash and cutaneous ulcerations may have the poorest prognosis,55,195 but this is not always the case.196 Late in the course, other cutaneous and subcutaneous changes occur. Thinning and atrophy of the skin and supporting structures may supervene, and alterations in pigmentation become more common. Although individual lesions may be hypopigmented, the child may exhibit generalized hyperpigmentation. Hyperkeratosis is relatively common and may occur in unusual areas such as the palms and the soles40 and the infrapatellar area.197
Dystrophic calcification (i.e., basic calcium phosphate, hydroxyapatite or fluoroapatite crystal formation) occurs in up to 40% of children, sometimes within 6 months onset, but rarely before symptomatic myositis.205 Calcium deposition may occur in subcutaneous plaques or nodules (Table 18–5) (Fig. 18–10), as large tumorous clumps in muscle groups or bridging joints (Fig. 18–11), as calcification within fascial planes, or as an extensive subcutaneous exoskeleton (Fig. 18–12).195,206 Calcinosis affecting the subcutaneous tissues can result in painful superficial ulceration of the overlying skin with recurrent extrusion of small flecks of calcium salts. These deposits may slowly resolve with time (Fig. 18–13). If deposition in subcutaneous tissues, along fascial planes, and within muscles is extreme, the child may literally be encased within a shell of calcium salts. This type of calcinosis is unlikely to resolve completely, and it results in severe disability. Aggressive treatment to achieve rapid and complete control of inflammation, especially early after onset, may minimize calcinosis.207,208
Vasculitis Visceral vasculitis occurs in a minority of children, usually soon after onset of the disorder. It signifies a poor prognosis and sometimes rapidly leads to death.27,28,55,209 This complication is characterized by diffuse abdominal pain, pancreatitis, melena, and hematemesis, which represent diffuse vasculitis of the mucosa of the gastrointestinal tract or an acute mesenteric infarction.210–214 Free intraperitoneal air radiographically indicates the presence of a perforation of the gastrointestinal tract. Multiple perforations of the duodenum are particularly difficult to recognize.210,212–214 Vasculitis of the gallbladder, urinary bladder, uterus, vagina, and testes can also occur.55,215 Anasarca, cholestasis, and pneumatosis intestinalis have been reported. Widespread vascular disease can involve the central and peripheral nervous systems,55 but severe central nervous system involvement is rare.216
Cardiopulmonary Disease
■ Figure 18–7 Advanced changes of the nail fold capillaries, with gross thickening and dropout areas in a child with dermatomyositis. (Courtesy of Dr. Jay Kenik.) (See color insert.)
The most frequently detected cardiac abnormalities are nonspecific murmurs and cardiomegaly with or without electrocardiographic changes.84 Pericarditis has also been described. Serious cardiac involvement (e.g., acute myocarditis, conduction defects, 1stdegree heart block) is
414
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
■ Figure 18–8 Acute dermatomyositis. A, Acute ulcerations over the olecranon processes in a child (A, See color insert.). B, Vasculitic ulcers over the elbow. C, Acute cutaneous ulcerations over pressure points in a 1.5-year-old boy.
rare but has been associated with death, and onset may be delayed until years after the initial diagnosis.55,84 Radioisotopic studies suggest that subclinical involvement of cardiac muscle may be more common than has been appreciated.217 Hypertension can occur and may be severe. It was present in 25% of patients in the series reported by Crowe and colleagues55 and is sometimes associated with or exacerbated by glucocorticoid therapy. Raynaud’s phenomenon is unusual but has been diagnosed in 2% to 15% of patients.55,173 Respiratory muscle weakness results in symptomatic, restrictive pulmonary disease in most children who are moderately to severely affected. However, asymptomatic pulmonary involvement may occur in up to 50% of children.218 Interstitial pneumonitis is rare and may be refractory to treatment.219,220 Aerobic and work capacity is reduced in these patients221 and correlates with parameters of disease activity, abnormalities on T1-weighted MRI, and disease duration.222,223 Interstitial lung disease developed in 3 of 8 Japanese children.224
Lipodystrophy Lipodystrophy is a clinically heterogeneous disorder that occurs in acquired and inherited forms.225 The association of lipodystrophy with JDM may be more common than
often appreciated (20% to 50%).47,226–228 This disorder may be generalized, localized and subtle, or unilateral.229 It is characterized by a slow but progressive loss of subcutaneous and visceral fat, often most noticeable over the upper body and face, that is accompanied by hirsutism, acanthosis nigricans, clitoral enlargement, hepatic steatosis, insulin resistance, abnormal glucose tolerance, and hypertriglyceridemia (Fig. 18–14). Huemer and colleagues230 reported lipodystrophy with a female predominance in 25% of 20 children with JDM and hypertriglyceridemia and insulin resistance in 50%. Severe lipodystrophy is associated with leptin deficiency.231 Misra and colleagues232 reported a 22% frequency of membranoproliferative glomerulonephritis in 35 patients with lipodystrophy and autoimmune diseases.
PATHOLOGY The distinctive pathologic lesions of JDM involve striated muscles, skin, and the gastrointestinal tract. The severity of clinical disease may or may not correlate with the intensity of the histologic findings.233,234 The histologic characteristics of JDM are contrasted with those in muscular dystrophy and neurogenic atrophy in Table 18–6.
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
415
■ Figure 18–9 (A) Vasculitic ulcers on fingers of a young boy with severe dermatomyositis. Ulcers on the knuckles (B) and on the elbow (C). These open ulcerations led to a Staphylococcus aureus bacteremia and endocarditis. (B, See color insert.)
TABLE 18–5 Forms of Dystrophic Calcification Form
Frequency (%)
Superficial plaques or nodules, usually on the extremities Deep, large, tumorous deposits, generally in the proximal muscles (i.e., calcinosis circumscripta) Intermuscular fascial plane deposition (i.e., calcinosis universalis) Severe, subcutaneous, reticular exoskeleton-like deposits Mixed forms: superficial plaques or nodules, tumorous deposits, intermuscular fascial plane deposition
33 20 16 10 22
Adapted from Bowyer SL, Blane CE, Sullivan DB, et al: Childhood dermatomyositis: factors predicting functional outcome and development of dystrophic calcification. J Pediatr 103: 882–888, 1983.
Skeletal Muscle Muscle fibers characteristically demonstrate group atrophy or necrosis at the periphery of the fascicle (Fig. 18–15).55 This perifascicular myopathy is often associated with a noninflammatory capillaropathy.55,235 Nonspecific changes include disruption of the myofibrils and tubular systems, central nuclear migration, and
prominent nuclei and basophilia.117,234 Concomitant degeneration and regeneration of muscle fibers occur and result in moderate variations in fiber size. Areas of focal necrosis are replaced during the healing phase by an interstitial proliferation of connective tissue and fat. An inflammatory exudate is often present. The inflammatory cells, which are often sparse and consist
416
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S ■ Figure 18–10 A, Widespread subcutaneous calcium deposits of various sizes developed in a 6-yearold boy who had dermatomyositis for 4 years.This is the same child shown in Figure 18–8C. B, A large area of tumorous calcification occurred over the knee with cutaneous ulceration and drainage in the same boy (Figures 18–8C and 18–10A).
■ Figure 18–11 A, Calcinosis circumscripta can be identified in the supracondylar areas of the knees. B, Massive tumorous calcium deposits developed unilaterally around the hip in a 7-year-old boy with the onset of dermatomyositis when he was 3 years old.
principally of lymphocytes and mononuclear cells, are located predominantly in the perimysium and perivascularly around the septae or in the fascicles (Fig. 18–16). This infiltrate is not in itself diagnostic because the muscular dystrophies may demonstrate an infiltrate around necrotic fibers. Macrophages, plasma cells, mast cells, and rarely, eosinophils or basophils are also present. Electron microscopy may confirm focal degeneration of the myofibrils, cytoplasmic masses, disorganization of sarcomeres, disruption of the Z lines and Z-disc streaming, actin-myosin filament disorganization, thickening of the capillary basement membranes, mitochondrial abnormalities, or an increase in vacuole formation. Lysosomes are more abundant in inflammatory myopathy than in
normal muscle. The relationship of these organelles to muscle necrosis is uncertain. Immunoglobulins can often be demonstrated on the sarcolemmal membrane by immunofluorescence microscopy, but this finding is of doubtful pathogenic significance.77,236 Damaged muscle fibers have an increase in calcium content,237,238 which may explain the uptake of technetium 99m diphosphonate by the muscles in inflammatory myopathy. The regenerative phase probably depends on the mononuclear myoblast that is derived from satellite cells.239 Regenerating fibers contain increased oxidative enzyme and alkaline phosphatase activities,240 which can be demonstrated noninvasively by 31 P-magnetic resonance spectroscopy (MRS).241 Li and
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
■ Figure 18–12 A, Calcinosis universalis developed in a 15-year-old girl with diffuse cutaneous ulcerations over the legs. Spicules of calcium salts were constantly extruded from these lesions. B, Interfascial deposition of calcium salts occurred during the healing phases in the leg of a child with dermatomyositis. C, Radiograph of a 12-year-old girl, who had the onset of acute, unremitting dermatomyositis at age 10, shows extensive radiopaque deposits of calcium salts throughout the interfascial planes of the musculature of the entire body.
■ Figure 18–13 Radiographs of the chest of the same child in Figures 18–8C and 18–10A and B. A, There are massive deposits of calcium salts around the right shoulder and in subcutaneous tissues of the chest. B, The calcium deposits have partially resolved 2 years later. C, Calcification around the left shoulder has almost completely resolved, but residual calcium deposits still exist in the subcutaneous areas of the left lateral thorax.
417
418
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S colleagues75 suggested that staining for MHC class I overexpression might be useful in evaluating muscle for evidence of JDM.
Blood Vessels
■ Figure 18–14 Paucity of subcutaneous tissue in two children with partial generalized lipoatrophy associated with dermatomyositis. A, Gottron’s papules, periungual capillary changes, and interosseous muscle atrophy are evident in the hand. B, Loss of subcutaneous fat is indicated by prominence of the veins on the volar surface of the forearm.
JDM should be considered a systemic inflammatory and noninflammatory vasculopathy rather than simply an inflammation of muscle and skin. A necrotizing vasculitis, presumably resulting from immune complex deposition, affects arterioles, capillaries, and venules of the striated muscles, gastrointestinal tract, skin, and subcutaneous tissues. Especially in the gastrointestinal tract, vasculitis leads to infarction, ulceration, and diffuse bleeding. The early studies of Banker and Victor209 identified this type of vasculitis as an important prognostic factor in survival. Investigations by Crowe and associates55 added distinctive features associated with persistent morbidity to an understanding of this complication. In their patients, muscle infarction and ulceration of the cutaneous and gastrointestinal tissues were associated with a zonal loss of the capillary bed, areas of focal infarction of muscle, non-necrotizing lymphocytic
TABLE 18–6 Comparison of Muscle Histopathology in Juvenile Dermatomyositis, Muscular Dystrophy, and Neurogenic Muscle Atrophy Histopathology
Dermatomyositis
Muscular Dystrophy
Neurogenic Atrophy
++
±
−
++ ++ ++ ++ ++ + + ± −
± ++ ++ ± − ++ + ++ −
− − − − − ± − ± ++
Focal necrosis and phagocytosis of muscle fibers Fiber regeneration Endomysial proliferation Random fiber atrophy Inflammatory cell infiltrates Vasculitis Central migration of nuclei Fat cell hyperplasia Fiber atrophy Motor unit atrophy − absent; ± possible; + present; ++ characteristic.
■ Figure 18–15 Muscle biopsy (hematoxylin and eosin stain; magnification × 250) with perivascular mononuclear inflammatory infiltrate, arterial wall thickening, and endothelial prominence.
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
419
■ Figure 18–16 A, Muscle biopsy (hematoxylin and eosin stain; magnification × 250) demonstrates atrophic muscle fibers (a) and a pale necrotic fiber (b). B, Muscle biopsy (hematoxylin and eosin stain; magnification × 430) demonstrates a partially necrotic myofiber with phagocytosis. C, Muscle biopsy (hematoxylin and eosin stain; magnification × 250) with an endomysial mononuclear cell inflammatory infiltrate.
vasculitis, and noninflammatory endarteropathy (Fig. 18–17). Conversely, severe vasculopathy was absent from the muscle specimens of children with limited disease.55 This spectrum of capillary endothelial damage had also been suggested in previous studies.77,209,242
Capillaries Widespread capillaropathy leads to intravascular coagulation, microvascular occlusion and infarction, and associated perifascicular myopathy. These capillary changes, although characteristic, are not specific and have been described in other connective tissue diseases, viral and rickettsial infections, malignancy, and normal wound healing.242,243 Changes in the capillaries can be identified clinically in the nail folds (see Figs. 18–6 and 18–7) and in tissues by light and electron microscopy.55,131–133,135,209 Endothelial swelling and necrosis, capillary thrombosis and obliteration, and endoplasmic tubuloreticular inclusions occur early in the course of disease.55,209,242,244–247
Arteries Inflammation of small muscular arteries and infarction of muscle results from an immune complex–mediated vasculitis.55 Other arterial lesions are unassociated with an inflammatory cell infiltrate. These abnormalities do not always correspond to those present in the capillaries, and severe capillaropathy may not be associated with discernible endarteropathy. IgM, IgG, and C3 were deposited in the perimysial veins in 9 of 11 patients studied by Whitaker and Engle.77 IgG may not always be present. Diffuse linear and occasionally granular vascular wall deposits of IgM, C3d, and fibrin were also observed in the areas of noninflammatory vasculopathy.55 Electron microscopy has not confirmed evidence of subendothelial deposition of immunoglobulins within vessel walls, as might have
been expected in classic immune complex disease, although circulating complexes and anticomplementary activity are often identified in the blood.84,248
Veins Intramural and perivascular mononuclear cell inflammatory infiltrates are frequently identified in the veins and may or may not be associated with immunoglobulin deposition.55,77 The endothelial cells may contain inclusions.
Skin A capillary endothelial change similar to that in muscle is usually observed in involved skin.55 Histopathologic examination of the skin demonstrates epidermal atrophy, liquefaction degeneration of basal cells, vascular dilatation, and lymphocytic infiltration of the dermis.249 An increase in the level of acid mucopolysaccharides has been reported in involved and uninvolved skin in approximately one third of patients.250 In a study of the histopathology of Gottron’s papules,251 basal layer vasculopathy, periodic acid–Schiff–positive basement membrane thickening, upper dermal mucin deposition, and a diffuse upper dermal mononuclear infiltrate were frequently identified. Epidermal hyperplasia consisting of acanthosis or papillomatosis often occurred, but epidermal atrophy was rare. In the healing phase of the disease, calcium phosphate crystals 252 may be deposited or formed in the skin, subcutaneous tissues, and interfascial planes of the muscle. Mechanisms for the excessive accumulation of calcium salts are unknown. With calcinosis, there may be an accompanying fibrosis and some degree of round cell and giant cell infiltration.
420
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S Gastrointestinal Tract Ulceration or perforation resulting from vasculopathy can occur in any part of the gastrointestinal tract, including the esophagus.55,209 Serious gastrointestinal disease of this type develops in approximately 10% of children with JDM.84 Pneumatosis intestinalis has been described.210,253 Except for the vascular disease, smooth muscle is not directly affected. Pancreatitis and hepatitis are rare.160
Heart Cardiac muscle is seldom affected by the primary pathologic process,254,255 but its involvement may be more common than clinically appreciated.217 A few cases of carditis have been described with areas of focal myocardial fibrosis and contraction-band necrosis. Interstitial myocarditis and narrowing of the coronary arteries have been reported.55
Kidneys Although microscopic hematuria may be present, especially at onset, histologic renal abnormalities are rare. In one report,9 histopathologic findings in 5 of 6 renal biopsies were abnormal and included cellular hyperplasia, capillary thickening, capsular adhesions, and hyperplasia involving the small blood vessels. Renal abnormalities have not been commonly cited in postmortem reports,14,55 and the possibility that overlap syndromes were responsible for some cannot be excluded. Progression to renal insufficiency has not been described.
DIFFERENTIAL DIAGNOSIS The differential diagnosis includes juvenile polymyositis, postinfectious myositis, primary myopathies, and inflammatory myositis accompanying other connective tissue diseases such as scleroderma or mixed connective tissue disease. The correct diagnosis is usually straightforward in the presence of the characteristic rash and weak, painful, or tender proximal muscles. Early in the disease course, especially in the absence of the characteristic rash, the differential diagnosis can be challenging. An expectation for the future is continued reclassification of the inflammatory and noninflammatory myopathies by identification of specific sets of immune-related genes.164,256
Juvenile Polymyositis
■ Figure 18–17 Electron microscopic sections of a muscle. A, Slight degree of endothelial cells swelling (A) in an arteriole (magnification × 63,337). B, Increased degree of endarteropathy (magnification × 5200). C, Virtual occlusion of the lumen (l) of a small arteriole by extreme swelling of the endothelial cells (magnification × 10,968), although there is no thrombosis or inflammation.
Juvenile polymyositis is uncommon.30,31,44 Age at onset and the sex ratio are comparable to JDM. The same environmental triggers and immunogenetic risk factors, such as following administration of D-penicillamine or growth hormone, or after bone marrow transplantation, have been associated with onset.22 Both proximal and distal muscles are weak. There are no associated cutaneous
C H A P T E R
abnormalities; the nail fold capillary pattern is normal. The disease of most patients pursues a chronic course and is often relatively unresponsive to glucocorticoids. Some cases of steroid-responsive polymyositis during the first year of life may result from maternal in utero infection.257,258 Severe muscle weakness with hypotonia and dysphagia are present at onset. Muscle biopsy is usually necessary for accurate diagnosis.
Postinfectious Myositis Acute, transient myositis may follow certain viral infections, especially influenza A and B100,103 and coxsackievirus B.97,98,129 Coxsackievirus B causes epidemic pleurodynia (i.e., Bornholm’s disease), characterized by fever and sharp pain in the muscles of the chest and abdominal wall. This syndrome is sometimes preceded by a moderate to severe headache, nausea, vomiting, and pharyngitis. The illness is most common in children and adolescents and usually lasts 3 to 5 days. Although myalgia is a characteristic complaint of acute influenza, myositis per se is rare (Table 18–7). In 1957, Lundberg102 described a contagious illness that occurred most commonly in preadolescent boys and was characterized by fever, headache, rhinitis, cough, nausea, and vomiting that lasted 2 to 3 days. This was followed by severe proximal calf pain and tenderness (i.e., myalgia cruris epidemica) that was exacerbated by movement. Complete recovery occurred after approximately 3 days. Laboratory studies demonstrated a slightly elevated erythrocyte sedimentation rate (ESR), moderate leukopenia with relative lymphocytosis, and a concomitant elevation of the serum levels of creatine kinase (CK) and aspartate aminotransferase (AST). Although no specific infectious agent was identified in this study, subsequent reports have confirmed influenza B as the most common causative agent. Treatment is supportive.
Other infectious causes of myositis include toxoplasmosis; trichinosis; cat-scratch fever; staphylococcal and streptococcal bacteremia; clostridial, mycoplasmal, Salmonella, or Serratia infection; schistosomiasis; and trypanosomiasis. Candidiasis and coccidioidomycosis are very uncommon causes. Toxoplasmosis may be associated with a syndrome that resembles dermatomyositis.115,259,260 Trichinosis, caused by ingestion of the larval cyst of the nematode Trichinella spiralis, is characterized initially by fever, diarrhea, and abdominal pain, followed in a week by periorbital edema and swelling and tenderness of muscles, especially those of the face, neck, and chest. Peripheral blood eosinophilia is often striking, and biopsy of affected muscles confirms the presence of the larvae and, later, calcified cysts. Treatment includes glucocorticoids to diminish inflammation and agents such as mebendazole or thiabendazole. Other unusual
18 J U V E N I L E D E R M AT O M Y O S I T I S
exposures include sarcocystis, malaria, cysticercosis, echinococcosis, and toxocariasis. Staphylococcal pyomyositis is an abscess in skeletal muscle that occurs after local muscle injury. It affects patients of all ages and is more common in boys than in girls. Lesions may be solitary or multiple and are usually located in the thigh, calf, buttock, arm, scapular areas, or chest wall. The abscess is tender and, if not too deep, warm. Patients usually have low-grade fever. Symptoms last for up to a week. Ultrasonography or gallium 67 citrate scanning may localize the lesion. Severe pustular acne occasionally may be associated with inflammatory disease of muscle and with an arthritis261 (see Chapter 9). Acute myopathy may also follow the use of isotretinoin for treatment of acne.262
Neuromuscular Diseases and Myopathies In the absence of characteristic skin changes, the differential diagnosis includes a wide variety of neuromuscular disorders (Tables 18–8 to 18–11).172 Early in the disease or before the development of cutaneous changes, muscular dystrophy or myotonia may be confused with JDM. The younger the child, the greater is the concern about these entities. Hypotonia in infancy, often associated with projectile vomiting, is a characteristic of the mitochondrial disorders involving the branched chain amino acids. Paroxysmal myoglobinuria or acute rhabdomyolysis occasionally may be encountered. Certain drugs or toxins, including alcohol, clofibrate, D-penicillamine, glucocorticoids, and hydroxychloroquine, can cause myopathy (see Chapter 5). The possibility of muscular dystrophy is suggested by a family history of myopathy and an insidious onset of slowly progressive, predominantly proximal muscle weakness, and by absence of the normal progression of achievement of developmental landmarks. Constitutional signs, muscle tenderness, and cutaneous abnormalities are absent. In Duchenne’s muscular dystrophy, there is a characteristic hypertrophy of the calves, a sign that occurs in other myopathies and occasionally in longstanding JDM. The hereditary nature of Duchenne’s muscular dystrophy is demonstrated by the presence of markedly elevated levels of serum CK in the patient and the patient’s mother. However, approximately one third of these disorders represent new mutations. Myoadenylate deaminase deficiency (MDD)263,264 occurs in an autosomal recessive primary form and an acquired disorder associated with rheumatic and neuromuscular diseases. Data suggest that homozygous MDD is relatively common because 2% of muscle biopsies are deficient in enzyme activity (less than 2% in the primary form; less than 15% in the secondary form). Muscle fatigue, stiffness, and cramping may be noticed after exercise and may begin in childhood (23%) or adolescence (26%), but these features do not occur in all deficient individuals. Patients with MDD may demonstrate a decreased muscle mass, hypotonia, and weakness. With forearm exercise, there is a failure of the plasma
TABLE 18–7 Acute Myositis Associated with Influenza B Infection ● ● ● ●
421
Onset during recovery phase of the viral illness Predominantly severe bilateral pain and tenderness of the gastrocnemius and soleus muscles Elevated serum muscle enzyme concentrations (e.g., creatine kinase, aspartate aminotransferase) Recovery in 3 to 5 days
422
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
TABLE 18–8 Classification of Neuromuscular Disorders I. Primary myopathies A. Muscular dystrophies 1. Sex-linked recessive a. Duchenne/Becker muscular dystrophy b. Emery-Dreifuss muscular dystrophy 2. Autosomal dominant a. Myotonic muscular dystrophy (Steinert) b. Facio-scapulo-humeral muscular dystrophy c. Limb girdle muscular dystrophy 3. Autosomal recessive a. Limb girdle muscular dystrophy b. Congenital muscular dystrophy with merosin deficiency c. Congenital muscular dystrophy (Fukuyama) B. Congenital myopathies 1. Sex-linked recessive a. Myotubular myopathy 2. Autosomal dominant a. Nemaline rod myopathy b. Central core disease 3. Autosomal recessive a. Myotubular myopathy b. Nemaline rod myopathy c. Congenital muscle fiber–type disproportion C. Myotonic disorders 1. Myotonia congenita (Thomsen) 2. Dystrophia myotonia (Steinert) 3. Paramyotonia congenita D. Metabolic myopathies 1. Disorders of glycogen metabolism a. Sex-linked recessive i. Glycogenosis IX (phosphoglycerate kinase deficiency) b. Autosomal recessive i. Glycogenosis II (Pompe, acid maltase deficiency) ii. Glycogenosis V (McArdle, myophosphorylase deficiency) iii. Glycogenosis VII (Tarui, phosphofructokinase deficiency) iv. Glycogenosis X (phosphoglycerate mutase deficiency) v. Glycogenosis XI (lactate dehydrogenase deficiency) 2. Mitochondrial myopathies a. Mitochondrial myopathy (Kearns-Sayre) b. Mitochondrial myopathy (MERRF) c. Mitochondrial myopathy (MELAS) 3. Familial periodic paralysis (hyperkalemic and hypokalemic) 4. Carnitine deficiency 5. Muscle carnitine palmityl-transferase deficiency 2 6. Secondary to endocrinopathies a. Addison’s disease b. Cushing’s syndrome c. Hypopituitarism d. Hypothyroidism 7. Myoadenylate deaminase deficiency
II. Inflammatory diseases A. Postinfectious 1. Viral syndromes a. Influenza B b. Coxsackievirus B c. Echovirus d. Poliomyelitis 2. Toxoplasmosis, sarcosporidiosis 3. Trichinosis, cysticercosis 4. Septic (staphylococci and other pyogenic organisms) 5. Tetanus 6. Gas gangrene B. Connective tissue diseases III. Genetic abnormalities A. Osteogenesis imperfecta B. Ehlers-Danlos syndrome C. Mucopolysaccharidoses IV. Trauma A. Physical (crush, rhabdomyolysis) B. Toxic (snakebite) C. Drugs 1. Glucocorticoids 2. Hydroxychloroquine 3. Diuretics, licorice 4. Amphotericin B 5. Alcohol 6. Vincristine 7. D-Penicillamine 8. Cimetidine 9. Chronic hemodialysis V. Neurogenic atrophies A. Spinal muscular and anterior horn cell dysfunction 1. Spinal muscular atrophy (Werdnig-Hoffmann, Kugelberg-Welander) 2. Familial dysautonomia (Riley-Day) 3. Arthrogryposis multiplex congenita 4. Amyotrophic lateral sclerosis B. Peripheral nerve dysfunction 1. Hereditary motor-sensory neuropathy (Charcot-Marie-Tooth, Dejerine-Sottas) 2. Hereditary motor-sensory neuropathy (axonal type) 3. Hereditary motor-sensory neuropathy (Charcot-Marie-Tooth-X) 4. Neurofibromatosis 5. Guillain-Barré syndrome C. Disorders of neuromuscular transmission 1. Congenital myasthenia gravis 2. Botulism 3. Tick paralysis 4. Organophosphate poisoning
Adapted by Brenda Banwell, M.D., The Hospital for Sick Children, University of Toronto.
ammonia level to rise along with inosine monophosphate. Electromyographic findings are nonspecific. The muscle in primary MDD is normal histologically except for the absence of adenosine monophosphate deaminase. Activity of this enzyme is normal in other tissues. Endocrinopathies, especially hyperthyroidism and hypothyroidism, hyperparathyroidism and hypoparathyroidism, diabetes mellitus, and myopathy associated with idiopathic or iatrogenic Cushing’s syndrome, should be considered in the differential diagnosis of a myopathy without evidence of cutaneous disease. Myasthenia gravis is rare, and the diagnosis is suggested by a decremental response to repetitive nerve stimulation, involvement of ocular and distal muscles, and improvement of the weakness after administration of cholinergic drugs. Primary neu-
rogenic atrophies, including infantile and juvenile spinal muscular atrophy, are associated with proximal muscle weakness and rarely may be confused with inflammatory myositis.
Myositis with Other Connective Tissue Diseases Children with systemic scleroderma (see Chapter 19), mixed connective tissue disease (see Chapter 21), or occasionally SLE (see Chapter 16) may have skin and muscle abnormalities at onset that suggest a diagnosis of JDM.38 The differentiation of these diseases is usually not difficult, however, because clinical features unique to each are almost always present. The laboratory evalua-
C H A P T E R
TABLE 18–9 Classification of Neuromuscular Disorders by Course
18 J U V E N I L E D E R M AT O M Y O S I T I S
TABLE 18–11 Classification of Neuromuscular Disorders by Age of Onset
Acute Disorders
Congenital Onset
Muscular dystrophy Paroxysmal myoglobinuria
Congenital muscular dystrophy Central core disease Nemaline myopathy Congenital hypoplasia Benign hypotonia
Chronic Disorders Dermatomyositis Muscular dystrophy (Duchenne’s, Dejerine-Landouzy) Central core disease and nemaline myopathy Congenital hypotonia Glycogen storage disease Myoadenylate deaminase deficiency Endocrine myopathy Nutritional myopathy Amyloidosis Episodic Disorders Paramyotonia congenita Familial periodic paralysis Hypokalemia Myasthenia gravis Myasthenia of malignancy
tion provides supportive or definitive diagnostic information in most instances. The child with JDM may have a malar dermatitis that is similar in distribution to the butterfly rash of SLE, but it often lacks relatively well-defined borders. Heliotrope suffusion and periorbital edema are not characteristic of SLE. Periungual capillary abnormalities are present in many connective tissue diseases, but Gottron’s papules are present only in children with JDM. Although early cutaneous abnormalities of scleroderma and JDM are quite different, the skin changes sometimes tend to become similar during the courses of these diseases. Myositis occurs in systemic scleroderma and mixed connective tissue disease and, to a limited extent, in SLE TABLE 18–10 Classification of Neuromuscular Disorders by Predominant Site of Involvement Proximal Involvement Dermatomyositis Steroid myopathy Thyrotoxic myopathy Sarcoid myopathy Muscular dystrophy Proximal familial neuromuscular diseases
423
Childhood Onset Muscular dystrophy (Duchenne) Glycogen storage diseases Myoadenylate deaminase deficiency Late Childhood and Adolescence Onset Muscular dystrophy (Dejerine-Landouzy) Myotonia congenita Periodic paralysis Any Age of Onset Dermatomyositis Steroid and hydroxychloroquine myopathies Myasthenia gravis Trichinosis
and systemic-onset juvenile rheumatoid arthritis. The myositis of JDM can be differentiated from that of other connective tissue diseases by its severity, the greater elevation of serum levels of muscle enzymes, and histologic examination of muscle obtained by biopsy. In uncomplicated chronic arthritis, acute rheumatic fever, SLE, or scleroderma, muscle biopsy demonstrates focal accumulations of lymphocytes, patchy fiber atrophy, and increased interstitial connective tissue but no significant vasculopathy.265,266 Muscle fiber degeneration and atrophy, sarcoplasmic degeneration, and microcyst formation occur in Sjögren’s syndrome.267 A necrotizing vasculitis with muscle fiber degeneration and neurogenic atrophy may be identified in polyarteritis. Laboratory evaluation confirms normal or only slightly elevated serum levels of the muscle enzymes in SLE and other connective tissue diseases compared with marked elevations in JDM. Systemic features of SLE, such as pericarditis and pleural effusions, are rare in JDM. Hepatosplenomegaly and lymphadenopathy occur in less than 5% of children with JDM. The arthritis of JDM, although not uncommon, is usually mild; that of SLE occurs more frequently and, although nonerosive, may be quite florid at onset and extremely painful.
Distal Involvement Myotonic dystrophy Distal muscular dystrophy Peroneal muscular atrophy Motor system diseases Proximal or Distal Involvement Floppy infant syndrome Myotonia congenita Dystrophic ophthalmoplegia Myasthenia gravis Periodic paralysis
Miscellaneous Disorders A dermatomyositis-like disease has been observed in children with agammaglobulinemia and common variable immune deficiency (see Chapter 33).84,119,268–270 Other rare causes of myositis in children include multicentric reticulohistiocytosis,271 leukemia,34 giant-cell myositis,272 and sarcoidosis.273 A number of uncommon forms of myositis have been described, especially in adults,274 including inclusion body myositis,275–277 eosinophilic myositis,278 and disease restricted to one muscle group or extremity, such as localized nodular myositis279,280 and proliferative myositis.281 Inclusion body myositis is resistant to all forms of therapy.
424
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
Rhabdomyolysis may follow an upper respiratory infection, trauma, or extreme muscular exertion.282 Onset is generally acute and is characterized by profound weakness, myoglobulinuria, very high serum levels of muscle enzymes, and occasionally, oliguria and renal failure. It may also occur after snakebite, in heat stroke, and in the familial malignant hyperpyrexia syndrome. Fibrodysplasia ossificans progressiva (i.e., myositis ossificans progressiva) is a rare, autosomal dominant inflammatory disorder (usually a new mutation) that results in painful swelling of muscle and fascia (e.g., ligaments, tendons, aponeuroses), followed by fibrosis, calcification, and ossification.283–289 The gene has been mapped to chromosome 4q27-31.290 The patient may present with a spontaneous joint contracture. The clinical diagnosis is often elusive until calcification and, later, ossification are evident on ultrasound or radiography.291,292 Biopsy findings of affected sites at an early stage may be misleading and be misinterpreted as malignant sarcoma.293,294 The back of the neck and posterior trunk are often involved initially, followed by the muscles of the limbs. Palmar and plantar fascia may be affected. The great toes are often congenitally short, and the thumbs are sometimes involved. The disease after onset in the first year of life is characterized by exacerbations and remissions and slowly progresses to severe debility, restriction of pulmonary capacity, and reduced life expectancy. There is no therapy for prevention or treatment, although bisphosphonates have been advocated.295 Data on overexpression of bone morphogenic protein-4296 or osteogenic transcription factor297 may provide additional modalities of therapy.298
LABORATORY EXAMINATION General Findings Nonspecific indicators of inflammation, such as the ESR and C-reactive protein (CRP) level, tend to correlate with the degree of clinical inflammation and are of diagnostic value in differentiating inflammatory myopathies such as JDM from noninflammatory disorders of muscle such as muscular dystrophy or myotonia.299,300 Leukocytosis and anemia are uncommon at onset, except in the child with associated gastrointestinal bleeding. Patients may have lymphopenia.301 Urinalysis is usually normal, although some children at onset have microscopic hematuria.9 Serum levels of factor VIII–related antigen (i.e., von Willebrand’s factor) reflect endothelial damage and are often elevated in children with JDM.83,155,302–304 Although abnormal factor VIII–related antigen levels occur in most children with active disease,305 they were not of value in one study in predicting a flare.83 There are few specific abnormalities of immunoglobulin (Ig) levels.306 IgE levels were elevated in one study of 22 Japanese children.307 Neopterin is a derivative of pyrimidine metabolism, and its serum concentration has been considered a marker of interferon-activated monocytes and macrophages. Levels of neopterin are raised in individuals with inflammation, infections, or malignant diseases. Determination of its concentration has been proposed as a useful laboratory marker of immune activation and disease activity.305,308 Levels of quinolinic acid may be another marker of activity.304,308 The serum level of myoglobin, a normal constituent of cardiac and skeletal muscle with a molecular mass of approximately 17 kD, is increased in approximately 50% of patients
with inflammatory myositis.309,310 This elevation does not always correlate with an increase in serum CK levels. Antibodies to myoglobin are present in 70% of patients and may interfere with its quantitation.311 Although myoglobin is much more nephrotoxic than hemoglobin, myoglobinuria in cases of JDM seldom reaches levels that are associated with renal damage. An adult with 30 kg of muscle mass normally releases 0.3 mg of myoglobin per day.312,313 The creatine/creatinine ratio is not a practical measure of myopathy in children. Creatine is synthesized in the liver from arginine and glycine by an aminotransferase to form ornithine and guanidoacetic acid. The latter is transmethylated by interaction with S-adenosylmethionine to form creatine and Sadenosylhomocysteine. Creatine circulates in the plasma in relatively low concentrations (less than 0.6 mg/dL in adults). It is stored in muscle as creatine phosphate and serves as the reserve energy pool for muscular activity. In muscle, creatine is converted to the anhydride creatinine at a constant rate of approximately 2% per day. Creatinine diffuses passively into the plasma and is excreted by the kidney. If the body pool of creatine decreases, the creatinine excretion per unit of time is also decreased. Endogenous creatinine excretion is an important index of body creatine stores and total muscle mass. Creatinuria in JDM is not simply a matter of failure of uptake by an inflamed muscle or a decreased muscle mass, but rather an inability to maintain normal membrane permeability. The urinary creatine/creatinine ratio is age-related in children with JDM.314 In those younger than 12 years, it is not a reliable guide to the activity of the myositis. A 24-hour creatinine excretion is an excellent indicator of muscle mass in children between the ages of 3 and 18 years. Boys begin to excrete significantly larger amounts of creatinine than girls at puberty. In males, excretion increases from approximately 0.36 g/day at 5 years of age to 1.6 g/day at 17 years. Creatine excretion, however, is much higher in the younger child; at no age in childhood is there a significant male/female difference in excretion. A 24-hour urine collection would have to document extremely large or infinitesimally low amounts of creatine before a creatine/creatinine ratio could be judged abnormal in young children.
Autoantibodies Rheumatoid factors (RFs) usually are absent in patients with JDM. Antinuclear antibodies (ANAs) have been reported in a variable frequency of 10% to 85%.11,13,315,316 Particularly important are antibodies directed against one of a number of extractable nuclear antigens that are soluble in saline at neutral or acid pH (Table 18–12).84,317–319 Myositis-specific antibodies (MSAs), such as those directed to the aminoacyl transfer RNA (tRNA) synthetases, have been described in only a minority of children.22,316,320 Their inclusion has been proposed for criteria for the classification of adult dermatomyositis or polymyositis.319,321–325 Myositis-associated antibodies (MAAs) occur in variants of JDM, often in association with overlap syndromes. Approximately 80% of children with JDM are negative for MSAs and MAAs.22
Myositis-Specific Antibodies MSAs are antibodies targeted to cytosolic RNAs or proteins involved in protein synthesis. Usually, only one MSA occurs in a patient, and it often can be detected before onset of the phenotype. Patients with a specific
C H A P T E R
TABLE 18–12 Autoantibodies in Dermatomyositis or Polymyositis Serologic Group
Specificity
Myositis-Specific Antibodies (MSAs) Anti-synthetases Jo-1 PL-7 PL-12 OJ KS EJ Anti-Mi-2a and b Anti-SRP
Histidyl-tRNA synthetase Threonyl-tRNA synthetase Alanyl-tRNA synthetase Isoleucine-tRNA synthetase; multienzyme complex Asparaginyl-tRNA synthetase Glycyl-tRNA synthetase 218-kD nuclear histone deacetylase helicases Cytoplasmic signal recognition peptide
Myositis-Associated Antibodies (MSAs) Anti-PM-Scl Anti-PMSI Anti-Ku Anti-Ro-52 Anti-KJ Anti-annexin XI Anti-U (1, 2, 3, 5) RNPs Anti-SSA/Ro
Nuclear/nucleolar exosome complex Nuclear DNA mismatch repair Acidic nuclear DNA-binding protein dimer Nuclear/cytoplasmic translation function Cytoplasmic translation function 56-kD nuclear protein Small nuclear RNP-associated peptides Nuclear protein complex
MSA are relatively homogeneous in clinical manifestations and prognosis.320 MSAs have been uncommon in childhood-onset disease and have only been described in approximately 10% of patients. Their specificities include antibodies to the Jo-1, Mi-2, and PL-7 antigens.326,327 Anti-synthetase antibodies usually occur in patients with an acute onset and rapid progression of disease, but they have been identified in others with slow progression and in asymptomatic adults.323 The anti-synthetases are associated with an increased frequency of HLA-DR3 and DRw52.160 DQA1*0501 or DQA1*0401 may also be present in patients with antibodies to Jo-1, PL12, and other MSAs. Anti-Jo-1 is the most common. It occurs in approximately 20% of adults and has been described in at least 10 children.328 It is specific for histidyl-transfer RNA synthetase, a cytoplasmic enzyme that catalyzes the esterification of histidine to its cognate tRNA. Patients who are anti-Jo-1 positive demonstrate a subset of multisystemic features that have been called the anti-synthetase syndrome. Onset of disease typically is acute and characterized by fever, arthritis, Raynaud’s phenomenon, and a myositis that is often severe. Interstitial pulmonary fibrosis may become the dominant feature of the course.323 Although polyarthritis, when it occurs, is generally mild, it can result in erosions and subluxations. A hyperkeratotic nonpruritic fissuring rash of the palms and lateral aspects of the fingers (i.e., mechanic’s hands) is another feature of this syndrome, especially in adults. Mi-2 is a 218-kD nuclear helicase that is involved in transcriptional activation and often associated with HLADR7 and HLA-DR53. Anti-Mi-2 has been identified in approximately 22 children with JDM, and in adults, it is
18 J U V E N I L E D E R M AT O M Y O S I T I S
425
strongly associated with a distinct pattern of rash22,326,327 and perhaps a more benign course.329 The areas of most intense involvement often are the V of the neck and the anterior chest area and the shawl area with involvement of the upper back and shoulders. Most children have not displayed this characteristic dermatitis, although they have malar erythema, Gottron’s papules, and cuticular overgrowth. The disease is responsive to glucocorticoid therapy. Another subset of patients is associated with antibodies to the signal recognition peptide (SRP), which have been identified in at least four children.22,330 Onset of disease is often severe and acute, with myositis of the proximal and distal muscles predominating. These patients may have severe weakness and a higher frequency of cardiac disease, and may respond poorly to glucocorticoid therapy. SRP is a cytoplasmic ribonucleolar protein complex that directs the passage of newly synthesized protein from the ribosome to the endoplasmic reticulum. Immunofluorescent tests therefore may demonstrate anticytoplasmic staining. HLA-DR5 and HLA-DRw52 are associated with this syndrome.
Myositis-Associated Antibodies The MAAs occur in somewhat less than 10% of children and consist of several distinct entities.22 Anti-PM-Scl is suggested by a nucleolar pattern on ANA testing.37,324 It occurs in the overlap syndrome of inflammatory myopathy and systemic scleroderma (i.e., scleromyositis). Clinical features include myositis, arthritis, digital sclerosis, and Raynaud’s phenomenon. The course of the disease is often benign and prolonged, but has a good prognosis. This antibody is associated with HLA-DR3. Antibodies reactive with PM-1 antigens are present in up to 60% of adults with polymyositis331 but occur in only a minority of children.84 PM-1 antibodies were identified in 4 of 18 patients in one series332 and in 3 of 21 in another.55
Other Antibodies Anti-U1RNP is characteristic of the overlap syndrome of mixed connective tissue disease (see Chapter 21). It is suggested by the presence of a high-titered ANA speckled pattern. There is a subgroup of patients (4 of 53 in one study) who have anti-Myo 22/25 antibodies to U3RNP.333 Anti-SSA/Ro antibody often occurs in association with anti-Jo-1 and other MSAs. The anti-Ku antibody has not often been described in patients from North America334 but was reported from Japan in approximately 50% of patients with an overlap syndrome. It has been reported in approximately 10 children with overlap syndromes.320 The antigen is a protein kinase that is involved in the phosphorylation of a number of transcription factors. Anti-annexin XI has been identified frequently in children with JDM and in those with other connective tissue diseases, and is associated with no particular clinical manifestations.320 It reacts against a 56-kD nuclear protein. Antibodies to endothelial cell antigens have been demonstrated in JDM,335 although these antibodies also
426
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
occur in other vasculopathies.336 The frequency of antibodies to myosin and muscle is the same for patients with inflammatory myopathy, muscular dystrophy, or denervation atrophy;337,338 therefore, these antibodies may result from muscle damage rather than being a primary phenomena. One half of the children have circulating immune complexes that may be involved in the pathogenesis of vascular injury.68,84,339 They may also interfere with the accurate serologic detection of antibodies and potential antigens, but serum complement determinations are normal.82,339 Antibodies to cardiolipin, found in a minority of children with JDM, may also reflect underlying vasculopathy.316
Specific Laboratory Diagnostic Studies The three investigations that are most useful in substantiating a diagnosis of JDM are measurement of serum levels of the muscle enzymes, electromyography, and histopathologic examination of a muscle specimen obtained by needle or open biopsy (Table 18–13).
Serum Muscle Enzymes Serum levels of the sarcoplasmic muscle enzymes are important for diagnosis and for monitoring the effectiveness of therapy. Considerable individual variation in the pattern of enzyme elevation is observed; it is therefore recommended that, at least early in disease, AST, CK, lactate dehydrogenase (LDH), and aldolase be measured to obtain a baseline evaluation.10 The degree of elevation in serum concentration ranges from 20 to 40 times normal for AST or CK. The CK level does not always correlate with disease activity;83 rarely, some children have normal serum levels of CK during the acute phase of the illness, and others have a persistent elevation late in the course without any other clinical indication of muscle inflammation.340 In the latter instance, evaluation of serum CK levels in family members may suggest an unrelated or unsuspected genetic abnormality. LDH and alanine aminotransferase (ALT) levels are increased in many children. Although relatively less specific, these enzymes often mirror global disease activity. Elevated ALT and LDH activities may also reflect liver disease associated with lipodystrophy and insulin resistance.230,341 Serum levels of all muscle enzymes usually decrease 3 to 4 weeks before improvement in muscle strength and rise 5 to 6
TABLE 18–13 Specific Diagnostic Studies at Onset of Juvenile Dermatomyositis Study Elevation of serum levels of the muscle enzymes Aspartate aminotransferase Creatine kinase Aldolase Lactate dehydrogenase Abnormal electromyography Abnormal muscle biopsy (inflammation)
Diagnostic Success (%) 90–98 90 85 65 65 95 80
weeks before clinical relapse. As a general rule, changes in CK levels occur first, often falling to the normal range within several weeks of instituting therapy; aldolase levels are the last to respond. Guzman and colleagues83 reported that flares of disease are best predicted by a combination of AST and LDH and that CK functions poorly as a predictor of exacerbation of myositis. Creatine Kinase CK catalyzes the transfer of a phosphoryl group from creatine phosphate to adenosine diphosphate to regenerate adenosine triphosphate in the mitochondria of muscle, brain, and heart. The adenosine triphosphate available to muscle is sufficient to sustain contractile activity for only a fraction of a second. In skeletal muscle, CK constitutes up to 20% of the soluble sarcoplasmic protein, and total CK activity is 225 to 12,000 units per gram of muscle. CK is a dimeric molecule with two subunits: M (muscle) and B (brain). Both consist of 360 amino acids with a molecular mass of 41 kD. Three isoenzymes exist. MM (CK-3) is found in muscle and myocardium, BB (CK-1) in brain, and MB (CK-2) in myocardium but also in regenerating muscle.342 There may be a persistent elevation of the MB band in muscle inflammation. The adult pattern of isozymes is achieved by the age of 4 years. Serum CK concentration is elevated in many cases of muscle injury, motor neuron diseases, vasculitis, metabolic disorders, endocrinopathies, toxic reactions, and infections. Very high levels are most commonly associated with muscular dystrophy and, somewhat less commonly, with JDM. Abnormalities of the junctional sites between the T-tubules and the sarcoplasmic reticulum in muscle cells of children with JDM may be the primary sites of leakage of the enzyme. These abnormal anastomoses are far more extensive in the perifascicular than in the centrofascicular myofibers. Enzyme levels are not increased in diseases in which there is no loss of sarcolemmal integrity (e.g., glucocorticoid myopathy, disuse atrophy).
Transaminases AST and ALT are cytosolic and mitochondrial enzymes with a wide tissue distribution. AST has two dimeric isoenzymes: one in the cytosol and the other in the mitochondria. The half-life in human plasma is 47 hours for ALT, 6 hours for mitochondrial AST, and 12 to 17 hours for cytosolic AST. Plasma levels decrease to normal adult ranges by 1 year of age.
Aldolase Aldolase (1,6-diphosphofructoaldolase) is found in myocardium, liver, cerebral cortex, kidneys, and erythrocytes, but it is present in much higher concentration in skeletal muscle. Aldolase is one of the principal glycolytic enzymes that catalyze the conversion of D-fructose-1,6-diphosphate to dihydroxyacetone phosphate and D-glyceraldehyde-3-phosphate. There are three cytosolic isoenzymes: aldolase A, which predominates in muscle; aldolase B in liver; and aldolase C in brain. Aldolase A is increased in active JDM and in some children with chronic disease.
Lactate Dehydrogenase LDH is abundant in myocardium and skeletal muscle. There are five isoenzymes: I (30%), II (40%), III (20%), IV (6%), and V (4%). In acute adult polymyositis, there is relatively less isoenzyme I and relatively more II, III, IV, and V. In chronic disease,
C H A P T E R
only isoenzymes I and II are disproportionately elevated. In contrast, patients with active muscular dystrophy exhibit an increase in types I and II and a decrease in III, IV, and V, especially in the younger patient.
Electromyography Electromyography occasionally is useful in confirming the diagnosis of JDM and in selecting the best site for performing a muscle biopsy. The electromyogram should be evaluated on one side of the body only so that the muscle biopsy, if necessary, can be obtained on the opposite extremity without an artifact created by a needle puncture. Electromyography can be troublesome in the young child, and mild sedation is often necessary. An electromyogram is not mandatory and need not be done unless the diagnosis is in doubt. The characteristic electromyographic changes of myopathy and denervation (Table 18–14) are associated with membrane instability (e.g., increased insertional activity, fibrillations, positive sharp waves) and random fiber destruction (e.g., decreased amplitude and duration of action potentials). The electrical changes in denervation probably result from segmental myonecrosis of the end plate, although the terminal axons may also be affected. Reinnervation may occur after the acute phase of the disease. Nerve conduction velocities and latencies are normal in JDM, unless severe muscle atrophy is present with a decrease in the number of muscle fibers in a motor unit and with electrical irritability of the sarcolemmal membrane or terminal axonal fibers. The value of electromyography in identifying continuing inflammatory activity of muscle during the course of JDM has not been adequately documented. Quantitative electromyography may be more informative in this regard.342–345 In following the course of the disease, increasing muscle strength correlates with less spontaneous activity and a decreasing proportion of high-frequency components. During the initial period of treatment, however, a temporary increase in high-frequency signals is expected.
Muscle Biopsy A muscle biopsy is indicated in the initial assessment of a child if the diagnosis is in any way uncertain; to evaluate “activity” of the disease, especially late in its course; or if histopathologic support for instituting long-term glucocorticoid therapy or immunosuppressive drugs is deemed necessary.346 Occasionally, a biopsy is indicated in a child who has failed to respond therapeutically to rule out disorders such as inclusion body myositis.31,39,276,347 A muscle biopsy also provides valuable prognostic information, based on the studies of Banker and coworkers209 and Crowe and coworkers.55 TABLE 18–14 Electromyography in Inflammatory Muscle Disease ● ● ●
Myopathic motor units (decreased amplitude, short duration, polyphasic) Denervation potentials (positive sharp waves), spontaneous fibrillations, and insertional activity High-frequency repetitive discharges
18 J U V E N I L E D E R M AT O M Y O S I T I S
427
The muscle to be biopsied, usually the deltoid or quadriceps, should be clinically involved as demonstrated by muscle testing on physical examination, electromyography, or MRI, but it should not be atrophied. MRI can be used to select an area of muscle that is likely to be affected by the disease and thereby increase the probability of obtaining informative tissue. If electromyography has been obtained within the previous 6 weeks, the biopsy should be performed on the opposite side of the body. Care should be taken to prepare the tissue in accordance with the instructions of the pathologist. A generous specimen (12 mm long, 4 mm wide, 2 mm thick) on open muscle biopsy should be obtained in a muscle clamp because muscle involvement is often spotty. The specimen is placed in a transport container and taken immediately to the pathology laboratory. One sample is frozen in isopentane for immunofluorescent and enzyme studies. Another on a separate pediatric clamp is fixed in glutaraldehyde for electron microscopy. Histopathologic results are more likely to be negative if the muscle specimen is inadequate in size, obtained from an inappropriate muscle, or taken late in the disease when the pathologic changes may no longer be specific. Occasionally, there is no evidence of inflammatory change even though characteristic abnormalities such as perifascicular atrophy are present.233 Although an open biopsy is most often performed, needle biopsy is preferred in some centers.348 Needle biopsy with a spring-activated, 14-gauge needle may offer a convenient and cost-effective alternative to a surgical procedure.349 Three or four cores can be obtained with only surface anesthesia by this procedure. The middle deltoid and vastus lateralis are the safest areas to biopsy by this technique from the standpoint of avoiding vascular or neurologic damage.
RADIOLOGIC EXAMINATION Radiographs in early JDM demonstrate increased soft tissue density caused by edema of the muscle and subcutaneous tissues and, somewhat later, atrophy of muscle. In chronic disease, areas of calcification in soft tissues can be documented by plain radiographs (see Figs. 18–10 to 18–12). Osteoporosis of long bones and of the vertebral bodies is often extensive (Fig. 18–18). Ultrasound studies of muscle demonstrate increased echogenicity (Fig. 18–19).350 Radionuclide scanning can detect early abnormal changes in blood flow in diseased muscles.351–354 This technique has limited clinical application, however, and has been superseded by MRI, which is more sensitive for localization of inflamed muscle. MRI dramatically documents the extent and focal nature of the muscle abnormalities (Fig. 18–20).350,355–360 The T1-weighted image demonstrates fibrosis, atrophy, and fatty infiltration. An increased signal is characteristic of fat, blood, edema, or other proteinaceous material. The short tau inversion recovery (STIR) image or T2weighted image with fat suppression demonstrates muscle edema and inflammatory changes by a hyperintense signal,361–364 and inflammation in skin, subcutaneous tissue, and fascia.364 Active myositis in general is best documented by the T2-weighted image, even when not reflected in enzyme elevations. Reversal of these changes occurs in response to treatment.356,357,365,366 However, exercise-induced changes may mimic active disease.363 Investigations have suggested that the course of children with JDM can be monitored with 31P-MRS as an indicator of bio-
428
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S magnesium and magnesium-ATP were also concordant and in part alleviated by immunosuppressive therapy.241 Dual energy x-ray absorptiometry (DEXA) has enabled a more accurate assessment of osteopenia and osteoporosis in children with JDM and other chronic rheumatic diseases (see Chapter 39).368,369 Z scores for these children should be compared with BMC data obtained from healthy, geographic, control children (male and female) with normal values adjusted for height, age, and ethnicity.370 Alternative evaluation has been proposed by comparisons with data obtained from quantitative calcaneal ultrasound.371
TREATMENT
■ Figure 18–18 Extreme osteopenia of the lumbar spine with multiple vertebral compression fractures in a 15-year-old girl with a long history of chronic polycyclic dermatomyositis and steroid treatment.
chemical defects in energy metabolism.361 Noninvasive 31P-MRS of thigh muscles in children with JDM was studied to characterize metabolic abnormalities during rest, exercise, and recovery.367 ATP and phosphocreatine levels were low, as were other measures of mitochondrial oxidative phosphorylation, and they correlated with clinical weakness and fatigue. Low levels of free
In the pre-steroid era, approximately one third of children with JDM died, one third recovered, and one third were disabled to a moderate or severe extent.9,11,20 The introduction of glucocorticoids revolutionized the treatment and prognosis for these children.6,11–13,48,169–174,176,315,372–375 The team approach and general supportive care, including bed rest and positioning early in the disease combined with individualized physical therapy, are essential. There is general agreement that glucocorticoids are always required, but the specifics of management vary considerably from physician to physician and patient to patient.376 The response of the child to the treatment program is judged on the basis of systemic signs and symptoms such as fever, general malaise, muscle tenderness, and pain (if present); repeated graded muscle examinations by the same observer; sequential serum levels of selected muscle enzymes, acute phase reactants, and other laboratory examinations if indicated (e.g., factor VIII-related antigen),83,303 and occasionally, other studies, such as MRI of muscle and ultrasonography.361,363,366,377 Indices of disease activity are being validated.378–383 Assessment of protocols of therapy is difficult because appropriately controlled clinical studies have not been performed.
■ Figure 18–19 A, Ultrasound view of the mid-thigh of a normal child.The skin surface is at the top; the convex arc is the femoral shaft. Fascial planes are visible in the muscle between the skin and bone. B, The same ultrasound view of the mid-thigh of a child with active dermatomyositis.The convex arc of the femoral shaft is difficult to see, and the fascial planes in the muscle are obliterated because of the intense increase in echogenicity in the inflamed muscle. (Courtesy of Dr. D. Stringer.)
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
429
and recovery.385 Systemic complications, particularly abdominal pain or gastrointestinal bleeding, require urgent surgical consultation and may be life threatening, especially early in the disease. Attention to nutritional status and intake of potassium and limitation of total caloric and sodium intake may help minimize the side effects of the glucocorticoid drugs.
Glucocorticoid Drugs ■ Figure 18–20 Cross-sectional magnetic resonance images of the proximal thigh of a child with chronic, severe dermatomyositis.The amount of adipose tissue is amplified, and the increased signal from the quadriceps muscles is highly suggestive of the diagnosis. (Courtesy of Dr. R. Cairns.)
General Supportive Care The approach to management should be based on the knowledge that in most instances, the disease is chronic, but often remits in 2 to 3 years. There is no evidence that any therapy is curative; rather, treatment is aimed at suppression of the immunoinflammatory response, maximal preservation of muscle function and joint range of motion, prevention of complications, and maintenance of general health and normal growth and development. In acute disease, attention must be directed at the adequacy of ventilatory effort and swallowing. Occasionally, weakness is so profound that respiratory assistance, nasogastric feeding, and frequent oral suctioning are required. Each patient should be monitored carefully for swallowing, adequacy of airway, and depth of breathing. Hypoxia may supervene insidiously. In the older child, vital capacity measurements can be a valuable objective measure of response to therapy. Although respiratory problems occur in approximately one third of severely affected children, ventilatory assistance is seldom required. Profound involvement of the thoracic and respiratory muscles occurs in a few children and leads rapidly to increasing dyspnea at rest, agitation, respiratory insufficiency, aspiration, or death.384 Skin care is especially important in children who develop fissures in the axillae and groin or ulcers of the skin over pressure points. Emollients and padding of pressure areas may help prevent breakdown and ulceration. These ulcerations become sites for secondary infections and abscesses, complications that are abetted by the administration of the glucocorticoid drugs. During the course of the disease, the dermatitis may become markedly photosensitive, and waterbased sunscreens with high SPF numbers (30+) are necessary. The rash may or may not respond to the use of low-potency topical glucocorticoid creams. They are generally not recommended because of the secondary atrophic effects that result from long-term application. Frequent counseling and education of the patient and parents are necessary to help allay anxiety and permit understanding of the necessarily slow pace of treatment
Early and adequate treatment with glucocorticoids is probably the single most important factor in improved prognosis during the last 50 years. Acute disease is treated with suppressive doses of the synthetic glucocorticoids (Table 18–15).11,13 Prednisone is preferred to other analogues, such as dexamethasone or triamcinolone, because these steroids may have a more potent myopathic effect (see Chapter 5). The drug is given in a dosage of approximately 2 mg/kg/day, usually in two to three divided doses for the first month of the disease and then, if indicated by the clinical response and a fall in the serum levels of the muscle enzymes, reduced toward a dosage of 1 mg/kg/day, which is also given initially in divided doses. Thereafter, the drug is gradually tapered as permitted by careful monitoring of improvement in muscle weakness and symptoms and by assay of serum levels of the muscle enzymes. Alternate-day glucocorticoid therapy is generally useful only late during the recovery phase of the disease. It is axiomatic that satisfactory clinical control is not attained until the serum enzymes have returned to normal or near-normal levels and have remained there during continued tapering of
TABLE 18–15 Medical Treatment of Juvenile Dermatomyositis Glucocorticoids Initial: oral prednisone, 2 mg/kg/day for 1 mo, or IV methylprednisolone, 30 mg/kg/day for 1–3 days; then: oral prednisone, 1 mg/kg/day, followed by a gradual taper in dose over approximately 2 yr Hydroxychloroquine 6 mg/kg/day in addition to prednisone for control of skin disease and to minimize glucocorticoid requirement Immunosuppressives Methotrexate: 0.35–0.65 mg/kg/wk, may be considered at onset of therapy or later if glucocorticoid response is inadequate Cyclosporine: 35 mg/kg/day Cyclophosphamide: 1 mg/kg/day orally or 500–750 mg/m2/mo Azathioprine: 1–3 mg/kg/day Intravenous Immunoglobulin 2 g/kg/mo Biologic Agent Etanercept: 0.4 mg/kg (maximum of 25 mg) SC twice weekly Autologous Hemopoietic Stem Cell Transplantation
430
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S TABLE 18–16 Characteristics of Steroid-Induced Myopathy ● ● ● ● ●
■ Figure 18–21 Course of a 5-year-old girl with an acute onset of dermatomyositis who eventually recovered completely. Dysphagia resolved within the first month, muscle strength returned to normal during the first year, and the rash ultimately subsided by 3 years, as did all other signs of the disease. (Courtesy of Dr. D.B. Sullivan.)
the steroids and a gradual increase in the physical activity of the child. It is necessary to maintain some children on very-low-dose glucocorticoids even many years after “control” of myositis. It is difficult to be certain when glucocorticoids can be discontinued without risking an exacerbation of the disease. The clinical response of the child to glucocorticoids is not entirely predictable. The fever should abate within a few days, and the serum levels of muscle enzymes should appreciably decrease in the first 1 to 2 weeks of therapy (Figs. 18–21 and 18–22). There may be no significant improvement in muscle strength for 1 to 2 months. Improvement in the dermatitis is unpredictable; the status of the rash at any point is generally not an indi-
Insidious onset Hip flexor weakness and atrophy Normal serum concentrations of muscle enzymes Minimal myopathic changes on electromyography Type II fiber atrophy on muscle biopsy
cation for a change in steroid therapy, because its course does not parallel that of the inflammatory myopathy. An extensive rash at onset or a generalized progression of the dermatitis is, however, a poor prognostic sign.13 Children who require high-dose steroids for long periods develop severe osteopenia and osteoporosis, often with vertebral compression fractures.368 It is unsettled whether supplementation with dietary calcium and vitamin D or administration of calcitonin can prevent this complication. The efficacy and long-term safety of alendronate is being evaluated.386 Cushing’s syndrome and growth retardation result in any child placed on suppressive doses for a period of months. The dosage of the glucocorticoid drug and duration of its use should therefore be kept as low as possible, commensurate with the clinical and laboratory responses to therapy. Steroid myopathy, although rare, may be misinterpreted as an exacerbation of the basic disease process (Table 18–16) (see Chapter 5). The manifestations of this syndrome are insidious onset of hip flexor weakness and atrophy with normal assays of the serum muscle enzymes and minimal myopathic changes on electromyography. Intravenous pulse glucocorticoid therapy has been proposed to gain rapid control of muscle inflammation and angiitis while minimizing the exposure of the child to long-term, high-dose daily steroids.22,387–392 An initial publication by Laxer and associates390 indicated a satisfactory response in six children; subsequently, two additional patients were treated. Both were boys who had mild early disease; a single intravenous pulse of methylprednisolone (30 mg/kg) led to a decrease in the enzyme levels and improved muscle strength over 2 months. There may be a subgroup of children with JDM in whom this initial approach to management will be successful and abrogate the need for long-term daily glucocorticoid treatment. Pachman and colleagues51 advanced the concept that early use of intravenous pulse therapy reduces future disability and the extent of calcinosis. As with any form of therapy, this approach to glucocorticoid administration is not always effective.393 Pulse therapy may be combined advantageously with methotrexate.392
Hydroxychloroquine
■ Figure 18–22 Course of an 8-year-old boy who initially responded to steroid treatment that was discontinued after 12 months. At 18 months, a gluteal abscess developed and an acute relapse occurred that required combined immunosuppressive therapy because of the development of clinically significant steroid toxicity. • , immunosuppression (Courtesy of Dr. D.B. Sullivan.)
Hydroxychloroquine has been recommended as a steroid-sparing agent and as a drug that is effective in treating the dermatitis of JDM.394 Olson and Lindsley395 reported significant improvement of the rash after 3 months and of muscle weakness after 6 months of treatment in a dose of 2 to 5 mg/kg/day in nine children. Anecdotal experience suggests that the addition of
C H A P T E R
hydroxychloroquine to a glucocorticoid regimen is warranted but has modest efficacy. It is alleged that patients with dermatomyositis may be prone to non–life-threatening cutaneous reactions from the antimalarial agents.396
Immunosuppressive Therapy Primary indications for the use of immunosuppressive drugs include glucocorticoid resistance or dependence. In steroid-resistant disease, there is inadequate improvement in muscle strength and a persistence of elevated serum levels of muscle enzymes in response to a closely monitored glucocorticoid program (prednisone, 1 to 2 mg/kg/day for at least 3 to 4 months). Steroid dependence occurs later in the course of the disease and is characterized by failure of the clinical manifestations of disease to remain suppressed during a gradual reduction of the glucocorticoid dose to an acceptable level, recurrence of progressive muscle weakness despite continuing therapy, or unacceptable steroid toxicity. At least four immunosuppressive agents have been employed in resistant disease: methotrexate,392,397–401 azathioprine,400,402 cyclophosphamide,55,403 and cyclosporine.33,34,52,404–407 The efficacy of these drugs is difficult to evaluate because, as with glucocorticoids or hydroxychloroquine, there have been no controlled trials. Of the immunosuppressive drugs, weekly oral or subcutaneous methotrexate is the preferred agent, with a minimal initial dose of 0.35 mg/kg/week. Careful monitoring of dosage and potential toxicity relative to age, height, and weight is mandatory. Benefit is usually evident within 1 to 2 months. No controlled study has been published on the consistency of response; however, a report of 22 adult patients suggested a favorable improvement in more than 75% without major toxicity or hepatic disease.399 An experimental protocol was also evaluated in adults that consisted of intravenous methotrexate (500 mg/m2), followed in 24 hours by oral leucovorin (50 mg/m2) every 6 hours for four doses, which was repeated every 2 weeks for 6 months. Preliminary data did not suggest that this regimen was superior to oral therapy with methotrexate and azathioprine.408 There is little published experience with azathioprine or cyclophosphamide. Crowe and colleagues55 recommended cyclophosphamide in children with a chronic ulcerative course that was unresponsive to glucocorticoids. Although there are no studies of the management of gastrointestinal ulceration, children with this complication warrant consideration of intravenous cyclophosphamide in addition to high-dose glucocorticoids. Intravenous cyclophosphamide therapy is difficult to use, however, and not always successful.409 Chlorambucil has been suggested as an alternative by some investigators but has its own unique toxicities.410 There have been several case reports and reviews of successful cyclosporine therapy.22,404–407,411,412 Some investigators have suggested that this drug should be considered as first-line therapy in the treatment of inflammatory muscle disease.413 Except for renal impairment, cyclosporine has less long-term toxicity than the traditional immunosuppressive drugs, and for this reason, its
18 J U V E N I L E D E R M AT O M Y O S I T I S
431
use should perhaps be considered earlier, rather than later, in the child who is nonresponsive to steroids or steroid dependent or who has evidence of interstitial lung disease.414 Topical FK506 (tacrolimus) has been proposed as therapy for the dermatitis of JDM.415 The efficacy of cyclosporine in the treatment of steroid-resistant or steroid-dependent disease has been reported by Heckmatt and associates.406 In this study, 14 children who had failed to respond fully to glucocorticoids and immunosuppressives were given a dose of 2.5 to 7.5 mg/kg/day (younger children require an increased dosage because drug clearance is age dependent). The investigators reported considerable benefit, with reduced prednisolone requirements in all, including discontinuation of glucocorticoid in 6. Two of 3 nonambulatory patients were able to walk after treatment, and 6 with limited ambulation subsequently regained good independent ambulation. Muscle strength improved but remained significantly reduced. The only notable side effects were hypertension and reversible decreases in renal function.416
Intravenous Immunoglobulin A number of studies and anecdotal reports describe the efficacy of intravenous immunoglobulin (IVIG).417–422 Clinical experience in children has been summarized in the review by Rider and Miller.22 Lang and coworkers417 reported results of administration of IVIG to five children who were steroid resistant or steroid dependent. All patients exhibited improved muscle strength and diminished rash over the 9-month period with infusions every 4 weeks of 1 g/kg/day on each of 2 consecutive days. A follow-up report summarized the experience of these investigators in a retrospective review of 18 patients.422 Controlled studies enrolling adults have been published,39 including those by the National Institutes of Health.423,424 In these reports, IVIG was of significant benefit, particularly if used early in the disease course and especially with respect to the skin changes. However, without controlled clinical trials of this therapeutic approach in children, no firm conclusions can be reached regarding efficacy.
Plasmapheresis Plasmapheresis has been of benefit in a few children.233,425–428 Studies suggest that the therapeutic effect may be minimal to nonexistent in adults.429 A report of a single patient with JDM who was treated by extracorporeal photopheresis (i.e., ultraviolet activation of methoxsalen to covalently crosslink lymphocyte DNA) is an experimental approach of interest.430
Biologic Agents and Stem Cell Transplantation The TNF-α inhibitors, including recombinant soluble human TNF-α receptor (p75)–Fc fusion protein and chimeric monoclonal antibodies to TNF-α, have been used with promising results in a few patients.376 In one trial of etanercept,431 9 of 10 children improved in activity, skin, and muscle scores and in functional ability. The use of rituximab, a monoclonal antibody to B cells, is in clinical trials.432 Infliximab produced encouraging results
432
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
in one report, but doses of 8 to 10 mg/kg were required to maintain a satisfactory response.433 Experience with autologous hemopoietic stem cell transplantation for JDM is limited, and these children are at high risk for developing severe viral infections.434
Physical and Occupational Therapy Physiotherapy should be initiated at the time of diagnosis. Although skeletal muscles are actively inflamed, the focus of attention should be on preventing loss of range of motion by giving twice- to thrice-daily passive range of motion exercises to all joints, with the use of gentle stretching to regain lost range. Splinting of knees, elbows, or wrists at night or during periods of rest helps to achieve these goals. During the healing phase, the physical therapy program is increased to normalize function as nearly as possible and to minimize development of contractures from muscle weakness or atrophy. Muscle strengthening should be added to the exercise program only after clinical evidence of acute inflammation has subsided.
Management of Calcinosis None of many approaches to the treatment of calcinosis has been consistently effective.252,435–447 Therapy has included colchicine, aluminum hydroxide, probenecid, diphosphonates, diltiazem, intravenous ethylenediaminetetraacetic acid, and warfarin. Colchicine may suppress local and systemic signs of inflammation associated with calcinosis.443 There is general agreement that early aggressive therapy with glucocorticoids results in decreased frequency and severity of calcinosis.51,208 Experimental data suggest that TNF blockade may be beneficial because overexpression of the TNF-α 308A allele is associated with a long duration of active disease and pathologic calcifications.163 Surgical excision of calcifications that mechanically interfere with function or have resulted in breakdown of skin may be indicated,448 although infections in such sites are a risk.449 The natural history of many of these calcific deposits is that they spontaneously begin to regress after months or years, coincident with inactivity of the muscle disease and increasing mobility of the patient. However, the one fourth of children who develop calcinosis in interfascial planes tend to have persistent lesions.195 Hypercalcemia and hypercalciuria have been reported during spontaneous resolution of calcinosis.442,450
Management of Lipodystrophy Recent data have emphasized the role of leptin deficiency in the abnormalities characteristic of lipodystrophy and the potential role of replacement therapy in treating this complication of dermatomyositis. This adipocyte hormone is critical in energy homeostasis and glycemic control. Oral and colleagues231 administered escalating doses of recombinant methionyl human leptin to nine female patients with leptin levels less than 4 ng/mL (0.32 nmol/mL) subcutaneously twice daily for 4 months. With treatment, triglyceride levels decreased along with hepatic volume and the resting metabolic rate improved. Peterson and colleagues451 extended these observations on three of these patients and confirmed a marked improvement in insulin sensitivity in the liver and muscle and in whole-body insulin-stimulated glucose metabolism.
COURSE OF THE DISEASE AND PROGNOSIS The course of JDM can be divided into four clinical phases (Table 18–17).196 The early prodromal phase is supplanted by a period of progressive muscle weakness and rash that then stabilizes for 1 to 2 years before recovery (Table 18–18). Most children pursue this type of a uniphasic course.11,13 The entire disease duration can be as brief as 8 months with complete recovery, or it can last 2 or more years with a chronically active disease and a continuing requirement for treatment with glucocorticoids. Family interviews documented the persistence of symptoms for at least 25% of 229 children at 36 months after diagnosis.452 Acute exacerbations and remissions without any stabilization of the initial course of the disease have occurred in approximately 20% of children.25 There has been a significant improvement in outcome over the past 3 decades (Table 18–19). In a multicenter study, 72% of patients had no or minimal disability an average of 7 years after onset.453 Risk factors for a poor prognosis include unremitting severe disease activity, cutaneous ulcerations, extensive calcinosis, dysphagia or dysphonia, advanced nail fold TABLE 18–17 Clinical Phases of Juvenile Dermatomyositis 1. Prodromal period with nonspecific symptoms (weeks to months)* 2. Progressive muscle weakness and rash (days to weeks) 3. Persistent weakness, rash, and active myositis (up to 2 years) 4. Recovery with or without residual muscle atrophy, contractures, and calcinosis *Durations of the clinical phases in part depend on treatment. Adapted from Spencer CH, Hanson V, Singsen BH, et al: Course of treated juvenile dermatomyositis. J Pediatr 105: 399–408, 1984.
TABLE 18–18 Clinical Features of Juvenile Dermatomyositis During the Course of the Disease Constitutional signs and symptoms (100%) Muscle weakness (90–95%) Pelvic girdle (95%) Shoulder girdle (75%) Neck flexors (60%) Pharyngeal muscles (12–85%) Distal muscles (30%) Facial and extraocular muscles (5%) Muscle contractures and atrophy (62–90%) Muscle pain and tenderness (30–80%) Induration of muscle (50–100%) Skin lesions (85–100%) Heliotrope rash of eyelids (10–15%) Erythematous rash of malar area and V of chest(30–40%) Subcutaneous and periorbital edema (60–90%) Periungual and periarticular rash (Gottron’s papules) (80–100%) Photosensitive rash (5–40%) Ulcerations (25%) Lipodystrophy (25% ) Raynaud’s phenomenon (2–15%) Acute arthritis and arthralgia (10–65%) Gastrointestinal signs and symptoms (10–60%) Mucosal ulcerations and pharyngitis (10–45%) Calcinosis (10–40%) Pulmonary disease (80%) Restrictive defect (80%) Fibrosis (<1%)
C H A P T E R
TABLE 18–19 Prognosis of Juvenile Dermatomyositis Outcome Normal to good functional outcome Minimal atrophy or contractures Calcinosis* Wheelchair dependence Death
Percent of Patients 65–80 25 20–40 5 5–10
*Children with calcinosis were also included in the other categories.
capillary abnormalities, a noninflammatory vasculopathy on muscle biopsy, and the presence of specific MSAs. Delay in treatment and inadequate treatment are also important risk factors. Determinants of physical function are receiving further elucidation.223 Newly developed outcome assessment tools for disease activity and physical function are being validated in adult and pediatric patients through international collaborative studies.180,182,454 The studies of Crowe and associates55 identified a group of children with noninflammatory vasculopathy who had extensive and chronic ulcerative cutaneous disease. These children and a similar group reported earlier by Banker and Victor7 were characterized by significant systemic complications, including fatal gastrointestinal hemorrhage. At a minimum, children with severe generalized erythroderma and cutaneous ulcerations often develop extensive calcinosis and significant overall functional impairment. Approximately 5% of children eventually develop a clinical disease that is more typical of systemic vasculitis.55,209 A small number of children late in the course may assume more of the characteristics of scleroderma with sclerodactyly and cutaneous atrophy13 or develop lipodystrophy with insulin resistance, acanthosis nigricans, or a recurrence of arthritis.230,341 Late progression has been reported with a recurrence of active disease after a prolonged remission455,456 or smoldering, persistent activity many years after onset with multiple physical or dermatologic sequelae.457–459 Of interest in this regard is the risk from pregnancy in women who have had or have JDM.460–462 Depending on the activity of the disease, residual muscle weakness, calcinosis, and general debility, pregnancy should be considered high risk for both mother and baby.
Calcinosis Historically, 20% to 40% of children with JDM have developed calcinosis (see Tables 18–5 and 18–19).13,195,453 Children with extensive calcinosis were often those who had suffered from a severe and unremitting course.195 In those children and to a lesser extent in others, calcinosis was responsible for more long-term disability with limitation of movement of involved muscles or contiguous joints than the residual effects of myositis. Calcific deposits may develop as early as 6 months or as late as 10 to 20 years after disease onset. Calcinosis is occasionally present at first presentation of the child to a physician. In this situation, it is surmised that the child had a prolonged but mild and therefore undiagnosed myositis before development of the calcinosis.205 Rarely, calcinosis is accompanied by hypercalcemia.442 Trauma may play a role in the generation of calcific deposits, because they also tend to occur in surgical incisions and over pressure points. A study by Moore and coworkers449 concluded that calcinosis was associated with preceding staphylo-
18 J U V E N I L E D E R M AT O M Y O S I T I S
433
coccal infection and high levels of IgE and IgE antistaphylococcal antibodies. Granulocyte chemotaxis to staphylococci was depressed, an effect that was mediated by unidentified serum factors.
Functional Disability In children with a typical uniphasic course, functional outcome is usually excellent, although minor flexion contractures and residual skin changes may persist (see Table 18–19). In others in whom the disease remains active beyond 3 years, there may be smoldering myositis and dermatitis with deposition of calcium salts and progressive loss of function. Adverse factors that influence outcome are listed in Table 18–20. Early and adequate steroid treatment has the greatest impact on a favorable outcome.10,13 Functional outcome appears best in children who have been diagnosed shortly after onset and treated vigorously, perhaps with initial steroid-pulse administration.390 Most survivors are able to function independently as adults, although some have flexion contractures and residual atrophy of skin or muscle.453,458 Four Canadian centers evaluated functional outcome in 65 children treated between 1984 and 1995 (46 girls and 19 boys) with a mean follow-up of 7.2 years (range, 3.2 to 13.9 years).453 A monocyclic course was characteristic of 37% and a chronic continuous or polycyclic one in 63%. Favorable outcomes were predominant; only 8% had moderate to severe disability, 34% had developed calcinosis, and there was one death. However, chronic disease and rash persisted in many patients in this cohort who were still on medical therapy more than 3 years after disease onset.
Psychosocial Outcome A study by Miller and associates463 suggested that a number of children who enter adulthood continue to have psychologic problems and learning disabilities based on unrecognized cerebral abnormalities that occurred at onset of disease. Another review of late outcome in JDM indicated that the educational achievements and employment status of 18 patients were better than those of the general adult population or a comparable group who had had chronic arthritis.464 Significant disability related to calcinosis or contractures developed in 3 patients, 6 had persistent muscle weakness, and 7 had recurrent rash. Raynaud’s phenomenon (33%), arthritis (22%), and TABLE 18–20 Factors that Adversely Influence Outcome Disease-Related Factors Rapid onset and extensive weakness Extensive cutaneous vasculitis with ulceration Gastrointestinal vasculitis Severe endarteropathy and infarction in muscle biopsy specimen Therapy-Related Factors Delay in diagnosis and institution of therapy Inadequate dose or duration of glucocorticoid therapy Minimal initial or sustained response to glucocorticoid therapy
434
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
subcutaneous nodules occurred in others, and minor increases in the serum concentrations of CK that did not correlate with the presence of muscle weakness or rash were observed in 7.
Death Long-term survival is somewhat better than 90%; in the pre-steroid era, this disease was associated with a mortality rate that approached 40%.13,55,463,465 Children who survived often had devastating residual problems of contractures and muscular atrophy. Fatalities most often occur within 2 years of onset and are often associated with progressive involvement of skin or muscle that is unresponsive to steroids. This observation suggests that the basic nature of the inflammatory disease, its early treatment and response, the presence of widespread vasculitis, and involvement of other organ systems (e.g., gastrointestinal tract, lungs) are major factors that should be assessed in estimating prognosis. Death most often results from respiratory insufficiency or pneumonitis or from acute gastrointestinal ulceration and bleeding. Surgical intervention in the latter group of children is often not successful. There were 6 deaths in the Ann Arbor series of 71 children.13 One young girl died 5 years after onset as a result of cardiorespiratory collapse. Necropsy examination documented no active myositis. Three patients died at 2 months, 6 months, and 4 years after onset from gastrointestinal hemorrhage. Another girl died from a subdural hematoma caused by a fall from a wheelchair. A boy succumbed shortly after onset of disease from respiratory insufficiency and hypoxia. In the series of Spencer and colleagues,196 7 of 66 patients died. Five deaths occurred early (1 to 11 months from diagnosis) and were related to sepsis (1), gastrointestinal perforation (2), and unresponsive muscle weakness and pneumonitis (2). One patient died of pulmonary fibrosis with cor pulmonale 9 years after disease onset, and one patient committed suicide 16 years after onset. In the report of 39 patients by Miller and associates,463 there were 10 deaths (26%); 8 of these were children seen before 1972. No child who had received intensive glucocorticoid treatment (and azathioprine in some cases) died. All deaths were associated with bowel perforation or aspiration pneumonitis and occurred an average of 2.5 years after onset. The improved outcome since 1972 (92% survival rate) was related to early and appropriate steroid regimens, better clinical assessment, followup with sequential serum muscle enzyme determinations, and optimal management of complications. Five of the 24 surviving patients were still on glucocorticoids at follow-up. Outcomes of children in geographic areas other than North America are being evaluated.459
R E F E R E N C E S 1. Hepp P: Uber einen Fall von acuter parenchymatoser myositis, welche Geschwulste bildete und Fluctuation vortauschte. Klin Wochenschr 24: 389, 1887. 2. Jackson H: Myositis universalis acuta infectiosa, with a case. Boston Med Surg J 116: 498, 1887. 3. Wagner E: Ein Fall von acuter Polymyositis. Dtsch Arch Klin Med 40: 241, 1887. 4. Unverricht H: Uber eine eigentumliche Form von acuter Muskelentzundung mit einem der Trichinose ahnelnden Krankheitsbilde. Munch Med Wochenschr 34: 1887. 5. Karelitz S, Welt SK: Dermatomyositis. Am J Dis Child 43: 1134, 1932. 6. Wedgwood RJ, Cook C, Cohen J: Dermatomyositis: Report of 26 cases in children with a discussion of endocrine therapy in 13. Pediatrics 12: 447, 1953.
7. Roberts HM, Brunsting LA: Dermatomyositis in childhood. A summary of 40 cases. Postgrad Med 16: 396, 1954. 8. Carlisle JW, Good RA: Dermatomyositis in childhood: Report of studies on 7 cases and a review of literature. Lancet 79: 266, 1959. 9. Bitnun S, Daeschner CW Jr, Travis LB, et al: Dermatomyositis. J Pediatr 64: 101, 1964. 10. Hanson V, Kornreich H: Systemic rheumatic disorders (“collagen disease”) in childhood: Lupus erythematosus, anaphylactoid purpura, dermatomyositis, and scleroderma. I. Bull Rheum Dis 17: 435–440, 1967. 11. Sullivan DB, Cassidy JT, Petty RE, et al: Prognosis in childhood dermatomyositis. J Pediatr 80: 555–563, 1972. 12. Rose AL: Childhood polymyositis. A follow-up study with special reference to treatment with corticosteroids. Am J Dis Child 127: 518–522, 1974. 13. Sullivan DB, Cassidy JT, Petty RE: Dermatomyositis in the pediatric patient. Arthritis Rheum 20: 327–331, 1977. 14. Hanson V: Dermatomyositis, scleroderma, and polyarteritis nodosa. Clin Rheum Dis 2: 445, 1976. 15. Scheueermann H: Zur Klinuk und Pathogenese der Dermatomyositis (Polymyositis). Arch Dermatol Syphilis 178: 414, 1939. 16. Hecht MS: Dermatomyositis in childhood. J Pediatr 17: 791, 1940. 17. Selander P: Dermatomyositis in early childhood. Acta Med Scand 246 (Suppl): 187, 1950. 18. Batten FE: Case of dermatomyositis in a child, with pathological report. Br J Child Dis 9: 247, 1912. 19. Pearson CM: Polymyositis. Annu Rev Med 17: 63–82, 1966. 20. Cook WE, Rosen FS, Banker BQ: Dermatomyositis and focal scleroderma. Pediatr Clin North Am 10: 979, 1963. 21. Martini A, Ravelli A, Aramini L, et al: Juvenile dermatomyositis. Pediatr Med Chir 12: 587–591, 1990. 22. Rider LG, Miller FW: Classification and treatment of the juvenile idiopathic inflammatory myopathies. Rheum Dis Clin North Am 23: 619–655, 1997. 23. Peloro TM, Miller OF III, Hahn TF, et al: Juvenile dermatomyositis: a retrospective review of a 30-year experience. J Am Acad Dermatol 45: 28–34, 2001. 24. Rennebohm R: Juvenile dermatomyositis. Pediatr Ann 31: 426–433, 2002. 25. Ramanan AV, Feldman BM: Clinical features and outcomes of juvenile dermatomyositis and other childhood onset myositis syndromes. Rheum Dis Clin North Am 28: 833–857, 2002. 26. Sallum AM, Kiss MH, Sachetti S, et al: Juvenile dermatomyositis: clinical, laboratorial, histological, therapeutical and evolutive parameters of 35 patients. Arq Neuropsiquiatr 60: 889–899, 2002. 27. Bohan A, Peter JB: Polymyositis and dermatomyositis (first of two parts). N Engl J Med 292: 344–347, 1975. 28. Bohan A, Peter JB: Polymyositis and dermatomyositis (second of two parts). N Engl J Med 292: 403–407, 1975. 29. Bohan A, Peter JB, Bowman RL, et al: Computer-assisted analysis of 153 patients with polymyositis and dermatomyositis. Medicine (Baltimore) 56: 255–286, 1977. 30. Hanissian AS, Masi AT, Pitner SE, et al: Polymyositis and dermatomyositis in children: an epidemiologic and clinical comparative analysis. J Rheumatol 9: 390–394, 1982. 31. Serratrice G, Schiano A, Pellissier JF, et al: Anatomoclinical expressions of polymyositis in the child. 23 cases. Ann Pediatr (Paris) 36: 237–243, 1989. 32. Bartley GB, Gibson LE: Blepharochalasis associated with dermatomyositis and acute lymphocytic leukemia. Am J Ophthalmol 113: 727–728, 1992. 33. Sherry DD, Haas JE, Milstein JM: Childhood polymyositis as a paraneoplastic phenomenon. Pediatr Neurol 9: 155–156, 1993. 34. Falcini F, Taccetti G, Trapani S, et al: Acute lymphocytic leukemia with dermatomyositis-like onset in childhood. J Rheumatol 20: 1260–1262, 1993. 35. Hatada T, Aoki I, Ikeda H, et al: Dermatomyositis and malignancy: case report and review of the Japanese literature. Tumori 82: 273–275, 1996. 36. Lang BA, Laxer RM, Thorner P, et al: Pediatric onset of Behçet’s syndrome with myositis: case report and literature review illustrating unusual features. Arthritis Rheum 33: 418–425, 1990. 37. Blaszczyk M, Jablonska S, Szymanska-Jagiello W, et al: Childhood scleromyositis: an overlap syndrome associated with PM-Scl antibody. Pediatr Dermatol 8: 1–8, 1991. 38. Rider LG, Okada S, Sherry DD: Epidemiologic features and environmental exposures associated with illness onset in juvenile idiopathic inflammatory myopathy. Arthritis Rheum 38 (Suppl): 362, 1995. 39. Riggs JE, Schochet SS Jr, Gutmann L, et al: Childhood onset inclusion body myositis mimicking limb-girdle muscular dystrophy. J Child Neurol 4: 283–285, 1989. 40. Isaacson G, Chan KH, Heffner RR Jr: Focal myositis. A new cause for the pediatric neck mass. Arch Otolaryngol Head Neck Surg 117: 103–105, 1991. 41. Agrawal BL, Giesen PC: Eosinophilic myositis. An unusual cause of pseudotumor and eosinophilia. JAMA 246: 70–71, 1981. 42. Medsger TA Jr, Dawson WN Jr, Masi AT: The epidemiology of polymyositis. Am J Med 48: 715–723, 1970. 43. Oddis CV, Conte CG, Steen VD, et al: Incidence of polymyositis-dermatomyositis: a 20-year study of hospital diagnosed cases in Allegheny County, PA 1963–1982. J Rheumatol 17: 1329–1334, 1990. 44. Symmons DP, Sills JA, Davis SM: The incidence of juvenile dermatomyositis: results from a nation-wide study. Br J Rheumatol 34: 732–736, 1995.
C H A P T E R 45. Kaipiainen-Seppanen O, Savolainen A: Incidence of chronic juvenile rheumatic diseases in Finland during 1980–1990. Clin Exp Rheumatol 14: 441–444, 1996. 46. Fujikawa S, Okuni M: A nationwide surveillance study of rheumatic diseases among Japanese children. Acta Paediatr Jpn 39: 242–244, 1997. 47. Mendez EP, Lipton R, Ramsey-Goldman R, et al: US incidence of juvenile dermatomyositis, 1995–1998: results from the National Institute of Arthritis and Musculoskeletal and Skin Diseases Registry. Arthritis Rheum 49: 300–305, 2003. 48. Rose AL, Walton JN: Polymyositis: a survey of 89 cases with particular reference to treatment and prognosis. Brain 89: 747–768, 1966. 49. Benbassat J, Geffel D, Zlotnick A: Epidemiology of polymyositis-dermatomyositis in Israel, 1960–76. Isr J Med Sci 16: 197–200, 1980. 50. Barwick DD, Walton JN: Polymyositis. Am J Med 35: 646, 1963. 51. Pachman LM: Inflammatory myopathy in children. Rheum Dis Clin North Am 20: 919–942, 1994. 52. Symmons DP, Jones M, Osborne J, et al: Pediatric rheumatology in the United Kingdom: data from the British Pediatric Rheumatology Group National Diagnostic Register. J Rheumatol 23: 1975–1980, 1996. 53. Shehata R, Al Mayouf S, al Dalaan A, et al: Juvenile dermatomyositis: clinical profile and disease course in 25 patients. Clin Exp Rheumatol 17: 115–118, 1999. 54. Pachman LM, Hayford JR, Hochberg MC, et al: New-onset juvenile dermatomyositis: comparisons with a healthy cohort and children with juvenile rheumatoid arthritis. Arthritis Rheum 40: 1526–1533, 1997. 55. Crowe WE, Bove KE, Levinson JE, et al: Clinical and pathogenetic implications of histopathology in childhood polydermatomyositis. Arthritis Rheum 25: 126–139, 1982. 56. Singalavanija S, Liamsuwan S, Limpongsanurak W, et al: Juvenile dermatomyositis in Thai children. J Med Assoc Thai 84: 1527–1533, 2001. 57. Whitaker JN: Inflammatory myopathy: a review of etiologic and pathogenetic factors. Muscle Nerve 5: 573–592, 1982. 58. Plotz PH, Dalakas M, Leff RL, et al: Current concepts in the idiopathic inflammatory myopathies: polymyositis, dermatomyositis, and related disorders. Ann Intern Med 111: 143–157, 1989. 59. Pachman LM: Juvenile dermatomyositis: immunogenetics, pathophysiology, and disease expression. Rheum Dis Clin North Am 28: vii, 579–602, 2002. 60. Abramson LS, Albertini RJ, Pachman LM, et al: Association among somatic HPRT mutant frequency, peripheral blood T-lymphocyte clonality, and serologic parameters of disease activity in children with juvenile onset dermatomyositis. Clin Immunol 91: 61–67, 1999. 61. Currie S, Saunders M, Knowles M, et al: Immunological aspects of polymyositis. The in vitro activity of lymphocytes on incubation with muscle antigen and with muscle cultures. Q J Med 40: 63–84, 1971. 62. Mastaglia FL, Currie S: Immunological and ultrastructural observations on the role of lymphoid cells in the pathogenesis of polymyositis. Acta Neuropathol (Berl) 18: 1–16, 1971. 63. Esiri MM, MacLennan IC, Hazleman BL: Lymphocyte sensitivity to skeletal muscle in patients with polymyositis and other disorders. Clin Exp Immunol 14: 25–35, 1973. 64. Lisak RP, Zweiman B: Mitogen and muscle extract induced in vitro proliferative responses in myasthenia gravis, dermatomyositis, and polymyositis. J Neurol Neurosurg Psychiatry 38: 521–524, 1975. 65. Currie S: Destruction of muscle cultures by lymphocytes from cases of polymyositis. Acta Neuropathol (Berl) 15: 11–19, 1970. 66. Johnson RL, Fink CW, Ziff M: Lymphotoxin formation by lymphocytes and muscle in polymyositis. J Clin Invest 51: 2435–2449, 1972. 67. Dawkins RL, Mastaglia FL: Cell-mediated cytotoxicity to muscle in polymyositis. Effect of immunosuppression. N Engl J Med 288: 434–438, 1973. 68. Haas DC, Arnason BG: Cell-mediated immunity in polymyositis. Creatine phosphokinase release from muscle cultures. Arch Neurol 31: 192–196, 1974. 69. Iannaccone ST, Bowen DE, Samaha FJ: Cell-mediated cytotoxicity and childhood dermatomyositis. Arch Neurol 39: 400–402, 1982. 70. Miller ML, Lantner R, Pachman LM: Natural and antibody-dependent cellular cytotoxicity in children with systemic lupus erythematosus and juvenile dermatomyositis. J Rheumatol 10: 640–642, 1983. 71. Haas DC: Absence of cell-mediated cytotoxicity to muscle cultures in polymyositis. J Rheumatol 7: 671–676, 1980. 72. Giorno R, Barden MT, Kohler PF, et al: Immunohistochemical characterization of the mononuclear cells infiltrating muscle of patients with inflammatory and noninflammatory myopathies. Clin Immunol Immunopathol 30: 405–412, 1984. 73. McDouall RM, Dunn MJ, Dubowitz V: Nature of the mononuclear infiltrate and the mechanism of muscle damage in juvenile dermatomyositis and Duchenne muscular dystrophy. J Neurol Sci 99: 199–217, 1990. 74. Englund P, Nennesmo I, Klareskog L, et al: Interleukin-1 alpha expression in capillaries and major histocompatibility complex class I expression in type II muscle fibers from polymyositis and dermatomyositis patients: important pathogenic features independent of inflammatory cell clusters in muscle tissue. Arthritis Rheum 46: 1044–1055, 2002. 75. Li CK, Varsani H, Holton JL, et al: MHC class I overexpression on muscles in early juvenile dermatomyositis. J Rheumatol 31: 605–609, 2004. 76. Falcini F, Calzolari A, Generini S, et al: Bcl-2, p53 and c-myc expression in juvenile dermatomyositis. Clin Exp Rheumatol 18: 643–646, 2000.
18 J U V E N I L E D E R M AT O M Y O S I T I S
435
77. Whitaker JN, Engel WK: Vascular deposits of immunoglobulin and complement in idiopathic inflammatory myopathy. N Engl J Med 286: 333–338, 1972. 78. Spencer CH, Jordon SC, Hanson V: Circulating immune complexes in juvenile dermatomyositis. Arthritis Rheum 23: 750, 1980. 79. Iannaccone ST, Bowen D, Yarom A, et al: In vitro study of cytotoxic factors against endothelium in childhood dermatomyositis. Arch Neurol 41: 862–864, 1984. 80. Kissel JT, Mendell JR, Rammohan KW: Microvascular deposition of complement membrane attack complex in dermatomyositis. N Engl J Med 314: 329–334, 1986. 81. Goncalves FG, Chimelli L, Sallum AM, et al: Immunohistological analysis of CD59 and membrane attack complex of complement in muscle in juvenile dermatomyositis. J Rheumatol 29: 1301–1307, 2002. 82. Scott JP, Arroyave C: Activation of complement and coagulation in juvenile dermatomyositis. Arthritis Rheum 30: 572–576, 1987. 83. Guzman J, Petty RE, Malleson PN: Monitoring disease activity in juvenile dermatomyositis: the role of von Willebrand factor and muscle enzymes. J Rheumatol 21: 739–743, 1994. 84. Pachman LM, Cooke N: Juvenile dermatomyositis: a clinical and immunologic study. J Pediatr 96: 226–234, 1980. 85. Lutz J, Huwiler KG, Fedczyna T, et al: Increased plasma thrombospondin-1 (TSP-1) levels are associated with the TNF alpha-308A allele in children with juvenile dermatomyositis. Clin Immunol 103: 260–263, 2002. 86. Bloom BJ, Miller LC, Blier PR: Soluble adhesion molecules in pediatric rheumatic diseases. J Rheumatol 29: 832–836, 2002. 87. Sallum AM, Marie SK, Wakamatsu A, et al: Immunohistochemical analysis of adhesion molecule expression on muscle biopsy specimens from patients with juvenile dermatomyositis. J Rheumatol 31: 801–807, 2004. 88. Rider LG, Artlett CM, Foster CB, et al: Polymorphisms in the IL-1 receptor antagonist gene VNTR are possible risk factors for juvenile idiopathic inflammatory myopathies. Clin Exp Immunol 121: 47–52, 2000. 89. Rider L, Ahmed A, Beausang L, et al: Evaluations of interleuken-1 receptor antagonist, sTNFR, sIL2R, and IL-10 in juvenile idiopathic inflammatory myopathies suggest a role for monocyte 1 macrophage and B lymphocyte activation. J Rheumatol 27 (Suppl): 82, 2002. 90. Uzel G, Pachman LM: Cytokines in juvenile dermatomyositis pathophysiology: potential and challenge. Curr Opin Rheumatol 15: 691–697, 2003. 91. Artlett CM, Ramos R, Jiminez SA, et al: Chimeric cells of maternal origin in juvenile idiopathic inflammatory myopathies. Childhood Myositis Heterogeneity Collaborative Group. Lancet 356: 2155–2156, 2000. 92. Reed AM, Picornell YJ, Harwood A, et al: Chimerism in children with juvenile dermatomyositis. Lancet 356: 2156–2157, 2000. 93. Reed AM, McNallan K, Wettstein P, et al: Does HLA-dependent chimerism underlie the pathogenesis of juvenile dermatomyositis? J Immunol 172: 5041–5046, 2004. 94. Artlett CM, Miller FW, Rider LG: Persistent maternally derived peripheral microchimerism is associated with the juvenile idiopathic inflammatory myopathies. Rheumatology (Oxf) 40: 1279–1284, 2001. 95. Reed AM, Diegelj M, Kredich D, et al: Geographic clustering of juvenile dermatomyositis in subjects. Arthritis Rheum 41 (Suppl): S265, 1998. 96. Tezak Z, Hoffman EP, Lutz JL, et al: Gene expression profiling in DQA1*0501+ children with untreated dermatomyositis: a novel model of pathogenesis. J Immunol 168: 4154–4163, 2002. 97. Ruff RL, Secrist D: Viral studies in benign acute childhood myositis. Arch Neurol 39: 261–263, 1982. 98. Bowles NE, Dubowitz V, Sewry CA, et al: Dermatomyositis, polymyositis, and Coxsackie-B-virus infection. Lancet 1: 1004–1007, 1987. 99. Christensen ML, Pachman LM, Maryjowski MC: Antibody to coxsackie B virus: increased incidence in sera from children with recently diagnosed juvenile dermatomyositis. Arthritis Rheum 26(Suppl): S24, 1983. 100. Christensen ML, Pachman LM, Schneiderman R, et al: Prevalence of Coxsackie B virus antibodies in patients with juvenile dermatomyositis. Arthritis Rheum 29: 1365–1370, 1986. 101. Pachman LM, Nigro R, Rowley A, et al: Polymerase chain reaction does not detect enteroviral or coxsackievirus B mRNA in fresh frozen muscle biopsies from 20 children with juvenile dermatomyositis. Arthritis Rheum 35 (Suppl): S65, 1992. 102. Lundberg A: Myalgia crures epedemica. Acta Paediatr 46: 18, 1957. 103. Middleton PJ, Alexander RM, Szymanski MT: Severe myositis during recovery from influenza. Lancet 2: 533–535, 1970. 104. Dietzman DE, Schaller JG, Ray CG, et al: Acute myositis associated with influenza B infection. Pediatrics 57: 255–258, 1976. 105. Mackay MT, Kornberg AJ, Shield LK, et al: Benign acute childhood myositis: laboratory and clinical features. Neurology 53: 2127–2131, 1999. 106. Lehmann HW, Kuhner L, Beckenlehner K, et al: Chronic human parvovirus B19 infection in rheumatic disease of childhood and adolescence. J Clin Virol 25: 135–143, 2002. 107. Landry M, Winkelmann RK: Tubular cytoplasmic inclusion in dermatomyositis. Mayo Clin Proc 47: 479–492, 1972. 108. Hanissian AS, Martinez AJ, Jabbour JT, et al: Vasculitis and myositis secondary to rubella vaccination. Arch Neurol 28: 202–204, 1973. 109. Cotterill JA, Shapiro H: Dermatomyositis after immunization. Lancet 2: 1158–1159, 1978. 110. Ehrengut W: Dermatomyositis and vaccination. Lancet 1: 1040–1041, 1978.
436
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
111. Kass E, Straume S, Munthe E: Dermatomyositis after B.C.G. vaccination. Lancet 1: 772, 1978. 112. Martini A, Ravelli A, Albani S, et al: Recurrent juvenile dermatomyositis and cutaneous necrotizing arteritis with molecular mimicry between streptococcal type 5 M protein and human skeletal myosin. J Pediatr 121: 739–742, 1992. 113. Albani S: Infection and molecular mimicry in autoimmune diseases of childhood. Clin Exp Rheumatol 12 (Suppl 10): S35–S41, 1994. 114. Massa M, Costouros N, Mazzoli F, et al: Self epitopes shared between human skeletal myosin and Streptococcus pyogenes M5 protein are targets of immune responses in active juvenile dermatomyositis. Arthritis Rheum 46: 3015–3025, 2002. 115. Hendrickx GF, Verhage J, Jennekens FG, et al: Dermatomyositis and toxoplasmosis. Ann Neurol 5: 393–395, 1979. 116. Phillips PE, Kassan SS, Kagen LJ: Increased toxoplasma antibodies in idiopathic inflammatory muscle disease. A case-controlled study. Arthritis Rheum 22: 209–214, 1979. 117. Schroter HM, Sarnat HB, Matheson DS, et al: Juvenile dermatomyositis induced by toxoplasmosis. J Child Neurol 2: 101–104, 1987. 118. Webster AD: Inflammatory disorders of muscle. Echovirus disease in hypogammaglobulinaemic patients. Clin Rheum Dis 10: 189–203, 1984. 119. Cassidy JT, Burt A, Petty R, et al: Selective IgA deficiency in connective tissue diseases. N Engl J Med 280: 275, 1969. 120. Carroll JE, Silverman A, Isobe Y, et al: Inflammatory myopathy, IgA deficiency, and intestinal malabsorption. J Pediatr 89: 216–219, 1976. 121. Leddy JP, Griggs RC, Klemperer MR, et al: Hereditary complement (C2) deficiency with dermatomyositis. Am J Med 58: 83–91, 1975. 122. Chou SM, Miike T: Ultrastructural abnormalities and perifascicular atrophy in childhood dermatomyositis with special reference to transverse tubular system—sarcoplasmic reticulum junctions. Arch Pathol Lab Med 105: 76–85, 1981. 123. Baringer JR, Swoveland P: Tubular aggregates in endoplasmic reticulum: evidence against their viral nature. J Ultrastruct Res 41: 270–276, 1972. 124. Katsuragi S, Miyayama H, Takeuchi T: Picornavirus-like inclusions in polymyositis—aggregation of glycogen particles of the same size. Neurology 31: 1476–1480, 1981. 125. Fidzianska A, Goebel HH: Tubuloreticular structures (TRS) and cylindric confronting cisternae (CCC) in childhood dermatomyositis. Acta Neuropathol (Berl) 79: 310–316, 1989. 126. Norton WL: Comparison of the microangiopathy of systemic lupus erythematosus, dermatomyositis, scleroderma, and diabetes mellitus. Lab Invest 22: 301–308, 1970. 127. Hashimoto K, Robison L, Velayos E, et al: Dermatomyositis. Electron microscopic, immunologic, and tissue culture studies of paramyxovirus-like inclusions. Arch Dermatol 103: 120–135, 1971. 128. Fukuyama Y, Ando T, Yokota J: Acute fulminant myoglobinuric polymyositis with picornavirus-like crystals. J Neurol Neurosurg Psychiatry 40: 775–781, 1977. 129. Gyorkey F, Cabral GA, Gyorkey PK, et al: Coxsackievirus aggregates in muscle cells of a polymyositis patient. Intervirology 10: 69–77, 1978. 130. Webb JN: Experimental immune myositis in guinea pigs. J Reticuloendothel Soc 7: 305–316, 1970. 131. Suenaga Y: Experimental pathological studies on homologous immunization with the skeletal muscle tissue of the rabbit. Zasshi Tokyo Ika Daigaku 25: 975–1007, 1967. 132. Morgan G, Peter JB, Newbould BB: Experimental allergic myositis in rats. Arthritis Rheum 14: 599–609, 1971. 133. Kakulas BA: In vitro destruction of skeletal muscle by sensitized cells. Nature 210: 1115–1118, 1966. 134. Currie S: Experimental myositis: the in-vivo and in-vitro activity of lymphnode cells. J Pathol 105: 169–185, 1971. 135. Smith PD, Butler RC, Partridge TS, et al: Current progress in the study of allergic polymyositis in the guinea pig and man. In Rose FC (ed): Clinical Neuroimmunology. Oxford, Blackwell Scientific Publications, 1979, p 146. 136. Grimley PM, Friedman RM: Arboviral infection of voluntary striated muscles. J Infect Dis 122: 45–52, 1970. 137. Seay AR, Griffin DE, Johnson RT: Experimental viral polymyositis: age dependency and immune responses to Ross River virus infection in mice. Neurology 31: 656–660, 1981. 138. Melnick JL, Godman GC: Pathogenesis of coxsackie virus infections: multiplication of virus and evolution of the muscle lesion in mice. J Exp Med 93: 247–266, 1951. 139. Ytterberg SR, Mahowald ML, Messner RP: Coxsackievirus B 1-induced polymyositis. Lack of disease expression in nu/nu mice. J Clin Invest 80: 499–506, 1987. 140. Ytterberg SR, Mahowald ML, Messner RP: T cells are required for coxsackievirus B1 induced murine polymyositis. J Rheumatol 15: 475–478, 1988. 141. Craighead JE: Pathogenicity of the M and E variants of the encephalomyocarditis (EMC) virus. I. Myocardiotropic and neurotropic properties. Am J Pathol 48: 333–345, 1966. 142. Hargis AM, Prieur DJ: Animal models of polymyositis/dermatomyositis. Clin Dermatol 6: 120–124, 1988. 143. Hargis AM, Winkelstein JA, Moore MP, et al: Complement levels in dogs with familial canine dermatomyositis. Vet Immunol Immunopathol 20: 95–100, 1988. 144. Brunsting LA, Christianson HB, Perry HO: Dermatomyositis; unusual features, complications, and treatment. AMA Arch Dermatol 74: 581–589, 1956.
145. Lambie JA, Duff IF: Familial occurrence of dermatomyositis. Ann Intern Med 59: 839, 1963. 146. Lewkonia RM, Buxton PH: Myositis in father and daughter. J Neurol Neurosurg Psychiatry 36: 820–825, 1973. 147. Harati Y, Niakan E, Bergman EW: Childhood dermatomyositis in monozygotic twins. Neurology 36: 721–723, 1986. 148. Rose T, Nothjunge J, Schlote W: Familial occurrence of dermatomyositis and progressive scleroderma after injection of a local anaesthetic for dental treatment. Eur J Pediatr 143: 225–228, 1985. 149. Rider LG, Gurley RC, Pandey JP, et al: Clinical, serologic, and immunogenetic features of familial idiopathic inflammatory myopathy. Arthritis Rheum 41: 710–719, 1998. 150. Andrews A, Hickling P, Hutton C: Familial dermatomyositis. Br J Rheumatol 37: 231–232, 1998. 151. Plamondon S, Dent PB, Reed AM: Familial dermatomyositis. J Rheumatol 26: 2691–2692, 1999. 152. Tsao CY, Mendell JR, Kissel JT: Dermatomyositis in two siblings and a brief review of familial dermatomyositis. J Child Neurol 17: 540–542, 2002. 153. Hirsch TJ, Enlow RW, Bias WB, et al: HLA-D related (DR) antigens in various kinds of myositis. Hum Immunol 3: 181–186, 1981. 154. Friedman JM, Pachman LM, Maryjowski ML, et al: Immunogenetic studies of juvenile dermatomyositis: HLA-DR antigen frequencies. Arthritis Rheum 26: 214–216, 1983. 155. Reed AM, Pachman LM, Hayford J, et al: Immunogenetic studies in families of children with juvenile dermatomyositis. J Rheumatol 25: 1000–1002, 1998. 156. Reed AM, Pachman L, Ober C: Molecular genetic studies of major histocompatibility complex genes in children with juvenile dermatomyositis: increased risk associated with HLA-DQA1 *0501. Hum Immunol 32: 235–240, 1991. 157. Reed AM, Stirling JD: Association of the HLA-DQA1*0501 allele in multiple racial groups with juvenile dermatomyositis. Hum Immunol 44: 131–135, 1995. 158. Vavrincova P, Havelka S, Cerna M, et al: HLA class II alleles in juvenile dermatomyositis. J Rheumatol (Suppl) 37: 17–18, 1993. 159. West JE, Reed AM: Analysis of HLA-DM polymorphism in juvenile dermatomyositis (JDM) patients. Hum Immunol 60: 255–258, 1999. 160. Arnett FC, Targoff IN, Mimori T, et al: Interrelationship of major histocompatibility complex class II alleles and autoantibodies in four ethnic groups with various forms of myositis. Arthritis Rheum 39: 1507–1518, 1996. 161. Shamim EA, Rider LG, Pandey JP, et al: Differences in idiopathic inflammatory myopathy phenotypes and genotypes between Mesoamerican Mestizos and North American Caucasians: ethnogeographic influences in the genetics and clinical expression of myositis. Arthritis Rheum 46: 1885–1893, 2002. 162. Werth VP, Berlin JA, Callen JP, et al: Mannose binding lectin (MBL) polymorphisms associated with low MBL production in patients with dermatomyositis. J Invest Dermatol 119: 1394–1399, 2002. 163. Pachman LM, Liotta-Davis MR, Hong DK, et al: TNFalpha-308A allele in juvenile dermatomyositis: association with increased production of tumor necrosis factor alpha, disease duration, and pathologic calcifications. Arthritis Rheum 43: 2368–2377, 2000. 164. Hoffman EP, Rao D, Pachman LM: Clarifying the boundaries between the inflammatory and dystrophic myopathies: insights from molecular diagnostics and microarrays. Rheum Dis Clin North Am 28: 743–757, 2002. 165. Fedczyna TO, Lutz J, Pachman LM: Expression of TNFalpha by muscle fibers in biopsies from children with untreated juvenile dermatomyositis: association with the TNFalpha-308A allele. Clin Immunol 100: 236–239, 2001. 166. de Carvalho M: Gabapentin for the treatment of spasticity in patients with amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord 2: 47–48, 2001. 167. Kuru S, Inukai A, Kato T, et al: Expression of tumor necrosis factor-alpha in regenerating muscle fibers in inflammatory and non-inflammatory myopathies. Acta Neuropathol (Berl) 105: 217–224, 2003. 168. Hill RH, Wood WS: Juvenile dermatomyositis. Can Med Assoc J 103: 1152–1156, 1970. 169. Schaller JG: Dermatomyositis. J Pediatr 83: 699–702, 1973. 170. Kornreich HK, Hanson V: The rheumatic diseases of childhood. Curr Probl Pediatr 4: 3–40, 1974. 171. Goel KM, Shanks RA: Dermatomyositis in childhood. Review of eight cases. Arch Dis Child 51: 501–506, 1976. 172. Dubowitz V: Muscle disorders in childhood. Major Probl Clin Pediatr 16: iii–282, 1978. 173. Winkelman RK: Dermatomyositis in childhood. J Dermatol 18: 13, 1979. 174. Malleson PN: Controversies in juvenile dermatomyositis. J Rheumatol (Suppl) 23: 1–2, 1990. 175. Pachman LM: Juvenile dermatomyositis. Pathophysiology and disease expression. Pediatr Clin North Am 42: 1071–1098, 1995. 176. Goel KM, King M: Dermatomyositis-polymyositis in children. Scott Med J 31: 15–19, 1986. 177. Rider L, Sonies B, Sapper D, et al: Swallowing and oral motor abnormalities are frequent in juvenile idiopathic inflammatory myopathy. Arthritis Rheum 39(Suppl): S191, 1996. 178. Resnick JS, Mammel M, Mundale MO, et al: Muscular strength as an index of response to therapy in childhood dermatomyositis. Arch Phys Med Rehabil 62: 12–19, 1981. 179. National Foundation: National Foundation Publication No. 60, National Foundation, 1946.
C H A P T E R 180. Huber AM, Hicks JE, Lachenbruch PA, et al: Validation of the Childhood Health Assessment Questionnaire in the juvenile idiopathic myopathies. Juvenile Dermatomyositis Disease Activity Collaborative Study Group. J Rheumatol 28: 1106–1111, 2001. 181. van der NJ, Kamphuis SS, Helders PJ: The Childhood Myositis Assessment Scale to assess muscle function in a patient with juvenile dermatomyositis. Arthritis Rheum 47: 694–695, 2002. 182. Bode RK, Klein-Gitelman MS, Miller ML, et al: Disease activity score for children with juvenile dermatomyositis: reliability and validity evidence. Arthritis Rheum 49: 7–15, 2003. 183. Huber AM, Feldman BM, Rennebohm RM, et al: Validation and clinical significance of the Childhood Myositis Assessment Scale for assessment of muscle function in the juvenile idiopathic inflammatory myopathies. Arthritis Rheum 50: 1595–1603, 2004. 184. Scheja A, Elborgh R, Wildt M: Decreased capillary density in juvenile dermatomyositis and in mixed connective tissue disease. J Rheumatol 26: 1377–1381, 1999. 185. Tse S, Lubelsky S, Gordon M, et al: The arthritis of inflammatory childhood myositis syndromes. J Rheumatol 28: 192–197, 2001. 186. Dolezalova P, Young SP, Bacon PA, et al: Nailfold capillary microscopy in healthy children and in childhood rheumatic diseases: a prospective single blind observational study. Ann Rheum Dis 62: 444–449, 2003. 187. Maricq HR, Spencer-Green G, LeRoy EC: Skin capillary abnormalities as indicators of organ involvement in scleroderma (systemic sclerosis), Raynaud’s syndrome and dermatomyositis. Am J Med 61: 862–870, 1976. 188. Spencer-Green G, Schlesinger M, Bove KE, et al: Nailfold capillary abnormalities in childhood rheumatic diseases. J Pediatr 102: 341–346, 1983. 189. Nussbaum AI, Silver RM, Maricq HR: Serial changes in nailfold capillary morphology in childhood dermatomyositis. Arthritis Rheum 26: 1169–1172, 1983. 190. Silver RM, Maricq HR: Childhood dermatomyositis: serial microvascular studies. Pediatrics 83: 278–283, 1989. 191. Ghali FE, Reed AM, Groben PA, et al: Panniculitis in juvenile dermatomyositis. Pediatr Dermatol 16: 270–272, 1999. 192. Woo TR, Rasmussen J, Callen JP: Recurrent photosensitive dermatitis preceding juvenile dermatomyositis. Pediatr Dermatol 2: 207–212, 1985. 193. Hamlin C, Shelton JE: Management of oral findings in a child with an advanced case of dermatomyositis: clinical report. Pediatr Dent 6: 46–49, 1984. 194. Ghali FE, Stein LD, Fine JD, et al: Gingival telangiectases: an underappreciated physical sign of juvenile dermatomyositis. Arch Dermatol 135: 1370–1374, 1999. 195. Bowyer SL, Blane CE, Sullivan DB, et al: Childhood dermatomyositis: factors predicting functional outcome and development of dystrophic calcification. J Pediatr 103: 882–888, 1983. 196. Spencer CH, Hanson V, Singsen BH, et al: Course of treated juvenile dermatomyositis. J Pediatr 105: 399–408, 1984. 197. Piantanida NA, Person DA, Piantanida EW: Infrapatellar hypertrichosis: an unusual cutaneous manifestation of juvenile dermatomyositis. Pediatr Dermatol 19: 132–135, 2002. 198. Euwer RL, Sontheimer RD: Amyopathic dermatomyositis (dermatomyositis sine myositis). Presentation of six new cases and review of the literature. J Am Acad Dermatol 24: 959–966, 1991. 199. Cosnes A, Amaudric F, Gherardi R, et al: Dermatomyositis without muscle weakness. Long-term follow-up of 12 patients without systemic corticosteroids. Arch Dermatol 131: 1381–1385, 1995. 200. Schmid MH, Trueb RM: Juvenile amyopathic dermatomyositis. Br J Dermatol 136: 431–433, 1997. 201. Eisenstein DM, Paller AS, Pachman LM: Juvenile dermatomyositis presenting with rash alone; late onset of myositis. Arthritis Rheum 39 (Suppl): 192, 1996. 202. Olsen NJ, Park JH, King LE Jr: Amyopathic dermatomyositis. Curr Rheumatol Rep 3: 346–351, 2001. 203. Eisenstein DM, Paller AS, Pachman LM: Juvenile dermatomyositis presenting with rash alone. Pediatrics 100: 391–392, 1997. 204. Plamondon S, Dent PB: Juvenile amyopathic dermatomyositis: results of a case finding descriptive survey. J Rheumatol 27: 2031–2034, 2000. 205. Wananukul S, Pongprasit P, Wattanakrai P: Calcinosis cutis presenting years before other clinical manifestations of juvenile dermatomyositis: report of two cases. Australas J Dermatol 38: 202–205, 1997. 206. Blane CE, White SJ, Braunstein EM, et al: Patterns of calcification in childhood dermatomyositis. Am J Roentgenol 142: 397–400, 1984. 207. Mukamel M, Horev G, Mimouni M: New insight into calcinosis of juvenile dermatomyositis: a study of composition and treatment. J Pediatr 138: 763–766, 2001. 208. Fisler RE, Liang MG, Fuhlbrigge RC, et al: Aggressive management of juvenile dermatomyositis results in improved outcome and decreased incidence of calcinosis. J Am Acad Dermatol 47: 505–511, 2002. 209. Banker BQ, Victor M: Dermatomyositis (systemic angiopathy) of childhood. Medicine (Baltimore) 45: 261–289, 1966. 210. Braunstein EM, White SJ: Pneumatosis intestinalis in dermatomyositis. Br J Radiol 53: 1011–1012, 1980. 211. Thompson JW: Spontaneous perforation of the esophagus as a manifestation of dermatomyositis. Ann Otol Rhinol Laryngol 93: 464–467, 1984. 212. Magill HL, Hixson SD, Whitington G, et al: Duodenal perforation in childhood dermatomyositis. Pediatr Radiol 14: 28–30, 1984. 213. Schullinger JN, Jacobs JC, Berdon WE: Diagnosis and management of gastrointestinal perforations in childhood dermatomyositis with particular reference to perforations of the duodenum. J Pediatr Surg 20: 521–524, 1985.
18 J U V E N I L E D E R M AT O M Y O S I T I S
437
214. Inamdar S, Slim MS, Bostwick H, et al: Treatment of duodenocutaneous fistula with somatostatin analog in a child with dermatomyositis. J Pediatr Gastroenterol Nutr 10: 402–404, 1990. 215. Jalleh RP, Swift RI, Sundaresan M, et al: Necrotising testicular vasculitis associated with dermatomyositis. Br J Urol 66: 660, 1990. 216. Ramanan AV, Sawhney S, Murray KJ: Central nervous system complications in two cases of juvenile onset dermatomyositis. Rheumatology (Oxf) 40: 1293–1298, 2001. 217. Buchpiguel CA, Roizenblatt S, Lucena-Fernandes MF, et al: Radioisotopic assessment of peripheral and cardiac muscle involvement and dysfunction in polymyositis/dermatomyositis. J Rheumatol 18: 1359–1363, 1991. 218. Trapani S, Camiciottoli G, Vierucci A, et al: Pulmonary involvement in juvenile dermatomyositis: a two-year longitudinal study. Rheumatology (Oxf) 40:216–220, 2001. 219. Schwarz MI, Matthay RA, Sahn SA, et al: Interstitial lung disease in polymyositis and dermatomyositis: analysis of six cases and review of the literature. Medicine (Baltimore) 55: 89–104, 1976. 220. Dickey BF, Myers AR: Pulmonary disease in polymyositis/dermatomyositis. Semin Arthritis Rheum 14: 60–76, 1984. 221. Takken T, Spermon N, Helders PJ, et al: Aerobic exercise capacity in patients with juvenile dermatomyositis. J Rheumatol 30: 1075–1080, 2003. 222. Hicks JE, Drinkard B, Summers RM, et al: Decreased aerobic capacity in children with juvenile dermatomyositis. Arthritis Rheum 47: 118–123, 2002. 223. Takken T, Elst E, Spermon N, et al: The physiological and physical determinants of functional ability measures in children with juvenile dermatomyositis. Rheumatology (Oxf) 42: 591–595, 2003. 224. Kobayashi I, Ono S, Kawamura N, et al: KL-6 is a potential marker for interstitial lung disease associated with juvenile dermatomyositis. J Pediatr 138: 274–276, 2001. 225. Garg A: Acquired and inherited lipodystrophies. N Engl J Med 350: 1220–1234, 2004. 226. Commens C, O’Neill P, Walker G: Dermatomyositis associated with multifocal lipoatrophy. J Am Acad Dermatol 22: 966–969, 1990. 227. Huang JL: Juvenile dermatomyositis associated with partial lipodystrophy. Br J Clin Pract 50: 112–113, 1996. 228. Quecedo E, Febrer I, Serrano G, et al: Partial lipodystrophy associated with juvenile dermatomyositis: report of two cases. Pediatr Dermatol 13: 477–482, 1996. 229. Kavanagh GM, Colaco CB, Kennedy CT: Juvenile dermatomyositis associated with partial lipoatrophy. J Am Acad Dermatol 28: 348–351, 1993. 230. Huemer C, Kitson H, Malleson PN, et al: Lipodystrophy in patients with juvenile dermatomyositis—evaluation of clinical and metabolic abnormalities. J Rheumatol 28: 610–615, 2001. 231. Oral EA, Simha V, Ruiz E, et al: Leptin-replacement therapy for lipodystrophy. N Engl J Med 346: 570–578, 2002. 232. Misra A, Peethambaram A, Garg A: Clinical features and metabolic and autoimmune derangements in acquired partial lipodystrophy: report of 35 cases and review of the literature. Medicine (Baltimore) 83: 18–34, 2004. 233. Bennington JL, Dau PC: Patients with polymyositis and dermatomyositis who undergo plasmapheresis therapy. Pathologic findings. Arch Neurol 38: 553–560, 1981. 234. Walton J: The inflammatory myopathies. J R Soc Med 76: 998–1010, 1983. 235. Carpenter S, Karpati G, Rothman S, et al: The childhood type of dermatomyositis. Neurology 26: 952–962, 1976. 236. Oxenhandler R, Adelstein EH, Hart MN: Immunopathology of skeletal muscle. The value of direct immunofluorescence in the diagnosis of connective tissue disease. Hum Pathol 8: 321–328, 1977. 237. Oberc MA, Engel WK: Ultrastructural localization of calcium in normal and abnormal skeletal muscle. Lab Invest 36: 566–577, 1977. 238. Bodensteiner JB, Engel AG: Intracellular calcium accumulation in Duchenne dystrophy and other myopathies: a study of 567,000 muscle fibers in 114 biopsies. Neurology 28: 439–446, 1978. 239. Mastaglia FL, Kakulas BA: A histological and histochemical study of skeletal muscle regeneration in polymyositis. J Neurol Sci 10: 471–487, 1970. 240. Cros D, Pearson C, Verity MA: Polymyositis-dermatomyositis: diagnostic and prognostic significance of muscle alkaline phosphatase. Am J Pathol 101: 159–176, 1980. 241. Niermann KJ, Olsen NJ, Park JH: Magnesium abnormalities of skeletal muscle in dermatomyositis and juvenile dermatomyositis. Arthritis Rheum 46: 475–488, 2002. 242. Banker BQ: Dermatomyositis of childhood, ultrastructural alterations of muscle and intramuscular blood vessels. J Neuropathol Exp Neurol 34: 46–75, 1975. 243. Eady RA, Odland GF: Intraendothelial tubular aggregates in experimental wounds. Br J Dermatol 93: 165–173, 1975. 244. Yunis EJ, Samaha FJ: Inclusion body myositis. Lab Invest 25: 240–248, 1971. 245. Mastaglia FL, Walton JN: An ultrastructural study of skeletal muscle in polymyositis. J Neurol Sci 12: 473–504, 1971. 246. Oshima Y, Becker LE, Armstrong DL: An electron microscopic study of childhood dermatomyositis. Acta Neuropathol (Berl) 47: 189–196, 1979. 247. Chou SM, Nonaka I, Voice GF: Anastomoses of transverse tubules with terminal cisternae in polymyositis. Arch Neurol 37: 257–266, 1980. 248. Jerusalem F, Rakusa M, Engel AG, et al: Morphometric analysis of skeletal muscle capillary ultrastructure in inflammatory myopathies. J Neurol Sci 23: 391–402, 1974.
438
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
249. Bowyer SL, Clark RA, Ragsdale CG, et al: Juvenile dermatomyositis: histological findings and pathogenetic hypothesis for the associated skin changes. J Rheumatol 13: 753–759, 1986. 250. Janis JF, Winkelmann RK: Histopathology of the skin in dermatomyositis. A histopathologic study of 55 cases. Arch Dermatol 97: 640–650, 1968. 251. Hanno R, Callen JP: Histopathology of Gottron’s papules. J Cutan Pathol 12: 389–394, 1985. 252. Sewell JR, Liyanage B, Ansell BM: Calcinosis in Juvenile dermatomyositis. Skeletal Radiol 3: 137, 1978. 253. Fischer TJ, Cipel L, Stiehm ER: Pneumatosis intestinalis associated with fatal childhood dermatomyositis. Pediatrics 61: 127–130, 1978. 254. Denbow CE, Lie JT, Tancredi RG, et al: Cardiac involvement in polymyositis: a clinicopathologic study of 20 autopsied patients. Arthritis Rheum 22: 1088–1092, 1979. 255. Haupt HM, Hutchins GM: The heart and cardiac conduction system in polymyositis-dermatomyositis: a clinicopathologic study of 16 autopsied patients. Am J Cardiol 50: 998–1006, 1982. 256. Greenberg SA, Sanoudou D, Haslett JN, et al: Molecular profiles of inflammatory myopathies. Neurology 59: 1170–1182, 2002. 257. Thompson CE: Infantile myositis. Dev Med Child Neurol 24: 307–313, 1982. 258. Roddy SM, Ashwal S, Peckham N, et al: Infantile myositis: a case diagnosed in the neonatal period. Pediatr Neurol 2: 241–244, 1986. 259. Pollock JL: Toxoplasmosis appearing to be dermatomyositis. Arch Dermatol 115: 736–737, 1979. 260. Topi GC, D’Alessandro L, Catricala C, et al: Dermatomyositis-like syndrome due to toxoplasma. Br J Dermatol 101: 589–591, 1979. 261. Noseworthy JH, Heffernan LP, Ross JB, et al: Acne fulminans with inflammatory myopathy. Ann Neurol 8: 67–69, 1980. 262. Fiallo P, Tagliapietra AG: Severe acute myopathy induced by isotretinoin. Arch Dermatol 132: 1521–1522, 1996. 263. Ashwal S, Peckham N: Myoadenylate deaminase deficiency in children. Pediatr Neurol 1: 185–191, 1985. 264. Gross M: Clinical heterogeneity and molecular mechanisms in inborn muscle AMP deaminase deficiency. J Inherit Metab Dis 20: 186–192, 1997. 265. Medsger TA Jr, Rodnan GP, Moossy J, et al: Skeletal muscle involvement in progressive systemic sclerosis (scleroderma). Arthritis Rheum 11: 554–568, 1968. 266. Brooke MH, Kaplan H: Muscle pathology in rheumatoid arthritis, polymyalgia rheumatica, and polymyositis: a histochemical study. Arch Pathol 94: 101–118, 1972. 267. Denko CW, Old JW: Myopathy in the Sicca syndrome (Sjögren’s syndrome). Am J Clin Pathol 51: 631–637, 1969. 268. Gotoff SP, Smith RD, Sugar O: Dermatomyositis with cerebral vasculitis in a patient with agammaglobulinemia. Am J Dis Child 123: 53–56, 1972. 269. Giuliano VJ: Case report: Polymyositis in a patient with acquired hypogammaglobulinemia. Am J Med Sci 268: 53–56, 1974. 270. Bardelas JA, Winkelstein JA, Seto DS, et al: Fatal ECHO 24 infection in a patient with hypogammaglobulinemia: relationship to dermatomyositis-like syndrome. J Pediatr 90: 396–399, 1977. 271. Anderson TE, Carr AJ, Chapman RS, et al: Myositis and myotonia in a case of multicentric reticulohistiocytosis. Br J Dermatol 80: 39–45, 1968. 272. Burke JS, Medline NM, Katz A: Giant cell myocarditis and myositis. Associated with thymoma and myasthenia gravis. Arch Pathol 88: 359–366, 1969. 273. Silverstein A, Siltzbach LE: Muscle involvement in sarcoidosis. Asymptomatic, myositis, and myopathy. Arch Neurol 21: 235–241, 1969. 274. Wortmann RL: Diseases of Skeletal Muscle. Philadelphia, Lippincott Williams & Wilkins, 2000. 275. Carpenter S, Karpati G, Heller I, et al: Inclusion body myositis: a distinct variety of idiopathic inflammatory myopathy. Neurology 28: 8–17, 1978. 276. Griggs RC, Askanas V, DiMauro S, et al: Inclusion body myositis and myopathies. Ann Neurol 38: 705–713, 1995. 277. Illa I, Dalakas MC: Dermatomyositis, polymyositis and inclusion body myositis: current concepts. Rev Neurol (Paris) 154: 13–16, 1998. 278. Layzer RB, Shearn MA, Satya-Murti S: Eosinophilic polymyositis. Ann Neurol 1: 65–71, 1977. 279. Cumming WJ, Weiser R, Teoh R, et al: Localised nodular myositis: a clinical and pathological variant of polymyositis. Q J Med 46: 531–546, 1977. 280. Heffner RR Jr, Barron SA: Polymyositis beginning as a focal process. Arch Neurol 38: 439–442, 1981. 281. Enzinger FM, Dulcey F: Proliferative myositis. Report of thirty-three cases. Cancer 20: 2213–2223, 1967. 282. Savage DC, Forbes M, Pearce GW: Idiopathic rhabdomyolysis. Arch Dis Child 46: 594–607, 1971. 283. Smith R, Russell RG, Woods CG: Myositis ossificans progressiva. Clinical features of eight patients and their response to treatment. J Bone Joint Surg Br 58: 48–57, 1976. 284. Hentzer B, Jacobsen HH, Asboe-Hansen G: Fibrodysplasia ossificans progressiva. Scand J Rheumatol 6: 161–171, 1977. 285. Wu T, Chen SS: Differential diagnosis between fibrodysplasia ossificans progressiva and childhood dermatomyositis with calcinosis. J Formos Med Assoc 92: 569–576, 1993. 286. Smith R, Athanasou NA, Vipond SE: Fibrodysplasia (myositis) ossificans progressiva: clinicopathological features and natural history. Q J Med 89: 445–446, 1996. 287. Blaszczyk M, Majewski S, Brzezinska-Wcislo L, et al: Fibrodysplasia ossificans progressiva. Eur J Dermatol 13: 234–237, 2003.
288. de Silva MV, Reid R: Myositis ossificans and fibroosseous pseudotumor of digits: a clinicopathological review of 64 cases with emphasis on diagnostic pitfalls. Int J Surg Pathol 11: 187–195, 2003. 289. Paim LB, Liphaus BL, Almeida da Silva CA: Fibrodysplasia ossificans progressiva (FOP). Indian Pediatr 40: 786–788, 2003. 290. Feldman G, Li M, Martin S, et al: Fibrodysplasia ossificans progressiva, a heritable disorder of severe heterotopic ossification, maps to human chromosome 4q27-31. Am J Hum Genet 66: 128–135, 2000. 291. Okayama A, Futani H, Kyo F, et al: Usefulness of ultrasonography for early recurrent myositis ossificans. J Orthop Sci 8: 239–242, 2003. 292. Hagiwara H, Aida N, Machida J, et al: Contrast-enhanced MRI of an early preosseous lesion of fibrodysplasia ossificans progressiva in a 21-month-old boy. Am J Roentgenol 181: 1145–1147, 2003. 293. Strzepek J, Gadek A: Diagnostic problems in osteosarcoma first diagnosed as myositis ossificans. Przegl Lek 59: 1044–1047, 2002. 294. Bernard M, Coumbaras M, Zeitoun F, et al: Radiological spectrum of myositis ossificans circumscripta evolution. J Radiol 84: 54–56, 2003. 295. Allgrove J: Use of bisphosphonates in children and adolescents. J Pediatr Endocrinol Metab 15 (Suppl 3): 921–928, 2002. 296. Olmsted EA, Kaplan FS, Shore EM: Bone morphogenetic protein-4 regulation in fibrodysplasia ossificans progressiva. Clin Orthop 408: 331–343, 2003. 297. Hegyi L, Gannon FH, Glaser DL, et al: Stromal cells of fibrodysplasia ossificans progressiva lesions express smooth muscle lineage markers and the osteogenic transcription factor Runx2/Cbfa-1: clues to a vascular origin of heterotopic ossification? J Pathol 201: 141–148, 2003. 298. International Fibrodysplasia Ossificans Progressiva Association (IFOPA): Available at: www.ifopa.org (accessed 2004). 299. Vignos PJ Jr, Goldwyn J: Evaluation of laboratory tests in diagnosis and management of polymyositis. Am J Med Sci 263: 291–308, 1972. 300. Haas RH, Dyck RF, Dubowitz V, et al: C-reactive protein in childhood dermatomyositis. Ann Rheum Dis 41: 483–485, 1982. 301. Viguier M, Fouere S, de la SP, et al: Peripheral blood lymphocyte subset counts in patients with dermatomyositis: clinical correlations and changes following therapy. Medicine (Baltimore) 82: 82–86, 2003. 302. Bowyer SL, Ragsdale CG, Sullivan DB: Factor VIII related antigen and childhood rheumatic diseases. J Rheumatol 16: 1093–1097, 1989. 303. Bloom BJ, Tucker LB, Miller LC, et al: Von Willebrand factor in juvenile dermatomyositis. J Rheumatol 22: 320–325, 1995. 304. Kobayashi S, Higuchi K, Tamaki H, et al: Characteristics of juvenile dermatomyositis in Japan. Acta Paediatr Jpn 39: 257–262, 1997. 305. De Benedetti F, De Amici M, Aramini L, et al: Correlation of serum neopterin concentrations with disease activity in juvenile dermatomyositis. Arch Dis Child 69: 232–235, 1993. 306. Lisak RP, Zweiman B: Serum immunoglobulin levels in myasthenia gravis, polymyositis, and dermatomyositis. J Neurol Neurosurg Psychiatry 39: 34–37, 1976. 307. Ishida T, Ohashi M, Matsumoto Y, et al: Connection of atopic disease in Japanese patients with juvenile dermatomyositis based on serum IgE levels. Clin Rheumatol 12: 41–48, 1993. 308. Rider LG, Schiffenbauer AS, Zito M, et al: Neopterin and quinolinic acid are surrogate measures of disease activity in the juvenile idiopathic inflammatory myopathies. Clin Chem 48: 1681–1688, 2002. 309. Askmark H, Osterman PO, Roxin LE, et al: Radioimmunoassay of serum myoglobin in neuromuscular diseases. J Neurol Neurosurg Psychiatry 44: 68–72, 1981. 310. Nishikai M, Reichlin M: Radioimmunoassay of serum myoglobin in polymyositis and other conditions. Arthritis Rheum 20: 1514–1518, 1977. 311. Nishikai M, Homma M: Circulating autoantibody against human myoglobin in polymyositis. JAMA 237: 1842–1844, 1977. 312. Dickerson JW, Widdowson EM: Chemical changes in skeletal muscle during development. Biochem J 74: 247, 1960. 313. Hallgren R, Karlsson FA, Roxin LE, et al: Myoglobin turnover—influence of renal and extrarenal factors. J Lab Clin Med 91: 246–254, 1978. 314. Clark LC Jr, Thompson HL, Beck EL, et al: Excretion of creatine and creatinine by children. Am J Dis Child 81: 774, 1951. 315. Ansell BM, Hamilton E, Bywaters EG: Course and prognosis in juvenile dermatomyositis. In Kakulas BA (ed): Second International Congress on Muscle Disease, Part 2. Publication 295. Amsterdam, Excerpta Medica, 1973. 316. Montecucco C, Ravelli A, Caporali R, et al: Autoantibodies in juvenile dermatomyositis. Clin Exp Rheumatol 8: 193–196, 1990. 317. Pachman LM, Maryjowski MC: Juvenile dermatomyositis and polymyositis. Clin Rheum Dis 10: 95–115, 1984. 318. Pachman LM: Juvenile dermatomyositis. Pediatr Clin North Am 33: 1097–1117, 1986. 319. Feldman BM, Reichlin M, Laxer RM, et al: Clinical significance of specific autoantibodies in juvenile dermatomyositis. J Rheumatol 23: 1794–1797, 1996. 320. Rider LG, Miller FW, Targoff IN, et al: A broadened spectrum of juvenile myositis. Myositis-specific autoantibodies in children. Arthritis Rheum 37: 1534–1538, 1994. 321. Cambridge G, Ovadia E, Isenberg DA, et al: Juvenile dermatomyositis: serial studies of circulating autoantibodies to a 56 kD nuclear protein. Clin Exp Rheumatol 12: 451–457, 1994. 322. Tanimoto K, Nakano K, Kano S, et al: Classification criteria for polymyositis and dermatomyositis [published erratum appears in J Rheumatol 22: 1807, 1995]. J Rheumatol 22: 668–674, 1995.
C H A P T E R 323. Friedman AW, Targoff IN, Arnett FC: Interstitial lung disease with autoantibodies against aminoacyl-tRNA synthetases in the absence of clinically apparent myositis. Semin Arthritis Rheum 26: 459–467, 1996. 324. Garcia-Patos V, Bartralot R, Fonollosa V, et al: Childhood sclerodermatomyositis: report of a case with the anti-PM/Scl antibody and mechanic’s hands. Br J Dermatol 135: 613–616, 1996. 325. Targoff IN, Miller FW, Medsger TA Jr, et al: Classification criteria for the idiopathic inflammatory myopathies. Curr Opin Rheumatol 9: 527–535, 1997. 326. Reichlin M, Mattioli M: Description of a serological reaction characteristic of polymyositis. Clin Immunol Immunopathol 5: 12–20, 1976. 327. Nishikai M, Reichlin M: Purification and characterization of a nuclear nonhistone basic protein (Mi-1) which reacts with anti-immunoglobulin sera and the sera of patients with dermatomyositis. Mol Immunol 17: 1129–1141, 1980. 328. Nishikai M, Reichlin M: Heterogeneity of precipitating antibodies in polymyositis and dermatomyositis. Characterization of the Jo-1 antibody system. Arthritis Rheum 23: 881–888, 1980. 329. Kowalska-Oledzka E, Stern LZ: Myositis specific antibodies: frequency in different populations. Folia Neuropathol 38: 101–103, 2000. 330. Targoff IN, Johnson AE, Miller FW: Antibody to signal recognition particle in polymyositis. Arthritis Rheum 33: 1361–1370, 1990. 331. Wolfe JF, Adelstein E, Sharp GC: Antinuclear antibody with distinct specificity for polymyositis. J Clin Invest 59: 176–178, 1977. 332. Pachman LM, Friedman JM, Maryjowski-Sweeney ML, et al: Immunogenetic studies of juvenile dermatomyositis. III. Study of antibody to organ-specific and nuclear antigens. Arthritis Rheum 28: 151–157, 1985. 333. Kubo M, Ihn H, Kuwana M, et al: Prevalence in myositis of antibodies recognizing anti-U3 RNA probably in a novel complex with 22/25 kD protein and not fibrillarin. Clin Exp Immunol 126: 339–344, 2001. 334. Mimori T, Akizuki M, Yamagata H, et al: Characterization of a high molecular weight acidic nuclear protein recognized by autoantibodies in sera from patients with polymyositis-scleroderma overlap. J Clin Invest 68: 611–620, 1981. 335. Cervera R, Ramirez G, Fernandez-Sola J, et al: Antibodies to endothelial cells in dermatomyositis: association with interstitial lung disease. BMJ 302: 880–881, 1991. 336. Edelsten C, D’Cruz D, Hughes GR, et al: Anti-endothelial cell antibodies in retinal vasculitis. Curr Eye Res 11 (Suppl): 203–208, 1992. 337. Caspary EA, Gubbay SS, Stern GM: Circulating antibodies in polymyositis and other muscle-wasting disorders. Lancet 2: 941, 1964. 338. Stern GM, Rose AL, Jacobs K: Circulating antibodies in polymyositis. J Neurol Sci 5: 181–183, 1967. 339. Behan WM, Behan PO: Complement abnormalities in polymyositis. J Neurol Sci 34: 241–246, 1977. 340. Pachman LM, Hayford JR, Chung A, et al: Juvenile dermatomyositis at diagnosis: clinical characteristics of 79 children. J Rheumatol 25: 1198–1204, 1998. 341. Tucker LB, Sadegi-Neged A, Schaller JG: The association of acquired lipodystrophy with juvenile dermatomyositis. Arthritis Rheum 33(Suppl): S146, 1990. 342. Sandstedt PE, Henriksson KG, Larrsson LE: Quantitative electromyography in polymyositis and dermatomyositis. Acta Neurol Scand 65: 110–121, 1982. 343. Haridasan G, Sanghvi SH, Jindal GD, et al: Quantitative electromyography using automatic analysis. A comparative study with a fixed fraction of a subject’s maximum effort and two levels of thresholds for analysis. J Neurol Sci 42: 53–64, 1979. 344. Partanen J, Lang H: EMG dynamics in polymyositis. A quantitative single motor unit potential study. J Neurol Sci 57: 221–234, 1982. 345. Smyth DP: Quantitative electromyography in babies and young children with primary muscle disease and neurogenic lesions. J Neurol Sci 56: 199–207, 1982. 346. Calore EE, Cavaliere MJ, Perez NM: Muscle pathology in juvenile dermatomyositis. Sao Paulo Med J 115: 1555–1559, 1997. 347. Tome FM, Fardeau M, Lebon P, et al: Inclusion body myositis. Acta Neuropathol (Suppl) (Berl) 7: 287–291, 1981. 348. Dorph C, Nennesmo I, Lundberg IE: Percutaneous conchotome muscle biopsy. A useful diagnostic and assessment tool. J Rheumatol 28: 1591–1599, 2001. 349. Campellone JV, Lacomis D, Giuliani MJ, et al: Percutaneous needle muscle biopsy in the evaluation of patients with suspected inflammatory myopathy. Arthritis Rheum 40: 1886–1891, 1997. 350. Stonecipher MR, Jorizzo JL, Monu J, et al: Dermatomyositis with normal muscle enzyme concentrations. A single-blind study of the diagnostic value of magnetic resonance imaging and ultrasound. Arch Dermatol 130: 1294–1299, 1994. 351. Smith WP, Robinson RG, Gobuty AH: Positive whole-body 67Ga scintigraphy in dermatomyositis. Am J Roentgenol 133: 126–127, 1979. 352. Guillet GY, Guillet JA, Blanquet P, et al: A new noninvasive evaluation of muscular lesions in dermatomyositis: thallium 201 muscle scans. J Am Acad Dermatol 5: 670–672, 1981. 353. Guillet J, Blanquet P, Guillet G, et al: The use of technetium Tc 99m medronate scintigraphy as a prognostic guide in childhood dermatomyositis. Arch Dermatol 117: 451, 1981. 354. Yonker RA, Webster EM, Edwards NL, et al: Technetium pyrophosphate muscle scans in inflammatory muscle disease. Br J Rheumatol 26: 267–269, 1987.
18 J U V E N I L E D E R M AT O M Y O S I T I S
439
355. Kaufman LD, Gruber BL, Gerstman DP, et al: Preliminary observations on the role of magnetic resonance imaging for polymyositis and dermatomyositis. Ann Rheum Dis 46: 569–572, 1987. 356. Hernandez RJ, Keim DR, Sullivan DB, et al: Magnetic resonance imaging appearance of the muscles in childhood dermatomyositis. J Pediatr 117: 546–550, 1990. 357. Keim DR, Hernandez RJ, Sullivan DB: Serial magnetic resonance imaging in juvenile dermatomyositis. Arthritis Rheum 34: 1580–1584, 1991. 358. Hernandez RJ, Keim DR, Chenevert TL, et al: Fat-suppressed MR imaging of myositis. Radiology 182: 217–219, 1992. 359. Hilario MO, Yamashita H, Lutti D, et al: Juvenile idiopathic inflammatory myopathies: the value of magnetic resonance imaging in the detection of muscle involvement. Sao Paulo Med J 118: 35–40, 2000. 360. Chan WP, Liu GC: MR imaging of primary skeletal muscle diseases in children. AJR Am J Roentgenol 179: 989–997, 2002. 361. Park JH, Vital TL, Ryder NM, et al: Magnetic resonance imaging and P-31 magnetic resonance spectroscopy provide unique quantitative data useful in the longitudinal management of patients with dermatomyositis. Arthritis Rheum 37: 736–746, 1994. 362. Park JH, Olsen NJ, King L Jr, et al: Use of magnetic resonance imaging and P-31 magnetic resonance spectroscopy to detect and quantify muscle dysfunction in the amyopathic and myopathic variants of dermatomyositis. Arthritis Rheum 38: 68–77, 1995. 363. Summers RM, Brune AM, Choyke PL, et al: Juvenile idiopathic inflammatory myopathy: exercise-induced changes in muscle at short inversion time inversion-recovery MR imaging. Radiology 209: 191–196, 1998. 364. Kimball AB, Summers RM, Turner M, et al: Magnetic resonance imaging detection of occult skin and subcutaneous abnormalities in juvenile dermatomyositis. Implications for diagnosis and therapy. Arthritis Rheum 43: 1866–1873, 2000. 365. Hernandez RJ, Sullivan DB, Chenevert TL, et al: MR imaging in children with dermatomyositis: musculoskeletal findings and correlation with clinical and laboratory findings. Am J Roentgenol 161: 359–366, 1993. 366. Huppertz HI, Kaiser WA: Serial magnetic resonance imaging in juvenile dermatomyositis—delayed normalization. Rheumatol Int 14: 127–129, 1994. 367. Park JH, Niermann KJ, Ryder NM, et al: Muscle abnormalities in juvenile dermatomyositis patients: P-31 magnetic resonance spectroscopy studies. Arthritis Rheum 43: 2359–2367, 2000. 368. Perez MD, Abrams SA, Koenning G, et al: Mineral metabolism in children with dermatomyositis. J Rheumatol 21: 2364–2369, 1994. 369. Stewart WA, Acott PD, Salisbury SR, et al: Bone mineral density in juvenile dermatomyositis: assessment using dual x-ray absorptiometry. Arthritis Rheum 48: 2294–2298, 2003. 370. Ellis KJ, Shypailo RJ, Hardin DS, et al: Z score prediction model for assessment of bone mineral content in pediatric diseases. J Bone Miner Res 16: 1658–1664, 2001. 371. Falcini F, Bindi G, Ermini M, et al: Comparison of quantitative calcaneal ultrasound and dual energy X-ray absorptiometry in the evaluation of osteoporotic risk in children with chronic rheumatic diseases. Calcif Tissue Int 67: 19–23, 2000. 372. Dubowitz V: Treatment of dermatomyositis in childhood. Arch Dis Child 51: 494–500, 1976. 373. Winkelmann RK: Dermatomyositis in childhood. Clin Rheum Dis 8: 353–368, 1982. 374. Ansell BM: Management of polymyositis and dermatomyositis. Clin Rheum Dis 10: 205–213, 1984. 375. Cassidy JT: Dermatomyositis in children. In Hicks RV (ed): Vasculopathies of Childhood. Littleton, MA, PSG Publishing, 1988, p 205. 376. Reed AM, Lopez M: Juvenile dermatomyositis: recognition and treatment. Paediatr Drugs 4: 315–321, 2002. 377. Rider LG: Assessment of disease activity and its sequelae in children and adults with myositis. Curr Opin Rheumatol 8: 495–506, 1996. 378. Chapman S, Southwood TR, Fowler J, et al: Rapid changes in magnetic resonance imaging of muscle during the treatment of juvenile dermatomyositis. Br J Rheumatol 33: 184–186, 1994. 379. Rider LG, Feldman BM, Perez MD, et al: Development of validated disease activity and damage indices for the juvenile idiopathic inflammatory myopathies. I. Physician, parent, and patient global assessments. Juvenile Dermatomyositis Disease Activity Collaborative Study Group. Arthritis Rheum 40: 1976–1983, 1997. 380. Feldman BM, Ayling-Campos A, Luy L, et al: Measuring disability in juvenile dermatomyositis: validity of the childhood health assessment questionnaire. J Rheumatol 22: 326–331, 1995. 381. Rider LG: Outcome assessment in the adult and juvenile idiopathic inflammatory myopathies. Rheum Dis Clin North Am 28: 935–977, 2002. 382. Rider LG, Giannini EH, Harris-Love M, et al: Defining clinical improvement in adult and juvenile myositis. J Rheumatol 30: 603–617, 2003. 383. Ruperto N, Ravelli A, Murray KJ, et al: Preliminary core sets of measures for disease activity and damage assessment in juvenile systemic lupus erythematosus and juvenile dermatomyositis. Rheumatology (Oxf) 42: 1452–1459, 2003. 384. Dubowitz LM, Dubowitz V: Acute dermatomyositis presenting with pulmonary manifestations. Arch Dis Child 28: 293, 1964. 385. Uziel Y, Nemet D, Maimon E, et al: Juvenile dermatomyositis: medical and psychosocial team approach. Clin Exp Rheumatol 20: 256, 2002.
440
C H A P T E R
18 J U V E N I L E D E R M AT O M Y O S I T I S
386. Bianchi ML, Cimaz R, Bardare M, et al: Efficacy and safety of alendronate for the treatment of osteoporosis in diffuse connective tissue diseases in children: a prospective multicenter study. Arthritis Rheum 43: 1960–1966, 2000. 387. Miller JJ III: Prolonged use of large intravenous steroid pulses in the rheumatic diseases of children. Pediatrics 65: 989–994, 1980. 388. Yanagisawa T, Sueishi M, Nawata Y, et al: Methylprednisolone pulse therapy in dermatomyositis. Dermatologica 167: 47–51, 1983. 389. Wollina U, Schreiber G: Prednisolone pulse therapy for childhood systemic lupus erythematosus with prominent dermatomyositis. A case report. Dermatologica 171: 45–48, 1985. 390. Laxer RM, Stein LD, Petty RE: Intravenous pulse methylprednisolone treatment of juvenile dermatomyositis. Arthritis Rheum 30: 328–334, 1987. 391. Huang JL: Long-term prognosis of patients with juvenile dermatomyositis initially treated with intravenous methylprednisolone pulse therapy. Clin Exp Rheumatol 17: 621–624, 1999. 392. Al Mayouf S, Al Mazyed A, Bahabri S: Efficacy of early treatment of severe juvenile dermatomyositis with intravenous methylprednisolone and methotrexate. Clin Rheumatol 19: 138–141, 2000. 393. Lang B, Dooley J: Failure of pulse intravenous methylprednisolone treatment in juvenile dermatomyositis. J Pediatr 128: 429–432, 1996. 394. Woo TY, Callen JP, Voorhees JJ, et al: Cutaneous lesions of dermatomyositis are improved by hydroxychloroquine. J Am Acad Dermatol 10: 592–600, 1984. 395. Olson NY, Lindsley CB: Adjunctive use of hydroxychloroquine in childhood dermatomyositis. J Rheumatol 16: 1545–1547, 1989. 396. Pelle MT, Callen JP: Adverse cutaneous reactions to hydroxychloroquine are more common in patients with dermatomyositis than in patients with cutaneous lupus erythematosus. Arch Dermatol 138: 1231–1233, 2002. 397. Malaviya AN, Many A, Schwartz RS: Treatment of dermatomyositis with methotrexate. Lancet 2: 485–488, 1968. 398. Sokoloff MC, Goldberg LS, Pearson CM: Treatment of corticosteroid-resistant polymyositis with methotrexate. Lancet 1: 14–16, 1971. 399. Metzger AL, Bohan A, Goldberg LS, et al: Polymyositis and dermatomyositis: combined methotrexate and corticosteroid therapy. Ann Intern Med 81: 182–189, 1974. 400. Jacobs JC: Methotrexate and azathioprine treatment of childhood dermatomyositis. Pediatrics 59: 212–218, 1977. 401. Fischer TJ, Rachelefsky GS, Klein RB, et al: Childhood dermatomyositis and polymyositis. Treatment with methotrexate and prednisone. Am J Dis Child 133: 386–389, 1979. 402. Benson MD, Aldo MA: Azathioprine therapy in polymyositis. Arch Intern Med 132: 447–451, 1973. 403. Niakan E, Pitner SE, Whitaker JN, et al: Immunosuppressive agents in corticosteroid-refractory childhood dermatomyositis. Neurology 30: 286–291, 1980. 404. Zabel P, Leimenstoll G, Gross WL: Cyclosporin for acute dermatomyositis. Lancet 1: 343, 1984. 405. Girardin E, Dayer JM, Paunier L: Cyclosporine for juvenile dermatomyositis. J Pediatr 112: 165–166, 1988. 406. Heckmatt J, Hasson N, Saunders C, et al: Cyclosporine in juvenile dermatomyositis. Lancet 1: 1063–1066, 1989. 407. Dantzig P: Juvenile dermatomyositis treated with cyclosporine. J Am Acad Dermatol 22: 310–311, 1990. 408. Villalba L, Hicks JE, Adams EM, et al: Treatment of refractory myositis: a randomized crossover study of two new cytotoxic regimens. Arthritis Rheum 41: 392–399, 1998. 409. Cronin ME, Miller FW, Hicks JE, et al: The failure of intravenous cyclophosphamide therapy in refractory idiopathic inflammatory myopathy. J Rheumatol 16: 1225–1228, 1989. 410. Sinoway PA, Callen JP: Chlorambucil. An effective corticosteroid-sparing agent for patients with recalcitrant dermatomyositis. Arthritis Rheum 36: 319–324, 1993. 411. Zeller V, Cohen P, Prieur AM, et al: Cyclosporin A therapy in refractory juvenile dermatomyositis. Experience and long-term followup of 6 cases. J Rheumatol 23: 1424–1427, 1996. 412. Reiff A, Rawlings DJ, Shaham B, et al: Preliminary evidence for cyclosporin A as an alternative in the treatment of recalcitrant juvenile rheumatoid arthritis and juvenile dermatomyositis. J Rheumatol 24: 2436–2443, 1997. 413. Grau JM, Herrero C, Casademont J, et al: Cyclosporine A as first choice therapy for dermatomyositis. J Rheumatol 21: 381–382, 1994. 414. Kobayashi I, Yamada M, Takahashi Y, et al: Interstitial lung disease associated with juvenile dermatomyositis: clinical features and efficacy of cyclosporin A. Rheumatology (Oxf) 42: 371–374, 2003. 415. Yoshimasu T, Ohtani T, Sakamoto T, et al: Topical FK506 (tacrolimus) therapy for facial erythematous lesions of cutaneous lupus erythematosus and dermatomyositis. Eur J Dermatol 12: 50–52, 2002. 416. Peters AM, Heckmatt JZ, Hasson N, et al: Renal haemodynamics of cyclosporin A nephrotoxicity in children with juvenile dermatomyositis. Clin Sci (Lond) 81: 153–159, 1991. 417. Lang BA, Laxer RM, Murphy G, et al: Treatment of dermatomyositis with intravenous gammaglobulin. Am J Med 91: 169–172, 1991. 418. Collet E, Dalac S, Maerens B, et al: Juvenile dermatomyositis: treatment with intravenous gammaglobulin. Br J Dermatol 130: 231–234, 1994. 419. Roifman CM: Use of intravenous immune globulin in the therapy of children with rheumatological diseases. J Clin Immunol 15: 42S–51S, 1995.
420. Sansome A, Dubowitz V: Intravenous immunoglobulin in juvenile dermatomyositis—four year review of nine cases. Arch Dis Child 72: 25–28, 1995. 421. Tsai MJ, Lai CC, Lin SC, et al: Intravenous immunoglobulin therapy in juvenile dermatomyositis. Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi 38: 111–115, 1997. 422. Al Mayouf SM, Laxer RM, Schneider R, et al: Intravenous immunoglobulin therapy for juvenile dermatomyositis: efficacy and safety. J Rheumatol 27: 2498–2503, 2000. 423. Cherin P, Herson S, Wechsler B, et al: Efficacy of intravenous gammaglobulin therapy in chronic refractory polymyositis and dermatomyositis: an open study with 20 adult patients. Am J Med 91: 162–168, 1991. 424. Cherin P, Piette JC, Wechsler B, et al: Intravenous gamma globulin as first line therapy in polymyositis and dermatomyositis: an open study in 11 adult patients. J Rheumatol 21: 1092–1097, 1994. 425. Brewer EJ Jr, Giannini EH, Rossen RD, et al: Plasma exchange therapy of a childhood onset dermatomyositis patient. Arthritis Rheum 23: 509–513, 1980. 426. Anderson L, Ziter FA: Plasmapheresis via central catheter in dermatomyositis: a new method for selected pediatric patients. J Pediatr 98: 240–241, 1981. 427. Dau PC, Bennington JL: Plasmapheresis in childhood dermatomyositis. J Pediatr 98: 237–240, 1981. 428. Dau PC: Plasmapheresis in idiopathic inflammatory myopathy. Experience with 35 patients. Arch Neurol 38: 544–552, 1981. 429. Miller FW, Leitman SF, Cronin ME, et al: Controlled trial of plasma exchange and leukapheresis in polymyositis and dermatomyositis. N Engl J Med 326: 1380–1384, 1992. 430. De Wilde A, DiSpaltro FX, Geller A, et al: Extracorporeal photochemotherapy as adjunctive treatment in juvenile dermatomyositis: a case report. Arch Dermatol 128: 1656–1657, 1992. 431. Miller ML, Smith RL, Abbott KA, et al: Use of etanercept in chronic juvenile dermatomyositis (JDM). Arthritis Rheum 46 (Suppl): S306, 2002. 432. Levine TD: A pilot study of rituximab therapy for refractory dermatomyositis. Arthritis Rheum 46(Suppl): S488, 2002. 433. Maillard SM, Wilkinson N, Riley P, et al: The treatment of persistent idiopathic inflammatory myositis (IIM) with anti-TNFα therapy. Arthritis Rheum 46 (Suppl): S307, 2002. 434. Wulffraat NM, Sanders LA, Kuis W: Autologous hemopoietic stem-cell transplantation for children with refractory autoimmune disease. Curr Rheumatol Rep 2: 316–323, 2000. 435. Dent CE, Stamp TC: treatment of calcinosis circumscripta with probenecid. Br Med J 1: 216–218, 1972. 436. Metzger AL, Singer FR, Bluestone R, et al: Failure of disodium etidronate in calcinosis due to dermatomyositis and scleroderma. N Engl J Med 291: 1294–1296, 1974. 437. Uttley WS, Belton NR, Syme J, et al: Calcium balance in children treated with diphosphonates. Arch Dis Child 50: 187–190, 1975. 438. Skuterud E, Sydnes OA, Haavik TK: Calcinosis in dermatomyositis treated with probenecid. Scand J Rheumatol 10: 92–94, 1981. 439. Lian JB, Pachman LM, Gundberg CM, et al: Gamma-carboxyglutamate excretion and calcinosis in juvenile dermatomyositis. Arthritis Rheum 25: 1094–1100, 1982. 440. Miller G, Heckmatt JZ, Dubowitz V: Drug treatment of juvenile dermatomyositis. Arch Dis Child 58: 445–450, 1983. 441. Ames EL, Posch JL: Calcinosis of the flexor and extensor tendons in dermatomyositis—case report. J Hand Surg Am 9: 876–879, 1984. 442. Wilsher ML, Holdaway IM, North JD: Hypercalcaemia during resolution of calcinosis in juvenile dermatomyositis. Br Med J (Clin Res Ed) 288: 1345, 1984. 443. Fuchs D, Fruchter L, Fishel B, et al: Colchicine suppression of local inflammation due to calcinosis in dermatomyositis and progressive systemic sclerosis. Clin Rheumatol 5: 527–530, 1986. 444. Martinez-Cordero E, Lopez-Zepeda J, Choza-Romero F: Calcinosis in childhood dermatomyositis. Clin Exp Rheumatol 8: 198–200, 1990. 445. Nakagawa T, Takaiwa T: Calcinosis cutis in juvenile dermatomyositis responsive to aluminum hydroxide treatment. J Dermatol 20: 558–560, 1993. 446. Ichiki Y, Akiyama T, Shimozawa N, et al: An extremely severe case of cutaneous calcinosis with juvenile dermatomyositis, and successful treatment with diltiazem. Br J Dermatol 144: 894–897, 2001. 447. Harel L, Harel G, Korenreich L, et al: Treatment of calcinosis in juvenile dermatomyositis with probenecid: the role of phosphorus metabolism in the development of calcifications. J Rheumatol 28: 1129–1132, 2001. 448. Shearin JC, Pickrell K: Surgical treatment of subcutaneous calcifications of polymyositis or dermatomyositis. Ann Plast Surg 5: 381–385, 1980. 449. Moore EC, Cohen F, Douglas SD, et al: Staphylococcal infections in childhood dermatomyositis—association with the development of calcinosis, raised IgE concentrations and granulocyte chemotactic defect. Ann Rheum Dis 51: 378–383, 1992. 450. Ostrov BE, Goldsmith DP, Eichenfield AH, et al: Hypercalcemia during the resolution of calcinosis universalis in juvenile dermatomyositis. J Rheumatol 18: 1730–1734, 1991. 451. Petersen KF, Oral EA, Dufour S, et al: Leptin reverses insulin resistance and hepatic steatosis in patients with severe lipodystrophy. J Clin Invest 109: 1345–1350, 2002. 452. Pachman LM, Ramsey-Goldman R, Lipton R, et al: The NIAMS juvenile dermatomyositis ( JDM) research registry: parent assessment of child’s symptoms at 36 months after diagnosis. Arthritis Rheum 46 (Suppl): S307, 2002.
C H A P T E R 453. Huber AM, Lang B, LeBlanc CM, et al: Medium- and long-term functional outcomes in a multicenter cohort of children with juvenile dermatomyositis. Arthritis Rheum 43: 541–549, 2000. 454. Miller FW, Rider LG, Chung YL, et al: Proposed preliminary core set measures for disease outcome assessment in adult and juvenile idiopathic inflammatory myopathies. Rheumatology (Oxf) 40: 1262–1273, 2001. 455. Miller JJ III: Late progression in dermatomyositis in childhood. J Pediatr 83: 543–548, 1973. 456. Lovell HB, Lindsley CB: Late recurrence of childhood dermatomyositis. J Rheumatol 13: 821–822, 1986. 457. Miller JJ III, Koehler JP: Persistence of activity in dermatomyositis of childhood. Arthritis Rheum 20: 332–337, 1977. 458. Collison CH, Sinal SH, Jorizzo JL, et al: Juvenile dermatomyositis and polymyositis: a follow-up study of long-term sequelae. South Med J 91: 17–22, 1998.
18 J U V E N I L E D E R M AT O M Y O S I T I S
441
459. Chowdhary V, Wakhlu A, Agarwal A, et al: Outcome in juvenile dermatomyositis. Indian Pediatr 39: 931–935, 2002. 460. Barnes AB, Link DA: Childhood dermatomyositis and pregnancy. Am J Obstet Gynecol 146: 335–336, 1983. 461. Gutierrez G, Dagnino R, Mintz G: Polymyositis/dermatomyositis and pregnancy. Arthritis Rheum 27: 291–294, 1984. 462. Pinheiro Gd, Goldenberg J, Atra E, et al: Juvenile dermatomyositis and pregnancy: report and literature review. J Rheumatol 19: 1798–1801, 1992. 463. Miller LC, Michael AF, Kim Y: Childhood dermatomyositis. Clinical course and long-term follow-up. Clin Pediatr (Phila) 26: 561–566, 1987. 464. Chalmers A, Sayson R, Walters K: Juvenile dermatomyositis: medical, social and economic status in adulthood. Can Med Assoc J 126: 31–33, 1982. 465. Taieb A, Guichard C, Salamon R, et al: Prognosis in juvenile dermatopolymyositis: a cooperative retrospective study of 70 cases. Pediatr Dermatol 2: 275–281, 1985.
C HAP T E R
19
THE SYSTEMIC SCLERODERMAS AND RELATED DISORDERS Francesco lullan and James T. Cassidy
;af DEFINITION AND CLASSIFICATION The word scleroderma means "hard skin." The diseases grouped under this term mean much more, although hardening of the skin is a feature that is common to all types of the disorder and is the most signal characteristic of these entities. A classification of the systemic and localized sclerodermas is in Table 19-1. Systemic scleroderma is subdivided by the extent of the skin disease into diffuse cutaneous systemic scleroderma (DCSS) and limited cutaneous systemic scleroderma (LCSS), previously designated as the CREST syndrome (calcinosis cutis, Raynaud's phenomenon, esophageal dysfunction, sclerodactyly, telangiestasia). The localized forms of the disease, such as morphea or linear scleroderma, often are regarded as more dermatologic than rheumatologic (see Chapter 20). Systemic scleroderma is rare in childhood. Many generalizations have been based primarily on the adult literature, and DCSS and LCSS are often discussed together in publications or inadequately separated.
with scleroderma and cutaneous histologic characteristics in another. In 1969, D'Angelo and coUeagues ltl reported postmortem abnormal involvement as percentages in excess of control subjects: skin, 98%; esophagus, 74%; lungs, 59%; kidneys, 49%; small intestine, 46%; pericardium 41%; large intestine, 39%; pleura, 29%; and myocardium, 26%. Other organs with less frequent involvement were the adrenal glands, lymph nodes, thyroid, and peripheral arteries.
DIFFUSE CUTANEOUS SYSTEMIC SCLERODERMA DCSS is a chronic, multisystem connective tissue disease characterized by sclerodermatous skin changes and widespread abnormalities of the viscera. Rodnan 11 defined DCSS as a disorder in which "symmetrical fibrous thickening and hardening (sclerosis) of the skin is combined with fibrous and degenerative changes in synovium, digital arteries, and certain internal organs, most notably the esophagus, intestinal tract, heart, lungs, and kidneys." Systemic sclerosis sine scleroderma has been described in adults
HISTORICAL REVIEW The early literature presents a confusing picture of scleroderma in a child because many cases were more compatible with a diagnosis of scleredema. In 1895, Lewin and Heller l reviewed 505 cases of scleroderma, mainly from the European literature. Goodman 2 observed that 88 occurred in children from birth to 19 years but that most cases were examples of circumscribed disease. Only 1 of 12 children reported as diffuse scleroderma was compatible with current concepts, with the rest being the "acute form," probably scleredema. 3 Another survey concluded that only 12 children with generalized scleroderma had been reported in the world literature through 1960. 4 In 1961, the Mayo Clinic added 63 additional pediatric cases in summarizing experience with 727 patients. 5 A survey (Padua database) of members of the Pediatric Rheumatology European Society and other pediatric rheumatology centers around the world (67 centers in 28 countries) reviewed 127 children with systemic scleroderma and 688 with localized scleroderma. 6.7 Pathologic studies lagged behind clinical reports, and there were no comprehensive descriptions until 1924, when Kraus H described pulmonary and cardiac fibrosis in a patient with scleroderma, and Matsui9 detailed necropsy findings in five patients
442
is a variant of limited cutaneous involvement and not a separate or distinct disorderY·13 Other than the absence of skin thickening, this disease has no significant differences in internal organ involvement, laboratory abnormalities, serum autoantibodies, or survival rate compared with LCSS.
Epidemiology DCSS has been reported worldwide and in all races. 14-22 It has an estimated annual incidence of 0.45 to 1.9 cases
per 100,000 persons in the general population and a prevalence of approximately 24 cases per 100,000.14.16.21-23 The frequency of this disorder increases with age and is maximal in the 30- to 50-year age group. Prevalence has been estimated at 27.6 cases per 100,000 adults (95% CI: 245-310). The disease is more frequent in African Americans and in Choctaw Native Americans. 22 African American women are more likely to develop diffuse disease, be diagnosed at a younger age, and have a poorer survival rate. 21 Onset in childhood is uncommon. Children younger than 10 years account for less than 2% of all cases, and
C HAP T E R
1=..
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
There is no racial predilection or peak age at onset determined for children.
1ABLE 19 -1 Classification of Systemic and Local Sclerodermas and Scleroderma-Like Disorders
There are several small series and case reports of children with DCSS totaling just over 115 patients, although there are undoubtedly many unreported cases: 33 ,34 Children for whom the diagnosis of DCSS is clear and for whom appropriate data are available in published series are summarized in Table 19_2.3.4.27.32.35-50 An additional 127 patients in the Padua database are included. DCSS occurs with equal frequency in boys and girls younger than 8 years old, whereas girls outnumber boys 3 to 1 when disease onset occurs in children who are older than 8 years. Among adults, the male-to-female ratio in the childbearing years is 3:1 to 5:1, whereas in an older age group Colder than 45 years) it is 1.8: 1.'1 One hypothesis is that factors such as the hormonal milieu, pregnancy-related events, or reproductive-specific exposures are responsible for these differences in disease susceptibility.
Systemic disease Cutaneous scleroderma Diffuse Limited Overlap syndromes Sclerodermatomyositis or with other connective tissue diseases Mixed connective tissue disease Localized disease Plaque morphea Generalized morphea BuIlous morphea Linear morphea Deep morphea Graft-versus-host disease ChemicaIly induced scleroderma-like disease Polyvinyl chloride Bleomycin Pentazocine Toxic oil syndrome Adjuvant disease Pseudosclerodermas Phenylketonuria Syndromes of premature aging Localized idiopathic tibroses Scleredema Diabetic cheiroarthropathy Porphyria cutanea tarda
Etiology and Pathogenesis
patients between 10 and 20 years for only 1.2% to 9OAlP-20,ZHI It has been estimated that approximately 3% of all patients with DCSS were children,32 DCSS constitutes 0.2% to 0.9% of the major connective tissue disorders in pediatric rheumatology clinics (see Table 1-4).
I!;. TABLE 19-2
The cause of DCSS is unknown, despite significant advances in understanding of potential pathogenetic mechanisms.52 The disease can be represented as a tripartite process in which dysfunction of the immune system, endothelium, and fibroblasts gives rise to a heterogeneous phenotype that is characterized prominently by fibrosis (Fig. 19-1). Autoimmunity is evident by the elaboration of circulating disease-specific autoantibodies. Raynaud's phenomenon, capillary dropout, endothelial injury, and abnormalities in vascular tone are manifestations of endothelial cell dysfunction. Fibroblast dysfunction is represented by fibrosis as the result of increased synthesis and deposition of extracellular matrix proteins. These three areas of abnormal func-
Scleroderma in Childhood Patients (n)
Study Jaffe and Winkelmann' Kennedy3s Kass et al4 Mukherjee et aP" Ve1ayos and Cohen37 Szymanski-Jagiello et aPH Goel and Shanks 39 Ansell et al4(' Gray and Altman 4l Cassidy et a1 32 Kornreich et al 27 Schlesinger and SchaIler42 Girouard et al 43 Spencer-Green et al44 Larregue et al45 Burge et al46 Suarez-Almazor et al 47 Lababidi et al 48 Martinez-Cordero et al49 Vancheeswaran et al'" Martini et al"
443
Total
Male
Female
3
2
Age at Onset (yr)
3---11
5 1
o
1
6
7
3
4
5-15
1
6 4 6-14
1
1
o
1
o
12 4
2 1
10
1 1
o
1
15 131 11 3
o
15
4 3
8
9 3
1
o o 1
3
o 9
3 9 2
2
o
4 5
1 1 1
3 4 6
4 30
7
7 11
127
2
97
·Patient has linear scleroderma with radiologic evidence of esophageal involvement. 'This series presumably includes children in an earlier report from the same institution (Kass et al'). 'Two of these patients had serum anti-RNP antibodies,
2·-10.5 13 6
3---15 3---12 N/A 6*-12 3---17 8-11 6-7 8-10 4--13
5-16 3---13 1-15
444
C HAP
T ER
19 SYSTEMIC SCLERODERMAS AND RELATED DISORDERS Endothelial cells
Fibroblast
• Figure 19-1
Possible pathogenic mechanisms in sderodenna.
tion, although apparently unassociated with each other, are closely linked by several immunologic alterations.
Immunologic Factors Many reports suggest that cellular immunity plays a major role in the initiation of scleroderma. These factors include the presence of mononuclear cell (MNC) infiltrates in early lesions, altered function of helper T and NK cells, and release of cytokines, chemokines, and growth factors (Table 19-3). MNC infiltration in various organ systems occurs early and consists of lymphocytes, plasma cells, and fibroblast-histiocytic cells around small blood vessels, eccrine sweat glands, and subcutaneous tissue.53 Activated T lymphocytes are prominent as evidenced by the expression of surface human leukocyte antigen (HLA) class II molecules. 54 The early infiltrates of MNCs release a number of cytokines and chemokines that affect endothelial cells and fibroblasts,
I! II
TABlE 19 J (ylokines dud Growlh fd( 1015 Involved in Ihe Regllidlioll 01 Ihe BlOloglI Behdviol of fiblobldsls
Biologic Effect
Cytoldne or Growth Factor
Increased collagen synthesis Decreased collagen synthesis Fibroblast proliferation
TGF-~, PDGF, IL-I, lL-4, IL-6 IFN-~, lFN-y, TNF-a, ~ lFN-~, IFN-y, TGF-~, PDGF,
Chemoattraction Glycosaminoglycan synthesis Fibronectin synthesis Endothelial cell injury
TNF, IL-I, IL-4 IFN-y, TGF-~, PDGF, TNF, IL-4 TGF-~, TNF, lL-I TGF-~, IL-4 IFN-y, TNF-a, ~, IL-2, NK cell, granzyme A
Reduction of synthesis Collagenase gene induction
TGF-~
TNF-a
IFN, interferon; IL, interlellkin; NK, natural killer cell; PDGF, platelet-derived growth factor; TNF, tumor necrosis factor; TGF, transforming growth factor.
Several growth factors have also been identified in scleroderma skin, including transforming growth factor-~ (TGF-~), connective tissue growth factor (CTGF), and adhesion molecules. TGF~ has pleomorphic cellular actions principally on fibroblasts and endothelial cells. 55 In vitro, it stimulates the synthesis of the extracellular matrix, including types I and III collagens,56 It also promotes fibrosis indirectly by inhibiting collagenase activityY Fibroblasts from scleroderma skin express in vivo and in vitro increased levels of TGF-~l and TGF-~2 receptor proteins compared with controls,58,59 Blockade of TGF-~l signaling with monoclonal antibodies inhibits upregulation of collagen synthesis in scleroderma fibroblasts. 58 Polymorphisms of the TGF~l gene have been reported in Japanese patients. 60 CTGF levels are greatly elevated in the dermis of patients with scleroderma and down-regulated by iloprost infusion,61 Although TGF-~l is known to initiate fibrosis, CTGF may play a greater role in maintaining and promoting fibrosis. 62 ,63 Levels of a number of cytokines (e.g., interleukin !Ill-I, IL-2, IL-4, IL-6, IL-8) are increased in the serum.64-66 Several interleukins (e.g., IL-4, IL-6, IL-8) have also been demonstrated in scleroderma skin, Serum levels and spontaneous production of IL-12, a potent inducer of type 1 helper (Thl) T cells, were increased in patients and associated with renal vascular damage. 67 Specific cytokines promote fibrosis; others, such as interferon-y (IFN-y), are potent suppressors of collagen synthesis. The effects of some cytokines are mixed. Tumor necrosis factor (TNF) decreases fibroblast production of types I and III collagen while promoting collagenase gene induction. 55 This cytokine also stimulates the proliferation of some fibroblasts and increases endothelial cell expression of adhesion molecules (Le., E-selectin, intracellular adhesion molecule-l [ICAM-ll and vascular cell adhesion molecule-l [VCAM-l]) and release of endothelin-I. The level of circulating soluble VCAM-l correlates with impaired left ventricular diastolic function. 68 Serum CD44 (sCD44), another adhesion molecule that regulates the migration of leucocytes, was elevated in scleroderma patients, particularly in those with limited cutaneous disease. 69 Genetic predispositions are being clarified; for example, the TNF-863A allele is associated with anticentromere antibody seropositivity.71l Chemokines, which are produced by white blood cells, are operative in the recruitment of specific types of leukocytes to involved skin and tissue. Serum levels of MCP-l, which attracts MNCs, and MIP-l, which attracts monocytes and helper T cells, are elevated?! A constitutive overexpression of MCP-l mRNA has been identified in scleroderma skin,72 Treatment with antibodies to MCP-l results in reduced chemotactic activity, indicating that this chemokine may be an important agent in the initiation of cutaneous inflammation. 73 Levels of chemokines ILS and growth-regulated oncogene-a (GRO-a), potent chemoattractants and activators of neutrophils, were found to be elevated in scleroderma and GRO-a correlated particularly with pulmonary involvement,74 Cell-mediated immunity to laminin, a constituent of basement membrane and to a lesser extent to type IV collagen, has also been demonstrated in patients with DCSS,"
Vascular Factors According to some authorities, endothelial cell injury is the central pathogenic event and predates fibrotic changes. The endothelial cell may be damaged by protease-dependent mechanisms that are independent of complement and immunoglobulin. Abnormalities of cutaneous mast cell number and type and of mast cell activation as a prefibrotic event have been documented. 76 Damage to the endothelial cell results in increased vascular permeability that is responsible for the edematous phase of the illness, which leads to activation of fibroblasts, increased collagen production, and resultant fibrosis. It also initiates activation of the coagulation path-
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
way, .contributing to an accumulation of platelets that release factors leading to proliferation and migration of myointimal cells.
Endothelial Cell Factors Evidence that the endothelial cell is damaged is provided by studies of the histology of the lesions in DCSS and by demonstration of elevated levels of factor VIII-related antigen,77 although this has not been a consistent observation. 78 Reduced plasma angiotensin-converting-enzyme activity may be an additional marker of endothelial injury.79 Endothelial cell apoptosis is accelerated. so The microvascular injury leads to arteriolar intimal fibrosis and narrowing of the vascular lumen, which results in ischemic damage. 81 Anti-endothelial cell antibodies are also present82.83 and lead to endothelial damage, vascular hyperpermeability, and myointimal cell proliferation. The link between vascular alterations and cellular immunity is represented by adhesion molecules. Three major families have been defined: selectins, integrins, and members of the immunoglobulin gene superfamily. Selectins mediate the initial contact of leukocytes with endothelial cells. Overexpression of E-selectin and P-selectin has been found in sera""S'i and in endothelial cells in the skin and minor salivary glands of scleroderma patients. 86 Integrins, a family of heterodimeric transmembrane glycoprotein molecules, serve as a means of communication between extracellular matrix molecules (e.g., collagen, laminin, fibronectin) through the cell membrane to the intracellular compartment. Expression of integrins VLA-2, VLA-4, and lFA-l is increased on endothelial cells, MNCs, fibroblasts, and dendritic cells in scleroderma skin. s6 ICAM-l and VCAM-l, members of the immunoglobulin gene superfamily on endothelial cells, and lFA-l and VLA-4, integrin receptors on lymphocytes, play a significant role in the interaction between lymphocytes and endothelial cells or fibroblasts. These molecules are increased on endothelial cells and fibroblasts in sclerodermatous skin87 .88 and facilitate MNC damage to endothelial cells and fibroblasts.
Abnormalities of Collagen Excessive accumulation of collagen in affected skin led to the hypothesis that there might be abnormalities of collagen type or metabolism. 22.89 There is an increased number of collagenproducing fibroblasts in the skin90; however, the ratios of various collagen types are normal. 91 Although reduced collagenase activity was found in one study,92 it was normal in another. 93 Abnormalities of glycosylation94 and hydroxylation95 of the collagen molecule may prevent normal feedback mechanisms from being effective in controlling synthesis and permit excessive deposition of collagen. A defect in the regulation of genes controlling apoptosis of fibroblasts (for example caspase 6, BCl2, and elastin) has been reported. 96
Genetic Background The rare familial occurrence of DCSS has been confmned in a mother and her 6-year-old son,41 in a second family with two affected sisters aged 12 and 16 years,97 and in monozygotic twins. 98 These reports and others have been summarized by Gray and Altman41 and Englert and colleagues. 99 There is little agreement about the potential associations of histocompatability antigens with DCSS. Initial studies indicated associations with class I alleles HLA-A9, lOll HLABS,101.102 and HLA-Bw35 103 and class II alleles HLA-DR3,102 HLA-DR5,104 and HLA-DRw15. 105 Associations with HLA-DR
445
and HLA-DQ alleles (DQB3.1, DQBl.1, DQB1.2, DQB1.3) have been reviewed by Whiteside and colleagues lO6 and Fox and Kang. 107 DRBl*1104 and DRB1*1101 confer odds ratios of 3.5 and 2.3 for the disease in adults. lOO Prolonged persistence of fetal progenitor cells and microchimerism in T cells has been associated with DQA1·0501. 109 ,110 Microchimerism, the presence within one individual of a very low level of cells derived from a different individual, was postulated as a possible cause of scleroderma from studies of chronic graft-versus-host disease (GVHD), a chimeric disorder in which donor T cells or NK cells react against the HLAs of the recipient. Microchimerism occurs in women who had previous pregnancies, individuals who have had blood transfuSions, and children with cells from the mother or a twin. Maternal cells can persist in an immunocompetent offspring even in adult life, and fetus-derived hemopoietic cells have persisted in the maternal circulation for many years postpartum. I I I Fetal DNA persists for even longer periods. ll2 Although microchimerism can be identified in normal subjects and in other diseases, it has been proposed as an important factor in pathogenesis of autoimmune diseases. 1l3 In scleroderma, chimeric cells are increased in number and, compared with normal subjects, are more similar to the maternal cells. Quantitative analysis of microchimerism has been reported in sclerodermatous skin. 114 Nelson and coworkers ll2 studied 40 mothers and documented high, persistent concentrations of male DNA in cells of the vascular compartment years after giving birth to a son. HLA class II compatibility of the child was more common in DCSS patients than in control subjects. An investigation by Artlett and colleagues m also concluded that fetal anti-maternal graftversus-host reactions might be involved in pathogenesis. Although this theory of a chronic graft-versus-host reaction is attractive, studies offer no data to explain the occurrence of scleroderma in men or women who have never had children.
Clinical Manifestations Early Signs and Symptoms Presenting signs and symptoms of DCSS in children are in Table 19-4. Onset of the disease is usually insidious and the course prolonged, punctuated by periods of inactivity or episodes of severe systemic complications, occasionally ending in remission or more often in chronic disability or death. 116 The onset is often characterized by the development of Raynaud's phenomenon; tightening, thinning, and atrophy of the skin of the hands and face; or the appearance of cutaneous telangiectases about the face, upper trunk, and hands. There is often a diagnostic delay of years because of the subtle nature of this presentation. A comprehensive general review of the assessment of patients with systemic sclerosis has been published as a symposium. 117
Skin Disease The onset of cutaneous abnormalities may be especially insidious, but these changes characteristically evolve in a sequence beginning with edema, followed by induration and sclerosis resulting in marked tightening and contractures, and eventually resulting in atrophy.
446
I!. II
C HAP T E R
TABLE t9-4
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
Presenting Signs dnd Symptoms in Children with Systemic Scleroderma
Sign Skin tightening Raynaud's phenomenon Soft tissue contracture Arthralgia Muscle weakness and pain Subclltaneous calcification Dysphagia Dyspnea
Jaffe et al (n=5) 1961
Goel et al (n=4) 1974
4 5 2
4 2 1 3 1
_I
-
-
-
-
Cassidy etll (n=13) 1977 15 11 10 9 4
3 3 3
Kornreich et al (n=13) 1977
Lanigue et al (n=3) 1983
13 5
3 3 2 2
-
-
-
Suarez-Almazor et al (n=4) 1985 4 4 4 1 2 1 1 1
Lababldl et al (n=5) 1991 3 3 3 5 2 -
2 2
Martini et al (n=118) 2003
Total %*
95 88 28 16 10 16 14
H4.4 72.4 61.1 32.2 17.1 10.2 15.5 14.1
'Percentage calculated only on series in which detailed information was provided. 'Dash indicates that information was not provided.
Edema Tense, nonpitting swelling of the skin and subcutaneous tissues of the digits, hands, arms, and face, or localized areas on the trunk, may be the initial manifestation of the disease. Edematous areas may be warm and tender with an erythematous border, but are often asymptomatic, Swelling may persist for weeks or months before subsiding or being replaced by sclerosis,
nail fold is often the most obvious early location of abnormal vessels (Fig. 19-5), and examination with an ophthalmoscope demonstrates capillary dropout, tortuous dilated loops, and occasionally distorted capillary architecture (Table 19-5 and Fig. 19-6).44.118,119 There is usually redundant cuticular growth; dystrophic changes in the nails have also been reported. llo Digital pitting, sometimes with ulceration and gangrene, occurs in the pulp of the fingertips as a result of ischemia and is one of the minor diagnostic criteria (Fig. 19-7).
Sclerosis During the sclerotic phase, the skin develops a waxy texture and becomes tight, hard, and bound to subcutaneous structures. This is particularly noticeable in skin of the dorsal surface of the digits, so-called acrosclerosis (Fig. 19-2), and face (Fig, 19-3); the characteristic immobile, expressionless, unwrinkled appearance of the skin may be the first clue to the diagnosis. The absence of forehead wrinkling and the presence of circumoral furrowing or diminished aperture of the mouth are particularly characteristic. Sclerotic changes usually follow a temporal sequence of development, beginning with bilateral, symmetric acrosclerosis, followed by involvement of the face, and finally by changes in the skin of the trunk and proximal limbs.
Atrophy The long-term consequence of edema and sclerosis is atrophy of skin and adnexa, These superficial abnormalities result in a shiny appearance of the skin accompanied by areas of hypo- or hyper-pigmentation and often by deposition of calcium salts in the subcutaneous tissues. Cutaneous lesions in all stages of evolution may be observed Simultaneously in the same child.
Telangiectases Telangiectases, fine macular dilatations of cutaneous or mucous membrane blood vessels, are characteristic (Fig. 19-4). Unlike "spider" angiomata that fill rapidly from central arterioles, telangiectatic vessels fill slowly and lack the characteristic central vessel. The periungual
• Figure 19-Z A, The hands of a 9-year-old girl with diffuse cutaneous systemic scleroderma.The skin over the dorsum of the fingers is taut and shiny. B, Five years later, the tightening is more evident, and flexion contractures have developed. (Courtesy of Dr, K. Den.)
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
447
• FIpre 19-3 Shiny skin on the face. (See color insert.) • Figure 19-5 Changes in the nail fold vessels with visible tortuosity, thickening, and pigmentary extrusion onto the cuticles.
Calcinosis Subcutaneous calcification, especially over the elbows, metacarpophalangeal joints, and knees, may occur, sometimes with ulceration of surrounding skin. Extensive periarticular calcification (i.e., calcinosis circumscripta) may be a late complication (Fig. 19-8). These lesions, if
extensive, lead to a severe reduction in joint mobility. Small, hard, subcutaneous nodules sometimes occur over the extensor surfaces of joints of the fingers and differ histologically from rheumatoid nodules by the absence of fibrinoid necrosis.1 21
Raynaud's Phenomenon The triple-phase sequence of blanching, cyanosis, and erythema, occurring spontaneously or in response to cold or to physical or emotional stress, was described by Raynaud, a French medical student, in 1862. 122 . 123 Raynaud's phenomenon denotes this tricolor change; Raynaud's disease is applied to patients in whom no underlying structural vascular disease can be detected. Raynaud's phenomenon occurs in 90% of children with DCSS and is often the initial symptom of the disorder, preceding other manifestations in some instances by years.124.125 It is characterized by obstructive digital arterial disease and sympathetic hyperactivity. In particular, the blanching phase of the disorder is well demarcated and uniformly white, beginning at the distal end of a digit and ending abruptly in the proXimal digit or at the metacarpophalangeal joint (Figs. 19-9, 19-10).126 These changes may be restricted to a single digit, be unilateral or bilateral, and usually spare the thumb. The color changes may be accompanied by paresthesias, numbness, or pain (especially during the erythematous phase). Raynaud's phenomenon is much more common in the fingers, but can be observed in toes and occasionally in ears, tip of the nose, lips, or tongue. Vascular spasm within viscera such as the esophagus and heart may accompany the peripheral anoxia. 127
• FIpre 19-4 Telangiectases. (See color insert.)
Juvenile idiopathic (or primary) Raynaud's disease, as in adults, principally affects girls (8()o!<J) and is frequently independent of association with connective tissue diseases. l28 Antinuclear antibody (ANA) seropositivity and abnormal nail fold capillaries are infrequent fmdings l29.130; however, even in instances in which skin changes of DCSS have not been demonstrated, ANAs or abnormalities of esophageal motility have been observed l 2'J and suggest the possibility of eventual disease progression to systemic
448
I!. III
C HAP T E R
lABLE 19 5
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
Abnormalities of Nail Fold tapillaries in Rheulllatic Dist)l(lers of (hildl\'ll
Disorder Systemic scleroderma Localized scleroderma Eosinophilic fasciitis Mixed connective tissue disease Raynaud's disease! Acrocyanosis Dermatomyositis* Systemic lupus erythematosus Juvenile rheumatoid arthritis
Deaeased Density
DllataUon and Tortuosity
+++ ++ + + +
thickening
ArborIzaUon
Dropout
+++
+++
+
+++
+ ++ + + +++ ++
+ ++
+
++
+++
++
++ +
'Markedly abnormal pattern correlates with development of pulmonary hypertension. 'Rare in children; abnormal patterns may predict future development of connective tissue disease. IAbnormal patterns correlate with histopathologic lesions of noninflammatory vasculopathy and chronic ulcerative course. -, Normal; + to +++, increasing degree of abnormality. Data from references 44, 118, 119, 131, 367, 368.
involvement. 131 Table 19-6 represents the relative proportions of children and adults with Raynaud's phenomenon or disease. The pathogenesis of Raynaud's phenomenon is not entirely clear. Raynaud suggested that the vasospasm resulted from increased sympathetic tone. The initial event is arterial vasoconstriction with resultant decrease in cutaneous blood flow leading to pallor of the affected part, often followed by systemic reduction in perfusion of vital organs. Cyanosis results from venous stasis; erythema is the result of reflex vasodilatation caused by mediators released during the ischemic phase. Vasoconstriction of the digital arterioles of patients with
Raynaud's phenomenon can be induced by immersion of the hand in ice water and relieved by warming. This reversibility is highly characteristic and diagnostically useful, although the cutaneous circulation may require minutes to hours to return to normal. Factors other than cold may precipitate the phenomenon, or it may occur spontaneously. The characteristic diagnostic changes in digital arteriolar blood flow can be documented by Doppler flow studies,132 plethysmography (Fig. 19-11), or arteriography (the latter is not usually necessary or indicated in DCSS and performed with some danger of precipitating acute catastrophic arteriolar spasm) (Fig. 19-12).1l3
• figure 19-6 A, There is a reduction in the number of nail fold capillaries and tortuosity of the remaining vessels in the microvasculature viewed with a microscope (magnification x 100). 8, Normal vessels. (Courtesy of Dr. J. Kenik.)
• Figure 19-7 A, Digital pitting of the fingertips. Notice the ulceration of tip of the right thumb (a17lJw) and shiny, tightly stretched skin over the fingertips bilaterally with pronounced flexion contractures at the metacarpophalangeal joints. (A, See color insert.) 8, Digital gangrene of the fourth right finger (a17lJw).
C HAP T E R
19
449
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
• Figure 19-9 Raynaud's phenomenon in a child with DCSS. Blanching of the digits distal to the proximal interphalangeal joints has occurred in response to cold and is accompanied by a dusky cyanosis of the hands. (See color insert.)
• f1Iare 19-8
Caldnosis drcumscripta affecting the thumb.
Capillary blood flow in the fingers and the fingerpad temperature of patients with Raynaud's phenomenon are reduced in warm and cool environments. 1.\4 The phenomenon is not always reversible. A vascular abnormality in tryptophane metabolism has been postulated, and smooth muscle from arteries of patients with scleroderma is hypersensitive to 5-hydroxytryptamine l31 and demonstrates a temperature-dependent loss of reactivity to catecholamine. l36 Locally repeated episodes of intravascular microcoagulation 137 and release of prostaglandins such as thromboxane A z and prostaglandin £1 may also contribute to vasospasm.1 38 Raynaud's phenomenon should be distinguished from two additional disorders: acrocyanosis and chilblains (pernio). Acrocyanosis, first described by Crocq in 1896,139 is an uncommon, painless, vasospastic disorder causing persistent coldness and bluish discoloration of the hands (and less commonly of the feet).14Q-144 Occasionally, excessive perspiration and edema of the hands and feet occur. This condition is exacerbated by cold but is essentially benign and unresponsive to treatment. Nail fold capillaries are abnormal, have dilated loops, and are decreased in number. Chilblains refers to episodic color changes after exposition to severe cold, typically with the development of nodules if recurrent, and represents spasm-induced vessel and tissue damage. 141
Musculoskeletal Disease Musculoskeletal symptoms are common and characteristically occur at or near onset. Among the 127 children with DCSS included in the Padua database, 36% had musculoskeletal symptoms during the course of the disease. Morning stiffness and pain of the small joints of the
• figure 19-10 Thermographic documentation of Raynaud's phenomenon. The apparent amputation of the distal portions of the digits indicates the abrupt transition from normal tissue to cold, blanched skin that is characteristic of this disorder.
I! III
TABLE 19-6 Raynaud's Phenomenon and Disease in Children ilnd Adulls
Percent Affected (0/0) Category Raynaud's disease Raynaud's phenomenon with Nonconnective tissue diseases Juvenile rheumatoid arthritis (or RA) Systemic lupus erythematosus Scleroderma Dermatomyositis ·In association with vasculitis.
Children
Adults
69
70
0.5
15
OS
7 4
19 10 1
3 1
450
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
TIM(
• Figure Ut-ll Plethysmographic study of the index finger of a child with Raynaud's phenomenon. Upper tradng demonstrates pulse amplitude at room temperature.Three abnormal waveforms are shown: anacrotic notching (a); peak notching (b); and high dicrotic notching (diminished) (c). Lower tradng demonstrates the effect of immersion of finger in an ice-water bath. The resultant changes persisted for more than 30 minutes.
hands, knees, and ankles may also be initial manifestations of the disease. Movement of a thickened tendon through its sheath, which is covered with fibrinous deposits, can often be palpated or detected with a stethoscope as an audible, coarse crepitus. Joint pain is usually mild and transient. Joint contractures of insidious onset and limitation of motion are most common at the proximal interphalangeal joints and elbows, but other joints can be affected. Objective evidence of articular inflammation is absent or mild in most instances, although small, bland synovial effusions occur. Muscle inflammation characterized by pain and tenderness occurs in up to one fifth of children, and proximal or distal muscle atrophy may be marked. Although pertinent studies are lacking in children, Clements and colleagues 146 in a group of 24 adults described "simple myopathy" in 20 patients and "complicated myopathy" (Le., resembling inflammatory muscle disease or mixed connective tissue disease) in the remainder. In the simple myopathy group, weakness of hip and shoulder girdle musculature was present in 81%; serum creatine kinase and aldolase levels were modestly elevated in less than one half. Electromyography documented polyphasic motor unit potentials of normal duration and amplitude in 18 of 19 patients, a decreased number of motor units in 5 of 19, but no insertional irritability, denervation, or diminished size or duration of motor unit potentials. In contrast, in patients with complicated myopathy, enzyme levels were similar to values found in patients with polydermatomyositiS, and electromyography exhibited polyphasic motor unit potentials that were short and of low amplitude and with insertional irritability or fibrillations and positive sharp waves.
• Figure 19-12 Digital arteriography outlines occlusions of proper digital arteries (arrowheads).
Gastrointestinal Disease Gastrointestinal involvement affects one fourth of the patients during the course of the disease; proximal disease usually precedes distal involvement. Lesions of the mouth include mucosal telangiectases, reduced interincisal distance caused by skin thickening and tightness, parotitis as part of the sicca syndrome, and loosening of the teeth because of changes in the periodontal membrane. The esophagus is involved, often quite early in the disease, in most children. Dysphagia may be one of the presenting signs. Although many patients are asymptomatic, symptoms On order of decreasing frequency) include heartburn with postural aggravation, dysphagia, delayed emptying, regurgitation with reflux into the throat, nocturnal aspiration, and cough with swallowing. Esophagitis with persistent ulceration and stricture along with progressive weight loss because of voluntary restriction of food intake may follow. Dilatation of the stomach or duodenum occurs uncommonly. At least in adults, gastric arteriovenous ectasia ("watermelon stomach") may develop. Smallbowel involvement usually develops in association with
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
esophageal or colonic disease. 1O Abdominal distention and pain with nausea and vomiting result from gut hypotonia that may occasionally be so severe that pseudo-obstruction occurs. 147 Pneumatosis intestinalis may develop. Malabsorptive diarrhea and delayed colon transit, when present, reflect long-standing disease. l48 Large-bowel disease, although not uncommon, is usually asymptomatic; however, it may cause severe constipation, bloating, or diarrhea. Primary biliary cirrhosis has not been reported in children.
Cardiac Disease Cardiopulmonary disease, although not common at presentation, is a leading cause of morbidity.47.149.15o Pericardial effusions are usually small and asymptomatic, although fever and retrosternal pain may accompany acute disease. 151 Changes in cardiac hemodynamics reflected by the presence of pedal edema, jugular venous distension, hepatomegaly, pulsus paradoxus, and pulsus alternans may be present in patients with chronic effusions. Tamponade from pericardial constriction and severe cardiomyopathy are however rare, although they can be one of the causes of early death and require prompt and aggressive immunosuppressive treatment. 149 Cardiac anoxia may result from the equivalent of Raynaud's phenomenon of the coronary arteries and is a potential precursor of myocardial fibrosis. Although coronary artery disease is uncommon, electrocardiographic changes and even angina pectoris may occur as a result of disease of the myocardial microvasculature. 1s2 Systemic and pulmonary hypertension may contribute to myocardial ischemia.
Pulmonary Disease Pulmonary parenchymal disease is almost universal, and although frequently asymptomatic, patients often have a dry, hacking cough or dyspnea on exertion. JS3 Occasionally, rales or a pleural friction rub are present. Clinical predictors of end-stage disease have been evaluated in adults. 154 The risk of alveolar and bronchiolar carcinoma is also increased in adults. 15s Pulmonary vascular disease results in progressive dyspnea with preserved lung volumes on pulmonary function testing. It can result from pulmonary fibrosis; however, the isolated form of this complication has a much worse prognosis. It can occasionally complicate DCSS associated with anti-fibrillin autoantibodies and is typical also of the LCSS subset of patients. 156 Interstitial pulmonary fibrosis, long recognized as a devastating complication, is being reclassified to reflect differences in histopathology and outcome. It has been postulated that fibrosis also results from pulmonary vascular hyper-reactivity similar to Raynaud's phenomenon. Furst and associates 157 demonstrated decreased pulmonary perfusion as measured by krypton 81m scans after cold challenge to the hands. Fahey and colleagues 158 noticed a low carbon monoxide diffusing capacity (Dieo) in patients with DCSS and Raynaud's phenomenon but failed to demonstrate a decrease with cold challenge as found in patients with idiopathic Raynaud's disease. Increased pulmonary uptake of gallium 67 occurred in most patients with early disease, a finding that sug-
451
gested an inflammatory process. VeselY6 and colleagues 159 measured serum concentrations of KL-6 in 6 children with interstitial lung disease, and in 6 without this abnormality, and compared their results to 20 healthy controls. Ki-6 was significantly higher in the children with pulmonary involvement and served as a clinically useful, noninvasive marker of fibrosis. This highmolecular-weight, mucin-like glycoprotein is expressed on type II pneumonocytes in alveoli and bronchiolar epithelial cells.
Renal Disease Overt renal disease is one of the most ominous features of DCSS. I60 Although little information is available, it is an impression that children may do better than adults in this regard. 4.32 ,47 In the Padua database, 9.4% of the children had renal involvement, and one developed renal crisis. 6 Medsger and colleagues l61 indicated that almost 50% of adult patients who developed renal disease did so within the first year after disease onset. As is the case in nephropathy from other causes, symptoms were usually absent initially. Proteinuria, the most common indicator of involvement of the kidneys, is often minimal, but it may be the only evidence of renal disease. Systemic hypertension occurs in up to one half of adult patients and is usually associated with proteinuria.162.163 The degree of hypertension ranges from mild or moderate in most patients to malignant hypertension in approXimately 25%. This complication often begins during the colder months of the year162 and may be heralded by the development of microangiopathic hemolytic anemia. 164 Onset is followed rapidly in most patients by death within a few weeks in the absence of intensive intervention. Renal or prerenal azotemia occurs in at least 25% of patients in the presence or absence of hypertension or proteinuria. 163 Renovascular Raynaud's phenomenon, demonstrated by decreased cortical blood flow, may be induced by immersion of the hands in cold water. 162 Even in the absence of angiographic evidence of vascular disease, xenon 133-demonstrated cortical blood flow may be impaired. 162 These reversible changes are mediated by the renin-angiotensin system, and plasma renin levels correlate with the presence of malignant hypertension.165
Central Nervous System Disease The most frequently described central nervous system (CNS) abnormality is cranial nerve involvement, especially of the sensory branch of the trigeminal nerve. 166-168 In contrast, peripheral neuropathies are uncommon 0.6%).169 A more subtle abnormality, diminished perception of Vibration, probably reflects the damping effect of cutaneous sclerosis on the transmission of the vibrations of a tuning fork l70 Clinical involvement of the CNS is usually a reflection of renal or pulmonary disease; however, cerebral arteritis has been described. 171
Sicca Syndrome (Sjogren's Syndrome) Xerostomia (Le., dry mouth) and keratoconjunctivitis sicca (i.e., dry eyes) are common in DCSS (see Chapter 21). Histologic evidence of salivary gland involvement was uniformly demonstrable in lip biopsies in a prospective study of Sjogren's syndrome in 17 adult patients with DCSS and 8 patients with LCSS.172 Xerostomia and sali-
452
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
vary gland enlargement were present in 84%. Scintigraphy of the salivary glands was abnormal in 88%, and sialography abnormal in 75%. Ocular symptoms of dryness or a foreign-body sensation occurred in 76%. Results of Schirmer's test were abnormal for 40%, and rose bengal staining of the cornea was positive in 55%.
Pathology An angiitis is regarded as the basic initial lesion with activated lymphocytes infiltrating around small blood vessels. There are increased numbers of T lymphocytes, plasma cells, and macrophages in the deep dermis and subcutaneous tissue and around small blood vessels, nerves, the pilosebaceous apparatus, and sweat glands. 53. m Marked hyalinization of blood vessel walls and proliferation of endothelium occur later. Raynaud's phenomenon, renal crisis, and pulmonary hypertension are all associated with a distinctive arteriosclerotic fibrotic lesion. 174 Another characteristic finding is mast cell hyperplasia in skin and viscera.17'i Hydrophilic glycosaminoglycan in the dermis may account in part for the accumulation of edema. 176 Later in the course, biopsies document homogenization of collagen fibers with loss of structural detail and an increased density and thickness of collagen deposition (Fig. 19-13A),75.177 With electron microscopy, the collagen appears embryonic with narrow fibrils and an immature cross-banding pattern (see Fig. 19-13B).178 The histologic characteristics of the skin in late disease include thinning of the epidermis, and loss of the rete pegs and atrophy of dermal appendages, often with a persistent inflammatory infiltrate of T lymphocytes. The
synovial membrane histologically resembles that of rheumatoid arthritis except for the abundance of fibrin and dense fibrosis.lOl.l79,lBO Biopsy specimens of muscle are abnormal in approximately one half of the patients. 181 The most prominent abnormalities are increased deposition of collagen and fat in interstitial perivascular sites of the perimysium and epimysium and focal, predominantly lymphocytic, perivascular infiltration. There is a relative loss of type II fibers. 1H2 Blood vessels are thickened and vessel lumens are narrowed. Immunofluorescence studies have demonstrated no abnormalities,IH2 Histopathologic changes in the vasa nervorum, neural dysfunction from fibrosis, and smooth muscle atrophy and fibrosis are similar throughout the gastrointestinal tract but are most prominent in the esophagus, where atrophic muscle is replaced by fibrous tissue. The smooth muscle of the lower two thirds of the esophagus is most commonly affected, but in some patients, striated muscle of the upper third may also be involved, The lamina propria and Auerbach plexus are infiltrated with mononuclear cells. Arterial walls are thickened, One half of patients in one necropsy series had evidence of myocardial fibrosis that was unrelated to coronary artery disease (Fig. 19-14),183 Other findings included contraction band necrosis (Le., myofibrillar degeneration) from transient ischemia in 31% (possibly the eqUivalent of Raynaud's phenomenon of the coronary arteries and a precursor to myocardial fibrosis). Necropsies in adults have demonstrated effusions or fibrous, fibrinous, and adhesive pericarditis in approximately 40%,184 a frequency similar to that detected by echocardiography.18> Convincing clinical evidence of pericarditis was present in only 3% to 16% of patients. l84 The main histologic abnormality in the lungs is diffuse alveolar, interstitial and peribronchial fibrosis, The thickened walls lead to a reduction of alveolar space (Le" compact sclerosis). Rupture of alveolar septae results in small areas of bullolls
• Figure It-13 A, The classic histopathologic features of the cutaneous disease are visible in this full-thickness section of skin from a patient with diffuse cutaneous systemic scleroderma.The epidermis is thin, and there is atrophy of the dermal appendages.The rete pegs are relatively obliterated (hematoxylin and eosin stain; magnification x 480). B, Electron microscopic studies indicate a relative reduction in the fiber size of newly synthesized collagen. Transverse sections of collagen fibers (left) are from the skin of a patient. Amarked variation in fiber size is apparent when compared with healthy skin (right). Many smaller collagen fibers are observed (alTOw) (normal diameter x 1200).The fine granular and whiskery material (GW) surrounding the sclerodermaI collagen probably represents mucopolysaccharides, and their visibility is enhanced by staining with ruthenium red, lead citrate and uranyl acetate (magnification x 38,610). (Courtesy of Dr. C. R. Wynne-Roberts.)
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
453
• FIpre 19-14 Fibrosis of the myocardium. H&E, X480.
• figure 19-15 Biopsy of a lung reveals striking fibrosis and disruption of the alveoli. H & E, x 480.
emphysema (Le., cystic sclerosis) (Fig. 19-15). Extensive bronchiolar hyperplasia, arteriolar endothelial proliferation, fibrous pleuritis, and pleural adhesions are also present. Young and Mark lA6 reported that 14 of 30 patients had moderate or marked abnormalities in the pulmonary vasculature at necropsy, and postulated that malignant pulmonary hypertension analogous to malignant renal hypertension was the cause of the rapidly progressive pulmonary failure that culminated in the death of 3 patients. The characteristic histopathologic change in the renal vasculature is concentric intimal proliferation of the interlobar and arcuate arteries, together with cortical infarcts and fibrinoid necrosis of the media. Vasculitis (other than the changes of malignant hypertension) is uncommon. The glomeruli exhibit a wide spectrum of abnormalities (Fig. 19-16), ranging from acute ischemic necrosis to thickening and sclerosis of the basement membrane. Swelling of the endothelial cells results in vascular narrowing. IS7 Deposition of immunoglobulin and complement in the renal vasculature has been reportedl87-189
but is generally sparse. On electron microscopy, intimal thickening of the vessels is associated with the presence of myointimal cells resembling those of smooth muscle, but with the capability to produce collagen and elastin. l90
• FIpre 19-16 Necropsy spedmen from a patient who died of renal failure and hypertension during the first few months of the disease. A, Virtual obliteration of the lumen of an arteriole by subintimal proliferation, thrombus formation, and mucoid hyperplasia of the media. B, Glomerulus from the same patient, showing fibrinoid necrosis and effacement of capillary loops without significant inflammatory cell infiltration. H & E, x 480.
Differential Diagnosis Proximal scleroderma is the most important characteristic of early disease. Raynaud's phenomenon is almost always present and accompanied by pronounced abnormalities of the nail fold capillaries. Esophageal dysmotility and abnormal pulmonary diffusion and function are often confirmed at onset. According to classification criteria of the American College of Rheumatology for adults, 191 definite DCSS requires the presence of the major criterion or two minor criteria (Table 19-7). This classification, published in 1980, was designed to be specific rather than sensitive to minimize false-positive
454
D:III
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
TABLE 19 7 Preliminary Criteria for the C1assifi< ation of Systemi( Stlerosis (Scleroderma)
Major Criterion
Proximal scleroderma: typical sclerodermatous skin changes (tightness, thickening, and nonpitting induration, excluding localized forms of scleroderma) involving areas proXimal to the metacarpophalangeal or metatarsophalangeal joints Minor CrIteria
Sclerodactyly: sclerodermatous skin changes limited to digits Digital pitting scars resulting from digital ischemia Bibasilar pulmonary fibrosis not attributable to primary lung disease From Subcommittee for Scleroderma Criteria of the American Rheumatism Association Diagnostic and Therapeutic Criteria Committee: Preliminary criteria for the classification of systemic sclerosis (scleroderma). Arthritis Rheum 23:581-590, 1980,
ascertainment. Subsequently, the widespread use of nail fold microscopy, more precise autoimmune serologic tests, and early detection of Raynaud's phenomenon in patients who, years later, developed DCSS, have raised the need for a more comprehensive classification. Leroy and Medsger proposed a new set of criteria 192 to identify patients with vascular abnormalities and serologic changes typical of scleroderma but who do not yet fulfill criteria for DCSS, or LCSS. Patients who exhibit Raynaud's phenomenon, and either nail fold capillary abnormalities or an antibody profile characteristic of DCSS or LCSS, are classified as having the limited form of scleroderma. Validation of new classification criteria for children is still in progress. Because DCSS usually involves internal organs, muscles and skin, the differential diagnosis includes many disorders such as juvenile dermatomyositis (see Chapter 18), mixed connective tissue disease, and other undifferentiated connective tissue diseases (see Chapter 21).193-t96 Chronic Graft-Versus-Host Disease Chronic GVHD is a complication of allogeneic bone marrow transplantation for the trealffient of marrow aplasia, leukemia or malignant diseases. GVHD results from the interaction between immunocompetent T lymphocytes from the donor and host cells bearing histocompatibility antigens that are recognized as for-
-, tl
TABLE 19 8
eign. The graft attempts to "reject" the host because, under the circumstances of the transplantation, the recipient is rendered immunologically incompetent by immunosuppression and x-irradiation. The resulting scleroderma-like disease may follow acute GVHD or occur de novo up to 100 days after transplantation. It is characterized by dermatitis, usually beginning with erythema of the face, palms, soles, and other regions that are distal in location, or is sometimes generalized involving only one side of the body in a harlequin-type syndrome. Hyperpigmentation and hypopigmentation follow. A hidebound skin and extreme tightening of the tendons, subcutaneum and periarticular structures may severely limit motion. Gastrointestinal disease with severe diarrhea, hepatitis and other visceral involvement is common. The salient features of chronic GVHD and DCSS are compared in Table 19-8. The histology in these two conditions is similar but not exactly the same.197 d-Penicillamine may be beneficial for some patients. Extracorporeal photopheresis after sensitization of host leukocytes by methylpsoralen, a procedure successfully used for the treatment of T-cell lymphoma, has therapeutic promise. 198 However, modifications of the chemotherapeutic preparation of the graft recipient may offer some hope of prevention of this complication. l99
Nephrogenic Fibrosing Dermopathy Scleroderma-like cutaneous fibrosis affecting the trunk and extremities, consisting of indurated plaques and papules, has been described in a number of patients, including children, after renal allograft transplantation" oo or institution of peritoneal or hemodialysis. 201 The face is usually spared. Systemic involvement is absent (as in scleromyxedema). On microscopic examination, there is a subtle proliferation of dermal fibroblasts in early lesions and florid proliferation of fibroblasts and dendritic cells in more advanced disease. Inflammatory infiltrates are usually absent. Collagen bundles with surrounding clefts are prominent, and dermal mucin and elastic fibers are variably increased. Electron microscopy confirms these findings, which in some cases resemble a sarcomatous process. Resolution after discontinuation of dialysis occurred in two of these patients.
Chemically Induced Scleroderma-like Disease Several chemicals have been implicated in the induction of scleroderma. 202 Polyvinyl chloride, initially used as an anesthetic agent,20j caused a scleroderma-like disease among workers. It is characterized by Raynaud's phenomenon; localized papular skin lesions, especially on fingers and hands, excluding the face and trunk; and osteolysis of the distal phalanges. Bleomycin, an antineoplastic agent, causes skin changes
Comparison of Systemic Sderoderma and Chronic Gr'lft- Versus Host Disease
System Affeded
Skin Raynaud's phenomenon Lungs Esophagus Small bowel Heart Kidney Keratoconjunctivitis sicca
Early DCSS (OAt)
Late DCSS (OAt)
GVHD (OAt)
n=13
n=36
n=12
100 100 77 77
8 69 15 15
100 92 96 86 92 61 75 69
100 42 58 17 0 42 8 50
DCSS, diffuse cutaneous systemic scleroderma; GVHD, graft-versus-host disease. Adapted from Clements PJ. Furst DE. Ho W, et al: Progressive systemic sclerosis-like disease follOWing bone marrow transplantation. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985. pp 376-381.
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
resembling scleroderma 204 and pulmonary fibrosis. 205 This syndrome is not accompanied by Raynaud's phenomenon and may improve on cessation of the drug. 206 Pentazocine, a nonnarcotic analgesic drug, has been reported to cause cutaneous scle'rosis with or without ulceration. 207 Predisposing factors may include diabetes mellitus and alcohol abuse. the toxic-oil syndrome, caused by ingestion of rapeseed cooking oil that contained unidentified contaminants, occurred in epidemic proportions in Spain in early 1981,208-210 affected approximately 20,000 persons, and resulted in at least 350 deaths. A report of 21 children indicated that complications may have been less severe in the younger group and that the ratio was closer to ~qual (2.5:1) than in adults (6:1).211 Onset of the disease was characterized by fever, eosinophilia, dyspnea caused by pulmonary edema, a pruritic rash, and malaise. Sclerodermatous skin lesiOns, alopecia, conjunctivitis sicca, Raynaud's phenomenon, myositis, neuropathy, joint contractures, dysphagia, and liver disease evolved over a period of months. Adjuvant disease, a systemic scleroderma-like condition, has followed cosmetic surgery involving injection of paraffin or silicone. m .m An inflammatory reaction in surrounding tissue occurs when silicone gel leaks from implants used for augmentation mammoplasty. A granulomatous reaction can also be demonstrated in regional lymph nodes. Silicone synovitis is well documented in patients after arthroplasty.214 A variety of atypical "connective tissue diseases," principally sclerodermalike conditions with chronic fatigue, myalgia, arthralgia, and arthritis, had been reported in women who had silicone breast implants. Causality had been based on extrapolation of epidentiologic data. 21S Recent reports216.217 have not demonstrated a causal relationship (U.S. Institute of Medicine, 1999). Pseudo~erodenmas
The term pseudoscleroderma describes a diverse group of disorders that are characterized by scleroderma-like fibrotic changes in the skin in association with other nonrheumatic diseases. This discussion is restricted to disorders of significance in the pediatric population.
Phenylketonuria A minority of children with phenylketonuria (Le., phenylalanine hyqroxylase deficiency) develop sclerodermatous skin lesions. 21 8-221 These lesions, which usually appear within the first year of life, are symmetric, poorly demarcated, and resemble morphea. They occur most frequently on the lower extremities and trunk. The lesion., may regress on introduction of a low-phenylalanine diet.222,m Although no differences in serum phenylalanine or tryptophan levels were found in children with phenylketonuria who did or did not have sclerodermatous changes, urinary excretion of 5-hydroxyindoleacetic acid, indoleacetic acid, and tryptamine was much higher in affected children. 222 The relationship of these biochemical abnormalities to the pathogenesis of the accompanying skin lesions or to that of scleroderma per se is unclear. The experimental use of a low-phenylalanine diet in patients with DC':;S produced inconclusive results. 222
Syndromes of Premature Aging Two rare autosomal recessive disorders accompanied by dwarfing, premature aging, and early death from atherosclerotic heart disease are associated with sclerodermatous skin changes. In progeria, the cutaneous changes usually develop before 1 year of age and are characterized by thickened, bound-down skin on the abdomen, flanks, proximal thighs, and upper buttockS.22+-226 During the second year of life, the skin becomes
455
thinner, subcutaneous vascularization is more evident, and alopecia and nail dystrophy develop. Werners syndrome most often presents in adolescence with generalized atrophy of muscle and subcutaneous tissue, graying of the hair, baldness, and scleroderma-like skin changes and ulcers involving the extremities.225.227.228 The histologic features of this disorder mimic those of scleroderma. Metastatic calcification may also develop.229
Localized Idiopathic Rbroses Several relatively rare disorders in children result in fibrosis of specific organs or structures. 23Q-232 Keloids are an obvious example. Retroperitoneal fibrosis usually occurs in the region of the sacral promontory and affects vital structures such as the great vessels and ureters. It is more common in males than in females and occurs in children and adults. The syndrome may be idiopathic or associated with administration of the serotonin inhibitor methysergide. Retractile mesenteritis, mediastinal fibrosis, fibrosing pericarditis, fibrosing carditis, and peritoneal fibrosis may represent similar disorders that have been related in some instances to administration of certain drugs, notably methysergide and some antihypertensives and anticonvulsants.233.234 Some variants of fibromatosis restricted to childhood are distinctive pathologically.230.2.35-237 Congenital torticollis or fibromatosis colli affects the lower sternomastoid muscle and is present at birth or shortly thereafter. It is associated with other anomalies such as congenital dislocations of the hip. Fibromatosis hyalinica multiplex is a morphologically distinctive type of familial multiplefibromatosis affecting children but not present at birth. Infantile digital fibromatosis affects predominantly the distal fingers or toes. A distinctive microscopic abnormality is the presence of eosinophilic cytoplasmic inclusions. Infantile myofibromatosis presents as solitary or multiple nodules limited to superficial soft tissues or associated with internal organ involvement. This disorder probably represents an inborn error of metabolism with possible autosomal dominant transmission. It is characterized microscopically by hyalinization of connective tissues of the skin, oral cavity, joint capsules, and bones. Microscopically, areas that resemble smooth muscle alternate with hemangioperieytoma-like foci with a more typical fibroblastic configuration. Central necrosis and intravascular growth may be present. Gardner's syndrome is a form of fibromatosis associated with multiple colonic polyps and osteomas. The fibrosis has a tendency to involve intra-abdontinal structures, such as the omentum and mesentery, or to occur after an operative procedure. Dupuytren s contracture is a nodular thickening of the palmar fascia and flexion contractures of the digits. Unassociated with disorders such as diabetes, it is rare in children. Lipogranulomatosis subcutanea of Rothmann-Makai produces scleroderma-like changes in the skin of the lower extremities with subcutaneous nodules. Morphea or linear scleroderma may be the initial diagnostic consideration. Systemic involvement is absent. The stiff-skin syndrome represents congenital scleroderma-like indentations of fascia, predominantly of the buttocks and thighs. Scleromyxedema is characterized by papular cutaneous lesions with induration of underlying subcutaneous tissues. The lesions occur predominantly on the hands, forearms, trunk, face, and neck. Histologic characteristics include a prominent fibrohistiocytic infiltrate and dense acid mucopolysaccharide deposits in the upper dermis. The disease in adults has been associated with monoclonal gammopathies.
Scleredema Scleredema, a nonsuppurative disorder that is primarily of historical interest, follows ~-hemolytic streptococcal infection and
456
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
is characterized by edematous induration of the face, neck, shoulders, thorax, and proximal extremities, but not the hands.238.239 Onset is characteristically insidious, and resolution spontaneously occurs after 6 to 12 months. Cardiac abnormalities suggesting the concurrence of acute rheumatic fever have been reported. Diagnosis is based on documentation of nonpitting, indurated edema or stiffness of the skin in the typical locations. Dysphagia may be present, but Raynaud's phenomenon and telangiectases are not. Histologically, the dermis is thickened; there are multiple fenestrations between swollen collagen bundles, a scant perivascular lymphocytic infiltrate, and minimal deposits of acid mucopolysaccharides within the fenestrations. Immunofluorescent staining is negative. Although some children with scleredema have poorly controlled insulindependent diabetes, the disorder is presumably distinct from diabetic cheiroarthropathy. Diabetic Chelroarthropathy Diabetic cheiroarthropathy, a syndrome of juvenile-onset diabetes mellitus, causes short stature and tightening of the skin and soft tissues, leading to contractures of the finger joints in children. 240 In one survey, 29% of diabetic children were found to have one or more flexion contractures (see Chapter 32). Porphyria Cutanea Tarda The development of scleroderma in adults with porphyria cutanea tarda has been reviewed.241.242 Plaque-like skin changes occurred predominantly on the face, neck, upper chest, and back. Some patients had features of other connective tissue diseases such as discoid lupus. There are no reports of this association in children.
Laboratory Examination Anemia, although uncommon, occurs in approximately one fourth of patients and is characteristic of the anemia of chronic disease or, less commonly, reflects vitamin B12 or folate deficiency resulting from chronic malabsorption. Microangiopathic hemolysis 164 or bleeding from mucosal telangiectases may also occur. Autoimmune hemolytic anemia is rare. 243 Leukocytosis is not prominent but correlates in degree with advanced visceral or muscle disease. Eosinophilia occurs in approximately 15% of patients. 244 Synovial fluid analysis was reported in one study to exhibit increased protein content and high numbers of polymorphonuclear leukocytes that had inclusions similar to those seen in rheumatoid arthritis. 245 Pericardial fluid has the characteristics of an exudate. 246 High-titers of ANAs are frequently identified; the predominant patterns on HEp-2 cell substrate are speckled and nucleolar. ANA seropositivity in the Padua database was 81%, a frequency lower than that reported in adults. 6 The prevalence of anti-Scl-70 (DNA-topoisomerase-l) was 30%, whereas anti-centromere antibodies (kinetochore) were less common than in adults (approximately 6%). In contrast, a large study of adults with DCSS indicated that 26% had anti-Scl-70 antibodies and 22% anticentromere antibodies. 247 No patient had reactivity to both antigens, an observation also confirmed by Kikuchi and Inagakj,248 Antibody to Scl-70, which occurred most
frequently in patients with DCSS, was associated with peripheral vascular disease, digital pitting, and pulmonary interstitial fibrosis. 249 Anti-centromere antibody occurred almost exclusively in patients with LCSS in association with calcinosis and telangiectases. Serologic and genetic markers help to predict particular complications. Patients with anti-topoisomerase-l autoantibodies or the HLA-DRS2a genotype are at increased risk for developing interstitial pulmonary fibrosis, irrespective of their apparent clinical subset. 250 In contrast, anti-RNA polymerase I or III antibodies are associated with renal involvement. 251 Anti-centromere antibodies in LCSS are an indicator of risk for isolated pulmonary hypertension and severe gastrointestinal involvement,252 and in children, they are a marker of Raynaud's phenomenon. 253 An association between the presence of antibody to Scl-70 and malignancy has been observed in adults. 254 Antibodies that are specific for a 70-kD mitochondrial antigen have been described in a small proportion of patients. 255 The associations of antibodies to the PM-Scl antigen have been reviewed. 256 Antineutrophil cytoplasmic antibodies have been reported with specificities to bactericidal or permeability protein and cathepSin G. 257
Cardiac Function Electrocardiographic abnormalities include 1st-degree heart block, right and left bundle branch block, premature atrial and ventricular contractions, nonspecific T-wave changes, and evidence of ventricular hypertrophy. 258 Disturbances of rhythm probably result from myocardial fibrosis or fibrosis of the sinus node and bundle branches. Clements and associates 259 identified paroxysmal supraventricular tachycardia in 13 of 46 patients with Holter monitoring, although 11 of the 13 had no cardiovascular symptoms. Ventricular tachycardia and premature atrial or ventricular contractions were less common. Thallium 201 radionuclide scans will often document abnormalities of myocardial perfusion, ventricular wall motion, chamber size, and left ventricular ejection fraction. 26o Echocardiographic abnormalities in addition to effusions include thickening of the left ventricular wall in 57% and diminished left ventricular compliance in 42%.261 Ultrasonic videodensitometric analysis has been introduced as an additional mechanism for evaluation. 262
Pulmonary Function Characteristic findings of involvement of the respiratory tract include a decrease in timed vital capacity and forced expiratory flow, an early decrease in diffusion, and an increase in functional residual volume.263.264 In one series, 11 of 15 children with DCSS had diminished pulmonary diffusion. 32 The two-dimensional echocardiogram is important in confirming early pulmonary hypertension by documentation of a dilated right ventricle with thickening of the ventricular wall and straightening of the septum. One-dimensional (M-mode) echocardiography is characterized by changes in the midsystolic movement of the pulmonary valve. Right heart catheterization provides definitive confirmation but is often unnecessary.
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
Steen and colleagues26s reported that only 38% of 77 adults with DCSS and 28% of 88 with LCSS had normal pulmonary function studies. Restrictive lung disease and isolated reduction of DLco were the most common abnormalities, occurring in 34 DCSS patients (18%) and 23 (26%) patients with LCSS. The earliest change was a decrease in the forced vital capacity with an FEY/FVC less than 70%. This abnormality was present in 8% of patients with diffuse disease and 16% of those with limited disease. Guttadauria and associates 266 also found a high prevalence of small airways disease (42%), usually in the absence of symptoms, chest radiographic changes, or other abnormalities of pulmonary function.
Renal Function Renal plasma flow is decreased in most patients, especially in the cortex, although normal glomerular filtration may be preserved by intrarenal shifts in blood flow. 162 Even in patients without clinical evidence of renal disease, plasma renin levels correlate with the degree of histologic abnormality of the renal arteries and arterioles. 267 Renal arteriography may document irregular arterial narrowing, tortuosity of the interlobular and arcuate arterioles, cortical hypoperfusion, and other changes of malignant hypertension. Kidney size is small to normal.
Radiologic Examination The most characteristic radiologic findings in the hands are a marked decrease in soft tissue and resorption of the tufts of the distal phalanges (acro-osteolysis) , particularly in patients with severe Raynaud's phenomenon (Fig. 19-17). Resorption of the distal tufts is particularly common in children. 32 ,268 Resorption may also occur in ribs, clavicles, distal radius and ulna, and other sites. 269 An incrtease in the thickness of the periodontal membrane results in radiolucent widening between the teeth and the jaw. 170 Periarticular or subcutaneous calcification, especially in the dominant hand, occurs in 15% to 25% of patients (see Figs. 19-8 and 19-9),32 Bony erosions can also develop at the distal interphalangeal and proximal interphalangeal joints. Involvement of the first carpometacarpal joint is particularly characteristic of DCSS. 271
457
Radiologic studies of the gastrointestinal tract often demonstrate characteristic abnormalities even in the absence of symptoms. A cine-esophagogram may document decreased or absent peristalsis in the lower part of the esophagus with distal dilatation and, frequently, a hiatal hernia with stricture and shortening of the esophagus (Fig. 19-18). The presence of air in the distal esophagus on the lateral chest radiograph suggests the diagnosis. Esophageal motility studies by manometry and pH probe monitoring of the distal esophagus for 12 to 24 hours provide more sensitive indicators of diminished lower sphincter tone and the presence of reflux. 272 The most frequent radiographic changes in the small bowel are dilatation of the second and third parts of the duodenum and the proximal jejunum (Fig. 19-19). Abnormalities in the colon are characterized by loss of colonic haustrations 273 and the presence of widemouthed diverticula or pseudosacculations on the antimesenteric border. Colonic transit is delayed. 148 Radiographic changes on chest x-ray films correlate poorly with pulmonary function. Bibasilar pulmonary fibrosis is one of the minor criteria for classification of DCSS (Fig. 19-20). It may be accompanied by rib notching and calcified pulmonary "granulomata" in LCSS. 265 High-resolution computed tomography (HRCT) may confirm pulmonary disease despite a normal chest radiograph. 274.275 In children, the most frequent HRCT findings are (in order) ground-glass opacification, subpleural micronodules, linear opacities, and honey combing. 276
Assessment of Disease Adlvlty and Severity Assessment of disease activity or severity is difficult. Regular follow-up and clinical review are the cornerstones of monitoring activity and progression. Serologic markers of activity have long been sought, and those which may be useful include soluble adhesion molecules such as sICAM-l, collagen propeptides,277 products of type I collagen breakdown,278 and immunologic markers such as s1L-2 receptor, neopterin, or vascular activation markers (e.g., E-selectin, thrombomodulin, von Willebrand factor) (Table 19-9).279 For organ-based complications such as pulmonary fibrosis, pulmonary hypertension, or renal involvement, objective assessment is easier. Sequential skin scores can be recorded. To provide a more global index of severity, a scoring system has been reported and is being validated. 280 Modified health assessment questionnaires have been developed in the United States281 and Europe. 282-ll;j They will undoubtedly be of considerable value, particularly because constitutional symptoms and functional impairment are among the most troublesome consequences of this disorder. The European Scleroderma Study Group has developed three different la-point indices of disease activity, one for scleroderma as a group, one for DCSS, and one for LCSS. 284 These assessments require further validation. No such tool exists for scleroderma in children.
• FIgIn 19-17 A, Radiograph of a boy with early resorption of the tufts of the distal phalanges (arrowheads). B, Magnified view of aao-osteolysis of the index finger (arrowhead).
Treatment No uniformly effective therapy is available. Management of DCSS presents one of the most difficult and frustrating
458
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
• Figure 19-18 Bariumcontrast examinations of the esophagus illustrate moderate dilatation and lack of a normal peristaltic pattern. A, Supine anteroposterior view. B, Lateral view.
challenges in all of rheumatology.285 Disease severity ranges from mild and stable to rapidly progressive and fatal. Management can be divided into three general areas: general supportive measures, therapy directed at controlling the underlying disease process (e.g., fibrosis, immunologic abnormalities, vasculopathy), and treatment of complications.
General Supportive Measures Supportive therapy is of utmost importance in managing a chronic, unpredictable, and potentially debilitating or fatal disease. Education of the child and parents should be undertaken early in an attempt to prevent unnecessary psychologic uncertainty and trauma. In general, "optimistic veracity" regarding complications, outcome, and treatment is appropriate. Patient support groups may be helpful and important, albeit difficult to assemble for such a rare disease. Patients should be instructed to avoid cold and trauma. Especially in cold climates, the family should keep the child warm by maintaining a satisfactory household temperature and by use of appropriate clothing, including well-insulated mittens (not gloves), boots, and a hat. The child should avoid excessive sun exposure and heat in the summer because of the susceptibility to hyperpigmentation of the skin and a relative inability to dissipate heat through sclerotic skin. General skin care should include avoidance of drying or irritating substances and daily application of lanolin or water-soluble cream as an emollient. The child should be encouraged to be as physically active as possible within the constraints of the disease. Active range of motion and gentle passive range of motion are essential to preserve maximal function. Dynamic splints may be necessary to treat or prevent contractures. Nonsteroidal anti-inflammatory drugs may relieve some of the musculoskeletal symptoms but may be detrimental to renal function. Subcutaneous calcifications, if ulcerating, may respond to incision and drainage.
Therapy of the Disease Process
• Figure 19-1. Upper gastrointestinal barium series with small bowel follow-through in a 3-year-old girl with dilatation of the jejunum and dosely approximated valvulae conniventes (the "dosed accordion" sign) due to thickening of the ileal mucosa.
Few of the drugs used as disease-modifying agents have undergone placebo-controlled evaluation, and the results from those that have are often disappointing. No drug has been of unequivocal benefit, and even if such data were available, therapeutic gain must be carefully balanced against toxicity and considered in the context of the natural history of the disorder. It is likely that organ-
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
459
• figure 19-20 A, Postero-
anterior radiograph of the chest illustrates a fine reticular pattern in both lower lobes. B, lateral view of the chest.
based strategies directed toward complications such as renal disease and pulmonary hypertension, fibrosis, or alveolitis, will have more immediate effect on morbidity and mortality than generalized disease-modifying strategies. Drugs can be grouped into two major categories: immunomodulatory therapy and antifibrotic therapy. Possible combinations of these approaches to treatment are suggested by the algorithm (see Fig. 19-23).
Immunomodulatory Therapy Cyclophosphamide Cyclophosphamide is used in treatment of pulmonary fibrosis. A number of retrospective series have suggested efficacy and delineated factors associated with responsiveness. 286 The toxicity of cyclophosphamide must be carefully balanced against efficacy; premature ovarian failure, opportunistic infections, and the pOSSibility of late secondary malignancies are important considerations in planning the route, dose, and duration of treatment. Methotrexate There has been some reluctance to use methotrexate because of concern that it might be deleterious in a disease exhibiting fibrosis and alveolitis. A report from the Netherlands 287 suggested clinical benefit documented by skin score and pulmonary function. Unfortunately, the result of a substantially larger study from North America was negative. 288 Mycophenolate Motetil In addition to efficacy in allograft rejection, the immunosuppressive agent mycophenolate
,"' I -
TABLE 19-9 Potential Laboratory Markers of Disease Aclivity in Systemic Scleroderma
BloIoglc Process
Serologic Markers
Immune Activity
Serum IL-2, IL-4, IL-6, IL-8 Serum IL-2R Serum TGF-p Serum E-selectin Plasma von Willebrand factor antigen Serum endothelin-I Serum procollagen III peptide breakdown fragments
Vascular damage Fibrosis
IL. inlerleukin; R, receplor; TGF, transforming growth factor.
mofetil has been advocated for scleroderma. The apparent safety and tolerability of this drug makes it a potential choice as an immunomodulatory drug for maintenance. 289 Its precise role needs to be defined by controlled clinical trials.
Glucocorticoids Glucocorticoids, the mainstay of treatment in many of the connective tissue diseases, are generally ineffective in DCSS, except for the early inflammatory stage of muscle involvement or in the edematous phase of the cutaneous disease. l46 Because case-control studies suggest that higher doses are associated with an increased frequency of renal crisis,290 use of a glucocorticoid should be accompanied by vigilant screening for signs of worsening renal function. Myocardial function may also be adversely affected. 291 Tumor Necrosis Factor Blockade In scleroderma, as in other autoimmune rheumatic diseases, there has been interest in the possible benefit of blocking TNF-a with neutralizing antibodies or soluble receptors. Because TNF-a antagonizes a number of profibrotic cytokines, including TGF-~l, it is uncertain whether its blockade would be wholly beneficial. A pilot study of 10 patients with early disease suggested that treatment with etanercept was well tolerated, although conclusions about efficacy would be premature.292 Cyclosporine Cyclosporine (or tacrolimus) has attractive therapeutic properties, especially for patients in whom there is evidence of marked immunologic activation. Open studies have suggested benefit for skin and visceral manifestations 293-m and for serum markers of collagen biosynthesis. 296 However, there remain major concerns about nephrotoxicity.297 Immunoablation with Autologous Peripheral Stem Cell Rescue One of the most aggressive approaches to therapy is immunoablation followed by reconstitution with autologous hemopoietic stem cell transplantation (HSCT).298 The rationale for this therapy is similar to that in other autoimmune diseases. If scleroderma is driven by an autoimmune process, ablation of self-reactive lymphocyte clones may block pathogenesis. If the immune system is reconstituted in the presence of the neoantigens responsible for autoimmunity, tolerance will be reestab-
460
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
lished. 299 However, even if such tolerization does not occur, the intensive immunosuppression may be directly beneficial. A multicenter study reported that HSCT improved the skin score for 69% of patients, did not affect lung function, but halted pulmonary hypertension. 30o However, disease progression occurred in 19%, and 17% died of complications related to the procedure. Because of this high mortality rate, HSCT must be carefully considered for patients,300·301 and unfortunately, it may only be rational therapy early in disease course (3 years or less from the first non-Raynaud's sign or symptom) before unmodifiable damage has resulted (e.g., fibrosis).
AntifibroticTherapies o-Penicillamine and Colchicine o-Penicillamine has traditionally been the drug most commonly used to treat DCSS in children and adults, although published results have been inconsistent,302-304 Unfortunately, most studies have been retrospective and poorly controlled. 302 A rigorously designed, double-blind, controlled trial demonstrated no difference between high-dose 050 to 1000 mg daily) and low-dose (125 mg on alternate days) regimens, providing no justification for using high doses. 304 Some investigators interpreted this study to indicate that o-penicillamine is ineffective,305 whereas others, considering the absence of a true placebo group, judged that a low dose was still effective. 306 There have been several reports that longterm colchicine administration (up to 0.5 mg four times a day in adults) resulted in softening of the skin.307.308 Sustained improvement in visceral disease was inconsistent, and it is not certain or demonstrated in controlled studies that o-penicillamine or colchicine is effective. Interferons Interferons (a, ~, or y) exert pleiotropic effects on fibroblasts in vitro, including downregulation of extracellular matrix gene expression (see Table 19-3). This understanding led to the hope that the interferons might have antifibrotic activity in vivo. A placebo-controlled trial of interferon-a for early DCSS demonstrated no benefit for skin sclerosis or pulmonary function. 309 Another open study suggested modest benefit,3lO Whether this agent will ultimately have any place in treatment remains uncertain. Recombinant Human Relaxin Another possible therapeutic approach is recombinant human relaxin, an antifibrotic hormone normally present in significant amounts only in pregnant woman. In vitro studies suggested that relaxin reduces synthesis of type I collagen by scleroderma fibroblasts.·m Unfortunately, final results from a large multicenter study did not confirm the benefit that had been reported in the earlier tria1.3 12
Therapy for Specific Complications Raynaud's Phenomenon Specific treatment of Raynaud's phenomenon may be necessary, although difficult, in addition to avoidance of precipitating circumstances such as cold or emotional stress. Pharmacologic side effects are often dose limiting,
responses to individual vasodilators are idiosyncratic, and substantial placebo responses and lack of mechanisms for objective assessment, confound therapeutic trials. The most commonly used vasodilator agents are probably the calcium channel blockers. Nifedipine is the most Widely recommended agent. It has been well tolerated in several controlled trials, has reduced the frequency and severity of Raynaud's phenomenon, and has promoted healing of cutaneous ischemic ulcers ..m.. .316 The drug is started in a low dose at bedtime. Full dosage in a 50-kg patient (approximately 10 mg, two to four times each day) is then achieved gradually to avoid precipitating postural hypotension. Some vasoactive agents may have additional disease modifying potential, such as those that influence the renin-angiotensin pathway (e.g., captopril). There is preliminary evidence that the selective angiotensin receptor antagonist losartan may decrease the severity of Raynaud's signs and symptoms and beneficially modulate levels of procollagen peptides and circulating isoforms of cell surface adhesion molecules. 317 Other agents include drugs that inhibit or suppress the sympathetic nervous system, thereby indirectly promoting vasodilation, and those that act directly on the smooth muscle of the vessel wall, such as reserpine, methyldopa, and ketanserin. 318- 323 One drug may be effective in one patient, whereas a different agent is effective in another. It is therefore worth trying several, one at a time, until the desired effect is obtained (Table 19-10). Griseofulvin has been beneficial in a few resistant patients, as have surgical sympathectomy and prostaglandin E1 infusions. 324-3 28 Intermittent infusions of prostacyclin or its analogues have been demonstrated to be effective329 but remain inconvenient and costly. Iloprost has been reported to be safe and effective in treatment of ischemic digits in children with DCSS and other connective tissue diseases. 33o It may be combined with another agent for enhanced therapeutic effect. Orally active formulations of prostacyclin or its analogues are an attractive alternative, but unfortunately, two large studies from Europe 331 and North America 332 have failed to demonstrate efficacy. Phenoxybenzamine is effective in doses of 10 to 40 mg/day, divided into three or four doses, although depression may be a significant side effect. 32.1H> Treatment should be initiated with a small dose (10 mg) at bedtime and increased gradually. Prazosin333 and locally applied nitroglycerin pasteJ34 may also be helpful. Talpos and colleagues335 and Winkelmann and coworkers 336 have employed serial plasmapheresis with good effect in some patients but with none in others. Biofeedback has also been used as the primary mode of therapy.337 Sympathectomy results in short-term benefit at best and is indicated only in the management of gangrene or intractable pain in the digits, and then only if temporary stellate ganglion block produces a beneficial result.
Renal Disease Until recently, prognosis for renal crisis was uniformly dismal. Immediate and effective lowering of the blood pressure in patients with malignant hypertension is mandatory. Any sudden change in plasma volume should be avoided because marked reductions in renal blood
C HAP T E R .~
,_II
IABLE 19 10
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
461
Plldrmacologic Agenls For Trealment of Raynaud's Phenomenon
~
References
Adlon
Drugs Aftectlng the Sympathetic Nl!IVOUS System Tolazoline Phenoxybenzantine Prazosin Reserpine Methyldopa Guanethidine
Blocks a-adrenergic receptors Blocks a-adrenergic receptors Blocks a-1 receptors Depletes norepinephrine from sympathetic nerves; IV administration results in short-lived vasodilation Central action Interferes with norepinephrine release at sympathetic neuroeffector junctions
369 370 333,370 318, 320, 369
Angiotensin converting enzyme inhibitor Slow calcium-channel blocker Vasodilator and platelet inhibitor Vasodilator Vasodilator Antagonizes vasoconstrictor effects of 5-hydroxytryptamine Vasodilator
321 313-316 325 326 334 322, 323 324, 327
319 371
DIrred-ActllllI Drugs C:ilptopril Nlfedipine Prostaglandin E, Prostaglandin I, Glyceryl trinitrate (topical) Ketanserin Griseofulvin
flow may precipitate acute clinical deterioration. The introduction of angiotensin-converting enzyme inhibitors (e.g., captopril) brought about a remarkable improvement in the outlook for prevention of vascular damage, effective long-term control of blood pressure, and stabiliz~tion of renal function. 338-341 It has also been observed anecdotally that systemic disease, especially cutaneous involvement, improved concomitantly in some patients. In cases of irreversible renal failure or uncontrollable hypertension, some success has followed the use of hemodialysis with or without bilateral nephrectomy and transplantation. 342 Dialysis may lead to improvement in the cutaneous abnormalities.
Pulmonary Disease Pulmonary complications are very serious, and there may be no effective long-term therapeutic approach to fibrosing alveolitis or primary pulmonary hypertension. Alveolitis is predominant early, and later progresses to fibrosis. Generally, cyclophosphamide is recommended when there is evidence of active alveolitis-usually determined by a ground-glass HRCT scan or by the presence of neutrophils in bronchoalveolar lavage. Following the experience in adults, it is common practice to combine cyclophosphamide (monthly intravenous infusions of 500 to 750 mg/m 2) with prednisone (0.3 to 0.5 mg/kg/day). Reassessments every 3 to 6 months are used to follow progress. Treatment is generally continued for at least 6 to 9 months. Controlled trials comparing cyclophosphamide with placebo are underway, and results of one open study were encouraging. 343 One of the most lethal complications is pulmonary hypertension (PHT), which can occur in the context of established interstitial fibrosis or without it in LCSS. It is accepted practice to screen all patients for evidence of PHT with a Doppler echocardiogram, together with pulmonary function tests for evidence of a reduction in diffusing capacity in the presence of normal spirometry. Continuous prostacyclin infusions (or analogues such as
epoprostenop44) are effective for primary PHT345 and possibly for pulmonary hypertension associated with scleroderma. 346 During the past decade, targeting mediators of immune or vasoactive reactions has been an innovative step in the treatment of connective tissue diseases. Endothelin-l, a potent vasoconstrictor and smooth muscle mitogen, is a possible target in patients with primary or secondary PHT. An endothelin-l receptor antagonist, bosentan, was demonstrated to be safe and effective in the treatment of PHT.347 It significantly improved exercise capacity and increased the time to clinical worsening in patients with severe disease. The oral formulation and the potential use of this agent for other vascular complications represent important arguments for its potential value in pediatric patients with DCSS. A combination of sildenafil and inhaled iloprost has been recommended for adults. 348
Gastrointestinal Disease Few studies in children address the most effective management for gastrointestinal disease. Except for symptomatic approaches, definitive therapy is materially lacking. Treatment of erosive esophagitis often is characterized by considerable delay in healing with the standard approaches of small, more frequent meals, with the last well before bedtime; antisecretory and promotility agents; and elevation of the head of the bed. Acid reflux and esophageal hypomotility are also factors leading to the development of pulmonary fibrosis. 349 Arteriovenous ectasia of the stomach may require multiple attempts at argon plasma coagulation. Malabsorption is difficult to manage. Diarrhea and bloating are most often caused by bacterial overgrowth and are treated by rotating antibiotics, prokinetics, and dietary and pancreatic supplements. Hyperalimentation may be necessary but has not been demonstrated to be a wise long-term choice. Surgical approaches in children have not been addressed except in case reports.
462
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
Course of the Disease and Prognosis
The most common causes of death in children are related to involvement of the cardiac, renal, and pulmonary systems (Table 19-12). Gastrointestinal complications and inanition may also become severe. Cardiac arrhythmias may result from myocardial fibrosis. Congestive heart failure is often a terminal event. Pulmonary interstitial disease and vascular lesions are probably universal, even if not clinically evident. Renal failure or acute hypertensive encephalopathy supervenes as a potentially fatal outcome in a few children. At least in adults, this event seems more likely to occur early in the course of the disease. Survivorship has not been determined in any large series of children because of the rarity of this disease. The age-specific mortality rate in one epidemiologic study for the 0- to 14-year age group was 0.04 per million person-years. 35 ! In a multinational surveY,352 8 of 135 patients died, but overall results were otherwise favorable. Therefore, an optimistic but realistic attitude should be taken in discussions with parents. Pulmonary hypertension has been associated with the deaths of at least two children. 183 ,353 As children with DCSS live into adulthood, complications associated with pregnancy become a concern.354.355
The systemic character of DCSS cannot be stressed too strongly, because the ultimate prognosis of the child depends primarily on the extent and nature of visceral involvement. The outcome has been poor but may be improving. Skin tightness and joint contractures inevitably lead to severe disability in some patients (Fig. 19-21).350 It is a curious but often-repeated observation that the skin may eventually soften years after onset. Progressive gastrointestinal involvement is typical, however, starting with the esophagus and proceeding distally, although the disease may stabilize in some patients for long periods (Table 19-11).
Mortality rates for adults were significantly increased in a study from Denmark (standardized mortality ratio [SMR] = 4.5; 95% CI: 3.5-5.7) and even more in patients younger than 35 years (SMR =13; 95% CI: 2.7-37).356 This study and another from Spain357 identified the extent of cutaneous sclerosis as an important determining factor in prognosis. Another report cited a 7-year survival rate from diagnosis of 72.5% for black women and 77.6% for white women. 21 Mortality seemingly increases throughout life and is higher for males than females and for nonwhites than whites. 23 Studies of patients of all ages demonstrated mean survival rates of 70% to 94% at 1 year, 34% to 73% at 5 years, and 35°!tl to 74% at 10 years. 161 Cardiac and renal disease were the most common con-
• RlIIIre 19-21 Hands of a 27-year-old woman with diffuse cutaneous systemic sderoderma that began in childhood.There was essentially no movement possible in these fingers because of joint contractures. Notice the extensive telangiectases (atrows).
1=..
lABlE 19 11
Organ Systelll Involvelllent During the Course 01 Systemic Sderodernld in Children
No. with InvolvemenVNo. Observed Organ System Skin Subcutaneous calcification Ulcerations Telangiectases Pigmentation Digital arteries (Raynaud's phenomenon) Musculoskeletal system Contractures Resorption of digital tufts Muscle weakness Muscle atrophy Gastrointestinal tract Abnormal esophageal motility Dilatation of duodenum Colonic sacculations Lungs Abnormal diffusion Abnormal vital capacity Heart Cardiomegaly Electrocardiographic abnormalities Congestive heart failure
Cassidy et aln
0/0
Martini et all
0/0
4/15 9/15 4/15 3/15 11/15
27 60 27 20 73
24/120 48/118
20 41
104/126
83
11/15 9/11 6/15 6/15
73 82 40 40
86/124 28/122
69 23
11115 1/15 3/5
73 7 60
40/120
33
11/15 10/15
73 67
36/74 56/94
49 60
2/15 4/15 2/15
13 27 13
5/124 8/124
4 6
C HAP T E R
!~..
I ABLE 19-12
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
463
Causes of Death in Children with Scleroderma
Sex Kornreich et aF'
Age at Onset (yr)
Disease Duration
4
10 yr 9 yr 23 mo 15 mo 22 mo 5 mo 6mo 10 yr 9 yr 2 yr 1 yr 2 yr
M F M
6 7 10 10 15 12 11 8
F F F F F F M F M
Cassidy et aP2
Bulkley 183 Suarez-Almazor et al4'
13 16 10
triblltors to mortality. Outcomes of 48 adults with "early" scleroderma indicated that sUlVivorship was 92% at 1 year, 75% at 3 years, and 68% at 5 years. 358 In another report, 20-year mortality rates for patients with renal involvement was 60%, compared with 10% for those without renal disease. 162 An overall10-year sUlVival rate close to 9Q01o was reported for a group of 106 patients from Italy who were predominantly female. 19 Median sUlVival in the Detroit tricounty area was approximately 11 years. 23
UNITED CUTANEOUS SYSTEMIC SCLERODERMA Definition LCSS is the designation for patients previously classified as having the CREST syndrome. Winterbauer359 first described this syndrome as a variant of systemic scleroderma. Very few instances of LCSS in children have been reported (Table 19-13).6.45--47 Whether it is a relatively mild form of DCSS or an entirely separate, although related, disorder is uncertain. 11.360 The combination of scl~roderma and calcinosis was designated as acrosclein the older literature or has been referred to as the rosis I 1btbierge-Weissenbach syndrome. 361
Epidemiology Overall, LCSS accounts for approximately one third to one half of the adult patients with scleroderma. 362 Limited
II
TABL E 19-13
Cause of Death Cerebral hemorrhage due to thrombocytopenia Cardiac failure Renal failure Cardiac failure Pulmonary emboli Cardiac failure Cardiac failure Cardiac failure Central nervous system disease, hypertension Pulmonary hypertension Pulmonary hypertension Cardiac failure and pulmonary hypertension
disease is more common among women and tends to occur at an earlier age than DCSS. A long interval between the onset of Raynaud's phenomenon and diagnostic skin changes is characteristic.
Clinical Manifestations Calcinosis is usually more severe in patients with LCSS than in DCSS (Fig. 19-22), Raynaud's phenomenon is more frequently complicated by digital ulceration and gangrene, and telangiectases are more widespread. 359 LCSS is by no means a mild disease, however, and severe systemic involvement, especially pulmonary fibrosis and hypertension, occurs, although renal disease is less frequent than in DCSS.
Diagnosis Cutaneous sclerosis is restricted to the distal segments of the digits, and telangiectases, Raynaud's phenomenon and calcinosis are prominent (Table 19-14). Isolated proximal scleroderma supports a diagnosis of DCSS rather than limited disease. In other ways, however, these two syndromes closely resemble each other, and clinical separation of these disorders may be entirely artifiCial. Incomplete forms (e.g., CRST, CRT) merge with diffuse disease and make differentiation of variants of systemic scleroderma difficult or impossible for a number of patients.
Limited Cutaneous Systemic Scleroderma (CREST Syndrome) and Variants in Children
Study Larregue et aI4S Burge et a1 46 Suarez-Almazor et al 47 Martini et al6
Sex F
M F F F F
M F F F
M
Age at Onset (yr) 6 6 10 6 10 8 10 7 9 7 7
Olnlcal Features CRST CREST RST CR RST CRS RST RE ES RE CR
C, calcinosis; E, esophageal dysmotility; R, Raynaud's phenomenon; S, sclerodactyly; T, telangiectases.
464
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
tromere antibody is associated with diseases other than LCSS,364 notably with primary biliary cirrhosis; occasionally with DCSS, Sjogren's syndrome, or isolated Raynaud's phenomenon; and rarely with rheumatoid arthritis, systemic lupus erythematosus, and other connective tissue diseases. Other ANAs (e.g., anti-ssDNA, anti-RNP) occasionally may be present. 365 Elevated levels of soluble CD31 were reported in patients with a relatively early age at onset and a lower frequency and severity of pulmonary fibrosis. Because this marker is associated with an anti-inflammatory effect by inhibiting transendothelial migration of leukocytes, it could represent a protective factor for the development of cutaneous and pulmonary fibrosis. 366
Treatment
• Figure 19-ZZ Striking subcutaneous caldfication with extrusion of calcareous material.
Course of the Disease and Prognosis
Immunologic Charaderlstlcs Antibody to centromere has been described as the serologic hallmark of LCSS, and its discovery historically supported the rationale for clinical differentiation of the CREST syndrome from DCSS.360.363 This antibody specificity is directed at the kinetochore of components of the mitotic spindle. It is evident, however, that anticen-
~. -
The management of children with LCSS is not materially different from that for diffuse disease, modified by each patient's specific organ involvement and severity (Figure 19-23).
It was initially believed that patients with this variant of scleroderma had a more benign course and lower mortality rate than those with DCSS, but this distinction has not been entirely substantiated. The mortality rate for LCSS, although somewhat less than in DCSS, is substantial with a lO-year survival rate of approximately 75% in adults. 362
TABLE 19 14 (olllp,uholl of Dilfll~e (1I1
Characteristic
DCSS
LCSS
EF
Distal sclerosis Proximal sclerosis Raynaud's phenomenon Telangiectases Nail fold capillaropathy Calcinosis Esophageal disease Pulmonary disease Cardiac disease Renal disease Muscle disease Arthritis Central nervous system disease Anti-dsDNA antibody Anti-nucleolar antibody Anti-centromere antibody Anti-Sm antibody Anti-RNP low titer Anti-RNP high titer Thrombocytopenia Peripheral eosinophilia
+++ ++++ ++++ +++ +++ ++ +++ ++++ ++++ +++ ++ + +
++++
+++ +
++ +
++++ ++++ +++ ++++ +++ +++ + +
+ +
MCID
++ + ++++ + ++ + ++ ++ + + +++ +++ + +
JDM
SLE
+
++
+++ ++
+
+
+ ++ +++ + ++ +++ +++
±
++++ +
+ ++++ + + ++++ +
++ ++ ++
++++
DCSS, diffuse cutaneous systemic scleroderma; EF, eosinophilic fasciitis; JOM, juvenile dermatomyositis; LCSS, limited cutaneous systemic scleroderma; MCTO. connective tissue disease; SLE. systemic lupus erythematosus; -. rare or absent; +, up to 25%; ++, up to 50%; +++, up to 75%; ++++. up to 100%.
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
465
Systemic scleroderma
Disease-modifying therapy Vasodilator (nifedipine, nicardipine, losartan)
Antifibrotic (? bosentan)
Risk stratification based on stage, subset, autoantibody profile
Screening for organ based complications
Organ-based treatments Gastroesophageal reflux (Iansoprazole)
Pulmonary hypertension (warfarin, i1oprost, bosentan)
Midgut disease (antibiotics)
Renal insufficiency (quinapril)
Myositis (glucocorticoid, MTX)
Hypertensive crisis (angiotensin converting enzyme inhibitor)
Fibrosing alveolitis (cyclophosphamide, glucocorticoid)
• FIgure 19-23 Treatment algorithm for systemic sclerodermas. ATG, antithymocyte globulin; CPM, cyclophosphamide; MFM, mycophenolate mofetil; MlX, methotrexate.
REFERENCES 1. Lewin G, Heller]: Die Sclerodermie. Berlin, August Hirschwald, 1895. 2. Goodman H: A case of scleroderma diffusa in a girl, nine years of age, with a review of the literature. .1 Cutan Pathol 36: 210, 1918. 3. faffe MO, Winkelmann RK: Generalized scleroderma in children. 'Acrosclerotic type. Arch Dermatol 83: 402, 1961. 4. !Cass H, Hanson V, Patrick J: Scleroderma in childhood. .1 Pediatr 68: 243, 1966. 5. Tuffanelli DL, Winkelmann RK: Systemic scleroderma. A clinical study of 727 cases. Arch Dermatol 84: 359, 1961. 6. Martini G, Foeldvari I, Russo R. et al: Systemic scleroderma syndromes in children: clinical and immunological characteristics of 181 patients. Arthritis Rheum 48(Suppl): S512, 2003. 7. Zulian F, De Oliveira SKF, Lehman TIl, et aI: Juvenile localized sclerodemla: clinical epidemiological features of 688 patients. Arthritis Rheum 48: S649, 2003. 8. Kraus EJ: Zur pathogenese der diffusen Sklerodermie. Zugleich ein Beitrag zur Pathologie der Epithelkorperchen. Virchows Archiv (Pathol Anat) 253: 710, 1924. 9. Matsui S: Anatomie pathologique et pathogenic de la scleropdermie generalisee. Presse Med 32: 142, 1924. 10. o'Angelo WA, Fries .IF, Masi AT, et aI: PathologiC observations in systemic sclerosis (scleroderma). A study of fifty-eight autopsy cases and fifty-eight matched controls. Am.l Med 46: 428--440, 1969. 11. Rodnan GP: When is scleroderma not scleroderma? The differential diagnosl< of progressive systemic sclerosis. Bull Rheum Dis 31: 7-10, 1981. 12. Poormoghim H, Lucas M, Fertig N, et al: Systemic sclerosis sine scleroderma: demographic, clinical, and serologic features and survival in forty-eight patients. Arthritis Rheum 43: 444-451, 2000. 13. Slobodin G, Rosner I, Rozenbaum M, et al: Systemic sclerosis sine scleroderma: is it always the same disease? Report of three patients and discussion. Rheumatol Int 22: 170-172, 2002. 14. Kurland LT, Hauser WA, Ferguson RH, et al: Epidemiologic features of diffuse connective tissue disorders in Rochester, Minn., 1951 through 1967, with special reference to systemic lupus erythematosus. Mayo Clin Proc 44: 649-663. 1969.
15. Caperton EM, Hathaway DE: Scleroderma with eosinophilia and hypergammaglobulinemia: the Shulman syndrome. Arthritl< Rheum 18: 391, 1975. 16. Medsger TA .Ir, Masi AT: Epidemiology of progressive systemic sclerosis. CUn Rheum Dis 5: 15, 1979. 17. Shinkai H: Epidemlology of progressive systemic sclerosis in Japan. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 79. 18. Asboe-Hansen G: Epidemiology of progressive systemic sclerosis in Denmark. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 78. 19. Giordano M, Valentini G, Ara M, et al: Epidemiology of progressive systemic sclerosis in Italy. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York. Gower Medical, 1985, pp 72-77. 20. Barnett A.I: Epidemiology of systemic scleroSis (scleroderma) in Australia. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 82. 21. Laing T.I, Gillespie BW. Toth MB, et al: Racial differences in scleroderma among women in Michigan. Arthritis Rheum 40: 734-742, 1997. 22. Mayes MD: Scleroderma epidemiology. Rheum Dis Clin North Am 29: 239-254, 2003. 23. Mayes MD, Lacey .TV .Ir, Beebe-Dimmer .I, et al: Prevalence, incidence, survival, and disease characteristics of systemic sclerosis in a large US population. Arthritis Rheum 48: 2246-2255. 2003. 24. Tuffanelli DL, LaPerriere R: Connective tissue diseases. Pediatr Clin North Am 18: 925-951, 1971. 25. Medsger TA .Ir, Masi AT: Epidemiology of systemic sclerosis (scleroderma). Ann Intern Med 74: 714-721, 1971. 26. Hanson V: Dermatomyositis, scleroderma, and polyarteritis nodosa. Clin Rheum Dis 2: 445, 1976. 27. Kornreich HK, King KK, Bernstein BH, et al: Scleroderma in childhood. Arthritis Rheum 20: 343-350, 1977. 28. Medsger TA .Ir: Epidemiology of systemic sclerosis. Clin Dermatol 12: 207-216, 1994. 29. Emery H: Pediatric scleroderma. Semin Cutan Med Surg 17: 41-47, 1998. 30. Black CM: Scleroderma in children. Adv Exp Med BioI 455: 35-48, 1999.
466
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
31. Sato S, Ishida W, Takeham K: A case of juvenile systemic sclerosis with disease onset at six months old, Clin Rheumatol 22: 162-163, 2003, 32, Cassidy JT, Sullivan DB, Dabich L, et al: Scleroderma in children, Arthritis Rheum 20: 351-354, 1977, 33, Foeldvari I: Scleroderma in children, Curr Opin Rheumatol 14: 699--703, 2002, 34, Murray KJ, Laxer RM: Scleroderma in children and adolescents, Rheum Dis Clin North Am 28: 603-624, 2002, 35, Kennedy WP: Cardiac death from progressive systemic scleroderma in a child, Can Med Assoc J 90: 33, 1964, 36, Mukherjee SK, Lahiri K, Sen MK: Scleroderma in a boy of nine, J Indian Med Assoc 47: 132-135, 1966, 37, Velayos EE, Cohen BS: Progressive systemic sclerosis, Diagnosis at the age of 4 years, Am J Dis Child 123: 57-60, 1972. 38, Szymanska-Jagiello W, Rondio H, Jakubowska K: Changes in the locomotor system in progressive systemic sclerosis in children, Mater Med Pol 4: 201-209, 1972. 39, Goel KM, Shanks RA: Scleroderma in childhood, Report of 5 cases, Arch Dis Child 49: 861-866, 1974, 40, Ansell BM, Nasseh GA, Bywaters EG: Scleroderma in childhood, Ann Rheum Dis 35: 189--197, 1976, 4I. Gray RG, Altman RO: Progressive systemic sclerosis in a family: case report of a mother and son and review of the literature, Arthritis Rheum 20: 35-41, 1977, 42, Schlesinger M, Schaller JG: Progressive systemic sclerosis of childhood, Arthritis Rheum 24(SupplJ: S104, 1981. 43. Girouard M, Pare C, Camerlain M: Juvenile scleroderma, Union Med Can 111: 546--551, 1982, 44, Spencer-Green G, Schlesinger M, Bove KE, et ai: Nailfold capillary abnormalities in childhood rheumatic diseases, J Pediatr 102: 341-346, 1983, 45, Larregue M, Canuel C, Bazex J, et al: [Systemic scleroderma in children, Apropos of 5 cases, A review of the literature], Ann Dermatol Venereol 110: 317-326, 1983, 46, Burge SM, Ryan TJ, Dawber RP: Juvenile onset systemic sclerosis, J R Soc Med 77: 793-794, 1984, 47, Suarez-Almazor ME, Catoggio LJ, Maldonado-Cocco JA, et al: Juvenile progressive systemic sclerosis: clinical and serologic findings, Arthritis Rheum 28: 699-702, 1985, 48, Lababidi HM, Nasr FW, Khatib Z: Juvenile progressive systemic sclerosis: report of five cases, J Rheumatol 18: 885-888, 1991. 49, Martinez-Cordero E, Fonseca MC, Aquilar Leon DE, et al: Juvenile systemic sclerosis, J Rheumatol 20: 405-407, 1993, 50, Vancheeswaran R, Black CM, David J, et al: Childhood-onset scleroderma: is it different from adult-onset disease, Arthritis Rheum 39: 1041-1049, 1996. 51. Steen YD, Oddis CV, Conte CG, et al: Incidence of systemic sclerosis in Allegheny County, Pennsylvania, A twenty-year study of hospital-diagnosed cases, 1963-1982, Arthritis Rheum 40: 441-445, 1997, 52, Jimenez SA, Derk CT: Following the molecular pathways toward an understanding of the pathogenesis of systemic sclerosis, Ann Intern Med 140: 37-50, 2004. 53, Fleischmajer R, Perlish JS, Reeves JR: Cellular infiltrates in scleroderma skin, Arthritis Rheum 20: 975-984, 1977, 54, Roumm AD, Whiteside n, Medsger TA Jr, et al: Lymphocytes in the skin of patients with progressive systemic sclerosis, Quantification, subtyping, and clinical correlations, Arthritis Rheum 27: 645-653, 1984, 55, Abraham OJ, Shiwen X, Black CM, et al: Tumor necrosis factor alpha suppresses the induction of connective tissue growth factor by transforming growth factor-beta in normal and scleroderma fibroblast~, J Bioi Chern 275: 15220-15225, 2000, 56, Varga J, Rosenbloom J, Jimenez SA: Transforming growth factor beta (TGF beta) causes a persistent increase in steady-state amounts of type I and type III collagen and fibronectin mRNAs in normal human dermal fibroblasts, Biochem J 247: 597-604, 1987, 57, Kikuchi K, Kadono T, Furue M, et al: Tissue inhibitor of metalloproteinase I (TIMP-I) may be an aUlocrine growth factor in sclerodenna fibroblasts, J Invest Dermatol 108: 281-284, 1997, 58, Ihn H, Yamane K, Kubo M, et al: Blockade of endogenous tmnsforming growth factor beta signaling prevents up-regulated collagen synthesis in scleroderma fibroblasts: association with increased expression of transforming growth factor beta receptors, Arthritis Rheum 44: 474-480, 2001. 59, Kubo M, Ihn H, Yamane K, et al: Up-regulated expression of transforming growth factor beta receptors in dermal fibroblasts in skin sections from patients with localized scleroderma, Arthritis Rheum 44: 731-734, 2001. 60, Ohtsuka T, Yamakage A, Yamazaki S: The polymorphism of tmnsforming growth factor-beta1 gene in Japanese patients with systemic sclerosis, Br J Dermatol 147: 458-463, 2002. 61. Stratton R, Shiwen X, Martini G, et al: !loprost suppresses connective tissue growth factor production in fibroblasts and in the skin of scleroderma patients, J Clin Invest 108: 241-250, 2001. 62, Sato S, Nagaoka T, Hasegawa M, et al: Serum levels of connective tissue growth factor are elevated in patients with systemic sclerosis: association with extent of skin sclerosis and severity of pulmonary fibrosis, J Rheumatol 27: 149--154, 2000, 63. Gore-Hyer E, Pannu J, Smith EA, et al: Selective stimulation of collagen synthesis in the presence of costimulatory insulin signaling by connective tissue growth factor in scleroderma fibroblasts, Arthrltis Rheum 48: 798-806, 2003.
64, Needleman BW, Wigley FM, Stair RW: Interleukin-I, interleukin-2, interleukin4, interleukin-6, tumor necrosL~ factor alpha, and interferon-gamma levels in sera from patients with scleroderma, Arthritis Rheum 35: 67-72, 1992, 65, Hasegawa M, Sato S, Ihn H, et al: Enhanced production of interleukin-6 ([L6), oncostatin M and soluble IL-6 receptor by cultured peripheral blood mononuclear cells from patients with systemic sclerosis. Rheumatology (Ox!) 38: 612-617, 1999, 66, Atamas SP, Yurovsky W, Wise R, et al: Production of type 2 cytokines by CD8+ lung cells is associated with greater decline in pulmonary function in patients with systemic sclerosis, Arthritis Rheum 42: 1168-1I78, 1999, 67, Sato S, Hanakawa H, Hasegawa M, et ale Levels of interleukin 12. a cytokine of type I helper T cells, are elevated in sera from patients with systemic sclerosis, J Rheumatol 27: 2838-2842, 2000, 68, Shahin AA, Anwar S, Elawar AH, et al: Circulating soluble adhesion molecules in patients with systemic sclerosis: correlation between circulating soluble vascular cell adhesion molecule-I (sVCAM-1) and impaired left ventricular diastolic function, Rheumatol Int 20: 21-24. 2000, 69, Komum K, Sato S, FUjimoto M, et al: Elevated levels of circulating CD44 in patients with systemic sclerosis: association with a milder subset. Rheumatology (Ox!) 41: 1149--1I54, 2002, 70, Sato H, Lagan AL, Alexopoulou C, et al: The TNF-863A allele strongly associates with anticentromere antibody positivity in scleroderma. Arthritis Rheum 50: 558-564, 2004, 71, Hasegawa M, Sato S, Takehara K: Augmented production of chemokines (monocyte chemotactic protein-l (MCP-I), macrophage inflammatory protein-lalpha (MIP-lalpha) and MIP-lbeta) in patients with systemic sclerosis: MCP-I and MIP-lalpha may be involved in the development of pulmonary fibrosis, Clin Exp Immunol 117: 159--165, 1999. 72, Galindo M, Santiago B, Rivero M, et al: Chemokine expression by systemic sclerosis fibroblast" abnormal regulation of monocyte chemoattmctant protein 1 expression, Arthritis Rheum 44: 1382-1386, 2001. 73, Distler 0, Pap T, Kowal-Bielecka 0, et al: Overexpression of monocyte chemoattractant protein 1 in systemic sclerosis: role of platelet-derived growth factor and effects on monocyte chemotaxis and collagen synthesis, Arthritis Rheum 44: 2665-2678, 2001. 74, Furuse S, Fujii H, Kahuragi Y, et al: Serum concentmtions of the CXC chemokines interleukin 8 and growth-regulated oncogene-alpha are elevated in patients with systemic sclerosis, J Rheumato! 30: 1524-1528,2003, 75, Huffstutter JE, DeLustro FA, LeRoy EC: Cellular immunity to collagen and laminin in scleroderma, Arthritis Rheum 28: 775-780, 1985, 76, Irani AM, Gruber BL, Kaufman LD, et al: Mast cell changes in scleroderma, Presence of MCT cells in the skin and evidence of mast cell activation, Arthritis Rheum 35: 933-939, 1992, 77, Kahaleh MB, Osborn I, leRoy EC: Increased factor VIII/von Willebrand factor antigen and von Willebrand factor activity in scleroderma and in Raynaud's phenomenon, Ann Intern Med 94: 482-484, 1981. 78, Woolf AD, WakerJey G, Wallington TB, et al: Factor VIII related antigen in the assessment of vasculitL~, Ann Rheum Dis 46: 441-447, 1987, 79, Matucci-Cerinic M, Pignone A, Lotti T, et al: Reduced angiotensin converting enzyme plasma activity in scleroderma, A marker of endothelial injury? J Rheumatol 17: 328-330, 1990, 80, 'Sgonc R, Gruschwitz MS, Dietrich H, et al: Endothelial cell apoptosis is a primary pathogenetic event underlying skin lesions in avian and human scleroderma, J Clin Invest 98: 785-792, 1996, 81. Fries JF: The microvascular pathogenesis of scleroderma: an hypothesis, Ann Intern Med 91: 788-789, 1979, 82, Salojin KY, Le Tonqueze M, Saraux A, et al: Arttiendothelial cell antibodies: useful markers of systemic sclerosis, Am J Med 102: 178-185, 1997, 83, Pignone A, Scaletti C, Matucci-Cerinic M, et ale Anti-endothelial cell antibodies in systemic sclerosis: significant association with vascular involvement and alveolo-capillary impairment. Clin Exp Rheumatol 16: 527-532. 1998. 84, Gruschwitz MS, Hornstein OP, von Den DP: Correlation of soluble adhesion molecules in the peripheral blood of scleroderma patients with their in situ expreSSion and with disease activity. Arthritis Rheum 38: 184-189, 1995, 85, Shimada Y, Hasegawa M, Takehara K, et al: Elevated serum L-seleetin levels and decreased L-selectin expression on CD8(+) lymphocytes in systemic sclerosis, Clin Exp Immunol 124: 474-479, 2001. 86, Gruschwitz M, von Den DP, Kellner I, et al: Expression of adhesion proteins involved in cell-cell and cell-matrix interactions in the skin of patients with progressive systemic sclerosis, J Am Acad Dermatol 27: 169--177, 1992, 87, Shi-Wen X, Panesar M, Vancheeswamn R. et al: Expression and shedding of intercellular adhesion molecule 1 and lymphocyte function-associated antigen 3 by normal and scleroderma fibroblasts, Effects of interferon-gamma, tumor necrosis factor alpha, and estrogen, Arthrltis Rheum 37: 1689--1697, 1994, 88, Koch AE, Kronfeld-Harrington LB, Szekanecz Z, et a1: In situ expression of cytokines and cellular adhesion molecules in the skin of patients with systemic sclerosis, Their role in early and late disease, Pathobio!ogy 61: 239--246, 1993, 89, Kuroda K, Okamoto 0, Shinkai H: Dermatopontin expreSSion is decreased in hypertrophic scar and systemic sclerosis skin fibroblasts and is regulated by transforming growth factor-betal. interleukin-4, and matrix collagen, J Invest Dermatol 112: 706--710, 1999, 90, Jelaska A, Ar'Jkawa M, Broketa G, et al: Heterogeneity of collagen synthesis in normal and systemic sclerosis skin fibroblasts, Increased proportion of high collagen-prodUcing cells in systemic sclerosis ftbroblasts, Arthritis Rheum 39: 1338-1346, 1996,
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
91. Lovell CR, Nicholls AC, Duance VC, et al: Characterization of dermal collagen in systemic sclerosis. Br J Dermatol 100: 359--369, 1979. 92. Brady AH: Collagenase in scleroderma. J Clin Invest 56: 1175-1180, 1975. 93. VillO J, Bauer EA, Eisen AZ: Scleroderma: increased biosynthesis of triplehelical type I and type III procollagens associated with unaltered expression c)f collagenase by skin fibroblasts in culture. J Clin Invest 64: 921--930, 1979. 94. 11eltonen L, Palotie A, Myllyla R, et al: Collagen biosynthesis in systemic scleroderma: regulation of posllranslational modifications and synthesis of procollagen in cultured fibroblasts, J Invest Dermatol 84: 14-18, 1985. 95, l3lumenkrantz N, Asboe-Hanson G: Subhydroxylated collagen in scleroderma. Acta Derm Venereol 58: 359--361, 1978. 96. Santiago B, Galindo M, Rivero M, et al: Decreased susceptibility to Fasinduced apoptosis of systemic sclerosis dermal fibroblasts. Arthritis Rheum 44: 1667-1676, 2001. 97. Burge KM, Perry HO, Stickler GB: "Familial" scleroderma. Arch Dermatol 99: 681-687, 1969. 98. De Keyser F, Peene I, Joos R, et al: Occurrence of scleroderma in monozygotic twins. J Rheumatol 27: 2267-2269, 2000. 99. Englert H, Small-McMahon J, Chambers P, et al: Familial risk estimation in systemic sclerosis. Aust N Z J Med 29: 36--41, 1999. 100. Clements PJ, Opelz G, Terasaki PI, et al: Association of HLA antigen a9 with progressive systemic sclerosis (scleroderma). Tissue Antigens 11: 357-361, 1978. 101. Van der Meulen J, Van der Voort-Beelen JM, D'Amaro J, et al: HLA-B8 in Raynaud's phenomenon. Tissue Antigens 15: 81, 1980. 102. Kallenberg CG, Van der Voort-Beelen JM, D'Amaro J, et al: Increased frequenty of B8/DR3 in scleroderma and association of the haplotype with impaired cellular immune response. Clin Exp Immunol 43: 478-485, 1981. 103. Lynch q, Singh G, Whiteside n, et al: Histocompatibility antigens in progressive systemic sclerosis (PSS: scleroderma). J Clin Immunol 2: 314-318, 1982. 104. Gladman DD, Keystone EC, Baron M, et al: Increased frequency of HLA-DR5 in scleroderma. Arthritis Rheum 24: 854-,<356, 1981. 105. Black CM, Welsh KI, Maddison PJ, et al: HLA antigens in scleroderma. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, pp 84-88. 106. Whiteside n, Medsger TA Jr, Rodnan G: Studies of HLA antigens in progressive systemic sclerosis. In Black CM, Meyers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 89. 107. Fox RI, Kang HI: Genetic and environmental factors in systemic sclerosis. Curr Opin Rheumatol 4: 857--861, 1992. 108. Lambert NC. Pang JM, Hermes HM, et al: HLA-DRB1 genes in risk of systemic sclerosis in women, and potential modulation of risk by microchirnerism from pregnancy. Arthritis Rheum 48 (Supp!): S356, 2003. 109. Megli FM, Matliazzi M, Di Tullio T, et al: Annexin V binding perturbs the cardiolipin fluidity gradient in isolated mitochondria. Can it affect mitochondrial function? Biochemistry 39: 5534-5542, 2000. 110. Arliell CM, O'Hanlon TP, Lopez AM, et al: HLA-DQA1 is not an apparent risk factor for microchimerism in patients with various autoimmune diseases and in healthy individuals. Arthritis Rheum 48: 2567-2572, 2003. 111. Burastero SE, Galbiati S, Vassallo A, et al: Cellular microchimerism as a lifelong physiologic status in parous women: an immunologic basis for its amplification in patients with systemic sclerosis. Althritis Rheum 48: 1109--1116, 2003. 112. Nelson JL, Furst DE, Maloney S, et al: Microchimerism and HLA-compatible relationships of pregnancy in scleroderma. Lancet 351: 559--562, 1998. 113. Sontheimer RD: A portable digital microphotography unit for rapid docul11entation of periungual nailfold capillary changes in autoimmune connective tissue diseases. J RheumalOl 31: 539--544, 2004. 114. Ohtsuka T, Miyamoto Y, Yamakage A, et al: Quantitative analysis of microchimerism in systemic sclerosis skin tissue. Arch Dermatol Res 293: 387-391, 2001. 115. ArtIelt CM, Smith]B, Jimenez SA: IdenlifiC'.ltion of fetal DNA and cells in skin lesions from women with systemic sclerosis. N EnglJ Med 338: 1186--1191, 1998. 116. Dabich L, Sullivan DB, Cassidy ]T: Scleroderma in the child. J Pediatr 85: 770-775, 1974. 117. Valentini G, Medsger TA Jr, Silman AJ. et al: The assessment of the patient with systemic sclerosis. Introduction. Clin Exp Rheumatol 21 (Suppl 29): S2-S4, 2003. 118. Maricq HR, Spencer-Green G, LeRoy EC: Skin capillary abnormalities as indicators of organ involvement in scleroderma (systemic sclerosis), Raynaud's syndrome and dermatomyositis. AmJ Med 61: 862--870, 1976. 119. Jacobs fI.U, Breslau PJ, Slaaf DW, et al: Nomenclature of Raynaud's phenomenon: a capillary microscopic and hemorheologic study. Surgery 101: 136-145. 1987. 120. Pallerson JW: Pterygium inversum unguis-like changes in scleroderma. Report of four C'.lses. Arch DermalOl 113: 1429--1430, 1977. 121. Moore CP, Willkens RF: The subcutaneous nodule: Its significance in the diagnosis of rheumatic disease. Semin Arthritis Rheum 7: 63-79, 1977. 122. Raynaud M: De l'asphyxie locale et de la gangrene symetrique des extremities. Paris, L Leclerc, Libraire-Editeur, 1862. 123. Raynaud M: On local asphyxia and symmetrical gangrene of the extremities (1862), Arch Gen Med 1: 189. 1874. 124. Yarom A, Levinson JE: Vasculopathy in scleroderma. In Hicks RV (ed): Vasculopathies in Childhood. Lillieton, MA, PSG Publishing, 1988, p 243125. Duffy CM, Laxer RM, Lee P, et al: Raynaud syndrome in childhood. J Pediatr 114: 73-78, 1989.
467
126. Follansbee WP, Curtiss EI, Medsger TA .lr, et al: Physiologic abnormalities of cardiac function in progressive systemic sclerosis with diffuse scleroderma. N Engl.l Med 310: 142-148, 1984. 127. Allen EV, Brown GE: Raynaud's disease: A critical review of minimal requisites for diagnosis. Am J Med Sci 183: 187, 1932. 128. Jones GT, Herrick At, Woodham SE, et al: Occurrence of Raynaud's phenomenon in children ages 12-15 years: prevalence and association with other common symptoms. Arthritis Rheum 48: 3518-3521, 2003. 129. Jung LK, Dent PB: Prognostic significance of Raynaud's phenomenon in children. Clin Pediatr (Phila) 22: 22-25, 1983. 130. Nigrovic PA, Fuhlbrigge RC, Sundel RP: Raynaud's phenomenon in children: a retrospective review of 123 patients. Pediatrics 111: 715-721, 2003. 131. Harper FE, Maricq HR, Turner RE, et al: A prospective study of Raynaud phenomenon and early connective tissue disease. A five-year report. Am J Med 72: 883--888, 1982. 132. Del Bianco E, Magini B, Muscarella G, et al: Raynaud's phenomenon (primary or secondary to systemic sclerosis). The usefulness of laser-Doppler flowmetry in the diagnosis. Int Angiol 20: 307-313, 2001. 133. Dabich L, Bookstein JJ, Zweifler A, et al: Digital arteries in patients with scleroderma. Arteriographic and plethysmographic study. Arch Intern Med 130: 708--714, 1972. 134. LeRoy EC, Downey JA, Cannon PJ: Skin capillary blood flow in scleroderma. J Clin Invest 50: 930-939, 1971. 135. Winkelmann RK, Goldyne ME, Linscheid RL: Influence of cold on catecholamine response of vascular smooth muscle strips from resistance vessels of scleroderma skin. Angiology 28: 330-339, 1977. 136. Winkelmann RK, Goldyne ME, Linscheid RL: HypersensitiVity of scleroderma cutaneous vascular smooth muscle to 5-hydroxytryptamine. Br J Dermatol 95: 51-56, 1976. 137. Kallenberg CG, Vellenga E, Wouda AA, et al: Platelet activation, fibrinolytic activity and circulating immune complexes in Raynaud's phenomenon. J Rheumatol 9: 878--884, 1982. 138. Majerus PW: Arachidonate metabolism in vascular disorders. .I Clin Invest 72: 1521-1525, 1983. 139. Crocq C: De I' "acrocyanose." Semaine Med 16: 208, i896. 140. Lewis T, Landis EM: Observations upon the vascular mechanism in acrocyanosis. Heart 15: 230, 1930. 141. Stern ES: The aetiology and pathology of acrocyanosis. Br J Dermatol Syphilis 49: 100, 1937. 142. Peacock JH: Vasodilatation in the human hand. Observations on primary Raynaud's disease and acrocyanosis of the upper extremities. Clin Sci 17: 575, 1957. 143. Peacock JH: A comparative study of the digital cutaneous temperatures and hand blood flows in the normal hand, primary Raynaud's disease and primary acrocyanosis. Clin Sci 18: 25, 1959. 144. Sivula A: Vascular reactions in acrocyanosis. Angiology 17: 269--274, 1966. 145. Goelle DK: Chilblains (perniosis). JAm Acad Dermatol 23: 257-262, 1990. 146. Clements P./, Furst DE, Campion DS, et al: Muscle disease in progressive systemic sclerosis: diagnostic and therapeutic considerations. Arthritis Rheum 21: 62-71, 1978. 147. Ortiz-Alvarez 0, Cabral D, Prendiville JS, et al: Intestinal pseudo-obstruction as an initial presentation of systemic sclerosis in two children. Br J Rheumatol 36: 280-284, 1997. 148. Wang SJ, Lan.JL, Chen DY, et al: Colonic transit disorders in systemic sclerosis. Clin Rheumatol 20: 251-254, 2001. 149. Quartier P, Bonnet D, Fournet JC, et al: Severe cardiac involvement in children with systemic sclerosis and myositis. J Rheumatol 29: 1767-1773, 2002. 150. Steen V: The heart in systemic sclerosis. Curr Rheumatol Rep 6: 137-140, 2004. 151. Follansbee WP, Zerbe TR, Med,ger TA Jr: Cardiac and skeletal muscle disease in systemic sclerosis (scleroderma): a high risk association. Am Heart J 125: 194-203, 1993. 152. Gupta MP, Zoneraich S. Zeitlin W, et al: Scleroderma heart disease with slow flow velocity in coronary arteries. Chest 67: 116-119, 1975. 153. Eid NS, Buchino JJ, Schikler KN: Pulmonary manifestations of rheumatic diseases. Pediatr Pulmonol Suppl 18: 91-92, 1999. 154. Morgan C, Knight C, Lunt M, et al: Predictors of end stage lung disease in a cohort of patients with scleroderma. Ann Rheum Dis 62: 146-150, 2003. 155. Hill CL, Nguyen AM, Roder D, et al: Risk of cancer in patients with scleroderma: a population based cohort study. Ann Rheum Dis 62: 728--731, 2003. 156. Denton CP, Black CM: Combination therapies for systemic sclerosis. Springer Semin Immunopathol 23: 109--129, 2001. 157. Furst DE, Davis JA, Clements PJ, et al: Abnormalities of pulmonary vascular dynamics and inflammation in early progressive systemic sclerosis. Arthritis Rheum 24: 1403-1408, 1981. 158. Fahey PJ, Utell MJ, Condemi JJ, et al: Raynaud's phenomenon of the lung. Am J Med 76: 263-269, 1984. 159. Vesely R, Vargova V, Ravelli A, et al: Serum level of KL-6 as a marker of interstitial lung disease in patients With juvenile systemic sclerosis. J Rheumatol 31: 795--800, 2004. 160. Rhew EY, Barr WG: Scleroderma renal crisis: new insights and developments. Curr Rheumatol Rep 6: 129--136,2004. 161. Medsger TA Jr. Masi AT: Survival with scleroderma. II. A life-table analysis of clinical and demographic factors in 358 male U.S. veteran patients. J Chronic Dis 26: 647-660, 1973.
468
C HAP T E R
19 SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
162. Cannon P], Hassar M, Case DB, et al: The relationship of hypertension and renal failure in sclerodenna (progressive systemic sclerosis) to stru<.'tural and functional abnormalities of the renal cortical circulation. Medicine (Baltimore) 53: 1-46, 1974. 163. Oliver ]A, Cannon P]: The kidney in sclerodenna. Nephron 18: 141-150, 1977. 164. LeRoy EC, Fleischmann RM: The management of renal scleroderma: experience with dialysis, nephrectomy and transplantation. Am] Med 64: 974-978, 1978. 165. Whitman HH, 1Il, Case DB, Laragh ]H, et al: Variable response to oral angiotensin-converting-enzyme blockade in hypertensive scleroderma patients. Arthritis Rheum 25: 241-248, 1982. 166. Burke M], Carty ]E: Trigeminal neuropathy as the presenting symptom of systemic sclerosis. Postgrad Med] 55: 423-425, 1979. 167. Teasdall RD, Frayha RA, Shulman LE: Cranial nerve involvement in systemic sclerosis (scleroderma): a report of 10 cases. Medicine (Baltimore) 59: 149-159, 1980. 168. Farrell DA, Medsger TA ]r: Trigeminal neuropathy in progressive systemic sclerosis. Am] Med 73: 57--62, 1982. 169. Lee P, Bruni], Sukenik S: Neurological manifestations in systemic sclerosis (scleroderma).] Rheumatol 11: 480-483, 1984. 170. Dahlgaard T, Nielsen VK, Kristensen ]K: Vibratory perception in patients with generalized sclerodenna. Acta Denn Venereol 60: 119-122, 1980. 171. Estey E, Lieberman A, Pinto R, et al: Cerebral arteritis in scleroclenna. Stroke 10: 595-597, 1979. 172. Alarcon-Segovia 0, Ibanez G, Hernandez-Ortiz], et al: Sjogren's syndrome in progressive systemic sclerosis (sclerodenna). Am] Med 57: 78-85, 1974. 173. Fleischmajer R, Perlish ]S, West WP: Ultrastructure of cutaneous cellular infiltmtes in scleroderma. Arch Dermatol 113: 1661-1666, 1977. 174. Rodnan GP, Myerowitz RL, ]usth GO: Morphologic changes in the digital arteries of patients with progressive systemic sclerosis (scleroderma) and Raynaud phenomenon. Medicine (Baltimore) 59: 393-408, 1980. 175. Claman HN: Mast cells and fibrosis. The relevance to scleroderma. Rheum Dis Clin North Am 16: 141-151, 1990. 176. Braun-Falco 0: Ober das Verhalten der interfibrillaren Grundsubstanz bei Sklerodennia. Dermatol Wochenschr 136: 1085, 1957 [cited by Rodnan GP: Progressive systemic sclerosis: clinical features and pathogenesis of cutaneous involvement (scleroderma). Clin Rheum Dis 5: 49, 1979]. 177. Rodnan GP, Lipinski E, Luksick]: Skin thickness and collagen content in progressive systemic sclerosis and localized scleroderma. Arthritis Rheum 22: 13D-140, 1979. 178. Hayes RL, Rodnan GP: The ultrastructure of skin in progressive systemic sclerosis (scleroderma). I. Dermal collagen fibers. Am] Pathol 63: 433-442, 1971. 179. Rodnan GP, Medsger TA ]r: MusculOSkeletal involvement in progressive systemic sclerosis (sclerodenna). Bull Rheum Dis 17: 419, 1966. 180. Clark ]A, Winkelmann RK, Ward LE: Serologic alterations in scleroderma and sclerodermatomyositis. Mayo Clin Proc 46: 104-107, 1971. 181. Medsger TA ]r, Rodnan GP, Moossy], et al: Skeletal muscle involvement in progressive systemic sclerosis (sclerodenna). Arthritis Rheum 11: 554-568, 1968. 182. Lindamood M, Mills 0, Steigerwald]: Skeletal muscle abnormalities in progressive systemic sclerosis. Arthritis Rheum 19: 807, 1976. 183. Bulkley BH: Progressive systemic sclerosis: Cardiac involvement. Clin Rheum Dis 5: 131, 1979. 184. Russell ML, Friesen 0, Henderson RD, et al: Ultrastructure of the esophagus in scleroderma. Arthritis Rheum 25: 1117-1123, 1982. 185. Smith JW, Clements P], Levisman], et al: Echocardiographic features of progressive systemic sclerosis (PSS). Correlation with hemodynamic and postmortem studies. Am] Med 66: 28-33, 1979. 186. Young RH, Mark G]: Pulmonary vascular changes in scleroderma. Am] Med 64: 998--1004, 1978. 187. Lapenas 0, Rodnan GP, Cavallo T: Immunopathology of the renal vascular lesion of progressive systemic sclerosis (sclerodenna). Am ] Pathol 91: 243--258, 1978. 188. MeGiven AR, De Boer WG, Barnett A]: Renal immune deposits in scleroderma. Pathology 3: 145-150, 1971. 189. Gerber MA: Immunohistochemical findings in the renal vascular lesions of progressive systemic sclerosis. Hum Pathol 6: 343--347, 1975. 190. ]armolyeh], Daoud AS, Landau], et al: Aortic media explants. Cell proliferation and production of mucopolysaccharides, collagen, and elastic tissue. Exp Mol Pathol 9: 171-188, 1%8. 191. Preliminary criteria for the classification of systemic sclerosis (scleroderma). Subcommittee for scleroderma criteria of the American Rheumatism Association Diagnostic and Therapeutic Criteria Committee. Arthritis Rheum 23: 581-590, 1980. 192. LeRoy EC, Medsger TA]r: Criteria for the classification of early systemic sclerosis. ] Rheumatol 28: 1573--1576, 2001. 193. Falcini F, Trapani S, Taccetti G, et al: [Systemic sclerosis in childhood]. Pediatr Med Chir 12: 593--599, 1990. 194. Ansell BM, Falcini F, Woo P: Sclerodenna in childhood. Clin Dermatol 12: 299-307, 1994. 195. Martini A: Juvenile systemic scleroderma. Curr Rheumatol Rep 3: 387-390, 2001. 196. Athreya BH: Juvenile sclerodenna. Curr Opin Rheumatol 14: 553--561, 2002.
197. Penas Pr~ ]ones-Caballero M, Aragues M, et al: Sclerodermatous graft-vs-host disease: clinical and pathological study of 17 patients. Arch Dermatol 138: 924-934, 2002. 198. Dall'Amico R, Rossetti F, lulian F, et al: PholOpheresis in paediatric patients with drug-resistant chronic graft-versus-host disease. Br] Haematol 97: 848-854, 1997. 199. Clements P], Furst DE, Ho W, et al: Progressive systemic sclerosis-like disease follOWing bone marrow transplantation. In Black CM, Myers AR (edsl: Systemic Sclerosis (Sclerodenna). New York, Gower Medical, 1985, pp 376-381. 200. Cowper SE, Su LD, Bhawan], et al: Nephrogenic fibrosing dermopathy. Am ] Dermatopathol 23: 383--393, 2001. 201. Cowper SE, Robin HS, Steinberg SM, et al: Scleromyxoedema-like cutaneous diseases in renal-dialysis patients. Lancet 356: 100D-1001, 2000. 202. Owens GR, Medsger TA: Systemic sclerosis secondary to occupational exposure. Am] Med 85: 114-116, 1988. 203. Selikoff 1], Hammond EC: Toxicity of vinyl chloride polyvinyl chloride. Ann NY Acad Sci 246: 1, 1975. 204. Cohen IS, Mosher MB, O'Keefe E], et al: Cutaneous toxicity of bleomycin therapy. Arch Dermatol 107: 553--555, 1973. 205. Luna MA, Bedrossian CW, Lichtiger B, et al: Interstitial pneumonitis associated with bleomycin therapy. Am] Clin Pathol 58: 501-510, 1972. 206. Finch WR, Buckingham RB, Rodnan GP, et al: Scleroderma induced by bleomycin. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, pp 114-121. 207. Palestine RF, Millns ]L, Spigel GT, et al: Skin manifestations of pentazocine abuse. ] Am Acad Dennatol 2: 47-55, 1980. 208. KIlbourne EM, Rigau-Perez ]G, Heath CW]r, et al: Clinical epidemiology of toxic-oil syndrome. Manifestations of a new illness. N Engl ] Med 309: 1408--1414, 1983. 209. Alonso-Ruiz A, lea-Mendoza AC, Salazar-Vallinas ]M, et al: Toxic oil syndrome: a syndrome with features overlapping those of various forms of scleroderma. Semin Arthritis Rheum 15: 20D-212, 1986. 210. Diaz-Perez ]L, lubizarreta], Gardeazabal], et al: [Familial eosinophilic fascitis induced by toxic oill. Med Cutan Ibero Lat Am 16: 51-58, 1988. 211. Izquierdo M, Mateo I, Rodrigo M, et al: Chronic juvenile toxic epidemic syndrome. Ann Rheum Dis 44: 98-103, 1985. 212. Miyjoshi K, Shiragami H, Yoshida K: Adjuvant disease of man. Clin Immunol (Tokyo) 5: 785, 1973. 213. Kumagai Y, Shiokawa Y, Medsger TA ]r, et al: Clinical spectrum of connective tissue disease after cosmetic surgery. Observations on eighteen patients and a review of the Japanese literature. Arthritis Rheum 27: 1-12, 1984. 214. Santavirta S, Konttinen YT, Bergroth V, et al: Aggressive granulomatous lesions associated with hip arthroplasty. Immunopathological studies.] Bone Joint Surg Am 72: 252-258, 1990. 215. Bridges A], Vasey FB: Silicone breast implants. History, safety, and potential complications. Arch Intern Med 153: 2638-2644, 1993. 216. Wolfe F: "Silicone related symptoms" are common in patients with fibromyalgia: no evIdence for a new disease. ] Rheumatol 26: 1172-1175, 1999. 217. Wolfe P, Anderson J: Silicone filled breast implants and the risk of fibromyalgia and rheumatoid arthritis [published erratum appears in] Rheumatol 27: 825, 2000]. ] Rheumatol 26: 2025-2028, 1999. 218. Kornreich HK, Shaw KN, Koch R, et al: Phenylketonuria and scleroderma. ] Pediatr 73: 571-575, 1968. 219. Lasser AE, Schultz Be, Beaff 0, et al: PhenylketonUria and sclerodenna. Arch Dermatol 114: 1215-1217, 1978. 220. Coskun T, Ozalp I, Kale G, et aI: Scleroderma-like skin lesions in two patients with phenylketonuria. Eur] Pediatr 150: 109-110, 1990. 221. Nova MP, Kaufman M, Halperin A: Scleroderma-like skin indurations in a child with phenylketonuria: a clinicopathologic correlation and review of the Iitemture.] Am Acad Dermatol 26: 329-333, 1992. 222. Nishimura N, Okamtoto M, Yasui M, et al: Intermediary metabolism of phenylalanine and tyrosine in diffuse collagen diseases. II. Influences on the low phenylalanine and tyrosine diet upon patients with collagen diseases. Arch Dennatol 80: 466, 1959. 223. Drummond KN, Michael AF, Good RA: Tryptophan metabolism in a patient with phenylketonuria and scleroderma: a proposed explanation of the indole defect in phenylketonuria. Can Med Assoc ] 94: 834-838. 1966. 224. Villee DB, Nichols G ]r, Talbot NB: Metabolic studies in two boys with classical progeria. Pediatrics 43: 207-216, 1969. 225. Fleischmajer R, Pollock ]L: Progressive systemic sclerosis: Pseudosclerodenna. Clin Rheum Dis 5: 243, 1979. 226. Jansen T, Romiti R: Progeria infantum (Hutchinson-Gilford syndrome) associated with scleroderma-like lesions and aero-osteolysis: a case report and brief review of the literature. Pediatr Dennatol 17: 282-285, 2000. 227. Epstein C], Martin GM, Schultz AL, et al: Werner's syndrome a review of its symptomatology, natural history, pathologic features, genetics and relationship to the natural aging process. Medicine (Baltimore) 45: 177-221, 1%6. 228. Bauer EA, Uitto], Tan EM, et al: Werner's syndrome. Evidence for preferential regional expression of a generalized mesenchymal cell defect. Arch Dermatol 124: 9D-101 , 1988. 229. Rocco VK, Hurd ER: Scleroderma and scleroderma-like disorders. Semin Anhritis Rheum 16: 22-69, 1986. 230. Rosenberg HS, Stenback WA, Spjut H]: The fibromatoses of infancy and childhood. Perspect Pediatr Pathol 4: 269-348, 1978.
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
231. Falanga V: Fibrosing conditions in childhood. Adv Dermatol 6: 145-158, 1991. 232. Young EM ]1'. Barr RJ: Sclerosing dermatoses. ] Cutan Pathol 12: 426-441, 1985. 233. Ormond ]K: Idiopathic retroperitoneal fibrosis: a discussion of the etiology. I Urol 94: 385-390, 1%5. 234. 'Marshall A], Baddeley H, Barritt OW, et al: Practolol peritonitis. A study of 16 cases and a survey of small bowel function in patients taking beta adrenergic blockers. Q] Med 46: 135-149, 1977. 235. Stout AI': The fibromatoses. Clin Orthop 19: 11, 1961. 236. Allen PW: The fibromatoses: a clinicopathologic classification based on 140 rases. Am] Surg Pathol 1: 255-270, 1977. 237. Enzinger FM, Weiss SW: Soft Tissue Tumors. St. Louis, CV Mosby, 1983. 238. Greenberg LM, Geppert C, Worthen HG, et al: Scleredema "adultorum" in children. Report of three cases with histochemical study and review of the world literature. Pediatrics 32: 1044, 1963. 239. Venencie PY, Powell FC, Su WP, et al: Scleredema: a review of thirty-three cases.] Am Acad Dermatol 11: 128--134, 1984. 240. Grgic A, Rosenbloom AL, Weber FT, et al: Joint contracture---common manIfestation of childhood diabetes mellitus. ] Pediatr 88: 584---588, 1976. 241. Grossman ME, Bickers DR, Poh-Fitzpatrick MB, et al: Porphyria cutanea tanla. Clinical features and laboratory findings in 40 patients. Am] Med 67: 277-286, 1979. 242. Doyle ]A, Friedman S]: Porphyria and scleroderma: a clinical and laboratory review of 12 patients. Australas] Dermatol 24: 109-114. 1983. 243. Doyle ]A, Connolly SM, Hoagland HC: Hematologic disease in scleroderma oyndromes. Acta Derm Venereol 65: 521-525, 1985. 244. Giordano M, Ara M, Valentini G, et al: Presence of eosinophilia in progressive systemic sclerosis and localized scleroderma. Arch Dermatol Res 271: 'HI-417, 1981. 245. Rodnan GP, Medsger TA: The rheumatic manifestations of progressive systemic sclerosis (scleroderma). Clin Orthop 57: 81-93, 1968. 246. Gladman DO, Gordon DA, Urowitz MB, et al: Pericardial fluid analysis in scleroderma (systemic sclerosis). Am] Med 60: 1064---1068, 1976. 247. Steen YD, Powell DL, Medsger TA ]1': Clinical correlations and prognosis hased on serum autoantibodies in patients with systemic sclerosis. Arthritis Rheum 31: 196-203, 1988. 248. Kikuchi M, Inagaki T: Bibliographical study of the concurrent existence of anticentromere and antitopoisomerase I antibodies. Clin Rheumatol 19: ,135-441, 2000. 249. liato S, Hamaguchi Y, Hasegawa M, et al: Clinical significance of anti-topoisomerase I antibody levels determined by ELISA in systemic sclerosis. Rheumatology (Ox£) 40: 1135--1140, 2001. 250. Briggs DC, Vaughan RW, Welsh Kl, et al: Immunogenetic prediction of pulIllonary fibrosis in systemic sclerosis. Lancet 338: 661-662, 1991. 251. Bunn CC, Denton CP, Shi-Wen X, et al: Anti-RNA polymerases and other autoantibody specificities in systemic sclerosis. Br] Rheumatol 37: 15-20, 1998. 252. MacGregor A], Davrashvili .1, Knight C, et al: Early pulmonary hypertension ill systemic sclerosis: risk of progression and consequences for survival. Arthritis Rheum 39(Suppl): S151, 1996 253. l-lossny E, Hady HA, Mabrouk R: Anti-centromere antibodies as a marker of l\aynaud's phenomenon in pediatric rheumatologic diseases. Pediatr Allergy Immunol 11: 250--255, 2000. 254. Weiner ES, Earnshaw WC, Senecal ]L, et al: Clinical associations of anticentromere antibodies and antibodies to topoisomerase I. A study of 355 patients. Arthritis Rheum 31: 378--385, 1988. 255. l'regeau DR, Leung PS, Coppel RL, et al: Autoantibodies to mitochondria in systemic sclerosis. Frequency and characterization using recombinant cloned l1utoantigen. Arthritis Rheum 31: 386-392, 1988. 256. addis CV, Okano Y, Rudert WA, et al: Serum autoantibody to the nucleolar antigen PM-Scl. Clinical and inununogenetic associations. Arthritis Rheum ;~5: 1211-1217, 1992. 257. Khanna 0, Aggarwal A, Bhakuni OS, et al: BactericidaVpermeability-increasing protein and cathepsin G are the major antigenic targets of antineutrophil cytoplasmic autoantibodies in systemic sclerosis.] Rheumatol 30: 1248-1252, 2003. 258. Armstrong GP, Whalley GA, Doughty RN, et al: Left ventricular function in "cleroderma. Br] Rheumatol 35: 983-988, 1996. 259. Clements 1'], Furst DE, Cabeen W, et al: The relatiomhip arrhythmias and conduction disturbances to other manifestations of cardiopulmonary disease in progressive systemic sclerosis (PSS). Am] Med 71: 38-46, 1981. 260. Alexander EL, Firestein GS, Weiss ]L, et al: Reversible cold-induced abnormalities in myocardial perfusion and function in systemic sclerosis. Ann Intern Med 105: 661-668, 1986. 261. Goltdiener .IS, Moutsopoulos HM, Decker ]L: Echocardiographic identification of cardiac abnormality in scleroderma and related disorders. Am] Med 66: 391-398, 1979. 262. Di Bello V, Ferri C, Giorgi 0, et al: Ultrasonic videodensitometric analysis in scleroderma heart disease. Coron Artery Dis 10: 103-110, 1999. 263. Garty BZ, Athreya BH, Wilmott R, et al: Pulmonary functions in children with progressive systemic sclerosis. Pediatrics 88: 1161-1167, 1991. 264. !'akini F, Pignone A, Matucci-Cerinic M, et al: Clinical utility of non invasive nlethods in the evaluation of scleroderma lung in pediatric age. Scand .I Rheumatol 21: 82-84, 1992.
469
265. Steen YD, Owens GR, Fino G], et al: Pulmonary involvement in systemic sclerosis (scleroderma). Arthritis Rheum 28: 759-767, 1985. 266. GUltadauria M, Ellman H, Kaplan 0: Progressive systemic sclerosis: Pulmonary involvement. Clin Rheum Dis 5: 151, 1979. 267. Kovalchik MT. Guggenheim S], Silverman MH, et al: The kidney in progressive systemic sclerosis: a prospective study. Ann Intern Med 89: 881-887, 1978, 268. Szymanska-]agiello W, Rondio H: Clinical picture of articular changes in progressive systemic sclerosis in children in the light of own ob"ervations. Reumatologia 8: 1-9, 1970. 269. Resnick 0, Niwayama G: Scleroderma (progressive systemic sclerosis). Tn Resnick 0, Niwayama G (eds): Diagnosis of Bone and Joint Disorders. Philadelphia, WB Saunders, 1988, p 1293. 270. Rowell BR, Hopper FE: The periodontal membrane in systemic sclerosis. Br .1 Dermatol 93 (Suppl 2): 23, 1975. 271. Resnick 0, Greenway G, Vint VC, et al: Selective involvement of the first carpometacarpal joint in scleroderma. A]R Am] Roentgenol 131: 283-286, 1978. 272. Weber 1', Ganser G, Frosch M, et al: Twenty-four hour intraesophageal pH monitoring in children and adolescenL~ with scleroderma and mixed connective tissue disease. .1 Rheumatol 27: 2692-2695, 2000. 273. Martel W, Chang SF, Abell MR: Loss of colonic haustration in progressive systemic sclerosis. Am .1 Roentgenol 126: 704---713, 1976. 274. Seely ]M, Jones LT, Wallace C, et al: Systemic sclerosis: using high-resolution CT to detect lung disease in children. AJR Am] Roentgenol 170: 691-697, 1998. 275. Shahin AA, Sabri YV, Mostafa HA, et al: Pulmonary function tests, high-resolution computerized tomography, alpha I-antitrypsin measurement, and early detection of pulmonary involvement in patients with systemic sclerosis. Rheumatol Int 20: 95--100, 2001, 276. Koh OM, Hansell OM: Computed tomogmphy of diffuse interstitial lung disease in children. Clin Radiol 55: 659-667, 2000. 277. Heickendorff L, Zachariae H, Bjerring 1', et al: The use of serologic markers for collagen synthesis and degradation in systemic sclerosis. ] Am Acad Dermatol 32: 584---588, 1995. 278. Hunzelmann N, Risteli], Risteli L, et al: Circulating type I collagen degradation products: a new serum marker for clinical severity in patients with scleroderma? Br] Dermatol 139: 1020--1025, 1998. 279. Denton CP, Bickerstaff MC, Shiwen X. et al: Serial circulating adhesion molecule levels reflect disease severity in systemic sclerosis. Br] Rheumatol 34: 1048--1054, 1995. 280. Medsger TA ]1': Asse"ment of damage and activity in systemic sclerosis, Curr Opin Rheumatol 12: 545--548, 2000. 281. Steen VD, Medsger TA ]1': The value of the Health Assessment Questionnaire and special patient-generated scales to demonstrate change in systemic sclerosis patients over time. Arthritis Rheum 40: 1984---1991, 1997. 282. Silman A. Akesson A, Newman], et al: Assessment of functional ability in patients with scleroderma: a proposed new disability assessment instrument. ] Rheumatol 25: 79-83, 1998. 283. Della RA, Valentini G, Bombardieri S, et al: European multicentre study to define disease activity criteria for systemic sclerosis. I. Clinical and epidemiological features of 290 patients from 19 centres. Ann Rheum Dis 60: 585-591, 2001. 284. Valentini G, Della RA. Bombardieri S. et al: European multicentre study to define disease activity criteria for systemic sclerosis. II. Identification of disease activity variables and development of preliminary activity indexes. Ann Rheum Dis 60: 592-598, 2001. 285. Foeldvari I, Wulffraat N: Recognition and management of scleroderma in children. Paediatr Drugs 3: 575-583, 2001. 286. Steen YD, Lanz JK ]1', Conte C, et al: Therapy for severe interstitial lung disease in systemic sclerosis. A retrospective study. Arthritis Rheum 37: 1290--1296, 1994. 287. van den Hoogen FH, Boerbooms AM, Swaak A], et al: Comparison of methotrexate with placebo in the treatment of systemic sclerosis: a 24 week randomized double-blind trial, followed by a 24 week observational trial. Br .1 Rheumatol 35: 364---372, 1996, 288. Pope ]E, Bellamy N, Seibold ]R, et al: A randomized, controlled trial of methotrexate versus placebo in early diffuse scleroderma. Arthritis Rheum 44: 1351-1358, 2001. 289. Stratton R], Wilson H, Black CM: Pilot study of anti-thymocyte globulin plus mycophenolate mofetil in recent-onset diffuse scleroderma. Rheumatology (Ox[) 40: 84---88, 2001. 290. Steen YD, Medsger TA .11': Case-control study of corticosteroids and other dmgs that either precipitate or protect from the development of scleroderma renal crisis. Arthritis Rheum 41: 1613-1619, 1998. 291. Antoniades L. Sfikakis PI', Mavrikakls M: Glucocorticoid effects on myocardial performance in patients with systemic sclerosis. Clin Exp Rheumatol 19: 431-437. 2001. 292. Ellman MH, MacDonald PA, Hayes FA: Etanercept as treatment for diffuse scleroderma: a pilot study. Arthritis Rheum 43(Supp1): S392, 2000. 293. Clements 1'], Lachenbruch PA, Sterz M, et al: Cyclosporine in systemic sclerosis. Results of a forty-eight-week open safety study in ten patients. Arthritis Ri,eum 36: 75--83, 1993. 294. Filaci G, Cutolo M, Scudeletti M, et al: Cydosporin A and Hoprost treatment of systemic sclerosis: clinical results and interleukin-6 serum changes after 12 months of therapy. Rheumatology (Ox£) 38: 992-996, 1999. 295. Morton S], Powell RJ: Cyclosporin and tacrolimus: their use in a routine clinical setting for scleroderma. Rheumatology (Oxf) 39: 865-869, 2000.
470
C HAP T E R
19 SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
296. Gisslinger H, Burghuber OC, Stacher G, et al: Efficacy of cyclosporin A in systemic sclerosis. Clin Exp Rheumatol 9: 38;}-390, 1991. 297. Casoli P, Carretti C, Tumiati B: Scleroderma renal crisis as a possible complication of cyclosporin A withdrawal. Clin Rheumatol 13: 126-128, 1994. 29H. Wulffraat NM, Sanders LA, Kuis W: Autologous hemopoietic stem-cell transplantation for children with refractory autoimmune disease. Curr Rheumatol Rep 2: 316-323, 2000. 299. Farge D, Marolleau jP, Zohar S, et al: Autologous bone marrow transplantation in the treatment of refractory systemic sclerosis: early results from a French multicentre phase I-ll study. Br j Haematol 119: 726-739, 2002. 300. Binks M, Passweg jR, Furst D, et al: Phase IIll trial of autologous stem cell transplantation in systemic sclerosis: procedure related mortality and impact on skin disease. Ann Rheum Dis 60: 577-584, 2001. 301. Martini A, Maccario R, Ravelli A, et at: Marked and sustained improvement two years after autologous stem cell transplantation in a girl with systemic sclerosis. Arthritis Rheum 42: 807-811, 1999. 302. Steen YD, Medsger TA jr, Rodnan GP: D-Penicillamine therapy in progressive systemic sclerosis (scleroderma} a retrospective analysis. Ann Intern Med 97: 652-659, 1982. 303. jimenez SA, Sigal SH: A 15-year prospective study of treatment of rapidly progressive systemic sclerosis with o-penicillamine. j Rheumatol 18: 1496-1503, 1991. 304. Clements Pj, Furst DE, Wong WK, et al: High-dose versus low-dose o-penicillamine in early diffuse systemic sclerosis: analysis of a two-year, double-blind, randomized, controlled clinical trial. Arthritis Rheum 42: 1194-1203, 1999. 305. Furst DE, Clements Pj: D-penicillamine is not an effective treatment in systemic sclerosis. Scand j Rheumatol 30: 189-191, 2001. 306. Medsger TA jr, Lucas M, Wildy KS, et al: o-Penicillamine in systemic sclerosis! Yes! Scand j Rheumatol 30: 192-194, 2001. 307. Steigerwald jC: Colchicine vs. placebo in the treatment of progressive systemic sclerosis. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 415. 308. Alarcon-Segovia D, Ramos-Niembro F, Ibanez DK, et al: Long-term evaluation of colchicine in the treatment of scleroderma. j Rheumatol 6: 705-712, 1979. 309. Black CM, Silman Aj, Herrick AI, et al: Interferon-alpha does not improve outcome at one year in patients with diffuse cutaneous scleroderma: results of a randomized, double-blind, placebo-controlled trial. Arthritis Rheum 42: 299-305, 1999. 310. Grassegger A, Schuler G, Hessenberger G, et al: Interferon-gamma in the treatment of systemic sclerosis: a randomized controlled multicentre trial. Br j Dermatol 139: 639-648, 1998. 311. Unemori EN, Pickford LB, Salles AL, et al: Relaxin induces an extracellular matrix-degrading phenotype in human lung fibroblasts in vitro and inhibits lung fibrosis in a murine model in vivo. j Clin Invest 98: 2739-2745, 1996. 312. SeiboldjR, KornjH, Sintms R, et al: Recombinant buman relaxin in the treatment of scleroderma. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 132: 871-879, 2000. 313. Smith CD, McKendry Rj: Controlled trial of nifedipine in the treatment of Raynaud's phenomenon. Lancet 2: 1299-1301, 1982. 314. Rodeheffer Rj, Rommel' jA, Wigley F, et al: Controlled double-blind trial of nifedipine in the treatment of Raynaud's phenomenon. N Engl j Med 308: 8H0-883 , 1983. 315. Sauza j, Kraus A, Gonzalez-Amaro R, et al: Effect of the calcium channel blocker nifedipine on Raynaud's phenomenon. A controlled double blind trial. j Rheumatol 11: 362-364, 1984. 316. Winston EL, PariseI' KM, Miller KB, et al: Nifedipine as a therapeutic modality for Raynaud's phenomenon. Arthritis Rheum 26: 1177-1180, 1983. 317. Dziadzio M, Denton CP, Smith R, et a1: Losartan therapy for Raynaud's phenomenon and scleroderma: clinical and biochemical findings in a fifteenweek, randomized, parallel-group, controlled trial. Arthritis Rheum 42: 2646-2655, 1999. :\18. Kontos HA, Wasserman Aj: Effect of reserpine in Raynaud's phenomenon. Circulation 39: 259-266, 1969. :\19. Varadi DP, Lawrence AM: Suppression of Raynaud's phenomenon by methyldopa. Arch Intern Med 124: 1;}-18, 1969. 320. McFadyen IJ, Housley E, MacPherson AI: Intraarteriai reserpine administration in Raynaud syndrome. Arch Intern Med 132: 526-528, 1973321. Miyazaki S, Miura K, Kasai Y, et al: Relief from digital vasospasm by treatment with captopril and its complete inhibition by serine proteinase inhibitors in Raynaud's phenomenon. Br Med j (Clin Res Ed) 284: 310--311, 1982. 322. Seibold jR, Jageneau AH: Treatment of Raynaud's phenomenon with ketanserin, a selective antagonist of the serotonin2 (5-HT2) receptor. Arthritis Rheum 27: 139-146, 1984. :\23. CoffmanjD, Clement DL, Creager MA, et al: International study of ketanserin in Raynaud's phenomenon. Amj Med 87: 264-268, 1989. 324. Naidoo P: Griseofulvin in Raynaud's phenomenon. Lancet 2: 1090, 1971. 325. Martin MF, Dowd PM, Ring EF, et al: Prostaglandin E1 infusions for vascular insufficiency in progressive systemic sclerosis. Ann Rheum Dis 40: 350--354, 1981. 326. Belch jj, McKay A, McArdle B, et al: Epoprostenol (prostacyclin) and severe arterial disease. A double-blind trial. Lancet 1: 315-317, 1983. 327. Giordano M, Ara M, Capelli L, et al: Griseofulvin in scleroderma. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, pp 446-448.
328. Drake DB, Kesler RW, Morgan RF: Digital sympathectomy for refractory Raynaud's phenomenon in an adolescent. J Rheumatol 19: 1286-128H, 1992. 329. Wigley FM, Wise RA, Seibold JR, et al: Intravenous i1oprost infusion in patients with Raynaud phenomenon secondary to systemic sclerosis. A multicenter, placebo-controlled, double-hlind study. Ann Intern Med 120: 199-206, 1994. 330. Zulian F, Corona F, Gerloni V, et al: Safety and efficacy of Hoprost for the treatment of ischaemic digits in paediatric connective tissue diseases. Rheumatology (Oxf) 43: 229-233, 2004. :\31. Black CM, Halkier-Sorensen L, Belchjj, et al: Oral i1oprost in Raynaud's phenomenon secondary to systemic sclerosis: a multicentre, placebo-controlled, dose-comparison study. Br j Rheumatol 37: 952-960, 1998. 332. Wigley FM, Korn jH, Csuka ME, et al: Oral iloprost treatment in patients with Raynaud's phenomenon secondary to systemic sclerosis: a multicenter, placebo-controlled, double-blind study. Arthritis Rheum 41: 670-677, 1998. 333. Russell lJ, Lessard jA: Prazosin treatment of Raynaud's phenomenon: a double blind single crossover study. j Rheumatol 12: 94-98, 1985. 334. Coppock jS, Hardman jM, Bacon PA, et al: Objective relief of vasospasm by glyceryl trinitrate in secondary Raynaud's phenomenon. Postgrad Med.J 62: 15-18, 1986. 335. Talpos G, Horrocks M, White jM, et al: Plasmapheresis in Raynaud's disease. Lancet 1: 416-417, 1978. 336. Winkelmann RK, McCune MA, Pineda AA, et a1: A controlled study of plasma exchange in scleroderma. Tn Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical. 1985, pp 449--453. 337. Surwit RS: Biofeedback: a pOSSible treatment for Raynaud's disease. Semin Psychiatry 5: 48;}-490, 1973338. Beckett VL, Donadio jV jr, Brennan LA jr, et al: Use of captopril as early therapy for renal sclerodenna: a prospective study. Mayo Clin Proc 60: 76;}-771 , 1985. 339. Whitman HH 1Il, Case DB, LeRoy EC: Management of hypeltensive scleroderma patients with converting-enzyme inhibition. Tn Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 428. 340. Steen YD, Costantino jP, Shapiro AP, et al: Outcome of renal crisis in systemic sclerosis: relation to availability of angiotensin converting enzyme (ACE) inhibitors. Ann Intern Med 113: 352-357, 1990. 341. Maddison P: Prevention of vascular damage in scleroderma with angiotensin-converting enzyme (ACE) inhibition. Rheumatology (Oxf) 41: 965-971, 2002. 342. Richardson jA: Hemodialysis and kidney transplantation for renal failure from scleroderma. Arthritis Rheum 16: 265-271, 1973. 343. White B, Moore WC, Wigley FM, et al: Cyclophosphamide is associated with pulmonary function and survival benefit in patients with scleroderma and alveolitis. Ann Intern Med 132: 947-954, 2000. 344. Kuhn KP, Byrne DW, Arbogast PG, et al: Outcome in 91 consecutive patients with pulmonary arterial hypertension receiving epoprostenol. Am j Respir Crit Care Med 167: 580--586, 2003. 345. Barst Rj, Rubin Lj, Long WA, et al: A comparison of continuous intravenous epoprostenol (prostacyclinJ with conventional therapy for primary pulmonary hypertension. The Primary Pulmonary Hypeltension Study Group. N Engl j Med 334: 296-302, 1996. 346. Badesch DB, Tapson VF, McGoon MD, et al: Continuous intravenous epoprostenol for pulmonary hypertension due [0 the scleroderma spectrum of disease. A randomized, controlled trial. Ann Intern Med 132: 425--434, 2000. 347. Rubin Lj, Badesch DB, Barst Rj, et al: Bosentan therapy for pulmonary arterial hypertension. N Engl j Med 346: 896-903, 2002. 348. Ghofrani HA, Wiedemann R, Rose F, et al: Combination therapy with oral sildenafil and inhaled iloprost for severe pulmonary hypertension. Ann Intern Med 136: 515-522, 2002. 349. Kinuya K, Nakajima K, Kinuya S, et al: Esophageal hypomotility in systemic sclerosis: close relationship with pulmonary involvement. Ann Nucl Med 15: 97-101, 2001. 350. Bottoni CR, ReinkeI' KA, Gardner RD, et al: Scleroderma in childhood: a 35year history of cases and review of the literature. j Pediatr Orthop 20: 442--449, 2000. 351. Hochberg MC, LopeZ-Acuna D, Gittelsohn AM: Mortality from systemic sclerosis (sclerodenna) in the United States, 1969-1977. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 61. 352. Foeldvari I, Zhavania M, Birdi N, et al: Favourable outcome in 135 children with juvenile systemic sclerosis: results of a multi-national survey. Rheumatology (Oxf) 39: 556-559, 2000. 353. Bulkley BH, Ridolfi RL, Salyer WR, et al: Myocardial lesions of progressive systemic sclerosis. A cause of cardiac dysfunction. Circulation 53: 48;}-490, 1976. 354. Silman Aj: Pregnancy and scleroderma. Am.J Reprod Immunol 28: 238-240, 1992. 355. Buyon]p: The effects of pregnancy on autoimmune diseases. j Leukoc Bioi 63: 281-287, 1998. 356. jacobsen S, Halberg P, Ullman S: Mortality and causes of death of 344 Danish patients with systemic sclerosis (scleroderma). Br j Rheurnatol 37: 750--7'i'i, 1998.
C HAP T E R
19
SYSTEMIC SCLERODERMAS AND RELATED DISORDERS
357. Simeon CP, Annadans L, Fonollosa V, et ai: Survival prognostic factors and markers of morbidity in Spanish patients with systemic sclerosis. Ann Rheum Dis 56: 723-728, 1997. 358. 8ulpitt KJ, Clements PJ, Lachenbruch PA, et al: Early undifferentiated connective tissue disease: III. Outcome and prognostic indicators in early scleroderma (systemic sclerosis). Ann Intern Med 118: 602-609, 1993 359. Winterbauer RH: Multiple telangiectasia, Raynaud's phenomenon, sclerodactyly and subcutaneous calcinosL.: A syndrome mimicking hereditary hemorrhagic telangiectasia. Bull Johns HopkinS Hosp 114: 361, 1964. 360. Fritzler MJ, Kinsella TD: The CREST syndrome: a distinct serologic entiry with anticentromere antibodies. Am J Med 69: 520--526, 1980. 361. l'hibierge G, Wiessenbach RJ: Concretions calcaires souscutanees et sclerodermie. Ann Dermatol Syph 2: 129, 1911. 362. Rodnan GP, Jablonska S: Classiflcation of systemic and localized scleroderma. In Black CM, Myers AR (eds): Systemic Sclerosis (Scleroderma). New York, Gower Medical, 1985, p 3. 363. Tan EM, Rodnan GP, Garcia I, et al: Diversity of antinuclear antibodies in progressive systemic sclerosis. Anti-centromere antibody and its relationship to CREST syndrome. Arthritis Rheum 23: 617-625, 1980.
471
364. Powell FC, Winkelmann RK, Venencie-Lemarchand F, et al: The anticentromere antibody: disease specificity and clirtical significance. Mayo Clin Proc 59: 700--706, 1984. 365. Furst DE, Clements PJ, Saab M, et al: Clinical and serological comparison of 17 chronic progressive systemic sclerosis (PSS) and 17 CREST syndrome patient. matched for sex, age, and disease duration. Ann Rheum Dis 43: 79~1, 1984. 366. Sato S, Komura K, Hasegawa M, et al: Clinical significance of soluble CD31 in patients with systemic sclerosis (SSe): association with limited cutaneous SSe. .I Rheumatol 28: 2460--2465, 2001. 367. Maricq HR, LeRoy EC, D'Angelo WA, et al: Diagnostic potential of in vivo capillary microscopy in scleroderma and related disorders. Arthritis Rheum 23: 183-189, 1980. 368. Maricq HR: Wide-field capillary microscopy. Arthritis Rheum 24: 1159-1165, 1981. 369. Coffman JD: Drug therapy: vasodilator drugs in peripheral vascular disease. N EnglJ Med 300: 713-717, 1979. 370. Gifford RWJr: The arteriospastic diseases: clinical significance and management. Cardiovasc Clin 3: 127-139, 1971. 371. Hansteen V, Lorentsen E: Vasodilator drugs in the treatment of peripheral arterial insufficiency. Acta Med Scand Suppl 556: 3-62, 1974.
C HAP T E R
20
LOCALIZED SCLERODERMAS Audrey M. Nelson and Ronald M. Laxer
~wp
The unifying characteristic of all of the types of scleroderma is an excessive accumulation of collagen in the tissues. 1.2 The localized sclerodermas are a group of disorders whose manifestations mostly are confined to the skin and subdermal tissues. They are rare conditions even in childhood, in which linear scleroderma is most common. The term morphea is applied to many of the types of localized scleroderma and may be preferred because the word scleroderma often leads to consideration of only the ominous consequences of systemic scleroderma.
tation or hyperpigmentation. These lesions occur most frequently on the trunk and less often on the extremities. The face is usually spared. Guttate morphea is much less common. These lesions are small, oval areas less than 1 cm in diameter. Atrophoderma of Pasini and Pierini is characterized by asymptomatic hyperpigmented atrophic patches on the trunk with well-demarcated, so-called cliff-drop borders. These lesions lack the typical inflammatory changes of plaque morphea.
Generalized Morphea
DEFINITION AND CLASSIFICATION Localized scleroderma includes a number of conditions that are often grouped together, and the individual syndromes therefore may seem ill defined. Commonly, localized scleroderma has been divided into three types: morphea, generalized morphea, and linear scleroderma.;}-s Eosinophilic fasciitis, which was originally considered to be a separate syndrome, may also be classified among the subtypes of localized scleroderma or morphea. Peterson and colleagues6 have proposed a revised expanded classification that divides localized scleroderma or morphea into five general types: plaque morphea, generalized morphea, bullous morphea, linear morphea, and deep morphea (Table 20-1).
Plaque Morphea Plaque morphea is the most common and benign form of the morphea syndromes. It is confined to the dermis with only occasional involvement of the superficial panniculus. Subtypes of plaque morphea include morphea en plaque, guttate morphea, atrophoderma of Pasini and Pierini, and keloid morphea. Some consider lichen sclerosus et atrophicus also to be a form of morphea. Morphea en plaque is the most common subtype and is characterized by the insidious onset of an oval or round, circumscribed area of induration with a central waxy, ivory-colored area surrounded by a violaceous halo (Fig. 20-1). The plaques are typically several centimeters in diameter and evolve from an erythematous inflammatory stage through a sclerotic indurated phase with surrounding inflammation and subsequently to softening and dermal atrophy with associated hypopigmen472
The term generalized morphea is applied when individual plaques of morphea become confluent or multiply and affect three or more anatomic sites.
Bullous Morphea Bullous morphea can occur with most subtypes, including typical plaque morphea and morphea profunda. The bullous lesions may possibly result from localized trauma or may be related to lymphatic obstruction from the sclerodermatous process. 7,H
Linear Sderoderma Linear scleroderma is the most common subtype in children and adolescents. 9•10 It is characterized by one or more linear streaks that typically involve an upper or lower extremity. It may also be associated with morphea plaques. With time, the streaks become progressively more indurated and can extend through the dermis, subcutaneous tissue, and muscle to the underlying bone (Fig. 20-2). The lesions frequently follow a dermatomal distribution and are unilateral in 85% to 95% of cases. IO· 11 When a linear lesion involves the face or scalp, it is referred to as en coup de sabre scleroderma (Fig. 20-3). This term was applied historically because the lesion was reminiscent of the depression caused by a dueling stroke from a sword. A number of associated disorders have been reported in such patients, including seizures, uveitis, dental abnormalities, ocular muscle dysfunction, and loss of eyebrows or eyelashes. 12- 19 Progressive hemifacial atrophy may occur in association with linear lesions on the head and scalp. When hemifacial atrophy
C HAP T E R
t.
IABLE 20 J
20
LOCALIZED SCLERODERMAS
473
C1dssificdlion of Morphed
P)i\que morphea Morphea en plaque Gunale morphea Atrophoderma of Pasini and Pierini Keloid morphea (nodular morphea) ILichen sclerosus et atrophicus]' Generalized morphea Bullous morphea Linear morphea Linear morphea (linear scleroderma) En coup de sabre scleroderma Progressive hemifacial atrophy Deep morphea Subcutaneous morphea Eosinophilic fasciitis Morphea profunda Disabling pansclerotic morphea of children
• Rgura Zo-l The morphea en plaque lesion is characterized by a central area of induration with a waxy, ivory-colored area surrounded by inflammation and hyperpigmentation. (See color insert.)
'The entry in brackets is not universally accepted. Reprinted with permission from Peterson LS, Nelson AM, Su WPD: Classification of morphea (localized scleroderma). Mayo Clin Proc 70: 1068--1076, 1995.
occurs without a definable lesion of en coup de sabre, the term Parry-Romberg syndrome is applied. 20 Whether this syndrome exists as a separate entity from localized scleroderma is controversial. Neurologic involvement has been described in both forms of progressive facial hemiatrophy.21
• FIgure Zo-Z A, Typical lesion of linear sderodenna involving the lower extremity.The skin changes are characterized by a waxy induration with surrounding inflammation and hYPerpigmentation distributed in a linear pattern. 8, Linear sderodenna results in undergrowth of the leg; taut, shiny skin; and shortening of the extensor tendon to the second toe on the right foot.
Deep Morphea The subtypes of deep morphea (Fig. 20-4) are the least common but most disabling variants and include subcutaneous morphea, eosinophilic fasciitis, morphea profunda, and disabling pansclerotic morphea of children. In
474
C HAP T E R
20
LOCALIZED SCLERODERMAS
• Figure Z0-3 A, En coup de sabre linear scleroderma of approximately 2 years' duration affects the chin just to the left of midline, resulting in a depression and mild asymmetry of the jaw. 8, En coup de sabre linear sderoderma involves the left face with hyperpigmentation, atrophy of subcutaneous tissues, and early hemifacial atrophy. (See color insert.)
subcutaneous morphea, the primary site of involvement is the panniculus or subcutaneous tissue,22,23 and the onset of sclerosis is often rapid over a period of several months. The plaques are hyperpigmented, symmetric, and somewhat ill defined, and the degree of inflammation is more pronounced than in the other subtypes. In morphea profunda, the entire skin feels thickened, taut, and bound down; however, the abnormality may be localized to a solitary, indurated plaque that usually involves the upper torso. 24 ,25 A rare but severe form of localized scleroderma is disabling pansclerotic morphea ofchildren, first described in 1980 by Diaz-Perez and colleagues. 26 This disorder typically begins before the age of 14 years and is characterized by generalized full-thickness involvement of the skin of the trunk, extremities, face and scalp with sparing of the fingertips and toes. Its course is relentlessly progressive. Ulcerative squamous cell carcinoma has developed in some of these children. 27 ,28 Eosinophilic fasciitis was described in 1974 by Shulman29.3o and in 1975 by Rodnan and colleagues,31 who called the condition "diffuse fasciitis with eosinophilia" and observed that these patients typically had hypergammaglobulinemia and eosinophilia. The fas-
cia is the predominant site of involvement. These lesions typically involve the extremities but spare the hands and feet, and have an appearance that is described as "peau d'orange." Histologic changes similar to eosinophilic fasciitis are found in most of the subtypes of localized scleroderma, strengthening the conjecture that this disorder is a subtype of localized scleroderma. In the pediatric literature, eosinophilic fasciitis frequently is described as involving the hands and feet, which is a departure from the syndrome in adults. Some of these pediatric cases may be more consistent with subcutaneous morphea or morphea profunda rather than eosinophilic fasciitis. Miller32 has described the combined syndrome of fasciitis and morphea in children and pointed out the linkage of these manifestations.
EPIDEMIOLOGY Localized scleroderma is more frequent than systemic scleroderma but still a rare condition. In the general populcition where systemic disease is estimated to have an incidence of 0.45 to 1.9 cases per 100,000,33.34 the incidence of the localized sclerodermas is 2.7 cases per
C HAP T E R
20
LOCALIZED SCLERODERMAS
475
ETIOLOGY AND PATHOGENESIS
• ~ ZG-4
Hands of a 7-year-old boy with deep morphea, demonstrating limitation of extension (A) and flexion (8) of the fingers. (A and 8, From Nelson AM: Localized scleroderma induding morphea, linear scleroderma, and eosinophilic fasditis. Curr Probl Pediatr 26: 318, 1996.)
100,000. 10 In a population-based study of localized scleroderma,1O plaque morphea accounted for 56%, generalized morphea for 13%, linear morphea for 20%, and deep m(~rphea for 11%. Linear and plaque morphea coexisted in 11% of the patients. Referral-based studies of localized scleroderma include a higher proportion of linear scleroderma. 4.9 The female-to-male ratio of localized scleroderma is 2.5:1, whereas in linear scleroderma, it is 1:1. Linear scleroderma is predominantly a pediatric disease. In a population-based study covering patients of all ages,1O 67% of those with linear scleroderma were diagnosed before the age of 18 years. The prevalence of morphea in children age 17 or younger was estimated to be 50 per 100,000. The mean age at onset of localized scleroderma in the pediatric population is approximately 7.9 years. 4 Uziel and colleagues4 reported 30 patients with localized scleroderma over a 7-year period and observed that this patient group numbered approximately the same as those with juvenile dermatomyositis and about one half of those with systemic lupus erythematosus seen during the same time. In a review of their pediatric rheumatology practice, Levinson and Bove35 identified 15 children with localized scleroderma, a frequency that was approximately 1 for every 20 children with juvenile rheumatoid arthritis. A study by Woo and colleagues36 reported that 2% of the patients attending their pediatric rheumatology clinic had localized scleroderma.
The cause and pathogenesis of the localized sclerodermas are unknown. The focus of much investigation is on abnormalities of regulation of fibroblasts, production of collagen, and immunologic abnormalities. In morphea, the collagen fibers become thickened and hyalinized. 37 Multiple studies have demonstrated increased levels of cytokines and other molecules that influence fibroblasts and collagen synthesis. 38 Autoimmunity, environmental factors, infection, and trauma have all been associated with localized disease. It seems certain that autoimmunity is important in the cause because of the multiplicity of abnormal serum antibodies that occurs in patients with localized scleroderma and because of the association of similar cutaneous abnormalities in patients with chronic graft-versus-host disease. 39.40 McNallan, Reed and colleagues41 described fetal chimeric cells in skin biopsies of linear scleroderma or morphea, further suggesting a similar pathogenesis to chronic graft-versus-host disease. A number of drugs and environmental toxins have resulted in scleroderma-like reactions, including bleomycin, ergot, bromocriptine, pentazocine, carbidopa and vitamin K/ 2A toxin contained in some lots of L-tryptophan was incriminated in 1990 in a large epidemic of a syndrome that was similar to eosinophilic fasciitis and morphea, called the eosinophilia-myalgia syndrome. 43 .44 A number of investigations have examined a putative association of morphea and Borrelia burgdorferi, the spirochete that causes Lyme disease. Since this association was first reported in 1985,45 many studies have documented evidence of infection with B. burgdorferi in patients with morphea who live in areas endemic for Lyme disease or have a history of tick bites46.47; however, patients with morphea who do not live in endemic areas have no evidence of prior exposure to B. burgdorferi. 4Pr-50 In the evaluation of patients with morphea, serologic testing for Lyme disease is not likely to be helpful unless the patient has been in an endemic area. Trauma or physical exertion has been implicated in the initiation of lesions and particularly with the onset of eosinophilic fasciitis. 25 .51,52 A review of childhood-onset scleroderma reported a history of trauma at the site of the lesion in 14 of 58 patients53 ; a similar history was not obtained in adults with morphea. The investigators also observed that three patients had developed typical lesions at the site of their measles, mumps, and rubella vaccination. Morphea also has been reported after irradiation of malignant tumors in adults at the site of irradiation and at a distance. 54 's5
CLINICAL MANIFESTATIONS The onset of localized scleroderma is subtle. The first manifestation is usually a localized area of erythema or waxy induration with a surrounding halo of erythema. A few patients have systemic symptoms such as arthralgias, synovitis, joint contractures, and carpal tunnel syndrome. 1O Plaque morphea mainly appears on the chest, abdomen, and back, whereas generalized morphea involves the extremities, chest, and back. Linear morphea
476
C HAP T E R
20
LOCALIZED SCLERODERMAS
affects the lower extremities more commonly than the arms, whereas the deep morphea subtypes affect both upper and lower extremities. The scalp is affected only in the en coup de sabre form of the disorder and in disabling pansclerotic morphea of childhood. 32 Most patients with generalized or deep morphea have bilateral involvement, whereas unilateral lesions are most frequent in plaque and linear disease. Arthralgias and mild synovitis with contractures out of keeping with the degree of synovitis are common presentations in patients with the deep morphea subtypes. Carpal tunnel syndrome has been associated with linear and deep morphea. 1O ,56,57 Raynaud's phenomenon has also been described, although it is rare, is usually unilateral, and occurs in patients who have extensive sclerosis of the hands and forearms,4,9,22,51,58 Calcinosis has been noted in areas involved with linear scleroderma. 59,60 The en coup de sabre type presents with a set of unusual manifestations unique to this group, including progressive hemifacial atrophy,61 ipsilateral uveitis, various dental abnormalities such as separation of the teeth, and involvement of the eyebrows and eyelashes. 62 Central nervous system (CNS) disorders, including seizures and manifestations of CNS vasculitis, have been reported6'Hi5; 47% of patients with craniofacial involvement in one study had neurologic abnormalities. 65 Evidence of internal organ involvement was described by Dehen and colleagues66 in 16 of 76 consecutive patients with morphea with or without associated linear scleroderma. Esophageal abnormalities were present in 7 of 41 patients, and abnormalities of pulmonary function occurred in 9 of 53 patients. Only 2 of the 16 patients with evidence of internal involvement had symptomatic or severe disease, and 1 of these patients was later determined to have systemic sclerosis. The esophageal and
pulmonary abnormalities were mild, were usually asymptomatic, and required no interventions. It was concluded that unless there were symptoms suggesting abnormalities in pulmonary, gastrointestinal, or other systems, there was no need to screen patients for internal organ involvement. 10,66
PATHOLOGY The histologic abnormalities of localized and systemic scleroderma are considered by most investigators to be indistinguishable. Before fibrosis, there may be an intense inflammatory infiltrate with lymphocytes, plasma cells, macrophages, eosinophils, and mast cells. Subsequently, there is an increase in collagen and fibroblasts that leads to increasing sclerosis. In advanced stages, the entire dermis may be replaced by compact collagen fibers. 4 The depth of involvement is important in differentiating the various morphea subtypes (Fig. 20-5).6 Plaque morphea is more superficial, with principal involvement of the dermis and occasionally the panniculus, whereas linear morphea involves the dermis, subcutaneous tissue, muscle, and underlying bone. Deep morphea syndromes tend to spare the superficial dermis and involve the deep dermis, subcutaneous tissue, fascia, or superficial muscle. Eosinophilic fasciitis involves the deep subcutaneous tissues with sclerosis and inflammatory infiltrates while sparing the dermis. Torres and colleagues67 reviewed a total of 51 skin biopsy specimens submitted to their laboratory from 1993 to 1995 from patients with a diagnosis of scleroderma, and they classified the cases into systemic scleroderma or localized disease. They concluded that localized and sys-
papjIl8:j:::::Q~;;';;!~P*K;YY;;;WYii/~;/::/ Reticular dermis
J
1
T
Atrophoderma of Pasini & Pierini Morphea en plaque
1
T
1
Bullous morphea
and Guttate Morphea
···Cc··< . ; -
r'!
I
T
Linear morphea and
I
T
Pansclerotlc morphea of childhood
!
T
-~~l~ ~ __ Facial hlemiatropht----t~!~r-------~ 'v-~" Generaliz~d morphea
,~
Panniculus '<)"
c\'-
and
Keloid
I
I
L.--==:=:::--....,,---------'""--------=tI
Muscle
:l
morphelf'-~l : : ....L.
'
11"'"3
~--,
I~_-
I
'~~' --~_I 1!i~~~~~~~~5~~~~f5=
MAVO
i
Subcutaneous morphea!
i
-
I
l
Morphea profunda
f
I
Eosinophilic ~ , fasciitls )- ~ - , ----..1---~
<.F
I'h:'-' - '''''\1 -'.'. ~ . ~
. !&\). - - - f." J. - . :~'L. --- ) i . \ . , '. :1.:.. . ')....L. \, ,,,,~ \\~'J;. \'\
'
I T
I
.l.
---
....
c.,\~
Regular involvement Occasional involvement
• Rlare ZO-S Schematic diagram of afull-thickness skin biopsy sample. demonstrating location of tissue involvement by the subtypes of localized scleroderma. (Courtesy of Dr. W. P. Daniel Su.)
C HAP T E R
temic scleroderma could be differentiated by the thickness of the dermis and amount of inflammatory infiltrate, both of which were greater in localized scleroderma.
DIFFERENTIAL DIAGNOSIS The most important condition to differentiate from localized scleroderma is systemic disease. Most of the localized disorders in children are linear scleroderma in which the lesions are discrete, limited to a single extremity, and easily differentiated from systemic involvement. The more difficult diagnostic challenge is to differentiate systemic scleroderma from the diffuse and deep forms of morphea with. distal involvement. These patients, in contrast to those with systemic scleroderma, rarely have Raynaud's phe;nomenon and do not develop symptomatic evidence of internal organ involvement. Occasionally, the deep forms of morphea may be confused with juvenile rheumatoid arthritis in that they can manifest with contractures of the hands, arthralgias, and sometimes synovitis and may have a positive rheumatoid factor test restllt. In cases such as these, further testing frequently documents the presence of antinuclear antibodies, anti-histone antibodies, hypergammaglobulinemia, and eosinophilia typical of deep morphea. Erosive joint diseas€ does not occur. Other conditions may mimic localized scleroderma (Table 20-2).6 Morphea has been reported to coexist with systemic scleroderma. Soma and colleagues 68 observed morphea in 9 (6.7%) of 133 patients who presented with systemic scleroderma. They considered mor-
CI
fABLE 20-2
Conditions That Mimi( Morphea
Eo~inophiJia-myalgia syndrome Graft-versus-host disease Agents that induce scleroderma-like diseases Vinyl chloride Bleomycin Pentazocine L.Tryptophan Scleredema adultorum Scleromyxedema Pre:l)1ature aging (Werner's syndrome) Poikiloderma Acrodermatitis chronica atrophicans Di'lbetic cheiroarthropathy Polyneuropathy, Organomegaly, Endocrinopathy, M protein, and Skin changes (POEMS syndrome) Winchester's syndrome Paohydermoperiostosis Phenylketonuria Localized idiopathic fibrosis Acromegaly Progeria Porphyria cutanea tarda Amyloidosis Ca~inoid syndrome Connective tissue hamartomas Growers' panatrophy Connective tissue panniculitis Focal lipoatrophy
Reprinted with permission from Peterson LS, Nelson AM, Su WPD: Classification of morphea (localized scleroderma). Mayo Clin Proc 70: 1068-1076. 1995.
20
LOCALIZED SCLERODERMAS
477
phea to be a part of the skin involvement of this disease. Morphea profunda may manifest as localized areas of induration. In patients in whom little inflammation is apparent, the initial concern may be a localized malignancy.24,2S However, histologic examination of biopsy specimens should easily differentiate morphea profunda from malignancy.
LABORATORY EXAMINATION The diagnosis of localized scleroderma is established on clinical grounds, usually by the physical appearance of the lesions and sometimes aided by biopsy of skin or subcutaneous tissues. Results of laboratory studies are frequently abnormal, raising the question of an associated systemic rheumatic disease. No laboratory abnormality is diagnostic. Results of routine laboratory tests such as a complete blood cell count, blood chemistries, and urinalysis are normal. The erythrocyte sedimentation rate may be increased in the subtypes of the disease with active inflammation, particularly in eosinophilic fasciitis. Eosinophilia and hypergammaglobulinemia are hallmarks of this disorder but also occur in linear scleroderma and the other deep subtypes. Eosinophilia and hypergammaglobulinemia tend to be markers of active disease, and levels normalize as the disease becomes less active. 3,S1 The serum level of immunoglobulin G is increased more often in patients with active and extensive disease and in those with joint contractures.SI Rheumatoid factors are present in 25% to 40% of patients. I1 ,69 The presence or absence of rheumatoid factor seropositivity does not correlate significantly with any particular clinical finding, although higher titers are usually associated with more severe cutaneous and articular involvement.SI Antinuclear antibodies can be present in any of the morphea subtypes, with a frequency ranging from 23% to 73%.4 Takehara and colleagues70 described antinuclear antibody seropositivity in 50% of patients with morphea, 100% of those with generalized morphea, and 67% of those with linear scleroderma. They did not find antibodies to centromere, Scl-70, nuclear ribonucleoproteins, Sm or La (55-B) antigens. Yamane and colleagues7J identified anti-UIRNP antibodies in 3% of patients with localized scleroderma. Anti-histone antibodies were associated with more extensive localized disease. 72 In a study by Rosenberg and colleagues,73 antibodies to denatured DNA were present in 56% of children; 41% had antibodies to high-mobility-group proteins, and 15% had anti-histone antibodies. Anticardiolipin antibodies were identified in 71% of patients with generalized morphea by Sato and colleaguesJ4 Antifibrillin-l antibodies were found in 28% of patients with localized sclerodermaJ5 Deficiency of the second component of complement has been described in patients with en coup de sabre lesions. 76 ,77 In their review of 76 patients with morphea, Dehen and colleagues66 also found that serum complement levels were frequently lower than normal. Elevations of serum aldolase levels in the presence of normal concentrations of creatine kinase have been observed in patients with eosinophilic fasciitis. 57,78,79
478
C HAP T E R
20
LOCALIZED SCLERODERMAS
These levels of enzymatic activity may correlate with activity of the disease.7BSerum concentrations of soluble interleukin-2 receptor have been increased in cases of localized scleroderma and may differentiate active from inactive disease,Bo.Bl although this finding is not supported by all studies. 53 Table 20-3 lists the immunologic abnormalities that have been reported in patients with localized scleroderma. Magnetic resonance imaging may be helpful in delineating areas of involvement: hyperintensity of signal within the fascia on T2-weighted images, fascial enhancement on TI-weighted images, and resolution with clinical improvement in eosinophilic fasciitis. 1o&-1l0 Perhaps the most useful application of magnetic resonance imaging is to document evidence of eNS or orbital involvement. 111 Skin imaging with high-frequency ultrasound has been found to be useful in monitoring therapy in a preliminary study,112 but the role of thermography in determination of disease activity is still unclear. 113 .114
III
TABLE 20- 3 S( l"rod"'lIld
hllllllJllohHli( Abnornldlili"s in LO(dliz('d
Elevated Levels of Orculatlng Cytoldnes or Recepton, or Both Interleukin 13 (82)' Tumor necrosis factor (82, 83) Interleukin 1 (83) Interleukin 6 (83, 84) Interleukin 6 receptor (85) Interleukin 2 (84) Interleukin 4 (84) Interleukin 2 receptor (81, 80) Interleukin 8 (86)
Elevated Levels of Soluble Cell Surface Molecules CD4 (87) CD8 (87) CD 23 (88) CD 30 (89)
Elevated Levels of Orculatlng Adhesion Molecules lCAM-1 (90) VCAM-1 (91)
Endothelial Activation Serum E-selectin (91)
Autoantibodies Antinuclear antibody (70, 73, 92-96) Anti-centromere (92, 97) Anti-histone (72, 73. 92, 98, 99) Anti-high mobility group protein (73) Anti-heat shock protein (73, 100) Anti Fc gamma receptor (101) Anti-mitochondrial 2-oxo-acid dehydrogenase complexes (102) Anti-DNA topoisomerase II alpha (103) Antiphospholipid (74) Lupus anticoagulant (74) Anti-U3SnRNP (104) Anti-UlRNP (71) Anti-ThfrO RNP (105) Anti-fibrillin (75) Anti-single-stranded DNA (73, 93, 95, 106) Rheumatoid factor (96, 107) 'Reference numbers are given in parentheses,
TREATMENT No controlled trials have demonstrated effectiveness of any modality of treatment in patients with localized scleroderma. All recommendations are based on anecdotal reports, case series, and personal experience. Decisions for management must be based on the realization that these disorders are benign in most patients and often spontaneously enter remission after 3 to 5 years.9.10.115.116 Morphea en plaque generally is of cosmetic concern only, and treatments with potentially significant toxicity therefore are not justified. In general, these lesions spontaneously remit with residual pigmentation as the only abnormality. Treatment mainly should be focused on topical therapies such as moisturiZing agents, topical glucocorticoids, or calcipotriene. ll7 Because there is a significant risk for disability in patients with linear morphea and the deep subtypes, systemic treatment should be considered. lO If there is evidence of active disease by laboratory measures such as eosinophilia, hypergammaglobulinemia, or an elevated sedimentation rate and evidence of cutaneous inflammation such as an erythematous halo about the lesions or rapid progression of the disease, glucocorticoids may be indicated, This approach is particularly justified in children with eosinophilic fasciitis. 29,llB There is increasing anecdotal evidence that methotrexate in the range of 0.5 to 1 mg/kg/week is beneficial in morphea. 1l8-120 Additional agents to be considered, particularly when the disease is more indolent, include D-penicillamine l21 and hydroxychloroquine or chloroquine.122.123 Multiple therapeutic successes in treating localized scleroderma have been reported with high-dose ultraviolet Al radiation therapy,124 low-dose ultraviolet Al phototherapy,125 and psoralen plus ultraviolet A therapy,126 making use of phototherapy in the treatment of localized scleroderma appear very promising. 127 This type of therapy, however, must be considered carefully, because the long-term side effects of ultraviolet Al and psoralen plus ultraviolet A therapy in children are unknown. Additional agents that have been used with anecdotal success include cimetidine,12B the antiallergenic drug tranilast,129 and oral calcitriol,130 although a doubleblind, placebo-controlled study subsequently documented no benefit of calcitriol in treating morphea. 131 A double-blind, placebo-controlled study of intralesional interferon-y concluded that this agent was ineffective ,132 Patients with significant involvement from one of the forms of localized scleroderma should have physical therapy directed at counteracting the development of flexion contractures. Surgical reconstruction may be considered, usually after the active phase of the disease has abated, and the child's growth is complete. 133- 136
COURSE OF THE DISEASE AND PROGNOSIS In contradistinction to systemic scleroderma, the prognosis for localized scleroderma is usually benign. The course is that of an early inflammatory phase, with progression to multiple or extensive lesions, then stabiliza-
C HAP T E R
tion, and finally improvement with softening of the skin and increased pigmentation about the lesions. In most instances, the mean duration of activity of disease is 3 to 5 years. 9.IO .46 In a population-based study of localized sclerodenna,lo 50% of the patients had documented skin softening of 50% or more or had disease resolution by 3.8 years after the diagnosis. The 50% resolution point occurred at 2.7 years in the plaque group, 5 years in the generalized and linear subtype groups, and 5.5 years in the deep groups. A small number of patients had active disease for more than 20 years. Prolonged disease activity was associated preponderantly with the linear type. During follow-up, 25% of patients with linear scleroderma and 44% of those with deep morphea had or developed significant disability. In this series, none of the patients progressed to systemic scleroderma; such progression occurs rarely.9.47.59,137 Farrington and colleagues79 reported the long-term outc:ome of 21 pediatric patients with biopsy-proven eosinophilic fasciitis. Two thirds of these patients developed residual cutaneous fibrosis. Children younger than 7 years had a twofold greater risk for progression to cutaneous fibrosis. All of the 14 patients who progressed to cutaneous fibrosis had involvement of three to four extremities; six had truncal involvement. Localized scleroderma should be viewed as a usually benign and self-limited condition with minimal risk of progression to systemic scleroderma. The spectrum of localized scleroderma and systemic scleroderma may be likened to that of discoid lupus erythematosus and systemic lupus erythematosus. It is important to identify the subtypes at risk for disability and to intervene early with appropriate anti-inflammatory medication and physical therapy to minimize the possibility of longterm disability.
REFERENCES 1. F41anga v: Fibrosing conditions in childhood, Adv DermatoI 6: 145-159. 1991. 2, Doyle JA. Connolly SM, Winkelmann RK: Cutaneous and subcutaneous inflammatory sclerosis syndromes, Arch Dermatol 118: 886-890, 1982, 3. Falanga V: Localized scleroderma. Med Clin North Am 73: 1143-1156, 1989. 4. Ul'iel Y, Krafchik BR, Silverman ED, et al: Localized scleroderma in childhood: a report of 30 cases, Semin Arthritis Rheum 23: 328-340, 1994. 5. Scl)achter RK: Localized scleroderma. Curr Opin Rheumatol 1: 505-511, 1989. 6. Peterson LS, Nelson AM, Su WPD: Subspecialty clinics: rheumatology and dermatology. Classification of morphea (localized scleroderma). Mayo Clin Proc 7(j, 1068-1076, 1995. 7, Daoud MS, Su WP, Leiferman KM, et al: Bullous morphea: clinical, pathologic, and immunopathologic evaluation of thirteen cases. J Am Acad Dermatol 30: 937-943. 1994. 8. Su wp. Greene SL: Bullous morphea profunda. Am J Dermatopathol 8: 144-147, 1986. 9. Clilri.tianson HB, Dorsey CS, O'Leary PA. et al: Localized sclerodemla: a clinicil study of two hundred thirty-five cases, Arch Dermatol 74: 629-039, 1956, 10. Peterson LS, Nelson AM. Su WPD, et al: The epidemiology of morphea (localized scleroderma) in Olmsted County 1960-1993. J Rheumatol 24; 73-80, 1997. 11. Kornreich HK, King KK, Bernstein BH, et al: Scleroderma in childhood. Arthritis Rheum 20 (Suppl 2): 343-350. 1977. 12. CljUng MH. Sum J. Morrell MJ, et al: Intracerebral involvement in scleroderma en coup de sabre: report of a case with neuropathologic findings. Ann Neurol 37 679-081, 1995. 13, Goldenstein-Schainberg C, Pereira RM, Gusukuma MC, et al: Childhood linear scleroderma "en coup de sabre" with uveitis. J Pediatr 117: 581-584, 1990. 14. J)oJvid J, Wilson J, Woo P: Scleroderma "en coup de sabre." Ann Rheum Dis 501: 260-262, 1991. 15. Seuw WK, Young W: Localized scleroderma in childhood: review of the literatmre and case report, Pediatr Dem 9: 240-244, 1987.
20
LOCALIZED SCLERODERMAS
479
16, Campbell WW, Bajandas FJ: Restrictive ophthalmopathy associated with linear scleroderma, J Neuroophthalmol 15: 95-97, 1995. 17. Tang RA, Mewis-Christmann L, Wolf J, et al: Pseudo-oculomotor palsy as the presenting sign of linear scleroderma. J Pediatr Ophthalmol Strabismus 23: 236-238, 1986. 18. Suttorp-Schulten MS, Koornneef L: Linear scleroderma associated with ptosis and motility disorders. Br J Ophthalmol 74: 694-695, 1990. 19. Serup J, Alsbirk PH: Localized scleroderma "en coup de sabre" and iridopalpebral atrophy at the same line. Acta Derm Venereol 63: 75-77, 1983. 20. Lewkonia RM, Lowry RB: Progressive hemifacial atrophy (Parry-Romberg syndrome): report with review of genetics and nosology. Am J Med Genet 14: 385-390, 1983. 21. Blaszczyk M, Krolicki L, Krasu M, et al: Progressive facial hemiatrophy: central nervous system involvement and relationship with scleroderma en coup de sabre. J Rheumatol 30: 1997-2004, 2003, 22, Jablonska S: Localized scleroderma. In Jablonska S (ed); Sclerodeffila and Pseudosclerodeffila. Warsaw, Polish Medical Publishers, 1975, pp 277-303. 23. Person JR, Su WP: Subcutaneous morphoea: a clinical study of sixteen cases. Br J Dermatol 100: 371-380, 1979. 24. Su WP, Person JR: Morphea profunda: a new concept and a histopathologic study of 23 cases. Am J Dermatopathol 3: 251-260, 1981. 25. Whittaker SJ, Smith NP, Jones RR: Solitary morphoea profunda. Br.J Dermatol 120: 431--440, 1989. 26. Diaz-Perez JL, Connolly SM, Winkelmann RK: Disabling pansclerotic morphea in children. Arch Dermatol 116: 169--173, 1980. 27. Wollina U, Buslau M, Weyers W: Squamous cell carcinoma in pansclerotic morphea of childhood. Pediatr Dermatol 19: 151-154, 2002. 28. Parodi PC, Riberti C, Draganic Stinco D, et al: Squamous cell carcinoma arising in a patient with long standing pansclerotic morphea. Br J Deffilatol 144: 417--419,2001. 29. Shulman LE: Diffuse fasclltis with hypergammaglobulinemia and eosinophilia: a new syndrome? J Rheumatol 1 (Suppl 1): 46, 1974. 30. Shuirnan LE: Diffuse fasclltis with eosinophilia: a new syndrome? Trans Assoc Am Physicians 88: 7o-B6, 1975. 31. Rodnan GP, Di Bartolomeo A, Medsgar TA Jr: Eosinophilic fasciitis. Report of six cases of a newly recognized scleroderma-like syndrome. Arthritis Rheum 18: 525, 1975. 32. Miller JJ Ill: The fasclltis-morphea complex in children. Am J Dis Child 146: 733-736, 1992. 33, Medsger TA Jr, Masi AT: Epidemiology of systemic sclerosis (scleroderma). Ann Intern Med 74: 714-721, 1971. 34. Mayes MD, Laing 1}, Gillespie BW, et al: Prevalence, incidence and survival rates of systemic sclerosis in the Detroit Metropolitan area. Arthritis Rheum 39: S150, 1996. 35. Levinson JE, Bove KE: Scleroderma. In Gershwin ME, Robbins DE (eds); Musculoskeletal Diseases of Children. Orlando. FL, Grune & Stratton, 1983, pp 195-208. 36. Woo P, David J, Vancheeswaran R, et al: Is scleroderma the same disease in childhood? Arthritis Rheum 35: S137, 1992. 37. Kahari VM, Sandberg M, Kalimo H, et al: Identification of fibroblasts responsible for increased collagen production in localized scleroderma by in situ hybridization. J Invest Dermatol 90: 664-670, 1988. 38. Liu B, Connolly MK: The pathogenesis of cutaneous fibrosis. Semin CUlan Med Surg 17: 3-11, 1998. 39. Janin A, Socie G, Devergie A, et al: Fasciitis in chronic graft-versus-host disease: a clinicopathologic study of 14 cases. Arm Intern Med 120: 993-998, 1994. 40. Aractingi S, Socie G, Devergie A: Localized scleroderma-like lesions on the legs in bone marrow transplant recipients: association with polyneuropathy in the same distribution. Br J Dermatol 129: 201-203, 1993, 41. McNallan K, Aponte C, EI Azhary R, et al: Immunophenotyping of chimeric cells and surrounding cellular infiltrates in linear sclerodenna and morphea skin tissue. Arthritis Rheum 48 (Suppl 9): S512, 2003, 42, Haustein UF, Haupt B: Drug-induced scleroderma and sclerodermiform conditions. Clin Dermatol 16: 353-366, 1998. 43. Martin RW, Duffy J, Engel AG: The clinical spectrum of the eosinophilia-myalgia syndrome associated with L-tryptophan ingestion, Clinical features in 20 patients and aspects of pathophysiology, Arm Intern Med 113: 124-134, 1990. 44, Mayeno AN, Lin F, Foote CS, et al: Characterization of "peak E." a novel amino acid associated with eosinophilia-myalgia syndrome, Science 250: 1707-1708, 1990. 45. Aberer E, Neumann R, Stanek G: Is localised scleroderma a Borrelia infection? Lancet 2: 278, 1985. 46. Aberer E, Stanek G, Ertl M, et al: Evidence for spirochetal origin of circumscribed scleroderma (morphea). Acta Derrn Venereol 67: 225-231, 1987, 47, Aberer E, Klade H, Stanek G, et al: Borrelia burgdorferi and different types of morphea. Dermatologica 182: 145-154, 1991. 48. Fan W, Leonardi CL, Penneys NS: Absence of Borrelia burgdorferi in patients with localized scleroderma (morphea). J Am Acad Dermatol 33: 682--Q84, 1995. 49, Dillon WI, Saed GM, Fivenson DP: Borrelia burgdorferi DNA is undetectable by polymerase chain reaction in skin lesions of morphea, scleroderma, or lichen sclerosus et atrophicus of patients from North America. J Am Acad Dermatol 33: 617--{j20, 1995.
480
C HAP T E R
20
LOCALIZED SCLERODERMAS
50. Weide B, Schittek B, Klyscz T, et al: Morphoea is neither associated with features of Borrelta burgdoifert infection, nor L. this agent detectable in lesional skin by polymerase chain reaction. Br J Dermatol 143: 780-785, 2000. 51. Falanga V, Medsger TA Jr, Reichlin M, et al: Unear scleroderma: clinical spectrum. prognosis, and laboratory abnormalities. Ann Intern Med 104: 8494l57, 1986. 52. Curtis AC, Jansen TG: The prognosL. of localized scleroderma. Arch Dermatol 78: 749-757, 1958. 53. Vancheeswaran R, Black CM, David J, et al: Childhood-onset scleroderma: is it different from adult-onset disease? Arthritis Rheum 39: 1041-1049, 1996. 54. Gollob MH, Dekoven JG, Bell MJ, et al: Post-radiation morphea. J Rheumatol 25: 2267-2269, 1998. 55. Ardern-Jones MR, Black MM: Widespread morphoea following radiotherapy for carcinoma of the breast. Clin Exp Dermatol 28: 160-162, 2003. 56. Michet CJ Jr, Doyle JA, Ginsburg WW: Eosinophilic fasciitis: report of 15 cases. Mayo Clin Proc 56: 27-34, 1981. 57. Winkelmann RK, Connolly SM, Doyle JA: Carpal tunnel syndrome in cutaneous connective tissue disease: generalized morphea, lichen sclerosus, fasciitis, discoid lupus erythematosus, and lupus panniculitis. J Am Acad Dermatol 7: 94-99, 1982. 58. Jarr'dtt M, Bybee JD, Ramsdell W: Eosinophilic fasciitis: an early variant of scleroderma. J Am Acad Dermatoi 1: 221-226, 1979. 59. Yamamoto A, Morita A, Shintani Y, et al: Localized linear scleroderma with cutaneous calcinosis. J of Dermatol 29: 112-114, 2002. 60. Jinnin M, Ihn H, Asano Y, et al: A case of linear scleroderma with muscle calcification. Br J of Dermatol 146: 1084-1086, 2002. 61. Blaszczyk M, Janniger CK, Jablonska S, et al: Childhood scleroderma and its peculiarities. Cutis 58: 141-144, 148-152, 1996. 62. Barton DH, Henderson HZ: Oral-facial characteristics of circumscribed scleroderma: case report. J Clin Pediatr Dent 17: 239-242, 1993. 63. Linman BH: Linear scleroderma: a response to neurologic injury? Report and literature review. J Rheumatol 16: 1135-1140, 1989. 64. Pupilla G, Andermann F, Dubeau F: Linear scleroderma and intractable epilepsy: neuropathologic evidence for a chronic inflammatory process. Ann Neurol 39: 277-278, 1996. 65. Marzano A, Menni S, Parodi A, et al: Localized scleroderma in adults and children, clinical and laboratory investigations of 239 cases. Eur J Dermatol 13: 171-176,2003. 66. Dehen L, Roujeau J, Cosnes A, et al: Internal involvement in localized scleroderma. Medicine 73: 241-245, 1994. 67. Torres JE, Sanchez JL: Histopathologic differentiation between localized and systemic scleroderma. Am J Dermatopathol 20: 242-245, 1998. 68. Soma Y, Tamaki T, Kikuchi K, et al: Coexistence of morphea and systemic sclerosis. Dermatology 186: 103-105, 1993. 69. Torok E, Ablonczy E: Morphoea in children. Clin Exp Dermatol 11: 607-612, 1986. 70. Takehara K, Moroi Y, Nakabayashi Y, et al: Antinuclear antibodies in localized scleroderma. Arthritis Rheum 26: 612-616, 1983. 71. Yamane K, Ihn H, Kubo M, et al: Anti-UIRNP antibodies in patients with localized scleroderma. Arch Dermatol Res 293: 455-459, 2001. 72. Sato S, Fujimoto M, Ihn H, et al: Clinical characteristics associated with antihistone antibodies in patients with localized scleroderma. J Am Acad Dermatol 31: 567-571, 1994. 73. Rosenberg AM, UZiel Y, Krafchik BR, et al: Antinuclear antibodies in children with localized scleroderma. J Rheumatol 22: 2337-2343, 1995. 74. Sato S, Fujimoto M, Hasegawa M, et al: Antiphospholipid antibody in localised scleroderma. Ann Rheum Dis 62: 771-774, 2003. 75. Arnett FC, Tan FK, Uziel Y, et al: Autoantibodies to the extracellular matrix microfibrillar protein, fibrillin I, in patients with localized scleroderma. Arthritis Rheum 42: 2656-2659, 1999. 76. Hulsmans RF, Asghar SS, Siddiqui AH, et al: Hereditary deficiency of C2 in association with linear scleroderma "en coup de sabre." Arch Dermatol 122: 76-79, 1986. 77. Venneker GT, van Meegan M, deKok-Nazaruk M, et all Incomplete functional deficiencies of the fourth (C4) and second (C2) components of complement in a patient with linear frontoparietal scleroderma and his family. Exp Clin Immunogenet 13: 104-111, 1996. 78. Fujimoto M, Sato S, Ihn H, et al: Serum aldolase level is a useful indicator of disease activity in eosinophilic fasciitis. J Rheumatol 22: 563-565, 1995. 79. Farrington ML, Haas JE, Nazar-Stewart V, et al: Eosinophilic fasciitis in children frequently progresses to scleroderma-like cutaneous fibrosis. J Rheumatol 20: 128-132, 1993. 80. 'Uziel Y, Krafchik BR, Feldman B, et al: Serum levels of soluble interleukin2 receptor. A marker of disease activity in localized scleroderma. Arthritis Rheum 37: 898-901, 1994. 81. Ihn H, Sato S, Fujimoto M, et al: Clinical significance of serum levels of soluble interleukin-2 receptor in patients with localized scleroderma. Br J Dermatol 134: 843-847, 1996. 82. Hasegawa M, Sato S, Nagaoka T, et al: Serum levels of tumor necrosis factor and interleukin-13 are elevated in patients with localized scleroderma. Dermatology 207: 141-147, 2003, 83. Alecu M, Geleriu L, Coman G, et al: The interleukin-1, interleukin-2, interleukin-6 and tumour necrosis factor alpha serological levels in localised and systemic sclerosis. Rom J Intern Med 36: 251-259, 1998,
84. Ihn H, Sato S, Fujimoto M, et al: Demonstration of interleukin-2, interleukin4 and interleukin-6 in sera from patients with localized scleroderma. Arch Dermatol Res 287: 193-197, 1995, 85. Nagaoka T, Sato S, Hasegawa M, et al: Serum levels of soluble interleukin 6 receptor and soluble gp130 are elevated in patients with localized scleroderma. J Rheumatol 27: 1917-1921,2000. 86. Ihn H, Sato S, Fujimoto M, et al: Demonstration of interleukin 8 in serum samples of patients with localized scleroderma. Arch Dennatol 130: 1327-1328, 1994. 87. Sato S, Fujimoto M, Kikuchi K, et al: Soluble CD4 and CD8 in serum from patients with localized scleroderma. Arch Dermatol Res 288: 358-362, 1996. 88. Sato S, Fujimoto M, Kikuchi K, et al: Elevated soluble CD23 levels in the sera from patients with localized scleroderma. Arch Dermatol Res 288: 74-78, 1996. 89. Ihn H, Yazawa N, Kubo M, et al: Circulating levels of soluble CD30 are increased in patients with localized scleroderma and correlated with serological and clinical features of the disease. J Rheumatol 27: 698-702, 2000. 90. Ihn H, Fujimoto M, Sato S, et al: Increased levels of circulating intercellular adhesion molecule-I in patients with localized scleroderma. J Am Acad Dermatol 31: 591-595, 1994. 91. Yamane K, Ihn H, Kubo M, et all Increased serum levels of soluble vascular cell adhesion molecule 1 and E-selectin in patients with localized scleroderma. J Am Acad Dermatol 42: 64-69, 2000. 92. Garcia-de la Torre I, Castello-Sendra J, Esgleyes-Ribot T, et al: Autoantibodies in Parry-Romberg syndrome: a serologic study of 14 patients. J Rheumato[ 22: 73-77, 1995. 93. Falanga V, Medsger TA Jr, Reichlin M: Antinuclear and anti-single-stranded DNA antibodies in morphea and generalized morphea. Arch Dermatol 123: 350-353, 1987. 94. Woo TY, Rasmussen JE: Juvenile linear scleroderma associated with serologic abnormalities. Arch Dermatol 121: 1403-1405, 1985. 95. Falanga V, Medsger TA, Reichlin M: High titers of antibodies to singlestranded DNA in linear scleroderma. Arch Dermatol 121: 345-347, 1985. 96. Hanson V, Kornreich HK, Drexler E, et al: Some immunologic considerations in focal scleroderma and progressive systemic sclerosis in children. Pediatr Res 8: 806-809, 1974. 97. Ruffatti A, Peserico A, Glorioso S, et al: Antkentromere antibody in localized scleroderma. J Am Acad Dermatol 15: 637-642, 1986. 98. Sato S, Fujimoto M, Ihn H, et al: Antigen specificity of antihistone antibodies in localized scleroderma. Arch Dermatol 130: 1273-1277, 1994. 99. Sato S, Ihn H, Soma Y, et all Antihistone antibodies in patients with localized scleroderma. Arthritis Rheum 36: 1137-1141, 1993. 100. Fujimoto M, Sato S, Ihn H, et all Autoantibodies to the heat-shock protein hsp73 in localized scleroderma. Arch Dermatol Res 287: 581-585, 1995. 101. Davis K, Boros P, Keltz M, et al: Circulating Fe gamma receptor-specific autoantibodies in localized and systemic scleroderma. J Am Acad Dennatol 33: 612-616, 1995. 102. Fujimoto M, Sato S, Ihn H, et al: Autoantibodies to mitochondrial 2-oxo-acid dehydrogenase complexes in localized scleroderma. Clin Exp Immunol 105: 297-301, 1996. 103. Hayakawa I, Hasegawa M, Takeham K, et al: Anti-DNA topoisomerase 11 alpha autoantibodies in localized scleroderma. Arthritis Rheum 50: 227-232, 2004. 104. Yimane K, Ihn H, Kubo M, et al: Anti-U3 snRNP antibodies in localised scleroderma. Ann Rheum Dis 60: 1157-1158, 2001. 105. Yamane K, Ihn H, Kubo M, et al: Antibodies to TIl/TO ribonucleoprotein in patients with localized scleroderma. Rheumatology (oxford) 40: 683-686, 2001. 106. Ruffatti A, Peserico A, Rondinone R, et al: Prevalence and characteristics of anti-single-stranded DNA antibodies in localized scleroderma. Comparison with systemic lupus erythematosus. Arch Dermatol 127: 1180-1183, 1991. 107. Hanson V, Drexler E, Kornreich H: Rheumatoid factor (anti-gamma-globulins) in children with focal scleroderma. Pediatrics 53: 945-947, 1974. 108. Nakajima H, Fujiwara S, Shinoda K, et al: Magnetic resonance imaging and serum aldolase concentration in eosinophilic fasciitis. Intern Med 36: 654-656, 1997, 109. AI-Shaikh A, Freeman C, Avruch L, et al: Use of magnetic resonance imaging in diagnosing eosinophilic fasciitis. Report of two cases. Arthritis Rheum 37: 1602-1608, 1994. 110. Liu P, Uziel Y, Chuang S, et al: Localized scleroderma: imaging features. Pediatr Radiol 24: 207-209, 1994. 111. Ramboer K, Dermaerel P, Baert AL, et al: Linear scleroderma with orhital involvement: follow up and magnetic resonance imaging [letter], Br J Ophthalmol81: 90-93, 1997. 112. Szymanska E, Nowicki A, Mlosek K, et al: Skin imaging with high frequency ultrasound-preliminary results. Eur J Ultrasound 12: 9-16, 2000. 113. Martini G, Murray KJ, Howell KJ, et al: Juvenile-onset localized scleroderma activity detection by infrared thermography. Rheumatology 41: 1178-1182, 2002. 114. Birdi N, Shore A, Rush P, et al: Childhood linear scleroderma: a possible role of thermography for evaluation. J Rheumatol 19: 968-973, 1992. 115. Chazen EM, Cook CD, Cohen J: Focal scleroderma. J Pediatr 60: 385-393, 1962. 116. Gaffney K, Kearns G, Moraes 0, et al: Eosinophilic fasciitis: a good response with conservative treatment. Ir J Med Sci 162: 256-257, 1993. 117. Cunningham BB, Landells 10, I.angman C, et al: Topical calcipotriene for morphea/linear scleroderma. J Am Acad Dermatol 39: 211-215, 1998.
C HAP T E R 118. Uziel Y, Feldman B, Krafchik BR, et al: Methotrexate and corticosteroid therapy for pediatric localized scleroderma. J Pediatr 136: 91-95, 2000. 119. Janzen L, Jeffery JR, Gough J, et al: Response to methotrexate in a patient ,with idiopathic eosinophilic fasciitis, morphea, IgM hypergammaglobulinemia. and renal involvement. J Rheumatol 22: 1967-1970, 1995. 120. Seyger MMB, van den Hoogen FHJ, de Boo T, et al: Low-dose methotrexate in the treatment of widespread morphea. J Am Acad Dennatol 39: 220-225, 1998. 121. Krafchik BR: Localized cutaneous scleroderma. Semin Dennatol ll: 65-72, 1992. 122. Lakhanpal S, Ginsburg WW, Michet CJ, et al: Eosinophilic fasciitis: clinical 'spe~trum and therapeutic response in 52 cases. Semin Arthritis Rheum 17: 221-231, 1988. 123. Weiss JS: Antimalarial medications in dermatology: a review. Dermatol Clin 9: 377-385, 1991. 124. Stege H, Berneburg M, Humke S, et al: High-dose UVA, radiation therapy for ;localized scleroderma. J Am Acad Dennatol 36: 938--944, 1997. 125. Kerscher M, Volkenandt M, Gruss C, et al: Low-dose UVA I phototherapy for treatment of localized sc1erodenna. J Am Acad Dennatol 38: 21-26, 1998. 126. Kanekura T, Fukumaru S, Matsushita S: Successful treatment of scleroderma with PUVA therapy. J Dermatol 23: 455-459, 1996. 127. !'isher G, Kang S: Phototherapy for scleroderma: biologic rationale, results, and promise. Curr Opin in Rheumatol 14: 723-726, 2002.
20
LOCALIZED SCLERODERMAS
481
128. Naschitz JE, Boss JH, Misselevich I, et al: The fasciitis-panniculitis syndromes. Clinical and pathologic features. Medicine (Baltimore) 75: 6-16, 1996. 129. Taniguchi S, Yorifuji T, Hamada T: Treatment of linear localized scleroderma with the anti-allergic drug, tranilast. Clin Exp Dermatol 19: 391-393, 1994. 130. Hulshof MM, Pavel S, Breedveld FC, et al: Oral calcitriol as a new therapeutic modality for generalized morphea. Arch Dermatol 130: 1290-1293, 1994. 131. Hulshof M, Bouwes Bavinck J, Bergman W, et al: Double-blind, placebocontrolled study of oral calcitriol for the treatment of localized and systemic sclerodenna. Am Acad Dermatol 43: 1017-1023,2000. 132. Hunzelmann N, Anders S, Fierlbeck G, et al: Double-blind, placebo-controlled study of intralesional interferon gamma for the treatment of localized scleroderma. J Am Acad Dermatol 36: 433-435. 1997. 133. Sengezer M, Deveci M, Selmanpakoglu N: Repair of "coup de sabre," a linear form of sclerodenna. Ann Plast Surg 37: 428--432, 1996. 134. Slavin SA, Gupta S: Reconstruction of scleroderma of the breast. Plast Reconstr Surg 99: 1736-1741, 1997. 135. Lyle W, Jerath R: Severe wrist contracture in a child with linear scleroderma. Ann Plast Surg 47: 465-467, 2001. 136. Lapiere J, Aasi S, Cook B, et al: Successful correction of depressed scars of the forehead secondary to trauma and morphea en coup do sabre by en bloc autologous dermal fat graft. Dermatol Surg 26: 793-797, 2000. 137. Mayorquin FJ, McCurley TL, Levernier JE, et al: Progression of childhood linear scleroderma to fatal systemic sclerosis. J Rheumatol 21: 1955-1957, 1994.
( HAP T E R
21
OVERLAP SYNDROMES James T. Cassidy and Ross E. Petty
;)f
A number of uncommon systemic connective tissue diseases (CTDs) present simultaneously with signs and symptoms that are characteristic of two or more of the major rheumatic disorders, such as juvenile rheumatoid arthritis (JRA), systemic lupus erythematosus (SLE), juvenile dermatomyositis (JDM), cutaneous systemic scleroderma (CSS), and vasculopathy. Children with these disorders are often difficult to categorize under existing classification criteria and are properly referred to as having overlap syndromes or undifferentiated CTD. Two of the defined disorders in these categories are discussed in this chapter: mixed connective tissue disease (MCTD) and Sjogren's syndrome. Overlap syndromes specific for the other major CTDs are discussed in their respective chapters.
MIXED CONNECfIVE TISSUE DISEASE Definition and Casslflcatlon MCTD was initially described by Sharp and colleagues in 1972 1 in 25 adults as a disorder with an excellent initial response to relatively low-dose glucocorticoid therapy and a favorable prognosis. The syndrome included clinical features of rheumatoid arthritis, scleroderma, SLE, and dermatomyositis in conjunction with a high antibody titer to an extractable nuclear antigen (ENA). However, reassessment of the original patients indicated that the inflammatory manifestations (e.g., arthritis, serositis, fever, myositis) tended to become less evident over time, whereas sclerodactyly and esophageal disease, less responsive to treatment with glucocorticoids, persisted and began to dominate the clinical picture. 2 Severe renal disease continued to remain an unusual feature. Although the concept of MCTD as a clinical entity separate from the other CTDs has remained controversial, classifications employing more precise serologic criteria and human leukocyte antigen (HLA) typing have confirmed the uniqueness of this disorder (Table 21-1).3-5
Epidemiology MCTD is one of the least common disorders in a pediatric rheumatology clinic. It had a frequency of 0.1% in a Finnish nationwide prospective study> and 0.3% in the u.s. Pediatric
482
Rheumatology Database? The median age at onset was approximately 11 years (range, 4 to 16 years). MCTD occurred three times more frequently in girls than in boys. There was one report of this disorder occurring in siblings. 8
Immunogenetic Background In white populations, the predominant HLA class II specificities associated with MCTD have been DR4 and DR2. 5,9-16 Patients with DR4 or DR2 have a region of homology of seven amino acids (numbers 26, 28, 30, 31, 32, 70, and 73) in the highly polymorphic antigen-binding segment of the DRBI gene (RIA-DRB]).15,17 These HLA specificities are also linked to antibodies to the uridine-rich small nuclear ribonucleoprotein (UlsnRNP) that are characteristic of the disorder. HLA types associated with CSS (DRS) or SLE (DR3) are uncommon. Unique T-cell-dependent immune responses also occur. 18,19 T-cell clones are directed against the 70-kd polypeptide of UlsnRNP (Ul-70kD) and are principally of the CD4+, Thl type. These data support the role of a specific HLA immunogenetic profile and T-cell reactivity in generation of the B-cell immune responses characteristic of the disorder.
Clnlcal Manifestations MCTD has been recognized with increasing frequency in childhood. 13 ,2°-32 These children present with features of more than one CTD, a speckled antinuclear antibody (ANA) pattern, and high titers of antibody to ribonucleoprotein (RNP) (see Table 21-1). Clinical characteristics of children from selected studies are summarized in Table 21-2. Polyarthritis (93%) and Raynaud's phenomenon (85%) are the most common manifestations at onset. The arthritis may be relatively painful; erosive disease is uncommon, but deformity may develop with flexion contractions or swan-neck deformities. The arthritis is often associated with rheumatoid factor (RF) seropositivity that is often present early in approximately two thirds of the children. Cutaneous changes include scleroderma-like disease in one half, the rash of SLE in one third, and that of JDM in one third of MCTD patients. Nail fold capillary abnormalities are similar to those in CSS (see Table 19-5).3-~""36
C HAP T E R
1m
_II IABI E 21
I
(Iinical ChardCleristics of Mixed (onnective
ll'>sue Disease
Ch'dcal Signs of It Lust Two of the following DlselSlS
Juvenile rheumatoid arthritis Systemic lupus erythematosus Juvenile dermatomyositis Systemic scleroderma
PotItIve SerologIc FIndIngs
PnIsence of HLA·DR4 or DR2 IDA, human leukocyte antigen; UlsnRNP, uridine-rich small nuclear rihonucleoprotein; Ul RNA, Ul ribonucleic acid.
Cardiopulmonary disease and esophageal dysmotility occur infrequently.37 Vasculitis can occur and be severe (i,e" transverse myelopathy), Although nephritis occurs in about one fourth of the patients with MCTD, it is less common and usually less severe than in those with SLE. However, children with MCTD may have more frequent and more severe renal disease than adults, more hematologic complications such as thrombocytopenia, and less pulmonary hypertension,38 In a prospective longitu-
-
II TABLE 21 2 Oisease Charaderisli<s of Children with Mixed (onneclive Iissue Disease
~cterlstlc
Arthritis Raynaud's phenomenon Sclerodermatous skin Ra$h of systemic lupus erythematosus Rash of dermatomyositis Fever Abnormal esophageal motility Cardiac disease Pericarditis Muscle disease Sicea syndrome Central nervous system disease Lung Abnormal diffusion Restrictive disease Hypertension Effusion Radiographic changes only Splenomegaly Hepatomegaly Renal disease Anli-dsDNA positive Anti-Sm positive Anli-RNP positive Rheumatoid factor positive
OVERLAP SYNDROMES
483
dinal study, 31 of 34 adults with high titers of RNP antibody had typical MCTD in which pulmonary disease, often initially asymptomatic, was common,39 Pulmonary hypertension was the most frequent serious complication. A comparison of the clinical and serologic characteristics of diffuse and limited CSS, eosinophilic fasciitis, MCTD, JDM and SLE is prOVided in Table 19-14.
Pathology
High-titer of antibodies to U1snRNP and the 70-kD A and C polypeptides Anti-UlRNA antibodies
11;
21
No. Reported*
No.
Presentt Percent 67 61 33 22
93 85 49 33
22 34 23 20 18 44 24 14
33 56 41 30 27 61 36 23
64 58
24 8 4 13 1 18 19 19 13 6
43 14 7 23 2 29 28 26 20
72
72
57
39
100 68
72 72
67 67 67 61 56 67 67 72
66 61 56 56 56 56 56 63 68 72
10
'The number of children in whom the characteristic was identified. 'The number in whom the abnormality was present. dsDNA, double-stranded DNA; RNP, ribonucleoprotein; Sm, Smith antigen. Data .from 10 reports with a total of 72 children: references 1, 20, 21, 22, 23, 24, 27, 211. 48, 49
Widespread intimal proliferation and medial hypertrophy of vascular walls have been described in four children with MCTD who died. 40 Renal biopsies in eight additional patients confirmed abnormalities of the glomerular basement membrane or vascular sclerosis. These investigators commented that although the histopathology of MCTD resembled that of CSS, the extent of fibrosis was less, and intimal vascular abnormalities in larger vessels such as the aorta and the coronary, pulmonary, and renal arteries were more prominent. Another study reported pulmonary hypertension and proliferative vasculopathy in the virtual absence of interstitial fibrosis in patients with MCTD in contrast to those with CSS.39
Differential Diagnosis MCTD in children is a syndrome that characteristically evolves over time from a more limited presentation of clinical disease to one with overlapping features of JRA, SLE, CSS, or JDM. Initial abnormalities are likely to include polyarthritis, Raynaud's phenomenon, and fever. Other manifestations develop sequentially, but not in any predictable order, or over any circumscribed period, The rashes of SLE or JDM are common at onset. Sclerodermatous skin changes are slow to develop but may become the most prominent feature of the disease late in its course. Moderately asymptomatic involvement, such as myositis with minimal weakness, mild atrophy, and minimal to moderate increases in the serum muscle enzyme concentrations, is common. Dysphagia and bowel dysmotility also may occur. Manifestations of the sicca syndrome with xerostomia, keratoconjunctivitis sicca, or parotid gland enlargement occur in one third of the children. 41 Although children do not usually complain of shortness of breath (depending on cognitive development or unconsciously self-imposed restriction of activity), they often have pulmonary functional impairments. Serologic findings may evolve over time. ANAs may be absent at onset or be found in very low titers in a speckled pattern. Development of high-titer anti-RNP antibodies that are RNAse-sensitive should alert the pediatric rheumatologist to the probable evolution of the child's illness into the pattern of MCTD. Criteria for MCTD have been evaluated for adulL<; but not for children (Table 21_3).42--45 These criteria are summarized in the review by Smolen and colleagues. 16 Shen and coworkers46 studied 50 patients from China during a 2- to 8-year period and indicated that the criteria of Sharp42 were the most reliable for the diagnosis of MCTD. Among 23 patients fulfilling these criteria, only 1 (4.3%) developed scleroderma. Among 23 patients satisfying
484
t.
C HAP T E R
lABL [21 3
2I
OVERLAP SYNDROMES
DidlJllOStit Cril('rid for Mixl'd (Olllll't livl' TisslIl' Dbl'dSI'
Major Crlterla 1. Severe myositis 2. Pulmonary involvement, with one or more of the following: OLeo < 70% normal, pulmonary hypertension, or proliferative vascular lesions on lung biopsy 3. Raynaud's phenomenon or esophageal hypomotility 4. Swollen hands or sclerodactyly 5. Highest observed anti-ENA titer ~ 1:10,000 and anti-RNP positive
Minor CrIteria 1. Alopecia 2. Leukopenia «4000 WBC/mm3) 3. Anemia (SlO g/dL for women, SI2 g/dL for men) 4. Pleuritis 5. Pericarditis 6. Arthritis 7. Trigeminal neuropathy 8. Malar rash 9. Thrombocytopenia (<100,000/mm3)
10. Mild myositis 11. History of swollen hands
Diagnosis
Exduslon
Definite
Definite
A. Four major criteria; anti-UIRNP positive with anti-ENA ~ 1:4000
Positive for anti-Sm antibodies (by immunodiffusion)
Probable
Probable A. Three major criteria None B. Two major criteria (including at least one or more of nos. 1, 2, and 3) plus two minor criteria Anti-UIRNP positive with anti-ENA ~ 1:1000 Possible Possible A. Three major criteria; no other None requirements B. Two major criteria; plus anti-UIRNP positive with anti-ENA ~ 1:100 C. One major and three minor criteria; plus anti-UIRNP positive with anti-ENA ~ 1:100
OLeo' single-breath carbon monoxide diffusing capacity of the lungs: ENA, extractable nuclear antigen: UIRNP, uridine-rich ribonucleoprotein. Adapted from Sharp GC: Diagnostic criteria for classification of Mcm. In Kasukawa R, Sharp GC: Mixed Connective Tissue Disease and Anti-Nuclear Antibodies. Amsterdam, Excerpta Medica, 1987, pp 23-32.
the criteria of Kasukawa and colleagues,43 7 (30.4%) developed another cm. Of the 27 who met the criteria of Alarcon-Segovia and colleagues,41 12 (44%) went on to fulfill classification criteria for another major rheumatic disease. The frequencies of HLA-DR4 and -DRS were significantly higher among the patients whose disease fulfilled Sharp's criteria. 42 Different conclusions were reached, however, in a comparative study of four diagnostic criteria from France by Amigues and coworkers. 47 These investigators analyzed the criteria of Sharp,42 Kasukawa and colleagues,43 Alarcon-Segovia and coworkers,44 and Kahn and associates4S in 45 patients with anti-U1RNP antibodies who were classified as having MCm, They found that the criteria of Alarcon-Segovia and coworkers44 had the highest sensitivity (62.5%) and specificity (86.2%), with an overlap of 16% with other major cms. These results were comparable to those obtained with the criteria of Kahn and associates. 4s
70K A 8'/8 D
m.w.
-94 -67 -43
Laboratory Examination Very high titers of ANAs are usually present initially, often in a speckled pattern on HEp-2 cell substrate. These antibodies react specifically with an RNAsesensitive component of ENA and RNP. Anti-RNP antibodies in high titer have been the serologic hallmark of MCTD, but these antibodies may be present in low titers in other diseases such as SLE. 16,21.48 Further investigations confirmed that the most characteristic specificities of the anti-RNP antibodies in MCTD were directed against a uridine-rich CUl), small nuclear RNP CsnRNP) complex (UlsnRNP) of the spliceosome consisting of UIRNA and the associated 70-kD A and C polypeptides (Fig. 21-1).49-51 The anti-U1snRNP profile of patients with MCTD is characterized by a high-titer antibody response, predominantly or solely of immunoglobulin (Ig) G antibodies, and specificity for an epitope different from that of SLE sera16; a false-positive anti-U1snRNP antibody response
-30
-20.1 -14.4
Westem blot results, with the patients' sera delineating a reaction with the 70-kD, A, B/B', and Dpolypeptides.
• FllIIIre 21-1
CHAPTER
may be observed in cases of SLE. 52 Later reports indicate that antibodies to UIRNA are even more closely associated clinically with disease activity during the course of MCtD than are UIRNP antibodies. 53-55 Substantial advances have been made in employing specific autclantibody activities against the UlsnRNP polypeptides for classification of disease. 15 ,56 Among adult patients with high titers of autoantibodies against the UI-70kD antigen, in contradistinction to patients with CTDs who did not demonstrate these antibody activities, there were significant clinical associations with Raynaud's phenomenon, swollen hands, sclerodactyly, telangiectasia, and abnormal esophageal motility. 14 In the study by DeRooij and colleagues,13 all five children who had antibodies against the UI-70kD antigen had clinical disease characterized by arthralgia or arthritis, swollen hands, Raynaud's phenomenon, and abnormalities of pulmonary funCtion. Children with antibodies against the 70-kD polypeptide uncommonly develop diffuse glomerulonephritis, cardiac disease, widespread cutaneous sclerosis or central nervous system disease. Clinical renal disease occurred, however, in 3 of 11 patients reported by Hoffman and colleagues9 and was confined to a subset of children who had IgG autoantibodies against the D polypeptide of UlsnRNP. All of these children were HLA-DR2 positive and were the same patients who developed an adverse or fatal outcome associated with antidsDNA antibodies. Children with anti-Sm antibodies may develop reactivity to the D polypeptide and clinically important renal disease. Their course may be accompanied by hypClcomplementemia and other features of nephritis more chal",lcteristic of SLE. Their sera may not be positive for antiSm antibodies by the relatively insensitive techniques of double immunodiffusion or counterimmunoelectrophoresis. Some MCTD sera, including those from patients without renal disease, also react with BIB' polypeptides, but probably to one or more epitopes different from those characteristic of the anti-Sm antibody activity found in SLE.56.57 Some patients have marked elevations of serum immunoglobulin levels, especially of IgG.22.27 Two children with selective 19A deficiency have been reported. 2o.48 In a study of adults with MCTD,58 48 anti-UI-70kD antibody-positive patients with MCTD were compared with 59 anti-Ul-70kD antibody-negative patients with classic SLE. Although levels of antiphospholipid antibodies were increased in the patients with MCTD compared with control subjects, levels of these antibodies were even higher in the patients with SLE who had clinical manifestations of the antiphospholipid antibody syndrome in whom deep vein thrombosis, pulmonary embolism, recurrent fetal loss, chorea, Iivido reticularis, severe thrombocytopenia, and avascular necrosis occurred. In a prospective investigation of 11 children with MCTD,9 antibpdies to UlsnRNP polypeptides were compared sequentially during the course of the disease. All patients had high-titer anti-ENA antibodies determined by hemagglutination (>1:1,000,000) and positive anti-RNP reactivity by immunodiffusion. Antigenic specificity identified by immunoblot analysis and enzyme-linked immunosorbent assay was to the 70-kD polypeptide in 11, A in 10, C in 2, BIB' in 9, and D in 3 patients. Four children had both IgG and IgM antibodies to the 70-kD protein, 5 to the A peptide, 2 to BIB', and only 1 to D. One patient developed low-titer anti-Sm antibodies, and 3 developed low-titer, transient anti-dsDNA seropositivity. Three had anti-Ro/SS-A antibodies. Anti-ENA titers and 70-kD reactivity decreased in patients during prolonged remission; positive reactions remained in those with continuing active disease and in patients receiving only symptomatic treatment. The predominant HLA antigens were DR2 in 6 of 9 and DR4 in 4 of 9; all
21
OVERLAP SYNDROMES
485
patients were either DR2 or DR4 positive, similar to findings in adults with MCTD.
Treatment There is no specific treatment for MCTD. Management should address the predominant problems of the child, such as arthritis, cutaneous disease, or visceral involvement. Many children respond satisfactorily to low-dose glucocorticoids, nonsteroidal anti-inflammatory drugs, or hydroxycWoroquine or a combination of these medications. 48 Patients with severe myositis or renal or visceral disease generally require high-dose glucocorticoids and sometimes require cytotoxic drugs (cyclophosphamide), especially for life-threatening complications such as pulmonary hypertension. Methotrexate has been advocated. 59 Autologous hemopoietic stem-cell transplantation has been attempted for refractory, life-threatening disease. 60
Course of the Disease and Prognosis The long-term outcomes of children with MCTD are varied and unpredictable. 48.61-M Deaths have been reported from disease resembling that of SLE accompanied by renal failure. In contrast to SLE, however, morbidity and mortality in MCTD are more often associated with development of pulmonary hypertension (7%)25.28.39.48.65 or gradually evolving restrictive disease 05%) with minimal fibrosis. 2J.25.38 Pulmonary dysfunction may be underestimated clinically, because it tends to develop insidiously.39 Another ominous development is severe thrombocytopenia (20%), which is often resistant to conventional therapy. This complication is more common in children than in adults. 48 In a retrospective review, Tiddens and colleagues61 reported 14 children with MCTD who met the criteria of Kasukawa and coworkers,43 with a mean follow-up of 9.3 years (range, 3.8 to 14.1 years) and a mean age at onset of 10.6 years (range, 5.2 to 15.6 years). Features of the disease characteristic of SLE and JDM tended to disappear over time, whereas those of CSS, Raynaud's phenomenon, and JRA persisted. At follow-up, thrombocytopenia persisted in 3 children, 4 had extensive limitation of range of joint movement, all had abnormal esophageal function, and none had active renal disease. No pulmonary hypertension was documented, although one half had restrictive disease on function studies. Glucocorticoids were judged successful in managing MCTD but were associated with osteonecrosis in 3 children and growth retardation in 1. The outcome of children with MCTD was evaluated in a study in three U.S. Midwestern clinics. 48 There were 21 girls and 6 boys, with a mean age at onset of 13 years (range, 5 to 18 years) and a mean duration of disease of 8 years (range, 1 to 18 years). Organ systems predominantly involved at onset and at follow-up at 8 years or more were the joints (24 and 11 patients, respectively), muscles (8 and 9), skin (16 and 8), lungs (10 and 13), heart (7 and 4), gastrointestinal tract (5 and 5), and kidneys (3 and 4). A characteristic onset involved Raynaud's phenomenon, arthritis, swollen hands, myositis, and the cutaneous features of JDM or SLE. Cutaneous disease, esophageal dysfunction, myositis, and arthritis were prominent during the entire course of the disease. These clinical features were similar to those of adults with MCTD, with less frequent and less severe pulmonary disease, and only one instance of pulmonary hypertension. Five patients developed severe thrombocytopenia.
486
C HAP T E R
21
OVERLAP SYNDROMES
All patients had positive immunodiffusion results for anti-RNP antibodies. Anti-ENA antibodies were found in titers up to 1:16,000,000 by hemagglutination and were often maintained at high levels for many years, but concentrations ultimately declined if the course stabilized or the patient entered remission. Transient, low-titer anti-dsDNA antibodies were present in 8 patients. Hypocomplementemia occurred in 8 and high titers of RF in 7. Outcomes were good or stable for 12, and 5 had prolonged remissions. Progressive disease developed in 7; 4 died of renal disease, diffuse intravascular coagulation, or cardiopulmonary failure. The 66 children from Japan studied by Yokota and colleagues 63 also had a homogeneous course and had clinical and laboratory characteristics that were distinct from those of children with SLE or other defined CfDs. The investigators confrrmed the diagnostic importance early in the course of Raynaud's phenomenon and high-titer anti-RNP antibodies. Other common clinical characteristics included swelling of the hands, polyarthralgia, facial erythema, RF seropositivity, hypergammaglobulinemia, and increased serum levels of the muscle enzymes. Michels64 reviewed the course of MCID in 224 children reported until 1996, including 33 patients from the Rheumakinderklinik in Garmisch-Partenkirchen, Germany. Because this review involved a number of studies over many years that were often retrospective and without serologic or genetic characterization by current standards, it predominantly reflects the clinical classifications and historical conclusions of the various centers. Nevertheless, this meta-analysis indicated that most of the children improved over time and that remissions occurred in 3% to 27% of the series. Raynaud's phenomenon and scleroderma-like skin changes were reported in up to 86%. Long-term problems included loss of range of joint motion in 29%, renal disease in up to 47%, pulmonary restrictive disease in up to 57%, and esophageal dysmotility in up to 29%. Cardiovascular disease included cardiomyopathy, pericarditis, and pulmonary hypertension. Central nervous system involvement was rare but could be severe. Seventeen (7.6%) of the 224 patients died of sepsis (7 patients), cerebral disease (3), heart failure (2), pulmonary hypertension (2), renal failure (2), or gastrointestinal bleeding (1). It was concluded that this mortality rate was in the range of that for other major systemic CTDs and that otherwise the long-term problems in patients who survived were minor. These conclusions are similar to observations from our clinic of children9 and adults. 65 Kotajima and colleagues62 compared two groups of Japanese patients with MCTD, one with onset when patients were younger than 16 years and another with onset when patients were 16 years old or older. Signs typical of SLE, such as facial erythema, photosensitivity, the presence of lupus erythematosus cells, lymphadenopathy, and cellular casts, were more common in the juvenile-onset group. Conversely, scleroderma-like symptoms, such as esophageal dysmotility, sclerodactyly, and pulmonary disease, were more common in the older group. The investigators also found that swelling of the hands occurred less frequently in children. The mortality rate was approximately 2.8% for the children. Compared with other CTDs, these outcomes were interpreted as relatively favorable. The long-term outcomes of 47 adults and children with MCTD who met the criteria of Kasukawa and coworkers43 and were followed for 3 to 29 years were studied by Burdt and colleagues. 65 All patients had antibodies to the 70-kD polypeptide of UlRNP, 81% to A, 79% to BIB', 48% to C, and 14% to D. AntiUIRNA was positive in 89% of the patients, and these antibody levels correlated with the activity of the disease. Initially, epitope spreading was observed as a feature of active MCfD, and with time, antibody reactivity was selectively reduced in patients in remission (Le., epitope contraction). HLA-DR4 and HLA-DR2 were present in 23 (85%) of 27 patients. Inflammatory features of the disease such as Raynaud's phenomenon and esophageal
hypomotility diminished over time, whereas pulmonary hypertension and central nervous system disease persisted despite treatment. Sclerodactyly was frequent (49%), but diffuse sclerosis occurred in only 19% of patients. Antibodies to centromere, Sci-70, and PMl!PM-Scl antigens were not detected. Renal disease developed in 5 patients (11%: World Health Organization class III in 2, class IV in 2, and classes III and V in 1). Eleven patients died 3 to 25 years after the onset of MCfD, with pulmonary hypertension the major contributory factor in 9 and often associated with the presence of anticardiolipin antibodies. A favorable outcome was documented in 62% of the patients, with 17% in remission (11 off therapy) leading normal lives without functional disabilities at the time of the study.
SJ6GREN'S SYNDROME Definition and Classification Sjogren's syndrome is a disorder in which decreased secretion of saliva and tears results in dryness of the mouth (Le., xerostomia) and eyes O.e., xerophthalmia or keratoconjunctivitis sicca).66 It is characterized serologically by the presence of autoantibodies to the nuclear antigens Ro/SS-A and La/SS-B (Table 21-4). Classification criteria have been proposed for adults67-7o and children. 71 If Sjogren's syndrome occurs as an isolated disorder, it is referred to as primary Sjogren's syndrome, which is rare in children. 72 A more frequent occurrence in childhood, secondary Sjogren's syndrome, most commonly accompanies SLE, as it does in adults,46 or less often accompanies another CTD. Children with primary or secondary Sjogren's syndrome reported up to 1988 were reviewed by Deprettere and associates. 22 ,73-82 Eleven of the 27 children had secondary Sjogren's syndrome, most commonly associated with SLE or MCTD and only rarely associated with JRA or CSS. The remaining children had primary Sjogren's syndrome. The authors of this study proposed classification criteria for this syndrome on the basis of their review: (1) keratoconjunctivitis (Schirmer or rose bengal test); (2) xerostomia (decreased basal and stimulated salivary flow); (3) minor salivary gland biopsy with lymphocytic infiltration in at least two foci of 4 mm2 ; and (4) RF ~ 1:160, ANA ~ 1:160, or ENA antibodies. Cimaz and colleagues72 reported a series of 40 patients from European centers with primary Sjogren's syndrome. The female-to-male ratio was 35:5. Age at onset was 9.3 to 12.4
._-
.. (ABLE 21 4
Sjogr('n's Syndrome
III
Childr('n
Designation
Charaderlzatlon
Definition
Keratoconjunctivitis sicca (i.e., dry eyes from decreased tear production by lacrimal glands) and xerostomia (i.e., dry mouth from decreased saliva production by salivary glands) Not associated with any other disease; rare in childhood Associated with a connective tissue disease, most often systemic lupus erythematosus or mixed connective tissue disease; uncommon in childhood Ro/SS-A (95%) and La/SS-B (85%)
Primary Sjogren's Syndrome Secondary Sjogren's Syndrome Autoantibodies
C HAP T E R
years (mean, 10.7 years). Follow-up periods were 0 to 7.5 years. At onset, parotid swelling preceded sicca symptoms, which were also less common. Oral glucocorticoids were required in 22 patients, hydroxychloroquine in 5, cyclosporine in 2, and cyclophosphamide in 1 patient. There were no major complications.
alnlcal Manifestations The characteristics of Sjogren's syndrome at onset are quite variable. The disorder often presents as recurrent parotid swelling that may be unilateral or bilateral, painful or painless (Fig. 21_2).83-86 Deficiency of saliva results in difficulty chewing and swallowing, abnormalities of taste, and severe dental caries. 87 Photophobia or irritation of the eyes results from involvement of the lacrimal glands, which produce insufficient tears. Dryness of other mucosal surfaces, including the nose, pharynx, and vagina, occurs. About 25% of adults with primary Sjogren's syndrome develop other systemic complications, such as interstitial pneumonitis, interstitial nephritis, myositis, achalasia with achlorhydria, isosthenuria or renal tubular acidosis, Hashimoto's thyroiditis,88 or splenic vasculitis. 89 Severe involvement of the central nervous system has been described in children and adults. 9D--n Optic neuropathy has occurred93 and may be caused by antiphospholipid antibodies in some patients. 94
• FIgure 21-2 Swelling of the parotid gland was present at the onset of Sj5gren's syndrome in this young girt Within 2 years, she developed the clinical features of systemic lupus erythematosus.
21
OVERLAP SYNDROMES
487
Pathology Histologic changes include widespread infiltration of lymphocytes and, to a lesser extent, of plasma and reticulum cells in salivary and lacrimal glands and in parenchymal organs. In some cases, germinal follicle formation occurs, followed by secondary atrophy and obliteration of secretory acini. Particularly in the salivary glands, there is a proliferation of ductal lining cells to form epimyoepithelial islands. This latter histologic finding is an important diagnostic feature of Sjogren's syndrome (Fig. 21-3). The diagnosis may be confirmed, if necessary, by biopsy of a minor salivary gland from the lower lip demonstrating periductal lymphocytic infiltrates. 95 Nail fold capillaries are normal unless another CTD is present,96 but they may be abnormal in adults with Raynaud's phenomenon. 97 Lymphoma may develop in adults with primary Sjogren's syndrome.98
Laboratory Examination The most striking laboratory abnormalities include polyclonal hypergammaglobulinemia with high-titer RF and ANAs directed at Ro/SS-A (95%) and La/SS-B (85%). AntiRo antibodies in Sjogren's syndrome are frequently accompanied by anemia, leukopenia, lymphopenia, cryoglobulinemia, and vasculitis.99-101 Specific HLA class II alleles (e,g., DR3, DQwl, DQw2) are associated with the development of anti-RolLa antibodies. lOz- 104 Antiphospholipid antibodies have been described in children but are usually of low titer and not associated with thrombotic phenomena. 105 A positive result on Schirmer's test indicates deficient tear flow «5 mm wetting of a filter paper strip in 15 minutes); rose bengal or fluorescein staining of the cornea may demonstrate superfiCial erosions. Spath and associates lOO documented the diagnostic utility of magnetic resonance imaging of the parotid gland. Demonstration of ectasia of the parotid ducts by sialographylO6 or
• Figure 21-3 Parotid gland tissue (hematoxylin and eosin stain, magnification x 40).There is obliteration of the nonnal adnar architecture by lymphocytes. Aregenerative epimyoepithelial island occupies the center of the field.
488
C HAP T E R
21
OVERLAP SYNDROMES
decreased uptake of technetium 99m pertechnetate also supports the diagnosis.
Treatment Therapy is nonspecific and not curative. 107 The systemic manifestations of Sjogren's syndrome are treated with nonsteroidal anti-inflammatory drugs or glucocorticoids. The sicca component is managed with adequate environmental humidity, artificial tears, nasal saline douches, and sour lemon drops to stimulate production of saliva. Pilocarpine tablets have been recommended for the sicca symptoms of the eyes and mouth. lOB The drug is well tolerated and safe but has significant parasympathomimetic side effects.
REFERENCES 1. Sharp GC, Irvin WS. Tan EM, et al: Mixed connective tissue disease-an apparently distinct rheumatic disease syndrome associated with a specific antibody to an extractable nuclear antigen (ENA). Am J Med 52: 148-159. 1972. 2. Nimelstein SH, Brody S, McShane 0, et al: Mixed connective tissue disease: a subsequent evaluation of the original 25 patients. Medicine (Baltimore) 59: 239-248, 1980. 3. Bennett RM, O'Connell OJ: Mixed connective tissue disease: a clinicopathologic study of 20 cases. Semin Arthritis Rheum 10: 25--51. 1980. 4. Lundberg I, Hedfors E: Clinical course of patients with anti-RNP antibodies. A prospective study of 32 patients. J Rheumatol 18: 1511-1519, 1991. 5. Hoffman RW, Sharp GC: Is anti-UI-RNP autoantibody positive connective tissue disease genetically distinct? J Rheumatol 22: 586-589, 1995. 6. Pelkonen PM, Jalanko HJ, Lantto RK, et al: Incidence of systemic connective tissue diseases in children: a nationwide prospective study in Finland. J Rheumatol 21: 2143-2146, 1994. 7. Bowyer S, Roettcher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group. J Rheumatol 23: 1968-1974, 1996. 8. Horn JR, Kapur JJ, Walker SE: Mixed connective tissue disease in siblings. Arthritis Rheum 21: 709-714. 1978. 9. Hoffman RW, Cassidy JT, Takeda Y, et al: VI-70-kd autoantibody-positive mixed connective tissue disease in children. A longitudinal clinical and serologic analysis. Arthritis Rheum 36: 1599-1602, 1993. 10. Genth E, Zarnowski H, Mierau R, et aI: HLA-DR4 and Gm(l,3;5,21J are associated with Ul-nRNP antibody positive connective tissue disease. Ann Rheum Dis 46: 189-196, 1987. 11. Black CM, Maddison PJ, Welsh KI, et al: HLA and immunoglobulin allorypes in mixed connective tissue disease. Arthritis Rheum 31: 131-134, 1988. 12. Harley JB, Sestak AL, Willis LG, et al: A model for disease heterogeneiry in systemic lupus erythematosus. Relationships between histocompatibility antigens, autoantibodies, and lymphopenia or renal disease. Arthritis Rheum 32: 826-836, 1989. 13. de Rooij OJ, Fiselier T, van de Putte LB, et al: Juvenile-onset mixed connective tissue disease: clinical, serological and follow-up data. Scand J Rheumatol 18: 157-160, 1989. 14. Hoffman RW, Rellenmaier LJ, Takeda Y, et al: Human autoantibodies against the 70-kd polypeptide of Ul small nuclear RNP are associated with HLA-DR4 among connective tissue disease patients. Arthritis Rheum 33: 666-673, 1990. 15. Kaneoka H, Hsu KC, Takeda Y, et al: Molecular genetic analysis of HLA-DR and HLA-DQ genes among anti-UI-70-kd autoantibody positive connective tissue disease patients. Arthritis Rheum 35: 83-94, 1992. 16. Smolen JS, Steiner G: Mixed connective tissue disease: to be or not to be? Arthritis Rheum 41: 768-m, 1998. 17. Lanchbury JS, Hall MA, Welsh Kl, et aI: Sequence analysis of HLA-DR4Bl subtypes: additional first domain variability is detected by oligonucleotide hybridization and nucleotide sequencing, Hum Immunol 27: 136-144, 1990 J8. Hoffman RW, Takeda Y, Sharp GC, et al: Human T celi clones reactive against V-small nuclear ribonucleoprotein autoantigens from connective tissue disease patients and healthy individuals. J Immunol 151: 6460--6469, 1993. 19, Talken BL, Lee DR, Caldwell CW, et al: Analysis of T cell receptors specific for UI-70kD small nuclear ribonucleoprotein autoantigen: the alpha chain complementariry detennining region three is highly conserved among connective tissue disease patients. Hum Immunol 60: 200-208, 1999. 20, Sanders DY, Huntley CC, Sharp GC: Mixed connective tissue disease in a child. J Pediatr 83: 642-645, 1973.
21. Singsen BH, Kornreich HK, Koster-King K, et al: Mixed connective tissue disease in children. Arthritis Rheum 20: 355--360. 1977. 22. Fraga A, Gudino J, Ramos-Niembro F, et aI: Mixed connective tissue disease in childhood. Relationship to Sjogren's syndrome. Am J Dis Child 132: 263-265, 1978. 23. Rosenthal M: Juvenile Sharp syndrome (mixed connective tissue disease). Helv Paediatr Acta 33: 251-258, 1978. 24. Peskett SA, Ansell BM, Fizzman P, et al: Mixed connective tissue disease in children. Rheumatol Rehabil 17: 245-248, 1978, 25. Rosenberg AM, Petty RE, Cumming GR, et al: Pulmonary hypertension in a child with mixed connective tissue disease. J Rheumatol 6: 700-704, 1979. 26. Michels H, Schuchmann L: MCTD (Sharp) syndrome in childhood. (author's trans]). KIin Padiatr 192: 389-392, 1980. 27. Oetgen WJ, Boice JA, Lawless OJ: Mixed connective tissue disease in children and adolescents. Pediatrics 67: 333-337, 1981. 28. Eberhardt K, Svantesson H, Svensson B: Follow-up study of 6 children presenting with a MCTD-Iike syndrome, Scand J Rheumatol 10: 62-64. 1981. 29. Allen RC, St Cyr C, Maddison PJ, et al: Overlap connective tissue syndromes. Arch Dis Child 61: 284-288, 1986, 30. Allen R: overlap syndrome. In Woo P, White PH, Ansell BM (eds): Paediatric Rheumatology Update. Oxford, Oxford Universiry Press, 1990. pp 209-216, 31. Mier R, Ansell B, Hall MA, et al: Long tenn follow-up of children with mixed connective tissue disease. Lupus 5: 221-226, 1996. 32. Yang YH, Tsai MJ, Lin SC, et al: Childhood mixed connective tissue disease. J Formos Med Assoc 99: 158-161, 2000. 33. Maricq HR, leRoy EC, D'Angelo WA, et al: Diagnostic potential of in vivo capillary microscopy in scleroderma and related disorders. Arthritis Rheum 23: 183-189, 1980. 34. Maricq HR: Wide-field capillary microscopy. Technique and rating scale for abnonnalities seen in scleroderma and related disorders. Arthritis Rheum 24: 1159-1165, 1981. 35. Gendi NS, Welsh Kl, Van Venrooij WJ, et al: HLA rype as a predictor of mixed connective tissue disease differentiation. Ten-year clinical and immunogenetic followup of 46 patients. Arthritis Rheum 38: 259--266, 1995. 36. Scheja A, Elborgh R, Wildt M: Decreased capillary densiry in juvenile dennatomyositis and in mixed connective tissue disease. J Rheumatol 26: 1377-1381. 1999, 37. Weber P, Ganser G, Frosch M, et al: Twenry-four hour intraesophageal pH monitoring in children and adolescents with scleroderma and mixed connective tissue disease. J Rheumatol 27: 2692-2695, 2000. 38. Saito Y, Terada M, Takada T, et al: Pulmonary involvement in mixed connective tissue disease: comparison with other collagen vascular diseases using high resolution CT. J Comput Assist Tomogr 26: 349--357, 2002, 39. Sullivan WO, Hurst OJ, Harmon CE, et al: A prospective evaluation emphasizing pulmonary Involvement in patients with mixed connective tissue disease. Medicine (Baltimore) 63: 92-107, 1984, 40. Singsen BH, Swanson VL, Bernstein BH, et al: A histologic evaluation of mixed connective tissue disease in childhood, AmJ Med 68: 710-717,1980. 41. Alarcon-Segovia 0: Symptomatic Sjogren's syndrome in mixed connective tissue disease. J Rheumatol 11: 582-583, 1984. 42. Sharp GC: Diagnostic criteria for classification of MCTD. In Kasukawa R. Sharp GC (eds): Mixed Connective Tissue Disease and Anti-nuclear Antibodies. Amsterdam, Excerpta Medica, 1987, pp 23-32. 43. Kasukawa R, Tojo T, Miyawaki S, et al: Preliminary diagnostic criteria le)r classification of mixed connective tissue disease, In Kasukawa R, Sharp GC (eds): Mixed Connective Tissue Disease and Anti-nuclear Antibodies. Amsterdam, Excerpta Medica, 1987, pp 41-48. 44. Alarcon-Segovia 0, Villarreal M: Classification and diagnostic criteria for mixed connective tissue disease. In Kasukawa R, Sharp GC (eds): Mixed Connective Tissue Disease and Anti-nuclear antibodies. Amsterdam. Excerpta Medica, 1987, pp 33-40. 45. Kahn MF, Appelboom T: Syndrome de Sharp. In Kahn MF, Peltier AP. Meyer 0, et al (eds): Les maladies systemiques, 3rd ed. Paris, Flammarion, 1991, pp 545--556. 46. Shen N, Chen S, Yang H, et al: Mixed connective tissue disease: a disease entiry? Chin Med J (Eng]) 111: 214-217, 1998. 47. Amigues JM, Cantagrel A, Abbal M, et al: Comparative study of 4 diagnosis criteria sets for mixed connective tissue disease in patients with anti-RNP antibodies. Autoimmuniry Group of the Hospitals of Toulouse. J Rheumatol 23: 2055--2062, 1996. 48. Cassidy]T, Hoffman RW, Wortmann OW, et al: Long-term outcome of children with mixed connective tissue disease (MCTD). J Rheumatol 27(Suppl 58): 100. 2000. 49. SavouretJF, Chudwin OS, Wara OW, et al: Clinical and laboratory findings in childhood mixed connective tissue disease: presence of antibody to ribonucleoprotein containing the small nuclear ribonucleic acid VI. .J Pediatr 102: 841--846, 1983. 50. Pellersson I, Wang G, Smith El, et al: The use of immunoblotring and immunoprecipitation of (U) small nuclear ribonucleoproteins in the analysis of sera of patients with mixed connective tissue disease and systemic lupus erythematosus. A cross-sectionai, longitudinal study. Arthritis Rheum 29: 986-996, 1986. 51. Margaux J, Hayem G, Palazzo E, et al: Clinical usefulness of antibodies to UlsnRNP proteins in mixed connective tissue disease and systemic lupus erythematosus. Rev Rhum Engl Ed 65: 378-386. 1998.
C HAP T E R 52. Habets W). Hoet MH, SUlekens PT, et al: Detection of autoantibodies in a quantitative immunoassay using recombinant ribonucleoprotein antigens. Clin Exp Immunol 76: 172-177, 1989. 53. Deutscher SL, Keene )D: A sequence-specific conformational epitope on VI RNA is recognized by a unique autoantibody. Proc Natl Acad Sci V S A 85: .~299-3303, 1988. 54. Van Venrooij W), Hoet R, Castrop), et al: Anti-(liI) small nuclear RNA antibodies in anti-small nuclear ribonucleoprotein sera from patients with connective tissue diseases. ) Clin Invest 86: 2154-2160, 1990. 55. Hoet RM, Koornneef I, de Rooij D), et al: Changes in anti-VI RNA antibody levels correlate with disease activity in patients with systemic lupus erythematosus overlap syndrome. Arthritis Rheum 35: 1202-1210, 1992. 56. Takeda Y, Wang GS, Wang R), et al: Enzyme-linked immunosorbent assay using isolated (0) small nuclear ribonucleoprotein polypeptides as antigens to investigate the clinical significance of autoantibodies to these polypeptides. Clin Immunol Immunopathol 50: 213-230, 1989. 57. Takano M, Golden SS, Sharp GC, et al: Molecular relationships between two nuclear antigens, ribonucleoprotein and Sm: purification of active antigens and their biochemical characterization. Biochemistry 20: 5929-5936, 1981. 58. Komatireddy GR, Wang GS, Sharp GC, et aI: Antiphospholipid antibodies among anti-Vl-70 kDa autoantibody positive patients with mixed connective tissue disease.) Rheumatol 24: 319-322, 1997. 59. Nakata S, Vematsu K, Mori T, et al: Effective treatment with low-dose methotrexate pulses of a child of mixed connective tissue disease with severe myositis refractory to corticosteroid. Nihon Rinsho Meneki Gakkai Kaishi 20: 178-183, 1997. 60. Wulffraat NM, Sanders LA, Kuis W: Autologous hemopoietic stem-cell transplantation for children with refractory autoimmune disease. Curr Rheumatol Rep 2: 316--323, 2000. 61. Tiddens HA, van der Net )), Graeff-Meeder ER, et al: Juvenile-onset mixed connective tissue disease: longitudinal follow-up.) Pediatr 122: 191-197, 1993. 62. Kotajima L, Aotsuka S, Sumiya M, et al: Clinical features of patients with juvenile onset mixed connective tissue disease: analysis of data collected in a nationwide collaborative study in )apan.) Rheumatol 23: 1088-1094, 1996. 63. Yokota S, Imagawa T, Katakura S, et al: Mixed connective tissue disease in childhood: a nationwide retrospective study in Japan. Acta Paediatr )pn 39: 273-276, 1997. 64. Michels H: Course of mixed connective tissue disease in children. Ann Med 29: 359-364, 1997. 65. Burdt MA, Hoffman RW, Deutscher SL, et al: Long-term outcome in mixed connective tissue disease: longitudinal clinical and serologic findings. Arthritis Rheum 42: 899-909, 1999. 66. Talal N: Sjogren's syndrome: historical overview and clinical spectrum of disease. Rheum Dis Clin North Am 18: 507-515, 1992. 67. Vitall C, Bombardieri S: Diagnostic critelia for Sjogren's syndrome: the state of tile art. Clin Exp Rheumatol 8 (Suppl 5): 13-16. 1990. 68. Vitali C, Bombardieri S, Moutsopoulos HM, et al: Preliminary criteria for the classification of Sjogren's syndrome. Results of a prospective concerted action supported by the European Community. Arthritis Rheum 36: 340--347, 1993. 69. Fox RI. Saito I: Criteria for diagnosis of Sjogren's syndrome. Rheum Dc, Clin North Am 20: 391-407, 1994. 70. Vitali C, Moutsopoulos HM, Bombardieri S: The European Community Study Group on diagnostic criteria for Sjogren's syndrome. Sensitivity and specificity of tests for ocular and oral involvement in Sjogren's syndrome. Ann Rheum Dis 53: 637-
21
OVERLAP SYNDROMES
489
80. Vermylen C, Meurant A, Noel H, et al: Sjljgren's syndrome in a child. Eur) Pediatr 144: 266--269, 1985. 81. Siamopoulou-Mavridou A, Drosos AA, Andonopoulos AP: Sjogren syndrome in childhood: report of two cases. Eur) Pediatr 148: 523-524, 1989. 82. Rocha G, Kavalec C: Sjogren's syndrome in a child. Can J Ophthalmol 29: 234-237, 1994. 83. Bernstein BH, Koster-King K, Singsen BH, et al: Sjogren's syndrome in childhood. Arthritis Rheum 20: 361, 1977. 84. Krause A, Alarcon-Segovia D: Primary juvenile Sjiigren's syndrome. J Rheumatol 15: 803, 1988. 85. Hara T, Nagata M, Mizuno Y, et al: Recurrent parotid swelling in children: clinical features useful for differential diagnosis of Sjogren's syndrome. Acta Paediatr 81: 547-549, 1992. 86. Fox R1: Sjogren's syndrome. In Kelley WN, Harris ED )r, Ruddy S, et al (eds): Textbook of Rheumatology. Philadelphia, WE Saunders, 1997,1'1' 955-968. 87. Nathavitharana KA, Tarlow M), Bedi R, et al: Primary Sjogren's syndrome and rampant dental caries in a 5-year-old child. Int) Paediatr Dent 5: 173-176, 1995. 88. Mirante A, Salgado M, Moura L, et al: Hashimoto's thyroiditis and mixed connective tissue disease in an 11 year-old girl. ) Pediatr Endocrinol Metab 10: 77-78, 1997. 89. Akin E, Tucker LB, Miller I.C, et al: Splenic vasculitis in juvenile onset mixed connective tissue disease. ) Rheumatol 25: 1444-1445. 1998. 90. Ohtsuka T, Saito Y, Hasegawa M, et al: Central nervous system disease in a child with plimary Sjogren syndrome. .J Pediatr 127: 961-963, 1995. 91. Niemela RK, Hakala M: Primary Sjljgren's syndrome with severe central nervous system disease. Semin Arthritis Rheum 29: 4-13, 1999. 92. loannidis )1', Moutsopoulos HM: Sjogren's syndrome: too many associations, too limited evidence. The enigmatic example of CNS involvement. Semin Arthritis R1leum 29: 1-3, 1999. 93. Rojas-Rodriguez), Garcia-Carrasco M, Ramirez ES, et aI: Optic neuropathy in a child with primary Sjogren's syndrome. Rev Rhum Engl Ed 65: 355-357, 1998. 94. Leo-Kottler B, Klein R, Berg PA, et al: Ocular symptoms in association with anti phospholipid antibodies. Graefes Arch Clin Exp Ophthalmol 236: 658-
C HAP T E R
22
VASCULITIS AND ITS CLASSIFICATION James T. Cassidy and Ross E. Petty
~
The term vasculitis indicates the presence of inflammation in a blood vessel wall. The inflammatory infiltrate may be one that is predominantly neutrophilic, eosinophilic, or mononuclear. Perivasculitis describes inflammation around the blood vessel wall but without mural involvement. Vasculopathy, a broader term, indicates an abnormality of blood vessels that may be inflammatory, degenerative, or result from intimal proliferation.
Hoffman3 eloquently points out the many pitfalls in classifications of vasculitis and writes that "there is one certainty about vasculitis classification systems. They will continue to change until these diseases can be categorized by a more complete understanding of their etiology and pathogenesis." Until that time, the choice of a system of classification is arbitrary and a matter of personal preference. The classification used this book accommodates the most significant histopathologic and clinical findings of the common vasculitides of childhood (Table 22-4).
CLASSIFICATION The vasculitides are the most difficult of all rheumatic diseases to classify.! A number of attempts have been made, and none has completely succeeded. 2,3 In part, this has resulted from inconsistent use of definitions of individual disorders, a problem that was addressed by a committee of the American College of Rheumatology (ACR) in 1990. 4 Subsequently, the Chapel Hill Consensus Conference in 1994 refined and modified the ACR classification (Table 22-1).5 Neither of these studies specifically addressed vasculitis in childhood, and from a practical point of view, they have limited usefulness in pediatrics.6-ll In these classifications, diseases are grouped by the size of the vessel affected. 9 In other classifications, the character of the vascular lesion or a combination of vessel size and histologic characteristics is considered. A comprehensive list of vasculitides (many of which are rare in childhood) is in Table 22-2. 10 In a classification by Savage and colleagues in 1997,11 the size of the affected vessels and the presence or absence of granulomata were used to classify vasculitis (Table 22-3). The consistency of histopathologic findings within any clinical diagnostic category is often limited.!2 Some disorders, such as Henoch-Schonlein purpura (HSP) and Kawasaki disease (KD), have consistent pathologic pictures; however, in polyarteritis nodosa, microscopic polyarteritis, granulomatous vasculitides, and giant cell arteritides, the histologic picture is often mixed, with lesions of different histologies occurring in patients with similar clinical syndromes. The term polyangiitis overlap syndrome has been suggested in recognition of the high frequency (40%) of patients who exhibit features of more than one distinct vasculitic syndrome.!3
492
GENERAL CLINICAL ASPECTS OF VASCULmS Childhood vasculitis is a complex but fascinating area of pediatric rheumatology. It is a clinical field that is often shared with other pediatric subspecialists, such as dermatologists, cardiologists, and nephrologists, a faLt that emphasizes the multisystem nature of these diseases. The type of pathologic change, site of involvement, size of vessel, and systemic extent of the vascular injury determine the clinical expression of the disease and its severity. Table 22-5 summarizes features that suggest a vasculitic syndrome. The onset of some vasculitides (e.g., HSP, KD) is usually abrupt, and diagnostic characteristics of the disease become apparent in a few days to a week. In many of the vasculitides, however, presentation is more indolent, and various signs and symptoms developing over weeks to months are characteristic. In this case, the diagnosis is often difficult and delayed, and requires a high index of suspicion and a thorough investigation of symptomatic and asymptomatic (but potentially affected) organs such as the heart, lungs, liver, and kidneys. Definitive diagnosis frequently requires a biopsy of one or more sites or angiographic studies, Table 22-6 summarizes the clinical and pathologic characteristics of the vasculitides in childhood that are discussed in greater detail in subsequent chapters.
EPIDEMIOLOGY The incidence and prevalence of vasculitis in children are not known. The relative frequencies of selected vasculitides among a group of 1000 patients (mostly adults) with
C HAP T E R
c.
TABLE ll- 1
22 V AS CUll TIS
AND ITS
C LAS S I FIe AT ION
493
Chapel Hill Consensus Conference on Nomenclature of Systemic Vasculitis
Latge-Vessel Vasculitis· Giant cell (temporal) arteritis Takayasu's arteritis
Granulomatous arteritis of the aorta and its major branches with a predilection for the extracranial branches of the carotid artery. It often involves the temporal artery. U~ual(y occurs in patients older than 50 years and is o.fien associated with polymyalgia rheumatica. t Granulomatous inflammation of the aorta and its major branches. Usual~y occurs in patients much younger than 50 years.
f--.
MedIum-Vessel Vasculitis· Polyarteritis nodosa
Necrotizing inflammation of medium-sized or small arteries without glomerulonephritis or vasculitis in arterioles, capillaries or venules. Arteritis involving large, medium-sized, and small arteries associated with the mucocutaneous lymph node syndrome. Coronary arteries are often involved. Aorta and veins may be affected. Usualry occurs in children.
Kuwasaki disease
_II-Vessel Vasculitis· Wegener's granulomatosis! Churg-Strauss syndrome! Microscopic polyangiitis! Henoch-Schonlein purpura Essential cryoglobulinemic vasculitis Cutaneous leukoeytoclastic angiitis
Granulomatous inflammation involving the respiratory tract associated with necrotizing vasculitis affecting small to medium-sized vessels. Necrotizing glomerulonephritL~ is common. Eosinophilic and granulomatous inflammation involving the respiratory tract accompanied by necrotizing vasculitis affecting small- to medium-sized vessels associated with asthma and eosinophilia. Necrotizing vasculitis with few or no immune deposits, affecting small vessels. Necrotizing arteritL~ involVing small- and medium-sized arteries may be present. Necrotizing glomerulonephritis is common. Pulmonary capillaritis often occurs. Vasculitis characterized by IgA-dominant immune deposits affecting small vessels. Typically involves skin, gut and glomeruli. Arthralgias and arthritis are common. Vasculitis with cryoglobulin immune deposits affecting small vessels associated with cryoglobulinemia. Skin and glomeruli are often involved. Isolated cutaneous leukocytoclastic angiitis without systemic vasculitis or glomerulonephritis.
'La'1le vessels: aorta and its latger branches directed toward major anatomic regions; medium vessels: renal, hepatic, coronary, and mesenteric arteries; small vessels: venules, capillaries, arterioles, and intraparenchymal distal arteries and arterioles. tEssl-'ntial components are in normal type; italicized (ype represents usual, but not essential, components. 'Str(~ngly associated with antinuclear cytoplasmic antibodies. Adapted from Jennene JC, Falk RJ, Andrassy K. et al: Nomenclature of systemic vasculitides. Proposal of an international consensus conference. Arthritis Rheum 37: 187-192. 1994.
[ . TABLE 2l-l
Classification of Vasculitis by Vessel Size
ulrge-vessel vasculitis Giant cell (temporal) arteritis Takayasu's arteritis Medium-sized vessel vasculitis Polyarteritis nodosa Kawasaki disease Primary granulomatous central nervous system vasculitis Small-vessel vasculitis ANCA-associated small-vessel vasculitis Microscopic polyangiitis Wegener's granulomatosis Churg-Strauss syndrome Drug-induced, ANCA-associated vasculitis Immune complex small-vessel vasculitis Henoch-Schonlein purpura Essential cryoglobulinemic vasculitis Hypocomplementemic urticarial vasculitis Vasculitis with systemic lupus erylhematosus, rheumatoid arthritis or Sjogren's syndrome Beh~et's syndrome Goodpasture's syndrome Serum sickness Drug-associated immune complex vasculitis Infection-associated immune complex vasculitis Paraneoplastic small-vessel vasculitis Lymphoproliferative neoplasm-induced vasculitis Myeloproliferative neoplasm-induced vasculitis Carcinoma-induced vasculitis Inflammatory bowel disease vasculitis ANCA. antinuclear cytoplasmic antibody. From Jennette Je. Falk. R.I: Small-vessel vasculitis. N Engl J Med 337: 1512-1523, 1997.
~~II TABLE ll~3
Classification 01 Primary Systemic VasLUlitis by Vessel Size and the Presence of Granulomata
Vessel Size Granulomatous Fonns
Nongranulomatous Forms
Large
Temporal arteritis Takayasu's arteritis
Medium
Polyarteritis nodosa Kawasaki disease Wegener's granulomatosis Microscopic polyangiitis Churg-Strauss syndrome Henoch-Schonlein purpura Cutaneous leukocyloclastic vasculitis Essential cryoglobulinemic vasculitis
Small
From Savage COS, Harper L. Adu D: Primary systemic vasculitis. Lancet 349: 554-558, 1997.
494
C HAP T E R
[II TABI E 22 4
22 V AS C U LI TI SAN D
C1a~sili( ation
ITS
C LAS S I F I CAT IO N
of PllmalY Sysh'mi( Vds(lIlitis in
lhildren
'~II
lABIE 22
~
fedtlll"~ llMI
SlIyy"sl
.I
Vasculiti( Syndrolll"
Clinical Features Leukocytoc1astic vasculitis Henoch-Schonlein purpura Hypersensitivity angiitis Hypocomplementemic urticarial vasculitis Mixed cryoglobulinemia Polyarteritis Polyarteritis nodosa Cutaneous polyarteritis Cogan's syndrome Soter's syndrome Kawasaki disease Microscopic polyangiitis Granulomatous vasculitis Wegener's granulomatosis Churg-Strauss syndrome (allergic granulomatosis) Lymphomatoid granulomatosis Primary angiitis of the central nervous system Giant cell arteritis Takayasu's arteritis Temporal (cranial) arteritis Sarcoidosis Other vasculitides Beh\;et's syndrome Mucha-Habermann disease Kohlmeier-Degos syndrome Cronkhite-Canada syndrome
,
II
TABLE 22 6
Fever, weight loss, fatigue of unknown origin Skin lesions (palpable purpura, vasculitic urticaria, livedo reticularis, nodules, ulcers) Neurologic lesions (headache, mononeuritis multiplex, focal central nervous system lesions) Arthralgia or arthritis, myalgia or myositis, serositis Hypertension Pulmonary infiltrates or hemorrhage
Laboratory Features Increased erythrocyte sedimentation rate or C-reactive protein level Leukocytosis, anemia Eosinophilia Antineutrophil cytoplasmic antibodies Elevated factor VIII-related antigen (von Willebrand factor) Cryoglobulinemia Circulating immune complexes Hematuria
Clini(dl dml Pdtholoyi( Chdldderisti(s of Some Vdsullilides in Childhood
Syndrome
Frequency
Vessels Affected
Charaderlstlc Pathology
Polyarteritis nodosa
Rare
Kawasaki disease
Common
Medium and small muscular arteries and sometimes arterioles Coronary and other muscular arteries
Focal segmental (often near bifurcations); fibrinoid necrosis; gastrointestinal, renal microaneurysms; lesions at various stages of evolution Thrombosis, fibrosis, aneurysms, especially coronaries
Arterioles and venules, often small arteries and veins Arterioles and venules
Leukocytoc1asis; mixed cells, eosinophils; IgA deposits in affected vessels (gastrointestinal tract) Leukocytoc1astic or lymphocytic, varying eosinophils, occasionally granulomatous; widespread lesions at same stage of evolution
Small arteries and veins, occasionally larger vessels Small arteries and veins, often arterioles and venules
Upper and lower respiratory tract, necrotizing granulomata, glomerulonephritis Necrotizing extravascular granulomata; lung involvement; eosinophilia
Granulomatous inflammation, giant cells; aortic arch and branches; aneurysms, dissection Granulomatous inflammation, giant cell arteritis; carotid artery and branches
Polyarteritis
Leukocytoclastlc Vasculitis Henoch-Schonlein purpura
Common
Hypersensitivity angiitis
Rare
Granulomatous Vasculitis Wegener's granulomatosis
Rare
Allergic granulomatosis
Rare
Giant Cell Arteritis Takayasu's arteritis
Uncommon
Muscular and elastic arteries
Temporal arteritis
Rare
Medium and large arteries
C HAP T E R
vasculitis are in Table 22-7. 14 The distribution is very different in the pediatric population, as presented in Table 22~. Without doubt, KD and HSP are the most common disorders in North America and Europe. In Asia, however, Takayasu's arteritis is common. Comprehensive studies of the frequencies of the vasculitides in Asia are not available, but it is expected that KD and Takayasu's arteritis constitute a larger proportion of vasculitis in that area of the world. Polyarteritis nodosa and cutaneous polyarteritis may also be more common in Japan and Turkey. In national diagnostic registries, the various forms of va$culitis account for 1% to 6% of pediatric rheumatic
III • rABt E 22 -, ~
7 Categories and Frequencies of Vasculitis in IOOU Patienls
Frequency (%)
of Vasculitis
Giant cell (temporal) arteritis V~sculitis with a connective tissue disease Vasculitis of unknown type Polyarteritis nodosa Hypersensitivity vasculitis Wegener's granulomatosis lienoch-Sch6nlein purpura T;jkayasu's arteritis Kawasaki disease Churg-Strauss syndrome
21.4 14.1 12.9 11.8 9.3 8.5 8.5 6.3 5.2 2.0
Data from Bloch DA, Michel BA, Hunder GG. et al. The American College of Rheumatology 1990 criteria for the classification of vasculitis. Patients and methods. Arthritis Rheum 33: 1068--1073, 1990.
REFERENCES 1. Ozen S: The spectrum of vasculitis in children. Best Pract Res Clin
Rheumatol 16: 411--425. 2002. 2. Lie]T: Nomenclature and classification of vasculitis: plus ca change, plus c'est la meme chose. Arthritis Rheum 37: 181-186, 1994. 3" Hoffman GS: Classification of the systemic vasculitides: antineutrophil cytoplasmic antibodies, consensus and controversy. C1in Exp Rheumatol 16: 111-115, 1998. 4. Hunder GG, Arend WP, Bloch DA. et al: The American College of Rheumatology 1990 criteria for the classification of vasculitis. Introduction. Arthritis Rheum 33: 1065-1067, 1990. 5. Jennette jC, Falk Rj, Andrassy K, et al: Nomenclature of systemic vasculitides. Proposal of an international consensus conference. Arthritis Rheum 37: 187-192, 1994. 6. Brogan PA, Dillon M]: Vasculitis from the pediatric perspective. CUIT Rheumatol Rep 2: 4II--416, 2000. 7. Yalcindag A, Sundel R: Vasculitis in childhood. CUIT Opin Rheumatol 13: 422--427, 2001. 8. Singh S, Dass R: Clinical approach to vasculitides. Indian j Pediatr 69: 881-888, 2002.
22
VASCULITIS AND ITS CLASSIFICATION
495
diseases. 15- 17 The proportions of children from the two registries that identified specific vasculitides are in Table 22-8, The wide differences in frequencies probably reflect referral patterns rather than determinable geographic differences. Although childhood vasculitis is uncommon, it is an important component of referrals to pediatric rheumatology clinics, and these children often require disproportionately large amounts of time and expertise, Diagnosis can be difficult, monitoring disease activity is problematic, and the outcome for some of the vasculitides may be serious or fataL
re
II
TABLE 22 - 8
Relalive Frequencies of Vasculitides
Vasculitides Kawasaki disease Henoch-Sch6nlein purpura Wegener's granulomatosis Polyarteritis nodosa Beh\;et's disease Takayasu's arteritis Unclassified
III
Childhood
U.S. Registry"
Canadian Reglsb'yt
N=434
%
N = 225
%
97 213 6 14
22.4 49.1 1.4 3.2
8 96
1.8 22.1
147 38 5 4 2 2 27
65.3 16.9 2.2 1.8 0.9 0.9 12.0
'Data from Bowyer S, Roettcher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. .I Rheumatol 23: 1968--1974, 1996. tData from Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry. .I Rheumatol 23: 1981-1987, 1996.
9. Weyand CM, Goronzy .1.1: Medium- and large-vessel vasculitis. N EnglJ Med 349: 160-169, 2003. 10. Jennette jC, Falk Rj: Small-vessel vasmlItis. N EnglJ Med 337: 1512-1523, 1997. 11. Savage CO, Harper L, Adu D: Primary systemic vasculitis. Lancet 349: 553-558, 1997. 12. Hoffman GS, Calabrese LH: Vasculitis 2003. Clin Exp Rheumatol 21 (Supp]): SI-SI39, 2003. 13. Leavitt RY, Fauci AS: Polyangiitis overlap syndrome. Classification and prospective clinical experience. Am .I Med 81: 79-85, 1986. 14. Bloch DA, Michel BA, Hunder GG, et al: The American College of Rheumatology 1990 criteria for the classification of vasculitis. Patients and methods. Arthritis Rheum 33: 1068--1073, 1990. 15. Bowyer S, Roencher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group. j Rheumatol 23: 1968--1974, 1996. 16. Symmons DP, jones M, Osborne .I, et al: Pediatric rheumatology in the United Kingdom: data from the British Pediatric Rheumatology Group National Diagnostic Register. j Rheumatol 23: 1975-1980, 1996. 17. Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry. .I Rheumatol 23: 1981-1987, 1996.
CHAPTER23 51
.. :
r~r
LEUKOCYTOCLASTIC VASCULITIS James T. Cassidy and Ross E. Petty
;'if
Necrotizing vasculitis affecting small blood vessels, especially the postcapillary venules, capillaries, and arterioles, usually caused by immune complex deposition, is referred to as leukocytoclastic vasculitis (Fig. 23-1 and Table 23-1).1-3 In these disorders, polymorphonuclear leukocytes infiltrate vessel walls, resulting in necrosis with scattered nuclear debris. This is the predominant inflammatory reaction in Henoch-Schonlein purpura (HSP), hypersensitivity angiitis, mixed cryoglobulinemia, and the vasculitis of other connective tissue diseases, such as systemic lupus erythematosus (SLE). Leukocytoclastic vasculitis is sometimes observed as a sequela of drug hypersensitivity, infectious endocarditis, and hematologic malignancies.
HENOCH-SCH6NLEIN PURPURA Definition and Casslficatlon HSP is one of the most common vasculitides of childhood. oWl It is characterized by nonthrombocytopenic purpura, arthritis and arthralgia, abdominal pain and gastrointestinal hemorrhage, and glomerulonephritis. This syndrome has a long and distinguished history with early references by Willan9 and Heberden. lO A diagnostic triad of purpuric rash, arthritis, and abnormalities of the urinary sediment was proposed by Schonlein in 1837,11 and Henoch described the association of purpuric rash, abdominal pain with bloody diarrhea, and proteinuria in 1874. 12 The term anaphylactoid purpura was applied by Gairdner in 1948. 13
Epidemiology HSP is predominantly a disease of childhood, although a similar syndrome has been reported in a few adults. I 4-17 It occurs most frequently between the ages of 3 and 15 years and is more common in boys than in girls 0.5:1).17,18 It is rare in children younger than 2 years. 19 An incidence of 13.5 cases per 100,000 children per year was observed in an unselected childhood population in Belfast, Northern Ireland,20 An incidence of 0.2 to 10 was estimated by Farley and colleagues. 21 In the latter study, incidence was highest among Hispanic children and in lower socio-
496
economic groups. In a study from Britain, although the combined incidence was 20.4, it was 70.3 in the 4- to 6year age group.22 Striking seasonal variations have been observed,21 with most cases occurring in winter, often preceded by an upper respiratory tract infection (30% to 50%).23,24
Etiology and Pathogenesis Many reports have implicated infection as a potential trigger for this disease, particularly with ~-hemolytic streptococcL13,25-28 Some investigators have, however, doubted this association. 23 ,24,29,3o Other preceding coincidences have been described, including inoculation with vaccines 31 ,32 related to or not related to viruses 33-36 (e.g" varicella,37-39 rubella, rubeola,37 hepatitis A and B),40,41 Mycoplasma pneumoniae,42.43 and Bartonella henselae. 44 As the term anaphylactoid purpura suggests, allergy has been regarded by some to be the basis for development of this disease after insect bites4 and exposure to drug and dietary allergens. 45 The universal vascular deposition of immunoglobulin A (IgA) suggests that HSP is an IgA-mediated immune response and may operate through the alternative complement pathway.46 In a study from Taiwan,47 serum levels of IgA anticardiolipin antibodies were elevated during the acute stage of the disease, along with transforming growth factor-~ (TGF-~) - secreting T cells (TGF-~ is a major factor in IgA production and may be involved in pathogenesis). Disorders of coagulation and its activation are also associated with development of HSP or an HSP-like vasculitis. 48 One study concluded that oxidant stress, especially lipid peroxidation, was involved in the origin of renal injury.49 Vasculitis may develop after antirheumatic therapy, including administration of methotrexate50 and anti-tumor necrosis factor (TNF) agents,SI
Genetic Background Familial clusters of the disease may occur, with siblings affected simultaneously or sequentially.21,52,53 Investigations from Spain have presented preliminary data on genetic associations. HLA-B35 was increased in
51
C HAP T E R
23 LEU K0
C Y Toe LAS TI C
V AS C U LI TIS
497
Olnlcal Manifestations Clinical characteristics of HSP are presented in Table 23-2. 15 ,59-61 The onset is often acute with the principal manifestations appearing sequentially over several days to weeks. Nonspecific constitutional signs, such as a lowgrade fever or malaise, are often present. 7,62
Cutaneous Involvement
• FIgare 23-1 Histopathologic demonstration of leukocytoclastic vasculitis. The characteristic "nuclear dust" is seen as granular, dark-stained material in the vessel wall. Hematoxylin and eosin stain; magnification x 480.
frequency in patients who developed nephritis.54 The incidence of HLA-DRBl·Ol was also increased compared with matched controls, and HLA-DRBl·07 was decreased.55 Although there were no general associations with the expression of intercellular adhesion molecule 1 (ICAM-I) in patients compared with controls, a KlE polymorphism at codon 469 was significantly decreased in those who did not develop severe gastrointestinal manifestations (and possibly in patients without renal sequelae).56 In a study from Israel, familial Mediterranean fever genotypes (MEFV) were more numerous (l0%) than expectedY Other genetic polymorphisms have also been implicated. 58 HSP has also been described in a number of patients with heterozygous C2 complement component deficiency (see Chapter 3).
'~III
1Mil E 23 I
Conditions Associated with leukocytodastic
\J,,~(ulitis
Henoch-Schonlein purpura Hypersensitivity angiitis Hypocomplementemic urticarial vasculitis Mixed cryoglobulinemia Cutaneous polyarteritis ANCA-associated small-vessel vasculitis' GOOdp:lsture's syndrome Rheumatic disorders' Systemic lupus erythematosus Juvenile dermatomyositis Mixed connective tissue disease Scleroderma Juvenile rheumatoid arthritis Mucha-Habermann Disease Relapsing polychondritis Kohlmeier-Degos syndrome Antiphospholipid antibody syndrome Malignancy-associated disease Sweers syndrome Cronkhite-Canada syndrome Stevens~'ohnson syndrome Erythema elevatum diutinum 'Leukocytodastic vasculitis may occur in cutaneous lesions found in some patients with ANCA-associated vasculitis and collagen vascular diseases, ANCA, antinuclear cytoplasmic antibody.
The presence of palpable purpura is essential to the diagnosis. 15 ,63.64 This rash is most prominent on dependent or pressure-bearing surfaces, especially the lower extremities and buttocks, but it may occur in other areas. The cutaneous lesions range from small petechiae to large ecchymoses to rare hemorrhagic bullae; they tend to occur in crops and progress in color from red to purple to brown (Figs. 23-2 and 23-3). Ulceration may occasionally develop in large ecchymotic areas. The rash is often preceded by maculopapular or urticarial lesions. Subcutaneous edema over the dorsa of the hands and feet and around the eyes, forehead, scalp, and scrotum may occur early in the disease, particularly in the very young child.
Gastrointestinal Disease Gastrointestinal manifestations occur in approximately two thirds of children, usually within a week after onset of the rash and almost always within 30 days; in 14% to 36% of cases, abdominal pain precedes other manifestations.60.6~7 Edema and submucosal and intramural hemorrhage resulting from vasculitis of the bowel wall occasionally lead to intussusception (usually confined to the small bowe!), gangrene, or overt perforation. Less common involvement includes acute pancreatitis,68,69 hepatobiliary involvement,70 ulcerative colitis, other forms of enteropathy,71 and steatorrhea. 72 In one study,60 abdominal pain was usually intermittent, colicky, and periumbilical. Rebound tenderness was uncommon. Vomiting occurred in 60%, hematemesis in 7%, and melena in 19% of children, although occult blood was present in the stools of one half of the patients. Massive gastrointestinal hemorrhage or intussusception occurred in less than 5% of children, but it could develop suddenly without preceding abdominal symptoms.
Renal Disease Glomerulonephritis affects up to one third of the children, but in less than 10% it is a serious and potentially lifethreatening complication. 4,20,21,24,30 ,37 In the Belfast study of 155,000 unselected children, 55 of 270 patients had evidence of nephritis at onset.20 Renal disease, like abdominal pain, seldom precedes the purpura, and in most instances, serious renal disease develops within 1 month of onset of the rash. The spectrum of manifestations ranges from microscopic hematuria and mild proteinuria to the less common nephrotic syndrome, acute nephritic syndrome, hypertension, or renal failure. Age at onset of more than 7 years, persistent purpuric lesions, severe abdominal symptoms, and decreased factor XIII activity
498
C HAP T E R
23 LEUKOCYTOCLASTIC VASCULITIS
t._.
. ' "'" lABEL 23 2
C1ini( ,11 Fe,llllles 01 Henodl SdlOnlein
PUrpUld
Percent of Patients with the alnlcal Feature (oAl) Emery etal, Rosenblum II
alnlcal Feature Purpura Arthralgia!arthritis Abdominal pain Renal involvement Subcutaneous edema Gastrointestinal bleeding Encephalopathy Orchitis
1977" (n = 43)
Winter, 198780 (n = 43)
lOa'
79 63 37 63
Balla etal, Saulsbury, 199111 199915 (n = 47) (n = 100)
Averalle (GAl)
97 65 100'
96 47 64 51 21
lOa'
82 63 40
99' 82 70 42 41
26
26
33 8 4
29 8 5
6
'Criterion used for inclusion in the series.
are associated with an increased risk of nephritis. 73 The initial 3 months are the most critical in determining the eventual extent of the illness. In a few children, however, nephritis may not occur until much later in the course, sometimes after a number of recurrences of the purpura. Renal involvement characteristically develops early, but end-stage disease may not be obvious for a number of years. In a small number of children, renal abnormalities
occur alone and clinically and immunopathogenically resemble Berger's nephropathy in adults.
Arthritis Arthralgia or arthritis involving only a few joints occurs in 50% to 80% of children with HSP. Large joints, such as the knees and ankles, are most commonly affected, but other
• Figure 23-Z Henoch-Schonlein purpura. A, These purpuric lesions appeared on the lower extremities of a 10-year-old boy who had an acute, self-limited illness characterized by fever, arthritis, melena, and transient hematuria. Notice the periarticular swelling around the ankle and knee. 8, Purpura on dorsum of the hand of a 14-year-old-boy with Henoch-Schonlein purpura. (See color insert.)
C HAP T E R
23
LEU K0 C Y Toe LAS TI C
V AS C U LI TI 5
499
• Rgure Z3-4 Leukocytodastic vasculitis in the skin of a patient with Henoch-Schiinlein purpura. Hematoxylin and eosin stain; magnification x 480.
streptococcal antigen (i.e., nephritis-associated plasmin receptor) was identified in the mesangium of 10 of 33 children by immunofluorescent microscopyY Levy and coworkers 24 proVided a comprehensive review of the renal pathology. The principal lesion is an endocapillary
• fIIure Z:l-3
Palpable purpuric lesions involve the heels and ankles of a patient with Henoch-Schiinlein purpura. (See color insert.)
areas, including wrists, elbows, and small joints of the fingers, may be involved. Characteristic findings include periarticular swelling and tenderness, usually occurring without erythema, warmth, or effusions but with considerable pain and limitation of motion. The joint disease is transient, although usually not migratory, and resolves within a few days to a week without residual abnormalities. Occasionally, arthritis may precede the appearance of the rash by 1 or 2 days.30.59
Other Manifestations Other manifestations of HSP include an isolated central nervous system vasculitis, seizures,74 coma and hemorrhage,75-78 Guillain-Barre syndrome,79 ataxia and central and peripheral neuropathY,80.81 ocular involvement,30,60,82 intramuscular, subconjunctival or pulmonary hemorrhages,83-&> interstitial pneumonitis,87 recurrent epistaxis, parotitis,24 carditis,4,88,89 and orchitis, epididymitis, or testicular torsion (2% to 38%),6.~,90-96
Pathology The pathologic lesion essential for diagnosis is leukocytoclastic vasculitis (Fig. 23--4).1,24,97,98 In the skin, this is demonstrated in the dermal capillaries and postcapillary venules. Deposition of IgA (principally IgAI) in these lesions is characteristic. 15 .46,99-101 In the kidney, proliferative glomerulitis ranges from focal and segmental lesions to severe crescentic disease (Fig. 23-5).100,102 Group A
• Rgure Z:l-5 A, Diffuse, mesangial. proliferative nephritis in HenochSchiinlein purpura. Hematoxylin and eosin stain; magnification x 480. B, Mesangial and capillary wall deposition of immunoglobulin A(immunoperoxidase stain) in Henoch-Schiinlein purpura.
500
C HAP T E R
23
LEUKOCYTOCLASTIC VASCULITIS
proliferative glomerulonephritis with an increase in endothelial and mesangial cells, All gradations of severity may be present in the same biopsy. There may be marked interstitial inflammatory disease, but vasculitis per se is usually not present. Fluorescence microscopy confirms deposits of immunoglobulin, principally IgA,46,loI,J03 but it is often accompanied by IgG, fibrin, C3, and properdin in most involved glomeruli. These deposits are invariably in mesangial cells, but peripheral capillary loops also are involved in more severe cases, Dense deposits in the mesangium and occasionally in the subendothelial paramesangial regions are present on electron microscopy. Thickening and splitting of the basement membrane, caused by the interposition of mesangial cell cytoplasmic material, are notable.
Differential Diagnosis The American College of Rheumatology criteria are in Table 23-3,64 HSP must be distinguished from immune thrombocytopenic purpura, acute poststreptococcal glomerulonephritis,28 SLE, septicemia, disseminated intravascular coagulation, the hemolytic-uremic syndrome, the papular-purpuric gloves-and-socks syndrome,104 and other types of vasculitis. 6,10'H07 The more common causes of an acute surgical abdomen with abdominal pain and gastrointestinal tract bleeding must be considered, A tender abdominal mass may indicate intussusception, and abdominal tenderness with an elevated serum amylase level may suggest acute pancreatitis, Punch biopsy of a cutaneous lesion may assist in the diagnosis of difficult cases by demonstrating a leukocytoclastic vasculitis characterized by deposition of IgA and C3. 46 A renal biopsy is indicated only in children with persistent or significant renal manifestations,24 Infantile acute hemorrhagic edema (IARE) affects infants between 4 and 24 months old with an acute onset of fever, purpura, ecchymoses, and inflammatory edema of the limbs and face. 10Il-121 Although spontaneous remission in 1 to 3 weeks and a benign course are characteristic, attacks may recur, Involvement of viscera (e,g., kidneys, gastrointestinal tract) is uncommon. Histopathology is typical of a leukocytoclastic vasculitis with
U:..
TABl [23 3
(rileri,\ 101 Cldssili
PurplIId
CrIterion·
DefInition
Palpable purpura
Slightly raised, "palpable" hemorrhagic skin lesions not related to thrombocytopenia Patient ~ 20 yr old at onset of first symptoms Diffuse abdominal pain that is worse after meals or the diagnosis of bowel ischemia, usually including bloody diarrhea Histologic changes showing granulocytes in the walls of arterioles or venules
Age
~
20 yr at onset
Bowel angina
Wall granulocytes on biopsy
'Por purposes of classification, a patient is said to have Henoch-Schtinlein purpura if at least two of these criteria are present, The presence of any two or more criteria yields a diagnostic sensitivity of 87.1% and specificity of 87,7%, Prom Mills lA, Michel BA, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of Henoch-Schtinlein purpura. Arthritis Rheum 33: 1114-1121, 1990.
occasional demonstration of perivascular IgA deposition,122 This disorder in older children overlaps clinically with HSP. HSP is uncommon in adults, with a reported incidence of 0,12 cases per 100,000 persons, Males are affected as commonly as females. 14,16,123--127 In a study of clinical features and outcome in an unselected population of 46 adults and 116 children with HSP,I211 cutaneous lesions were the principal initial manifestation in both groups, However, adults had a lower frequency of abdominal pain and fever and a higher frequency of joint symptoms and severe renal involvement. Adults often reqUired more aggressive therapy with glucocorticoids or cytotoxic agent~, or both. Outcome was relatively good in both age groups, with a complete recovery in 94% of children and 89010 of adults, Another study indicated that leukocytosis, thrombocytosis, and elevated levels of serum Creactive protein were more common in children, whereas elevated serum IgA and cryoglobulin levels were more common in adults. 124 HSP in adults may represent a more severe form of the disease with a higher frequency of significant renal involvement but without the other manifestations of HSP,124-127
Laboratory Examination There are no diagnostic laboratory abnormalities. 5M3 ,G4 The platelet count is normal or increased, differentiating this form of purpura from that caused by thrombocytopenia, A moderate leukocytosis of up to 20,000 white blood cells/mm 3 (20 x 109/L) with a left shift is identified in some children. Normochromic anemia is often related to gastrointestinal blood loss, confirmed by a positive stool guaiac examination in 80% of the children who have abdominal complaints. Antinuclear antibody and rheumatoid factor are not characteristic. Although renal disease may occur in the absence of overt urinary findings and minimal abnormalities such as hematuria are not necessarily associated with a severe glomerular lesion, these laboratory abnormalities usually demonstrate a direct correlation with the severity of the proliferative changes, Patients occasionally have decreased concentrating ability and creatinine clearance. Proteinuria, sometimes severe enough to result in hypoalbuminemia, may occur,59,129 Although levels of Clq, C3, and C4 are usually normal,130 activation of the alternative complement pathway is demonstrated in one half of the children during the acute illness by the presence of C3d, low levels of total hemolytic complement, and decreased concentrations of properdin and factor B in the serum,130,131 Plasma levels of von Willebrand factor antigen are elevated, indicating endothelial cell damage. 132 ,133 Circulating IgA-containing immune complexesLH-136 and cryoglobulins 137 may be present. Serum IgA and IgM concentrations are increased in one half of the patients during the acute phase of the disease,138 An increased number of circulating IgA-producing cells was found in one study in definite HSP cases hut not in other forms of leukocytoclastic vasculitis. 139 Antibodies to neutrophil cytoplasmic antigens usually are not detected.
Radiologic Examination Plain radiographs delineate decreased bowel motility with dilated loops of bowel in children with abdominal involvement. Ultrasound studies can identify specific gastrointestinal
C HAP T E R
abnormalities in children with abdominal complaints,65 and magnetic resonance imaging and magnetic resonance angiography can define the extent of cerebral vasculitis. 14D-142 Occasionally, intussusception is identified on a barium study and is relieved by it if performed early in the course.63.143 Epididymal enlargement, subcutaneous swelling or hydrocele, or rarely, testicular torsion can be confirmed if necessary by scrotal ultrasonography.91
T......ent Treatment is supportive with maintenance of good hydration, nutrition, and electrolyte balance and with control of pain with simple analgesics such as acetaminophen and, if necessary, control of hypertension.144 Although glucocorticoids dramatically decrease the severity of joint and cutaneous disease, they are not usually indicated for management of these manifestations. 15 Short-term glucocorticoid therapy is effective in relieving the pain of severe orchitis. Prednisone has been advocated in children with severe gastrointestinal disease or hemorrhage. 25 ,60,145.146 However, studies do not demonstrate a clear advantage of prednisone over supportive therapy (e.g., nasogastric suction, parenteral nutrition, antibiotics). The severity of disease may prompt the use of intravenous methylprednisolone.146.147 Glucocorticoids have no universally accepted role in thetreatrnent of the renal disease,15.37,73,148-153 although adequate therapeutic trials have not been reported. Intravenous methylprednisolone may be effective if started early in the course of clinical renal disease.1S! There have been few studies of the efficacy of cytotoxic drugs in management of the nephritis. 15,154-157 Warfarin and heparin have also been used with disputable effect,154 as have cyclosporine,158.159 intravenous immunoglobulin, and plasma exchange. 16.160-162 Renal transplantation has been successful in some children with renal failure. 163,164 In pooled data 163,165 there was a 35% risk of recurrence 5 years after transplantation and an 11% risk of graft loss.
23
LEUKOCYTOCLASTIC VASCULITIS
501
the occurrence of numerous exacerbations associated with nephropathy suggests a poor prognosis for renal function. 102.172 Additional poor prognostic factors 172 are decreased factor XIII activity, hypertension and renal failure at onset, and if a renal biopsy had been performed, an increased number of glomeruli with crescents, macrophage infiltration, and tubulointerstitial disease. The reported outcome of children with renal disease is highly variable. 173 With minimal lesions, more than 75% recover within 2 years; in contrast, two thirds of children with crescentic glomerulitis in more than 80% of glomeruli progress to renal failure within the first year. The worst outcome was associated with the presence of the nephrotic or nephritic syndrome at onset. 148 Almost one half of such children had active renal disease or renal insufficiency at follow-up periods of 6 or more years. Thus the extent of the renal disease has been the ultimate determinant of long-term outcome.162.174 Overall, less than 5% of children progressed to end-stage renal failure. HSP accounts for less than 1% of children with renal failure from all causes. In a follow-up evaluation of 64 children, 162 the renal survival rate at 10 years was 73%, and initial renal insufficiency was the best predictor of the future course of nephritis. The long-term mortality rate is less than 1%.20 Patients who have had clinical nephritis should be followed for at least 5 years. 14B,167.172. 173 .176 In one study, 8 of 12 patients had mesangial IgA deposition at follow-up of 2 to 9 years despite an apparent clinical remission of their renal disease. Later, 16 of 44 pregnancies in adults were complicated by proteinuria or hypertension, even in the absence of active renal disease. 173 Of 18 patients in the Belfast study who had a nephrotic or nephritic syndrome at onset 20 and who were followed a mean of 8.3 years, 1 had died and 3 had persistent urinary abnormalities but no azotemia. The overall mortality rate was less than 1%, and the morbidity rate was 1.1%. This somewhat optimistic outcome is tempered by a study of 16 children from Minnesota that indicated the longer a child was followed, the more likely it was that renal disease would become clinically evident. 175
HYPERSENSITIVITY ANGIITIS Course of the Disease and Prognosis In two thirds of children, HSP runs its entire course witl;lin 4 weeks of onset.144.148,166.167 Younger children gen~rally have a shorter course and fewer recurrences than older patients. One third to one half of the children have at least one recurrence that commonly consists of a rash and abdominal pain, with each episode usually being similar but briefer and milder than the preceding one. 15 Most exacerbations take place within the initial 6week period but may occur as late as 2 years after onset. They may be spontaneous or coincide with repeated respiratory tract infections. The severity of the cutaneous leukocytoclastic vasculitis does not correlate with visceral involvement. 98 Prognosis is excellent for most children. 168-170 Significant morbidity or mortality is associated with gastrointestinal tract lesions in the short term and with nephritis in the long term.14.162.171 The development of major indications of renal disease within the first 3 months after onset or
Vascular inflammation in hypersensitivity angiitis occurs more typically in smaller vessels than in those involved in the classic form of polyarteritis nodosa and, in this regard, resembles HSP. Previously, it was the most frequently encountered form of vasculitis after the administration of therapeutic antisera. 177 The Chapel Hill International Consensus Conference did not use the term hypersensitivity vasculitisY8 It was proposed instead that microscopic polyarteritis and cutaneous leukocytoclastic vasculitis were best equated with common usage of this designation. The terminology used to describe leukocytoclastic vasculitis resulting from an allergic reaction remains confuSing. The American College of Rheumatology criteria define hypersensitivity vasculitis (Table 23-4)179 as palpable purpura, with or without a maculopapular rash precipitated by a medication or other agent, and a biopsied lesion characterized by a neutrophilic perivascular or extravascular infiltration in small vessels (like those affected in HSP).180.181
502
II
C HAP T E R
TABI E 23 -4
23
LEU K0 C Y Toe LAS TI C
Criterid for
Didgllo~is
of
V AS CULl TIS
Iiyper~ell~itivity
Vd~(uliti~
CrIterion·
Deflnhlon
Age at onset >16 yr Medication at disease onset
Development of symptoms after age 16 yr Medication that may have been a precipitating factor was taken at the onset of symptoms Slightly elevated purpuric rash over one or more areas of skin; does not blanch with pressure and is not related to thrombocytopenia Flat and raised lesions of various sizes over one or more areas of the skin Histologic changes showing granulocytes in a perivascular or extravascular location
Palpable purpura
Maculopapular rash Biopsy, including arteriole and venule
'For purposes of classificatlon, a patient is said to have hypersensitivity vasculitis if at least three of these criteria are present. The presence of any three or more criteria yields a diagnostic sensitivity of 71.0% and specificity of 83.9%. The age criterion Is not applicable for children. From Calabrese LH, Michel BA, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of hypersensitiVity vasculitis. Atthritis Rheum 33: 1106-1113, 1990.
A report of serum sickness-like arthritis in Finland estimated its frequency at 4.7 cases per 100,000 children younger than 16 years, establishing it as one of the most common causes of acute arthritis in childhood. 182 In this study, the arthritis was transient, usually lasting only a few weeks, and most commonly affected the ankles, metacarpophalangeal joints, wrists, and knees (Fig. 23-6). Occasionally, pulmonary, renal, and other vasculature systems were affected (Fig. 23-7). Leukocytosis usually occurs in cases of hypersensitivity angiitis and is sometimes accompanied by eosinophilia and circulating immune complexes. 182 The erythrocyte sedimentation rate is often normal. IgG antibodies to the putative antigen may be demonstrable. Synovial fluid examination in one report demonstrated 8800 to 59,000 leukocytes/mm3 (8 to 59 x 109L), of which 38% to 80% were polymorphonuclear leukocytes. 182 Biopsy of a cutaneous lesion confirms that small venules and capillaries are the predominantly involved vessels. Inflammatory lesions are at a similar stage of development in all areas of involvement, and the cellular infiltrate contains large numbers of neutrophils and eosinophils. Systemic treatment is directed at the relief of symptoms because the course, although often acute and variable, is self-limiting. Removal of the precipitating agent, if identified, is the first step in treatment. In the absence of systemic features, management is usually symptomatic. Antihistamines and nonsteroidal anti-inflammatory drugs (NSAIDs) alleviate cutaneous symptoms and arthralgias, respectively. Glucocorticoid therapy may be indicated in children with severe cutaneous symptoms or systemic vasculitis. Historically semm sickness, a classic example of immune complex-mediated disease in humans, was encountered after the administration of heterologous antisemm to treat or prevent specific infections such as diphtheria and tetanus. Although current indications for use of heterologous antiserum are uncom-
• Figure 23-6 Diffuse and periarticular swelling of the hand in a boy with acute serum sickness.
mon, it has been superseded as a cause of serum sickness by myriad drugs, notably cefaclor, penicillin, sulfonamides. quinolones, allopurinol, thiazide diuretics, NSAIDs, phenytoins, iodides, antithyroid drugs, and rarely, other medications, such as
• figure 23-7 Hypersensitivity angiitis is demonstrated in a lung biopsy spedmen from a young drug addict with ashort history of increasing dyspnea on exertion and purpura.This section shows prominent infiltration by inflammatory cells and eoslnophils of the alveolar walls and around blood vessels. Hematoxylin and eosin stain; magnification x 480.
C HAP T E R
23 1
LEUKOCYTOCLASTIC VASCULITIS
503
preted to indicate that HSP and hypersensitivity angiitis were similar, but separable, clinical disorders. Although only seven of the younger patients had hypersensitivity angiitis (attesting to the rarity of this disorder in pediatric patients), this study was unable to document any clinical differences between them and the older patients. In contrast, the 60 younger patients with HSP had less extensive and severe disease than adults, especially in relation to renal involvement.
streplokinase, recombinant human growth hormone, cytokines, and monoclonal antibodies.182.183 TIle clinical syndrome begins 7 to 14 days after primary exposure to the antigen and is characterized by fever, arthralgia, frank arthritis (sometimes), myalgia, lymphadenopathy, and a rash that may be purpuric, linear, urticarial, or ecchymotic and distributed predominantly over the lower legs, although the trunk and arms may be involved (Fig. 23-8). Kunnamo and colleagues 182 described a patchy discoloration over the affected joints, with urticaria predominantly on the trunk. With the use of equine antithymocyte globulin in the treatment of bone marrow failure, 30 of 35 patients developed senUll sickness characterized by malaise, headache, fever, cutaneous emptions, artl1ralgias, arthritis, myalgias, gastrointestinal complaints, and lymph node enlargement, beginning 7 to 9 days after infusion and lasting 10 to 14 days. 177 In the study by Michel and associates180 in which HSP and hypersensitivity angiitis were directly compared in patients 20 years old or younger, transient arthralgias and oligoarthritis, myalgias, cutaneous nodules, ulcerations, livido, gangrene, and eosinophilia were more common in hypersensitivity angiitis. In HSP, gastrointestinal bleeding, hematuria, and palpable purpura were more frequently encountered. These results were inter-
Children, usually girls, with this rare syndrome have recurrent episodes of urticaria that are associated with pruritus and a burning sensation. The urticaria resolves over 2 to 4 days, leaVing only residual pigmentation (Fig. 23-9).184-186 Other skin lesions include purpura, papules, and vesicles. Fever, nausea, vomiting, and abdominal pain may accompany each cutaneous exacerbation. Arthralgias occur in approximately 60% and
• figure 23-8 Skin lesions of a patient with hypersensitivity vasculitis.
• Figure Z3-9 Linear bands of urticarial lesions in a 6-year-old girt with hypocomplementemic urticarial vasculitis. These lesions were transient and recurrent. (See color insert.)
HYPOCOMPLEMENTEMIC URTICARIAL VASCULITIS
504
C HAP T E R
23
LEU K0 C Y Toe LAS TIC
V AS C U LI TI S
arthritis, usually of small joints, in 30% of patients. The arthritis has a brief duration and has no long-term residua. Abdominal and chest pain occur in 25%, pulmonary disease (e.g., cough, dyspnea, hemoptysis) in 30%, and glomerulonephritis in 15% of patients. Less commonly, uveitis and episcleritis, fever, angioedema, Raynaud's phenomenon, pseudotumor cerebri, and seizures may be associated findings. Rapidly progressive glomerulonephritis and pulmonary hemorrhage have been described. 187 Hypocomplementemic urticarial vasculitis (HUV) has also been associated with SLE,1138-190 Sjogren's syndrome, hepatitis Band C antigenemia, drug reactions, and excessive exposure to sun. 191 A separate group of patients with urticarial vasculitis have normal complement levels. I86 ,l89 Patients with hypocomplementemia tend to have more severe systemic symptoms,185 The pathogenesis of this type of vasculitis is unknown, but an immune complex process with activation of the classic complement pathway is suggested by lesional deposits of immunoglobulins and complement and circulating immune complexes. 192 Levels of early and late components of the complement cascade may be depressed, with the degree of hypocomplementemia paralleling the severity of the disease. Antinuclear antibody, rheumatoid factor, and cryoglobulins are usually absent. An IgG antibody to the collagen-like region of C1q has been described (HUV, 100%; SLE, 35%)192 Levels of C3, C4, and Clq are reduced in 18% to 50%. Skin biopsy documents a leukocytoclastic vasculitis, predominantly a venulitis, with abundant neutrophilic and occasionally eosinophilic infiltrates. 18s ,189,191 Immunofluorescence microscopy confirms the deposition of IgM and C3 in affected vessels. A mild membranoproliferative glomerulonephritis may accompany the cutaneous disease. Management consists of supportive measures and treatment of any associated disorders. Antihistamines, dapsone, hydroxycWoroquine, colchicine, and indomethacin have been used with variable success. 18S Systemic glucocorticoids or other immunosuppressive drugs may be required in severely involved children. The course of the disease is usually benign but may vary depending on the associated disorders and extent of systemic involvement.
MIXED CRYOGLOBUUNEMIA Vasculitis with essential mixed cryoglobulinemia (types II and III) clinically resembles other leukocytoclastic vasculitides. This disease is defined by the Chapel Hill Consensus Conference as vasculitis with immune deposits in capillaries, venules, or arterioles of the skin and kidney that are associated with cIYoglobulins in the serum. 178 Purpura on the distal extremities, often precipitated by exposure to cold, is accompanied by arthralgia, with or without frank arthritis,193 and by glomerulonephritis in up to one half of the patients. 194 Patients also may have pulmonary involvement. High levels of mixed cryoglobulins (i.e., IgG and IgM that reversibly precipitate on cooling), sometimes with hepatitis BI94 or coccidioidin antigen,19S are the serologic hallmark of the disorder. Cryoglobulinemic vasculitis is rare in children l93 and results in an acute vascular inflammation after localiza-
tion of mixed cryoglobulins in vessel walls,196 Low levels of C4 with normal or slightly low C3 levels are distinctive. Infection with hepatitis B or C virus must always be excluded. 194,197 Progressive renal disease that resembles membranoproliferative glomerulonephritis is the principal cause of long-term morbidity.195 Outcome is related to severity of the systemic disease. Treatment with combinations of glucocorticoids, cyclophosphamide, plasmapheresis, and intravenous immunoglobulin may be of benefit in some patients. 198
OTHER LEUKOCYTOCLASTIC AND PERIVASCUUTIC SYNDROMES AND SIMILAR DISORDERS In several other types of vasculitis, the histopathology of the lesions has a leukocytoclastic element. Some of these disorders, such as cutaneous polyarteritis and sarcoidosis, are discussed in Chapters 24 and 26. The differential diagnosis of systemic vasculitis is often challenging in that other forms of secondary vasculitis and vasculitic mimics, such as thromboangiitis obliterans (Buerger's disease) and infectious angiitis (including syphilis and Lyme disease), must be considered. Other syndromes are more difficult to classify: vasculitis associated with retroperitoneal fibrosis, inflammatory bowel disease, primary biliary cirrhosis, and transplant rejection. Children may present with a vasculopathy from ergot poisoning, neurofibromatosis, coarctation-hypoplasiadysplasia, and embolism.
Antlneutrophll Cytoplasmic Antlbody-Assodated Small-Vessel Vasculitis Antibodies to neutrophil cytoplasmic antigens (ANCAs) are associated with three major vaSCUlitides, all of which are uncommon in children: Wegener's granulomatosis, microscopic polyarteritis, and the Churg-Strauss syndrome (see Chapter 26).199-202 Wegener's granulomatosis and the Churg-Strauss syndrome are characterized by granulomatous inflammation involving the respiratory tract and necrotizing vasculitis affecting small- and medium-sized vessels (i.e., capillaries, venules, arterioles, veins, and arteries).203,204 Cutaneous involvement is common and consists of petechiae, palpable purpura, skin ulcers, papules, and nodules. I99 ,205 Skin lesions rarely precede the appearance of other features of these illnesses. Rapid diagnosis of ANCA-associated vasculitis is important because appropriate immunosuppressive treatment often limits life-threatening injury to vital organs.
Goodpasture's Syndrome In 1919, Goodpasture 206 described a patient with pulmonary hemorrhage and severe crescentic proliferative glomerulonephritis, leading to death. Constitutional symptoms (e.g., fever, chills, increased sweating) occur in one fourth of patients, and pulmonary complaints (e.g., dyspnea, weakness, chest pain, wheeZing) are common. 207 Pulmonary hemorrhage is often the initial manifestation and may precede renal abnormalities anywhere from weeks to years, or clinical progression may be rapid, Serum antibodies to the alpha 3 chain of type IV
C HAP T E R
collagen (NCI domain) in alveolar and glomerular basement membranes are diagnostic,20H,209 These antibodies can be demonstrated as linear staining of the basement membranes in lung tissue and glomemli by immunofluorescence microscopy. This disease predominantly affects young men with a male/ female ratio of 9: 1. It has only occasionally been reported in children or adolescents. 207.2JO-214 In children, idiopathic pulmonary hemosiderosis, Wegener's granulomatosis, hemolyticuremic syndrome, and SLE are considerations in differential diagnosis. Goodpasture's syndrome has occurred after therapy with D-penicillamine, in cases of heavy metal poisoning, and in patients with a variety of rheumatic diseases. Treatment includes the prompt use of glucocorticoids, plasmapheresis, and immunosuppressive agents,2JO-213,215 The rationale for this combined approach is that circulating anti-basement membrane antibodies are removed from the vascular compartment by plasmapheresis, and their synthesis is limited by immunosuppressive agents. Nonetheless, the survival rate is low. Death is caused by asphyxia, pulmonary hemorrhages, or uremia.
VaSCUlitis Assodated with Connective
23
LEUKOCYTOCLASTIC VASCULITIS
505
(see Chapter 27) develop papular or pustular lesions that may ulcerate from a vasculitis with a predominant lymphocytic infiltrate, but a leukoeytoclastic vasculitis may be present.
Mucha-Habermann Disease Mucha-Habermann disease, or pityriasis Iichenoides et varialiformis acuta (PLEVA), is a form of cutaneous vasculitis of unknown origin. At presentation, the dermatitis has the appearance of chronic or recurrent chickenpox-like lesions that become atrophic and scarred. The rash is accompanied by fever and joint pain and swelling (Fig. 23-10). Histologically, the lesions are characterized by a lymphocytic inflammation of capillaries and venules of the upper dermis. PLEVA has been described in two children with chronic arthritis resembling juvenile rheumatoid arthritis, one of whom developed severe acrosclerosis and scleroderma late in her course. 221 A third patient with similar cutaneous findings had been reported by Lister and Hollingworth.m
nssue Disorders
Relapsing Polychondrltls
Leukocytoclastic vasculitis may occur in SLE, usually presenting as palpable purpuriC lesions or urticaria,IHB,216 and in dermatomyositis, mixed connective tissue disease, scleroderma, and rarely in juvenile rheumatoid arthritis (see Chapters 9, 16, 18, 19, and 21 ). Vascular involvement is particularly well recognized in juvenile dermatomyositis, in which small-vessel vasculitis has been identified in striated muscle, skin, subcutaneous tissue, and the gastrointestinal tract. 105,217,218
Relapsing polychondritis is a rare, idiopathic, widespread inflammation of cartilage that is associated with uveitis, deafness, vestibular involvement, and aortic valve insufficiency.m It has seldom been observed in children,224-228 Differential diagnosis includes Wegener's granulomatosis and a variety of infections. Inflammation affects the cartilage of the ear or hyaline cartilage of the joints and then spreads to involve cartilage of the upper respiratory tract, including the nose, trachea, and bronchi (Fig. 23-11 and see Fig. 26-6). Episodic oligoarthritis occurs in approximately 80% of affected patients. The synovial fluid is noninflammatory, and the synovial membrane demonstrates minimal inflammation. 226 The course is initially episodic but becomes progressive in most patients. Relapsing polychondritis has been described in one patient with HSP.229 Intrauterine transmission has been suggested in another, but this association appears to be exceptional. 230
Familial Mediterranean Fever and
Beh~et's
Disease
A number of reports describe the occurrence of HSP in patients with familial Mediterranean fever (see Chapters 24 and 34).219,220 In some patients, the onset of vasculitis may be the first indication of this disorder. Similarly, patients with Beh~et's disease
• figure Z3-10 An ll-year-old, black girl with destructive acrosclerosis and Mucha-Habermann disease. A, Hand. B, Forearm. The characteristic cutaneous lesions of Mucha-Habermann disease are visible, along with advanced ischemic digital changes. (8, See color insert.)
506
C HAP T E R
23 LEU K 0 C Y TO CLAS TI C V AS CUll TIS • Figure 23-11 Histopathology of relapsing polychondritis. A, Biopsy spedmen of ear cartilage. Hematoxylin and eosin stain; magnification x 250. B, Oose-up view. Hematoxylin and eosin stain; magnification x 600. Acute inflammation and perichondritis are evident, with numerous lymphocytes and plasma cells and smaller numbers of polymorphonudear cells. The edge of the aural elastic cartilage is being destroyed by the cellular exudate.
Glucocorticoicls suppress the disease, Death is often the result of respiratory obstruction.m
and HSP (see Chapters 26 and 21). Occasionally, vasculitis precedes the diagnosis of a lymphoproliferative syndrome,
KiihlmeleF-Degos Syndrome
Sweet's Syndrome
The Kohlmeier-Degos syndrome has a number of other names, including malignant atrophic papulosis, papulosis atrophicans maligna, and progressive arterial occlusive disease. 23 1.232 It is a rare, often fatal vasculitis of cutaneous, gastrointestinal, and central nervous system small- and medium-sized arteries that results in progressive occlusion by fibrosis, leading to infarction. It occurs almost exclusively in young to middle-aged men and has been reported in three teenaged boys.232 Its presentation and course in adults have been reviewed by Snow and Muller. 233 Some investigators regard it as a lupus variant. 234
Acute febrile neutrophilic dermatosis (Sweet's syndrome) is rare in children and consists of an inflammatory perivasculitis with a dermal infiltrate of mature neutrophiles characterized clinically by spiking fever and tender, raised, pseudovesicular, erythematous plaques or nodules on the face and extremities and sometimes on the trunk,2-H-247 Arthritis occurs in one third of adult patients 248 and has been described in an 8-month-old boy,217 Musculoskeletal pain, including arthritis or multifocal osteomyelitis, has also been reported in children. 247 ,li9,15" This syndrome may be secondary to malignancy and its treatment, Behc,;et's syndrome, or miscellaneous disorders.
Antlphosphollpld Antibody Syndrome Patients with the antiphospholipid antibody syndrome have repeated episodes of intravascular coagulation and thrombosis (see Chapter 16). In the absence of identifiable systemic disease, these patients may be erroneously diagnosed as having idiopathic vasculitis. 235 ,2.16 Atrial myxoma may simulate vasculitis by embolization237-2.w and should be considered in the diagnosL'i of an obscure vasculitis-like syndrome. This phenomenon has only rarely been reported in children or adolescents, 24G-242 Echocardiography demonstrates the lesion 242 and surgical removal is curative. Infectious endocarditis produces a similar but not identical clinical syndrome.
Vasculitis Assodated with Malignancy Lymphoproliferative disease rarely is accompanied by a leukocytoclastic vasculitis limited to the skin, Lymphocytic lymphoma and Waldenstrom's macroglobulinemia, both rare in children, can result in a cryoglobulinemic vasculitis,243 Lymphoma may also be associated with Wegener's granulomatosis, Sjogren's syndrome,
Cronkhite-Canada Syndrome Juvenile gastrointestinal polyposis is rarely associated with a widespread vasculitis of small- and medium-sized arteries, the Cronkhite-Canada syndrome. 251 ,m The syndrome is characterized by cutaneous anergy, skin hyperpigmentation, alopecia, and onychatrophia and by intestinal polyps and malabsorption, This disorder has a poor prognosis and few affected children live more than 2 years. 253 The differential diagnosis includes infantile necrotizing enterocolitis, which also may be associated with a vasculitis. 2<;4
Stevens-Johnson Syndrome The Stevens-Johnson syndrome is a severe, systemic, widespread form of mucocutaneous erythema multiforme and merges into toxic epidermal necro~ysis (depending on the surface area involved). Numerous erosive, vesiculobullous, hemorrhagic, and papular lesions develop acutely on the mucosa and skin of the face, hands, tnmk, and feet (Pig. 23-12). Anal, geni-
C HAP T E R
• fItwe Z3-1Z
Vesicular erythematous lesions of erythema multiforme occurred in ayoung boy with Stevens-Johnson syndrome.The characteristic bullous lesions also developed around body orifices.
tal, and ocular orifices are often affected, and scarring may result. Onset is usually abrupt and is associated with fever, profound constitutional symptoms, and the appearance of periarticular swelling and pain or frank arthritis. TI1e respiratory and gastrointestinal tracts can be involved in severe disease. Histopathologic studies demonstrate a perivasculitis that results in stomatitis, conjunctivitis, or corneal ulcerations, with no evidence of a necrotizing vasculitis. The cause of the syndrome is unknown, but a preceding infectious illness, particularly with Mycoplasma pneumoniae, is frequently documented. 255 •2'6 Antibiotics, especially trimethoprim-sulfamethoxazole257 and cefac1or, NSAIDs, and anticonvulsant medications have also been in1plicated. 258 Expert supportive care is usually the sale treatment necessary for this self-limited disease. Extensive mucosal and cutaneous disease is best managed in a burn unit. In severe cases, glucocorticoids may be considered necessary but increase the risk of infection. 259
Erythema Elevatom Dlutlnum Erythema e1evatum diutinum is a rare form of chronic, localized, cutaneous leukocytoclastic vasculitis, characterized by edematous papules and plaques (e.g., yellow, red, purple) occurring mainly over extensor surfaces. A similar lesion, granuloma faciale, is localized to the face. Fibrinoid necrosis of the upper and middle dermal vessels develops in both conditions. 2(" Systemic involvement is unusuaJ.261 Treatment with dapsone or intralesional steroids may be beneficial. 262
REFERENCES 1. Lie Jf: l1Iustrated histopathologic classification criteria for selected vasculitis syndromes. American College of Rheumatology Subcommittee on Cla$sification of Vasculitis. Arthritis Rheum 33: 1074-1087. 1990. 2. Yakindag A, Sundel R: Vasculitis in childhood. Curr Opin Rheumatol 13: 422-427. 2001. 3. Ozen S: The spectrum of vasculitis in children, Best Pract Res Clin Rh"umatol 16: 411-425, 2002. 4. Kobayashi O. Wada H. Okawa K, et al: Schiinlein-Henoch's syndrome in children, Contrib Nephrol 4: 48--71, 1975, 5. Tizard EJ: Henoch-Sch6nlein purpura, Arch Dis Child 80: 380-383. 1999, 6. Mur-Jli NS, George R, John GT, et al: Problems of classification of Henoch Schi;nlein purpura: an Inclian perspective. Clin Exp Derrnatol 27: 260--263. 2002. 7. Ballinger S: Henoch-Sch6nlein purpura. Curr Opin Rheumatol 15: 591-594. 2003. 8. Mnlsek S. Kruger M, Greiner P, et al: Henoch-Sch6nlein purpura. Lancet 363: 11 16. 2004.
23
LEU K 0 C Y Toe LAS TIC
VAS C U L IT I S
507
9. Willan R: On Cutaneous Diseases. London, J Johnson. 1808. 10. Heberden W: Commentaries on the History and Cure of Diseases. London, 1896. 11. Sch6nlein JL: Allegemeine und Specielle Pathologie und Therapie. 3rd ed. Herisau, Germany, Literatur-Comptoir, 1837. 12. Henoch EH: About a peculiar form of purpura. by Eduard H. H. Henoch, Berlin. Reprinted in AmJ Dis Child 128: 78-79,1974. 13. Gairdner 0: The Sch6nlein-Henoch syndrome (anaphylactoid purpura). Q J Med 17: 95. 1948. 14. Rieu P, Noel LH: Henoch-Sch6nlein nephritis in children and adults. Morphological features and clinicopathological correlations. Ann Med Interne (Paris) 150: 151-159, 1999. 15. Saulsbury Ff: Henoch-Sch6nlein purpura in children. Report of 100 patient, and review of the literature. Medicine (Baltimore) 78: 395-409, 1999. 16. RoslOker G: Sch6nlein-Henoch PUrpUr-d in children and adults: diagnosis, pathophysiology and management. BioDrugs 15: 99-138, 2001. 17. Saulsbury IT: Epidemiology of Henoch-Sch6nlein purpum. Cleve ClinJ Med 69 (Suppl 2): SU87-5U89. 2002. 18, Calvino MC, Uorca J, Garcia-Porrua C, et al: Henoch-Sch6nlein purpura in children from northwestern Spain: a 20-year epidemiologic and clinical study. Medicine (Baltimore) 80: 279-290, 2001. 19. Al Sheyyab M, EI Shanti H, Ajlouni S, et al: The clinical spectrum of HenochSch6nlein purpura in infants and young children. Eur J Pediatr 154: 969-972, 1995. 20. Stewart M, Savage JM, Bell B, et al: Long term renal prognosis of HenochSch6nlein purpura in an unselected childhood population. Eur.r Pediatr 147: 113-115. 1988. 21. Farley TA, Gillespie S, Rasoulpour M, et al: Epidemiology of a cluster of Henoch-Sch6nlein purpura. Am J Dis Child 143: 798-803, 1989. 22. Gardner-Medwin JM, Dolezalova P, Cummins C, et al: Incidence of HenochSch6nlein purpura. Kawasaki disease, and rare vasculitides in children of different ethnic origins. Lancet 360: 1197-1202, 2002. 23. Atkinson SR. Barker OJ: Seasonal distribution of Henoch-Sch6nlein purpura. Br.r Prev Soc Med 30: 22-25, 1976. 24. Levy M. Brayer M, Arsan A. et al: Anaphylactoid purpura nephritis in childhood: natural history and immunopathology. Adv Nephrol Necker Hosp 6: 183-228, 1976. 25. Allen OM, Diamond LK, Howell DA: Anaphylactoid purpur-J in children (Sch6nlein-Henoch syndrome): review with a follow-up of the renal complications. Am J Dis Child 99: 833-854, 1960. 26. Al Sheyyab M, Batieha A, EI Shanti H, et al: Henoch-Sch6nlein purpur-J and streptococcal infection: a prospective case-control study. Ann Trop Paediatr 19: 253-255, 1999. 27. Masuda M, Nakanishi K, Yoshizawa N, et al: Group A streptococcal antigen in the glomeruli of children with Henoch-Sch6nlein nephritis. Am J Kidney Dis 41: 366--370, 2003. 28. Matsukura H, Oht,uki A, Fuchizawa T, et al: Acute poststreptococcal glomerulonephritis mimicking Henoch-Sch6nlein purpum. Clin Nephrol 59: 64-65. 2003. 29. Ayoub EM. Hoyer .J: Anaphylactoid purpura: streptococcal antihody titers and beta1c-globulin levels. J Pediatr 75: 193-201, 1969. 30, Ansell BM: Henoch-Sch6nlein purpura with particular reference to the prognosis of the renal lesion. Br J Dermatol 82: 211-215, 1970. 31. Courtney PA, Patterson RN, Lee RJ: Henoch-Sch6nlein purpura follOWing meningitis C vaccination. Rheumatology (Oxf) 40: 345-346, 2001. 32. Lambert EM, Liebling A, Glusac E, et al: Henoch-Sch6nlein PUrpUr-d following a meningococcal vaccine. Pediatrics 112: e491, 2003. 33. Minohara Y: Studies on the relationship between anaphylactoid purpura and human parvovirus B19. Kansenshogaku Zasshi 69: 928-933. 1995. 34. Watanabe T, ada Y: Henoch-Sch6nlein purpum nephritis associated with human parvovirus B19 infection. Pediatr Int 42: 94-96, 2000. 35. Eisenstein EM: Lack of evidence for herpeSVirus. retrovirus, or parvovirus infection in Henoch-Sch6nlein purpura. Clin Exp Rheumatol 20: 734. 2002. 36. Heegaard ED. Taaning EB: Parvovirus B19 and parvovirus V9 are not associated with Henoch-Sch6nlein purpura in children. Pediatr Infect Dis J 21: 31-34, 2002. 37. Meadow SR, Glasgow EF, White RH, et al: Sch6nlein-Henoch nephritis. Q.r Med 41: 241-258, 1972. 38. Pedersen FK, Petersen EA: Varicella followed by glomerulonephritis. Treatment with corticosteroids and azathioprine resulting in recurrence of varicella. Acta Paediatr Scand 64: 8116-890, 1975. 39. Kalyoncu M, Odemis E, Yaris N, et al: Association of Henoch-Sch6nlein purpura with varicella zoster. Indian Pediatr 40: 274-275, 2003. 40. Maggiore G, Martini A. Grifeo S, et al: Hepatitis B virus infection and Schonlein-Henoch purpura. Am.l Dis Child 138: 681--{j82, 1984, 41. Islek I, Kalayci AG, Gok F, et al: Henoch-Sch6nlein purpur-J associated with hepatitis A infection. Pediatr Int 45: 114-116. 2003. 42. Liew SW, Kessel I: Mycoplasmal pneumonia preceding Henoch-Sch6nlein purpura [letter!. Arch Dis Child 49: 912-913, 1974. 43. Kaneko K, Fujinaga S. Ohtomo Y, et 'II: Mycoplasma pneumoniae-associated Henoch-Sch6nlein purpura nephritis. Pediatr Nephrol 13: 1000--1001, 1999. 44. Ayoub EM. McBride J, Schmiederer M. et al: Role of Bartonella henselae in the etiology of Henoch-Sch6nlein purpur-J. Pediatr Infect Dis J 21: 28-31, 2002. 45. Ackroyd JF: Allergic purpura, including purpura due to foods, drugs and infections. Am J Med 14: 605-632, 1953.
508
C HAP T E R
23
LEU K 0 C Y Toe LAS TIC
V AS C U LI TIS
46, Giangiacomo], Tsai CC: Dermal and glomerular deposition of IgA in anaphylactoid purpura, Am] Dis Child 131: 981-983, 1977, 47, Yang YH, Huang MT, Lin SC, et al: Increased transforming growth factor-beta (TGF-beta)-secreting T cells and 19A anti-cardiolipin antibody levels during acute stage of childhood Henoch-Schonlein purpura, Clin Exp Immunol 122: 285-290, 2000, 48, Brendel-Muller K, Hahn A, Schneppenheim R, et al: Laboratory signs of activated coagulation are common in Henoch-Schonlein purpum, Pediatr Nephrol 16: 1084-1088, 2001. 49, Demircin G, Oner A, Unver Y, et al: Erythrocyte superoxide dismutase activity and plasma malondialdehyde levels in children with Henoch-Schonlein purpura, Acta Paediatr 87: 848-852, 1998. 50. YOnlk A, Cizmecioglu F, Yukselgungor H, et al: Methotrexate-induced systemic vasculltis, Med Pediatr Oneal 38: 139-140, 2002. 51. Livermore PA, Murmy K]: Anti-tumour necrosis factor therapy associated with cutaneous vasculitis. Rheumatology (Oxf) 41: 1450-1452, 2002. 52, Levy-Khademi F, Korman SH, Amitai Y: Henoch-Schonlein purpum: simultaneous occurrence in two sibllngs. Pediatr Dermatol 17: 139-140, 2000, 53. Grech V, Vella C: Henoch-Schonlein purpura with nephritis in two siblings following infectious mononucleosis. Ann Trop Paediatr 22: 297-298, 2002, 54. Amoll MM, Thomson W, Hajeer AH, et al: HLA-B35 association with nephritis in Henoch-SchOnlein purpum. .I Rheumatol 29: 948-949, 2002, 55, Amoll MM, Thomson W, Hajeer AH, et al: HLA-DRBrOl association with Henoch-Schonlein purpum in patients from nonhwest Spain. ] Rheumatol 28: 1266-1270, 2001. 56, Amoll MM, Maney DL, Calvino MC, et al: Polymorphism at codon 469 of the intercellular adhesion molecule-l locus is associated with protection against severe gastrointestinal complications in Henoch-Schonlein purpura. .1 Rheumatol 28: 1014-1018, 2001. 57, Gershoni-Baruch R, Broza Y, Brik R: Prevalence and significance of mutations in the famillal Mediterranean fever gene in Henoch-Schonlein purpura. ] Pediatr 143: 658-661, 200358. Dudley .1, Aflfi E, Gardner A, et al: Polymorphism of the ACE gene in Henoch-Schonlein purpura nephritis. Pediatr Nephrol 14: 218-220, 2000. 59. Emery H, Larter W, Schaller ]G: Henoch-Schonlein vasculitis. Arthritis Rheum 20: 385-388, 1977. 60. Rosenblum ND, Winter HS: Steroid effects on the course of abdominal pain in children with Henoch-Schonlein purpura. Pediatrics 79: 1018-1021, 1987. 61. Bagga A, Kabra SK, Srivastava RN, et al: Henoch-Schonlein syndrome in northern Indian children. Indian Pediatr 28: 1153--1157, 1991. 62. Coppo R, Amore A, Gianogllo B: Cllnical features of Henoch-Schonlein purpura, ltallan Group of Renal Immunopathology. Ann Med Interne (Paris) 150: 143--150, 1999. 63, Saulsbury FT: Henoch-SchOnlein purpura. Pediatr Dermatol I: 195-201, 1984. 64. Mills ]A, Michel BA, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of Henoch-Schonlein purpura. Arthritis Rheum 33: 1114-1121, 1990. 65. Choong CK, Beasley SW: Intra-abdominal manifestations of HenochSchOnlein purpum, .I Paediatr Child Health 34: 405-409, 1998, 66. Nathan K, Gunasekaran TS, Berman.JH: Recurrent gastrointestinal HenochSchOnlein purpura, .I Clin Gastroenterol 29: 86-89, 1999. 67, Chen SY, Kong MS: Gastrointestinal manifestations and compllcations of Henoch-Schonlein purpura, Chang Gung MedJ 27: 175-181, 2004. 68. Garner ]A: Acute pancreatitis as a complication of anaphylactoid menochSchonlein) purpura. Arch Dis Child 52: 971-972, 1977. 69. Levy-Weil FE, Sigal M, Renard P, et al: Acute pancreatitis in rheumatoid purpura. Apropos of 2 cases. Rev Med Interne 18: 54-58, 1997. 70, Chao HC, Kong MS, Lin 5.1: Hepatobiliary involvement of Henoch-Schonlein purpura in children. Acta Paediatr Taiwan 41: 63--68, 2000, 71. Gow KW, Murphy]], III, Blair GK, et al: Multiple entero-entero fistulae: an unusual compllcation of Henoch-Schonlein purpura . .I Pediatr Surg 31: 80~11, 1996. 72. Feldt RH, Stickler GB: The gastrointestinal manifestations of anaphylactoid purpura in children. Mayo Clln Proc 37: 465-473, 1962. 73. Kaku Y, Nohara K, Honda S: Renal involvement in Henoch-Schoniein purpura: a multivariate analysis of prognostic factors. Kidney Int 53: 1755-1759, 1998, 74, Lewis IC, Philpott MG: Neurological compllcations in the Schonlein-Henoch syndrome. Arch Dis Child 31: 369-371, 1956. 75. Bakkaloglu SA, Ekim M, Turner N, et al: Cerebral vasculltis in HenochSchOnlein purpura. Nephrol Dial Transplant IS: 246-248, 2000. 76. Imai T, Okada H, Nanba M, et al: Henoch-Schonlein purpura with intracerebral hemOlThage. Brain Dev 24: 115-117, 2002. 77. Chiaretti A, Caresta E, Piastra M, et al: Cerebral hemorrhage in HenochSchiinlein syndrome. Childs Nerv Syst 18: 365-367, 2002. 78, Paollni S, Ciappena P, Pianella MC, et al: Henoch-Schonlein syndrome and cerebellar hemorrhage: report of an adolescent case and Ilterature review, Surg Neurol 60: 339-342, 2003. 79, Goraya .IS, ]ayashree M, Ghosh D, et al: Guillain-Barre syndrome in a child with Henoch-Schonlein Purpura. Scand] Rheumatol 27: 310-312, 1998. 80. Kaplan ]M, Quintana P, Samson J: Facial nerve palsy with anaphylactoid purpura. Am] Dis Child 119: 452--453, 1970. 81. Bulun A, Topaloglu R, Duzova A, et al: Ataxia and peripheral neuropathy: rare manifestations in Henoch-Schonlein purpura. Pediatr Nephrol 16: 1139-1141, 2001.
82. Wu TT, Sheu SJ, Chou LC, et al: Henoch-Schonlein purpura with bilateral centml retinal anery occlusion. BrJ Ophthalmol 86: 351-352, 2002, 83. Olson ]C, Kelly K.1, Pan CG, et al: Pulmonary disease with hemorrhage in Henoch-Schonlein purpura. Pediatrics 89: 1177-1181, 1992. 84. Paller AS, Kelly K, Sethi R: Pulmonary hemorrhage: an often fatal eampllcation of Henoch-Schoenlein purpura. Pediatr Dermatol 14: 299-302, 1997, 85, Vats KR, Vats A, Kim Y, et al: Henoch-Schonlein purpura and pulmonary hemorrhage: a report and Ilterature review. Pediatr Nephrol13: 530-534, 1999. 86. Besbas N, Duzova A, Topaloglu R, et al: Pulmonary haemorrhage in a 6-year-old boy with Henoch-Schonlein purpura. Clln Rheumatol 20: 293--296, 2001. 87. Chaussain M, de Boissieu D, Kallfa G, et al: Impairment of lung diffusion capacity in Schonlein-Henoch purpura . .I Pediatr 121: 12-16, 1992, 88. Imai T, Matsumoto S: Anaphylactoid purpura with cardiac involvement. Arch Dis Child 45: 727-729, 1970. 89, Cimaz R, Boccazzi A, Milone V, et al: Pericarditis as a presenting feature of Henoch-Schonlein purpura. Clin Exp Rheumatol 18: 785, 2000. 90, Sahn DJ, Schwartz AD: Schonlein-Henoch syndrome: observations on some atypical clinical presentations. Pediatrics 49: 614-616, 1972. 91. Naiman ]L, Harcke T, Sebastianelli], et al: Scrotal imaging in the HenochSchonlein syndrome. .I Pediatr 92: 1021-1022, 1978. 92. O'Regan 5, Robitaille P: Orchitis mimicking testicular torsion in HenochSchonlein's purpura . .I Urol 126: 834-835, 1981. 93. Clark WR, Kramer SA: Henoch-Schonlein purpura and the acute scrotum, ] Pediatr Surg 21: 991-992, 1986. 94. Mintzer CO, Nussinovitch M, Danziger Y, et al: Scrotal involvement in HenochSchonlein purpura in children. Scand] Urol Nephrol 32: 138-139. 1998, 95. Choong CS, Liew KL, Liu PN, et al: Acute scrotum in Henoch-Schonlein purpura. Zhonghua Yi Xue Za Zhi (Taipei) 63: 577-580, 2000, 96. loannides AS, Turttock R: An audit of the management of the acute scrotum in children with Henoch-Schonlein Purpura, ] R Coli Surg Edinb 46: 98-99, 2001. 97, Sinniah R, Feng PH, Chen BT: Henoch-Schonlein syndrome: a clinical and morphological study of renal biopsies. Clin Nephrol 9: 219-228, 1978. 98. Cribier B, Couilliet 0, Meyer P, et al: The severity of histopathological changes of leukocytoclastic vasculitis is not predictive of extracutaneous involvement. AmJ Dermatopathol 21: 532-536, 1999. 99, Yoshikawa N, Hjima K, Ito H: IgA nephropathy in children, Nephron 8.:\: 1-12, 1999. 100. Vogler C, Eliason SC, Wood EG: Glomerular membranopathy in children with 19A nephropathy and Henoch Schonlein purpura. Pediatr Dev Pathol 2: 227-235, 1999. 101. Conley ME, Cooper MD, Michael AF: Selective deposition of immunoglobulin Al in immunoglobulin A nephropathy, anaphylactOid purpura nephritis, and systemic lupus erythematosus. ] Clin Invest 66: 1432-1436, 1980. 102. Szeto CC, Choi PC, To KF, et al: Grading of acute and chronic renal lesions in Henoch-Schonlein purpura, Mod Pathol 14: 635-640, 2001. 103, Nakamoto Y, Asano Y, Dohi K, et al: Primary IgA glomerulonephritis and Schonlein-Henoch purpura nephritis: Clinicopathological and immunohistological characteristics. QJ Med 47: 495-516, 1978, 104, Hakim A: Concurrent Henoch-Schonlein purpura and papular-purpuric gJoves-and-socks syndrome. Scand] Rheumatol 29: 131-132, 2000. 105. Dillon MJ, Ansell BM: Vasculitis in children and adolescents. Rheum Dis Clin North Am 21: 1115-1136, 1995. 106. Gibson LE, Su WP: Cutaneous vasculitis. Rheum Dis Clin North Am 21: 1097-1113, 1995, 107. Blanco R, Martinez-Taboada VM, RodrigueZ-Valverde V, et al: Cutaneous vasculitis in children and adults. Associated diseases and etiologic factors in 30.:\ patients. Medicine (Baltimore) 77: 403--418, 1998. 108. Dubin BA, Bronson OM, Eng AM: Acute hemorrhagic edema of childhood: an unusual variant of leukocytoclastic vasculitis, J Am Acad Dermatol 23: 347-350, 1990. 109. Saraclar Y, Tinaztepe K, Adalioglu G, et al: Acute hemorrhagic edema of infancy (AHEJ)-a variant of Henoch-Schiinlein purpura or a distinct clinical entity? ] Allergy Clin Immunol 86: 473--483, 1990, 1l0. Legrain V, Lejean 5, Taieb A, et al: Infantile acute hemorrhagic edema of the skin: study of ten cases. ] Am Acad Dermatol 24: 17-22, 1991. 111. Krause I, Lazarov A, Rachmel A, et al: Acute haemorrhagic oedema of infancy, a benign variant of leucocytoclastic vasculitis. Acta Paediatr 85: 114-117, 1996. 112, Gonggryp LA, Todd G: Acute hemorrhagic edema of childhood (AHEl. Pediatr Dermatol 15: 91-96, 1998. 113. Cunningham BB, Eramo L, Caro W: Acute hemorrhagic edema of childhood present at birth, Pediatr Dermatol 16: 68, 1999. 114. Millard T, Harris A, MacDonald D: Acute infantile hemorrhagic oedema, JAm Acad Dermatol 41: 837-839, 1999. 115. Braun-Falco M, Abeck 0: Acute infantile haemorrhagic oedema. Lancet 360: 210, 2002. 116, Kuroda K, Yabunami H, HLsanaga Y: Acute haemorrhagic oedema of infancy associated with cytomegalovirus infection, BrJ Dermatol147: 1254-1257,2002, 117. Caksen H, Odabas D, Kosem M, et al: Report of eight infants with acute infantile hemorrhagic edema and review of the literature. J Dermatol 29: 290-295, 2002, 118. Garty BZ, Ofer 1, Finkelstein Y: Acute hemorrhagic edema of infancy, Isr Mecl Assoc.1 4: 228-229, 2002,
C HAP T E R 119. Shah D, Goraya ]S, Poddar B, et al: Acute infantile hemorrhagic edema and Henoch-Schonlein purpura overlap in a child. Pediatr Dermatol 19: 92-93, 2002. 120. Macea ]M, Santi CG, Sotto MN, et al: Multiple erythematous plaques on a child. Acute hemorrhagic edema (AHE) of infancy. Arch Dermatol 139: 531-536, 2003. 121. Poyrazoglu HM, Per H, Gunduz Z, et al: Acute hemorrhagic edema of infancy. Pediatr Int 45: 697-700, 2003, 122. Goraya ]S, Kaur S: Acute infantile hemorrhagic edema and HenochSchonlein purpura: is 19A the missing link? ] Am Acad Dermatol 47: 801-802, 2002. 123. Coppo R, Cirina P, Amore A, et al: Properties of circulating 19A molecules in Henoch-Schonlein purpura nephritis with focus on neutrophil cytoplasmic antigen 19A binding (IgA-ANCA): new insight into a debated issue. Italian Group of Renal Immunopathology Collaborative Study on HenochSchonlein purpura in adults and in children. Nephrol Dial Transplant 12: 2269-2276, 1997. 124. Lin S], Huang]L: Henoch-Schonlein purpura in Chinese children and adults. Asian Pac] Anergy Immunol 16: 21-25, 1998. 125. !Ian Y, Naparstek Y: Henoch Schonlein purpura in children and adults: is it one entity? Semin Arthritis Rheum 32: 139-140, 2002. 126. Garcia-Porrua C, Calvino MC, Uorca ], et al: Henoch-Schonlein purpura in children and adults: clinical differences in a defined population. Semin Arthritis Rheum 32: 149-156, 2002. 127. Pillebout E, Thervet E, Hill G, et al: Henoch-Schonlein Purpura in adults: outcome and prognostic factors. ] Am Soc Nephrol 13: 1271-1278, 2002. 128. Blanco R, Martinez-Taboada VM, Rodriguez-Valverde V, et al: HenochSchonlein purpura in adulthood and childhood: two different expressions of the same syndrome, Arthritis Rheum 40: 859-864, 1997. 129. Muller D, Greve D, Eggert P: Early tubular proteinuria and the development of nephritis in Henoch-Schonlein purpura. Pediatr Nephrol 15: 85-89, 2000. 130. Garcia-Fuentes M, Martin A, Chantler C, et al: Serum complement components in Henoch-Schonlein purpura. Arch Dis Child 53: 417-419, 1978. 131. Spitzer RE, Urmson JR, Farnett ML, et al: Alteration of the complement system in children with Henoch Schonlein purpura. Clin lmmunol Immunopathol 11: 52-59, 1978. 132, De Mattia D, Penza R, Giordano P, et al: von Willebrand factor and factor XlII in children with Henoch-SchOnlein purpura. Pediatr Nephrnl9: 603-605, 1995. 133. Casonato A, Pontara E, Bertomoro A, et al: Abnormally large von Willebrand faL10r multimers in Henoch-Schonlein purpura. Am] Hematol 51: 7-11, 1996. 134. Levinsky RJ, Barratt TM: 19A immune complexes in Henoch-Schonlein purpura. Lancet 2: 1100-1103, 1979. 135. Kauffmann RH, Herrmann WA, Meyer CJ, et al: Circulating 19A-immune complexes in Henoch-Schonlein purpura. A longitudinal study of their relationship to disease activity and vascular deposition of 19A. Am J Med 69: 859-866, 1980. 136. Hall RP, Lawley TJ, Heck JA, et al: 19A-containing circulating immune complexes in dermatitis herpetiformis, Henoch-Schonlein purpura, systemic lupus erythematosus and other diseases. Clin Exp lmmunol 40: 431-437, 19HO.
137. Garcia-Fuentes M, Chander C, Williams DG: Cryoglobulinaemia in HenochSchonlein purpura. Br Med J 2: 163-165, 1977. 138. Trygstad CW, Stiehm ER: Elevated serum 19A globulin in anaphylactoid purpura. Pediatrics 47: 1023-1028, 1971. 139. Casanueva B, Rodriguez-Valverde V, Luceno A: Circulating 19A prodUcing cells in the differential diagnosis of Henoch-Schonlein purpura. J Rheumatol 15: 1229-1233, 1988. 140. Ha TS, Cha SH: Cerebral vasculitis in Henoch-Schonlein purpura: a case report with sequential magnetic resonance imaging. Pediatr Nephrol 10: 634-636, 1996. 141. Chen CL, Chiou YH, Wu CY, et al: Cerebral vasculitis in Henoch-Schonlein purpura: a case report with sequential magnetic resonance imaging changes and treated with plasmapheresis alone. Pediatr Nephrol 15: 276-278, 2000. 142. Eun SH, Kim SJ, Cho DS, et al: Cerebral vasculitis in Henoch-Schonlein purpura: MRJ and MRA findings, treated with plasmapheresis alone. Pediatr lnt 45: 484-487, 2003. 143. Navarro 0, Dugougeat F, Kornecki A, et al: The impact of imaging in the management of intussusception owing to pathologic lead points in children. A review of 43 cases. Pediatr Radiol 30: 594-603, 2000. 144. Dillon MJ: Henoch-Schonlein purpura (treatment and outcome). Cleve Clin J Med 69 (Suppl 2): SIIl21~IIl23, 2002. 145. Szer IS: Gastrointestinal and renal involvement in vasculitis: management strategies in Henoch-Schonlein purpura. Cleve Clin J Med 66: 312-317, 1999. 146. Wang L, Huang FC, Ko SF, et al: Successful treatment of mesenteric vasculitis caused by Henoch-Schonlein purpura with methylprednisolone pulse therapy. Clin Rheumatol 22: 140-142, 2003. 147. Leung SP: Use of intravenous hydrocortisone in Henoch-Schonlein purpura. J Paediatr Child Health 37: 309-310, 2001. 148. Counahan R, Winterborn MH, White RH, et al: Prognosis of HenochSchonlein nephritis in children. Br Med J 2: 11-14, 1977. 149. Mollica F, Li VS, Garozzo R, et al: Effectiveness of early prednisone treatment in preventing the development of nephropathy in anaphylactoid purpura. Eur J Pediatr 151: 140-144, 1992.
23
LEU K 0 C Y Toe LAS TI C VA S C U LI TI S
509
ISO. Saulsbury FT: Corticosteroid therapy does not prevent nephritis in HenochSchonlein purpura, Pediatr Nephrol 7: 69-71, 1993151. Niaudet P, Habib R: Methylprednisolone pulse therapy in the treatment of severe forms of Schonlein-Henoch purpura nephritis. Pediatr Nephrol 12: 238-243, 1998. 152. Reinehr T, Burk G, Andler W: Does steroid treatment of abdominal pain prevent renal involvement in Henoch-Schonlein purpura? J Pediatr Gastroenterol Nutr 31: 323-324, 2000. 153. Kawasaki Y, Suzuki J, Nozawa R, et al: Efficacy of methylprednisolone and urokinase pulse therapy for severe Henoch-Schonlein nephritis. Pediatrics 111: 785-789, 2003. 154. Iijima K, lto-Kariya S, Nakamura H, et aI: Multiple combined therapy for severe Henoch-SchOnlein nephritis in children. Pediatr Nephrol 12: 244-248, 1998. 155. Foster B], Bernard C, Drummond KN, et al: Effective therapy for severe Henoch-Schonlein purpura nephritis with prednisone and azathioprine: a clinical and histopathologic study. ] Pediatr 136: 370-375, 2000. 156. Flynn JT, Smoyer WE, Bunchman TE, et al: Treatment of Henoch-Schonlein purpura glomerulonephritis in children with high-dose corticosteroids plus oral cyclophosphamide. Am J Nephrol 21: 128-133, 2001. 157. Singh S, Devidayal, Kumar L, et al: Severe Henoch-Schonlein nephritis: resolution with azathioprine and steroids. Rheumatol Int 22: 133-137, 2002. 158. Huang DC, Yang YH, Lin YT, et al: Cyclosporin A therapy for steroiddependent Henoch-SchOnlein purpura. 1 Microbiol lmmunol Inlect 36: 61--64, 2003. 159. Someya T, Kaneko K, Fujinaga S, et al: Cyclosporine A for heavy proteinuria in a child with Henoch-SchOnlein purpura nephritis. Pediatr Int 46: 111-113, 2004. 160. Rostoker G, Desvaux-Belghiti D, Pilatte Y, et al: High-dose immunoglobulin therapy for severe 19A nephropathy and Henoch-Schonlein purpura, Ann Intern Med 120: 476-484, 1994. 161. Rostoker G, Desvaux-Belghiti D, Pilatte Y, et al: Immunomodulation with low-dose immunoglobulins for moderate 19A nephropathy and HenochSchonlein purpura. Preliminary results of a prospective uncontrolled trial. Nephron 69: 327-334, 1995. 162. Scharer K, Krmar R, Querfeld U, et al: Clinical outcome of Schonlein-Henoch purpura nephritis in children. Pediatr Nephrol 13: 816-823, 1999, 163. Hasegawa A, Kawamura T, Ito H, et aI: Fate of renal grafts with recurrent Henoch-Schonlein purpura nephritis in children. Transplant Proc 21: 2130-2133, 1989. 164. Cameron ]S: Recurrent primary disease and de novo nephritiS following renal transplantation. Pediatr Nephrol 5: 412-421, 1991. 165. Meulders Q, Pirson Y, Cosyns JP. et al: Course of Henoch-Schonlein nephritis after renal transplantation. Report on ten patients and review of the literature, Transplantation 58: 1179-1186, 1994. 166. Meadow SR: The prognosis of Henoch Schoenlein nephritis. Clin Nephrol 9: 87-90, 1978. 167. Coakley lC, Chambers U: Should we follow up children with HenochSchonlein syndrome? Arch Dis Child 54: 903-904, 1979. 168. Pabunruang W, Treepongkaruna S, Tangnararatchakit K, et al: HenochSchonlein purpura: clinical manifestations and long-term outcomes in Thai children.] Med Assoc Thai 85 (Suppl 4): S121~1218, 2002. 169. Thervet E, Pillebout E, Guillevin L: Outcome after childhood HenochSchonlein purpura. Lancet 361: 81, 2003. 170. Beevers DG: Outcome after childhood Henoch-Schonlein purpura. Lancet 361: 81, 2003, 171. Ronkainen J, Nuutinen M, Koskimies 0: The adult kidney 24 years after childhood Henoch-Schonlein purpura: a retrospective cohort study. Lancet 360: 666-670, 2002. 172. Kawasaki Y, Suzuki J. Sakai N, et al: Clinical and pathological features of children with Henoch-Schoenlein purpura nephritis: risk factors associated with poor prognosis. Clin Nephrol 60: 153-160, 2003. 173. Goldstein AR, White RH, Akuse R, et aI: Long-term follow-up of childhood Henoch-Schonlein nephritis. Lancet 339: 280-282, 1992. 174. Ronkainen J, A1a-Houhaia M, Huttunen NP, et al: Outcome of HenochSchoenlein nephritis with nephrotic-range proteinuria. Clin Nephrol 60: 80-84, 2003. 175. Bunchman TE, Mauer SM, Sibley RK, et al: Anaphylactoid purpura: characteristics of 16 patients who progressed to renal failure. Pediatr Nephrol 2: 393-397, 1988. 176. Algoet C, Proesmans W: Renal biopsy 2-9 years after Henoch-Schonlein purpura. Pediatr Nephrol 18: 471-473, 2003. 177. Bielory L, Gascon P, Lawley TJ, et al: Human serum sickness: a prospective analysis of 35 patients treated with equine anti-thymocyte globulin for bone marrow failure. Medicine (Baltimore) 67: 40-57, 1988. 178. Jennette ]C, Falk Rl, Andrassy K, et al: Nomenclature of systemic vasculitides. Proposal of an international consensus conference. Arthritis Rheum 37: 187-192, 1994. 179. Calabrese LH, Michel BA, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of hypersensitivity vasculitis. Arthritis Rheum 33: 1108-1113, 1990. 180. Michel BA, Hunder GG, Bloch DA, et al: Hypersensitivity vasculitis and Henoch-Schonlein purpura: a comparison between the 2 disorders. J Rheumatol 19: 721-728, 1992. 181. Tamburro ]E, Esterly NB: Hypersensitivity syndromes. Adolesc Med 12: vii, 323-341, 2001.
510
C HAP T E R
23
LEU K0 C Y Toe LAS TI C
V AS C U LI TI S
182. Kunnamo 1, Kallio P, Pelkonen P, et al: Serum-sickness-Jike disease is a common cause of acute arthritis in children. Acta Paediatr Scand 75: 964-969, 1986, 183. Murray DL, Singer DA, Singer AB, et al: Cefaclor-a cluster of adverse reactions. N Engl J Med 303: 1003, 1980. 184. Soter NA: Chronic urticaria as a manifestation of necrotizing venulitis. N Engl .J Med 296: 1440--1442, 1977: 185. Mehregan DR, Hall MJ, Gibson LE: Urticarial vasculitis: a histopathologic and clinical review of 72 cases. J Am Acad Dermatol 26: 441--448, 1992, 186. Black AK: Urticarial vasculitis, Clin Dermatol 17: 565-569, 1999. 187. Martini A, Ravelli A, Albani S, et aI: Hypocomplemememic urticarial vasculitis syndrome with severe systemic manifestations. J Pediatr 124: 742-744, 1994, 188. Provost IT, Zone .JJ, Synkowski 0, et al: Unusual cutaneous manifestations of systemic lupus erythematosus: I. Urticaria-like lesions. Correlation with clinical and serological abnonnalities. J Invest Dermatol 75: 495--499, 1980. 189. Davis MD, Daoud MS, Kirby B, et al: Clinicopathologic correlation of hypocomplementemic and normocomplementemic urticarial vasculitis. 1 Am Acad Dermatol 38: 899-905, 1998. 190. DeAmicis T, Mofid MZ, Cohen B, et al: Hypocomplemememic urticarial vasculitis: report of a 12-year-old girl with systemic lupus erythematosus. 1 Am Acad Dermatol 47: S273-8274, 2002. 191. O'Donnell B, Black AK: Urticarial vasculitis. Int Angiol 14: 166-174, 1995. 192. Wisnieski 11, Jones SM: IgG autoantibody to the collagen-like region of Clq in hypocomplementemic urticarial vasculitis syndrome, systemic lupus erythematosus, and 6 other musculoskeletal or rheumatic diseases. J Rheumatol 19: 884-888, 1992, 193. Weinberger A, Berliner S, Pinkhas J: Articular manifestations of essemial cryoglobulinemia. Semin Arthritis Rheum 10: 224--229, 1981. 194. Levo Y, Gorevic PO, Kassab HJ, et al: Association between hepatitis B virus and essential mixed cryoglobulinemia. N Engl J Med 296: 1501-1504, 1977, 195. Gamble CN, Ruggles SW: The immunopathogenesis of glomerulonephritis associated with mixed cryoglobulinemia. N Engl J Med 299: 81-84, 1978. 196. Cattaneo R, Fenini MG, Facchetti F: The cryoglobulinemic vasculitis. Ric Clin Lab 16: 327-333, 1986, 197. Revenga AF, Diaz DR, Iglesias DL, et al: Cryoglobulinemic vasculitis associated with hepatitis C virus infection. A report of eight cases. Acta Derm Venereol 75: 234--236, 1995, 198. Tavoni A, Mosca M, Ferri C, et al: Guidelines for the management of essential mixed cryoglobulinemia, Clin Exp Rheumatol 13 (Suppl 13): S191-S195, 1995, 199. Jennette JC, Falk R.J: Small-vessel vasculitis. N Engl1 Med 337: 1512-1523, 1997. 200. Nash MC, Dillon MJ: Antineutrophil cytoplasm antibodies and vasculitis. Arch Dis Child 77: 261-264, 1997. 201, Wong SN, Shah V, Dillon M.J: Antineutrophil cytoplasmic antibodies in Wegener's granulomatosis. Arch Dis Child 79: 246-250, 1998. 202, Rovel-Guitera P, Diemert MC, Charuel JL, et al: IgA antineutrophil cytoplasmic antibodies in cutaneous vasculitis. Br J Dermatol 143: 99-103, 2000, 203. Jennette .JC, Falk RJ, Wilkman AS: Anti-neutrophil cytoplasmic autoantibodies -a serologic marker for vasculitides. Ann Acad Med Singapore 24: 248-253, 1995, 204. Rottem M, Fauci AS, Hallahan CW, et al: Wegener granulomatosis in children and adolescents: clinical presentation and outcome, J Pediatr 122: 26-31, 1993, 205. Davis MD, Daoud MS, McEvoy MT, et al: Cutaneous manifestations of ChurgStrauss syndrome: a clinicopathologic correlation, J Am Acad Dermatol 37: 199-203, 1997. 206. Goodpasture EW: The significance of certain pulmonary lesions in relation to the etiology of influenza. Am J Med Sci 158: 863, 1919. 207, Harrity P, Gilbert-Barness E, Cabalka A, et al: Isolated pulmonary Goodpasture syndrome, Pediatr Pathol 11: 635--646, 1991. 208, Thorner PS, Baumal R, Eddy A, et al: Characterization of the NCI domain of collagen type IV in glomerular basement membranes (GBM) and of antibodies to GBM in a patient with anti-GBM nephritis. Clin Nephrol 31: 160--168, 1989. 209. Kalluri R, Melendez E, Rumpf KW, et aI: Specificity of circulating and tissuebound autoantibodies in Goodpasture syndrome. Proc Assoc Am Physicians 108: 134--139, 1996. 210. Siegler RL, Bond RE, Morris AH: Treatment of Goodpasture's syndrome with plasma exchange and immunosuppression. Clin Pediatr (Phila) 19: 488--491, 1980. 211. Levin M, Rigden SP, Pincott JR, et al: Goodpasture's syndrome: treatment with plasmapheresis, immunosuppression, and anticoagulation. Arch Dis Child 58: 697-702, 1983. 212. Gilvarry J, Doyle GF, Gill DG: Good outcome in anti-glomerular basement membrane nephritis. Pediatr Nephrol 6: 244--246, 1992. 213, McCarthy LJ, Cotton J, Danielson C, et al: Goodpasture's syndrome in childhood: treatment with plasmapheresi. and immunosuppression. J Clin Apheresis 9: 116-119, 1994, 214. Rydel JJ, Rodby RA: An 18-year-old man with Goodpasture's syndrome and ANCA-negative central nervous system vasculitis. Am J Kidney Dis 31: 345-349, 1998. 215, Gianviti A, Trompeter RS, Barratt TM, et al: Retrospective study of plasma exchange in patients with idiopathic rapidly progressive glomerulonephritis and vasculitis, Arch Dis Child 75: 186-190, 1996,
216. Gyselbrecht L, De Keyser F, Ongenae K, et al: Etiological factors and underlying conditions in patients with leucocytoclastic vasculitis, Clin Exp Rheumatol 14: 665--668, 1996, 217. Banker BQ, Victor M: Dermatomyositis (systemic angiopathy) of childhood. Medicine (Baltimore) 45: 261-289, 1966. 218. Crowe WE, Bove KE, Levinson JE, et al: Clinical and pathogenetic impllcations of histopathology in childhood polydermatomyositis, Arthritis Rheum 25: 126-139, 1982. 219. Tinaztepe K, Gucer S, Bakkaloglu A, et al: Familial Mediterranean fever and polyarteritis nodosa: experience of five paediatric cases. A causal relationship or coincidence? Eur J Pediatr 156: 505-506. 1997. 220. Ozdogan H, Arisoy N, Kasapcapur 0, et al: Vasculitis in familial Mediterranean fever. J Rheumatol 24: 323-327, 1997, 221. EllsworthjE, Cassidy JT, Ragsdale CG, et al: Mucha-Habennann disease in children-the association with rheumatic diseases. J Rheumatol 9: 319-324, 1982. 222. Lister PO, Hollingworth P: Arthritis associated with leucocytoclastic angiitis. Ann Rheum Dis 39: 526-527, 1980, 223. McAdam LP, O'Hanlan MA, Bluestone R, et al: Relapsing polychondritis: prospective study of 23 patients and a review of the literature. Medicine (Baltimore) 55: 193-215, 1976. 224. Blau EB: Relapsing polychondritis and retroperitoneal fibrosis in an 8-yearold boy. Am J Dis Child 130: 1149-1152, 1976. 225. Michet CJ Jr, McKenna CH, Luthra HS, et al: Relapsing polychondritis, Survival and predictive role of early disease manifestations. Ann Intern Med 104: 74--78, 1986. 226. Balsa A, Expinosa A, Cuesta M, et al: Joint symptoms in relapsing polychondritis, Clin Exp Rheumatol 13: 425--430, 1995. 227. Masaoka A, Yamakawa Y, Niwa H, et al: Pediatric and adult tracheobronchomalacia. Eur J Cardiothorac Surg 10: 87-92, 1996. 228. Sacco 0, Fregonese B, Oddone M, et al: Severe endobronchial obstruction in a girl with relapsing polychondritis: treatment with Nd YAG laser and endobronchial silicon stent. Eur Respir J 10: 494--496, 1997. 229, Varonos S, Kostaki M, Tsapra H, et al: Polychondritis associated with Sch{inleinHenoch purpura: report of a case, Clin Exp Rheumatol 12: 443-445, 1994, 230. Papo T, Wechsler B, Bletry 0, et al: Pregnancy in relapsing polychondritis: twenty-five pregnancies in eleven patient•. Arthritis Rheum 40: 1245-1249, 1997, 231. Degos R, Delort J, Tricot R: Papulose atrophiante maligne (syndrome cutaneo-intestinal morteD. Bull Soc Med Hop Paris 64: 803, 1948. 232. Strole WE Jr, Clark WH Jr, Isselbacher KJ: Progressive arterial occlusive disease (Kohlmeier-Degos), A frequently fatal cutaneosystemic disorder. N Engl J Med 276: 195-201, 1967. 233. Snow JL, Muller SA: Degos syndrome: malignant atrophic papulosis. Semin Dermatol 14: 99-105, 1995. 234. Ball E, Newburger A, Ackerman AB: Degos' disease: a distinctive pattern of disease, chiefly of lupus erythematosus, and not a specific disease per se. Am J Dermatopathol 25: 308-320, 2003. 235. Watanabe T, Onda H: Henoch-Sch{inlein purpura with antiphospholipid antihodies following an intluenza vaccination, Pediatr Nephrol 16: 458--459, 2001. 236. Monastiri K, Selmi H, Tabarki B, et al: Primary antiphospholipid syndrome presenting as complicated Henoch-Sch6nlein purpura. Arch Dis Child 86: 132-133, 2002. 237. Weerasena NA, Groome 0, PollockJG, et al: Atrial myxoma as the cause of acute lower limb ischaemia in a teenager. Scott Med J 34: 440--441, 1989. 238. Tonz M, Laske A, Carrel T, et al: Convulsions, hemiparesis and central retinal artery occlusion due to left atrial myxoma in child. Eur J Pediatr 151: 652--654, 1992. 239, Hung PC, Wang HS, Chou ML, et al: Multiple cerebral aneurysms in a child with cardiac myxoma. J Formos Med Assoc 91: 818-821, 1992. 240, Park JM, Garcia RR, Patrick JK, et al: Right atrial myxoma with a nonembolic intestinal manifestation. Pediatr Cardiol 11: 164--166, 1990. 241. Coughlin WF, Knott PE: Right atrial myxoma. A cause of septic puimonary emboli In an adolescent female. .I Adolesc Health Care 11: 351-354, 1990. 242. Pasaoglu I, Demircin M, Ozkutlu S, et ai: Right atrial myxoma in an infant. Jpn Heart J 32: 263-266, 1991. 243. Wooten MD, Jasin HE: Vasculitis and Iymphoproliferative diseases, Semin Arthritis Rheum 26: 564--574, 1996. 244. Bajwa RP, Marwaha RK, Garewal G, et al: Acute febrile neutrophilic dermatosis (Sweet's syndrome) in myelodysplastic syndrome. Pediatr Hematol Oncol 10: 343--346, 1993. 245. Garty BZ, Levy I, Nitzan M, et al: Sweet syndrome associated with G-CSF treatment in a child with glycogen storage disease type lb. Pediatrics 97: 401--403, 1996, 246, Shimizu T, Yoshida I, Eguchi H, et al: Sweet syndrome in a child with aplastic anemia receiving recombinant granulocyte colony-stimulating factor, J Pediatr Hematol Oncol 18: 282-284, 1996. 247. Tuerlinckx D, Bodart E, Despontin K, et al: Sweet's syndrome with arthritis in an 8-month-old boy, J Rheumatol 26: 440--442, 1999. 248, Moreland LW, Brick .JE, Kovach RE, et al: Acute febrile neutrophilic dermatosis (Sweet syndrome): a review of the literature with emphasis on musculoskeletal manifestations, Semin Arthritis Rheum 17: 143-153, 1988. 249. Boatman BW, Taylor RC, Klein LE, et al: Sweet's syndrome in children. South Med.J 87: 193-196, 1994. 250. Nurre LD, Rabalais GP, Callen JP: Neutrophilic dermatosis-associated sterile chronic multifocal osteomyelitis in pediatric patients: case report and review. Pediatr Dermatol 16: 214--216. 1999,
C HAP T E R 251. Cronkhite LW ]r, Canada WJ: Generalized gastrointestinal polyposis; an unusual syndrome of polyposis, pigmentation, alopecia and onychatrophia. N Engl J Med 252: 1011-1015, 1955. 252. Parsa C: Cronkhite-Canada syndrome associated with systemic vasculitis: an autopsy srudy. Hum Pathol 13: 758-760, 1982. 253. Kucukaydin M. Patiroglu TE, Okur H, et al: Infantile Cronkhite-Canada syndrome?-Case report. Eur] Pediatr Surg 2: 295-297, 1992. 254. Gray ES. Lloyd DJ, Miller SS. et al: Evidence for an immune complex vasculitis in neonatal necrotising enterocolitis. J Clin Pathol 34: 759-763, 1981. 255. Levy M. Shear NH: Mycoplasma pneumoniae infections and Stevens-Johnson syndrome. Report of eight cases and review of the literarure. Clin Pediatr (Phila) 30: 42-49, 1991. 256. Tay YK, Huff ]C, Weston WL: Mycoplasma pneumoniae infection is associated with Stevens-Johnsoo syndrome. not erythema multiforme (von Hebra). JAm Acad Dermatol 35: 757-760. 1996.
23 LE UKOCYTOC LASTIC
VASCU LITIS
S11
257. Myers MW, lick H: Hospitalization for serious blood and skin disorders following use of co-trimoxazole. Br J Clin Pharmacol 43: 446-448, 1997. 258. Messenheimer JA: Rash in adult and pediatric patients treated with lamotrigine. Can] Neurol Sci 25: SI4-518, 1998. 259. Prendiville ]S, Hebert AA, Greenwald M], et al: Management of StevensJohnson syndrome and toxic epidermal necrolysis in children. J Pediatr 115: 881-887, 1989. 26o. Pacheco LS, Sotto MN: Factor XIIIa+ dermal dendrocytes in erythema elevarum diutinum and ordinary cutaneous leukocytoclastic vasculitis lesions. ] Cutan Pathol 27: 136-140, 2000. 261. Rodriguez-Serna M, Fortea JM, Perez A, et al: Erythema elevatum diutinum associated with celiac disease: response to a gluten-free diet. Pediatr Dermatol 10: 125-128, 1993. 262. Katz SI, Gallin JI, Hertz KC, et al: Erythema elevatum diutinum: skin and systemic manifestations, immunologic studies, and successful treatment with dapsone. Medicine (Baltimore) 56: 443-455, 1977.
C HAP T E R
24
POLYARTERITIS NODOSA AND RELATED VASCULITIDES James T. Cassidy and Ross E. Petty
~bf
Polyarteritis nodosa (PAN) and cutaneous polyarteritis (CPA) are rare vasculitides in childhood. I They may represent diverse manifestations of the same disease, although some authorities regard them as separate entities linked by similarities of cutaneous lesions and histopathology. PAN is typically a multisystem disease resulting from vascular inflammation predominantly in skin, abdominal viscera, kidneys, central nervous system, and muscles. CPA is restricted to vessels of the skin, muscle, joints, and peripheral nerves. A history of developments from 1761 to 1995 in the description of idiopathic vasculitis has been summarized by Matteson. 2 A recent worldwide survey of 21 pediatric centers identified 110 patients with necrotizing vasculitis. 3 These cases were classified as polyarteritis nodosa (63, 57.2%), cutaneous polyarteritis (33, 30%), vasculitis associated with hepatitis B surface antigen (5,4.6%), and ANCA-positive microscopic polyarteritis (9, 8.1%). The female to male ratio was 56:54; the mean age at the time of the study was 9.05 +/- 3.57 years. Remarkable clinical differences were present among these vasculitic categories, and there were better survival and lower relapse rates when each was compared to similar disorders with onset in the adult years.
substantial number of children, they were not validated because no control group was included in the analysis.
Epidemiology Incidence and Prevalence PAN is uncommon at any age but occurs most frequently in the fifth and sixth decades of life; it is rare in childhood, particularly in North America. The correct classification in reported cases is often uncertain; early reviews of the subject usually included children with infantile PAN, which is now viewed as a severe manifestation of Kawasaki disease. This chapter focuses on recent studies, but the historically important reports of Fager and associates8 and Frohnert and Sheps 9 should also be consulted. Reimold and colleagues lO estimated that fewer than 150 cases of PAN in infants and children had been reported up to 1976. Since that time, there have been several additional small series.3.7.II-16
Age at Onset, Sex Ratio, and Geographic Distribution
POLYARTERITIS NODOSA Deflnltlon and Oasslflcatlon The classic form of PAN described in 1866 by Kussmaul and Maier' as periarteritis nodosa is a chronic, relapsing, febrile disease with protean manifestations resulting from inflammation of small- and medium-sized muscular arteries, often leading to aneurysm formation ("nodose lesions"). Jennette and colleagues" defined PAN as a necrotizing inflammation of small- or medium-sized arteries without glomemlonephritis or vasculitis in arterioles, capillaries, or venules. The American College of Rheumatology criteria for the classification of PAN are given in Table 24-1. 6 A number of the criteria are inappropriate for use in pediatrics and have not been evaluated in children or adolescents. Ozen and colleagues7 proposed diagnostic criteria for PAN (Table 24-2) in a group of 31 Turkish children 1 to 14 years old. Although these criteria described characteristics of the disease in a
512
PAN occurs with approximately equal frequency in boys and girls and has a peak age at onset of 9 to 11 years, although it can occur in very young children. It is probably the childhood equivalent of PAN in the adult. Distribution is worldwide, but the largest and latest reports come from Turkey7 and Japan. 16
Etiology and Pathogenesis The cause of PAN is unknown, but infectious processes are sometimes implicated. Destmction of the vascular wall may be the direct consequence of immune complex deposition and complement activation. 17 In adults, an association with hepatitis B infection has been described in 10% to 54% of patients, with the highest frequencies found among inner-city populations. IS In some patients, a relation between hepatitis B-associated antigen and immune complex formation has been proved. 19 PAN occurring after infection with hepatitis C has also been
CHAPTER
24
POLYARTERITIS NODOSA AND RELATED VASCULITIDES
_ I ABLE 24 -I
"
--
American College of Rheumatology 1990 '. rlteria for the Classification of Polyarteritis Nodosa
Crlterlon* 1. Weight loss 2. Uvedo reticularis 3. Testicular pain 4. Myalgia 5. Mononeuropathy or polyneuropathy 6. Diastolic blood pressure > 90 mm Hg 7. Increased blood urea nitrogen (BUN) or creatinine 8. Hepatitis B virus 9. Arteriographic change 10. Biopsy
Deflnhlon Loss of 4 kg or more of body weight not caused by dieting or other factors Mottled reticular pattern of the skin of portions of the extremities or torso Pain or tenderness of testicles not due to other causes Diffuse myalgia, excluding shoulder and hip girdle, or muscle weakness or tenderness of leg muscles Mononeuropathy, multiple mononeuropathies, or polyneuropathy BUN> 40 mg/dL or serum creatinine > 1.5 mg/dL not due to other causes Presence of hepatitis B surface antigen or antibody in serum Aneurysms or occlusions of visceral arteries not due to other causes Granulocytes and mononuclear leukocytes in wall of small or medium-sized artery
513
PAN12 or CPN4; in others, such an association is not apparent. PAN as a paraneoplastic event has not been reported in children, although it occasionally occurs in adults,25 It has been described following a drug reaction. 26
Animal Models ofVasculitis In animals, infectious agents, particularly viruses, have resulted in polyarteritis-like disease. 27 This finding supports the view that vasculitis in humans may be related to an initial or a sustained infectious process.
Equine Encephalitis and Anemia A vasculitis of the media of small arteries occurring in the horse is of viral origin and transmitted by an arthropod vector,27 Principal clinical features include an acute onset with high fever, conjunctival injection, subcutaneous edema of the extremities, anorexia, diarrhea, and death of two thirds of the animals. Pregnant mares abort. Hemolytic anemia and hypergammaglobulinemia are consistently present, and mild glomerulitis accompanied by immune complexes and depletion of C3 is common. A persisting viremia coexists with antibodies to the virus. The gross and microscopic lesions of this disease resemble the fibrinoid lesions of human polyarteritis.
Dennatitis-Nephropathy Syndrome in Swine 'Por classification purposes, an adult is said to have polyarteritis nodosa if at least three criteria are met (sensitivity. 82.2%; specificity. 86.6%). From Lightfoot RW Jr. Michel BA. Bloch DA. et al: The American College of Rheumatology 1990 criteria for the classification of polyarteritis nodosa. Arthritis Rheum 33: 1088-1093, 1990.
reported. 2o ,21 These associations have not been recognized in children, although a severe PAN-like vasculitis after infection with parvovirus B1922 or cytomegalovirus 23 has been described. The relation of PAN to a preceding group A streptococcal infection remains a matter of conjecture,IO·12.24 In some series, upper aitway infection with group B Streptococcus is a frequent event before onset of
Systemic vasculitis that involves the skin and kidneys has been described in swine. 28 It may result from infection with porcine reproductive and respiratory syndrome virus,
Vasculitis in Deer In 1996, systemic vasculitis associated with a high mortality rate was described in mule deer. 29 Intranuclear adenovirus was identified in six deer that were examined. This disease was similar to bluetongue virus infection and epizootic hemorrhagic disease virus infection in white-tailed deer.
Central Nervous System Vasculitis in Turkeys
""--
fABLE 24-2 Proposed Criteria for a Diagnosis of Polyarteritis Nodosa in Childhood
Infectious and fatal vasculitis of turkeys in commercial lots has been associated with Mycoplasma synoviae3° and with a toxin elaborated by Mycoplasma gallesepticum. 3u2
Major Crlterla*
Renal disease Musculoskeletal findings
Genetic Background
Minor CrIterIa
Genetic factors have not been identified for PAN, and familial occurrence is rare. The disease is so uncommon that human leukocyte antigens and other genetic markers have not been adequately studied.
Cutaneous findings Gastrointestinal involvement Peripheral neuropathy Central nervous system disease Hypertension Cardiac disease Lung disease Constitutional symptoms Increased acute phase reactants Presence of hepatitis B surface antigen Diagnosis of polyarteritis nodosa requires the presence of five criteria. including at least one major criterion, Antinuclear antibody and anti-dsDNA must be absent. From Ozen S. Besbas N, Saatci U. et al: Diagnostic criteria for polyarteritis nodosa in childhood. J Pediatr 120: 206--209. 1992.
Clinical Manifestations No single pattern of clinical presentation characterizes this disease, but the insidious onset of unexplained fever, weight loss, skin lesions, abdominal pain, and musculoskeletal pain should suggest the diagnosis (Table 24-3), The fever is usually remittent and may be low or high grade. Musculoskeletal pain with tenderness of muscle, and in two thirds of the patients overt arthritis, may be localized or diffuse. Panniculitis has been described.
514
C HAP T E R
24
POLYARTERITIS NODOSA AND RELATED VASCULITIDES
' . TABLE 24 3 C1ini(dl Abnormdlities in HI Children with Polyarteritis Nodosd
Clinical Finding Fever Arthritis/arthralgia Abdominal pain Myalgia Skin abnormalities Rash Edema Petechiae Mucous membrane abnormalities Nervous system abnormalities Seizures Other, including peripheral neuropathy Cardiac disease Respiratory disease Cervical lymphadenopathy Splenomegaly Renal
Frequency (%)
84 74 68 67
69 20 17 9 16 10 21 7
<5 <5 25
• Figure 24-1
Splinter hemorrhages under the nails of a teenage girl with
polyarteritis nodosa.
Data from references 7, 12-17.
Secondary hypertrophic osteoarthropathy is a rare cause of musculoskeletal pain in patients with PAN.33 Nonspecific abdominal pain occurs in two thirds of the patients and is presumably related to ischemia of the mesenteric and other intra-abdominal arteries. 34 Infarction of the gut, gallbladder, or pancreas has occurred. 35 Patients with PAN may have signs that suggest primary renal disease or renovascular hypertension. 36 Kidney involvement (e.g., proteinuria, nephrotic syndrome, nephritis, hypertension) has been described in 26 childrenY Central nervous system involvement ultimately develops in 50% to 70% of children. Clinical findings include the organic brain syndrome, psychosis, focal neurologic defects, aneurysms, unilateral blindness, seizures, and hemiparesis. Isolated central nervous system vasculitis and that related to Henoch-Schonlein purpura produce similar findings. A 1999 report 38 documents monocular blindness in a 9-year-old girl who presented with temporal arteritis. Magnetic resonance imaging demonstrated multiple focal cortical and subcortical ischemic changes; arteriography documented small aneurysms at bifurcations of small- and medium-sized arteries. A severe sensorimotor peripheral neuropathy (Le., mononeuritis multiplex) is characteristic of the disease. Cutaneous involvement includes painful subcutaneous nodules (particularly in the calf and fool), purpura, splinter hemorrhages (Fig. 24-1), livedo reticularis and digital ischemia (Fig. 24-2), edema, peripheral gangrene, and ulceration. A multiplicity of other signs-including testicular or epididymal swelling, pain, or tenderness and serous otitis media-rarely occur. Episcleritis has occurred, especially in older children. 39 Cardiovascular abnormalities in a study of children with PAN included mild mitral or tricuspid valve regurgitation, diminished left-ventricular systolic ejection, and pericardial thickening in one patient,40 None of the 15 children in this study had symptoms referable to the cardiovascular system, and electrocardiograms were normal.
Pathology The characteristic histopathologic features include necrotizing arteritis with the formation of nodules along the walls of small- and medium-sized muscular arteries, especially in the mesenteric vasculature, and identification of fibrinoid necrosis of the entire thickness of the vessel walls (Fig. 24-3). These lesions tend to be segmental with a predilection for bifurcations of vessels. Biopsy specimens document vasculitis in all stages of development, from acute to chronic, interspersed with areas of normal vessel wall. Aneurysm formation is higWy characteristic of this type of vasculitis. Destruction of the internal elastic lamina is common. Immunofluorescence microscopy usually documents little evidence of complement or immunoglobulin deposition.
Differential Diagnosis Although PAN can be suspected from the typical clinical presentation, diagnosis can only be secured by pathologic demonstration of characteristic vascular lesions or by radiologic documentation of aneurysms. 41 ,42 The American College of Rheumatology criteria in Table 24-1 describe the characteristics of the disease. The Chapel Hill nomenclature has also been validated but does not address sufficiently the diagnosis of Wegener's granulomatosis and microscopic polyangiitis. 43 Onset is often insidious. The presence of a cutaneous lesion or subcutaneous nodule should prompt a careful, deep excisional (not punch) biopsy that includes a muscular vessel. These lesions are often transient and should therefore be examined early in their course. In the presence of peripheral neuropathy, a muscle or nerve biopsy (sural) may be diagnostic. Selective angiography of the celiac, mesenteric, or renal vasculature is likely to demonstrate one or many aneurysms located at the bifurcations of the small- to medium-sized arteries.
C HAP T E R
24
POLYARTERITIS NODOSA AND RELATED VASCULITIDES
'~II
TABLE 24 -4
515
Laboratory Abnormalities in Polyarteritis Nodosa
Laboratory FInding Elevated erythrocyte sedimentation rate Elevated C-reactive protein level Leukocytosis Anemia Proteinuria Hematuria Increased BUN or creatinine level Antinuclear antibody present ANCA present Rheumatoid factor present
Frequency (0At)*
75
90 90
60 80 65
50 25 -15
o
'Estima1ed from published cases and series. ANCA. antineutrophil cytoplasmic antibody; BUN. blood urea nitrogen.
• FItm'e 24-2
Marked digital cyanosis, livedo reticularis, and pain were characteristics of acute polyarteritis nodosa in this 9-year-old boy. Despite muscle and kidney involvement, he made a complete recovery over the 5-year follow-up period. (See color insert.)
mental ureteral obstruction, have been reported in at least four children.52-54
Treatment The key to a correct and timely diagnosis is often a high index of clinical suspicion. Diagnostic possibilities to be considered include other forms of vasculitis, microscopic polyangiitis, relapsing polychondritis, bacterial endocarditis or microembolism, chronic viral infections (e.g., parvovirus B19), and sarcoidosis. The latter can lead to vasculitis of the small, medium, or large vessels and includes granulomatous, nongranulomatous, necrotizing, and perivascular inflammatory involvement (see Chapter 26).44
Laboratory Examination Common features of PAN include leukocytosis, thrombocytosis, anemia, and elevation of the erythrocyte sedimentation rate, C-reactive protein, and serum immunoglobulin levels, together with an abnormal urinary sediment, are frequent (Table 24-4). Circulating immune complexes may be present, but their assay is seldom of clinical value. 45 Rheumatoid factor and antinuclear antibody are seldom detected. Antineutrophil cytoplasmic antibodies may be present, but they are more common in Wegener's granulomatosis or microscopic polyarteritis (polyangiitis).46 Levels of factor VIII-related antigen47 and f3-thromboglobulin48 reflect the activity of the vascular inflammation and may be useful in follOWing the effects of treatment. A greenish tint of the plasma caused by a ceruloplaSmin-like, acute phase protein has been described. 49 Ischemic coronary arteritis may be evident on electrocardiography.
Radiologic Examination Angiographic demonstration of small aneurysms involving the renal, celiac, or mesenteric arteries is higWy characteristic of PAN (Figs. 24-4 and 24-5).50,51 Magnetic resonance angiography may confirm the presence of abnormalities of the vascular system without the need for contrast studies. Unusual but characteristic abnormalities of intravenous excretory urography, including fixed seg-
Glucocorticoid therapy is required in most children. Prednisone (l to 2 mg/kg/day) improves life expectancy and decreases the frequency of hypertension and renal involvement. 36.55 Oral cyclophosphamide (2 mg/kg/day)56 or azathioprine (2 mg/kg/day) may be useful if glucocorticoids fail. Intravenous pulse cyclophosphamide may be indicated in the more severe forms of the disease with aneurysmal involvement of the celiac and mesenteric vessels. 56 Plasmapheresis has not improved survival.57 Conflicting data regarding the effectiveness of glucocorticoid therapy have resulted in part from failure to distinguish among the several forms of polyarteritis in reports of therapeutic responsiveness. Historically, there was concern that glucocorticoids could suppress the inflammatory vasculitis without permitting adequate wound healing with the resultant formation of aneurysms, or that rapid healing of the vascular lesions could lead to occlusion and peripheral anoxia or necrosis of tissue. Direct evidence that these potential actions of glucocorticoids affect outcome in PAN is substantially lacking. Although the evidence is limited, the efficacy of intravenous immunoglobulin in the management of Kawasaki disease suggests that this therapy might also have a role in management of children in whom glucocorticoids fail and before cytotoxic agents are used. One case report supports this possibility: Intravenous immunoglobulin was effective in a 2-year-old boy with necrotizing vasculitis. 58 Severe vascular insufficiency affecting the extremities and causing gangrene may respond to iloprost. 22 In patients in whom there is an association with streptococcal infection, prophylaxis with penicillin for prevention of recurrences has been advocated. 12
Course of the Disease and Prognosis The reported outcome of children with PAN varies a great deal from study to study, perhaps in part related to
516
C HAP T E R
24
POLYARTERITIS NODOSA AND RELATED VASCULITIDES
• Figure Z4-3 A, This biopsy of a medium-sized muscular artery exhibits marked fibrinoid necrosis of the vessel wall (alTOw) characteristic of polyarteritis nodosa (PAN). 8, The muscle biopsy specimen from a 6-year-old boy with PAN demonstrates vasculitis with marked mixed cellular infiltration in and around the walls of the arteriole (a) and venules (v). Rbrinoid necrosis of the arteriole is visible. C, Infiltration of mononuclear cells around the central arteriole, which is surrounded by a dense fibrous scar.The lumen is occluded by a dense thrombus. 0, PAN in a boy with hematuria that developed 3 years after the onset of disease. A renal biopsy specimen demonstrated deposits of immunoglobulin G(IgG) in the media of the afferent arterioles (Le., glomerulus at the upper left comer). A-E, H & E, x 480.
the criteria used for classification; this reflects not only improvement in prognosis with recent treatment protocols, but also differences in the severity of the disease in various series. It is difficult to rationalize these differences but important to bear them in mind when considering management of any child. Even in children with substantial systemic involvement characterized by fever, calf pain, subcutaneous nodules, elevated acute phase reactants, and multisystem vasculitis demonstrated by
biopsy or arteriography, outcome is often without lifethreatening sequelae. B In one study,14 9 children, most of whom had classic PAN, were followed for a mean of 4 years and treated with high-dose prednisone (approximately 2 mg/kg/day). Although seriolls complications (e.g" myocardial infarction, systemic hypertension, impaired renal function) occurred, the course was, in general, chronic with no mortality. In contrast, the report of Fink 12 described 8 children with PAN, followed between 1959 and 1974,
C HAP T E R
24
POLYARTERITIS NODOSA AND RELATED VASCULITIDES
517
• FIgIn 24-4 A, Characteristic renal aneurysms (arrow) visualized by angiography in a lS-year-old boy who presented with hypertension, myocardial infarction, and hematuria. B, The lesions eventually resolved, as demonstrated on this repeat angiogram some years after a course of prednisone therapy.
who developed severe hypertension, seizures, and renal failure, and 3 died. All 10 patients in a series from )apan l6 survived, although serious sequelae were observed in one third.
CUTANEOUS POLYARTERITIS There is no strict definition of CPA and therefore no accurate estimate of the number of reported cases, although it is rare in children. Kumar and colleagues59 reported 10 children from India with benign cutaneous PAN who
• FIgIIre 24-5 Celiac angiography documents aneurysms (arrows) in multiple vessels of an 18-year-old boy with polyarteritis nodosa.
were 1 to 10 years old at diagnosis. The disease was limited to fever, skin, and joints (fever in 10; peripheral gangrene in 8; livido reticularis in 4; ulceration, nodules, and vesiculobullous lesions alone or in combination in 10; necrotic patches over limbs and truck in 3; and arthralgia or effusions in 7). Three children had cryoglobulinemia. In a 1998 case report and literature review, Ginarte and coworkers60 reported 45 cases. Mocan and colleagues61 identified an additional child who met their definition of CPA: tender subcutaneous nodules with high fever, arthralgias, and myalgias but without major organ system involvement. The child improved markedly with monthly methylprednisolone administration. The most characteristic clinical findings are purpura (Fig. 24-6) and multiple tender subcutaneous nodules, particularly on the lower extremities and sometimes on the soles of the feet. Livedo reticularis is common. Ulceration may occur and is associated with a more protracted course and peripheral neuropathy, at least in adults. 62 Biopsy of the dermis and subdermal structures demonstrates panarteritis. CPA usually responds to glucocorticoids (prednisone, 0.5 to 1.0 mg/kg/day), although in some children, the course may be prolonged and recurrences frequent, sometimes spanning a decade or more. 63 . 64 When streptococcal infection is implicated as a triggering event,13.65-{i7 prophylaxis with penicillin may be effective.63 Intravenous immunoglobulin has been used successfully to treat a 9-year-old boy65 and a 2-year-old boy58 with poststreptococcal disease. Children with CPA usually have a favorable outlook, although the disease is often characterized by relapses over a period of many years. In the series of children with CPA described by Kumar and colleagues,59 most of the patients did well despite developing digital gangrene.
518
C HAP T E R
24
POLYARTERITIS
N ODOSA
AND RELATED VASCU LITIDES
,~.
TABI E 24-5
Clini(al Manil..,slations of Cogan's Syndronll!
Inflammatory eye disease' Interstitial keratitis Other (conjunctivitis, episc!eritis/sc!eritis, vitritis, retinal vasculitis, retinitis/choroiditis) Vestibuloauditory dysfunction' Hearing impairment, vertigo, tinnitus, ear pain Systemic vasculitis Aortitis Aortic valve insufficiency Vasculitis of small- and medium-sized arteries Constitutional Fever, weight loss, arthritis, abdominal pain, hepatosplenomegaly, lymphadenopathy
• figure Z4-6 Acute arthritis involving the ankle and tarsometatarsal joints and vascular necrotic purpura occur in a5-year-old girl with cutaneous polyarteritis.
There are three reports of neonates with CPA born to mothers with the disease.6&-70 All three had livedo reticularis and subcutaneous nodules. Necrosis of fingers and toes occurred in two. Treatment with glucocorticoids was advocated, but concomitant heparin may be required for acral arterial insufficiency.7o The transient nature of the infants' lesions suggests an unknown transplacental effect rather than a genetic pathogenesis.
OTHER RARE VASCULITIDES Cogan's Syndrome Cogan's syndrome (Le., nonsyphilitic interstitial keratitis with vestibuloauditory dysfunction)71 is a rare disease that most frequently affects young adults. It has been identified in a number of children. 72-81 The cause is unknown, and associations with Borrelia burgdorferi,72 Chlamydia trachomatis,73 or Chlamydia psittaci 74 infection are unconvincing. The ocular and auditory-vestibular abnormalities may result from organ-specific autoimmune processes. R2 The major clinical features are listed in Table 24-5. In addition to photophobia, vertigo, and cochleovestibular hearing IOSS,R3 patients may have widespread vasculitis,7'; including aortitis and aortic valve insufficiency, that resembles Takayasu's arteritis; both disorders have been reported in one adult. R4 In some patients, involvement of other vessels, such as the renal artery, may suggest a systemic vasculitis such as PAN. 85.86 Most patients with active disease have leukocytosis, thrombocytosis, anemia, and an elevated erythrocyte sedimentation rate. Early treatment with systemic glucocorticoids is beneficial, although hearing loss may persist. Methotrexate and pulse cyclophosphamide have been advocated as an effective approach to patients who have not had a satisfactory response to glucocorticoids. R7-li9
Cystic Fibrosis and Vasculitis (Soter's Syndrome) Soter and colleagues90 described two males with cystic fibrosis who developed palpable purpura from a cutaneous necrotizing venulitis. Confirmatory reports have
'Both elements are required to make the diagnosis, Adapted from St. Clair EW, McCallum RM: Cogan's syndrome. Curr Opin Rheumatol 11: 47-52, 1999.
subsequently been published,9I-94 The venulitis can be recurrent92 and potentially associated with thrombosis,9~ Antineutrophil cytoplasmic antibodies (ANCAs) have been described. 96
Familial Mediterranean Fever and Vasculitis A possible association between PAN and familial Mediterranean fever (FMF), a disease with a documented genetic basis (see Chapter 34), has been reported,97-lo3 Tinaztepe and colleagues99 reviewed 16 cases of FMF and PAN in the literature and described another 5 patients. Tekin and colleagues 101 examined 23 children with FMFassociated vasculitis (HSP in 11, PAN in 2, protracted febrile attacks in 10). Most of these patients had mutations in the MEFV gene. FMF may also predispose to renal vascular involvement,104 All reports of the coincidence of FMF and PAN have come from Turkey, where FMF and PAN are more common than in North America or Northern Europe. It is not evident whether vasculitis is part of FMF (at least in the Middle East) or whether the two disorders occur by coincidence in the same individual. Environmental and associated genetic factors are possibly involved. The identification of the gene causing FMF and the periodic febrile syndromes should permit screening of patients with PAN to further delineate this association (see Chapter 34).
REFERENCES 1. Dillon MJ: Childhood vasculitis. Lupus 7: 259-265. 1998. 2. Matteson EL: A history of early investigation in polyarteritis nodosa, Arthritis Care Res 12: 294-302, 1999. 3. Ozen S, Anton J, Atisoy N, et al: Juvenile polyalteritis: result. of a multicenter survey of 110 children. J Pediatr 145: 517-522, 2004. 4. Kussmaul A, Maier K: Uber eine bisher nichl beschriebene eigenthumliche Arterienerkrankung (Periarteritis nodosa). die mit Morbus Brightii und rapid fortshreitender allgemeiner Muskellahmung einhergehL On a preViously undescribed peculiar arterial disease (Periarteritis nodosa). accompanied by Bright's disease and rapidly progressive general muscle weakness. Dtsch Arch KIln Med 1: 484, 1866. 5. Jennette JC. Falk RJ, Andrassy K, et al: Nomenclature of systemic vasculitides. Proposal of an international consensus conference. Arthritis Rheum .P: 187-192, 1994.
CHAPTER
24
POLYARTERITIS NODOSA AND RELATED VASCULITIDES
6. Ughtfoot RW ]r, Michel BA, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of polyarteritis nodosa. ArthritL' Rheum 33: 108B-I093, 1990. 7. Ozen S, Besbas N, Saatci U, et al: Diagnostic criteria for polyarteritis nodosa in childhood. ] Pediatr 120: 206-209, 1992. 8. Fager DB, Bigler ]A, Simonds ]P: Polyarteritis nodosa in infancy and childhood. ] Pediatr 39: 65, 1951. 9. Frohnert PP, Sheps SG: Long-term follow-up study of periarteritis nodosa. Am] Med 43: B-14, 1967. 10. Reimold EW, Weinberg AG, Fink CW, et al: Polyarteritis in children. Am] Dis Child 130: 534-541, 1976. 11. Blau EB, Morris RF, Yunis E]: Polyarteritis nodosa in older children. Pediatrics 60: 227-234, 1977. 12. Fink CWo Polyarteritis and other diseases with necrotizing vasculitis in childhood. Arthritis Rheum 20: 37B-384, 1977. 13. Petty RE, Magilavy DB, Churg], et al: Polyarteritis in childhood. A clinical description of eight cases. Arthritis Rheum 20 (Supp)): 39, 1977. 14. Magilavy DB, Petty RE, Cassidy ]T, et al: A syndrome of childhood polyarteritis. ] Pediatr 91: 25--30, 1977. 15. Enlinger RE, Nelson AM, Burke EC, et al: Polyarteritis nodosa in childhood a clinical pathologic study. Arthritis Rheum 22: 820-825, 1979. 16. Maeda M, Kobayashi M, Okamoto S, et al: Clinical observation of 14 cases of childhood polyarteritis nodosa in Japan. Acta Paediatr ]pn 39: 277-279, 1997. 17. McCluskey RT, Fienberg R: Vasculitis in primary vasculitides, granulomatoses, and connective tissue diseases. Hum Pathol 14: 305--315, 1983. 18. GuiIlevin L, Lbote F, Cohen P, et al: Polyarteritis nodosa related to hepatitis B virus. A prospective study with long-term observation of 41 patients. Medicine (Baltimore) 74: 23B-253, 1995. 19. Gocke OJ, Hsu K, Morgan C, et al: Association between polyarteritis and Australia antigen. Lancet 2: 1149-1153, 1970. 20. Cacoub P, Lunel-Fabiani F, Du LT: Polyarteritis nodosa and hepatitis C viOlS infection. Ann Intern Med 116: 605--606, 1992. 21. Deny P, Guillevin L, Bonacorsi S, et al: Association between hepatitis C virus and polyarteritis nodosa. Clin Exp Rheumatol 10: 319, 1992. 22. Zulian F, Costantini C, Montesco MC, et al: Successful treatment of gangrene in systemic necrotizing vasculitis with iloprost. Br] Rheumatol 37: 22B-230, 1998. 23. Fernandes SR, Bertolo MB, Rossi CL, et al: Polyarteritis nodosa and cytomegalovirus: diagnosis by polymerase chain reaction. Clin Rheumatol 18: 501-503, 1999. 24. Sheth AP, Olson]C, Esterly NB: Cutaneous polyarteritis nodosa of childhood. .I Am Acad Dermatol 31: 561-566, 1994. 25. Seelen MA, de Meijer PH, Arnoldus EP, et al: A patient with multiple myeloma presenting with severe polyneuropathy caused by necrotizing vasculitiS. Am] Med 102: 485--486, 1997. 26. Schrudt B], Callen ]P: Polyarteritis nodosa anributable to minocycline treatment for acne vulgaris. Pediatrics 103: 503-504, 1999. 27. Bryans .IT, Doll ER, Jones TC: The lesions of equine viral arteritis. Cornell Vet 47: 52-68, 1957. 28. Thibault S, Drolet R, Germain MC, et al: Cutaneous and systemic necrotizing vasculitis in swine. Vet Pathol 35: 10B-116, 1998. 29. Woods LW, Swift PK, Barr BC, et al: Systemic adenovirus infection associated with high mortality in mule deer (Odocoileus hemionus) in California. Vet Pathol 33: 125--132, 1996. 30. Chin RP, Meteyer CU, Yamamoto R, et al: Isolation of Mycoplasma synoviae from the brains of commercial meat turkeys with meningeal vasculitis. Avian Dis 35: 631-637, 1991. 31. Thomas L: The neurotoxins of M. neurolyticum and M. ga/lisepticum. Trans Assoc Am Physicians 79: 38B--398, 1966. 32. Chin RP, Daft BM, Meteyer CU, et al: Meningoencephalitis in commercial meat turkeys associated with Mycoplasma gallisepticum. Avian Dis 35: 986--993, 1991. 33. Woudward AH, Andreini PH: Periosteal new bone formation in polyarteritis nudosa: a syndrome involving the lower extremities. Arthritis Rheum 17: 1017-1025, 1974. 34. Oguzkurt L, Cekirge S, Balkanci F: Inferior suprarenal artery aneurysm in polyarteritis nodosa. Pediatr Radiol 27: 234-235, 1997. 35. Adu 0, Bacon PA: Classical polyarteritis nodosa, microscopic polyart.eritis, and Churg-Strauss syndrome. In Maddison P], Isenberg DA, Woo P, et al (eds): Oxford Textbook of Rheumatology, 2nd ed. Oxford, England, Oxford University Press, 1998, p. 1351. 36. Furlong T], Ibels LS, Eckstein RP: The clinical spectrum of necrotizing glomerulonephritis. Medicine (Baltimore) 66: 192-201, 1987. 37. Besbas N. Ozen S, Saatci U, et al: Renal involvement in polyarteritis nodosa: evaluation of 26 Turkish children. Pediatr Nephrol 14: 325--327, 2000. 38. Bert R], Antonacci VP, Berman L, et al: Polyarteritis nodosa presenting as temporal arteritis in a 9-year-old child. A]NR Am] Neuroradiol 20: 167-171, 1999. 39. McCluskey P], Watson PG, Lightman S, et al: Posterior scleritis: clinical features, systemic associations, and outcome in a large series of patients. Ophthalmology 106: 2380-2386, 1999. 40. Gunal N, Kara N, Cakar N, et al: Cardiac involvement in childhood polyarteritis nodosa. Int J Cardiol 60: 257-262, 1997.
519
41. Hunder GG, Arend WP, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of vasculitis. Introduction. Arthritis Rheum 33: 1065--1067, 1990. 42. Lie ]T: Illustrated histopathologic classification criteria for selected vasculitis syndromes. American College of Rheumatology Subcommittee on Classification of Vasculitc,. Arthritis Rheum 33: 1074-1087, 1990. 43. Sorensen SF, Slot 0, Tvede N, et al: A prospective study of vasculitis patients collected in a five year period: evaluation of the Chapel Hill nomenclature. Ann Rheum Dis 59: 47&-482, 2000. 44. Fernandes SR, Singsen BH, Hoffman GS: Sarcoidosis and systemic vasculitis. Semin Arthritis Rheum 30: 33-46, 2000. 45. Levin M, Holland PC, Nokes TJ, et al: Platelet immune complex interaction in pathogenesis of Kawasaki disease and childhood polyarteritL,. Br Med ] (Clin Res Ed) 290: 1456-1460, 1985. 46. Nash MC, Dillon MJ: Antineutrophil cytoplasm antibodies and vasculitis. Arch Dis Child 77: 261-264, 1997. 47. Woolf AD, wakeriey G, Wallington TB, et al: Factor VlII related antigen in the assessment of vasculitis. Ann Rheum Dis 46: 441-447, 1987. 48. Burns ]C, Glode MP, Clarke SH, et al: Coagulopathy and platelet activation in Kawasaki syndrome: identification of patients at high risk for development of coronary artery aneurysms. ] Pediatr 105: 206-211, 1984. 49. Dillon M]: Primary vasculitis in children. In Maddison P], Isenberg DA, Woo P, et al (eds): Oxford Textbook of Rheumatology, 2nd. ed. Oxford, England, Oxford University Press, 1998, pp. 1402-1413. 50. Mclain LG, Kelsch RC, Bookstein ]]: Polyarteritis nodosa diagnosed by renal arteriography. ] Pediatr 80: 1032-1035, 1972. 51. Yousefzadeh OK, Chow KC, Benson CA: Polyarteritis nudosa: regression of arterial aneurysms following immunosuppressive and corticosteroid therapy. Pediatr Radiol 10: 139-141, 1981. 52. Fisher RS, Howard HH: Unusual ureterograms in a case of periarteritis nodosa. ] Urol 60: 398, 1948. 53. Glanz I, Grunebaum M: Ureteral changes in polyarteritis nodosa as seen during excretory urography.] Urol 116: 731-733, 1976. 54. Khanfar NM, Morgenstern BZ: 15-year-old boy with abdominal pain and hypertension. Mayo Clin Proc 71: 713-716, 1996. 55. Sack M, Cassidy]T, Bole GG: Prognostic factors in polyarteritis. ] Rheumatol 2: 411--420, 1975. 56. Fauci AS, Katz P, Haynes BF, et al: Cyclophosphamide therapy of severe systemic necrotizing vasculitis. N Engl] Med 301: 235--238, 1979. 57. Guillevin L, Fain 0, Lhote F, et al: Lack of superiority of steroids plus plasma exchange to steroids alone in the treatment of polyarteritis nudosa and Churg-Strauss syndrome. A prospective, randomized trial in 78 patients. Arthritis Rheum 35: 20B-215, 1992. 58. Gedalia A, Correa H, Kaiser M, et al: Case report: steroid sparing effect of intravenous gamma globulin in a child with necrotizing vasculitis. Am] Med Sci 309: 226-228, 1995. 59. Kumar L, Thapa BR, Sarkar B, et al: Benign cutaneous polyarteritis nudosa in children below 10 years of age-a clinical experience. Ann Rheum Dis 54: 134-136, 1995. 60. Ginarte M, Pereiro M, Toribio J: Cutaneous polyarteritiS nodosa in a child. Pediatr Dennatol 15: 103-107, 1998. 61. Mocan H, Mocan MC, Peru H, et al: Cutaneous polyarteritis nodosa in a child and a review of the literature. Acta Paediatr 87: 351-353, 1998. 62. Daoud MS, Hutton KP, Gibson LE: Cutaneous periarteritis nodosa: a clinicopathological study of 79 cases. Br] Dermatol 136: 706-713, 1997. 63. Till SH, Amos RS: Long-term follow-up of juvenile-onset cutaneous polyarteritis nodosa associated with streptococcal infection. Br] Rheumatol 36: 909-911, 1997. 64. Albornoz MA, Benedetto AV, Korman M, et al: Relapsing cutaneous polyarteritis nodosa associated with streptococcal infections. Int J Dermatol 37: 664-666, 1998. 65. Uziel Y, Silverman ED: Intravenous immunoglobulin therapy in a child with cutaneous polyarteritis nodosa. Clin Exp Rheumatol 16: 187-189, 1998. 66. David], Ansell BM, Woo P: Polyarteritis nodosa associated with streptococcus. Arch Dis Child 69: 685--688, 1993. 67. Siberry GK, Cohen BA, Johnson B: Cutaneous polyarteritis nodosa. Reports of two cases in children and review of the literature. Arch Dennatol 130: 884-889, 1994. 68. Boren R], Everett MA: Cutaneous vasculitis in mother and infant. Arch Dermatol 92: 56B-570, 1965. 69. Miller ]], III, Fries ]F: Simultaneous vasculitis in a mother and newborn infant. ] Pediatr 87: 443-445, 1975. 70. Stone MS, Olson RR, Weismann ON, et a1: Cutaneous vasculitis in the newborn of a mother with cutaneous polyarteritis nodosa.] Am Acad Dermatol 28: 101-105, 1993. 71. Cogan DG: Syndrome of nonsyphilitic interstitial keratitis and vestibuloauditory symptoms. Arch Ophthalmol 33: 144, 1945. 72. Schwegmann ]P, Enzenauer R]: Cogan's syndrome mimicking acute Lyme arthritis. Am] Orthop 24: 426--428, 1995. 73. Haynes BF, Kaiser-Kupfer MI, Mason P, et al: Cogan syndrome: studies in thirteen patients, long-term follow-up, and a review of the literature. Medicine (Baltimore) 59: 426--441, 1980. 74. Darougar S, John AC, Viswalingam M, et al: Isolation of Chlamydia psitraci from a patient with interstitial keratitis and uveitis associated
520
75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90.
C HAP T E R
24
POLYART E R I TI S NOD 0 S A AND
with otological and cardiovascular lesions. Br] Ophthalmol 62: 709-714, 1978. Kundell SP, Ochs HD: Cogan syndrome in childhood. ] Pediatr 97: 96-98, 1980. Cheson BD, Blunting AZ, Alroy]: Cogan's syndrome: a systemic vasculitis. Am] Med 60: 549-555, 1976. Podder S, Shepherd RC: Cogan's syndrome: a rare systemic vasculitis. Arch Dis Child 71: 163-164, 1994. Bachynski B, Wise J: Cogan's syndrome: a treatable cause of neurosensory deafness. Can} Ophthalmol 19: 145-147, 1984. Andler W, Hulse M, Bruch PM, et al: Cogan's syndrome in childhood (author's transl). Monatsschr Kinderheilkd 125: 161-164, 1977. Kasapcopur 0, Ashraf M, Caliskan S, et al: Cogan's syndrome: a rare vasculitis in childhood. .1 Rheumatol 27: 1824-1825, 2000. Olfat M, Al Mayouf SM: Cogan's syndrome in childhood. Rheumatol Int 20: 246-249, 2001. St Clair EW, McCallum RM: Cogan's syndrome. Curl' Opin Rheumatol 11: 47-52, 1999. Ndiaye IC, Hassi S.1, Wiener-Vacher SR: Cochleovestibular impairment in pediatric Cogan's syndrome. Pediatrics 109: E38, 2002. Raza K, Karokis 0, Kitas GO: Cogan's syndrome with Takayasu's arteritis. Br , Rheumatol 37: 369-372, 1998. 'Vella .1P, O'Caliaghan.1, Hickey 0, et al: Renal artery stenosis complicating Cogan's syndrome. Clin Nephrol47: 407-408, 1997. Gaubitz M, Lubben B, Seidel M, et al: Cogan's syndrome: organ-specific autoimmune disease or systemic vasculitis? A report of two cases and review of the literature. Clin Exp Rheumatol 19: 463-469, 2001. Richardson B: Methotrexate therapy for hearing loss in Cogan's syndrome. Arthritis Rheum 37: 1559-1561, 1994. Riente L, Taglione E, Berrettini S: Efficacy of methotrexate in Cogan's syndrome. ] Rheumatol 23: 1830-1831, 1996. Watanabe K, Nishimaki T, Yoshida M, et al: Atypical Cogan's syndrome successfully treated with corticosteroids and pulse cyclophosphamide therapy. Fukushima} Med Sci 46: 49-54, 2000. Soter NA, Mihm MC .11', Colten HR: Cutaneous necrotizing venulitis in patients with cystic fibrosis. ] Pediatr 95: 197-201, 1979.
R E L ATE D V AS C U LI TI DES
91. .1ohn EG, Meden!s R, Rao S: Cutaneous necrotizing venulitis in patients with cystic fibrosis . .1 Pediatr 97: 505, 1980. 92. Fradin MS, Kalb RE, Grossman ME: Recurrent cutaneous vasculitis in cystic fibrosis. Pediatr Dennatol 4: 108-111, 1987. 93. Finnegan M], Hinchcliffe}, Russell-.1ones D, et al: Vasculitis complicating cystic fibrosis. Q.1 Med 72: 609-621, 1989. 94. Hodson ME: Vasculitis and arthropathy in cystic fibrosis . .1 R Soc Med 85 (Suppl 19): 38-40, 1992. 95. Collinson}, Haworth C, O'Caliaghan C: Recurrent venous thrombosis in a patient with cystic fibrosis. Pediatr Pulmonol 20: 410-412, 1995. 96. Sediva A, Bartunkova.1, Kolarova I, et al: Antineutrophil cytoplasmic autoantibodies (ANCA) in children with cystic fibrosis.] Autoimmun 11: 185-190, 1998. 97. Glikson M, Galun E, Schlesinger M, et al: Polyarteritis nodosa and familial Mediterranean fever: a report of 2 cases and review of the literature. .1 Rheumatol 16: 536-539, 1989. 98. Kocak H, Cakar N, Hekemoglu B, et al: The coexistence of familial Mediterranean fever and polyarteritis nodosa: report of a case. Pediatr Nephrol 10: 631, 1995. 99. Tinaztepe K, Gucer S, Bakkaloglu A, et al: Familial Mediterranean fever and polyarteritis nodosa: experience of five paediatric cases. A causal relationship or coincidence? Eur] Pediatr 156: 505-506, 1997. 100. Ozdogan H, Arisoy N, Kasapcapur 0, et al: Vasculitis in familial Mediterranean fever. .1 Rheumatol 24: 323-327, 1997. 101. Tekin M, Yakinkaya F, Tumer N, et al: Clinical, laboratory and molecular characteristics of children with Familial Mediterranean Fever-associated vasculitis. Acta Paediatr 89: 177-182,2000. 102. Oguzkurt P, Akcoren Z, Kale G, et al: Polyarteritis nodosa involving the hepatobiliary system in an eight-year-old girl with a previous diagnosis of familial Mediterranean fever. Eur.1 Pediatr Surg 10: 145-147. 2000. 103. Ozen S, Ben Chetrit E, Bakkaloglu A, et al: Polyarteritis nodosa in patients with familial Mediterranean fever (FMF): a concomitant disease or a feature of FMF? Semin Arthritis Rheum 30: 281-287, 2001. 104. Tekin M, Yalcinkaya F, Tumer N, et al: Familial Mediterranean fever-renal involvement by diseases other than amyloid. Nephrol Dial Transplant 14: 475-479, 1999.
25
.
C HAP T E R
KAWASAKI DISEASE Robert P. Sunde. and Ross E. Petty
~
HISTORICAL BACKGROUND Kawasaki disease (KD) is one of the most common vasculitides of childhood. It is an acute, self-limited, systemic inflammatory process with the potential to cause severe complications and significant morbidity and mortality. Expeditious treatment can largely prevent these complications, underscoring the importance of early and accurate diagnosis. The diagnosis is based on clinical criteria (Table 25-1), and correct identification of KD can still be as exacting a challenge today as it has been for almost 40 years. This vasculitis bears the eponym Kawasaki disease because of the painstaking description of this novel febrile illness in 50 children by Tomisaku Kawasaki in 1967. 1 A case series reported in 1960 by Itoga 2 identified what was most likely the same condition, although he thought that it was a variant of Stevens-Johnson syndrome. Scattered case reports of young children who died of ruptured or thrombosed coronary artery aneurysms have appeared in the medical literature since 1871,3 and this infantile polyarteritis nodosa, a uniformly fatal condition diagnosed at autopsy, apparently represented the most severe cases of this same disease.' Although KD has existed for more than 100 years, only with the publication of diagnostic criteria has the condition been widely recognized.
DEFINmON AND DIAGNOSTIC CRITERIA Guidelines for the diagnosis of KD have not changed since they were proposed in 1967. Diagnosis requires the presence of fever lasting 5 days or more without any other explanation in addition to at least four of the five physical findings S described in Table 25-1: conjunctivitis, lymphadenopathy, rash, mucosal changes, and changes in the extremities. As with all clinical criteria, these are imperfect guidelines, with less than 100% sensitivity and specificity. If a child has the characteristic clinical features and develops coronary artery aneurysms, the diagnosis is certain. However, most children who fulfill diagnostic criteria for KD do not have demonstrable abnormalities of the coronary arteries. Children who do not meet the criteria may have an incomplete or atypical form of KD (discussed later). Alternatively, some patients who fulfill five or six
of the criteria may have other conditions. In a study of patients referred because of possible KD, Burns and colleagues6 found that the standard clinical diagnostic criteria for KD were fulfilled in 18 (46%) of 39 patients in whom other diagnoses were established. In up to 40% of cases, Kawasaki disease may coexist with infectious conditions. 7 The original diagnostic guidelines for KD were published before the recognition of cardiac involvement and were not intended to identify children at risk for developing coronary artery abnormalities. It is not surprising that many children who develop coronary artery aneurysms never meet criteria for KD.8 A review of 127 patients treated for KD found that 36% did not meet criteria but that the proportion of aneurysms was higher among this group than among those who had the full clinical syndromeY The youngest patients are most likely to have atypical features and to develop aneurysms-up to 60% of children younger than 12 months of age developed aneurysms in one series. 10 The diagnosis of KD should be considered for any infant with prolonged, unexplained fever. However, alternative explanations for the child's symptoms must be carefully excluded before treating empirically with MG. Treating KD is seldom an emergency, especially when patients present after only 5 or 6 days of fever. Observation of children who do not fulfill criteria may be the best course of action. Spontaneous improvement and resolution of fever suggest a viral illness, whereas persistence of fever or development of additional signs of KD favors treatment for KD.
EPIDEMIOLOGY Kawasaki disease is an illness of early childhood. Eightyfive percent of affected patients are younger than the age of 5 years, with an average age of approximately 2 years. ll Occurrence beyond early childhood is uncommon,12 although there are reports of KD occurring in older children 13 and adults. 14 The overall incidence of the disease is approximately 10 cases per 100,000 children per year in the United States. IS The rate is much higher in Asia, and more than 100,000 cases of KD have been registered in Japan since the initial description of the
521
522
~~
C HAP T E R
TABLE 25 I
25 K AWAS A KID I SEA S E
Criteria for the Diagnosis of Kawasaki Disease
Fever for more than 5 days (4 days if treatment with intravenous immunoglobulin eradicates fever) plus at least four of the following clinical signs not explained by another disease process: • Bilateml conjunctival injection (80% to 90%)" • Changes in the oropharyngeal mucous membranes, including one or more of injected and/or fissured lips, stmwberry tongue, injected pharynx (80% to 90%) • Changes in the peripheral extremities, including erythema and/or edema of the hands and feet (acute phase) or periungual desquamation (convalescent phase) (80%) • Polymorphous rash, primarily truncal; nonvesicular (> 90%) • Cervical lymphadenopathy with at least one node > 1.5 cm (50%) 'Numbers in parentheses indicate the approximate percentage of children with Kawasaki disease who demonstrate the criterion, 'Moditled from Centers for Disease Control: Revised diagnostic criteria for Kawasaki disease, MMWR Morb Mortal WkJy Rep 39: 27-28. 1990.
disease in 1967. 16 Among japanese children younger than 5 years, the attack rate is 90 cases per 100,000 children per year. 17 Children of japanese descent who reside outside japan also face a higher risk of KD than do white children. 18 African Americans are at intermediate risk,ll In one large area of Great Britain, the annual incidence rate was 5.5 cases per 100,000 for children younger than 5 years old; the incidence for children of Asian ancestry was more than double that for white and African American children. 19 In japan, KD is more common in boys than in girls (male/female ratio of 1.36: 1). Among boys, the highest incidence occurs between the ages of 9 and 11 months (227.3 cases per 100,000 children), and among girls, the highest incidence occurs between the ages of 3 and 8 months 033.9 cases per 100,000 children).2o In North America, the peak age at onset of KD is somewhat older, and children in the 2- to 3-year-old age group are most commonly affected. In an Australian study, only 20% of children were younger than 1 year at diagnosis, and 25% were older than 5 years. The reasons for the geographic differences in age at onset are unclear. 21 Several reports document a seasonal incidence of KD,21 and in North America, cases have tended to occur between November and May.22 Clustering of cases in time and geographic area further suggests an unrecognized vector. Although distinct epidemics of KD were documented in japan up to 1987, none has occurred since that time. 23 In japan, siblings of affected children have a risk of contracting KD that is approximately 10 times higher than the risk in the general population,24 but cases among children sharing the same home in other countries are uncommon. 2S The 3% recurrence rate l6 and increased incidence of KD occurring simultaneously in twins also support the theory that an infectious agent causes KD.
ETIOLOGY AND PATHOGENESIS The cause of KD remains unknown. 26 Many of its epidemiologic and clinical manifestations suggest an infectious origin. The fever, exanthem, lymphadenopathy,
conjunctivitis, and lesions of the oral mucosa are reminiscent of a bacterial or viral illness, and bear a partiClIlarly close resemblance to scarlet fever. If an infectious agent causes KD, it is of very low communicability, or almost all cases of infection are subclinical. Repeated attempts to identify a particular infectious trigger have been unsuccessfu1. 27 Consequently, researchers have also evaluated the possibility that vasculitis in KD is caused by antigens that trigger an immune response to endothelial cells, rather than by direct infection of the vessels. Whether such a trigger may be a conventional antigen or a so-called superantigen is debated. 28 Superantigens are produced by several bacteria, notably certain strains of Staphylococcus and Streptococcus, and they are capable of stimulating large numbers of T cells in an antigennonspecific manner by interaction with the Bchain of the T cell receptor. An indicator of the effect of superantigens is skewing of the T cell receptor V~ repertoire. Overrepresentation of T cells bearing V~2 among lymphocytes in coronary artery aneurysms and intestinal mucosa from patients with KD supports the hypothesized role of superantigens in the pathogenesis. 29.3o Brogan and colleagues31 have demonstrated that major histocompatibility complex (MHC) class II-positive endothelial cells are capable of activating CD4+ and CD8+ T cells in the presence of superantigens. A protective effect of maternal antibodies to superantigens has been suggested,32 In a murine model of KD, Duong and associates 33 demonstrated the superantigenic activity of Lactobacillus casei cell wall extracts. Mycobacterial antigens also may function as superantigens, and children with KD have unique reactions to these organisms. Inflammatory changes occur at the site of a previous bacillus Calmette-Guerin (BCG) immunization,34 as do temporarily positive responses to mycobacterial antigens in vivo and in vitro during the acute phase of the disease. 3s Lymphocytes from convalescent patients react with the 65-kD heat-shock protein from mycobacteria. 36 Whether these responses represent a specific response to mycobacterial antigens or cross-reactivity with other antigens is not clear. A predominance of immunoglobulin A (IgA)-secreting plasma cells in the blood vessel walls of children with fatal KD suggests that an organism that gained entry through mucosal surfaces underlies the disease. v No single pathogen is regularly demonstrable, although associations with Epstein-Barr virus,38 rotavirus,39 and other viruses 40-42 and with bacteria43,44 have been reported. This suggests the possibility that the vascular injury in KD may be the result of direct cell-mediated attack of endothelial cells that are infected with an unidentified infectious agent,4S Additional clues to the cause of KD come from the humoral factors, including anti-endothelial cell antibodies, circulating immune complexes,18 and antineutrophil c:ytoplasm antibodies (ANCAs) that are demonstrated in a large proportion of patients. 46 Whed1er these are involved in pathogenesis or are epiphenomena is not known. High levels of serum IgE in children with infantile polyarteritis nodosa and KD have been interpreted as evidence of immune dysregulation in this disorder. 47 Levels of a wide variety of cytokines, including serum tumor necrosis factor-a (TNF-a) and interleukin-6 (IL-6),48 and growth
C HAP T E R
factors, such as vascular endothelial growth factor,49 are elevated, generally in proportion to the severity of the illness. As with many other vasculitides, a reasonable working hypothesis is that KD is a stereotyped, pathologic immune response to one or a variety of environmental or infectious triggers, to which certain individuals are predisposed by virtue of their genetic constitution. The predilection for childhood onset may reflect the presence of developmental antigens that are targets for the inflammatory response only early in life, subtle maturational defects in immune responsiveness,5o or the timing of exposure to environmental triggers.
GENmC BACKGROUND Studies from Japan indicate that approximately 1% of patients with KD have a family history of an affected sibling,5l and concordance for KD was 13.3% in dizygotic twins and 14.1 % in monozygotic twins. 52 There is a significantly increased frequency of the history of KD in the parents of children with the disease. 53 These observations indicate that there is a genetic predisposition to this disease, although the fact that affected twin pairs became ill within 2 weeks of each other also suggests an important role for an environmental agent. 52 The exact genetic factors that may underlie the disorder are not known. Candidate genes include those at the histocompatibility locus and those for TNF and other proteins involved in immunoregulation (e.g., polymorphism of an immune response gene, SLC11Al). Human leukocyte antigen (HLA) genes for B5, B44, Bw51, DR3, and DRB3*0301 have been associated with KD in whites; B54, Bw15, and Bw35 in Japanese; and Bw51 in Israelis. S4 There has been no reported association of any HLA antigen with the risk of coronary artery disease. 5s Similarly ambiguous are studies of the TNF-a gene (7NF). In white children with KD, there was increased frequency of the lymphotoxin-a + 250 AIA allele and the TNF-a-308 A/G high secretor allele. This TNF-a polymorphism was particularly increased in children with coronary artery abnormalities. 56 These differences were not observed in Japanese or Korean children,57 however, and their pathogenic significance remains uncertain. Yet to be confirmed are reports of a polymorphism of the promoter of the CD14 gene (CD14)'8 and genotype II of the angiotensin I converting enzyme59 that have been associated with the development of coronary artery lesions in KD.
2S K AWAS A KID I SEA S E
523
a useful framework for considering the natural history of the disease. With current therapy, the three-phase pattern of disease is considerably altered, and the first two phases are markedly abbreviated. After IVIG administration, the acute phase usually persists no more than 1 or 2 days, followed by another 3 to 4 weeks of the subacute phase.
Acute Febrile Phase KD characteristically begins abruptly, often preceded by symptoms of an upper respiratory or gastrointestinal illness. The child becomes febrile and is usually very irritable. Over the next 3 to 4 days, cervical adenitis, conjunctivitis, changes in the lips and oral mucosa, a pleomorphic rash, and erythema and edema in the hands and feet develop (in no particular order). Untreated, these manifestations persist for an average of 12 days and then subside. If carditis occurs, it often does so early and may be manifested by tachycardia, an S3 gallop, and subtle or occasionally marked signs of congestive heart failure. Abdominal pain and hydrops of the gallbladder may occur at this time.
Subacute Phase After the acute phase, the child may be entirely asymptomatic if given IVIG. Untreated, the fever gradually resolves by the third or fourth week. Sites of earlier involvement, such as the tongue, begin to heal during this period, and the child develops a so-called glossy tongue. Desquamation of the skin of the digits and perineum (which may begin earlier) may be the only clinically apparent residual feature of the illness. Some children develop arthritis of one or several joints during the late acute and subacute phases. Coronary artery aneurysms most commonly first develop during the subacute phase, occasionally earlier, but rarely later in children treated with IVIG.
Convalescent Phase Most children are asymptomatic during the convalescent phase. The acute phase response has usually returned to normal, unless there are complications. Horizontal ridging of the nails, known as Beau's lines and characteristic of many acute inflammatory conditions, may appear during this period.
CLINICAL MANIFESTAnONS
Disease Course The disease course can be divided into three phases (Fig. 25-1): 1. Acute febrile period of approximately 10 to 14 days 2. Subacute phase of approximately 2 to 4 weeks, ending with a return to normal of the platelet count and erythrocyte sedimentation rate 3. Convalescent or recovery period lasting months or years, during which time vessels affected by the disease undergo healing, remodeling, and scarring
There are no precise markers of the transition from one phase to another, but this concept of progression provides
Clinical Charaderlstics of the Classification CrIteria The diagnosis of KD is complicated by the fact that the signs and symptoms of the condition are common, and each of them is nonspecific. Although there is often uncertainty in diagnosing KD, the characteristic clinical manifestations of the disease help distinguish KD from its mimics, and the occurrence of several of these criteria in the same patient at the same time helps secure the diagnosis.
Fever Fever, often up to 40"C or higher, is the most consistent manifestation of KD. The fever is typically persistent and
524
C HAP T E R
25 K AWAS A KID I 5 E A 5 E • Figure Z5-1 Kawasaki disease can be viewed as an illness with acute, subacute, and recovery phases. the temporal characteristics outlined here are typical of the course of the disease. (Adapted from ref. 1)
Fever
Platelet Count Arthritis
CardioVascular Skin
Lips & Mucous Membrane Conjunctiva
Cervical Adenitis
Duration (weeks) minimally responsive to antipyretic agents, tending to remain above 38,5" C during most of the illness. It reflects elevated levels of pro-inflammatory cytokines, such as TNF-a and interleukin-l, which are also thought to mediate the underlying vascular inflammation. 60 The diagnosis of KD should be considered in all children with prolonged, unexplained fever, irritability, and laboratory signs of inflammation, especially in the presence of mucositis and rash. Conversely, the diagnosis must be suspect in the absence of fever, although the physician must be sure that elevated temperatures were not missed by anxious or inexperienced parents.
mimics of KD. Exceptionally, there may be a conjunctival exudate,62 conjunctival scarring,63 or changes in the retina and vitreous. 64
Changes in the Lips and Oral Mucosa Oral mucosal changes often become evident as the mucositis of KD evolves. Vertically cracked, red lips and a strawberry tongue are characteristic; the latter is caused by sloughing of filiform papillae and denuding of the inflamed glossal tissue (Fig. 25-2). Discrete oral lesions, such as vesicles or ulcers, and tonsillar exudate suggest a viral or bacterial infection rather than KD. 6
Conjunctivitis Bilateral, nonexudative bulbar conjunctivitis occurs in more than 85% of patients with KD. Conjunctival injection spares the limbus, the zone immediately around the iris. Inflammation of the palpebral conjunctiva is not prominent. Purulent discharge is especially unusual and suggests an alternative diagnosis. During the first week of illness, about three fourths of children are photophobic, an effect of anterior uveitis. 61 Slit-lamp examination may be helpful diagnostically in ambiguous situations; the presence of uveitis provides further evidence for the diagnosis because it is uncommon in
Exanthem The cutaneous manifestations of KD are protean. Although the rash usually begins on the trunk, there is often a perineal confluence during the first days of the illness, followed by desquamation in the diaper area by day 6 in most cases. Macular, morbilliform or targetoid lesions of the trunk and extremities are most characteristic. The rash is seldom pruritic, and vesicular or bullous lesions are rare. Psoriasis has been reported in several children with KD (Fig. 25-3).65 Pastia's lines, faint petechial lines in the skin folds of the antecubital fossae
C HAP T E R
2S KAWAS A KID I SEA S E
525
• Rgure ZS-Z A, The intense reddening, swelling, and vertical cracking of the lips are characteristic of Kawasaki disease (KD). Anonspecific facial rash is also seen in this 2-year-old boy with acute KD. B, Notice the strawbeny tongue of acute KD with hypertrophied papillae on an erythematous base and the peeling of the facial skin.
and inguinal and axillary regions, suggest the alternate diagnosis of scarlet fever rather than KD.
Lymphadenopathy Anterior cervical lymphadenopathy occurs during the acute phase of the disease, is usually unilateral, and may appear to involve only a single node. However, ultrasound or computed tomographic imaging of the neck typically reveals grapelike clusters of enlarged nodes similar to those seen in Epstein-Barr virus infections rather than the isolated adenopathy typical of bacterial adenitis. 66 Occasionally, a node enlarges rapidly and may be mistaken for bacterial infection. After 3 or 4 days, it usually shrinks with or without specific therapy. Diffuse lymphadenopathy and splenomegaly are not typical of KD and should raise suspicions of a viral illness.
Extremity Changes Indurated edema of the dorsum of the hands and feet and a diffuse red-purple erythema of the palms and soles
• FIgure ZS-3 The polymorphous exanthem of Kawasaki disease is shown in this photograph taken during the acute phase of the disease. Its appearance is not diagnostic, and its character may evolve as the disease progresses, although it is rarely purpuric, vesicular or bullous.
occur early and last for 1 to 3 days. Sheetlike desquamation typically occurs 10 days or more after the start of the fever and begins at the tips of the fingers and, less commonly, the toes just below the distal edge of the nails (Fig. 25-4). Flaky desquamation may occur in the perineum or elsewhere. Because skin peeling occurs late, it is useful more for retrospective confirmation of the diagnosis than for making therapeutic decisions.
Atypical Kawasaki DIsease Children suspected of haVing KD who do not fulfill diagnostic criteria may have incomplete or atypical disease. Early reports have suggested a grim prognosis for children with atypical KD; one review cited a 41% mortality rate, although only children with coronary artery aneurysms were included in this series. 67 However, when clinical judgment of reliable observers is used to define atypical KD, signs, symptoms, and outcome parallel those of children who fulfill the diagnostic criteria. In a study of 242 patients hospitalized for KD in Japan during a 9-year period, 25 00%) ultimately failed to meet diagnostic criteria. 68 Three criteria were met in 17 (680/0) of the 25 patients, and 7 (28%) met two criteria. Only one patient ultimately developed transient dilatation of a coronary artery. A particularly high level of suspicion is needed in infants younger than 1 year old. In a retrospective review of 45 cases of KD, 5 (45%) of 11 infants had atypical disease, compared with 4 02%) of 33 older children.69 Unfortunately, infants are the group at the highest risk for developing coronary artery aneurysms, and in this study, coronary artery complications occurred in seven infants (64%), compared with three older children (9%), including all five infants with atypical disease. 69 Overall, among the 2221 children younger than 5 years who were analyzed in the 1995 to 1996 Japanese nationwide survey of KD, the odds ratio for development of cardiac sequelae in infants younger than 1 year was 1.54. 70 A retrospective survey reported that 8.5% of patients younger than 12 months developed coronary artery abnormalities, compared with 1.8% of those 12 months old or older. 71
526
C HAP T E R
25
KAWASAKI DISEASE
• Figure 25-4 A, Edema of the hand and early peeling of the skin begins around the nail margins during the subacute phase of Kawasaki disease. B, Desquamation of the skin of the hand occurs later in the subacute and early recovery phase of the disease. In many children. the degree of desquamation is much less than is depicted here.
Other Clinical Manifestations of Kawasaki Disease In addition to the manifestations encompassed in the classification criteria, a number of other abnormalities occur with various frequencies (Table 25-2).
Cardiovascular Disease The most significant and characteristic complications of KD involve the cardiovascular system. Up to 25% of untreated patients develop coronary artery aneurysms, making KD the leading cause of acquired heart disease among children in the developed world (Figs. 25-5 and 25-6). Giant coronary artery aneurysms, with an internal diameter larger than 8 mm, are associated with the highest risk of morbidity and mortality. Up to one third of such aneurysms become obstructed, leading to myocardial infarction, arrhythmias, or sudden death. 72 Treatment with IVIG decreases the incidence of giant aneurysms by
If. II
TABLE 25 Z
more than 95%, and the overall incidence of aneurysms by 85%. Coronary aneurysms may cause morbidity early in the course due to rupture or thrombosis, resulting in sudden death or myocardial infarction,73 Development of de novo coronary artery abnormalities more than 2 weeks after the acute illness is extremely unusual, although ongoing vascular scarring of existing lesions may result in progressive coronary insufficiency. Approximately one half of coronary artery aneurysms demonstrated by echocardiogram ultimately resolve, usually those under 6 mm in diameter,74 but persistent vasodilatory abnormalities have been observed in arteries where aneurysms have resolved. 75 At onset, there is usually a tachycardia commensurate with the degree of fever. Early myocarditis occurs in at least one half of patients76 and is characterized by arrhythmias and signs of congestive heart failure. Pericarditis may also occur. Depressed myocardial contractility, occasionally progressing to congestive heart failure, can occur during the acute illness. 77 Clinically,
M,lI1ifesl"liolls of K"wasilki Disease
FInding Organ System
Common
Uncommon
Suggests Alternate Diagnosis
Skin
Targetoid, urticarial, morbilliform rashes, livedo reticularis Pleural effusion Urethritis, pyuria Irritability, lethargy, anterior uveitis, sensorineural hearing loss Diarrhea, vomiting, hydrops of gallbladder, hepatomegaly Anemia, thrombocytosis, leukocytosis
Psoriasiform rash
Pustular, vesicular rashes
Lungs Urinary tract Nervous system Gastrointestinal system Hematologic system Reticuloendothelial system Mucosa
Anterior cervical lymphadenopathy
Musculoskeletal system Cardiac system
Extremity edema, arthritis Tachycardia, gallop rhythm, myocarditis, pericarditis
Nodules, interstitial infiltrates Hematuria, proteinuria, orchitis Seizure, stroke, cranial nerve palsy Intestinal hemorrhage, mptured viscus Thrombocytopenia, consumptive coagulopathy, hemophagocytic syndrome Posterior cervical, axillary lymphadenopathy
Mucositis, glossitis, conjunctivitis
'Except during the convalescent phase.
Raynaud's phenomenon Coronary artery aneurysm, aortic root dilatation, valvulitis
Lymphocytosis' Diffuse lymphadenopathy, splenomegaly Discrete oral lesions, exudative conjunctivitis
C HAP T E R
25 K AWAS A KID I SEA S E
527
• fItue Z5-5 Echocardiographic demonstration of aneurysms of three coronary arteries in a child with Kawasaki disease. A, aneurysms; CIRC, drcumflex; LAD, left anterior descending coronary artery; RVOT, right ventricular outflow tract. (Courtesy of Dr. Dennis Crowley.)
this is often manifested by an 53 gallop that may become more prominent with hydration but that typically resolves after treatment with IVIG. Long-term abnormalities of cardiac contractility are very uncommon in children treated during the acute phase of KD. 78 Although involvement of the coronary arteries is the most characteristic manifestation of the vasculitis of KD, other medium-sized muscular arteries may be involved. Aneurysms of brachial and femoral arteries may be palpable clinically or demonstrable angiographically (Fig. 25-7). In severe cases, peripheral arterial obstmction may lead to ischemia and gangrene. This vasculitis usually spares visceral arteries, although there are reports of gastrointestinal obstmction79 and acute abdominal
catastrophe80 occurring as a result of vasculitis. This complication generally accompanies other manifestations of critical disease, such as giant coronary artery aneurysms and aneurysms in peripheral arteries.
Central Nervous System Complications One of the most consistent clinical observations of children with KD, particularly infants and very young children, is their extreme irritability. This probably represents the effect of aseptic meningitis and associated headache. 8! Numerous other central nervous system complications have been reported, including cerebrovascular accident82 and facial nerve paralysis. 83
A, Angiography of the coronary vessels in a 7-month-old boy with Kawasaki disease shows a huge aneurysmal dilatation of the right coronary artery (annw). 8, Aneurysm of the left coronary artery in a 3-year-old girl with Kawasaki disease (annw). (A and 8, Courtesy of Dr. Zuidi Lababidi.) • figure Z5-6
528
C HAP T E R
25 K AWAS A KID I SEA S E
• Figure Z5-7 Angiographic study of a 2-year-old boy with severe Kawasaki disease resulting in multiple aneurysms of the coronary, axillary, iliac, and femoral arteries.The study revealed large aneurysms of the aorta and iliac (A) and femoral (B) arteries (arrows). Aneurysms that were palpable in the axilla and groin in this patient later resolved. (A and B, Courtesy of Dr. G. Culham.)
Ocular Disease Although conjunctivitis is the most characteristic ocular abnormality in KD, additional abnormalities are common,H.j Asymptomatic uveitis occurs in approximately three fourths of children. It is more common in those older than 2 years. It is bilateral and begins a little later than the conjunctivitis, peaking between days 5 and 8 of illness. 6t Posterior synechiae are rare,HS and the signs listed in Table 25-3 usually disappear without visually significant sequelae and require no specific therapy.
Musculoskeletal Disease
TABLE 25::1 frequency of (l
Ocular SlgR or Symptom Injection of bulbar conjunctivae Nongranulomatous iridocyclitis Superficial punctate keratitis Vitreous opacities Papilledema Subconjunctival hemorrhage
RespiratoryTract Disease Cough, coryza, or hoarseness frequently occur early in the course of the disease and suggest a viral upper respiratory tract infection. These conditions may be accompanied by otitis media. 6 The basis of a persistent sensorineural hearing loss that occurs in some children after KD is unkown. H7
Gastrointestinal Tract Disease and Other Abnormalities
Arthralgia and myalgia are quite common, and approximately one-fourth of patients have arthritis, most commonly affecting the knees and ankles. 86 It may occur at any time during the disease course but has been described most commonly during the recovery phase.
~.
Treatment with IVIG does not appear to prevent this manifestation of the disease.
Frequency (%)
89 78 22 12 11
Abdominal pain is common, and approximately one fourth of children with KD have profuse, watery diarrhea during the acute febrile period. Abdominal distention may occur and may mimic mesenteric vasculitis or intussusception. The relatively common occurrence of hydrops of the gallbladder demonstrated by ultrasonographyHH sometimes aids in the diagnosis of incomplete or atypical KD. The specificity of this finding is limited, however, and a dilated, engorged gallbladder may be seen in cases of streptococcal and staphylococcal infections, among other mimics of KD. Occasionally, the gallbladder becomes large enough to be seen as a bulge in the anterior abdominal wall. Hepatosplenomegaly may occur in the absence of heart disease or may reflect cardiac failure.
3
Genitourinary Tract Involvement From Kumagai N, Ohno S: Kawasaki disease. In Pepose JS, Holland GM, Wilhelmus KR (eds): Ocular Immunity and Infection. SI. Louis, Mosby, 1996. pp 391-396.
Scrotal pain and swelling due to testicular inflammation are characteristic of pediatric vasculitides, including
C HAP T E R
Henoch-Schonlein purpura, polyarteritis nodosa, and KD. Meatitis and dysuria also occur frequently during the acute phase of KD, and priapism has been described. 89 Hemolytic-uremic syndrome, immune complex-mediated glomerulonephritis, and acute interstitial nephritis have each been reported in a few cases. 90 ,91 Acute renal failure is a rare complication that appears to be caused by different factors in different patients, although it most commonly can be ascribed to complications of treatment with certain preparations of MG.92
DIFFERENTIAL DIAGNOSIS KD is often difficult to differentiate from viral exanthems of childhood, particularly early in the disease course or in children with incomplete or atypical KD (Table 25-4),93 The differential diagnosis also includes poststreptococcal scarlet fever, toxic shock syndrome, drug reactions, and systemic-onset juvenile idiopathic arthritis. Viral illnesses such as measles (especially when atypical or occurring after vaccination), Epstein-Barr virus, and adenovirus infections share many of the signs of mucocutaneous involvement, but they typically have less evidence of systemic inflammation and generally lack the extremity changes of KD. Toxin-mediated illnesses, especially scarlet fever and toxic shock syndrome, lack the ocular and articular involvement typical of KD, Drug reactions, such as those in Stevens-Johnson syndrome or serum sickness, may mimic KD but have subtle differences in the ocular and mucosal manifestations. In particularly severe or prolonged KD, the possibility of a chronic vasculitis such as polyarteritis nodosa must be considered carefully.
PATHOLOGY The basic pathologic lesion is a systemic necrotiZing vasculitis with fibrinoid necrosis of the medium-sized muscular arteries; the coronary arteries are the predominant
I:: •
fABlE 25-4
Differential Diagnosis of Kawasaki Disease
Infedlous Conditions Adenovirus Measles Parvovirus Human herpesviruses (HHV) (e.g., herpes simplex vinls, cytomegalovinls, HHV-6, HHV-7) Rocky Mountain spotted fever Leptospirosis Streptococci Staphylococci
Immlllle Reactions Stevens-Johnson syndrome Serum sickness
Rheumatic Diseases Systemic-onset juvenile idiopathic arthritis Polyarteritis nodosa
25
KAWASAKI DISEASE
529
sites of involvement,94 An early neutrophilic infiltrate occurs in all layers of the heart, including the valves. Pathologic examination of autopsy specimens demonstrates inflammation beginning in the microvasculature (i.e., arterioles, capillaries, vasa vasorum, and venules) and subsequently spreading to larger vessels, especially the coronary arteries. 95 In these lesions, infiltrating cells are mostly macrophages and IgA-secreting plasma cells,96 findings that may be unique to KD.97 As with other vasculitides, vessel damage appears to result from an aberrant immune response leading to endothelial cell injury. Endothelial cells express a variety of markers of activation, presumably as a result of the high levels of proinflammatory cytokines that characterize the acute phase of disease. 98 Some children have a lymphocytic myocarditis, with cellular infiltrates or myofibrosis on endomyocardial biopsy that may persist for years in cases of untreated KD,99 Evolution of the cardiac lesions is detailed in the snldy of Fujiwara and Hamashima. lOo Coronary artery vasculitis predominated early in the disease but was absent in those who died after 28 days of illness. Aneurysms, thrombosis, and stenosis did not appear until 12 days of disease or later. Pericarditis, myocarditiS, and endocarditis were universal findings early in the disease but diminished as fibrosis of the myocardium became the predominant lesion in children whose death occurred 40 days or more after onset. In a study of 262 children, Suzuki and colleagues 101 documented an equal frequency of aneurysms in right and left coronary arteries and a higher frequency of segmental stenosis and occlusions in the right coronary artery,
LABORATORY EXAMINATION There are no specific diagnostic tests for KD, but at onset, evidence of inflammation is manifested by elevation of acute phase reactants (e.g., C-reactive protein [CRP), erythrocyte sedimentation rate, Ill-antitrypsin), leukocytosiS, and a left shift in the white blood cell differential count. Occasionally, significant neutropenia occurs early,102 possibly indicating the presence of particularly severe disease. Toxic granulation of neutrophils is more frequent in children with KD than in those with other febrile illnesses. 103 Although platelet counts may be abnormally low at disease onset, by the second week of illness, they characteristically rise and may reach 1,OOO,OOO/mm3 (reactive thrombocytosis) in the most severe cases. Children with KD often present with a normocytic, normochromic anemia; hemoglobin concentrations greater than two standard deviations below the mean for age are found in one half of patients within the first 2 weeks of illness. 6 Microscopic examination of urine commonly reveals white blood cells, although urine culture results are negative. The pyuria is of urethral origin and therefore is missed on urinalyses obtained by bladder aspiration or catheterization. The white blood cells are mononuclear and are not detected by dipstick tests for leukocyte esterase. Measurement of liver enzymes often documents elevated transaminase levels or mild hyperbilirubinemia due to intrahepatic congestion. A few children develop obstructive jaundice from hydrops of the gallbladder.
530
C HAP T E R
25 K AWAS A KID I SEA S E
Cerebrospinal fluid (CSF) analysis typically displays a mononuclear pleocytosis without hypoglycorrhachia or elevation of CSF protein. In a chart review of 46 children with KD, 39% were documented to have elevated CSF white blood cell counts. SI The median count was 22.5 cells/mm3 with 6% neutrophils and 91.5% mononuclear cells, although cell counts as high as 320/mm3 with up to 79% neutrophils were reported. Arthrocentesis of involved joints typically demonstrates 50 to 300,000 white blood cells/mm3 , consisting primarily of neutrophils. Children with KD develop significant perturbations in serum lipid profiles beginning dUring the subacute phase of illness, including elevated concentrations of triglycerides and low-density lipoproteins and depressed levels of high-density lipoproteins. l04 These abnormalities are probably caused by the widespread endothelial injury. As with other sequelae of KD, normalization may take years in untreated children but typically occurs within weeks or months after IVIG therapy. Antibodies to neutrophil cytoplasmic antigens (ANCAs)105 and to endothelial cells (AECAs)l06 may be present late but not early in the disease. 107 Consequently, they are of little diagnostic value. Other autoantibodies are usually absent. Elevated levels of von Willebrand factor antigen indicate the presence of damaged endothelium. lOs Activation products of C3 and C4 have been demonstrated on erythrocytes (C3g) and in the plasma (C4d),109 suggesting the participation of complement in at least some of the manifestations of the disease.
TREATMENT General Approach The child with suspected or definite KD should be admitted to the hospital for observation, monitoring of cardiac status, and management of systemic manifestations (Table 25-5). Initial evaluation of the heart should include an electrocardiogram to identify arrhythmias, signs of ischemia, or myocarditiS and a baseline echocardiogram to detect coronary artery vasculitis, ectasia, or aneurysms. If the diagnosis is relatively certain (even if diagnostic criteria are not met), and other diagnoses have been considered and excluded, treatment should be initiated with aspirin and IVIG (Fig. 25-8),
Goals of Therapy Recommendations for the management of KD are predicated on the goals of therapy and must reflect the natural history of the disease, the effectiveness of therapy, and the costs, risks, and benefits. In addition to control of the acute inflammation and its symptoms, the goal of therapy is to prevent long-term sequelae and, most importantly, coronary artery abnormalities. The consequences of failure to appropriately treat a child with KD are so important that, within reason, after very careful evaluation, error on the side of premature or unnecessary therapy is preferable to delayed or missed therapy for a child for whom the diagnosis is uncertain.
II: ~
Inilial Evaluation and Management of Kawasaki Db-ease
TABLE 25-5
Make Diagnosis Admit to Hospital Evaluate
Treat
Cardiac starus (ECHO, ECGl CNS status Fluid and electrolyte status Urinalysis
Aspirin If patient is febrile: 80-100 mglkglday in 4 doses If patient is afebrile: 3-5 mglkglday in 1 to 4 doses IVlG: 2 g/kg Keep in hospital until afebrile for 24 hr or if there are complications. Repeat IVlG once. If no clinical response, consider intravenous methylprednisolone: 30 mg/kg Maintain low-dose aspirin until ESR and platelet count are normal if there have been no coronary artery abnormalities; for 2 years, if coronary abnormalities have been but are no longer present; "forever" if coronary artery disease persists.
Ophthalmologic status Monitor cardiac status Monitor ESR and platelet count at 2-week intervals until stable, then I-month intervals until normal Repeat echocardiogram at 6--8 weeks
CNS, central nervous system; ECG. electrocardiogram: ECHO, echocardiogram: ESR. erythrocyte sedimentation rate: IVIG, intravenous immunoglobulin.
The American Academy of Pediatrics and the American Heart Association recommend that children with KD should be treated with aspirin and IVIG during the first 10 days of the illness. I J() Subsequent management remains controversial, however, and depends on the presence or absence of coronary artery abnormalities. The Japanese Ministry of Health criteria,JIl,lI2 published in 1984, use angiography or echocardiography to define coronary arteries as abnormal if the internal lumen diameter is greater than 3 mm in children younger than 5 years or greater than 4 mm in children at least 5 years old; the internal diameter of a segment measures at least 1.5 times that of an adjacent segment; or the coronary artery lumen is clearly irregular. Although coronary artery dimensions in normal children have been shown to increase linearly with body size, as measured by body surface area (BSA) or length,ll3 these criteria are not based on body size. Evaluation of coronary arteries in KD using age-, size-, and sex-adjusted indices suggests that the incidence of abnormalities is higher than was generally recognized. Using mean BSA-adjusted criteria, dimensions of the proximal left anterior descending and right coronary arteries were significantly larger (P < .01) in patients with KD than in normal subjects. lJ4 Among patients classified as having normal coronary arteries on all echocardiograms by the Japanese Ministry of Health criteria, 27% had at least one BSA-adjusted coronary artery dimension more than two standard deviations above the mean. Thus coronary artery dilation appears to be more prevalent than previously reported, highlighting the need for systematic longitudinal surveillance of this population. Treatment strategies also depend on the implications of coronary artery dilatation. Long-term outcome studies
C HAP T E R
are somewhat reassuring. Fifty percent of coronary artery aneurysms regress angiographically, and among such children, there was no increase in morbidity and mortality rates, even after more than 2 decades. 72 Nonetheless, abnormalities of more subtle markers of endothelial health are a matter of concern. Vessels show histologic 1l5 and functional 1l6 abnormalities at the sites of healed aneurysms, and vascular reactivity to endogenous vasodilators is abnormal in children who have had KD, regardless of whether they have detectable coronary artery abnormalities. 1l7 This lends credence to the report of an increased standardized mortality ratio of 2.35 among male patients with cardiac sequelae. 1I8 Acute phase reactants and platelet counts do not return to normal for up to 2 months after apparently successful treatment, suggesting that vasculitis and endothelial inflammation may not fully resolve even when fever is controlled. It is reasonable to ask whether persistent KD requires additional therapy and whether initial treatment should be more robust than IVIG alone, aiming for anatomically and functionally normal vessels.
Aspirin Aspirin was the first medication to be used for treatment of KD because of its anti-inflammatory and antithrombotic effects. 119 Anti-inflammatory regimens using high-close (>80 mg/kg/day) or lower-dose (30 mg/kg/day) aspirin have been recommended during the acute phase of the illness. After the fever resolves, the dose is usually reduced to an antiplatelet dose of 3 to 5 mg/kg/day. These doses, well below the anti-inflammatory level, have the effect of inhibiting platelet adhesion to endothelium by curtailing platelet release of thromboxane A2 without suppressing prostacyclin production by endothelial cells. 120 This effect is believed to be beneficial in preventing thrombosis when platelet counts are elevated, although no studies have demonstrated such a benefit clinically. In the event of aspirin sensitivity, another antiplatelet agent, such as dipyridamole, should be considered. Unless coronary artery abnormalities are detected by echocardiogram, aspirin is discontinued after results of laboratory studies return to normal, usually within 2 months of the onset. There have been no published comparisons of aspirin with other anti-inflammatory agents, and it is unclear whether salicylates are uniquely efficacious in this condition. A meta-analysis found that high-dose and lowerdose aspirin regimens were associated with a similar incidence of coronary artery abnormalities at 30 and 60 days after disease onset. 121 Although the necessity of using high-dose aspirin might be questioned because of the rapid response to MG, all of the trials showing the benefit of IVIG were conducted with children who also were receiving anti-inflammatory doses of aspirin. For other effects, however, such as treatment of prolonged arthritis, alternative anti-inflammatory agents, such as ibuprofen, may be used. The risks of aspirin appear to be similar to those reported in other settings: chemical hepatitis, transient hearing loss, and rarely, Reye's syndrome. These risks
25
KAWASAKI DISEASE
531
may be increased in KD. Aspirin binding studies have suggested that the hypoalbuminemia of children with KD predisposes them to toxic levels of free salicylate, despite measured (bound) values within the therapeutic range. 122 At least one case of Reye's syndrome has been reported after 6 days of aspirin therapy for KD.123 In view of the potential risks and lack of obvious benefits of high-dose aspirin regimens, the dose should be reduced after acute inflammation has subsided and antiplatelet effects are required. Physicians should not disregard the risk of Reye's syndrome, and aspirin should be rapidly discontinued if there is any concern about varicella or influenza. Persistent fever usually indicates ongoing vasculitis and should prompt consideration of retreatment with MG or salvage therapy, as discussed later (see Fig. 25-8).
Intravenous Immunoglobulin Before 1984, approximately 20% of children with KD developed coronary artery aneurysms, and in the absence of effective therapy, up to 1.5% of them succumbed to the disease. Aspirin was used to relieve fever and musculoskeletal symptoms but had no benefit in preventing or treating coronary artery involvement. As with many important medical advances, discovery of the efficacy of IVIG in treating KD was fortuitous. In the early 1980s, MG was reported to be useful for the treatment of idiopathic thrombocytopenic purpura. It was reasoned that KD might also involve platelet abnormalities (because thrombocytosis is a hallmark of the disease), and in 1984, Furusho and coworkers 124 reported that high-dose IVIG appeared to decrease the incidence of coronary artery abnormalities. To verify these findings, Newburger and colleagues125 brought together six large pediatric centers in North America in a 19-month-long, randomized, controlled clinical trial. One hundred sixtyeight children with KD were enrolled in one of two arms. One half received IVIG (400 mg/kg/day on 4 consecutive days) plus high-dose aspirin (100 mg/kg/day), and one half received aspirin alone. IVIG reduced the incidence of coronary artery abnormalities by 78%, and no child suffered serious adverse effects from the therapy, confirming the remarkable therapeutic potential of MG. The initial IVIG treatment regimen was based on thencurrent protocols for treating immune thrombocytopenic purpura. The question of whether this protocol was optimal for KD was addressed in 1991. 126 Children were randomized to receive the traditional four-dose regimen or a single dose of 2.0 g/kg of IVIG infused over 8 to 12 hours. Children receiving the larger, single dose fared better, and evidence of a dose-response pattern for MG suggested that further modifications of this treatment regimen might lead to additional improvements in outcome. Meta-analyses have documented the additional benefit of larger doses of IVIG.127 This apparent dose-response effect forms the theoretical basis for the current practice of IVIG retreatment of patients who have persistent or recrudescent fever after initial IVIG therapy. In these circumstances, retreatment with 2 g/kg of MG is recommended if other explanations for persistent fever have been excluded. 110
532
C HAP T E R
2S K AWAS A KID IS E A S E • Figure 25-8 Management algorithm for Kawasaki disease. KD, Kawasaki disease; IVIG, intravenous immunoglobulin; CRP, C reactive protein.
Child fulfills criteria for KD, or has incomplete manifestations but is <1 year of age, or has had more than B days of fever without an alternate explanation
IVIG 2 g/kg over 8-12 hours plus aspirin 20-25 mg/kg q 6h Obtain echocardiogram
~
Fever persists and no alternative explanation found
Defervesces
Repeat IVIG 2 g/kg over 8-12 hours Resume high dose aspirin
1
Fever persists and no alternative explanation found
V
Decrease aspirin to 3-5 mg/kg once daily until all lab stUdies normal (usually 4-8 weeks) Defervesces
r--------""1'-----
Intravenous methylprednisolone 30 mg/kg (maximum 1.5 g) on three consecutive days Fever persists and/or CRP rises and/or aneurysms progress Consult referral center; consider additional dose of IVIG. additional steroids, or salvage therapy with plasmapheresis or immunosuppression
Although standard therapy with MG and aspirin given within the first 10 days of illness greatly reduces the risk of coronary artery involvement, approximately 5% of children still develop coronary artery aneurysms according to Japanese Ministry of Health criteria, and a larger number demonstrate coronary artery ectasia. Many researchers have attempted to identify the children at highest risk for developing coronary artery abnormalities to stratify the intensity of tl1erapy. In general, younger patients, especially infants younger than 6 months, are at higher risk. l28 Stockheim and colleagues l29 reported that older children also were at additional risk. In their retrospective series, 21% of patients older than 8 years had coronary artery abnormalities. They attributed this increased incidence to a delay in diagnosis and treatment among older children in whom KD is not usually seen, although an age-related difference in the disease cannot be excluded. In a retrospective series from Japan, Fukunishi and colleagues 130 found higher serum levels of CRP, lactate dehydrogenase, and bilirubin to be predictive of failure to respond to MG. In a Canadian study, Han and colleagues 131 could not identify any difference in laboratory parameters between responders and nonresponders. According to Mori and coworkers,132 a rise in the white
blood cell count and CRP level after MG infusion are independent predictors of coronary artery abnormalities. lVlG is most effective in reducing the risk of coronary artery disease when administered within 10 days of the onset of fever. Unfortunately, the diagnosis may remain in doubt as this deadline approaches. In general, when a reasonable alternative explanation for prolonged fever is not apparent, many pediatricians elect to treat with IVlG, preferring the possibility of unnecessary use of medication to that of leaving the child with possible KD untreated. In ambiguous cases, the physician is best guided by the epidemiology of the disease. More than 50% of infants with KD present atypically, and they have a very high incidence of aneurysms. Empiric treatment in very young children is often warranted. Coronary artery abnormalities are only one expression of the Widespread vasculitis of Kawasaki disease, and IVlG appears to benefit these other manifestations as well. The lipid abnormalities seen early in the course normalize after treatment and disease control. 133 Similarly, echocardiographic data suggest that another common manifestation, depressed myocardial contractility, may be more rapidly reversed by lVlG.
C HAP T E R
The mechanism of action of IVIG is uncertain, with studies adding induction of neutrophil apoptosis134 and reversal of inhibited lymphocyte apoptosis 135 to a long list of immunomodulatory effects of IVIG (Table 25-6), Regardless, the response is generally prompt, and temperature returns to normal in most children even before the end of the infusion, with rapid clearing of the rash, mucositis, and conjunctivitis. Irritability and emotional lability, however, may persist for up to several weeks before resolving. IVIG is generally well tolerated, although headache up to 72 hours after the infusion is not uncommon and may require low-dose opiates for relief. 1.~6 Despite its advantages, MG is an expensive and potentially toxic inteIVention. The greatest long-term issue is potential transmission of blood-borne pathogens. Elaborate sterilization procedures, including lyophilization, pasteurization, and addition of solvent detergents, are generally effective in rendering the product free of lipid-soluble viruses. Nonetheless, apparent technical deficiencies led to more than 100 cases of hepatitis C in recipients of a single brand of MG in 1994, although none was a child with KD.137 Overall, however, significant toxicity is rare, and benefits clearly outweigh risks for children with confirmed KD. No cases of MG-transmitted infections have been reported since the institution of current purification and processing practices in 1995, and no cases of MG-transmitted HIV have ever been reported. Cost-benefit analysis documents that MG treatment of KD is one of the most cost-effective medical therapies available, leading to short- and long-term savings. 138
All recommendations concerning the role of IVIG are limited to treatment during the first 10 days of illness. This is not to say that treatment after 10 days of illness is ineffective or contraindicated; it is merely inadequately studied. In a report of 16 children with coronary artery aneurysms treated a mean of 17 days after onset of fever, there was a trend toward increased resolution of abnormalities by echocardiogram. 139 The American Academy of Pediatrics cautiously recommends IVIG for children beyond the 10th day of illness with "manifestations of continuing inflammation," and such an approach appears pru-
,~.
lABLE 25-6 Potential Effects 01 Intravenous Immunoglobulin Kawasaki Disease
III
Spedtlc Effects • Provides antibodies against infectious agent • Provides antibodies against circulating toxin • Provides anti-idiotypic antibodies
NonspedfIc Effects • • • • • • • • • • •
Blockades Fc receptors Accelerates clearance of activated complement fragments Alters solubility characteristics of circulating immune complexes Decreases soluble adhesion molecules (e.g., E-selectin, ICAM-I) Upregulates activity of natural killer cells Reverses immunoregulatory abnormalities by increasing suppressor T cells and decreasing helper T cells and circulating B cells Downregulates transcription of cytokine genes Neutralizes activity of pro-inflammatory cytokines Causes feedback inhibition of autoantibody synthesis Reverses inhibited lymphocyte apoptosis Induces neutrophil apoptosis
2S
KAWASAKI DISEASE
533
dent. 110 Questions have arisen concerning the efficacy of early treatment of KD. One group reported an increased failure rate, but Tse and colleagues l4o reported that IVIG given on or before the fifth day of illness resulted in fewer coronary artery abnormalities at the I-year follow-up assessment.
Glucocorticoids A subgroup of patients with KD is resistant to IVIG therapy; these children are at greatest risk for development of coronary artery aneurysms and long-term sequelae of the disease. No effective treatment for these patients with refractory disease has been established. Glucocorticoids, the treatment of choice in other forms of vasculitis and therefore a logical candidate for treating such children, have been considered unsafe in KD. This conclusion was based primarily on the study of Kato and coworkers, 141 which demonstrated an extraordinarily high incidence of coronary artery aneurysms (11 of 17 patients) in a group that received oral prednisolone at a dose of 2 to 3 mg/kg/day for at least 2 weeks, followed by 1.5 mg/kg/day for an additional 2 weeks. Because subjects were not stratified according to risk factors for the development of aneurysms and no information about methods of treatment group allocation was given, these data are difficult to interpret. A smaller group of seven patients in the same study received prednisolone plus aspirin, and none of these patients developed aneurysms. Studies support a role for glucocorticoids. In a randomized trial enrolling 100 children treated with intravenous prednisolone (followed by an oral taper) or low-dose IVIG (300 mg/kg/day for 3 consecutive days), Nonaka and colleagues 142 reported a shorter duration of fever in the prednisolone-treated group and no significant difference in the prevalence of coronary aneurysms. Shinohara and colleagues l43 reviewed the experience of almost 300 patients with acute KD seen between 1982 and 1998 who were treated before the 10th day of illness and had coronary lumen diameters of less than 4 mm. Prednisolone therapy was associated with a significantly shorter duration of fever and a lower prevalence of coronary artery aneurysms. No adverse reactions were recorded for any therapy. This study is limited by the fact that medications were administered in various combinations dUring routine care, without a prospective protocol. Kato's groupl44 also found a beneficial effect of glucocorticoids in KD. Children who had failed to respond to two doses of IVIG were randomized to receive a third dose of IVIG or pulsed methylprednisolone. Patients who received methylprednisolone had a significantly shorter duration of fever, and although transient coronary artery dilatation was associated with glucocorticoid therapy, there was no overall difference in the incidence of coronary artery abnormalities between groups. A question has been raised about whether the addition of steroids at the time of diagnosis might decrease the rate of nonresponse and benefit even children apparently "cured" by IVIG but in whom subtle measures of endothelial health remained abnormal. Data from one study 145 suggests that such an approach may be preferable. Subjects were randomized to receive IVIG at a
534
C HAP T E R
25 K AWAS A KID I SEA S E
dosage of 2 g/kg given over 10 hours, with or without intravenous methylprednisolone at a dosage of 30 mg/kg (maximum, 1.5 g) over 3 hourS. 145 All patients received aspirin at a dosage of 20 to 25 mg/kg every 6 hours until afebrile for 48 hours and then received 3 to 5 mg/kg/day. Treatment of patients with acute KD with intravenous methylprednisolone plus IVIG resulted in faster resolution of fever, more rapid improvement in markers of inflammation, and shorter length of hospitalization than for those receiving IVIG alone. Adverse effects occurred infrequently. Similar results were reported in a study using dexamethasone with IVIG. 146 Neither study had sufficient statistical power to detect the influence of glucocorticoid therapy on coronary artery outcomes. Additional trials are being conducted to address this issue. Corticosteroids are the preferred therapy for children with persistent KD despite at least two doses of MG (total of at least 4 g/kg), and they also may have a role in the initial treatment of KD.
Other Therapeutic Approaches Many therapies that are effective in other forms of vasculitis have been used in KD. Pentoxifylline is used for treatment of peripheral vascular disease, largely because of its effects on erythrocyte flexibility and blood viscosity. It is also an effective vasodilator and inhibitor of platelet aggregation and neutrophil activation, leading to trials in treating KD. The largest study reported compared children treated with IVIG and aspirin alone with those for whom low-dose 00 mg/kg/day) or high-dose (20 mg/kg/day) pentoxifylline was added l47 Coronary artery aneurysms were detected in 110/0 and in 14% of two groups of children who received IVIG alone or in conjunction with low-dose pentoxifylline, respectively, but were not observed in those who received combination IVIG plus high-dose pentoxifylline therapy. No adverse effects were observed in 79 patients. Further studies are needed to determine whether pentoxifylline has a role in the treatment of KD. One of the effects of pentoxifylline is inhibition of TNF, and anti-TNF agents such as etanercept or infliximab also may be beneficial. Case reports indicate that infliximab may have a role in patients with KD who have not responded to IVIG and corticosteroids. 148 More definitive studies are planned. A dramatic response to plasmapheresis has been reported,149 but the technical limitations and potential hazards of this therapy are considerable, and it should be reserved for children with active inflammation who have failed all available medical interventions, including multiple doses of IVIG, intravenous methylprednisolone, and TNF inhibition. The role for immunosuppressive agents in KD, such as cyclophosphamidel 50 or cyclosporine,151 is extremely limited. Their onsets of action typically are delayed for days or weeks, whereas even the most severe cases of KD are usually recovering by that time. If presumed KD is prolonged beyond 3 or 4 weeks, consideration should be given to an alternative diagnosis, including chronic vasculitides such as polyarteritis nodosa. 152 There have been conflicting reports of the efficacy of abciximab, a monoclonal antibody that inhibits platelet glycoprotein lIb/IlIa receptor. In one report,153 there was an increased reso-
lution of aneurysms in patients with KD who received abciximab compared with those who received conventional treatment. However, a second studyl54 could not duplicate these findings. Many other agents, including antioxidants,155 elastase inhibitors,l56 immunosuppressants. and monoclonal antibodies, are being evaluated. Ultimately, with more precise genetic characterization of children who develop KD and a better understanding of the factors that affect outcomes in this disease, it may be possible to tailor therapy to an individual child's needs. As effective as IVIG may be, until the cause of KD is understood and preventive strategies can be implemented, the search for better treatments is not likely to cease.
TREATMENT OF RELAPSES Fever returns within 48 hours of treatment with MG in approximately 100/0 of children, indicating failure to suppress the underlying inflammatory process. Because earlier studies 125 suggested that peak IgG levels correlate with response and because prolonged fever is an independent risk factor for the development of coronary artery aneurysms, these children should be retreated with a second dose of IVIG (2 g/kg). Those who fail to respond to a second dose-up to a third of patients in some studies71-usually respond to intravenous methylprednisolone at a dosage of 30 mg/kg/day for 1 to 3 daysyl
FOLLOW-UP AND MONITORING OF CARDIAC STATUS There is no universal agreement about the timing and frequency of echocardiographic monitoring of patients with KD. Most protocols are somewhat arbitrary but account for the development of coronary artery aneurysms, which occur most frequently between the second and the eighth weeks after the onset of fever. Aneurysms can be present in the first week of the illness but rarely occur de novo after the eighth week. The recommendations of the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the American Heart Association 157 are that the initial echocardiogram should be obtained at the time a diagnosis of KD is suspected and that each child with KD should have a second echocardiogram obtained 6 to 8 weeks after onset of the disease. In some centers, an additional echocardiogram is obtained 3 weeks after disease onset. Patients should also have repeated clinical examinations during the first 2 months to detect arrhythmias, congestive heart failure, valvular insufficiency, or myocarditis. 158 Low-dose (3 to 5 mg/kg/day) aspirin should be continued until the erythrocyte sedimentation rate and platelet counts have normalized. Further follow-up is conducted on an individualized basis, with more frequent studies performed for children with demonstrated coronary artery abnormalities (Table 25-7). Children whose coronary arteries have always been normal (risk level I) or are normal by echocardiographic criteria 1 to 2 months after the acute illness (risk level II) are regarded as healthy, and no further intervention is recommended after the 8-week follow-up assessment. In view of the chronic abnormalities in endothelial function,
C HAP T E R
Ii %_-..
IABLE 25-7
25
KAWASAKI DISEASE
535
Recommendations for long-Term Follow-Up
Risk Level
Pharmacological Therapy
Physical AdIvIty
I (no coronary artery changes at any stage of illness) II (transient coronary artery ectasia disappears within 1" (r-8 weeks) III (I small-medium coronary artery aneurysm/major coronary artery)
None beyond 1" 6-8 weeks
No restrictions beyond 1"'6-8 weeks
None beyond 1" 6-8 weeks
No restrictions beyond 1" 6-8 weeks
Low-dose aspirin (3....5 mg/kg aspirin/day), at least until aneurysm regression documented
IV (~1 large or giant coronary artery aneurysm, or multiple or complex aneurysms in same coronary artery, without obstruction)
Long-term antiplatelet therapy and warfarin (target INR 2.~2.5) or low-molecularweight heparin (target: antifactor Xa level 0.5-1.0 U!mL) should be combined in giant aneurysms
V (coronary artery obstruction)
Long-term low-dose aspirin; warfarin or low-molecularweight heparin if giant aneurysm persists; consider use of ~-blockers to reduce myocardial 0, consumption
Follow-Up and DIagnostic Testing
Invasive Testing
Cardiovascular risk assessment counseling at 5 yr intervals Cardiovascular risk assessment counseling at 3 to 5 yr intervals
None recommended
For patients < 11 yrs old, no restriction beyond 1" 6-8 weeks; patients 11-20 yrs old, physical activity gUided by biennial stress test, evaluation of myocardial perfusion scan; contact or high-impact sports discouraged for patients taking antiplatelet agents Contact of high-impact sports should be avoided because of risk of bleeding; other physical activity recommendations gUided by stress test!evaluation of myocardial perfusion scan outcome
Annual cardiology follow-up with echocardiogram + EeG, combined with cardiovascular risk assessment, counseling; biennial stress test/ evaluation of myocardial perfusion scan
Angiography, if noninvasive test suggests ischemia
Biannual follow-up with echocardiogram + ECG; annual stress test/ evaluation of myocardial perfusion scan
1'" angiography at
Contact or high-impact sports should be avoided because of risk of bleeding; other physical activity recommendations guided by stress test! myocardial perfusion scan outcome
Biannual follow-up with echocardiogram and ECG; annual stress test! evaluation of myocardial perfusion scan
None recommended
6-12 mo or sooner if clinically indicated; repeated angiography if noninvasive test, clinical, or laboratory findings suggest ischemia; elective repeat angiography under some circumstances Angiography recommended to address therapeutic options
From Newburger JW et al. Diagnosis, treatment and long-term management of Kawasaki disease: a statement for health professionals from the committee on rheumatic fever, endocarditis and Kawasaki disease, Council on Cardiovascular Disease in the Young: American Heart Association. Pediatrics 114: 170&-1733.2004.
however, many physicians consider a history of KD to be a risk factor for the development of coronary artery disease later in life,I;9 They counsel modification of other risk factors and continue to monitor children on an infrequent but regular basis (e.g., once every 5 years). Children with coronary artery abnormalities are often treated with antithrombotic agents such as low-dose aspirin (3 to 5 mg/kg) for as long as the abnormalities persist. Single small- to medium-sized aneurysms (risk level III) usually resolve as determined by echocardiographic criteria, although this is not always the case. Healing occurs by fibrointimal proliferation, often accompanied by calcification, and vascular reactivity does not return to normal despite a grossly normal appearance. 160 This point is highlighted by a report of sudden death in a 35-year-old child 3 months after dilated coronary arteries had regained a normal echocardiographic appearance .161 Autopsy revealed obliteration of the lumen of the left anterior descending coronary artery due to fibrosis,
with evidence of ongoing active inflammation in the epicardial arteries. Such reports emphasize the need for confirmation of complete response to therapy in children who have had KD. Larger aneurysms--especially so-called giant aneurysms with an internal diameter of at least 8 mm-represent a significant risk for morbidity and mortality, including a 35% chance of infarction (risk level IV).72 These children are treated with anticoagulation using warfarin. Trials with lowmolecular-weight heparin or other antiplatelet agents are ongoing. When injured coronary arteries become obstructed (risk level V), in addition to anticoagulation, various therapies have been attempted to restore circulation, although control of vascular inflammation with sufficient MG or corticosteroids, or both, is an essential prerequisite to arterial reperfusion. Thereafter, treatments may include thrombolytic therapy for arterial thrombosis or vasodilators if tissue viability is primarily threatened by vasospasm,
536
C HAP T E R
25 K AWAS A KID IS E AS E
Urokinase, streptokinase, and tissue-type plasminogen have all been used for the lysis of coronary artery thromboses. Similarly, peripheral arterial obstruction may be corrected by thrombolysis, after which perfusion is maintained with heparin followed by a chronic oral anticoagulant regimen. If these treatments fail, a variety of invasive approaches have been suggested, including percutaneous transluminal coronary angioplasty162 and coronary artery bypass grafting. 163 A number of children with coronary artery disease due to KD have required cardiac transplantation. 164
DISEASE COURSE AND PROGNOSIS Although standard therapy with IVIG and aspirin given within the first 10 days of illness greatly improves outcomes, approximately 5% of children still develop coronary artery aneurysms, and more demonstrate coronary artery eetasia. 126 The mortality rate has dropped steadily as the diagnosis and treatment have improved. It is about 0.1 % in the United States and Japan. 165 Recurrent disease after full recovery from a first episode of KD is rare but does occur. In Japan, the recurrence rate is 2.9%, with a higher incidence of cardiac complications during the second episode. 166 In the United States, the rate of recurrence is lower.
PERSPECTIVE The brief history of KD is much like a condensed version of the history of medicine. In barely one-half century, this acute vasculitis of childhood has gone from being a uniformly fatal disease to a treatable exanthem of childhood. It began as infantile polyarteritis nodosa, diagnosable only at autopsy. Recognition of the mucocutaneous manifestations permitted development of diagnostic criteria and identification of effective treatment. Therapy has been modified, first empirically and then on the basis of a better understanding of the factors that affect outcomes in this disease. Next on the horizon is the possibility of tailoring therapy to an individual child's needs. Ultimately, with microarray and proteomic analysis, identifying the cause of KD and determining ways to prevent it appear to be within reach.
REFERENCES J. Kawasaki T: Acute febrile mucocutaneous syndrome with lymphoid involve-
2. 3. 4. 5. 6.
ment with specific desquamation of the fingers and toes in children. ArelUgi 16: 178-222. 1967. Hoga S. Yamagishi M: Steroid treatment for mucocutaneous ocular syndrome in childhood. Chiryo (Japan) 42: 1174---1179. 1960. Shulman ST: The first reponed case of Kawasaki disease? ] Pediatr 135: 532. 1999. Matteson EL: Notes on the history of eponymic idiopathic vasculitis: the diseases of Henoch and Schonlein, Wegener, Churg and Strauss. Honon, Takayasu, Beh~et, and Kawasaki. Arthritis Care Res 13: 237-245, 2000. Centers for Disease Control and Prevention: CDC revised diagnostic criteria for Kawasaki disease. MMWR Morb Mortal Wkly Rep 39: 27-28, 1990. Burns ]C, Mason WH, Glode M, Shulman ST, et al: Clinical and epidemiologic characteristics of patients referred for evaluation of possible Kawasaki disease. United States Multicenter Kawasaki Disease Study Group. ] Pediatr J18: 680-686, 1991.
7. BenseJer SM, McCrindle BW, Silverman ED, et al: Infections and Kawasaki disease: implications for coronary artery outcome. Arthritis Rheum 48 (Supp!): S516, 2003. 8. Hsieh YC, Wu MH, Wang ]K, et al: Clinicai features of atypical Kawasaki disease. J Microbiol Immunol Infect 35: 57--60. 2002. 9. Witt MT, Minich LL, Bohnsack]F, Young PC: Kawasaki disease: more patients are being diagnosed who do not meet American Heart Association criteria. Pediatrics 104: e1O, 1999. 10. Rosenfeld EA. KE Corydon, Shulman ST: Kawasaki disease in infants less than one year of age.] Pediatr 126: 524---529, 1995. 11. Holman RC, Curns AT, Belay ED, et al: Kawasaki syndrome hospitalizations in the United States, 1997 and 2000. Pecliatr 112: 495-501, 2003. 12. Chang RK: The incidence of Kawasaki disease in the United States did not increase between 1988 and 1997. Pediatrics 111: 1124---1125.2003. U. Stockheim ]A, Innocentini N, Shulman ST: Kawasaki disease in older children and adolescent•. ] Pediatr 137: 250-252, 2000. 14. Tomiyama], Hasegawa Y, Kumagai Y, et al: Acute febrile mucocutaneous lymph node syndrome (Kawasaki disease) in adults: case repon and review of the literature. Jpn] Med 30: 285-289, 1991. 15. Newburger jW, Taubert KA, Shulman SOT, et al: Summary and abstracts of the Seventh International Kawasaki Disease Symposium. December 4---7, 2001, Hakone, Japan. Pediatr Res 53: 153-157, 2003. 16. Yanagawa H, Nakamura Y, Yashiro M, et al: Results of the nationwide epidemiologic survey of Kawasaki disease in 1995 and 1996in]apan. Pediatrics 102: E65, 1998. 17. Yanagawa H, Nakamura Y, Yashiro M, et al: Incidence sUlvey of Kawasaki disease in 1997 and 1998 in Japan. Pediatrics 107:E33, 2001. 18. Shulman ST, McAuley JB, Pachrnan LM, et al: Risk of coronary abnormalities due to Kawasaki disease in urban area with small Asian population. Am ] Dis Child 141: 420-425, 1987. 19. Gardner-Medwin ]M, Dolezalova P, Cummins C, Southwood TR: Incidence of Henoch-Schonlein purpura, Kawasaki disease, and rare vasculitides in children of different ethnic origins. Lancet 360: 1197-1202, 2002. 20. Yanagawa H, Yashiro M, Nakamura Y, et al: Epidemiologic pictures of Kawasaki disease in Japan: from the nationwide incidence survey in 1991 and 1992. Pediatrics 95: 475-449, 1995. 21. Royle ]A, Williams K, Elliott E, et a1. Kawasaki disease in Australia, 1993-9~. Arch Dis Child 78: 33-39, 1998. 22. Bell OM. Morens OM, Holman RC, et al: Kawasaki syndrome in the United States 1976 to 1980. Am] Dis child 137: 211-214, 1983. 23. Yanagawa H, Nakamura Y, Ojima T. et al: Changes in epidemic patterns of Kawasaki disease in Japan. Pediatr Infect Dis] 18: 64--66. 1999. 24. Fujita Y, Nakamura Y, Sakata K, et al: Kawasaki disease in families. Pediatrics 84: 666--669, 1989. 25. Hewitt M, Smith L], Joffe HS, Chambers TL: Kawasaki disease in siblings. Arch Dis child 64: 398-389, 1989. 26. Newburger ]: Treatment of Kawasaki disease: corticosteroids revisited. ] Pediatr 135: 411-413, 1999. 27. Rowley AH, Wolinsky SM. Reiman DA, et al: Search for highly conserved viral and bacterial nucleic acid sequences corresponding to an etiologic agent of Kawasaki disease. Pediatr Res 36: 567-571, 1994. 28. Rowley AH: The etiology of Kawasaki disease: superantigen or conventional antigen? Pediatr Infect Dis] 18: 69-70, 1999. 29. Leung DY, Giorno RC, Kazemi LV, et al: Evidence for superantigen involvement in cardiovascular injury due to Kawasaki syndrome.] Immunol 155: 5018-5021, 1995. 30. Yamashiro Y, Nagata S, Oguchi S, Shimizu T: Selective increase of VR2+ T-cells in the small intestinal mucosa in Kawasaki disease. Pediatr Res 39: 264---266, 1996. 31. Brogan PA, Shah V, Klein N, Dillon M]: V beta-restricted T-cell adherence to endothelial cells: A mechanism for superantigen-dependent vascular injUry. Arthritis Rheum 50: 589-597, 2004. 32. Nomura Y, Yoshinaga M, Masuda K, et al: Maternal antibody against toxic shock syndrome toxin-1 may protect infants younger than 6 months of age from developing Kawasaki syndrome. J Infect Dis 185: 1677-1680. 2002. 33. Duong 'IT, Silverman ED, Bissessar MY, Yeung RS: Sllperantigenic activity is responsible for induction of coronary arteritis in mice: an animal model of Kawasaki disease. [nt Immunol 15: 79-89. 2003. 34. Hsu YH, Wang YH, Hsu WY. Lee YP: Kawasaki disease characterized byerythema and induration at the Bacillus Calmette-Guerin and pUrified protein derivative inoculation sites. Pediatr Infect Dis J 6: 576-578, 1987. 35. Bertotto A, Spinozzi F, Vagliasindi C, et al: Tuberculin skin test reactivity in Kawasaki disease. Pediatr Res 41: 560-562, 1997. 36. Yokota S, Tsubaki K, Kuriyama T, et al: Presence in Kawasaki disease of antibodies to mycobacterial heat-shock protein HSP65 and autoantibodies to epitopes of human HSp65 cognate antigen. Clin Immllnol Immunopathol 67: 163-170, 1993. 37. Rowley AH, Shulman ST, Spike BT, et al: Oligoclonal [gA response in the vascular wall in acute Kawasaki disease.] Imrnunol 166: 1334---1343, 20<1l. 38. Kikuta H, Saklyama Y, Matsumoto S, et al: Detection of Epstein-Barr virus DNA in cardiac and aortic tissues from chronic, active Epstein-Barr vinlS infection associated with Kawasaki disease-like coronary artery aneurysms. ] Pediatr 123: 90-92, 1993.
C HAP T E R 39. Ma!..,uno M, Utagawa E, Sugiura A: ASsociation of rotavirus infections with Kawasaki syndrome, ] Infe<.1 Dis 148: 177, 1983, 40. Okano M, Thiele GM, Sakiyama Y, et al: Adenovirus infection in patients with Kawasaki disease. ] Med Virol 32: 53-57, 1990, 41. Holm ]M, Hansen LK, Oxhoj H: Kawasaki disease associated with parvovirus B19 infection. Eur] Pediatr 154: 633-634, 1995. 42. Burns ]C, Geha RS, Schneeberger EE, et al: Polymerase activity in lymphocyte culture supernatants from patients with Kawasaki disease. Nature 323: 81-Hl16, 1986. 43. Nonnann E, Naas], Gnarpe J, et al: Demonstration of Chlamydia pneumoniae in cardiovascular tissues from children with Kawasaki disease. Pediatr Infect Dis J 18: 72-73, 1999. 44. Akiyama T, Yashiro K: Probable role of Streptococcus pyogenes in Kawasaki disease. Eur J Pediatr 152: 82-92, 1993, 45, Nonoyama S: Immunological abnormalities and endothelial cell injury in Kawasaki disease. Acta Paediatr ]pn 33: 752-755, 1991, 46. Savage CO, Tizard], Jayne 0, et al: Antineutrophil cytoplasm antibodies in Kawasaki disease. Arch Dis Child 64: 360-363, 1989. 47. Furukawa S, Matsubara T, Okumura K, Yabuta K: Decreased expression of CD23 on peripheral blood macrophages/monocytes during acute Kawasaki disease with coronary artery lesions, Int Arch Allergy Immunol 102: 335-339, 1993. 48, Lin CY, Lin CC, Hwang B, Chiang B: Serial changes of serum interleukin-6, interleukin-8, and tumor necrosis factor alpha among patients with Kawasaki disease,] Pediatr 121: 924-926, 1992. 49. Yasukawa K, Terai M, Shulman ST, et al: Systemic production of vascular endothelial growth factor and fms-like tyrosine kinase-1 receptor in acute Kawasaki disease. Circulation 105: 766-769, 2002, 50, Kuijpers T, Wiegman A, van Uer RA, et al: Kawasaki disease: a maturational defect in immune responsiveness. J Infect Dis 180: 1869-1877, 1999. 51. Yanagawa H, Yashiro M, Nakamura Y, et al: Nationwide surveillance of Kawasaki disease in Japan, 1984 to 1993. Pediatr Infect Dis] 14: 69-71, 1995. 52. Harada F, Sada M, Kamiya T, et al: Genetic analysis of Kawasaki syndrome. Am] Hum Genet 39: 537-539, 1986. 53. Uehara R, Yashiro M, Nakamura Y, Yanagawa H: Kawasaki disease in paren!.., and children. Acta Paediatr 92: 694-697, 2003. 54. Huang Y, Lee YJ, Chen MR, et al: Polymorphism of transmembrane region of MICA gene and Kawasaki disease. Exp Clin Immunogenet17: 130-137,2000. 55. Barron KS, Silverman ED, Gonzales ]C, et al: Major histocompatibility complex class II alleles in Kawasaki syndrome-lack of consistent correlation with disease or cardiac involvement.] Rheumatol 19: 1790-1793, 1992. 56. Quasney MW, Bronstein DE, Cantor RM, et al: Increased frequency of alleles associated with elevated tumor necrosis factor-alpha levels in children with Kawasaki disease. Pediatr Res 49: 686-690, 2001. 57. Ahn SY, Jang GC, Shin ]S, et al: Tumor necrosis factor-alpha levels and promoter polymorphism in patients with Kawasaki disease in Korea. Yonsei Med] 44: 1021-1026, 2003. 58. Nishimura S, Zaitsu M, Hara M, et al: A polymorphism in the promoter of the CDI4 gene (CD14/-159) is associated with the development of coronary artery lesions in patients with Kawasaki disease.] Pediatr 143: 357-362, 2003. 59. Takeuchi K, Yamamoto K, Kataoka S, et al: High incidence of angiotensin I converting enzyme genotype II in Kawasaki disease patients with coronary aneurysm. Eur J Pediatr 156: 266-268, 1997. 60. Barron KS: Kawasaki disease: etiology, pathogenesis, and treatment. Cleve Clin J Med 69 (Suppl 2): SII69-S\I78, 2002, 61. Smith LB, Newburger]W, Burns]C: Kawasaki syndrome and the eye. Pediatr Infect Dis] 8: 116-118, 1989. 62. Ammerman SO, Rao MS, Shope TC, Ragsdale CG: Diagnostic uncertainty in atypical Kawasaki disease, and a new finding: exudative conjunctivitis. Pediatr Infect Dis 4: 210-211, 1985. 63. Ryan EH, Walton OS: Conjunctival scarring in Kawasaki disease: a new finding? J Pediatr Ophthalmol Strabismus 20: 106-108, 1983. 64. Jacob JL, Polomeno RC, Chad Z, Lapointe N: Ocular manifestations of Kawasaki disease (mucocutaneous lymph node syndrome). CanJ Ophrhalmol 17: 199-202, 1982. 65. Eberhard BA, Sundel RP, Newburger ]W, et al: Psoriatic eruption in Kawasaki disease. J Pediatr 137: 578--580, 2000, 66. Tashiro N, Matsubara T, Uchida M, et al: Ultrasonographic evaluation of cervical lymph nodes in Kawasaki disease, Pediatrics. 109: E77-E87, 2002, 67. Levy M, Koren G: Atypical Kawasaki disease: analysis of clinical presentation and diagnostic clues. Pediatr Infect Dis J 9: 122-126, 1990. 68. Fukushige], Takahashi N, Ueda Y, Ueda K: Incidence and clinical features of incomplete Kawasaki disease. Acta Paediatr 83: 1057-1060, 1994. 69. Joffe A, Kabani A, Jadavji T: Atypical and complicated Kawasaki disease in infants. Do we need criteria? West] Med 162: 322-327, 1995. 70. Yanagawa H, Tuohong Z, Oki I, et al: Effects of gamma-globulin on the cardiac sequelae of Kawasaki disease. Pediatr Cardiol 20: 248--251, 1999. 71. Burns JC, Capparelli EV, Brown ]A, et al: Intravenous gamma-globulin treatment and retreatmenr in Kawasaki disease. US/Canadian Kawasaki Syndrome Study Group. Pediatr Infect Dis] 17: 1144-1148, 1998, 72. Kato H, Sugimura T, Akagi T, et al: Long-term consequences of Kawasaki disease. A 10- to 21-year follow-up study of 594 patients. Circulation 94: 1379-1385, 1996, 73. Kato H, !chinose E, Kawasaki T: Myocardial infarction in Kawasaki disease: clinical analyses in 195 cases.] Pediatr 108: 923-927, 1986.
2S K AWAS A KID I SEA S E
537
74. Kato H, Ichinose E, Yoshioka F, et al: Fate of coronary aneurysms in Kawasaki disease: serial coronary angiography and long-term follow-up study. Am] Cardiol 49: 1758--1766 , 1982. 75. lemura M, Ishii M, Sugimura T, et al: Long term consequences of regressed coronary aneurysms after Kawasaki disease: vascular wall morphology and function. Heart 83: 307-311, 2000. 76, Matsuura H, Ishikita T, Yamamoto S, et al: Gallium-67 myocardial imaging for the detection of myocarditis in the acute phase of Kawasaki disease (mucocutaneous lymph node syndrome): the usefulness of single photon emission computed tomography. Hr Heart] 58: 385-392, 1987, 77. Newburger JW, Sanders SP, Burns ]C, et al: Left ventricular contractility and function in Kawasaki syndrome. Effect of intravenous gamma-globulin. Circulation 79: 1237-1246, 1989. 78, Moran A, Newburger ]W, Sanders SP, et al: Abnormal myocardial mechanics in Kawasaki disease: rapid response to gamma globulin. Am Heart] 139: 217-223, 2000. 79. Mele T, Evans M: Intestinal obstruction as a complication of Kawasaki disease. ] Pediatr Surg 31: 985-986, 1996. 80. Zulian F, Falcini F, Zancan L, et al: Acute surgical abdomen as presenting manifestation of Kawasaki disease.] Pediatr 142: 731-735, 2003. 81. Dengler LD, Capparelli EV, Bastian ]F, et al: Cerebrospinal fluid proftle in patients with acute Kawasaki disease. Pediatr Infect Dis] 17: 478--481,1998. 82. Tabarki B, Mahdhaoui A, Selmi H, et aI: Kawasaki disease with predominant central nervous system involvement. Pediatr Neurol 25: 239-241, 2001. 83. Larmlde M, Santos-Munoz A, Rutiman R: Kawasaki disease with facial nerve paralysis, Pediatr Dermatol 20: 511-513, 2003. 84. Kumagai N, Ohno S: Kawasaki disease. In Pepose ]S, Holland GN, Wilhelmus KR (eds): Ocular Immunity and Infection. SI. Louis, Mosby, 1996, pp 391-396. 85. Blatt AN, Vogler L, Tychsen L: Incomplete presentations in a series of 37 children with Kawasaki disease: the role of the pediatric ophthalmologist. J Pediatr Ophthalmol Strabismus 33: 114-119, 1996. 86, Melish ME: Kawasaki syndrome: a 1986 perspective. Rheum Dis Clin North Am 13: 7-17, 1987. 87, Sundel RP, Cleveland SS, Beiser AS, et al: Audiologic profiles of children with Kawasaki disease. Am J Otol 13: 512-515, 1992. 88, Suddleson EA, Reid B, Woolley MM, Takahashi M: Hydrops of the gallbladder associated with Kawasaki syndrome, J Pediatr Surg 22: 956-959, 1987. 89, Waring NP, Ortenberg J, Galen WK, et al: Priapism in Kawasaki disease. JAMA 261: 1730-1731, 1989. 90. Ferriero OM, Wolfsdorf JI: Hemolytic uremic syndrome associated with Kawasaki disease. Pediatrics 68: 405-406, 1981. 91. Salcedo ]R, Greenberg L, Kapur S: Renal histology of mucocutaneous lymph node syndrome (Kawasaki disease). Clin Nephrol 29: 47-51, 1988, 92. Lande MB, Gleeson JG, Sundel RP: Kawasaki disease and acute renal failure. Pediatr Nephrol 7: 593, 1993. 93. Yanagihara R, Todd ]K: Acute febrile mucocutaneous lymph node syndrome. Am J Dis Child 134: 603-610, 1980. 94. Naoe S, Takahashi K, Masuda H, Tanaka N: Kawasaki disease. With particular emphasis on arterial lesions. Acta Pathol Jpn 41: 785-797, 1991. 95. Fujiwara H, Hamashima Y: Pathology of the heart in Kawasaki disease. Pediatr 61: 100-107, 1978, 96. Rowley AH, Eckerley CA, Jack HM, et al: 19A Plasma cells in vascular tissue of patients with Kawasaki syndrome. ] Immunol 159: 5946-5955, 1997. 97. Jennette ]C: Implications for pathogenesis of patterns of injury in small- and medium-sized vessel vasculitis. Cleve Clin] Med 69 (Suppl 2): SU33-S1I38, 2002. 98. Leung DY, Cotran RS, Kurt-]ones E, et al: Endothelial activation in the pathogenesis of Kawasaki disease. Trans Assoc Am Physicians 102: 131-138, 1989. 99. Yonesaka S, Nakada T, Sunagawa Y, et al: Endomyocardial biopsy in children with Kawasaki disease. Acta Paediatr Jpn 31: 706-711, 1989. 100, Fujiwara H, Hamashima Y: Pathology of the heart in Kawasaki disease. Pediatrics 61: 100-107, 1978. 101. Suzuki A, Kamiya T, Kuwahara N, et al: Coronary arterial lesions of Kawasaki disease: cardiac catheterization findings of 1100 cases. Pediatr Cardiol 7: 3-9, 1986, 102, Hara T, Mizuno Y, Veda K, et al: Neutropenia in Kawasaki disease. Eur J Pediatr 148: 580, 1989. 103, Rowe PC, Quinlan A, Luke BK: Value of degenerative change in neutrophils as a diagnostic test for Kawasaki syndrome. J Pediatr 119: 370-374, 1991. 104. Salo E, Pesonen E, Viikari J: Serum cholesterol levels during and after Kawasaki disease, .J Pediatr 119: 557-561, 1991. 105. Soppi E, Salo E, Pelkonen P, Antibodies against neutrophil cytoplasmic components in Kawasaki disease. Acta Path Microbiol Immunol &and 100: 269-272, 1992, 106. Grunebaum E, Blank M, Cohen S, et al: The role of anti-endothelial cell antibodies in Kawasaki disease-in vitro and in vivo studies. Clin Exp Immunol 130: 233-240, 2002. 107. Guzman J, Fung M, Petty RE: Diagnostic value of anti-neutrophil cytoplasmic and anti-endothelial cell antibodies in early Kawasaki disease. ] Pediatr 124: 917-920, 1994. 108, Irazuzta ]E, E1bl F, Rees AR: Factor VIlI related antigen (von Willebrand's factor) in Kawasaki disease, Clin Pediatr (Phila) 29: 347-348, 1990. 109. Laxer RM, Schaffer FM, Myones BL, et al: Lymphocyte abnormalities and complement activation in Kawasaki disease. Prog Clin Bioi Res 250: 175-184,1987.
538
C HAP T E R
25 K AWAS A KID I SEA S E
110. American Academy of Pediatrics: Kawasaki syndrome. In Pickering LK (ed): Red Book: 2003 Report of the Committee on Infectious Diseases. Elk Grove Village, IL, American Academy of Pediatrics, 2003, pp 392-395. II 1. Ishihara H, Watanabe S, Izumida N, et al: Detection of coronary arterial lesions by two dimensional echocardiography in patients with Kawasaki disease. Bull Tokyo Med Dent Univ 31: 139-143, 1984. 112. Report of the Subcommittee on Standardization of Diagnostic Criteria and Reporting of Coronary Artery Lesions in Kawasaki Disease. Tokyo, Japan, Ministry of Health and Welfare, 1984. 113. Oherhoffer R, Lang D, Feilen K: The diameter of coronary arteries in infants and children without heart disease. Eur] Pediatr 148: 389-392, 1989. 114. de Zorzi A, Colan SD, Gauvreau K, et al: Coronary artery dimensions may be misclassified as normal in Kawasaki disease.] Pediatr 133: 254--258, 1998. II5. Tanaka N, Naoe S, Masuda H, Ueno T: Pathological srudy of sequelae of Kawasaki disease (MCLS) with special reference to the heart and coronary arterial lesions. Acta Pathol ]pn 36: 1513-1527, 1986. II6. Furuyama H, Odagawa Y, Katoh C, et al: Assessment of coronary function in children with a history of Kawasaki disease using (l5)0-water positron emission tomogmphy. Circulation 105: 2878-2884, 2002. II7. Dhillon R, Clarkson P, Donald AE, et ai: Endothelial dysfunction late after Kawasaki disease. Circulation 94: 2103-2106, 1996. 118. Nakamura Y, Yanagawa H, Harada K, et al: Mortality among persons with a history of Kawasaki disease in Japan: the fifth look. Arch Pediatr Adolesc Med 156: 162-165. 2002. 119. Kusakawa S. Tatara K: Efficacies and risks of aspirin in the treatment of the Kawasaki disease. Prog Clin BioI Res 250: 401-413, 1987. 120. Akagi T, Kato H, Inoue 0, Sato N: Salicylate treatment in Kawasaki disease: high dose or low dose? Eur] Pediatr 150: 642-646, 1991. 121. Durongpisitkul K, Gururaj \1], Partin ]M, Martin CF: The prevention of coronary artery aneurysm in Kawasaki disease: a meta-analysis on the efficacy of aspirin and immunoglobulin treatment. Pediatrics 96: 1057-1061. 1995. 122. Koren G, Silverman E, Sundel R, et al: Decreased protein binding of salicyIates in Kawasaki disease. ] Pediatr 118: 456-459, 1991. 123. Lee ]H, Hung HY, Huang FY: Kawasaki disease with Reye syndrome: report of one case. Zhonghua Min Guo Xiao Er Ke Yi Xue Hui Za Zhi 33: 67-71, 1992. 124. Furusho, K., Kamiya T, Nakano H, et al: High-dose intravenous gamma-globulin for Kawasaki disease. Lancet 2: 1055-1058, 1984. 125. Newburger lW, Takahashi M, Bums ]C, et al: The treatment of Kawasaki syndrome with intravenous gamma globulin. N Engl] Med 315: 341-347, 1986. 126. Newburger lW, Takahashi M, Beiser AS, et al: A single intravenous infusion of gamma globulin as compared with four infusions in the treatment of acute Kawasaki syndrome. N Engl] Med 324: 1633-1639, 1991. 127. Terai M, Shulman ST: Prevalence of coronary artery abnormalities in Kawasaki disease is higWy dependent on gamma globulin dose but independent of salicylate dose.] Pediatr 131: 888-893, 1997. 128. Newburger]: Kawasaki disease: Who is at risk? J Pediatr 137: 149-152, 2000. 129. Stockheim A, Innocentin N, Shulman S: Kawasaki disease in older children and adoiescents. ] Pediatr 137: 250--252, 2000. 130. Fukunishi M, Kikkawa M, Hamana K, et al: Prediction of non-responsiveness to intravenous high dose gamma-globulin therapy in patients with Kawasaki disease at onset. J Pediatrics 137: 172-176, 2000. 131. Han RK, Silverman ED, Newman A, McCrindle BW: Management and outcome of persistent or recurrent fever after initial intravenous gamma globulin therapy in acute Kawasaki disease. Arch Pediatr Adolesc Med 154: 694-699, 2000. 132. Mori M. lmagawa T, Yasui K, et al: Predictors of coronary artery lesions after intravenous gamma-globulin treatment in Kawasaki disease. ] Pedlatr 137: 177-180, 2000. 133. Newburger lW, Burns ]C, Beiser AS, Loscalzo ]: Altered lipid profile after Kawasaki syndrome. Circulation 84: 625-<531, 1991. 134. Tsujimoto H, Takeshita S, Nakatani K, et al: Intravenous immunoglobulin therapy induces neutrophil apoptosis in Kawasaki disease. Clin Immunol 103: 161-168, 2002. 135. Yi Q), U CR, Yang XQ: Effect of intravenous immunoglobulin on inhibiting peripheral blood lymphocyte apoptosis in acute Kawasaki disease. Acta Paediatr 90: 623-627, 2001. 136. Sherer Y, Levy Y, Langevitz P, et al: Adverse effects of intravenous immunoglobulin therapy in 56 patients with autoimmune diseases. Pharmacology 62: 133-137, 2001. 137. Bresee ]S, Mast EE, Coleman P], et al: Hepatitis C virus infection associated with administration of intravenous immune globulin. A cohort study. ]AMA 276: 1563-1567, 1996. 138. Klassen TP, Rowe PC, Gafni A: Economic evaluation of intravenous immune globulin therapy for Kawasaki syndrome. ] Pediatr 122: 538-542, 1993. 139. Marasini M, Pongiglione G, Gazolo D, et al: Late intravenous gamma globulin treatment in infants and children with Kawasaki disease and coronary artery abnormalities. Am] Cardiol 68: 796-797, 1991.
140. Tse SM, Silverman ED, McCrindle BW, Yeung RS: Early treatment with intravenous immunoglobulin in patients with Kawasaki disease. ] Pediatr 140: 450-455, 2003. 141. Kato H, Koike S, Yokoyama T: Kawasaki disease: effect of treatment on coronary artery involvement. PediatriCS 63: 175-179, 1979. 142. Nonaka Z, Maekawa K, Okabe T, et ai. Randomized controlled study of intravenous prednisolone an gamma globulin treatment in 100 cases with Kawasaki disease. In Fifth International Kawasaki Disease Symposium. Philadelphia, Elsevier Science, 1994. 143. Shinohara M, Sone K, Tomoroasa T, Morikawa A: Corticosteroids in the treatment of the acute phase of Kawasaki disease. ] Pediatr 135: 465-469, 1999. 144. Hashino K, Ishii M, lemura M, et al: Re-treatment for immune globulinresistant Kawasaki disease: a compardtive study of additional immune globulin and steroid pulse therapy. Pediatr lnt 43: 2Il-217, 2001. 145. Sundel RP, Baker AL, Fulton DR, Newburger ]W: Corticosteroids in the initial treatment of Kawasaki disease: report of a randomized trial. ] Pediatr 142: 611-616, 2003. 146. ]ibiki T, Terai M, Kurosaki T, et al: Efficacy of intravenous immune globulin therapy combined with dexamethasone for the initial treatment of acule Kawasaki disease. Eur] Pediatr 163: 229-233, 2004. 147. Furukawa S, Matsubara T, Umezawa Y, et al: Pentoxifylline and intravenous gamma globulin combination therapy !()r acute Kawasaki disease. Eur ] Pediatr 153: 663-667, 1994. 148. Weiss ]E, Eberhard BA, Chowdhury D, Gottlieb BS: lnfliximab as a novel therapy for refractory Kawasaki disease. J Rheumatol 31: 808--H1O, 2004. 149. Mori M, Tomono N, Yokota S: Coronary arteritis of Kawasaki disease unresponsive to high-dose intravenous gamma-globulin treatment successfully treated with plasmapheresis. Nihon Rinsho Meneki Gakkai Kaishi 18: 282-288, 1995. 150. Wallace CA, French lW, Kahn S], Sherry DO: Initial intravenous gamma-globulin treatment failure in Kawasaki disease. Pediatrics 105: E78, 2000. 151. Raman V, Kim], Sharkey A, Chatlla T: Response of refractory Kawasaki disease to pulse steroid and cyclosporin A therapy. Pediatr Infect Dis J 20: 635-<537, 2001. 152. Miyamae T, Nakasima S, Tomono N, et al: Two infants with classic polyarteritis nodosa but not Kawasaki disease. Nihon Rinsho Meneki Gakkai Kaishi 23: 445-453, 2000. 153. Williams RV, Wilke VM, Tani LY, Minich LL: Does abciximab enhance regression of coronary aneurysms resulting from Kawasaki disease? Pediatrics 109: E4,2002. 154. Takahashi M, Weinter K: Changes in coronary artery aneurysms following treatment with abciximab. Presented at the Seventh International Kawasaki Disease Symposium. 2001. Hakone, Japan. 155. Shen CT, Wang MK: Antioxidants may mitigate the deterioration of coronary arteritis in patients with Kawasaki disease unresponsive to high-dose intravenous gamtna-globulin. Pediatr Cardioi 22: 419-422, 2001. 156. Zaitsu M, Hamasaki Y, Tashiro K, et al: Ulinastatin, an elastase inhibitor, inhibits the increased mRNA expression of prostaglandin H2 synthase-type 2 in Kawasaki disease.] Infect Dis 181: 1101-1109. 2000. 157. Dajani AS, Taubert KA, Takahashi M, et al: Guidelines for long-term management of patients with Kawasaki disease. Report from the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease, Council on Cardiovascular Disease in the Young, American Heart Association. Circulation 89: 916-922. 1994. 158. Fulton DR, Newburger lW: Long-term cardiac sequelae of Kawasaki disease. Curr Rheumatol Rep 2, 324--329. 2000. 159. Cheung YF, Yung TC, Tam SC, et al: Novel and traditional cardiovascular risk factors in children after Kawasaki disease: implications for premature atherosclerosis. ] Am Coli Cardiol 43: 120--124 2004. 160. Dhillon R, Clarkson P, Donald AB, et al: Endothelial dysfunction late after Kawasaki disease. Circulation 94: 2103-2106, 1996. 161. McConnell ME, Hannon DW, Steed RD, Gilliland MG: Fatal obliterative coronary vasculitis in Kawasaki disease.] Pediatr 133: 259-261, 1998. 162. Kuramochi Y, Ohkubo T, Takechi N, Ogawa S: Feasibility of percutaneous transluminal coronary angioplasty to patients with Kawasaki disease as an early management strategy. Pediatr Cardiol 22: 183-187, 2001. 163, Gotteiner N, Mavroudis CV, Backer CL, et al: Coronary artery bypass grafting for Kawasaki dLqease. Pediatr Cardiol 23: 62-67, 2002. 164. Checchia PA, Pahl E, Shaddy RE, Shulman ST: Cardiac transplantation for Kawasaki disease. Pediatrics 100, 695-<599, 1997. 165. Chang RK: Hospitalizations for Kawasaki disease among children in the United States, 1988-1997. Pediatrics 109: e87, 2002. 166. Nakamura Y, Yanagawa H, Ojima T, et al: Cardiac sequelae of Kawasaki disease among recurrent cases. Arch Dis Child 78: 163-165, 1998.
26
C H A P T E R
G RANULOMATOUS VASCULITIS , G IANT C ELL A RTERITIS , AND S ARCOIDOSIS Carol B. Lindsley and Ronald M. Laxer
The granulomatous vasculitides include Wegener’s granulomatosis (WG), Churg-Strauss syndrome, lymphomatoid granulomatosis, and primary angiitis of the central nervous system (CNS). These diseases are categorized as histiocytic disorders in a pathophysiologic model of macrophage-monocyte proliferation. All are rare in childhood and adolescence, and much of what we know comes from studies of adults with the diseases. Of these four, WG is the most common.
WEGENER’S GRANULOMATOSIS WG was first described in the 1930s.1,2 It is a systemic disease characterized by granulomatous vasculitis involving the upper and lower respiratory tracts and associated with glomerulonephritis. Although rare and occurring predominantly in middle-aged adults, the disease is reported in the pediatric population.3,4
Classification The 1990 American College of Rheumatology (ACR) criteria for the classification of WG are listed in Table 26–1.5 The Chapel Hill Consensus Conference on the Nomenclature of Systemic Vasculitis has defined WG as “granulomatous inflammation involving the respiratory tract and necrotizing vasculitis affecting small to mediumsized vessels.” If lesions are limited to the upper respiratory passages and the manifestations of vasculitis are minimal, the syndrome is called localized WG or midline granuloma. The limited form of WG does not include glomerulonephritis.6
Epidemiology Very little is known about the incidence, prevalence, or demographic characteristics of WG in children and adolescents. In a large study of patients who were hospitalized between 1986 and 1990,7 the 5-year incidence was
3.2 cases per 100,000 persons. Those with disease onset before 20 years of age accounted for 3.3%, an incidence in that age group of approximately 0.1 cases per 100,000 persons. In the pediatric patient, WG is generally a disease of the second decade of life, with a mean age at onset of 15.4 years in one of the largest studies.4 There is no clear sex predominance in the pediatric age range,8 although in adults, males outnumber females 1.7:1.5 In a British study,9 13 of 17 patients were female, and age at disease onset ranged from 2 weeks to 14 years, with a mean of 6.3 years. In a prospective study of 23 children between 9 and 19 years old at onset, 61% had generalized disease, and 39% had the limited form of the disease.4
Etiology and Genetic Factors The cause of WG is unknown. Theories of causation have included autoimmune, hypersensitivity, or allergic reactions to unknown antigens and sensitization of the respiratory tract to bacterial pathogens. Nasal carriage of Staphylococcus aureus has been associated in adults with high rates of relapse.10 Using T lymphocyte clones derived from two patients with WG, reactivity to S. aureus, but not to other bacteria, was demonstrated, suggesting a role for this organism in pathogenesis.11 The familial occurrence of WG has only occasionally been reported.12–14 Associations with antigens of the histocompatibility system (human leukocyte antigen [HLA]) have been inconsistent. A large study from the Netherlands reported a highly significant decrease in the frequency of HLA-DR13/DR6.15 Increased expression of some of the polymorphic forms of the Fc γ receptors have been associated with WG in some studies but not in others. Homozygosity for the R131 form of Fc γ RIIa and the F158 form of Fc γ RIIIa were associated with an increased risk of relapse in adults with WG.16 These forms of the Fc γ receptors limit antigen clearance and may therefore predispose to disease associated with chronic infection with organisms such as S. aureus. Chemokine receptor 5
539
540
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S TABLE 26–1 Four Criteria for Classification of Wegener’s Granulomatosis
Criterion*
Description
Nasal or oral inflammation
Painful or painless oral ulcers or purulent or bloody nasal discharge Nodules, fixed infiltrates or cavities Microhematuria (>5 RBC/high-power field) or RBC casts Granulomatous inflammation within the wall of an artery or in the perivascular or extravascular area of an artery or arteriole
Abnormal-appearing chest radiograph Abnormal urinary sediment Granulomatous inflammation
*Diagnosis of Wegener’s granulomatosis requires the presence of two of the four criteria. The presence of any two or more criteria has a sensitivity of 88.2% and a specificity of 92.0%. RBC, red blood cell. Adapted from Leavitt RY, Fauci AS, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of Wegener’s granulomatosis. Arthritis Rheum 33: 1101, 1990. Copyright © 1990 John Wiley & Sons, Inc. Reprinted with permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
(CCR5) and its ligands are abundantly present in pulmonary lesions and may exert an important pathogenetic role in some patients.17 Interleukin-10 polymorphisms may also have a role in susceptibility.18
Clinical Manifestations The triad of paranasal sinus involvement, pulmonary infiltration, and renal disease is characteristic of WG (Table 26–2).4,7,19–21 At onset, nonspecific complaints— including fever, malaise, and weight loss—are common, and most patients present with upper and lower respiratory disease. Upper respiratory tract signs and symptoms such as rhinorrhea, nasal mucosal ulcerations, epistaxis, persistent cough, hoarseness, and paranasal sinus pain or drainage are common. Damage to the nasal cartilage, characteristic of long-standing disease, may result in a saddle-nose deformity, suggesting the possible diagnosis of relapsing polychondritis (Fig. 26–1). Overall, nasal, sinus, tracheal (including tracheal stenosis), and ear abnormalities were reported in 91% of patients with childhood onset of the disease.4,22 Lung disease occurs in 74% of children.4 Lower respiratory tract symptoms include cough, dyspnea, and hemoptysis. Nodular pulmonary infiltrates are often visible on radiographs and may be a presenting manifestation.23,24 Blurred vision, eye pain, conjunctivitis, episcleritis, persistent otitis media, hearing loss, arthralgia, and myalgia are also common.25 Mild, transient arthralgia occurs in 30% to 78% of patients.4 Renal disease occurs in 61% of children4 and often leads to renal failure, which can develop and worsen rapidly.23 Skin lesions may resemble HenochSchönlein purpura20 or may be ulcerative, vesicular, papular, or nodular; these lesions occur in 9% of patients at disease onset and in up to one half during the course of the disease.4 CNS involvement (e.g., neuropathy, cranial nerve palsies, seizures) occurs less frequently than in adult patients.26 Cardiac disease is uncommon but may include myocardial infarction, arrhythmias, and valvulitis; in one 16-year-old boy, a right ventricular granuloma was reported.27 Gastrointestinal symptoms include nonspecific pain, nausea, and vomiting.20
Diagnosis A definite diagnosis of WG requires demonstration of typical changes on biopsy of lung, skin, or kidney. However, a characteristic clinical presentation, together with antibodies directed at neutrophil cytoplasmic antigens (ANCAs) with a cytoplasmic (c) pattern on immunoTABLE 26–2 Manifestations of Wegener’s Granulomatosis in Childhood Patients Affected (%) (n = 23) Abnormality Ear, nose, throat Sinusitis Nasal disease Otitis media Subglottic stenosis Hearing loss Ear pain Oral lesions Arthralgia/arthritis Pulmonary disease Infiltrates Nodules Hemoptysis Pleuritis Glomerulonephritis Rash (purpura, vesicles, papules, nodules) Ocular disease Dacryocystitis Eye pain Proptosis Scleritis/episcleritis Conjunctivitis Visual loss Corneal ulcers Fever Weight loss CNS disease (cranial nerve palsy, seizure) Peripheral neuropathy Pericarditis
Onset 87
Total 91
61 48 39 4 26 22 4 30 22
83 65 48 48 39 22 9 78 74
9 13 9 9 9 9
61 43 26 13 61 52
13
48 4 4 0 4 0 0 0
26 17 17 13 9 9 4
22 13 4
43 26 17
0 9
9 9
CNS, central nervous system. Data from Rottem M, Fauci AS, et al: Wegener granulomatosis in children and adolescents: clinical presentation and outcome. J Pediatr 122: 26, 1993.
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
541
cally demonstrates proteinuria, microscopic hematuria, and red blood cell casts in up to 50% of patients.5,23 Gross hematuria is uncommon.4 Pulmonary function tests may show an decrease in the diffusion capacity of carbon monoxide (DLCO), which may be the earliest sign of pulmonary hemorrhage. ANCAs are most important autoantibodies in WG. ANCAs may be cytoplasmic (cANCA) or perinuclear (pANCA) on immunofluorescence microscopy (Fig. 26–2). Each pattern has a different antigenic specificity and correlates somewhat with the type of vascular injury.28 ANCAs directed to neutrophil cytoplasmic enzyme protein PR3 (cANCA) were present in more than 90% of patients.29–31 Enzyme-linked immunosorbent assay (ELISA) testing should follow immunofluorescence testing of peripheral blood neutrophils. Multicenter data suggest capture ELISA is a highly sensitive assay for detection of PR3-ANCA in WG.32 Membrane-bound PR3 appears to colocalize with the adhesion molecule CD116/CD18 (i.e., β integrin).33 There is some debate about whether titers of ANCA correlate with disease activity. Although these antibodies have not been associated with other types of connective tissue disease, they have been demonstrated in inflammatory bowel diseases (usually pANCA), infection-associated vasculitis, and paraneoplastic syndromes (Table 26–3).28,31,32 Antinuclear antibodies of unknown specificity are uncommon. Rheumatoid factors are present in approximately 50% of adult patients.23 Serum levels of immunoglobulin A (IgA) may be increased.7 Hypercoagulability may be an increased risk because of antiphospholipid antibodies and factor V Leiden mutations.34
Pathology ■ Figure 26–1 The saddle-nose deformity resulted from granulomatous destruction of the nasal cartilage in this 14-year-old girl with Wegener’s granulomatosis.The process was painless and occurred over several months. Later, she experienced pulmonary hemorrhage and radiologic changes that led to the diagnosis.
fluorescent staining and the presence of antibodies to proteinase-3 (PR3) antigen, may obviate the need for a tissue diagnosis. The differential diagnosis includes other causes of granulomatous vasculitis, such as mycobacteria, fungi, or helminths.23 Other forms of vasculitis that can manifest as pulmonary-renal syndromes, such as Goodpasture’s syndrome, systemic lupus erythematosus, mixed connective tissue disease, or microscopic polyarteritis nodosa, should be considered. In young children, chronic granulomatous disease may need to be excluded.
Laboratory Examination White blood cell counts are usually normal or moderately elevated. Patients usually have anemia, thrombocytosis, and marked elevation of the erythrocyte sedimentation rate or C-reactive protein level. Elevation of blood urea nitrogen and serum creatinine levels indicates the presence of significant renal disease. Urinalysis characteristi-
Granulomatous involvement of medium-sized arteries and veins is characteristic. Leukocytoclastic vasculitis may be seen involving small vessels as well. Granulomata show acute and chronic inflammation with central necrosis and histiocytes, lymphocytes, and giant cells (Fig. 26–3).23 Renal glomeruli are infiltrated with lymphocytes and histiocytes (Fig. 26–4). The most commonly reported renal lesions are extracapillary proliferation (with or without fibrinoid necrosis) and crescent formation, found in a focal and segmental pattern, followed by necrotizing glomerulonephritis.35,36 Glomerular sclerosis occurs quickly and is often seen on the initial biopsy, even with good renal function. Renal granulomata are rare.37 Immunofluorescent microscopy is characteristic of a “pauci-immune” pattern with scanty deposition of immunoglobulins and complements.7,35 Dense subendothelial deposits are visible on electron microscopy.25
Radiologic Examination Approximately two thirds of children with WG have abnormalities on chest radiographs, including nodules (granulomata), cavitation, and infiltrates38,39 (Fig. 26–5). The lesions may be solitary or bilateral. Infiltrates are often fleeting and may be asymptomatic. In one study,4 one third of all abnormal radiographs were those of patients without pulmonary symptoms. Pleural effusions and pneumothorax may also occur. Sinus radiographs
542
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S ■ Figure 26–2 Indirect immunofluorescence microscopy staining patterns produced by antibodies directed at neutrophil cytoplasmic antigens (ANCAs) with a cytoplasmic staining pattern (cANCAs) (A) and those with a perinuclear staining pattern (pANCAs) (B) on alcohol-fixed neutrophils. (A and B, From Jennette JC, Falk RJ: Antineutrophil cytoplasmic autoantibodies and associated diseases: a review. Am J Kidney Dis 15: 517, 1990.)
may demonstrate thickening of the sinus lining or opacification of the frontal or maxillary sinuses. Computed tomography (CT) of the sinuses or high-resolution CT of the chest can help delineate the lesion and may be necessary to exclude the presence of abnormalities.40
Treatment After a firm diagnosis is established, treatment with glucocorticoids in combination with cyclophosphamide is indicated. The disease was fatal in almost all children reported before the use of a combination of glucocorticoid and cytotoxic agents.19 This protocol has induced remission in more than 90% of patients.19,23,25,41 In criti-
cally ill patients, intravenous methylprednisolone followed by daily high-dose glucocorticoids and cyclophosphamide should be used initially.42 Cyclophosphamide (2 mg/kg/day) with prednisone at a dose of 1 mg/kg/day for 4 weeks and then tapered to an alternate-day regimen induced remission in 97% of patients in one large series.4 This regimen was continued for approximately 1 year past remission, and then the cyclophosphamide was tapered by 25-mg decrements every 2 months if there was no relapse. The median requirement for cytotoxic therapy was 28 months. Studies of therapy are summarized in Table 26–4. Therapy with a combination of glucocorticoid and methotrexate resulted in remission in 69% of adults with
TABLE 26–3 Common Disease Associations with Neutrophil Cytoplasmic Antigens Antigen
ANCA Pattern
Disease Association
PR3
cANCA
MPO
pANCA
BPI Actin
ANCA pANCA
Wegener’s granulomatosis Churg-Strauss syndrome Microscopic polyarteritis Churg-Strauss syndrome Ulcerative colitis Sclerosing cholangitis Crohn’s disease Cystic fibrosis Autoimmune hepatitis type I
Frequency (%) 30–90 25–50 25–75 50–75 40–80 65–85 10–40 80–90 70–75
ANCA, antibody directed at neutrophil cytoplasmic antigen; cANCA, cytoplasmic ANCA; pANCA, perinuclear ANCA. Data from Hoffman GS, Leavitt RY, Kerr GS, et al: Treatment of Wegener’s granulomatosis with glucocorticoids and methotrexate. Arthritis Rheum 35: 1322–1329, 1992, and from Specks U, Wheatley CL, McDonald TJ, et al: Anticytoplasmic autoantibodies in the diagnosis and follow-up of Wegener’s granulomatosis. Mayo Clin Proc 64: 28–40, 1989.
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
543
■ Figure 26–3 Lung biopsy specimen from a patient with Wegener’s granulomatosis. Necrotizing granulomata and fibrous tissue have obliterated the normal alveolar architecture. Hematoxylin and eosin stain; magnification × 480.
■ Figure 26–5 Patchy, lower lobe infiltrates and several well-defined granulomata (left lower lobe, right upper and middle lobes) are evident in the lungs of an adolescent with Wegener’s granulomatosis.
WG that was not immediately life threatening.29 Methotrexate has also been suggested as an alternative to cyclophosphamide in children with WG.43,44 Because of long-term toxicity of cyclophosphamide, the use of methotrexate for maintenance should be considered. Trimethoprim-sulfamethoxazole has been effective in some patients45,46 and should be considered as adjunctive therapy. Leflunomide has been used as an agent to maintain remission.47 Acute hemorrhagic respiratory failure may be responsive to diluted surfactant.48 Tracheal stenosis may require tracheostomy or stent placement.22 A pilot study using etanercept indicated that it might be beneficial in some patients who have failed standard treatment49; a multicenter study is ongoing. Baseline data from that study reveal significant differences between limited and severe subsets of WG, with the former being younger, more likely female, and having longer disease duration, greater likelihood of recurrent disease, and higher prevalence of destructive upper respiratory tract disorders.50 Rituximab was effective in one patient and may be considered in patients who have failed other therapeutic options.51 Current therapeutic recommendations are summarized in Table 26–5. For patients with pulmonary hemorrhage, intensive care unit management with ventilatory support may be required. The role of plasmapheresis in these desperate situations is unknown, but it is frequently used in an attempt to stabilize the patient. ■ Figure 26–4 A, Renal biopsy specimen from a child with Wegener’s granulomatosis.The glomerulus on the right shows areas of hypercellularity and fibrinoid necrosis with interstitial inflammation. Hematoxylin and eosin stain; magnification × 480. B, Positive immunofluorescent stain for fibrin in a renal biopsy specimen from a patient with Wegener’s granulomatosis. Magnification × 480.
Course of the Disease and Prognosis In adults treated with prednisone and cyclophosphamide, relapses were common and disease- and treatmentrelated morbidity substantial, which led to the search for
544
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S TABLE 26–4 Studies of the Treatment of Wegener’s Granulomatosis
Study
Regimen
Result
Fauci et al, 198323
CTX, 2 mg/kg/day PO Prednisone, 1 mg/kg/day PO CTX every month IV
Remission in 93%
Hoffman et al, 199043 Hoffman et al, 199244 de Groot et al, 199653
Guillevin et al, 199754
Langford et al, 199955
Response followed by relapse in 72% Improvement in 1 of 9
TMP-SMX Induction: CTX, 2 mg/kg/day PO or IV Prednisone, 1 mg/kg/day PO Maintenance: MTX, 0.3 mg/kg/wk SC 86% responded or TMP-SMX, 960 mg bid PO 58% responded or MTX + prednisone 91% responded or TMP-SMX + prednisone 0% responded Induction: CTX, 0.7 g/m2 Methylprednisolone IV × 3 days followed by oral prednisone, 1 mg/kg/day Maintenance: CTX, IV + prednisone 59% relapsed CTX, PO + prednisone 13% relapsed Induction: CTX, 2 mg/kg/day PO prednisone, 1 mg/kg/day PO 100% responded Maintenance: MTX, 0.3 mg/kg/wk PO, increasing to maximum of 20–25 mg/wk Prednisone tapered to alternate-day dose
CTX, cyclophosphamide; MTX, methotrexate; TMP-SMX, trimethoprim-sulfamethoxazole.
alternative treatment regimens.44 Approximately 25% of children with WG have at least one serious infection during the course of the illness.4 Subglottic stenosis is four times as common and nasal deformity twice as common in children as in adults; surgical intervention may be necessary to maintain the airway. In one third of children, irreversible renal insufficiency develops. Treatment-
related morbidity includes cystitis and infertility in 22%.4 Malignancies did not occur in any of 23 children followed for a mean of 8.7 years.4 Relapses of disease requiring retreatment occurred in approximately one half of the patients. Persistent sinus pain and dysfunction, hearing loss, and pulmonary insufficiency may ensue. Twentytwo children younger than 15 years died of the disease in
TABLE 26–5 Current Recommended Therapy of Wegener’s Granulomatosis Phase
Drug
Regimen*
Induction
Prednisone
1–2 mg/kg/day PO in two or three divided doses (max, 80 mg); exceptionally ill patients receive methylprednisolone, 30 mg/kg/day (max, 1 g) for 1–3 d IV plus 2 mg/kg/day PO; exceptionally ill patients may receive up to 4 mg/kg/day PO for 3 days, followed by 2 mg of cyclophosphamide as IV pulse (0.75 g/m2 ) monthly is an alternative After 4 wk, prednisone is consolidated and tapered as long as the patient remains well. plus 2 mg/kg/day PO (taper after 1 yr of disease control or remission) or 0.3–1 mg/kg SC once each week (max, 30–40 mg) or 2 mg/kg/day
Cyclophosphamide* Maintenance
Prednisone Cyclophosphamide Methotrexate Azathioprine
*Dose may need to be adjusted based on white blood cell count and urinalysis. Data from references 23, 312, 313, 314.
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
545
TABLE 26–6 American College of Rheumatology Criteria for Classification of Churg-Strauss Syndrome Criterion*
Description
Asthma Eosinophilia History of allergy
History of wheezing or diffuse high-pitched rales on expiration Eosinophils > 10% of white blood cell differential count History of seasonal allergy (e.g., allergic rhinitis) or documented allergies, including food, contactants, and others (except for drug allergies) Mononeuropathy, multiple mononeuropathies or polyneuropathy (i.e., glove/stocking distribution) attributable to a systemic vasculitis Migratory or transitory pulmonary infiltrates on radiographs attributable to a systemic vasculitis History of acute or chronic paranasal sinus pain or tenderness or radiographic opacification of the paranasal sinuses Biopsy including artery, arteriole, or venule, showing accumulation of eosinophils in extravascular areas
Mononeuropathy or polyneuropathy Pulmonary infiltrates Paranasal sinus abnormality Extravascular eosinophils
*For classification purposes, a patient is said to have Churg-Strauss syndrome if at least four of these criteria are present. The presence of any four or more criteria has a sensitivity of 85% and a specificity of 99.7%. From Masi AT, Hunder GG, Lie JT, et al: The American College of Rheumatology 1990 criteria for the classification of Churg-Strauss syndrome (allergic granulomatosis and angiitis). Arthritis Rheum 33: 1094, 1990. Copyright © 1990 John Wiley & Sons, Inc. Reprinted with permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
a 10-year period ending in 1988 in the United States.7 In the series reported by Rottem and associates,4 one patient died of severe lung disease and cor pulmonale, and one died of sepsis. Long-term follow-up is needed to appreciate the extent of the disease. Significant morbidity and relapses can occur with childhood-onset disease.52
Churg-Strauss Syndrome (Allergic Granulomatosis)
Definition and Classification Criteria Criteria for the classification of Churg-Strauss syndrome were developed by the ACR in a study of 20 patients and 787 controls (Table 26–6).57,58 This disease characteristically affects middle-aged males59 and is rare in children. Ten patients who were 16 years old or younger have been reported (Table 26–7).56,60–67 The cause is unknown.
Clinical Manifestations
53–55
Churg and Strauss originally described the disorder that bears their names in 1951 in a report of 13 patients with severe asthma associated with fever, eosinophilia, and vasculitis affecting various organ systems. They later broadened the initial description to include eosinophilic pneumonitis, angiitis, allergic granulomata, and necrotizing angiitis. Granulomatous extravascular and vascular changes are characteristic.
Churg-Strauss syndrome usually occurs in conjunction with a long history of asthma. Other allergic manifestations such as chronic allergic rhinitis may be present, particularly in the prodromal or early phase, which may last several years.2 Eosinophilia, pulmonary infiltrates, and vasculitis follow. Not all components may be present at one time; the disease may first occur as a localized disorder. In the ACR classification study57 (see Table 26–6),
TABLE 26–7 Reported Cases of Churg-Strauss Syndrome in Childhood Study
Sex
Age (yr)
F
7
M
9
Farooki et al60 Petty et al61
M M
12 16
Frayha62
F
13
Treitman et al Heine et al64 Jessuran et al65
M F M
14 4 14
Rabusin et al66
M
2
Mpofu et al67
M
7
56
Churg and Strauss
63
Clinical Manifestations Asthma, eosinophilia, pneumonitis, hypertension, skin nodules and purpura, cardiac failure, nephritis, death Asthma, eosinophilia, pneumonitis, hypertension, skin nodules and purpura, cardiac failure, nephritis, peripheral neuropathy Pericarditis, myocarditis, eosinophilia, history of wheezing, death Fever, asthma, painful calf nodules, peripheral neuropathy, hypertension, eosinophilia Fever, asthma, peripheral neuropathy, celiac aneurysms, paresthesias, eosinophilia, death Asthma, sinusitis, pulmonary basilar infiltrates, cardiomegaly, eosinophilia, death Fever, pneumonia, pseudotumor, eosinophilia Cough, weight loss, skin lesions, eosinophilia, mediastinal granulomata with central necrosis, eosinophils; arteries infiltrated with histiocytes, eosinophils, and giant cells Asthma, eosinophilia, food allergies, eosinophilic myocarditis, eosinophilic infiltrates in skin, lung; death Fever, asthma, eosinophilia, skin lesions, proteinuria, pulmonary infiltrates
546
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
all patients had pulmonary infiltrates and neuropathy (especially mononeuritis multiplex), and 83% had paranasal sinus infection. The vasculitis may be clinically indistinguishable from polyarteritis nodosa or hypersensitivity angiitis.58 In one pediatric patient, microaneurysms typical of those occurring in polyarteritis nodosa were present in the hepatic artery and celiac axis.62 Overlap features have been described in other pediatric patients, including the presence of pANCA in a patient with ocular pseudotumor.64 Additional skin manifestations include purpura (representing leukocytoclastic vasculitis), maculopapular rash, cutaneous nodules (extravascular granulomata),60 livedo reticularis, ulcers, and bullae.67 Cardiac involvement was prominent in the patients described by Churg and Strauss and included granulomatous pericardial disease68 and eosinophilic myocarditis. Renal disease is usually mild and rarely progresses.59,69 Myalgia, arthralgia, and arthritis can occur. Hypertension and ocular involvement, in addition to pseudotumor cerebri, occur in some patients.70 Neuropathy with mononeuritis multiplex is an important and common cause of morbidity.71 Gastrointestinal ulceration and granulomata of the omentum have been reported.62 Testicular pain can be an early clinical manifestation.72
Pathology Histopathologic studies confirm vasculitis of small arteries and veins associated with necrotizing extravascular granulomata and eosinophilic infiltrates. These eosinophilic exudates with necrosis, fibrinoid changes, and granulomatous proliferation of epithelioid and giant cells are characteristic (Fig. 26–6).68
Differential Diagnosis The differential diagnosis includes other forms of vasculitis such as WG, polyarteritis nodosa, and HenochSchönlein purpura. The diagnosis is based on the presence of two or more of the clinical criteria described in Table 26–3 and confirmed by biopsy of renal, skin, or lung tissue. Transbronchial biopsy is less invasive than transthoracic biopsy for obtaining lung tissue.63 The diagnosis should be considered in an asthmatic patient with fever, deteriorating clinical course, and increasing eosinophilia. A syndrome similar to the Churg-Strauss syndrome has been reported after the use of the leukotriene antagonist zafirlukast in adult patients with asthma.63 This disorder was characterized by pulmonary infiltrates, eosinophilia, neuropathy, sinusitis, rash, fever, and muscle pain. Acute dilated cardiomyopathy was also characteristic of the zafirlukast-associated syndrome but is not a typical feature of the Churg-Strauss syndrome.
Laboratory Examination Elevation of acute phase reactants accompanies active disease. Peripheral blood eosinophilia (with eosinophils accounting for 10% or more of leukocytes) and elevation of serum levels of IgE are typical. Chest radiographs may reveal diffuse pulmonary infiltrates (Fig. 26–7), and pulmonary function tests demonstrate poor lung diffusing capacity and low PO2. ANCAs are common,74,75 but other autoantibodies are usually not present.
Treatment There is at least an initial response to high-dose glucocorticoids, but immunosuppressive agents such as cyclophosphamide or methotrexate often are required.58 The use of interferon-α was reported to be beneficial in four patients with Churg-Strauss syndrome that were resistant to a combination of glucocorticoids and cyclophosphamide or methotrexate.76 Plasmapheresis may also have a role in the management of the Churg-Strauss syndrome.77
Course of the Disease and Prognosis The long-term follow-up of 96 patients with the ChurgStrauss syndrome reported by Guillevin and colleagues77 revealed a 24% mortality rate. In this study, development of severe myocardial or gastrointestinal disease was associated with a poor outcome. The disease course is often very prolonged, however, and therapy with glucocorticoids with or without other drugs is usually required for years.
Lymphomatoid Granulomatosis
■ Figure 26–6 Lung biopsy specimen from a young girl with allergic granulomatosis. No definite vasculitis is identified, but there are necrotizing granulomata with giant cells (arrow). H & E, × 480.
Lymphomatoid granulomatosus, first described in 1972 by Liebow and coworkers,78 is a rare necrotizing pulmonary vasculitis that is often fatal. It may initially resemble WG but then progress to lymphoma.78–81 It is sometimes associated with immunodeficiencies (e.g., Wiskott-Aldrich syndrome, X-linked lymphoproliferative syndrome), human
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
547
■ Figure 26–7 Chest radiographs of a young man with allergic granulomatosis.The initial film on the left demonstrates an enlarged cardiac silhouette and relatively normal pulmonary fields.The film on the right was taken during an acute episode of left- and right-sided heart failure. Notice the cardiac enlargement and disappearance of the normal pulmonary vascular markings, indicating acute pulmonary hypertension and cor pulmonale, in this case on the basis of vasculitis.
immunodeficiency virus (HIV) infection, and malignancy.82–86 The disease has been reported in several adult patients who had undergone renal transplantation.87–89 Descriptions of this disease in children have been rare and include an infant,82 a 16-year-old boy,79 a child with leukemia in remission,86 and a child with the WiskottAldrich syndrome.84 The primary clinical manifestations include fever, pneumonia, lymphadenopathy, and (particularly in children) failure to thrive and recurrent infections, including otitis media and sinusitis. Other organs and systems involved include kidneys, liver, skin, and the CNS. Midline sinus or upper respiratory disease is rare. The characteristic lesions are angiocentric, necrotizing granulomata with cellular infiltrates consisting of immunoblasts, lymphocytes, histiocytes, plasma cells, and eosinophils.59 Manifestations include thrombosis and necrosis of the adjacent parenchyma (i.e., necrotic nodules involving vascular infiltration surrounded by areas of parenchymal necrosis).
children. A series of children were identified with biopsyproven PACNS but involving very small vessels that were not detected by angiographic study.93
Clinical Manifestations The clinical signs and symptoms vary. Headache occurs in up to 75% of patients at disease onset.94,95 Other common neurologic manifestations include transient ischemic attacks, paresis, seizures, visual loss, neurocognitive impairment, and progressive encephalopathy. Systemic signs and symptoms are absent. Most patients have an abrupt onset of symptoms several weeks before diagnosis.94,95 The study of Calabrese and Mallek96 included a 10-year-old girl and a 10-year-old boy, both with hemiparesis. Intracranial hemorrhage may be the initial manifestation.97,98 There appears to be an increased risk in the postpartum period.95,99 The diagnosis should be considered in children with acute onset of unexplained neurologic deficits.94,100
Primary Angiitis of the Central Nervous System
Laboratory Examination
Primary angiitis of the central nervous system (PACNS), first called granulomatous angiitis,90 is now recognized as a distinct form of vasculitis.91 It occurs predominantly in whites of either sex between the ages of 35 and 50 years. There are fewer than 50 reports of children with PACNS. The marked variation observed in the disease course led Calabrese and associates92 to subdivide the disease into PACNS and benign angiitis of the central nervous system (BACNS). The benign form is a mild, monophasic illness that usually responds well to glucocorticoids and resolves with minimal residua over a period of several weeks. BACNS is rare in
Leukocytosis is common and is occasionally associated with thrombocytosis and antinuclear antibodies. Antiphospholipid antibodies do not occur. Complement activation has been reported.101 Cerebrospinal fluid analysis is usually characterized by an increased protein level and mild pleocytosis.95 Disease is restricted to small and mediumsized arteries and venules of the brain and spinal cord.102 Infiltration of mononuclear cells and granuloma formation have been found. If a patient is suspected of having PACNS but the radiographic studies are not supportive, a tissue biopsy should be obtained.
548
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
Radiologic Examination The diagnosis is usually made by angiography, which demonstrates beading or alternating ectasia and stenosis of intracranial arteries (Fig. 26–8).103 Abu-Shakra95 found abnormalities in 73% of patients by CT, in 77% by magnetic resonance imaging (MRI), and in three of four patients studied by single photon emission CT and pertechnetate isotope brain scans. Other studies have shown a higher frequency of MRI abnormalities.104
Treatment and Outcome PACNS was initially thought to be uniformly fatal, but with the availability of improved imaging techniques allowing earlier diagnosis, an improved outcome has been reported.105,106 Nonetheless, morbidity is significant, and recurrences are frequent. When patients were treated with glucocorticoids and cyclophosphamide, Woolfenden and colleagues104 reported that patients developed no new lesions but old lesions did not resolve. In another study, glucocorticoids alone or in combination with cyclophosphamide were used if visual loss, stroke, or severe neurocognitive disease developed.95 Eighty percent of this group stabilized, but 12% experienced relapse after a median of 2 months. Two studies95,104 showed significant permanent neurologic deficits, including paresis, visual
loss, and seizures in patients treated with glucocorticoids or glucocorticoids and cyclophosphamide.
Moyamoya Disease First described in 1969,107 moyamoya (“puff of smoke” in Japanese) is a term applied to the appearance of the cerebral angiograms in patients with hemiplegia associated with supraclinoid carotid stenosis and multiple cerebral telangiectasia. This angiographic appearance is related to the development of multiple collateral vessels. Approximately 25% of patients are younger than 10 years,108 and 90% present with hemiplegia.108,109 Other manifestations include alteration of consciousness, speech or visual disturbances, and seizures.110 Characteristic vessel abnormalities apparent on angiography include multiple collateral vessels and anastomoses of meningeal vessels with internal carotid vessels (i.e., rete mirabile) (Fig. 26–9). Diffusion-weighted MRI offers a noninvasive means of early detection of ischemic lesions and monitoring of the clinical course.111 Surgical extracranialintracranial bypass procedures have been useful.112 Calcium channel blockers may be helpful in some patients.113 The prognosis may be worse in patients younger than 16 years, and recovery or stabilization was observed in only 40% of that age group.108 Congenital arterial dysplasia and changes due to nonspecific vascular injury have been hypothesized as causes.114–116 A number of clinical associations have been described, including Down syndrome,108,117 occlusive peripheral vascular disease,116 primary pulmonary hypertension,118 and other cerebrovascular malformations.119 The familial occurrence of moyamoya disease has been described.120 In adults, further associations include myopathy, renal artery stenosis, Fanconi’s anemia, hemoglobinopathies, meningitis, and arteritis.108
GIANT CELL ARTERITIS A number of clinically different diseases are characterized by identical histologic abnormalities of a giant cell arteritis. The two major vasculitides of this type that occur in childhood and adolescence are Takayasu’s arteritis (TA) and temporal arteritis.
Takayasu’s Arteritis
■ Figure 26–8 Magnetic resonance angiogram of a 12-year-old girl who presented with mild, right-sided hemiparesis and headache.Test results were strongly positive for antinuclear antibody, negative for anti-dsDNA antibody, and positive for antibody to myeloperoxidase (pANCA). An obstruction to flow was demonstrated in the right posterior cerebral artery.The patient had no evidence of extracerebral vasculitis. She was treated with intravenous methylprednisolone and cyclophosphamide and has remained asymptomatic without progression of the vascular lesion.
TA, a chronic, inflammatory, large-vessel vasculitis affecting the aorta and its major branches, is most common in young women, particularly of Japanese origin. It was first described as pulseless disease.121,122 Takayasu, a Japanese ophthalmologist, first described the characteristic arteriovenous abnormalities of the retina in 1908.123 It is a panarteritis that initially involves the adventitia and then progresses to the intima.
Definition and Classification The ACR classification criteria are shown in Table 26–8.124 The presence of all six criteria has a sensitivity of 90.5% and a specificity of 97.8% for TA. In the patient popula-
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
549
■ Figure 26–9 Arteriography of the left vertebral artery shows stenosis of the basilar artery (arrow) just above the tentorium.The extensive collaterals are the characteristic “puff of smoke” pattern of moyamoya. A, Anteroposterior view. B, Lateral view. (A and B, Courtesy of Dr. O. Flodmark.)
tion analyzed for validation of these criteria, 18 (29%) of 63 patients were younger than 20 years at diagnosis. TA is categorized according to the distribution of affected vessels (Table 26–9).125,126 Type I affects the aortic arch; type II affects the thoracic and abdominal aorta; type III affects the aorta above and below the diaphragm; and type IV affects the aorta and the pulmonary artery.
Epidemiology Although rare in individuals before the age of 16 years, TA is the most common giant cell arteritis of childhood; worldwide, it follows Kawasaki disease and HenochSchönlein purpura as the most frequent of the vasculitides of childhood (see Table 22–8).127–129 In Japan, 20% of patients with TA are younger than 19 years, and 2% are younger than 10 years. Reports from South Africa,130 Canada,131 and Mexico132 document its occurrence in children around the world. Blacks, Asians, Hispanic Americans, and Sephardic Jews are most at risk. The
female/male ratio is about 8:1 in adults but closer to 4:1 to 2:1 in children.129,133,134 There are unexplained geographic variations in the presentation of TA. Obstructive lesions are the most common in the United States, Europe, and Japan, whereas aneurysms appear to be more common in India, Thailand, and Africa. Involvement of the brachiocephalic arteries occurs in patients in most geographic areas; the abdominal aorta is most often involved in reports from Thailand135 and the descending aorta in those from India.136,137 The significance of these differences, if real, is unknown.
Genetic Backg\round TA has been reported in families and in monozygotic twin sisters.138 Associations with specific HLA antigens in different racial groups have been reported. In Japanese patients, the haplotype A11 B40,139 the complotype Aw24-DW52C4A2-C4BQ0-Dw12,139 and B5201 and B3902140 have been TABLE 26–9 Takayasu’s Arteritis in Childhood—Patterns of Involvement
TABLE 26–8 Classification Criteria for Takayasu’s Arteritis Subclavian or aortic bruit Age < 40 yr at onset Decreased brachial artery pulse Blood pressure difference of > 10 mm between arms Claudication of extremities Arteriographic evidence of narrowing or occlusion of aorta, its primary branches or large arteries in the proximal, upper, or lower extremities From Arend WP, Michel BA, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of Takayasu arteritis. Arthritis Rheum 33: 1129, 1990.
Type
Affected Vessels
I
Aortic arch only Aortic arch and descending thoracic aorta Aortic arch, thoracic and abdominal aorta Aortic arch and abdominal aorta Descending thoracic aorta only Descending thoracic and abdominal aorta Diffuse aortic involvement Diffuse aortic and pulmonary artery involvement
II III IV
Frequency (%)
Data from published reports of Takayasu’s arteritis in children.
5 19 16 19 7 19 0 2
550
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
documented, whereas in patients from northern India, there is a marked increase in the frequency of B5 compared with an ethnically matched control population (relative risk = 4.3). B51 and B52 were equally represented in this population.141 A weaker association with DR8 was found in the same group. Possible associations with DR6 (DRB1*1301) were observed in a Mexican population.142
Etiology and Pathogenesis The cause of TA is unknown. An association with tuberculosis in some areas of the world has been suggested but not proved.143,145 In India, patients with TA have an increased humoral immune response to the 65-kD heatshock protein from Mycobacterium tuberculosis.146 Whether this indicates a role for tuberculosis in the etiopathogenesis of TA or is an indication that there is cross-reactivity with other heat-shock proteins or cross-reacting selfantigens is not clear.
Clinical Manifestations Hypertension appears to be the most common complaint, followed by headache.134 However, fever, back pain, claudication, or visual complaints may also be the presenting manifestation. An early inflammatory phase may manifest with nonspecific myalgias, arthralgias, fever, and chills, together with a marked acute phase laboratory response. It is often challenging to determine whether the manifestations are caused by “active” disease or the consequences of active disease.
Laboratory Examination Laboratory findings are nonspecific and include elevation of the erythrocyte sedimentation rate, moderate anemia, mild leukocytosis, and hypergammaglobulinemia. Antinuclear antibodies are rare, and rheumatoid factors are only occasionally present. ANCAs are seldom found,147,148 but antibodies to endothelial cells are frequently detected.147 Electrocardiograms may confirm left ventricular hypertrophy, which is strongly associated with the presence of hypertension.149
Radiologic Examination Chest radiographs may demonstrate cardiomegaly or an irregular contour of the aortic arch and descending aorta. There may be areas of calcification or widening related to prestenotic dilatation (Figs. 26–10 and 26–11). Arteriography of the aorta and its major branches may demonstrate multiple sites of segmental involvement.150 Transfemoral digital subtraction angiography avoids the risk of catheter manipulation in the presence of severe aortic disease.151 The usefulness of such studies may be limited by cardiac failure and motion artifacts. Sonography and Doppler echocardiography may be useful in demonstrating flow turbulence.152,153 Echocardiography may be needed to assess the aortic root in coronary artery disease. In one study, duplex color-flow Doppler demonstrated decreased pulses and vascular bruits and was superior to MRI in defining mural thickening in involved vessels.154
MR angiography, CT, and ultrasonography may all document arterial early inflammatory changes occurring before the development of obstructive lesions.155
Treatment Treatment requires high-dose glucocorticoids (1 to 2 mg/ kg/day) for an initial period of 4 to 8 weeks, followed by a judicious reduction of the dose. The response to glucocorticoids depends on the activity of the disease; not all patients respond.156 Cytotoxic drug therapy has been used, but its efficacy has not been established. Low-dose oral methotrexate may be an effective steroidsparing agent.157 Pilot data suggest anti–tumor necrosis factor agents may be useful. A remission lasting longer than 1 year and tapering of glucocorticoids to a very low dose or none were achieved in 52% of 48 patients.158 Aggressive treatment of hypertension is critical. Use of angiotensin-converting enzyme (ACE) inhibitors should be withheld unless other antihypertensive therapy has failed, because these drugs may diminish renal function. Antiplatelet agents (i.e., low-dose aspirin or dipyridamole) may be helpful in preventing thrombosis in abnormal vessels. Surgical interventions for severe renal hypertension, cerebral hypoperfusion, claudication, or aneurysm formation have had various degrees of success. Percutaneous transluminal renal angioplasty was successful in 80% of Indian children subjected to the procedure.159 Percutaneous expandable renal and aortic stents have been placed with success.155,160–162 When feasible, renal autotransplantation may be the definitive procedure for some patients if other measures fail to control renovascular hypertension.144,155
Course of the Disease and Prognosis The course is prolonged and variable, and relapses occur despite therapy. A prospective study in adults demonstrated an 83% 5-year survival rate and a 58% 10-year survival rate.126 In a study of 11 patients who met ACR criteria for the diagnosis of TA, 1 patient died and 5 patients required renal artery transplantation, resulting in preserved renal function in 4.156 The length of time between disease onset and institution of therapy is critical to the outcome, because glucocorticoid therapy is unlikely to alter vascular lesions in which fibrosis with narrowing has already taken place.
Temporal (Cranial) Arteritis Classic giant cell arteritis affecting the temporal artery is a common form of vasculitis in elderly patients but is rare in childhood and adolescence. Inflammation involves primarily the carotid artery and its branches. Headache, localized pain, and tenderness over the temporal arteries are occasionally accompanied by jaw or face claudication. Disease of the ophthalmic and central retinal vessels may cause blindness. Studies suggest that the arterial damage results from an antigen-driven immune response.163 Temporal arteritis is often associated with polymyalgia rheumatica in the adult population but not in children.
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
551
■ Figure 26–10 Four studies document the lesions of Takayasu’s arteritis. A, Angiography shows dilatation, irregularity, and stenosis of the right innominate artery and its branches (arrow).The left subclavian artery is not visualized because of occlusion. B, This gallium scan shows increased uptake of the isotope in the region of the aortic arch (arrow), supporting the diagnosis of inflammation of this structure. C, Magnetic resonance imaging shows the dilated aortic arch (long arrow) and localized aneurysms of the thoracic portion of the descending aorta (short arrows). D, Stenotic region of the abdominal aorta (arrow). (B, Courtesy of Dr. H. Nadel; D, Courtesy of Dr. G. Culham.)
Juvenile temporal arteritis is a non–giant cell, nonnecrotizing vasculitis of the temporal arteries of unknown cause that was initially described by Lie and associates in 1975.164 It is a specific form of temporal arteritis that differs from the classic disease. It has been reported in seven children164,165 and occasionally in adults.166 There are no systemic symptoms. The lesions are observed accidentally as pea-sized nodules in the temporal area. The condition is usually bilateral. Patients with juvenile temporal arteritis have a benign course that does not require glucocorticoid therapy. Excisional biopsy has been curative, with no evidence of recurrence. Similar involvement of the occipital artery has also been described.167
SARCOIDOSIS Sarcoidosis is an uncommon multisystem disease of unknown origin. Two patterns of clinical manifestations are reported in children. Early descriptions of the disorder were primarily of older children or adolescents who had lung disease, lymphadenopathy, weight loss, fever, and hypercalcemia but little or no joint disease.168–174 In 1966, Harris and colleagues175 described patients younger than 4 years at onset of sarcoidosis characterized by the clinical triad of skin, joint, and eye disease. Subsequent series have largely confirmed these two distinct patterns of disease,176–179 although many exceptions have been reported.180,181
552
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S Genetic Background There have been numerous reports of a familial occurrence of sarcoidosis in parent-offspring or sibling pairs.192–198 In an international registry, a familial pattern was identified in 11 of 53 children with sarcoid arthritis.185 Patterns of inheritance in a study from the United Kingdom of 80 black patients from 11 families supported multigenic inheritance.199 The incidence of familial disease was three times greater in Los Angeles than in London, and a pattern consistent with autosomal recessive inheritance was evident.200 Studies of HLA antigens suggest associations of sarcoidosis with DQB1*0603, DQB1*0604, and DPB1*0201 (Glu positive).201
Clinical Manifestations Sarcoidosis may be asymptomatic, and the diagnosis may therefore be delayed. Only 11 of 18 children in one study were symptomatic; hilar adenopathy seen on routine chest radiographs was the first indication of the disease.170 Symptomatic children were younger, had more extensive disease, and had a more complicated course. Duration of symptoms before diagnosis in one study was 7.6 months.179 In this group, 75% had more than one area of involvement. Six of the seven asymptomatic children were well at follow-up. In general, symptoms and presentation vary greatly.202
Lymphadenopathy and Hepatosplenomegaly
■ Figure 26–11 A, Dilatation of the abdominal aorta. B, Ultrasound study (sagittal section) shows thickening of the aortic wall (arrow) in a child with active Takayasu’s aortitis. (A, Courtesy of Dr. G. Culham; B, Courtesy of Dr. A. R. Buckley.)
Lymphadenopathy is the most common initial manifestation of sarcoidosis. The lymph nodes are usually firm, mobile, and nontender.170,179 Retroperitoneal adenopathy is common.203 In a study of children with sarcoid arthritis, 9 of 12 children had hepatosplenomegaly.180
Skin Disease
Epidemiology Sarcoidosis is primarily a disease of adults in the 20- to 40-year age range and is almost twice as common in women as in men.182,183 Approximately 3% of cases in a 1953 study were in patients younger than15 years.168 In a study from eastern Hungary, however, 15% of patients were children between 8 and 14 years old.174 In 1983, there were 325 pediatric cases in the English language literature, 15 (5%) of which included arthritis.184 In an international registry study of children with sarcoidosis associated with joint disease, the mean age at onset was 10.6 years (range, 0.1 to 16 years). Thirty-eight (72%) of 53 patients had onset before the age of 5 years.185 The prevalence of sarcoidosis is high among Japanese children,182,186 and the disease is more common in African American children than in white children, although the racial distribution varies with geographic location.168,170,173,179,187–191 Within the United States, approximately 80% of reported patients live in the southeastern part of the country, particularly in rural areas.170,172,179 The onset of sarcoidosis is more likely between December and May.179
Skin disease occurs in approximately 30% of children with sarcoidosis.172,204 The lesions may be maculopapular, vesicular, papular, or nodular (including erythema nodosum). An association between ichthyosiform cutaneous lesions and severe joint disease has been made.205 Nodular subcutaneous sarcoid lesions also occur and may represent isolated musculocutaneous involvement.206,207 Sarcoidosis should always be included in the differential diagnosis of unusual skin lesions (Fig. 26–12).
Eye Disease Ocular abnormalities are common causes of morbidity in sarcoidosis and may be the presenting manifestations.208 In a study of uveitis associated with systemic diseases, 3 of 340 children were found to have sarcoidosis.209 However, in a population of children with sarcoidosis and joint disease, the incidence of eye involvement is higher. All of the 12 patients reported by Hafner and associates180 had iridocyclitis; 8 had residual eye damage. Uveitis may be more common in early-onset disease than in later-onset disease. In children between 8 and 15 years old, 32% had eye involvement.169,170,172 In another study,210
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
553
■ Figure 26–13 Conjunctival nodules are evident in this everted lower eyelid of a child with sarcoidosis.
than 4 years, 22% had lung disease in one study.178 In children with sarcoid arthritis, pulmonary disease occurred in almost one half, and in about 15%, it was severe.180 Hilar and paratracheal adenopathy and parenchymal involvement are common.187 Parenchymal disease, characterized by small, irregular interstitial infiltrates, pleural effusions, and atelectasis,218 is less common in children than in adults, in whom it occurs in one fourth of patients.187 Restrictive disease is most characteristic.179 ■ Figure 26–12 Many of the lesions visible on the legs of this child with sarcoidosis are nodular.The child also has sarcoid arthritis in the knees and ankles.
77% of children with sarcoidosis younger than 5 years had eye disease. Posterior uveitis also occurs. In the sarcoid registry, 44 of 53 children had uveitis, which was bilateral in 43.185 The anterior uveal tract, posterior uveal tract, or both were involved.185 Posterior or panuveitis may be associated with CNS involvement.211 Other manifestations are diverse and include lacrimal gland swelling, orbital granulomata, conjunctival granulomata (Fig. 26–13), vitritis, chorioretinitis, optic nerve involvement, and proptosis.210,212–214 Interstitial keratitis is uncommon. Eye involvement may precede joint or pulmonary manifestations.215 Young children may be asymptomatic, even though blind at the time of diagnosis.169,170,216 The pattern of eye involvement differs from that of juvenile rheumatoid arthritis. The tightly packed corneal accumulations of lymphocytes (i.e., keratic precipitates) are often larger and in the peripheral cornea may be confluent (i.e., a “snowbank” appearance).217 Similar accumulations may be present at the iris-pupil margin (i.e., Koeppe’s nodules).217
Pulmonary Disease Chronic cough is the second most common presenting symptom of sarcoidosis.179 Of 53 children in the international registry, 5 (9%) had lung disease.185 Although bronchopulmonary disease is less common in children younger
Neurologic Abnormalities Sarcoidosis has a plethora of neurologic manifestations.219 In one study of children with sarcoid arthritis, 30% had CNS abnormalities, including encephalopathy and seizures.180 In adults, CNS involvement is reported in 5% to 10%,220–226 most commonly affecting the brain and peripheral nerves.223–225 Of the cranial nerves, the seventh is most frequently affected, but involvement of the optic, acoustic, vagus, and glossopharyngeal nerves has been described.226,227 Mass lesions are much more common in the cerebrum than in the posterior fossa219,227 and may be clinically silent.228 Obstructive hydrocephalus, aseptic meningitis, myelopathy, general and focal seizures, pituitary-hypothalamic lesions, and spinal cord involvement also occur.227–230 Three cases of intraspinal sarcoid without systemic manifestations have been described.220 Children are more likely to have seizures and spaceoccupying lesions than adults and less likely to have cranial nerve palsies.231
Musculoskeletal Disease Musculoskeletal involvement varies from less than 5% to 50% in reported series of children with sarcoidosis.179 Fewer than a dozen cases of sarcoid arthritis were described in children before 1970.232 Musculoskeletal manifestations of sarcoidosis include arthralgia, arthritis, bone abnormalities, and muscle involvement. The arthritis is characterized by a boggy synovial thickening with large effusions of joints and tendon sheaths. There is
554
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
often minimal pain, stiffness, or limitation of motion.232 Prolonged synovitis may not be associated with radiographic evidence of erosion or osteoporosis. Although initial joint involvement may be transient and affect only a few joints, a polyarticular pattern generally evolves over a period of several years.205,232 As the duration of disease increases, the arthritis more closely resembles that of juvenile rheumatoid arthritis, with increasing limitation of motion, stiffness, fusiform swelling of the fingers, and a high incidence of cervical spine involvement. This type of deforming polyarthritis has been described with increasing frequency.217,233 Eight of 12 patients in one study had progressive articular disease.180 Transient polyarthritis occurs in 6% to 25% of adults but later may become chronic and destructive (Fig. 26–14).234 Osseous sarcoid is uncommon in children, although small bones of the hands and feet may be involved. Vertebral sarcoid can occur and manifest as back pain.235 Up to 50% of adults have muscle disease,236,237 although it may be symptomatic in less than 5%.238 Muscle involvement in children is rare but was described as a cause of myalgia with associated electromyographic abnormalities in a 9-year-old boy with symptoms for 5 years.239 The spectrum of muscle involvement includes nodular lesions and inflammatory myositis.183,240,241
Other Manifestations Gastrointestinal tract involvement is uncommon, but when it occurs, the stomach is most frequently affected.242 Intestinal obstruction with gastric and small bowel involvement243 and rectal prolapse218 have been reported. Bilateral parotid swelling has been described,244 and in one study,180 one third of patients had a sicca syndrome, including lacrimal and submandibular involvement. Sarcoidosis can manifest as a testicular mass.245 In one study,180 8 of 12 children had pericardial effusions and chronic pericarditis. Involvement of the myocardium can also occur.153,246,247 Renal involvement is rare in adults
and children.170,176–178,248–256 It may be asymptomatic or manifest as polyuria (from interstitial nephritis) or even eneuresis.252,253 These signs have been associated with bilateral enlargement of the kidneys.254 Nephrocalcinosis may be precipitated by hypercalciuria.179,256
Differential Diagnosis Sarcoidosis should be suspected in any child with unexplained lymphadenopathy, hepatomegaly, uveitis, enlargement of the salivary or lacrimal glands, cystic bone lesions of the hand and feet, or cutaneous infiltrates or papules, especially if the patient has an associated arthritis. Several rare autosomal dominant familial syndromes should be considered in the differential diagnosis.257,258 Blau259 described 11 family members with a multisystem granulomatous disease characterized by uveitis, joint and skin disease, and camptodactyly associated with elevation of serum levels of ACE. They had negative KveimSiltzbach skin test results and no lung disease. Later studies disclosed comma-shaped and wormlike bodies on electron microscopy of the granulomas.260 Additional similar families have been reported.258,261–264 Linkage analysis studies have demonstrated a susceptibility locus for Blau’s disease on the pericentromeric region of chromosome 16 (16p12-16q21),265,266 and it is believed to result from mutations in the CARD15 gene.267 Jabs268 described familial granulomatous synovitis associated with uveitis and cranial neuropathies but without skin disease in four members of one family. The relationship of this syndrome to that described by Blau and of both syndromes to sarcoidosis is not certain. They may be variants of the same disorder. A wide spectrum of vasculitides, including small-, medium-, and large-vessel disease, granulomatous vasculitis, and leukocytoclastic vasculitis has been associated with sarcoidosis.269–271 Aortic arch disease identical to that of TA,269 carotid and subclavian arteritis,272 and abdominal aortic aneurysms have been reported in patients with
■ Figure 26–14 Synovial biopsy from a child with sarcoid arthritis. A, Giant cells (arrows). B, Noncaseating granuloma. A and B, H & E, magnification × 480.
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
sarcoidosis.273 Necrotizing sarcoid granulomatosis consisting of sarcoid-like granulomas, granulomatous vasculitis, and necrosis may represent a variant and has been reported in two children.274
Laboratory Examination No laboratory test is diagnostic of sarcoidosis. Leukopenia, eosinophilia, increased levels of immunoglobulins, and elevated acute phase reactants are common. Hypercalciuria, with or without nephrocalcinosis, occurs in most patients.179 The serum calcium level may be normal, even in multisystem disease.180 Hoffman274 described a 15-yearold who presented with hypercalcemic crisis, and hypercalcemia has been reported in an infant with sarcoidosis.275 Hypercalciuria can occur with normal serum levels of calcium.276 Hypercalcemia appears to result from abnormal pulmonary alveolar macrophages. The cells and homogenates of lymph nodes are capable of synthesizing 1,25-dihydroxyvitamin D from 25-hydroxyvitamin D, and these cells appear to be insensitive to regulatory feedback by hypercalcemia.277,278 Serum levels of ACE produced by epithelial cells in granulomata279 are elevated in up to 80% of children with sarcoidosis,280 a frequency similar to that seen in adults.281–284 ACE is less frequently elevated in very young children with sarcoidosis. Serum levels appear to correlate with disease activity and may be helpful in adjusting therapy.280,285 Levels of ACE in normal children are higher than in adults, and pediatric standards are important in assessing results.286 The Kveim-Siltzbach skin test (i.e., sarcoid granulomata resulting from intradermal injection of extract of spleen from a patient with sarcoidosis) is antiquated, and the standard test reagent is no longer available.287,288 Cutaneous delayed-type hypersensitivity to previously administered antigens is absent in approximately one half of patients. Such anergy is characteristic but not diagnostic of sarcoidosis. Anergy to purified protein derivatives of M. tuberculosis often occurs.
555
identify characteristic lytic bone lesions, which have been reported in up to 19% of patients170,173,190,191 and are most common in the metacarpal bones or vertebrae (Figs. 26–15 and 26–16). CT and ultrasonography are helpful in defining the nature and extent of orbital lesions.213,295 Demineralization of the orbital wall with bony destruction indicates sinus extension. MRI is particularly useful for the detection of basilar and meningeal abnormalities and also nodular lesions in the bone marrow.296,297 A common pattern of involvement includes orbits, parotids, and submandibular glands (“panda pattern”).298 A combination of gallium and thallium 201 myocardial scintigraphy is best for defining cardiac disease.299
Treatment Glucocorticoids have been the standard treatment for patients who are symptomatic and have significant organ involvement.169 In one study of sarcoid arthritis,180 glucocorticoids were effective in 11 of 12 patients, but most required 0.2 to 0.3 mg/kg of prednisolone per day as a maintenance dose, and all required azathioprine or methotrexate in addition. Low-dose methotrexate was reported to be effective, safe, and glucocorticoid sparing in a study of seven children.233 No adverse effects were found in this study. Other immunomodulating drugs,
Investigation of Pulmonary Disease Measurement of pulmonary function indicates that up to 50% of patients with sarcoidosis have restrictive lung disease, even if asymptomatic.179 Bronchial alveolar lavage (BAL) yields a threefold to fivefold increase in the number of lymphocytes and macrophages in children with sarcoidosis.179,289 The CD4+/CD8+ ratio is elevated. Macrophages demonstrate an increased release of hydrogen peroxide. With glucocorticoid therapy, the mononuclear cell abnormalities slowly normalize.290,291 Tessier and colleagues292 reported 11 children with pulmonary sarcoidosis who underwent BAL. Elevated levels of interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α, and transforming growth factor-β mRNA corresponded with disease activity and severity.
Radiologic Examination Chest radiography is the standard screening procedure for sarcoidosis of the lungs, although CT may better define details of specific lesions.293,294 Plain radiographs
■ Figure 26–15 Sarcoid arthritis involving the wrists and proximal interphalangeal joints in a 2-year-old boy.The destructive, lacelike, permeative lesion in the proximal end of the left fifth proximal phalanx is consistent with the osseous changes of sarcoidosis (arrow).
556
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S ual disease232 or may cause destructive polyarthritis.180 The latter group of patients also had a high frequency of cardiac, lung, and CNS involvement at 8 to 19 years of followup.180 Ocular sequelae are often very significant, and synechiae and band keratopathy may develop early.
R E F E R E N C E S
■ Figure 26–16 Sarcoid arthritis. A, Fenestrated lesions of bone and scalloping of osseous defects (arrow). B, Profound granulomatous destruction of phalanx resembling the changes seen in tuberculosis.
including cyclosporin, cyclophosphamide, and chloroquine, have demonstrated some benefit in adults.247,300–302 Ocular involvement with sarcoidosis usually responds well to glucocorticoids administered topically, locally, or systemically. Inhaled corticosteroids may be adequate for maintenance therapy of pulmonary sarcoidosis.303 Case reports have demonstrated the efficacy of infliximab.304–306
Course of the Disease and Prognosis Mortality rates for sarcoidosis vary with the geographic region, sex of the patient, and race. Mortality rates in most studies were higher for females than males295 and for African Americans than whites.247,307,308 Japanese children with bilateral hilar adenopathy had complete clearing of these abnormalities over a 2-year period.186 In Kendig’s series309 of 28 children, 5 had severe sequelae; restrictive lung disease occurred in 3 (1 died) and blindness in 2. Of 60 children and youths with sarcoidosis reported by Patishall and colleagues,179 47% had persistent abnormalities demonstrated on chest radiographs at 5-year follow-up. Another 35% had physical abnormalities, and 40% were normal. Abnormal lung function was demonstrated in 68% of 19 patients with childhood-onset sarcoidosis followed for a mean of 21 years; 5 patients were blind, had Bell’s palsy, vertebral lesions causing back pain, or cor pulmonale.310 Several studies have identified severe growth delay in children with early- or late-onset joint disease.181,311 Sarcoid arthritis may result in minimal morbidity and resid-
1. Wegener F: Ubers generalisierte septische Gefasserkrankungn. Verh Dtsch Ges Pathol 29: 202, 1936. 2. Klinger H: Grenzformen der Periarteritis nodosa. Z Pathol 42: 455, 1931. 3. Fahey J, Leonard E, Churg J, et al: Wegener’s granulomatosis. Am J Med 17: 168–179, 1954. 4. Rottem M, Fauci AS, Hallahan CW, et al: Wegener’s granulomatosis in children and adolescents: clinical presentation and outcome. J Pediatr 122: 26–31, 1993. 5. Leavitt RY, Fauci AS, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of Wegener’s granulomatosis. Arthritis Rheum 33: 1101–1107, 1990. 6. Carrington CB, Liebow AA: Limited forms of angiitis and granulomatosis of Wegener’s type. Am J Med 41: 497–527, 1966. 7. Cotch MF, Hoffman GS, Yerg DE, et al: The epidemiology of Wegener’s granulomatosis. Estimates of the five-year period prevalence, annual mortality, and geographic disease distribution from population-based data sources. Arthritis Rheum 39: 87–92, 1996. 8. Orlowski JP, Clough JD, Dyment PG: Wegener’s granulomatosis in the pediatric age group. Pediatrics 61: 83–90, 1978. 9. Belostotsky VM, Shah V, Dillon MJ: Clinical features in 17 paediatric patients with Wegener granulomatosis. Pediatr Nephrol 17: 754–761, 2002. 10. Stegeman CA, Travaert JW, Sluiter WJ, et al: Association of chronic nasal carriage of Staphylococcus aureus and higher relapse rates in Wegener’s granulomatosis. Ann Intern Med 120: 12–17, 1994. 11. Mayet WJ, Marker-Hermann E, Schlaak J, et al: Irregular cytokine pattern of CD4+ T lymphocytes in response to Staphylococcus aureus in patients with Wegener’s granulomatosis. Scand J Immunol 49: 585–594, 1999. 12. Stoney PJ, Davies W, Ho SF, et al: Wegener’s granulomatosis in two siblings: a family study. J Laryngol Otol 105: 123–124, 1991. 13. Hay EM, Beaman M, Ralston AJ, et al: Wegener’s granulomatosis occurring in siblings. Br J Rheumatol 30: 144–145, 1991. 14. Nowack R, Lehmann H, Flores-Suarez LF, et al: Familial occurrence of systemic vasculitis and rapidly progressive glomerulonephritis. Am J Kidney Dis 34: 364–373, 1999. 15. Hagen EC, Stegeman CA, D’Amaro J, et al: Decreased frequency of HLADR13DR6 in Wegener’s granulomatosis. Kidney Int 48: 801–805, 1995. 16. Dijstelbloem HM, Scheepers RH, Oost WW, et al: Fc gamma receptor polymorphisms in Wegener’s granulomatosis: risk factors for disease relapse. Arthritis Rheum 42: 1823–1827, 1999. 17. Zhou Y, Huang D, Farver C, Hoffman G: Relative importance of CCR5 and antineutrophil cytoplasmic antibodies in patients with Wegener’s granulomatosis. J Rheumatol 30: 1541–1547, 2003. 18. Bartfai Z, Gaede KI, Russell KA, et al: Different gender-associated genotype risks of Wegener’s granulomatosis and microscopic polyangiitis. Clin Immunol 109: 330–337, 2003. 19. Moorthy AV, Cheswney RW, Segar WE, et al: Wegener’s granulomatosis in childhood: prolonged survival following cytotoxic therapy. J Pediatr 91: 616–618, 1977. 20. Hall SL, Miller LC, Duggan E, et al: Wegener’s granulomatosis in pediatric patients. J Pediatr 106: 739–744, 1985. 21. Isaeva LA, Fedorova AN, Lysenka GA: Wegener’s granulomatosis in children. Pediatriia 51: 67–70, 1972. 22. Watters K, Russell J: Subglottic stenosis in Wegener’s granulomatosis and the Nitinol stent. Laryngoscope 113: 2222–2224, 2003. 23. Fauci AS, Haynes BF, Katz P, et al: Wegener’s granulomatosis: prospective clinical and therapeutic experience with 85 patients for 21 years. Ann Intern Med 98: 76–85, 1983. 24. Borchers T, Dirsch O, Schaper J, et al: Nodular pulmonary vasculitis in a twelve-year-old boy. Pediatr Pulmonol 37: 181–185, 2004. 25. Wolff SM, Fauci AS, Horn HG, et al: Wegener’s granulomatosis. Ann Intern Med 81: 513–525, 1974. 26. Drachman DA: Neurological complications of Wegener’s granulomatosis. Arch Neurol 8: 145, 1963. 27. Kosovsky PA, Ehlers KH, Rafal RB, et al: Case report: MR imaging of cardiac mass in Wegener’s granulomatosis. J Comput Assist Tomogr 15: 1028–1030, 1991. 28. Jennette JC, Falk RJ: Diagnostic classification of anti-neutrophil cytoplasmic autoantibody-associated vasculitides. Am J Kidney Dis 18: 184–187, 1991. 29. Hoffman GS, Leavitt RY, Kerr GS, et al: Treatment of Wegener’s granulomatosis with glucocorticoids and methotrexate. Arthritis Rheum 35: 1322–1329, 1992. 30. Specks U, Wheatley CL, McDonald TJ, et al: Anticytoplasmic autoantibodies in the diagnosis and follow-up of Wegener’s granulomatosis. Mayo Clin Proc 64: 28–40, 1989.
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
31. Nolle B, Specks U, Ludermann J, et al: Anticytoplasmic autoantibodies: their immunodiagnostic value in Wegener’s granulomatosis. Ann Intern Med 111: 28–40, 1989. 32. Csernok E, Holle J, Hellmich B, Willem J, et al: Evaluation of capture ELISA for detection of antineutrophil cytoplasmic antibodies directed against proteinase 3 in Wegener’s granulomatosis: first results from a multicenter study. Rheumatology 43: 174–180, 2003. 33. David A, Kacher Y, Specks U, Aviram I: Interaction of proteinase 3 with CD11b/CD18 (β2 integrin) on the cell membrane of human neutrophils. J Leukoc Biol 74: 551–557, 2003. 34. von Scheven E, Lee C, Berg BO: Pediatric Wegener’s granulomatosis complicated by central nervous system vasculitis. Pediatr Neurol 19: 317–319, 1998. 35. Roback SA, Herdman RC, Hoyer J, et al: Wegener’s granulomatosis in a child. Am J Dis Child 118: 608–614, 1969. 36. Rubin DF, Peterson P, Meltzer JI: Renal lesions in Wegener’s granulomatosis. Ann Intern Med 82: 849–850, 1975. 37. Bajema IM, Hagen EC, van der Woude FJ, Bruijn JA: Wegener’s granulomatosis: a meta-analysis of 349 literary case reports. J Lab Clin Med 129: 17–22, 1997. 38. Felson B: Less familiar roentgen patterns of pulmonary granulomas. Am J Roentgenol Radium Ther Nucl Med 81: 211–223, 1959. 39. McGregor MBB, Sandler G: Wegener’s granulomatosis: a clinical and radiological survey. Br J Radiol 37: 430–439, 1964. 40. Connally B, Manson D, Eberhard A, et al: Am J Roentgenol 167: 901–904, 1996. 41. Baliga R, Chang CH, Bidani AK: A case of generalized Wegener’s granulomatosis in childhood: successful therapy with cyclophosphamide. Pediatrics 61: 286–290, 1978. 42. Harrison HL, Linshaw MA, Lindsley CB, et al: Bolus corticosteroids and cyclophosphamide for initial treatment of Wegener’s granulomatosis. JAMA 244: 1599–1600, 1980. 43. Hoffman GS, Leavitt RY, Fleisher TA, et al: Treatment of Wegener’s granulomatosis with intermittent high dose intravenous cyclophosphamide. Am J Med 89: 403–410, 1990. 44. Hoffman GS, Kerr GS, Leavitt RY, et al: Wegener’s granulomatosis: an analysis of 158 patients. Ann Intern Med 116: 488–498, 1992. 44a. Gottlieb BS, Miller LC, Ilowite NT: Methotrexate treatment of Wegener’s granulomatosis in children. J Pediatr 129: 604–607, 1996. 45. DeRemee RA: The treatment of Wegener’s granulomatosis with trimethoprim/sulfamethoxazole: illusion or vision? Arthritis Rheum 31: 1068–1074, 1988. 46. Leavitt RY, Hoffman GS, Fauci AS: Response: the role of trimethoprim/sulfamethoxazole in the treatment of Wegener’s granulomatosis. Arthritis Rheum 31: 1073–1074, 1988. 47. Metzler C, Fink C, Lamprecht P, et al: Maintenance of remission with leflunomide in Wegener’s granulomatosis. Rheumatology (Oxf ) 43: 315–320, 2004. 48. Hermon MM, Golej J, Emminger W, et al: Acute hemorrhagic respiratory failure caused by Wegener’s granulomatosis successfully treated by bronchoalveolar lavage with diluted surfactant. Wien Klin Wochenschr 115: 793–796, 2003. 49. Stone JH, Talor M, Stebbing J, et al: Test Characteristics of immunofluorescence and ELISA tests in 856 consecutive patients with possible ANCA-associated conditions. Arthritis Care Res 13: 424–434, 2000. 50. The Wegener’s Granulomatosis Etanercept Trial Research Group. Limited versus severe Wegener’s granulomatosis: baseline data on patients in the Wegener’s Granulomatosis Etanercept Trial. Arthritis Rheum 48: 2299–2309, 2003. 51. Specks U, Fervenza FC, McDonald TJ, Hogan MC: Response of Wegener’s granulomatosis to anti-CD20 chimeric monoclonal antibody therapy. Arthritis Rheum 44: 2836–2840, 2001. 52. Knoch DW, Lucarelli MJ, Dortzbach RK, Smith ME: Limited Wegener granulomatosis with 40 years of follow-up. Arch Ophthalmol 121: 1640–1642, 2003. 53. de Groot K, Reinhold-Keller E, Tatsis E, et al: Therapy for the maintenance of remission in sixty-five patients with generalized Wegener’s granulomatosis. Arthritis Rheum 39: 2052–2061, 1996. 54. Guillevin L, Cordier JF, Lhote F, et al: A prospective randomized trial comparing steroids with pulse cyclophosphamide versus steroids and oral cyclophosphamide in the treatment of generalized Wegener’s granulomatosis. Arthritis Rheum 40: 2187–2198, 1997. 55. Langford CA, Taler-Williams C, Barron KS, Sneller MC: A staged approach to the treatment of Wegener’s granulomatosis: induction of remission with glucocorticoids and daily cyclophosphamide switching to methotrexate for remission maintenance. Arthritis Rheum 42: 2666–2673, 1999. 56. Churg J, Strauss L: Allergic granulomatosis, allergic angiitis, and periarteritis nodosa. Am J Pathol 27: 277–301, 1951. 57. Masi AT, Hunder GG, Lie JT, et al: The American College of Rheumatology 1990 criteria for the classification of Churg-Strauss syndrome (allergic granulomatosis and angiitis). Arthritis Rheum 33: 1094–1100, 1990. 58. Lie JT: The classification of vasculitis and a reappraisal of allergic granulomatosis and angiitis (Churg-Strauss syndrome). Mt Sinai J Med N Y 53: 429–439, 1986. 59. Lanhan JG, Elkon KB, Pusey CD, Hughes GR: Systemic vasculitis with asthma and eosinophilia: a clinical approach to the Churg-Strauss syndrome. Medicine (Baltimore) 63: 65–81, 1984. 60. Farooki ZQ, Brough AJ, Green EW: Necrotizing arteritis. Am J Dis Child 128: 837–840, 1974.
557
61. Petty RE, Magilavy DB, Cassidy JT, et al: A syndrome of childhood polyarteritis. J Pediatr 91: 25–30, 1977. 62. Frayha RA: Churg-Strauss syndrome in a child. J Rheumatol 9: 807–809, 1982. 63. Treitman P, Herskowitz JL, Bass HN: Churg-Strauss syndrome in a 14-yearold boy diagnosed by transbronchial lung biopsy. Clin Pediatr 30: 502–505, 1991. 64. Heine A, Beck R, Stropahl G, et al: Inflammatory pseudotumor of the anterior orbit. A symptom of allergic granuloma. Ophthalmologe 92: 870–873, 1995. 65. Jessurun J, Azevedo M, Saldan M: Allergic angiitis and granulomatosis (Churg-Strauss syndrome): report of a case with massive thymic involvement in a nonasthmatic patient. Hum Pathol 17: 637–639, 1986. 66. Rabusin M, Lepore L, Constantinides F, Bussani R: A child with severe asthma. Lancet 351: 32, 1998. 67. Mpofu C, Bakalinova D, Kazi MA, Dawson KP: Churg-Strauss syndrome in childhood. Ann Trop Paediatr 15: 341–344, 1995. 67a. Davis MD, Daoud MS, McEvoy MT, Su WP: Cutaneous manifestations of Churg-Strauss syndrome: a clinicopathologic correlation. J Am Acad Dermatol 37: 199–203, 1997. 68. Case 46-1980: interstitial eosinophilic pneumonitis, pleuritis and angiitis. N Engl J Med 304: 611–612, 1981. 69. Chumbley LC, Harrison EG Jr, DeRemee RA: Allergic granulomatosis and angiitis (Churg-Strauss syndrome): report and analysis of 30 cases. Mayo Clin Proc 52: 477–484, 1977. 70. Cury D, Braekley AS, Payne BF: Allergic granulomatous angiitis associated with posterior uveitis and papilloedema. Arch Ophthalmol 55: 261–266, 1956. 71. Hattori N, Ichimura M, Nagamatsu M, et al: Clinicopathological features of Churg-Strauss syndrome-associated neuropathy. Brain 122: 427–439, 1999. 72. Louthrenoo W, Norasetthada N, Kjunamornpong S, et al: Childhood ChurgStrauss syndrome. J Rheumatol 26: 1387–1393, 1999. 73. Wechsler ME, Garpestad E, Flier SR, et al: Pulmonary infiltrates, eosinophilia and cardiomyopathy following corticosteroid withdrawal in patients with asthma receiving zafirlukast. JAMA 279: 455–457, 1998. 74. Keogh KA, Specks U: Churg-Strauss syndrome: clinical presentation, antineutrophil cytoplasmic antibodies, and leukotriene receptor antagonists. Am J Med 115: 284–290, 2003. 75. Guillevin L, Visser H, Noel LH, et al: Antineutrophil cytoplasm antibodies in systemic polyarteritis nodosa with and without hepatitis B virus infection and Churg-Strauss syndrome—62 patients. J Rheum 20: 1345–1349, 1993. 76. Tatsis E, Schnabel A, Gross WL: Interferon-alpha treatment of four patients with the Churg-Strauss syndrome. Ann Intern Med 129: 370–374, 1998. 77. Guillevin L, Cohen P, Gayraud M, et al: Churg-Strauss syndrome. Clinical study and long-term follow-up of 96 patients. Medicine (Baltimore) 78: 26–37, 1999. 78. Liebow AA, Carrington CRB, Friedman PJH: Lymphomatoid granulomatosis. Hum Pathol 3: 457–558, 1972. 79. Fauci AS, Haynes BF, Costa J, et al: Lymphomatoid granulomatosis: prospective clinical and therapeutic experience over 10 years. N Engl J Med 306: 68–74, 1982. 80. Koss MN Liselotte H, Langloss JM, et al: Lymphomatoid granulomatosis: a clinicopathologic study of 42 patients. Pathology 18: 283–288, 1986. 81. Katzenstein AA, Carrington CB, Liebow AA: Lymphomatoid granulomatosis. A clinicopathologic study of 152 cases. Cancer 43: 360–373, 1979. 82. Lehman TJ, Church JA, Isaacs H: Lymphomatoid granulomatosis in a 13month-old infant. J Rheumatol 16: 235–238, 1989. 83. Grierson H, Purtilo DT: Epstein-Barr virus infections in males with the Xlinked immunoproliferative syndrome. Ann Intern Med 106: 538–545, 1987. 84. Ilowite NT, Fligner CL, Ochs HD, et al: Pulmonary angiitis with atypical lymphoreticular infiltrates in Wiskott-Aldrich syndrome: possible relationship of lymphomatoid granulomatosis and EBV infection. Clin Immunol Immunopathol 41: 479–484, 1986. 85. Anders KH, Guerra WF, Tomiyasu U, et al: The neuropathology of AIDS. ULCA experience and review. Am J Pathol 124: 537–558, 1986. 86. Shen SC, Heuser ET, Landing NH, et al: Lymphomatoid granulomatosis-like lesions in a child with leukemia in remission. Hum Pathol 12: 276–280, 1981. 87. Gardiner GW: Lymphomatoid granulomatosis of the larynx in a renal transplant recipient. J Otolaryngol 8: 549–555, 1979. 88. Waltern M, Thomson NM, Dowling J, et al: Lymphomatoid granulomatosis in a renal transplant recipient. Aust N Z J Med 9: 434–436, 1979. 89. Hammer S, Mennemeyer R: Lymphomatoid granulomatosis in a renal transplant recipient. Hum Pathol 7: 111–116, 1976. 90. Newman W, Wolf A: Non-infectious granulomatous angiitis involving the central nervous system. Trans Am Neurol Assoc 56: 114–117, 1952. 91. Cravioto H, Feigin I: Noninfectious granulomatous angiitis with a predilection for the nervous system. Neurology 9: 599–609, 1959. 92. Calabrese LH, Furlan AJ, Gragg LA, Popos TJ: Primary angiitis of the central nervous system: diagnostic criteria and clinical approach. Cleve Clin J Med 59: 293–306, 1992. 93. Benseler SM, Deveber G, Tsang LM, et al: CNS vasculitis in children. [abstract #138]. Pediatr Rheumatol Online J, http://www.pedrheumonlinejournal.org (accessed July 2003). 94. Lanthier S, Lortie A, Michaud J, et al: Isolated angiitis of the CNS in children. Neurology 57: 837–842, 2001.
558
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
95. Abu-Shakra M, Khraishi M, Grosman H, et al: Primary angiitis of the CNS diagnosed by angiography. Q J Med 87: 351–358, 1994. 96. Calabrese LH, Mallek JA: Primary angiitis of the central nervous system: report of 8 new cases, review of the literature and proposal for diagnostic criteria. Medicine (Baltimore) 67: 20–39, 1988. 97. Biller J, Loftus CM, Moore SA, et al: Isolated central nervous system angiitis first presenting as spontaneous intracranial hemorrhage. Neurosurgery 20: 310–315, 1987. 98. Forman HP, Levin S, Stewart B, et al: Cerebral vasculitis and hemorrhage in an adolescent taking diet pills containing phenylpropanolamine: case report and review of literature. Pediatrics 83: 737–741, 1989. 99. Farine D, Andreyko J, Lysikiewicz A, et al: Isolated angiitis of brain in pregnancy and puerperium. Obstet Gynecol 63: 586–588, 1984. 100. Gallagher KT, Shaham B, Reiff A, et al: Primary angiitis of the central nervous system in children: 5 cases. J Rheumatol 28: 616–623, 2001. 101. Langlois PF, Sharon GE, Gawryl MS: Plasma concentrations of complementactivation complexes correlate with disease activity in patients diagnosed with isolated central nervous system vasculitis. J Allergy Clin Immunol 83: 11–16, 1989. 102. Hellman DB, Roubenoff R, Healy RA, et al: Central nervous system angiography: safety and predictors of a positive result in 125 consecutive patients evaluated for possible vasculitis. J Rheumatol 19: 568–572, 1992. 103. Moore PM: Diagnosis and management of isolated angiitis of the central nervous system. Neurology 39: 167–173, 1989. 104. Woolfenden AR, Tong DC, Marks MP, et al: Angiographically defined primary angiitis of the CNS. Is it really benign? Neurology 51: 183–188, 1998. 105. Cupps TR, Moore PM, Fauci AS: Isolated angiitis of the CNS: prospective diagnostic and therapeutic experience. Am J Med 74: 97–105, 1983. 106. Sigal LH: Isolated CNS angiitis. Neurology 39: 167–173, 1989. 107. Suzuki J, Takaku A: Cerebrovascular “moyamoya” disease: disease showing abnormal net-like vessels in base of brain. Arch Neurol 20: 288–299, 1969. 108. Van Erven PM, Gabreels FJ, Thijssen HO, Renier WO: The Moya-Moya syndrome: a report of two children. Clin Neurol Neurosurg 84: 179–189, 1982. 109. Carlson CB, Harvey FH, Loop J: Progressive alternating hemiplegia in early childhood and basal arterial stenosis and telangiectasia (moyamoya syndrome). Neurology 23: 734–744, 1973. 110. Magee R, Marshall M, Schaub M, Terrio L: Speech-language patterns in a child with moya moya disease. Percept Mot Skills 79: 1183–1192, 1994. 111. Chabbert V, Ranjeva JP, Sevely A, et al: Diffusion and magnetisation transfer weighted MRA in childhood moya-moya. Neuroradiology 40: 267–271, 1998. 112. Suzuki Y, Negoro M, Shibuya M, et al: Surgical treatment for pediatric moyamoya disease: use of the superficial artery for both areas supplied by the anterior and middle cerebral arteries. Neurosurgery 40: 324–329, discussion 329–330, 1997. 113. McLean MJ, Gebarski SS, van der Spek AFL, Goldstein GW: Response of moyamoya disease to verapamil. Lancet 1: 163–164, 1985. 114. Harvey FH, Alvord EC Jr: Juvenile arteriosclerosis and other cerebral arteriopathies of childhood. Acta Neurol Scand 48: 479–509, 1972. 115. Galligioni F, Andrioli GC, Marin G, et al: Hypoplasia of the internal carotid artery associated with cerebral pseudoangiomatosis: report of four cases. Am J Roentgenol Radium Ther Nucl Med 112: 251–262, 1971. 116. Goldberg HJ: Moyamoya associated with peripheral vascular occlusive disease. Arch Dis Child 49: 964–966, 1974. 117. Schrager GO, Cohen SJ, Vigman MP: Acute hemiplegia and cortical blindness due to moya moya disease: report of a case in a child with Down’s syndrome. Pediatrics 60: 33–37, 1977. 118. Kapusta L, Daniels O, Renier WO: Moya-Moya syndrome and primary pulmonary hypertension in childhood. Neuropediatrics 21: 162–163, 1990. 119. Kowada M, Momma F, Kikuchi K: Intracranial aneurysm associated with cerebrovascular moyamoya disease. Report of a case and review of 13 cases. Br J Radiol 52: 236–237, 1979. 120. Kitahara T, Ariga N, Yamaura A, et al: Familial occurrence of moya-moya disease: report of three Japanese families. J Neurol Neurosurg Psychiatry 42: 208–214, 1979. 121. Shimizu K, Sano K: Pulseless disease. Clin Surg (Tokyo) 3: 377, 1948. 122. Shimizu K, Sano K: Pulseless disease. J Neuropathol Clin Neurol 1: 37, 1951. 123. Takayasu M: Case with unusual changes of the central vessels in the retina. Acta Soc Ophthalmol Jpn 12: 554, 1908. 124. Arend WP, Michel BA, Bloch DA, et al: The American College of Rheumatology 1990 criteria for the classification of Takayasu arteritis. Arthritis Rheum 33: 1129–1134, 1990. 125. Lupi-Herrera E, Sanches-Torres G, Marushamer J, et al: Takayasu’s arteritis. Clinical study of 126 cases. Am Heart J 93: 94–103, 1977. 126. Ishikawa K: Survival and morbidity after diagnosis of occlusive thromboaortopathy (Takayasu’s disease). Am J Cardiol 47: 1026–1032, 1981. 127. Wagenvoort CA, Harris LE, Brown AL, et al: Giant cell arteritis with aneurysm formation in children. Pediatrics 32: 861–867, 1963. 128. Warshaw JB, Spach MS: Takayasu’s disease (primary aortitis) in childhood: case report with a review of the literature. Pediatrics 35: 620–626, 1965. 129. Hong CY, Yun YS, Choi JY, et al: Takayasu arteritis in Korean children: clinical report of seventy cases. Heart Vessels 71 (Suppl): 91–96, 1992. 130. Hahn D, Thomson PD, Kala U, et al: A review of Takayasu’s arteritis in children in Gauteng, South Africa. Pediatr Nephrol 12: 668–675, 1998.
131. D’Souza SJ, Tsai WS, Silver MM, et al: Diagnosis and management of stenotic aorto-arteriopathy in childhood. J Pediatr 132: 1016–1022, 1998. 132. Dabague J, Reyes PA: Takayasu arteritis in Mexico: a 38-year clinical perspective through literature review. Int J Cardiol 54 (Suppl): S103–S109, 1996. 133. Hall S, Barr W, Lie JT, et al: Takayasu arteritis: a study of 32 North American patients. Medicine (Baltimore) 64: 89–99, 1985. 134. Jain S, Sharma N, Singh S, et al: Takayasu arteritis in children and young Indians. Int J Cardiol 75 (Suppl 1): S153–S157, 2000. 135. Vinijchaikul K: Primary arteritis of the aorta and its main branches (Takayasu’s arteriopathy). A clinicopathologic autopsy study of eight cases. Am J Med 43: 15–27, 1967. 136. Gupta S: Surgical and haemodynamic considerations in middle aortic syndrome. Thorax 34: 470–478, 1979. 137. Gupta S, Goswami B, Ghosh DC, et al: Middle aortic syndrome as a cause of heart failure in children and its management. Thorax 36: 63–65, 1981. 138. Numano R, Isohisa I, Kishi U, et al: Takayasu’s disease in twin sisters. Possible genetic factors. Circulation 58: 173–177, 1978. 139. Numano F: Hereditary factors of Takayasu arteritis. Heart Vessels Suppl 7: 68–72, 1992. 140. Kimura A, Kitamura H, Date Y, Numano F: Comprehensive analysis of HLA genes in Takayasu arteritis in Japan. Int J Cardiol 54 (Suppl): S61–S69, 1996. 141. Mehra NK, Rajalingam R, Sagar S, et al: Direct role of HLA-B5 in influencing susceptibility to Takayasu aortoarteritis. Int J Cardiol 54 (Suppl): S71–S79, 1996. 142. Girona E, Yamamoto-Furusho JK, Cutino T, et al: HLA-DR6 (possibly DRB1*1301) is associated with susceptibility to Takayasu arteritis in Mexicans. Heart Vessels 11: 277–280, 1996. 143. Pantell RH, Goodman BW Jr: Takayasu’s arteritis: the relationship with tuberculosis. Pediatrics 67: 84–88, 1981. 144. Morales E, Pineda C, Martinez-Lavin M: Takayasu’s arteritis in children. J Rheumatol 18: 1081–1084, 1991. 145. Milner LS, Jacobs DW, Thomson PD, et al: Management of severe hypertension in childhood Takayasu’s arteritis. Pediatr Nephrol 5: 38–41, 1991. 146. Aggarwal A, Chag M, Sinha N, Naik SP: Takayasu’s arteritis: role of Mycobacterium tuberculosis and its heat shock protein. Int J Cardiol 55: 49–55, 1996. 147. Eichorn J, Sima D, Thiele B, et al: Anti-endothelial cell antibodies in Takayasu arteritis. Circulation 94: 2396–2401, 1996. 148. Garcia-Torres R, Noel LH, Reyes PA, et al: Absence of ANCA in Mexican patients with Takayasu’s arteritis. Scand J Rheumatol 26: 55–57, 1997. 149. Wiggelinkhuizen J, Cremin BJ: Takayasu arteritis and renovascular hypertension in childhood. Pediatrics 62: 209–217, 1978. 150. Hall S, Nelson AM: Takayasu’s arteritis and juvenile rheumatoid arthritis. J Rheumatol 13: 431–433, 1986. 151. Yamato M, Kecky JW, Hiramatsu K, et al: Takayasu arteritis: radiographic and angiographic findings in 59 patients. Radiology 161: 329–334, 1986. 152. Fournier AM, Dickinson ZC, Kelly R: Doppler ultrasonography as a guide to management of Takayasu’s arteritis. J Rheumatol 15: 527–528, 1988. 153. Hunder GG, Weyland CM: Sonography in giant-cell arteritis. N Engl J Med 337: 1385–1386, 1997. 154. Buckley A, Southwood T, Culham G, et al: The role of ultrasound in evaluation of Takayasu’s arteritis. J Rheumatol 18: 1073–1080, 1991. 155. Kissin EY, Merkel PA: Diagnostic imaging in Takayasu arteritis. Curr Opin Rheumatol 16: 31–37, 2004. 156. D’Souza SJA, Tsai WS, Silver MM, et al: Diagnosis and management of stenotic aorto-arteriopathy in childhood. J Pediatr 132: 1016–1022, 1998. 157. Shetty AK, Stopa AR, Gedalia A: Low-dose methotrexate as a steroid-sparing agent in a child with Takayasu’s arteritis. Clin Exp Rheumatol 16: 335–336, 1998. 158. Kerr GS: Takayasu’s arteritis. Rheum Dis Clin North Am 21: 1041–1058, 1995. 159. Sharma S, Rajani M, Shrivastava S, et al: Non-specific aorto-arteritis (Takayasu’s disease) in children. Br J Radiol 64: 690–698, 1991. 160. Becker GJ: Intravascular stents. General principles and status of lowerextremity arterial applications. Circulation 83 (Suppl): I122–I136, 1991. 161. Palmaz JC: Intravascular stents: tissue-stent interactions and design considerations. Am J Roentgenol 160: 613–618, 1993. 162. Tyagi S, Sharma VP, Arora R: Stenting of the aorta for recurrent, long stenosis due to Takayasu’s arteritis in a child. Pediatr Cardiol 20: 215–217, 1999. 163. Weyand CM, Wagner AD, Bjornson J, et al: Correlation of the topographical arrangement and the functional pattern of tissue-infiltrating macrophages in giant cell arteritis. J Clin Invest 98: 1642–1649, 1996. 164. Lie JT: Bilateral juvenile temporal arteritis. J Rheumatol 22: 774–776, 1995. 165. Lie JT, Gordon LP, Titus JL: Juvenile temporal arteritis: biopsy study of four cases. JAMA 234: 496–499, 1975. 166. Fujimoto M, Sato S, Hayashi N, et al: Juvenile temporal arteritis with eosinophilia: a distinct clinicopathological entity. Dermatology 192: 32–35, 1996. 167. Bollinger A, Leu H-J, Brunner U: Juvenile arteritis of extracranial arteries with hypereosinophilia. Klin Wochenschr 64: 526–529, 1986. 168. McGovern JP, Merritt DH: Sarcoidosis in childhood. Adv Pediatr 8: 97–135, 1956.
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
169. Jasper PL, Denny FW: Sarcoidosis in children with special emphasis on the natural history and treatment. J Pediatr 73: 499, 1968. 170. Siltzbach LE, Greenberg GM: Childhood sarcoidosis—a study of 18 patients. N Engl J Med 279: 1239–1512, 1968. 171. Kendig EL: Sarcoidosis among children: a review. J Pediatr 61: 269–278, 1962. 172. Kendig EL: The clinical picture of sarcoidosis in children. Pediatrics 54: 289–292, 1974. 173. Beier FR, Lahey ME: Sarcoidosis among children in Utah and Idaho. J Pediatr 65: 350–359, 1964. 174. Mandi L: Thoracic sarcoidosis in childhood. Acta Tuberc Scand 45: 256–270, 1964. 175. Harris C, Gibson WM, Schuchter SL, et al: Rare diagnosis: sarcoid arthritis in four children. JAMA 197: 31, 1966. 176. Sahn EE, Hampton MT, Garen PD, et al: Preschool sarcoidosis masquerading as juvenile rheumatoid arthritis: two case reports and a review of the literature. Pediatr Dermatol 7: 208–213, 1990. 177. Gluck J, Miller JJ III, Summerlin WT: Sarcoidosis in a young child. J Pediatr 81: 354–357, 1972. 178. Hetherington S: Sarcoidosis in young children. Am J Dis Child 136: 13–15, 1982. 179. Pattishall EN, Strope GL, Spinola SM, et al: Childhood sarcoidosis. J Pediatr 108: 169–177, 1986. 180. Hafner R, Vogel P: Sarcoidosis of early onset. A challenge for the pediatric rheumatologist. Clin Exp Rheumatol 11: 685–691, 1993. 181. Cron RQ, Wallace CA, Sherry DD: Childhood sarcoidosis-does age of onset predict clinical manifestations? J Rheumatol 24: 1654–1656, 1997. 182. Bresnitz EA, Strom BL: Epidemiology of sarcoidosis. Epidemiol Rev 5: 124–156, 1983. 183. Fanburg BL, Pitt EA: Sarcoidosis. In Murray JF, Nadel JA (eds): Textbook of Respiratory Medicine. Philadelphia, WB Saunders, 1988, p 1495. 184. Rosenberg AM, Yee EH, MacKenzie JW: Arthritis in childhood sarcoidosis. J Rheumatol 10: 987–990, 1983. 185. Lindsley CB, Petty RE: Childhood sarcoid arthritis registry. Arthritis Rheum 36: S123, 1993. 186. Niitu Y, Horikawa M, Suetake T, et al: Pulmonary sarcoidosis among children. Sai Shin Med 27: 1347, 1972. 187. Merten DF, Kirks DR, Grossman HP: Pulmonary sarcoidosis in children. Am J Roentgenol 135: 673–679, 1980. 188. Kendig EL Jr: Sarcoidosis in children: personal observations on age distribution. Pediatr Pulmonol 6: 69–70, 1989. 189. Schabel SI, Stanley HJ, Shelley BE Jr: Pediatric sarcoidosis. J S C Med Assoc 76: 419–422, 1980. 190. Reed WB: Sarcoidosis: a review and report of eight cases in children. J Tenn Med Assoc 62: 27–36, 1969. 191. Schmitt FE, Appelman H, Threatt B: Sarcoidosis in children. Radiology 106: 621–625, 1973. 192. Beskow R, Wiman LG: Familial occurrence of sarcoidosis. In Levinsky L, MacHolda F (eds): Proceedings of the Fifth International Conference on Sarcoidosis. Prague, Czechoslovakia, Universita Karlova, 1971, p 280. 193. James DG, Piyasena HG, Neville E, et al: Possible genetic influences in familial sarcoidosis. Postgrad Med J 50: 664–670, 1974. 194. Jorgensen G: Untersuchungen zur Genetik der Sarkoidose. Göttingen, Germany, Habil Schrift, 1965. 195. Buck AA, McKusick VA: Epidemiologic investigations of sarcoidosis. Am J Hygiene 74: 174–188, 1961. 196. Keating JP, Weissbluth M, Ratzan SK, et al: Familial sarcoidosis. Am J Dis Child 126: 644–647, 1973. 197. Schweizer AT, Kanaar P: Sarcoidosis with polyarthritis in a child. Arch Dis Child 42: 671–674, 1967. 198. Prieur AM, Henkes CJ, Bessis JL, et al: Familial sarcoid arthritis. Arch Fr Pediatr 39: 311–313, 1982. 199. Headings VE, Weston D, Young RC Jr, et al: Familial sarcoidosis with multiple occurrences in eleven families: a possible mechanism of inheritance. Ann N Y Acad Sci 278: 377–385, 1976. 200. Sharma OP, Neville E, Walker AN, et al: Familial sarcoidosis: a possible genetic influence. Ann N Y Acad Sci 278: 386–400, 1976. 201. Schurmann M, Bein G, Kirsten D, et al: HLA-DQB1 and HLA-DPB1 genotypes in familial sarcoidosis. Respir Med 92: 649–652, 1998. 202. Shetty AK, Gedalia A: Sarcoidosis in children. Curr Probl Pediatr 30: 149–176, 2000. 203. Zsolway K, Sinai LN, Magnusson M, Tunnessen WW Jr: Two unusual pediatric presentations of sarcoidosis. Arch Pediatr Adolesc Med 152: 410–411, 1998. 204. Clark SK: Sarcoidosis in children. Pediatr Dermatol 4: 291–299, 1987. 205. Mallory SB, Paller AS, Ginsburg BC, et al: Sarcoidosis in children: differentiation from juvenile arthritis. Pediatr Dermatol 4: 313–319, 1987. 206. Kuramoto Y, Shindo Y, Tagami H: Subcutaneous sarcoidosis with extensive caseation necrosis. J Cutan Pathol 15: 188–190, 1988. 207. Ramanan AV, Denning DW, Baildam EM: Cutaneous childhood sarcoidosisa rare disease refractory to treatment. Rheumatology (Oxf) 42: 1570–1571, 2003. 208. Obenauf CD, Shaw HE, Sydnor CR, Klintworth GK: Sarcoidosis and its ophthalmic manifestations. Am J Ophthalmol 86: 648–655, 1978.
559
209. Kansi JJ, Shun-Shin GA: Systemic uveitis syndromes in childhood: an analysis of 340 cases. Ophthalmology 91: 1247–1252, 1984. 210. Khan JA, Hoover DL, Giangiacomo J, et al: Orbital and childhood sarcoidosis. J Pediatr Ophthalmol Strabismus 23: 190–194, 1986. 211. Brinkman CJJ, Pogany KP: Neurosarcoidosis and uveitis. In Dernouchamps JP, Veroughstraete L, Caspers-Velu L, Tassignon MJ (eds): Recent advances in uveitis. In Proceedings of the Third International Symposium on Uveitis. Brussels, Belgium, Amsterdam; New York: Krigler Publications, 1992, p 375. 212. Hoover DL, Khan J, Giangiacomo J: Pediatric ocular sarcoidosis. Surv Ophthalmol 30: 215–228, 1986. 213. Bronson LJ, Fisher YL: Sarcoidosis of the paranasal sinuses with orbital extension. Arch Ophthalmol 94: 243–244, 1976. 214. Srinivasan E: A case of Boeck’s sarcoidosis involving the orbit. Arch Ophthalmol 25: 493, 1941. 215. Lennarson P, Barney NP: Interstitial keratitis as presenting ophthalmic sign of sarcoidosis in a child. J Pediatr Ophthalmol Strabismus 32: 194–196, 1995. 216. Leigh MW: Sarcoidosis. In Behrman RE (eds): Nelson’s Textbook of Pediatrics, 16th ed. Philadelphia, WB Saunders, 2000, p 2143. 217. Lindsley CB, Godfrey WA: Childhood sarcoidosis manifesting as juvenile rheumatoid arthritis. Pediatrics 76: 765–768, 1985. 218. Harris RO III, Spock A: Childhood sarcoidosis: pulmonary infiltrates as an early sign in a very young child. Clin Pediatr 17: 119–121, 1978. 219. Aszkanazy CL: Sarcoidosis of the central nervous system. J Neuropathol Exp Neurol 11: 392–400, 1952. 220. Jallo GI, Zagzag D, Lee M, et al: Intraspinal sarcoidosis: diagnosis and management. Surg Neurol 48: 514–520, 1997. 221. Bernstein J, Rival J: Sarcoidosis of the spinal cord as the presenting manifestation of the disease. South Med J 71: 1571–1573, 1978. 222. Campbell JN, Black P, Ostrow PT: Sarcoid of the cauda equina: case report. J Neurosurg 47: 109–112, 1977. 223. Stern BJ, Krumholz A, Johns C, et al: Sarcoidosis and its neurological manifestations. Arch Neurol Psychiatry 42: 909–917, 1985. 224. Wood EH, Bream CA: Spinal sarcoidosis. Radiology 73: 226–233, 1959. 225. Rubinstein I, Hiss J, Baum GL: Intramedullary spinal cord sarcoidosis. Surg Neurol 21: 272–274, 1984. 226. Leiba H, Siatkowski RM, Culbertson WW, Glaser JS: Neurosarcoidosis presenting as an intracranial mass in childhood. J Neuroophthalmol 16: 269–273, 1996. 227. Weinberg S, Bennett H, Weinstock I: Central nervous system manifestations of sarcoidosis in children: case report and review. Clin Pediatr 22: 477–481, 1983. 228. Kone-Paut I, Portas M, Wechsler B, et al: The pitfall of silent neurosarcoidosis. Pediatr Neurol 20: 215–218, 1999. 229. Day AL, Sypert GW: Spinal cord sarcoidosis. Ann Neurol 1: 79–85, 1977. 230. Caroscio J, Yahr MD: Progressive myelopathy due to sarcoid. Clin Neurol Neurosurg 82: 217–222, 1980. 231 Baumann RJ, Robertson WC Jr: Neurosarcoid presents differently in children than in adults. Pediatrics 112: e480–e486, 2003. 232. North FA, Fink CW, Gibson WM, et al: Sarcoid arthritis in children. Am J Med 48: 449–455, 1970. 233. Gedalia A, Molina JF, Ellis GS, et al: Low-dose methotrexate therapy for childhood sarcoidosis. J Pediatr 130: 25–29, 1997. 234. Gumpel JM, Johns CJ, Shulman LE: The joint disease of sarcoidosis. Ann Rheum Dis 26: 194–205, 1967. 235. Stump K, Spock A, Grossman H: Vertebral sarcoidosis in adolescents. Pediatr Radiol 121: 153–155, 1976. 236. Silverstein A, Siltzbach LE: Muscle involvement in sarcoidosis. Arch Neurol 21: 235–241, 1969. 237. Stjernberg N, Cajander S, Truedsson H, et al: Muscle involvement in sarcoidosis. Acta Med Scand 209: 213–216, 1981. 238. Mayock RL, Bertrand P, Morrison DE, Scott JH: Manifestations of sarcoidosis: analysis of 145 patients with a review of nine series selected from the literature. Am J Med 35: 67–89, 1963. 239. Celle ME, Veneselli E, Rossi GA, et al: Childhood sarcoidosis presenting with prevalent muscular symptoms: report of a case. Eur J Pediatr 156: 340–341, 1997. 240. Banker BQ: Other inflammatory myopathies. In Engel AG, Banker BQ (eds): Myology. New York, McGraw-Hill, 1986, p 507. 241. Dubowitz V: Inflammatory myopathies. In Dubowitz V (ed): Muscle Biopsy: A Practical Approach. London, Bailliere Tindall, 1985, p 605. 242. Levine MS, Ekberg O, Rubesin SE, et al: Gastrointestinal sarcoidosis: radiographic findings. Am J Roentgenol 153: 293–295, 1989. 243. Noel JM, Katona IM, Pineiro-Carrero VM: Sarcoidosis resulting in duodenal obstruction in an adolescent. J Pediatr Gastroenterol Nutr 24: 594–598, 1997. 244. Cohen DL: Sicca syndrome: an unusual manifestation of sarcoidosis in childhood. Am J Dis Child 137: 289–290, 1983. 245. Evans SS, Fisher RG, Scott MA, et al: Sarcoidosis presenting as bilateral testicular masses. Pediatrics 100: 392–394, 1978. 246. Mastui Y, Iwai K, Tachibana T, et al: Clinicopathological study of fatal myocardial sarcoidosis. Ann N Y Acad Sci 80: 278: 455–469, 1976. 247. Demeter SL: Myocardial sarcoidosis unresponsive to steroids. Treatment with cyclophosphamide. Chest 94: 202–203, 1988.
560
C H A P T E R
26 G R A N U L O M A T O U S V A S C U L I T I S , G I A N T C E L L A RT E R I T I S , & S A R C O I D O S I S
248. Muther RS, McCarron DA, Bennett WM: Renal manifestations of sarcoidosis. Arch Intern Med 141: 643–645, 1981. 249. Morris KP, Coulthard MG, Smith PJ, et al: Renovascular and growth effects of childhood sarcoid. Arch Dis Child 75: 74–75, 1996. 250. Cameron HM: Renal sarcoidosis. J Clin Pathol 9: 136–141, 1956. 251. Keech MK: Generalized sarcoidosis with renal involvement. Proc R Soc Med 44: 728–729, 1951. 252. Berger KW, Relman AS: Renal impairment due to sarcoid infiltration of the kidney: report of a case proved by renal biopsies before and after treatment with cortisone. N Engl J Med 252: 44–49, 1955. 253. Longcope WT, Freiman DG: A study of sarcoidosis; based on a combined investigation of 160 cases including 30 autopsies from The Johns Hopkins Hospital and Massachusetts General Hospital. Medicine (Baltimore) 31: 1–132, 1952. 254. Bautista A: Childhood sarcoidosis involving joints and kidneys. Am J Dis Child 119: 259–263, 1970. 255. Correa P: Sarcoidosis: association with glomerulonephritis. Arch Pathol 57: 523–529, 1954. 256. Nocton JJ, Stork JE, Jacobs G, Newman AJ: Sarcoidosis associated with nephrocalcinosis in young children. J Pediatr 121: 937–940, 1992. 257. James DG: A comparison of Blau’s syndrome and sarcoidosis. Sarcoidosis 11: 100–101, 1994. 258. Manouvrier-Hanu S, Puech B, Piette F, et al: Blau syndrome of granulomatous arthritis, iritis, and skin rash: a new family and review of the literature. Am J Med Genet 76: 217–221, 1998. 259. Blau EB: Familial granulomatous arthritis, iritis and rash. J Pediatr 107: 689–693, 1985. 260. de Chadaverian JP, Raphael SA, Murphy GF: Histologic, ultrastructural, and immunocytochemical features of the granulomas seen in a child with the syndrome of familial granulomatous synovitis, uveitis and rash. Arch Pathol Lab Med 117: 1050–1052, 1993. 261. Raphael SA, Blau EN, Zhang WH, et al: Analysis of a large kindred with Blau syndrome for HLA, autoimmunity and sarcoidosis. Am J Dis Child 147: 842–848, 1993. 262. Moraillon J, Hayem F, Bourrillon A, et al: Blau syndrome or familial form of sarcoidosis with onset in infancy. Ann Dermatol Venereol 123: 29–30, 1996. 263. Pastores GM, Michels VV, Stickler GB, et al: Autosomal dominant granulomatous arthritis, uveitis, skin rash, and synovial cysts. J Pediatr 117: 403, 1990. 264. Mau U, Baykal HE, Erb C, et al: Blau syndrome (familial non HLA-B27-associated acute anterior uveitis with arthritis and skin manifestations): a rare syndrome in a family with 10 members over 4 generations. Med Genet 7: 180, 1995. 265. Tromp G, Kuivaniemi H, Raphael S, et al: Molecular characterization of Blau syndrome: genetic linkage to chromosome 16. Am J Hum Genet Suppl 55: 205, 1994. 266. Tromp Kuivaniemi H, Raphael S, et al: Genetic linkage of familial granulomatous inflammatory arthritis, skin rash and uveitis to chromosome 16. Am J Hum Genet 59: 1097–1107, 1996. 267. Wang X, Kuivaniemi H, Bonavita G, et al: CARD 15 mutations in familial granulomatosis syndromes: a study of the original Blau syndrome kindred and other families with large-vessel arteritis and cranial neuropathy. Arthritis Rheum 46: 3041–3045, 2002. 268. Jabs DA, Houk JL, Bias WB, Arnett FC: Familial granulomatous synovitis, uveitis, and cranial neuropathies. Am J Med 78: 801–804, 1985. 269. Bottcher E: Disseminated sarcoidosis with a marked granulomatous arteritis. Arch Pathol 68: 419–423, 1959. 270. Churg A, Carrington CB, Gupta R: Necrotizing sarcoid granulomatosis. Chest 76: 406–413, 1979. 271. Gross KR, Malleson PN, Culham G, et al: Vasculopathy with renal artery stenosis in a child with sarcoidosis. J Pediatr 108: 724–726, 1986. 272. Rose CD, Eichenfield AH, Goldsmith DP, et al: Early onset sarcoidosis with aortitis-“juvenile systemic granulomatosis ?” J Rheumatol 17: 575–579, 1990. 273. Gedalia A, Shetty AK, Ward K, et al: Abdominal aortic aneurysm associated with childhood sarcoidosis. J Rheumatol 23: 757, 1996. 274. Hoffmann AL, Milman N, Nielson HE, et al: Childhood sarcoidosis presenting with hypercalcaemic crisis. Sarcoidosis 11: 141–143, 1994. 274a. Henrich D, Gordjani N, Trusen A, et al: Necrotizing sarcoid granulomatosis: a rarity in childhood. Pediatr Pulmonol 35: 407–411, 2003. 275. Stanworth SJ, Kennedy CTC, Chetcuti PAJ, et al: Hypercalcaemia and sarcoidosis in infancy. J R Soc Med 85: 177–178, 1992. 276. James DG, Kendig EL Jr: Childhood sarcoidosis. Sarcoidosis 5: 57–59, 1988. 277. Mason RS, Frankel T, Chan YL, et al: Vitamin D conversion by sarcoid lymph node homogenate. Ann Intern Med 100: 59–61, 1984. 278. Reichel H, Koeffler HP, Barbers R, et al: Regulation of 1,25-dihydroxyvitamin D3 production by cultured alveolar macrophages from normal human donors and from patients with pulmonary sarcoidosis. J Clin Endocrinol Metab 65: 1201–1209, 1987. 279. Bresnihan B: Sarcoidosis. In Maddison PJ, Isenberg DA, Woo P, Glass DN (eds): Oxford Textbook of Rheumatology. New York, Oxford University Press, 1993, p 928. 280. Rodriguez DE, Shin BC, Abernathy RS, Kendig EL Jr: Serum angiotensinconverting enzyme activity in normal children and in those with sarcoidosis. J Pediatr 99: 68–72, 1981.
281. Studdy P, Bird R, James DG, et al: Serum angiotensin converting enzyme (SACE) in sarcoidosis and other granulomatous disorders. Lancet 2: 1331–1334, 1978. 282. Silverstein E, Friedland J, Lyons HA, et al: Elevation of angiotensin converting enzyme in granulomatous lymph nodes and serum in sarcoidosis: clinical and possible pathogenic significance. Ann N Y Acad Sci 278: 498–513, 1976. 283. DeRemee RA, Rohrbach MS: Serum angiotensin-converting enzyme activity in evaluating the clinical course of sarcoidosis. Ann Intern Med 92: 361–365, 1980. 284. Lieberman J: The specificity and nature of serum-angiotensin-converting enzyme (serum ACR) elevations in sarcoidosis. Ann N Y Acad Sci 278: 488–497, 1976. 285. Katz P, Fauci AS, Yeager H, et al: Serum angiotensin-converting enzyme in granulomatous diseases of unknown cause. Ann Intern Med 94: 359–360, 1981. 286. Beneteau-Burnate B, Baudin B, Morgant G, et al: Serum angiotensin-converting enzyme in healthy and sarcoidotic children: comparison with the reference interval for adults. Clin Chem 36: 344–346, 1990. 287. Mitchell DN, Cannon P, Dyer NH, et al: Further observations on Kveim test in Crohn’s disease. Lancet 2: 496–498, 1970. 288. Siltzbach LE: An international Kveim test study. Acta Med Scand Suppl 425: 178–190, 1964. 289. Hunninghak GW, Kawanami O, Ferrans VJ, et al: Characterization of the inflammatory and immune effector cells in the lung parenchyma of patients with interstitial lung disease. Am Rev Respir Dis 123: 407–412, 1981. 290. Baughman RP, Lower EE: The effects of corticosteroid or methotrexate therapy on lung lymphocytes and macrophages in sarcoidosis. Am Rev Respir Dis 142: 1268–1271, 1990. 291. Chadelat K, Baculard A, Grimfeld A, et al: Pulmonary sarcoidosis in children: serial evaluation of bronchoalveolar lavage cells during corticosteroid treatment. Pediatr Pulmonol 16: 41–47, 1993. 292. Tessier V, Chadelat K, Baculard A, et al: BAL in children: a controlled study of differential cytology and cytokine expression profiles by alveolar cells in pediatric sarcoidosis. Chest 109: 1430–1438, 1996. 293. Brauner MW, Grenler P, Mompoint D, et al: Pulmonary sarcoidosis: evaluation with high-resolution CT. Radiology 172: 467–471, 1989. 294. Keesling CA, Frush DP, O’Hara SM, Fordham LA: Clinical and imaging manifestations of pediatric sarcoidosis. Acad Radiol 5: 122–132, 1998. 295. Nichols CW, Mishkin M, Yanoff M: Presumed orbital sarcoidosis: report of a case followed by computerized axial tomography and conjunctival biopsy. Trans Am Ophthalmol Soc 76: 67–75, 1978. 296. Gedalia A, Shetty AK, Ward KJ, et al: Role of MRI in diagnosis of childhood sarcoidosis with fever of unknown origin. J Pediatr Orthop 17: 460–462, 1997. 297. Hayes W, Sherman J, Stern B, et al: MR and CT evaluation of intracranial sarcoidosis. AJR Am J Roentgenol 149: 1043–1049, 1987. 298. Sakurai Y, Nakajima M, Kamisue S, et al: Preschool sarcoidosis mimicking juvenile rheumatoid arthritis. The significance of gallium scintigraphy and skin biopsy in the differential diagnosis. Acta Paediatr Jpn 39: 74–78, 1997. 299. Hirase Y, Ishida Y, Hayashida K, et al: Myocardial involvement in patients with sarcoidosis: an analysis of 75 patients. Clin Nucl Med 19: 522–526, 1994. 300. Martinet Y, Pinkston P, Saltini C, et al: Evaluation of the in vitro and in vivo effects of cyclosporine on the lung T-lymphocyte alveolitis of active pulmonary sarcoidosis. Am Rev Respir Dis 138: 1242–1248, 1988. 301. The Research Committee of the British Tuberculosis Association: Chloroquine in the treatment of sarcoidosis. Tubercle 48: 257–272, 1967. 302. Mathur AM, Kremer JM: Immunopathology, rheumatic features and therapy of sarcoidosis. Curr Opin Rheumatol 4: 76–80, 1992. 303. Kiper N, Anadol D, Ozcelik U, Gocmen A: Inhaled corticosteroids for maintenance treatment in childhood pulmonary sarcoidosis. Acta Paediatr 90: 953–956, 2001. 304. Ulbricht KU, Stoll M, Bierwirth J, et al: Successful tumor necrosis factor alpha blockade treatment in therapy-resistant sarcoidosis. Arthritis Rheum 48: 3542–3543, 2003. 305. Roberts SD, Wilkes DS, Burgett RA, Knox KS: Chest 124: 2028–2031, 2003. 306. Baughman RP, Lower EE, du Bois RM: Sarcoidosis. Lancet 361: 1111–1118, 2003. 307. Young RC Jr, Hackney RL, Harden KA: Epidemiology of sarcoidosis: ethnic and geographic considerations. J Natl Med Assoc 66: 386–388, 1974. 308. Gillum RF: Sarcoidosis in the United States—1968–1984. Hospitalization and death. J Natl Med Assoc 80: 1179–1184, 1988. 309. Kendig EL Jr, Brummer DL: The prognosis of sarcoidosis in children. Chest 70: 351–353, 1976. 310. Marcille R, McCarthy M, Barton JW, et al: Long-term outcome of pediatric sarcoidosis with emphasis on pulmonary status. Chest 102: 1444–1449, 1992. 311. Fink CW, Cimaz R: Early onset sarcoidosis: not a benign disease. J Rheumatol 24: 174–177, 1997. 312. Haubitz M, Schellong S, Gobel U, et al: Intravenous pulse administration of cyclophosphamide versus daily oral treatment in patients with antineutrophil cytoplasmic antibody-associated vasculitis and renal involvement: a prospective, randomized study. Arthritis Rheum 41: 1835–1844, 1998. 313. Langford CA, Talar-Williams C, Barron KS, Snellar MC: Use of a cyclophosphamide-induction methotrexate-maintenance regimen for the treatment of Wegener’s granulomatosis: extended follow-up and rate of relapse. Am J Med 114: 463–469, 2003. 314. Jayne D, Rasmusses N, Andrassy K, et al: A randomized trial of maintenance therapy for vasculitis associated with antineutrophil cytoplasmic autoantibodies. N Engl J Med 349: 36–44, 2003.
27
(
HAP T E R
BEH<;ET'S DISEASE Seza Ozen and Ross E. Petty
;}f
In 1937, the Turkish dermatologist Hulusi Beh~et1 described the syndrome that bears his name: the clinical triad of aphthous stomatitis, genital ulceration, and uveitis. Superficial thrombophlebitis was identified as the fourth criterion in 1946. 2 Matteson3 identified even earlier reports of this condition, including those from Japan. The history of Beh~et's disease (BD) has been reviewed by Kaklamani and colleagues. 4
DEFINITION AND CLASSIFICATION There is still no unanimous agreement about the definition of this syndrome, and several sets of diagnostic criteria have been proposed. Those of the International Study Group (ISG)5 are most widely used and are listed in Table 27-1. If only one of the criteria is present along with oral ulcerations, the term incomplete or partial Behret's disease is applied. Criteria proposed by Mason and Barnes6 (Table 27-2) and O'Duffy and Goldstein7 emphasize the broader spectrum of disease. The ISG criteria have a specificity of 96% and sensitivity of 91%; the Mason and Barnes criteria have a specificity of 84% and sensitivity of 86%.8 All have been applied to the diagnosis of BD in children, although none has been validated.
EPIDEMIOLOGY The geographic distribution of disease prevalence retraces the historical route of the Silk Road from Japan to the eastern edge of the Mediterranean Sea and through areas of the Ottoman Empire. 9 However, BD is by no means confined to these areas and is rarely reported in children from India 10 and more commonly identified in children from Europe 1! and North America, reflecting emigration patterns in the 20th century. The frequency of BD in French children younger than 15 years is approximately 1 case in 600,000,12 Although BD is undoubtedly more common in Turkey and other parts of the Middle East, there are no published data concerning incidence or prevalence; Ozen and coworkers 13 have proposed that the prevalence is not more than 10 cases per 100,000 children. Several reviews document BD in childhood and adolescence. 12,!4-21 Lang and colleagues!4 reviewed 37 cases of BD beginning in children before age 17 that were published in the English language literature.
Thirty-five of the patients met criteria for complete BD by the criteria of Mason and Barnes. An international study of the clinical features of BD in 86 children from France, Turkey, Iran, and Saudi Arabia used the ISG criteria. 12 Eighteen Greek children who met the ISG criteria for BD were described by Vaiopoulos and colleagues in 1999.!5 There have also been series reported from Saudi Arabia, 16 Turkey,17 Korea,18 Italy, 19 Japan,20 and IsraeJ.21 In most series, boys and girls are affected with equal frequency, in contrast to BD in adults, in which men are affected almost twice as frequently as women. Overall, 5.4% to 7.6% of all patients had the onset of BD in childhood. 19,22 The age at onset of disease ranges widely (Table 27-3). BD has been reported in neonates born to mothers with BD.23-25
GENETIC BACKGROUND The higher frequency of BD in Japan and the countries of the Middle East and the high frequency of familial occurrence suggest that there may be a genetic or envi-
I!: II
TABLE 27-t Criteria of the International Study Group for the Diagnosis of Behcefs Disease
CrIterion
Desalptlon
Recurrent oral ulceration
Minor aphthous, major aphthous, or herpetiform ulceration recurring at least three times in one 12-month period, observed by physician or patient
Plus two ofthe following: Recurrent genital ulcers
Eye lesions
Skin lesions
Pathergy
Aphthous ulceration or scarring observed by physician or patient Anterior uveitis, posterior uveitis, or cells in vitreous on slit-lamp examination, or retinal vasculitis observed by an ophthalmologist Erythema nodosum observed by physician or patient: pseudofolliculitis or papulopustular lesions; or acneiform nodules observed by physician in postadolescent patient not on corticosteroid treatment Skin reaction to a needle prick observed by physician at 24-48 hr
From Criteria for the diagnosis of Beh~et's disease, International Study Group for Beh~et's Disease. Lancet 335: 1078--1080, 1990,
561
562
C HAP T E R
27
BEH<;:ET'S DISEASE
compared with healthy controls, patients with systemic illnesses, or patients with recurrent oral ulcers but no other manifestations of the disease. 33 Despite several studies suggesting a role for herpes simplex virus type 1, parvovirus B19, and streptococci, no microbial cause has been established. An antibodymediated process may be responsible, as suggested by the observation of transient neonatal BD in the offspring of mothers with the disorder. 23-25
l i TABLE 27 2 Mason and Barnes Criteria lor a IJidgnosb 01 Beh(el's Dbedse
Major a1terla"
Minor alterla
Buccal ulceration Genital ulceration Eye lesions Skin lesions
Gastrointestinal lesions Thrombophlebitis Cardiovascular lesions Arthritis Central nervous system lesions Family history of Beh~el's disease
'Diagnosis requires the presence of at least three major criteria or two major plus two minor criteria. From Mason RM. Barnes CG: Beh~el's syndrome with arthritis. Ann Rheum Dis 28: 9;-103, 1969.
ronmental component to the disease. A study of adult Turkish patients showed that the risk of the sibling of a patient with BD also having BD was 4.2%.26 In an international study of BD in European children,27 15% had parents or siblings with the disease. Human leukocyte antigen (HLA) B51 has been associated with the syndrome in adults in most populations. It is likely that B51 confers significant risk of BD (relative risk, 6.3 to 6.44), particularly in patients with a family history of the disease. 28 A study of microsatellite markers in Japanese and European adults with BD suggested that the pathogenic gene is HLA-B51 and not other genes located in its vicinity.29 MICA alleles may confer additional risk. 30 No associations with HLA-DR-DQ antigens have been reported.
ETIOLOGY AND PATHOGENESIS The cause of BD is unknown. It is speculated that a microbial agent is responsible for inducing an aberrant immune response in a genetically predisposed individual. Evidence for a microbial cause of BD is reviewed by Verity and colleagues. 31 Lehner3 2 has suggested that immunity to microbial heat-shock proteins that share homology with human 65 kD mitochondrial heat-shock protein may be important in pathogenesis. The y/8 T cells from patients with BD are highly responsive to four peptides from the human 60-kD heat-shock protein
u:
~
1ABLE 27 - J
CLINICAL MANIFESTATIONS The clinical manifestations of BD are varied and often emerge over a period of several years. In a study of 40 Korean children, the mean interval between the first and the second major manifestations was 7 years. 18 The usual course of BD in any organ system is that of exacerbations and remissions, with the overall activity generally declining with time. The frequencies of the major and minor manifestations are given in Table 27-3.
Mucocutaneous Disease Ulcerations Most BD patients have oral ulceration; it usually occurs at disease onset and may persist for much of the course of the disease. 12.20,21 Crops of extremely painful ulcers appear on the lips, tongue, palate, and elsewhere in the gastrointestinal tract (Fig. 27-1). They last for 3 to 10 days (sometimes longer), recur at various intervals, and usually heal without scarring. The exception to this is neonatal disease, in which extensive scarring may result, 24 Recurrent, painful ulcerations of the glans penis, prepuce, scrotum, and perianal area in the male and of the vulva and vagina in the female are characteristic. These ulcers usually occur after oral ulcers and, unlike them, may scar with healing.
Other Skin Lesions Other skin lesions occur in more than 90% of children with BD.12.21 They may be in the form of nodular lesions like erythema nodosum, purpura, papulopustular (acnei-
Beh( el's Disease ill Childhood
CrIteria (%) Study Lang et alB Bahabri et a!'" Kone-Paut et ajl4 EIdem et a!" Kim et all" Pivetti-Pezzi et aP9 Fujikawa and Suemitsu lO Uziel et al'l Total
n
M:F
37 12 65 20 40 16 31 15 236
19:18 7:5 33:32 15:5 16:24 9:7 14:17 7:8 120:116
'Includes pathergy. GU, genital ulceration: M:F, sex ratio: OU, oral ulceration.
OU
GU
5/dn
IJveItis
8.7 11.5 8.4 15.1 10.6
100 100 100 100 100
75 65 96 65 82
84' 83' 92' 35'
72'
30 92 45 80 27
6.6'
100
33
100'
53
Onset (yr)
I'athergy 57 80 ?
40
C HAP T E R
figure 27-1 insert.)
Oral aphthous lesion in a girl with
Beh~et's
27
BEHC;:ET'S DISEASE
563
disease. (See color
form) lesions, ulcers, or folliculitis. 21 .22 Pathergy, an unusual cutaneous pustular reaction occurring 24 to 48 hours after a needle puncture, is highly characteristic but not pathognomonic of the syndrome. 34 Pathergy test positivity varies considerably from one series to another and occurs most commonly (50% to 70%) in patients from the Middle East. 4
Ocular Disease Eye lesions occur in 30% to 61% of children with BD. 12,1(,-21 In a series of 20 children, posterior uveitis was the most common ocular manifestation, occurring in 11 patients, although anterior uveitis occurred in 3 patients, and 2 had papilledema. 17 The eye may be painful and red. Hypopyon may occur, and severe uveitis may lead to blindness. In older studies,35 blindness in untreated patients exceeded 90%. Complications of uveitis may include glaucoma and cataract. Corneal ulceration, cystoid macular degeneration, retinal vasculitis, retinal detachment, and retrobulbar neuritis are rare events. Ocular disease is much more common in boys than girls l2 ,17
Central Nervous System Disease The reported frequency of central nervous system (CNS) disease in children varies from 5% to 15% (Fig. 27_2).12,20.21 It may be parenchymal or nonparenchymal. 36 Four neurologic syndromes are recognized in patients with BD37: • Encephalomyelitis (e.g., pyramidal, extrapyramidal, cerebellar, spinal cord abnormalities, seizures) • Aseptic meningitis (e.g., headaches, meningitis) • Benign intracranial hypertension (e.g., pseudotumor cerebri, papilledema, dural sinus thrombosis) • Organic psychiatric disturbances (e.g., psychoses, depression, dementia) Several of these manifestations may occur simultaneously. The most common CNS disorder at presentation is meningoencephalitis with headache, stiff neck, focal neurologic abnormalities, and pleocytosis of the cere-
Figure 27-2 Magnetic resonance imaging demonstrates sagittal sinus thrombosis in a patient with Beh~et's disease. (Courtesy of Dr. A. Dine.)
brospinal fluid. 38 Neuro-BD develops in 5% to 15% of affected children. 12
Musculoskeletal Disease Polyarthritis or oligoarthritis occurs in 50%12 to 75%21 of children with BD. It most commonly affects the knees, ankles, wrists, and elbows but may occur in other joints as well. 12.21 The disease is usually oligoarticular, but polyarthritis is observed in at least one third of patients. 12 The disease does not usually result in erosions or joint destruction. There is probably no association of BD with sacroiliac arthritis, at least in childhood. 12,39 Acute, localized myositis is uncommon, is rarely multifocal,40 and has been reported in only a few children. 14 ,21 Generalized myositis may also occur, and both forms are reviewed by Sarui and associates. 4o It may be confused with vasculitis or venous thrombosis, especially when it affects the gastrocnemius muscle. 14
Vascular Disease Adamantiades 2 recognized the importance of vasculitis in BD, which is the only vasculitis that involves the arterial and venous systems. It is characteristically associated with arterial or venous thromboses. Superficial or deep venous thromboses are common in adults but occur only in 5% to 15% of children. 12 Most thromboses develop in veins, especially those of the lower extremities. Arteritis and arterial aneurysms may occur,41-43 and involvement of the pulmonary artery and central retinal artery has been reported. 12 Pulmonary artery thrombosis is rare, but it is one of the most severe features of the disease and is
564
C HAP T E R
27
BEH<;:ET'S DISEASE
associated with high morbidity and mortality rates. 44.45 Patients who develop superficial thrombophlebitis are more likely to develop major venous occlusions and must be carefully monitored. Dilatation and dropout of periungual capillaries was reported in 75% of adults in one study.46
Uncommon Manifestations Gastrointestinal Disease Diarrhea, abdominal pain, and ulceration of the ileum, cecum, and colon I2 ,20,21 appear to be more common in Japanese patients. 47 Gastrointestinal manifestations are characterized by exacerbations and remissions. Gastrointestinal tract lesions indistinguishable from those of Crohn's disease or ulcerative colitis may develop in patients with BD. Hepatic vein occlusion may result in Budd-Chiari syndrome. 48
Renal Disease Renal involvement is probably more common than was initially recognized. The most common manifestations are amyloidosis (occurring in patients as young as 13 years) and a wide spectrum of glomerulonephritides, but renal artery disease, renal vein thrombosis, and interstitial nephritis are also documented. 49 Renal involvement may occur in the form of vascular disease, including renal artery aneurysms or renal vein thrombosis.
Other Uncommon Manifestations A number of cardiac complications (e.g., endocarditis, myocarditis, pericarditis, arrhythmias) have been reported in adults but are rare in children. Dilatation of the proximal aorta, interatrial septal aneurysm, mitral valve prolapse, and mitral valve regurgitation were the most common abnormalities reported in one survey.50 Pulmonary abnormalities in BD have been reviewed by Erkan and colleagues.51 Recurrent dyspnea, cough, chest pain, and hemoptysis suggest pulmonary hemorrhage.
PATHOLOGY The underlying pathologic lesion is an occlusive vasculitis in arterioles and veins. In the skin, the lesions may be necrotizing but do not exhibit fibrinoid degeneration. 5 Inflammation in the synovium is nonspecific, with a predominantly neutrophil infiltration. Muscle biopsies have demonstrated a wide range of abnormalities, from perivascular infiltrates and fibrosis 14 to muscle necrosis,sz There is some controversy regarding the histopathology of the pathergy reaction. A serial study of the lesion revealed a superficial and deep perivascular mixed inflammatory cell infiltrate, with neutrophils peaking at 24 hours. 5' True vasculitis was not present. However, leukocytoclastic vasculitis and a neutrophilic vascular reaction with endothelial swelling has also been reported. 54 The differences in the reported series may be explained by the variability of the immune response or by ethnic factors.
LABORATORY EXAMINATION No laboratory findings are diagnostic of BD. There is a generalized increase in acute phase reactants with active disease, Levels of immunoglobulins and circulating immune complexes are increased.55 The frequencies of autoantibodies such as antinuclear antibody and rheumatoid factor are not increased, but antibodies to ocular and oral mucosal antigens have been observed.56 Antibodies to neutrophil cytoplasmic antigen 57 are uncommon and, when present, are associated with retinal vasculitis. Anticardiolipin antibodies are uncommon, and one study5H suggests that there is no primary role of these antibodies in BD. Elevated levels of von Willebrand factor antigen and decreased thrombomodulin levels were associated with vasculitis and active disease. Concentrations of protein C and protein S were normal. 59 Increased serum levels of tumor necrosis factor-C( (TNF-C()60 and soluble TNF-C( receptor6 1 may serve as markers of active disease. The pathogenic significance of elevated soluble interleukin-2 receptor (sIL-2R),60 IL-10,61 IL-12,61 and IL-862 requires further evaluation. Neutrophils are prominent infiltrating cells in the skin and eye lesions of BD, and inconsistent abnormalities of their function have been reported. Carletto and colleagues63 studied neutrophil function in 15 adults and found that although superoxide production and adhesion were normal, migration was significantly increased in patients with active disease compared with those with inactive disease or with controls. Enhanced migration into inflammatory sites facilitates their participation in the leukocytoclastic vasculitis or neutrophilic vasculitis that may occur in BD and may be at least partly responsible for pathergy. In other studies,!>'! levels of reactive oxygen species were increased. The role of neutrophil abnormalities in the pathogenesis of BD requires further study. Synovial fluid analysis is characterized by a predominance of neutrophils in relatively low numbers «15,000/mm' [<15 x 109/L]) and low glucose levels but no other distinguishing features. 61
IMAGING STUDIES The most important imaging studies are angiography to delineate the extent and character of the vascular lesions and magnetic resonance imaging to evaluate the effects of disease on the CNS. Magnetic resonance angiography is an alternative to direct angiography, although it is less accurate for imaging smaller arteries. 66 In a study of 98 adult patients,67 brain stem or basal ganglia lesions were demonstrated, especially during an attack. Similar findings were evident by single photon emission computed tomography.68
DIAGNOSIS In general, aphthous stomatitis is the presenting sign; other components of the syndrome may not appear for decades. The diagnosis is principally clinical, with laboratory tests supplying only supporting evidence. The differential diagnosis includes inflammatory bowel disease; aphthous stomatitis, erythema nodosum, arthritis, and uveitis occur in both diseases. In BD, the likelihood of occurrence of posterior uveitis is greater than in inflammatory bowel disease.
C HAP T E R
TREATMENT BD is difficult to treat, and treatment depends largely on the site and severity of involvement. There are no controlled studies evaluating treatment of BD in children, and physicians must therefore depend on the experience gained in treating adults. 36.69 Yazici and colleagues 36 suggest that young men have the worst prognosis and should be treated most aggressively. Whether this applies to pediatric patients is not certain.
Oral and Genital Ulcers Several approaches may be tried for oral and genital ulcers, including mouthwashes with sucralfate suspension or topical corticosteroids and systemic colchicine. During the acute stage of ulceration, a short course of oral prednisone helps provide fast relief. 69 For severe ulceration, thalidomide is very useful, but the high risk of peripheral neuropathy and its contraindication during pregnancy limits its usefulness. 7o A number of regimens have been used. An initial dose of thalidomide of 50 mg/day for adolescents is reduced to 50 mg twice per week if the patient responds. Alternatively, the drug may be started in a dose up to 1 mg/kg/day and tapered to 1 mg/kg taken 2 days each week or every other day. Dapsone (100 mg/day) has been reported to benefit the mucocutaneous lesions?! Long-term colchicine administration was effective in controlling the frequency and severity of oral and genital ulceration in one study. n A double-blind study enrolling adults with BD showed that colchicine significantly reduced the frequency of genital ulcers, erythema nodosum, and arthritis among women and arthritis among men. n
Ocular Inflammation Eye disease should be followed by an experienced ophthalmologist. Azathioprine (2 mg/kg/day) has been beneficial in trials surveyed in the Cochrane Controlled Trials Register,73 and it is one of the first choices of treatment for the severe uveitis of BD?4 The addition of cyclosporin to glucocorticoids has been recommended for the treatment of sight-threatening uveitis in adults. 75 The results of treatment with cyclosporin alone (5 mg/kg/day for 2 years) in 16 patients was generally favorable, and complete clinical remission was attained in 14 patients within 6 months. 76 Combination therapy with azathioprine and interferon-a or methotrexate have been successful for severe uveitis. 77 Reports have shown that infliximab is an effective alternative for all organ manifestations of disease, including severe panuveitis. 78
Treatment of Other Manifestations Prednisone is usually chosen as the initial therapy for other manifestations of BD. Methylprednisolone may be given intravenously initially at 10 to 20 mg/kg per dose, depending on the severity of the disease, or prednisone may be given orally in a dose of 1 to 2 mg/kg/day, followed by reducing the schedule to a dose of 0.2 mg/kg each day or every other day. Steroid-sparing or alternative
27
BEH<;:ET'S DISEASE
565
immunosuppressive treatment options include sulfasalazine, which has been reported to benefit the associated gastrointestinal disease?9 Treatment with low-dose methotrexate80 and chlorambucil (0,1 mg/kg/day)8I,82 has been successful in suppressing CNS disease. Short-term, high-dose therapy with this drug may be useful in treatment of intractable ocular disease. 83 Thalidomide was given to adults with mucocutaneous BD who did not have major organ involvement in doses of 100 mg/day with significant improvement in oral and genital ulceration within 4 to 8 weeks. 84 Peripheral neuropathy may be a limiting side effect of this drug, and thalidomide is a potent teratogen that must not be used in women of child-bearing age without contraception. New approaches to therapy for BD have been reviewed elswhere. 73 ,85 The frequency of postoperative complications such as suture detachment may be increased in BD patients because of an exagg~rated inflammatory response to simple penetrating trauma. However, this is not a contraindication to surgery.
COURSE OF THE DISEASE AND PROGNOSIS BD tends to run a very long, relapsing course. Young age at onset and male sex are both indicators of a prolonged disease course. The ocular and CNS manifestations in particular can be extremely incapacitating. 2! The young child who presents with only recurrent oral mucocutaneous lesions may develop genital ulcerations and gastrointestinal tract disease during adolescence. 86 Potentially fatal lesions include occlusion or aneurysms of arteries supplying the CNS or heart, pulmonary hemorrhage, and bowel perforation. 87.88 In a series of 65 patients, the mortality rate was 3%.!2
REFERENCES 1. Beh~et H: Uber rezidivierende Aphthose, durch ein Virus verusachte Geschwure am Mund, am Auge und an den Genitalien. Dermatol Wochenschr 105: 1151-1157. 1937. 2. Adamantiades B: Le thrombophlebite comme quatrieme symptome de I'irltis recidivante a hypopyon. Ann Ocul (Paris) 179: 143-148, 1946. 3. Matteson EL: Notes on the history of eponymic idiopathic vasculitis: the diseases of Henoch and Schbnlein, Wegener, Churg and Strauss, Horton, Takayasu, Beh~et, and Kawasaki. Arthritis Clin Res 13: 237-245, 2000. 4. Kaklamani VG, Vaiopoulos G, Kaklamanis PG: Beh~et's disease. Semin Arthritis Rheum 27: 197-217, 1998. 5. International Study Group for Beh~et's Disease: Criteria for diagnosis of Beh~et's disease, Lancet 335: 107(}-1080. 1990. 6, Mason RM, Barnes CGL: Beh~et's syndrome with arthritis. Ann Rheum Dis 28: 95-103, 1969, 7. O'Duffy JD, Goldstein NP: Neurologic involvement in seven patients with Beh~et's disease, Am J Med 61: 17(}-178, 1976, 8. Rigby AS, Chamberlain MA. Bhakta B: Classification and assessment of rheumatic diseases, Part 1. Beh~et's disease, Baillieres Clin Rheumatol 9: 375-395, 1995, 9. Dilsen N, History and development of Beh~et's disease. Rev Rhum (Engl Ed) 63: 512-519, 1996, 10, Sen DK: Endogenous uveitis in Indian children: analysis of 94 cases. J Pediatr Ophthalmol 14: 25-32, 1977. 11. Kone-Paut, 1, BernardJL: La maladie de Beh~et chez I'enfant en France. Arch Fr Pediatr 50: 561-565, 1993. 12, Kone-Paul I, Yurdakul S, Bahabri SA, et al: Epidemiological features of Beh~et's syndrome in children: an international collaborative survey of 86 cases. J Pediatr 132: 721-725, 1998. 13, Ozen S, Karaaslan Y. Ozdemir 0, et al: Prevalence of juvenile chronic arthritis and Familial Mediterranean Fever in Turkey, A field study. .I Rheumatol 25: 2445-2449, 1998, 14, Lang BA, Laxer RM, Thorner P, et al: Pediatric onset of Beh~et's syndrome With myositis: case report and literature review illustrating unusual features, Arthritis Rheum 33: 41&-425, 1990,
566
C HAP T E R
27
BEH<;:ET'S DISEASE
15, Vaiopoulos G, Kaklamani VG, Markomichelakis N, et al: Clinical features of juvenile Adamantiades-Beh~et's disease In Greece. Clin Exp Rheumatol 17: 256-259, 1999. 16. Bahabri SA, AI-Mazyed A, AI-Balaa S, et at: Juvenile Beh~et's disease in Arab children, Clin Exp Rheumatol 14: 331-335, 1996. 17, EIdem B, Onur C, Ozen S: Clinical features of pediatric Beh~et's disease. J Pediatr Ophthalmol Strabismus 35: 159--161, 1998. 18, Kim DK, Chang SN, Bang D, et al: Clinical analysis of 40 cases of childhoodonset Beh~et's disease. Pediatr Dermatol 11: 95-101, 1994. 19, Pivetti-Pezzi P, Accorinti M, Abdulaziz MA. et al: Beh~et's disease in children, Jpn J Ophthalmol 39: 309--314, 1995. 20, FUjikawa S, Suemitsu T: Beh~et disease in children: a nationwide retrospective survey in Japan. Acta Paediatr Jpn 39: 285-289, 1997, 21. Uziel Y, Brik R, Padeh S, et al: Juvenile Beh~et's disease in Israel. Clin Exp Rheumatol 16: 502-505, 1998. 22. Sarica R, Azizlerli G, Kose A, et al: Juvenile Beh~et's disease among 1784 Turkish Beh~et's patients, Int J Dermatol 35: 109--111, 1996. 23. Stark AC, Bhakta B, Chamberlain MA, et al: Life-threatening transient neonatal Beh~et's disease, Br J Rheumatol 36: 700-702, 1997. 24. Fam AG, Siminovitch KA, Carette S, From L: Neonatal Beh\;et's syndrome in an infant of a mother with the disease. Ann Rheum Dis 40: 509--512, 1981. 25. Lewis MA, Priestly Bt: Transient neonatal Beh~et's disease. Arch Dis Child 61: 805--806, 1986. 26. Gill A, InsanO M, Ocal L, et al: Familial aggregation of Beh~et's disease in Turkey. Ann Rheum Dis 59: 622-625, 2000. ' 27, Kone-Paut I, Geisler I, Wechsler B, et al: Familial aggregation in Beh~et's disease. J Pediatr 135: 89--93, 1999, 28, Chajek-Shaul T, Pisanty S, Knobler H, et al: HLA-B51 may serve as an immunogenetic marker for a subgroup of patients with Beh~et's syndrome, Am J Med 83: 666-672, 1987. 29. Mizuki N, Ota M, Yabuki K, et al: Localization of the pathogenic gene of Beh~et's Disease by microsatellite analysis of three different populations. Invest Ophthalmol Vis Sci 41: 3702-3708, 2000. 30. Wallace GR, Verity DH, Delamalne LJ, et al: MIC-A allele profiles and HLA class I associations in Beh~et's disease. Immunogenetics 49: 613-617, 1999, 31. Verity DH, Wallace GR, Vaughan RW, Stanford MR: Beh~et's disease: from Hippocrates to the third millennium. Br J Ophthalmol 87: 1175-1183, 2003, 32, Lehner T: The role of heat shock protein, microbial and autoimmune agents in the aetiology of Beh~et's disease, Int Rev Immunol 14: 21-32, 1997, 33, Hasan A, Fortune F, Wilson A, et al: Role of gamma delta T cells in pathogenesis and diagnosis of Beheet's disease, Role of gamma delta T cells in pathogenesis and diagnosis of Beh\;et's disease, Lancet 347: 1631-1632, 1996, 34. Yazici H, Tuzun Y, Pazarli H, et al: The combined used of HLA-B5 and the pathergy test as diagnostic markers of Beheet's disease in Turkey, J Rheumatol 7: 206-210, 1980, 35, Mamo JG: The rate of visual loss in Beh~et's disease, Arch Ophthalmol 84: 451-452, 1970. 36, Yazici H, Yurdakul S, Hamuryudan V: Beh~et's syndrome, CUff Opin Rheumatol 13: 18-22, 2001, 37, Kone-Paut I, Chabrol B, Riss JM, et al: Neurologic onset of Beheet's disease: a diagnostic enigma in childhood, J Child Neurol 12: 237-241, 1997, 38, O'Duffy JD, Goldstein NP: Neurologic involvement in seven patients with Beheet's disease, Am J Med 61: 170-178, 1976, 39. Chamberlain MA, Robertson RJ: A controlled study of sacroiliitis in Beh\;et's disease, Br J Rheumatol 32: 693-698, 1993, 40, Sarui H, Maruyama T, Ito I, et al: Necrotising myositis in Beheet's disease: characteristic features on magnetic resonance imaging and a review of the literature, Ann Rheum Dis 61: 751-752, 2002, 41. Enoch BA, Castillo-Olivares JL, Khoo TCL, et al: Major vascular complications In Beh~et's syndrome, Postgrad Med J 44: 453-459, 1968, 42, Davies JD: Beheet's syndrome with haemoptysis and pulmonary lesions, J Pathol 109: 351-356, 1973, 43. Grenier P, Bletry 0, Cornud F, et al: Pulmonary involvement in Beheet's disease, Am J Roentgenol 137: 565-569, 1981. 44, Erkan F: Pulmonary involvement In Beheet disease, Curr Opin Pulm Med 5: 314-318, 1999. 45. Agdad Kose A, Kayabali M, et al: Pulmonary artery involvement in Beheet's disease, Adv Exp Med BioI 528: 419-422, 2003, 46. Vaiopoulos G, Pangratis N, Samarkos M, et al: Nailfoid capillary abnormalities in Beheet's disease, J Rheumatol 22: 1108-1111, 1995, 47, Shimizu T, Ehrlich GE, Inaba G, Hayashi K: Beheet's disease (Beh~et's syndrome), Semin Arthritis Rheum 8: 223-260, 1979, 48, Bayraktar Y, Ozaslan E, van Thiel DH: Gastrointestinal manifestation of Beh\;et's disease, J Clin Gastroenterol 30: 144-154, 2000, 49, Akpolat T, Akkoyunlu M, Akpolat I, et al: Renal Beheet's disease: a cumulative analysis, Semin Arthritis Rheum 31: 317-337, 2002. 50, GOrgiin C, Ercan E, Ceyhun C, et al: Cardiovascular involvement in Beh~et's disease, Jpn Heart J 43: 389--399, 2002. 51, Erkan F, Glil A, Tasali E: Pulmonary manifestations of Beh~et disease. Thorax 56: 572-578, 2001. 52, Arkin CR, Rothschild BM, Florendo NT, Popoff N: Beh~et syndrome with myositis: a case report with pathologic fmdlngs. Arthritis Rheum 23: 600-604, 1980,
53. Ergun T, Gurbuz 0, Harvell J, et al: The histopathology of pathergy: a chronologic study of skin hyperreactivity in Beh~et's disease. Int J Dermatol 37: 929--933, 1998. 54. Jorizzo JL, Soloman AR, Cavallo T: Belwet's syndrome: inununopathological and histopathological assessment of pathergy lesions is useful in diagnosis and follow-up. Arch Pathol Lab Med 109: 747-751, 1985. 55. Scully C, Boyle P, Yap PL: Immunoglobulins G, M, A, 0, and E in Beh~et's syndrome. Cloin Chim Acta 120: 237-242, 1982. 56, K10k AM, de Vries J, Rothova A, et al: Antibodies against ocular and oral antigens in Beheet's disease associated with uveitis, Curr Eye Res 8: 957-962, 1989, 57. Vaiopoulos G, Hatzinicolaou P, Tsiroyanni A, et al: Antineutrophil cytoplasmic antibodies in Adamantiades-Behl;:et's disease. Br J Rheumato! 33: 406-407, 1994. 58, Tokay S, Direskeneli H, Yurdakul S, Akoglu T: Anticardiolipin antibodies in Beheet's disease: a reassessment. Rheumatology (Ox!) 40: 192-195,2001. 59, Demirer S, Sengiil N, Yerdel MA, et al: Haemostasis in patients with Beh~et's disease. Eur./ Vasc Endovasc Surg 19: 570-574, 2000, 60. Sayinalp N, Ozcebe 01, Ozdemir 0, et al: CYlOkines in Beh~et's disease. J Rheumatol 23: 321-322, 1996. 61. Turan B, Gallati H, Erdi H, et aI: Systemic levels of the T cell regulatory cytokines IL-1O and IL-12 in Beh~et's disease; soluble TNFR-75 as a biological marker of disease activity, J Rheumatol 24: 128-132, 1997. 62. A1-DaJaan A, AI-Sedairy S, A1-Balaa S, et al: Enhanced interleukin 8 secretion in drculation of patienL' with Beh~et's disease. J Rheumatol 22: 904-907, 1995. 63, Cadetto A, Pacor ML, Biasi D, et al: Changes of neutrophil migration without modification of in vitro metabolism and adhesion in Beheet's disease, J Rheumatol 24: 1332-1336, 1997, 64. Takeno M, Kariyone A, Yamashita N, et al: Excessive function of peripheral blood neutrophils from patients with Beh~et's disease and from HLA-B51 transgenic mice, Arthritis Rheum 38: 426-433, 1995. 65. Yurdakul S, Yazici H, Tuzun Y, et al: The arthritis of Beh~et's disease: a prospective srudy. Ann Rheum Dis 42: 505-515, 1983. 66. Akpolat T, Danaci M, Belet U, et al: MR imaging and MR angiography in vascular Beh~et's disease, Magn Reson Imaging 18: 1089--1096, 2000. 67. Akman-Demir G, Bahar S, Coban 0, et al: Cranial MRI in Beh~et's clisease: 134 examinations of 98 patients. Neuroradiology 45: 851--859, 2003. 68, Nobili F, Cutolo M, Sulli A, et al: Brain functional involvement by perfusion SPECT in systemic sclerosis and Beh~et's disease. Ann N Y Acad Sci 966: 409-414, 2002, 69. Ozen S: The spectrum of vasculitis In children, Baillieres Best Pract Res Clin Rheumatol 16: 411-425, 2002. 70, Kari JA, Shah V, Dillon MJ: Beheet's disease in UK children: clinical features and treatment including thalidomide, Rheumatology 40: 933-938, 2001. 71. Sharquie KE, Najim RA, Abu-Raghif AR: Dapsone in Beh~et's disease: a double blind placebo controlled cross over srudy. ./ Dermatol 29: 267-279, 2002, 72. Yurdakul S, Mat C, Tiizlin Y, et al: A double-blind trial of colchicine in Behl;:et's syndrome. Arthritis Rheum 44: 2686-2692, 2001. 73. Saenz A, Ausejo M, Shea B, et al: Pharmacotherapy for Beheet's Syndrome. Cochrane Database Syst Rev 2: CDOOI084, 2004. 74. Yazici H, Pazarli H, Barnes GG, et al: A controlled trial of azathioprine in Beh~et's syndrome. N Engl J Med 322: 281-285, 1990, 75. Whitcup SM, Salvo EC Jr, Nussenblatt RB: Combined cyclosporine and corticosteroid therapy for sight-threatening uveitis in Beheet's disease, Am J OphthaJmol 118: 39-45, 1994, 76. Pacor ML, Biasi 0, Lunardi C, et al: Cyclosporin in Beh~et's disease: resulL' in 16 patients after 24 months of therapy. Clin Rheumatol 13: 224-227, 1994, 77, Schirmer M, Calamia KT, Direskeneli H: International conference on Beh~et's disease. Seoul, Korea, 2000. J Rheumatol 28: 636-639, 2000. 78. Sfikakis PP, Beh~et's disease: a new target for anti-rumour necrosis factor treatment. Ann Rheum Dis 6l(Suppl 2): ii51-il53, 2002. 79, Yazici H, Yurdakul S, Hamuryudan V. Beh~et's syndrome. In Maddison P./, Isenberg DA, Woo P, Class ON (eds): Oxford Textbook of Rheumatology, 2nd ed. Oxford, Oxford University Press, 1998, pp 1394-1402. 80. Hirohata S, Suda H, Hashimoto T: Low-dose weekly methotrexate for progressive neuropsychiatric manifestations in Beh~et's disease, J Neurol Sci 159: 181-185, 1998. 81. Rakover Y, Adar H, Tal I , et al: Beheet disease: long-term followup of three children and review of the literature. Pediatrics 83: 986-992, 1989. 82. Matteson EL, O'Duffy JD: Treatment of Beh~et's disease with chlorambucil. In O'Duffy JD, Kokmen E (eds): Beh~et's Disease: Basic and Clinical Aspects, Proceedings of the Fifth International Conference on Beh~et's Disease. New York, Marcel Dekker, 1992, p 575. 83. Tessler HH, Jermings T: High-dose short-term cWorambucil for intractable sympathetic ophthalmia and Beheet's disease, Br./ Ophthalmol 74: 353-357, 1990. 84. Hamuryudan V, Mat C, Saip S, et al: Thalidomide in the treatment of the mucocutaneous lesIons of the Beh~et syndrome. A randomized, doubleblind, placebo-controlled trial. Ann Intern Med 128: 443-450, 1998, 85, Sakane T, Takeno M: Novel approaches to Beh~et's disease. Expert Opin Invest Drugs 9: 1993-2005, 2000, 86, Mundy TM, Miller JJ Ill: Beheet's disease presenting as chronic aphthous stomatitis In a child. Pediatrics 62: 205-208, 1978. 87. Ammann AJ, Johnson A, Fyfe GA, et al: Beh~et syndrome. J Pediatr 107: 41-43, 1985. 88. O'Duffy JD: Prognosis in Beheet's syndrome. Bull Rheum Dis 29: 972, 1978.
C H A P T E R
28
I NFECTIOUS A RTHRITIS AND O STEOMYELITIS Ross E. Petty and Ronald M. Laxer
The relation of infectious agents to arthritis is an area of great interest to the rheumatologist.1 Important discoveries have led to an understanding of the origin, pathogenesis, treatment, and cure of at least one infection-related arthritis—Lyme disease—and have given impetus to investigations of other possible arthritogenic infectious agents. Arthritis related to infection can be regarded as septic, reactive, or postinfectious.2 Septic arthritis occurs when a viable infectious agent is present or has been present in the synovial space. Although direct bacterial infection of the joint constitutes the most widely recognized form of septic arthritis, direct infection with viruses, spirochetes, or fungi also occurs. Reactive arthritis is a response to an infectious agent that is or has been present in some other part of the body, usually the upper airway, gastrointestinal tract, or genitourinary tract. By definition, viable infectious agents are not recoverable from the synovial space in patients with reactive arthritis, which may be regarded as an autoimmune disorder resulting from immunologic cross-reactivity between articular structures and infectious antigens. The reactive arthritis group merges pathogenically with diseases such as the seronegative spondyloarthropathies (i.e., Reiter’s syndrome and possibly juvenile ankylosing spondylitis). Postinfectious arthritis may be considered a special type of reactive arthritis in which immune complexes containing nonviable components of an initiating infectious agent may be present in the inflamed joint. Lyme disease is discussed in Chapter 29, the reactive arthritides in Chapter 30, and post-streptococcal arthritis in Chapter 31. The precise relation of infection to arthritis is complex and by no means completely understood.3 As techniques for the demonstration of infectious organisms improve, the frequency with which they are detected in synovial fluid or membrane is increasing, lending authority to the suspicion that some or many of the chronic arthritides of children are related to infectious diseases. Perhaps many of the so-called reactive arthritides will be found to represent diseases in which pathogens are present in the joint and are, by definition, septic. In some of the viral arthritides that fit the concept of reactive arthritis (i.e.,
568
joint disease follows the onset of the acute illness by days or weeks), viral antigen or living virus can be isolated from synovial fluid lymphocytes or membrane when appropriate techniques are used. The same has been true for Lyme disease, in which early attempts to demonstrate Borrelia were unsuccessful, although the organisms have since been demonstrated by silver stain in several different laboratories. The lesson implicit in all of these observations is that in chronic arthritides, which we currently consider aseptic, concerted investigations for infectious agents using the most powerful techniques of molecular biology may yet demonstrate the causative agent in the joint space. Although study of infectious agents such as viruses as possible initiators of some forms of arthritis in children has attracted much attention, it is important to remember that intra-articular and systemic bacterial infections remain the most important curable causes of arthritis in childhood.
SEPTIC ARTHRITIS Epidemiology Septic arthritis of bacterial origin accounts for approximately 6.5% of all childhood arthritis.4 It has been suggested that its frequency might be increasing,5,6 although the retrospective review by Fink and Nelson7 of 591 cases of septic arthritis in children in Dallas from 1955 to 1984 did not document an increase in incidence. In a 1997 report, 12.7% of 1158 patients with septic arthritis were children younger than age 10 years.8
Sex Ratio and Age at Onset Septic arthritis is slightly less common in girls than in boys, who account for 55% to 62% of patients in reported series.9–11 Septic arthritis is found most often in the very young12 and the very old; septic arthritis may occur in the neonate, is most common in children younger than 2 years, and diminishes in frequency throughout childhood.7,13
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
TABLE 28–1 Microorganisms Involved in Septic Arthritis and Osteomyelitis Age
Usual Organisms
Neonate
Group B Streptococcus Staphylococcus aureus Gram-negative bacilli Staphylococcus aureus Streptococcus species Haemophilus influenzae Staphylococcus aureus Streptococcus pneumoniae Group A Streptococcus Staphylococcus aureus Streptococcus pneumoniae Group A Streptococcus Neisseria gonorrhoeae
Infant Child Adolescent
Familial and Geographic Clustering There does not appear to be a genetic predisposition to septic arthritis. Typical cases of presumed septic arthritis in which no pathogen is identified tend to occur in the summer and fall,5 but geographic clustering has not been reported. In spirochetal arthritis, such as Lyme disease, there are marked geographic and seasonal outbreaks.
Etiology and Pathogenesis A wide range of microorganisms can cause septic arthritis in children; Staphylococcus aureus and non–group A β streptococci are most common overall.13–16 However, different organisms are more common at some ages and in certain circumstances2 (Table 28–1). Haemophilus influenzae type b had been the most common infection identified in children younger than 2 years, but vaccination of infants for H. influenzae has significantly decreased the frequency of infection with this organism.17–21 After the age of 10 years, it is rarely a cause of septic arthritis. Streptococcus pneumoniae is a frequent cause of infection in children younger than 2 years and is not uncommon in the older child.22,23 After 2 years of age, S. aureus is the most frequently occurring organism.18 Group A streptococci and enterococci account for a small proportion of all cases of septic arthritis in childhood and are most prevalent in the 6- to 10-year-old age group. Salmonella arthritis constitutes approximately 1% of all cases of septic arthritis, and it is commonly associated with sickle cell disease.24 Infection with Mycobacterium tuberculosis is an unusual cause of septic monarthritis in childhood. Other rare causes of infectious arthritis in children include Streptobacillus moniliformis (rat-bite fever), Pseudomonas aeruginosa, Bacteroides species, Campylobacter fetus, Serratia species, Corynebacterium pyogenes, Pasteurella multocida, and Propionibacterium acnes. In some parts of the world, Kingella kingae (formerly called Moraxella kingii) is emerging as an important pathogen in children with septic arthritis affecting knees, hips, or ankles.25,26 Most infections with this organisms occur in children younger than 5 years, and 60% occur in children younger than 2 years.27 In the neonate, S. aureus (40% to 50%) or group B Streptococcus (20% to
AND
OSTEOMYELITIS
569
25%) are the most common causative organisms.7,11,28 Enterobacteriaceae, gonococcus, and Candida species are also significant pathogens in the neonate. Septic arthritis usually arises by hematogenous spread from a focus of infection elsewhere in the body.29 Direct extension of an infection from overlying soft tissues (e.g., cellulitis, abscess) or bone (e.g., osteomyelitis)30 or traumatic invasion of the joint accounts for only 15% to 20% of cases. Joint damage results from several mechanisms. Proliferation of bacteria in the synovial membrane results in accumulation of polymorphonuclear (PMN) leukocytes and the inflammatory effects outlined in Chapter 4. Synovial fluid contains high levels of pro-inflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-1β)31 that mediate cartilage damage by metallopreoteinases.32 The ensuing damage to cartilaginous surfaces of the bone and the supporting structures of the joint may be severe and permanent if treatment is not urgently initiated. Although trauma or extra-articular infection preceding onset of septic arthritis is common in case histories, knowledge of the etiologic significance of these factors is incomplete.33 In one series, upper respiratory tract infections preceded septic arthritis in approximately 50% of patients, and approximately one third had received antibiotics within 1 week of onset.5 A history of a mild, nonpenetrating injury to the affected extremity was elicited in approximately one third of patients. Intravenous drug users are at particular risk for septic arthritis of the sacroiliac and sternoclavicular joints, usually caused by gram-negative organisms.34 Chronic inflammatory arthritis such as juvenile idiopathic arthritis may predispose to joint infection.35
Clinical Manifestations Septic arthritis is usually accompanied by systemic signs of illness (e.g., fever, vomiting, headache)36 and may be a component of a more generalized infection that may include meningitis, cellulitis, osteomyelitis, or pharyngitis.37 Joint pain is usually severe, and the infected joint and periarticular tissues are swollen, hot, and sometimes erythematous. Passive and active motion of the joint is severely, often completely, restricted (i.e., pseudoparalysis). Osteomyelitis frequently accompanies bacterial arthritis, and the presence of bone pain (as opposed to joint pain) should alert the examiner to this possibility. Other sites of hematogenous spread, although less common, are nonetheless important (Table 28–2).5,9,11
Affected Joints The joints of the lower extremity are most commonly the sites of infection. Knees, hips, ankles, and elbows account for 90% of infected joints in children. Septic arthritis affecting the small joints of the hands or feet is rare (Table 28–3).7,10,11 Pyogenic sacroiliac joint disease can occur.38
Multiple Infected Joints Although septic arthritis is most often a monarthritis, two or more joints are infected simultaneously or during the course of the same illness in a few children. In the large
570
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
OSTEOMYELITIS
TABLE 28–2 Extra-articular Sites of Infection in Children with Septic Arthritis Sites of Infection
Nelson and Koontz9 n = 117 (%)
Osteomyelitis Meningitis Cellulitis, abscess Respiratory tract Middle ear Urine Genital tract Pericardium Pleura
12 4 — 19 — — 4 — —
Welkon et al5 n = 95 (%)
Speiser et al11 n = 86 (%)
12 4 — — 20 — — — —
26 11 9 9 3 1 1 1 1
TABLE 28–3 Frequency of Infected Joints in Septic Arthritis Infected Joint Knee Hip Ankle Elbow Shoulder Wrist PIP, MCP, MTP Other
Fink and Nelson7 n = 591 (%) 40 23 13 14 4 4 1 1
Welkon et al5 n = 95 (%) 46 25 15 5 4 — — 5
Speiser et al11 n = 86 (%) 30 29 17 11 2 1 10 —
Wilson and DiPaola10 n = 61 (%) 29 40 21 3 3 1 — 1
Overall n = 833 (%) 39 25 14 12 4 3 2 1
MCP, metacarpophalangeal; MTP, metatarsophalangeal; PIP, proximal interphalangeal.
clinical experience reported by Fink and Nelson,7 septic arthritis was monarticular in 93.4% but affected two joints in 4.4%, three in 1.7%, and four in 0.5% of patients. Certain immune deficiencies, such as chronic granulomatous disease or acquired immunodeficiency syndrome (AIDS), may predispose to septic arthritis in multiple joints.
Diagnosis It is essential that every child with acute unexplained monarthritis undergo aspiration of the affected joint immediately because septic arthritis continues to be associated with considerable morbidity and mortality.17,39,40 Synovial fluid examination is outlined in Table 28–4. TABLE 28–4 Synovial Fluid Examination in a Child with Suspected Septic Arthritis Synovial fluid aspiration must be done under strictly aseptic conditions to minimize the risk of bacterial contamination. Septic arthritis is strongly suggested by the following: ■ ■ ■ ■ ■ ■ ■
Visual inspection finds cloudy, serosanguineous or greenish fluid. Cell count reveals elevated numbers of neutrophils (50,000– 300,000). Synovial fluid viscosity is low. Gram stain is positive. Synovial fluid glucose is low (< 30 mg/dL). Lactate dehydrogenase level is high (> 500 IU). Culture is positive for about 70% of those tested.
If an anaerobic organism or mycobacterium is suspected, enriched culture medium and special anaerobic culture conditions are necessary. Children in whom septic arthritis is considered should also have cultures of blood and of any potential source of infection (e.g., cellulitis, abscess, cerebrospinal fluid) performed. Rapid antigen latex agglutination tests for H. influenzae, group B and C streptococci, Neisseria meningitidis, and S. pneumoniae are available in most clinics. The polymerase chain reaction (PCR) has proved useful in detecting evidence of infectious agents in synovial fluid.41–44 In a group of children with septic arthritis in whom the bacterial agent was identified, Fink and Nelson7 reported that synovial fluid was culture positive in 307 (79%) of 389 patients (Table 28–5).5,10,11 The remaining 21% had positive cultures from sites other than the joint: blood (10%), cerebrospinal fluid (3.8%), blood and cerebrospinal fluid (2.3%), and vagina (1.3%). One of five children with culture-positive septic arthritis had a negative synovial fluid culture but a positive culture from elsewhere, most often the blood. Although an organism can be identified in one third to two thirds or more of patients by culturing of all appropriate sites, no causative organisms are ever identified in approximately one third of children with pyogenic arthritis.45 In these patients, the diagnosis of septic arthritis is based on a typical history and the demonstration of frank pus by arthrocentesis.
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
OSTEOMYELITIS
571
TABLE 28–5 Laboratory Confirmation of Septic Arthritis in Children Laboratory Confirmation Confirmed diagnosis (culture positive) Positive synovial fluid Gram stain Positive synovial fluid culture Positive blood culture
Fink and Nelson7 n = 591 (%) 66 33 79 33
Welkon et al5 n = 95 (%) 64 — 84 46
Speiser et al11 n = 86 (%) 84 19–54* 36–70* 46
Wilson and DiPaola10 n = 61 (%) 92 — 71-80† 41
*Depends on prior administration of antibiotics. † Depends on procedure (aspiration = 80, arthrotomy = 71).
Synovial Fluid Analysis The characteristics of the synovial fluid depend somewhat on the duration and severity of the disease and previous administration of antibiotics. Synovial fluid may appear normal, turbid, or grayish green with bloody streaks (see Table 28–4). The synovial fluid white blood cell (WBC) count is often markedly elevated, with 90% PMNs. Speiser and colleagues11 reported that synovial WBC counts in septic arthritis were less than 50,000/mm3 (50 × 109/L) in 15% of children, 50,000 to 100,000/mm3 in 34%, and more than 100,000/mm3 (100 × 109) in 51%. Fink and Nelson7 found a relatively low WBC count (less than 25,000/mm3 or 25 × 109) in one third of their patients. The protein content is high (more than 2.5 g/dL), and the glucose concentration compared with plasma glucose is usually low in septic arthritis, although it may be normal. A Gram stain identifies the organism in one half of untreated patients but in only one fifth of those who have received antibiotics. A Gram stain provides rapid confirmation of bacterial infection and tentative identification of the organism (if the findings are positive), permitting rational antibiotic therapy. Special procedures such as counterimmunoelectrophoresis, latex agglutination, or evaluation by PCR may sometimes identify bacterial antigens in a culture-negative fluid (i.e., blood, urine, or cerebrospinal fluid). These techniques have the advantage of providing antigenic identification much more rapidly than cultures, but they do not provide antibiotic sensitivities.
the affected area early in the course may demonstrate only increased soft tissue and capsular swelling. Juxta-articular osteoporosis reflects inflammatory hyperemia and is evident within several days after onset of infection. Cartilage loss and narrowing of the joint space develop as the disease progresses. These changes are followed by marginal erosions and eventually by ankylosis (Fig. 28-2).48 Computed tomography (CT) and especially magnetic resonance imaging (MRI) are additional confirmatory techniques. In the hip, accumulation of fluid within the joint displaces the gluteal fat lines laterally, or the obturator sign (i.e., displacement of the margins of this muscle medially) may be present. Traction applied to the leg during the radiographic procedure normally induces a radiolucent outline of the femoral head, the “vacuum” phenomenon. This lucency does not occur in the presence of increased intra-articular fluid.48
Ultrasonography The ultrasonic detection of an effusion in the hip of a child being treated for osteomyelitis of the femur indicates the presence of septic arthritis of that joint.48 Although the antibiotics administered for osteomyelitis and septic arthritis are similar, an effusion in the hip joint is sufficiently hazardous to the blood supply of the femoral head that open drainage is indicated.
Radionuclide Scans Blood Studies At least two blood cultures should always be performed for a child suspected of having septic arthritis. An elevated WBC count with a predominance of PMNs and bands and a markedly elevated erythrocyte sedimentation rate (ESR) or C-reactive protein (CRP) level—although of limited help in specific diagnosis—provide a baseline whereby the efficacy of subsequent treatment can be judged. CRP is a better predictor than ESR. If the CRP value was less than 1 mg/dL, the likelihood that the patient did not have septic arthritis was 87%.46 Concentrations of other acute phase reactants are usually increased but provide no additional useful information.
Radiologic Examination A number of imaging techniques are of value in evaluating a child with septic arthritis (Fig. 28–1).47–51 Radiographs of
During the first few days of disease, when plain radiographs show only soft tissue changes, technetium 99m scans reflect hyperemia of the infected area on blood flow studies and increased uptake of the isotope on both sides of the joint.49 This technique is useful in the early detection of joint or bone inflammation or infection, but it does not differentiate the two with certainty. Radionuclide scans with gallium 67 or the patient’s indium 111–labeled granulocytes can be used to identify accumulations of PMNs in infected sites, including joints with septic arthritis, but are not routinely needed.
Magnetic Resonance Imaging Delineation of soft tissue structures by MRI is superior to that provided by CT.48,51 Signal-intensity alterations in the bone marrow are characteristic but not diagnostic of septic arthritis (i.e., low intensity on fat-suppressed,
572
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
OSTEOMYELITIS ■ Figure 28–1 Flowchart of recommendations for evaluating septic arthritis in children. (Adapted from Jaramillo D, Treves ST, Kasser JR, et al: Osteomyelitis and septic arthritis in children: appropriate use of imaging to guide treatment. AJR Am J Roentgenol 165: 399–403, 1995.)
Plain radiographs
Suspected osteomyelitis
Osseous abnormality
Yes
No Reassess; evaluate for osteomyelitis
Hip sonography
Joint effusion
No
Yes Aspiration, therapy
MR imaging for possible osteomyelitis and abscess
No
Response in 48 hr
gadolinium-enhanced, T1-weighted spin-echo images and high signal intensity on fat-suppressed, T2-weighted, fast spin-echo images).52 Articular cartilage and growth cartilage in children are depicted along with other fibrous structures, muscle, blood vessels, and synovial fluid. An abnormal collection of fluid or debris, often displacing the joint capsule, eroding into other tissues, or in children, leading to subluxation, supports the possibility of septic arthritis.
Yes
Follow-up radiographs
Treatment The child with septic arthritis requires hospitalization and consultation with an orthopedic surgeon and specialist in infectious diseases. Nonsteroidal anti-inflammatory drugs may be used to help minimize the effects of inflammation, control fever, and contribute to pain relief. Intravenous dexamethasone was shown to reduce duration of symptoms and minimize joint damage in a
■ Figure 28–2 A, Questionable joint space widening of the right hip of a 10-year-old girl with fever and an irritable hip. B, Repeat xray film taken 20 days later demonstrated epiphyseal demineralization and erosion (arrow).
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
randomized, double-blind study.53 Dexamethasone (0.2 mg/kg/dose) or placebo was given immediately before the first dose of parenteral antibiotics and every 8 hours for a total of 12 doses. At the end of 12 months, 50 patients were evaluable in each group. Those who received dexamethasone had significantly shorter duration of symptoms and a significantly better outcome, with residual dysfunction in only 2%, compared with 26% in those who received placebo. A clinical practice guideline for the management of septic arthritis in children has been proposed.54 It was demonstrated that this approach was effective in minimizing bone scans, minimizing the rate of joint drainage, accelerating the change to oral antibiotic administration, and shortening the duration of hospital stay. There were no differences in outcomes such as readmission to hospital, recurrence of infection, or the development of residual joint damage.
Antibiotics In a child with septic arthritis, intravenous antibiotics should be administered as promptly as possible. The choice of antibiotic depends on the presence of predisposing factors, the age of the child, and the organisms that are suspected on the basis of the Gram stain or rapid antigen detection tests (although it is hazardous to narrow initial treatment based solely on these results because either can be wrong). If the Gram stain and results of rapid antigen detection are negative or not available, an approach based on age outlined in Table 28–6 is suggested.38 The demonstration of an organism or antigen may support or contradict the generalizations outlined in this table and should influence the physician in selection of the initial antibiotic treatment.39,55 It is prudent to monitor intravenous antibiotic efficacy with serum bactericidal titer determinations at frequent intervals initially and at follow-up for compliance, especially if home therapy is instituted. After satisfactory control of the infectious process with intravenous antibiotic administration is achieved, treatment by the oral route in the hospital or on an outpatient basis may be appropriate.56–58 Such a program should be undertaken only after careful consideration and consultation with an expert in pediatric infectious disease.
If the cultures are negative, intravenous antibiotics should be continued for a minimum of 21 days.2 If the child’s clinical state is improving (i.e., temperature returning to normal, pain diminishing, range of motion improving) and the WBC count and ESR are falling, the initial antibiotics should be maintained. If the patient does not appear to be responding, additional intravenous
TABLE 28–6 Recommended Empiric Antibiotic Therapy by Age Age
Recommended Empiric Antibiotic Therapy
Neonate Infant Child Adolescent
Cloxacillin + gentamicin Cefuroxime, cefotaxime Cefazolin Ceftriaxone or cefixime + azithromycin
AND
OSTEOMYELITIS
573
antibiotic coverage should be instituted. Because of various patterns of antibiotic susceptibility and resistance, guidelines regarding antibiotic choice and duration of treatment are constantly changing, and the physician is urged to review the most current recommendations.
Aspiration and Drainage The usefulness of repeated aspiration and drainage of an infected joint has been hotly debated. There is no dispute that an initial diagnostic arthrocentesis must be performed. Any joint that appears to be under pressure from an effusion can probably benefit from aspiration, if only for pain relief. Studies of the importance of repeated aspirations under other circumstances, however, have failed to show a consistent benefit. Similarly, open drainage is not better than closed needle aspiration (except for specific joints such as the hip) and is attended by significantly increased morbidity. Irrigation of the joint at the time of aspiration has no demonstrated additional benefit. Occasionally, arthroscopic examination is indicated.59 Intra-articular administration of antibiotic is unnecessary because therapeutic synovial fluid antibiotic levels are readily achieved,60 and it may induce chemical synovitis in the infected joint.
Special Cases Neonatal Septic Arthritis In addition to S. aureus, group B Streptococcus can be the offending organism in the neonate.61 It is a rare but potentially very serious infection in this age group and may have a subtle presentation. Most affected newborns show no fever, toxemia, or leukocytosis. Any infant who has swelling in the region of the thigh or holds the leg flexed, abducted, or externally rotated must be investigated promptly. Problems in early recognition of disease undoubtedly contribute to the often disastrous outcome of this involvement.62
Septic Hip Joint Septic arthritis of the hip is such an important problem that it merits special attention.63,64 The femoral head is intracapsular, and the arterial supply passes through the ligamentum teres through the intracapsular space. Increased intracapsular pressure can therefore interrupt the blood supply to the femoral head, with disastrous consequences to its viability, and the subsequent development of avascular necrosis.65 Metaphyseal osteomyelitis readily leads to septic arthritis of the hip joint in the infant because nutrient blood vessels pass from the metaphysis through the epiphyseal growth plate and terminate in the distal ossification center. Septic arthritis of the hip joint is most common in infants and very young children; 70% of patients are 4 years old or younger.66 The typical clinical picture is that of an infant or young child who may have an unexplained fever, is irritable, and refuses to move an extremity, bear weight, or walk. Any movement of the hip is extremely painful, and the affected leg is held in a
574
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
position of partial flexion, abduction, and external rotation at the hip. Occasionally, the child has lower abdominal pain or tenderness, sometimes with paralytic ileus. In very young or premature infants, a number of risk factors predispose to septic hip joint athritis.67 In a study of septic arthritis of the hip of 16 infants younger than 4 weeks, 11 were premature, 7 had an umbilical catheter, and 12 had septicemia. In contrast, of 13 children with septic arthritis between the ages of 1 month and 3 years, none was premature or had an umbilical catheter, and only 5 were septicemic. A high frequency of preceding or accompanying osteomyelitis of the femur or pelvis has also been observed. The association of septic arthritis of the hip and femoral venipuncture has been recorded68 and may account in part for the high frequency of arthritis of this site in the premature neonate. Management of septic arthritis of the hip requires open drainage to minimize intra-articular pressure.69,70 Traction and immobilization for the first 2 to 3 days of treatment provides pain relief but should be followed by passive and then active physiotherapy to prevent loss of range of motion. Prognosis is guarded even with the best treatment, especially in the neonate. The anatomy of the shoulder joint is not unlike that of the hip with respect to vascular supply. Septic arthritis of this joint, although rare, should be treated similarly.71
Gonococcal Arthritis In reported series of septic arthritis in children and adolescents, disease caused by N. gonorrhoeae appears to be uncommon. When it occurs it is most common in the adolescent, although it occasionally occurs in the neonate in association with disseminated infection.72 It is more common in girls than in boys and is particularly likely just after menstruation or with pregnancy.73 Gonococcal arthritis usually develops in patients with primary asymptomatic genitourinary gonorrhea or with a gonococcal infection of the throat or rectum. The patient presents with a systemic illness characterized by fever and chills.74 A vesiculopustular rash, sparsely distributed on the extremities, commonly yields organisms on culture or Gram stain of the smear. Gonococcal arthritis may have an initial migratory phase and may be accompanied by tenosynovitis. In contrast to most patients with septic arthritis, those with gonococcal arthritis may present with a purulent arthritis of several joints. For a patient with suspected gonococcal arthritis, it is important to culture samples from the genital tract, throat, rectum, and any vesicles in addition to the affected joint. The possibility of sexual abuse should be considered and appropriately investigated.
OSTEOMYELITIS
arises on a background of pulmonary tuberculosis as indolent, chronic monarthritis, often of the knee or wrist, that eventually results in extreme destruction of the joint and surrounding bones. Rarely, it manifests as acute arthritis.78 Al Matar and colleagues79 have observed two young children with tuberculous monarthritis that mimicked oligoarticular juvenile idiopathic arthritis. They were unresponsive to nonsteroidal anti-inflammatory drugs (NSAIDs) and intra-articular corticosteroids. One had a history of exposure, but the other child had no identifiable contact with tuberculosis. Joint infection occurs by hematogenous dissemination of the organism from adjoining osteomyelitis. Pott’s disease is a consequence of vertebral osteomyelitis (Fig. 28–3). Tuberculous dactylitis may occur with cystic expansion and destruction of bone (i.e., spina ventosa) (Fig. 28–4).80 A family or environmental history of pulmonary tuberculosis and a positive purified protein derivative (Mantoux) skin test result suggest the possibility of tuberculous arthritis. Although synovial fluid cultures are positive in approximately three fourths of patients, synovial membrane biopsy and culture are preferred and confirm the diagnosis in almost all patients (Fig. 28–5). The synovial WBC count is classically less than 50,000/mm3 (50 × 109/L), with a high proportion of mononuclear cells. Genus-specific PCR is invaluable in the diagnosis.44 Rarely, a polyarthritis accompanies tuberculosis (i.e., Poncet’s disease); it probably represents a reactive arthritis because culture of the inflamed joints fails to demonstrate tubercle bacilli.81,82 Leprosy can result in articular changes and inflammatory disease, including polyarthritis and subcutaneous nodules.83 Clinical differentiation from juvenile idiopathic arthritis may be difficult, especially if the possibility of leprosy is not considered in a nonendemic area.84 Mycobacterium leprae is not always easily identified in synovial biopsies.85,86
Tuberculous Arthritis Tuberculous arthritis is seldom encountered in North America or Europe, although its frequency may be increasing because of immunosuppressive therapy, drug-resistant strains of tuberculosis, and the human immunodeficiency virus (HIV) epidemic.75–77 Tuberculous arthritis is by no means rare in other parts of the world. Typically, arthritis
■ Figure 28–3 Pott’s disease of the vertebral column in an adolescent boy with pulmonary tuberculosis produced destruction of the disk space and vertebral end-plate erosion (arrow).
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
OSTEOMYELITIS
575
found that periarthritis without effusion was most common and that small joints and the spine were not affected. Whether this reflects differences in the infecting organism or in ascertainment is not known. No association with HLA-B27 has been demonstrated.88 Synovial fluid WBC counts are only modestly elevated, with a slight predominance of mononuclear cells.90,93 Joint fluid culture is positive for the organism in some patients. Tetracyclines, aminoglycosides, rifampin, and trimethoprim-sulfamethoxazole, often in combination, provide effective treatment of the acute infection, although permanent sequelae may result.87,91
Mycoplasma and Arthritis
■ Figure 28–4 Advanced osseous destruction occurred in the foot of a child with tuberculous dactylitis (i.e., spinal ventosa).
Myalgia and arthralgia are common during pulmonary infection with Mycoplasma pneumoniae. Objective oligoarticular, polyarticular, or migratory arthritis has also been described.94 Two children developed arthritis after an upper respiratory mycoplasmal infection,95 confirmed by enzyme-linked immunosorbent assay (ELISA) and Western blot analysis.
Bartonella Infection and Arthritis Arthritis Associated with Brucellosis Human Brucella infections are uncommon in North America, but there are European,87,88 Israeli,89 and South American90,91 reports of substantial numbers of patients with this infection complicated by arthritis. The species most frequently implicated are Brucella melitensis87,90,91 and, less commonly, Brucella canis.92 Unpasteurized milk is a source of infection. The systemic illness is often mild in children but is usually characterized by undulant fever, gastrointestinal complaints, lymphadenopathy, and sometimes dermatitis. In 88 children from Israel,89 the classic triad of fever (91%), arthralgia or arthritis (83%), and hepatosplenomegaly (63%) was characteristic of most patients. In a large series of cases from Peru,90 almost one third were children, and one third had arthritis. In the birth to 15-year age group, peripheral arthritis of a hip or knee was most common. Spondylitis and sacroiliac arthritis became predominant after 15 years of age. Gomez-Reino and colleagues87
■ Figure 28–5 Synovial biopsy specimen of chronic inflammation in tuberculous joint disease in which a giant cell (arrow) is indicated.
There have been rare reports of Bartonella henselae infection causing arthritis (i.e., cat-scratch disease).96–98 In two of the children, the disease mimicked systemic juvenile idiopathic arthritis97,98; the third child had polyarthritis and subcutaneous nodules.96
Arthritis in Immunocompromised Patients Chronic inflammatory arthritis in patients with a primary immunodeficiency is discussed in Chapter 33. Typical septic arthritis has been reported infrequently in immunodeficient children.99 Mycoplasma is the most common cause of severe chronic erosive arthritis in patients with congenital immunodeficiency syndromes100 and has been recovered from joints of patients with acquired immunodeficiency syndrome (AIDS).101 Ureaplasma urealyticum has been identified in patients with agammaglobulinemia.102 Candida albicans is occasionally responsible for arthritis in immunosuppressed patients.103
576
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
Course of the Disease and Prognosis The outcome in septic arthritis is somewhat guarded because, even with early and appropriate antibiotic treatment, permanent damage is common. The child usually recovers from the acute illness, but with the passage of time, reduction in range of motion, pain, and eventually degeneration of the surfaces of the affected joint may require surgical intervention. It is estimated that residual dysfunction occurs in 10% to 25% of children, although the changes (e.g., limited joint mobility, joint instability of chronic subluxation) may not be apparent until years later.5
OSTEOMYELITIS Although osteomyelitis, like septic arthritis, is most often encountered and treated by specialists in orthopedics and infectious diseases, its frequent association with septic arthritis and the diagnostic problems that it presents require that it be included in this discussion.6,56,104–109
Definition and Classification Osteomyelitis is an intraosseous infection with bacteria or, rarely, fungi. It is classified as acute, subacute, or chronic. Acute osteomyelitis is of recent onset and short duration. It is most often hematogenous in origin but may result from trauma such as a compound fracture or puncture wound. It can be metaphyseal, epiphyseal, or diaphyseal in location. Subacute osteomyelitis is of longer duration and is usually caused by less virulent organisms. Chronic osteomyelitis results from ineffective treatment of acute osteomyelitis and is characterized by necrosis and sequestration of bone.
Epidemiology Acute osteomyelitis is somewhat less common than acute septic arthritis. An incidence of 16.7 cases per year was reported from an institution at which acute septic arthritis occurred at a rate of 28.4 cases per year.7 However, it may be more common than septic arthritis in developing countries of the world.110 It occurs twice as often in boys as in girls7,110 and is more common in younger children. It can occur in the neonate.61
OSTEOMYELITIS
without underlying disease.121 Salmonella osteomyelitis is a complication of sickle cell anemia but also occurs in normal children.122 In the neonate, group B streptococci,123,124 gram-negative organisms,125–127 and Candida in addition to S. aureus are all potential causes of osteomyelitis. B. melitensis uncommonly results in osteomyelitis, but when it does, it has a predilection for the vertebral bodies.92 Tuberculous osteomyelitis may mimic chronic pyogenic disease, Brodie’s abscess, tumor, or other types of granuloma.128–130 B. henselae (organism of cat-scratch disease) has been identified as the causative agent in a few patients with osteomyelitis.131–133
Clinical Manifestations Fever, severe bone pain, and tenderness with or without local swelling should suggest the possibility of acute osteomyelitis. Although a history of prior trauma is elicited in approximately one third of young patients, its significance is uncertain. In the infant, fever may be minimal, and localization of the pain may be difficult on physical examination.134 Pseudoparalysis of a limb is often evident. The examiner may find clinical evidence of a preceding systemic infection. The site of infection is usually metaphyseal, and bony tenderness is elicited by pressure near or over the infected area. There may also be an area of overlying cellulitis, especially in the infant, in whom the thin cortex allows pus to erode into the periosteal structures. The presence of a joint effusion adjacent to the site of bone infection may reflect septic arthritis or a sterile noninflammatory “sympathetic” effusion.135 Osteomyelitis in children has a predilection for the metaphysis of rapidly growing bone. Many explanations have been suggested for this tendency. The anatomic differences in vasculature in this area in children and its easily compromised blood supply may in part explain the clinical observation (see Chapter 2). In one anatomic model, bacteremia and, in some cases, preceding microtrauma were sufficient to initiate disease.136 The bones of the lower extremity are affected in two thirds of patients, those of the upper extremity account for approximately 25%, but those of the skull, face, spine, and pelvis are the site of infection in fewer than 10% (Table 28–7).7, 110,137–139 Less than 10% of children have two or more simultaneously infected bones; in some cases, five or more bones are involved as part of a severe septicemic illness, usually caused by staphylococci. This type of involvement must be distinguished from chronic recurrent multifocal osteomyelitis (discussed later).
Etiology and Pathogenesis S. aureus (50% to 80%) and the group A streptococci (5% to 10%) are the predominant organisms at all ages.7,111 Even before specific immunization, H. influenzae seldom caused osteomyelitis (2% to 10%) and should now be even less common.21,112 In certain circumstances, specific or unusual organisms (15%) are found. For example, infection of the calcaneus or other bone in the foot associated with a puncture wound through athletic footwear is likely to be caused by P. aeruginosa.29,33,113,114 Osteomyelitis caused by S. pneumoniae22,115 usually occurs in children with associated diseases such as sickle cell anemia,116–118 asplenia,119 or hypogammaglobulinemia,120 although it has been observed in young infants
Brodie’s Abscess Subacute osteomyelitis, usually of staphylococcal origin, may develop after a penetrating injury or by hematogenous spread of an infection to the metaphysis. It is characterized clinically by localized soft tissue swelling and tenderness with marked pain that may awaken the child at night (Table 28–8). Radiographs demonstrate only soft tissue swelling in the first week, but metaphyseal osteolytic lesions are evident by the second week of the illness. They are most common in the proximal or distal ends of the tibia (Fig. 28–6).140,141 Culture of the abscess may be negative. Treatment includes intravenous antibiotics, an NSAID, and immobilization.
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
TABLE 28–7 Comparison of Affected Sites in Septic Osteomyelitis and Chronic Recurrent Multifocal Osteomyelitis Bone
Osteomyelitis* (%)
CRMO† (%)
25 <1 6 1 27 4
28 13 10 10 9 9
4 6 11 2 <1 <1 <1 <1 1 6
6 4 3 3 3 1 1 1 1 0
Tibia Clavicle Fibula Spine Femur Metatarsus, metacarpus, phalanx Radius Pelvis Humerus Ulna Sternum Mandible Scapula Rib Talus Calcaneus
AND
OSTEOMYELITIS
577
early detection of increased blood flow and uptake in the infected bone (see Fig. 28–7C).29,49,142 A bone scan is particularly helpful in localizing osteomyelitis in the neonate or infection of the axial skeleton and in searching for subclinical areas of infection in multifocal osteomyelitis. A positive result is not necessarily diagnostic of osteomyelitis, but a negative scan is unlikely for a child with bacterial osteomyelitis, except in the very early stages of the illness. MRI is often superior to other modalities in identifying changes in the marrow (see Fig. 28–7D).38,143–145 T1- or T2-weighted images, fat suppression, and gadolinium enhancement can confirm a focal area of increased inflammatory exudate (i.e., protons or water). A major advantage of MRI in early disease over plain radiographs, ultrasonography,146–148 or CT is the delineation of soft tissue or subperiosteal pus (see Fig. 28–8).149
Treatment *Data for osteomyelitis from Fink CW, Nelson JD: Septic arthritis and osteomyelitis in children. Clin Rheum Dis 12: 423, 1986, and from Cole WG, Dalziel RE, Leitl S: Treatment of acute osteomyelitis in childhood. J Bone Joint Surg Br 64: 218, 1982. † Data for chronic recurrent multifocal osteomyelitis (CRMO) from Sonozaki H, Miyanaga Y, et al: Clinical features of 53 cases with pustulotic arthro-osteitis. Ann Rheum Dis 40: 547, 1981. CRMO, chronic recurrent multifocal osteomyelitis.
Diagnosis As in septic arthritis, it is essential that every reasonable attempt be made to identify the organism and determine its antibiotic susceptibility.104 Aspiration of subperiosteal pus is the diagnostic procedure of choice and, together with cultures of the blood, synovial fluid, or infected wound, should yield an organism in approximately 70% to 80% of cases. A bone biopsy may be desirable or necessary if other sites of culture prove negative. The elevated WBC count and ESR are nonspecific and provide little help with diagnosis; they are useful in assessing effectiveness of therapy. Radiographic evaluation may delineate soft tissue swelling very early, but osteoporosis is not evident until days 10 to 14, and diagnostic findings may not be clear until days 10 to 21 (Fig. 28–7).47 Radionuclide scanning (i.e., technetium 99m polyphosphonate or diphosphonate) provides a sensitive if nonspecific method for the
In the absence of specific indications to the contrary,57 the initial antibiotic choices in the treatment of acute osteomyelitis should be effective against methicillinresistant S. aureus (see Table 28–6). Intravenous antibiotics for 4 to 6 weeks have been traditionally recommended, with subsequent oral coverage if appropriate. Recommendations have included a shortened course of intravenous treatment.149–151 Surgical treatment, which should be kept to a minimum,152 includes drainage of subperiosteal and soft tissue abscesses and débridement of associated lesions. Immobilization of the extremity for relief of pain is often necessary; otherwise, weight bearing may be permitted as tolerated by the patient.
Course of the Disease and Prognosis The most dreaded complications of acute osteomyelitis are chronic osteomyelitis and impaired bone growth.153,154 Chronic osteomyelitis should be suspected in a child whose systemic symptoms have responded slowly or incompletely to antibiotics or in whom there is a late recurrence of pain at the affected site. Radiographic studies at that time show a radiolucent involucrum (i.e., granulation tissue) surrounding dead (sclerotic) sequestered bone.
Differential Diagnosis and Related Disorders TABLE 28–8 Brodie’s Abscess Characteristic
Findings or Procedures
Symptoms Signs
Pain may be severe; child awakens at night. Evidence of a penetrating injury of hematogenous spread First week: soft tissue swelling Second week: metaphyseal osteolytic lesion with surrounding sclerosis Sterile joint effusion; curettage samples and cultures may be negative Immobilization, nonsteroidal anti-inflammatory drugs, antibiotics
Investigations Treatment
Chronic Recurrent Multifocal Osteomyelitis The syndrome known as chronic recurrent multifocal osteomyelitis (CRMO), first described by Giedion in 1972,140 clinically mimics septic osteomyelitis. The cause is unknown; cultures for bacteria are usually negative, although it has been speculated that a viral, chlamydial, mycoplasmal, or fastidious slow-growing organism could be responsible for this syndrome.155–161 Clinically, patients with CRMO experience an acute or insidious onset of multifocal bone pain accompanied by fever. The course of the disease is characterized by periodic painful relapses and remissions. In a review of the
578
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
OSTEOMYELITIS
■ Figure 28–6 Brodie’s abscess is revealed in radiographs of the knee of a 16-month-old boy with acute hematogenous osteomyelitis that was inadequately treated 1 month earlier. A, Central sequestration with a surrounding, ill-defined lytic margin.The patient was appropriately treated with antibiotics at this stage. B, One month later, the sequestrum has been removed by osteoclasts. C, One month later, the radiograph shows a well-defined lesion with sclerotic borders. (A to C, Courtesy of Dr. B. Wood.)
anatomic distribution of 181 lesions in 35 patients, Gamble and Rinsky157 found that long bones such as the tibia were most often affected. The ribs, clavicles, and vertebral bodies may also be involved162 (see Table 28–7). Radiographic changes are similar to those of septic osteomyelitis, with osteolytic lesions identified early in the illness that gradually become surrounded by sclerosis and enlargement of the affected bones (see Fig. 28–9). Unusual presentations may lead to diagnostic delay.162–164 Bone scans demonstrate the presence of multiple areas of involvement and occasional involvement of the epiphysis.165,166 Biopsy does not confirm an infectious organism but is often necessary to eliminate other diagnostic possibilities. Histologic examination shows necrosis and new bone formation together with acute and chronic inflammatory cells with fibrosis.156 Antibiotics are unnecessary and ineffective. NSAIDs and, occasionally, glucocorticoids bring symptomatic relief. Interferon-α has been used to treat one child with chronic recurrent multifocal osteomyelitis.167 Prognosis is uncertain, however, and the course of this painful disorder may be prolonged. The lesions of CRMO usually heal without specific treatment but may recur months or years later. In one study,168 7 of 12 children with CRMO followed for 2 to 20 years had residual deformity, most commonly leglength inequality, and only 2 patients had a complete recovery. Other reports have documented similar long-term deformities, although the outcome is satisfactory in most patients.165,169,170 In a large series from Japan, Sonozaki and coworkers169 identified inflammatory oligoarthritis in one fourth of patients and sacroiliac joint disease resembling ankylosing spondylitis in a small proportion. Bjorksten and associates171 described a cutaneous complication, palmoplantar pustulosis, in six of nine patients, and two of the seven patients reported by Laxer and colleagues156 had
palmoplantar pustulosis and psoriasis. The relation of these skin changes to the bony lesions of this syndrome is not known.172 Pyoderma gangrenosum has been described as a complication of CRMO.173 Caffey’s disease (i.e., infantile cortical hyperostosis), a rare disorder of infancy, may be confused with osteomyelitis. It is characterized by fever, irritability, and swelling associated with hyperostosis of the mandible, clavicles, and long bones174 (see Chapter 36).
SAPHO Syndrome Synovitis, acne, pustulosis, hyperostosis, and osteomyelitis constitute the SAPHO syndrome and may be another variant of CRMO.175 SAPHO usually has an insidious onset of local pain, swelling of affected bones (usually symmetrical) and enthesitis, sometimes with fever and elevated ESR. In this syndrome, characteristic skin lesions accompany the bony lesions of CRMO. In one review,176 260 children with CRMO syndrome were identified, and 10 additional patients were reported. The average age at onset was 10 years (range, 2.9 to 14 years). More than 80% were female. Sixty-one of the patients had skin lesions: palmoplantar pustulosis (37), other types of pustulosis (3) psoriasis (14), acne conglobata (4), Sweet syndrome (2), and pyoderma gangrenosum (1). The reason for the link between osteoarticular and cutaneous manifestations is unknown. There is no known genetic association, and the frequency of HLA-B27 is not different from that in the general population.
Arthritis Associated with Acne The association between arthritis and acne has been reviewed by Davis and associates.177 Most patients are
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
OSTEOMYELITIS
579
■ Figure 28–7 A, Anteroposterior radiograph of the pelvis of a 14-year-old body with fever and an irritable left hip.This x-ray film (A) and close-up of the left hip (B) are normal. C, Bone scan documents increased uptake of technetium 99m in the area of the left proximal femur (arrow). D, Magnetic resonance imaging demonstrates an increased marrow signal in the same area of the left hip (arrow), which indicates acute osteomyelitis.
male and have onset of musculoskeletal complications during adolescence. The syndrome includes severe truncal acne followed in several months by fever and arthralgia or arthritis, most often involving hips, knees, and shoulders. Myopathy may also accompany this disorder178 (see Chapter 18). We have observed one such boy with pain and swelling in the sternoclavicular, knee, and ankle joints and sacroiliac tenderness. Synovial histology was characterized by nonspecific proliferative synovitis. Sacroiliac joint radiographs confirmed early bilateral inflammatory changes. The patient was positive for HLAB27. It is possible that this syndrome is another example of reactive arthritis. Although arthritis lasts for only a few months in some patients, recurrences over many years have been documented.177,179 Treatment with NSAIDs and
with antibiotics for control of the acne is indicated. Isotretinoin can cause an acute arthritis.180–183
Diskitis There is considerable dispute about whether diskitis is basically an infectious process. Infection of an intervertebral disk space from osteomyelitis of an adjoining vertebral body is rare.184 However, acute diskitis unassociated with vertebral osteomyelitis is a self-limited inflammation of an intervertebral disk that may be caused by pathogens of low virulence, although bacteria or viruses are seldom recovered by aspiration. S. aureus and Enterobacteriaceae or Moraxella organisms are responsible in some patients. Diskitis occurs throughout childhood, but one half of
580
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
Suspected osteomyelitis
Plain radiographs
AND
Reassess
Negative
Osseous abnormality
No
Tc-MDP bone scan
Yes
Positive
Contrastenhanced MR imaging
Preoperative evaluation of spine (always) pelvis physis
Yes
No
No
Response in 48 hr
teristic tripod position during sitting or other unusual posturing.188 The child, who almost always has a low-grade fever usually refuses to walk, stand, or bend over; and may complain of abdominal pain. Palpation of the spine produces well-localized tenderness, usually in the lower lumbar region. The ESR is usually moderately elevated. Plain radiographs of the affected area often appear normal until late in the disease (Fig. 28–10). A technetium 99m bone scan is valuable diagnostically (Fig. 28–11). The L4-L5 interspace is most often affected (44%), followed by L3-L4 (37%), L2-L3 (7%), and L5-S1 (6%).185,186,189,190 The cervical spine may also be involved. In one study, disk space narrowing occurred in 82% of children, and a bone scan was positive in 72%.191 MRI may be valuable in differentiating infection from other conditions, including idiopathic disk calcification.192–194 Aspiration of the disk space or disk biopsy should not be routinely necessary. Treatment is supportive with immobilization. If bacterial infection is suspected, intravenous antibiotics should be instituted until results of blood cultures are available.
Whipple’s Disease
Therapy
MR imaging for possible abscess
OSTEOMYELITIS
Yes
Follow-up radiograph
■ Figure 28–8 Flowchart of the recommendations for evaluating acute hematogenous osteomyelitis in children (Adapted from Jaramillo D, Treves ST, Kasser JR, et al: Osteomyelitis and septic arthritis in children: appropriate use of imaging to guide treatment. AJR Am J Roentgenol 165: 399–403, 1995.)
the cases present before 4 years of age (peak age, 1 to 3 years).185,186 The sex ratio is approximately equal, although one review observed that diskitis occurred more frequently in girls.187 Clinical signs may be subtle. Diskitis is characterized by vague back pain and stiffness, often resulting in a charac-
Whipple’s disease, first described in 1907,195 is rare in childhood; approximately five children younger than 15 years have been reported.196 It is characterized by abdominal pain, weight loss, diarrhea, and in 65% to 90% of patients, arthralgias or arthritis.197–199 Whipple’s disease occurs 10 times more frequently in males than in females and is most common in middle age, although it has been identified in a 3-month-old boy200 and a 7-year-old boy201; there is also one report of central nervous system disease in a young boy.196 Migratory, peripheral joint pain and inflammation lasting hours to months occur over a period of many years, often in association with fatigue, weight loss, and anemia. Joint swelling with increased synovial fluid and restriction of range of motion may occur,202 although residual deformity does not.203 The joints most frequently affected are ankles, knees, shoulders, and wrists,202 and spondylitis has been reported in 20%.203 Periodic acid–Schiff–positive material and bacteria are detectable in macrophages infiltrating the upper small intestine and abdominal lymph nodes. Although the role of the bacterium (Tropheryma whippelii), if any, is unknown, antimicrobial therapy (e.g., tetracycline, penicillin, streptomycin) has greatly improved the outcome in this disease.
ARTHRITIS CAUSED BY VIRUSES
■ Figure 28–9 The patient had chronic, recurrent, multifocal osteomyelitis affecting both distal tibiae. Radiographs show lytic lesions of both distal tibiae (arrows) and periosteal new bone apposition (arrowhead).
A classification of viruses known to be associated with arthritis in humans is shown in Table 28–9.204 The togaviruses account for most of the viral arthritides. In general, viral arthritides occur much more often in adults than in children.205 Arthralgia is more common than objective arthritis, and both are usually migratory and of short duration (1 to 2 weeks), disappearing without residual joint disease. Small joints are most often affected by rubella, hepatitis B, and members of the alphavirus group (e.g., Ross River, chikungunya), whereas one or two large joints (usually the knees) are
C H A P T E R
■ Figure 28–10 disk space (arrow).
28 I N F E C T I O U S A RT H R I T I S
AND
OSTEOMYELITIS
581
A, Normal disk space is demonstrated on a lateral view of the lumbar spine. B, Diskitis has caused collapse of the
most often affected by mumps, varicella, and other viruses. In some viral arthritides, virus (e.g., rubella, varicella, herpes simplex, cytomegalovirus) can be isolated from the joint space; in others, only virus-containing immune complexes (e.g., hepatitis B, adenovirus 7) are found; and in still others, neither virus nor viral antigen can be recovered from the joint.206 Whether this represents limitations of recovery and culture techniques or the fact that culture-negative viral arthritis is “reactive” rather than “septic” is unknown.
common in young women. These symptoms are unusual in preadolescent children and in males, however, and are much less frequent after rubella immunization than after natural infection. Arthritis is more common after natural infection, and it is more severe and lasts longer.208
TABLE 28–9 Viruses That Cause Arthritis in Humans Virus
Rubella Virus Rubella-associated arthropathy was recognized by Osler207 and was one of the most commonly identified virusassociated arthritides in North America. Musculoskeletal symptoms after natural rubella infection are relatively
■ Figure 28–11 Technetium 99m bone scan of a 2.5-year-old girl with back pain demonstrates increased uptake of the isotope in the inferior end plate of L2 and superior end plate of L3 (arrow), which is characteristic of diskitis. (Courtesy of Dr. H.Y. Nadel.)
Togaviruses Rubivirus Rubella Alphaviruses Ross River Chikungunya O’nyong-nyong Mayaro Sindbis Ockelbo Pogosta Parvoviruses Hepadnaviruses Hepatitis B Adenoviruses Adenovirus 7 Herpesviruses Epstein-Barr Cytomegalovirus Varicella-zoster Herpes-simplex Paramyxoviruses Mumps Enteroviruses Echovirus Coxsackievirus B Orthopoxvirus Variola virus (smallpox) Vaccinia virus
Comment
Global; most reports from North America and Europe Australasia Africa, Asia Africa South America Africa, Asia, Australia Sweden Finland B19 associated with fifth disease Global Rare Rare; suggested role in RA Rare Rare Rare Rare Rare Rare Nonexistent today Rare
Adapted from Petty RE, Tingle AJ: Arthritis and viral infection. J Pediatr 113: 948, 1998.
582
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
Arthralgia usually begins within 7 days of the appearance of the rash or 10 to 28 days after immunization. The joints of the fingers and, later, the knees are most frequently affected. Joint pain may be accompanied by warmth, erythema, and effusion, and tenosynovitis is common. Carpal tunnel syndrome has also been reported. These findings usually disappear within 3 to 4 weeks but occasionally persist for months or even years. In a study of natural rubella infection in 37 teenage students, 52% of girls and 8% of boys developed objective arthritis,208 and an additional 13% and 48%, respectively, experienced arthralgia. In a group of young women who received RA 27/3 rubella vaccine, 14% developed an acute polyarthritis. Arthralgia and arthritis are most common in adults, although approximately 25% of prepubertal females develop arthralgia and 10% an arthritis.209,210 Chronic arthritis in adult women has been reported after vaccination in 5% to 11%.211,212 It has been suggested that reinfection contributes to arthritis in susceptible hosts.213,214 Rubella virus has been recovered from the synovial fluid of patients with rubella arthritis in many215,216 but not all instances.205,217 The virus was isolated from synovial or peripheral blood mononuclear cells in 7 of 19 children with juvenile rheumatoid arthritis but from no control subjects.218 Other investigations have shown no association.219
Parvovirus Parvoviruses are the latest candidates in the list of viruses putatively involved in the cause of rheumatoid arthritis in adults.220,221 Parvovirus RA-1 has been isolated from the synovial membrane of one patient with classic rheumatoid arthritis.222 A second parvovirus, B19, is the agent responsible for erythema infectiosum (i.e., fifth disease or slapped cheek syndrome).223,224 This illness is sometimes accompanied by an arthritis not unlike that of rubella infection.223,225–227 Arthralgia, symmetric joint swelling, and morning stiffness have been described in adults, especially women, after parvovirus B19 infection.227 Carpal tunnel syndrome, hepatitis, and angioedema have been described. This syndrome may be more widespread than previously thought and considerably underdiagnosed. In some patients, symptoms have persisted for years. An early study of erythema infectiosum in 364 patients indicated that joint pain, most often affecting knees and wrists, was present during the first week in 77% of adults and 8% of patients younger than 20 years.228 Subsequently, it was determined that arthritis was most common in patients who were HLA-DR4 positive.229 Whether a chronic arthritis results from parvovirus B19 infection is still controversial.206,230 Parvovirus infection is common and widespread. Parvovirus B19 genome consists of a linear, 5.6-kD, single-stranded DNA. There is only one serotype of parvovirus B19. Human parvovirus B19 has been implicated as the causative agent in erythema infectiosum, aplastic crises, some cases of hemophagocytic syndrome, and hydrops fetalis.231 Erythema infectiosum, or fifth disease, is a common exanthem of older children that lasts for a few days to a week and manifests with a low-grade fever, an erythematous facial rash (“slapped cheeks”), and a lacy, reticular rash on the extremities. These manifestations often recur with malaise, irritability, and arthralgia. Only a few cases of children with documented B19-associated arthritis have been reported.225,232–234 Joint symptoms tend to be mild and transient. In the children reported by Reid and coworkers,225 there was symmetric involvement of the small joints of the hands and feet. In one child, the arthritis preceded development of the rash. In another, arthritis persisted for 3 months. Antinuclear antibodies and rheumatoid factors were absent in all
OSTEOMYELITIS
of the patients in this series. Rivier and colleagues232 described a 5-year-old boy with arthritis of one knee that lasted for 6 weeks after typical erythema infectiosum. Nocton and associates234 described acute arthritis in 20 children with parvovirus B19 infection. The arthritis was associated with constitutional symptoms in one half of the children and was of brief duration (less than 4 months) in 14. Six children had persistent arthritis lasting up to 13 months; criteria for a diagnosis of juvenile idiopathic arthritis would have been met in this group. Laboratory results were generally normal, except for serologic evidence of the B19 infection.
The precise relation of the viral infection to arthritis has not been clarified. The virus has not been grown from synovial fluid or blood from patients with joint symptoms, although B19-specific DNA has been identified by hybridization in the synovial fluid of adults235 and by PCR amplification in synovial tissue.236 However, Soderlund and colleagues237 demonstrated genomic B19 DNA in the synovium of joints that had suffered trauma even more frequently than in those of children with chronic arthritis. Inflammatory synovitis may not be identifiable by arthroscopy. Although infection gives rise first to IgM antibodies238 and then to IgG antibodies, there is no evidence that the arthropathy represents an immune complex disease. Demonstration of IgM antibodies, however, is essential to diagnosis. The prevalence of IgG antibodies in the general population is too high to be diagnostically helpful, unless a fourfold increase concurrent with the clinical symptoms is demonstrated.239–241
Hepatitis B Arthritis-Dermatitis Syndrome In adults, up to 20% of infections with hepatitis B virus are characterized by a period of rash and arthritis that resembles serum sickness.242 In a review of reported cases of arthritis associated with hepatitis B infection,243 the age of the patients ranged from 14 to 56 years and the maleto-female ratio was 1.5:1. The dermatitis is characterized by a maculopapular rash, sometimes with petechiae, or urticaria and is most prominent on the lower extremities. The arthritis usually begins abruptly and symmetrically and affects the interphalangeal joints in 82%, knees in 30%, and ankles in 24% of patients. Although erythema and warmth are present, synovial effusions are uncommon. Joint symptoms last for 4 weeks on average, respond well to NSAIDs, and disappear without sequelae. The ESR is usually normal, although serum and synovial fluid complement levels are low in the early stages of the illness.244 Synovial fluid has been reported to show a mononuclear cell predominance.245 Electron microscopic evidence of hepatitis antigen in the synovial membrane has been reported.246 Hepatitis C virus is lymphotrophic and is associated with rheumatic symptoms resulting from mixed cryoglobulinemia.205
Alphaviruses Epidemic polyarthritis caused by infection with one of the alphaviruses is the most common virus-associated arthritis in Australia, the islands of the South Pacific, Africa, and Asia.247 These viruses are transmitted by
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
arthropods, usually the mosquito, and incite an illness characterized by arthritis and a rash that may be macular, papular, vesicular, or purpuric. Although there are some virus-specific differences in these illnesses, they usually are mild in children and occur with equal frequency in males and in females. In Ross River virus disease, the wrist is most commonly affected, often accompanied by tenosynovitis and enthesitis at the insertion of the plantar fascia into the calcaneus.247,248 The synovial fluid is said to be highly characteristic, with a predominance of vacuolated macrophages and very few PMNs. In chikungunya, the knee is the most commonly involved joint and back pain and myalgia are prominent. The arthritis lasts 1 to 2 weeks and is followed by complete recovery. Diagnosis rests on the clinical presentation and an elevated level of antibodies to the specific virus. Viral antigen has not been recovered from synovial fluid.
Herpesviruses Four of the herpesviruses have been associated with arthritis. Herpes simplex virus type 1 has been isolated from the synovial fluid of one patient with arthritis and disseminated herpes simplex infection.249 Epstein-Barr virus has long been thought by some investigators to have a primary role in the cause or pathogenesis of rheumatoid arthritis, although direct evidence is lacking.250–252 Arthritis is a rare complication of infectious mononucleosis.253–255 Cytomegalovirus is occasionally associated with arthritis and has been isolated from synovial fluid in one instance.249 Varicella-zoster infection is uncommonly complicated by arthritis.256–263 However, there have been instances of bacterial septic arthritis complicating chickenpox.261,264,265 In one instance, varicella-zoster virus was grown from synovial fluid of an 8-year-old girl with acute, painless monarthritis occurring 3 days after the onset of chickenpox.259 The synovial fluid cells were predominantly lymphocytes.256,259,266 Occasionally, chickenpox is associated with the emergence of psoriatic arthritis.267 Acute monarthritis has been reported in association with herpes zoster in two adults.268,269
Mumps Virus The paramyxovirus (mumps) rarely causes arthritis. In a 1984 review,270 only 32 cases were well documented. Since then, two additional patients have been reported.271,272 The male-to-female ratio is 3.6:1, and the peak age of occurrence is 21 to 30 years. Four patients younger than 11 years and seven between the ages of 11 and 20 years have been described. Arthritis occasionally preceded but usually followed parotitis by 1 to 3 weeks. In children, the arthritis was mild, affected few joints, and lasted 1 to 2 weeks. In postadolescent males, arthritis was often accompanied by orchitis and pancreatitis.270 It is reported that the arthritis responds to ibuprofen or prednisone but not to aspirin.270 The pathogenesis is unknown, and no attempts at recovery of mumps virus from synovium or synovial fluid have been reported. Arthritis has not occurred after mumps immunization.
AND
OSTEOMYELITIS
583
Human Immunodeficiency Virus AIDS resulting from HIV infection may be complicated by septic arthritis, usually of fungal origin.273,274 Although the precise nature of rheumatic syndromes associated with HIV infection remains controversial, a number of stereotypic presentations have been documented in adults.275 These include Reiter’s syndrome, reactive arthritis, psoriasiform arthritis, and an undifferentiated spondyloarthropathy,276–280 often more severe than in patients without HIV infection. Arthralgias occur initially with viremia; lower extremity oligoarthritis or persistent polyarthritis can supervene later. A study of 270 patients concluded that the most frequent pattern of joint involvement was one of an acute onset, short duration, few if any recurrences, and no erosive sequelae.281 We have observed chronic oligoarthritis in two young children with HIV infection (one transplacental and one related to blood products).
Other Viruses There are reports of arthritis associated with adenovirus type 7 infections, although virus was not isolated from the synovial fluid and diagnosis was confirmed only on clinical and serologic grounds.281–283 Echoviruses284–286 and coxsackie B viruses282,287 have been rarely implicated as the cause of arthritis. Smallpox (variola virus infection), now eradicated from the world, was often accompanied by arthritis, especially in children younger than 10 years. Arthritis also followed cowpox vaccination.288 Human T cell leukemia virus type 1 has been associated with a number of rheumatic disorders in adults, including arthritis and Sjögren’s syndrome.289 A 1998 report290 outlined an outbreak of Sindbis virus–induced Pogosta disease (e.g., fever, rash, joint symptoms) in Finland.
Syndromes Presumably Related to Viral Infection Transient or toxic synovitis of the hip is an idiopathic disorder often preceded by a nonspecific upper respiratory tract infection. It occurs most commonly in boys (70%) between 3 and 10 years old (Table 28–10).291 Pain in the hip, thigh, or knee may be of sudden or gradual onset and lasts for an average of 6 days. Bilateral involvement occurs in approximately 4% of cases. There is loss of internal rotation of the hip, and it may be held in the TABLE 28–10 Transient Synovitis of the Hip Feature
Description
Age at onset Sex ratio Symptoms Signs
3–10 years old Boys > girls Antecedent respiratory infection; limp, knee pain Decreased range of motion of hip; low-grade fever Slightly elevated to normal erythrocyte sedimentation rate; occasional widening of joint space, capsular distention Self-limited course of 1–2 weeks; relief with joint aspiration (coxa plana develops in 5%)
Investigations Treatment
584
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
flexed, abducted position. The ESR and WBC count are usually normal.291,292 Radiographs often appear normal or may document widening of the joint space with lateral displacement of the femoral head because of effusion. These findings can be confirmed by CT or ultrasound studies.291 Radionuclide scanning may demonstrate a transient decrease in uptake of technetium 99m phosphate. Signal intensity is normal with MRI and differentiates toxic synovitis of the hip from a septic process,52 which is often the principal differential diagnosis.293 After a diagnostic ultrasound scan to confirm the presence of fluid, the hip joint should be aspirated to exclude bacterial sepsis.294 The synovial fluid has a normal or minimally increased cell count but may be under high pressure.291 After aspiration, the pain and range of motion are dramatically improved, at least temporarily. Treatment includes the use of analgesics or NSAIDs, bed rest, and skin traction with the hip in 45 degrees of flexion to minimize intracapsular pressure.291 Long-term sequelae include Legg-Calvé-Perthes disease in about 1.5% of cases,295,296 coxa magna, and osteoarthritis. Recurrences are often accompanied by low-grade fever.
ARTHRITIS ASSOCIATED WITH OTHER INFECTIONS
OSTEOMYELITIS
■ Figure 28–12 Destruction of the first metatarsophalangeal joint was caused by sporotrichosis.
eign material. CT is indicated if plain radiographic results are negative. Because this disorder may develop months after the initial injury, foreign-body synovitis may be ignored as a diagnostic possibility. Treatment should be directed at appropriate surgical exploration and removal of the foreign material.312
Arthritis Caused by Spirochetes Lyme Disease
Fungal Arthritis Arthritis caused by fungal infection is rare297 and is almost unknown in children beyond the neonatal period. Fungi that have been reported as causing arthritis or osteomyelitis are C. albicans,298 Sporothrix schenkii,299,300 Actinomyces israelii,301 Aspergillus fumigatus,302 Histoplasma capsulatum,303 Cryptococcus neoformans,303 Blastomyces dermatididis,304 Coccidioides immitis,305 Paracoccidioides brasiliensis,306 Nocardia asteroides,307 and Pseudallescheria boydii.308 Candidal arthritis,298 often with accompanying osteomyelitis, is a recognized entity in the newborn309,310 and occurs occasionally in immunocompromised patients,273,274 and in patients with prosthetic joints.311
Sporotrichosis and Plant Thorn synovitis Infection with Sporothrix schenckii is a rare but significant occupational hazard of gardeners, night-crawler farmers, and field workers.299,300 Monarthritis or, less commonly, polyarthritis resembling rheumatoid arthritis has been reported (Fig. 28–12). Synovial biopsy is often necessary to make the diagnosis (Fig. 28–13). Other fungal infections are even less common causes of bone or joint infection in children. The interested reader is referred to the review by Goldenberg and Cohen300 and to other selected references.302–308,310,311 Synovitis caused by the penetration of a plant thorn into the joint space or surrounding structures is probably a reaction to the foreign material rather than an outright infection, although the circumstances of the injury may suggest the latter (Table 28–11). The synovial effusion is inflammatory, and culture occasionally yields a relatively nonvirulent organism. In the case of rose thorn penetration, S. schenckii is the probable cause. More commonly, the thorn of the palm tree or blackthorn is implicated.312 There are signs of local inflammation, and radiographs demonstrate periosteal new bone formation, a radiolucent defect in bone, or the presence of radiopaque for-
The geographic and temporal clustering of cases of what was thought to be juvenile rheumatoid arthritis in Old Lyme, Connecticut, led to the discovery and description of the cause, pathogenesis, and cure of Lyme disease. This epidemiologic work is one of the most important developments of the past quarter century in rheumatology and provides a model for approaching the question of the infectious cause of other chronic arthritides of childhood. It led directly to a vaccine for prevention of the illness. Chapter 29 provides a complete discussion of classic Lyme disease.
Other Spirochetes and Arthritis Arthritis rarely complicates leptospirosis (Leptospira icterohemorrhagica)313 and syphilis (Treponema pallidum).314,315 Congenital syphilis causes juxtaepiphyseal osteochondritis and periarthritis
■ Figure 28–13 Sporothrix schenckii is identified in a Gram-stained preparation of the synovial fluid aspirate.
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
TABLE 28–11 Plant Thorn Synovitis Feature
Description
Onset
Days to months after periarticular injury by penetrating foreign body (palm or blackthorn) Local inflammation; joint effusion Lytic lesion on radiographs, periosteal new bone apposition (may need CT scan); synovial fluid culture occasionally positive Surgical removal of foreign material
Signs Investigations Treatment
in infancy and syphilitic dactylitis in early childhood (Fig. 28–14). Clutton’s joints—relatively painless, recurrent, nonprogressive, symmetric synovitis of the knees—develop later.316
Parasites and Arthritis There have been case reports of arthritis accompanying a wide range of parasitic infestations,317 including Giardia intestinalis (lamblia),318 Endolimax nana,319 Toxocara canis,320 schistosomiasis,321 and others.322 In general, the joint disease presumably is reactive or postinfectious rather than septic and pursues a benign course with a good prognosis.
POSTINFECTIOUS ARTHRITIS Arthritis-Dermatitis Syndrome Associated with Small-Bowel Bypass A syndrome characterized by recurrent episodes of polyarthritis, often with an associated pustular vasculitis, occurred in 5% to 10% of adults who had undergone surgical bypass of the distal jejunum and proximal ileum for treatment of morbid obesity.323 This syndrome included arthritis or arthralgia in all patients, cutaneous lesions in 75%, paresthesia in 35%,
AND
OSTEOMYELITIS
585
Raynaud’s phenomenon in 29%, fever in 14%, and pericarditis in 3%.324 Morning stiffness occurred, and severe periarthritis with warmth and swelling most commonly affected knees, ankles, fingers, wrists, shoulders, and elbows. The ESR was usually elevated, and cryoglobulins were detected in one third of patients. Rheumatoid factors and antinuclear antibodies were seldom detected, and complement levels were usually normal.324 The pathogenesis of this syndrome is not certain, although circulating immune complexes have been implicated and sometimes shown to contain antibodies to Escherichia coli and Bacillus fragilis.325,326 Effective treatment included phenylbutazone, prednisone, and (if a blind-loop syndrome was suspected) tetracycline. To our knowledge, this syndrome has not been reported in children.
Musculoskeletal Manifestations of Systemic Bacterial Infections Bacterial infection of ventricular shunts for the management of hydrocephalus may result in arthritis and nephritis.327 Rheumatoid factors may be demonstrable in the sera of these patients. Meningococcemia is complicated by arthritis in up to 10% of cases.328 It is usually oligoarticular and occurs most often during the recovery phase, when immune complexes can be demonstrated in the synovium.329 It can also be complicated by acute septic arthritis in the early stage of the disease. H. influenzae type B meningitis may lead to a sterile arthritis.330 Infective endocarditis frequently causes arthralgia or arthritis331,332 and signs suggesting vasculitis (e.g., Osler nodes, Janeway lesions, Roth spots). The musculoskeletal signs and symptoms (e.g., arthralgia, arthritis, myalgia, low back pain) may precede other manifestations of infective endocarditis by weeks.332 The arthritis is characteristically polyarticular and symmetric, affecting both large and small joints. An immune complex–mediated pathogenesis is thought to be responsible, and the presence of hypocomplementemia,333 circulating immune complexes,334 and sometimes rheumatoid factors333 supports this theory. Specificity of the RFs is directed to the patient’s IgG in combination with the infecting organism.
R E F E R E N C E S
■ Figure 28–14 Lesions of congenital syphilis were identified in a 6-month-old girl brought to the child abuse clinic because of multiple fractures that occurred during the previous 10 days. Her rapid plasma reagin (RPR) test result was 1:256. A, Bilateral, symmetric, destructive metaphysitis lesions of the proximal ends of the tibiae (Wimberger’s sign) (solid arrow) and periosteal new bone apposition (open arrow) can be seen. B, With penicillin therapy, the lesions have almost healed 2 months later.
1. Rose CD, Eppes SC: Infection-related arthritis. Rheum Dis Clin North Am 23: 677–695, 1997. 2. Krogstad P, Smith AL: Osteomyelitis and septic arthritis. In Feigin RD, Cherry JD (eds): Textbook of Pediatric Infectious Diseases, 4th ed. Philadelphia, WB Saunders, 1998, pp 683–704. 3. Koopman WJ: Host factors in the pathogenesis of arthritis triggered by infectious organisms. Overview. Rheum Dis Clin North Am 19: 279–292, 1993. 4. Kunnamo I, Kallio P, Pelkonen P, et al: Clinical signs and laboratory tests in the differential diagnosis of arthritis in children. Am J Dis Child 141: 34–40, 1987. 5. Welkon CJ, Long SS, Fisher MC, et al: Pyogenic arthritis in infants and children: a review of 95 cases. Pediatr Infect Dis 5: 669–676, 1986. 6. Christiansen P, Frederiksen B, Glazowski J, et al: Epidemiologic, bacteriologic, and long-term follow-up data of children with acute hematogenous osteomyelitis and septic arthritis: a ten-year review. J Pediatr Orthop B 8: 302–305, 1999. 7. Fink CW, Nelson JD: Septic arthritis and osteomyelitis in children. Clin Rheum Dis 12: 423–435, 1986. 8. Ryan MJ, Kavanagh R, Wall PG, et al: Bacterial joint infections in England and Wales: analysis of bacterial isolates over a four year period. Br J Rheumatol 36: 370–373, 1997. 9. Nelson JD, Koontz WC: Septic arthritis in infants and children: a review of 117 cases. Pediatrics 38: 966–971, 1966. 10. Wilson NI, Di Paola M: Acute septic arthritis in infancy and childhood: 10 years’ experience. J Bone Joint Surg Br 68: 584–587, 1986.
586
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
11. Speiser JC, Moore TL, Osborn TG, et al: Changing trends in pediatric septic arthritis. Semin Arthritis Rheum 15: 132–138, 1985. 12. Wang CH, Huang FY: Septic arthritis in early infancy. Chung Hua Min Kuo Hsiao Erh Ko I Hsueh Hui Tsa Chih 31: 69–75, 1990. 13. Bonhoeffer J, Haeberle B, Schaad UB, Heininger U: Diagnosis of acute hematogenous osteomyelitis and septic arthritis: 20 years’ experience at the University Children’s Hospital, Basel. Swiss Med Wkly 131: 39–40, 575–581, 2001. 14. Ryan MJ, Kavanagh R, Wall PG, Hazleman BL: Bacterial joint infections in England and Wales: analysis of bacterial isolates over a four year period. Br J Rheumatol 36: 370–373, 1997. 15. Luhmann JD, Luhmann SJ: Etiology of septic arthritis in children: an update for the 1990s. Pediatr Emerg Care 15: 40–42, 1999. 16. Wang CL, Wang SM, Yang YJ, et al: Septic arthritis in children: relationship of causative pathogens, complications, and outcome. J Microbiol Immunol Infect 36:41–46, 2003. 17. Shetty AK, Gedalia A: Septic arthritis in children. Rheum Dis Clin North Am 24: 287–304, 1998. 18. Luhmann JD, Luhmann SJ: Etiology of septic arthritis in children: an update for the 1990s. Pediatr Emerg Care 15: 40–42, 1999. 19. Bowerman SG, Green NE, Mencio GA: Decline of bone and joint infections attributable to Haemophilus influenzae type b. Clin Orthop 341: 128–133, 1997. 20. Peltola H, Kallio MJ, Unkila-Kallio L: Reduced incidence of septic arthritis in children by Haemophilus influenzae type-b vaccination. Implications for treatment. J Bone Joint Surg Br 80: 471–473, 1998. 21. Howard AW, Viskontas D, Sabbagh C: Reduction in osteomyelitis and septic arthritis related to Haemophilus influenzae type b vaccination. J Pediatr Orthop 19: 705–709, 1999. 22. Bradley JS, Kaplan SL, Tan TQ, et al: Pediatric pneumococcal bone and joint infections. The Pediatric Multicenter Pneumococcal Surveillance Study Group (PMPSSG). Pediatrics 102: 1376–1382, 1998. 23. Ispahani P, Weston VC, Turner DP, et al: Septic arthritis due to Streptococcus pneumoniae in Nottingham, United Kingdom, 1985–1998. Clin Infect Dis 29: 1450–1454, 1999. 24. Wright J, Thomas P, Serjeant GR: Septicemia caused by Salmonella infection: an overlooked complication of sickle cell disease. J Pediatr 130: 394–399, 1997. 25. Yagupsky P: Kingella kingae: from medical rarity to an emerging paediatric pathogen. Lancet Infect Dis 4: 358–367, 2004. 26. Centers for Disease Control and Prevention (CDC): Kingella kingae infections in children—United States, June 2001–November 2002. MMWR Morb Mortal Wkly Rep 26: 244, 2004. 27. Yagupsky P, Dagan R, Howard CB, et al: Clinical features and epidemiology of invasive Kingella kingae infections in southern Israel. Pediatrics 92: 800–804, 1993. 28. Dan M: Septic arthritis in young infants: clinical and microbiologic correlations and therapeutic implications. Rev Infect Dis 6: 147–155, 1984. 29. Ezra E, Wientroub S: Primary subacute haematogenous osteomyelitis of the tarsal bones in children. J Bone Joint Surg Br 79: 983–986, 1997. 30. Jackson MA, Burry VF, Olson LC: Pyogenic arthritis associated with adjacent osteomyelitis: identification of the sequela-prone child. Pediatr Infect Dis J 11: 9–13, 1992. 31. Sáez-Lorens X, Mustafa MM, Ramilo O, et al: Tumor necrosis factor alpha and interleukin 1 beta in synovial fluid of infants and children with suppurative arthritis. Am J Dis Child 144: 353–356, 1990. 32. Jafari HS, Sáez-Lorenz X, Paris M, et al: Dexamethasone attenuation of cytokine-mediated articular cartilage degradation in experimental lapine Haemophilus arthritis. J Infect Dis 168: 1186–1193, 1993. 33. Laughlin TJ, Armstrong DG, Caporusso J, et al: Soft tissue and bone infections from puncture wounds in children. West J Med 166: 126–128, 1997. 34. Roca RP, Yoshikawa TT: Primary skeletal infections in heroin users: a clinical characterization, diagnosis and therapy. Clin Orthop 238–248, 1979. 35. Morrey BF, Bianco AJ Jr, Rhodes KH: Septic arthritis in children. Orthop Clin North Am 6: 923–934, 1975. 36. Caksen H, Ozturk MK, Uzum K, et al: Septic arthritis in childhood. Pediatr Int 42: 534–540, 2000. 37. Pioro MH, Mandell BF: Septic arthritis. Rheum Dis Clin North Am 23: 239–258, 1997. 38. Tokuda K, Yoshinaga M, Nishi J, et al: Three cases of pyogenic sacroiliitis, and factors in the relapse of the disease. Acta Paediatr Jpn 39: 385–389, 1997. 39. Mader JT, Mohan D, Calhoun J: A practical guide to the diagnosis and management of bone and joint infections. Drugs 54: 253–264, 1997. 40. Weston VC, Jones AC, Bradbury N, et al: Clinical features and outcome of septic arthritis in a single UK Health District 1982–1991. Ann Rheum Dis 58: 214–219, 1999. 41. Li F, Bulbul R, Schumacher HR Jr, et al: Molecular detection of bacterial DNA in venereal-associated arthritis. Arthritis Rheum 39: 950–958, 1996. 42. Louie JS, Liebling MR: The polymerase chain reaction in infectious and postinfectious arthritis. A review. Rheum Dis Clin North Am 24: 227–236, 1998. 43. Wilbrink B, van der Heijden I, Schouls LM, et al: Detection of bacterial DNA in joint samples from patients with undifferentiated arthritis and reactive arthritis, using polymerase chain reaction with universal 16S ribosomal RNA primers. Arthritis Rheum 41: 535–543, 1998. 44. van der Heijden I, Wilbrink B, Schouls LM, et al: Detection of mycobacteria in joint samples from patients with arthritis using a genus-specific poly-
OSTEOMYELITIS
45. 46. 47. 48. 49. 50. 51. 52. 53. 54.
55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80.
merase chain reaction and sequence analysis. Rheumatology (Oxf) 38: 547–553, 1999. Lyon RM, Evanich JD: Culture-negative septic arthritis in children. J Pediatr Orthop 19: 655–659, 1999. Levine MHJ, McGuire KJ, McGowan KL, Flynn JM: Assessment of the test characteristics of C-reactive protein for septic arthritis in children. J Pediatr Orthop 23: 373–377, 2003. Jaramillo D, Treves ST, Kasser JR, et al: Osteomyelitis and septic arthritis in children: appropriate use of imaging to guide treatment. Am J Roentgenol 165: 399–403, 1995. Poznanski AK, Conway JJ, Shkolnik A, et al: Radiological approaches in the evaluation of joint disease in children. Rheum Dis Clin North Am 13: 57–73, 1987. Sundberg SB, Savage JP, Foster BK: Technetium phosphate bone scan in the diagnosis of septic arthritis in childhood. J Pediatr Orthop 9: 579–585, 1989. Buchmann RF, Jarmillo D: Imaging of articular disorders in children. Radiol Clin North Am 42: 151–168, 2004. Marin C, Sanchez-Alegre ML, Gallego C, et al: Magnetic resonance imaging of osteoarticular infection in children. Curr Probl Diagn Radiol 33: 43–59, 2004. Lee SK, Suh KJ, Kim YW, et al: Septic arthritis versus transient synovitis at MR imaging: preliminary assessment with signal intensity alterations in bone marrow. Radiology 211: 459–465, 1999. Odio CM, Ramirez T, Arias G, et al: Double blind, randomized, placebo-controlled study of dexamethasone therapy for hematogenous septic arthritis in children. Pediatr Infect Dis J 22: 883–888, 2003. Kocher MS, Mandiga R, Murphy JM, et al: A clinical practice guideline for treatment of septic arthritis in children: efficacy in improving process of care and effect on outcome of septic arthritis of the hip. J Bone Joint Surg Am 85: 994–999, 2003. Cimmino MA: Recognition and management of bacterial arthritis. Drugs 54: 50–60, 1997. Sonnen GM, Henry NK: Pediatric bone and joint infections. Diagnosis and antimicrobial management. Pediatr Clin North Am 43: 933–947, 1996. Wall EJ: Childhood osteomyelitis and septic arthritis. Curr Opin Pediatr 10: 73–76, 1998. Newton PO, Ballock RT, Bradley JS: Oral antibiotic therapy of bacterial arthritis. Pediatr Infect Dis J 18: 1102–1103, 1999. Stanitski CL, Harvell JC, Fu FH: Arthroscopy in acute septic knees. Management in pediatric patients. Clin Orthop 241: 209–212, 1989. Nelson JD: Antibiotic concentrations in septic joint effusions. N Engl J Med 284: 349–353, 1971. Frederiksen B, Christiansen P, Knudsen FU: Acute osteomyelitis and septic arthritis in the neonate, risk factors and outcome. Eur J Pediatr 152: 577–580, 1993. Betz RR, Cooperman DR, Wopperer JM, et al: Late sequelae of septic arthritis of the hip in infancy and childhood. J Pediatr Orthop 10: 365–372, 1990. Bennett OM, Namnyak SS: Acute septic arthritis of the hip joint in infancy and childhood. Clin Orthop 123–132, 1992. Klein DM, Barbera C, Gray ST, et al: Sensitivity of objective parameters in the diagnosis of pediatric septic hips. Clin Orthop 338: 153–159, 1997. Vidigal EC Jr, Vidigal EC, Fernandes JL: Avascular necrosis as a complication of septic arthritis of the hip in children. Int Orthop 21: 389–392, 1997. Griffin PP, Green WT Sr: Hip joint infections in infants and children. Orthop Clin North Am 9: 123–134, 1978. Fabry G, Meire E: Septic arthritis of the hip in children: poor results after late and inadequate treatment. J Pediatr Orthop 3: 461–466, 1983. Chacha PB: Suppurative arthritis of the hip joint in infancy. A persistent diagnostic problem and possible complication of femoral venipuncture. J Bone Joint Surg Am 53: 538–544, 1971. Campagnaro JG, Donzelli O, Urso R, et al: Treatment of the sequelae of septic osteoarthritis of the hip during pediatric age. Chir Organi Mov 77: 233–245, 1992. Kim HK, Alman B, Cole WG: A shortened course of parenteral antibiotic therapy in the management of acute septic arthritis of the hip. J Pediatr Orthop 20: 44–47, 2000. Bos CF, Mol LJ, Obermann WR, et al: Late sequelae of neonatal septic arthritis of the shoulder. J Bone Joint Surg Br 80: 645–650, 1998. Scopelitis E, Martinez-Osuna P: Gonococcal arthritis. Rheum Dis Clin North Am 19: 363–377, 1993. Brogadir SP, Schimmer BM, Myers AR: Spectrum of the gonococcal arthritisdermatitis syndrome. Semin Arthritis Rheum 8: 177–183, 1979. Cohen M: Gonococcal arthritis. Bull Rheum Dis 47: 4–6, 1998. Rook G, Lydyard P, Stanford J: Mycobacteria and rheumatoid arthritis. Arthritis Rheum 33: 431–435, 1990. Ruggieri M, Pavone V, Polizzi A, et al: Tuberculosis of the ankle in childhood: clinical, roentgenographic and computed tomography findings. Clin Pediatr (Phila) 36: 529–534, 1997. Shih HN, Hsu RW, Lin TY: Tuberculosis of the long bone in children. Clin Orthop 335: 246–252, 1997. Zahraa J, Johnson D, Lim-Dunham JE, et al: Unusual features of osteoarticular tuberculosis in children. J Pediatr 129: 597–602, 1996. Al Matar M, Cabral DA, Petty RE: Isolated tuberculous monarthritis mimicking oligoarticular juvenile rheumatoid arthritis. J Rheumatol 28: 204–206, 2001. Wessels G, Hesseling PB, Beyers N: Skeletal tuberculosis: dactylitis and involvement of the skull. Pediatr Radiol 28: 234–236, 1998.
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
81. Southwood TR, Hancock EJ, Petty RE, et al: Tuberculous rheumatism (Poncet’s disease) in a child. Arthritis Rheum 31: 1311–1313, 1988. 82. Sood R, Wali JP, Handa R: Poncet’s disease in a north Indian hospital. Trop Doct 29: 33–36, 1999. 83. Pernambuco JC, Cossermelli-Messina W: Rheumatic manifestations of leprosy: clinical aspects. J Rheumatol 20: 897–899, 1993. 84. Markusse HM, Smelt AH, Teepe RG: Unusual arthritis: be on the alert for leprosy. Clin Rheumatol 8: 266–268, 1989. 85. Atkin SL, el-Ghobarey A, Kamel M, et al: Clinical and laboratory studies of arthritis in leprosy. Br Med J 298: 1423–1425, 1989. 86. Louie JS, Kornasky JR, Cohen AH: Lepra cells in synovial fluid of a patient with erythema nodosum leprosum. N Engl J Med 289: 1410–1411, 1973. 87. Gomez-Reino FJ, Mateo I, Fuertes A, et al: Brucellar arthritis in children and its successful treatment with trimethoprim-sulphamethoxazole (co-trimoxazole). Ann Rheum Dis 45: 256–258, 1986. 88. Ertem GT, Tanyel E, Tulek N, et al: Osteoarticular involvement of brucellosis and HLA-B27 antigen frequency ion Turkish patients. Diagn Microbiol Infect Dis 48: 243–245, 2004. 89. Gottesman G, Vanunu D, Maayan MC, et al: Childhood brucellosis in Israel. Pediatr Infect Dis J 15: 610–615, 1996. 90. Gotuzzo E, Alarcon GS, Bocanegra TS, et al: Articular involvement in human brucellosis: a retrospective analysis of 304 cases. Semin Arthritis Rheum 12: 245–255, 1982. 91. Thapar MK, Young EJ: Urban outbreak of goat cheese brucellosis. Pediatr Infect Dis 5: 640–643, 1986. 92. Young EJ: Human brucellosis. Rev Infect Dis 5: 821–842, 1983. 93. Press J, Peled N, Buskila D, Yagupsky P: Leukocyte count in the synovial fluid of children with culture proven brucellar arthritis. Clin Rheumatol 21: 191–193, 2002. 94. Broughton RA: Infections due to Mycoplasma pneumoniae in childhood. Pediatr Infect Dis 5: 71–85, 1986. 95. Cimolai N, Malleson P, Thomas E, et al: Mycoplasma pneumoniae associated arthropathy: confirmation of the association by determination of the antipolypeptide IgM response. J Rheumatol 16: 1150–1152, 1989. 96. Al-Matar MJ, Petty RE, Cabral DA, et al: Rheumatic manifestations of Bartonella infection in 2 children. J Rheumatol 29: 184–186, 2002. 97. Hayem F, Chacar S, Hayem G: Bartonella henselae infection mimicking systemic onset juvenile chronic arthritis in a 2-1/2 year-old girl. J Rheumatol 233: 1263–1265, 1996. 98. Tsukahara M, Tsuneoka H, Tateishi H, et al: Bartonella infection associated with systemic juvenile rheumatoid arthritis. Clin Infect Dis 32:E22–E23, 2001. 99. Arlievsky N, Li KI, Munoz JL: Septic arthritis with osteomyelitis due to Streptococcus pneumoniae in human immunodeficiency virus-infected children. Clin Infect Dis 27: 898–899, 1998. 100. Franz A, Webster AD, Furr PM, et al: Mycoplasmal arthritis in patients with primary immunoglobulin deficiency: clinical features and outcome in 18 patients. Br J Rheumatol 36: 661–668, 1997. 101. Gilbert MS, Aledort LM, Seremetis S, et al: Long term evaluation of septic arthritis in hemophilic patients. Clin Orthop 328: 54–59, 1996. 102. Asmar BI, Andresen J, Brown WJ: Ureaplasma urealyticum arthritis and bacteremia in agammaglobulinemia. Pediatr Infect Dis J 17: 73–76, 1998. 103. Swanson H, Hughes PA, Messer SA, et al: Candida albicans arthritis one year after successful treatment of fungemia in a healthy infant. J Pediatr 129: 688–694, 1996. 104. Morrissy RT, Shore SL: Bone and joint sepsis. Pediatr Clin North Am 33: 1551–1564, 1986. 105. Scott RJ, Christofersen MR, Robertson WW Jr, et al: Acute osteomyelitis in children: a review of 116 cases. J Pediatr Orthop 10: 649–652, 1990. 106. Hoffman EB, de Beer JD, Keys G, et al: Diaphyseal primary subacute osteomyelitis in children. J Pediatr Orthop 10: 250–254, 1990. 107. Faden H, Grossi M: Acute osteomyelitis in children. Reassessment of etiologic agents and their clinical characteristics. Am J Dis Child 145: 65–69, 1991. 108. Trobs R, Moritz R, Buhligen U, et al: Changing pattern of osteomyelitis in infants and children. Pediatr Surg Int 15: 363–372, 1999. 109. Nade S: Acute haematogenous osteomyelitis in infancy and childhood. J Bone Joint Surg Br 65: 109–119, 1983. 110. Cole WG, Dalziel RE, Leitl S: Treatment of acute osteomyelitis in childhood. J Bone Joint Surg Br 64: 218–223, 1982. 111. Dahl LB, Hoyland AL, Dramsdahl H, et al: Acute osteomyelitis in children: a population-based retrospective study 1965 to 1994. Scand J Infect Dis 30: 573–577, 1998. 112. Stricker T, Frohlich S, Nadal D: Osteomyelitis and septic arthritis due to Citrobacter freundii and Haemophilus influenzae type b. J Paediatr Child Health 34: 90–91, 1998. 113. Jaakkola J, Kehl D: Hematogenous calcaneal osteomyelitis in children. J Pediatr Orthop 19: 699–704, 1999. 114. Elliott SJ, Aronoff SC: Clinical presentation and management of Pseudomonas osteomyelitis. Clin Pediatr (Phila) 24: 566–570, 1985. 115. Jacobs NM: Pneumococcal osteomyelitis and arthritis in children. A hospital series and literature review. Am J Dis Child 145: 70–74, 1991. 116. Seeler RA, Metzger W, Mufson MA: Diplococcus pneumoniae infections in children with sickle cell anemia. Am J Dis Child 123: 8–10, 1972. 117. Sadat-Ali M: The status of acute osteomyelitis in sickle cell disease. A 15-year review. Int Surg 83: 84–87, 1998.
AND
OSTEOMYELITIS
587
118. Narchi H: Osteomyelitis in sickle cell haemoglobinopathy with elevated fetal haemoglobin. Ann Trop Paediatr 20: 70–75, 2000. 119. Mallouh A, Talab Y: Bone and joint infection in patients with sickle cell disease. J Pediatr Orthop 5: 158–162, 1985. 120. Kauffman CA, Watanakunakorn C, Phair JP: Pneumococcal arthritis. J Rheumatol 3: 409–419, 1976. 121. Hadari I, Dagan R, Gedalia A, et al: Pneumococcal osteomyelitis. An unusual cluster of cases. Clin Pediatr (Phila) 24: 143–145, 1985. 122. Sucato DJ, Gillespie R: Salmonella pelvic osteomyelitis in normal children: report of two cases and a review of the literature. J Pediatr Orthop 17: 463–466, 1997. 123. Perkins MD, Edwards KM, Heller RM, et al: Neonatal group B streptococcal osteomyelitis and suppurative arthritis. Outpatient therapy. Clin Pediatr (Phila) 28: 229–230, 1989. 124. Barton LL, Villar RG, Rice SA: Neonatal group B streptococcal vertebral osteomyelitis. Pediatrics 98: 459–461, 1996. 125. Knudsen CJ, Hoffman EB: Neonatal osteomyelitis. J Bone Joint Surg Br 72: 846–851, 1990. 126. Gutman LT: Acute, subacute, and chronic osteomyelitis and pyogenic arthritis in children. Curr Probl Pediatr 15: 1–72, 1985. 127. Deshpande PG, Wagle SU, Mehta SD, et al: Neonatal osteomyelitis and septic arthritis. Indian Pediatr 27: 453–457, 1990. 128. Vohra R, Kang HS, Dogra S, et al: Tuberculous osteomyelitis. J Bone Joint Surg Br 79: 562–566, 1997. 129. Chen SC, Huang SC, Wu CT: Nonspinal tuberculous osteomyelitis in children. J Formos Med Assoc 97: 26–31, 1998. 130. Wang MN, Chen WM, Lee KS, et al: Tuberculous osteomyelitis in young children. J Pediatr Orthop 19: 151–155, 1999. 131. Keret D, Giladi M, Kletter Y, et al: Cat-scratch disease osteomyelitis from a dog scratch. J Bone Joint Surg Br 80: 766–767, 1998. 132. Ratner LM, Kesack A, McCauley TR, et al: Disseminated Bartonella henselae (cat-scratch disease): appearance of multifocal osteomyelitis with MR imaging. AJR Am J Roentgenol 171: 1164–1165, 1998. 133. Robson JM, Harte GJ, Osborne DR, et al: Cat-scratch disease with paravertebral mass and osteomyelitis. Clin Infect Dis 28: 274–278, 1999. 134. Willis RB, Rozencwaig R: Pediatric osteomyelitis masquerading as skeletal neoplasia. Orthop Clin North Am 27: 625–634, 1996. 135. Perlman MH, Patzakis MJ, Kumar PJ, et al: The incidence of joint involvement with adjacent osteomyelitis in pediatric patients. J Pediatr Orthop 20: 40–43, 2000. 136. Morrissy RT, Haynes DW: Acute hematogenous osteomyelitis: a model with trauma as an etiology. J Pediatr Orthop 9: 447–456, 1989. 137. Tyrrell PN, Cassar-Pullicino VN, Eisenstein SM, et al: Back pain in childhood. Ann Rheum Dis 55: 789–793, 1996. 138. Chelsom J, Solberg CO: Vertebral osteomyelitis at a Norwegian university hospital 1987–1997: clinical features, laboratory findings and outcome. Scand J Infect Dis 30: 147–151, 1998. 139. Krogsgaard MR, Wagn P, Bengtsson J: Epidemiology of acute vertebral osteomyelitis in Denmark: 137 cases in Denmark 1978–1982, compared to cases reported to the National Patient Register 1991–1993. Acta Orthop Scand 69: 513–517, 1998. 140. Giedion A, Holthusen W, Masel LF, et al: Subacute and chronic “symmetrical” osteomyelitis. Ann Radiol (Paris) 15: 329–342, 1972. 141. Green NE, Beauchamp RD, Griffin PP: Primary subacute epiphyseal osteomyelitis. J Bone Joint Surg Am 63: 107–114, 1981. 142. Pennington WT, Mott MP, Thometz JG, et al: Photopenic bone scan osteomyelitis: a clinical perspective. J Pediatr Orthop 19: 695–698, 1999. 143. Cohen MD, Cory DA, Kleiman M, et al: Magnetic resonance differentiation of acute and chronic osteomyelitis in children. Clin Radiol 41: 53–56, 1990. 144. Mandell GA: Imaging in the diagnosis of musculoskeletal infections in children. Curr Probl Pediatr 26: 218–237, 1996. 145. Umans H, Haramati N, Flusser G: The diagnostic role of gadolinium enhanced MRI in distinguishing between acute medullary bone infarct and osteomyelitis. Magn Reson Imaging 18: 255–262, 2000. 146. Howard CB, Einhorn M, Dagan R, et al: Ultrasound in diagnosis and management of acute haematogenous osteomyelitis in children. J Bone Joint Surg Br 75: 79–82, 1993. 147. Chao HC, Lin SJ, Huang YC, et al: Color Doppler ultrasonographic evaluation of osteomyelitis in children. J Ultrasound Med 18: 729–734, 1999. 148. William RR, Hussein SS, Jeans WD, et al: A prospective study of soft-tissue ultrasonography in sickle cell disease patients with suspected osteomyelitis. Clin Radiol 55: 307–310, 2000. 149. Azouz EM, Greenspan A, Marton D: CT evaluation of primary epiphyseal bone abscesses. Skeletal Radiol 22: 17–23, 1993. 150. Nelson JD: Toward simple but safe management of osteomyelitis. Pediatrics 99: 883–884, 1997. 151. Karwowska A, Davies HD, Jadavji T: Epidemiology and outcome of osteomyelitis in the era of sequential intravenous-oral therapy. Pediatr Infect Dis J 17: 1021–1026, 1998. 152. Hamdy RC, Lawton L, Carey T, et al: Subacute hematogenous osteomyelitis: are biopsy and surgery always indicated? J Pediatr Orthop 16: 220–223, 1996. 153. Gledhill RB: Subacute osteomyelitis in children. Clin Orthop 96: 57–69, 1973. 154. Reinehr T, Burk G, Michel E, Andler W: Chronic osteomyelitis in childhood: is surgery always indicated? Infection 28: 282–286, 2000. 155. Speer DP: Chronic multifocal symmetrical osteomyelitis. Am J Dis Child 138: 340, 1984.
588
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
156. Laxer RM, Shore AD, Manson D, et al: Chronic recurrent multifocal osteomyelitis and psoriasis—a report of a new association and review of related disorders. Semin Arthritis Rheum 17: 260–270, 1988. 157. Gamble JG, Rinsky LA: Chronic recurrent multifocal osteomyelitis: a distinct clinical entity. J Pediatr Orthop 6: 579–584, 1986. 158. Van Howe RS, Starshak RJ, Chusid MJ: Chronic, recurrent multifocal osteomyelitis. Case report and review of the literature. Clin Pediatr (Phila) 28: 54–59, 1989. 159. Kourtis AP, Ibegbu CC, Snitzer JA, et al: Recurrent multifocal osteomyelitis due to Mycobacterium avium complex. Clin Infect Dis 23: 1194–1195, 1996. 160. Handrick W, Hormann D, Voppmann A, et al: Chronic recurrent multifocal osteomyelitis—report of eight patients. Pediatr Surg Int 14: 195–198, 1998. 161. Girschick HJ, Huppertz HI, Harmsen D, et al: Chronic recurrent multifocal osteomyelitis in children: diagnostic value of histopathology and microbial testing. Hum Pathol 30: 59–65, 1999. 162. Martin JC, Desoysa R, O’Sullivan MM, et al: Chronic recurrent multifocal osteomyelitis: spinal involvement and radiological appearances. Br J Rheumatol 35: 1019–1021, 1996. 163. Quelquejay C, Job-Deslandre C, Hamidou A, et al: Chronic recurrent multifocal osteomyelitis in children. J Radiol 78: 115–121, 1997. 164. Bousvaros A, Marcon M, Treem W, et al: Chronic recurrent multifocal osteomyelitis associated with chronic inflammatory bowel disease in children. Dig Dis Sci 44: 2500–2507, 1999. 165. Manson D, Wilmot DM, King S, et al: Physeal involvement in chronic recurrent multifocal osteomyelitis. Pediatr Radiol 20: 76–79, 1989. 166. Mandell GA, Contreras SJ, Conard K, et al: Bone scintigraphy in the detection of chronic recurrent multifocal osteomyelitis. J Nucl Med 39: 1778–1783, 1998. 167. Gallagher KT, Roberts RL, MacFarlane JA, et al: Treatment of chronic recurrent multifocal osteomyelitis with interferon gamma. J Pediatr 131: 470–472, 1997. 168. Duffy CM, Lam PY, Ditchfield M, et al: Chronic recurrent multifocal osteomyelitis: review of orthopaedic complications at maturity. J Pediatr Orthop 22: 501–505, 2002. 169. Sonozaki H, Mitsui H, Miyanaga Y, et al: Clinical features of 53 cases with pustulotic arthro-osteitis. Ann Rheum Dis 40: 547–553, 1981. 170. Peters W, Irving J, Letts M: Long-term effects of neonatal bone and joint infection on adjacent growth plates. J Pediatr Orthop 12: 806–810, 1992. 171. Bjorksten B, Gustavson KH, Eriksson B, et al: Chronic recurrent multifocal osteomyelitis and pustulosis palmoplantaris. J Pediatr 93: 227–231, 1978. 172. Ravelli A, Marseglia GL, Viola S, et al: Chronic recurrent multifocal osteomyelitis with unusual features. Acta Paediatr 84: 222–225, 1995. 173. Schaen L, Sheth AP: Skin ulcers associated with a tender and swollen arm. Pyoderma gangrenosum (PG) in association with chronic recurrent multifocal osteomyelitis (CRMO). Arch Dermatol 134: 1146–1150, 1998. 174. Marshall GS, Edwards KM, Wadlington WB: Sporadic congenital Caffey’s disease. Clin Pediatr (Phila) 26: 177–180, 1987. 175. Eyrich GK, Harder C, Sailer HF, et al: Primary chronic osteomyelitis associated with synovitis, acne, pustulosis, hyperostosis and osteitis (SAPHO syndrome). J Oral Pathol Med 28: 456–464, 1999. 176. Beretta-Piccoli B, Sauvain MJ, Gal I, et al: Synovitis, acne, pustulosis, hyperostosis, osteitis (SAPHO) syndrome in childhood: a report of ten cases and review of the literature. Eur J Pediatr 159, 594–601, 2000. 177. Davis DE, Viozzi FJ, Miller OF, et al: The musculoskeletal manifestations of acne fulminans. J Rheumatol 8: 317–320, 1981. 178. Noseworthy JH, Heffernan LP, Ross JB, et al: Acne fulminans with inflammatory myopathy. Ann Neurol 8: 67–69, 1980. 179. Cros D, Gamby T, Serratrice G: Acne rheumatism. Report of a case. J Rheumatol 8: 336–339, 1981. 180. Hughes RA: Arthritis precipitated by isotretinoin treatment for acne vulgaris. J Rheumatol 20: 1241–1242, 1993. 181. Dubourg G, Koeger AC, Huchet B, et al: Acute monarthritis in a patient under isotretinoin. Rev Rhum Engl Ed 63: 228–229, 1996. 182. Erhardt E, Harangi F: Two cases of musculoskeletal syndrome associated with acne. Pediatr Dermatol 14: 456–459, 1997. 183. De Francesco V, Stinco G, Campanella M: Acute arthritis during isotretinoin treatment for acne conglobata. Dermatology 194: 195, 1997. 184. Sapico FL, Montgomerie JZ: Pyogenic vertebral osteomyelitis: report of nine cases and review of the literature. Rev Infect Dis 1: 754–776, 1979. 185. Fischer GW, Popich GA, Sullivan DE, et al: Diskitis: a prospective diagnostic analysis. Pediatrics 62: 543–548, 1978. 186. Hensey OJ, Coad N, Carty HM, et al: Juvenile discitis. Arch Dis Child 58: 983–987, 1983. 187. Alexander CJ: The aetiology of juvenile spondylarthritis (discitis). Clin Radiol 21: 178–187, 1970. 188. Crawford AH, Kucharzyk DW, Ruda R, et al: Diskitis in children. Clin Orthop 266: 70–79, 1991. 189. Spiegel PG, Kengla KW, Isaacson AS, et al: Intervertebral disc-space inflammation in children. J Bone Joint Surg Am 54: 284–296, 1972. 190. Rocco HD, Eyring EJ: Intervertebral disk infections in children. Am J Dis Child 123: 448–451, 1972. 191. Wenger DR, Bobechko WP, Gilday DL: The spectrum of intervertebral discspace infection in children. J Bone Joint Surg Am 60: 100–108, 1978. 192. Heinrich SD, Zembo MM, King AG, et al: Calcific cervical intervertebral disc herniation in children. Spine 16: 228–231, 1991. 193. Song KS, Ogden JA, Ganey T, et al: Contiguous discitis and osteomyelitis in children. J Pediatr Orthop 17: 470–477, 1997.
OSTEOMYELITIS
194. Fernandez M, Carrol CL, Baker CJ: Discitis and vertebral osteomyelitis in children: an 18-year review. Pediatrics 105: 1299–1304, 2000. 195. Whipple GH: A hitherto undescribed disease characterized anatomically by deposits of fat and fatty acids in the intestinal and mesenteric lymphatic tissues (intestinal lipodystrophy). Bull Johns Hopkins Hosp 18: 382, 1907. 196. Tan TQ, Vogel H, Tharp BR, et al: Presumed central nervous system Whipple’s disease in a child: case report. Clin Infect Dis 20: 883–889, 1995. 197. Maizel H, Ruffin JM, Dobbins WO III: Whipple’s disease: a review of 19 patients from one hospital and a review of the literature since 1950. Medicine (Baltimore) 49: 175–205, 1970. 198. LeVine ME, Dobbins WO III: Joint changes in Whipple’s disease. Semin Arthritis Rheum 3: 79–93, 1973. 199. Misbah SA, Ozols B, Franks A, et al: Whipple’s disease without malabsorption: new atypical features. Q J Med 90: 765–772, 1997. 200. Aust CH, Smith EB: Whipple’s disease in a three-month-old infant. Am J Clin Pathol 37: 66, 1962. 201. Barakat AY, Bitar J, Nassar VH: Whipple’s disease in a seven-year-old child. Report of a case. Am J Proctol 24: 312–315, 1973. 202. Caughey OE, Bywaters EGL: The arthritis of Whipple’s syndrome. Ann Rheum Dis 22: 327, 1963. 203. Kelly JJ, Weisiger BB: The arthritis of Whipple’s disease. Arthritis Rheum 6: 615, 1963. 204. Petty RE, Tingle AJ: Arthritis and viral infection. J Pediatr 113: 948–949, 1988. 205. Phillips PE: Viral arthritis. Curr Opin Rheumatol 9: 337–344, 1997. 206. Ytterberg SR: Viral arthritis. Curr Opin Rheumatol 11: 275–280, 1999. 207. Osler W: The Principles and Practice of Medicine, 8th ed. New York, Appleton, 1918, p 348. 208. Tingle AJ, Allen M, Petty RE, et al: Rubella-associated arthritis. I. Comparative study of joint manifestations associated with natural rubella infection and RA 27/3 rubella immunisation. Ann Rheum Dis 45: 110–114, 1986. 209. Best JM, Banatvala JE, Bowen JM: New Japanese rubella vaccine. Comparative trials. Br Med J 3: 221–224, 1974. 210. Polk BF, Modlin JF, White JA, et al: A controlled comparison of joint reactions among women receiving one of two rubella vaccines. Am J Epidemiol 115: 19–25, 1982. 211. Tingle AJ, Chantler JK, Pot KH, et al: Postpartum rubella immunization: association with development of prolonged arthritis, neurological sequelae, and chronic rubella viremia. J Infect Dis 152: 606–612, 1985. 212. Grillner L, Hedstrom CE, Bergstrom H, et al: Vaccination against rubella of newly delivered women. Scand J Infect Dis 5: 237–241, 1973. 213. Tingle AJ, Yang T, Allen M, et al: Prospective immunological assessment of arthritis induced by rubella vaccine. Infect Immun 40: 22–28, 1983. 214. Tingle AJ, Pot KH, Yong FP, et al: Kinetics of isotype-specific humoral immunity in rubella vaccine-associated arthropathy. Clin Immunol Immunopathol 53: S99–S106, 1989. 215. Ogra PL, Herd JK: Arthritis associated with induced rubella infection. J Immunol 107: 810–813, 1971. 216. Chantler JK, Ford DK, Tingle AJ: Persistent rubella infection and rubellaassociated arthritis. Lancet 1: 1323–1325, 1982. 217. Spruance SL, Metcalf R, Smith CB, et al: Chronic arthropathy associated with rubella vaccination. Arthritis Rheum 20: 741–747, 1977. 218. Chantler JK, Tingle AJ, Petty RE: Persistent rubella virus infection associated with chronic arthritis in children. N Engl J Med 313: 1117–1123, 1985. 219. Bosma TJ, Etherington J, O’Shea S, et al: Rubella virus and chronic joint disease: is there an association? J Clin Microbiol 36: 3524–3526, 1998. 220. Naides SJ: Parvovirus B19 infection. Rheum Dis Clin North Am 19: 457–475, 1993. 221. Ware R: Human parvovirus infection. J Pediatr 114: 343–348, 1989. 222. Simpson RW, McGinty L, Simon L, et al: Association of parvoviruses with rheumatoid arthritis of humans. Science 223: 1425–1428, 1984. 223. White DG, Woolf AD, Mortimer PP, et al: Human parvovirus arthropathy. Lancet 1: 419–421, 1985. 224. Anderson MJ, Jones SE, Fisher-Hoch SP, et al: Human parvovirus, the cause of erythema infectiosum (fifth disease)? Lancet 1: 1378, 1983. 225. Reid DM, Reid TM, Brown T, et al: Human parvovirus-associated arthritis: a clinical and laboratory description. Lancet 1: 422–425, 1985. 226. Semble EL, Agudelo CA, Pegram PS: Human parvovirus B19 arthropathy in two adults after contact with childhood erythema infectiosum. Am J Med 83: 560–562, 1987. 227. Smith CA, Woolf AD, Lenci M: Parvoviruses: infections and arthropathies. Rheum Dis Clin North Am 13: 249–263, 1987. 228. Ager EA, Chin TD, Poland JD: Epidemic erythema infectiosum. N Engl J Med 275: 1326–1331, 1966. 229. Klouda PT, Corbin SA, Bradley BA, et al: HLA and acute arthritis following human parvovirus infection. Tissue Antigens 28: 318–319, 1986. 230. Speyer I, Breedveld FC, Dijkmans BA: Human parvovirus B19 infection is not followed by inflammatory joint disease during long term follow-up. A retrospective study of 54 patients. Clin Exp Rheumatol 16: 576–578, 1998. 231. Anderson LJ: Role of parvovirus B19 in human disease. Pediatr Infect Dis J 6: 711–718, 1987. 232. Rivier G, Gerster JC, Terrier P, et al: Parvovirus B19 associated monarthritis in a 5-year-old boy. J Rheumatol 22: 766–767, 1995. 233. Schwarz TF, Roggendorf M, Suschke H, et al: Human parvovirus B19 infection and juvenile chronic polyarthritis. Infection 15: 264–265, 1987.
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
234. Nocton JJ, Miller LC, Tucker LB, et al: Human parvovirus B19-associated arthritis in children. J Pediatr 122: 186–190, 1993. 235. Dijkmans BA, Elsacker-Niele AM, Salimans MM, et al: Human parvovirus B19 DNA in synovial fluid. Arthritis Rheum 31: 279–281, 1988. 236. Foto F, Marsh JL, Scharosch LL: Synovial tissue analysis in patients with chronic parvovirus B19 arthropathy. Arthritis Rheum Suppl 34: S50, 1991. 237. Soderlund M, von Essen R, Haapasaari J, et al: Persistence of parvovirus B19 DNA in synovial membranes of young patients with and without chronic arthropathy. Lancet 349: 1063–1065, 1997. 238. Naides SJ, Scharosch LL, Foto F, et al: Rheumatologic manifestations of human parvovirus B19 infection in adults. Initial two-year clinical experience. Arthritis Rheum 33: 1297–1309, 1990. 239. Erdman DD, Usher MJ, Tsou C, et al: Human parvovirus B19 specific IgG, IgA, and IgM antibodies and DNA in serum specimens from persons with erythema infectiosum. J Med Virol 35: 110–115, 1991. 240. Schwarz TF, Hottentrager B, Roggendorf M: Prevalence of antibodies to parvovirus B19 in selected groups of patients and healthy individuals. Zentralbl Bakteriol 276: 437–442, 1992. 241. Yamashita K, Matsunaga Y, Taylor-Wiedeman J, et al: A significant age shift of the human parvovirus B19 antibody prevalence among young adults in Japan observed in a decade. Jpn J Med Sci Biol 45: 49–58, 1992. 242. Alarcon GS, Townes AS: Arthritis in viral hepatitis. Report of two cases and review of the literature. Johns Hopkins Med J 132: 1–15, 1973. 243. Inman RD: Rheumatic manifestations of hepatitis B virus infection. Semin Arthritis Rheum 11: 406–420, 1982. 244. Alpert E, Isselbacher KJ, Schur PH: The pathogenesis of arthritis associated with viral hepatitis. Complement-component studies. N Engl J Med 285: 185–189, 1971. 245. Duffy J, Lidsky MD, Sharp JT, et al: Polyarthritis, polyarteritis and hepatitis B. Medicine (Baltimore) 55: 19–37, 1976. 246. Schumacher HR, Gall EP: Arthritis in acute hepatitis and chronic active hepatitis. Pathology of the synovial membrane with evidence for the presence of Australia antigen in synovial membranes. Am J Med 57: 655–664, 1974. 247. Fraser JR: Epidemic polyarthritis and Ross River virus disease. Clin Rheum Dis 12: 369–388, 1986. 248. Aaskov JG, Chen JY, Hanh NT, et al: Surveillance for Ross River virus infection using blood donors. Am J Trop Med Hyg 58: 726–730, 1998. 249. Friedman HM, Pincus T, Gibilisco P, et al: Acute monoarticular arthritis caused by herpes simplex virus and cytomegalovirus. Am J Med 69: 241–247, 1980. 250. Bluestein HG, Hasler F: Epstein-Barr virus and rheumatoid arthritis. Surv Immunol Res 3: 70–77, 1984. 251. Lotz M, Roudier J: Epstein-Barr virus and rheumatoid arthritis: cellular and molecular aspects. Rheumatol Int 9: 147–152, 1989. 252. Baboonian C, Halliday D, Venables PJ, et al: Antibodies in rheumatoid arthritis react specifically with the glycine alanine repeat sequence of Epstein-Barr nuclear antigen-1. Rheumatol Int 9: 161–166, 1989. 253. Adebonojo FO: Monarticular arthritis: an unusual manifestation of infectious mononucleosis. Clin Pediatr (Phila) 11: 549–550, 1972. 254. Pollack S, Enat R, Barzilai D: Monarthritis with heterophil-negative infectious mononucleosis. Case of an older patient. Arch Intern Med 140: 1109–1111, 1980. 255. Berger RG, Raab-Traub N: Acute monarthritis from infectious mononucleosis. Am J Med 107: 177–178, 1999. 256. Ward JR, Bishop B: Varicella arthritis. JAMA 212: 1954–1956, 1970. 257. Friedman A, Naveh Y: Polyarthritis associated with chickenpox. Am J Dis Child 122: 179–180, 1971. 258. Mulhern LM, Friday GA, Perri JA: Arthritis complicating varicella infection. Pediatrics 48: 827–829, 1971. 259. Brook I: Varicella arthritis in childhood. Reports of 2 cases and 4 others found in the literature. Clin Pediatr (Phila) 16: 1156–1157, 1977. 260. Fierman AH: Varicella-associated arthritis occurring before the exanthem. Case report and literature review. Clin Pediatr (Phila) 29: 188–190, 1990. 261. Schreck P, Bradley J, Chambers H: Musculoskeletal complications of varicella. J Bone Joint Surg Am 78: 1713–1719, 1996. 262. Stebbings S, Highton J, Croxson MC, et al: Chickenpox monarthritis: demonstration of varicella-zoster virus in joint fluid by polymerase chain reaction. Br J Rheumatol 37: 311–313, 1998. 263. Chen MK, Wang CC, Lu JJ, et al: Varicella arthritis diagnosed by polymerase chain reaction. J Formos Med Assoc 98: 519–521, 1999. 264. Buck RE: Pyarthrosis of the hip complicating chickenpox. JAMA 206: 135–136, 1968. 265. Sethi AS, Schloff I: Purulent arthritis complicating chickenpox. Clin Pediatr (Phila) 13: 280, 1974. 266. Priest JR, Urick JJ, Groth KE, et al: Varicella arthritis documented by isolation of virus from joint fluid. J Pediatr 93: 990–992, 1978. 267. Shore A, Ansell BM: Juvenile psoriatic arthritis—an analysis of 60 cases. J Pediatr 100: 529–535, 1982. 268. Cunningham AL, Fraser JR, Clarris BJ, et al: A study of synovial fluid and cytology in arthritis associated with herpes zoster. Aust N Z J Med 9: 440–443, 1979. 269. Devereaux MD, Hazelton RA: Acute monarticular arthritis in association with herpes zoster. Arthritis Rheum 26: 236–237, 1983. 270. Gordon SC, Lauter CB: Mumps arthritis: a review of the literature. Rev Infect Dis 6: 338–344, 1984. 271. Fontebasso M: Mumps polyarthritis. J R Coll Gen Pract 35: 152, 1985. 272. Moffatt CD: Mumps arthropathy. J R Coll Gen Pract 36: 230, 1986.
AND
OSTEOMYELITIS
589
273. Lipstein-Kresch E, Isenberg HD, Singer C, et al: Disseminated Sporothrix schenckii infection with arthritis in a patient with acquired immunodeficiency syndrome. J Rheumatol 12: 805–808, 1985. 274. Ricciardi DD, Sepkowitz DV, Berkowitz LB, et al: Cryptococcal arthritis in a patient with acquired immune deficiency syndrome. Case report and review of the literature. J Rheumatol 13: 455–458, 1986. 275. Calabrese LH: Human immunodeficiency virus (HIV) infection and arthritis. Rheum Dis Clin North Am 19: 477–488, 1993. 276. Raphael SA, Wolfson BJ, Parker P, et al: Pyomyositis in a child with acquired immunodeficiency syndrome. Patient report and brief review. Am J Dis Child 143: 779–781, 1989. 277. Labrune P, Blanche S, Catherine N, et al: Human immunodeficiency virus-associated thrombocytopenia in infants. Acta Paediatr Scand 78: 811–814, 1989. 278. Calabrese LH, Estes M, Yen-Lieberman B, et al: Systemic vasculitis in association with human immunodeficiency virus infection. Arthritis Rheum 32: 569–576, 1989. 279. Espinoza LR, Aguilar JL, Berman A, et al: Rheumatic manifestations associated with human immunodeficiency virus infection. Arthritis Rheum 32: 1615–1622, 1989. 280. Steinbach LS, Tehranzadeh J, Fleckenstein JL, et al: Human immunodeficiency virus infection: musculoskeletal manifestations. Radiology 186: 833–838, 1993. 281. Berman A, Cahn P, Perez H, et al: Human immunodeficiency virus infection associated arthritis: clinical characteristics. J Rheumatol 26: 1158–1162, 1999. 282. Rahal JJ, Millian SJ, Noriega ER: Coxsackievirus and adenovirus infection. Association with acute febrile and juvenile rheumatoid arthritis. JAMA 235: 2496–2501, 1976. 283. Luder AS, Naphtali V, Ben Porat E, et al: Still’s disease associated with adenovirus infection and defect in adenovirus directed natural killing. Ann Rheum Dis 48: 781–786, 1989. 284. Blotzer JW, Myers AR: Echovirus-associated polyarthritis. Report of a case with synovial fluid and synovial histologic characterization. Arthritis Rheum 21: 978–981, 1978. 285. Kujala G, Newman JH: Isolation of echovirus type 11 from synovial fluid in acute monocytic arthritis. Arthritis Rheum 28: 98–99, 1985. 286. Ackerson BK, Raghunathan R, Keller MA, et al: Echovirus 11 arthritis in a patient with X-linked agammaglobulinemia. Pediatr Infect Dis J 6: 485–488, 1987. 287. Roberts-Thomson PJ, Southwood TR, Moore BW, et al: Adult onset Still’s disease or coxsackie polyarthritis? Aust N Z J Med 16: 509–511, 1986. 288. Holtzman CM: Postvaccination arthritis. N Engl J Med 280: 111–112, 1969. 289. Nishioka K, Nakajima T, Hasunuma T, et al: Rheumatic manifestation of human leukemia virus infection. Rheum Dis Clin North Am 19: 489–503, 1993. 290. Turunen M, Kuusisto P, Uggeldahl PE, et al: Pogosta disease: clinical observations during an outbreak in the province of North Karelia, Finland. Br J Rheumatol 37: 1177–1180, 1998. 291. Wingstrand H: Transient synovitis of the hip in the child. Acta Orthop Scand Suppl 219: 1–61, 1986. 292. Hardinge K: The etiology of transient synovitis of the hip in childhood. J Bone Joint Surg Br 52: 100–107, 1970. 293. Kocher MS, Zurakowski D, Kasser JR: Differentiating between septic arthritis and transient synovitis of the hip in children: an evidence-based clinical prediction algorithm. J Bone Joint Surg Am 81: 1662–1670, 1999. 294. Del Beccaro MA, Champoux AN, Bockers T, et al: Septic arthritis versus transient synovitis of the hip: the value of screening laboratory tests. Ann Emerg Med 21: 1418–1422, 1992. 295. Spock A: Transient synovitis of the hip joint in children. Pediatrics 24: 1042, 1959. 296. Wynne-Davies R, Gormley J: The aetiology of Perthes’ disease. Genetic, epidemiological and growth factors in 310 Edinburgh and Glasgow patients. J Bone Joint Surg Br 60: 6–14, 1978. 297. Cuellar ML, Silveira LH, Citera G, et al: Other fungal arthritides. Rheum Dis Clin North Am 19: 439–455, 1993. 298. Silveira LH, Cuellar ML, Citera G, et al: Candida arthritis. Rheum Dis Clin North Am 19: 427–437, 1993. 299. Crout JE, Brewer NS, Tompkins RB: Sporotrichosis arthritis: clinical features in seven patients. Ann Intern Med 86: 294–297, 1977. 300. Goldenberg DL, Cohen AS: Arthritis due to tuberculous and fungal microorganisms. Clin Rheum Dis 4: 211, 1978. 301. Hart PD, Russell EJ, Remington JS: The compromised host and infection. II. Deep fungal infection. J Infect Dis 120: 169–191, 1969. 302. Tack KJ, Rhame FS, Brown B, et al: Aspergillus osteomyelitis. Report of four cases and review of the literature. Am J Med 73: 295–300, 1982. 303. Bayer AS, Choi C, Tillman DB, et al: Fungal arthritis. V. Cryptococcal and histoplasmal arthritis. Semin Arthritis Rheum 9: 218–227, 1980. 304. Sanders LL: Blastomycosis arthritis. Arthritis Rheum 10: 91–98, 1967. 305. Bayer AS, Yoshikawa TT, Galpin JE, et al: Unusual syndromes of coccidioidomycosis: diagnostic and therapeutic considerations; a report of 10 cases and review of the English literature. Medicine (Baltimore) 55: 131–152, 1976. 306. Castaneda OJ, Alarcon GS, Garcia MT, et al: Paracoccidioides brasiliensis arthritis. Report of a case and review of the literature. J Rheumatol 12: 356–358, 1985. 307. Dinulos JG, Darmstadt GL, Wilson CB, et al: Nocardia asteroides septic arthritis in a healthy child. Pediatr Infect Dis J 18: 308–310, 1999. 308. Ansari RA, Hindson DA, Stevens DL, et al: Pseudallescheria boydii arthritis and osteomyelitis in a patient with Cushing’s disease. South Med J 80: 90–92, 1987.
590
C H A P T E R
28 I N F E C T I O U S A RT H R I T I S
AND
309. Klein JD, Yamauchi T, Horlick SP: Neonatal candidiasis, meningitis, and arthritis: observations and a review of the literature. J Pediatr 81: 31–34, 1972. 310. Poplack DG, Jacobs SA: Candida arthritis treated with amphotericin B. J Pediatr 87: 989–990, 1975. 311. MacGregor RR, Schimmer BM, Steinberg ME: Results of combined amphotericin B–5-fluorcytosine therapy for prosthetic knee joint infected with Candida parapsilosis. J Rheumatol 6: 451–455, 1979. 312. Sugarman M, Stobie DG, Quismorio FP, et al: Plant thorn synovitis. Arthritis Rheum 20: 1125–1128, 1977. 313. Sutliff WD, Shepard R, Dunham WB: Acute Leptospira pomona arthritis and myocarditis. Ann Intern Med 39: 134, 1953. 314. Reginato AJ, Schumacher HR, Jimenez S, et al: Synovitis in secondary syphilis. Clinical, light, and electron microscopic studies. Arthritis Rheum 22: 170–176, 1979. 315. Reginato AJ: Syphilitic arthritis and osteitis. Rheum Dis Clin North Am 19: 379–398, 1993. 316. Argen RJ, Dixon AS: Clutton’s joints with keratitis and periostitis. A case report with histology of synovium. Arthritis Rheum 6: 341, 1963. 317. Bocanegra TS, Vasey FB: Musculoskeletal syndromes in parasitic diseases. Rheum Dis Clin North Am 19: 505–513, 1993. 318. Woo P, Panayi GS: Reactive arthritis due to infestation with Giardia lamblia. J Rheumatol 11: 719, 1984. 319. Burnstein SL, Liakos S: Parasitic rheumatism presenting as rheumatoid arthritis. J Rheumatol 10: 514–515, 1983. 320. Williams D, Roy S: Arthritis and arthralgia associated with toxocaral infestation. Br Med J (Clin Res Ed) 283: 192, 1981. 321. Atkin SL, Kamel M, el-Hady AM, et al: Schistosomiasis and inflammatory polyarthritis: a clinical, radiological and laboratory study of 96 patients infected by S. mansoni with particular reference to the diarthrodial joint. Q J Med 59: 479–487, 1986.
OSTEOMYELITIS
322. Corman LC: Acute arthritis occurring in association with subcutaneous Dirofilaria tenuis infection. Arthritis Rheum 30: 1431–1434, 1987. 323. Shagrin JW, Frame B, Duncan H: Polyarthritis in obese patients with intestinal bypass. Ann Intern Med 75: 377–380, 1971. 324. Stein HB, Schlappner OL, Boyko W, et al: The intestinal bypass: arthritis-dermatitis syndrome. Arthritis Rheum 24: 684–690, 1981. 325. Wands JR, LaMont JT, Mann E, et al: Arthritis associated with intestinalbypass procedure for morbid obesity. Complement activation and characterization of circulating cryoproteins. N Engl J Med 294: 121–124, 1976. 326. Ginsberg J, Quismorio FP Jr, DeWind LT, et al: Musculoskeletal symptoms after jejunoileal shunt surgery for intractable obesity. Clinical and immunologic studies. Am J Med 67: 443–448, 1979. 327. Pinals RS, Tinnessen WW Jr: Shunt arthritis. J Pediatr 91: 681, 1977. 328. Schaad UB: Arthritis in disease due to Neisseria meningitidis. Rev Infect Dis 2: 880–888, 1980. 329. Greenwood BM, Whittle HC, Bryceson AD: Allergic complications of meningococcal disease. II. Immunological investigations. Br Med J 2: 737–740, 1973. 330. Rush PJ, Shore A, Inman R, et al: Arthritis associated with Haemophilus influenzae meningitis: septic or reactive? J Pediatr 109: 412–415, 1986. 331. Churchill MA Jr, Geraci JE, Hunder GG: Musculoskeletal manifestations of bacterial endocarditis. Ann Intern Med 87: 754–759, 1977. 332. Levo Y, Nashif M: Musculoskeletal manifestations of bacterial endocarditis. Clin Exp Rheumatol 1: 49–52, 1983. 333. Williams RC Jr, Kunkel HG: Rheumatoid factor, complement and conglutinin aberrations in patients with subacute bacterial endocarditis. J Clin Invest 41: 666, 1962. 334. Bayer AS, Theofilopoulos AN, Eisenberg R, et al: Circulating immune complexes in infective endocarditis. N Engl J Med 295: 1500-1505, 1976.
29
C HAP T E R
LYME DISEASE
..
Hans-Iko Huppertz and Frank Dressler
~
Lyme arthritis was first described in 1977 by Steere and colleagues 1 in a cluster of children thought to have juvenile rheumatoid arthritis. They lived in and around Old Lyme, Connecticut. Subsequent studies documented that the disease was caused by the spirochete Borrelia burgdorftrP,3 and that arthritis was only one of many possible manifestations of this infection, now known as Lyme borreliosis or Lyme disease.4--8 Clinical case descriptions of various manifestations of this disease date back more than a century. Acrodermatitis chronica atrophicans was described in Germany in 1883.9 Erythema migrans, the early skin manifestation of Lyme borreliosis, was reported in Sweden in 1909. 10 The first case of neuroborreliosis and its association with a tick bite was reported in France in 1922,u Among the cases of lymphocytic meningitis and inflammatory polyneuritis studied by Bannwarth 12 in Germany in 1941, several patients described had "rheumatism," probably the first report of what is now called Lyme arthritis. Successful treatment with penicillin was described in 1946. 13 Erythema migrans was transferred by skin biopsy to healthy human volunteers in 1955. 14 These observations suggested an infectious cause.
DEFINmON AND CLASSIFICATION Lyme disease is a complex disease with cutaneous, articular, neurologic, and other systemic manifestations that results from infection with the spirochete B. burgdorferi transmitted by the bite of a tick of the genus Ixodes. Various components of the disease (e.g., erythema migrans, arthritis, neuroborreliosis) may occur in isolation. The term Lyme borreliosis is often used in Europe; Lyme disease is the most frequent term used in North America.
EPIDEMIOLOGY Geographic Distribution Lyme disease has been documented only in the temperate zones of the northern hemisphere. 4 ,ls In North America, the disease is recognized most commonly in the northeastern, mid-Atlantic and north-central United States; it occurs less commonly on the West Coast and in Ontario, Canada. 16,17 Lyme borreliosis is rare or absent in
the other parts of the United States and Canada. In Europe, the disease is most common in central Europe but occurs endemically from southern Sweden to the northern Mediterranean and from Portugal to Russia. Although sporadic cases of Lyme disease have been reported in eastern Russia, China, Korea, and Japan, it appears to be much less common in Asia than in the endemic areas of North America or Europe.
Inddence and Prevalence The Centers for Disease Control and Prevention have reported a rapid increase in the frequency of Lyme disease in the United States since 1982. Between 1991 and 2000, the incidence nearly doubled, and 23,763 cases were reported in 2002. 17,18 In 2000, the highest incidence (111 cases per 100,000) was in Connecticut. All incidence rates are per 100,000 of the general population, The highest local incidence (1200 cases per 100,000) was on the island of Nantucket, Massachusetts. 16,17 Data from the Slovenian National Registry document annual incidences of Lyme disease of 115 to 168 cases per 100,000 between 1998 and 2002 (F. Strle, personal communication, 2003). A study in southern Sweden reported an incidence of 69 cases per 100,000; Lyme arthritis was present in 7% of all cases of Lyme disease. 19 In a population-based study in Wl1rzburg, Germany, the incidence was 111 per 100,000, with higher rates among children younger than 16 years. 20 In a community-based Connecticut cohort study of 201 consecutive cases in children in whom Lyme disease had been newly diagnosed, 13 (6%) had arthritis, and 5% had facial palsy.21 In Europe, Lyme arthritis and neuroborreliosis have been reported in similar frequencies, and in the Wl1rzburg study, arthritis was more common. 20 Compared with adults, children more frequently had manifestations other than isolated erythema migrans. 2o Early onset of cutaneous disease and neural involvement are closely related to tick activity in the spring to autumn months; there is no seasonal pattern for late manifestations such as Lyme arthritis. 4•s.20 ,22
Sex Ratio and Age at Onset Both sexes are affected equally. Cases have been reported among all age groups, with peaks occurring in school-age children and people between 40 and 74 years of age. 19,20
591
592
CHAPTER
29
LYME DISEASE
GENETIC BACKGROUND Although Lyme disease may affect several members of the same family, genetic factors appear to have a limited influence on its occurrence. Nonetheless, host factors influence the course of the disease. In American patients, the development of chronic Lyme arthritis and antibiotic unresponsiveness has been associated with the presence of human leukocyte antigen (HLA)-DR4. HLA-DR2 is an additional risk factor, especially in patients who are HLADR4 negative. 23 These results have not been confirmed in European patients.
ETIOLOGY AND PATHOGENESIS Etiology Lyme disease is the most common vector-borne infection in North America and Europe and is transmitted by hardbodied ticks of the genus Ixodes 4,24 (Fig. 29-1). Transmission by other ticks or flying hematophagous insects has been suggested but not proved. Ticks of the genus Ixodes include Ixodes ricinus in central Europe, Ixodes persulcatus in eastern Europe and Asia, Ixodes scapularis in the northeastern and north-central United States and Ontario, and Ixodes pacificus in the western United States. 4 Ticks require a warm and humid environment and are affected by climatic variability.25 Infection is acquired in tick habitats, including forests, shaded valleys, gardens, lawns, and inner-city parks. After gaining access to unprotected skin, ticks crawl to the preferred feeding locations on the popliteal region, thigh, groin, breast, axilla, neck, or head. Ixodes ticks feed only once during each of the three stages of their life cycle. Most human infections occur after the painless bite of nymphs
(Fig. 29-2). Ixodes ticks also transmit tick-borne encephalitiS virus, Ehrlichia, Anaplasma phagoeytophila, and Babesia organisms. Coinfection of these organisms with B. burgdorferi has been reported in rare cases.26--2H
Mluoblology Lyme disease is caused by infection with one of several species of B. burgdorferi sensu lato. These spirochetes have a protoplasmic cylinder surrounded by a cell membrane, a periplasmic flagellum, and an outer membrane. 29 They are microaerophilic and grow best at 33° C in a special liquid medium. They grow slowly, with doubling times between 12 and 24 hours. B. burgdorferi sensu lato has been subdivided into several species, of which only B. burgdorferi sensu stricto has been found to cause human disease in North America; Borrelia garinii and Borrelia afzelii also have been identified regularly in patients in Europe. 4.5 In general, diversity in B. burgdorferi organisms has been greater in Europe and Asia than in North America. Concurrent infection with more than one species of B. burgdorferi was described in a patient with acrodermatitis and erythema migrans,30 as was cultureconfirmed reinfection in patients with several episodes of erythema migrans. 31 B. burgdorferi species differ genomically. Even within a species, different strains express proteins of different molecular weights as identified on gel electrophoresis. The major proteins identified in sonicates of B. burgdorJeri are the 41-kD flagellar antigen; the 60-kD GroEL heat-shock protein; the three major outer surface proteins (Osp) OspA (30 to 32 kD), OspB (34 to 36 kD), and OspC (21 to 25 kD); the 39-kD BmpA protein; and the 83- to 100-kD antigen. Other proteins remain unidentified. The linear chromosome and 11 plasmids of B. burgdorferi sensu stricto strain B31 have been sequenced. 32 The natural reservoirs of B. burgdorferi are mice and voles, although hedgehogs and birds may also serve this function. The life cycle of Ixodes ticks lasts 2 years (see Fig. 29-2),33 The eggs hatch and larvae develop in the spring of the first year. The larvae feed once that summer on their preferred host (Le., mice and voles) and so become infected with B. burgdorferi. The next spring, the larvae molt into nymphs, which feed on the preferred host before becoming mature ticks, at which time larger animals (e.g., deer) act as hosts. Mating occurs while the female tick feeds. The female then detaches and lays her eggs on the ground.
Pathogenesis
• Rgure 21-1 Ixodes scapularis, a member of the Ixodes ricinus complex. Clockwise, beginning upper left: nymph, larva, and adult female.
Lyme arthritis provides a fascinating model for other arthritides because the causative organism and the clinical picture are well known. However, knowledge of the pathogenesis of this disease remains fragmented. B. burgdorferi excreted through tick salivary glands spread locally in the skin and can frequently be found at the advancing edge of erythema migrans. They attach to human cells by binding to various integrins, such as the fibronectin and vitronectin receptors. 34 Binding of the organism to platelets may playa role in its hematogenous
C HAP T E R
Maintenance Hosts
29
LYME DISEASE
593
Incidental Hosts
\ \
\
\
\ \
\
\ \
~ c.~
Established Colony
Dispersal: new colonies may be formed or established colonies may be supplemented
• ,.... ZI-Z Life cyde of Ixodes sCilpularis. I, Engorged adult female; 2, adult female laying eggs; 3, questing and engorged larva; 4, questing and engorged nymph; 5, questing adult. Aquesting tick successfully finds a host (solid alTOws).The tick engorges and drops from the host (dashed alTOws).The tick develops to the next stage (curved alTOws). (From Anderson JF, Magnarelli LA: Avian and mammalian hosts for spirochete-infected ticks and insects in a Lyme disease focus in Connecticut. In Steere AC, Malawista AC, Craft JE, et al [eds): Rrst International Symposium on Lyme Disease, New Haven, G.Vale JBioi Med 57:638, 1984.)
spread. 3S B. burgdorfiri organisms are presumed to reach the synovium through the bloodstream. It is probable that their presence in synovium is required at the onset of arthritis. Survival of B. burgdorfiri for decades in the lesions of acrodermatitis chronica atrophicans indicates that the spirochetes are able to evade the host immune response. There is also evidence that B. burgdorfiri may survive intracellularly in endothelial cells, fibroblasts, and synovial cells. 36-38 B. burgdorfiri has stimulatory effects on B cells,39 and a dominant Thl helper T cell response has been found in synovial fluid of patients with Lyme arthritis. 40 ,4J A B. burgdorfiri-specific CD8+ cytotoxic T cell response has also been reported for patients with Lyme arthritis. 42 These cells were found only after the disappearance of arthritis. 42 B. burgdorfiri also stimulate synovial y/o T cells from patients with Lyme arthritis, leading to high and prolonged expression of Fas ligand associated with cytolytic activity.43.44 A number of cytokines are induced, including interleukin (IL)-l and IL_6,45.46 tumor necrosis factor,47 and interferon-y and IL-1O. 48 Molecular mimicry may also play a role in the pathogenesis of and some of the manifestations of Lyme disease. Sequence homologies have been identified between B. burgdorfiri flagellin and human myelin basic protein, as well as cross-reactivity between flagellin and
a human axonal protein. 49,So In American patients, antibody reactivity to OspA and OspB occurred late in the course of infection in patients with chronic Lyme arthritis. 5J Helper T cells from patients with treatment-resistant Lyme arthritis demonstrated dominant recognition of an OspA peptide of B. burgdorfiri. 52 The human leukocyte function-associated antigen-l (LFA-1) was implicated as a candidate autoantigen in treatment-resistant Lyme arthritiS. 53 However, later work from the same group cast doubt that LFA-l is a relevant autoantigen. 54
CLINICAL MANIFESTATIONS Many persons infected with B. burgdorfiri are asymptomatic. Very often, a tick bite is not recalled. In symptomatic patients, the cutaneous, nervous, and musculoskeletal systems are most frequently involved.4-7 Symptoms of Lyme disease can be divided into early and late manifestations (Table 29-1). Early signs of infection become evident within weeks or a few months of the tick bite, whereas late organ involvement begins several months or even years later. Early symptoms are usually self-limiting, whereas late manifestations may become chronic and occasionally lead to irreversible damage of involved organs. Most patients present with disease that affects only one organ system.
594
I!
CHAPTER
29
LYME DISEASE
III TABLE 29 I
Mdjor Clinilal ManifeslaIions of Lyme Disease in Children dnd Adoleslenls
Organ System
Early l,yme Disease
Late l,yme Disease
Skin
Erythema migrans Borrelial lymphocytoma"
Nervous system
Cranial nerve palsy Lymphocytic meningitis Arthralgia
Acrodermatitis chronica atrophicans" Chronic encephalomyelitis" Arthritis
Musculoskeletal system Other
Carditis"
"Rare in childhood.
Cutaneous Disease The earliest and most common skin manifestation, erythema migrans, typically occurs days to several weeks after infection as an enlarging, warm, but usually painless erythematous rash at the site of the bite, and it lasts for days or weeks (Fig, 29_3).4-7,55 In its classic form, this lesion begins as a red macule or papule and expands peripherally with partial central clearing occurring later. Sometimes, this clearing does not appear; in other cases, the clearing is so complete that erythema migrans becomes a mere curved red streak. In children, the neck and head are the most frequently affected sites, It is also common in the groin, axilla, or thigh and may become quite large (up to 30 cm). Secondary lesions can occur at
sites distant from the tick bite. Erythema migrans may be accompanied by flulike symptoms of fever, chills, arthralgia, musculoskeletal pain, headaches, malaise, and fatigue. Lyme disease may also begin as a flulike illness in the absence of erythema migrans. 56 Weeks to months after infection, borrelial lymphocytoma, also known as lymphadenosis cutis benigna CLe" a purple swelling most commonly at an earlobe, the scrotum, or a nipple), is occasionally reported in European patientsY Acrodermatitis chronica atrophicans, a late skin manifestation, rarely affects European children and then only years after infection. 55'sH In the early phase of acrodermatitis, the affected limb develops inflammatory changes with a red or bluish discoloration. Later, cutaneous atrophy becomes apparent. A peripheral neuropathy can accompany the skin lesion. Lymphocytoma and acrodermatitis are extremely uncommon in North America.
Nervous System Disease: Neuroborrellosls Early neuroborreliosis most frequently presents as lymphocytic meningitis or a cranial nerve palsy weeks to months after infection. 59,6o It may be accompanied by fever, headache, nausea, vomiting, radicular paresthesias, or pain. Unilateral or bilateral facial nerve palsy is the most common focal neurologic manifestation, but cranial nerves III, IV, VI, or VIII may also be affected. Signs of meningitis may be mild or absent in spite of increased protein and lymphocytes in the cerebrospinal fluid. A painful meningoradiculoneuritis is the most common neurologic manifestation in European adults but is relatively uncommon in children. Months to years after infection, a small number of patients develop progressive encephalomyelitis or an encephalopathy.60.6! Other rare neurologic manifestations include the Guillain-Barre syndrome,62.63 pseudotumor cerebri,M optic neuritis,65 cerebral vasculitis,66,67 and neurogenic bladder. 6H
Musculoskeletal Disease
• Rgure Z9-3 Erythema migrans.The site of the tick bite is visible near the center of the lesion (top).Typical "bull's eye" lesion (bottom). (See color insert.)
After erythema rnigrans, arthritis is the most common manifestation of Lyme borreliosis in many series of pediatric patients and is perhaps slightly more common in North America than in Europe.4-B,22,69-7! There is little evidence that the musculoskeletal symptoms of Lyme borreliosis otherwise differ between Europe and North America. Myalgia and myositis occur less often, and enthesitis is not a common feature, although B. burgdorferi have been identified in a few patients with enthesitis or nodular fasciitis. 72- 74 Arthralgia and myalgia develop as early as days to weeks after infection, sometimes concurrent with erythema migrans or flulike symptoms. However, arthritis appears typically months to years after infection. 75 The two largest series of pediatric patients with Lyme arthritis include 90 children from Connecticut7 1 and 109 from Germany,75 62 of whom have been described in detaiJ.22 Monarthritis of a knee occurred in approximately two thirds of all children. n ,7! Both knees or other large joints may also be affected. 22 ,71.75 Polyarticular involvement of small joints was rare. At onset, the arthritis was usually episodic, with relatively painless swelling lasting only a few days and disappearing without ther-
C HAP T E R
apy. Recurrent episodes of arthritis may become prolonged, and chronic arthritis (duration of more than 3 months) has been reported in up to 18% of patients. 22 Among 109 German children with Lyme arthritis, 70 had monarthritis, 32 oligoarthritis, and 7 polyarthritis. 7s The pattern of oligoarticular involvement differed from that found in patients with early-onset oligoarticular juvenile idiopathic arthritis or juvenile rheumatoid arthritis. Ocular involvement with keratitis and anterior and intermediate uveitis may occur in children with Lyme arthritis. 76 Lyme arthritis was reported in a woman after autologous chondrocyte transplantation. n The investigators hypothesize that B. burgdorferi was asymptomatically present in the patient's joint before the chondrocyte transplant procedure. Myositis has only rarely been described in adult patients,n and only myalgia has been reported in children. 78 A dermatomyositis-like picture has also occurred in an adult patient. 79 B. burgdorferi was isolated from a child with subacute multifocal osteomyelitis. so Fibromyalgia may follow Lyme borreliosis and does not respond to antibiotic therapy.S]
Other Manifestations Involvement of other organ systems is much more uncommon. Carditis is rare in children and most commonly manifests as a reversible atrioventricular block,82 Ocular involvement, including conjunctivitis, keratitis, iridocyclitis, intermediate uveitis, choroiditis, or optic neuritis, has been described in children with Lyme arthritis. 76,83,84 Even more uncommonly, patients may develop hepatitis. 8s There have been anecdotal reports of transplacental transmission of B. burgdorferi,86 but this has not been confirmed in controlled studies. 87 The offspring of 5 of 19 pregnant women who had Lyme disease during pregnancy had one or more of the follOWing abnormal outcomes: prematurity, syndactyly, rash, cortical blindness, developmental delay, or intrauterine fetal death. 88,89 Whether any of these complications is attributable to infection with B. burgdorferi or with other spirochetes (more probably with 'Treponema pallidum) is not certain. There is no evidence that maternal infection presents a significant risk to the fetus.
PATHOLOGY The synovitis of Lyme arthritis resembles that of juvenile idiopathic arthritis or juvenile rheumatoid arthritis, with villous hypertrophy, synovial cell hyperplasia, and infiltration of lymphocytes and plasma cells. 9o Lymphoid follicles may also be present. Endarteritis is a characteristic finding in patients with Lyme synovitis. In one study, spirochetes were detected in 2 of 17 synovia, mainly in a perivascular distribution. 90 Other studies using special silver stains have also identified B. burgdorferi in synovium or synovial fluidYl.9Z The organism has been recovered from the margins of the erythema migrans lesion and cardiac tissue. 93--9S Although cardiomyopathy may result from the initial myocarditis, valvular endocarditis does not develop. Myositis may in part account for the myalgia and fatigue
29
LYME DISEASE
595
that occur in this disease, and DNA from B. burgdorferi has been identified in the muscle of such patients. 96
LABORATORY EXAMINATION Nonspedflc Abnormalities The erythrocyte sedimentation rate is elevated in one half of the patients, especially during the early phase of Lyme arthritis. Meningoencephalitis results in a mild cerebrospinal fluid lymphocytic pleoeytosis. 97 The mean synovial fluid white blood cell count ranges from less than 1000/mm3 to greater than 50,000/mm3, with a predominance of neutrophils in samples with high cell counts. 22
Confirmation of Infedion with Borrelia bUIgdorfell Laboratory methods to document infection with B. burgdorferi include direct tests, such as culture or the polymerase chain reaction (PCR) to detect borrelial sequences, and indirect tests, such as serology (Table 29-2).98 The latter tests are most frequently used and universally available. Although attempts have been made to standardize the laboratory evaluation of North American patients with Lyme disease, the approach suggested by the American College of Physicians99 ,lOo has not been widely adopted for European patients. Some of the common problems with standardization of test procedures for the diagnosis of Lyme disease have been reviewed. 101
Direct Methods to Detect Infection Culture of B. burgdorferi usually takes 2 weeks to a few months, requires immediate suspension of the test material in special medium, and has high rates of recovery only from skin biopsies of patients with dermatologic manifestations of the disease. 93 Culture of the organism
I!;~
TABLE 29 2
Laboratory Diagnosis of Lyme Arthritis
Method
Assessment
Culture of Borrelia burgdorferi Histochemistry using silver stain or monoclonal antibodies Polymerase chain reaction for borrelial DNA
Requires weeks; rarely successful Rarely successful in synovial tissue
Enzyme immunoassay or immunofluorescence assay Immunoblot Lymphocyte-proliferation assay with borrelial antigens
Efficiency varies Widely: Urine, 5-85% Synovium, 6-90% (higher in membrane than in fluid) High sensitivity, low specificity Confirmatory test with high specificity Sensitivity and specificity < 80% Limited availability
All diagnostic tests bear the risk of false-negative or false-positive results, No test is of value in a patient with low pretest probability of having Lyme arthritis, Adapted from Huppertz HI: Lyme arthritis. In Wahn U, Seger R, Wahn V (eds): Paediatrische A11ergologie and Immunologie. Munich, Urban & Fischer, 1999, pp 598-602.
596
C HAP T E R
29
LYME DISEASE
from blood and synovial fluid has been relatively unsuccessful. The possibility of obtaining positive cultures is somewhat better from the cerebrospinal fluid of patients with early neuroborreliosis. 10 2 Methods such as silver staining of spirochetes in tissue specimens or staining with monoclonal antibodies are not routinely performed and are prone to artifacts. The PCR can demonstrate DNA of B. burgdorferi in tissues or body fluids, including synovial fluid. 103-109 In a large North American study of synovial fluid from patients with Lyme arthritis,1 03 PCR results were positive for 96% of patients not previously treated with antibiotics and 37% of those who had been treated. In a later study from the same group, borrelial sequences were undetectable in synovial specimens from patients with chronic Lyme arthritis after appropriate antibiotic therapy.104 The precise role of PCR in routine diagnosis is unclear. False-positive or false-negative results may occur. Optimization of PCR includes using more than one primer pair, targeting genes situated on the bacterial chromosome and the plasmids, performing nested PCR, and analYZing synovial fluid and urine. !Os There is evidence that PCR with the use of synovial tissue may have a higher positivity rate than with synovial fluid 108 and may remain positive in patients with ongoing arthritis whose synovial fluid is negative by PCR after antibiotic treatment. 109 PCR results for urine may be positive for healthy humans whose sera contain B. burgdorferi-specific antibodies. llo
Indirect Methods to Detect Infection Specific antibodies can be demonstrated after B. burgdorferi infection by a variety of tests including enzymeimmunoassay (EIA), immunofluorescence, hemagglutina-
tion, and Western blottinglll (Fig. 29-4), It is recommended that a sensitive EIA be used as a screening test and that all results in the indeterminate or positive ranges be confirmed by Western blotting. ll2 Typical IgM and IgG responses of patients with Lyme arthritis and the North American criteria for a positive IgG blot are shown in Figure 29-4. 112•113 In early Lyme borreliosis, IgM blots are considered positive if at least two of the following three bands are present: 2!-kD OspC, 39-kD BmpA, and 4!-kD flagellin. 112.114 Antigens from different strains of B. burgdorferi have different molecular weights, and for this reason, North American criteria cannot easily be applied in Europe or Asia, where there is greater strain diversity. Even so, a two-test approach is the best available method for diagnosis of Lyme disease in European children. 22 In a German pediatric Lyme arthritis study,22 at least six specific bands were required for a positive IgG Western blot, similar to the American criteria. Commonly, patients with Lyme disease have 10 or more IgG bands. (see Fig. 29-4), including the ones mentioned earlier. Specific blot-positivity criteria have been established for each of the three pathogenic species in Europe, with different positivity criteria for each strain. 115 .116 A North American study found that kinetic EIAs detecting IgG responses to recombinant B. burgdorferi antigen visE! or to a conserved internal sequence of visE! were equally sensitive and specific in assessing patients with Lyme arthritis and more sensitive in assessing patients with early skin or neurologic manifestations of Lyme borreliosis. ll7 European studies have also shown the usefulness of EIAs or Western blots using recombinant vIsE! peptide antigen in children with Lyme arthritis l18 or adults with neuroborreliosis.11 9 Within the first weeks after infection, all serologic results may be negative because specific IgM
Western blots of immunoglobulin M(lgM) and IgG antibody responses of 25 patients with Lyme arthritis to a sonicated whole-cell lysate of Borrelia burgdorferi sensu stricto strain G39/40. Molecular masses (in kilodaltons) are indicated on the left side. Characteristic of Lyme arthritis, strong IgG responses against many antigens are demonstrated. In North America, the criteria for a positive IgG blot are the detection of at least five of the following 10 bands: 18 kD, 21-kD Osp C, 28 kD, 30 kD,39kD BmpA, 41-kD flagellin, 45 kD, 58 kD, 66 kD, and 93 kD. All patients met these criteria. Aminority of patients also have IgM responses to asmaller number of antigens. Molecular masses depend on the strain used for testing. (Adapted from Dressler F, Whalen JA, Reinhardt BN, et al: Western blotting in the serodiagnosis of Lyme disease. JInfect Dis 167; 392, 1993.) • figure Z9-4
C HAP T E R
r II -
IABLE 29- 3 AssesslIlent 01 Patients with Suspected I )lme Arthritis
Patient living in or having visited an endemic area Presence of arthritis documented No other obvious cause of arthritis Serology positive (enzyme immunoassay [EIA] and Western blot): No further laboratory test needed Stalt therapy Serology negative (EIA and Western blot): Rule out other diagnosis Refer for specialized evaluation
antibodies usually do not appear until 3 to 4 weeks after infection and IgG antibodies cannot be detected until 4 to 8 weeks after infection. Not infrequently, EIA may give false-positive results related to cross-reactive antibodies such as rheumatoid factors or after infection with Epstein-Barr virus l20 or other spirochetes, including Treponema pal/idum, Treponema denticola, Borrelia hermsii, and leptospirosis.1 21 Antibodies to B. burgdorferi occasionally are found in children with juvenile rheumatoid arthritiS,122,ll3 systemic lupus erythematosus,124 and other illnesses (e.g., bacterial endocarditis, mumps, Rocky Mountain spotted fever, other rickettsial diseases). Serologic tests cannot distinguish patients with active infection from those with a previous infection who have responded to therapy. In particular, about 10% of patients with late manifestations of Lyme disease continue to demonstrate an IgM response in addition to the IgG response. Because IgG titers tend to remain elevated for years,125 serology cannot be used to monitor treatment success or failure. Contrary to claims that the rate of decline of antibodies to the conserved internal sequence of vlsE1 could be useful to monitor treatment efficacy, this was not true in German children with Lyme arthritis. liB In an endemic area, serologic tests should be performed only in patients with clinical signs suggestive of the disease because a positive result in a patient with a low pretest probability of Lyme disease is much more likely to represent a false-positive rather than a true-positive result. 101 ,126 Given these restrictions, serology is useful diagnostically in patients with suspected Lyme arthritis because almost all patients are clearly IgG seropositive!2,71 Table 29-3 provides a simplified overview of the laboratory evaluation. Seronegative Lyme arthritis has been describedl27-129 but must be considered so uncommon that vigorous attempts must be undertaken by centers with special experience with this disease to rule out another diagnosis. In these cases, the cellular immune response induced by B. burgdorferi can be evaluated,127-130 and this is the route that is recommended when clinical symptoms and serologic results are discordant. 129 However, lymphoproliferative assays are not well standardized and have high rates of false positivity and false negativity.127-129
DIAGNOSIS Clinical characteristics of patients with arthritis that suggest a diagnosis of Lyme arthritis include residence in or travel to an endemic area, a preceding tick bite, episodic
29
LYME DISEASE
597
oligoarthritis involving the knee joint, the absence of arthralgias preceding the onset of arthritis, and an adolescent age at onset. Specific criteria have been combined to form a clinical diagnostic score that would confirm or exclude Lyme arthritis in two thirds of children with arthritis l31 (Table 29-4), For a diagnosis of Lyme arthritis, arthritis (Le., swelling and effusion or painful limitation of motion in the absence of trauma) must be observed by a physician. Arthralgias alone or reports by the patient or the parents that the joint was swollen are not objective signs of arthritis in this context. Not infrequently, however, the clinical presentation of Lyme arthritis may be indistinguishable from that of other rheumatic diseases of childhood with arthritis as a principal manifestation, making laboratory tests mandatory in all patients with arthritis living in or having traveled to an endemic area. lol The Centers for Disease Control and Prevention established the follOWing criteria for the diagnosis of Lyme disease l32 : • The presence of erythema migrans larger than 5 cm in diameter, or at least one clinical sign (i.e., arthritis, meningitis, radiculoneuritis, mononeuritis, or carditis) • The presence of specific antibodies to B. burgdorferi These criteria were developed for epidemiologic research and may not always be applicable in the clinical setting. For example, borrelial lymphocytoma may initially occur in the absence of specific antibodies. Moreover, the mere combination of an objective sign
~I
TABLE 29-4 Score
Diagnosis 01 lyme Arthritis Using a Clinical
Criterion Episodic arthritis Arthralgia before onset of arthritis Age at onset of arthritis Initial arthritis in knee joint History of tick bite Number of joints involved
Score· +4
-3 +0.3 x age (yr) +2 +2 -0.4 x number of large joints affected
Scoring example: A 10-year-old boy without
prior arthralgia or history of a tick bite developed arthritis in a knee. The arthritis resolved after 10 days but recurred after a 3-month interval.! Episodic arthritis present No initial arthralgias Age at onset x 0.3 Initial arthritis in knee No history of tick bite 1 large joint affected Total score
+4 +0 +3 +2 +0
-0.4 +8.6
'If a crilerion is recognized. ilS indicated value is added to or subtracted from the total score for the patient. If it is not identified. the item is scored as O. Values of 6 or greater indicate the presence of Lyme arthritis. and values of 2.5 or less exclude the diagnosis. 'The patient's serum was later shown to contain IgG antibodies to Borrelia burgdorferi by enzyme immunoassay and immunoblot. He was treated with ceftriaxone for 2 weeks, Arthritis disappeared dUring therapy and did not recur in the subsequent 2 years of follow-up. Adapted from Huppertz HI, Bentas W, Haubitz I. et al: Diagnosis of pediatric Lyme arthritiS using a clinical score. Eur J Pediatr 157: 304, 1998,
598
C HAP T E R
29
LYME DISEASE
with specific antibodies may include chance associations between two not-infrequent events: arthritis of some kind may affect 1 in 1000 children, and in endemic areas, 3% or more of healthy blood donors may be positive for specific antibodies to B. burgdorferi. 133 Serology can provide evidence of current or prior infection but cannot absolutely confirm a pathogenic link between the infection and the clinical manifestation. The child may have been infected with B. burgdorferi as documented by serology; however, arthritis may result from other known or unknown causes. Overdiagnosis of Lyme disease in children has also been reported. 134 Synovial fluid analysis is of little help in establishing a diagnosis of Lyme arthritis because the white blood cell count and type of cells vary greatly.107.135 However, synovial fluid analysis can exclude septic arthritis and other infection-associated arthritides, yields material for testing by PCR, and confirms the presence of inflammation. Although a positive PCR result from an experienced laboratory indicates a persistent infection, a negative result does not exclude the diagnosis. Synovial tissue may be more suitable than synovial fluid for PCR testing, especially after antibiotic treatment has failed to produce a remission of symptoms. 108.109 A lumbar puncture should be performed in patients with suspected neuroborreliosis, including those with facial palsy. Lymphocytic pleocytosis and elevated cerebrospinal fluid protein levels are characteristic. 59.60 The reliable standard in diagnosing neuroborreliosis in European patients remains detection of intrathecal antibody production.59.60.136 However, specific antibody production frequently occurs only after several weeks to months of infection and has been less commonly demonstrable in American patients.
TREATMENT Antibiotic Regimens Recommendations for the treatment of Lyme disease (Table 29-5) vary according to disease manifestations. In the treatment of patients with erythema migrans or neuroborreliosis, amoxicillin plus probenecid is as effective as doxycycline. 137 Cephalosporins were only marginally
P'Il
TABLE 29-5
better than penicillin G.J3B-142 Cefuroxime axetil was equally efficacious as doxycycline in adults or as amoxicillin in children with erythema migrans.143.144 Whereas doxycycline and ceftriaxone have been equally effective,144 macrolide antibiotics are inferior to other antibioticS. 145-149 It is not known whether these results are applicable to patients with Lyme arthritis, for whom a variety of antibiotics have been recommended, including parenteral penicillin G, oral penicillins, amoxicillin with or without probenecid, ceftriaxone, cefotaxime, cefuroxime, erythromycin, roxithromycin (plus cotrimoxazole), aZithromycin, tetracycline, doxycycline, and others.22. 150 When initial treatment with ceftriaxone fails, 4 weeks of therapy with amoxicillin, with roxithromycin plus cotrimoxazole in young children, or with doxycycline in adolescents is recommended. Before the physician assumes failure of antibiotic therapy, at least two courses of sufficient duration and well-documented compliance are required. When confronted with failure of a treatment program with appropriate antibiotics, the correctness of a diagnosis of Lyme arthritis should be questioned and reconfirmed. Among 51 German patients followed for at least 12 months after initiation of antibiotic treatment, 8 patients still had arthritis, and 4 had arthralgias. 15I Risk factors for a prolonged course of disease were female gender, age older than 10 years, and intra-articular steroids before antibiotic treatment. Duration of therapy is a matter of debate. Because B. burgdorferi is a slow-growing organism, treatment should be continued for at least 10 days. In adults with erythema migrans, extension of treatment with doxycycline from 10 days to 20 days provided no additional benefit. 152 In a retrospective study of Swedish children with early neuroborreliosis, antibiotic treatment for 10 days was considered sufficient. 153 There is also no proof that treatment extending beyond 1 month is of any additional benefit. The success of antibiotic treatment must be determined clinically because serologic results remain positive for a long time after resolution of all manifestations. 125 An approach to treatment is shown in Figure 29-5. In young children, erythema migrans is treated with amoxicillin (50 mg/kg/day) in three doses for 10 days to 3 weeks. In children 9 years or older, doxycycline (200 mg/day) is given once daily for 10 days to 3 weeks. In all
Treatment Recommendations for Lyme Disease in Children and Adolescents
Manifestation
Drugs·
Dose·
Duration
Erythema migrans
Amoxicillin Doxycycline! Ceftriaxone Cefotaxime Amoxicillin Doxycycline! Roxithromycin
50 mg/kglday in 3--4 doses 200 mglday in 1-2 doses 50-100 mglkglday in 1 dose 150 mglkglday in 3 doses 50 mg/kg/day in 3-4 doses 200 mg/day in 1-2 doses 5 mg/kglday plus cotrimoxazole, 6 mglkglday in 2 doses
10-30 days! 10-30 days! 2-4 wk 2-4 wk 4 wk 4 wk
Early disseminated and late disease
4 wk
'Ceftriaxone and cefotaxime are administered intravenously; amoxicilJin and doxycycline are taken orally. Maximum daily dose of amoxicillin - 2 g; doxycycline = 200 mg; ceftriaxone = 2 g; cefotaxime - 6 g. !Doxycycline should not be administered to patients younger than 10 years of age. !Continue treatment for another 10 days if erythema migrans is still present at the end of 10 days.
C HAP T E R
First antibiotic treatment:
ceftriaxone 50 mglkg (maximum 2
I
if
a"'''''. pa";." aile, 6 - " "
Second antibiotic treatment:
<10 years: roxithromycine (5 mg/kg) + cotrimoxazol (6 mg/kg) >10 years: doxycycline 200 mg/day each for 4 weeks
!Intraartlcular steroids (triamcinolone hexacetonide) ,
j
29
LYME DISEASE
599
response to the antibiotic requires immediate interruption of administration of the drug, a Jarisch-Herxheimer reaction is a favorable, self-limited sign, and treatment can be continued. Although nonsteroidal anti-inflammatory agents are frequently given to patients with Lyme arthritis, often before the correct diagnosis is made, their efficacy has not been established; however, nonsteroidal anti-inflammatory agents can be used as analgesics or used after antibiotics have failed. Treatment failures in patients with Lyme arthritis or late neuroborreliosis are often associated with prior administration of glucocorticoids. 138 In such instances, repetition of antibiotic treatment with the same or another antibiotic is recommended. Intra-articular steroids, sulfasalazine, methotrexate, or arthroscopic synovectomy l56 with a further course of antibiotics (in that order) are treatment options.
if arthriti. ".,.Ots
Prevention
IRepeat Intraarticular steroids I if arthritis persists
Sulfasalazlne, methotrexate,
or synovectomy (including repeat ceftriaxone) Sequential treatment of Lyme arthritis (Adapted from Huppertz HI: Lyme arthritis./nWahn U, Seger R, WahnV ledsl: Paediatrische A1lergologie und Immunolgie, 3rd ed. Urban & Rscher Miinchen, 1999, pp 598-602.) • figure 29-5
other forms of the disease, including Lyme arthritis, ceftriaxone (50 mg/kg!day) is administered intravenously for 14 days. Because only one 20-minute infusion is required per day, treatment can be provided in an outpatient setting using an indwelling venous access line. Patients with Lyme arthritis have also been treated successfully with oral antibiotics, including amoxicillin or doxycycline, for 4 weeks. This approach to treatment is more convenient for the patient and more cost-effective than intravenous regimens, but it should not be used in patients with a Baker cyst, neuroborreliosis, or carditis. 154 In case of allergy to penicillin, amoxicillin, or cephalosporins, macrolide antibiotics are recommended in children younger than 9 years, although these drugs are less effective than the ~-lactam antibiotics. 146,149 This disadvantage may be overcome by a combination of roxithromycin and cotrimoxazol. 155 Infection during pregnancy should be treated with antibiotics not posing a risk to the fetus (i.e., amoxicillin, intravenous penicillin G, or cephalosporins). During antibiotic treatment, up to 10% of patients with arthritis develop a Jarisch-Herxheimer reaction, with fever, a nonpruritic, nonpalpable rash, and severe pain. This complication usually develops after the first few doses of antibiotics but may occur up to 10 days after beginning treatment. It must be distinguished from allergic reactions to the administered drug. 22 In our experience, most reactions thought to be allergic are Jarisch-Herxheimer reactions. Whereas an allergic
Recommendations for the prevention of Lyme disease have been published by the American Academy of Pediatrics,lS7 and the subject has been reviewed in detail elsewhere. 158
Avoidance Avoiding tick bites in endemic areas is difficult. Reduction of tick numbers on residential properties and gardens can be achieved through landscaping measures that create a drying barrier between forest and lawn, the use of acaricides, and the removal of deer from specified areas. 15S Appropriate clothing with light-colored long trousers tucked into socks makes it more difficult for ticks to attach to a human host. Tick repellents containing N;N-diethylmeta-toluamide (DEET) or permethrin applied to clothing can reduce tick attachment for several hours. 24 DEET may also be applied directly to skin, but the use of repellents on skin should be limited because toxic side effects can occur. Ticks should be removed promptly because B. burgdorferi resides in the tick's midgut and proliferation starts only after the host's blood has entered the tick's gut. Thereafter, B. burgdorferi organisms spread by the acarial hemolymph to the tick's salivary glands. Because this takes 24 to 36 hours, a daily search for and removal of ticks is helpful in endemic areas. 24 There is evidence that B. afzelii may be transmitted more quickly than B. burgdorferi sensu strictO. 159 Ticks should be grasped with tweezers or fingernails as close to their point of attachment as possible and pulled steadily away from the skin to allow the tick to detach its mouth parts. 24 Mouth parts that remain in the skin do not pose a risk for further transmission of B. burgdorferi but may lead to a superficial bacterial infection. The site of the tick bite should be disinfected after the tick is removed. The use of prophylactic antibiotics after a tick bite is controversial. A single dose of 200 mg of doxycycline after 1. scapularis bites was found to reduce the occurrence of erythema migrans. 160 However, most tick bites remain unnoticed, and in most geographic areas, the frequency of antibiotic side effects exceeds the estimate of preventable disease manifestations. Failures of
600
CHAPTER
29
LYME DISEASE
prophylactic treatment also have been described. 161.162 Prompt antibiotic treatment of the early manifestations of Lyme disease usually prevents late manifestations such as arthritis.
Immunization Two human vaccines have been developed with a recombinant fragment of OspA of B. burgdorferi sensu stricto. These vaccines were first evaluated in North American adults and found to be safe. After three injections, vaccine efficacy in adults was 76% to 92%.162.163 One of these vaccines was safe and efficacious in North American children,164 and recommendations for the use of the vaccine were published. 165 However, these vaccines were never intended for use in Europe or Asia because of the greater strain variation on these continents. The only licensed vaccine was withdrawn from the American market due to economic reasons.
COURSE OF THE DISEASE AND PROGNOSIS The prognosis for children with erythema migrans or early neuroborreliosis is excellent when the disease has been promptly treated with appropriate antibioticS. 153 .166-168 Even in children with Lyme arthritis who have not been treated, manifestations usually diminish and eventually disappear over time. 84 Among 90 children with Lyme arthritis treated with appropriate antibiotics, 4 had ongoing musculoskeletal complaints 7 years later. 71 Of 51 German children with Lyme arthritis examined 1 year after initiation of antibiotic treatment, 8 patients had chronic arthritis, and 4 had persistent arthralgias in joints previously affected by arthritis. 15l In rare cases, flares of arthritis in a previously affected joint have been observed several years after antibiotic treatment and the initial disappearance of arthritis. Erosion of cartilage is rare in children. Arthralgia in joints previously affected by arthritis may persist for several months, but in contrast to similarly affected adults, children generally do not fulfill diagnostic criteria for fibromyalgia, and arthralgias usually do not restrict the physical or educational performance of adolescents. Late neurologic complications have been described in untreated children with Lyme arthritis84 but have not been observed after appropriate treatrnent. 169 However, transient neurocognitive abnormalities may occur. 61 Late development of keratitis has occurred in treated and in untreated children. 84 Lyme disease is not fatal and rarely results in significant persistent organ damage, but coinfection with tick-borne encephalitis virus or with Ehrlichia or Babesia organisms may lead to a more severe disease course. 26-28 A follow-up study of American adult patients found more musculoskeletal disease and verbal memory impairment in patients than in controls. 170 Poor prognosis of Lyme arthritis in American adults has been associated with the presence of antibodies to OspA and the DRBl*0401and DRB1*0101 alleles. 53.54 The risk of treatment failure seems to increase with increasing age and when intra-articular steroids were given before antibioticS. 138.1';1
Szer and colleagues84 studied 46 American children (25 boys) with chronic Lyme arthritis with onset of disease between 1976 and 1979. None had been treated with antibiotics for the first 4 years of disease. Almost all (98%) had arthralgias during the early phase, with a median time from disease onset to development of arthritis of 3 months (range, 2 to 24 months). The number of children with recurrent episodes declined each year. Older children tended to have arthritis of longer duration. At the end of the study, 12 children (31%) still had occasional brief episodes of joint pain. One child had marked fatigue, and two developed keratitis. All 46 children had persistently positive IgG antibody responses. IgM responses were more frequent and IgG titers higher in children with recurrent symptoms than in those who became asymptomatic.
REFERENCES 1. Steere AC, Malawista SE, Snydman DR, et al: Lyme arthritis. An epidemic of oligoarticular arthritis in children and adults in three Connecticlit communities. Arthritis Rheum 20: 7-17, 1977. 2. Burgdorfer W, Barbour AG, Hayes SF, et al: Lyme disease-a tick-borne spirochetosis? Science 216: 1317-1319, 1982. 3. Steere AC, Grodzicki RL, Kornblatl AN. et al: The spirochetal etiology of Lyme disease. N Engl j Med 308: 733-740, 1983. 4. Stanek G. Strle F: Lyme borreliosis. Lancet 362: 1639-1647, 2003. 5. Steere AC: Lyme disease. N Engl J Med 345: 115-125, 2001. 6. i-Iuppertz HI: Childhood Lyme borreliosis in Europe. Em j Pediatr 149: 814-821, 1990. 7. Dressler F: Lyme borreliosis in European children and adolescents. Clin Exp Rheumatol 12 (Suppl 10): S49- S54, 1994. 8. Sigal LH: Musculoskeletal manifestations of Lyme arthritis. Rheum Dis Clin North Am 24: 323-351. 1998. 9. Buchwald A: Ein Fall von diffuser idiopathischer Hautatrophie. Arch Dermatol Syph 10: 553, 1883. 10. Afzelius A: Verhandlungen der dermatologischen Gesellschaft zu Stockholm. Arch Dermatol Syph 101: 404, 1910. 11. Garin C. Bujadoux A: Paralysie par les tiques. j Med Lyon 71: 765, 1922. 12. Bannwarth A: Chronische lymphocytare Meningitis, entzOndliche Polyneuritis und "Rheumatismus." Arch Psychiatr Nervenkr 113: 284, 1941. 13. Svartz N: Penicillinbehandling vid dermatitis atrophicans Herxheimer. Nord Med 32: 2783, 1946. 14. Binder E, Doepfmer R, Hornstein 0: Obertragung des Erythema chronicum migrans von Mensch zu Mensch in zwei Passagen. KIin Wochenschr 33: 727, 1955. 15. Steere AC: Lyme disease: a growing threat to urban populations. Proc Nat! Acad Sci USA 91: 2378-2383, 1994. 16. Centers for Disease Control and Prevention (CDC); Lyme disease-United States, 1994. MMWR Morb Mortal Wkly Rep 44: 459, 1995. 17. Centers for Disease Control and Prevention (CDC); Lyme disease-United States, 2000. MMWR Morb Mortal Wkly Rep 51: 29. 2002. 18. Centers for Disease Control and Prevention (CDC): West Nile virus activityUnited States, juiy 31-August 6, 2003. MMWR Morb Mortal Wkly Rep 52: 741. 2003. 19. Berglund j, Eitrem R, Ornstein K, et al: An epidemiologic study of Lyme disease in Southern Sweden. N Engl J Med 333: 1319. 1995. 20. Huppenz HI, Bohme M, Standaert SM, et al: Incidence of Lyme borreliosis in the WOrzburg region of Germany. Eur j Clin Microbiol Infect Dis 18: 697-703, 1999. 21. Gerber MA, Shapiro ED, Burke GS, et al: Lyme disease in children in southeastern Connecticut. N Engl J Med 335: 1270-1274, 1996. 22. Huppertz HI. Karch H, Suschke Hj, et a1: Lyme arthritis in European children and adolescents. Arthritis Rheum 38: 361-368, 1995. 23. Steere AC, Dwyer E, Winchester R: Association of chronic Lyme arthritis with HLA-DR4 and HLA-DR2 alleles. N Eng! J Med 323: 219-223, 1990. 24. Fish 0: Environmental risk and prevention of Lyme disease. Am J Med 98 (Suppl 4A): 2s-HS, 1995. 25. Subak 5: Effects of climate on variability in Lyme disease incidence in the Northeastern United States. Am j Epidemiol 157: 531-538, 2003 26. Cimperman J, Maraspin V, Lotric-Furlan S. et al: Concomitant infection with tick-borne encephalitis virus and Borrelia burgdorferi sensu lata in patients with acute meningitis or meningoencephalitis. Infection 26: 160-164. 1998. 27. Krause Pj, Telford SR. Spielman A, et al: Concurrent Lyme disease and babesiosis: evidence for increased severity and duration of disease. JAMA 275: 1657-1660, 1996.
C HAP T E R 28. Nadelman RB, Horowitz HW, Hsieh T, et al: Simultaneous human granulocytic ehrlichiosis and Lyme borreliosis. N Engl] Med 337: 27-30, 1997. 29. Barbour AG, Hayes SF: Biology of Borrelia species. Microbiol Rev 50: 381-400, 1986. 30. Wienecke R, Neubert U, Volkenandt M: Cross immunity among types of Borrelia burgdorftri. Lancet 342: 435, 1993. 31. Nowakowski], Schwartz I, Nadelman R, et al: Culture-confirmed infection and reinfection with Borrelia burgdorfm. Ann Intern Med 127: 130-132, 1997. 32. Fmser CM, Casjens S, Huang WM, et al: Genomic sequence of a Lyme disease spirochaete, Borrelia burgdorftri. Nature 390: 580-586, 1997. 33. Anderson JF, Magnarelli LA: Avian and mammalian hosts for spirocheteinfected ticks and insects in a Lyme disease focus in Connecticut. In Steere AC. Malawista AC, Craft ]E, et al (eds): First International Symposium on Lyme Disease, New Haven. Yale] Bioi Med 57: 638, 1984. 34. Coburn ], Magoun L, Bodary SC, et al: Integrins alpha(v)-beta3 and alpha5betal mediate attachment of Lyme disease spirochetes to human cells. Infect Immun 66: 1946-1952, 1998. 35. Coburn ]C, Leong]M, Erban ]K: Integrin alpha lIb-beta III mediates binding of the Lyme disease agent Borrelia burgdorftri to human platelets. Proc Nat! Acad Sci USA 90: 7059-7063, 1993. 36. Ma Y, Sturrock A, Weis ]J: Intracellular localization of Borrelia burgdorftri within human endothelial cells. Infect Immun 59: 671--678, 1991. 37. Klempner M, Noring R, Rogers RA: Invasion of human skin fibroblasts by the Lyme disease spirochete, Borrelia burgdorferi. J Infect Dis 167: 1074-1081, 1993. 38. Girschick HJ, Huppertz HI, ROssmann H, et al: Intracellular persistence of Borrelia burgdorferi in human synovial cells. Rheumatol Int 16: 125-132, 1996. 39. Yang L, Ma Y, Schoenfeld R, et al: Evidence of B-Iymphocyte mitogen activity In Borrelia burgdorferi--infected mice. Infect Immun 60: 3033-3041, 1992. 40. Yin Z, Braun], Neure L, et al: T cell cytokine pattern in the joints of patients with Lyme arthritis and It~ regulation by cytokines and anticytokines. Anhritis Rheum 40: 69-79, 1997. 41. Gross OM, Steere AC, Huber BT: T helper 1 response is dominant and localized ro the synovial fluid in patients with Lyme arthritis. J Immunol 160: 1022-1028, 1998. 42. Busch DH, ]assoy C, Brinckmann U, et al: Detection of Borrelia burgdorftrispecific CD8+ cytotoxic T-cells in patients with Lyme arthritis. J Immunol 157: 3534-3541, 1996. 43. Glatzel A, Entschladen F, Zollner TM, et a1: The responsiveness of human V III yO-T cells to Borrelia burgdorftri is largely restricted to synovial fluid cells from patients with Lyme arthritis. J Infect Dis 186: 1043-1046, 2002. 44. Roessner K, Wolfe], Shi C, et al: High expression of Fas ligand by synovial fluid-derived yO-T cells in Lyme arthritis. J Immunol 170: 2702-2710, 2003. 45. Miller LC, Isa S, Vannier E, et al: Live Borrelia burgdorftri preferentially activate interleukin-lbeta gene expression and protein synthesis over the interleukin1 recepror antagonist.] Clin Invest 90: 906-912, 1992. 46. Hahicht GS, Katona U, Benach JL: Cytokines and the pathogenesis of neuroborreliosis: Borrelia burgdorftri induces glioma cells to secrete interleukin-6. ] Infect Dis 164: 568-574, 1991. 47. Defosse DL, Johnson RC: In vitro and in vivo induction of tumor necrosis factor alpha by Borrelia burgdorftri. Infect Immun 60: 1109-1113, 1992. 48. Pohl-Koppe A, Balashov KE, Steere AC, et al: Identification of a T cell subset capable of both IFN-gamma and IL-I0 secretion in patients with chronic Borrelia burgdorftri infection.] Immunol 160: 1804-1810, 1998. 49. Weigelt W, Schneider T, Lange R: Sequence homology between spirochaete flagellin and human myelin basic protein. Immunol Today 13: 279-280, 1992. 50. Sigal LH: Cross-reactivity between Borrelia burgdorftri flagellin and a human axonal 64000 molecular weight protein.] Infect Dis 167: 1372-1378, 1993. 51. Kalish R, Leong ]M, Steere AC: Association of treatment-resistant chronic Lyme arthritis with HLA-DR4 and antibody reactivity to OspA and OspB of Borrelia burgdorftri. Infect Immun 61: 2774-2779. 1993. 52. Kamradt T, Lengl-]anssen B, Strauss AF, et al: Dominant recognition of a Borrelia burgdorftri outer surface protein A peptide by T helper cells in patients with treatment-resistant Lyme arthritis. Infect Immun 64: 1284-1289,
1996. 53. Gross OM, Forsthuber T, Tary-Lehmann M, et al: Identification of LFA-l as a candidate autoantigen in treatment-resistant Lyme arthritis. Science 2Bl: 703-706, 1998. 54. Steere AC, Falk B, Drouin EE, et al: Binding of outer surface protein A and human lymphocyte function-associated antigen I peptides to HLA-DR molecules associated with antibiotic treatment-resistant Lyme arthritis. Arthritis Rheum 48: 534-540, 2003. 55. Malane MS, Grant-Kels ]M, Feder HM, et al: Diagnosis of Lyme disease based on derrnatologic manifestations. Ann Intern Med 114: 490-498, 1991. 56. Feder HM, Gerber MA, Krause PJ, et al: Early Lyme disease: a flu-like illness without erythema migrans. Pediatrics 91: 456-459, 1993. 57. Pohl-Koppe A, Wilske B, Weiss M, et al: Borrelia lymphocytoma in childhood. Pediatr Infect Dis J 17: 423-426, 1998. 58. Btzonova I, Wollenberg A, Prinz JC: Acrodermatitis chronica atrophicans affecting all four limbs in an 11-year-old girl. Br J Dermatol 147: 375-378, 2002. 59. Christen H], Hanefeld F, Eiffert H, et al: Epidemiology and clinical manifestation., of Lyme borreliosis in childhood. A prospective multicentre study with special regard to neuroborreliosis. Acta Paediatr 82 (Suppl 386): 1-75, 1993.
29
LYME DISEASE
601
60. Hansen K, Lebech AM: The clinical and epidemiologic prome of Lyme neuroborreliosis in Den.mark 1985-1990. Brain 115: 399-423, 1992. 61. Bloom B], Wyckoff PM, Meissner HC, et al: Neurocognitive abnormalities in children after classic manifestations of Lyme disease. Pediatr Infect Dis] 17: 189-196, 1998. 62. Shapiro EE: Guillain-Barre syndrome in a child with serologic evidence of Borrelia burgdorftri infection. Pediatr Inf Dis J 17: 264-265, 1998. 63. Horneff G, Huppertz H-I, Muller K et al: Demonstration of Borrelia burgdorfiri infection in a child with Guillain Barre syndrome. Eur ] Pediatr 152: 81Q-812, 1993. 64. Kan L, Sood SK, Maytal]: Pseudotumor cerebri in Lyme disease: a case report and review of the literature. Pediatr Neurol 18: 439-431, 1998. 65. Scott IV, Silva-Lepe A, Siatkowski RM: Chiasmal optic neuritis in Lyme disease. Am] Ophthalmol 123: 136-138, 1997. 66. Lock G, Berger G, Grobe H: Neuroborreliosis: progressive encephalomyelitis with cerebral vasculitis. Monatsschr Kinderheilkd 137: 101-104, 1989. 67. Oksi J, Kalimo H, Marttila R], et al: In.flamrnatory brain changes in Lyme borreliosis. A report on three patients and review of literature. Brain 119: 2143-2154, 1996. 68. Chancellor MB, McGinnis DE, Shenot P], et al: Urinary dysfunction in Lyme disease.] Urol 149: 26-30, 1993. 69. Huppertz ill, Karch H: Die Lyme-Anhritis im Kindesalter: Monarthritis des Kniegelen.ks, klinisch nicht unterscheidbar von Monarthritiden unbekannter Ursache. Monatsschr Kinderheilkd 139: 759-764, 1991. 70. Hammers-Berggren S, Andersson U, Stiernstedt G: Borrelia arthritis in Swedish children: clinical manifestations in 10 children. Acta Paediatr 81: 921-924, 1992. 71. Gerber MA, Zemel 15, Shapiro ED: Lyme arthritis in children: clinical epidemiology and long-term outcomes. Pediatrics 102: 905-908, 1998. 72. Reimers CD, de Koning], Neubert U, et al: Borrelia burgdorftri myositis: report of eight patients. ] Neurol 240: 278-283, 1993. 73. Haupl T, Hahn G, Rittig M, et al: Persistence of Borrelia burgdorfm in ligamentous tissue from a patient with chronic Lyme borreliosis. Arthritis Rheum 36: 1621-1626, 1993. 74. Schnarr S, Wahl A, ]Orgens-Saathoff B, et al: Nodular fasciitis, erythema migrans, and oligoarthritis: manifestation of Lyme borreliosis caused by Borrelia aftelii. Scand] Rheumatol 31: 184-186, 2002. 75. Huppertz HI, Michels H: Pattern of joint involvement in children with Lyme arthritis. Br J Rheumatol 35: 1016-1018, 1996. 76. Huppertz HI, MOnchmeier D, Lieb W: Ocular manifestations in children and adolescents with Lyme arthritis. Br] Ophthalmol83: 1149-1152, 1999. 77. Marlovits S, Khanakah G, Striessnig G, et al: Emergence of Lyme arthritis after autologous chondrocyte transplantation. Arthritis Rheum 50: 259-264, 2004. 78. Christofaro RL, Appel MH, Gelb RF, Williams CL: Musculoskeletal manifestations of Lyme disease in children. ] Pediatr Orthop 7: 527-530, 1987. 79· Hoffmann ]C, Stichtenoth DO, Zeidler H, et al: Lyme disease in a 74-yearold forest owner with symptoms of dermatomyositis, Arthritis Rheum 38: 1157-1160, 1995. 80. Oksi], Mertsola ], Reunanen M, et al: Subacute multiple-site osteomyelitis caused by Borrelia burgdorftri. Clin Infect Dis 19: 891-8%, 1994. 81. Sigal LH, Patella S]: Lyme arthritis as the incorrect diagnosis in pediatric and adolescent flbromyalgia. Pediatrics 90: 523-528, 1992. 82. COX J, Krajden M: Cardiovascular manifestations of Lyme disease. Am Heart ] 122: 1449-1455, 1991. 83. Aaberg TM: The expanding ophthalmologic spectrum of Lyme disease. Am J Ophthalmol 107: n-80, 1989. 84, Szer IS, Taylor E, Steere AC: The long-term course of Lyme arthritis in children. N Engl J Med 325: 159-163, 1991. 85. Edwards KS, Kanengiser S, Li KI, et al: Lyme disease presenting as hepatitis and jaundice in a child. Pediatr Iruect Dis] 9: 592-593, 1990. 86. Weber K, Bratzke H], Neubert U, et al: Borrelia burgdorftri in a newborn despite oral penicilline for Lyme borreliosis during pregnancy. Pediatr Infect Dis J 7: 286-289, 1988. 87. Silver HM: Lyme disease during pregnancy. Infect Dis Clin North Am 11: 93-97, 1997. 88. Markowitz LE, Steere AC, Benach ]L, et al: Lyme disease during pregnancy. ]AMA 255: 3394-3396, 1986. 89. Nadal D, Hunziker UA, Bucher HU, et al: lruants born to mothers with antibodies against Borrelia burgdorftri at delivery. Eur] Pediatr 148: 426-427, 1989. 90. Johnston YE, Duray PH, Steere AC, et al: Spirochetes found in synovial microangiopathic lesions. Am J Pathol 118: 26-34, 1985. 91. Snydman DR, Schen.kein DP, Berardi VP, et al: Borrelia burgdorftri in joint fluid in chronic Lyme arthritis. Ann Intern Med 104: 798-800, 1986. 92. De Koning ], Bosma RE, Hoogkamp-Korstanje ]AA: Demonstration of spirochaetes in patients with Lyme disease with a modified silver stain. ] Med Microbiol 23: 261-267, 1987. 93. Berger BW, Johnson RC, Kodner C, Coleman L: Cultivation of Bomlia burgdorfiri from erythema migrans lesions and perilesional skin. ] Clin Microbiol 30: 359-361, 1992. 94. Stanek G, Klein], Bittner R, Glogar D: Isolation of Borrelia burgdorftri from the myocardium of a patient with longstanding cardiomyopathy. N Engl] Med 322: 249-252, 1990. 95. Lardier! G, Salvi A, Camerini F: Isolation of Borrelia burgdorfm from myocardium. Lancet 342: 490, 1993.
602
C HAP T E R
29
LYME DISEASE
96. Frey M, Jaulhac B, Piemont Y, et al: Detection of Bomdia burgdorf"i DNA in muscle of patients with chronic myalgia related to Lyme disease. Am J Med 104: 591-594, 1998. 97. Eppes SC, Nelson DK, Lewis LL, Klein JD: Characterization of Lyme meningitis and comparison with viral meningitis in children. Pediatrics 103: 1957-1960, 1999. 98. Golightly M: Laboratory considerations In the diagnosis and management of Lyme borrellosis. Am J Clin Pathol 99: 168-174, 1993. 99. American College of Physicians: Guidelines for laboratory evaluation in the diagnosis of Lyme disease. Clinical guideline, part 1. Ann Intern Med 127: 1106-1108, 1997. 100. Tugwell P, Dennis DT, Weinstein A, et al: Laboratory evaluation in the diagnosis of Lyme disease. Clinical guideline, part 2. Ann Intern Med 127: 1109-1123, 1997. 101. Sigal LH: Pitfalls In the diagnosis and management of Lyme disease. Arthritis Rheum 41: 195-204, 1998. 102. Karlsson M, Hovind-Hougen K, Svenungsson B, et aI: Cultivation and characterization of spirochetes from cerebrospinal fluid of patients with Lyme borreliosis. J Clin Microbiol 28: 47~79, 1990. 103. Nocton JJ, Dressler F, Rutledge BJ, et al: Detection of Borrtlia burgdorftri DNA by polymerase chain reaction in synovial fluid from patients with Lyme arthritis. N Engl J Med 330: 229-234, 1994. 104. Carlson D, Hernandez J, Bloom BJ, et al: Lack of Borrtlia burgdorftri DNA in synovial fluid samples from patients with antibiotic treatment-resistant Lyme arthritis. Arthritis Rheum 42: 2705-2709, 1999. 105. Priem S, Rittig MG, Kamradt T, et al: An optimized PCR leads to rapid and highly sensitive detection of Borrtlia burgdorftri in patients Witll Lyme borreliosis. J Clin Microbiol 35: 685-690, 1997. 106. Huppertz HI, Sclunldt H, Karch H: Detection of Borrtlia burgdorf"i by nested polymerase chain reaction in cerebrospinal flUid and urine of children with neuroborreliosis. Eur J Pediatr 152: 414-417, 1993. 107. Karch H, Huppertz HI: Repeated detection of Borrelia burgdorftri DNA in synovial fluid of a child with Lyme arthritis. Rheumatol Int 12: 227-229, 1993. 108. Jaulhac B, Chary-Valckenaere I, Sibilia J, et al: Detection of Borrtlia burgdorfm by DNA amplification in synovial tissue samples from patients with Lyme arthritis. Arthritis Rheum 39: 736-745, 1996. 109. Priem S, Burmester GR, Kamradt T, et al: Detection of Borrelia burgdorfm by polymerase chain reaction in synovial membrane, but not in synovial fluid from patients with persisting Lyme arthritis after antibiotic therapy. Ann Rheum Dis 57: 118-121, 1998. 110. Karch H, Huppertz HI, Bbhme M, et al: Demonstration of Borrtlia burgdorftri DNA in urine samples from healthy humans whose sera contain B. burgdorftrispecific antibodies. J Clin Microbiol 32: 2312-2314, 1994. 111. Brown SL, Hansen SL, Langone lJ: Role of serology In tbe diagnosis of Lyme disease. JAMA 282: 62-66, 1999. 112. Recommendations for test performance and interpretation from the Second National Conference on Serologic Diagnosis of Lyme Disease. MMWR Morb Mortal WkIy Rep 44: 590--591, 1995. 113. Dressler F, Whalen JA, Reinhardt BN, et aI: Western blotting in the serodiagnosis of Lyme disease. J Infect Dis 167: 39~00, 1993. 114. Engstrom SM, Shoop E, Johnson RC: Immunoblot Interpretation criteria for serodiagnosis of early Lyme disease. J Clin Microbiol 33: 419-427, 1995. 115. Hauser D, Lehnert G, Lobentanzer R, et al: Interpretation criteria for standardized Western blots for three European species of Borrtlia burgdorftri sensu lato. J Clin Microbiol 35: 143}-1444, 1997. 116. Hauser D, Lehnert G, Wilkske B: Validity of interpretation criteria for standardized Western blots (inlmunoblots) for serodiagnosis of Lyme borrellosis based on sera collected throughout Europe. J Clin Microbiol 37: 2241-2247, 1999. 117. Bacon RM, Biggerstaff BJ, Schriefer ME, et al: Serodiagnosis of Lyme disease by kinetic enzyme-linked inlmunosorbent assay using recombinant vlsE1 or peptide antigens of Borrtlia burgdorfn-i compared with 2-tiered testing using whole celllysates. J Infect Dis 187: 1187-1199, 2003. 118. Heikkila T, Huppertz HI, Seppala I, et al: Recombinant or peptide antigens in the serology of Lyme arthritis in children. J Infect Dis 187: 1888-1894, 2003. 119. Schulte-Spechtel D, Lehnert G, Uegl G, et al: Significant improvement of the recombinant Borrtlia-specific immunoglobulin G immunoblot test by addition of visE and a DbpA homologue derived from Borrtlia garinii for diagnosis of early neuroborreliosis. J Clln Microbiol 41: 1299-1303, 2003. 120. Levine D, Tilton RC, Parry MF et al: False positive EBNA IgM and IgG antibody tests for Infectious mononucleosis In children. Pediatrics 94: 892-894, 1994. 121. Berardi VP, Weeks KE, Steere AC: Serodiagnosis of early Lyme disease: analysis of IgM and IgG antibody responses by using an antibody-capture enzynje immunoassay. J Infect Dis 158: 754--760, 1988. 122. Sood SK, Rubin LG, Blader ME, Uowlte NT: Positive serology for Lyme borreliosis in patients with juvenile rheumatoid arthritis In a Lyme borrellosis endemic area: analysi' by immunoblol. J Rheumatol 20: 739-741, 1993. 123. Saulsbury FT, Katzmann JA: Prevalence of antibody to Borrelia burgdorf"i in children with juvenile rheumatoid arthritis. J Rheumatol17: 119}-1194, 1990. 124. Weiss NL, Sadock VA, Sigel LH, et al: False positive seroreactivity to Borrtlia burgdorftri in systemic lupus erythematosus: value of inlmunoblot analysis. Lupus 4: 131-137, 1995.
125. Rose CD, Fawcett PT, Gibney KM, Doughty RA: Residual serologic reactivity in children with resolved Lyme arthritis.] Rheumatol 23: 367-369, 1996. 126. Nichol G, Dennis DT, Steere AC, et al: Test-treatment strategies for patients suspected of having Lyme disease: a cost-effectiveness analysis. Ann Intern Med 128: 37-48, 1998. 127. Dressler F, Yoshinari NH, Steere AC: The T-cell proliferative assay In the diagnosis of Lyme disease. Ann Intern Med 15: 53}-539, 1991. 128. Zoschke DC, Skemp AA, Defosse DL: Lymphoproliferative responses to B. burgdorf"i In Lyme disease. Ann Intern Med 114: 285-289, 1991. 129. Huppertz HI, Mbsbauer S, Busch DH, et al: Lymphoproliferative responses to Borrtlia burgdorfm in the diagnosis of Lyme arthritis in children and adolescents. Eur J Pediatr 155: 297-302, 1996. 130. Rutkowski S, Busch DH, Huppertz HI: Lymphocyte proliferation assay in response to Borrelia burgdorferi in patients with Lyme arthritis: analysis of lymphocyte subsets. Rheumatol Int 17: 151-158, 1997. 131. Huppertz HI, Bentas W, Haubitz I, et al: Diagnosis of pediatric Lyme arthritis using a clinical score. Eur] Pediatr 157: 304-308, 1998. 132. Wharton M, Chorba TL, VOg!, RL, et al: Case definitions for public health surveillance. MMWR Morb Mortal Wkly Rep 39: 19, 1990. 133. Bbhme M, Schweneke S, Fuchs E, et al: Screening of blood donors and recipients for Borrelia burgdorftri antibodies: no evidence of B. burgdorfm infection transmitted by transfusion. Infusionstherapie 19: 204--207, 1992. 134. Zahid Qureshi M, New 0, Zulqarni NJ, Nachman S: Overdiagnosis and overlreatment of Lyme disease in children. Pediatr Infect Dis J 21: 12-14, 2002. 135. Huppertz HI, Karch H, Heesemann J: Diagnostic value of synovial fluid analysis in children with reactive arthritis. Rheumatol Int 15: 167-170, 1995. 136. Kaiser R, Rauer S: Analysis of the intrathecal inlmune response in neuroborreliosis to a sonicate antigen and three recombinant antigens of Borrtlia burgdorf"i sensu stricto. Eur J Clin Microbiollnfect Dis 17: 159-166, 1998. 137. Dattwyler RJ, Volkman DJ, Conaty SM, et aI: Amoxicillin plus probenecid versus doxycycline for treatment of erythema migrans borreliosis. Lancet 336: 1404-1406, 1990. 138. Dattwyler RJ, Halperin JJ, Volkman DJ, et al: Treatment of late Lyme borreliosis-randomised comparison of ceftriaxone and penicillin. Lancet 1: 1191-1194, 1988. 139. Weber K, Preac-Mursic V, Wilske B, et al: A randomized trial of ceftriaxone versus oral penicillin for the treatment of early European Lyme borreliosis. Infection 18: 91-96, 1990. 140. Pfister HW, Preac-Mursic V, Wilske B, et al: Cefotaxime versus penicillin G for acute neurologic manifestations in Lyme borreliosis. A prospective randomized study. Arch Neurol 46: 119Q-..1194, 1989. 141. MUllegger RR, Millner MM. Stanek G, Spork KD: Penicillin G sodium and ceftriaxone In the treatment of neuroborreliosis in children-a prospective study. Infection 19: 279-283, 1991. 142. Massarolti EM, Luger SW, Rahn DW, et al: Treatment of early Lyme disease. Am J Med 92: 396-403, 1992. 143. Nadelman RB, Luger SW, Frank E, et al: Comparison of cefuroxime and doxycycline in the treatment of early Lyme disease. Ann Intern Med 117: 273-280, 1992. 144. Eppes SC, Childs ]A: Comparative study of cefuJ'Oxinle axetil versus amoxicillin in children with early Lyme disease. Pediatrics 109: 117}-1177, 2002. 145. Dattwyler RJ, Luft BJ, Kunkel MJ, et al: Ceftriaxone compared with doxycycline for the treatment of acute disseminated Lyme disease. N Engl J Med 337: 289-294, 1997. 146. Hansen K, Hovmark A, l.ebeque AM, et al: Roxithromycin in Lyme borreliosis: discrepant results of an in vitro and in vivo animal susceptibility study and a clinical trial In patients with erythema migrans. Acta Derm Venerol 72: 297-300, 1992. 147. Weber K, Wilske B. Preac-Mursic V, Thurmayr R: Azithromycin versus penicillin V for the treatment of early Lyme borreliosis. Infection 21: 367-372. 1993. 148. Strle F, Preac-Mursic V, Cimperman J, et aI: Azithromycln versus doxycycline for treatment of erythema migrans: clinical and microbiologic findings. Infection 21: 83-88, 1993. 149. Luft BJ, Dattwyler RJ, Johnson Re, et al: Azithromycin compared with amoxicillin in the treatment of erythema migrans. A double-blind. randomized. controlled trial. Ann Intern Med 124: 785-791, 1996. 150. Steere AC, Levin RE, Molloy PJ, et al: Treatment of Lyme arthritis. Anhritis Rheum 37: 878-888. 1994. 151. Bentas W, Karch H, Huppertz HI: Lyme arthritis in children and adolescents: outcome 12 months after initiation of antibiotic therapy. J Rheumatol 27: 2025-2030, 2000. 152. Wormser GP, Ramanathan R, Nowakowski J, et al: Duration of antibiotic therapy for early Lyme disease. Ann Intern Med 138: 697-704, 2003. 153. Thorstrand C, Belfrage E, Bennet R, et al: Successful treatment of neuroborreliosis with ten day reginlens. Pediatr Infect Dis J 21: 1142-1145, 2002. 154. Eckman MH, Steere AC, Kalish RA, Pauker SG: Cost effectiveness of oral as compared with intravenous antibiotic therapy for patients with early Lyme disease or Lyme arthritis. N Engl J Med 337: 357-363, 1997. 155. Gasser R, Wendelin I, Reisinger E, et al: Roxithromycin In the treatment of Lyme disease-update and perspectives. Infection 23 (Supp\): 539-S43, 1995. 156. Schoen RT, Aversa JM, Rahn DW, et al: Treatment of refractory chronic Lyme arthritis with arthroscopic synovectomy. Arthritis Rheum 34: 1056-1060, 1991. 157. American Academy of Pediatrics, Committee on Infectious Diseases: Prevention of Lyme disease. Pediatrics 105: 142-147. 2000.
C HAP T E R
29
LYME DISEASE
603
158. Hayes EB, Piesman J: How can we prevent Lyme disease? N Engl J Med 348: 2424-2430, 2003. 159. Crippa M, Rais 0, Gem L: Investigations on the mode and dynamics of trans-
164. Feder HM, Beran J, van Hoecke C, et aI: Immunogenicity of a recombinant
mission and infectivity of Bo,.,..lia burgdorftri stnsU stricto and Bo,.,..lia aft'lii in lxotin ricinus ticks. Vector Borne Zoonotic Dis 2: 3-9, 2002. 160. Nadelman RB, Nowakowski J, Fish 0, et aI: Prophylaxis with single-dose doxycycline for the prevention of Lyme disease after an Ixotits scapularis tick bile. N Engl J Med 345: 79-84, 2001. 161. Warshafsky S, Nowakowski J, Nadelman RB, et al: Efficacy of antibiotic prophylaxis for prevention of Lyme disease. J Gen Intern Med 11: 329--333,
165.
1996.
166. 167. 168.
162. Steere AC, Sikand VK, Meurice F, et al: Vaccination against Lyme disease
with recombinant Bo,.,..lia burgdorftri outer-surface lipoprotein A with adjuvant. N Eng! J Med 339: 209--215, 1998. 163. Sigal LH, Zahradnik JM, Lavin P, et al: A vaccine consisting of recombinant Bo,.,..lia burgdorftri outer-surface protein A to prevent Lyme disease. N Engl J Med 339: 216-222, 1998.
169. 170.
Bo,.,..lia burgdorftri outer surface protein A vaccine against Lyme disease in children. J Pediatr 135: 575--579, 1999. Recommendations for the use of Lyme disease vaccine. Recommendations of the AdVisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 48: I-I7, 21-25, 1999. Adams WV, Rose CD, Eppes SC, Klein JD: Cognitive effect' of Lyme disease in children: a 4 year followup study. J Rheumatol 26: 1190-1194, 1999. Salazar lC, Gerber MA, Goff CWo Long-term outcome of Lyme disease in children given early treatment. 1 Pediatr 122: 591-593, 1993. Seltzer EG, Gerber MA, Cartter M, et al: Long-term outcomes of persons with Lyme disease. lAMA 283: 609-616, 2000. Adams WV, Rose CD, Eppes SC, KleinlD: Cognitive effects of Lyme disease in children. Pediatrics 94: 185--189, 1994. Shadick NA, Phillips CB, Logigian EL, et al: The long-term clinical outcomes of Lyme disease. A population-based retrospective cohort study. Ann Intern Med 121: 560-567, 1994.
C HAP T E R
30
REACTIVE ARTHRITIS Ruben Burgos-Vargas and Janltzla Vazquez-Mellado
~
The reactive arthritides constitute a diverse group of inflammatory arthritides, including acute rheumatic fever and arthritis after genitourinary tract or gastrointestinal tract infections with specific organisms. They are probably among the most common of the childhood rheumatic diseases worldwide. Viral and postinfectious arthritides are discussed in Chapter 28. Acute rheumatic fever and poststreptococcal arthritis are considered in Chapter 31. This chapter reviews disorders related to enteric or genitourinary bacterial infections.
DEFINmON In this chapter, use of the term reactive arthritis is restricted to the human leukocyte antigen (HLA)B27-associated arthritides, a group of conditions traditionally classified with the spondyloarthropathy group and triggered by enteric and genital bacterial infections. Reactive arthritis is a form of nonseptic arthritis developing after an extra-articular infection with one of the so called arthritogenic bacteria, particularly Chlamydia, Yersinia, Salmonella, Shigella, or Campylobacter, 1-3 Reiter's syndrome is a presentation of reactive arthritis defined by the triad of arthritis, conjunctivitis, and urethritis (or cervicitis). The term had been frequently used in the pediatric rheumatology literature in the past.
CLASSIFICATION AND DIAGNOSTIC CRITERIA The diagnosis of reactive arthritis is a clinical challenge. The criteria used in the literature and in clinical practice to make a diagnosis of reactive arthritis have ranged from a brief episode of undifferentiated arthritis to criteria such as those of the 1995 Berlin Third International Workshop on Reactive Arthritis 4 (Table 30-1), which require the presence of typical peripheral arthritis (a predominantly lower limb, asymmetric oligoarthritis) in addition to evidence of a preceding infection (either a history of diarrhea or urethritis within the preceding 4 weeks or laboratory confirmation of infection with an arthritogenic organism in the absence of clinical symptoms) (Table 30-2). The value of these criteria is yet to be determined, particularly with regard to the retrospective diagnosis of infection, The problem of definition and classification of reactive arthritis has been reviewed
604
by Pacheco-Tena and colleagues. s In general, the value of diagnostic tests depends on the pretest probability of the diagnosis. 6
EPIDEMIOLOGY The occurrence of reactive arthritis is related to the prevalence of HLA-B27 and probably to the rate of infections by arthritogenic bacteria in the general population, However, reactive arthritis may occur in HLA-B27-negative patients. Reports of Yersinia-triggered reactive arthritis in children have usually come from countries in which infection with Yersinia is common. In epidemiologic studies summarized by Keat,3 reactive arthritis was estimated to occur in 1% of patients with sexually-acquired infections, 2.4% of those with Shigella or Campylobacter infection, 3.2% with Salmonella infections, and up to 33% of adult patients with yersiniosis. Reactive arthritis develops in 5% to 10% of children with yersiniosis. 7 ,8 In a report of a Salmonella outbreak in Germany,9 no case of reactive arthritis occurred among 286 infected children, although 6 children had brief arthralgia. The relative frequency of reactive arthritis among patients in four registries of pediatric rheumatology clinics in the United States, 10, 11 United Kingdom,12 and Canada B ranged between 8.6% and 41.1%, This wide variation is consistent with differences in the stringency of diagnostic and classification criteria used in each study. The American and Canadian data lO ,JI,13 included four different but related categories: Reiter's syndrome, probable Reiter's syndrome, reactive arthritis, and probable reactive arthritis. In contrast, the British study12 included only the Reiter's syndrome category. Reports from other sources suggest that clinical recognition of reactive arthritis may be increasing. I 4-19 Most cases of reactive arthritis occur in boys between the ages of 8 and 12 years, but sex and age distribution vary according to the causative organism. In an Italian study of children with Yersinia-triggered reactive arthritis, most cases occurred between 3 and 7 years of age, and there was a slight predominance of females, 19 Enteric infections are responsible for reactive arthritis at all ages, but reactive arthritis after genital infections with Chlamydia occurs more frequently during adolescence.
C HAP T E R
II • -
1 !\B! E 30-1 The Berlin Diagnosli( Criteria for I<~
TypIcal PerIpheral ArthrItIs Predominantly lower limb, asymmetric oligoarthritis plus
Evidence of PrecedIng Infection If there is a clear history of diarrhea or urethritis within the preceding 4 weeks, laboratory confirmation is desirable but not essential. Where no clear clinical infection is identified, laboratory confirmation of infection is essential.
ExdusJon CrIterIa Patients with other known causes of monarthritis or oligoarthritis (e.g., other defined spondyloarthropathies, septic arthritis, crystal arthritis, Lyme disease, streptococcal reactive arthritis) should be excluded. From Kingsley G, Sieper ]: Third International Workshop on Reactive Arthritis: an overview. Ann Rheum Dis 55: 564-570, 1996. Reproduced with permission from BM] Publishing Group.
GENmC BACKGROUND Although the susceptibility to the primary infection is not related to any genetic marker, reactive arthritis most frequently occurs in HLA-B27-positive individuals. The HLA-B27 association and most clinical manifestations relate reactive arthritis and enthesitis-related arthritis (juvenile ankylosing spondylitis). The frequency of HLAB27 in children with reactive arthritis varies widely. In some children, particularly those with mild forms of Yersinia-, Campylobacter-, and Chlamydia-related reactive arthritis or those with nasopharyngeal infection, the frequency of HLA-B27 is similar to that of the general population. 7 ,8,20.21 In other small series, the prevalence of HLA-B27 may reach 60%. Artamonov and colleagues21 found a relative risk of 149 in HLA-B27-positive children with Reiter's syndrome after gastrointestinal infection. An association with the tumor necrosis factor c1 allele that is independent of B27 was reported in a predominantly adult Finnish population with reactive arthritis. 22 In a sim-
II II -
TAB! f 30 -2 Laboratory Tests for Documenting Preceding Infl ! lion in Reactive Arthritis
Stool culture Urethral culture Serology: antibodies against specific arthritogenic bacteria Urethral swab for detection of bacterial DNA by polymerase chain reaction (PCR)" Synovial fluid or synovial membrane for detection of bacterial DNA by PCR" Immunofluorescence microscopy for detection of bacteria in synoviumt Stimulation of synovial fluid lymphocytes with antigens from arthritogenic bacteria t •A potential diagnostic test. tRes~arch tools. not suitable for routine diagnostic use. From Kingsley G, Sieper ]: Third International Workshop on Reactive Arthritis: an overview. Ann Rheum Dis 55: 564-570, 1996. Reproduced with permission from BM] Publishing Group.
30
REACTIVE ARTHRITIS
605
ilar population, TAP2J, a polymorphism of transporters associated with antigen processing (TAP2), was more common in HLA-B27-positive patients with reactive arthritis. 23 No other genetic associations have been identified.
ETIOLOGY AND PATHOGENESIS Arthritogenic Bacteria Several bacteria may cause reactive arthritis. In preadolescent patients, reactive arthritis after Salmonella, Shigella, Yersinia, and Campylobacter enteric infections is much more frequent than that after genital infections with Chlamydia. In children, gastroenteritis precedes the onset of arthritis in 80% of cases. Shigellajlexneri,24-27 Yersinia enterocolitica,28 Salmonella enteritidis,29 Salmonella oranienburg,27 and Salmonella typhimurium30 have all been isolated from children with postdysenteric Reiter's syndrome. In at least two youths and three children, Chlamydia trachomatis31 has been identified in synovial flUid. Respiratory tract infection with Mycoplasma pneumoniae32 or Chlamydia pneumoniae33 has preceded the development of reactive arthritis in a few children, and the latter agent was responsible for approximately 10% of cases of reactive arthritis in a Finnish study.33 Enteritis caused by Clostridium dif!icile,34,35 the protozoan Giardia lamblia,36 and Cryptosporidium37 has occasionally been associated with reactive arthritis, although the extent of these associations and their relationship to HLA-B27 are not known. In the study by Kocar and colleagues,34 about one half of the arthritis patients with C. difficile toxin A were HLA-B27 positive, suggesting that this disease falls into the same category as post-Yersinia arthritis. However, this finding requires further confirmation.
Role of HLA-B27 The role of HLA-B27 in the pathogenesis of reactive arthritis is still unknown. 38-41 The arthritogenic peptide theory postulates a CD8-positive cross-reactive T cell response to endogenous or exogenous peptides presented by HLA-B27-positive antigen-presenting cells. 4o ,42-46 These peptides induce a CD8-positive T cell response that cross-reacts with bacterial epitopes. 45 ,46 The role of T cells in the inflammatory process that characterizes reactive arthritis is paramount. 45 ,47,4B The promiscuous peptide theory suggests that class II molecules present B27-derived peptides to CD4-positive cells. 39 ,43,49 In this case, these peptides would induce a CD4-positive response, which then cross-reacts with B27 peptides. Although the serum antibody response against arthritogenic bacteria in patients with reactive arthritis lasts longer than that in patients with the same infection who do not develop arthritis, the role of antibodies in the pathogenesis of reactive arthritis is probably minimal. Other hypotheses include defective oxidation of cysteine at the B27 B pocket and the existence of B27-linked genes. 39,40,50 It has been postulated that misfolding of B27 in the cytoplasm may be an important mechanism in the
606
C HAP T E R
30
REACTIVE ARTHRITIS
pathogenesis of spondyloarthropathyY Additional findings suggest a role for heat-shock proteins and bacterial peptidoglycan in the pathogenesis or reactive arthritis. 52,53 Arthritogenic bacteria invade the mucosa and replicate within polymorphonuclear cells and macrophages. 54-56 The transport of bacteria and their products from the mucosa to the joint is complex and involves adhesion molecules on phagocytic cells. Studies in murine fibroblasts transfected with B27 indicated that the expression of this antigen inhibited invasion by arthritogenic bacteria. 57,58 This phenomenon could not be replicated in human cells, however,59-61 although persistence of the organism within the cell was prolonged in cells expressing B27. 62 It has been possible to identify various bacterial components (including lipopolysaccharide, DNA, and RNA) in synovial fluid cells and synovial membranes of patients with reactive arthritis. 63-72
CLINICAL MANIFESTATIONS The course and severity of reactive arthritis vary considerably. Several stages of reactive arthritis may be recognized. A clinical infection precedes the appearance of arthritis, enthesitis, or extra-articular disease (singly or in combination) by 1 to 4 weeks. After an active period of weeks to months, the arthritis subsides, and the patient may then enter a sustained remission or follow a pattern characterized by recurrent episodes of disease activity lasting a number of years, which may evolve into ankylosing spondylitis (Le., enthesitis-related arthritis).
Charaderlstlcs of the Primary Infection An appreciation of the characteristics of the preceding infectious illness helps to identify patients with reactive arthritis. In retrospect, diarrhea had preceded the onset of disease in most children with reactive arthritis.
Shigella Enteritis A period of high fever, with or without watery diarrhea and cramping abdominal pain lasting 48 to 72 hours, may be followed in 7 to 21 days by the sudden onset of oligoarthritis, most commonly affecting the knees and ankles. The arthritis is nonmigratory and lasts several weeks to 3 or 4 months. Diagnosis requires a careful history, demonstration of a sterile synovial fluid culture, the presence of agglutinins to Shigella flexneri serotype 2 or 2a,26,27 and an attempt to isolate the organism from the stool. Because of the long interval between the diarrhea and the joint complaints, blood cultures are positive in less than 4% of patients.
Salmonella Infection The acute onset of oligoarthritis, most often affecting the knees and ankles, may follow an enteric infection with S. typhimurium or S. enteritidis by 1 to 3 weeks.73.74 The enteric infection may be mild, but the onset of arthritis is usually accompanied by low-grade fever. Because Salmonella infection can result in osteomyelitis and sep-
tic arthritis as well as reactive arthritis, it is important to make certain that the synOVial fluid is sterile. The erythrocyte sedimentation rate (ESR) is usually elevated, and the leukopenia that may accompany the acute infection is generally followed by leukocytosis. Stool cultures are usually pOSitive, even late in the disease course, but seroconversion to Salmonella Hand 0 antigens occurs in only 50% of patients.
Yerslnla Infection Reactive arthritis of peripheral joints or the spine follows infection with Yersinia in susceptible children. 19 ,4-76 The interval between infection and onset of arthritis in 18 children with Yersinia-triggered reactive arthritis reported by Taccetti and colleagues 19 was 7 to 30 days. The diarrhea preceding reactive arthritis was very mild, much more so than in the usual Yersinia enterocolitis. Contact with the organism is through infected drinking water or milk. Yersinia enteroeolitiea causes gastroenteritis in young children and a syndrome of abdominal pain similar to that of appendicitis in older children and adolescents. In a study of children who were hospitalized because of Yersinia infection, 35% had arthritis lasting 3 to 22 months (average, 6.5 months).7 Of those with arthritis, 85% were positive for HLA-B27. Yersinia can occasionally cause septic arthritis.
Campylobaeter Infection In an epidemic of Campylobaeter jejuni enteritis in Finland, 2.6% of patients (all adults) developed oligoarthritis or polyarthritis 4 days to 4 weeks after infection. SynOVial fluid cultures were negative, and 33% of the patients with arthritis were positive for HLA-B27,77
Chlamydia Infection Genitourinary tract infection with C. traehomatis is often asymptomatic but may cause dysuria, frequency, and a urethral or vaginal discharge. Reactive arthritis may also be related to upper respiratory tract infections with C. pneumoniae. 33 Artamonov and colleagues21 found evidence of nasopharyngeal infection in 45 of 52 children with reactive arthritis. Although the prevalence of HLAB27 was higher than that in the control population (relative risk = 2.5), it was lower than that in those who developed reactive arthritis after intestinal infection.
Musculoskeletal Disease Acute arthritis is characteristic of reactive arthritis, but some children present with only slight to moderate joint pain and swelling occurring over several weeks. 7,193 1.7oHlJ Enthesitis may occur alone or with arthritis, tenosynovitis, or bursitis (Figs. 3G-1 and 3G-2). In other children, arthralgias antedate the onset of arthritis for a variable period. The initial episode of arthritis usually affects the knees or ankles. The pattern of arthritis in the metatarsophalangeal joints and the proXimal and distal interphalangeal joints of the feet may be that of a dactylitis and involve two or
C HAP T E R
30
REACTIVE ARTHRITIS
607
dren with Reiter's syndrome developed oligoarthritis, 7 had polyarthritis, and 1 had monarthritis, and axial symptoms occurred in 6. Five patients followed for a mean of 83.5 months developed radiographic sacroiliitis. Symptoms remitted in most patients, but some had a sustained or a fluctuating course. In a group of nine Greek children with Salmonella-triggered reactive arthritis, disease was active at 4 to 13 months, and there were one to four recurrences in four patients during 48 to 78 months but no axial symptoms. 74
Constitutional Signs and Symptoms
• figure 30-1 There is slight swelling of the midfoot and dactylitis involving the right second toe and the fourth and fifth left toes in an adolescent with Salmonella-triggered reactive arthritis of 6 months'duration.
Apart from residual disease resulting from the infection itself, children with reactive arthritis may continue to have fever, weight loss, fatigue, and muscle weakness during active periods of disease. Polyarthralgia, muscle pain, and joint stiffness affecting peripheral joints and the axial skeleton sometimes accompany these symptoms. Myocarditis and pericarditis have been described during the active phase of the disease in children with Salmonella enteritidis-triggered reactive arthritis. 82
Mucocutaneous and Ocular Disease three joints in one or more digits in combination with tenosynovitis and bursitis. Arthritis of the small joints of the hands has also been described in cases of reactive arthritis caused by Yersinia and Salmonella. 7.!9.3!.7?>--76 The synovial fluid effusion is usually marked, but proliferative synovitis is uncommon. In addition to involvement of peripheral joints and entheses, there may be inflammation of joints of the axial skeleton resulting in spinal and sacroiliac pain, stiffness, and reduced mobility of the lumbar and cervical spine. In a study of 11 children with reactive arthritis followed for 0.9 to 6.7 years, Hussein78 observed recurrent episodes of arthritis in most patients: 4 children had severe arthritis, and 5 had sacroiliitis, but none had significant disability. Cuttica and colleagues79 found that, at a mean follow-up of 28.6 months, 18 of 26 chil-
• FIgIn 30-Z Achilles tendinitis and swelling of the retrocalcaneal bursa of the right foot of the patient in Figure 30-1.
Painless, shallow ulcers of the oral mucosa and palate are common and often ignored because they are asymptomatic. Aphthous stomatitis occurs in some patients. Urethritis and cervicitis are rare manifestations, occurring more frequently in adolescents with sexually acquired reactive arthritis caused by Chlamydia but also developing in patients with reactive arthritis of other causes. 3! These conditions are often mild, and girls tend to have no symptoms; disease is detected only because of the presence of sterile pyuria. Diarrhea occurs in association with bacterial infection but may also be part of a generalized episode of mucositis. Skin lesions in reactive arthritis include erythema nodosum in some children with Yersinia-triggered reactive arthritis as well as circinate balanitis (Fig. 30-3) and keratoderma blennorrhagicum (Figs. 30-4 and 30-5), with or without other stigmata of Reiter's disease.9.3J.75.83 The latter may be clinically and histologically indistinguishable from psoriasis (Fig. 30-6). Several other nonspecific skin lesions have been described in patients with reactive arthritis and Reiter's syndrome. Mucocutaneous involvement in reactive arthritis tends to parallel disease activity in the peripheral joints. Conjunctivitis, one of the components of Reiter's syndrome, occurs in about two thirds of children at onset. In Yersinia-triggered reactive arthritis, conjunctivitis may be purulent and severe. 84 Acute iridocyclitis in these cases is characterized by aqueous flare and cells, small keratic precipitates, cells in the vitreous, and occasionally by fibrinous exudates, posterior synechiae, and macular edema in a unilateral or bilateral pattern. Acute anterior uveitis has also been described in reactive arthritis triggered by S. typhimurium. 85 Although there are few studies of the visual prognosis in children with reactive arthritis, the percentage of patients with permanent ocular sequelae appears to be low.
608
C HAP T E R
30
REACTIVE ARTHRITIS
• Figure 38-3 Circinate balanitis in an adolescent with Chlamydiatriggered reactive arthritis.The shallow ulcers on the glans penis are usually painless.
• Figure 30-5 Dorsal (A) and lateral (B) views of the foot of a 16-year old patient with chronic reactive arthritis.The third toe of the right foot is swollen. The mid-foot and ankle are also swollen. Several nails are dystrophic and cutaneous lesions of keratodenna blennorrhagicum are evident.
• Figure 38-4 Keratodenna blennorrhagicum.This scaly eruption on the soles of the feet of an 18-year-old youth with reactive arthritis is difficult to distinguish from psoriasis.
• Figure 30-6 Keratodenna blennorrhagicum resembling psoriasis on the face of a 9-year-old giri with Chlamydia-induced reactive arthritis who became infected with Chlamydia as a result of sexual abuse. (Courtesy of Dr. Mario Magana-Garcia.)
C HAP T E R
30
REACTIVE ARTHRITIS
609
LABORATORY EXAMINATION
RADIOLOGIC EXAMINATION
In the early inflammatory phase, there may be a slight decrease in the hemoglobin concentration and hematocrit, as well as mild leukocytosis and neutrophilia. The platelet count and serum levels of immunoglobulins (Ig) M, G, and occasionally A may be elevated. The ESR and C-reactive protein levels correlate with disease activity. In patients with severe disease-particularly in those with polyarthritis and polyenthesitis, fever, weight loss, fatigue, mucositis, or dermatitis-each of the aforementioned laboratory abnormalities may be extreme. In particular, the hemoglobin concentration may fall to 8 to 10 g/dL and the platelet count rise well above 400,000 mm 3. The ESR and C-reactive protein values may remain elevated for a protracted period. Autoantibodies (e.g., rheumatoid factor, antinuclear antibodies) are usually absent. Synovial fluid analysis helps to distinguish between reactive arthritis and septic arthritis. 86 With the exception of epidemics and some isolated reports, the clinical and laboratory confirmations of infection as a trigger in children with reactive arthritis are seldom made. When available, cultures obtained at the time of the infection may be helpful. Salmonella, Yersinia, Shigella, and Campylobacter may be isolated from the gut during an episode of diarrhea, or Chlamydia may be cultured from the urethra, but negative results do not exclude the diagnosis of infection-related arthritis. Because Salmonella and Chlamydia may also be present in asymptomatic carriers, these organisms can occasionally be cultured from patients who have arthritis that is not directly related to these organisms. Reactive arthritis may be diagnosed in the appropriate clinical setting because of the presence of high titers of serum antibodies against arthritogenic bacterial antigens. 4.5 Hemagglutination tests are sometimes useful in documenting recent infections with Salmonella or Yersinia. 7,19.31,73,75.76 The sensitivity and the specificity of circulating IgA and IgM antibodies to Salmonella, Yersinia, and Campylobacter detected by enzyme-linked immunoassay are acceptable, but results must be compared with those in the control population. IgG antibodies are useful if levels change significantly; a rising titer of IgA antibodies may be detected. Lymphoproliferation assays performed on cells from peripheral blood or synovial fluids also have some utility as diagnostic tools. 4.5 Unfortunately, these tests are not easy to perform and often demonstrate nonspecific responses to several antigens. Yersinia or Chlamydia antigens may be detected in intestinal or genital smears or biopsies. By using electron and immunofluorescence microscopy and immunohistochemistry, it has been possible to identify intra-articular chlamydial elemental bodies,31,63 Yersinia 60-kD heatshock protein and the urease ~ subunit,64,65 and Salmonella lipopolysaccharide. 66 Likewise, bacterial DNA or RNA from several bacterial species, including Chlamydia sp, Salmonella sp, Shigella sp, and Campylobacter sp has been identified in synovial fluid cells or the synovial membrane by polymerase chain reaction.6Il-n.78 The role of these tests as diagnostic tools is restricted.
Radiographic abnormalities early in the disease consist only of nonspecific soft tissue swelling, juxta-articular osteopenia, and less frequently, slight periosteal irregularities at tendon attachments. 87 The occurrence of subchondral cysts, erosions, and sometimes extensive destruction of joints such as the hips, proximal and distal interphalangeal joints of the hands and feet, and less commonly, joints of the wrist indicate the severity of the synovitis that can be present in reactive arthritis. Ultrasonographic studies may delineate synovial sheath and tendon thickening and the accumulation of synovial fluid within the tendon sheath and bursae. Magnetic resonance imaging is an excellent method for studying peripheral and axial joints and entheses. 80 In certain anatomic areas, particularly the foot, osteopenia may be extensive and affect entire bones. Various entheses, especially those at the attachment of the plantar fascia to the calcaneus, develop erosions and marked bony proliferation and spur formation (Fig. 30-7). These abnormalities may also be apparent in the navicular bone, greater trochanter, and ischium. Subchondral cysts and bony erosions of the hip and the metacarpophalangeal, metatarsophalangeal, and proximal interphalangeal joints of the hands and feet in a unilateral or asymmetric distribution characterize more extensive and unremitting disease. An association between joint erosions and occult inflammation of the gut has been described in patients with reactive arthritis. 88 Sacroiliac and spinal involvement in children with reactive arthritis is rare (Fig. 30-8). However, in an extensive study of the utility of magnetic resonance imaging of the sacroiliac joints in children with reactive arthritis, but no sacroiliac joint symptoms,89 approximately one half had acute or chronic sacroiliitis.
• Figure :18-7 The lateral radiograph of a foot of a 16-year-old boy, whose first symptom started after a I-week episode of diantlea at the age of 8 years, demonstrates marked osteopenia and probable ankylosis of the intertarsal joints. During the course of his disease, he experienced recurrent episodes of arthritis and enthesitis that were sometimes associated with diantlea. Salmonella organisms were isolated from his stools on two oc:c:asions, and bacterial DNA was identified from the knee that was consistent with the stool cultures.
610
C HAP T E R
30
REACTIVE ARTHRITIS
also be differentiated from reactive arthritis. Dactylitis and skin manifestations such as keratoderma b/ennorrhagicum and nail changes may make it difficult to separate children with reactive arthritis from those who have developed psoriatic arthritis.
TREATMENT
• Figure 30-8 Sacroiliac joints of a 16-year-old youth with reactive arthritis. On the left. there are erosions and marked reactive sderosis that are most prominent on the iliac side of the joints. Changes on the right are minimal.
DIFFERENTIAL DIAGNOSIS Differentiation of reactive arthritis from other types of arthritis is often difficult. Table 30-3 lists the most common conditions that need to be excluded. Reactive and infectious arthritides not associated with HLA-B27 share similar symptoms of arthritis after infection, although the primary site of infection is usually the upper airway rather than the gastrointestinal or genitourinary tracts. The second group of diseases possibly related to infection includes those in which mucocutaneous symptoms and arthritis predorninate. 9o The third group includes certain orthopedic conditions and pain syndromes that can mimic arthralgias or enthesopathy and have foot swelling and tenderness. Other forms of childhood arthritis must
'~II
TABLE ]() 3
Disease activity, functional status, and quality of life should be evaluated in children with reactive arthritis. Although no specific instruments have been developed for this disease, the use of validated measures of health status designed for other chronic arthritides of childhood should be helpfu1 91 ,92 (see Chapter 7). In addition to the arthritis, a number of other issues are relevant to quality of life. In developing countries, where reactive arthritis is most common, resources for treating reactive arthritis are limited. Socioeconomic and educational factors determine the prognosis of many chronic diseases. Awareness of the attributes of each culture leads to a better understanding of the effects of the disease on the child, family, and society. There are no special nutritional recommendations for children with reactive arthritis. Any measure taken to avoid bacterial contamination of food, from slaughtering of the animals to refrigeration, cooking, and serving, is essential to avoid enteric infections. Reactive arthritis is an epidemic disease which is probably endemic in some areas of the world. Programs that improve the sanitary conditions of the community to prevent the spread of infectious diseases are required. Counseling of the family about the risk of recurrences in case of enteric infections is advisable. This applies to adolescents with regard to sexual activities and the risk of sexually transmitted diseases.
Differential Diagnosis of Readive Arthritis
Infectious Arthritides Viral arthritis Poststreptococcal arthritis, rheumatic fever Lyme disease Septic arthritis, tuberculosis, gonococcal arthropathy
Disease PossIbly Related to Infectlon Irritable hip or toxic synovitis of the hip SAPHO syndrome (I.e., synovitis, acne, pustulosis, hyperostosis, and osteomyelitis) Beh~et's disease Kawasaki disease
Orthopedic and Ampllftcatlon Pain Syndromes Legg-Calve-Perthes, Osgood-Schlatter disease "Growing pains" (I.e., benign nocturnal pains of childhood) Idiopathic pain syndromes (e.g., fibromyalgia, reflex sympathetic dystrophy)
Juvenile Idiopathic Arthritis Oligoarthritis Polyarthritis Psoriatic arthritis Enthesitis related arthritis Arthritis associated with Crohn's disease or ulcerative colitis
Pharmacologic Therapy The inflammatory manifestations of reactive arthritis require the administration of nonsteroidal anti-inflammatory drugs (NSAIDs) in most patients and glucocorticoids in some. The requirement for anti-inflammatory drug treatment of reactive arthritis tends to be intermittent rather than constant. However, in patients in whom reactive arthritis becomes chronic, medications, sometimes including sulfasalazine, should be maintained for long periods. Except for the use of antibiotics in selected cases, there is no clear evidence that any drug alters the course of the disease. Recommended doses and therapeutic regimens of NSAIDs in children with reactive arthritis are similar to those used in other forms of childhood arthritis. Because episodes of reactive arthritis tend to be self-limiting, lasting from 3 to 6 months, NSAIDs may be discontinued in many children with onset of a remission. Glucocorticoids may be reqUired for children with severe and disabling polyarthritis and polyenthesitis. Oral or intravenous glucocorticoids generally control arthritis. In contrast, enthesitis responds poorly, may require higher doses of drug than usual, and may demand a longer period of treat-
C HAP T E R
ment. Fever, fatigue, and anemia tend to disappear, and the C-reactive protein levels and ESR tend to improve after several weeks of treatment. Despite this and perhaps as a consequence of the episodic nature of the disease, glucocorticoid dose reduction and withdrawal in children with reactive arthritis usually are achieved easily. The required doses of prednisone or prednisolone vary between 10 and 20 mg/day and doses of deflazacort between 12 and 24 mg/day. To minimize the risk of adverse events, it is advisable to administer the drug as a single dose in the morning and to start reducing the dose after 2 to 3 weeks. As an alternative to oral therapy, intravenous methylprednisolone offers several advantages. One recommended approach is a step-down regimen of 3 consecutive days of 15 to 30 mg/kg, with a maximum of 1 g to be administered during the first day. Doses on days 2 and 3 are reduced to approximately 75% and 50%, respectively. Depending on clinical response, this schedule may be repeated at 4-week intervals. The need for this approach to suppression of the disease rarely extends beyond the first 3 months of illness. Intra-articular and intralesional administration of glucocorticoids are recommended in special situations. In patients with slight or mild synovitis, the intra-articular administration of triamcinolone hexacetonide or analogues of methylprednisolone or hydrocortisone produces rapid and sustained relief. It is not unusual, however, for synovial fluid to reaccumulate in less than a week. There is no reported experience regarding the injection of synovial sheaths, bursae, or entheses in children with reactive arthritis. Injection of entheses may result in postinjection pain and local soft tissue calcification or atrophy. Because of the possible presence of occult inflammation of the gut in patients with HLA-B27-associated arthritis, including reactive arthritis,93.94 and their responsiveness to sulfasalazine,95-98 this drug is often recommended in the management of resistant arthritis and enthesitis. The administration of sulfasalazine at doses ranging from 30 to 50 mg/kg/day (maximum, 1.5 to 2.0 g/day in adolescents) reduces the number of painful and swollen joints, pain intensity, and ESR. 95-98 The response to sulfasalazine is usually good, and most patients enter remission after 3 to 6 months of therapy. This period corresponds, however, to the natural history of the disease, and in many trials, the 6-month placebo response equals that of sulfasalazine. It is advisable to sustain the same dose of the drug (or in some cases a lower dose) through an additional period of 3 to 6 months after remission has been achieved to avoid a flare. The frequency of adverse events with sulfasalazine ranges from 10% to 20%. These mainly consist of dyspepsia or slight and transitory increases of aspartate transaminase and alanine aminotransferase levels. Rarely, toxicity necessitates sulfasalazine withdrawal. Some beneficial effect of the drug has also been observed in patients with iritis and skin manifestations such as keratoderma blennorrhagicum. There is a limited experience with the use of methotrexate in any of the spondyloarthropathies 99 and only anecdotal reports of good responses to methotrexate at weekly doses ranging from 7.5 to 15 mg in patients with reactive arthritis. Our experience with this drug in treating reactive arthritis has not been as
30
REACTIVE ARTHRITIS
611
favorable. It is possible that the dose of methotrexate needed to reduce disease activity in children with reactive arthritis is greater than that used for juvenile idiopathic arthritis. In contrast, its effects on iritis and keratoderma blennorrhagicum in children have been satisfactory. Iritis usually responds to topical or systemic glucocorticoids, but severe, resistant ocular inflammation occasionally reqUires other immunosuppressive drugs. No antibiotic regimen has been clearly efficacious in reactive arthritis. Data from double-blind and open comparative trials of various tetracycline derivatives (except for one using lymecycline) or ciprofloxacin indicate no significant differences compared with placebo during the short term and long term. A double-blind trial comparing lymecycline and placebo1°O demonstrated a significant difference between groups with regard to the time needed to recover from arthritis in patients with Chlamydia-related but not enteritis-related reactive arthritis. Antibiotic treatment, however, did not change the natural history of reactive arthritis in these patients 101 It has been suggested that amoxicillin alone or in combination with clavulanic acid may be useful for chiidren. 102
The use of tumor necrosis factor-a (TNF-a) blockers, specifically infliximab, has been beneficial in adults with reactive arthritis. 103 Although no data are yet available for children with reactive arthritis, TNF-a blockers may prove to be useful. 104
Physical Therapy and Rehabilitation In the acute inflammatory phase, treatment of reactive arthritis is similar to that for other forms of chronic arthritis. Rest, ice or hot packs, and aids for walking may be useful. For children with plantar fasciitis, custom-made insoles relieve pain caused by enthesitis at the heel and metatarsal heads and help preserve the longitudinal arch of the foot. The use of night resting splints helps avoid joint contractures associated with tendonitis or tenosynovitis. Active and passive stretching of joints, as well as muscle strengthening, should be prescribed when inflammation is being controlled and pain permits. Children with chronic and recurrent reactive arthritis tend to develop fibrous ankylosis first, followed by bone ankylosis of the midtarsal joints and subluxation of the metatarsophalangeal joints, and they therefore require special attention to insoles and shoes. Knee, hip, and axial disease benefits from activities such as biking and swimming.
Orthopedic Surgery Arthroscopic synovectomy is potentially beneficial for children with recurrent synovitis of the knee or small joints of the hands and feet. Early soft tissue release of contractures at the hip, knee and at the metatarsophalangeal and interphalangeal joints increases functional capacity and may reduce the risk of severe impairment thereafter. Adolescents with severe hip or knee disease may require arthroplasties and joint replacements in the long term.
COURSE OF THE DISEASE AND PROGNOSIS The course of disease in children with reactive arthritis varies. Most children have only a single episode of monarthritis or oligoarthritis. Such may typify the case
612
C HAP T E R
30
REACTIVE ARTHRITIS
in reactive arthritis triggered by Yersinia JO ,21,3 1 ,79 or Campylobacter. 23 Others have recurrent episodes of oligoarthritis or an extended form of disease affecting multiple joints and entheses that may account for most of those attending specialized clinics. Although remission may still occur in some patients, many others have disease that evolves into an identifiable spondyloarthropathy with sacroiliitis. Very few data indicate which factors are likely to influence the outcome of reactive arthritis or undifferentiated spondyloarthropathy in children. It is a clinical impression that most children with reactive arthritis recover in a matter of several months; however, a few eventually develop chronic spondyloarthropathy. There are unfortunately no reports of long-term outcome of reactive arthritis in children. Children with reactive arthritis (including Yersinia- and Salmonella-triggered reactive arthritis) who are positive for HLA-B27 have more severe involvement. 21 ,24,34 ,76 Extra-articular disease, including iridocyclitis and Reiter's syndrome, occurs more frequently among children with reactive arthritis who are B27 positive. In one report, three of five HLAB27-positive children with Salmonella-triggered reactive arthritis developed psoriasis.77 The number of joints involved at onset and systemic features such as fever or anemia, the number of episodes of disease activity in a given period, and their duration could also influence the outcome of reactive arthritis. The specific agent responsible for the infection may also play a role in determining ultimate outcome. For example, the clinical prognosis of Chlamydia- or Yersinia-triggered reactive arthritis is less severe than that described after Shigella or Salmonella infection. Whether this is a direct influence of the infectious agent or represents different frequencies of association with HLA-B27 is uncertain,
REFERENCES 1. Aho K, Leirisalo-Repo M, Repo H: Reactive anhritis, Clin Rheum Dis 11: 25--40. 1985. 2. Toivanen A: Reactive arthritis, In Klippel JH. Dieppe P (eds): Rheumatology. London. Mosby. 1994, pp 4,9,1-4.9,8. 3. Keat A: Reiter's syndrome and reactive anhritis in perspective. N Engl J Med 309: 1606--1615, 1983. 4. Kingsley G, Sieper J: Third international workshop on reactive arthritis. An overview. Ann Rheum Dis 55: 564--584, 1996. 5. Pacheco-Tena C, Burgos-Vargas R, V:l.zquez-Mellado J, et al: A proposal for the classification of patients for clinical and experimental studies on reactive arthritis, J Rheumatol 26: 1338-1346, 1999. 6. Sieper J, Rudwaleit M, Braun J, et al. Diagnosing reactive arthritis: role of clinical setting in the value of serologic and microbiologic assays. Arthritis Rheum 46: 319-327, 2002. 7. Leino R, Makela AL, Tiilikainen A, et al: Yerslnla arthritis in children. Scand J Rheumatol 9: 245-249, 1980. 8. Hoogkamp-Korstanje JA. Stolk-Engelaar VM: Yerslnla enterocoltttca infection in children. Pediatr Infect Dis J 14: 771-775, 1995. 9. Rudwaleit M, Richter S, Braun J, et al. Low incidence of reactive arthritis in children following a salmonella outbreak. Ann Rheum Dis 60:1055-1057, 2001. 10. Denardo BA, Tucker LB, Mlller LC, Schaller JG: Demography of a regional pediatric rheumatology patient population. J Rheumatol 21: 1553-1561, 1994. 11. Bowyer S, Roettcher P, for the Pediatric Rheumatology Database Research Group: Pediatric Rheumatology Clinic populations in the United States: results of a 3 year survey. J Rheumatol 23: 1968-1974, 1996. 12. Symmons DPM, Jones M, Osborne J, et al: Pediatric rheumatology in the United Kingdom: data from the British Pediatric Rheumatology Group National Diagnostic Register. J Rheumatol 23: 1975-1980, 1996.
13. Malleson PN, Fung MY, Rosenberg AM, for the Canadian Pediatric Rheumatology Association: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry. J Rheumatol 23: 1981-1987, 1996. 14. Rosenberg AM: Analysis of a pediatric rheumatology clinic population. J Rheumatol 17: 827--fl30, 1990. 15. Gare BA, Fasth A: The natural history of juvenile chronic arthritis: a population based cohort study. I. Onset and disease process. J Rheumatol 22: 295-307, 1995. 16. Oen KG, Cheang M: Epidemiology of chronic arthritis in childhood. Semin Arthritis Rheum 26: 575-591, 1996. 17. Kiessling U, Doring E, Listing .I, et al: Incidence and prevalence of juvenile chronic anhritis in East Berlin, 1980--1988. .I Rheumatol25: 1837-1843, 1998. 18. Ozen S, Karaaslan Y, Ozdemir 0, et al: Prevalence of juvenile chronic arthritis and familial Mediterranean fever in Turkey: a field study. .I Rheumatol 25: 2445-2449. 1998. 19. Taccetti G, Trapani S. Ermini M, Falcini F: Reactive arthritis triggered by Yerslnla enterocoltttca: a review of 18 pediatric cases. Clin Exp Rheumatol 12: 681-684, 1994. 20. Johnsen K, Ostensen M, Melbye AC, Melby K: HLA-B27-negative arthritis related to Campylobacter jejunt enteritis in three children and two adulls. Acta Med Scand 214: 165-168, 1983. 21. Artamonov VA, Akhmadi S, Polianskaia IS: The clinical and immunogenetic characteristics of reactive arthritis in children, Ter Arkh 63: 22-24, 1991. 22. Tuokko .I, Koskinen S, Westman P, et al: Tumour necrosis factor microsatellites in reactive arthritis. BrJ Rheumatol 37: 1203-1206, 1998. 23. Tuokko .I, Pushnova E, Yli-Kerttula U, et al: TAP2 alleles in inflammatory arthritis. Scand J Rheumatol 27: 225-229, 1998. 24. Czonka GW: The course of Reiter's syndrome. Br MedJ 1: 1088-1090, 1958. 25. Florman AL, Goldstein HM: Arthritis, conjunctivitis and urethritis (so-called Reiter's syndrome) in a 4-year-old boy. .I Pediatr 33: 172-177, 1948. 26. Davies NE, Haverty JR, Boatwright M: Reiter's disease associated with shigellosis. South Med J 62: 1011-1014, 1969. 27. Singsen BH, Bernstein BH, Koster-King KG, et al: Reiter's syndrome in childhood. Arthritis Rheum 20 (Supp\): 402-407, 1977. 28. Russell AS: Reiter's syndrome in children following infection with Yersinia enterocolitica and shigella. Arthritis Rheum 20 (Supp\): 471-472, 1977. 29. Iveson JMI, Nanda BS, Hancock JAH, et al: Reiter's disease in three boys. Ann Rheum Dis 34: 364--368, 1975. 30. Jacobs AG: A case of Reiter's syndrome in childhood. Br MedJ 2: 155, 1961. 31. Rosenberg AM, Petty RE: Reiter's disease in children. Am .I Dis Child 133: 394--398, 1979. 32. Cimolai N, Malleson P, Thomas E, Middleton PJ: Mycoplasma pneumotliae associated anhropathy: confirmation of the association by determination of the antipolypeptide IgM response. .I Rheumatol 16: 1150--1152, 1989. 33. Hannu T, Puolakkainen M, Leirisalo-Repo M: Chlamydia pneumoniae as a triggering infection in reactive arthritis. Rheumatology (Oxf) 38: 411-414, 1999, 34. Cron RQ, Gordon PV: Reactive arthritis to Clostridium dijficile in a child. West J Med 166: 419-421, 1997. 35. Kocar IH, Caliskaner Z, Pay S, Turan M: Clostridium dlffictle infection in patients with reactive arthritis of undetermined etiology. Scand .I Rheumatol 27: 357-362, 1998. 36. Letts M, Davidson D, Lalonde F: SynOVitis secondary to giardiasis in children. Am .I Orthop 27: 451-454, 1998. 37. Cron RQ, Sherry DD: Reiter's syndrome associated with cryptosporidial gastroenteritis. .I Rheumatol 22: 1962-1963, 1995. 38. Ikeda M, Yu DT: The pathogenesis of HLA-B27 althritis: role of HLA-B27 in bacterial defense. Am .I Med Sci 316: 257-263, 1998. 39. Wordsworth P, Brown M: HLA-B27, ankylosing spondylitis and the spondyloarthropathies. In Calin A, Taurog JD (eds): The Spondyloarthritides. Oxford, England, Oxford University Press, 1998, p 179. 40. Taurog JD: HLA-B27 subtypes, disease susceptibility, and peptide binding. In Calin A, Taurog JD (eds): The Spondyloanhritides. Oxford. England, Oxford University Press, 1998, p 267. 41. Wordsworth P: Genes in the spondyloanhropathies. Rheum Dis Clin North Am 24: 845--fl63, 1998. 42. Benjamin R, Parham P: Guilt by association: HLA-B27 and ankylosing spondylitis. Immunol Today 11: 137-142, 1990. 43. Sieper .I, Braun J: Pathogenesis of spondyloarthropathies. Persistent bacterial antigen, autoimmunity, or both? Arthritis Rheum 38: 1547-1554, 1995. 44. Sieper .I, Braun J: Triggering mechanisms and T-cell responses in the spondyloarthropathies. In Calin A, Taurog JD (eds): The Spondyloarthril.ides. Oxford, England, Oxford University Press, 1998, p 195. 45. Hermann E, Yu DTY, Meyer zum B(jschenfelde K-H, Fleischer B: HLA-B27 restricted CD8 T cells derived from synovial fluids of patients with reactive arthritis and ankylosing spondylitis. Lancet 342: 646--650, 1993. 46. Sieper .I, Kingsley G, Marker-Hermann E: Aetiological agents and immune mechanisms in emerogenic reactive arthritis. Baillieres Clin Rheumatol 10: 105-121, 1996. 47. Kingsley G, Panayi GS: Antigenic responses in reactive arthritis. Rheum Dis Clin Nonh Am 18: 4966, 1992. 48. Marker-Hermann E, Hohler T: Pathogenesis of human leukocyte antigen B27-positive arthritis: information from clinical materials. Rheum Dis Clin North Am 24: 865--fl81, 1998,
C HAP T E R 49. D~venport MP: The promiscuous B27 hypothesis. Lancet 346: 500-501, 1995. 50. S<;ofield RH, Warren WL, Koelsch G, Harley JB: A hypothesis for the HLAB.;!7 immune dysregulation in spondyloarthropathy: contributions from e~teric organism, B27 structure peptides bound by B27, and convergent evolution. Proc Natl Acad Sci USA 90: 9330-9334, 1993. 51. D~ngoria, NS, Delay, ML, Kingsbury, OJ et al: HLA-B27 misfolding is associated with aberrant intermolecular disulfide bond formation (dimerization) in. tlle endoplasmic reticulum. ] Bioi Chern 277: 23459-23468, 2002. 52. B\lrgos-Vargas R, Howard A, Ansell BM: Antibodies to peptidoglycan in juvenile onset ankylosing spondylitis and pauciarticular onset juvenile arthritis associated with chronic iridocyclitis. J Rheumatol 13: 760-762. 1986, 53. Life P, Hassell A, Williams K, et al: Responses to gram negative enteric bacterial antigens by synovial T cells from patients with juvenile chronic arthritis: recognition of heat shock protein Hsp60. J Rheumatol 20: 1388--1396, 1993. 54. Grantors K: Do bacterial antigens cause reactive arthritis? Rheum Dis Clin N~)rth Am 18: 37-48, 1992. 55. Wuorela M, Granfors K: Infectious agents as triggers of reactive arthritis. Am J Med Sci 316: 264--270, 1998. 56, Uksila J, Toivanen P, Granfors K: Enteric infection and arthritis: Bacteriological aspects. In Collin A, Taurog JD (eds): The Spondyloarthritides. Oxford, England, Onord University Press, 1998, p 167. 57. Kapasi K, Inman RD: HLA B27 expression modulates gram-negative bacterial invasion into transfected L cells. J Immunol 148: 3554--3559, 1992. 58, Kapasi K, Inman RD: MEl epitope of HLA B27 confers class I-mediated mpdulation of gram-negative bacterial invasion. J Immunol 153: 833-840. 1994. 59. Oltiz-A1varez 0, Yu DT, Petty RE, Finlay BB: HLA-B27 does not affect invasi'm of arthritogenic bacteria into human cells. J Rheumatol 25: 1765-1771, 1998. 60. Huppertz H-I, Heesemann J: The influence of HLA B27 and interferon-yon the invasion and persistence of Yersinia in primary human fibroblasts. Med Microbiol Immuno! 185: 163--170, 1996. 61. La,itio P, Virtala M, Salmi M, et al: HLA-B27 modules intracellular survival of Salmonella enteritidis in human monocytic cells. Eur J Immunol 27: 1331-1338, 1997. 62. Granfors K: Host-microbe interaction in reactive arthritis: does HLA-B27 have a direct effect? J Rheumatol 25: 1659-1661, 1998. 63. SChumacher HR, Magge S, Cherian PV, et al: Light and electron microscopic stlildies on synovial membrane in Reiter's syndrome: inlmunocytochemical identification of cl1lamydial antigens in patients with early disease. Arthritis Rheum 31: 937-946, 1988, 64. G"mfors K, Jalkanen S, von Essen R, et al: Yersinia antigens in synovial fluid cells from patients with reactive arthritis. N Eng! J Med 320: 216-221, 1989. 65. Hammer M, Zeidler H, Klinlsa S, et al: Yersinia enterocolitica in the synovial membrane of patients with yersinia induced arthritis. Arthritis Rheum 33: 1795-1800, 1990. 66. G~anfors K, Jalkanen S, Lindberg AA, et aI: Salmonella lipopolysaccharide in the synOVial cells from patients with reactive arthritis. Lancet 335: 685-688, 1990. 67. Taylor-Robinson 0, Gilroy CB, Thomas BJ, Keat AC: Detection of Chlamydia trnchomatis DNA in joints of reactive arthritis patients by polymerase chain reaction. Lancet 340: 81-82, 1992. 68. Garcia CO, Paira S, Burgos-Vargas R, et aI: Detection of Salmonella DNA in synovial membrane and synovial fluid from Latin American patients with reactive arthritis using the polymerase chain reaction. Arthritis Rheum 39 (S1;Jpp1): S185, 1996. 69. Bmun J, Tuszewski M, Eggens U, et al: Nested polymerase chain reaction stmtegy sinlultaneously targeting DNA sequences of multiple bacterial species in inflammatory joint diseases. I. Screening of synovial fluid samples of patients with spondyloarthropathies and other arthritides. J Rheumatol 24: 1092-1100, 1997. 70. Pacheco-Tena C, Alvarado de la Barrera C, LOpeZ-Vidal Y, et al: Bacterial DNA in synOVial fluid cells of patients with juvenile onset spondyloarthropathies. Rheumatology. 40: 920-927, 2001. 71. Cuchacovich R, Japa S, Huang WQ, et al. Detection of bacterial DNA in Latin American patients with reactive arthritis by polymerase chain reaction and sequencing analysis. J Rheumatol 29: 1426-1429, 2002. 72. Cox C), Kempsell KE, Gaston JS: Investigation of infectious agents associated with arthritis by reverse transcription PCR of bacterial rRNA. Arthritis Res Ther 5: RI-R8, 2003. 73. Carroll WL. Balistreri WF, Brilli R, et .II: Spectrum of Salmonella-associated arthritis. Pediatrics 68: 717-720, 1981.
30
REACTIVE ARTHRITIS
613
74. Kanakoudi-Tsakalidou F, Pardalos G, Prastidou-Gertsi P, et al: Persistent or severe course of reactive arthritis following Salmonella enteritidis infection. Scand J Rheumatol 27: 431-434, 1998. 75, lakovleva AA, Mitchenko AF, lushchenko GV, et aI: Yersinia arthritis in children. Vestn Akad Med Nauk SSSR 6: 71-75, 1989. 76. Jezequel C, Prigent]Y, Loiseau-Corvez MN, et al: Reactive arthritis caused by Yersinia in children. Report of 4 cases. Ann Pediatr (Paris) 38: 318--322, 1991. 77. Hannu T, Kauppi M, Tuomala M, et a1: Reactive arthritis following and outbreak of Campylobacter jejuni infection. J Rheumatol 31: 528--530, 2004, 78. Hussein A: Spectrum of post-enteric reactive arthritis in childhood. Monatsschr Kinderheilkd 135: 93--98, 1987. 79. CUllica R), Scheines EJ, Garay SM, et al: Juvenile onset Reiter's syndromea retrospective study of 26 patients. Clin Exp Rheumatol 10: 285-288, 1992. 80. Lockie GN, Hunder GG: Reiter's syndrome in children: a case report and review. Arthritis Rheum 14: 767-772, 1971. 81. Friis J: Reiter's disease with childhood onset having special reference to HLA B27. Scand J Rheumatol 9: 250-252, 1980. 82. Huppertz HI, Sandhage K: Reactive arthritis due to Salmonella enteritidis complicated by carditis. Acta Paediatr 83: 1230-1231, 1994. 83. Zivony 0, Nocton J, Wortmann 0, Esterly N: Juvenile Reiter's syndrome: a report of four cases. J Am Acad Dermatol 38: 32-32, 1998. 84, Saari KM, Milici M, Piliviinsalo T, et al: Acute anterior uveitis and conjunctivitis following Yersinia infection in children. Int Ophthalmol 9: 237-241, 1986, 85. Fischel JD, Lipton J: Acute anterior uveitis in juvenile Reiter's syndrome. Clin Rheumatol 15: 83-85, 1996. 86. Huppertz HI, Karch H, Heesemann J: Diagnostic value of synovial fluid analysis in children with reactive arthritis. Rheumatol Int 15: 167-170, 1995. 87. Azouz EM, Duffy CM: Juvenile spondyloarthropathies: clinical manifestations and medical inlaging. Skel Radiol 24: 399-408, 1995. 88. De Keyser F, Elewaut 0, De Vos M, et al: Bowel inflammation and the spondyloarthropathies. Rheum Dis Clin North Am 24: 785-813, 1998. 89, Bollow M, Biedermann T, Kannenberg J, et al: Use of dynamic magnetic resonance imaging to detect sacroiliitis in HLA-B27-positive and negative children with juvenile arthritides. J Rheumatol 25: 556-564, 1998. 90. Bauman C, Cron RQ, Sherry DO, FrancL, )S: Reiter syndrome initially misdiagnosed as Kawasaki disease. J Pediatr 128: 366-369. 1996. 91. Duffy CM, Arsenault L, Duffy KN, et al: The Juvenile Arthritis Quality of life Questionnaire: development of a new responsive index for juvenile rheumatoid arthritis and juvenile spondyloarthropathies. J Rheumatol 24: 738--746, 1997. 92. Tucker LB, De Nardo BA, Abetz LN, et .II: The Childhood Arthritis Health Profile (CARP): validity and reliability of the condition specific scales. Arthritis Rheum 38: S183, 1995. 93. Mielants H, Veys EM, Joos R, et .II: Late onset pauciarticular juvenile chronic arthritis. Relation to gut inflammation. J Rheumatol 14: 459-465, 1987. 94. Mielants H, Veys EM, Cuvelier C, et al: Gut inflammation in children with late onset pauciarticular juvenile chronic arthritis and evolution to adult spondyloarthropathy-a prospective srudy. J Rheumatol 20: 1567-1572, 1993. 95. Joos R, Veys EM, Mielants H, et al: Sulfasalazine treatment in juvenile chronic arthritis: an open srudy. J Rheumatol 18: 880-884, 1991. 96. Suschke H]: Treatment of juvenile spondyloarthritis and reactive arthritis with sulfasalazine. Monatsschr Kinderheilkd 140: 658-660, 1992. 97, Job-DesIandre C, Menkes C): Treatment of juvenile spondyloarthropathies with sulfasalazine. Rev Rhum Ed Fr 60: 489-491, 1993. 98. BurgOS-Vargas R, Vazquez-Mellado J, Pacheco-Tena C, et .II: A 26-week randomized, double blind, placebo-controlled exploratory study of sulfasalazine in juvenile onset spondyloarthropathies. Ann Rheum Dis 61: 941-942, 2002. 99. Creemers MC, Franssen MHJ, van de PUlle LB, et al: Methotrexate in severe ankylosing spondylitis: an open study. .J Rheumatol 22: 1104--1107. 1995. 100. Lauhio A, Leirisalo-Repo M. Lahdevirta J, et al: Double-blind, placebo-controlled srudy of three-month treatment with lymecycline in reactive arthritis, with special reference to Chlamydia arthritis. Arthritis Rheum 34: 6-14, 1991. 101. Laasila K, Laasonen L, Leirisalo-Repo M: Antibiotic treatment and long term prognosis of reactive arthritis. Ann Rheum Dis 62: 655-658, 2003. 102. Astrauskiene 0: Efficacy of empirically prescribed amoxicillin and amoxicillin + clavulanic acid in children's reactive arthritis: a randomised trial. Clin Exp Rheumatol 21: 515-521, 2003. 103. am KS, Niinisalo H, Korpilahde T, VirolainenJ: Treatment of reactive arthritis with inflixinlab. Scand J Rheumatol 32: 122-124, 2003. 104. Burgos-Vargas R: Juvenile onset spondyloarthropathies: therapeutic aspects. Arch Dis Child. 88: 312-318, 2003.
C HAP T E R
31
ACUTE RHEUMATIC FEVER AND POST-STREPTOCOCCAL REACTIVE ARTHRITIS Ella M. Ayoub and Khaled Alsaeld
'Jf ACUTE RHEUMATIC FEVER Definition and Classification Acute rheumatic fever (ARF) is a rheumatic disease characterized by an inflammatory process that affects several organs of the body. It is one of the few rheumatic diseases for which the cause has been identifiedtonsillopharyngitis due to the group A /3-hemolytic Streptococcus pyogenes. The streptococcal infection and the onset of the clinical manifestations of ARF are separated by a period of latency of 2 to 3 weeks. During this time, the patient is asymptomatic. The clinical presentations include arthritis, carditis, chorea, a characteristic rash, and subcutaneous nodules. Arthritis is the most common but least specific of these manifestations, whereas carditis is the most specific and serious. The pathologic process underlying the inflammatory reaction in the various organs is a diffuse vasculitis mediated by an immune reaction to the streptococcal infection. This nonpurulent complication of group A streptococcal disease can be prevented by appropriate treatment of the streptococcal pharyngitis.
Epidemiology Incidence and Prevalence ARF was prevalent worldwide until the middle part of the 20th century. The advent of industrialization and improved public hygiene in Western Europe and North America was associated with a sharp decline in the incidence of this disease. During the early part of the 20th century, incidence rates of 100 to 200 cases per 100,000 members of the general population were documented in the United States. 1 Although this rate still prevails in developing countries,2 current estimates of the incidence of ARF in children in the United States document a markedly lower incidence rate of 0.5 to 3 cases per 100,000 children. 3 Between 1985 and 1990, a marked 614
resurgence of the disease occurred in several areas of the United States.4-13 This dramatic reappearance of what had been an increasingly rare disease was followed by a persistently higher rate in the incidence of ARF in these geographic areas. I 4-16 However, the focal nature of these episodes has not significantly affected the overall prevalence of the disease in the United States.
Age at Onset and Sex Ratio The age-related incidence of ARF follows that of group A streptococcal pharyngitis and peaks between the ages of 6 and 15 years. ARF is rarely encountered in the United States in children younger than 5 years. 17- 19 Among adults at high risk for streptococcal pharyngitis, such as military recruits and persons working in crowded settings, the incidence of the disease is higher. There is no difference in the incidence of ARF between males and females.
Geographic and Racial Distribution ARF used to be considered a disease of temperate climates but is now more common in countries with tropical climates, particularly in developing countries. In the United States, the highest seasonal incidence is in the spring, follOWing the peak season of streptococcal pharyngitis in the winter. In other countries, a season of peak frequency is less well defined. Despite the decline of ARF in industrialized countries, its prevalence in developing areas of the world remains very high. About 6 million children suffer from rheumatic heart disease in India and in Sri Lanka, where the annual incidence of the disease is estimated at 100 to 1SO cases per 100,000 members of the general population. 3,2o,21 Factors that have been invoked in explaining the decreased incidence of the disease in the United States include less crowding in homes and schools and the increased availability of health care to children. 22,23 Observations during the recent resurgence of ARF suggest that these factors may not be of major importance because this disease was now occurring primarily
C HAP T E R
31
ACUTE RHEUMATIC FEVER
in children from middle- to high-income families with ready access to medical care. 4 Differences in the incidence of ARF among racial and ethnic groups have been described. In New Zealand, the disease is more common among the Maori population compared with local non-Maoris of similar socioeconomic status. 24 ARF in the United States is more prevalent among African Americans and Hispanics than among whites. 3 Although genetic factors can account for these racial and ethnic differences, environmental factors may also be instrumental in explaining these observations. 2
Etiology and Pathogenesis ARF is a complication of a group A streptococcal tonsillopharyngitis in a predisposed human host; streptococcal pyoderma does not lead to this nonpurulent complication. 25 There is no experimental model for this disease. Specific factors that influence its evolution include the characteristics of the etiologic organism, the site of the streptococcal infection, and a genetic predisposition of the host (Fig. 31-1). Less than 2% to 3% of previously healthy persons who acquire streptococcal pharyngitis develop ARF. This complication can be prevented by prompt identification and treatment of the streptococcal infection.
Etiologic Agent ~-Hemolytic streptococci have been divided into 20 serogroups (A to Hand K to V) by Lancefield26 on the basis of immunochemical differences in their cell-wall
PAmOGENESIS OF RHEUMATIC FEVER Group A Streptococcus Rheumatogenic Strains Serotypes M3, MI8 Mucoid Colonies
Tinue/Organ Inflammation Joints
Heart
Brain
Vascular
Connective Tissue
Susceptible Host Positive for HLA-OR 4,2,1,3,7 ORBI*16 allele Allotype 08/17
Immune Reaction Cross-Reactive Antibody and/or Cell-Mediated Immunity
• ~ 31-1 Interactions between the group A Streptococcus and the human host that lead to acute rheumatic fever. HLA, human leukocyte antigen. (Adapted from Ayoub EM: Acute rheumatic fever. In Emmanouilides Ge, Riem~mschneiderTA, Allen HD, et al reds): Moss and Adams' Heart Disease in Infants, Children, and Adolescents, Induding the Fetus and Young Adult,VollI, 5th ed. Baltimore, Williams & Wilkins, 1995, p 1400.)
& POST-STREPTOCOCCAL
615
polysaccharide. The group A Streptococcus is the most common bacterial pathogen that is associated with tonsillopharyngitis, and it is the only member of these groups of streptococci that can initiate ARF. Several cellular components and extracellular products produced by this streptococcus in vivo and in vitro have been identified. The streptococcal bacterium consists of a cytoplasm enclosed in a membrane composed predominantly of lipoproteins. This structure is surrounded by a rigid cell wall made up of three components. The primary component is a peptidoglycan that imparts rigidity to the cell wall. A complex of this component and the cell-wall polysaccharide elicits arthritis and a recurrent nodular reaction when injected into the skin of experimental animals. 27- 29 Integrated into the peptidoglycan is the cell-wall polysaccharide or group-specific carbohydrate whose immunochemical structure determines the serogroup specificity. This polysaccharide has been reported to share antigenic determinants with a glycopeptide present in mitral valve tissue. 3o Traversing through and extending outside the cell wall as hairlike fimbriae is the M protein, part of a mosaic that also includes the Rand T proteins. The M protein is a coiled protein with an a-helical structure consisting of a free, distal, hypervariable aminoterminus and a proximal carboxylterminus anchored to the cell wallY This protein is the type-specific antigen of the group A Streptococcus. About 100 M proteins have been identified by differences in immunochemical composition of the variable aminoterminus. A major biologic property of the M protein resides in its capacity to inhibit phagocytosis of the streptococcus, a property that is neutralized by antibody to the aminoterminal region. Immunity to group A streptococcal infections is therefore type specific, predicated on formation of antibodies to the various M proteins. Additional attributes include the association of certain serotypes with potential pathogenicity and virulence. Data procured during a resurgence of ARF confirmed that serotypes 3 and 18, particularly strains that produced mucoid colonies when cultured on blood agar, were primarily associated with the disease. 32,33 These two serotypes and the M1 serotype were also associated with severe, invasive group A streptococcal disease, including the streptococcal toxic shock syndrome. 34 Studies have indicated that bacterial strains that have conserved parts of the carboxylterminal portion of the M-protein molecule exposed on their cell surface (class I strains) were associated with ARF, whereas strains that did not have this characteristic (class II) were not. 35 It was reported that phages and phage-like elements were the source for variation in the genome of an M18 isolate, recovered from a patient with ARF, and an Ml strain. 36 The pathogenetic importance of the M proteins is supported by data indicating that several epitopes of the M-protein molecule cross-react antigenically with human myocardium, myosin, and brain tissue, ostensibly leading to tissue inflammation. 37,38 M protein also functions as a "superantigen."39 These findings indicate that this streptococcal molecule can induce an inflammatory response in certain tissues by eliciting "autoimmune" antibodies and tissue inflammation by nonspecific stimulation of cell-mediated immunity as a superantigen. The cellular component of the group A streptococci that has been implicated in the pathogenesis of arthritis is the hyaluronate capsule. Like the M protein, this moiety appears to carry epitopes that elicit antibodies that cross-react with human cartilage and synovial hyaluronate. 4o Some studies have documented that components of the M3 and M18 epitopes aggregate type IV collagen, a component of the human basement membrane. This reaction is effected in M3 strains by the production of a collagen-binding factor. M18 strains bind collagen through
616
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
the hyaluronic acid capsule. Patients with ARF have higher levels of anti-collagen IV antibodies than controls41 j mice immunized with recombinant M3 protein produced anti-collagen antibodies. In addition to the cellular components, extracellular products of the group A streptococci have important biologic activities and are of practical value in the diagnosis of group A streptococcal infections and their nonpurulent complications. Most of these products are proteins with enzymatic properties, and they possess specific biologic and antigenic activity. The streptococcal pyrogenic exotoxins (SPEs) A, B, C, and F (Le., the mitogenic factor [MF)) and SSA) are of particular interest because they act as superantigens that induce proliferation of T lymphocytes in vitro and the synthesis and release of several lymphokines in vivo. 37-43 This biologic activity reflects the ability of SPEs to bind simultaneously to class II major histocompatibility antigens of antigen-presenting cells and to the V~ region of the T-cell receptor. Production of these exotoxins is associated in vivo with a febrile response, alteration of membrane permeability, and enhancement of susceptibility to endotoxin-induced lethal shock,44 Selective activation of lymphocytes has been ascribed to different SPEs. SPE A activates T cells bearing T-cell receptor ~-chain segments V~8, V~12, and V~14, whereas SPE B activates T cells bearing segments V~2 and V~8. 45 SPE B has been identified as a cysteine protease that inhibits phagocytosis and enhances dissemination of the organism in vivo. It also induces apoptosis of phagocytic cells. 46 The frequencies of the spe genes and their expression vary among group A streptococci; speA is found in 45% of strains, speB in almost all strains, and speC in 30% of strains. SPE A is expressed by 43% and SPE B by 76% of strains. 34,47,4B The frequencies of the speA genes and their products are similar among M1 and M3 serotypesY The association of certain serotypes with various clinical manifestations of streptococcal infections, such as toxic shock syndrome, has been ascribed to the capacity of the infecting strain to produce one of the SPES.34,45,47 However, the ubiquity of the production of these toxins makes confirmation of the specificity of these associations questionable. 48
'~I
Streptococcal Antigen
AnlilJen~
AnUbody Test
Extracellular Produd Streptolysin 0 Streptokinase Hyaluronidase Deoxyribonuclease B Nicotinamide adenine dinucleotidase Multiple antigens
Antistreptolysin 0 (ASO) Antistreptokinase Antihyaluronidase Anti-DNAse B Anti-NADase Streptozyme
Cellular Component M protein Group-speCific polysaccharide
Type-speCific antibody Anti-A-carbohydrate
Adapted from Ayoub EM: Streptococcal antibody tests in rheumatic fever. Clin Immunol Newsletter 3: 107-111, 1982.
the ASO test. The other tests, which are no longer readily available, and the streptozyme test, which was widely used at one time, lacked standardization and reproducibility and should not be relied on for evidence of antecedent group A streptococcal infection.50 The pattern of the antibody response to the streptococcal antigens is illustrated in Figure 31-2. Antibodies peak approximately 3 weeks after the acute infection. Because of the period of latency between the infection and the onset of the clinical manifestations of ARF, serum obtained at the time of clinical presentation should document the necessary evidence for antecedent group A streptococcal infection. However, as outlined in Table 31-2, only about 85% of patients with ARF mount an ASO response. Another streptococcal antibody test, such as anti-DNAse B, can provide evidence for an antecedent streptococcal infection in patients in whom an ASO response has not been diagnostic.
600 500
Streptococcal AntlbodyTests The specific antigenicity of most of the streptococcal extracellular products led to the establishment of antibody tests for these products. These tests are used to confirm evidence of a group A streptococcal infection, primarily in patients with ARF and glomerulonephritis. The first and still most universally used is the antistreptolysin 0 (ASO) test, which was designed by Todd49 to measure neutralizing antibodies to purified streptolysin 0 in patients with scarlet fever and ARF. This test proved helpful in providing evidence for antecedent group A streptococcal infection, particularly when throat cultures were negative. Subsequently, tests were developed to assay for antibodies to other streptococcal antigens (Table 31-1). The anti-DNAse B test, which assays for antibodies to the most ubiquitous of four deoxyribonuclease isozymes produced by the group A streptococcus (A, B, C, D), proved to be as reliable and reproducible as
lAin I 31 1 (,IOUp A 511"plo(O« al alld (OIH>~pondin<J Anlibody le~ls
f!? 400
Q)
F>.
300
+= c::
200
"8 .0 «
100 2 3 4 5 6 7 8 Interval Following Acute Infection (months)
9
• Figure J1-2 Pattern of antibody response to the extracellular antigens of the group A Streptococcus after tonsillopharyngeal infection in humans. (From Ayoub EM: Streptococcal antibody tests in rheumatic fever. elin Immunol Newsletter 3: 107-111, 1982.)
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
617
1'1 .. TABLE
31-2 Frequency of Patients with Acute Rheumatic Fever with Elc'vated Titers of ASO or anti-DNase B
-
Group
ASO
Antl-DNAse B
ASO and antI-DNAse B
Normal controls Acute rheumatic fever Sydenham's chorea (isolated)
19% 83% 67%
19"/0 82% 40%
30% 92% 80%
ASO, antistreptolysin 0; DNase B, deoxyribonuclease B. Adapted from Ayoub EM, Wannamaker LW: Evaluation of the streptococcal deoxyribonuclease Band diphosphopyridine nucleotidase antibody tests in acute rheumatic fever and acute glomerulonephritis. Pediatrics 29: 527-538, 1962, and from Ayoub EM, Wannamaker LW: Streptococcal antibody titers in Sydenham's chorea. Pediatrics 38: 946-956,
1966.
Tests for antibodies to the cell-wall components of group A streptococci are available but not widely used. Determination of type-specific antibody to the different M proteins is employed in epidemiologic studies to determine previous exposure or immunity to specific M serotypes. Testing for antibody to the group-specific carbohydrate is available in some laboratories. Because this antibody tends to persist for prolonged periods in patients with rheumatic valvular disease, it may help to confirm the rheumatic cause of mitral valve disease in a patient without a history of ARF.51-54
Mechanism ofTissue Injury Initial suggestions that tissue injury in ARF was caused by direct mvasion by the streptococcus or the effect of its extr;lcellular toxins were subsequently replaced by the theory that an immune mechanism was responsible for the inflammatory response in the affected organs. The potential role of an immunologic process as the cause of tissue mjury was predicated on the observation that the clinical manifestations of ARF occurred after a period of latency of about 3 weeks from the incitmg group A streptococcal infection. Evidence for involvement of an immune mechaniSm m pathogenesis was first advanced by Kaplan and coworkers. 55.56 These mvestigators and others described the presence of common antigenic determinants among the cellular components of the group A streptococci and myocardial tissues. Structures that share cross-reactive antigenic determinants included components of the M protem and myocardial sarcolemma,55-{jO cell-wall carbohydrate, and valvular glycoprotem,30 streptococcal protoplast mem. . . . . . 31-3 GroupA strePtococcal romponents and rorrtsponding human tissues that have been reported to exhibit immunologic cross-reactivity. (From Ayoub EM, Schiebler GL: Acute rheumatic fever. In Kelley VC led): Practice of Pediatrics, Vol 8. NewYork, Harper & Row, 1987, P7.)
brane and neuronal tissue of the subthalamic and caudate nuclei,61.62 and the hyaluronate capsule and articular cartilage. 4o Based on these studies, it was concluded that antibodies formed against these streptococcal antigens cross-reacted with the corresponding tissues and ostensibly led to inflammation m the heart, joints, and brain (Fig. 31-3).37.38 As attractive as the process of "antigenic mimicry" is in explaining the inflammatory reaction in ARF, there are several flaws in this hypothesis. The most compelling of these arguments is the presence of high levels of cross-reactive antibodies in the sera of patients who do not have any manifestations of acute carditis or arthritis. An alternative explanation was provided by subsequent studies that documented a potential role for cell-mediated immunity in inducing tissue damage. These studies confirmed that peripheral blood lymphocytes from patients with acute rheumatic carditis were cytotoxic to human myocardial cells in tissue culture. 63 Addition of plasma from the same patients abrogated this cytotoxic effect. The latter observation suggested that the cross-reactive antibodies elicited by group A streptococci had a protective rather than a detrimental effect on the host. Based on these arguments, the prevalent hypothesis for explaining tissue injury in this disease is that an immunologic mechanism involving the humoral or the cellular immune system may be responsible for tissue inflammation inARF.
Genetic Background Early postulates regarding the epidemiology of rheumatic fever suggested that persons who acqUired this disease
Ca~sule
Hyaluronic acid
Cell wall M-Proteln, M and M-AsSocla ted Protein
Groug Carbohy,grate N-acetyl-Qlucosamlne Rhannose
Protop-Iast Membrane Protein, Lipid, Glucose
• JOint-4(
t
Myocardium
.. Valvular ~ tissue Myocardial sarcolemma Subthalamic & Caudate Nudel
t
618
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
had a peculiar susceptibility to it. This postulate was based on the observation that 30% to 8oo!o of patients who had had ARF developed a recurrence of the disease after subsequent group A streptococcal pharyngitis, whereas only about 2% of normal persons would develop ARF after such an infection. 64 Studies by several investigators documented the familial occurrence of the disease. 65-67 Citing their studies, these investigators concluded that susceptibility to ARF is inherited as a single recessive gene. More substantial evidence for a genetic association was provided by Khanna and associates,68 who reported that a B-cell alloantigen, designated D8/17, was present in 99% of patients with ARF but in only 14% of normal controls, data that have been confirmed in subsequent studies. 69 Further support for the role of genetic factors in susceptibility was provided by studies on the association of this disease with inheritance of the major histocompatibility antigens (HLAs),7Q--78 The results of these investigations, summarized in Table 31-3, document a significant association of susceptibility with class II HLA antigens. These associations with rheumatic heart disease are more evident and consistent among clinically homogeneous patients. 79 Studies have documented an association between ARF and HLA class II alleles. These include an association with DRBl'1680 and DRBl'07. 81 Early investigators proposed that susceptibility to ARF was related to a state of hyper-reactivity to streptococcal antigens. Studies of hyper-responsiveness to a number of streptococcal and nonstreptococcal antigens suggested a hyperimmune response to streptococcal extraceIlular products, particularly streptolysin 0, although subsequent reports did not confirm these findings. 82 ,83 Later studies on the immune response to the group A streptococcal group-specific carbohydrate documented an unusual pattern of hyperimmune response to this antigen in patients with rheumatic valvular disease,5H4,84 This response was associated with inheritance of HLA-DR2 and HLA-DR4 antigens. 7o This finding is of particular relevance in view of data that indicate that the immune response to streptococcal ceIl-waIl antigen is under genetic control in experimental animals and humans. 85--ll7
• lABLE 31 3
Clinical Manifestations Arthritis, carditis, Sydenham's chorea, erythema marginatum, and subcutaneous nodules constitute the major clinical manifestations of ARF (Fig. 31-4). A patient may present with only one, two, or more of these manifestations, as well as with various degrees of severity of each. Although the severity and frequency of these manifestations vary considerably from patient to patient, their overall frequencies in various populations are similar (Table 31-4). Minor manifestations of ARF include fever, arthralgia, abnormal acute phase reactants, and a prolonged PR interval.
Arthritis Arthritis occurs in about 70% of patients. Although it is the most common of the major manifestations, it is relatively less specific than the others because it is encountered in such a large number of other rheumatic diseases. As such, it is the most common cause of a misdiagnosis of ARE Despite its lower specificity, the arthritis of ARF has characteristics that can help in its differentiation from that due to other causes. The arthritis affects primarily large joints, particularly the knees, ankles, wrists, and elbows. Small peripheral joints are only occasionally involved, and axial disease occurs rarely, if ever. The arthritis of ARF is characteristically migratory and additive; it is usually initially a monarthritis but can be polya.rticular. 88 Symptoms in an affected joint may resolve spontaneously within hours of onset, only to reappear in a different joint. The affected joint manifests the cardinal signs of inflammation with swelling, erythema, warmth, and pain. The latter symptom is the most prominent. It occurs at rest and is accentuated by passive or active movement of the joint. The severity of pain induces guarding of the joints, which may lead to a pseudoparalysis.
Carditis Cardiac inflammation develops in about 50% of the patients. The high frequency of this manifestation
RelJOrled Asso( idtions of HLA DR Antigens dnd Alleles With Rheunldti( Fever
Study
location
No. of Patients
Ethnldty
Ayoub et aFo
Florida, USA
Anastasiou-Nana et aJ71 ]hinghan et al n Rajapakse et aI73 Mahara j et aF4 Taneja et aF5 Guilherme et aF6 Ozkan et aln
Utah, USA New Delhi, India Riyadh, Saudi Arabia Durban, South Africa New Delhi, India San Paulo, Brazil Istanbul, Turkey
24 48 33 134 40 120 54 40 107
White African American White Indian Arab African American Indian Brazilian (mulano) Turkish
Weidebach et aF"
Sao Paulo, Brazil
24
Brazilian (mulatto)
Ahmed et al llO
Florida, USA
18
White
HLA·DR Antigen/Allele DRBl*16 DR2 DR4 DR3 DR4 DRI DQw2 DR7 DR3 DR7 DR16 DRw53 DRBl·16
Percent Positive Controls
Patients
32 23 32 26 12 3 32 26 23 33 34
63 54 52 50 65 13 63 58 49 57 83
4
15
Adapted from Ayoub EM: Rheumatic fever. In Rich RR, Fleisher TA, Schwartz BD, et al (eds): Clinical Immunology: Principles and Practice. 51. Louis, Mosby-Year Book, 1996. p 1134.
C HAP T E R
31
ACUTE RHEUMATIC FEVER
& POST-STREPTOCOCCAL
619
• FIgIre 31-4 Major manifestations of acute rheumatic fever. This diagram Illustrates the expected occurrence of each of the major manifestations of acute rheumatic fever. The relative duration in weeks is indicated on the abscissa.The maximum clinical activity of each finding is represented by the peak of the shaded area. The expected frequency of each clinical manifestation is represented by the relative height of each shaded area. Arthritis and carditis usually are manifestations of acute disease. (horea, although it may be an early manifestation, usually occurs about 3 months after the inciting episode of pharyngitis. It may be unaccompanied by other manifestations of the disease. Erythema marginatum is present for a longer period during and after the initial acute attack. This manifestation, although it is often associated with severe disease, is relatively uncommon in children.
reported from developing countries probably reflects a bias toward hospitalization of patients with severe heart disease. Carditis is the most common cause of morbidity and mortality. As with other manifestations, the severity of the carditis is highly variable. 89 In some patients, such as those with Sydenham's chorea, signs of carditis may be subtle, and cardiac involvement may be missed unless its diagnosis is pursued vigorously with echocardiographic examination.90 Other patients may present with acute pancarditis and severe, life-threatening congestive heart failure. Carditis usually occurs in tandem with other major manifestations, such as arthritis. If it is not present initially, carditis may follow arthritis within 1 week; the onset of carditis beyond this interval is rare. Inflammation of the heart in ARF usually involves the myocardium and endocardium. Pericarditis is a sign of pancarditis: involvement of all cardiac layers in the inflammatory process. It is an ominous development associated with a high mortality rate. Unlike its occurrence in other rheumatic diseases, isolated pericarditis is rare in patients with ARF. Myocarditis occurs during the initial stage of cardiac involvement; it is recognized clinically by the presence of tachycardia at rest in an afebrile patient and obliteration of the normal respiratory variation in heart rate. Myocarditis may be associated with heart block,91 cardiac arrhythmias, and a prolonged PR interval on electrocardiography. Endocarditis affects principally valvular tissue and leads to the hallmark lesion of rheumatic carditis, valvular insufficiency. The mitral valve is affected alone or in
,t._-
..I TABLE 31-4
conjunction with other valves in 94% of patients. Isolated mitral valve disease occurs in 65% of patients, isolated aortic disease in 6%, and simultaneous involvement of both valves in 29% of patients. The pulmonic and tricuspid valves are only occasionally affected. Mitral insufficiency or regurgitation is identified clinically by the presence on auscultation of a high-frequency, smooth, holosystolic, apical murmur. This murmur radiates to the left axilla and is best heard with the patient in a left lateral decubitus position. A mid- to late-diastolic flow murmur of relative mitral stenosis (Le., Carey-Coombs murmur) may be heard in patients with severe mitral insufficiency. The murmur of aortic insufficiency is a high-frequency, diastolic murmur that starts with the aortic component of the second heart sound. It is best heard with the diaphragm of the stethoscope over the third left intercostal space with the patient in the upright position and leaning forward. The murmur of mild aortic insufficiency is faint and often difficult to hear. Murmurs of severe insufficiency are loud and accompanied by a diastolic thrill. In these patients, an increased pulse pressure due to aortic runoff is associated with bounding peripheral pulses (Le., Corrigan pulse). Mitral and aortic valve stenoses result from valvular scarring and develop during the chronic stages of the disease.
Acute heart failure due to severe myocarditis or valvular insufficiency occurs in about 5% of children with ARF. The clinical manifestations vary greatly and include cough, chest pain, dyspnea, orthopnea, and anorexia. Tachycardia, cardiomegaly, and hepatomegaly with tenderness of the liver are present on physical examination.
Frequency of Major Manifeslations of A( ute Rheumatic Fever in Patients in U.S. dnd Non-U.S Patients
U.S. Patients
Manifestation Arthritis Carditis Sydenham's chorea Erythema marginatum Subcutaneous nodules
1958-1962 75% 48% 16% 6% 7%
Non-IJ.S. Patients
1962-1980
1985-1989
1960-1980
53% 78% 5% 2% 5%
65% 59% 20% 6% 5%
30-79% 41-93% 1-12% 0-16% 1-9"10
Adapted from Ayoub EM: Resurgence of rheumatic fever in the United States. The changing pictures of a preventable disease. Postgrad Med 92: 133-142, 1992.
620
C HAP T E R
31
ACUTE RHEUMATIC FEVER
& POST-STREPTOCOCCAL
Sydenham's Chorea Also known as St. Vitus' dance, this manifestation of inflammatory involvement of the basal ganglia and caudate nucleus of the central nervous system occurs in about 15% of patients. A higher frequency of Sydenham's chorea was documented by several centers during the recent resurgence of ARF in the United States. 60 The latency period between the inciting streptococcal pharyngitis and the onset of clinical signs of chorea is longer than that of the other major manifestations of the disease, averaging 2 to 4 months, and sometimes extending to as long as 12 months. A patient with Sydenham's chorea presents with persistent involuntary and purposeless movements of the extremities that are usually symmetric and with muscular incoordination. 92 These movements are jerky and most prominent in the face, trunk, and distal extremities. These symptoms disappear during sleep. On examination, the patient grimaces and fidgets constantly. The protruded tongue darts in and out and resembles a bag of worms (Le., wormian tongue). Speech is halting and explosive, and a steady tone cannot be maintained for even a short time. Extension of the arms above the head leads to pronation of the hands (Le., pronator sign); extension of the arms anteriorly results in hyperextension of the fingers (Le., spoon or dishing sign). When the patient is asked to squeeze the examiner's fingers, the examiner feels irregular contractions of the hand muscles (i.e., milkmaid's grip or milking sign). Handwriting, particularly drawing vertical straight lines, is clumsy and irregular because of the loss of fine muscle coordination. The patient has difficulty putting on clothes or buttoning a shirt. Such attempts lead to easy frustration and emotional upsets. Parents and teachers often complain about the child's clumsiness, inability to concentrate on tasks, or emotional lability. Some patients are misdiagnosed as having a behavior problem, an attention-deficit disorder, or a "tic." These symptoms usually resolve spontaneously in 2 to 3 weeks, but in severe cases, they may persist for several months and sometimes for years.
Erythema Marglnatum Erythema marginatum is characteristic of rheumatic fever and occurs in about 5% of patients. This rash is nonpruritic and macular with a serpiginous erythematous border (Fig. 31-5). The individual lesions are about 0.4 cm in diameter and are usually located on the trunk and proximal inner aspects of the limbs, particularly where they join the trunk. The rash is rare on the face or other exposed areas. It is accentuated by warmth, such as the application of warm towels or a bath. Erythema marginatum is difficult to detect in patients with dark skin.
Subcutaneous Nodules The subcutaneous nodules of ARF that were most common in patients who developed chronic rheumatic heart disease and were a sign of severe involvement are now rare. They are usually located on the extensor surfaces of the joints, particularly the elbows, knees, ankles, and
• FI...... :11-5 Rash of erythema marginatum in an adolescent boy with acute rheumatic fever occurred with its characteristic serpiginous and erythematous margins. (See color insert.)
knuckles, and occasionally on the occiput and spine. The overlying skin is not discolored. Their size varies from 0.5 to 2 cm, and they are freely movable. In many respects, they clinically and histologically resemble benign rheumatoid nodules.
Minor Manifestations of the Disease The minor manifestations of fever, arthralgia, and elevated acute phase reactants are nonspecific and encountered in a number of other rheumatic diseases. The severity and duration of fever vary; the patient may have a temperature of 38.5 0 C to 40 0 C during the acute phase of the disease. Arthralgia (Le., pain without objective changes in the joint) should be differentiated from arthritis. Abnormally elevated acute phase reactants are indicators of tissue inflammation and are present during the acute stage of the disease. A prolonged PR interval on the electrocardiogram is another nonspecific finding. It occurs frequently in ARF but does not alone constitute an adequate criterion for carditis; PR prolongation also does not correlate with the ultimate development of chronic rheumatic heart disease.
Pathology The inflammation that occurs in ARF is the result of a diffuse vasculitis. The organs most commonly affected are the joints, heart, brain, and peripheral vascular system. The vasculitis affects the smaller vessels and is characterized by proliferation of endothelial cells. This vasculitic process is reflected in the rash of ARF; inflammation of collagen occurs primarily in the arthritis, valvulitis, and pericarditis. The synovitis of ARF is typified by a mononuclear cell infiltrate with fibrinoid degeneration. The cartilage is usually not involved. 93 .94 Inflammation of the heart, the most serious complication of the disease, usually involves the myocardium and endocardium. Unlike other rheumatic diseases, such as
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
systemic lupus erythematosus (SLE) or juvenile rheumatoid arthritis (JRA), sole involvement of the pericardium is distinctly uncommon in ARF. Valvular endocarditis is the more common and characteristic inflammatory process and the principal cause of chronic cardiac disease. Acute inflammation leads to valvular insufficiency, and persistence of the inflammation results in scarring and stenosis (Fig 31-6). The mitral valve is the most commonly involved, and mitral insufficiency is the hallmark of rheumatic carditis. The aortic and tricuspid valves are affected less frequently. A review of the cardiac pathology of ARF by Roberts 9S indicated that isolated mitral valve disease was of rheumatic origin in 76% of cases, whereas aortic valve disease was ascribable to ARF in only 13% of cases. The simultaneous presence of mitral and aortic disease was related to a rheumatic cause in 97% of cases. Serum cardiac troponin I, a sensitive and specific marker of myocardial injury, is not elevated in cases of ARF with cardiac involvement, indicating that congestive heart failure in ARF is related to valvar insufficiency rather than specific myocardial inflammation. 96 The histologic changes in acute rheumatic carditis are not specific, and the degree of abnormality does not necessarily correlate with the severity of carditis. 93 .94 In the early stage, when dilatation of the myocardium is present, histologic changes can be minimal. Despite this, cardiac function may be severely impaired and associated with a high rate of mortality. Progression of the inflammation leads to an exudative and proliferative reaction in the myocardium characterized by edematous changes followed by a cellular infiltrate of lymphocytes and plasma cells with few granulocytes. CD4-positive cells predominate in the lymphocytic infiltrate. 97 Degenerating collagen fibers are visible throughout the tissue as eosinophilic, granular deposits consisting of a mixture of fibrin, globulin, and other substances. This stage is followed by the formation of the Aschoff body.98.99 This lesion consists of a perivascular infiltrate of large cells with polymorphous nuclei and basophilic cytoplasm arranged in a rosette around an avascular center of fibrinoid. The Aschoff body is pathognomonic of rheumatic carditis and occurs most commonly in patients with subacute or chronic carditis. It may develop in any area of the myocardium but is not present in other tissues. Tissue edema and cellular infiltrates characterize the inflammation of valvular tissue. This inflammatory process also involves the chordae tendineae. Verrucae may form at the edge of the leaflets, preventing the valves from complete closure. Persistent inflammation for several years results in fibrosis and calcification of the valve that lead to stenosis. The pathophysiology of Sydenham's chorea is centered in the basal ganglia. lOo Magnetic resonance imaging volumetric studies have indicated focal striatal enlargement and response of the chorea to dopamine antagonists. Histologic studies have documented cellular infiltration and neuronal loss in the basal ganglia. 101 •102 Subcutaneous nodules are characterized by a central area of fibrinoid necrosis surrounded by loosely demarcated zones of scattered mononuclear cells. Edema and vascular islands are present, but palisading of epithelioid cells is not well developed. Interstitial collagen fibers and scar
621
formation occupy the outermost layers without formation of a capsule. The histology is not pathognomonic but resembles that of the Aschoff body. Descriptions of the pathologic features of erythema marginatum are scant.
Diagnosis Classification Criteria No specific test is available for the definitive diagnosis of ARF. The diagnosis continues to be based on guidelines of clinical and laboratory criteria that were initially promulgated by T. Duckett Jones and subsequently revised by several committees of the American Heart Association. The latest modification of the Jones criteria is outlined in Table 31-5. 103 The purpose of these guidelines is to assist in the diagnosis of an initial attack of rheumatic fever and to minimize overdiagnosis of this disease. As stated under these guidelines, the presence of two major manifestations or one major plus two minor manifestations provides the basis for the diagnosis of ARF-if supported by evidence of antecedent group A streptococcal infection. The latter is a sine qua non for establishing the diagnosis. A positive throat culture or rapid antigen test can confirm an antecedent group A streptococcal pharyngitis. However, the period of latency between the inciting pharyngitis and the onset of ARF reduces the frequency of positive cultures to less than one third of patients. 104 More reliable evidence can be obtained by the use of the streptococcal antibody tests listed in Table 31-1. Because of the latency period, serum obtained at the time of the initial evaluation of the patient coincides with the peak of the antibody response (see Fig. 31-2). An elevated ASO or anti-DNAse B level is expected in about 85% of patients (see Table 31-2). When both tests are performed (considered by many to be a reasonable and conservative approach to diagnostic specificity), more than 90% of patients have an elevated titer
I! II
TABLE 31 -5 Guidelines for the Diagnosis of an Initl
Major Manifestations·
Minor Manifestations·
Carditis Polyarthritis Sydenham's chorea Erythema marginatum Subcutaneous nodules
Clinical Fever Arthralgia Laboratory Elevated acute phase reactants: Erythrocyte sedimentation rate C-reactive protein level Prolonged PR interval
Supporting Evidence of Antecedent Group A Streptococcal Infection· Elevated or rising streptococcal antibody titers Positive throat culture or rapid streptococcal antigen tests -The presence of two major manifestations or of one major and two minor manifestations indicates a high probability of acute rheumatic fever if supported by evidence of preceding group A streptococcal infection. Adapted from Dajani AS. Ayoub EM, Bierman FZ, et ai: Guidelines for the diagnosis of rheumatic fever: Jones Criteria. updated 1992. JAMA 87: 302-307, 1992.
622
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
for one of these tests. If the result of the ASO test is negative, a DNAse B titer should be performed. A fourfold (two-tube) increase or decrease in titers should be demonstrated over time because normal children in many geographic areas may have elevated titers. lOS Results of these antibody tests may be normal for most patients with chronic rheumatic heart disease, and a high proportion of patients with Sydenham's chorea may have normal ASO or anti-DNAse B titers (see Table 31-2). Neither the ASO nor the other streptococcal antibody tests are diagnostic of ARF; they are used to provide supportive evidence for antecedent streptococcal infection. The three acute phase reactants most commonly used in the diagnosis of ARF are the peripheral blood leukocyte count, the erythrocyte sedimentation rate (ESR), and the C-reactive protein (CRP) level. The leukocyte count is the most variable and least dependable. It is normal in about one half of the patients with ARF. The ESR is markedly elevated in patients with acute disease but may be normal even with severe congestive failure. 106 The CRP level is also elevated in patients with acute disease,107 and unlike the ESR, it is not affected by congestive heart failure. These tests are most useful in following the course of the disease and its response to treatment. Serum cardiac troponin 1 levels, known to be associated with myocardial injury, are not elevated in active rheumatic carditis. 89,96,108 Other studies that are useful in diagnosis include chest radiography, electrocardiography, and echocardiography. A chest radiograph can detect cardiac enlargement or pericardial fluid. These findings are best confirmed by echocardiographic studies, which can also define the presence of myocarditis by assessing myocardial contractility and the nature and extent of valvular lesions. 109 Electrocardiography is most useful in confirming abnormalities in conduction and rhythm during acute myocardial inflammation.
Differential Diagnosis Other rheumatic diseases account for most of the disorders that are misdiagnosed as ARF. JRA can be confused with ARF without carditis (see Chapters 9 to 12), Characteristics indicating a diagnosis of JRA rather than ARF include an onset of oligoarticular arthritis in a child before the age of 5 years; absence of erythema of the joint; a protracted, recurrent course with an incomplete response to nonsteroidal anti-inflammatory drug (NSAID) therapy; and particularly the absence of evidence for antecedent group A streptococcal infection. Post-streptococcal reactive arthritis poses some difficulty in differentiation from ARF. Clinical findings that should assist in the diagnosis of this disorder are discussed later in this chapter. Other conditions in which joint involvement is common that should be differentiated from ARF include SLE, Kawasaki disease, mixed connective tissue disease, other reactive arthritides, and serum sickness. no Infectious arthritis, particularly gonococcal arthritis, and brucellosis in endemic areas, may present a problem in differential diagnosis. Leukemia and hemoglobinopathy with bone infarcts can be mis-
taken for ARF. Appropriate evaluations and laboratory studies should assist proper diagnosis. Patients with carditis and pericarditis may develop secondary infections by a variety of bacterial, viral, rickettsial, or mycoplasmal agents. Endocardial involvement occurs in patients with bacterial endocarditis and in patients with SLE and Libman-Sacks endocarditis. A murmur and systolic clicks are present in patients with mitral valve prolapse. Some children with Kawasaki disease develop clinically obvious myocarditis and valvular disease during the early stages of illness. In these patients, the lack of evidence for antecedent group A streptococcal infection allows an initial differentiation from ARF. Differentiation of Sydenham's chorea from other neurologic disorders requires careful evaluation. 11l Imaging studies of the central nervous system are usually normal for patients with Sydenham's chorea. Other neurologic conditions that may be confused with Sydenham's chorea include congenital or acquired "habitual" tic disorders, attention-deficit disorders, and obsessive-compulsive behavior. 1l2 ASO and anti-DNAse B tests should provide evidence for antecedent streptococcal infection in more than 800/0 of children with Sydenham's chorea and assist in its differentiation from other neurologic disorders. Some children with pediatric autoimmune neuropsychiatric disorders are associated with streptococcal infections (PANDAS) and develop antineuronal antibodies. l13 ,1l4 Chorea is also a characteristic symptom in children with the antiphospholipid antibody syndrome (see Chapter 16).
Treatment The initial treatment of ARF should address the eradication of streptococci that initiated this complication and the inflammatory process that has affected the various organs. Patients with ARF should be promptly evaluated for cardiac involvement. Subsequent management includes prophylaxis to prevent recurrence of streptococcal infections and treatment of residual cardiac disease when present.
Eradication of Streptococci Patients should receive a streptococcal eradicating regimen of antimicrobials even though their throat culture or rapid antigen test is negative (Table 31-6).11S Penicillin is the primary agent of choice administered intramuscularly as a single dose or orally for 10 days. The intramuscular route is preferable in children with cardiac involvement because of its greater dependability and efficacy. Patients allergic to penicillin should receive erythromycin orally for 10 days.
Treatment of Clinical Manifestations Carditis Acute carditis requires immediate attention. 116 For mild to moderate carditis, aspirin is administered in a dose of 80 to 100 mg/kg/day in four divided doses. This schedule is maintained for 4 to 8 weeks, depending on clinical response, and then reduced gradually and discontinued
C HAP T E R I~
'_I
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
623
fABLE 31 -6 Antibiotic Regimens lor Primary Prevention (Strepto(O((al Eradication) dnd Secondary Prevention 01 RheUllJdti( Fever
Antibiotic
Dose
Route
DUl'lItion
600,000-1,200,000 va 250 mg every 8 hours 20-40 mg/kg/day (maximum, 1.0 g/day) in 2-4 divided doses
Intramuscular Oral Oral
Single dose 10 days 10 days
600,000-1,200,000 V 250 mg every 12 hours 1.0 g once daily 250 mg bid
Intramuscular Oral Oral Oral
Every 4 weeksb
PrImary PreventIon Benzathine penicillin G Penicillin V Erythromycin (estolate or ethylsuccinate) 5econdaly Prevention Benzathine penicillin G Penicillin V Sulfadiazine Erythromycin ethylsuccinate
• See text for details, b May be given every 3 weeks in high-risk situation, Adapted from Dajani A, Taubert K, Ferrieri P, et al: Treatment of acute streptococcal pharyngitis and prevention of rheumatic fever: A statement for health professionals, Pediatrics 96: 758-764, 1995,
during the next 4 weeks, Other NSAIDs may be as effective as aspirin,117,118 but have not yet been recommended by the expert committee of the American Heart Association, Glucocorticoid therapy is reserved for patients with severe carditis and congestive heart failure-particularly those with pancarditis, in whom it may be life saving, The use of glucocorticoids rather than aspirin in patients with heart failure is also justified to avoid solute overload from aspirin. It should be emphasized that neither form of therapy has been demonstrated to influence the subsequent evolution of valvular disease, 119-t21 Unlike most rheumatic diseases, the use of intravenous methylprednisone as a single anti-inflammatory agent is inferior to conventional treatment with oral prednisone in the control of severe rheumatic carditis. 122 Prednisone is given orally in a dose of 2 mg/kg once daily. The duration of daily steroid therapy should rarely exceed 2 weeks, and the drug should be tapered and withdrawn during the next 2 to 3 weeks. One week before termination of therapy, aspirin should be instituted (following the regimen described earlier) to avoid the rebound of symptoms and acute phase reactants that occurs when steroid therapy is abruptly terminated. The ESR and CRP level are essential in monitoring the response to anti-inflammatory therapy. In patients with heart failure and a falsely low ESR, a rise in this reactant may occur with recovery; the CRP level is more reliable in monitoring the response in these patients. Ancillary therapy for cardiac failure includes the judicious use of drugs such as digitalis; inotropic agents such as dobutamine, dopamine, or arnrinone; vasodilators (captopril or enalapril); and diuretics. General aspects of intial management include bed rest for patients with acute carditis. This recommendation was overly emphasized in the past, led to prolonged confinement in bed and cardiac neurosis, and should be discouraged. Gradual resumption of normal activity should be allowed after the acute carditis subsides.
Echocardiographic follow-up is predicated on the type and severity of the initial carditis and its response to therapy.123
Arthritis The arthritis characteristically pursues a self-limiting course, rarely lasting more than 1 week in anyone joint. A hallmark of the arthritis in this disease is its exquisite sensitivity to salicylates. A dose of aspirin of 50 to 75 mg/kg/day given in three to four divided doses is usually effective. This therapy is continued for no more than 2 weeks and is thereafter gradually withdrawn. A rapid resolution of the fever and a decline in the ESR usually parallel resolution of the arthritis. A lack of improvement of the arthritis within about 5 days of salicylate therapy should prompt a reconsideration of the correctness of the diagnosis. No data are available regarding the efficacy of other NSAIDs in the treatment of ARE Steroids should not be used in patients with isolated arthritis.
Chorea Mild manifestations of Sydenham's chorea require only bed rest and avoidance of physical and emotional stress. Although anticonvulsant drugs may help control severe symptoms, the response to these agents is unpredictable. Phenobarbital, haloperidol, carbamazepine, and valproate have been used with varying success. Anti-inflammatory agents are not needed for the treatment of chorea.
Prophylaxis of Rheumatic Heart Disease Medical management after the acute stage of the disease centers on prevention of recurrences of rheumatic fever and continued treatment of residual heart disease, including prevention of bacterial endocarditis. Antimicrobial prophylaxis against streptococcal pharyngitis has proved highly effective in reducing recurrences
624
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
of rheumatic fever and in preventing cumulative heart damage. Regimens for streptococcal prophylaxis recommended by the American Heart Association are outlined in Table 31-6. Because recurrences of rheumatic fever are most common during the 5 years after the initial attack,17,37,93 intramuscular benzathine penicillin prophylaxis is preferable and should be given once monthly in areas of low incidence of rheumatic fever and every 3 weeks in areas endemic to this disease. Oral prophylaxis is acceptable for patients without cardiac involvement. Although sulfonamides are ineffective in eradicating streptococcal infections, these agents are as effective, if not more effective, than oral penicillin for prophylaxis against recurrent streptococcal infections. The American Heart Association has revised recommendations on the duration of prophylaxis. Current protocols are based on the risk of reinfection and the development of streptococcal pharyngitis. 124 This risk is highest in school-aged children and in persons working in crowded conditions and military recruits, and those in close contact with children, such as parents, teachers, and health providers. Therefore patients with carditis should receive prophylaxis well into adulthood, preferably for life, whereas it may be discontinued at the age of 21 years in those with no cardiac involvement (although all such patients should receive prophylaxis for a minimum of 5 years regardless of age).125 Prophylaxis should not be discontinued after surgical valve repair.
Endocarditis Prophylaxis Supplemental doses of antibiotic should be prescribed for surgical or dental procedures in children with known rheumatic heart disease. Specific recommendations vary, depending on the procedure and age of the patient. 126
Chronic rheumatic valvular heart disease.Verrucal endocardial thickening was present along the line of dosure of the valve leaflets
• Figure 31-6 (arrow).
adults but does not occur in children. 128 It is more commonly associated with SLE. Sydenham's chorea and erythema marginatum are also self-limited with no permanent residua. Patients who escape severe heart disease can be assured of a benign course and a good prognosis.
POST-STREPTOCOCCAL REACTIVE ARTHRITIS Course of the Disease and Prognosis Major morbidity in rheumatic fever is associated exclusively with the degree of cardiac damage. Severe carditis, which leads to chronic residual valvular disease (see Fig. 31-6), primarily occurs in children in developing countries. The availability of cardiac surgery has alleviated to a considerable extent the crippling effect of this complication. Mortality is rare and occurs predominantly in patients with pancarditis. A better understanding of the relationship of streptococcal infection to the occurrence of initial attacks and recurrences of rheumatic fever has led to the institution of prophylactic regimens that have prevented subsequent attacks of the disease and reduced the cumulative heart damage produced by these exacerbations. The study by Tompkins and colleagues 127 emphasizes the singular value of prophylaxis by confirming that signs of rheumatic valvular disease resolve in about 80% of patients who receive continuous, long-term prophylaxis. This information is of particular importance in encouraging patients with rheumatic heart disease to adhere to the prescribed regimen of prophylaxis. Rheumatic arthritis is self-limited. A rare form of nonerosive but deforming arthropathy ascribed to rhel\.matic fever (Le., Jaccoud's arthritis) has been reported in
Definition and Classification The occurrence of arthritis after group A streptococcal infection in children who did not fulfill criteria for the diagnosis of ARF was described first by Crea and Mortimer in 1959. 129 Subsequently, a number of other studies reported on this entity which was designated post-streptococcal reactive arthritis (PSRA).80,13G-137 In contrast to the arthritis of ARF, arthritis observed in these patients was nonmigratory, protracted in course, and responded poorly to aspirin or other NSAIDs. Despite these clinical differences, several investigators have maintained that PSRA is an extension of the spectrum of ARF. 131,138 Some studies, however, suggest that this syndrome differs significantly in pathogenesis and clinical characteristics from the arthritis of rheumatic fever. 139
Epidemiology Eighty-six patients with PSRA have been described. 80 Although it is difficult to assess accurately, the incidence of this disease in North-Central Florida is estimated to be 1 to 2 cases per 100,000 children at risk per year; 17 of
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
625
455 patients with rheumatic diseases encountered over a period of 2 years had PSRA. 80· 140 This incidence was twice that for ARF during the same period. The age of the patients varied from 5 to 16 years with a mean of 9.7 years. A slightly but not significantly higher incidence of the disease occurred in males (56% versus 44%). There was no ethnic preponderance.
Etiology and Pathogenesis Evidence for group A streptococcal infection should be documented in all patients. In contrast to ARF, in which throat cultures or rapid antigen tests are positive in one third of patients, results are positive in about 75% of patients with PSM. This difference can be ascribed to the shorter latency Oess than 10 days) for this disease.132.133.136 Streptococcal pharyngitis is associated with an ASO and an anti-DNAse B response in most patients. 80 Skin infection does not elicit an ASO response. The high frequency of elevated ASO titers in patients with PSRA suggests that streptococcal pharyngitis is the primary inciting cause of the disease. 141
Genetic Background Studies of the relationship of PSRA with HLA-B27 failed to document a significant association80 ; only 3 of 18 (16.7%) white American patients were positive. This frequency contrasts with reactive arthritis in children, in which 93% are HLA-B27 positive. 142 Further studies, however, documented a significant association of PSRA with HLA class II alleles. Compared with normal controls and patients with ARF, these patients had a significant increase in the frequency of DRBl*01. 80 In ARF, there is an increased frequency of the DRB1*16 allele. These associations with DRB1 alleles suggest a common pathogenetic mechanism for PSRA and ARF.
alnlcal Manifestations In addition to a pharyngitis that is present in 66% of patients,80·140 approximately 30% report the occurrence of low-grade fever, and a similar number describe a nonscarlatinal rash that precedes onset of the arthritis. About one half of the children complain of morning stiffness of varying duration. Most patients present with arthritis involving one or more joints. About 10% complain only of arthralgia. The frequency of joint involvement is illustrated in Figure 31-7. The arthritis is asymmetric and nonmigratory in 70% to 80% and involves joints of the lower extremities in almost all patients. One half of the patients also have arthritis involving the upper extremities. 80 Axial disease occurs in 25%; in our experience, these patients account for any possible association with HLA-B27. Cardiac disease was present in 5.8% of 86 patients described in the literature. 80 In almost all cases, valvular disease was only detected, if at all, several months after onset. Most of these patients had not been placed on penicillin prophylaxis. The delay in onset of cardiac abnormalities should be contrasted with the carditis associated with ARF, in which cardiac involvement usually occurs during the acute stage of the disease and in tandem with the arthritis.
• Figure 31-7 Frequency of joint involvement in patients with poststreptococcal reactive arthritis.Values represent the number of patients with involvement of that joint. MCp, metacarpopharyngeal; PIp, proximal interphalangeal. (From Ahmed 5, Ayoub EM, Scomik !C, et al: Poststreptoc.occal reactive arthritis: dinical characteristics and assodation with HLA-DR alleles. Arthritis Rheum 41: 109~ll02, 1998. Copyright © 1998 John Wiley & Sons, Inc. Reprinted with permission ofWiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.)
Some investigators have described patients who presented with "silent carditis," indicating that this complication was not clinically detectable and required echocardiographic studies for confirmation. 133 This suggests that the frequency of carditis in this disease may be higher than that reported to date. Extra-articular manifestations include vasculitis, tenosynovitis, and glomerulonephritis. 143 Acquired Brown's syndrome, the inability to elevate the affected eye in full adduction resulting from inflammatory tenosynovitis of the superior oblique tendon, has been reported in one child. 144
Diagnosis and Differential Diagnosis Proposed criteria for the diagnosis of PSRA are in Table 31-7. 140 The differential diagnosis includes most of the
I!.'II
TABLE 31-7 Proposed Criteria for the Diagnosis of Post-strepto(O«:al Reactive Arthritis
A. Characteristics of the arthritis 1. Acute-onset arthritis, symmetric or asymmetric, usually nonmigratory, can affect any joint 2. Persistent or recurrent 3. Poorly responsive to aspirin or nonsteroidal anti-inflammatory drugs B. Evidence of antecedent group A streptococcal infection C. Does not fulfill the modified Jones criteria for the diagnosis of acute rheumatic fever Adapted from Ayoub EM, Ahmed s: Update on complications of Group A streptococcal infections. Curr Probl Pediatr 27: 90-101. 1997.
626
C HAP T E R
3 I A CUT E R HE U MATI C FE V ER &
~ II
Po S T - S T REP Toe 0 C CAL
TAB! E 1I 8 (linicdl ,lIId I dbOldtO'y (hdlddemli< s of f'ost strepto(o«ili Red
Characteristics
PSRA
ARF
Antecedent group A streptococcal infection Onset of arthritis after infection Migratory arthritis Axial arthritis Heart involvement Response to ASA Association with HLA-B27 Association with HLA-DR~ aJleles
Yes <2 weeks No Yes
Yes
6% Not dramatic No DRBl*Ol
2-3 weeks Yes No 50% Dramatic No DRBl*16
ARF, acute rheumatic fever; ASA, 5-aminosalicylic acid; PSRA, post-streptococcal reactive arthritis. Adapted from Ahmed S, Ayoub EM, Scorrtik JC, et al: Poststreptococcal reactive arthritis: clinical characteristics and association with HLA-DR alleles. Arthritis Rheum 41: 1096-1102, 1998. Copyright © 1998 John Wiley & Sons, Inc. Reprinted with permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.
same arthritides outlined for ARE The similarity in cause, and in some of the clinical manifestations of both diseases, poses unresolved difficulty in differentiating these two entities, However, as outlined in Table 31-8, clinical and laboratory differences should permit the separation of this entity from ARF and other reactive arthritides in most children.
Laboratory Examination The leukocyte count is normal in the majority of patients. However, the ESR is elevated in 75%. As in ARF, this test is helpful in assessing the course of the arthritis. Streptococcal antibody tests are more dependable than in ARF for documenting evidence for an antecedent group A streptococcal infection. The ASO level is elevated in 88% and the anti-DNAse B in 80% of patients. At least one of these antibodies should be elevated in almost all patients at the time of presentation or shortly thereafter. so Because of the shorter period of latency between the streptococcal infection and the onset of arthritis, these patients have a higher frequency of positive throat cultures or rapid antigen tests for group A streptococci than patients with ARF.
by then, prophylaxis is discontinued. If carditis occurs during this interval, the patient is considered to have had ARF and should continue to receive prophylaxis in accordance with previously stated recommendations. However, discontinuing prophylaxis after only 1 year potentially leaves the patient at risk for the development of carditis. Our preference is to institute a prophylactic regimen similar to that proposed by the American Heart Association for patients with ARF who have arthritis but no carditis-that is, to continue prophylaxis until the patient reaches the age of 21 years and for a minimum of at least 5 years. 124 All patients should be reevaluated for evidence of carditis for a period extending over 5 years.
Course of the Disease and Prognosis Unlike the arthritis of ARF, the course of the arthritis in PSRA is often protracted, lasting 5 days to 8 months, with a mean duration of 66 days from onset to resolution. 80 Some patients continue to have arthralgia for several months after remission of the arthritis. This prolonged course is not altered significantly by the administration of NSAIDs or antimicrobials.
ACKNOWLEDGMENT
Treatment NSAIDs (e.g., naproxen, ibuprofen, tolmetin) are the principal drugs used in treatment. Aspirin probably offers no particular advantage. The value of disease-modifying drugs, such as methotrexate, has not been assessed. Physical therapy should be instituted for relief of joint pain and stiffness. As recommended for patients with group A streptococcal pharyngitis and its complications, antimicrobial therapy should be prescribed at the time of initial diagnosis to eradicate streptococci from the tonsillopharyngeal tissue (see Table 31-6). Antimicrobial prophylaxis to prevent recurrences, and possibly subsequent cardiac disease, has been recommended by some investigators, but this issue is controversiaJ.l45 Because carditis can occur in this disease, albeit probably at a lower rate than in patients with rheumatic fever, the American Heart Association has suggested prophylaxis for 1 year. 124 If carditis is not detected
Dr. Elia Ayoub died at home on April 4, 2004, after a long illness, comforted by the presence of his family and close associates. He had revised this chapter with his former fellow and colleague, Dr. Khaled Alsaeid, during the first few months of 2004. Elia was a world expert on rheumatic fever, an outstanding clinician and scholar, and a dear friend. He will be greatly missed by the pediatric rheumatology community, the University of Florida College of Medicine, and his many patients.
REFERENCES 1. Stollerman GH: Rheumatogenic group A streptococci and the return of rheumatic fever. Adv Intern Med 35: 1-25, 1990. 2. Steer AC, Carapetis JR, Nolan TM, et al: Systematic review of rheumatic heart disease prevalence in children in developing countries: the role of environmental factors. J Paediatr Child Health 38: 229-234, 2002, 3, Markowitz M: The decline of rheumatic fever: role of medical intervemion. Lewis W, Wannamaker Memorial Lecture. J Pediatr 106: 545-550, 1985.
C HAP T E R
31
ACUTE RHEUMATIC FEVER
4. Veasy LG, Wiedmeier SE, Orsmond GS, et al: Resurgence of acute rheumatic f"ver in the intermountain area of the United States. N Engl J Med 316: 421-427, 1987. 5. Wald ER, Dashefsky B, Feidt C, et al: Acute rheumatic fever in western l'ennsylvania and the tristate area. Pediatrics 80: 371-374, 1987. 6. (:ongeni B, Rizzo C, Congeni J, et al: Outbreak of acute rheumatic fever in northeast Ohio. J Pediatr 111: 176-179, 1987. 7. tlDsier DM, Craenen JM. Teske DW, et al: Resurgence of acute rheumatic fever. Am J Dis Child 141: 730-733, 1987, 8. Griffiths SP, Gersony WM: Acute rheumatic fever in New York City 0969 to 1988): a comparative study of two decades, J Pediatr 116: 882-887, 1990. 9. Westlake RM, Graham TP, Edwards KM: An outbreak of acute rheumatic 'fever in Tennessee. Pediatr Infect Dis J 9: 97-100, 1990, 10. 'Eckerd JM, Mcjunkin JE: Recent increase in incidence of acute rheumatic fever in southern West Virginia. W V MedJ 85: 32~325, 1989. 11. :Mason T, Fisher M, Kujala G: Acute rheumatic fever in West Virginia. Not just :a disease of children. Arch Intern Med 151: 13~136, 1991. 12. Papadinos T, Escanmilla J, Garst P, et al: Acute rheumatic fever at a Navy training center-San Diego, California. MMWR Morb Mortal Wkly Rep 37: 101-104, 1988. 13. 'Sampson GL, Williams RG, House MD, et al: Acute rheumatic fever among Army trainees-Fort Leonard Wood, Missouri, 1987-1988. MMWR Morb Mortal WkIy Rep 37: 519-522, 1988. 14. 'Zangwill KM, Wald ER, Londino AV Jr: Acute rheumatic fever in western Pennsylvania: a persistent problem into the 1990s. J Pediatr 118: 561-563, 1991. 15. Loeffler AM, Neches WH, Ortenzo M, et al: Identification of cases of acute rheumatic fever managed on an outpatient basis. Pediatr Infect Dis .I 14: 975-978, 1995. 16, Veasy LG, Tani LY, Hill HR: Persistence of acute rheumatiC fever in the intermountain area of the United States. J Pediatr 124: 9-16, 1994. 17. Bland EF, DuckettJT: Rheumatic fever and rheumatic heart disease; a twenty year report on 1000 patients followed since childhood. Circulation 4: 836-843, 1951. 18. Stollerman G: Factors determining the attack rate of rheumatic fever. JAMA 177: 82~28, 1961. 19, Feinstein AR, Spagnuolo M: The clinical patterns of acute rheumatic fever: a reappraisal. Medicine (Baltimore) 41: 279-305, 1962. 20. Agarwal BL: Rheumatic heart disease unabated in developing countries. Lancet 2: 910-911, 1981. 21. WHO Study Group: Rheumatic fever and rheumatic heart disease. Report of a WHO Study Group. World Health Organ Tech Rep Ser 764: 1-58, 1988. 22, Gordis L: Changing risk of rheumatic fever. In Shulman S (ed); Management of Pharyngitis in an Era of Declining Rheumatic Fever. The 86th Conference on Pediatric Research. Columbus, OH, Ross Laboratories, 1983, pp 7-13. 23. Gordis L: The virtual disappearance of rheumatic fever in the United States: lessons in the rise and fall of disease. T. Duckett Jones memorial lecture. Circulation 72: 1155-1162, 1985. 24. Caughey DE, Douglas R, Wilson W, et al: HL-A antigens in Europeans and Maoris with rheumatic fever and rheumatic heart disease. J Rheumatol 2: 319-322, 1975, 25, Wannamaker LW: Differences between streptococcal infections of the throat and of the skin (second of two parts). N Engl J Med 282: 78-85, 1970, 26, Lancefield RC: A serological differentiation of human and other groups of hemolytic streptococci. .I Exp Med 57: 571-595, 1933. 27, Schwab]H, Cromartie W): Immunological studies on a C polysaccharide complex of group A streptococci having a direct toxic effect on connective tissue. J Exp Med 111: 295-307, 1960. 28, Cromartie WJ, Craddock JG, Schwab]H, et al: Arthritis in rats after systemic injection of streptococcal cells or cell walls. J Exp Med 146: 1585-1602, 1977. 29, Esser RE, Stimpson SA, Cromartie WJ, et al: Reactivation of streptococcal cell wall-induced arthritis by homologous and heterologous cell wall polymers, Arthritis Rheum 28: 1402-1411, 1985, 30. Goldstein I, Halpern B, Robert L: Immunologic relation between Streptococcus A polysaccharide and the structural glycoproteins of heart valve, Nature 213: 44-47, 1967. 31. Fischetti VA: Streptococcal M protein, Sci Am 264: 58-65, 1991. 32. Johnson DR, Stevens DL, Kaplan EL: Epidemiologic analysis of group A streptococcal serotypes associated with severe systemic infections, rheumatic fever, or uncomplicated pharyngitis. .I Infect Dis 166: 374-382, 1992. 33. Smoot jC, Korgenski EK, Daly JA, et al: Molecular analysis of group A Streptococcus type emm18 isolates temporally associated with acute rheumatic fever outbreaks in Salt Lake City, Utah. .I Clin Microbiol40: 1805-1810, 2002. 34. Talkington DF, Schwartz B, Black CM, et al: Association of phenotypic and genotypic characteristics of invasive Streptococcus pyogenes isolates with clinical components of streptococcal toxic shock syndrome. Infect Immun 61: 3369-3374, 1993. 35. Bessen D, Jones KF, Fischetti VA: Evidence for two distinct classes of streptococcal M protein and their relationship to rheumatic fever, .I Exp Med 169: 269-283, 1989. 36. Smoot JC, Barbian KD, Van Gompel JJ, et al: Genome sequence and comparative microarray analysis of serotype M18 group A Streptococcus strains associated with acute rheumatic fever outbreaks. Proc Nat! Acad Sci USA 99: 4668-4673, 2002.
& POST-STREPTOCOCCAL
627
37, Ayoub EM: Cross-reacting antibodies in the pathogenesis of rheumatic myocardial and valvular disease. In Wannamaker LW, Matsen JM (eds): Streptococci and Streptococcal Diseases. Recognition, Understanding and Management, 1st ed. New York, Academic Press, 1972, pp 451-464. 38. Ayoub EM, Kaplan E: Host-parasite interaction in the pathogenesis of rheumatic fever. J Rheumatol Suppl 30: 6-13. 1991. 39, Tomai M, Kotb M, Majumdar G, et al: Superantigenicity of streptococcal M protein. J Exp Med 172: 359-362, 1990. 40. Sandson j, Hamerman D, Janis R, et al: Immunologic and chemical similarities between the streptococcus and human connective tissue. Trans Assoc Am Physicians 81: 249-257, 1968. 41. Dinkla K, Rohde M, Jansen WT, et al: Rheumatic fever-associated Streptococcus pyogenes isolates aggregate collagen. .I Clin Invest 111: 1905-1912, 2003, 42, Hauser AR, Stevens DL, Kaplan EL, et al: Molecular analysis of pyrogenic exotoxins from Streptococcus pyogenes isolates associated with toxic shocklike syndrome. J Clin Microbiol 29: 1562-1567, 1991. 43, Bhatnagar A, Grover A, Ganguly NK: Superantigen-induced T cell responses in acute rheumatic fever and chronic rheumatic heart disease patients. Clin Exp Immunol 116: 100-106, 1999. 44. Stevens DL: Streptococcal toxic-shock syndrome: spectrum of disease, pathogenesis, and new concepts in treatment. Emerg Infect Dis 1: 69-78, 1995. 45. Murray DL, Ohlendorf DH, ScWievert PM: Staphylococcal and streptococcal superantigens: their role in human diseases. ASM News 61: 229-235, 1995. 46, Kuo CF, Wu JJ, Tsai PJ, et al: Streptococcal pyrogenic exotoxin B induces apoptosis and reduces phagocytic activity in U937 cells. Infect Immun 67: 126-130, 1999, 47, Yu CE, Ferretti 11: Molecular epidemiologic analysis of the type A streptococcal exotoxin (erythrogenic toxin) gene (speA) in clinical Streptococcus pyogenes strains. Infect Immun 57: 3715-3719, 1989. 48. Chaussee MS, Liu J, Stevens DL, et al: Genetic and phenotypic diversity among isolates of Streptococcus pyogenes from invasive infections. J Infect Dis 173: 901-908, 1996. 49. Todd EW: AntIhaemolysin titers in haemolytic streptococcal infections and their significance in rheumatic fever, Br J Exp Pathol 13: 248-259, 1932. 50. Gerber MA, Wright LL, Randolph MF: Streptozyme test for antibodies to group A streptococcal antigens. Pediatr Infect Dis J 6: 36-40, 1987. 51. Dudding BA, Ayoub EM: Persistence of streptococcal group A antibody in patients with rheumatic valvular disease. J Exp Med 128: 1081-1098, 1968. 52. Ayoub EM, Shulman ST: Pattern of antibody response to the streptococcal group A carbohydrate in rheumatic patients with or without carditiS. In Read SE, Zabriskie JB (eds): Streptococcal Disease and the Immune Response. New York, Academic Press. 1980, pp 649-659. 53. Appleton RS, Victorica BE, Tamer D, et al: Specificity of persistence of antibody to the streptococcal group A carbohydrate in rheumatic valvular heart disease. J Lab Clin Med 105: 114-119, 1985, 54. Ayoub EM: Immune response to group A streptococcal infections. Pediatr Infect Dis .I 10: Sl5-S19, 1991. 55, Kaplan MH, Meyeserian M: An immunological cross-reaction between group-A streptococcal cells and human heart tissue. Lancet 1: 706-710, 1962. 56. Kaplan MH, Svec KH: Immunologic re!ation of streptococcal and tissue antigens. Ill. Presence in human sera of streptococcal antibody cross-reactive with heart tissue. Association with streptococcal infection, rheumatic fever, and glomerulonephritis. J Exp Med 119: 651-666, 1964, 57. Widdowson JP, Maxted WR, Pinney AM: An M-associated protein antigen (MAP) of group A streptococci, .I Hyg (Land) 69: 553-564, 1971. 58. Zabriskie JB, Freirner EH: An immunological relationship between the group, A streptococcus and mammalian muscle, J Exp Med 124: 661-678, 1966, 59. van dR, I, Zabriskie ]B, McCarty M: Group A streptococcal antigens crossreactive with myocardium. Purification of heart-reactive antibody and isolation and characterization of the streptococcal antigen. J Exp Med 146: 579-599, 1977, 60. Dale JB, Beachey EH: Multiple, heart-cross-reactive epitopes of streptococcal M proteins. J Exp Med 161: 11~122, 1985. 61. Husby G, van dR, I, Zabriskie]B, et al: Antibodies reacting with cytoplasm of subthalamic and caudate nuclei neurons in chorea and acute rheumatic fever. J Exp Med 144: 1094-1110, 1976. 62. Kirvan CA, Swedo SE, Heuser jS, et al: Mimicry and autoantibody-mediated neuronal cell signaling in Sydenham chorea. Nat Med 9: 914-920, 2003. 63. Hutto .I, Ayoub EM: CytotOXicity of lymphocytes from patients with rheumatic carditis to cardiac cells in vitro. In Read SE, Zabriskie JB (eds): Streptococcal Disease and Immune Response. New York, Academic Press, 1980, pp 733-738, 64. Rammelkamp CHJ: Epidemiology of streptococcal infections. Harvey Lect 51: 11~142, 1956. 65. Read SE, Reid H, Poon-King T, et al: HLA and predisposition to the nonsuppurative sequelae of group A streptococcal infections. Transplant Proc 9: 54~546, 1977. 66, Wilson MG, Schweitzer MD, Lubschez R: The familial epidemiology of rheumatic fever: genetic and epidemiologic studies. J Pediatr 22: 468-492, 1943. 67, Wilson MG, Schweitzer M: Pattern of hereditary susceptibility in rheumatic fever. Circulation 10: 699-704, 1954,
628
C HAP T E R
31
ACUTE RHEUMATIC FEVER & POST-STREPTOCOCCAL
68. Khanna AK, Buskirk DR, Williams RC jr, et al: Presence of a non-HLA B cell antigen in rheumatic fever patients and their families as defined by a monoclonal antibody. J Clin Invest 83: 1710-1716, 1989. 69. Harel L, Zeharia A, Kodman Y, et al: Presence of the d8/17 B-cell marker in children with rheumatic fever in Israel. Clin Genet 61: 293--298, 2002. 70. Ayoub EM, Barrett OJ, Maclaren NK, et al: Association of class II human histocompatibility leukocyte antigens with rheumatic fever. j Clin Invest 77: 2019-2026, 1986. 7!. Anastasiou-Nana MI, Anderson jL, Car/quist JF, et al: HLA-DR typing and lymphocyte subset evaluation in rheumatic heart disease: a search for immune response factors. Am Heart J 112: 992-997, 1986. 72. Jhinghan B, Mehra NK, Reddy KS, et al: HLA, blood groups and secretor status in patients with established rheumatic fever and rheumatic heart disease. Tissue Antigens 27: 172-178, 1986. 73. Rajapakse CN, Halim K, AI Orainey I, et al: A genetic marker for rheumatic heart disease. Br Heart J 58: 659-662, 1987. 74. Maharaj B, Hammond MG, Appadoo B, et al: HLA-A, B, DR, and DQ antigens in black patients with severe chronic rheumatic heart disease. Circulation 76: 259-261, 1987. 75. Taneja V, Mehra NK, Reddy KS, et al: HLA-DR/DQ antigens and reactivity to B cell alloantigen 08/17 in Indian patients with rheumatic heart disease. Circulation 80: 335-340, 198'). 76. Guilherme L, Weidebach W, Kiss MH, et al: Association of human leukocyte class II antigens with rheumatic fever or rheumatic heart disease in a Brazilian population. Circulation 83: 1995-1998, 1991. 77. Ozkan M, Carin M, Sonmez G, et al: HLA antigens in Turkish race with rheumatic heart disease. Circulation 87: 1974-1978, 1993. 78. Weidebach W, Goldberg AC, Chiarella JM, et al: HLA class II antigens in rheumatic fever. Analysis of the DR locus by restriction fragment-length polymorphism and oligotyping. Hum Immunol 40: 253--258, 1994. 79. Guedez Y, Kotby A, EI Demellawy M, et ai: HLA class II associations with rheumatic heart disease are more evident and consistent among clinically homogeneous patients. Circulation 99: 2784-2790, 1999. 80. Ahmed S, Ayoub EM, Scomik JC, et al: Poststreptococcal reactive arthritis: clinical characteristics and association with HLA-DR alleles. Arthritis Rheum 41: 1096--1102, 1998. 8!. Stanevicha V, Eglite j, Sochnevs A, et al: HLA class II associations with rheumatic heart disease among clinically homogeneous patients in children in Latvia. Arthritis Res Ther 5: R340-R346, 2003. 82. Stetson CA: The relation of antibody response to rheumatic fever. In McCarty M (ed); Streptococcal Infections. New York, Columbia University Press, 1954. 83. Ayoub EM, Wannamaker LW: Evaluation of the streptococcal deoxyribonuclease Band diphosphopyridine nucleotidase antibody tests in acute rheumatic fever and acute glomerulonephritis. Pediatrics 29: 527-538, 1962. 84. Shulman ST, Ayoub EM, Victorica BE, et al: Differences in antibody response to streptococcal antigens in children with rheumatic and non-rheumatic mitral valve disease. Circulation 50: 1244-1251, 1974. 85. Braun DG, Eichmann K, Krause RM: Rabbit antibodies to streptococcal carbohydrates. Influence of primary and secondary immunization and of possible genetic factors on the antibody response. J Exp Med 129: 809-830, 1969. 86. Klapper DG, Kindt 1]: Idiotypic cross-reactions among anti-streptococcal antibodies in an inbred rabbit population. Scand J Immunol 3: 483--490, 1974 87. Sasazuki T, Kaneoka H, Nishimura Y, et al: An HLA-linked immune suppression gene in man. J Exp Med 152: 297s-313s, 1980. 88. Carapetis JR, Currie B]: Rheumatic fever in a high incidence population: the importance of monoarthritis and low grade fever. Arch Dis Child 85: 223--227, 2001. 89. Williams RV, Minich LL, Shaddy RE, et al: Evidence for lack of myocardial injury in children with acute rheumatic carditis. Cardiol Young 12: 519-523, 2002. 90. Kamblock J, Payot L, lung B, et ale Does rheumatic myocarditis really exists? Systematic study with echocardiography and cardiac troponin I blood levels. Eur Heart J 24: 855-862, 2003. 91. Malik jA, Hassan C, Khan GQ: Transient complete heart block complicating acute rheumatic fever. Indian Heart J 54: 91-92, 2002. 92. Aron AM, Freeman JM, Carter S: The natural history of Sydenham's chorea. Review of the literature and long-term evaluation with emphasis on cardiac sequelae. Am J Med 38: 83--95, 1965, 93. Taranta A: Rheumatic fever. In McCarty OJ, Koopman WJ (eds): Arthritis and Allied Conditions, 12th ed. Philadelphia, Lea & Febiger, 1993, pp 1357-1368. 94. Taranta A, Markowitz M: Rheumatic Fever, 2nd ed. Dordrecht, KIuwer Academic Publishers, 1993. 95. Roberts we: Anatomically Lsolated aortic valvular disease, The case against its being of rheumatic etiology, Am J Med 49: 151-159, 1970. 96. Gupta M, Lent RW, Kaplan EL, et al: Serum cardiac troponin I in acute rheumatic fever. Am J Cardiol 89: 779-782, 2002. 97. Raizada V, Williams RC Jr, Chopra P, et al: Tissue distribution of lymphocytes in rheumatic heart valves as defined by monoclonal anti-T cell antibodies. AmJ Med 74: 90-96,1983. 98. Aschoff L: Zur myocarditisfrage. Disch Ges 8: 46--53, 1904. 99. Murphy GE: Nature of rheumatic heart disease with special reference to myocardial disease and heart failure. Medicine (Baltimore) 39: 289--381, 1960.
100. Faustino PC, Terreri MT, da Rocha AJ, et al: Clinical, laboratory, psychiatric and magnetic resonance findings in patients with Sydenham chorea. Neuroradiology 45: 456--462, 2003. 101. Giedd jN, Rapoport JL, Kruesi Mj, et al: Sydenham's chorea: magnetic resonance imaging of the basal ganglia. Neurology 45: 2199-2202, 1995. 102. Greentleld]G, Wolf.ohnJM: The pathology ofSydenham's chorea. Lancet 2: 603--006, 1992. 103. Dajani AS, Ayoub EM, Bierman FZ, et al: Guidelines for the diagno.is of rheumatic fever: Jones Criteria, updated 1992. JAMA 87: 302-307, 1992. 104. Ayoub EM: Streptococcal antibody test. in rheumatic fever. Clin Immunol Newsletter 3: 107-111,1982. 105. Sethi S, Kaushik K, Mohandas K, et al: Anti-streptolysin 0 titer. in nomlal healthy children of 5-15 years. Indian Pediatr 40: 1068-1071, 2003. 106. Haber HL, Leavy JA, Kessler PO, et al: The erythrocyte sedimentation rate in congestive heart failure. N EngI J Med 324: 353-358, 1991. 107. Golbasi Z, Ucar 0, Keles T, et al: Increased levels of high sensitive C-reactive protein in patients with chronic rheumatic valve disease: evidence of ongoing inflammation. Eur J Heart Fail 4: 593--595, 2002, 108. Gran B, Coban H, Karaaslan S, et al: Serum cardiac troponin-1 in active rheumatic carditis. Indian J Pediatr 68: 943--944, 2001. 109. Saxena A: Diagnosis of rheumatic fever: current status of jones criteria and role of echocardiography. Indian] Pediatr 67: 283--286, 2000. 110. Sondheimer HM, Lorts A: Cardiac involvement in inflammatory disease: systemic lupus erythematosus, rheumatic fever, and Kawasaki disease. Adolesc Med 12: 69-78, 2001. 11 1. Carapetis jR, Currie B]: Rheumatic chorea in northern Australia: a clinical and epidemiological study. Arch Dis Child 80: 353--358, 1999. 112, Swedo SE, Rapoport ]1, Cheslow DL, et aI: High prevalence of obsessivecompulsive symptoms in patients with Sydenham's chorea. Am j Psychiatry 146: 246--249, 1989. 113. Swedo SE, Leonard HL, Mittleman BB, et al: Identification of children with pediatric autoimmune neuropsychiatric disorders associated With streptococcal infections by a marker associated with rheumatic fever. Am J Psychiatry 154: 110-112, 1997. 114. Swedo SE, Leonard HL, Garvey M, et al: Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections: clinical description of the first 50 cases. Am J Psychiatry 155: 264-271, 1998. 115. Shulman ST: Acute streptococcal pharyngitis in pediatric medicine: current issues in diagnosis and management. Paediatr Drugs 5 (Suppl 1); 13--23, 2003. 116. Albert DA, Harel L, Karrison T: The treatment of rheumatic carditis: a review and meta-analysis, Medicine (Baltimore) 74: 1-12, 1995. 117. Karademir S, Oguz D, Senocak F, et al: Tolmetin and salicylate therapy in acute rheumatic fever: Comparison of clinical eftlcacy and side-effects. Pediatr Int 45: 676--679, 2003. 118. Hashkes P.I, Tauber T, Somekh E, et al: Naproxen as an alternative to aspirin for the treatment of arthritis of rheumatic fever: a randomized trial. J Pediatr 143: 399-401, 2003. 119. United Kingdom and United States joint Report: The treatment of acute rheumatic fever in children: a cooperative clinical trial of ACTH, cortisone and aspirin. Circulation 11: 343--371, 1955. 120, United Kingdom and United States joint Report: The evolution of rheumatic heart disease in children: five year report of a cooperative clinical trial of ACTH, cortisone and aspirin. Circulation 22: 503--515, 1960. 121. United Kingdom and United States Joint Report: The natural history of rheumatic fever and rheumatic heart disease. Ten-year report of a cooperative clinical trial of ACTH, cortisone, and aspirin. Circulation 32: 457--476, 1965. 122. Camara EJ, Braga JC, Alves-Silva LS, et al: Comparison of an intravenous pulse of methylprednisolone versus oral corticosteroid in severe acute rheumatic carditis: a randomized clinical trial. Cardiol Young 12: 119-124, 2002. 123, Figueroa FE, Fernandez MS, Valdes P, et al: Prospective comparison of clinical and echocardiographic diagnosis of rheumatic carditis: long term follow up of patients with subclinical disease. Heart 85: 407--410, 2001. 124. Dajani A, Taubert K, Ferrieri P, et al: Treatment of acute streptococcal pharyngitLs and prevention of rheumatic fever: a statement for health professionals. Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the Council on Cardiovascular Disease in the Young, the American Heart Association. Pediatrics %: 758-764, 1995. 125. Danjani AS, Bisno At, Chung KJ, et al: Prevention of rheumatic fever. A statement for health profe.sionals by the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the Council on Cardiovascular Disease in the Young, the American Heart Association. Circulation 78: 1082-1086, 1988. 126. Committee on Infectious Diseases American Academy of Pediatrics: Red Book 2003 Report of the Committee on Infectious Diseases, 26th ed. Elk Grove Village, IL, AAP, 2003. 127. Tompkins DG, Boxerbaum B, Liebman j: Long-term prognosis of rheumatic fever patients receiving regular intramuscular benzathine penicillin. Circulation 45: 543--551, 1972. 128. Zvailler NJ: Chronic postrheumatic-fever (Jaccoud's) arthritis. N Engl .I Med 267: 10-14, 1962. 129. Crea MA, Mortimer EA Jr: The nature of scarlatinal arthritis. Pediatrics 23: 879-884, 1959. 130. Goldsmith DP, Long SSe Streptococcal disease of childhood-a changing syndrome. Arthritis Rheum 25 (Supp!): S18, 1982.
C HAP T E R
31
ACUTE RHEUMATIC FEVER
131. D
& POST-STREPTOCOCCAL
629
139. Tutar E, Atalay S, Yilmaz E, et al: Poststreptococcal reactive arthritis in children: is it really a different entity from rheumatic fever? Rheumatol Int 22: 8~3, 2002. 140. Ayoub EM, Ahmed S: Update on complications of group A streptococcal infections. CUff Probl Pediatr 27: 90-101. 1997. 141. Fischetti VA, Windels M: Mapping the immunodeterminants of the complete streptococcal M6 protein molecule. Identification of an immunodominant region. J 1mmunol 141: 3592-3599. 1988. 142. McDermott M, McDevitt H: The immunogenetics of rheumatic diseases. Bull Rheum Dis 38: 1-10, 1988. 143. 19lesias-Gamarra A, Mendez EA, Cuellar ML, et al: Poststreptococcal reactive arthritis in adults: long-term follow-up. AmJ Med Sci 321: 173--177, 2001. 144. Faust AO, Gillenwater JM, Saulsbury FT: AcqUired Brown's syndrome in a child with poststreptococcal reactive arthritis. J Rheumatol 28: 2748-2749, 2001. 145. Lehman TJ, Edelheit BS: Clinical trials for post-streptococcal reactive arthritis. Curr RheumatoI Rep 3: 363--364, 2001.
( HAP T E R
32
~USCULOSKELETAL ~ANIFESTATIONS OF SYSTEMIC
DISEASE James T. Cassidy and Ross E. Petty
~jf
A great many nonrheumatic systemic disorders cause musculoskeletal signs or symptoms. Sometimes these are trivial; occasionally, they are the predominant presentations of the underlying disease. This chapter outlines systemic disorders that may present in the guise of an inflammatory rheumatic disease. It is not our intent to describe comprehensively the clinical and laboratory manifestations or management of such disorders, which can be found in standard textbooks dealing with the specific diseases.
NUTRmONAL ABNORMALITIES The category of nutritional musculoskeletal diseases and metabolic bone diseases encompasses a variety of disorders in which there is a defined or suspected nutritional deficiency or excess that results in signs or symptoms suggesting a rheumatic disease. 1
Rickets Rickets is an ancient term, introduced into the English literature around 1650, that includes several diseases associated with defective ossification of bone matrix (Table 32-1).2 It can result from deficiency of the active form of vitamin D O,25-dihydroxyvitamin D3), from a deficiency of phosphate, or rarely, from a deficiency of calcium. Some types (Le., hypophosphatemic rickets and rickets associated with hypophosphatasia) are associated with defective mineralization and are classified as osteochondrodysplasias, as discussed in Chapter 40. Disorders such as cystinosis that result in renal tubular acidosis may present as rickets with pain in the joints and metaphyseal enlargement (Fig. 32-1). The affected child presents with joint pain and tenderness over the bones. Bowing of the long bones and splaying of the rib cage are characteristic features. Proximal muscle weakness, particularly of the lower extremities, is occasionally prominent. Defective bone
630
growth results from suppression of calcification and maturation of epiphyseal cartilage. The result is a wide, frayed, irregular zone of uncalcified osteoid at the epiphyseal line-the rachitic metaphysis (Fig. 32-2). Most cases of rickets worldwide result from exclusion from the sun for social or cultural reasons or from insufficient dietary intake of vitamin D.3.4 Vitamin ~defi ciency rickets is seldom encountered in developed countries but may occur in infantile and adolescent forms in the rest of the world. loS In the presence of sufficient dietary vitamin, impaired absorption because of celiac disease, inflammatory bowel disease, scleroderma, or liver disease may result in vitamin D-deficiency rickets. Renal disease or administration of anticonvulsant medications in children deprived of sunlight may also cause the disorder. Glucocorticoids are antagonistic to vitamin D for calcium homeostasis. The normal source of vitamin D3 in humans is the skin, in which ultraviolet rays of sunlight convert 7-dehydrocholesterol into the vitamin prohormone. 6 This compound is subsequently transformed to the 25-hydroxy form in the liver and to active 1,25-dihydroxyvitamin in the kidney (Fig. 32-3). A deficiency of 1,25-dihydroxyvitamin D3 may result from a nutritional deficiency, from hepatic failure to convert vitamin D to 25-hydroxyvitamin D, or from failure of the kidney to convert 25-hydroxyvitamin D to 1,25-dihydroxyvitamin D3 ,
Hypophosphatemic vitamin ~resistant rickets, when expressed in infancy, leads to short stature, bowing of the legs, and ectopic calcificationP This disorder is inherited as an X-linked recessive or autosomal dominant trait, although sporadic cases occur. The basic defect is impaired parathormone-dependent proximal renal tubular reabsorption of phosphate. A low serum phosphate concentration with a normal calcium level is characteristic. Type I vitamin ~dependent rickets is an autosomal recessive defect in renal I-ex-hydroxylase that results in failure of hydroxylation of 25-hydroxyvitamin D to 1,25dihydroxyvitamin D3. The onset of typical features of rickets occurs before the age of 2 years. Type II vitamin
C HAP T E R
• _ . . ABI [ 32 ~ I
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
Causes of Rickets
Cause or Biochemical Abnormality Vitamin D deficiency Calcium deficiency Vitamin D resistance VitalTlin D dependence Type 1 Type 2 Hypophosphatasia
Exclusion from light or insufficient dietary vitamin D Impaired calcium absorption in celiac disease, inflammatory bowel disease, scleroderma, or liver disease Impaired parathormone-dependent prOXimal renal tubular reabsorption of phosphate Defect in renal I-IX-hydroxylase End-organ unresponsiveness to I,2S-dihydroxyvitamin D3 Decreased serum alkaline phosphatase
631
~dependent rickets is rare and characterized by defec-
tive intracellular interaction between 1,25-dihydroxyvitamin D3 and its receptor. Symptoms of rickets begin in early infancy before 1 year of age. Alopecia and absence of eyelashes occur frequently in this disorder. 8 Hypophospbatasia, a rare autosomal recessive disorder, has onset in infancy as severe rickets and fractures. 2.9 Band keratopathy, proptosis and papilledema develop. There may be early loss of teeth. Chondrocalcinosis and pseudogout may be associated features. There is a marked depression in the concentration of alkaline phosphatase. No treatment is effective.
Scurvy Ascorbic acid (vitamin C) is required for the formation of normal collagen and chondroitin sulfate. lO In scurvy, a deficiency of dietary vitamin C, poor collagen synthesis leads to intradermal and subperiosteal hemorrhage. Subperiosteal hemorrhage results in bone pain; the child, usually an infant, assumes the flexed posture of pseudoparalysis and is irritable when picked up. II Hemarthroses may also occur. In severe cases, "scorbutic beads," resulting from subluxation of the sternum at the costochondral junctions, may be visible on physical examination. Radiographs demonstrate subperiosteal new-bone apposition.
Hypervitaminosis A A large number of physiologic functions, organogenesis, and embryogenesis are affected by vitamin A and the derivative retinoids. 12 Excess intake of vitamin A or retinoids causes pain in the extremities, irritability, apathy, alopecia, and delayed growth. 13•14 Cortical hyperostosis (e.g., metatarsal bones, ulnas, spine) is a typical radiographic finding. Abnormal epiphyseal growth and periosteal new-bone apposition occur occasionally.
Fluorosis Fluorosis is endemic in certain areas of the world and results in chronic rheumatic symptoms in children. 15 High levels of fluoride may occur naturally in the water supply or may result from pollution. Dental fluorosis is an early sign of toxicity.16 Knee pain is often an early symptom, followed by limb, hand, or spinal abnormalities that suggest a chronic inflammatory arthropathy. Radiographs demonstrate increased bone density and later show calcification of the spinal ligaments, intervertebral disks, and entheses.J7 Cord compression can result from narrowing of the spinal canal.
Endemic Osteoarthritis • FJgwe 12-1 Vitamin D-deficient rickets in a toddler. A, Radiographs of knees demonstrate rachitic metaphyseal changes, indistinct cortices, and poorly defined trabeculation. The zone of provisional calcification is almost completely absent, the axial height of the epiphyseal plate is markedly increased, and cupping is evident. 8, X-ray films taken 6 months later demonstrate progressive healihg with replacement of vitamin D.
Kasbin-Beck disease and Mseleni joint disease are examples of an early-onset polyarticular arthritis that is little known in the developed world. These disorders may represent the result of a geographically restricted nutritional deficiency or toxin or be related to a heritable epiphyseal dysplasia in a genetically isolated population.
632
C HAP T E R
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
• Figure 3Z-Z Metabolism of vitamin D. Previtamin D3is fonned in the skin and isomerizes to vitamin D3or other biologically inert isomers.Vitamin Dbinding protein (DBP) has an affinity only for vitamin D3, which is translocated to the drculation.Vitamin D is then hydroxylated in the liver and kidney to the active metabolite, 1,25(OH)P3' (From Bhalla AK: Osteoporosis and osteomalada. In Maddison Pl, Isenberg DA, Woo P, Glass DN [eds): Oxford Textbook of Rheumatology. Oxford, England, Oxford University Press, 1993, p 1005.)
Kashin-Beck (Urov). Disease Reports from northeastern Russia, Siberia, northern China, and North Korea have described an endemic progressive arthropathy in children, unassociated with systemic or visceral manifestations, that has an uncertain cause but may result from mycotoxins in fungus-infected grain or from selenium deficiency.1I~-26 The result is an epiphyseal dysplasia from a zonal necrosis of chondrocytes of the epiphyses and metaphyses. 27 These abnormalities continue to increase in severity as long as the child lives in the endemic area and eats foods made with the contaminated grain. Excessive amounts of iron in the water and diet may contribute further to the polyarthritis. Experimental animals fed grain infected with Fusarium species develop a similar form of epiphyseal dysplasia. 21
Kashin-Beck disease causes symmetric polyarthritis and progressive enlargement and limitation of motion involving multiple joints (Le., interphalangeal, wrist, knee, and ankle).28--30 In the school-age child, aching and muscle weakness are the initial symptoms. Joint effusions and laboratory indices of inflammation are absent early in the disease. The eventual dwarfing, epiphyseal deformity, and short digits resemble those encountered in the lysosomal storage diseases.
Mseleni Joint Disease A chronic polyarthritis affects a large proportion of the Tsonga population of the Mseleni area of northern Zululand on the eastern seaboard of South AfricaY-33
C HAP T E R
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
633
tis; in the hips, which bear the brunt of the disease, protrusio acetabuli occurs in females (Fig. 32-4).34 Short metacarpals, ulna, and radius, and a deformity of the distal end of the ulna, are also present. The diagnosis is usually obvious in the geographic and racial context, but the differential diagnosis includes cretinism, brucellosis, hemochromatosis, alkaptonuria, and Legg-Calve-Perthes disease. Hips, knees, and ankles are the predominant sites of involvement in 66% of the women, 25% of the men, 7% of the girls, and 4% of the boys. Hands, wrists, shoulders, and elbows are less commonly affected.
METABOLIC DISEASES Abnormalities of Uric Add Metabolism Gout
• ~ 3Z-3 Cystinosis. An 18-month-old girl presented with joint pain primarily involving the large joints and profound muscle weakness due to cystinosis. A. Hands demonstrate swelling predominantly in the metaphyseal area of the radius and ulna, not the wrist joint proper. 8, Radiographs document metaphyseal resorption that is typical of rickets.
Onset of joint pain in childhood or adolescence is the first symptom of the disease. Restriction of movement and limitation of mobility develop at a variable rate. Mild stunting of growth is common, and a few patients develop severe dwarfing (Table 32-2). The life span is not shortened. Characteristic radiographic abnormalities include irregularity of the surface, density, and shape of the epiphyses that progresses to a secondary osteoarthri-
.~.
TABLE 32-2
Mseleni Joint Disease and Kashin-Beck Disease
a-.dertstlc
Mselenl Joint Disease
Kashln-Beck Disease
First noted Inherited Sex ratio Stunting of growth Posture
6 yr-adult Probably not More females Slight to severe
6-10 yr Probably not More males Moderate
Lumbar lordosis, genu valgum
Precocious osteoarthritis Radiology
Yes
Lumbar lordosis, neck extended, knees flexed Yes
Fragmented epiphyses, flared metaphyses, brachymetacarpia, protrusio acetabuli, platyspondyly
Dysplastic interphalangeal, wrist, knee, ankle joints; intra-articular loose bodies
The term gout refers to a group of disorders characterized by hyperuricemia and deposition of monosodium urate monohydrate crystals in tissues. 35-37 Its major clinical manifestations include an acute monarthritis, most commonly in the first metatarsophalangeal joint; chronic erosive arthritis associated with subcutaneous periarticular deposits of urate (tophi); and nephrolithiasis, often leading to chronic renal failure. Serum urate levels increase normally at puberty, particularly in males, from approximately 3.5 mg/dL (0.21 mmollL) in childhood to an upper limit of 7 mg/dL (0.42 mmollL) in adult males, and 6 mg/dL (0.36 mmollL) in adult females. Above these concentrations, the serum becomes saturated with urate. Gout may result from increased production or decreased excretion of uric acid (Table 32-3), Diagnosis is confirmed by demonstration with compensated polarized light microscopy of negatively birefringent, needleshaped monosodium urate crystals in synovial fluid. Treatment of the acute attack with nonsteroidal antiinflammatory drugs (NSAIDs) such as indomethacin, or with colchicine, is usually effective. After the acute episode has subsided, allopurinol is the drug of choice for prevention of recurrences. 38,39 Gouty arthropathy is rare in children. In a review of alleged cases, Treadwell 40 identified 66 patients younger than 20 years who were reported between 1769 and 1960 and added two additional cases. In many early publications, the exact diagnosis is in doubt. The onset of gouty arthritis in a 14-year-old boy as a result of chronic compensated hemolysis of unknown cause has been described. 41 Yarom and colleagues42 reported two children with marked hyperuricemia, mild renal failure, and acute, episodic, painful swelling of one joint, often the first metatarsophalangeal joint, knee, ankle, elbow, or a proximal interphalangeal joint of the hand. Gout has also been reported in children with glycogen storage disease,43 malignancY,44 and renal failure. 45-47
Lesch-Nyhan Syndrome The Lesch-Nyhan syndrome, first described in 1967 as an X-linked recessive disorder of uric acid metabolism and
634
C HAP T E R
32
Mu SCU LOSKELETA L MANIFESTATIONS OF SYSTEMIC
0
I SEASE
• Hgure 3Z-4 Mseleni joint disease. A, Irregularity and deformity of the distal ends of the ulnar and radius with distraction of the radius from the ulna. 8, Marked deformation of the femoral heads. C. Platyspondyly. (Courtesy of Dr. G. Lockitch.)
central nervous system dysfunction, results from a deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT) (Table 32-4).4~50 It is characterized by the childhood onset of choreoathetosis, spasticity, mental
II
TABLE 32-3
c'llIses of Hyperuricemia dnd Gout
Inaeased Uric Add Produdlon Primary Lesch-Nyhan syndrome Becker's syndrome (phosphoribosyl pyrophosphate synthetase superactivity) Secondary Glycogenosis type I (glucose-6-phosphate dehydrogenase deficiency) Myeloproliferative disorders Lymphoproliferative disorders Severe psoriasis Gaucher's disease Cytotoxic dmgs Hypoxia Chronic hemolysis Secondary polycythemia
Deaeased Uric Add Excretion Reduced glomemlar filtration rate Reduced fractional urate excretion Down syndrome Lead nephropathy Analgesic nephropathy Amyloidosis Sickle cell anemia Sarcoidosis Hypothyroidism Hyperparathyroidism Increased levels of organic acids Type I glycogen storage disease Maple symp urine disease Dmgs Diuretics Salicylates (low dose) Levodopa
retardation, severe growth retardation, self-mutilation, hyperuricemia with increased uric acid synthesis, and uric acid crystalluria. Severity of the disorder is determined by the degree of HPRT deficiency resulting from unique mutations in each family.'i! Affected boys do not develop acute gouty arthritis, however, at least not until the adolescent or adult years. Treatment with allopurinol effectively prevents the rheumatic complaints but does not alter the central nervous system disease, for which there is no effective therapy. An incomplete hereditary deficiency of the enzyme (Le., Kelley-Seegmiller syndrome) may occur in adolescent or adult males as severe gouty arthritis with renal calculi, but it lacks the dramatic neurologic and mutilating characteristics of the complete enzyme deficiency.
Phosphorlbosyl Pyrophosphate Synthetase Superactlvlty An X-linked mutation resulting in excessive activity of
phosphoribosyl pyrophosphate synthetase (PPRPS), the enzyme that converts ribose-5-phosphate to PP-ribosephosphate, results in increased purine production and gout in children and young adults, sometimes with neurologic deficits and sensorineural deafness. 52, 'i3 Allopurinol effectively controls this disorder (Becker's syndrome).
I=: II
TABLE 32 -4
Lesch-Nyhan Syndrome
Clinical characteristics
Genetics Biochemical defect Laboratory findings
Progressive development of choreoathetosis, spasticity, and mental retardation with self-mutilation X-linked recessive Deficiency of hypoxanthine-guanine phosphoribosyltransferase Hypemricemia and uric acid crystalluria
C HAP T E R
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
Glucose-6-Phosphatase Deficienc.y Glycogen storage disease type I (Le., von Gierke's disease) may be associated with the onset of gouty arthritis in childhood. 54,55 Children with this disorder are stunted and have marked hepatosplenomegaly, progressive mental retardation, abnormalities of platelet function, and hypoglycemia. Hyperuricemia results from increased catabolism of adenosine triphosphate and decreased urate excretion.
Caldum Pyrophosphate Deposition Disease Crystals of calcium pyrophosphate dihydrate (CPPD) in synovial fluid and joint structures are associated with a ohronic inflammatory and degenerative joint disease (Le., pseudogout).37,56 The wrists, knees, shoulders, and ankles are most commonly affected. Synovial fluid CPPD crystals are positively birefringent when viewed through a compensated polarized light microscope and are shorter than urate crystals. Chondrocalcinosis, the depositipn of CPPD crystals in hyaline cartilage and fibrocartilage, is a disorder of the adult. Radiographs demonstrate linear calcifications in the menisci of the knee and in other cartilaginous structures such as the triangular cartilage of the wrist.57,58 In descriptions of familial chondrocalcinosis, however, there have been rare reports of adolescents in whom the disorder presented as an acute, self-limited polyarthritis, often precipitated by exercise or trauma. 59 The characteristics of the clinical disease have vatied, however, depending on the kindred, age at onset, severity, and the presence of an associated osteoarthritis or chondrodysplasia.
Ochronosis Ochronosis (Le., alkaptonuria) is an autosomal recessive defect in homogentisic acid oxidase involved in the metabolism of phenylalanine and tyrosine. This abnormality results in the accumulation of homogentisic acid in tissues, and pigmentation of cartilage (e.g., ears, sclerae, heart valves), calcification and ossification of the intervertebral disks, accelerated osteoporosis and osteoarthritis, and vascular disease. 60.61 A secondary defect, inhibition of lysyl hydroxylation, leads to impairment of collagen cross-linking. Arthritis becomes symptomatic only in midadult life and has not been reported in children. Black urine or staining of the diapers is often the sign that prompts referral of the child with this metabolic defect.
Hyperllpoprotelnemla Defects in lipoprotein metabolism are associated with a high risk of premature atherosclerosis, coronary artery disease, and musculoskeletal abnormalities. 62 Articular and tendinous swelling accompany essential familial hypercholesterolemia and hypertriglyceridemia; both of these conditions are inherited as autosomal dominant traits.62~4 Homozygotes are most severely involved. In type II hyperlipoproteinemia (i.e., familial hypercholesterolemia), the Achilles, patellar, and extensor tendons of the hands are the principal locations of xanthomata. 65 .66
635
These lesions are associated with recurrent episodes of an acute migratory polyarthritis. In type IV hyperlipoproteinemia (Le., hypertriglyceridemia), the hands, knees, and ankles are primarily affected by mild chronic or migratory oligoarthritis. 67 The onset is often acute, and fever and an elevated white blood cell count may occur. The arthritis is self-limited but may be misdiagnosed as acute rheumatic fever, especially if the tendon xanthomata are mistaken for nodules. Large and small joints may be involved by symmetric oligoarthritis. Xanthomata of the tendons also occur in sitosterolemia, a syndrome resulting from accumulation of sterols derived from vegetable sources. The xanthomata initially appear in childhood and usually involve the extensor tendons of the hands and, later, the patellar, Achilles, and plantar tendons. Plasma sterol levels are elevated, and cholesterol levels may be increased. 68.69
HEMATOLOGIC DISORDERS Hemoglobinopathies Homozygous sickle cell disease7o .71 and p-thalassemian -74 result in severe rheumatic manifestations. Musculoskeletal symptoms are caused by secondary skeletal changes that accompany hematopoietic expansion of the bone marrow and repeated episodes of avascular necrosis.75 Sickle cell anemia is an autosomal dominant trait and occurs almost exclusively in black children.76 Acute arthritis and long bone pain may be severe and incapacitating during sickle cell crises. In the infant, dactylitis and periostitis of the small bones of the hands may cause painful swollen extremities and the hand-foot syndrome. 70 Each acute episode lasts 1 to 3 weeks and is characterized by diffuse, symmetric, painful swelling of the hands or feet. In a 1997 review, dactylitis occurred before 1 year of age in 41 00%) of 392 children and was a predictor of severe disease in later life. 77 Osteonecrosis may occur in any bone and leads to marked abnormalities of growth and deformity. The hip is particularly vulnerable and is the usual site of the septic arthritis caused by Salmonella or other species to which these children are unduly susceptible. Differentiation of sickle cell bone infarction from osteomyelitis is sometimes difficult and is aided by scintigraphy, ultrasonography, and magnetic resonance imaging. 78 The thalassemias are a group of heterogeneous syndromes of inherited hypochronic anemias of widely differing degrees of severity. Thalassemia minor is characterized by anemia, hepatosplenomegaly, and recurrent brief episodes of joint pain, swelling, and effuSion, especially in the ankles.n.73.79
Hemophilia Recurrent intra-articular hemorrhage is a hallmark of classic hemophilia A (Le. factor VIII deficiency) and is one of the most important causes of morbidity in this X-linked recessive coagulopathy.80 The frequency of episodes of hemarthrosis is related to the plasma concentration of factor VIII. Levels higher than 5% of normal are associated
636
C HAP T E R
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
with hemarthrosis in approximately one half of the cases, whereas hemarthroses almost invariably occur in children with lower levels of factor VIII. B1 A similar association is found in von Willebrand's disease. B2 Hemarthrosis can occur even before the child starts walking, and the frequency of episodes increases during the early childhood years. The joints most commonly affected are the knees, elbows, and ankles. B3 Bleeding into the small joints of the hands, feet, or spine is unusual. Hemorrhage into soft tissues, especially muscle, may be severe and can mimic hemarthrosis.B4,Bs Acute hemarthrosis is signaled by onset over a few minutes to an hour of increasing pain, a feeling of fullness in the joints, and loss of range of motion. The joint is warm and distended. Resorption of the hemarthrosis takes place over several days with effective factor VIII replacement. Intra-articular bleeds, however, tend to be recurrent, and lead to secondary proliferation of synovium with hemosiderosis that produces a diffuse increase in the density of the soft tissues on radiographs that is highly suggestive of the diagnosis. These debilitating changes may develop in as little as 1 to 2 years. Radiographic abnormalities range from changes in the density of soft tissues to epiphyseal overgrowth, osteoporosis, subchondral cyst formation and bony sclerosis, squaring of the patella, narrowing of the joint space, and eventually, osteoarthritis (Fig. 32-5).B6 Widening of the femoral intercondylar notch is a characteristic finding. Intensive physical therapy with strengthening of the muscles around affected joints helps to prevent hemarthroses. 77 ,B7 Management of the acute bleed consists of factor VIII replacement,B3,BB application of ice to the affected joint, splinting, and rest. Agents that affect coagulation (e.g., NSAIDs) should be avoided. Joint aspiration has only a limited therapeutic role and must be preceded by factor VIII administration. B6 Arthrocentesis accompanied by intra-articular glucocorticoid is some-
times dramatically effective in redUcing the severity and frequency of hemarthroses,89 as is prophylactic administration of factor VIII. Surgical synovectomy with or without a sclerosing agent (radioactive or chemical) has a place in treating the older child with early destmctive changes. 90,91
DISORDERS OF ENDOCRINE AND EXOCRINE GLANDS Diabetes Mellitus With the exception of diabetes mellitus, musculoskeletal disease is rarely associated with endocrinopathies in childhood. Rosenbloom and coworkers"7-70 described a syndrome of juvenile-onset insulin-dependent diabetes mellitus, short stature, and contractures of the finger joints: diabetic cheiroarthropathy or stiff:hand syndrome (Fig. 32-6 and Table 32-5\ In a survey of 229 diabetics aged 7 to 18 years, 29% had flexion contractures of one or more joints of the fingers, most often the proximal interphalangeal joints of the fifth or fourth fingers. 92 In a few children, flexion contractures occurred in other joints (e.g" wrists, elbows, ankles, toes, knees), and spinal motion was decreased. In most instances, the child was unaware of any joint limitations and had no pain. Functional disability was uncommon. The prevalence of joint contractures increased from less than 10% in those with diabetes for less than 1 year to close to 50% in children with disease for longer than 9 years. 92 However, there did not appear to be a correlation with the severity of the diabetes or adequacy of its control. Tightening of the skin over the distal phalanges mimicked the acrosclerosis of scleroderma. The precise relation of diabetes mellitus to contractures is unknown. Studies have demonstrated increased gluco-
• Figure 32-5 Hemophilic: arthropathy. A. Normal ankle. B. Recurrent hemarthroses resulted In arthritis characterized by a loss of joint space and development of atalar osteophyte. (Courtesy of Dr. R. Cairns.)
C HAP T E R
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
637
• FItI-e 3Z-6
Diabetic cheiroanhropathy with soft tissue contraetures that limit extension of the metacarpophalangeal and interphalangeal joints of the hands (.4) and flekor tendon contractures in the palm &). In this 16-year-old boy, multiple flexion contractures without evidence of intra-articular or muscle inflammation had failed to respond to physical therapy over a2-year period. Adiagnosis of insulin-dependent diabetes mellitus was made, and 2 months after institution of insulin therapy, flexion contraetures were considt!rably improved.
sylation of collagen in this syndrome. 93 Perhaps increased cross-linking of collagen leads to the contractures. NSAIDs have no beneficial effect on this particular disorder. The frequency of juvenile rheumatoid arthritis QRA) may be increased in children with diabetes mellitus 94 (see Chapter 9). Although Rosenbloom and colleagues95 found that their patients did not have thickening of the palmar fascia, the incidence of Dupuytren's contractures is ir)creased in adults with diabetes mellitus.% Occasionally, hemochromatosis occurs with diabetes mellitus and leads to an arthropathy that results in a characteristic bony enlargement of the second and third metacarpophalangeal joints; other joints are affected less commonly.97 This disorder has not been documented in childhood. Juvenile hemochromatosis is a rare autosomal recessive disorder Clq2D that results in iron overload lending to hypogonadism and cardiorp.yopathy.98,99 Diabetic osteopathy is characterized by pain and osteoporosis of the distal metatarsal heads that may progress to erosion or even complete resorption of the ends of the hones. 100 This cause of this phenomenon is unknown, and the disorder has not been recorded in childhood.
Other Disorders of Endoulne Glands Hyperparathyroidism is rare in children; in adults, it may be characterized by fever, abdominal pain, musculoskeletal pain, osteoporosis,101 mental disturbances, and
'~1Ii TAB~[ 32-5
Musculoskeletal Complications of Diabetes
Mplhtus
headaches. Elevation of serum parathormone levels confirms the diagnosis. 102 Pseudobypoparatbyroidism and pseudopseudohypoparatbyroidism are classified as forms of acromelic dysplasia (see Chapter 40). Hyperthyroidism and hypothyroidism can be associated with diffuse musculoskeletal pain and muscle weakness, although these disorders and their complications appear to be rare in childhood. Hashimoto's thyroiditis can complicate systemic lupus erythematosus,103 and autoimmune hyperthyroidism (Le., Graves' disease) is occasionally associated with ]RA. Thyroid acropachy is a rare form of hyperostosis of the phalanges, metacarpals, and metatarsals that is associated with hyperthyroidism, pretibial myxedema, exophthalmos, and clubbing. 104
CYSTIC FIBROSIS Musculoskeletal disease occurs in a small proportion of children with cystic fibrosis (Table 32-6).105 Some children develop hypertrophic osteoarthropathy, others episodic arthritis, and less commonly, erosive arthritis. Secondary hypertrophic osteoarthropathy occurs in approximately 5% of children with cystic fibrosis.I06,107 The occurrence of ]RA, including rheumatoid factor-positive disease, has been reportedYlB Episodic arthritis appears to be the most common musculoskeletal complication. I09-112 In one study,l09 3 boys and 2 girls, aged 2 to 20 years, had
,-11 Ii -
Rosenbloom's syndrome Dupuytren's contracture Hemochromatosis Diabetic osteolysis
Short stature, tight skin over distal digits, diabetic cheiroarthropathy Rare in childhood Rare in childhood Not recorded in childhood
TABLE 32-6 Musculoskelplal Disordprs Rpportpd Cystic Fibrosis
Hypertrophic osteoarthropathy Episodic arthritis with rash Juvenile rheumatoid arthritis Sarcoidosis
III
638
C HAP T E R
32
Muscu LOSKELE TAL MAN IFESTA no NS OF SYSTEMIC DISEASE
episodes of arthritis lasting 1 to 10 days that recurred at intervals of weeks to months. One or more joints were affected during each episode. A pruritic nodular rash occurred in all 5 children. Results of serologic studies for rheumatoid factors and antinuclear antibodies were negative, and radiographs demonstrated no abnormalities. The cause and pathogenesis of this self-limited arthropathy are unknown, but it may be a reaction to chronic bacterial infection in the lung. There is no evidence that the use of fluoroquinolones in the management of cystic fibrosis leads to arthropathy.1l3 A child with cystic fibrosis complicated by sarcoidosis affecting the knee joints has been reported. 114
HYPEROSTOSIS Hyperostosis, the abnormal subperiosteal or endochondral deposition of bone, is characteristic of a number of unrelated disorders (Table 32-7). Hypertrophic osteoarthropathy is a type of hyperostosis in which there is clubbing of the fingers and toes, painful subperiosteal apposition of new bone along the shafts of the long bones, and occasionally, arthritis. In children, it may be a sporadic or hereditary disease, or it may develop secondary to suppurative lung disease, inflammatory bowel disease, malignancy, or tumor metastatic to lung, pleura, or mediastinum (see Chapter 39). Radiographs are characterized by a distinctive periosteal new-bone apposition, soft tissue swelling, and joint effusions. Bone scintigraphy demonstrates increased isotope uptake in the areas of new-bone formation. Asymptomatic, isolated clubbing can occur in children with cyanotic congenital heart disease. Familial clubbing can develop without associated systemic disease and is usually asymptomatic. The Goldbloom syndrome is a form of idiopathic, periosteal new-bone formation associated with fever, constitutional symptoms, severe pain in the extremities, and elevated serum immunoglobulin levels and an increased erythrocyte sedimentation rate. 1l5 ,116 Radiographs demonstrate typical periosteal new-bone apposition along the long bones. The child may develop limited motion in contiguous joints and refuse to walk if the lower extremities are involved. The disorder runs a chronic course over several months; a spontaneous recovery is expected. NSAIDs are sometimes useful for symptomatic control.
I!: II
TABI E J2 7 ClUS('S of S('( ond,uy "Ylwrtrophi( Ost"oarthrolJdthy
Malignant metastases to chest (lungs, pleura, mediastinum) Osteosarcoma Neuroblastoma Lymphoma Chronic suppurative pulmonary disease Cystic fibrosis Cyanotic congenital heart disease Gastrointestinal disease Inflammatory bowel disease Biliary cirrhosis or ductal atresia Thyroid acropachy
There is no known cause, but the disorder may follow an infectious disease or viral syndrome. Several of the osteochondrodysplasias characterized by increased bone density should be considered in the differential diagnosis of hyperostosis. These include diaphyseal dysplasia (i.e., Camurati-Engelmann disease), craniodiaphyseal dysplasia (Le., leontiasis ossea), cherubism,jamilial infantile cortical hyperostosis (i.e., Caffey's disease), and melorheostosis (see Chapter 40). Children may also develop localized areas of periostitis from repeated episodes of musculoskeletal trauma. l17,1 18 Pachydermoperiostosis is a rare, autosomal recessive disorder characterized by digital clubbing, excessive oiliness, and hypertrophic skin changes (Le., cutis verticis gyrata), occasional gynecomastia and female hair distribution, enlargement of the extremities, painful and swollen joints, and hypertrophic osteoarthropathy.119,120
PANCREATITIS WITH ARTHRITIS Acute or chronic pancreatitis or pseudocyst formation from trauma to the pancreas may be accompanied by disseminated fat necrosis, leading to the development of subcutaneous nodules and osteolytic lesions resembling multicentric osteomyelitis or arthritis. 121-124 The nodules are tender, erythematous, widely disseminated, and similar to those of erythema nodosum. They are often accompanied by systemic illness and fever. Joint pains and effusions may develop 2 to 3 weeks later. The arthropathy is usually self-limited and remits spontaneously, Soft tissue swelling is evident on radiographs, which show multiple sites of periosteal new-bone apposition and diaphyseal lytic lesions. Because the bony lesions are delayed in appearance by a few months, the initiating abdominal trauma may have been forgotten. Diagnosis is confirmed during the acute illness by elevation of the serum lipase and amylase concentrations. A bone scan may demonstrate increased uptake of isotope in the metaphyses or diaphyses because of the infarctions that have resulted from the disseminated intravascular fat. Differential diagnosis includes histiocytic cytophagic panniculitis,125 erythema induratum (often related to tuberculosis),126 and aI-antitrypsin dejiciency.127 A form of lobular panniculitis, Weber-Christian disease, may mimic rheumatic disease. It usually occurs in adult women but has been described in infancy and young children. Widespread subcutaneous nodules, lymphadenopathy and splenomegaly, fever, malaise and arthralgia are characteristic. The course may be limited in time or recurrent. 128
SPHINGOLIPIDOSES In the sphingolipidoses, lipid accumulates in cells as a result of specific enzyme deficiencies. 129 Of the many different sphingolipidoses, three have prominent musculoskeletal signs and symptoms (Table 32-8). Farber's lipogranulomatosis is an autosomal recessive disorder marked in the neonatal period by a hoarse cry and irritability.130,13 1 Painful red masses develop along tendon sheaths and over pressure points, as well as around the
C HAP T E R
I:.
IABLI:. 32-8
32
Muscu LOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
639
Sphingolipidoses
GeneUcs
Musculoskeletal AbnormallUes
Farber's disease
AR
Gaucher's disease
AR
Fabry's disease
XR
Painful red masses along tendons at wrists, elbows, knees, and ankles Osteoporosis with pathologic fractures of femur and vertebrae Recurrent fever and severe distal arthritis with burning pain; rash
AR, autosomal recessive; XR, X-linked recessive.
joints, especially the wrists, small joints of the hands and feet, elbows, knees, and ankles. Nodules have also been described in conjunctivae, ears, and nares. Epiglottal and laryngeal swelling results in repeated pulmonary infections, leading to death by about 2 years of age. Delayed motor development and mental retardation are prominent. The basic process underlying this disease is the cytoplasmic accumulation of a glycolipid ceramide in fibroblasts, histiocytes, macrophages, and neurons, attributable to a deficiency of lysosomal acid ceramidase. The central nervous system, retina, respiratory tract, heart, liver, spleen, lymph nodes, synovium, and bone are all affected to various degrees. Radiographic changes in the skeleton consist of osteoporosis, juxta-articular erosions, and, disruption of the normal trabecular pattern. In Gaucher's disease, an autosomal recessive disorder, glucocerebroside accumulates in the reticuloendothelial cells of the bone marrow, spleen, liver, lymph nodes, and viscera as a result of deficiency of glucocerebrosidase. Hepatosplenomegaly and pathologic fractures of the femur or vertebrae suggest the diagnosis. Premature osteoarthritis of weight-bearing joints is an important feature of the juvenile form of this disease. One of the diagnostic hallmarks of Gaucher's disease is widening of the distal femur. Characteristic areas of rarefaction and osteoporosis are visible in the peripheral and axial skeleton, including the skull. Fabry's disease is characterized by the progressive accumulation of birefringent deposits of triglycosylceramide in the endothelial, perithelial, and smooth muscle cells of blood vessels and in ganglion and perineural cells of the autonomic nervous system. 2 The disease results from an X-linked recessive deficiency of ceramide trihexosidase. Affected boys in late childhood or adolescence have recurrent attacks of fever and severe arthritis and a characteristic burning, tingling pain in the extremities that is aggravated by hot weather or exercise. The fingers, elbows, and knees may become swollen, and a characteristic deformity limiting extension of the fingers develops. Other bones may also be involved, and secondary effects of osteonecrosis become increasingly important, especially in weight-bearing joints such as the hips. A typical rash consisting of purple papules, angiokeratoma c01poriS diffusum universale, accompanies the other features of Fabry's disease. Enzyme activity may be assayed on skin fibroblasts and leukocytes. Female heterozygotes may develop milder forms of this disorder.
• Rgure 32-7 Multicentric reticulohistiocytosis in a 14-year-old boy.There is marked swelling and subluxation of the wrist and swelling of the distal interphalangeal joints of the fingers and the interphalangeal joint of the thumb. Cutaneous nodules are visible over the thumb.
Renal, cardiac, or cerebral disease leads to death in the mid-adult years in untreated patients. Recombinant (lgalactosidase enzyme replacement therapy and renal allograft transplantation, if renal failure has developed, correct the metabolic defect. 132 A number of other rare disorders present in a manner similar to that of the diseases discussed earlier, but they have not been clearly identified as involving a lysosomal degradative enzyme. One such entity, multicentric reticulohistiocytosis, or lipoid dermatoarthritis, is a rare, mutilating, symmetric polyarthritis. 133,I34 An important diagnostic clue is the presence of clear histiocytic cutaneous nodules (Fig. 32-7). Stiffness and contractures appear early, and the joints (with a predilection for the interphalangeal and metacarpophalangeal joints) are swollen and tender. Biopsy of the lesions of the skin, mucous membranes, or synovium demonstrates lipid-laden histiocytes and foamy multinucleated giant cells. Most described cases have been in adults and are not familial.
REFERENCES C./: Metabolic bone disease: an update. Instr Course Lect 52: 769-784, 2003. Scriver CR, Beaudet AL, Sly WS, Valle D (eds): The Metabolic and Molecular Bases of Inherited Disease, 7th ed. New York, McGraw-Hili, 1995. Abrams SA: Nutritional rickets: an old disease returns. Nutr Rev 60: 111-115, 2002. Wharton B. Bishop N: Rickets. Lancet 362: 1389-1400, 2003. Mishra S, Yadav TP, Nangia S, et al: Vitamin D-dependent rickets type II. Indian Pediatr 33: 334--336, 1996. Holick MF: Vitamin D: A millennium perspective. J Cell Biochem 88: 296-307, 2003, Ono T, Seino Y: Medical management and complications of X-linked hypophosphatemic vitamin D resistam ricket•. Acta Paediatr Jpn 39: 503-507, 1997. Sultan A, Vitale P: Vitamin D-dependem rickets type II with alopecia: two case reports and review of the literature. 1m J Dermatol 42: 682-685, 2003. Sato S, Matsuo N: Genetic analysis of hypophosphatasia. Acta Paecliatr Jpn 39: 528-532, 1997.
I. Mankin HJ, Mankin
2. 3. 4. 5, 6, 7.
8.
9.
640
C HAP T E R
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
10, Jacob RA: Three eras of vitamin C discovery, Subcell Biochem 25: 1-16, 1996, 11. Ratanachu-Ek S, Sukswai P, Jeerathanyasakun Y, et al: Scurvy in pediatric patients: a review of 28 cases, J Med Assoc Thai 86 (Suppl 3): S734-S740, 2003, 12, Perrotta S, Nobili B, Rossi F, et al: Vitamin A and infancy, Biochemical, functional, and clinical aspects, Vitam Horm 66: 457-591, 2003, 13, Pease CN: Focal re~ardation and arrestment of growth of bones due to vitamin A intoxication, JAMA 182: 980--985, 1962, 14, Christensen WR, Liebman C, Sosman MC: Skeletal and periarticular manifestations of hypervitaminosis 0, Am J Roentgenol Radium TIler Nucl Med 65: 27-41, 1951. 15, Barot VV: Occurrence of endemic fluorosis in human population of North Gujarat, India: human health risk. Bull Environ Contam Toxicol61: 303-310, 1998. 16, Levy SM: An update on fluorides and fluorosis, J Can Dent Assoc 69: 286-291, 2003, 17, Fisher RL, Medcalf 1W, Henderson Me: Endemic fluorosis with spinal cord compression, A case report and review, Arch Intern Med 149: 697-700, 1989, 18, Nesterov AI: The clinical course of Kashin-Beck disease, Arthritis Rheum 26: 29-40, 1964, 19, Takamori T: Kashin-Beck's disease, Tokyo, University School of Medicine, The Professor Tokio TakamOri Foundation, 1968, 20, Sokoloff L: Endemic forms of osteoarthritis, Clin Rheum Dis 11: 187-202, 1985, 21. Lee YW, Mirocha C], Shroeder OJ, et al: TOP-I, a toxic component causing tibial dyschondroplasia in broiler chickens, and trichothecenes from Fusarium roseum 'Graminearum.' Appl Environ Microbiol 50: 102-107, 1985, 22, Sokoloff L: Kashin-Beck disease, Rheum Dis Clin North Am 13: 101-104, 1987. 23. Tomlinson R: Beijing conference reviews Kashin-Beck disease, BMJ 318: 485, 1999. 24. Zhang WH, Neve .I, XU JP, et al: Selenium, iodine and fungal contamination in Yulin District (People's Republic of China) endemic for Kashin-Beck disease. Int Orthop 25: 188--190,2001. 25. Suetens C, MorenO-Reyes R, Chasseur C, et al: Epidemiological support for a mulrifactorial aetiology of Kashin-Beck disease in Tibet. Int Orthop 25: 180--187, 2001. 26. Moreno-Reyes R, Mathieu F, Boelaert M, et al: Selenium and iodine supplementation of rural Tibetan children affected by Kashin-8eck osteoarthropathy. AmJ Clin Nutr 78: 137-144, 2003. 27. Pasteels JL, Liu FD, Hinsenkamp M, et al: Histology of Kashin-Beck lesions. Int Orthop 25: 151-153, 2001. 28, Mathieu F, Begaux F, Lan IT, et al: Clinical manifestations of Kashin-Beck disease in Nyemo Valley, Tibet. Int Orthop 21: 151-156, 1997. 29, Xiong G: Diagnostic, clinical and radiological characteristics of Kashin-Beck disease in Shaanxi Province, PR China. Int Orthop 25: 147-150, 2001. 30, Yu W, Wang Y, Jiang Y, et al: Kashin-Beck disease in children: radiographic findings in the wrist. Skeletal Radiol 31: 222-225, 2002. 31. Lockitch G, Fellingham SA, Wittman W, et al: Mseleni joint disease: the pilot clinical survey. S Afr MedJ 47: 2283-2293, 1973, 32. Lockitch G, Fellingham SA, Elphlnstone CD: Mseleni joint disease: a radiological study of two affected families. S Afr Med J 47: 2366-2376, 1973. 33, Rodriguez-Merchan EC: Effects of hemophilia on articulations of children and adults. Clin Orthop 7-13, 1996. 34, Miller ST, Sleeper LA, Pegelow CH, et al: Prediction of adverse outcomes in children with sickle cell disease. N Engl] Med 342: 83-89, 2000. 35. Becker MA: Clinical aspects of monosoclium urate monohydrate crystal deposition disease (gout). Rheum Dis Clin North Am 14: 377-394, 1988. 36. Simkin PA: Gout and hyperuricemia, Curr Opin Rheumatol 9: 268-273, 1997. 37, Agudelo CA, Wise CM: Crystal-associated arthritis. Clin Geriatr Med 14: 495-513, 1998. 38. Wortmann RL: Effective management of gout: an analogy, Am] Med 105: 513-514, 1998. 39. Emmerson BT: The management of gout. N EnglJ Med 334: 445-451, 1996. 40. Treadwell BL: Juvenile gout. Ann Rheum Dis 30: 279-284, 1971. 41. Liberman UA, Samuel R, Halabe A, et al: Juvenile metabolic gout caused by chronic compensated hemolytic syndrome. Arthritis Rheum 25: 1264-1266, 1982. 42. Yarom A, Rennebohm RM, Strife F, et al: Juvenile gouty arthritis. Two cases associated with mild renal insufficiency. Am ] Dis ChUd 138: 955-957, 1984, 43. Hoyningen-Huene CB: Gout and glycogen storage disease in preadolescent brothers. Arch Intern Med 118: 471-477, 1966. 44. Morley q, Houston lB, Morris-Jones P: Acute renal failure and gout as presenting features of acute lymphoblastic leukaemia, Arch Dis Child 51: 723-725, 1976, 45. Warren OJ, Sinlmonds HA, Gibson T, et al: Familial gout and renal failure. Arch Dis Child 56: 699-704, 1981. 46. Calabrese G, Simmonds HA, Cameron ]S, et al: Precocious familial gout with reduced fractional urate clearance and normal purine enzymes. QJ Med 75: 441-450, 1990. 47. Foreman ]W, Yudkoff M: Familial hyperuricemia and renal insufficiency. Child Nephrol Urol 10: 115-118, 1990.
48. Lesch M, Nyhan WL: A familial disorder of uric acid metabolism and central nervous system function. Am] Med 36: 561-570, 1964. 49, Seegmiller]E, Rosenbloom FM, Kelley WN: Enzyme defect associated with a sex-linked human neurological disorder and excessive purine synthesis. Science 155: 1682-1684, 1967. 50. Kelley WN, Greene ML, Rosenbloom FM, et al: Hypoxanthine-guanine phosphoribosyltransferase deficiency in goul. Ann Intern Med 70: 155-206, 1969. 51. Nyhan WL: The recognition of Lesch-Nyhan syndrome as an inborn error of purine metabolism. .I Inherit Metab Dis 20: 171-178, 1997. 52. Becker MA, Puig JG, Mateos FA, et al: Inherited superactiviry of phosphoribosylpyrophosphate synthetase: association of uric acid overproduction and sensorineural deafness. Am .I Med 85: 383-390, 1988. 53. Ahmed M, Taylor W, Smith PR, et al: Accelerated transcription of PRPS1 in X-linked overactivity of normal human phosphoribosylpyrophosphate synthetase.] Bioi Chern 274: 7482-7488, 1999, 54. Alepa FP, Howell RR, Klinenberg JR, et al: Relationships between glycogen storage disease and tophaceous gout, Am .I Med 42: 58-66, 1967. 55, Chou]y, Matern 0, Mansfield BC, et al: Type [ glycogen storage diseases: disorders of the glucose-6-phosphatase complex. Curr Mol Med 2: 121-143, 2002. 56. Rosenthal AK: Calcium crystal-associated arthritides. Curr Opin Rheumatol 10: 273-277, 1998. 57. Reginato AJ, Reginato AM: Diseases associated with deposition of calcium pyrophosphate or hydroxyapolite, In Ruddy S, Harris ED ]r, Sledge CB (eds): Kelley's Textbook of Rheumatology, 6th ed, Philadelphia, WB Saunders, 2000, pp 1377-1390. 58, Steinbach LS, Resnick 0: Calcium pyrophosphate dihydrate crystal deposition disease revisited. Radiology 200: 1-9, 1996. 59, Richardson BC, Chafetz Nl, Ferrell LD, et al: Hereditary chondrocalcinosis in a Mexican-American family. Arthritis Rheum 26: 1387-1396, 1983. 60, O'Brien WM, LaDu BN, Bunim ]]: Biochemical, pathologic and clinical aspects of alcaptonuria, ochronosis and ochronotic arthropathy. Review of the world literature 0584-1962). AmJ Med 34: 813, 1963. 61. Dom K, Piltevils T: Ochronotic arthropathy: the black hip, Case report and review of the literature. Acta Orthop Belg 63: 122-125, 1997. 62, Davignon], Genest] ]r: Genetics of lipoprotein disorders. Endocrinol Metal> Clin North Am 27: 521-550, 1998. 63, Franklin FA ]r, Dashti N, Franklin CC: Evaluation and management of dysIipoproteinemia in children. Endocrinol Metal> Clin North Am 27: 641~54, 1998, 64. Cleeman .II: Detection and evaluation of dyslipoproteinemia. Endocrinol Metab Clin North Am 27: 597~11, ix, 1998. 65. Shapiro ]R, Fallat RW, Tsang RC, et al: Achilles tendinitiS and tenosynOVitis, A diagnostic manifestation of familial type 11 hyperlipoproteinemia in children. AmJ Dis Child 128: 486-490,1974. 66. Ose L: Familial hypercholesterolemia from children to adults, Cardiovasc Drugs Ther 16: 289-293, 2002. 67. Buckingham RB, Bole GG, Bassett DR: Polyarthritis associated with type IV hyperlipoproteinemia. Arch Intern Med 135: 286-290, 1975. 68. Be1amarkh PF, Deckelbaum RJ, Stare 1:T, et al: Response to diet and cholestyramine in a patient with sitosterolemia, Pediatrics 86: 977-981, 1990. 69. Bjorkem I, Boberg KM: Inborn errors in bile acid biosynthesis and storage of sterols other than cholesterol. In Scriver CS, Beaudet AL, Sly WS, et al (eds): The Metabolic and Molecular Bases of Inherited Disease, 7th ed. New York, McGraw-Hili, 1995, pp 2073-2102. 70. Weinberg AG, Currarino G: Sickle cell dactylitis: histopathologic observations. Am] Clin Pathol 58: 518-523, 1972. 71. Fixler .I, Styles L: Sickle cell disease. Pediatr Clin North Am 49: 1193-1210, vi, 2002. 72, Gerster JC, Dardel R, Guggi S: Recurrent episodes of arthritis in thalassemia minor, .I Rheumatol 11: 352-354, 1984. 73. Arman MI, Butun B, Doseyen A, et al: Frequency and features of rheumatic findings in thalassaemia minor: a blind controlled study. Br J Rheumatol 31: 197-199, 1992. 74. Lo L, Singer ST: Thalassemia: current approach to an old disease. Pediatr Clin North Am 49: 1165-1191,2002. 75. Jean-Baptiste G, De Ceulaer K: Osteoarticular disorders of haematological origin. Baillieres Best Pract Res Clin Rheumatol 14: 307-323, 2000. 76. Lane PA: Sickle cell disease, Pediatr Clin North Am 43: 639-664, 1996. 77. Gilbert MS, Radomisli TE: Therapeutic options in the management of hemophilic synovitis. Clin Orthop 343: 88-92, 1997. 78. Rifai A, Nyman R: Scintigraphy and ultrasonography in differentiating osteomyelitis from bone infarction in sickle cell disease. Acta Radiol 38: 139-143, 1997. 79. Vichinsky EP: The morbidity of bone disease in thalassemia. Ann N Y Acad Sci 850: 344-348, 1998, 80. Ljung R: Paediatric care of the child with haemophilia. Haemnphilia 8: 178-182, 2002. 81. Arnold WO, Hilgartner MW: Hemophilic arthropathy, Current concepts of pathogenesis and management. ] Bone Joint Surg Am 59: 287-305, 1977. 82. Ahlberg A, Sllwer.J: Arthropathy in von Willebrand's disease, Acta Orthop Scand 41: 539-544, 1970.
C HAP T E R
32
MUSCULOSKELETAL MANIFESTATIONS OF SYSTEMIC DISEASE
83. Ljlmg RC: Can haemophilic arthropathy be prevented? Br J Haematol 101: 215-219, 1998. 84. Alcalay M, Deplas A: Rheumatological management of patients with hemophilia. Part 11: Muscle hematomas and pseudotumors. Joint Bone Spine 69: 556--559, 2002. 85. Rodriguez-Merchan EC: Management of musculoskeletal complications of hemophilia. Semin Thromb Hemost 29: 87-96, 2003. 86. Handelsman JE: The knee joint in hemophilia. Orthop Clin North Am 10: llW--173, 1979. 87. Buzzard BM: Physiotherapy for the prevention of articular contraction in haemophilia. Haemophilia 5 (Suppl 1): 10--15, 1999. 88. Gruppo RA, Brown 0, Wilkes MM, et al: Comparative effectiveness of fuli-
length and B-domain deleted factor YI1I for prophylaxis-a meta-analysis, Haemophilia 9: 251--260, 2003. 89. Kisker CT, Burke C: Double-blind studies on the use of steroids in the treatment of acute hemarthrosis in patients with hemophilia. N Engl J Med 282: 639--642, 1970. 90. Hilgartner MW: Current treatment of hemophilic arthropathy. Curr Opin Pediatr 14: 46--49, 2002. 91. Jburneycake JM, Miller KL, Anderson AM, et al: Arthroscopic synovectomy ir children and adolescents with hemophilia. J Pediatr Hematol Oncol 25: 126--731, 2003. 92. Grgic A, Rosenbloom AL, Weber FT, et al: Joint contracture----
the non-enzymatic glucosylation of collagen in children with poorly controlled insulin dependent diabetes. Pediatr Res IS: 626, 1981. 94. Rudolf MC, Genel M, Tamborlane WV Jr, et al: Juvenile rheumatoid arthritis In children with diabetes mellitus, J Pediatr 99: 519-524, 1981. 95. Rosenbloom AL, Silverstein JH, Lezotte DC, et al: Limited joint mobility in childhood diabetes mellitus indicates increased risk for microvascular disease. N Engl J Med 305: 191--194, 1981. 96. Vijanto JA: Dupuytren's contracture: a review. Semin Arthritis Rheum 3: 155-176, 1973. 97. de Seze S, Solnica J, Mitrovic 0, et al: Joint and bone disorders and I'ypoparathyroidism in hemochromatosis. Semin Arthritis Rheum 2: 71--94, 1972. 98. Pe Gobbi M, Roetto A, Piperno A, et al: Natural history of juvenile haemochromatosis. Br J Haematol 117: 973--979, 2002. 99. Camaschella C, Roetto A, De Gobbi M: Juvenile hemochromatosis. Semin Hematol 39: 242-248, 2002.
100. Clouse ME, Gramm HF, Legg M, et al: Diabetic osteoarthropathy. Clinical and roentgenographic observations in 90 cases. Am J Roentgenol Radium Ther Nucl Med 121: 22--34, 1974. 101. Mazzuoli GF, D'Erasmo E, Pisani 0: Prinlary hyperparathyroidism and osteoporosis. Aging (Milano) 10: 225-231, 1998. 102. Bhalla AK: Musculoskeletal manifestations of primary hyperparathyroidism. Clin Rheum Dis 12: 691-705, 1986. 103. Eberhard BA, Laxer RM, Eddy AA, et al: Presence of thyroid abnormalities in children with systemic lupus erythematosus. J Pediatr 119: 277-279, 1991. 104. Kinsella RA Jr, Back OK: Thyroid acropachy. Med Clin North Am 52: 393--398, 1968. 105. Botton E, Saraux A, Laselve H, et al: Musculoskeletal manifestations in cystic fibrosis, Joint Bone Spine 70: 327-335, 2003. 106. Athreya BH, Borns 1', Rosenlund ML: Cystic fibrosis and hypertrophic osteoarthropathy in children. Report of three cases. Am J Dis Child 129: 634-637, 1975. 107. Nathanson I, Riddlesberger MM Jr: Pulmonary hypertrophic osteoarthropathy in cystic fibrosis. Radiology 135: 649--651, 1980.
641
108. Sagransky OM, Greenwald RA, Gorvoy]D: Seropositive rheumatoid arthritis in a patient with cystic fibrosis, Am J Dis Child 134: 319-320, 1980. 109. Newman AJ, Ansell BM: Episodic arthritis in children with cystic fibrosis, Pediatr 94: 594-596, 1979. 110. Schidlow DV, Goldsmith 01', Palmer J, et al: Arthritis in cystic fibrosis. Arch Dis Child 59: 3n-379, 1984. 111. Dixey J, Redington AN, Butler RC, et al: The arthropathy of cystic fibrosis. Ann Rheum Dis 47: 218-223, 1988. 112. Pertuiset E, Menkes CJ, Lenoir G, et al: CystiC fibrosis arthritis. A report of five cases. Br J Rheumatol 31: 535-538, 1992.
113. Warren RW: Rheumatologic aspects of pediatric cystic fibrosis patients treated with fluoroquinolones. Pediatr Infect Dis J 16: 118-122, 1997, 114. Soden M, Tempany E, Bresnilian B: Sarcoid arthropathy in cystic fibrosis. Br J Rheumatol 28: 341-343, 1989. 115. Goldbloom RB, Stein PB, Eisen A, et al: Idiopathic periosteal hyperostosis with dysproteinemia. A new clinical entity. N Engl J Med 28: 873, 1966. 116. Kuwashinla S, Nishinlura G, Harigaya A, et al: A young infant with Goldbloom syndrome. Pediatr Int 41: 110--112, 1999. 117, Grossfeld SL, Van Heest A, Arendt E, et al: Pitcher's periostitis. A case report. Am J Sports Med 26: 303--307, 1998. 118. Craver RD, Correa-Gracian H, Heinrich S: Florid reactive periostitis. Hum Pathol 28: 745-747, 1997. 119. Sinha GP, Curtis 1', Haigh 0, et al: Pachydermoperiostosis in childhood. Br J Rheumatol 36: 1224-1227, 1997. 120, Loredo R, Pathria MN, Salonen 0, et al: Magnetic resonance inlaging in pachydermoperiostosis, Clin Imaging 20: 212-218, 1996. 121. Shackelford pG: Osseous lesions and pancreatitis, Am J Dis Child 131: 731-732, 1977. 122. Goluboff N, Cram R, Ramgotra B, et al: Polyarthritis and bone lesions complicating traumatic pancreatitis in two children. Can Med Assoc J 118: 924-928, 1978. 123. Buntain WL, Wood JB, Woolley MM: Pancreatitis in childhood, J Pediatr Surg 13: 143--149, 1978. 124. Lopez A, Garcia-EstanJ, Marras C, et al: Pancreatitis associated with pleuralmediastinal pseudocyst, panniculitis and polyarthritis. Clin Rheumatol 17: 335-339, 1998. 125, Alegre VA, Winkelmann RK: Histiocytic cytophagic panniculitis. J Am Acad Dermatol 20: 177-185, 1989, 126, Schneider]W, Jordaan HF, Geiger DH, et al: Erythema induratum of Bazin. A clinicopathological study of 20 cases and detection of Mycobacterium
tuberculosis DNA in skin lesions by polymerase chain reaction. Am J Dermatopathol 17: 350--356, 1995. 127. Smith KC, Pittelkow MR, Su WP: Panniculitis associated with severe alpha 1antitrypsin deficiency, Treatment and review of the literature. Arch Dernlatol 123: 1655-1661, 1987, 128, White]W Jr, Winkelmann RK: Weber-Christian panniculitis: a review of 30 cases with this diagnosis. J Am Acad Dermatol 39: 56--62, 1998. 129. Kolter T, Sandhoff K: Recent advances in the biochemistry of sphingolipidoses. Brain Pathol 8: 79-100, 1998. 130. Farber S, Cohen J, Uzman LL: Lipogranulomatosis; a new lipo-glycoprotein storage disease, J Mt Sinai Hosp NY 24: 816--837, 1957. 131. Toppet M, Vamos-Hurwitz E, Jonniaux G, et al: Farber's disease as a ceramidosis: clinical, radiological and biochemical aspects. Acta Paediatr Scand 67: 113--119, 1978. 132. Brady RO: Therapy for the sphingolipidoses. Arch Neurol 55: 1055--1056, 1998. 133. Zayid 1, Farraj S: Familial histiocytic dermatoarthritis. A new syndrome. Am J Med 54: 793--800, 1973. 134. Uhl M, GutfleischJ, Rother E, et al: Multicentric reticulohistiocytosis. A report of 3 cases and review of literature, Bildgebung 63: 126--129, 1996.
( HAP T E R
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES Nlco M. Wulffraat, Lleke A. M. Sanders and Wletse Kuls
I-WI' Genetic disorders of the immune system enable us to study the relation between the clinical expression of immunodeficiencies and the underlying immune defect. Although infections are the most common and early clinical expressions of primary immunodeficiencies, autoimmune diseases and malignancies often occur in immunodeficient patients. In recent years, the genetic basis of immunodeficiencies has become better understood, and the molecular abnormalities of these diseases have begun to be unraveled. An increasing number of associated genetic defects have been identified by a variety of techniques, such as positional cloning and complementation. Understanding the molecular basis of immunodeficiencies and the consequences for the immune system helps to explain why autoimmune diseases develop in certain circumstances. With this knowledge, we can link observations of immunodeficient patients with autoimmune diseases and a known gene defect to patients with comparable autoimmune diseases for whom there is no clear understanding of pathogenesis. In this paradigm, autoimmune diseases are regarded as subtle immunodeficiencies (Table 33-1). The expectation is that this approach will help clarify the pathogenesis of many autoimmune diseases. This chapter focuses on genetically determined primary immunodeficiencies in which autoimmune disorders may occur.
DISORDERS OF INNATE IMMUNITY ASSOCIATED WITH RHEUMATIC DISEASES Defective Control of L,mphocyte Survival Apoptosis (Le., programmed cell death) is an essential phYSiologic mechanism to regulate embryonic development, cell differentiation, and tissue turnover. Of the several mechanisms leading to apoptosis, the best studied is the death pathway initiated by the interaction of CD95 (Fas/APO-l) and its ligand. l --4 The molecular pathway of this process was defined. After binding of Fas ligand to the extracellular part of the Fas molecule, the so-called death domain of this molecule associates with Fas-associated death domain (FADD) and procaspase-8 and
642
-10.5-41 This mutation results in a syndrome characterized
by lymphoproliferation of CD4-CD8- T cells and associated with autoimmune manifestations. The severity of the disease depends on mutations in the Fas-encoding gene and the genetic background of the mice. Mutations of the Fas-ligand gene (Le., GLD mutation of the protein) also result in lymphoproliferation. 8 The human counterpart of this murine abnormality, the autoimmune lymphoproliferative syndrome (ALPS, also called Canale-Smith syndrome), is a lymphoproliferative disorder with accumulation of CD4-CD8- T cells and B cells that produces variable autoimmunity. 5.9. \() Subtypes of ALPS (Le., types la, Ib, II and III) result from defects in different parts of the Fas pathway. In this homozygous Fas deficiency, lymphoproliferation is already present at birth. Stimulated lymphocytes do not express Fas and are insensitive to treatment by an agonist anti-Fas antibody.9-lJ Apoptotic lymphocytes can be detected in the peripheral blood and in the spleen, indicating that other death pathways are operating. Treatment with the antimalarial drug pyrimethamine caused improvement in six of seven ALPS patients (two type la and five type I1I).12 For the more severe cases, allogeneic bone marrow transplantation can be performed.9.13.14 Heterozygous Fas gene (FAS) mutations are more common. ll .1S-17 More than 60 different mutations have been found in the Fas gene, with most affecting the intracellular domain of the Fas molecule and leading to defective Fas-mediated apoptosis. Patients who are heterozygous for a Fas gene mutation are characterized by lymphadenopathy at an early age, autoimmune hemolytic anemia, and thrombocytopenia. Glomerulonephritis, the Guillain-Barre syndrome, and urticaria occur less frequently. Parents of affected children have Fas mutations without clinical symptoms of lymphoproliferation or autoimmunity. This important observation indicates that for the disease to be expressed, the single-allele Fas mutation must be combined with another gene defect (Le., digenic disease).9 Mutations in the Fas-ligand gene (FASLG) can also result in lymphoproliferative diseases associated with autoimmunity. Mutations have been detected in a patient with systemic lupus erythematosus (SLE).18 Molecular cloning and sequencing indicated that the genomic DNA of this patient contained an 84-bp deletion within exon 4
C HAP T E R
~~.
IABIE B-1
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
Classification oflmmunodeficien
Disetders of Innate Immunity Abnormalities of Fas-mediated apoptosis Phagocytic abnormalities Complement abnormalities
Disorders of Adaptive Immunity Abnormalities of T and B lymphocytes Abnormalities of T lymphocytes Abnormalities of B lymphocytes
of the Fas-ligand gene, resulting in a predicted 28-amino acid in-frame deletion. To complicate the picture further, patients are described with lymphoproliferation and autoimmunity with normal Fas expression and Fasindllced apoptosis of lymphocytes, indicating the presence of defects in other death pathways.19 Knowledge of the basic mechanisms controlling cell sunrival or death and the identification of genetic defects in death pathways have led to new concepts of the pathogenesis of autoimmune disease. In addition to Fasinduced apoptosis, many other death pathways are being discovered. A challenge for future research is to evaluate the role of these pathways in preventing autoimmune disease and to recognize defects leading to disease.
Rh.-matlc Diseases Assodated with Disorders
r II ,-
643
TABU B l Classification of Deficiencies of Innat I' Immunity Associated with Rheum"ti< Disease
Disorders of Innate immunity
Rheumatic DIsease AssocIation
Phagocytic Defects Chronic granulomatous disease Familial lipochrome histiocytosis Chediak-Higashi disease Streaking leukocyte syndrome
DLE,S1£ Arthritis SLE-like disease in animals Polyarthritis
Complement Defldencles Deficiency Deficiency Deficiency Deficiency Deficiency Deficiency
of Clq of C1r of Cis of Cl INH of C4 of C2
SLE-like GTN, RTS SLE, OLE, GTN SLE, OLE SLE, OLE 51£, Sjogren's syndrome 51£, OLE, PM, HSP, vasculitis, GTN, Hodgkin's disease, jRA,RA
Deficiency of C3 Deficiency of C5 Deficiency of C6 Deficiency of C7 Deficiency of C8
SLE, vasculitis, GTN, arthralgias, SLE SLE SLE, DLE SLE, sclerodactyly, RA, vasculitis SLE, ]RA
DLE, discoid lupus erythematosus; GTN, glomerulotubulonephritis; HSP, Henoch-Sch6nlein purpura; ]RA, juvenile rheumatoid arthritis; PM, polymyositis; RA, rheumatoid arthritis; RTS, Rothmund-Thomson syndrome (congenital poikiloderma); SLE, systemic lupus erythematosus. Adapted from Ruddy S: Complement deficiencies and rheumatic diseases. In Kelley WN, Harris ED Jr, Ruddy S, Sledge CB Ceds): Texthook of Rheumatology, 4th ed. Philadelphia, WE Saunders, 1993, p 1283.
of Phagocytes There is an increasing awareness of the association of rheumatic diseases with abnormalities of phagocytic cell function and the complement pathways (Table 33-2).
Chronic Granulomatous Disease Chronic granulomatous disease (CGO) is a rare (1 case in 250,000 people in the general population), inherited primali)' immunodeficiency of phagocytic leukocytes characterized by recurrent, life-threatening bacterial, fungal, and yeast infections of the subcutaneous tissues, airways, lymph nodes, liver, and bones. 2o The disorder results from absence or malfunction of the reduced form of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase enzyme system that produces superoxide in the prdfessional phagocytic cells (Le., neutrophils, monocytes, macrophages, and eosinophils). Deficiency of this oxidase (which is required for the production of microbicidal oxygert metabolites) renders the phagocytes unable to kill ingested microorganisms. NADPH oxidase consists of several subunits, each encoded by a separate gene. 20 Treatment of CGO consists of antimicrobial prophylaxis and the use of recombinant human interferon-I'. 20,21 In more severe cases, stem cell transplantation can be performed. 22 Seventy percent of patients have the X-linked form of the disease, which is caused by mutations in CYER, the gene that encodes the ~ subunit of cytochrome b558' also cal~ed gp91phox. Mutations in three other subunits cause
autosomally inherited forms of CGO. This concerns the a subunit of cytochrome b 558 (p22phox), needed for stabilization of the cytochrome in the plasma membrane of phago(.]'tes, and the cytoplasmic proteins p47phox and p67phox that translocate to the cytochrome during cell activation, a process needed for induction of the bactericidal enzymatic activity after phagocytosis of microorganisms. 23 The molecular basis of CGO has been extensively reviewed. 24 Clinical variability in CGO is considerable and mainly associated with the degree of residual respiratoli)' burst activity. A mucocutaneous syndrome characterized by discoid lupus erythematosus (OLE), photosensitive dermatitis, and recurrent aphthous stomatitis occurs in up to one third of carriers of the CYER gene of CGO. 2S-29 Occasionally, other rheumatic complaints are documented, and mothers of some patients with CGD have autoantibodies to nuclear antigens. There have been a few descriptions of children with CGO who developed convincing clinical, serologic, and pathologic evidence of SLE25-28; others have features of OLE and photosensitivity.27,3()'-32 However, the frequency of defects in neutrophil function among patients with OLE who lack a family history of CGO is very low. 27 The pathogenesis of these cutaneous lesions of lupus in patients with CGO or carriers is unknown. It has been proposed that a partial defect in bactericidal ability leads to chronic antigen persistence and immune activation, possibly provoking autoantibody formation. Ultraviolet irradiation may be an environmental trigger.
644
C HAP T E R
33 IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
A girl of Chinese ancestry with autosomal recessive CGD (p47phox) was described as having polyarthritis since the age of 4 years that involved the large joints (Le., wrists, ankles, knees, and hip) and small joints (Le., metacarpophalangeal and metatarsophalangeal joints).33 The patient had early morning stiffness and joint swelling with synovial thickening, stiffness, and tenderness. Infections were excluded. She had rheumatoid factor (RF), antinuclear antibodies (ANAs), and anti-dsDNA antibodies. Serum complement levels were not depressed. The patient did not develop SLE-Iike symptoms in the 22-month follow-up period,33 although such symptoms occurred later (R. E. Petty, personal communication, 1998). Severe, deforming arthritis of hands and wrists has dominated the clinical picture. Bland peripheral arthritis and bursitis in two boys with CGD and erosive polyarthritis in a young girl with the disease have been reported. However, apart from susceptibility to septic arthritis and osteomyelitis, children with CGD only rarely have had a rheumatic disease. Suppurative and granulomatous infections in CGD patients are established soon after birth, first at body surfaces normally in contact with bacteria and fungi (Le., skin, airways, and gUt).20 From these areas, infectious organisms may be carried to lymph nodes and internal organs, such as the liver. Failure to contain the infection may result in bacteremia that enables additional infectious foci to develop. The major clinical manifestations of CGD are pyoderma, pneumonia, gastrointestinal involvement, lymphadenitis, liver abscesses, and osteomyelitis. 2o. 34 In contrast to normal children, in whom osteomyelitis usually involves the metaphyseal areas of long bones, patients with CGD more often develop infections of the small bones of the hands and feet. 2o ,34 Often, multiple sites are infected. Aspiration of pus is mandatory for identification of the pathogenic microorganism. A variety of bacterial pathogens have been isolated from the lesions of patients with CGD. 20 Staphylococcus aureus, Staphylococcus epidermidis, and enterobacteria predominate. Common gram-negative organisms include Escherichia coli, Salmonella, Pseudomonas aeruginosa, Pseudomonas cepacia, Klebsiella-Aerobacter, Proteus, Serratia marcescens, Salmonella arizona, and Legionella. Infections with Nocardia species and Mycobacteria (BCG strain) are of special importance. Fungal pathogens isolated most commonly in CGD are Aspergillus species and, to a lesser extent, Candida albicans. The response to viral pathogens is normal, and parasitic infections, except for Pneumocystis carinii, are rare. Patients with CGD may be considerably shorter than expected based on parental height.20,35 This phenomenon is not fully explained. Infections often suppress growth temporarily, but catch-up growth is normal. Protein-calorie malnutrition does not seem to explain the shorter stature. Patients have a normal pubertal growth spurt.
Familial Lipochrome HIstIocytosis A syndrome of familial lipochrome histiocytosis has been described in three female siblings: one developed migratory polyarthritis, RF seropositivity, and rheumatoid nodules at the age of 15 years; a second had recurrent episodes of monarthritis of brief duration; and two had a photosensitive rash. 36 All had increased susceptibility to bacterial infection, hypergammaglobulinemia, and lipochrome pigment in histiocytes of lymph nodes and liver. Subsequent studies revealed defects in polymorphonuclear neutrophil function that were identical to those of patients with CGDY Unlike children with typical CGD, these girls did not develop granulomata, and in this way they resembled girls with Job'S syndrome. 38 The two most severely affected girls died of pulmonary infection when 19 and 34 years old, The exact relation of this disorder to CGD remains to be determined. This syndrome must be differentiated from the
hyper IgE syndrome, which is also associated with autoimmune phenomena. 39 The hyper-IgE syndrome is a primary immunodeficiency with elevated immunoglobulin E, eosinophilia, vasculitis, autoimmunity, and central nervous system symptoms.
ChedIak-HIgashI syndrome The rare Chediak-Higashi syndrome is characterized by susceptibility to bacterial infection beginning in early childhood and is associated with the presence of large cytoplasmic granules in neutrophils. The hemophagocytic Iymphohistiocytosis syndrome that characterizes the evolution of the Chediak-Higashi syndrome results from defects affecting intracellular trafficking. 40 ,41 Although there have been no reports of rheumatic disease in patients with this disorder or their relatives, it is intriguing that the same leukocyte anomaly occurs in Aleutian mink that develop a spontaneous lupus-like disease.
ArthrItIs, Pyoderma Gangrenosum, and StreakIng Leukocyte Factor A 2-year-old boy developed massive monarticular joint effusions and later had severe pyoderma gangrenosum after minor trauma. 42 The sterile pyoarthritic episodes, leading to the repetitive erroneous diagnosis of septic arthritis, were ultimately selflimited or could be controlled by prednisone. The pyoderma responded to FK506. A second case was reported by]acobs. 43 Pyoderma gangrenosum can also be seen in inflammatory bowel disease. 44 The mechanism of this disorder remains unknown, although a serum factor that enhanced random migration of leukocytes with a secondary increase in chemotaxis was described,
Rheumatic Diseases Associated with Complement Deficiencies Primary deficiencies of complement are inherited as autosomal recessive traits with the exception of Cl inhibitor deficiency (causing hereditary angioneurotic edema [HANEl, an autosomal dominant disease, and properdin deficiency, an X-linked disorder). The heterozygous state can usually be detected by measuring the level of the particular complement protein in serum. The clinical manifestations of complement deficiencies vary.45--53 Some patients are asymptomatic but most suffer from rheumatic diseases, particularly a syndrome resembling SLE. The clinical findings include early onset of skin lesions resembling DLE, alopecia, photosensitivity, and mild renal and pleuropericardial involvement. The two other main clinical presentations are increased susceptibility to infection (i.e., repeated bacterial infections with pathogens such as Streptococcus pneumoniae and Neisseria meningitidis and viral infections), so and angioedema in the case of BANE. Frequent infections are the predominant manifestation of deficiencies of C3 and factors I and H, the absence of which leads to consumption of C3. Primary or secondary C3 deficiency leads to infections with encapsulated bacteria such as S. pneumoniae, underlining the importance of C3 as a mediator of opsonization. Deficiencies of components of the membrane attack complex C5-C8 particularly predispose to recurrent infections with Neisseria species. 49 .S4-S7
C HAP T E R
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
SLE-like rheumatic disorders are the major clinical manifestations of classic pathway complement deficiencies. 4S ,47,49,58 The frequency and severity of disease vary with each deficiency. SLE was observed in 28 of 30 Clqdeficient persons, 12 of 16 with C4 deficiency, approximately one third with C2 deficiency, but in only 4 of 24 patients with C3 deficiency. These observation~ i~ply a physiologic protective activity of the early activaUon of the classic complement pathway against the development of the immune complex-mediated syndrome SLE. Binding of Cl to immune complexes activates the classic complement pathway, resulting in the cleavage of C4 and C3 to C4b and C3b, which bind covalently to immune complexes, leading to two important effects. 47 ,49,59,60 First, binding of C3b (and, to a lesser extent, of C4b) promotes the solubility of immune complexes. Second, immune complexes are bound by means of C3b and C4b to CRI receptors on peripheral blood cells, mainly erythrocytes, and transported to the liver and spleen. There immune complexes are transferred to fixed macrophages, after which the erythrocytes return to the circulation.5~1 The observation that erythrocytes transport immune comple~es in lupus is demonstrated by the depression of CRI levels in active disease. This defect is also found in other diseases that are accompanied by complement fixation on red blood cells, whether by immune complexes or directly by red blood cell antibodies. 59,61 It has been proposed that failure of the mononuclear phagocytic system to effectively remove immune complexes from the cir
645
inhibitor deficiency, which results in a secondary subtotal deficiency of C2 and C4, is also associated with an increased incidence of autoimmune immune complex disease. 46 ,49 The frequency of complement deficiencies in the general population is probably very low. 65.66 C2 deficiency is still the most frequently recognized component deficiency. Heterozygous deficiency for C2 is estimated to be 1%. In patients with SLE, the rate of homozygous complement component deficiency is also low (estimated to be about 1 case in 2000 women in the United Kingdom). Nevertheless, particularly in children with early-onset (preschool age) SLE-like disease and in instances of familial SLE, hereditary primary deficiencies of complement should be considered.
(1 Deficiency The absence of Clq is the most common abnormality of the first component of complement. 61 About 30 patients with homozygous Clq deficiency have been reported. 67 Of these, 28 suffered from SLE, 1 had DLE alone, and a 38-year-old man was healthy. Several characteristic clinical features of SLE are associated with Clq deficiency.61,67 Disease onset tends to be early, with a median age at onset of 7 years (range, 6 months to 42 years). Rash was present in 25 cases. The SLE may be severe, and 11 had glomerulonephritis. Six patients had central nervous system disease (5 of 6 with grand mal seizures). Seventeen of 23 patients had ANAs, and autoantibodies to extractable nuclear antigens were present in 10 patients (anti-RNP in 6; anti-Sm in 6; anti-Ro in 4). Anti-dsDNA antibodies are unusual in the context of Clq deficiency. Therapy with hydroxychloroquine and oral steroids may relieve some symptoms. Thalidomide may benefit the skin lesions. 68 The authors have treated a Clq deficient girl with fresh-frozen plasma for 7 years. She presented at age 3 years with fever, tonsillitis, and arthralgia and later developed mesangioproliferative glomerulonephritis characterized by IgM, IgG, C3, and C5 (but not Clq) along the glomerular basement membrane. Other evidence of SLE emerged: photosensitive malar dermatitis, alopecia, oral ulcers, Raynaud's phenomenon, livedo reticularis, and severe vasculitis of the palms, fingers, soles, and toes. She had anemia, leukopenia, and thrombocytopenia, and test results for antibodies to dsDNA, Sm, RNP, and cardiolipin were positive. Total hemolytic complement was less than 5% of normal, but the functional activity of the alternative complement pathway was intact. Levels of complement factors C3 and C4 were increased even during active disease; the C2 level was normal. Family investigation confirmed that the patient was homozygous for a gene that resulted in the production of low-molecular-weight Clq that was unable to activate the classic complement pathway,69 Fresh-frozen plasma infusions resulted in clinical and hematologic remission until the onset of menarche, when symptoms of active lupus required reinstitution of prednisone. Results of tests for autoantibodies have remained positive, despite clinical remission. Antibodies to Clq have not been observed. Inherited deficiency of C1r, often with concomitant CIs deficiency, has been reported in eight children, five of whom had
646
C HAP T E R
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
SLE and many of whom also had multiple episodes of upper respiratory tract infections, skin infections, meningitis, unexplained fevers, and glomerulonephritis. sl ,61 BANE, caused by a deficiency in Clq esterase inhibitor, has no known human leukocyte antigen (HLA) association but is associated with Sjogren's syndrome or lupus-like disease in some families. s1 ,s2.7o The authors observed a girl with familial Clq esterase deficiency who developed typical DLE with antibodies to Ro and La but not to dsDNA.
C2 Deficiency Deficiency of the second component of complement is the most common genetic deficiency of complement.65.66.71 Heterozygous C2 deficiency may occur in approximately 1% of the normal population, 1.4% of adults with rheumatoid arthritis, 3.7% of children with juvenile rheumatoid arthritis QRA), and 6% of patients with SLE. 66 About 60% of homozygous and 13% of heterozygous C2-deficient persons have had associated SLE-like disease, manifested by photodermatitis, alopecia, fever, arthritis, and renal disease. The lupus-like disease tends to be somewhat milder than would otherwise be expected, with less clinically significant nephritis but more florid cutaneous lesions. 50.51 Steinsson and colleaguesn successfully treated a 43-yearold woman with homozygous C2 deficiency and SLE with infusions of fresh-frozen plasma and were able to discontinue previously required medications (I.e., prednisone and azathioprine).
C3 Deficiency Almost all reported patients with homozygous C3 deficiency have been infants or young children with severe bacterial infections (e.g., meningitis, pneumonitis, peritonitis, osteomyelitis).51.73.74 Other associations described in children have included SLE, vasculitis, arthralgia, and glomerulonephritis. Renal transplantation in a C3-deficient patient with glomerulonephritis has been successful.
C4 Deficiency At least seven homozygous C4-deficient children have been recorded.49.51.62.64 Five had SLE, one had glomerulonephritis, and three had serious infections. SLE is associated with an extended HLA haplotype that includes the C4A null allele. 62.64
Deficiency of Late Complement Components Deficiency of C5, although unusual, is more common in adults and older children. 46 Both of the reported C5-deficient adolescents had N. meningitidis meningitis, but neither had rheumatic complaints. Absence of c6 has been reported in six children younger than 18 years, most of whom had N. meningitidis meningitis. 75 Although no child with C6, C7, or C8 deficiency and a rheumatic disease has been reported, adults with these deficiencies have developed DLE, Sjogren's syndrome, or SLE. 75-78 We reported a 13-year-old boy with a 6-month history of recurrent fever, an exanthem involving the trunk and
extremities, and arthritis of the wrists, knees, and metacarpophalangeal and proximal interphalangeal joints. 79 He had a deficiency of the ~ subunit of C8. Infection, particularly meningococcal infection, was excluded. Deficiencies of C9 and of components of the alternate complement pathway are rare. 51 ,56
Mannose-Blndlng Lectlns Mannose-binding lectin (MBL) is a serum protein with structural similarities to Clq that has a specific role in the innate immunity.so,sl MBL binds to mannose in the cell surface of bacteria and yeasts, facilitating opsonization by phagocytes and initiating complement activation through the classic pathway. This happens by activation of the mannan-binding lectin-associated serine protease 2 (MASP-2), which then cleaves complement factors C4 and C2, generating the C3 convertase C4bC2b. Activation of C3 initiates the alternative pathway and the formation of the membrane-attack complex. An inherited MASP-2 deficiency has been described in a man with ulcerative colitis since age 13, SLE since age 29, and severe pneumococcal pneumonias. 82 The human MBL2 gene on chromosome 10 has three variant alleles. Mutations in these alleles may cause low serum MBL levels that are associated with an increased risk of infections, although individuals with MBL deficiency may be asymptomatic. sl .83 Because Kawasaki disease is an acute vasculitis with a possible infectious cause, a possible role of the MBL gene in white patients with Kawasaki disease was investigated. 84 Children with Kawasaki disease had a higher frequency of MBL gene mutations compared with healthy children. Similar studies of MBL gene polymorphisms in SLE, Sjogren's syndrome, and sarcoidosis reported contrasting results. S5-90
DISORDERS OF ADAPTIVE IMMUNITY ASSOCIATED WITH RHEUMATIC DISEASES Adaptive immunity is mediated by lymphocytes and their products. Deficiencies are classified according to abnormalities of T or B lymphocytes, or both. Acquired abnormalities of adaptive immunity are common in children with rheumatic diseases. It is thought that these laboratory abnormalities (e.g., hypergammaglobulinemia, altered lymphocyte numbers) reflect a response to the disease rather than a primary abnormality, although the validity of this conclusion is by no means certain. Rare but instructive examples of the association of primary immunodeficiencies and rheumatic diseases are discussed in the following paragraphs (Table 33-3). The association of immunodeficiency and rheumatic disease has been reviewed by several investigators. 91 - 93
Primary Abnormalities of T and B Lymphocytes Some diseases are characterized clinically and immunologically by defects in T and B lymphocytes (Tables 33-4 and 33-5). Research has led to gene identification in a substantial number of these disorders.
C HAP T E R
,~.
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
fABLE 33-3 Disorders of Adaptive Immunity Associated with Rheumatic Disease
Rheumatic Disease AssocIations
D1liorden of Adaptive Immunity CoInblned Immunodeftdendes
Chronic arthritis, vasculitis Chronic arthritis
Wiskott-Aldrich syndrome Immunodeficiency with thymoma (Good's syndrome) Nezelof's syndrome
Chronic arthritis, SLE
Humorallmmunodefldendes ]RA, SLE, RA, others Chronic arthritis, SLE ]RA, SLE
Seilective IgA defiCiency Hypogammaglobulinemia IgG subclass deficiencies
Ii Ii
TABLE 53- 5
647
Primary Humoral Immunodeficiencies
X-linked agammaglobulinemia (Bruton's agammaglobulinemia) X-linked hypogammaglobulinemia with growth hormone deficiency X-linked hyper-IgM Autosomal recessive hyper-IgM IgG subclass deficiencies Selective 19A deficiency Kappa light chain deficiency Antibody deficiency with normal immunoglobulins (Nezelof's syndrome) Common variable immunodeficiency Hyper-IgD syndrome Hyper-IgE syndrome
JRA, juvenile rheumatoid arthritis; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus.
Severe Combined Immunodefidency Severe combined immunodeficiency (SCID) is a rare disorder characterized by severe congenital defects in cellular and humoral immunity.91,94,95 The incidence is approximately 1 case in 75,000 births. Because of absent T cell-mediated immunity, affected children develop severe lung infections with P. carin ii, chronic candidiasis, persistent diarrhea, and failure to thrive, usually within the first year of life. Affected children have lymphoid aplasia, and the thymus usually cannot be detected radiographically. Laboratory tests confirm the presence of agammaglobulinemia (although some maternal IgG can be detected in the first months of life) and T cell lymphopenia with absent in vitro responses to mitogens.
I~.
TABLE 33-4 Primary Immunodeficiencies of T and B Lymphocytes
Inheritance
Gene or Gene Product
DIseue
Pattern
scm scm
XL
y chain
AR
fAK3, RAG1, RAG2,
Adenosine deaminase deficiency Purine nucleoside phosphorylase deficiency MHC class II deficiency
AR
AR
Wiskott-Aldrich syndrome
XL
Reticular dysgenesis !\taxia telangiectasia Omenn's syndrome CD3yand CD3E deficiency CD8 deficiency
AR AR AR AR AR
AR
DiGeorge syndrome
AR
Cartilage-hair hypoplasia
AR
DNA-PK Adenosine deaminase Purine nucleoside phosphorylase CIlTA (also called MHC2TA), RFX5 WASP (also called WAS) AIM RAGJ, RAG2 ? ZAP70 (zeta
chain-associated protein kinase 70 kD) Candidate gene exists Candidate gene exists
AR, autosomal recessive: MHC, major histocompatibility complex; SCID. severe combined immune deficiency; XL, X-linked.
In the X-linked form of SCID, which accounts for 50% to 60% of cases, B lymphocytes are present, but natural killer (NK) cells are absent. In these patients (T-B+ SCID), the agammaglobulinemia is a direct consequence of deficient T cell help. The X-linked form results from a gene (IL2RG) defect located at Xq12-13. 96.97 This gene encodes for the common chain present in the interleukin (IL) receptors IL-2R, IL-4R, IL-7R, IL-9R, and IL-15R. In the autosomal recessive form of SCID, B cells are lacking, and NK cells may be present Cr-B- SCID). In adenosine deaminase (ADA) deficiency, a variety of ADA gene mutations have been described. A lack of ADA in precursor lymphocytes results in a maturation arrest by accumulation of deoxy-ATP, which inhibits cell division. 98 The clinical course is fatal within the first 2 years of life. JAK3 kinase deficiency causes autosomal recessive SCID by a mutation in the JAK-3 kinase gene (JAK3).9 This enzyme mediates post-IL-2R signaling. Another form of autosomal recessive SCID is deficiency of ZAP-70, a key signal transduction molecule in T cells. 9 ZAP-70 is involved in TCR down-modulation, rendering T cells anergic. A spontaneous point mutation of the gene encoding an SH2 domain of ZAP-70 causes chronic autoimmune arthritis in mice that resembles human rheumatoid arthritis in many aspects. 99 However, arthritis has so far not been described in its human counterpart. Naturally acquired immunologic self-tolerance is not entirely accounted for by this T cell anergy. The T cell repertoire of healthy individuals harbors self-reactive lymphocytes with a potential to cause autoimmune disease, and these lymphocytes are under dominant control by a unique subpopulation of CD4+ T cells called regulatory T cells. lOG The only curative treatment for SCID is allogeneic hemopoietic stem cell transplantation. As in patients with ADA deficiency, patients with the X-linked IL-2R deficiency may benefit from gene therapy.101 This approach has been initially very successful but is associated with an increased risk of acute lymphatic leukemia. 102 There are no reports of rheumatic diseases in children with SCID, which may be because they usually die before the age of 2 years unless they undergo transplantation, but it may also illustrate the essential role of T cells in the
648
C HAP T E R
33 IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
initiation of an autoimmune disorder. In scm due to purine nucleoside phosphorylase (PNP) deficiency, autoimmune thyroiditis, idiopathic thrombopenia, SLE, and cerebral vasculitis were described before stem cell transplantation was performed. 103 After stem cell transplantation, autoimmune hematologic phenomena have been described in patients with graft-versus-host disease (GVHD), a positive Coombs test result, and autoimmune hemolytic anemia. Chronic GVHD of the skin leads to skin changes that resemble those that occur in systemic scleroderma. Patients with ADA deficiency frequently have deformities of the ribs abnormalities and flat ilia, without joint problems.
Combined Immunodeficiency The term combined immunodeficiency is applied to a group of disorders of variable clinical severity associated with defects in cellular and humoral immunity.91,104 The Wiskott-Aldrich syndrome (WAS) is characterized by a progressive abnormality in T and B lymphocyte function. IOS It is thought to be caused by defects in immunoregulatory peptides involved in cell-cell interactions. There is a low expression of sialophorin (CD43). The syndrome is associated with mutations of the gene located at Xpll.23. 106 The precise function of the WAS protein this gene encodes is uncertain. A female WAS patient has been described in whom the gene mutation was present on one X chromosome. The fact that she was nevertheless affected was explained by a nonrandom inactivation of the X chromosomes. Discovery of the gene mutation has also led to the identification of related male adults with only thrombocytopenia. Children with WAS have persistent eczema, thrombocytopenia with a low platelet volume, and recurrent ear, nose, and throat infections. They often have chronic cytomegalovirus infection. Laboratory abnormalities vary widely. The characteristic immunoglobulin pattern includes normal IgG and elevated IgA and IgE levels, with absence of antibodies to polysaccharide antigens (e.g., pneumococcal capsular antigen) and absent blood group isohemagglutinins. There is a high incidence of Coombspositive hemolytic anemia, vasculitis, and (mostly transient) arthritis. I06-111 The syndrome is a premalignant condition with a high frequency of thymomas and sarcomas later in life, although the precise incidence of these malignancies is unknown because asymptomatic persons with the WAS gene mutation may be undetected.
T Cell Immunodefldendes In the T cell immunodeficiencies, T lymphocytes are, in contrast with SCID, present in the peripheral blood, although their numbers are reduced, This is a heterogeneous and often poorly defined group of disorders. Various functional and genetic defects have been described (see Table 33-4). Clinically, patients with these diseases do not have life-threatening infections in the first months of life but show a more gradually developing immunodeficiency. An imbalance between T and B lymphocytes may explain the high incidence of autoimmune disorders, as well as infections, allergies, and malignancies. Severe and chronic
autoimmune manifestations, mostly involVing blood cells, develop between 1 and 12 years of age. 104 Vasculitis, autoimmune hepatitis, and thyroiditis have been described. In younger patients without severe ongoing infections, bone marrow transplantation may be performed. The DiGeorge syndrome (DGS) is a T cell disorder of variable severity caused by mutations in the 22qll region. There have been several case reports of chronic polyarthritis in DGS. lI2-lIS In a cohort of 80 patients with DGS and a proven chromosome 22q11.2 deletion, 3 patients were found to have polyarticular ]RA. ll5 Cartilage-hair hypoplasia (CHH) consists of generalized hypermobility, bony dysplasia, short-limbed dwarfism, fine and sparse hair, short fingernails, and various immune defects, including neutropenia, and combined immunodeficiency or mild humoral deficiency.1l6,117 However, inflammatory rheumatic diseases have not been reported. The disease-causing gene has been identified as the RMRP gene on the short arm of chromosome 9. Four children have been described with a similar syndrome of short stature due to spondylometaphyseal dysplasia and severe infections due to a combined humoral and cellular immune deficiency.lls They all had normal RMRP gene expression, thereby excluding CHH. ADA activity was normal. Three children had idiopathic thrombocytopenia, and hypothyroidism occurred in three, vitiligo in two, oligoarticular ]IA in one, and Crahn's disease in one. l1S The immunologic abnormalities were comparable to those seen in CHH. 117 One patient died of encephalitis of unknown cause.
Primary Humoral Immunodefldendes These antibody-deficiency syndromes result from impaired intrinsic B cell development or ineffective B cell responses to T cell-derived signals (see Table 33-5), The association of primary humoral immunodeficiencies and rheumatic disease is a well-known phenomenon.
Selective IgA Deficiency Selective IgA deficiency (sIgA-D) is the most common primary immunodeficiency. In Western countries, the prevalence ranges between 1 case in 330 to 2200 persons.119--121 It is characterized by serum IgA levels of less than 0.01 to 0.05 giL. IgA is also absent in secretions, although the secretory component of IgA is normally present in saliva. Patients with sIgA-D identified by routine immunodiffusion assays have trace amounts of circulating IgA detectable by the more sensitive radioimmunoassay.12o Although the term sIgA-D denotes an isolated deficiency of 19A, this immunoglobulin is also deficient in 20% of patients with IgG subclass deficiency and in 40% of those with a defective anti-polysaccharide antibody response. Antibodies of the IgM or the IgG class directed against IgA are commonly found in sera from patients with sIgA_D. 122,123 The cause of sIgA-D is largely unknown. That anti-lgA autoantibodies may play a role in the induction of 19A deficiency (Table 33-6) is supported by the observation that 19A deficiency is more common in children of 19A deficient mothers with anti19A antibodies than in children of sIgA-D fathers, although males and females are affected with equal frequency. 119.124 Transplacental passage of maternal anti-lgA antibodies may therefore interfere with the developing 19A-producing system. 124 The fact that plasma cells producing anti-lgA could not be detected locally along the mucosal linings has led to the hypoth-
C HAP T E R
33
esis that sIgA-D results from systemic exposure to endogenous IgA. sIgA-D with anti-IgA antibodies can be regarded as an autoimmune disorder. m Moreover, anti-IgA antibodies are more common in IgA-deficient patients with autoimmune and rheumatic diseases than in asymptomatic IgA-deficient patients. 126 In most patients, B cells expressing IgA on their surface and in the cytoplasm are still present in the blood, albeit in low numbers. 127 Exposure to an oral vaccine induces a normal mucosal immune response by B cells that secrete antigen-specific IgG or IgM. Nevertheless, a B cell maturation defect may be present because, in contrast to normal persons, B cells from IgA-deficient persons also express surface IgM and IgD. Comparable to common variable immunodeficiency (CVID), T cellular proliferative responses to mitogens are decreased in a pni'portion of patients with IgA deficiency.127 Defective helper T cell function and defective suppressor T cells inhibiting IgA production have been described. The sIgA-D can be familial, and an autosomal dominant inheritance pattern is found in some families.128.129 The mother and three siblings of an IgAdeficient girl with JRA lacked serum IgA. The mother had very high levels of anti-IgA antibodies detected by a hemagglutination assay. The incidence of sIgA-D is increased in families of persons with CVID or hypogammaglobulinemia. The sIgA-D may also precede CVID. As in CVID, the putative gene defect resides on chromosome 6 between the HLA-B and the HLA-DQ regions (discussed later).130
Deficiency of sIgA is usually congenital and permanent, although transient cases have been described. 127 In some patients with ]RA and sIgA-D, the IgA deficiency developed before antirheumatic drugs were used.122·123.129 Drug-induced 19A deficiency is well known, however. In particular, nonsteroidal anti-inflammatory drugs such as diclofenac and sulfasalazine, parenteral gold, and D-penicillamine are associated with IgA deficiency that is sometimes reversible on discontinuation of the drug. 131- 134
Disease Associations The clinical spectrum of sIgA-D varies from asymptomatic healthy persons to those with infectious, allergic, or autoimmune disorders (Table 33-6). The frequency of autoimmune disease as reported in large studies of IgAdeficient individuals ranges from 7% to 36%. Among the rheumatic diseases, ]RA, SLE, and rheumatoid arthritis are the most frequent associations (see Table 33-7). AS$ociations of sIgA-D with other rheumatic diseases such as sarcoidosis, scleroderma, dermatomyositis, and
'~, I
TABLE ~~-6 Frequency of Anti-lgA Antibodies in Selective IgA DefiCIency
Antl-IgA antibodies
DIagnosis Asymptomatic blood donors Miscellaneous diseases Recurrent infections RA-like arthritis JM-like arthritis SLE-like disease _r
649
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
Number
Number
Percent
27 8 10 4 13 10
5 2 3 2 10 10
19 25 30 50 77 100 -----l
JRA. juvenile rheumatoid arthritis; RA, rheumatoid arthritis; SLE, systemic lupus erythemalOsus. AdlIpted Petry RE, Palmer NR, Cassidy JT, et al: The association of autoimmune disl'ases and anti-lgA antibodies in patients with selective 19A deficiency. Clio Exp Immunol 37: 83, 1979.
I! It
lABI L
n
-1
Selective IgA Deficiency and Rheullldti( Diseases
Study Cassidy et ai, 1977,168 1979120 Huntley et ai, 1967 169 Panush et ai, 1972 170 Pelkonen et ai, 1975171 Barkley et ai, 1979172 Salmi et ai, 1973 131 Cassidy et ai, 1969 173 Cleland and Bell, 1978136 Cassidy et ai, 1969 173 Cassidy, 1981 174 Jay et ai, 1981 17s Spirer et ai, 1979 176 Cassidy, 1981 174 Good et ai, 1977 140 Barkley et ai, 1979 172 Cassidy et ai, 1969 m
Rheumatic Disease AssocIations
Numberl Totar
Chronic arthritis (fRA-like)
18/477
Chronic Chronic Chronic Chronic Chronic SLE SLE
2/23 3/176
arthritis arthritis arthritis arthritis arthritis
(IRA-like) (fRA-like) (fRA-like) (JRA-like) (JRA like)
Dermatomyositis Scleroderma (systemic) Scleroderma (systemic) Scleroderma (local) Ankylosing spondylitis Ankylosing spondylitis Juvenile ankylosing spondylitis Mixed connective tissue disease
111300 2/582 5/115 10/50 2 3 1/15 1 1 2 1 1 1/14
JRA, juvenile rheumatoid arthritis; SLE, systemic lupus erythematosus. 'Number/total is the number of 19A-deficient patients over number of patients with specific rheumatic disease.
Kawasaki disease are more sporadic and may reflect an ascertainment bias. In general, the rheumatic disease in these patients responds to the conventional antirheumatic therapy. Chronic Arthritis The prevalence of sIgA-D in JRA varies from 2% to 4%.124,128,130 In general, the clinical picture, sex ratio, and age at onset of arthritis do not differ from those in children with]RA and normal or elevated levels of IgA. The distribution of onset types is also similar: oligoarticular onset in 64%, polyarticular onset in 32%, and systemic onset in 40f0l27.129 (Figs. 33-1 to 33-3). In most patients, the course of the disease is mild and remains oligoarticular, with little or no functional limitations. Erosive arthritis, however, has been described in up to 28%.135 In one studY,113 the arthritis appeared more severe in patients with transient 19A deficiency or in patients with borderline IgA values. In another studY, 129 patients with sIgA-D associated with oligoarticular JRA had a higher frequency of ANAs and uveitis, although this could not be confirmed in a subsequent study by Pelkonen and colleagues. 127 Systemic Lupus Erythematosus The prevalence of sIgA-D in patients with SLE is 1% to 4%, which is 20 to 30 times higher than that in the normal population. 121 ,136 In general, the clinical manifestations of SLE and the response to therapy do not differ in patients with or without sIgA-D, although in a series of 10 children with sIgA-D and SLE, there was more neuropsychiatric disease, but nephritis was absent,122 Resolution of the sIgA-D may occur after immunosuppressive therapy, as may be the case with low IgG-associated SLE and sIgA-D-associated JRA. 137,138 Other Rheumatic Diseases Deficiency of sIgA has been described sporadically in other systemic rheumatic diseases such as dermatomyositis, sarcoidosis, scleroderma,
650
C HAP T E R
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
and ankylosing spondylitis. 139,14o However, these associations may merely reflect an ascertainment bias.
Hypogammaglobulinemia The term hypogammaglobulinemia is applied to a number of disorders characterized by decreased levels of serum IgG and the inability to produce specific antibodies when exposed to an antigen. Unlike SCID and combined immunodeficiency, there usually are no specific T cell abnormalities. Among the primary hypogammaglobulinemias are X-linked agammaglobulinemia (Le., Bruton's agammaglobulinemia), CVID (also called lateonset hypogammaglobulinemia), autosomal early-onset agammaglobulinemia, and the hyper-IgM syndrome (Table 33-8); see also Table 33-5). Mutations in the leas gene were found in a minority of adults with CVID.141 The ICOS protein is a costimulatory molecule involved in T cell activation, Drug-induced hypogammaglobulinemia has been reported in patients exposed to various anticonvulsants and antirheumatic drugs. • figure 33-1 Hands of an ll-year-old girt with chronic arthritis, tenosynovitis, and selective immunoglobulin Adeficiency. Hand and wrist involvement gradually retumed to normal, but a minimally symptomatic effusion of her right knee persisted.
X-Linked Agammaglobulinemia X-linked agammaglobulinemia is characterized by recurrent severe bacterial infections from the age of 6 to 12 months onward. 142 Only boys are affected with recurrent otitis media, pneumonia, meningitis, and septic arthritis
• Figure 33-2 A, Hand of I P. when 27 years old. A chronic, deforming, erosive arthritis of the wrists and small joints of the hands was slowly progressive from the onset.These deformities and subluxation of metacarpophalangeal (MCP) joints are evident.The second proximal interphalangeal (PIP) joint had been surgically fused in a functional position. 8, Hand of an 18-year-old girt with selective immunoglobulin Adeficiency and systemic lupus erythematosus, with onset of arthritis at the age of 7 years. Destruction of joints is already far advanced, with subluxations of ulnar side of wrist, MCP joints 1 to 3, and PIP joints 4 and 5. Erosions, destruction of articulating surfaces, microfractures and bony collapse, and extreme juxta-articular osteoporosis have occurred.
C HAP T E R
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
651
~ 33-3 X-ray films of the knee of a patient with selective immunoglobulin A deficiency at 25 years of age. A, Anteroposterior view. 8, Lateral view. Coarsening of the trabecular architecture and slight osteoporosis are evident, along with a moderate decrease in the cartilaginous space.There is sharpening of the tibial tubercles and the posterior aspect of the lateral femoral condyle. Erosions are not present despite 16 years of continuous joint effusion.
•
from extracellular encapsulated organisms such as S. pneumoniae and Haemopbilus influenzae. The defective gene resides at Xq21.3-22 and causes a block in differentiation at the pre-B cell stage. 143 B cells are therefore absent. Serum IgG levels are usually less than 2 giL; IgM, 19A, and IgE are absent. Patients should be treated vigorously with antibiotics and with life-long intravenous immunoglobulin (IVIG).
Common Variable Immunodeficiency CvrD is a heterogeneous primary immunodeficiency characterized by hypogammaglobulinemia and recurrent infections, predominantly with bacterial agents. Recurrent
'.::.11
IABLE 33-8
sinopulmonary infections often cause bronchiectasis. There is a high frequency of gastrointestinal disease, such as giardiasis, and autoimmune diseases. Patients with CvrD have an increased risk (up to 10%) of malignancies of the lymphoid system or the gastrointestinal tract. 144-146 The incidence of CvrD is about 5 cases per million people. The age at onset is usually between 30 and 45 years, with a wide range (l to 71 years),145-147 and about 25% have onset before the age of 15 years. The number of B lymphocytes may be normal or decreased. When purified B cells from patients with CvrD are cultured in with normal allogeneic T cells, the defect in immunoglobulin production cannot be corrected,146.148 although B cells from most patients with
Chara
Sex Genetics Age at onset of symptoms B lymphocytes Peripheral lymphoid tissue Plasma cells in nodes S\lrface immunoglobulins Serum IgG Serum IgA, IgM Natural antibodies Specific antibodies T lymphocytes Clinical characteristics Severe bacterial infections Viral infections Malabsorption Autoimmune diseases JRA-like arthritis Control with Ig replacement
X-linked
Common Variable
Males X-linked recessive (Xq21.3-22) 6moto2yr pre-B cell Hypoplastic Rare Absent <1 giL Very low Very low Absent Numbers increased, subsets normal
Equal sex distribution Variable (? 6p21.3) 2 yr to adulthood B cell Normal to enlarged Decreased or none Present <5 giL Variable Variable Variable (autoantibodies) Variable number and function
Yes Yes Sometimes No
Sometimes; less severe Yes Frequent Frequent Infrequent; frequency unknown Partial
30%
Partial
652
C HAP T E R
33
IMMuNoDEFrcrEN crES AND THE RHE UM ATIC Dr SEASES
CVID can synthesize at least some immunoglobulin in the presence of an appropriate in vitro stimulus. When T cells from patients with CVID are co-cultured with normal B cells, immunoglobulin secretion is normal. Although this disease is regarded as an intrinsic B cell defect, in vitro T lymphocyte proliferative responses to mitogens are decreased or absent in about one half of patients. 146,148 The precise B cell defect is still unknown, and the hypogammaglobulinemia may also result from a lack of appropriate T cell-derived stimulation necessary for normal B cell maturation, as is shown by the example of leas gene mutations. 141 Such a T cell abnormality may account for the observed predisposition of patients with CVID to malignancies and autoimmune disorders. Specifically, patients' CD4+ cells produce less lL-2, possibly because of a selective defect in the ability to activate the IL-2 gene normally.148 Some patients with CVID have expanded activated (Le., CD57+ and DR+) CD8+ populations, a pattern comparable to that in patients infected with cytomegalovirus, Epstein-Barr virus, or human immunodeficiency virus. It was speculated that a chronic viral infection in a genetically predisposed person could induce CVID (discussed later). A similar pathogenesis is thought to apply to the X-linked lymphoproliferative syndrome. Susceptibility to CVID and sIgA-D is determined by a gene located at 6p21.3. This region of chromosome 6 contains the 21-hydroxylase A and tumor necrosis factor genes, and it lies between the HLA-B and HLA-DQ domains. Autoimmune Disease Autoimmune disease occurs in 20% to 30% of patients with CVID,145,146 whereas there are only a few reports of autoimmune disease in X-linked agammaglobulinemia. 149 Thrombocytopenia and hemolytic anemia are most common. 150 The paradoxical association of antibody-mediated autoimmunity and hypogammaglobulinemic states may be explained by defects in the antiidiotypic network, which normally regulates expression of naturally occurring autoantibodies. The prevalence of arthritis in hypogammaglobulinemia ranges from 10% to 30%. It is divided into septic and aseptic forms.
and limitation of motion 151 (Figs. 33-4 and 33-5). In about 50% of patients, arthritis is the presenting symptom. 152 Uluhan and colleagues 153 described a patient with a systemic-onset JRA and CVID who later developed neutropenia (found in up to 10% of patients with CVID and X-linked agammaglobulinemia), autoimmune hemolytic anemia, and a cellular immune deficiency. Despite immunoglobulin infusions, the patient died at the age of 22 years of infection. Other Rheumatic Diseases Whereas the prevalence of sIgA-D in patients with SLE is 20 to 30 times higher than that in the normal population, only nine cases of SLE with persistent hypogammaglobulinemia have been described. 1S4--156 One of these patients developed fatal extrapulmonary tuberculosis after a 24 year history of hypogammaglobulinemia and SLE.154-156 Cronin and colleagues 137 reported 18 patients with SLE who developed low IgG levels. There was no significant proteinuria that could account for a loss of IgG. The low IgG was transient in 10 patients and related to cytotoxic drugs in 8; only 4 had recurrent infections. This low number suggests that the association of SLE and CVID may be coincidental. It is also difficult to understand an association of an autoimmune disease such as SLE, in which B cell activation, elevated serum IgG, and circulating immune complexes are prominent, with another disorder characterized by deficient antibody production. Dermatomyositis-like Syndrome There is considerable doubt that the dermatomyositis-like syndrome that sometimes complicates hypogammaglobulinemia has the same pathogenesis as idiopathic dermatomyositis of childhood. Clinical characteristics of the reported cases of this syndrome are summarized in Table 33-9. It is characterized by subcutaneous edema, myalgia, muscle wasting, and sometimes by polyarthritis followed by contractures of the large joints. It is not clear whether the myopathy is proximal, dis-
Septic Arthritis Septic arthritis caused by common and rare microorganisms occurs relatively frequently in patients with hypogammaglobulinemia.132.148 Causative microorganisms are S. aureus, S. pneumoniae, H. influenzae, Mycobacteria species, and Mycoplasma species (including Ureaplasma). Mycoplasma species are difficult to culture. Improved detection techniques (e.g., specific culture fluids, electron microscopy, polymerase chain reactions) may identify the presence of these microorganisms in a substantial number of cases heretofore been regarded as aseptic. 132•145 Aseptic Arthritis The association of rheumatic diseases and CVID or X-linked agammaglobulinemia is firmly established, with reported frequencies varying between 7% and 42%. In a large series of 103 patients with CVID, 3 were reported as having chronic arthritis similar to oligoarticular JIA. I46 Onset of arthritis occurs between 3 and 15 years of age; it is often subtle and is characterized by small to moderate effusions, soft tissue thickening,
• Figure 33-4 Needle biopsy of the synovium of R. B., who developed chronic synovitis of the left knee and was found to have common variable immunodeficiency.There is marked hypertrophy of the subsynoviallayers with hyperplasia of vascular endothelium and compaction of collagen. A nonspedfic infiltrate of mononudear cells is visible, but there are no aggregations of round cells. Plasma cells are absent. Rbrin is present on the synovial surface. (From Petty RE, Cassidy JT, Tubergen DG: Association of arthritis with hypogammaglobulinemia. Arthritis Rheum 20 ISuppl): 441, 1977.)
C HAP T E R
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
653
tal or diffuse in most instances. Cutaneous manifestations in~lude heliotrope discoloration of the eyelids, rash on the extensor surfaces of the metacarpophalangeal and proximal interphalangeal joints, and sometimes a nonspecific rash. 157 Electromyographic evidence of a myopathy was documented in one patient, and abnormalities on muscle biopsy consistent with a diagnosis of dermatomyositis were present in seven. Focal calcification was present in one. Central nervous system disease and deafness frequently accompanied the myositis, and a fatal outcome was common. Other associations have also been described, including leukemia and lymphoma. Viral isolations (e.g., echovirus, adenovirus) from the cerebrospinal fluid and sometimes from muscle and other sites raise the question of a viral cause of this syndrome and support a role for persistent latent viral infection in its pathogenesis. I3').I58.159 One patient was successfully treated with IVIG. A direct relation to idiopathic dermatomyositis remains unproved.
IgG Subclass Deficiencies and Rheumatic Diseases
• figure 33-5 Thermograph of the knees of R. B. at 20 years of age show increased heat production on the left knee, the site of the inflammatory arthritis. (The paper dip on the left leg is used for standardization in thermography.)
. : . tABLE 33-9
IgG subclass deficiencies have been reported in a number of patients.16o-162 A lO-year-old boy with JRA and Hodgkin's disease had low concentrations of IgA (0.19 giL) and IgG2 (0.02 giL). Another 10-year-old boy had SLE, an undetectable IgA level, and an IgG2 concentration of 0.02 giL. Oxelius 163 also reported IgG2 subclass deficiency in SLE. Heiner and colleagues found low IgG4 levels (less than 30 mglL) in 12 of 112 patients with "disseminated collagen vascular disease" but provided no further details. 164 In a study of 450 patients with subclass deficiencies, IgG2 deficiency was associated most frequently with vasculitis and cytopenias.165.166 Another study found an increased frequency of HenochSchonlein purpura and glomerulonephritis. 167
Dermatomyositis-like Disease dnd Hypogammaglobulinemia
Sex
Age
Olnlcal DescrIption
177
M
2 yr
178 179 180 180 liB
M
2 yr
157
M
5 yr
159
M
24 yr
159
M
12 yr
159
M
3 yr
158
M
11 yr
182
F
3 mo
U!3
M
29 yr
Brawny edema of distal extremities; leg stiffness, contractures; gastrocnemius biopsy specimen consistent with dermatomyositis Patient with classic dermatomyositis and hypogammaglobulinemia Dermatomyositis, lymphoma Adenovirus 12 isolated Echovirus 9 isolated Intermittent arthritis (knees) since 2 yr; at 17 yr, heliotrope rash on eyelids, MCP and PIP rash, muscle weakness, contractures, elevated LDH. Biopsy: perivascular, endomysial, perimysial lymphocytic infiltrate; negative immunofluorescence; cerebral vasculitis. Death. Brawny edema of extremities; violaceous, telangiectatic rash over extensor aspects of legs; muscle wasting, contraclures of elbows, wrists, knees. Elevated ALT and AST. CK normal. Muscle biopsy: perivascular lymphocytic infiltration. Death. Muscle weakness, edema, rash on extensor surface of joints; contractures of elbows, knees. GSF: echovirus 9. Muscle biopsy: perivascular round cell infiltration. Death. Joint stiffness, contractures; weakness and atrophy of muscles; deafness. Muscle biopsy: perivascular lymphocytic infiltration. Arthritis in knees and ankles, myalgia; increased AST level, elevated aldolase level. EMG: myopathy. EGG: right bundle branch block. GSP: echovirus 33. Polyarthritis, edema, encephalitis. Muscle biopsy: mononuclear infiltration. Leukocyte migration inhibited by echovirus-11 vaccine. Fever, muscle weakness, cellulitis, violaceous edematous eyelids, mucosal ulceration; elevated CK. Skin biopsy: leukocytoclastic vasculitis. Chronic arthritis at 19 mo; in adulthood myalgia, proximal muscle weakness, elevated CK. Gastrocnemius biopsy: active inflammatory myopathy. Echovirus 11 from blood, urine, CSF, muscle.
ALT, alanine transaminase; AST, aspartate transaminase: CK, creatine kinase; CSP, cerebrospinal fluid: ECG, electrocardiogram; EMG, electromyogram; LDH. lactate debydrogenase; Mep. metacarpophalangeal; PIP, proximal interphalangeal.
654
C HAP T E R
33
IMMUNODEFICIENCIES AND THE
REFERENCES 1. Chinnaiyan AM, O'Rourke K, Tewari M, Dixit VM: FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis. Cell 81: 505-512, 1995. 2. Dhein J, Walczak H, Baumler C, et al: Autocrine T-cell suicide mediated by APO-l!(Fas/CD95). Nature 373: 438-441, 1995. 3. Hsu H, Xiong J, Goeddel DV: The TNI' receptor I-associated protein TRADD signals cell death and NF-kappa B activation. Cell 81: 495-504, 1995. 4. Stanger BZ, Leder P, Lee TH, et al: RIP: a novel protein containing a death domain that interacts with Fas/APO-1 (CD95) in yeast and causes cell death. Cell 81: 513-523, 1995. 5. Rieux-Laucat 1', Fischer A, Deist FL: Cell-death signaling and human disease. Curr Opin Immunol 15: 325-331, 2003. 6. Wang J, Zheng L, Lobito A, et al: Inherited human caspase 10 mutations underlie defective lymphocyte and dendritic cell apoptosis in autoimmune Iymphoproliferative syndrome type II. Cell 98: 47-58, 1999. 7. Chun HJ, Zheng L, Ahmad M, et al: Pleiotropic defects in lymphocyte aclivation caused by caspase-8 mutations lead to human immunodeficiency. Nature 419: 395-399. 2002. 8. Merino R, Fossati L, Izui S: TIle lupus-prone BXSB strain: the Yaa gene model of systemic lupus erythematosus. Springer Semin Immunopathol 14: 141-157, 1992. 9. Fischer A, Cavazzana-Calvo M, De Saint BG, et al: Naturally occurring primary deficiencies of the immune system. Annu Rev Immunol 15: 93-124, 1997. 10. Rieux-Laucat 1', Le Deist 1', Hivroz C, et al: Mutations in Fas associated with human Iymphoproliferative syndrome and autoimmunity. Science 268: 1347-1349, 1995. 11. Rieux-Laucat 1', Blachere S, Danielan S, et al: Lymphoproliferative syndrome with autoimmunity: a possible genetic basis for dominant expression of the clinical manifestations. Blood 94: 2575-2582, 1999. 12. van der Werff Ten Bosch, Schotte P, Ferster A, et al: Reversion of autoimmune Iymphoproliferative syndrome with an antimalarial drug: prelinlinary results of a clinical cohort study and molecular observations. Br J Haematol 117: 176-188, 2002. 13. Benkerrou M, Le Deist 1', de Villartay JP, et al: Correction of Fas (CD95) deficiency by haploidentical bone marrow transplantation. Eur J Immunol 27: 2043-2047, 1997. 14. Sleight BJ, Prasad VS, Delaat C, et al: Correction of autoimmune lymphoproliferative syndrome by bone marrow transplantation. Bone Marrow Transplant 22: 375-380, 1998. 15. de Villartay]p, Rieux-Laucat 1', Fischer A, Le Deist 1': Clinical effects of mutations to CD95 (Pas): relevance to autoimmunity? Springer Semin Immunopathol 19: 301-310, 1998. 16. DrappaJ, Vaishnaw AK, Sullivan KE, et al: Pas gene mutations in the CanaleSmith syndrome, an inherited Iymphoproliferative disorder associated with autoimmunity. N Engl J Med 335: 1643-1649, 1996. 17. Sneller MC, Wang J, Dale JK, et al: Clincial, immunologic, and genetic features of an autoimmune Iymphoproliferative syndrome associated with abnormal lymphocyte apoptosis. Blood 89: 1341-1348, 1997. 18. Dianzani U, Bragardo M, DiFranco D, et al: Deficiency of the Fas apoptosis pathway witJ10ut Fas gene mutations in pediatric patients with autoimmunity/lymphoproliferation. Blood 89: 2871-2879, 1997. 19. Wu J, Wilson J, He J, et al: Fas ligand mutation in a patient with systemic lupus erythematosus and Iymphoproliferative disease. J Clin Invest 98: 1107-1113, 1996. 20. Seger RA, Ezekowitz RA: Treatment of chronic granulomatous disease. Immunodeficiency 5: 113-130, 1994. 21. Ma HR, Mu SC, Yang YH, et al: Therapeutic effect of interferon-gamma for prevention of severe infection in X-linked chronic granulomatous disease. J Formos Med Assoc 102: 189-192, 2003. 22. Seger RA, Gungor T, Belohradsky BH, et al: Treatment of chronic granulomatous disease with myeloablative conditioning and an unmodified hemopoietic allograft: a survey of the European experience, 1985-2000. Blood 100: 4344--4350, 2002. 23. Roos D: X-CGDbase: a database of X-CGD-causing mutations. Immunol Today 17: 517-521, 1996. 24. Meischl C, Roos D: The molecular basis of chronic granulomatous disease. Springer Semin Immunopathol 19: 417-434, 1998. 25. Badolato R, Notarangelo LD, Plebani A, Roos D: Development of systemic lupus erythematosus in a young child affected with chronic granulomatous disease follOWing withdrawal of treatment with interferon-gamma. Ri1eumatology (Oxl') 42: 804-805, 2003. 26. Brandrup F, Koch C, Petri M, et al: Discoid lupus erythematosus-like lesions and stomatitis in female carriers of X-linked chronic granulomatous disease. Br J Dermatol 104: 495-505, 1981. 27. Humbert JR, Fishman CB, Weston WL, et al: Frequency of the carrier state for X-linked chronic granulomatous disease among females with lupus erythematosus. Clin Genet 10: 16-20, 1976. 28. Manzi S, Urbach AH, McCune AB, et a1. Systemic lupus erythematosus in a boy with chronic granulomatous disease: case report and review of the literature. Arthritis Rheum 34: 101-105, 1991. 29. Schaller J: lllness resembling lupus erythematosus in mothers of boys with chronic granulomatous disease. Ann Intern Med 76: 747-750, 1972.
RH E UMATIC
DISEASES
30, Hafner J, Enderlin A, Seger RA, et al: Discoid lupus erythematosus-like lesions in carriers of X-linked chronic granulomatous disease. Br J Dermatol 127: 446-447, 1992. 31. Romera Modamio G, Martin Mateos MA, Gonzalez Ensenat MA, Pastor Gomez MA: Chronic granulomatous disease: a case study of a symptomatic carrier. J Investig Allergol Clin Immunol 7: 57-61, 1997. 32. Rupec RA, Petropoulou T, Belohradsky BH. et a1. Lupus erytllematosus tumidus and chronic discoid lupus erythematosus in carriers of X-linked chronic granulomatous disease. Eur J Dermatol 10: 184--189, 2000, 33. Lee BW, Yap HK: Polyarthritis resembling juvenile rheumatoid arthritis in a girl with chronic granulomatous disease. Arthritis Ri1eum 37: 773-776, 1994. 34. Sponseller PD, Malech HL, McCarthy EF Jr, et al: Skeletal involvement in children who have chronic granulomatous disease. J Bone Joint Surg Am 73: 37-51, 1991. 35. Payne NR, Hays NT, Regelmann WE, et al: Growth in patients with chronic granulomatous disease, J Pediatr 102: 397-399, 1983, 36, Landing BH, Shirkey HS: A syndrome of recurrent infection and inftltration of viscera by pigmented lipid histiocytes. Pediatrics 20: 431-438, 1957, 37. Rodey GE, Park BH, Ford DK, et al: Defective bactericidal activity of peripheral blood leukocytes in lipochrome histiocytosis, Am J Med 49: 322-327, 1970. 38. Dreskin SC, Goldsmith PK, Gallin JI: Immunoglobulins in the hyperimmunoglobulin E and recurrent infection (job's) syndrome. Deficiency of antiStaphylococcus aureus immunoglobulin A. J Clin Invest 75: 26-34, 1985. 39. Renner ED, Puck JM, Holland SM, et a1. Autosomal recessive hyperimmunoglobulin E syndrome: a distinct disease entity. J Pediatr 144: 93-99, 2004. 40. Certain S, Barrat F, Pastural E, et a1. Protein truncation test of LYST reveals heterogenous mutations in patients with Chediak-Higashi syndrome. Blood 95: 979-983, 2000. 41. Dufourcq-Lagelouse R, pastural E, Barrat FJ, et al: Genetic basis of hemophagocytic Iymphohistiocytosis syndrome [review]. Int J Mol Med 4: 127-133, 1999. 42. Jacobs JC, Goetzl EJ: "Streaking leukocyte factor." arthritis, and pyoderma gangrenosum. Pediatrics 56: 570-578. 1975. 43. Jacobs JC: The differential diagnosis of arthritis in childhood. In Pediatric Ri1eumatology for the Practitioner, 2nd ed. New York, Springer-Verlag. 1993, 44, Brouard M, Chavaz P, Borradori L: Acute pusrulosis of the legs in diverticulitis with sigmoid stenosis: an overlap between bowel-associated dermatosis-arthritis syndrome and pustular pyoderma gangrenosum. J Eur Acad Dermatol Venereol 18: 89-92, 2004. 45. Rarnoff wo: Inherited deficiencies of complement in rheumatic diseases, Rheum Dis Clin North Am 22: 75-94, 1996. 46. Rosenfeld SI, Kelly ME, Leddy JP: Hereditary deficiency of the fifth component of complement in man. I. Clinical, immunochemical, and family studies. J Clin Invest 57: 1626-1634, 1976, 47. Ruddy S: Rheumatic diseases and inherited complement deficiencies. Bull Rheum Dis 45: 6-8, 1996. 48. Sullivan KE: Complement deficiency and autoimmunity, CUlT Opin Pediatr 10: 600-606, 1998. 49. Walport MJ: Complement and systemic lupus erythematosus. Arthritis Res 4 (Suppl 3): S279-S293, 2002. 50, Ross SC, Densen P: Complement deficiency states and infection: epidemiology, pathogenesis and consequences of neisserial and other infections in an immune deficiency. Medicine (Baltinlore) 63: 243-273, 1984. 51. Agnello V: Lupus diseases associated with hereditary and acquired deficiencies of complement. Springer Semin Immunopathol 9: 161-178, 1986. 52. Atkinson JP: Complement system on the attack in autoinlmunity. J Clin Invest 112: 1639-1641, 2003. 53. Fries LF, O'Shea JJ, Frank MM: Inherited deficiencies of complement and complement-related proteins. Clin Immuno! Immunopathol 40: 37-49. 1986. 54. Leggiadro RJ: Systemic meningococcal infection and complement deficiency. Pedlatr Infect Dis J 22: 760-761, 2003. 55. Glass D. Raum D, Balavitch D, et al: Inherited deficiency of the sixth component of complement: a silent or null gene. J Immunol 120: 538-541, 1978. 56. Nagata M, Hara T, Aoki T, et al: Inherited deficiency of ninth component of complement: an increased risk of meningococcal meningitis. J Pediatr 114: 260-264, 1989, 57. Tedesco F. Roncelli L, Petersen BH, et al: Two distinct abnormalities in patients with C8 alpha-gamma deficiency, Low level of C8 beta chain and presence of dysfunctional C8 alpha-gamma subunit. J Clin Invest 86: 884-888, 1990. 58. Pickering MC, Walport MJ: Links between complement abnormalities and systemic lupus erythematosus. Ri1eumatology (Oxf) 39: 133-141, 2000, 59. Gatenby PA, Barbosa JE, Lachmann PJ: Differences between C4A and C4B in the handling of immune complexes: the enhancement of CR1 binding is more important than the inhibition of immunoprecipitation. Clin Exp Immunol79: 158-163, 1990. 60. Pickering MC, Botto M, Taylor PR, et al: Systemic lupus erythematosus. complement deficiency, and apoptosis, Adv Immunol 76: 227-324, 2000, 61. Reid KB: Deficiency of the first component of human complement. Immunodetk Rev 1: 247-260, 1989. 62. Rupert KL, Moulds JM, Yang Y, et al: The molecular basis of complete complement C4A and C4B deficiencies in a systemic lupus erythematosus patient
C HAP T E R
63.
64.
65. 66.
67. 68.
69.
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
'!Vith homozygous C4A and C4B mutant genes. J Immunol 169: 157~1578, l002. lleveille JD, Moulds JM, Ahn C, et al: Systemic lupus erythematosus in three «'thnic groups: I. The effects of HLA class II, C4, and CRI alleles, socioeco· nomic factors, and ethnicity at disease onset. LUMINA Study Group. Lupus in minority populations, nature versus nurture. Arthritis Rheum 41: 1I61-1172, 1998. fielder AH, Walport MJ, Batchelor JR, et al: Family study of the major histo· compatibility complex in patients with systemic lupus erythematosus: imporlance of null alleles of C4A and C4B in determining disease susceptibility. Br Med J (Clin Res Ed) 286: 425-428, 1983. Gibson DJ, Glass D, Carpenter CB, Schur PH: Hereditary C2 deficiency: diagnosis and HLA gene complex associations. J Immunol 116: 1065-1070, 1976. Glass D, Raum D, Gibson D, et al: Inherited deficiency of the second component of complement. Rheumatic disease associations. J Clin Invest 58: 853-861, 1976. Bowness P, Davies KA, Norsworthy PJ, et al: Hereditary Clq deficiency and systemic lupus erythematosus. Q J Med 87: 455-464, 1994. Sato EI, Assis LS, Lourenzi VP, Andrade LE: Long·term thalidomide use in refractory cutaneous lesions of systemic lupus erythematosus. Rev Assoc Med Bras 44: 289-293, 1998. Hoekzema R, Swaak AJ, Brouwer MC, et al: Significance of low molecular weight Clq in systemic lupus erythematosus. Ann Rheum Dis 49: 698-704,
1990. 70. Suzuki Y, Hinoshita F, Yokoyama K, et al: Association of Sjogren's syndrome with hereditary angioneurotic edema: report of a case. Clin Immunol Immunopathol 84: 95-97, 1997. 71. Raum D, Glass D, Agnello Y, et al: Congenital deficiency of C2 and factor B. N Engl) Med 299: 1313, 1978. 72. Steinsson K, Erlendsson K, Valdimarsson H: Successful plasma infusion treatment of a patient with C2 deficiency and systemic lupus erythematosus: clinical experience over forty-five months. Arthritis Rheum 32: 906-913, 1989. 73. Osofsky SG, Thompson BH, Lint TF, Gewurz H: Hereditary deficiency of the third component of complement in a child with fever, skin rash, and arthralgias: response to transfusion of whole blood. J Pediatr 90: 18~186, 1977. 74. Roord )J, Daha M, Kuis W, et al: Inherited deficiency of the third component of complement associated with recurrent pyogenic infections, circulating immune complexes, and vasculitis in a Dutch family. Pediatrics 71: 81-87. 1983. 75. Leddy)P, Frank MM, Gaither T, et al: Hereditary deficiency of the sixth com· ponent of complement in man. I. lmmunochemical, biologic, and family studies. ) Clin Invest 53: 544-553, 1974. 76. Boyer )T, Gall EP, Norman ME, et al: Hereditary deficiency of the seventh component of complement. ) Clin Invest 56: 905-913, 1975. 77. )asin HE: Absence of the eighth component of complement in association with systemic lupus erythematosus·like disease.) Clin Invest 60: 709-715, 1977. 78. Zeitz HJ, Lint TF. Gewurz A, Gewurz H: Component deficiencies. 7. The seventh component. Prog Allergy 39: 289-294, 1986. 79, Wulffraat NM, Sanders EA, Fijen CA, et al: Deficiency of the beta subunit of the eighth component of complement presenting as arthritis and exanthem. Arthritis Rheum 37: 1704-1706, 1994, BO, Turner MW: Mannose-binding lectin: the pluripotent molecule of the innate immune system, Immunol Today 17: 532-540, 1996, 81. Aittoniemi J, Baer M, Soppi E, et al: Mannan binding lectin deficiency and concomitant immunodefects, Arch Dis Child 78: 245-248. 1998. 82.. Stengaard-Pedersen K, Thiel S, Gadjeva M, et a1: Inherited deficiency of mannan-binding lectin·associated serine protease 2, N Engl) Med 349: 554-560, 2003. 83', Koch A, Melbye M, Sorensen P, et al: Acute respiratory tract infections and mannose·binding lectin insufficiency during early childhood, )AMA 285: 1316-1321, 2001. 84" Biezeveld MH, Kuipers 1M, Geissler), et al: Association of mannose-binding lectin genotype with cardiovascular abnonnalities in Kawasaki disease, Lancet 361: 1268-1270, 2003, 85. Foley P), Mullighan CG, McGrath DS, et al: Mannose-binding lectin promoter and structural gene variants in sarcoidosis, Eur J Clin Invest 30: 549-552, 2000. 86. Zimmermann·Nielsen E, Baatrup G, Thorlacius-Ussing 0, et al: Complement activation mediated by mannan-binding lectin in plasma from healthy individuals and from patients with SLE, Crohn's disease and colorectal can· cer. Suppressed activation by SLE plasma. Scand ) Immunol 55: 105-110, 2002. 87. Wang ZY, Morinobu A, Kanagawa S, Kumagai S, Polymorphisms of the mannose binding lectin gene in patients with Sjogren's syndrome. Ann Rheum Dis 60: 483-486, 2001. 88. Tsutsumi A, Sasaki K, Wakamiya N, et al: Mannose-binding lectin gene: polymorphisms in)apanese patients with systemic lupus erythematosus, rheumatoid arthritis and Sjogren's syndrome, Genes Immun 2: 99-104, 2001. 89, Seelen MA, Trouw LA, van der Hoorn]W, et al: Autoantibodies against mannose·binding lectin in systemic lupus erythematosus, Clin Exp Immunol 134: 335-343. 2003.
655
90. Garcia-Laorden MI, Rua-Figueroa I, Perez·Aciego P, et a1. Mannose binding lectin polymorphisms as a disease-modulating factor in women with systemic lupus erythematosus from Canary Islands, Spain. ) Rheumatol 30: 74~746, 2003. 91. Buckley RH: Primary immunodeficiency diseases due to defects in lymphocytes. N Engl J Med 343: 1313-1324, 2000. 92, Sleasman]W: The association between immunodeficiency and the development of autoimmune disease. Adv Dent Res 10: 57-61, 1996. 93. Stein CM: Immunodeficient states and associated rheumatic manifestations. Curr Opin Rheumatol 8: 52-56, 19%, 94. Rosen FS, Cooper MD, Wedgwood R): The primary immunodeficiencies. N Engl ) Med 333: 431-440, 1995, 95. Fischer A:. Human primary immunodeficiency diseases: a perspective, Nat Immunol 5: 23-30, 2004. 96, De Saint BG, Arveiler B, Oberle I, et al: Close linkage of the locus for X chromosome-linked severe combined immunodeficiency to polymorphic DNA markers in Xqll-q13, Proc Natl Acad Sci USA 84: 7576-7579, 1987, 97. Noguchi M, Yi H, Rosenblatt HM, et aI. Interleukin-2 receptor gamma chain mutation results in X-linked severe combined immunodeficiency in humans. Cell 73: 147-157, 1993. 98. Hirschhorn R: Adenosine deaminase deficiency, Immunodefic Rev 2: 175-198, 1990, 99, Sakaguchi N, Takahashi T, Hata H, et al: Altered thymic T-cell selection due to a mutation of the ZAP-70 gene causes autoimmune arthritis in mice. Nature 426: 454-460, 2003, 100. Hori S, Takahashi T, Sakaguchi S: Control of autoimmunity by naturally arising regulatory CD4+ T cells. Adv Immunol 81: 331-371, 2003, 101, Hacein-Bey·Abina S, I.e Deist F, Carlier F, et al: Sustained correction of X·linked severe combined immunodeficiency by ex vivo gene therapy, N Engl) Med 346: 1185-1193, 2002, 102, Hacein·Bey-Abina S, Von Kalle C, Schmidt M, et al: LM02·associated clonal T cell proliferation in two patients after gene therapy for SCID·XI. Science 302: 415-419, 2003, 103, Markert ML: Purine nucleoside phosphorylase deficiency, Immunodefic Rev 3: 45-81, 1991. 104, Berthet F, I.e Deist F, Duliege AM, et al: Clinical consequences and treatment of primary immunodeficiency syndromes characterized by functional T and B lymphocyte anomalies (combined immune deficiency). Pediatrics 93: 265-270, 1994. 105, Cooper MD, Chae HP, Lowman )T, et al: Wiskott·Aldrich syndrome. An immunologic deficiency disease involving the afferent limb of immunity. Am ) Med 44: 499-513, 1968. 106. Kwan SP, Hagemann n, Radtke BE, et al: Identification of mutations in the Wiskott-Aldrich syndrome gene and characterization of a polymorphic di· nucleotide repeat at DXS6940, adjacent to the disease gene, Proc Nat! Acad Sci USA 92: 4706-4710, 1995. 107. Schaller )G: Immunodeficiency and autoimmunity, Birth Defects Orig Artie Ser 11: 173-184, 1975. 108. Dupuis·Girod S, Medioni J, Haddad E, et al: Autoimmunity in Wiskott· Aldrich syndrome: risk factors, clinical features, and outcome in a single-center cohort of 55 patients, Pediatrics 111 (Pt I): e622-e627, 2003. 109. Lau YL, Wong SN, Lawton WM: Takayasu's arteritis associated with WiskottAldrich syndrome. ) Paediatr Child Health 28: 407-409, 1992. 110, Schurman SH, Candotti F: Autoimmunity in Wiskott-Aldrich syndrome. Curr Opin Rheumatol 15: 446-453, 2003, 111. Akman 10, Ostrov BE, Neudorf S: Autoimmune manifestations of the Wiskott-Aldrich syndrome. Semin Arthritis Rheum 27: 218-225, 1998, 112, Rasmussen SA, Williams CA, Ayoub EM, et a1. Juvenile rheumatoid arthritis in velo-cardio·facial syndrome: coincidence or unusual complication? Am ) Med Genet 64: 546-550, 1996, 113, Pelkonen P, Lahdenne P, Lantto R, Honkanen Y: Chronic arthritis associated with chromosome deletion 22qll.2 syndrome. J Rheumatol 29: 2648-2650, 2002, 114, Davies K, Stiehm ER, Woo P, Murray K): Juvenile idiopathic polyarticular arthritis and 19A deficiency in the 22q11 deletion syndrome. ) Rheumatol 28: 2326-2334, 2001. 115. Sullivan KE, McDonald-McGinn DM, Driscoll DA, et al: Juvenile rheumatoid arthritis-like polyarthritis in chromosome 22q I 1.2 deletion syndrome (DiGeorge anomalad/velocardiofacial syndrome/conotruncal anomaly face syndrome). Arthritis Rheum 40: 430-436, 1997. 116, Gatti RA, Platt N, Pomerance HH, et al: Hereditary Iymphopenic agamma· globulinemia associated with a di,tinctive form of short-limbed dwarfism and ectodermal dysplasia. J Pediatr 75: 675-684, 1969. 117. Polmar SH, Pierce GF: Cartilage hair hypoplasia: immunological aspects and their clinical implications, Clin Immunollmmunopathol 40: 87-93, 1986. 118. Roifman CM, Melamed I: A novel syndrome of combined immunodeficiency, autoimmunity and spondylometaphyseal dysplasia, Clin Genet 63: 522-529, 2003. 119, Oen K, Petty RE, Schroeder ML: Immunoglobulin A deficiency: genetic stud· ies. Tissue Antigens 19: 174-182, 1982. 120, Cassidy )T, Oldham G, Platts·MiIls TA: Functional assessment of a B cell defect in patients with selective 19A deficiency. Clin Exp Immunol 35: 296-305, 1979.
656
C HAP T E R
33
IMMUNODEFICIENCIES AND THE RHEUMATIC DISEASES
121. Liblau RS, Bach ]F: Selective 19A deficiency and autoimmunity. Int Arch Allergy Immunol 99: 16-27, 1992. 122. Petty RE, Palmer NR, Cassidy]T, et al: The association of autoimmune diseases and anti-lgA antibodies in patients with selective 19A deficiency. Clin Exp Immunol 37: 83-88, 1979. 123. Warrington R], Rutherford W], Sauder P], Bees WC: Homologous antibody to human immunoglobulin (Ig)-A suppresses in vitro mitogen-induced 19A synthesis. Clin Immunol Immunopathol 23: 698-704, 1982. 124. Petty RE, Sherry DO, ]ohannson]: Anti-lgA antibodies in pregnancy. N Engl ] Med 313: 1620-1625, 1985. 125. Mochizuki S. Smith CI, Hallgren R, Hammarstrom L: Systemic immunization against 19A in immunoglobulin deficiency. Clin Exp Immunol 94: 334--336, 1993. 126. Gershwin ME, Blaese RM, Steinberg AD, et al: Antibodies to nucleic acids in congenital immune deficiency states. ] Pediatr 89: 377-381, 1976. 127. Pelkonen P, Savilahti E, Makela AL: Persistent and transient 19A deficiency in juvenile rheumatoid arthritis. Scand] Rheumatol 12: 273-279, 1983. 128. Cassidy .IT, Nordby GL: Human serum immunoglobulin concentrations: prevalence of immunoglobulin deficiencies, .I Allergy Clin Immunol 55: 35-48, 1975. 129, Cassidy .IT, Burt A, Petty R, Sullivan 0: Selective 19A deficiency in connective tissue diseases, N Engl] Med 280: 275, 1969. 130. French MA, Dawkins RL: Central MHC genes, 19A deficiency and autoimmune disease. Immunol Today 11: 271-274, 1990. 131. Salmi IT, Schmidt E, Laaksonen AL, et al: Levels of serum immunoglobulins in juvenile rheumatoid arthritis. Ann Clin Res 5: 395-397, 1973. 132. Itescu S: Adult immunodeficiency and rheumatic disease. Rheum Dis Clin North Am 22: 53-73, 1996. 133. van Rossum MA, Fiselier 1], Franssen MJ, et al: Effects of sulfasalazine treatment on serum immunoglobulin levels in children with juvenile chronic arthritis. Scand] Rheumatol 30: 25-30. 2001 134, Leickly FE, Buckley RH: Development of 19A and IgG2 subclass deficiency after sulfasalazine therapy. .I Pediatr 108: 481-482, 1986. 135. Cassidy .IT, Petty RE, Sullivan DB: Abnormalities in the distribution of serum immunoglobulin concentrations in juvenile rheumatoid arthritis . .I Clin Invest 52: 1931-1936, 1973. 136. Cleland LG, Bell DA: The occurrence of systemic lupus erythematosus in two kindreds in association with selective IGA deficiency. .I Rheumatol 5: 288-293, 1978, 137. Cronin ME, Balow .IE, Tsokos GC: Immunoglobulin deficiency in patients with systemic lupus erythematosus. Clln Exp Rheumatol 7: 359--364, 1989. 138. Petty RE, Cassidy .IT, Sullivan DB: Reversal of selective 19A deficiency in a child with juvenile rheumatoid arthritis after plasma transfusions. Pediatrics 51: 44-48, 1973139, Webster AD: InfJammatoty disorders of muscle. Echovirus disease in hypogammaglobulinaemic patients. Clin Rheum Dis 10: 189--203, 1984. 140. Good AE, Cassidy]T, Mutchnick MG, et al: Ankylosing spondylitis with selective 19A deficiency and a circulating anticoagulant. .I Rheumatol 4: 297-302, 1977. 141. Grimbacher B, Hutloff A, Schlesier M, et al: Homozygous loss of lCOS is associated with adult-onset common variable immunodeficiency. Nat Immunol 4: 261-268, 2003. 142. Bruton OC: A decade with agammaglobulinemia,] Pediatr 60: 672-676, 1962. 143. Kwan SP, Kunkel L, Bruns G, et al: Mapping of the X-linked agammaglobulinemia locus by use of restriction fragment-length polymorphism. .I Clin Invest 77: 649-652, 1986. 144, Simonte S], Cunningham-Rundles C: Update on primary immunodeficiency: defects of lymphocytes. Clin Immunol 109: 109--118, 2003. 145. Lee AH, Levinson AI, Schumacher HR ]r: Hypogammaglobulinemia and rheumatic disease. Semin Arthritis Rheum 22: 252-264, 1993. 146. Cunningham-Rundles C: Clinical and immunologic analyses of 103 patients with common variable immunodeficiency.] Clin Immunol9: 22-33, 1989, 147. Conley ME, Park CL, Douglas SO: Childhood common variable immunodeficiency with autoimmune disease. .I Pediatr 108: 915-922, 1986, 148. Sneller MC, Strober W, Eisenstein E, et al: NIH conference, New insights into common variable immunodeficiency. Ann Intern Med 118: 720-730, 1993. 149. Fu JL, Shyur SO, Lin HY, Lai YC: X-linked agammaglobulinemia presenting as juvenile chronic arthritis: report of one case. Acta Paediatr Taiwan 40: 280-283, 1999. 150. Cunningham-Rundles C: Hematologic complications of primary immune deficiencies. Blood Rev 16: 61-64, 2002. 1S1. Petty RE, Cassidy ]T, Tubergen DG: Association of arthritis with hypogammaglobulinemia. Arthritis Rheum 20: 441-445,1977. 152. Lederman HM, Winkelstein JA: X-linked agammaglobulinemia: an analysis of 96 patients. Medicine (Baltimore) 64: 145-156, 1985. 153. Uluhan A, Sager D, ]asin HE: Juvenile rheumatoid arthritis and common variable hypogammaglobulinemia. ] Rheumatol 25: 1205-1210, 1998.
154. Duzgun N, Duman M, Sone! B, et al: Lupus vulgaris in a patient with systemic lupus erythematosus and persistent IgG deficiency. Rheumatol Int 16: 213-216, 1997. 155. Swaak A], van den Brink HG: Common variable immunodeficiency in a patient with systemic lupus erythematosus. Lupus 5: 242-246, 1996. 156. Duzgun N, Peksari Y, Sonel B, et al: Localization of extrapulmonary tuberculosis in the synovial membrane, skin, and meninges in a patient with systemic lupus erythematosus and IgG deficiency. Rheumatol Int 22: 41-44. 2002, 157. Bardelas ]A, Winkelstein ]A, Seto OS, et al: Fatal ECHO 24 infection in a patient with hypogammaglobulinemia: relationship to dermatomyositis-like syndrome. .I Pediatr 90: 396-399, 1977. 158, Webster AD, Tripp ]H, Hayward AR, et al: Echovirus encephalitis and myositis in primary immunoglobulin deficiency. Arch Dis Child 53: 33-37. 1978. 159, Wilfert CM, Buckley RH, Mohanakumar T, et a!. Persistent and fatal centrdlnervous-system ECHO virus infections in patients with agammaglobulinemia. N EnglJ Med 296: 1485-1489, 1977. 160, Lacombe C, Aucouturier P, Preud'Homme JL: Selective IgG1 deficiency, Clin Immunol Immunopathol 84: 194--201, 1997. 161. Plebani A, Ugazio AG, Avanzini MA, et a!. Semm 19G subciass concentrations in healthy subjects at different age: age normal percentile charts. Eur .r Pediatr 149: 164--167, 1989. 162. Shackelford PG, Granoff OM, Polmar SH, et a!. Subnormal serum concentrations of IgG2 in children with frequent infections associated with varied patterns of immunologic dysfunction. .r Pediatr 116: 529--538, 1990. 163. Oxelius VA: Immunoglobulin G (IgG) subciasses and human disease. Am .I Med 76: 7-18, 1984. 164. Heiner DC, Lee SI, Short JA: IgG4 subclass defkiency syndromes. Monogr Allergy 20: 149--156, 1986, 165. Aucouturier P, Lacombe C, Bremard C, et a!. Serum IgG subciass levels in patients with primary immunodeficiency syndromes or abnormal susceptibility to infections. Clin Immunol Immunopathol 51: 22-37, 1989, 166. Jimenez A, Lopez-Trascasa M, Fontan G: Incidence of selective IgG2 deficiency in patients with vasculitis. Clin Exp Immunol 78: 149--152, 1989. 167. Rostoker G, Pech MA, Del Prato S, et a!. Serum IgG subclasses and IgM imbalances in adult 19A mesangial glomemlonephritis and idiopatIlic Henoch-Schilnlein purpura. Clin Exp 1mmunol 75: 30-34, 1989. 168. Cassidy .IT, Petty RE, Sullivan DB: Occurrence of selective 19A deficiency in children with juvenile rheumatoid arthritis. Arthritis Rheum 20 (Supp!): 181, 1977. 169. Huntley CC, Thorpe DP, Lyerly AD, et al: Rheumatoid arthritis with 19A deficiency. Am] Dis Child 113: 411-418, 1967. 170. Panush RS, Bianco NE, Schur PH, et al: Juvenile rheumatoid arthritis. Cellular hypersensitivity and selective 19A deficiency, Clin Exp ImmunollO: 103-115. 1972. 171. Pelkonen P, Salilahti E, Westeren L, et al: 19A deficiency in juvenile rheumatoid arthritis. ScandJ Rheumatol 8 (Supp!): 4. 1975. 172. Barkley DO, Hohermuth HJ, Howard A, et al: 19A deficiency in juvenile chronic arthritis. .I Rheumatol 6: 219--224, 1979. 173. Cassidy]T, Burt A, Petty RE, et al: Prevalence of selective 19A deficiency in patients with connective tissue disease. N EnglJ Med 280: 275, 1969. 174. Cassidy .IT: Selective 19A deficiency and chronic arthritis in children. In Moore TO (ed): Arthritis in Childhood. Report of the Eightieth Ross Conference on Pediatric Research, Columbus, OH, Ross Laboratories, 1981, P 82. 175. Jay S, Helm S, Wray BB: Progressive systemic scleroderma with 19A deficiency in a child. Am .I Dis Child 135: 965-966, 1981. 176, Spirer Z, !lie I, Pick A, et al: Localized scleroderma follOWing varicella in a 3-year-old girl with IgA deficiency. Acta Paedlatr Scand 68: 783-785. 1979, 177. Janeway CA, Gitlin D, Craig .1M, et al: "Collagen disease" associated with congenital agammaglobulinemia, Trans Assoc Am Physicians 69: 93-97, 1956. 178. Good RA. Rotstein .I, Mazzitello WF: The simultaneous occurrence of rheumatoid arthritis and agammaglobulinemia, ] Lab Clin Med 49: 343-357. 1957. 179, Page AR, Hansen AE, Good RA: Occurrence of leukemia and lymphoma in patients with agammaglobulinemia. Blood 21: 197-206, 1963. 180. Janeway CA, Rosen FS, Merler E, et al: The antibody deficiency syndromes. In Janeway CA, Rosen FS, Merler E (eds): The Gamma Globulins. Boston. Little, Brown, 1967, p 75. 181. Gotoff SP, Smith RD, Sugar 0: Dermatomyositis with cerebral vasculitis in a patient with agammaglobulinemia. Am .I Dis Child 123: 53-56, 1972, 182. Maguire JR, Perez-Atayde AR, Geha RS: Vasculitis presenting in an infant with agammaglobulinemia, Ann Allergy 57: 14--16, 42-45, 1986. 183. Crennan ]M, Van Scoy RE, McKenna CH, et al: Echovims polymyositis In patients with hypogammaglobulinemia. AmJ Med 81: 35-42,1986.
34
C HAP T E R
PERIODIC FEVER SYNDROMES IN CHILDREN Susannah Brydges, Balu Athreya and Daniel L. Kastner
~
Fever is one of the most common signs of illness in children. Most episodes are acute, of short duration, and usually caused by upper respiratory infections. When febrile episodes are prolonged beyond 2 to 3 weeks, infection is still the most common etiologic factor. However, after acute infectious causes have been excluded, conditions such as chronic infections, rheumatic diseases, and malignancy enter the differential diagnosis. Classically, a sustained or daily fever of at least 101 0 F (38.3 0 C), lasting more than 3 weeks, that eludes diagnosis despite a defined period of investigation,1-3 is denoted fever ofunknown origin (FUG). With the advent of new diagnostic tools, some advocate the use of the term FUG for children with unexplained fever for as few as 8 days.4 Repeated febrile episodes lasting for a few days to a few weeks are common in young infants and children attending day-care centers and kindergarten. Such episodes are often caused by repeated viral infections, although parents frequently worry about immune system defects. Infections in immunodeficient children are often caused by unusual or opportunistic pathogens (although children with hypogammaglobulinemia may contract the same infections as healthy children but at an increased rate) (see Chapter 33). Immunocompromised children often develop failure to thrive and other clinical signs of underlying pathology. Congenital organ malformations, such as bronchopulmonary sequestration or urethral vatves, can also predispose immunocompetent children to repeated febrile episodes. Frequent localization of infections to the same organ system should raise the suspicion of anatomic defects. If repeated infections due to immunodeficiency or organ malformation can be excluded, unexplained bouts of fever with a characteristic frequency and constellation of symptoms fall under the term recurrent or periodic fever syndrome. Such disorders are defined as three or more episodes of unexplained fever in a 6-month period, occurring at least 7 days apart. 5 These conditions may demonstrate strict periodicity or recur with variable intervals between attacks. Specific genetic mutations have been linked to some syndromes in this group, although the pathophysiology of others remains obscure.
INFECTIOUS CAUSES OF PERIODIC FEVER SYNDROMES Among the known causes of recurrent fevers (Table 34-1), infectious agents have been recognized for years and are still the most important causative factors in various parts of the world. Although malaria is probably the most common cause of cyclical fever, it is excluded from the periodic fever rubric because its cycles are counted in days (Le., every other day or every third day) instead of weeks or months. In the following sections, three infectious conditions that meet the previous criteria for periodic fever syndromes are described: brucellosis or undulant fever, rat-bite fever, and relapsing fever.
Brucellosis Brucella melitensis and Brncella abortus cause brncellosis or undulant fever, Mediterranean fever, or goat-milk fever. 6 Brucellosis is contracted by ingestion of contaminated, unpasteurized milk or milk products or through contact with infected animals by means of skin abrasions.7·8 Inhalation of the organism also can lead to infection, but this route is rare. 9 Human infections are rare except in areas of the world lacking proper pasteurization. After a 1- to 8-week incubation, a prodromal period of fatigue, headache, and myalgia begins. 10 Fever starts gradually and may peak above 40"C. It may occur daily or be intermittent, a pattern responsible for the name undulantfever. Other symptoms include weight loss, abdominal pain, lymphadenopathy, hepatosplenomegaly, and chronic monarthritis (see Chapter 28). Cutaneous involvement such as ulcerations, petechiae, and erythema nodosum may occur. Nervous system disease is uncommon. The febrile episodes can continue for a year or more, but the mortality rate is low (less than 2%), and death is associated with rare cases of endocarditis. A history of contact with farm animals or unpasteurized milk products should suggest brucellosis, confirmable by serology or culture of the organism from blood or cutaneous ulcers. 6
Rat-Bite Fever Rat-bite fever is caused by Spirillum minus or Streptobacillus moniliformis. ll In the spirillary type of rat-bite fever, also called Sodoku. in Japan, the skin at the site of the bite becomes indurated, erythematous, and suppurative 4 to 28 days after exposure (10 days on average). The patient has associated
657
658
,~.
C HAP T E R
tABLE 34 I
34
Rl'UJrrent
PERIODIC FEVER SYNDROMES IN CHILDREN
01
Pl'rlOdic h'ver Syndronlt's in
Childrl'n
Infectious Diseases
Brucellosis Rat-bite fever Relapsing fever Rheumatic Diseases Beh~et's
disease Systemic lupus erythematosus Relapsing polychondritis Crohn's disease Hereditary Autolnflammatory Syndromes
Familial Mediterranean fever (FMF) Cryopyrinopathies Familial cold autoinflammatory syndrome (FCAS) Muckle-Wells syndrome (MWS) Chronic infantile neurologic cutaneous and articular syndrome (CINCA), also called neonatal onset multisystem inflammatory disease (NOMID) Tumor necrosis factor receptor-associated periodic syndrome (TRAPS) Hyperimmunoglobulinemia D with periodic fever syndrome (HIDS) Cydlc Hematopoiesis (CH)
several days, the initial episode subsides dramatically, with profuse sweating, hypothermia, and profound weakness. The clinical disease often recurs, although with decreasing duration of illness and longer intervals between attacks. The causative organism can be identified in blood smears. 22
RHEUMATIC DISORDERS Among the rheumatic diseases, Bebr,;et's disease and relapsing polycbondritis can cause unexplained febrile episodes that sometimes conform to the criteria for the periodic fever syndromes. Beh~et's disease (see Chapter 27) is relatively uncommon in children; its exacerbations and remissions may suggest periodicity. The concomitant presence of oral and genital ulcers, uveitis (acute anterior and posterior), or cutaneous vasculitis should suggest this diagnosis. Relapsing polychondritis is even less common in children, but the associated clinical features are very suggestive: intense pain, redness, and tenderness of auricular cartilage (pinnae of the ear) or the cartilage of the nose and trachea. Systemic lupus erythematosus (see Chapter 16) may also manifest initially as an FUO or recurrent bouts of fever with rash and arthralgia. Unexplained, recurrent fever may also occur in Crohn's disease (see Chapter 15), although abdominal pain, (sometimes bloody) diarrhea, anemia, and growth delay should suggest this disorder. Patients with systemic-onset juvenile rheumatoid arthritis or sarcoidosis may present with fever, but the clinical presentation does not usually conform to that of the periodic fever syndromes.
Hereditary form AcqUired form Idiopathic CondOlons
Periodic fever with aphthous stomatitis, pharyngitis, and adenitis (PFAPA)
fever, and there is localized lymphadenopathy at the draining site. All signs and symptoms subside in 3 to 4 days, but after an asymptomatic period of several days, there may be recurrent cycles of fever, rash, and constitutional symptoms for several months or years. Complications include endocarditis, myocarditis, hepatitis, splenomegaly, and meningitis; 6% to 10% of untreated patients die. 12 Spirillum organisms can be identified in blood or biopsies from a lesion or adjacent lymph nodes. The streptobacillary type (the main form in the United States) also occurs after a rat bite. After an incubation period of less than 7 days, there is an abrupt onset of fever, chills, myalgia, headache, and pharyngitis. 13 ,14 A few days after the onset of fever, a morbilliform rash appears that may include the palms and soles. IS This rash may be maculopapular or petechial. A migratory polyarthritis is common. 13,14 If untreated, recurrent bouts of fever, rash, and arthritis may last for several months. About 10% of untreated cases are fatal.J2 Streptobacillus can be cultured from blood. l1
Relapsing Fever Borrelia recurrentis is transmitted person to person by ticks or lice and is responsible for recurrent or relapsing fever, 16 There are minor clinical differences between the louse-borne and tick-borne types. The onset is sudden, with high fever, headache, photophobia, myalgia, and arthralgia. Often, there is an associated diffuse, erythematous, macular rash. Petechiae may occur. Unlike rat-bite fever, there is no arthritis; there may be prominent central nervous system effects such as lethargy, stupor, meningeal signs, and cranial neuropathy. 16-21 After
HEREDITARY PERIODIC (RECURRENT) FEVER SYNDROMES A third class of periodic fevers is included among the inherited autoinflammatory syndromes, a newly recognized category affecting primarily the innate immune system. Patients present with episodic or, less commonly, fluctuating but continuous inflammation. Unlike patients with autoimmune disorders, patients with autoinflammatory disease lack high titers of autoantibodies or increased levels of self-reactive T cells. 23-25 At least six distinct disorders are grouped among the hereditary periodic fever syndromes, based on clinical findings and mendelian patterns of inheritance (Table 34-2). These hereditary periodic fever syndromes manifest with a broad spectrum of clinical findings, and there can be wide variation in sever-
'~III
IlIherilalJ<e Patterns of the Hereditary Pl'riodi< Fevel Syndrollles
TABL E 34- 2
Autosomal Dominant Pattern
Tumor necrosis factor receptor-associated periodic syndrome (TRAPS) Familial cold autoinflammatory syndrome (FCAS) Muckle-Wells syndrome (MWS) Chronic infantile neurologic cutaneous and articular syndrome (CINCA), also called neonatal onset multisystem inflammatory disease (NOMID) Autosomal Recessive Pattern
Familial Mediterranean fever (FMF) Hyperimmunoglobulinemia D with periodic fever syndrome (HIDS)
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
ity, even among patients with the same diagnosis. Treatment options are increasing for these disorders and, if begun early, may avert serious sequelae.
History Probably the first description of one of these conditions as a distinct hereditary disorder was published in 1940, when Kile and Rusk 26 reported an extended family with recurrent cold urticaria (see Chapter 35). Patients with this autosomal dominant condition had fever and an urticaria-like rash on exposure to cold temperatures, In 1945, SiegeP7 described the first series of patients with the condition that was later named familial Mediterraneanfever (FMF) by investigators in Israel. 28 FMF is an autosomal recessive illness characterized by bouts of fever, serositis, synovitis, or rash, sometimes resulting in systemic amyloidosis. The next of the periodic fever syndromes to be described, the Muckle-Wells syndrome (MWS) (see Chapter 35), was first reported in 1962 and is distinguished by its dominant pattern of inheritance, sensorineural hearing loss, and amyloidosis,29 In 1982, another dominantly inherited periodic fever in some ways clinically resembling FMF was reported as familial Hibernian (Irish) fever. 30 Subsequent identification of the causative genetic abnormality, coupled with the recognition thai this condition may occur in other ethnic groups, gave rise to the current nomenclature, tumor necrosisfactor (TNF) receptor-+associated periodic syndrome (TRAPS).23 Another FMF-like condition, the recessively inherited hYPerimmunoglobulinemia D with periodic fever syndrome (HIDS), was described in the Netherlands in 1984.31 The first descriptions of the inflammatory condition that is variously called neonatal onset multisystem inflammatory disease (NOMID)32 or chronic infantile neurologic cutaneous and articular syndrome (CINCA)33 were published in the early 1980s, The positional cloning of the gene for FMF in 199734,3'5 ushered in the era of molecular genetics for these disorders, eventually leading to the identification of four genes underlying the six hereditary recurrent fevers (Table 34-3). These molecular genetic insights led to the elucidation of some rather unexpected pathophysiologic connections among these illnesses,
Familial Mediterranean Fever Genetics and Pathogenesis FMF is the most common mendelian autoinflammatory syndrome, resulting from autosomal recessive mutations in the MEFV (Mediterranean fever) locus. 34,3'5 This dis-
t..
TABLE 34-3
order occurs most frequently among Sephardic Jewish, Arab, Armenian, and Turkish populations, with carrier frequencies as high as 1:3 to 1:5 in population-based surveys,3H2 FMF occurs at lower frequencies in other Mediterranean populations and ethnicities. 36.37M-48 More than 50 mutations have been described in MEFV, which encodes an immunoregulatory protein called pyrin or marenostrin.34.35 A list of mutations for FMF and the other hereditary periodic fever syndromes is available at http://fmfigh .cnrsfr/infeversl. Pyrin contains an N-terminal motif called the PYRIN domain,49--'54 a member of the death domain-fold superfamily of protein structural motifs, which includes the death domain (DD), death effector domains (DEDs), and caspase-recruitment domains (CARDS).'5'5--'59 The PYRIN domain of pyrin interacts with the PYRIN domain of a protein called apoptosis-associated specklike protein with a CARD (ASC).'5'5,~3 This interaction places pyrin upstream in several pathways regulating interleukin-1~ (IL-1~) secretion, nuclear factor-KB (NF-KB) activation, and apoptosis. 61 -63 ASC binds pyrin through cognate PYRIN domain interactions and oligomerizes with caspase-1 (also called IL-1~ converting enzyme [ICE]) through CARD-CARD binding. 6I ,64-67 The interaction of pyrin with ASC appears to be a key regulatory step in the ASC-caspase-1-IL-1~ cascade. Although the precise molecular mechanism of FMF-associated mutations remains to be elucidated, it is likely that these pyrin variants lead to accentuated innate immune responses, as demonstrated in pyrin-deficient mice. 61
Clinical Manifestations The first clinical episode usually occurs during childhood or adolescence, with 90% of patients having had onset by age 20 (Table 34-4).68-70 There is often a modest male predominance. 68 ,69,71 FMF attacks last between 12 and 72 hours and consist of inflammation involving the peritoneum, pleura, joints, or skin, sometimes in combination. Between episodes, patients usually are completely well and remain so for a few days to a few months. In children, fever may be the only sign of FMF, although other symptoms generally develop progressively with time. 70 .72 Abdominal symptoms often accompany the fever and range from mild discomfort and distention to severe pain
Molecular Genetics of the Hereditary Periodic Fever Syndromes
Chromosomal
Gene Symbol
$yndrome
location
FMF FCAS MWS CINCAINOMID TRAPS
16pl3.3 lq44 lq44 lq44 12p13
MEFV
HIDS
12q24
MVK
CIASI CIASI CIASI TNFRSFIA
Protein Pyrin (marenostrin) Cryopyrin Cryopyrin Cryopyrin 55-kD TNF receptor (p5S) Mevalonate kinase
659
Proposed Protein FundlonZ5 Regulation of IL-l~ production, NF-1d3 Regulation of IL-l~ production, NF-KB Regulation of IL-l~ production, NF-KB Regulation of IL-l~ production, NF-KB Inflammatory signal transdUclion
activalion, activation, activation, activation.
apoptosis apoptosis apoptosis apoptosis
Isoprenoid biosynthesis
Clj\/CAINOMID. chronic infantile neurologic cutaneous and articular syndrome, also called neonatal onset multisystem inflammatory disease; FCAS, familial cold aUloinflammatory sl'ndrome; FMF, familial Mediterranean fever; HlDS, hyperimmunoglobulinemia 0 with periodic fever syndrome; IL-l~, interleukin-l~; MWS, Muckle-Wells syndrome; NF-KB, nuclear factor-KB; TNF, tumor necrosis factor: TRAPS. tumor necrosis factor receptor-associated periodic syndrome.
660
t= II
C HAP T E R
TABLE 34--4
34
PERIODIC FEVER SYNDROMES IN CHILDREN
Summary of Clinical Findings Asso( ialed with
Olnlal Manlfestadon FMF Duration of attacks Cutaneous
Abdominal
MWS
ONWNOMID
TRAPS
HIDS
12-72 hr
12-24 hr
2-3 days
Continuous
often >7 days
3-7 days
Erysipeloid erythema
Cold-induced urticaria-like rash Nausea
Urticaria-like rash
Urticaria-like rash
Sometimes abdominal pain Rare Polyarthralgia, oligoarthritis
Uncommon
Migratory rash, underlying myalgia Peritonitis, diarrhea, or constipation
Nonmigratory maculopapular rash on trunk, limbs; urticaria Severe pain, vomiting, diarrhea >constlpation, rarely peritonitis Rare Symmetric polyarthritis, arthralgia
Ocular
Conjunctivitis
Neurologic
Headache
Headache
Lymph/ spleen Vasculitis
Splenomegaly> lymphadenopathy HSP, polyarteritis nodosa Variable risk depending on MEFV, SAA genotypes, family history, gender, compliance with treatment
Not seen
Rare
Not seen
Not seen
Rare
Occurs in =25%
Amyloidosis
Fevers
FCAS
Peritonitis, constipation> diarrhea Frequent Monarthritis, occasionally protracted in knee or hip Rare
Pleural AJ1hropathic
~Iereditary Periodic
Not seen Polyarthralgia
Conjunctivitis, episderitis Sensorineural deafness
Rare Epiphyseal overgrowth, contracnlres, intermittent or chronic arthritis Conjunctivitis, uveitis, vision loss Headache, deafness, aseptic meningitis, mental retardation Adenopathy, hepatosplenomegaly Occasional May develop in a portion of patients reaching adulthood
Frequent Arthralgia, arthritis in large joints
Conjunctivitis, periorbital edema Rare
Rare
Splenomegaly > lymphadenopathy HSP, lymphocytic vasculitis Occurs in =10%
Cervical adenopathy
Headache
Cutaneous vasculitis, rarely HSP Rare
CINCA/NOMID, chronic infantile neurologic cutaneous and articular syndrome, also called neonatal onset multisystem inflammatory disease; FCAS, familial cold autoinflammatory syndrome; FMF. familial Mediterranean fever; HIDS, hyperimmunoglobulinemia D with periodic fever syndrome; HSP, Henoch-Schonlein purpura; MWS. Muckle-Wells syndrome; TRAPS, tumor necrosis factor receptor-associated periodic syndrome.
with rigidity.68 Constipation is more common than diarrhea, and in extreme cases, peristalsis may cease and result in paralytic ileus. Pain can be generalized or focused in a quadrant, sometimes mimicking acute appendicitis. Pleural pain is generally unilateral, occurring with decreased breath sounds. Less commonly, a small effusion, friction rub, or atelectasis may be present. 73 Joint manifestations are common and sometimes the first sign of the disease in children. 74 Arthralgia occurs more frequently than arthritis. Arthritis in adults usually is monarticular, although children may have involvement of several joints, symmetrically or asymmetrically, with pain and large effusions. 75.76 Synovial aspirates from joints are sterile, but they may demonstrate elevated leukocyte counts that are usually associated only with septic arthritis. Rarely, arthritis in the knee and hip may have a protracted course.77 In these cases, radiographic changes may include severe juxta-articular osteoporosis, erosions, and osteonecrosis. Cutaneous findings are less common than serosal or synovial involvement. Most commonly, there is an erysipeloid erythematous rash (Fig. 34-1) on the dorsum of the foot, ankle, or lower leg. 78.79 The rash may occur alone or in conjunction with other manifestations. Biopsies of the rash are characterized by a prominent mixed cellular infiltrate. 79 Findings less commonly associated with FMF include episodes of unilateral acute scrotal pain in prepubescent boys,8CH32 febrile myalgia,83.84 and diverse cutaneous
manifestations including Henoch-Sch6nlein purpura.68.72.85 Rarely, pericarditis is observed. 86 Beh~et's disease,87-9t polyarteritis nodosa,92-98 microscopic polyarteritis,98 and glomerulonephritis99-102 may occur more frequently in FMF patients than in the general population. Although headache and febrile seizures may occur
• Figure 34-1 Erysipeloid erythema on the right ankle of a patient with familial Mediterranean fever. This painful rash usually lasts for several days and may occur alone with fever or in conjunction with arthritis of the adjacent joint. (See color insert.)
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
in p¢diatric patients, other neurologic symptoms are rare. 'There may also be a higher than expected frequency of inflammatory bowel disorders among FMF patients. lo3.J04
Laboratory Investigations During attacks, concentrations of acute phase reactants such as C-reactive protein (CRP), serum amyloid A (SAA), and complement increase. Leukocytosis and an increased erythrocyte sedimentation rate (ESR) are commonly obse~ed.
The continuous elevation of these acute phase serum proteins during and even between attacks lO5-107 sometimes eventuates in the development of AA systemic amyloidosis, the most serious sequela of FMF. SAA deposition occurs in several organs,68 and renal failure occurred by age 40 in many patients before effective treatment was available. The risk of amyloidosis increases with a positive family history of this complication, male sex, the ala. genotype at the serum amyloid Al (SAAl) locus, and poor compliance with colchicine therapyyl8-112 In most studies, homozygosity for the M694V mutation also predisposes patients to amyloidosis, as well as to arthritis and erysipeloid erythema,74,110.112-114 although the association with amyloidosis has not been observed in Turkey.1I5 For reasons that are not clear, the risk of amyloidosis appears greater in the Middle East than in the United States. An early indicator of impaired renal function is microalbuminuria, and periodic urinalyses are an important part of continuing care for FMF patients. After proteinuria occurs, amyloidosis can be confirmed by biopsy of the kidney or rectum. Although kidney biopsy is more sensitive, rectal biopsy is preferred because it is safer, less invasive, and still has a sensitivity of 75%.11 6
Diagnosis The diagnosis of FMF is based on the presence of short (12 to 72 hours), recurrent (three or more) febrile episodes, with abdominal, chest, joint, or skin manifestation$ and no discernible infectious cause. 1I7 A favorable resBonse to colchicine, appropriate ethnicity, positive famUy history, and onset before the age of 20 also support the diagnosis. Because physicians in the Western Hemisphere are not' as familiar with FMF as clinicians in regions with a higliler prevalence, genetic testing has become a valuable adjlJnct to clinical diagnosis, especially in North America and, Europe. Based on its autosomal recessive inheritanqe, patients with FMF were expected to be homozygous for a single mutation or heterozygous for two diffhent mutations. However, the reality is not always tha~ simple. Certain mutations, most notably the substitution of alanine for valine at residue 726 (V726A) and the sUbftitution of glutamine for glutamic acid at position 148 (E148Q), are sometimes found in cis in so-called comple~ alleles,37.44 and it is possible for some patients to have three or even four demonstrable mutations. Depending on the laboratory, DNA samples are often screened only for the most common mutations, and patients with rare mutations therefore may appear to
661
have no mutations or only one. Even complete sequencing of MEFV exon 10 (where most mutations lie) will fail to diagnose patients with mutations in other regions of the gene. Sequencing of the entire MEFV coding sequence fails to identify any abnormalities in a small number of patients who respond well to colchicine and exhibit FMF symptoms, suggesting that there may be more than one gene causing FMFYS-120 To further complicate the issue, some rare mutations appear inherited in a dominant fashion,121 and approximately 30% of patients with clinical signs of FMF have only one demonstrable mutation. 37.113,120,122-124 A diagnosis of FMF should never be excluded based solely on the results of genetic testing, However, the clinical and ethnic spectra of FMF have definitely expanded with the availability of genetic testing,36 suggesting that a combination of clinical evaluation with genetic testing for selected patients is the most sensible diagnostic approach.
Treatment Colchicine therapy is highly effective for most patients in preventing febrile episodes and systemic amyloidosiS. 12 5-129 Approximately 95% of patients demonstrate a marked improvement in symptoms, whereas almost 75% have a near-complete remission. Continuous therapy is generally more effective in controlling the attacks of FMF than intermittent treatment at the time of attacks, and daily therapy has the important added benefit of reducing the subclinical inflammation between episodes that potentially leads to amyloidosis.I05-107 Colchicine is generally safe in children, although colchicine pharmacokinetics may differ in younger patients, and doses adjusted for body weight may be greater in children than those used in adults. The recommended adult colchicine dose is 1.2 to 1.8 mg/day. Dosage should be started as low as possible (one half of a O.6-mg tablet once daily) and slowly increased, titrating to maximize efficacy and minimize side effects, but usually not exceeding 1.8 mg/day.130,131 A gradual increase in dose often prevents or lessens diarrhea, the most common adverse effect. Some patients develop lactose intolerance due to colchicine, and a lactose-free diet may help to control gastrointestinal symptoms. 132 Simultaneous treatment with colchicine and other drugs that are metabolized by-or that inhibit-the CYP3A4 liver enzyme system, such as erythromycin and cimetidine, can increase colchicine blood levels to toxic concentrations. 132- 135 There are no established alternatives in patients who are unresponsive to or cannot tolerate therapeutic doses of colchicine, although the role of pyrin, the FMF protein, in cytokine regulation suggests a possible future role for biologics.
Outcome and Prognosis Among FMF patients with end-stage renal amyloidosis, the survival rate on hemodialysis is lower than among agematched controls, perhaps because of poor vascular access and hemodynamic instability.I36-140 Studies have confirmed little difference in patient and graft survival between FMF and control kidney transplant recipients 14I ,142;
662
C HAP T E R
34
PERIODIC FEVER SYN DROMES IN CHILDREN
transplantation (with oral colchicine administration to prevent amyloidosis in the transplanted kidney) is the preferred treatment for renal failure. 143
The Cryopyrlnopathles A second group of inherited recurrent fevers are the cryopyrinopathies (see Chapter 35): familial cold autoinflammatory syndrome (FCAS), MWS, and CINCA!NOMID). All three disorders arise from autosomal dominant mutations in the eIASl IOCUS,I44-148 which encodes a protein that contains an N-terminal PYRIN domain. Although these three syndromes had been recognized as separate entities, there is a continuous range in severity, with a number of cases falling between the clinical boundaries defined in the literature. All three disorders share an urticaria-like rash (Fig. 34-2) and involve episodic or fluctuating but continuous inflammation, which varies from fairly mild to debilitating, depending on the particular genetic lesion and other factors (see Table 34--4).144-151
Tumor Neaosls Fador Receptor-Assodated Periodic Syndrome One of the first clinical descriptions of TRAPS was that of a large family of Irish/Scottish ancestry, with an illness denoted as familial Hibernian fever. 30,ls2 With the discovery of mutations in the TNFRSF1A gene,23 which encodes the 55-kD TNF receptor in this family and in several other families of non-Irish ancestry, the current TRAPS nomenclature was proposed.
Genetics and Pathogenesis TRAPS is inherited as an autosomal dominant trait, although in some cases, a clear pattern of inheritance cannot be discerned because of reduced penetrance in mutation-positive relatives or, rarely, because of de novo mutation. TRAPS has occurred in patients of many ethnicities. It is the second most common periodic fever disorder, with more than 40 known mutations in 1NFRSF1A. 23,IS3-i64 The 55-kD TNF receptor is widely expressed on cell surfaces and mediates a number of proinflammatory effects on ligand binding. Signaling through the receptor
• Figure 34-2 Generalized, urticaria-like rash in a patient with MuckleWells syndrome. Unlike true urticaria, in which mast cells are present, the inflammatory infiltrate is composed primarily of neutrophils. (See color insert.)
during an immune response usually leads to the shedding of the extracellular domains of both the p55 and p75 TNF receptors. 16S ,166 This process contributes to a pool of soluble receptors that compete with the membrane-bound receptor for its ligand (Le., TNF) and cause an attenuation of the inflammatory response. 167 Several TRAPS-associated TNFRSF1A mutations impair normal receptor shedding, which has been hypothesized to result in repeated signaling and prolongation of the inflammatory response. Impaired receptor shedding has been observed by flow cytometry in patients with some but not all mutations. 23 ,ISS,161,162 Moreover, impaired cleavage does not seem to correlate with disease severity, suggesting that there must be additional mechanisms by which TNFRSF1A mutations cause autoinflammatory disease.
Clinical Manifestations The clinical manifestations of TRAPS are more similar to FMF than to the cryopyrinopathies (see Table 34--4). TRAPS causes episodic fever and inflammation with serosal, synovial, and cutaneous manifestations. Distinguishing characteristics of TRAPS include longer attacks (1 to 4 weeks or more) and conspicuous eye and skin symptoms. 168,169 TRAPS attacks may be precipitated by minor trauma or infection or by stress and physical exertion. During attacks, patients exhibit vigorous acute phase responses that sometimes persist into the intercritical period, albeit at lower intensity,169 Cutaneous symptoms associated with TRAPS are often distinctive, consisting of migratory, macular areas of erythema that occur on the torso (Fig, 34-3) or on an extremity.I68,169 These cutaneous lesions are warm and tender and consist of superficial and deep perivascular infiltrates of mononuclear cells. When lesions occur on the limbs, there may be an associated myalgia due to inflammation of the underlying fascia. 170 Other types of rash may also occur, including annular patches and generalized serpiginous plaques. 168,169
• Figure 34-3 Migratory, erythematous macular rash on the abdomen and chest of a patient with the tumor necrosis factor receptor-assodated periodic syndrome (TRAPS).The rash extends from the midline to the right lateral chest wall. Notice the surgical scars from previous exploratory laparotomies. (See color insert.)
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
Clihical attacks may include peritoneal inflammation or pleurisy, or both. Ocular inflammation with periorbital edeIlla or conjunctivitis is common. 169 Arthralgia is more prominent than arthritis, and it generally involves single joints, especially the hips, knees, and ankles. Scrotal inflammation may occur. IS2 Amyloidosis, although less common than in untreated FMF, affects about 10% of patients and can lead to renal or hepatic failure. IS 5-IS7,169.171 The risk of amyloidosis appears to be greater among patients with cysteine mutations. ISS A positive family history of amyloidosis may increase the risk for other relatives.
Laboratory Investigations Levels of SAA, CRP, and serum complement components are increased, and most patients exhibit leukocytosis and thrombocytosis, with an accelerated ESR.
Diagnosis More than 40 point mutations in TNFRSFl have been described. 23,IS3-164,172 The specific diagnosis is defined by mutations in this locus. Patients without mutations are considered to have a TRAPS-like phenotype,lss,162 possibly caused by mutations in related genes.
Treatment Treatment depends on the severity of the underlying disease. For some patients with relatively infrequent episodes, tapering doses of prednisone at the time of attacks may be effective and relatively safe. For patients with more severe disease, the recombinant TNF receptor antagonist, etanercept (0.4 mg/kg, with a maximum of 25 mg, twice each week), is effective in preventing attacks. 24,169,173 Some patients require dosing three times each week or 0.6 mg/kg twice weekly to achieve an adequate response. Colchicine usually has no effect on symptoms or the development of amyloidosis.1 69 .171 The prognosis depends on the development of amyloidosis. More aggressive anti-TNF therapy may be indicated in patients with a positive family history of amyloidosis or mutation at cysteine residues to suppress subClinical inflammation.
Hyperlmmunoglobullnemla Dwith Periodic Fever Syndrome HIDS is an autosomal recessive disease l74 that was initially described in several patients of Dutch heritage. 31 HIDS is caused by mutations in the MVK gene, which encodes mevalonate kinase.l75-177 HIDS occurs mainly in patients of northern European ancestry, and approximately 50% of patients are of Dutch ancestry. 178-183
Genetics and Pathogenesis Mevalonate kinase is a peroxisomal enzyme that catalyzes the conversion of mevalonic acid to 5-phosphomevalonic acid in the synthesis of sterols (Le., cholesterol, steroid hormones, vitamin D, and bile salts) and' nonsterol isoprene compounds (Fig. 34-4).177.184.185
663
Isoprenes are involved in a variety of cellular functions, including electron transport, protein glycosylation and synthesis, and prenylation of adenine transduction proteins. Mutations associated with HIDS lead to markedly reduced mevalonate kinase enzymic activity,17S,176 whereas the mutations in the clinically more severe mevalonic aciduria result in the absence of enzymic activity.186 Excessive production of proinflammatory cytokines by HIDS mononuclear cells may result from excessive accumulation of mevalonic acid substrate or be related to deficiencies in isoprenoids synthesized through the mevalonate pathway.187 If the latter possibility is correct, small GTP-binding proteins, which undergo prenylation, may be the link between the mevalonate pathway and the febrile attacks of HIDS.I88
Clinical Manifestations HIDS manifests in early childhood, often by 6 months of age (see Table 34-4). Attacks last about 3 to 7 days, usually separated by 1- to 2-month, symptom-free intervals. Episodes are often heralded by chills and headache, a rising fever, abdominal pain, nausea, and vomiting, sometimes precipitated by immunizations, surgery, trauma, and mild infections.180 The mevalonate kinase enzyme in patients with HIDS-associated mutations loses activity at supraphysiologic temperatures, perhaps explaining the association of immunizations, upper respiratory infections, and other inflammatory provocations with attacks. 189 Some patients develop a nondestructive arthritis, usually in the large joints, that is associated with attacks. 180 ,190-192 This arthritis is often polyarticular, unlike that associated with FMF. Protracted joint manifestations are rare. During attacks, widespread, erythematous macules develop that are sometimes painful,lso.193 The rash is usually not migratory, differentiating it from the rash associated with TRAPS, and it has no predilection for the lower legs, unlike that of FMF. The HIDS rash may be a diffuse maculopapular eruption (Fig. 34-5) extending to the palms and soles, or it can be nodular, urticarial, or morbilliform. Skin biopsies show perivascular inflammatory cells and deposits of antibody or complement component C3, or both. Oral and vaginal aphthous ulcers are common. Henoch-Schonlein purpura l90 and erythema elevatum diutinum (a benign type of necrotiZing vasculitis)179 have been reported. Cervical lymphadenopathy is a common manifestation of HIDS, as are severe headache and splenomegaly.ISO Pleurisy is uncommon. Clinical findings are summarized in Table 34-4.
Laboratory Investigations Most patients have elevated serum immunoglobulin D levels, but how this observation contributes to the clinical disease is poorly understood. Some patients with periodic fever and MVK mutations have normal IgD levels, suggesting that an elevated IgD concentration may be an epiphenomenon.l76,188,I94.19s Patients also exhibit an accelerated ESR, leukocytosis, and elevated levels of CRP I80 ,182,194 during and, less commonly, between attacks. Elevated levels of mevalonic acid are usually detected in urine during attacks. 17S ,176,1%,197
664
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
A_O_s/_C_OA_U~oA
A"'YI CoA [
3-hydroxy3-methylglutaryl CoA
[
.JLU
OH
CoA
}
_ _ YI CoA
J
' - - - -
Mevalonate
['----OH~_O OH
OH
]
.. Mevalonate kinase
Mevalonate-P
[
O_H~_H
P_____'O,]
Mevalonate-PP
Isopentenyl-PP
[....._
Geranyl-PP
~_C_H_2
P02 P03
...-J_ -. 1,0p""yl,dwno-.RNA
(~~-p--------~pO'
]
Famesyl-PP
[
....
Dolichol PO PO ..... Heme A 2 3 Ubiquinone ----' Famesylated proteins
J
Squalene
Cholesterol
Steroid honnones ..... Vitamin D Bile acids Lipoproteins
OH • Figure 34-4 The mevalonate pathway. Patients with the hyperimmunoglobulinemia 0 with periodic fever syndrome have mutations in mevalonate kinase that result in enzyme activity that is markedly diminished but not absent. Patients with clinically more severe mevalonic aciduria have mutations leading to an almost total loss of enzyme activity.
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
665
Periodic Fever due to (ydle Hematopoiesis Cyclic hematopoiesis (CH), or cyclic neutropenia, is a rare disorder consisting of febrile episodes due to periodic granulocytopenia, interspersed between intervals of relatively normal granulocyte counts. CH may occur as a sporadic congenital disorder, an autosomal dominant inherited disease, or an acquired condition. 204-207 Cyclic neutropenia is a more common term for this condition, although cyclic hematopoiesis is more descriptive, because other formed elements of blood in addition to neutrophils also demonstrate cyclic variations in numbers. 206 Although the fevers associated with CH are sometimes caused by infectious agents, patients with CH may develop fevers in the absence of apparent infection,5 perhaps due to the large-scale apoptotic death of bone marrow precursors that underlies the variation in circulating mature forms. 207
• FIture 34-5
Adiffuse maculopapular rash also extended over the palms and soles of a patient with the hyperimmunoglobulinemia 0 with periodic fever syndrome. (See color insert.)
Diagnosis A diagnosis may require several lines of inquiry, including clinical observation, genetic testing, serum IgD measurement, and assay of mevalonate in urine. Modest elevations in IgD should be interpreted with caution, because this phenomenon is common in several other conditions, including chronic infections, acquired immunodeficiency syndrome (AIDS), Hodgkin's disease, and other periodic fever syndromes. 180 ,198-200 Most laboratories that perform genetic testing for HIDS screen for the V377I mutation (Le., substitution of isoleucine for valine at residue 377), because most HIDS patients are heterozygous for this mutation.]83,196 However, even with complete sequencing of the coding region of MVK, genetic testing may be inconclusive. About 25% of patients do not have MVK mutations, suggesting the existence of other genetic phenotypes. 20 ] Based on a combination of diagnostic indicators, three types of HIDS with very similar symptoms have been proposed. Classic RIDS is defined by elevated IgD levels, a mutation in MVK, and mevalonic acid in the urine during attacks. Variant RIDS patients also have recurrent fevers and elevated IgD levels, but they lack mutations in Ml/K and do not excrete high levels of mevalonate. 201 Dutch-type periodic fever patients have normal levels of IgD but are positive for mutations in MVK and excrete increased levels of mevalonate in urine. 194
Treatment Various treatment" have been proposed. A few patients may respond to colchicine. Glucocorticoids, immunoglobulin, and cyclosporine have all been tried with various success rates. Two small studies demonstrated improvement with etanercept l88 and simvastatin. 202 HIDS is not generally associated with a shortened lifespan,l80 although HIDS-associated amyloidosis has been reported. 203
Genetics and Pathogenesis The clinical features of autosomal dominant, familial CH are indistinguishable from those of the sporadic form, suggesting that the sporadic variety may represent unrecognized familial cases or de novo mutations in CH genes. Inherited CH is caused by mutations in the neutrophil elastase-2 gene (EIA2J,208-21O which encodes neutrophil elastase (NE). Mutations in the growth factor independent-1 gene (GFIl),211 which encodes a transcription factor that controls expression of NE, causes severe congenital neutropenia, a noncyclical disorder. CH has also been related to mutations in dogs in an adaptor protein (AP3Bl) bound to NE,21O,212 although a causal connection has yet to be established between CH and AP3B1 in humans. The current hypothesis is that CH is a mistrafficking disorder of NE. Computational modeling suggests that NE is a membrane protein, and that EIA2 mutations affect primarily the transmembrane domains, leading to excessive deposition of NE in intracellular granules. 212 In contrast, AP3Bl mutations modify the adaptor protein in such a way that there is excessive transport to the plasma membrane. 212 Adult-onset CH may be a benign neoplasm with clonal proliferation of large granular lymphocytes. m -215
Clinical Manifestations Clinical manifestations of CH start in early childhood, with the earliest reported case occurring in the first few weeks of life (Table 34-5).206 The cycle length is typically 21 days (range, 14 to 36 days), and each febrile cycle lasts 3 to 10 days.206,216 In older persons, the cycles may not be evident. During attacks, the absolute neutrophil count (ANC) is less than 200/dL (0.2 x 109 L) and may be O. While patients are neutropenic, they are highly
I!: II
TABLE 3/.-5
Clinical Features of Cyclic
Ilenldtop()le~is
1. Typical cycles recur approXimately every 21 days. 2, Absolute neutrophil count is less than 0.2 x 109fL. 3. Absolute neutrophil count is low normal to mildly neutropenic between cycles,
666
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
susceptible to infections from normal flora, resulting in recurrent oral ulcers, gingivitis, fever, and lymphadenopathy. Although these infections are usually mild to moderate in severity, severe infections due to Clostridium or Escherichia coli, with abdominal pain and vomiting rapidly progressing to necrotizing enterocolitis, may occur. Clostridium septicum infection has caused enterocolitis, myonecrosis, and death.217-220 Other uncommon and less serious symptoms include bone pain, fatigue, malaise, diarrhea, and headache. Symptoms improve rapidly as neutrophil counts recover. Children are well between attacks, with ANCs in the low normal to mildly neutropenic range. Blood monocyte counts cycle in opposite fashion, so that the peak monocyte count coincides with the nadir of the ANC. 21S Reticulocytes, platelets, and eosinophils also may oscillate with neutrophils. 221 An acute phase response may be observed during the neutropenic episodes. Results of bone marrow examination are characterized by intramedullary destruction of promyelocytes and defects in granulopoiesis 222 due to accelerated apoptosis. 223
Diagnosis Based on extensive family studies,206 the diagnosis of autosomal dominant CH can be established with reasonable accuracy based on the following criteria: regular, cyclic fluctuations in peripheral blood neutrophil counts, with a periodicity ranging from 19 to 21 days, and documentation of neutrophil counts less than 0.2 x 109/L during periods of neutropenia. Complete blood counts should be determined two or three times each week for at least 6 weeks. 206 Genetic testing may play an adjunctive role, especially in families in which formes fruste are suspected or when there is no family history but is a suspicion of de novo mutation.
rare. Early loss of permanent teeth associated with chronic gingivitis is common to all forms of neutrope. t'Ion WIt. h mal'Ignancy h as been nl'a . 221>-230 N0 assocla observed.
Periodic Fever with Aphthous Stomatitis, Pharyngitis, and Adenitis Periodic fever with aphthous stomatitis, pharyngitis, and adenitis (PFAPA) syndrome (Le., Marshall's syndrome) was described in 1987. 231 It is a relatively benign and common condition that has been reported in several areas of the world.232.233 The underlying cause is unknown. No infectious cause has been discovered, although the clinical features suggest it. PFAPA does not appear to be inherited, because most cases occur sporadically. PFAPA can be considered idiopathic.
Clinical Manifestations The onset of PFAPA is usually before the age of 5 years. In an American series23 4, febrile episodes occurred approximately every 28 days (range, 26 to 30) and lasted for a mean of 5 days, although a French group reported a longer interval between flares. 235 Children are healthy between episodes and grow normally. Malaise, fatigue, and oral lesions may herald the onset of a cycle. In the largest series reported,234 70% of patients had aphthous stomatitis, characterized by shallow ulcers in the buccal mucosa and pharynx that lasted for 3 to 5 days and healed without scarring. Seventy-two percent had pharyngitis, consisting of an intensely red, ulcerated pharynx, without exudate. Although cervical adenitis is a major feature of the disease in 88% of patients, lymph nodes in other locations usually are not enlarged. Hepatosplenomegaly is rare. Arthralgia is reported in 10% to 15%. The maximum temperature of 40 C to 41' C is usually reached within the first day and may end abruptly or settle down over 1 or 2 days. 0
Treatment Treatment with granulocyte colony-stimulating factor (G_CSF)224-226 or granulocyte-macrophage colony-stimulating factor (GM-CSF)227 may be effective. The recommendation is to administer G-CSF subcutaneously at doses of 1 to 5 Ilglkglday. Symptoms are controlled by this treatment, and the cycles are shortened, with an increase in the nadir ANc. 225 Infections must be treated promptly and aggressively. E. coli and Clostridium species precipitate serious and often fatal illness. Appropriate cultures should be obtained, particularly if a child develops abdominal pain with diarrhea and vomiting. Typhlitis (i.e., inflammation of the cecum) and perforating enterocolitis should always be considered.
Outcome and Prognosis Prognosis appears to be good, except for the increased mortality rate associated with infection. 206 With age, the cycles are less prominent, and symptoms improve. Sinusitis and bone pain become more common, whereas fever, lymphadenopathy, and skin infections become
Laboratory Investigations During episodes, there is an increase in the total white blood cell count and elevation of acute phase reactants. Neutropenia usually is not present, but mild elevations in serum IgG, IgM, and IgA may occur. Elevated levels of IgD were reported in one study236 but not in another. 234 Increased serum levels of interferon-y, TNF, and IL-6 have been observed with fevers,234 suggesting that perturbations in the cytokine network may be responsible for this disease.
Diagnosis Based on a 10-year registry, which includes 94 PFAPA patients,234 the following diagnostic criteria were suggested (Table 34-6): recurrent febrile cycles with an onset before age 5 and at least one of the following features: aphthous stomatitis, cervical adenitis, or pharyngitis. Patients are asymptomatic and grow normally between cycles.
C HAP T E R
' : II
34
PERIODIC FEVER SYNDROMES IN CHILDREN
IABLE 34-6 Clinical Featu~~s of the Sy.n~rome of P~riodic Fever with Aphthous Stomahtls. Pharyngitis, and Cervical Adenopathy
1. 'Regularly recurring fevers at an early age at onset «5 years of
age) 2. Constitutional symptoms in the absence of upper respiratory infection with at least one of the following: a. Aphthous stomatitis b. Cervical adenitis Co Pharyngitis 3. Exclusion of cyclic neutropenia and known hereditary periodic fever syndromes 4. Asymptomatic intervals between episodes 5. Normal growth and development
Treatment Several treatments have been proposed, including nonsteroidal anti-inflammatory drugs CNSAIDs), colchicine, glucocorticoids, and cimetidine. In many patients, glucocorticoids started early during a cycle seem to be effectiv¢ in aborting attacks. 234 ,235 Although a single dose is often effective, some children may require treatment for 3 to 4 days, Steroid therapy often aborts attacks, but it results in a shorted interval between episodes. 234 Acetaminophen or NSAIDs with or without antibiotics may be of modest benefit,237 although the cycles will return. Cimetidine may be effective at preventing recurrences. 238,239 Some investigators have noticed that tonsillectomy and adenoidectomy may eliminate attacks. 240
Outcome and Prognosis Prognosis seems to be excellent. In a lO-year registry,234 cyclic episodes ceased after a mean of 4.5 years from the on~etj approximately one third of patients stopped having episodes. In other patients, the symptoms became less intense and less frequent with the passage of time. Two patients continued to have episodes even after 17 years of follow-up. Neither developed malignancies or autoimmune disorders.
REFERENCES 1. Petersdorf RG, Beeson PB: Fever of unexplained origin: repon on 100 cases. Medicine (Baltimore) 40: 1, 1961. 2. Durack DT, Street AC: Fever of unknown origin-reexamined and redefined. Curr Clin Top Infect Dis 11: 35, 1991. 3. Miller LC, Sisson BA, Tucker LB, et al: Prolonged fevers of unknown origin in children: pallerns of presentation and outcome.] Pediatr 129: 419, 1996. 4. Lorin M, Feigin R: Fever of undetermined origin. In Oski F, McMillan]A (ed): Oski's Pediatrics: Principles and Practice, 3rd ed. Philadelphia, Uppincoll Williams & Wilkins, 1999, pp 1116--1119. 5. John CC, Gilsdorf ]R: Recurrent fever in children. Pediatr Infect Dis] 21: 1071, 2002. 6. Al Dahouk S, Tomaso H. Nockler K, et al: Laboratory-based diagnosis of brucellosis-a review of the literature. Part n. Serological tests for brucellosis. Clin Lab 49: 577, 2003. 7. Glass WI: Brucellosis as an Occupational Disease in New Zealand. N Z Med ] 63: 301, 1964. 8. Foley BV, Clay MM, O'Sullivan D]: A study of a brucellosis epidemic. lr] Med Sci 3: 457, 1970. 9. Williams E: Brucellosis and the British public. Lancet 1: 1220, 1970. 10. Mantur BG, Akki AS, Mangalgi SS, et al: Childhood brucellosis-a microbiological, epidemiological and clinical study. ] Trop Pediatr 50: 153, 2004. 11. Ojukwu IC. Christy C: Rat-bite fever in children: case report and review. Scand] Infect Dis 34: 474. 2002.
667
12. Rat-bite fever-New Mexico. 1996. MMWR Morb Mortal Wkly Rep 47: 89, 1998. 13. Parker F]. Hudson NP: The etiology of Haverhill fever (erythema arthriticum epidemicum). Am] Pathol 2: 357. 1926. 14. Raffin B]. Freemark M: Streptobacillary !"dt-bite fever: a pediatric problem. Pediatrics 64: 214. 1979. 15. Place EH. SUllon LE: Erythema arthriticum epidemicum (Haverhill fever). Arch Intern Med 54: 659. 1934. 16. Obermeir 0: Vorkommen feinster eine eigenbewegung zeigender faden im blute von rekurrenskranken. Zentralbl Med Wiss 11: 145. 1873. 17. Barbour AG, Hayes SF: Biology of Borrelia species. Microbiol Rev 50: 381, 1986. 18. Paster B]. Dewhirst FE. Weisburg WG. et aI: Phylogenetic analysis of the spirochetes.] Bacteriol173: 6101. 1991. 19. Barbour AG, Fish D: The biological and social phenomenon of Lyme disease. Science 260: 1610. 1993. 20. Fukunaga M. Takahashi Y, Tsuruta Y, et al: Genetic and phenotypic analysis of Borrelia miyamotoi sp. nov., isolated from the ixodid tick Ixodes persulcatus, the vector for Lyme disease in Japan. Int] Syst Bacteriol 45: 804. 1995. 21. Barbour AG. Maupin GO, Teltow G]. et al: Identification of an uncultivable Borrelia species in the hard tick Amblyomma americanum: possible agent of a Lyme disease-like illness. ] Infect Dis 173: 403. 1996. 22. Cadavid D. Barbour AG: Neuroborreliosis during relapsing fever: review of the clinical manifestations. pathology, and treatment of infections in humans and experimental animals. Clin Infect Dis 26: 151, 1998. 23. McDermott MF, Aksentijevich I, Galon ], et al: Germline mutations in the extracellular domains of the 55 kDa TNF receptor. TNFRI. define a family of dominantly inherited autoinflammatory syndromes. Cell 97: 133, 1999. 24. Galon]. Aksentijevich I. McDermoll MF, et al: TNFRSFIA mutations and autoinflammatory syndromes. Curr Opin Immunol 12: 479, 2000. 25. Hull KM. Shoham N. Chae J]. et al: The expanding spectrum of systemic autoinflammatory disorders and their rheumatic manifestations. Curr Opin Rheumatol 15: 61. 2003. 26. Kile RM. Rusk HA: A case of cold unicaria with an unusual family history. ]AMA 114: 1067. 1940. 27. Siegal S: Benign paroxysmal peritonitis. Ann Intern Med 23: 1, 1945. 28. Heller H. Sohar E. Sherf L: Familial Mediterranean fever. Arch Intern Med 102: 50. 1958. 29. Muckle 1], Wellsm: Urticaria, deafness, and amyloidosis: a new heredofamilial syndrome. Q] Med 31: 235. 1962. 30. Williamson LM. Hull D. Mehta R, et al: Familial Hibernian fever. Q] Med 51: 469, 1982. 31. van der Meer ]W, Vossen ]M. Radl,J, et al: Hyperimmunogiobulinaemia D and periodic fever: a new syndrome. Lancet 1: 1087, 1984. 32. Hassink SG, Gold.mith DP: Neonatal onset multisystem inflammatory disease. Arthritis Rheum 26: 668, 1983. 33. Prieur AM, Griscelli C: Arthropathy with rash. chronic meningitis. eye lesions, and mental retardation. ] Pediatr 99: 79, 1981. 34. The International FMF Consortium: Ancient missense mutations in a new member of the RoRet gene family are likely to cause familial Mediterranean fever. Cell 90: 797, 1997. 35. The French FMF Consortium: A candidate gene for familial Mediterranean fever. Nat Genet 17: 25, 1997. 36. Samuels]. Aksentijevich I. Torosyan Y. et al: Familial Mediterranean fever at the millennium. Clinical spectrum. ancient mutations, and a survey of 100 American referrals to the National Institutes of Health. Medicine
668
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
46. Shinozaki K, Agematsu K, Yasui K, et al: Familial Mediterranean fever in 2 Japanese families. ] Rheumatol 29: 1324, 2002. 47. Konstantopoulos K, Kanta A, Deltas C, et al: Familial Mediterranean fever associated pyrin mutations in Greece. Ann Rheum Dis 62: 479, 2003. 48. La Regina M, Nucera G, Dlaco M, et al: Familial Mediterranean fever is no longer a rare disease in Italy. Eur] Hum Genet 11: 50, 2003, 49. Bertin], DiStefano PS: The pYRlN domain: a novel motif found in apoptosis and inflammation proteins. Cell Death Differ 7: 1273, 2000. 50, Fairbrother W], Gordon NC, Humke EW, et al: The pYRlN domain: a member of the death domain-fold superfamily. Protein Sci 10: 1911, 2001. 51. Kastner DL, O'Shea]J: A fever gene comes in from the cold. Nat Genet 29: 241, 2001. 52. Martinon F, Hofmann K, Tschopp ]: The pyrin domain: a possible member of the death domain-fold family implicated in apoptosis and inflammation. Curr BioI 11: R118, 2001. 53. Pawlowski K, pio F, Chu Z, et al: PAAD-a new protein domain associated with apoptosis, cancer and autoimmune diseases. Trends Biochem Sci 26: 85,2001. 54. Staub E, Dahl E, Rosenthal A: The DAPIN family: a novel domain links apoptotic and interferon response proteins. Trends Biochem Sci 26: 83, 2001. 55. Richards N, Schaner 1', Diaz A, et al: Interaction between pyrin and the apoptotic speck protein (ASC) modulates ASC-induced apoptosis. ] Bioi Chern 276: 39320, 2001. 56. Eliezer D: Folding pyrin into the family. Structure (Camb) 11: 1190, 2003. 57, Hiller S, Kohl A, Fiorito F, et al: NMR structure of the apoptosis- and inflammation-related NALpl pyrin domain. Structure (Camb) 11: 1199, 2003. 58. Liepinsh E, Barbals R, Dahl E, et al: The death domain fold of the ASC pYRlN domain, presenting a basis for pYRlN/pYRlN recognition. ] Mol BioI 332: 1155, 2003. 59, Liu T, Rojas A, Ye Y, et al: Homology modeling provides insights into the binding mode of the pAAD/DAPIN/pyrin domain, a fourth member of the CARD/DD/DED domain family. Protein Sci 12: 1872, 2003. 60, Masumoto], Taniguchi S, Ayukawa K, et al: ASC, a novel 22-kDa protein, aggregates during apoptosis of human promyelocytic leukemia HL-60 ceUs, ] Bioi Chem 274: 33835, 1999. 61. Chae ]], Komarow HD, Cheng], et al: Targeted disruption of pyrin, the FMF protein, causes heightened sensitivity to endotoxin and a defect in macrophage apoptosis, Mol Cell 11: 591, 2003. 62, Dowds TA, Masumoto], Chen FF, et al: Regulation of cryopyrinlPypafl signaling by pyrin, the familial Mediterranean fever gene product. Biochem Biophys Res Commun 302: 575, 2003. 63. Masumoto], Dowds TA, Schaner 1', et al: ASC is an activating adaptor for NFkappa Band caspase-8-dependent apoptosis. Biochem Biophys Res Commun 303: 69, 2003. 64. Martinon F, Bums K, Tschopp]: The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 10: 417, 2002. 65. Srinivasula SM, Poyet jL, Razmara M, et al: The pYRlN-CARD protein ASC is an activating adaptor for caspase-l.] BioI Chem 277: 21119, 2002. 66. Wang L, Manji GA, Grenier ]M, et al: PYPAF7, a novel PYRlN-containing Apafl-like protein that regulates activation of NF-kappa Band caspaseI-dependent cytokine processing. ] Bioi Chern 277: 29874, 2002. 67. Stehlik C, Lee SH, Dorfleutner A, et al: Apoptosis-associated speck-like protein containing a caspase recruitment domain is a regulator of procaspase-l activation. ] Immunol 171: 6154, 2003. 68. Sohar E, Gafni], Pras M, et al: Familial Mediterranean fever. A survey of 470 cases and review of the literature. Am] Med 43: 227, 1967. 69. Schwabe AD, Peters RS: Familial Mediterranean fever in Armenians. Analysis of 100 cases. Medicine (Baltimore) 53: 453, 1974. 70. Majeed HA, Rawashdeh M, el-Shanti H, et al: Familial Mediterranean fever in children: the expanded clinical profile. Q] Med 92: 309, 1999, 71. Barakat MH, Karnik AM, Majeed HW, et al: Familial Mediterranean fever (recurrent hereditary polyserositis) in Arabs-a study of 175 patients and review of the literature. Q] Med 60: 837, 1986. 72. Gedalia A, Adar A, Gorodischer R: Familial Mediterranean fever in children. ] Rheumatol Suppl 35: 1, 1992. 73. Brauman A, Gilboa Y: Recurrent pulmonary atelectasis as a manifestation of familial Mediterranean fever. Arch Intern Med 147: 378, 1987. 74, Brik R, Shinawi M, Kasinetz L, et al: The musculoskeletal manifestations of familial Mediterranean fever in children genetically diagnosed with the disease, Arthritis Rheum 44: 1416, 2001. 75, Majeed HA, Rawashdeh M: The clinical patterns of arthritis in children with familial Mediterranean fever. Q] Med 90: 37, 1997. 76. Ince E, Cakar N, Tekin M, et al: Arthritis in children with familial Mediterranean fever. Rheumatol Int 21: 213, 2002. 77. Sneh E, Pras M, Michaeli D, et al: Protracted arthritis in familial Mediterranean fever. Rheumatol Rehabil 16: 102, 1977, 78, Azizi E, Fisher BK: Cutaneous manifestations of familial Mediterranean fever. Arch Dermatol 112: 364, 1976, 79. Barzilai A, Langevitz 1', Goldberg I, et al: Erysipelas-like erythema of familial Mediterranean fever: clinicopathologic correlation. ] Am Acad Dermatol 42: 791,2000. 80. Eshel G, Vinograd I, Barr ], et al: Acute scrotal pain complicating familial Mediterranean fever in children. Br] Surg 81: 894, 1994.
81. Livneh A, Madgar I, Langevitz 1', et aI: Recurrent episodes of acute scrotum with ischemic testicular necrosis in a patient with familial Mediterranean fever.] Urol 151: 431, 1994. 82. Majeed HA, Ghandour K, Shahin HM: The acute scrotum in Arab children with familial Mediterranean fever. Pediatr Surg Int 16: 72, 2000. 83, Langevitz 1', Zemer 0, Livneh A, et al: Protracted febrile myalgia in patients with familial Mediterranean fever. J Rheumatol 21: 1708, 1994. 84, Majeed HA, A1-Qudah AK, Qubain H, et al: The clinical panerns of myalgia in children with familial Mediterranean fever. Semin Arthritis Rheum 30: 138, 2000. 85. Rawashdeh MO, Majeed HA: Familial Mediterranean fever in Arab children: the high prevalence and gene frequency. Eur] Pediatr 155: 540, 1996. 86, Kees S, Langevitz 1', Zemer 0, et ale Attacks of pericarditis as a manifestation of familial Mediterranean fever (FMF). Q] Med 90: 643, 1997. 87. Schwartz T, Langevitz 1', Zemer 0, et al: Beh,et's disease in familial Mediterranean fever: characterization of the association between the two diseases. Semin Arthritis Rheum 29: 286, 2000. 88. Touitou I, Magne X, Molinari N, et al: MEFV mutations in Beh,et's disease. Hum Mutat 16: 271, 2000. 89. Livneh A, Aksentijevich I, Langevitz 1', et ,II: A single mutated MEFV allele in Israeli patients suffering from familial Mediterranean fever and Beh~'et's disease (FMF-BD), Eur] Hum Genet 9: 191, 2001. . 90. Akpolat T, Yilmaz E, Akpolat I, et al: Amyloidosis in Beh,et's disease and familial Mediterranean fever, Rheumatoiogy (Oxf) 41: 592, 2002. 91. Ben-Chetrit E, Cohen R, Chajek-Shaul T: Familial Mediterranean fever and Beh,et's disease---are they associated? ] Rheumatol 29: 530, 2002. 92. Glikson M, Galun E, Schlesinger M, et al: Polyarteritis nodosa and familial Mediterranean fever: a report of 2 cases and review of the literature. ] Rheumatol 16: 536, 1989. 93. Schlesinger M, Oren S, Fano M, et al: Perirenal and renal subcapsular haematoma as presenting symptoms of polyarteritis nodosa. Postgrdd Med ] 65: 681, 1989. 94. Ozdogan H, Arisoy N, Kasapcapur 0, et al: Vasculitis in familial Mediterranean fever. J Rheumatol 24: 323, 1997. 95. Tinaztepe K, Gucer S, Bakkaloglu A, et al: Familial Mediterranean fever and polyarteritis nodosa: experience of five paediatric cases. A causal relationship or coincidence? Eur J Pediatr 156: 505, 1997. 96. Ozen S: New interest in an old disease: familial Mediterranean fever. Clin Exp Rheumatol 17: 745, 1999. 97. Tekin M, Yalcinkaya F, Tumer N, et al: Familial Mediterranean fever-renal involvement by diseases other than amyloid, Nephrol Dial Transplant 14: 475, 1999, 98. Ozen S, Ben-Chetrit E, Bakkaloglu A, et al: Polyaneritis nodosa in patient. with familial Mediterranean fever (FMF): a concomitant disease or a feature of FMF? Semin Arthritis Rheum 30: 281, 2001. 99. Said R, Nasrallah N, Hamzah Y, et ale IgA nephropathy in patients with familial Mediterranean fever. Am] Nephrol8: 417,1988. 100. Said R, Hamzeh Y, Tarawneh M, et al: Rapid progressive glomerulonephritis in patients with familial Mediterranean fever. AmJ Kidney Dis 14: 412, 1989. 101. Said R, Hamzeh Y, Said S, et al: Spectrum of renal involvement in familial Mediterranean fever. Kidney Int 41: 414, 1992. 102. Akpolat T, Akpolat I, Karagoz F, et ai: Familial Mediterranean fever and glomerulonephritis and review of the literature. Rheumatol Int 24: 43, 2004, 103. Cattan D, Notarnicola C, Molinari N, et al: Inflammatory bowel disease in non-Ashkenazi Jews with familial Mediterranean fever. Lancet 355: 378, 2000, 104. Fidder HH, Chowers Y, Lldar M, et al: Crohn disease in patients with familial Mediterranean fever. Medicine (Baltimore) 81: 411, 2002. 105. Tunca M, Kirkali G, Soyturk M, et al: Acute phase response and evolution of familiai Mediterranean fever. Lancet 353: 1415, 1999. 106, Korkmaz C, Ozdogan H, Kasapcopur 0, et al: Acute phase response in familial Mediterranean fever, Ann Rheum Dis 61: 79, 2002. 107. Duzova A, Bakkaloglu A, Besbas N, et al: Role of A-SAA in monitoring subclinical inflammation and in colchicine dosage in familial Mediterranean fever. Clin Exp Rheumatol 21: 509, 2003. 108. Cazeneuve C, Ajrapetyan H, Papin S, et al: Identification of MEFV-independent modifying genetic factors for familial Mediterranean fever. Am ] Hum Genet 67: 1136, 2000, 109. Akar N, Hasipek M, Akar E, et al: Serum amylOid Al and tumor necrosis factor-alpha alleles in Turkish familial Mediterranean fever patient. with and without amyloidosis. Amyloid 10: 12, 2003. 110. Gershoni-Baruch R, Brik R, Zacks N, et al: The contribution of genotypes at the MEFV and SAAI loci to amyloidosis and disease severity in patient. with familial Mediterranean fever. Arthritis Rheum 48: 1149, 2003. 111. BakkaJoglu A, Duzova A, Ozen S, et al: Influence of Serum Amyloid A (SAA!) and SAA2 gene polymorphisms on renal amyloidosis, and on SAAlCreactive protein values in patients with familial Mediterranean fever in the Turkish population.] Rheumatol 31: 1139, 2004, 112. MedIej-Hashim M, Deiague V, Chouery E, et ale Amyloidosis in familial Mediterranean fever patients: correlation with MEFV genotype and SAA1 and MICA polymorphisms effect., BMC Med Genet 5: 4, 2004. 113. Cazeneuve C, Sarkisian T, Pecheux C, et al: MEFV-gene analysis in Armenian patients with familial Mediterranean fever: diagnostic value and unfavorable renal prognosis of the M694V homozygous genotype-genetic and therapeutic implications. Am] Hum Genet 65: 88,1999.
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
114. Kone Paut 1, Dubuc M, Sportouch], et al: Phenotype-genotype correlation in 91 patients with familial Mediterranean fever reveals a high frequency of clltaneomucous features. Rheumatology (Oxf) 39: 1275, 2000. 115. Yalcinkaya F, Cakar N, Misirlioglu M, et al: Genotype-phenotype correlation in a large group of Turkish patients with familial Mediterranean fever: evidence for mutation-independent amyloidosis. Rheumatology (Oxf) 39: 67, 2000.
116. Ulum A, Sohar E: The diagnosis of amyloidosis. Ancillary procedures. Lancet 1: 721, 1962. 117. Livneh A, Langevitz P, Zemer 0, et al: Criteria for the diagnosis of familial Mediterranean fever. Arthritis Rheum 40: 1879, 1997. 118. hkarsu AN, Saatci D, Ozen S, et al: Genetic linkage study of familial Mediterranean fever (FMF) to 16p13.3 and evidence for genetic heterogeneity in the Turkish population. ] Med Genet 34: 573, 1997. 119. I'>omingo C, Touitou I, Bayou A, et al: Familial Mediterranean fever in the 'Chuetas' of Mallorca: a question of Jewish origin or genetic heterogeneity. Eur] Hum Genet 8: 242, 2000. 120. Cazeneuve C, Hovannesyan Z, Genevieve 0, et al: Familial Mediterranean fever among patients from Karabakh and the diagnostic value of MEFV gene analysis in all c1assimlly affected populations. Arthritis Rheum 48: 2324, 2003. 121. Uooth DR, Gillmore ]0, Lachmann H], et al: The genetic basis of autosomal dominant familial Mediterranean fever. Q] Med 93: 217, 2000. 122. Akar N, Misiroglu M, Yalcinkaya F, et al: MEFV mutations in Turkish patients suffering from familial Mediterranean fever. Hum Mutat 15: 118, 2000. 123. Mansour I, Delague V, Cazeneuve C, et al: Familial Mediterranean fever in Lebanon: mutation spectrum, evidence for cases in Maronites, Greek ortho
147. 148.
149. 150. 151. 152. 153.
154. 155.
156. 157. 158. 159. 160. 161.
162. 163.
164. 165. 166. 167. 168. 169. 170. 171. 172. 173.
669
neonatal-onset multisystem inflammatory disease (NOMID): a new member of the expanding family of pyrin-associated autoinflammatory diseases. Arthritis Rheum 46: 3340, 2002. Dode C, Le Du N, Cuisset L, et al: New mutatIons of CIASI that are responsible for Muckle-Wells syndrome and familial cold urticaria: a novel mutation underlies both syndromes. Am] Hum Genet 70: 1498, 2002. Feldmann], Prieur AM, Quartier P, et al: Chronic infantile neurological cutaneous and articular syndrome is caused by mutations in ClASl, a gene highly expressed in polymorphonuclear cells and chondrocytes. Am] Hum Genet 71: 198, 2002. Granel B, Philip N, Serratrice], et al: ClASl mutation in a patient with overlap between Muckle-Wells and chronic infantile neurological cutaneous and articular syndromes. Dermatology 206: 257, 2003. Rosen-Wolff A, Quietzsch], Schroder H, et al: Two German CINCA (NOMID) patients with different clinical severity and response to anti-inflammatory treatment. Eur] Haematol 71: 215, 2003. Neven B, Callebaut I, Prieur AM, et al: Molecular basis of the spectral expression of ClASl mutations associated with phagocytic cell-mediated autoinflammatory disorders CINCAINOMID, MWS, and FCU. Blood 103: 2809, 2004. McDermott EM, Smillie OM, Powell R]: Clinical spectrum of familial Hibernian fever: a 14-year follow-up study of the index case and extended family. Mayo Clin Proc 72: 806, 1997. Dode C, Papo T, Fieschi C, et al: A novel missense mutation (C30S) in the gene encoding tumor necrosis factor receptor 1 linked to autosomaldominant recurrent fever with localized myositis in a French family. Arthritis Rheum 43: 1535, 2000. Aganna E, Aksentijevich 1, Hilman GA, et al: Tumor necrosis factor receptorassociated periodic syndrome (TRAPS) in a Dutch family: evidence for a TNFRSFIA mutation with reduced penetrance. Eur] Hum Genet 9: 63, 2001. Aksentijevich 1, Galon], Soares M, et al: The tumor-necrosis-factor receptor-associated periodic syndrome: new mutations in TNFRSFIA, ancestral origins, genotype-phenotype studies, and evidence for further genetic heterogeneity of periodic fevers. Am] Hum Genet 69: 301, 2001. ]adoul M, Dode C, Cosyns ]P, et al: Autosomal-dominant periodic fever with AA amyloidosis: novel mutation in tumor necrosis factor receptor 1 gene [rapid communication]. Kidney Int 59: 1677, 2001. Simon A, Dode C, van der Meer lW, et al: Familial periodic fever and amyloidosis due to a new mutation in the TNFRSFIA gene. Am] Med 110: 313, 2001. Simon A, van Deuren M, Tighe P], et al: Genetic analysis as a valuable key to diagnosis and treatment of periodic fever. Arch Intern Med 161: 2491, 2001. Aganna E, Zeharia A, Hitman GA, et a1: Art Israeli Arab patient with a de novo TNFRSFIA mutation causing tumor necrosis factor receptor-associated periodic syndrome. Arthritis Rheum 46: 245, 2002. Dode C, Andre M, Bienvenu T, et al: The enlarging clinical, genetic, and population spectrum of tumor necrosis factor receptor-associated periodic syndrome. Arthritis Rheum 46: 2181, 2002. Nevala H, Karenko L, Stjemberg S, et al: A novel mutation in the third extracellular domain of the tumor necrosis factor receptor 1 in a Finnish family with autosomal-dominant recurrent fever. Arthritis Rheum 46: 1061, 2002. Aganna E, Hammond L, Hawkins PN, et al: Heterogeneity among patients with tumor necrosis factor receptor-associated periodic syndrome phenotypes. Arthritis Rheum 48: 2632, 2003. Kriegel MA, Huffmeier D, Scherb E, et al: Tumor necrosis factor receptor-associated periodic syndrome characterized by a mutation affecting the cleavage site of the receptor: implications for pathogenesis. Arthritis Rheum 48: 2386, 2003. Kusuhara K, Nomura A, Nakao F, et al: Tumour necrosis factor receptorassociated periodic syndrome with a novel mutation in the TNFRSFIA gene in a Japanese family. Eur] Pediatr 163: 30, 2004. Reddy P, Slack ]L, Davis R, et al: Functional analysis of the domain structure of tumor necrosis factor-alpha converting enzyme. ] Bioi Chern 275: 14608, 2000. Cui X, Hawari F, Alsaaty S, et ai: Identification of ARTS-l as a novel TNFRlbinding protein that promotes TNFRI ectodomain shedding. ] Clin Invest 110: 515, 2002. Engelmann H, Aderka D, Rubinstein M, et al: A tumor necrosis factor-binding protein pUrified to homogeneity from human urine protects cells from tumor necrosis factor toxicity.] Bioi Chern 264: 11974, 1989. Toro JR, Aksentijevich 1, Hull K, et al: Tumor necrosis factor receptor-associated periodic syndrome: a novel syndrome with cutaneous manifestations. Arch Dermatol 136: 1487, 2000. Hull KM, Drewe E, Aksentijevich 1, et al: The TNF receptor-associated periodic syndrome (TRAPS): emerging concepts of an autoinflamrnatory disorder. Medicine (Baltimore) 81: 349, 2002. Hull KM, Wong K, Wood GM, et al: Monocytic fasciitis: a newly recognized clinical feature of tumor necrosis factor receptor dysfunction. Arthritis Rheum 46: 2189, 2002. Dode C, Hazenberg BP, Pecheux C, et al: Mutational spectrum in the MEFV and TNFRSFIA genes in patients suffering from AA amyloidosis and recurrent inflammatory attacks. Nephrol Dial Transplant 17: 1212, 2002. Weyhreter H, Schwartz M, Kristensen TO, et al: A new mutation causing autosomal dominant periodic fever syndrome in a Danish family. ] Pediatr 142: 191, 2003. Nigrovic PA, Sundel RP: Treatment of TRAPS with etanercept: use in pediatrics. Clin Exp Rheumatol 19: 484, 2001.
670
C HAP T E R
34
PERIODIC FEVER SYNDROMES IN CHILDREN
174. Drenth ]P, Mariman EC, Van der Velde-Visser SD, et al: Location of the gene causing hyperimmunoglobulinemia D and periodic fever syndrome differs from that for familial Mediterranean fever. International Hyper-IgD Study Group. Hum Genet 94: 616, 1994. 175. Drenth]P, Cuisset L, Grateau G, et al: Mutations in the gene encoding mevalonate kinase cause hyper-IgD and periodic fever syndrome. International Hyper-IgD Study Group. Nat Genet 22: 178, 1999. 176. Houten SM, Kuis W, Duran M, et al: Mutations in MVK, encoding mevalonate kinase, cause hyperimmunoglobulinaemia D and periodic fever syndrome. Nat Genet 22: 175, 1999. 177. Valle D: You give me fever. Nat Genet 22: 121, 1999. 178. Topaloglu R, Saatci U: Hyperimmunoglobulinaemia D and periodic fever mimicking familial Mediterranean fever in the Mediterranean. Postgrad Med ] 67: 490, 1991. 179. Miyagawa S, Kitamura W, Morita K, et al: Association of hyperimmunoglobulinaemia D syndrome with erythema elevatum diutinum. Br] Dermatol 128: 572, 1993. 180. Drenth ]P, Haagsma C], van der Meer ]W: Hyperimmunoglobulinemia D and periodic fever syndrome, The clinical spectrum in a series of SO patients. International Hyper-IgD Study Group, Medicine (Baltimore) 73: 133, 1994. 181. Grose C, Schnetzer ]R, Ferrante A, et al: Children with hyperimmunoglobulinemia D and periodic fever syndrome, Pediatr Infect Dis] IS: 72, 1996, 182, Drenth ]P, van der Meer ]W: Hereditary periodic fever. N Engl] Med 345: 1748, 2001. 183. Simon A, Mariman EC, van der Meer]W, et al: A founder effect in the hyperimmunoglobulinemia D and periodic fever syndrome. Am] Med 114: 148, 2003, 184. Goldstein]L, Brown MS: Regulation of the mevalonate pathway. Nature 343: 425, 1990. 185. Brown MS, Goldstein ]L: The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell 89: 331, 1997. 186, Hoffmann GF, Charpentier C, Mayatepek E, et al: Clinical and biochemical phenotype in 11 patients with mevalonic aciduria. Pediatrics 91: 915, 1993. 187. Frenkel], Rijkers GT, Mandey SH, et al: Lack of isoprenoid products raises ex vivo interleukin-lbeta secretion in hyperimmunoglobulinemia D and periodic fever syndrome. Arthritis Rheum 46: 2794, 2002, 188. Takada K, Aksentijevich I, Mahadevan V, et al: Favorable preliminary experience with etanercept in two patients with the hyperimmunoglobulinemia D and periodic fever syndrome. Arthritis Rheum 48: 2645, 2003. 189. Houten SM, Frenkel], Rijkers GT, et al: Temperature dependence of mutant mevalonate kinase activity as a pathogenic factor in hyper-IgD and periodic fever syndrome. Hum Mol Genet 11: 3115, 2002. 190, Haraldsson A, Weemaes CM, De Boer AW, et al: Immunological studies in the hyper-immunoglobulin D syndrome,] Clin Immunol 12: 424, 1992, 191. Drenth]p, Prieur AM: Occurrence of arthritis in hyperimmunoglobulinaemia D. Ann Rheum Dis 52: 765, 1993. 192. Loeliger AE, Kruize AA, Bljilsma]W, et al: Arthritis in hyperimmunoglobulinaemia D, Ann Rheum Dis 52: 81, 1993. 193, Drenth ]P, Boom BW, Toonstra], et al: Cutaneous manifestations and histologic findings in the hyperimmunoglobulinemia D syndrome. International Hyper IgD Study Group. Arch Dermatol130: 59, 1994. 194, Frenkel], Houten SM, Waterham HR, et al: Mevalonate kinase deficiency and Dutch type periodic fever. Clin Exp Rheumatol 18: 525, 2000. 195. Saulsbury !'T: Hyperimmunoglobulinemia D and periodic fever syndrome (HIDS) in a child with normal serum IgD, but increased serum IgA concentration.] Pediatr 143: 127,2003. 196. Frenkel], Houten SM, Waterham HR, et al: Clinical and molecular variability in childhood periodic fever with hyperimmunoglobulinaemia D. Rheumatology (Oxf) 40: 579, 2001. 197. Kelley RI, Takada I: Hereditary periodic fever. N Engl] Med 346: 1415,2002. 198. Hiemstra I, Vossen ]M, van der Meer]W, et al: Clinical and immunological studies in patients with an increased serum IgD level.] Clin Immunol 9: 393, 1989. 199. Boom BW, Daha MR, Vermeer B], et al: IgD immune complex vasculitis in a patient with hyperimmunoglobulinemia D and periodic fever. Arch Dermatol 126: 1621, 1990. 200, Medlej-Hashim M, Petit I, Adib S, et al: Familial Mediterranean fever: association of elevated IgD plasma levels with specific MEFV mutations. Eur) Hum Genet 9: 849, 2001. 201. Simon A, Cuisset L, Vincent MF, et al: Molecular analysis of the mevalonate kinase gene in a cohort of patients with the hyper-IgD and periodic fever syndrome: its application as a diagnostic tool. Ann Intern Med 135: 338, 2001. 202. Simon A, Drewe E, van der Meer ]W, et aI: Simvastatin treatment for inflammatory attacks of the hyperimmunoglobulinemia D and periodic fever syndrome, Clin Pharmacol Ther 75: 476, 2004. 203. Obici L, Manno C, Muda AD, et al: First report of systemic reactive (AA) amyloidosis in a patient with the hyperimmunoglobulinemia D with periodic fever syndrome. Arthritis Rheum 50: 2966, 2004. 204. Morley AA, Carew ]P, Baikie AG: Familial cyclical neutropenia. Br ] Haematol 13: 719, 1967.
205. Dale DC, Bolyard AA, Hammond WP: Cyclic neutropenia: natural history and effects of long-term treatment with recombinant human granulocyte colony-stimulating factor. Cancer Invest 11: 219, 1993. 206, Palmer SE, Stephens K, Dale DC: Genetics, phenotype, and natural history of autosomal dominant cyclic hematopoiesis. Am) Med Genet 66: 413, 1996. 207. Dale DC, Bolyard AA, Aprikyan A: Cyclic neutropenia. Semin Hematol 39: 89, 2002. 208. Horwitz M, Benson KF, Person RE, et al: Mutations in ELA2, encoding neutrophil elastase, define a 21-day biological clock in cyclic haematopoiesis. Nat Genet 23: 433, 1999. 209. Dale DC, Person RE, Bolyard AA, et al: Mutations in the gene encoding neutrophil elastase in congenital and cyclic neutropenia. Blood 96: 2317,2000, 210. Benson KF, Li FQ, Person RE, et al: Mutations associated with neutropenia in dogs and humans disrupt intracellular transport of neutrophil elastase. Nat Genet 35: 90, 2003. 211. Person RE, Li FQ, Duan Z, et al: Mutations in proto-oncogene GFil cause human neutropenia and target ELA2, Nat Genet 34: 308, 2003. 212, Horwitz M, Benson KF, Duan Z, et al: Hereditary neutropenia: dogs explain human neutrophil elastase mutations. Trends Mol Med 10: 163, 2004. 213, Loughran TP ]r, Clark EA, Price TH, et al: Adult-onset cyclic neutropenia is associated with increased large granular lymphocytes. Blood 68: 1082, 1986. 214, Loughran TP ]r, Hammond WPT: Adult-onset cyclic neutropenia is a benign neoplasm associated with clonal proliferation of large granular lymphocytes. ] Exp Med 164: 2089, 1986. 215, Berliner N, Horwitz M, Loughran TP )r: Congenital and acquired neutropenia. Hematology (Am Soc Hematol Educ Program) 2004: 63. 216, Souid AK: Congenital cyclic neutropenia, Clin Pediatr (Phila) 34: 151, 1995, 217. Felitti V]: Primary invasion by Clostridium spbenoldes in a patient with periodic neutropenia. Calif Med 113: 76, 1970. 218, Geelhoed GW, Kane MA, Dale DC, et al: Colon ulceration and perforation in cyclic neutropenia. ] Pediatr Surg 8: 379, 1973, 219, Hopkins DG, Kushner ]P: Clostridial species in the pathogenesis of necrotizing enterocolitis in patients with neutropenia. Am] Hematol14: 289,1983, 220. Bar-]oseph G, Halberthal M, Sweed Y, et al: Clostridium sept/cum infection in children with cyclic neutropenia. ] Pediatr 131: 317, 1997. 221. Lange RO: Cyclic hematopoiesis: human cyclic neutropenia, Exp Hematol 11: 435, 1983, 222. Parmley RT, Presbury G], Wang WC, et al: Cyclic ultrastructural abnormalities in human cyclic neutropenia, Am] Pathol 116: 279, 1984, 223. Aprikyan AA, Liles WC, Rodger E, et al: Impaired survival of bone marrow hematopoietic progenitor cells in cyclic neutropenia, Blood 97: 147, 2001. 224. Hammond WPT, Price TH, Souza LM, et al: Treatment of cyclic neutropenia with granulocyte colony-stimulating factor. N Engl] Med 320: 1306, 1989. 225. Bonilla MA, Dale D, Zeidler C, et al: Long-term safety of treatment with recombinant human granulocyte colony-stimulating factor (r-metHliG-CSFl in patients with severe congenital neutropenias, Br] Haematol 88: 723, 1994. 226. Heussner P, Haase D, Kanz L, et al: G-CSF in the long-term treatment of cyclic neutropenia and chronic idiopathic neutropenia in adult patients, Int ] Hematol 62: 225, 1995, 227, Kurzrock R, Talpaz M, Gutterman ]U: Treatment of cyclic neutropenia with very low doses of GM-CSF. Am) Med 91: 317, 1991. 228. Pernu HE, Pajari UH, Lanning M: The importance of regular dental treatment in patients with cyclic neutropenia. Follow-up of 2 cases, ] Periodontol 67: 454, 1996, 229, da Fonseca MA, Fontes F: Early tooth loss due to cyclic neutropenia: longterm follow-up of one patient. Spec Care Dentist 20: 187, 2000, 230, Nakai Y, Ishihara C, Ogata S, et a1: Or.tl manifestations of cyclic neutropenia in a Japanese child: case report with a 5-year follow-up, Pediatr Dent 25: 383, 2003, 231. Marshall GS, Edwards KM, Butler ], et al: Syndrome of periodic fever, pharyngitis, and aphthous stomatitis. ] Pediatr 110: 43, 1987, 232. Atas B, Caksen H, Arslan S, et al: PFAPA syndrome mimicking familial Mediterranean fever: report of a Turkish child. ] Emerg Med 25: 383, 2003. 233. Berlucchi M, Meini A, Plebani A, et al: Update on tre'.ttment of Marshall's syndrome (PFAPA syndrome): report of five cases with review of the literature. Ann Otol Rhinol Laryngol 112: 365, 2003, 234. Thomas KT, Feder HM ]r, Lawton AR, et al: Periodic fever syndrome in children,) Pediatr 135: 15, 1999, 235, Ovetchkine P, Bry ML, Reinert P: [Marshall syndrome: results of a retrospective national survey). Arch Pediatr 7 (Suppl 3): 578s, 2000. 236. Padeh S, Brezniak N, Zemer D, et al: Periodic fever, aphthous stomatitis, pharyngitis, and adenopathy syndrome: clinical characteristics and outcome, ] Pediatr 135: 98, 1999, 237. Feder HM ]r, BialeckI CA: Periodic fever associated with aphthous stomatitiS, pharyngitis and cervical adenitis, Pediatr Infect Dis] 8: 186, 1989. 238. Feder HM )r: Cimetidine treatment for periodic fever associated with aphthous stomatitis, pharyngitis and cervical adenitis. Pediatr Infect Dis) 11: 318, 1992. 239. pillet P, Ansoborlo S, Carrere A, et al: (P)FAPA syndrome: value of cimetidine. Arch Pediatr 7: 54, 2000. 240. Abramson )S, Givner LB, Thompson )N: Possible role of tonsillectomy and adenoidectomy in children with recurrent fever and tonsillopharyngitis. Pediatr Infect Dis] 8: 119, 1989,
35
C HAP T E R
CRYOPYRIN-ASSOCIATED PERIODIC SYNDROMES Benedlde Neven, Anne-Marie PrIeur and Ross E. Petty
Among the episodic or periodic fever syndromes are three clinically identifiable disorders that are caused by dontinantly inherited abnormalities in cryopyrin resulting from missense mutations in the cold autoinflammatory syndrome 1 gene (CIASJ): familial cold autoinflammatory syndrome (FCAS), Muckle-Wells syndrome (MWS), and chronic infantile neurologic cutaneous and articular syndrome (CINCA), also called neonatal-onset multisystem inflammatory disease (NOMID) (see Chapter 34). Cryopyrin-associated syndromes resemble the episodic fever syndromes in many ways, although the periodicity of the fever is much less marked in these disorders. Phenotypically, they represent a spectrum of diseases.
FCAS is the mildest condition, and CINCA is the most severe (Fig. 35-1), They are all characterized by an early onset of recurrent episodes of fever with marked systemic inflammation, a characteristic nonpruritic urticarial rash with perivascular polymorphonuclear cell cutaneous infiltrates, and a broad spectrum of joint manifestations, ranging from arthralgias in FCAS to recurrent arthritis in MWS or the mild form of CINCA and the permanent arthropathies in severe CINCA. Sensorineural hearing loss can develop with increasing age in MWS and CINCA. Neurologic involvement occurs in CINCA and is caused by a chronic aseptic meningitis with polymorphonuclear cells infiltrate in the cerebrospinal fluid (CSF).
• ..... 35-1 ainical characteristics of familial cold autoinflammatory syndrome (FCAS), Muckle-Wells syndrome (MWS), and chronic: infantile neurologic cutaneous and articular «(INCA) syndrome, also called neonatal-onset multisystem inflammatory disease (NOMID).These three autoinflammatory diseases represent a spectrum of severity. FCAS is the mildest condition, and (INCAlNOMID is the most severe.
671
672
C HAP T E R
35
CRYOPYRIN-AssOCIATED PERIODIC SYNDROMES
GENETICS AND ETIOLOGY FCAS, MWS, and CINCA are associated with missense mutations of CIASl gene. I-5 (Fig. 35-2). More than 30 mutations have been identified, and they are to some extent associated with different phenotypes in different families l .4.6 This gene encodes for the cryopyrin protein, also called NALP3 or PYPAF1,1" which is a member of the family of death domain proteins. Its expression is limited to immune cells and chondroeytes. I.BCryopyrin contains an N-terminal PYRIN domain, a central NACHT domain with a nucleotide binding site, and a C-terminal leucinerich repeat,I,B The term NALP is derived from NACHT, LRR, and PYD. The function of this protein is unclear, but it may play a central role in control of inflammation, cytokine processing, and apoptosis. 9 Cryopyrin interacts with an apoptosis-associated speck-like protein with a caspase-recruitment domain (ASC) by means of a homotypic interaction between its pyrin domain and the pyrin domain of ASC.' This interaction mediates procaspase-1 activation, and caspase-l activates pro-interleukin (IL)-l~ in its proinflammatory active form, IL-1~. The interaction of cryopyrin with ASC also mediates the activation of nuclear factor-KB (NF-lCB).' All mutations identified in FCAS, MWS, and CINCA are localized in exon 3, irrespective of disease severity.I-5,B.9
This exon encodes the NACHT domain and its flanking protein regions. Preliminary data suggest a genotype/phenotype correlation. s Genetic heterogeneity is suggested in that only 60% of patients with a clinical presentation of FCAS, MWS, or CINCA have a mutation of the CIASl gene. Janssen and colleagues lO demonstrated enhanced IL-l~ and IL-18 production in CINCA that is probably responsible for the clinical manifestations. 11
CLINICAL MANIFESTATIONS Familial Cold Autolnflammatory Syndrome FCAS (Mendelian Inheritance in Man [MIMl number 120100) was first described in 1940. 12 This syndrome is characterized by recurrent short and self-limited episodes of fever, rash, and arthralgia precipitated by cold exposure. Conjunctivitis is also frequently observed. Other commonly reported symptoms are muscle pain, profuse sweating, drowsiness, headache, extreme thirst, and nausea. Early onset of the disease, at birth or within the first 6 months of life, is characteristic. The duration of the attacks is usually less than 24 hours. Most of the patients describe a correlation between the severity of the crisis and the intensity of cold exposure. Attacks are more
• Figure 35-2 Mutations In cryopyrin-associated diseases. CINCA, chronic infantile neurologic cutaneous and articular syndrome; FCAS, familial cold autolnflammatory syndrome; MWS, Muckle-Wells Syndrome.
C HAP T E R
35
CRYOPYRIN-AsSOCIATED PERIODIC SYNDROMES
673
• Figure 35-4 The typical patella overgrowth in chronic. infantile neurologic cutaneous and articular (CINCA) syndrome results in a gross deformity of the joint.
• ~ 35-3 The rash of chronic infantile neurologic cutaneous and articular (ONCA) syndrome. Appearance of the rash varies in character and severity. (See color insert.)
frequent in winter, during damp and windy days, or after exposure to air conditioning. Leukocytosis and increased acute phase reactants accompany episodes of inflammation. Deafness and amyloidosis usually are not observed, even if several members of the patient's family reported late-onset renal amyloidosis.I,13,14
Muckle-Wells Syndrome MWS (MIM 191900) was described in 1962 by Muckle and Wells in one family with urticaria (see Fig. 34-2), deafness, and renal amyloidosis.I 5 The frequency of this disease is difficult to estimate; large series have not been published. This autosomal dominantly inherited disease is characterized by
recurrent episodes of fever and rash associated with articular, auditory, and ophthalmologic manifestations, and frequently, AA amyloidosis. Precipitating factors are not always identified; some patients report cold, tiredness, and hunger. The course of the disease varies from typical recurrent attacks of inflammation to more persistent symptoms. Fever is not always present. Joint manifestations can be mild with brief episodes of arthralgias, but recurrent episodes of synovitis affecting predominantly large joints can be observed,16 Conjunctivitis is common, and episderitis and iridocyditis have been reported. 17 Sensorineural deafness is frequently observed. Onset can occur any time from childhood to adulthood. AA amyloidosis, due to chronic inflammation, is the primary complication. 1s-22 Headache and papilledema occurred in some cases, and facial palsy has been reported in one case. Clinical manifestations of mild forms of MWS resemble those of FCAS, and more severe phenotypes overlap with CINCA,8,23
• FIgure 35-5 Radiographs of the knee in a child with chronic infantile neurologic cutaneous and articular (CINCA) syndrome at 6 months (A), 2 years (8), and 3.5 years old (0. The most characteristic changes occur in the metaphysic and epiphysis, with overgrowth and irregularity of ossification.
674
C HAP T E R
35
CRYOPYRIN-AssOCIATED PERIODIC SYNDROMES
nia, frontal bossing, and saddle-back nose are frequently observed. CSF examination demonstrates variable hypercellularity with increased polymorphonuclear leukocytes, elevated protein levels, and an increased opening pressure. Computed tomography or magnetic resonance imaging can be normal or document mild ventricular dilatation and enlarged subdural fluid spaces, suggesting mild cerebral atrophy. Some patients develop progressive calcifications of the falx cerebri and dura mater. Ocular disease consists of an anterior uveitis in one half of the patients and posterior uveitis in another 20%. Optic atrophy may develop. Ocular manifestations can progress to blindness, and one patient in four has a significant ocular disability.27.28 Perceptive deafness is frequently observed in older patients
TREATMENT • Figure 35-6 Papilledema in a case of chronic infantile neurologic cutaneous and articular syndrome (CINCA).
Chronic Infantile Neurologic Cutaneous and Articular Syndrome CINCA is the most severe phenotype in the cryopyrin spectrum of diseases, although the clinical manifestations vary in severity. Permanent joint and central nervous system involvement are present in CINCA but absent in FCAS and MWS. 24 Prematurity and dysmaturity are characteristic of one third of patients. Umbilical cord anomalies were observed in a few children. 24-26 Urticaria is usually present at birth or during the first months of life. The rash is nonpruritic and papular (Fig. 35-3), It varies in intensity from patient to patient, with time, and with disease activity. Fever is intermittent, and it can be absent or very mild in some cases. Bone and joint involvement vary in severity. In approximately two thirds of the patients, joint manifestations are limited to arthralgia and transient swelling without effusion during flare-ups. In one third of patients, joint abnormalities are severe and usually begin within the first year of life. The metaphyses and epiphyses of the long bones are affected, and bony overgrowth can result in gross deformity of the joints, articular and bone pain, and loss of range of motion (Fig. 35-4). The knees, ankles, wrists, and elbows are most commonly affected in a symmetric pattern, but small joints can also be involved. Radiologic manifestations, when present, are distinctive (Fig. 35-5). The most characteristic changes occur in the metaphysis and epiphysis with overgrowth and irregularity of ossification. There may be bowing, shortening, and widening of the long bones with a periosteal reaction. A chronic aseptic meningitis occurs in almost all patients. Chronic headaches, sometimes with vomiting and papilledema, are common (Fig. 35-6). Spastic diplegia and epilepsy may develop. Progressive cognitive impairment occurs in severely affected patients. Closure of the anterior fontanelle may be delayed, and macrocra-
There is no established treatment for the pyrin-associated autoinflammatory syndromes, but newer biologic agents are promising. Nonsteroidal anti-inflammatory drugs and prednisone offer temporary clinical relief. Colchicine is ineffective. Hawkins and colleagues29 reported the effectiveness of IL-l blockade in three patients with MSW who had a CIASl mutation. Dramatic and rapid improvement in the erythrocyte sedimentation rate, C-reactive protein level, and serum amyloid A levels were observed, and fever, rash, conjunctivitis, and arthralgia cleared. However, there were no changes in sensorineural hearing loss or ophthalmic abnormalities. No adverse effects were reported in a 3-month follow-up period. Frenkel and colleagues30 reported the effective use of anakinra in three children with CINCA syndrome who lacked a mutation in the CIASl gene. In these children, fever, rash, and arthritis all responded to therapy. This report suggests that blockade of IL-l is effective in patients with normal or abnormal CIAS 1 genes, indicating the possibility that its effect is mediated through another mechanism. Antitumor necrosis factor-a therapy was useful in one child with CINCA syndrome. 31 The clinical response included improvement of the arthropathy over a 6-month period.
REFERENCES 1.
2.
3. 4.
5.
6.
Hoffman HM, Mueller ]1, Broider DH, et al: Mutation of a new gene encodIng a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet 29: 301-305, 2ool. Feldmann], Prieur A-M, Quartier P, et al: Chronic infantile neurological cutaneous and articular syndrome is caused by mutations in CIASl, a gene highly expressed In polymorphonuclear cells and chondrocytes. ] Hum Genet 71: 198-203, 2002. Dode C, Le Du N, Cuisset L, et al: New mutations of CIASI that are responsible for Muckle-Wells syndrome and familial cold urticaria: a novel mutation underlies both syndromes. Am] Hum Genet 70: 1498-1506.2002. Aganna E, Martinon F, Hawkins PN, et al: Association of mutations in the NALP3/CIASl/PYPAFl gene with a broad phenotype including recurrent fever, cold sensitivity, sensorineural deafness, and AA amyloidosis. Arthritis Rheum 46: 2445-2452, 2002. Aksentijevlch I, Nowak M, Mallah M, et al: De novo CIASI mutations, cytokine activation, and evidence for genetic heterogeneity in patients with neonatal-onset multisystem inflammatory disease (NOMID): a new member of the expanding family of pyrin-associated autoinflammatory diseases. Arthritis Rheum 46: 3340-3348, 2002. Hull KM, Shoham N, Chae ]J, et al: The expanding spectrum of systemic autoinflammatory disorders and their rheumatic manifestations. CUlT Opin Rheumatol 15: 61...{i9, 2003,
C HAP T E R
35
CRYOPYRIN -ASSOCIATED PERIODIC SYNDROMES
7. Manji GA, Wang L, Geddes BJ, et al: PYFAFl, a PYRIN-containing Apafl-like Pfotein that assembles with ASC and regulates activation of NF-kappa B. J Bioi Chem 277: 11570--11575, 2002. 8. FrldmannJ, Prieur AM, Quartier P, et al: Chronic infantile neurological cutan~ous and articular syndrome is caused by mutations in CIASl, a gene highly expressed in polymorphonuclear cells and chondrocytes. Am J Hum Genet 71: 198-203, 2002. 9. Tschopp J, Martinon F, Burns K: NALPs: a novel protein family involved in inflammation. Nat Rev Mol Cell BioI 4: 95-104, 2003. 8. Neven B, Callibaut I, Prieur AM, et al: Molecular basis of the spectral expression of CJASI mutations associated with phagocytic cell-mediated auto-inflammatory disorders (CINCA/NOMID, MWS, FCU). Blood 103: 2809-2815, 2004. 9. Rosen-Wolff A, Quietzsch J, Schroeder H, et al: Two German CINCA (NOMID) patients with different clinical severity and response to anti-inflamqlatory treatment. Eur J Haematol 71: 215-219, 2003. 10. Janssen R, Verhard E, Lankester A, et al: Enhanced interluekin-l beta and interleukin-18 release in a patient with chronic infantile neurologic, cutaneous, articular syndrome. Arthritis Rheum 50: 3329-3333, 2004. 11. Agostini L, Martinon F, Burns K, et al: NALP3 forms an 11-1 beta-processing i"flammasome with increased activity in Muckle-Wells autoinflammatory dis',rder. Immunity 20: 319-325, 2004. 12. lKile RL, Rusk H. A case of cold urticaria with unusual family history. JAMA 114: 1067-1068, 1940. 13. Boffman HM, Gregory SG, Mueller jL, et al: Fine structure mapping of CIASl: identification of an ancestral haplotype and a common FCAS mutation, L353P. Hum Genet 112: 209-216, 2003. 14. Johnstone RF, Dolen WK, Hoffman HM: A large kindred with familial cold autoinflammatory syndrome. Ann Allergy Asthma Immunol 90: 233-237, 2003. 15. Muckle TJ, Wells M: Urticaria, deafness, and amyloidosis: a new heredofamilial syndrome. Q J Med 31: 235-248, 1962. 16. Wans RA, Nicholls A, Scott DG: The arthropathy of the Muckle-Wells syn~rome. Br J Rheumatol 33: 1184-1187, 1994. 17. Prost A, Barriere H, Legent DF, et al: Intermittent rheumatism revealing a familjal syndrome. Arthritis--urticarian eruptions-deafness: Muckle-Wells syndrome without kidney amylosis. Rev Rhum Mal Osteoartic 43: 201-208, 1976. 18. ~erratrice G, Pouget J: Muckle-Wells syndrome or association of joint pain attacks, urticarial outbreaks and sensory deafness!. Rev Rhum Mal Osteoartic 54: 105-108, 1987.
675
19. Nicolas JF, Garnier jL, Marchand C, Thivolet J: Muckle-Wells syndrome: a new familial case. Demonstration of abnormalities of lymphocyte subpopulations. Ann Dermatol Venereol116: 827-829, 1989. 20. Nazzari G, Desirello G, Crovato F: Recurrent urticarial skin eruption since infancy. Muckle-Wells syndrome (MWS). Arch Dermatol 131: 81--82, 84-85, 1995. 21. Throssell D, Feehally J, Trembath R, Walls J: Urticaria, arthralgia, and nephropathy without amyloidosis: another variant of the Muckle-Wells syndrome? Clin Genet 49: 130--133, 1996. 22. Lieberman A, Grossman ME, Silvers DN: Muckle-Wells syndrome: case report and review of cutaneous pathology. J Am Acad Dermatol 39 (Pt D: 290--291, 1998. 23. Granel B, Philip N, Serratrice J, et al: CIASl mutation in a patient with overlap between Muckle-Wells and chronic infantile neurological cutaneous and articular syndromes. Dermatology 206: 257-259, 2003. 24. Prieur AM, Griscelli C, Lampert F, et al: A chronic, infantile, neurological, cutaneous and articular (CINCA) syndrome. A specific entity analysed in 30 patients. Scand J Rheumatol 66 (Supp!) 57-68, 1987. 25. Torbiak RP, Dent PB, Cockshott WP: NOMID--a neonatal syndrome of multisystem inflammation. Skeletal Radiol 18: 359-364, 1989. 26. Prieur AM: A recently recognised chronic inflammatory disease of early onset characterised by the triad of rash, central nervous system involvement and arthropathy. Clin Exp Rheumatol 19: 103-106, 2001. 27. Lee AG, Warren RW: Optic disc edema in neonatal onset multisystem inflammatory disease (NOMlD). J Neuroophthalmol 19: 180--1, 1999. 28. Dollfus H, Hafner R, Hofmann HM, et al: Chronic infantile neurological cutaneous and articular/neonatal onset multisystem inflammatory disease syndrome: ocular manifestations in a recently recognized chronic inflammatory disease of childhood. Arch Ophthalmol 118: 1386-1392, 2000. 29. Hawkins PN, Lachmann HJ, Aganna E, McDermott MF: Spectrum of clinical features in Muckle-Wells syndrome and response to Anakinra. Arthritis Rheum 50: 607-612, 2004. 30. Frenkel J, Wulffraat N, Kuis W: Anakinra in mutation-negative NOMID/CINCA syndrome: comment on the articles by Hawkins et al and Hoffman and Pate. Arthritis Rheum 50: 3738-3739, 2004. 31. Federico G, Rigante D, Pugliese AL, et al. Etanercept induces improvement of an arthropathy in chronic infantile neurological cutaneous articular (CiNCA) syndrome. ScandJ Rheumatol32: 312-314, 2003.
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
-.
Peter N. Malleson and David D. Sherry
IIfIIr
Musculoskeletal pain of noninflammatory origin is a common cause of morbidity in childhood and a frequent cause of referral to pediatric rheumatologists, orthopedic surgeons, sports medicine specialists, and other physicians and ancillary health care providers, particularly physiotherapists. Noninflammatory causes of pain are much more common than inflammatory ones such as juvenile idiopathic arthritis, and their identification and differentiation from other causes of musculoskeletal pain such as infection or malignancy are essential to institute appropriate therapy and to avoid inappropriate investigations. This chapter focuses on specific noninflammatory conditions associated with musculoskeletal pain. It does not discuss the primary disorders of bone and connective tissues, which are the subject of a separate chapter.
PAIN ASSOCIATED WITH HYPERMOBILITY Generalized Hypemoblllty The term benign bypermobility syndrome is applied to those children with musculoskeletal pain associated with generalized hypermobility of the joints (or "double-jointedness") without any associated congenital syndrome or abnormality of connective tissue.1-3 Some syndromes associated with hyperrnobility are listed in Table 36-1; in most of these, it is immediately apparent that the child does not have straightforward benign hypermobility syndrome, but conditions such as Marfan or Stickler's syndrome can be overlooked if they are not actively considered in the differential diagnosis. The criteria for hypermobility have evolved over the years, and currently most authors use either the nine-point Beighton scale or the modified criteria of Carter and Wilkinson (Table 36-2).4,5 Estimates of the frequency of hypermobility range from 8% to 20% in white populations; Chinese and West Africans have a much higher prevalence of hypermobility.5--9 Girls are hyperrnobile about twice as often as boys. A family history of hypermobility is common. Although many children fulfill the criteria for benign hypermobility described earlier, having widespread joint 676
laxity, many more children are lax at only a few joints. 1O It is the authors' clinical experience that this more limited joint laxity is also a cause of musculoskeletal pain. It is not clear whether the pains are caused by the laxity at those few joints or whether an imbalance between lax and less lax joints is important. Although it is well recognized that extreme joint laxity can be associated with early and severe degenerative joint disease, with consequent joint pain, it is less obvious why hyperrnobile children with no evidence of structural joint damage have joint pain. Altered proprioception in hypermobile knee joints has been reported. 11 Hyperrnobility is frequently associated with intermittent pains that often appear to be precipitated by certain activities. Children aged 3 to 10 years are most strongly affected, because the prevalence of hypermobility decreases with age.'"--9,12,13 It is relatively uncommon for children with benign hypermobility to be significantly disabled by the pain, although there is some evidence (not supported by all studies) that hypermobility is associated with fibromyalgia. 9,14-16 Mild joint effusions may be observed. 6 Reassurance is the initial treatment of hypermobility. Supportive footwear is helpful for many. Some children benefit from a post-activity or evening dose of acetaminophen or a nonsteroidal anti-inflammatory agent. A very few children require a change of activity, although it is important that the alteration of activity not be interpreted by the family or child as a recommendation to stop participating in physical activities. Older, more severely affected children may be helped by formal physical therapy, including bracing of troublesome joints. 17 Although hypermobility is generally innocuous, there are several less benign associations with it (see Table 36-1). Infants with hypermobility/floppiness often have delayed development of major motor skills, and this may lead to unnecessary anxiety and physiotherapy.lH,19 Joint pain, if not identified as related to hypermobility, may also lead to unnecessary laboratory investigations and treatments in a mistaken belief that the pains are rheumatic in origin. 20 A benign bleeding tendency has been reported in children with hypermobile thumbs. 21 Both bladder and bowel problems due to "non-neurogenic
C HAP T E R
tl~
IABtE.36-1
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
Selected Conditions Associated With Hypermobility'
MIJ'fan Syndrome Tall and thin Ann span greater than height Lower ratio of upper-body segment to lower-body segment (long legs); normal ratio is 0.85 in whites and 0.92 in blacks Arachnodactyly Pectus excavatum or carinatum Kyphoscoliosis Dislocation of the lens of the eye Aortic root dilatation Heart murmurs, midsystolic click Hernias Autosomal dominant disorder due to mutations of fibrillin gene on chromosome 15
HQIIIOCYStinuria Marfanoid habitus Major risk of thrombotic events Autosomal recessive disorder usually associated with cystathionine j3-synthase deficiency due to mutations of gene on long arm of chromosome 21
Stklder's Syndrome Marfanoid habitus Typical facial appearance: malar hypoplasia, depressed nasal bridge, epicanthal folds, micrognathia Cleft palate (Pierre Robin sequence) Severe myopia (may lead to retinal detachment) Sensorineural hearing loss Mitral valve prolapse Aliltosomal dominant disorder due to mutations of type II collagen gene on chromosome 12
EJIIers.Danlos Syndromes Skin abnormalities: thin, hyperelastic, cigarette paper scars, easy bruising Dislocation of joints Rarely, artery aneurysms; hollow organ rupture Heterogeneous conditions; at least nine types with different inheritance patterns
Osteogenesis Imperfecta Blue sclerae bones with multiple fractures and deformities Short stature Spinal deformity Different types; usually autosomal dominant inheritance Involves abnormalities of type I collagen F~agile
Williams' Syndrome Short stature Characteristic elfin facial appearance Hoarse voice Friendly and loquacious Developmental delay Supravalvular stenosis Occasionally hypercalcemia Il1litially hypermobile but later become hypomobile without pain Sporadic and inherited cases due to deletion of elastin allele on chromosome 7
Down Syndrome (Trisomy 21) Hypotonia Developmental delay Characteristic facial appearance; epicanthal folds Short stature Endocardial cushion defects Broad hands with simian creases Brushfield (depigmented) spots of the iris Usually inherited in a sporadic fashion 'For further details about these conditions, the reader is referred to Jones KL, Smith's Recognizable Patterns of Human Malformation, 5th ed. Philadelphia, WB Saunders. 1997, Chapter 37, and Beighton p, McKusick's Heritable Disorders of Connective Tissue, 5th ed., 51. Louis, Mosby. 1993.
677
678
I! II
C HAP T E R
lABl E l6-2
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
(rilerid for HYPl'rll1olJllily
Modified CrIteria of Carter and Wllldnson· Three of five are required to establish a diagnosis of hypermobility: Touch thumb to volar forearm Hyperextend metacarpophalangeal joints so fingers parallel forearm >10 hyperextension of elbows >10' hyperextension of knees Touch palms to floor with knees straight
Belghton 5calet ~6 point~
defines hypermobility: Touch thumb to volar forearm (one point each for right and left) Extend fifth metacarpophalangeal joint to 90° (one point each for right and left) >10° hyperextension of elbow (one point each for right and left) >10° hyperextension of knee (one point each for right and left) Touch palms to floor with knees straight (one point)
Other nonalterla features of many children with hypermoblllty: Put heel behind head Excessive internal rotation to hip Excessive ankle dorsiflexion Excessive eversion of the foot Passively touch elbows behind the back 'Carter C. Wilkinson]: Persistent joint laxity and congenital dislocation of the hip. J Bone Joint Surg Br 46: 40-45. 1964. lBeighton P, Solomon L, Soskolne C: Articular mobility in an African population. Ann Rheum Dis 32: 413-418. 1973.
sphincter dysfunction" may be more common in children with hypermobility.22 Although hypermobility may enable a child to be a good gymnast or ballet dancer, injuries may be more frequent in hypermobile athletes. 23- 25 Therefore, having the benign hypermobility syndrome may actually mitigate against a successful professional career in ballet dancing26 or against strenuous training, such as in military recruits. 27 Temporomandibular joint dysfunction may be a consequence of hypermobility.28-30 Proprioception of the knee, reported to be diminished in hypermobile females, may lead to poor biomechanicalloading and microtrauma. l1 In addition to altered proprioception, there is some recent evidence suggesting a disturbance in the autonomic nervous system, though the significance of this finding is unclear. 31 Patients with anterior knee pain (often called chondromalacia patellae, although the evidence for any pathology of the patella in children is usually lacking) are more likely to be hypermobile than are control patients. 32 Back pain is also more common in hypermobile patients, especially those who spend more time sitting or standing; individuals whose activities require frequent changes of position experience less back pain. 32 .33 Premature osteoarthritis has been suggested as a consequence of hypermobility, but the evidence is not convincing. 34 One study showed no difference in bone density between those with and those without hypermobility.3s Attempts to determine whether benign hypermobility is a risk factor for later joint disease may be confounded in crosssectional studies by the fact that hypermobility in late
adult life appears to be a marker of fitness, with flexible older adults having less osteoarthritis and osteopenia. 36 Children who "crack their knuckles" are frequently hypermobile. Parents are often concerned that this activity might lead to joint damage, but it is probably not a cause of later osteoarthritis. 37 Children with benign hypermobility syndrome do not seem to be at an increased risk for aortic dilatation or mitral valve prolapse..ls..l8 ,39
Pes Planus The flexible flat foot is normal in very young children. Most infants have no longitudinal arch, and the development of this arch is part of normal growth. 40 In children, particularly those with hypermobility, the arch may exist when they are toe standing or lying (and they may actually appear to have a high arch) but disappears on weight-bearing. Usually, flexible flat feet are not a cause of significant discomfort. A study in adults with flexible feet found no relationship between pain scores and arch configuration. 41 A patient with flat feet and hindfoot valgus is shown in Figure 36-1. The occasional adolescent with a short Achilles tendon and hypermobile flat foot may have pain from excessive weight-bearing on the talar head. 42 This is rare, however, being present in only 25 of 3619 male soldiers. 43 Treatment is controversial. Wenger and associates 44 reported a prospective randomized trial of 98 children with flat feet who received either no treatment or corrective orthopedic shoes, a Helfet heel-cup, or a custom-molded plastic insert. There was significant improvement in most children, irrespective of treatment group. Orthotic devices that involve only the hindfoot do not reduce the mediolateral ground forces in adults with pes planus. 45 Mosca recommended aggressive heel cord stretching and adjusting of the shoes; if that failed, then a soft foot orthosis could be triedY Surgery to lengthen the heel cord is indicated only in the most extreme cases in skeletally mature adolescents. 42 ,46 Pes planus (and pes cavus) in the athlete may be associated with overuse injury more commonly than is a nor-
• Rgure 36-1 Kuchta.)
Pes planus with left hindfoot valgus. (Courtesy of Dr. G.
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
mal-appearing arch. 47 However, neither pes planus nor pes cavus (which can also be a cause of foot discomfort) has been shown to be a significant predictor of injury in army recruits,48 and one study reported fewer stress fractures in those with low arches. 49 No therapeutic studies in these populations are available. In contrast to the mobile flat foot, a rigid flat foot is always pathologic. It may result from tarsal coalition, in which a bone or cartilaginous bridge is present between tarsal bones, usually the talus and the calcaneus or the navicular and the calcaneus (Fig. 36-2). As the bridge ossifies, motion is restricted and pain may result; 25% of children with tarsal coalition have symptoms. 50 Computed tomography or magnetic resonance imaging (MRI) usually shows the coalition, and surgical intervention is required if conservative management fails.
679
condition may be associated with a congenital abnormality of the patella (unifaceted or bipartite patella), of the femoral condyles (shallow intercondylar groove), or of the patellar ligament (lateral attachment). Femoral anteversion, patella alta, and an increased Q-angle are also commonly found. Repeated episodes of dislocation lead to premature degeneration of the articular cartilage of the patellofemoral joint. The patellar apprehension sign-contraction of the quadriceps muscle when the examiner attempts to displace the patella laterally-suggests the diagnosis. Treatment consists of short-arc active and resistive exercises to strengthen the musculature around the knee joint, especially the vastus medialis, and center the patella. If these interventions fail, surgical realignment of the extensor system may be indicated.52
Hypomobllity Genu Recurvatum Genu recurvatum, like pes planus, may be part of a generalized hypermobility syndrome or may occur as an isolated phenomenon. Symptomatic genu recurvatum occurs most commonly in adolescent girls and is associated with popliteal pain and an increased incidence of anterior cruciate ligament injury.27.51 Symptoms are worse with standing or walking and are relieved by rest. Athletes may have particular difficulty.27 Treatment includes correction of bicpmechanical faults by use of orthotics; improving knee proprioception and muscle control (especially quadriceps strength) as well as gait; and maintaining good knee alignment during functional activities. 51
Recurrent Patellar Dislocation As part of the hypermobility syndrome or as an isolated
phenomenon, the patella may dislocate laterally. This is accompanied by a sensation of giving way with sudden pain and the inability to straighten the leg. The knee is held in a position of about 25 degrees of flexion. The
.: FIgure J6-Z Computed tomographic scan illustrates the bony bridge between the calcaneus and talus on the right side.The left hindfoot is normal.This feature may not be detectable on plain radiographs. (Courtesy of Dr. R. Caims.)
Although it has not been studied in a formal fashion, it is the authors' impression that a few children with slightly limited joint mobility (either as part of an underlying syndrome or as one end of the normal spectrum of joint mobility) may also present with arthralgias. It is not uncommon for children with back and lower-extremity arthralgias to have tight hamstring, quadriceps, and calf muscles. 53 Bowyer reported that four of five children with hip contractures due to an underlying spondyloarthropathy experienced hip pain. 54 The authors and others have seen children with a variety of individually relatively uncommon disorders, including hyalinosis and familial fibrosing serositis,55.56 who seemed to be in pain because of very stiff joints (Table 36-3). Most children with
I!:.
TABLE 36-3 Seleded Conditions Associated with HYPolllobility or Joint Contraclures'
Diabetes mellitus (diabetic cheiroarthropathy) Tightening of skin and soft tissues of fingers Short stature Scleroderma and scleroderma-like conditions Mucopolysaccharidoses and mucolipidoses with dysostosis multiplex Autosomal recessive inheritance (except in Hunter's syndrome, which is X-linked) Hyalinosis Familial fibrosing serositis Progressive contractures of fingers and toes Fibrosing pleuritis and constrictive pericarditis Probably autosomal recessive inheritance Camptodactyly syndromes (several familial conditions including B1au's syndrome) Flexion contractures of fingers Beals' contractural arachnodactyly syndrome Marfanoid features Crumpled ears Cardiac abnormalities unusual Linked to fibrillin-like gene on chromosome 5 (autosomal dominant inheritance) Winchester's syndrome Multicentric osteolysis particularly of fingers, starting in infancy Autosomal recessive inheritance 'For further details about the inherited conditions listed here, 1he reader is referred to Jones KL: Smith's Recognizable Patterns of Human Malformation, 5th ed. Philadelphia, WB Saunders, 1997, Chapter 37, and Beighton P: McKusick's Heritable Disorders of Connective Tissue, 5th ed., St. LOUis, Mosby, 1993.
680
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
marked stiffness due to conditions such as arthrogIYposis, Williams' syndrome, and cerebral palsy do not seem to complain unusually of arthralgias, so careful evaluation for other explanations for pain needs to be undertaken in children with joint contractures.
PAIN SYNDROMES RELATED TO OVERUSE Patellofemoral Pain Pain in the anterior aspect of the knee, originating in the patellofemoral joint, is quite common (Table 36-4).57,58 It is characterized by pain and tenderness in the region of the medial patellar facet and crepitation on movement of the joint. There are several possible causes of this disorder. 59-63 If the syndrome is accompanied by fissuring and fibrillation of the posterior surface of the patella, the term chondromalacia patellae is used. It is most common in teenage girls. There is insidious development of retropatellar knee pain. Pain occurs at first with activity that stresses the quadriceps (particularly eccentric contraction), such as deep knee bends or climbing or descending stairs, and is lessened by rest. Pain recurs with prolonged sitting with the knee flexed and can be relieved by extension of the leg. On physical examination, knee flexion may be accompanied.by patellar crepitus. Pain may be reproduced by compreSSIon of the patella and by palpation along its inferomedial edge. Pain may also be elicited if the upward movement. of the patella is restrained while the quadriceps. m~scle IS contracted. A small effusion may be present. Fmdmgs on standard radiographs and laboratory investigations are normal, but MRI can be diagnostic, showing decreased signal intensity, irregularity with focal thinning of the articular cartilage, or even cartilage denudation and subchondral cyst formation. Arthroscopy (if performed) may reveal ridging and strands of degenerating cartilage on the retropatellar sur. face, blistering, or frank denudation of cartilage. Patellofemoral pain syndromes can be chronic and difficult to treat, and no controlled studies have been done. Activities that provoke pain should be curtailed initially, and then reintroduced with a carefully graded increase of the activity in association with specific exercises aimed at strengthening the muscles around the knee, especially the vastus medialis muscle. A variety of different braces or strapping techniques are available, whose main .fu~ction is to maintain the movement of the patella wlthm the patellar groove. They are of limited value but may provide
J~.
TABl E 3b 4
Age at onset Sex ratio Symptoms
Signs
Pdlellofemordl Pdin
Syndroll1e~
Adolescence to young adulthood Girls> boys Insidious onset of exertional knee pain, difficulty descending stairs and walking downhill, need to sit with legs straigbt ("theater sign") Patellar tenderness on compression; quadriceps weakness; inhibition sign; small joint effusion
some help. Correction of pronated feet by custom-fitted orthoses and weight reduction in the overweight teenager may help. Surgical procedures, including shaVing off the patellar irregularities, are of questionable benefit.
Plica Syndromes The medial and lateral plicae arise from incomplete involution of the synovial membranes that separate the compartments of the knee during embIYologic development. These folds do not usually cause symptoms; however, if they become thickened, they can cause pain in the knee (Table 36-5).64 Occasionally the membranes do not involute at all, leaving a complete septum. A nonperforated septum can result in unusual swelling that may mimic a soft tissue tumor. 65 The mediopatellar plica syndrome is the more common condition associated with pathologic plicae and causes pain with flexion of the knee; erosion of the cartilage apposing the plica may eventually occur. There is often an area of tenderness over the superomedial border of the femoral condyle and locking or snapping during movement of the joint. The thickened plica can often be palpated as a thickened band on the medial aspect of the knee. MRI may show a thickened synovial plica occasionally accompanied by synovitis or cartilage erosion. The diagnosis is confirmed by arthroscopy; resection of the band can be performed at the same time and is often curative.
Stress Fractures and Physeal and Apophyseal Injuries Stress injuries occur when bones, physes, or apophyses (sites of growth cartilage where tendons insert) are damaged by their exposure to repetitive, nonviolent loads. They are particularly common in the adolescent athlete. 66 The intensity and duration of the sport is significantly related to the fracture rate. 67 The onset of pain and localized weakness due to reflex inhibition is usually insidious, occurring in an athletic adolescent who is performing a repetitive activity, such as long-distance running. Physical examination reveals localized bone tenderness with reproduction of pain on resisted movement. Early radiographs may be normal, or they may show only soft tissue swelling. Later radiographs demonstrate callus formation (Fig. 36-3), physeal widening, or apophyseal fragmentation. If there is diagnostic concern
I: II
TABlE 36 '}
Age at onset Symptoms Signs Arthroscopy
MediolJdlellar Pli(,l Syndrollle
Adolescence Medial knee pain, intermittent aching increased with activity or motion, giving way on weight-bearing, locking Medial palpable band, snapping on motion Fibrous band with hemorrhage and inflammation identified
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
681
• fIture 36-3
Serial radiographs document the evolution of astress fracture in a lO-year-old giri who was the hopscotch champion of her block.Two weeks after the onset of leg pain, the fracture line is evident. After 4 weeks, the fracture callus is seen. (Courtesy of Dr. Robert N. Hensinger.)
because of persistent bone pain or pain with activity, bone scintigraphy is the imaging technique of choice. There is significant overlap between these conditions and the osteochondroses (see later text and Table 36-7). The classic stress fracture is the march fracture of the metatarsals seen in members of the military. The most common site of a stress fracture in children, however, is the proximal third of the tibia (see Fig. 36-3), with metatarsal fractures being uncommon. Femoral fractures also occur with some frequency in young children. Some of these may be associated with nonaccidental trauma, and this possibility should be seriously considered if the child is not yet walking. 68
Lower-Limb Conditions Pelvic Apophysitis Pain may occur where abdominal and hip muscles insert into the pelvis. Particular conditions involve the insertion of the sartorius into the anterior superior iliac spine, the insertion of the rectus femoris into the anterior inferior iliac spine, the insertion of the iliopsoas into the lesser trochanter, and the insertion of the abdominal musculature into the iliac crest apophysis. 69 These conditions are associated with adolescent athletes, especially runners, but are usually not associated with a single traumatic event; they cause a dull pain in the general area of the hip on activity and may therefore be confused with intra-articular hip disease. The occurrence of pain on resisted contractions of involved muscles, in which the hip joint itself is not allowed to move, indicates that the pain is not caused by intrinsic hip disease. Additionally, a relatively common benign cause of pelvic pain in adolescents is apophysitis of the iliac crest. In this condition, pelvic brim or lumbar pain may occur spontaneously and is reproduced by direct palpation of the affected area of the iliac crest.70 The condition settles spontaneously after a few weeks or rest, much like Osgood-Schlatter disease. Once healed, the adolescent should gradually return to strenuous activity. Recognition of this entity prevents unnecessary investigations and interventions.
Osgood-Schlatter Disease Osgood-Schlatter disease 71 ,72 is an osteochondrosis caused by repeated trauma to the tibial tuberosity. Essentially, it is a microavulsion fracture that results when the infrapatellar tendon pulls out from the tibial tuberosity.73 It commonly occurs in athletic adolescents (Table 36-6).74 There is a complaint of pain over the tibial tubercle that is exacerbated by exercise or kneeling. On examination, tenderness and, often, swelling of the tibial tubercle and patellar tendon insertion are present. Radiographs-which should be obtained to exclude other conditions including infection, neoplasia, avulsion fracture, and stress fracture-may show soft tissue swelling and an enlarged and fragmented tubercle (Fig. 36-4). However, it is normal for the tibial tubercle to appear irregular in adolescence, and it may be difficult to differentiate normal development from disease. Ultrasonography can help in identifying the lesion. 75 Laboratory studies reveal no evidence of chronic inflammation. There is no association with the human leukocyte antigen HLA-B27, so it is not a forme fruste of enthesitis-related juvenile idiopathic arthritis. 76 Rest or use of a basketball knee protector is usually sufficient treatment. The outcome is usually very good.?7
Sinding-Larsen-Johansson Disease Sinding-Larsen-Johansson disease is an osteochondrosis caused by repeated trauma to the inferior pole (secondary ossification center) of the patella (Fig. 36-5); essentially
11:'.
TABLE 36-6
Age at onset Sex ratio Symptoms Signs Investigations
Osgood-Schlaller Disease
Athletic adolescents Boys> girls Pain over the tibial tubercle exacerbated by exercise, unable to kneel because of pain Tenderness and swelling over the attachment of the infrapatellar tendon Radiograph shows soft tissue swelling, enlarged and sometimes fragmented tubercle
682
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
Sever's Disease Sever's disease is a common cause of heel pain that usually occurs in physically active individuals in early adolescence. The cause is uncertain. Liberson and colleagues80 suggested that radiographic findings of a dense calcaneal apophysis are normal, occurring as commonly in control subjects as in subjects with heel pain, but that radiographic lucencies or fragmentation changes are more commonly associated with pain. Based on radiographic changes, computed tomographic scans, and histologic examinations, they hypothesized that the cause of Sever's disease is a process of remodeling of the calcaneal apophysis secondary to repetitive stresses of traction from the Achilles tendon and plantar fascia and impact from weight-bearing. This remodeling process is usually a normal, subclinical event but it becomes symptomatic when it is excessive.
Upper Limb Conditions
• Figure 36-4 Radiograph of the knee of a boy with Osgood-Schlatter
disease. In addition to fragmentation of the apophysis, the soft tissues overlying the tibial tuberde are thickened. (Courtesy of Dr. R. Cairns.)
it is an avulsion fracture that results when the infrapatellar tendon pulls out from the patella. 78 ,79 It may be confused with Osgood-Schlatter disease or infrapatellar tendinitis, in which pain at the inferior pole of the patella may also occur. Treatment consists of rest and temporary reduction in physical activities that involve the legs. Most cases resolve within 3 to 12 months.
Upper limb stress injuries occur less commonly than lower limb conditions. Shoulder pain on active and resisted movement is associated with widening of the proXimal humeral physis. Known as little league shoulder, it occurs in children participating in sports activities, such as baseball pitching, that involve repetitive overarm movements,Rl These overarm movements can also be associated with pain at the elbow, caused by an apophyseal injury to the medial epicondyle (golfer's elbow). A similar injury is tennis elbow, in which the pain is caused by apophysitis of the lateral epicondyle at the insertion of the common wrist extensors. These elbow syndromes can be helped by the use of a "tennis elbow strap," which is applied fairly tightly around the forearm just distal to the tender site. It is believed to help diminish the pain by inhibiting full muscle expansion, thereby diminishing intrinsic muscle force on the lateral epicondyle. Glucocorticoid injections into the site of maximal tenderness may be required if the symptoms do not respond to conservative measures. Little league elbow is another stress injury in which there is a painful fragmentation of the capitellum of the distal humerus. It is probably the same condition as Panner's osteochondrosis, which usually affects preadolescent boys. The whole ossific nucleus becomes flattened and fragmented, but the condition resolves with no long-term sequelae. 82 A variety of bony injuries around the elbow occur in young gymnasts; those involving the articular surface of the joint necessitate permanent discontinuation of competitive gymnastics, whereas those involving the olecranon have a good outcome. R3 ,84
Soft nssue Stress Injuries
3~ Radiograph of the knee of a boy with Sindling-LarsenJohansson disease.The lower pole of the patella has been separated from the patella.
• Figure
Repetitive trauma can cause soft tissue injuries in addition to bony lesions. Tenosynovitis is relatively common, particularly in the athletic adolescent and in relatively inactive children who, without proper preparation, overengage in an activity (the "weekend warrior").
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
Shin Splints The term shin splints is applied to a pain syndrome in the posteromedial aspect of the lower half of the shin, usually in o~der adolescents. It is associated with running, jogging, or walking; is worse with the activity; and is relieved by rest. s; It is caused by periostitis at the insertion of the sole~s muscle. 86 The diagnosis is confirmed by the demonstration of tenderness at the posteromedial border of the distal tibia. Stress fractures can mimic shin splints. Rarely, an anterior compartment syndrome resulting from hemorrhage or edema, accompanied by ischemic changes in the foot, is confused with shin splints. Shin splints are best prevented by adequate stretching and strengthening and avoidance of jogging or walking on hard surfaces. Treatment is symptomatic, with rest, stretching, ice, nonsteroidal anti-inflammatory drugs, proper footwear, and a graduated rehabilitation program. S;,86 Orthotics may help patients with pronated feet or excessive subtalar motion.
Tenosynovitis Although tenosynovitis commonly accompanies rheumatic disease, it can also occur as a result of unaccustomed repetitive movement, especially around the ankle (Achilles tenosynovitis, anterior tibial tenosynovitis) or at the wrists, or as stenosing tenosynovitis of the abductor pollicis longus or extensor pollicis brevis (de Queroain's disease). Treatment requires rest, with splinting if needed, and possibly injection of glucocorticoid into the tendon sheath. For a complete discussion of these and other overuse syndromes, the reader is referred to the work of Sheon and associates. 87
OStEOCHONDROSES As indicated earlier, there is a great deal of commonality among many of the stress-related injuries and the osteochondroses, and dividing the discussion into individual conditions is somewhat arbitrary, Some members of this heterogeneous group of disorders (apophyseal injuries) were described earlier; others, of particular significance to pediatric rheumatologists, are discussed briefly in the following sections. Some of the conditions commonly considered to be osteochondroses are probably variants of normal ossification, some are probably the result of stress injury, and others may be caused by avascular necrosis without any obvious precipitating trauma, perhaps occurring because of some inherent predisposition to vascular insufficiency. This uncertainty of the etiology has led to confusion and contradictory nomenclature and classification. Brower defined osteochondrosis as "a condition in which the primary or secondary ossification center in the growing child undergoes aseptic necrosis with gradual resorption of dead bone and replacement by reparative osseous tissue."88 Resnick uses a somewhat broader definition, stating that an osteochondrosis is any of "a group of disorders that share certain features: predilection for the immature skeleton; involvement of an epiphysis,
683
apophysis, or epiphysioid bone; and a radiographic picture that is dominated by fragmentation, collapse, sclerosis, and frequently, reossification with reconstitution of the osseous contour."S9 From a clinical point of view, osteochondroses might be defined as idiopathic, acquired, localized disorders of cartilage and bone, often characterized by localized pain. The osteochondroses usually affect a single site but may be bilateral; they are characteristically observed in children between the ages of 3 and 12 years and are much more common in boys than in girls. In large part, these disorders have been defined by their radiologic appearance, although, as already discussed, it is now clear that some of these findings are normal variants. In Table 36-7, the osteochondroses are listed according to the sites affected, and the putative causes are indicated.
li'~
TABtE 36-7
The Osteochondroses
Eponym
Mechanism
Basal phalanges Second metacarpal head Lunate Carpal navicular Distal ulna Capitellum of humerus
Thiemann Mauclaire Kienoock Prieser Burns Panner
Head of humerus
Hass
Trauma Trauma Osteonecrosis Trauma Trauma Trauma? Avascular necrosis Trauma
Area Affected Upper Extremity
Lower Extremity Second metatarsal head Fifth metatarsal base Tarsal navicular Talus Calcaneal apophysis Distal tibia Tibial tubercle Proximal tibia Intercondylar spines Primary patellar center Secondary patellar center Greater trochanter Femoral epiphysis Femoral epiphysis
Freiberg Iselin Kohler Diaz Sever Liffert-Arkin OsgoodSchlatter Blount Caffey Kohler Sinding-LarsenJohansson Mandl Legg-CalvePerthes Meyer's dysplasia
Osteonecrosis Trauma Normal variation Trauma Normal variation Trauma Trauma Trauma ? ?
Repeated trauma ? Osteonecrosis Normal variation
AxIal Skeleton Vertebral body Disk Vertebral epiphysis Iliac crest Symphysis pubis Ischiopubic synchondrosis Ischial apophysis
Calve Schmorl Scheuermann Buchman Pierson Van Neck Milch
? ? Repeated trauma ? ? Normal variant ?
Adapted from Brower AC; The osteochondroses. Orthop elin North Am 14: 99, 1983: Resnick D: The osteochondroses. In Resnick D, Niwayama G (eds): Diagnosis of Bone and Joint Disorders. Philadelphia, WB Saunders, 1981, p 2874: and other sources.
684
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
Legg-Calve-Perthes Disease Legg-Calve-Perthes disease was independently and simultaneously described by Legg,90 Calve,91 and Perthes. 92 It is an idiopathic avascular necrosis of the femoral head that occurs in children between about 5 and 10 years of age. It is approximately four times more common in boys than in girls, and it affects both hips in 10% to 15% of cases. There is an increased incidence among family members of an index case; Asians and whites are more frequently affected than are Native Australians, Native Americans, and blacks; and the disease occurs more frequently in urban than in rural areas.9.~ It has been observed that children with Legg-Calve-Perthes disease have delayed skeletal maturation and reduced height. 94 The cause of Legg-Calve-Perthes disease is unknown, but it is generally accepted that the condition occurs in individuals whose vascular supply to the femoral head is particularly vulnerable to interruption. There is evidence that children in the Legg-Calve-Perthes age range have a less extensive anastomotic vascular network to the femoral epiphysis than at other ages, and that boys have a less complete network than do girls. There is also increasing evidence of a thrombotic predisposition in some children. Data from two studies suggest that the factor V Leiden gene, responsible for resistance to protein C activation and therefore associated with an increased risk of thrombosis, is found more commonly in individuals with Legg-Calve-Perthes disease. 95.% However, another study failed to find any evidence of a thrombotic diathesis. 97 A recent study found both an increased frequency of the ~-fibrinogen gene G-455-A polymorphism and an increased exposure to passive smoke in children with Legg-Calve-Perthes disease, compared with controls. 98 Children who were perinatally infected with human immunodeficiency virus (HIV) appeared to be at increased risk of developing Legg-Calve-Perthes disease. 99 Affected children present with a limp and varying degrees of hip pain. Radiographs taken shortly after the onset of symptoms are often normal, but later radiographs show a progression through four stages: (1) an initial stage, at which there may be a small ossific nucleus, widening of the joint space, irregularity of the physis, and a subchondral radiolucent area; (2) a fragmentation stage, at which the bony epiphysis begins to fragment and there are patchy areas of increased radiolucency and radiodensity; (3) a reossification stage, at which normal bone density returns, radiodensities develop in previously radiolucent areas, and abnormalities of the shape of the femoral head and neck appear; and (4) the healed stage, at which the bone density is normal but the head is left with residual deformities. These changes may take several years to complete. Bone scans and MRI studies have a greater sensitivity for the detection of early disease than do plain radiographs (Fig. 36-6). The prognosis of Legg-Calve-Perthes disease depends on how extensive is the involvement of the epiphyseal head. Based on a study of 48 patients (51 involved hips) who were monitored for a mean of 50.2 years, the best prognostic indicator for long-term outcome appears to be the shape of the femoral head at skeletal maturity: normal or flattened spherical heads cause little problem, but
irregular heads are associated with a poor outcome. 100 The greater the extent of the necrosis, the more likely the femoral head is to become widened and flattened and uncontainable within the acetabulum. These changes predispose the individual to decreased range of movement at the hip and later changes of osteoarthritis. The aim of all forms of treatment for this condition is to maintain the femoral head well covered within the acetabulum, so as to minimize the deformity of the head. There is considerable uncertainty about the benefits of various surgical procedures compared with nonsurgical interventions such as splints.JOJ.102 A study by Lahdes-Vasarna and associates lOJ demonstrated that femoral heads with less than 50% involvement had a significantly better radiographic outcome than did those with greater than 500/0 involvement, and that hips treated by femoral varus osteotomies did only a little better than those treated with containment splints. The more severely affected hips may benefit from operative intervention. A recent study of 610 children with Legg-Calve-Perthes disease found that only 24% of untreated patients had a spherical femoral head at follow-up. 103
Scheuermann's Disease Scheuermann's disease (juvenile kyphosis) may be considered to be an osteochondrosis of the ring apophysis of the vertebral body, perhaps caused by repeated trauma, although the cause is far from clear and there is no consistency to its definition. It is most common in girls between 13 and 17 years of age, who present with pain in the midthoracic or lumbar spine; it may, however, be painless but causes a round-shouldered appearance and dorsal kyphosis. Radiographically, it is associated with anterior wedging of one to three adjacent vertebral bodies by at least 5 degrees each. 104 The kyphosis is thoracic in 75% of patients and thoracolumbar in most of the rest, except for the rare child with disease limited to the lumbar spine. 105 A prominent fixed dorsal kyphosis with a compensatory increase in the lumbar lordosis is characteristic. Tightness of the pectoral and hamstring muscles has been noted. 106 A standing lateral radiograph of the spine reveals anterior vertebral wedging, irregularity of vertebral end plates (Schmorl's nodes), increased anteroposterior diameter of the affected vertebral bodies, and increased dorsal kyphosis (Fig. 36-7). Treatment usually consists of simple analgesia and, occasionally, the use of a back brace to prevent flexion. The outcome is usually very good. Surgical interventions are restricted to patients with severe kyphoses (greater than 60 or 70 degrees). A significant correction of the degree of kyphosis can usually be obtained immediately postoperatively, but the curve tends to worsen again over time. 107 There is no correlation between functional outcome and radiographic outcome. J07 A cohort study of all 63 patients treated in a single institution by exercise and observation only, by Milwaukee bracing, or by surgical fusion using Harrington rods failed to find any statistical difference in quality of life, pain, function, or degree of curve between the different treatment modalities at a mean of 14 years after treatment. 108 Patients with curves exceeding 70
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
685
• Figure 36-6 Progression of Legg-Calve-Perthes disease. A, On the right side, the capital femoral epiphysis is flattened and sderotic; on the left, it is fragmented.The femoral metaphyses are widened, especially on the left side. B, Technetium 99m bone scan of another patient shows absence of uptake of the isotope in the right capital femoral epiphysis (arrow) but normal uptake on the left side. C, Magnetic resonance image of aseptic necrosis of the right femoral head, thought to be related to prolonged glucocorticoid intake, in a boy with dermatomyositis. The entire femoral head and the marrow in the metaphysis are abnormal. (A and C, courtesy of Dr. R. Cairns; B, courtesy of Dr. H. Nadel.)
degrees at follow-up did have inferior function. These data suggest that operative intervention should be reserved only for very large curves.
Ki5hler's Disease Kohler's disease is an osteochondrosis of the tarsal navicular bone. It tends to affect children between 4 and 9 years of age, who present with onset of insidious foot pain and a limp. The affected child characteristically bears weight on the lateral aspect of the foot. 109 Some authorities consider the radiographic findings of navicular narrowing, increased density, and sometimes fragmentation to be a variant of normal ossification (Fig. 36-8). There is no evidence that treatment affects outcome, which is almost always complete recovery; any
persistent or late-onset foot complaints are usually caused by other, unrelated pathology yo
Freiberg's Disease Freiberg's disease is an osteochondrosis of the second metatarsal head occurring most frequently in adolescent girls. Other metatarsal heads are occasionally affected. It causes localized pain on weight-bearing, sometimes with swelling in the region of the second metatarsal head. This condition has usually been ascribed to trauma. ll1 A retrospective study of 31 patients, however, elicited a history of trauma in only 15%, and pedobarographic studies failed to show abnormally high pressure at the affected metatarsal head. 112 The researchers observed that, in 85% of cases, the affected metatarsal was the
686
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
• Figure :16-. Freiberg's disease affecting the left second metatarsal head. (Courtesy of Dr. R. Cairns.)
heads and rest; casting may be necessary for pain control. 46 The outcome is usually good.
• Fllure :16-7 Lateral radiograph of the spine illustrating the abnonnalities of the anterior margins of the vertebral bodies that are characteristic of Scheuennann's disease and result in anterior wedging. (Courtesy of Dr. R. Calms.)
longest in the foot, and that this might, in some poorly explained way, predispose the metatarsal head to vascular insufficiency and infarction. Radiographs reveal increased density or flattening of the affected metatarsal head (Fig. 36-9). Treatment is usually supportive, with shoe inserts to reduce weight-bearing on the metatarsal
thiemann's Disease Thiemann's disease is an osteochondrosis caused by osteonecrosis of the phalangeal epiphyses, possibly secondary to trauma. Thiemann's disease is characterized by progressive, painless enlargement-during adolescence-of the epiphyses of the proximal interphalangeal joints of the hands and the interphalangeal joints of the first toes. 113 ,114 Flexion contractures of the large joints also occur. Radiographically, there is irregularity of the epiphyses of the digits. Results of tests for acute phase reactants are normal. Disability is minimal. The condition may be familial. ll5- 117
TRAUMA-INDUCED CONDITIONS In some patients with the apophyseal stress injuries described earlier, the injury may be more severe and may be associated with avulsion. In these situations, surgical intervention may be required. This next section does not attempt to cover all types of musculoskeletal conditions caused by trauma; rather, it focuses on conditions that are more likely to come to the attention of the pediatric rheumatologist. For more detailed information, the reader is referred to a standard orthopedic textbook.
Osteochondritis Dissecans
Flgure:l6-8 Kohler's disease.The tarsal navicular on the left (arrow) is flattened and sderotic. (Courtesy of Dr. R. Cairns).
Osteochondritis dissecans is a condition that has been described as a primary avascular necrosis of bone with a secondary involvement of the overlying cartilage,118 but it has also been characterized as a disturbance of endochondral ossification. 119 Osteochondritis dissecans manifests clinically with activity-related pain, and sometimes with recurrent bland effusions, although there may only be localized tenderness on examination. If there has been complete separation of a fragment, there may also
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
be joint-locking. It is related to trauma, but it is more frequently caused by repeated microtrauma and overuse than by a single traumatic episode.u s In a study of 51 knees in 38 patients with osteochondritis of the lateral condyle using MRI, there were some associated meniscal tears or discoid menisci, suggesting that altered mechanics is an important etiologic factor. 12o Bilateral lesions are common (approximately 20%). Occasional familial cases suggest that a genetic predisposition to vascular insufficiency of the subchondral bone may be of etiologic importance .116 Although the condition is usually recognized by plain radiography, because of the appearance of a subchondral fracture, the use of MRI has led to an increased recognition that pure cartilaginous separation can occur, even if radiographs appear normal (Fig. 3~1O). The inner aspect of the distal femoral medial condyle is the most commonly affected area (Fig. 3~11); however, osteochondritis dissecans of the dome of the talus (Fig. 3~12), the capitellum, or the patdla also occurs fairly commonly. MRI is almost as accurate as arthroscopy at staging the condition l21 and predicting outcome. 122 It has become the method of choice for staging the lesion. Intact cartilage, contrast enhancement of the lesion, and absence of osseous "cystic" defects are designated stage 1. Treatment can be conservative (avoidance of weight-bearing for a few weeks), precluding the need for arthroscopy. A cartilage defect with or without complete separation of the fragment, fluid around an undetached fragment, an osseous "cystic" lesion, and a dislodged fragment are stage 2 findings, which probably require arthroscopy and surgical intervention. 11S Persistence of symptoms despite non-weight-bearing in individuals with stage 1 findings also usually requires arthroscopic investigation. Surgical interventions include trying to relocate the fragments, by various fixation methods, and removing the loose bodies. Patients with open physes and small lesions tend to have
• 'figure 36-10 In this magnetic: resonance image, the area from which the osteochondral fragment has arisen is seen as a black area in the posterior part of the articular surface of the femur (curved arrow).The loose fragment of cartilage is indicated by the straight arrow. (Courtesy of Dr. R. Caims.)
687
• RlIUre 36-11 Osteochondritis dissecans is evident in the posterior aspect of the medial femoral condyle (arrow). The loose fragment of cartilage and bone is not evident. (Courtesy of Dr. R. Caims.)
a better prognosis than do those with closed physes or larger lesions. I22
Traumatic Arthritis Joint swelling associated with trauma occurs in older school-aged children and adolescents. An effusion arising immediately after an injury is more likely to be associated
• RlIUre 36-1Z dome of the talus.
Osteochondritis dissecans is seen on the medial side of the
688
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
with intra-articular hemorrhage, fracture, or joint derangement than is swelling that develops over several hours. A history of injury is often elicited in children with juvenile rheumatoid arthritis, because a minor injury brings a swollen joint to parental attention. Trauma, especially minor trauma, is not an explanation for joint swelling in a young child, and internal joint derangements (meniscal tears) are very rare. Juvenile rheumatoid arthritis is far more common than traumatic arthritis in the very young. Transient joint swelling can result, however, from patellar subluxation or from repetitive trauma associated with overuse syndromes or structurally abnormal joints in older children.
Slipped Capital Femoral Epiphysis Although slipped capital femoral epiphysis (SCFE) and acute chondrolysis of the hip are discussed here, their actual relationship to trauma is unclear, and often no significant history of trauma is elicited, although it seems probable that abnormal mechanical loading contributes to their occurrence. The condition most commonly affects overweight boys in early to middle adolescence. Children with SCFE have a higher average body mass index than other children. 123 There are ethnic differences in the frequency of the condition, which occurs more commonly among blacks and Polynesians. 124 The explanation for the racial differences is unclear. It has been postulated that individuals with an increased acetabular depth are at increased risk; but a study of healthy adults showed that the racial differences in acetabular depth did not match the known racial prevalence of SCFE.125 The mean age at onset of SCFE is about 11 years in girls and 12.5 years in boys, but the range is large, and children as young as 8 years of age can be affected. 124 The condition usually manifests with limp and pain in the affected hip. In the case of a subacute or a chronicon-subacute slip, the onset of symptoms is insidious. Occasionally an acute slip develops after a moderate injury, resulting in more severe pain and inability to walk. Radiographs are usually diagnostic, assuming a good lateral (axial) view of the hip is obtained; they show a posterior and downward slip of the femoral head on the neck caused by a separation through the growth plate between the zones of hypertrophic and calcified cartilage. Occasionally, symptoms can precede radiographic changes. If the condition is suspected, MRI can be helpful in diagnosing a "pre-slip."126 Once the diagnosis is suspected, the child should be given crutches and told to avoid weight-bearing until he or she can be seen urgently by the orthopedic surgeon. In general, the slip is classified as mild if it is less than one third the diameter of the femoral head and severe if it is greater than this amount. More recently, new classification schemes have been described that are probably of more prognostic utility than the traditional ones. 127 It has also been suggested that decreased radionuclide uptake on bone scans by the physis of the greater trochanter on the affected side, and later its premature closure on radiographs, is a predictive sign for the development of acute chondrolysis. 128 Although SCFE usually manifests with the
involvement of one hip only, the other hip frequently becomes involved with time. Eventually, between 300/0 and 45% of affected individuals will have evidence of bilateral involvement.124.129 Very mild slips may be treated conservatively, but there is always a risk of slip progression, so careful observation is mandatory. If the slip progresses surgical intervention is required. I3O Treatment is by fixation, usually with a single central screw. Acute chondrolysis is a complication of both treated and untreated SCFE and occurs in about 6% of cases; avascular necrosis occurs in about 5% of cases. 129,131
Acute Chondrolysls of the Hlp Acute chondrolysis of the hip is an unusual condition in which there is an insidious onset of hip pain and limitation of movement in association with radiographic evidence of progressive loss of articular cartilage. It can occur as an apparently idiopathic event or secondary to other hip pathology, particularly SCFE or Legg-Calve-Perthes disease. Idiopathic chondrolysis usually occurs in adolescents and may be more frequent in black girls. 13I Some children reportedly have minimal symptoms on long-term followup, with radiographs showing apparent restoration of joint cartilage, often with lateral overgrowth of the femoral head and lateral acetabular osteophyte formation. 132 However, a much worse outcome has also been described; in one study, all 11 patients, a mean of 13 years after presentation, had constant pain and stiffness with severe radiologic damage. 133 It has been suggested that idiopathic chondrolysis may be caused by synovitis of the hip, perhaps from a spondyloarthropathyl31,134; however, other studies have failed to show histologic evidence of significant synovial inflammation. 132 MRI shows cartilage loss (especially centrally), small hip joint effusions, and regional muscle wasting without synovial enhancement. Chondrolysis associated with a SCFE appears to have a better long-term prognosis than does idiopathic chondrolysis. 127
Nonacddental Injury An abused infant or child is sometimes thought to have a rheumatic disease because of a refusal to walk or bear weight, the presence of a joint effusion, or the presence of skin lesions that are believed to be vasculitic in origin. These children may be misdiagnosed as having juvenile idiopathic arthritis if a proper history is not obtained. Beating a child over the hands and knuckles results in brawny induration of the dorsal surfaces of the hands as well as thickened, dense round bones and bone chips on radiographs. Physical abuse should be considered in the child who has a history of repeated visits to the emergency department because of poorly explained trauma, the occurrence of allegedly spontaneous bruises, or hemarthrosis without an underlying bleeding disorder. Radiographic demonstration of multiple fractures and periosteal new bone formation is characteristic. The syndrome termed Munchausen's syndrome by proxy is a bizarre form of child abuse induced by the parent to bring the child to medical attention, usually to meet the pathologic psychological needs of the mother.
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
Rarely, a child with ecchymoses and deep bruising due to Henoch-Schonlein purpura or autoerythrocyte sensitization syndrome is thought to have been abused. 135 Awareness of this condition can save such children and their families much suffering.
Frostbite Arthropathy Frostbite is a cold-induced necrosis of the superficial tissues caused by freezing. The acral or exposed areas, including the fingers, toes, nose, and ears, are predominantly affected. Immediately after exposure, the diagnosis of frostbite can be made by the characteristic appearance of swollen, red fingers or toes. The history of cold exposure should be obvious, although the parents may not have been aware of the increased susceptibility of the very young child to cold injury. Vasomotor changes suggesting Raynaud's phenomenon may persist for months. In the growing child, frostbite produces a characteristic stunting of growth of the small bones and aero-osteolysis (Fig. 36-13).136-138 Secondary symptoms of osteoarthritis may develop in early adulthood.
689
Congenital Indifference to Pain In the rare syndromes of congenital indifference to pain, affected children develop swollen but painless joints associated with induration or necrosis of the toes and fingers (Fig. 36-14). Children with this condition, which may be an autosomal recessive trait, also frequently have anhydrosis, self-mutilating behaviors, and mental retardation. This neurogenic arthropathy (Charcot'sjoint) results in severely damaged, unstable joints that may eventually require arthrodesis to maintain some degree of function. One study suggested that substance P containing nerve fibers may be absent in this condition, confirming the importance of substance P in nociception within normal diarthrodial joints. 139 In three unrelated children with this condition, different mutations of the tyrosine kinase receptor gene for nerve growth factor were found, indicating that abnormalities of this gene may be responsible for this condition. 140 This finding was confirmed in other studies. Neurogenic arthropathy in children can also be caused by other lesions, for example as a consequence of spinal dysraphism. 141
PAIN SYNDROMES AFFECI1NG THE BACK, CHEST, OR NECK
• fItpn 3&-U The effects of frostbite. A and B, Necrosis of the distal epiphyses of all digits except the thumbs and left second and third fingers has resulted in growth failure.
Noninflammatory disorders of the back, chest, or neck are important causes of chronic or recurrent pain in children. Pain can arise from a wide variety of causes, including congenital, developmental, or acquired defects and nonmusculoskeletal diseases. Back pain in the general pediatric population is very frequent in developed countries. A study from Denmark reported that at least 50% of teenagers had one episode of back pain 142 j a frequency of 74% was reported from Switzerland143 and 44% from Iceland 144 (where 21% had weekly back pain). The prevalence is even higher in athletes144.145 but, perhaps surprisingly, is lower (only 32%) in those with idiopathic scoliosis. 146 Volinn 147 reviewed the literature and concluded that low back pain was twofold to fourfold higher in Swedish, German, and Belgium populations than in southern Chinese, Indonesian, Nigerian, and Filipino farmers. In contrast, back pain is a relatively infrequent reason for referral to a pediatric orthopedic surgeon,148 accounting for 2% of referrals. Among those children seen by an orthopedic surgeon because of low back pain, no specific cause could be found in almost half; Scheuermann's disease was present in 15%, spondylolysis in 13%, infection in 8%, disk prolapse in 6%, and tumor in 6%.148 A fairly common cause of back pain in very young children is diskitis (see Chapter 28). A number of studies have explored what factors might contribute to back pain in children for which no specific cause could be found. The weight of backpacks carried by schoolchildren is a plausible contributing factor, but recent data do not support the hypothesis that this causes back pain. 149 There is some evidence that long hours sitting at a computer or in front of the television may be an adverse factor. However, back pain (like most pain conditions) is multifactorial in origin, and psychosocial factors such as
690
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
• Figure 36-14 Radiographs of the effects of congenital indifference to pain. A, This chila has lost the tips of several fingers (atrow). B Hypertrophic and destructive changes are visible in the calcaneus, talus, and mldfoot (atrow). Cand D There is marked loss of the medial compartment space and calcifications typical of Charcot's arthropathy (arrow in D). Oinically, the knees were warm and contained large effusions. J
J
mood, parental support, and health perception seem to be at least as important as mechanical factors such as body mass index or backpack load. 149 Relatively little is known about the most effective way to treat back pain in children. In a chiropractic study, 40% of adult patients rated themselves as "much improved" after 7 days of treatment, and 82% after 30 days; those with chronic back pain were less likely to improve quickly.150 Whether children would improve at the same rate without treatment is unknown. In a longitudinal study, more than 50% of children who reported back pain initially denied ever having had back pain
when reinterviewed 2 years later, indicating that for many children back pain resolves spontaneously. J5J A controlled study suggested that a back education program may be an effective way of reducing pain prevalence for at least 1 year. J52 Chest pain is also a fairly frequent complaint in the pediatric population. Rowe and coworkers l53 reported that 6 of 1000 emergency department visits in a children's hospital were for chest pain. Chest pain occurred with equal frequency in boys and girls. Of 366 patients, 28% were diagnosed with chest wall pain; only 1% (5 of 336) had cardiac causes (Table 36-8). Precordial catch syn-
C HAP T E R
.~
_I
IABl E 36-8
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
Causes of Chest Pain in Children Presenting
to all Emergellcy Oepartmen
t
Cat8aorY
%
Conditions Induded
Chest wall
28
Lung/pleura
19
Tmuma Psychogenic
15 5
Otl1er
21
Unknown
12
Costochondritis, Tietze's syndrome, musculoskeletal pain, breast tenderness Asthma, infection, embolism, pleurisy, pneumothorax Contusion, abrasion Depression, anxiety, conversion disorder, hyperventilation Esophagitis, gastritis, upper respiratory tract infection, constipation, cardiac causes Chest pain of undetermined cause
Adapted and reprinted from Rowe BH, Dulberg CS, Peterson RG, et aI: Chamcteristics of children presenting with chest pain to a pediatric emergency department. Can Med Assoc J 143: 388, 1990, by permission of the publisher.
drome, previously known as Texidor twinge, is a very under-recognized condition in which there is a history of recurrent, well-localized, sharp chest wall pain of sudden on$et lasting a few seconds to minutes only, with negative findings on examination or laboratory testing. 154
SponcIyIolysls and Spondylolisthesis Spondylolysis is a defect in the pars interarticularis, most commonly of the fifth lumbar vertebra (L5). In one study of 185 children with spondylolysis, 193 defects were detected: 1 at L2, 6 at L3, 16 at L4, 168 at L5, and 2 at L6. 155 If there is bilateral spondylolysis, the affected vertebra may slip anteriorly, giving rise to spondylolisthesis; slips do not seem to happen with unilateral lesions. 156 Pars lesions are rare before the age of 5 years and usually occur in adolescents. The condition occurs in 6% of the population and frequently is asymptomatic. 156-158 Thirty children with pars lesions who were diagnosed during a prospective study of 500 first-grade children were monitored for 45 years. 156 There was no association between slippage and low back pain. The rate of progression of spondylolisthesis slowed with each passing decade, and none of the subjects developed a slip of 40%. The overall well-being of the group, as measured by the Medical Outcome Study Short-Form Health Survey (SF-36), did not differ from normative data. Spondylolysis is usually caused by a stress or fatigue fracture of the pars interarticularis, but there may be a genetic component, because it is more common in first-order reiatives. 158 Certain high-risk sports, including gymnastics, American football (especially in interior linemen), fast bowling in cricket, dance, and weight-lifting, have been associated with spondylolysis. Either condition can give rise to low back pain that may occur with activity and radiate down the posterior thigh. Occasionally, pain can be elicited by palpation of the involved vertebra, and a defect may be noted if a slip has occurred. Results of the neurologic examination are usually normal, but there may be tightness and spasm of the hamstring muscles. Having the child hyperextend the back and lift a leg often exacerbates the pain. 159 The diagnosis is usually clear from
691
oblique and lateral radiographs (Fig. 36-15), although bone scintigraphy may be needed in selected cases. 159 Treatment is usually conservative, with rest, analgesics, and a back corset or bracing to limit extension.155.157,158 Continuation in sports usually is allowable once the pain has subsided and range of motion has been regained. '60 If surgery is performed, the outcome appears to be excellent, with the great majority of athletes being able to return to their chosen sport. 161
Intervertebral Disk Herniation Disk herniation is rare in childhood, with estimates of the incidence in the Japanese population ranging from 1.69 per 100,000 in the 10- to 12-year-old age group, to 3.15 per 100,000 in the 13- to 15-year-old age group, and 9.63 per 100,000 in high school students aged 16 to 18 years. 162 These estimates are similar to those for the adult population in the United States. 163 Disk herniation accounted for 6% of 61 children younger than 16 years of age who were seen in one orthopedics clinic because of low back pain. l48 In another study of 1920 patients who underwent surgery for herniated disks, only 10 (0.5%) were children, and none was younger than 12 years of age. l64 Only 5 children aged 9 years or younger have been reported with disk herniation. 165 Boys may be slightly more frequently affected than girIS. 166-168 The vast majority of herniated disks are at L4-L5 and LS-S1. They are subligamental and usually posterolateral in position.l64 Although they often seem to be sports or injury related, the exact cause of the herniations is not clear. One study showed marked degenerative changes in 11 of 15 disks examined,'69 but another study of
• R.un 36-15 Spondylolysis affecting the pars interarticularis of L5 (alTOw) is visible in this oblique view radiograph of the lumbar spine of a child with low back pain.The posterior elements on the oblique view form the ·Scotty dog," and the pars interarticularis appears as the dog's neck.There is also spondylolisthesis with an anterior slip of L5 on S1.
692
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
10 adolescent disks failed to show such changes. 1M The onset of pain is usually insidious, with stiffness of the lower lumbar spine with or without radicular pain down the leg (which is the presenting complaint in some patients). Coughing, sneezing, and bending may aggravate the pain. Examination reveals limitation of forward bending and straight-leg raising and, in 25%, weakness of the plantar flexors. Plain radiographs are seldom helpful, although the disk space is narrowed in 200!o of patients and there may be a slight posterior calcification if there is a slipped apophysis; diagnosis is evident on MRI or computed tomographic scans. Conservative treatment with rest, analgesics, and physical therapy is successful in up to 80% of cases. 1S7 The long-term success of surgery in adolescents is quite goOd.167.168 However, in one study of 129 children younger than 18 years of age,170 at a mean follow-up of 12.4 years the outcome was excellent in only 40%, good in 47%, and poor in 13%, with reintervention required in 10%. (See also the discussion of diskitis in Chapter 28.)
Slipping Rib Syndrome The slipping rib syndrome is produced by trauma to the costal cartilages of the 8th to 10th ribs. 171 These cartilages attach to each other by fibrous tissue rather than to the sternum. Interruption of this fibrous tissue by trauma permits a rib to impinge on the adjacent rib, causing a click and sharp pain under the ribs. The symptoms may be precipitated in a number of ways, including forward flexion, deep breathing, and raising of the ipsilateral arm. The physician can reproduce the symptoms by hooking fingers under the inferior margins of the affected ribs and pulling anteriorly. Treatment consists of injection of local anesthetic or, if it does not heal, surgical excision of the subluxating cartilaginous rib tip.172
Costochondrltls Costochondritis, characterized by anterior chest wall pain that is reproduced by palpation of one or more of the costal cartilages, has been reported to be quite common in adolescents, constituting the reason for 4% of outpatient visits to one adolescent clinic. 173 It may result from trauma or idiopathic inflammation. 174 One, or occasionally, two or more costochondral junctions (usually the second or third) are painful and tender. The associated pain is usually acute and stabbing, often related to position or deep breathing. The syndrome can be self-limited or chronic and intermittent, lasting from a few months to a few years. Local anesthetic injections and anti-inflammatory medication may provide symptomatic relief. The term Tietze's syndrome is usually applied to a costochondritis in which, in addition to pain and tenderness of one or more costal cartilages, there is swelling overlying the affected costal cartilage.
Torticollis Torticollis, or wryneck, can accompany juvenile rheumatoid arthritis as a manifestation of cervical spine disease, develop due to a neurologic abnormality, or be caused by
an idiopathic shortening of a sternocleidomastoid muscle. Squints, if uncorrected surgically, may lead to a head tilt that can become permanent, with consequent asymmetry of the facial structures. Acute torticollis is transient if associated with trauma or cervical adenitis. The phenothiazine group of drugs and psychogenic disorders may also cause an acute torticollis. 17s Although acute torticollis has usually been considered to be caused by an atlantoaxial rotatory subluxation or atlantoaxial rotatory fixation, a careful imaging study of 33 children failed to demonstrate any such abnormalities. 176 Intensive physiotherapy177 and botulinum toxin178 should be administered before consideration of surgical release.
MISCELLANEOUS CONDITIONS
Growing Pains The term growing pains has significant positive and negative connotations. The positive aspect is that most parents accept it as a benign condition. Frequently, an immediate family member or family friend was diagnosed with growing pains that resolved without sequelae. The negative aspect is that this diagnosis has been too frequently applied to children who actually have a serious rheumatic or malignant disease. The term is a misnomer, because growth itself is not associated with the cause of the pain. The authors prefer the term benign nocturnal pains of childhood; however, no generally accepted alternative term exists. 179-181 The term should be restricted to identification of a fairly narrow spectrum of complaints (Table 36-9). Children who have unusual symptoms or abnormal findings on examination should not be diagnosed as having growing pains. Surveys of school-aged children have indicated that as many as 10% to 20% have had growing pains.179.181.182 Most growing pains occur in preschool- to school-aged children. The pain is sometimes crampy, usually localized to the lower extremities, and often located deep in the thigh, shin, or calf or behind the knee. Benign pain in the groin, back, or upper extremity is far less frequent. Growing pains may be precipitated by exercise and are usually relieved by massage. They occur in the evening or at night and often interrupt sleep. A few children have pain almost every night. Growing pains are never associ-
Ill'.
IABI E 3(, 'I
1_
of Childhood)
Age at onset Sex ratio Symptoms
Signs Investigations
GrowinlJ Pains (BenilJn NOt Illmal Pams
4 to 12 years Probably equal, slightly more girls in some series Deep aching, cramping pain in thigh or calf, usually in the evening or during the night; never present in the morning; bilateral; responds to massage and analgesia Physical examination results are normal Laboratory and radiographic studies (if done) have normal results
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
ated with a limp, and symptoms disappear by morning. Children with such pain have completely normal patterns of activity and normal physical examinations during and after the episode. Results of laboratory studies and radiography are normal. The pathophysiology of the pain is unknown. Long experience with such pains in many children, however, has proved that they do not portend serious illness. Successful management of growing pains includes education of the child and family about the benign nature of the problem. Gentle massage with or without analgesics is usually effective. In children with frequent attacks, administration of an evening dose of either acetaminophen or a nonsteroidal anti-inflammatory drug may prove preventive. Passive stretching can also be of benefit. 183
Pernlosls (Chilblains) Perniosis is a cold-induced condition in which there are painful or pruritic, erythematous or violaceous, papular or nodular lesions, usually on the fingers or toes. 184,185 A history of wet cold exposure and typical skin changes establish the diagnosis. It can recur within a few minutes after re-exposure to cold. Perniosis may occur as an idiopathic condition, or it may be associated with an underlying systemic disease such as systemic lupus erythematosus. The histology of idiopathic perniosis consists of a predominantly T-cell papillary and deep infiltrate with a perieccrine accumulation, associated with dermal edema and necrotic keratinocytes. l86 Histology can help differentiate idiopathic pernio from lupus, but immunohistochemistry appears not to be helpful. 186 If the condition is severe, treatment with nifedipine may be beneficial. Ca~alTunnel Synd~rne
Carpal tunnel syndrome is a rare condition in children. A revtew of 64 cases found that about 50% were secondary to various lysosomal storage diseases and 25% were idiopathic. 187 The children often had long-standing, rather nonspecific symptoms, such as complaints of poor manual dexterity, before diagnosis. By the time of diagnosis, clinical findings of weakness and thenar wasting are often marked. Therefore, although the disease is uncommon, it is important to have a low threshold of suspicion, particularly in children with skeletal abnormalities. A controlled study of the clinical diagnostic tests in adults with electrophysiologically proven carpal tunnel syndrome found that the wrist-flexion test (Phalen maneuver) was the most sensitive, whereas the nerve-percussion or compression test (Tinel's sign) was least sensitive but most specific. l88 The tourniquet test was not very accurate, and these researchers suggested that it not be used in clinical practice. Because the results of all of these clinical tests are often normal, electrophysiologic testing is essential in any child in whom the diagnosis is suspected. 187 Importantly electrophysiologic tests often show bilateral abnormalities in children even if only one hand is affected clinically. Open operative release is the only treatment documented to be effective in children.
693
Neuralgic Arnyot~phy Neuralgic amyotrophy (brachial plexus neuropathy) is a rare condition with an annual incidence rate of 1 to 2 per 100,000 individuals. 189 It is more common in adults, but it does occasionally affect older children. It almost always manifests as an acute onset of pain in a shoulder, followed by localized muscle wasting, without restricted passive range of motion. 190-194 In a study of 40 affected individuals, there was bilateral involvement in 17.5% of cases. 195 In about half of the cases, it was preceded by a fever and symptoms of an upper respiratory tract infection. Seven of the cases were recurrent; in a number of individuals, there was evidence of a mononeuritis multiplex, with involvement of cranial or other nerves outside the brachial plexus. Some cases are familial. l90 ,191 It can occur after vaccination. The overall prognosis is good, but recovery may take many months, and physiotherapy is needed to minimize the risk of permanent joint contracture. Brachial neuritis follows a similar pattern but is accompanied by paralysis of the affected part. In a similar fashion, lumbosacral plexus neuropathy can occur, leading to leg pain and paralysis. l96 Characteristic electromyographic findings of damage to both the nerve roots and the peripheral nerves usually confirm the diagnosis.
Eryth~rnelalgla
Erythromelalgia (erythro = red, mel = limb, algia = pain) is a rare condition that manifests as episodic burning pain with accompanying erythematous, warm, swollen hands or feet (or both) that is eased by cold and elevation. 197 Affected patients refuse to stop using ice or cold water for their painful extremities. Exercise or heat (even wearing socks) can precipitate an attack. This condition occurs in three forms: erythromelalgia associated with thrombocythemia, primary erythromelalgia, and secondary erythromelalgia. l98 The most common form is that associated with thrombocythemia, which can occur in isolation or in association with polycythemia vera or myelofibrosis and is sensitive to treatment with aspirin. Primary erythromelalgia begins in childhood or adolescence and can be familial (autosomal dominant). 199 It affects girls more often than boys. It is almost always symmetrical and is resistant to treatment. Secondary erythromelalgia is more common in adults; it is associated with an underlying condition other than one associated with a platelet dysfunction and therefore is not aspirin responsive, It usually responds to treatment of the underlying disorder. Additionally, an epidemic form was reported in southern China that was caused by a poxvirus. zOO Treatment includes avoidance of exposure to heat, elevation of the extremity, and application of cold during acute attacks. If aspirin is unsuccessful, a host of treatments have been tried, each with a few case reports of benefit, including nifedipine, verapamil, nicardipine, pergolide, bromocriptine, busulfan, propranolol, posterior pituitary extract, epinephrine, biofeedback, hypnosis, sympathectomy, clonazepam, nitroprusside, prostaglandin El' amputation, and stereotactic destruction of the ventroposteromedial and centromedian regions of the thalamus.
694
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
Restless Legs Syndrome Restless legs syndrome is a feeling of discomfort in, and an inability to keep from moving, the legs at night after resting or going to bed. It was reported in 17% of children attending two community-based general pediatric clinics. 20J It is relieved by activity and, therefore, frequently leads to insomnia. Periodic movements of the limb during sleep are common. These last less than 1 minute and rarely cause the child to awaken. Restless legs syndrome is more common in older adults but has been reported in children.202.203 The cause of this condition is unknown. It has been postulated that some children with "growing pains" really have the restless legs syndrome, but this would not seem to be true for the majority of such children. 204 Hyperactivity/attention deficit disorder may be more common in children with restless legs syndrome. 20J Recently, a study of a large French Canadian family mapped a locus conferring susceptibility to restless legs syndrome to chromosome 12q.205 Reducing caffeine intake is the first therapeutic step. A wide variety of medications, including pramipexole, clonidine, carbidopa/levodopa, and levodopa, have been reported to be helpful. 202.206
REFERENCES 1. Kirk JA, Ansell BA, Bywaters EGL: The hypermobility syndrome: musculoskeletal complaints associated with generalized hypermobility. Ann Rheum Dis 26: 419-425, 1967. 2. Jessee EF, Owen DSJ, Sagar KB: The benign hypermobile joint syndrome. Arthritis Rheum 23: 1053-1056, 1980. 3. Biro F, Gewanter HL, Baum J: The hypermobility syndrome. Pediatrics 72: 701-706, 1983. 4. Beighton P, Solomon L, Soskolne C: Articular mobility in an African population. Ann Rheum Dis 32: 413-418, 1973. 5. Carter C, Wilkinson J: Persistent joint laxity and congenital dislocation of the hip. J Bone Joint Surg Br 46: 40-45, 1964. 6. Gedalia A, Person DA, Brewer EJ, Giannini EH: Hypermobility of the joints in juvenile episodic arthritis/arthralgia. J Pediatr 107: 873-876, 1985. 7. Birrell FN, Adebajo AO, Hazleman BL, Silman AJ: High prevalence of joint laxity in West Africans. Br J RheumalOl 33: 56-59, 1994. 8. Cheng JC, Chan PS, Hui PW: Joint laxity in children. J Pediatr Orthop 11: 752-756, 1991. 9. Mikkelsson M, Salminen JJ, Kautiainen H: Joint hypermobility is not a contributing factor to musculoskeletal pain in pre-adolescents. J Rheumatol 23: 1963-1967, 1996. 10. Larsson L-G, Baum J, Mudholkar GS: Hypermobility: features and differential incidence between the sexes. Arthritis Rheum 30: 1426-1430, 1987. 11. Hall MG, Ferrell WH, Sturrock RD, et al: The effect of the hypermobility syndrome on knee joint proprioception. Br J Rheumatol 34: 121-125, 1995. 12. El-Garf AK, Mahmoud GA, Mahgoub EH: Hypermobility among Egyptian children: prevalence and features. J Rheumatol 25: 1003-1005, 1998. 13. Gedalia A, Press ]: Articular symptoms in hypermobile schoolchildren: a prospective study. J Pediatr 119: 944-946, 1991. 14. Acasuso-Dlas M, Collantes-Estevez E: Joint hypermobility in patients with fibromyalgia syndrome: Arthritis Care Res 11: 39-41, 1998. 15. Gedalia A, Press J, Klein M, Buskila 0: Joint hypermobility and fibromyalgia in schoolchildren. Ann Rheum Dis 52: 494-496, 1993. 16. Hud,on N, Fitzcharles MA, Cohen M, et al: The association of soft-tissue theumatism and hypermobility. Br J RheumalOl 37: 382-386, 1998. 17. Barton LM, Bird HA: Improving pain by the stabilization of hyperlax joints. J Orthop Rheumatol 9: 46-51, 1996. 18. Henady S, lvanans T: Hypermobile joints: a benign cause of transitory motor delay in infancy. Clin Pedialr 17: 790--795, 1978. 19. Davidovitch M, Tirosh E, Tal Y: The relationship between joint hypermobility and neurodevelopmental attributes in elementary school children. J Child Neurol 9: 417-419, 1994. 20. Lewkonia HM, Ansell BM: Articular hypermobility simulating chronic rheumatic disease. Arch Dis Child 58: 988-992, 1983. 21. Kaplinsky C, Kenet G, Seligsohn U, Rechavl G: Association between hyperllexibility of the thumb and an unexplained bleeding tendency: is it a rule of thumb? Br J Haematol 101: 260--263, 199B. 22. de Kort LM, Verhulst JA, Engelbert RH, et aI: Lower urinary tract dysfunction in children with generaliZed hypermobility of joints. J Uro1170: 1971-1974, 2003.
23. Everman DB, Robin NH: Hypermobility syndrome. Pediatr Rev 19: 111-117, 1998. 24. Klemp P, Stevens JE, Isaacs S: A hypermobility study in bailet dancers. J Rheumatol 11: 692-696, 1984. 25, Decoster LC, Vailas JC, Undsay RH, Williams GR: Prevalence and features of joint hypermobility among adolescent athletes. Arch Pediatr Adolesc Med 151: 989-992, 1997, 26. McCormack M, Briggs J, Hakim A, Grahame R: Joint laxity and the benign joint hypermobility in student and professional bailet dancers. J Rheumatol 31: 173-178, 2004. 27. Cowan ON, Jones BH, Frykman PN: Lower limb morphology and risk of overuse injury among male infantry trainees. Med Sci Sports Exerc 28: 945--952, 1996. 28. Adair SM, Hecht C: Association of generalized joint hypermobility with history, signs, and symptoms of temporomandibular joint dysfunction in children. Pediatr Dent 15: 323-326, 1993. 29. Harinstein 0, Buckingham RB, Braun T. et al: Systemic joint laXity (the hypermobile joint syndrome) Is associated with temporomandibular joint dysfunction. Arthritis Rheum 31: 1259-1264, 198B. 30, Westling L, Mattiasson A: General joint hypermobility and temporomandibular joint derangement in adolescents. Ann Rheum Dis 51: 87-90, 1992. 31. Gazit Y, Nahir AM, Grahame R, Jacob G: Dysautonomia in the joint hypermobility syndrome. Am J Med 115: 33-40, 2003. 32. al-Rawi Z, Nessan AH: Joint hypermobility in patients with chondromalacia patellae. Br J Rheumatol 36: 1324-1327, 1997. 33. Larsson LG, Mudholkar GS, Baum], Srivastava OK: Benefits and liabilities of hypermobility in the back pain disorders of industrial workers. J Intern Med 238: 461-467, 1995. 34. Klemp P: Hypermobility. Ann Rheum Dis 56: 573-575, 1997. 35. Mishra MB, Ryan P, Atkinson P: Extra-articular features of benign joint hypermobility syndrome. Br J Rheumatol 35: 861--B66, 1996. 36. Dolan AL, Hart OJ, Doyle DV, et al: The relationship of joint hypermobility, bone mineral density, and osteoarthritis in the general population: the Chlngford Study. J Rheumatol 30: 799-803, 2003. 37. Unger OL: Does knuckle cracking lead to arthritis of the fingers? Arthritis Rheum 41: 949-950, 1998. 38. Marks JS, Sharp J, Brear SC, Edwards JD: Normal joint mobility In mitral valve prolapse. Ann Rheum Dis 42: 54-55, 1983. 39. Grahame R, Edwards JC, Pitcher 0, et al: A clinical and echocardiographic study of patients with the hypermobility syndrome. Ann Rheum Dis 40: 541-546, 1981. 40. Staheli LT, Chew DE, Corbett M: The longitudinal arch: a survey of eight hundred and eighty-two feet in normal children and adults. J Bone Joint Surg Am 69: 426-428, 1987. 41. Hogan MT, Staheli LT: Arch height and lower limb pain: an adult civilian study. Foot Ankle Int 23: 43-47, 2003. 42. Mosca VS: Flexible flatfoot and skewfoot. Instr Course Lect 45: 347-354, 1996. 43. Harris RI, Beath T: Hypermobile llat-foot with short tendoachilles. J Bone Joint Surg Am 30: 116-138, 1948. 44. Wenger DR, Mauldin 0, Speck G, et al: Corrective shoes and inserts as treatment for llexible flatfoot in infants and children. J Bone Joint Surg Am 71: 800--810, 1989. 45. Miller CO, Laskowski ER, Suman VJ: Effect of corrrective rear-foot orthotic devices on ground reaction forces during ambulation. Mayo C1in Proc 71: 757-762, 1996, 46. Sullivan ]A: The child's foot. In Mortis&)' RT, Weinstein SI (eds): Lovell and Winter's Pediatric OrthopaedicS. Philadelphia, Lippincott-Raven, 1996, pp 1077-1135. 47. Sneyers q, Lysens R, Feys H, Andries R: Influence of malalignment of feet on the plantar pressure pattern in running. Foot Ankle Int 16: 624-632, 1995. 48. Rudzki S]: InjUries in Australian army recruits. Part Ill: the accuracy of a pretraining orthopedic screen in predicting ultimate injury outcome. Mil Med 162: 481-483, 1997. 49. GlIadi M, Milgrom C, Stein M, et al: The low arch, a protective factor in stress fractures. Orthop Rev 14: 81-84, 1985. 50. Mosier KM, Asher M: Tarsal coalitions and peroneal spastic flat foot: a review. J Bone Joint Surg Am 66: 976-984, 1984. 51. Loudon JK, Goist HL, Loudon KL: Genu recurvatum syndrome. J Orthop Sports Phys Ther 27: 361-367, 1998. 52. Thabit GO, Micheli L]: Patellofemoral pain in the pediatric patient. .J Orthop Sports Phys Ther 27: 567-585, 1992. 53. Tjernstrom B, Rehnberg L: Back pain and arthralgia before and after lengthening: 75 patients questioned after 6 0-11) years. Acta Orthop Scand 65: 328-332, 1994. 54. Bowyer S: Hip contractures as the presenting sign in children With HLA-B27 arthritis. J Rheumatol 22: 165-167, 1995. 55. Glover MT, Lake BO, Atherton OJ: Infantile systemic hyalinosis: newly recognized disorder of collagen? Pediatrics 87: 228-234, 1991. 56. Verma UN, Misra R, Radhakrisnan S, et al: A syndrome of fibrosing pleuritis. pericarditis, and synovitis with infantile contractures of fingers and toes in 2 sisters: "familial fibrosing serositis." J Rheumatol 22: 2349-2355, 1995. 57. Yates CK, Grana WA: Patellofemoral pain in children, Clin Orthop 255: 36-43, 1990. 58. O'Neill DB, Micheli LJ, Warner JP: Patellofemoral stress: a prospective analysis of exercise treatment in adolescents and adults. Am J Sports Med 20: 151-156, 1992.
C HAP T E R
36 NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
59. Ra~in EL: Chondromalacia of the patella. Bull Rheum Dis 34: 1--6, 1984. 60. Fairbank ]C, Pynsent PB, van poortyliet ]A, Phillips H: Mechanical factors in the incidence of knee pain in adolescents and young adults. ] Bone Joint Surg Br 66: 685--693, 1984. 61. Outerbridge RE: The etiology of chondromalacia patellae. J Bone Joint Surg 43: 752, 1961. 62. Ingall J: "Chondromalacia patellae": patellar malalignment syndrome. Orthop Clin North Am 10: 117-127, 1979. 63. Goodfellow], Hungerford DS, Woods C: Patello-femoral joint mechanics and Nthology: 2. Chondromalacia patellae.] Bone]oint Surg Br 58: 291-299, 1976. 64. Reid GD, Glasgow M, Gordon DA, Wright TA: Pathological plicae of the kmee mistaken for arthritis.] Rheumatol 7: 573-576, 1980. 65. De Mot P, Brys P, Samson I: Non perforated septum supra-patellaris mimicking a soft tissue tumour. ]BR-BTR 86: 262-264, 2003. 66. Peck DM: Apophyseal injuries in the young athlete [reviewl. Am Fam Physician 51: 1891-1895, 1997. 67. Ohta-Fukushima M, Mutoh Y, Takasugi S, et al: Characteristics of stress fractures in young athletes under 20 years. ] Sports Med Phys Fitness 42: 198-206. 2002. 68. Schwend RM, Werth C, Johnston A: Femur shaft fractures in toddlers and young children: rarely from child abuse. ] Pediatr Orthop 20: 475-481, 2000. 69. Micheli L]: The traction apophysitises. Clin Sports Med 6: 389-404, 1987. 70. Taussig G, Aufaure P, Pilliard D: ApophySitis of the iliac crest: a little known cause of low back pain and pelvic pain in children. Rev Chir Orthop Reparatrice Appar Mot 79: 58--61, 1993. 71. Schlatter C: Verlezungen des schnabelfbrmigen forsatzes der oberen tibiaepiphyse. Beitr Klien Chir 38: 874, 1903. n. Osgood RB: Lesions of the tibial tubercle occurring during adolescence. aoston Med] 8: 114, 1903. 73. Ogden ]A. Southwick Wo: Osgood-Schlatter's disease and tibial tuberosity development. Clin Orthop 116: 180-189, 1976. 74. Kujala UM, Kvist M, Heinonen 0: Osgood-Schlatter's disease in adolescent alhletes. retrospective study of incidence and duration. Am] Sports Med 13: 236-241, 1985. 75. l;anning P, Heikkinen E: Ultrasonic features of the Osgood-Schlatter lesion. ] Pediatr Orthop 11: 538-540, 1991. 76. Sherry DD. Petty RE, Tredwell S, Schroeder ML: Histocompatibility antigens in Osgood-Schlatter disease. ] Pediatr Orthop 5: 302-305, 1985. 77. Krause BL. Williams P, Caterall A: Natural history of Osgood-Schlaner disease. J Pediatr Orthop 10: 65--68, 1990. 78. Gardiner ]S, Mcinerney VK, Avella DG, Valdez NA: Pediatric update: 13. Injuries to the inferior pole of the patella in children. Orthop Rev 19: 643--649, 1990. 79. Medlar RC, Lyne ED: Sinding-Larsen-]ohansson disease: its etiology and natural history. J Bone Joint Surg Am 60: 1113-1116, 1978. 80. Wberson A, Lieherson S, Mendes DG, et al: Remodeling of the calcaneus apophysis in the growing child. J Pediatr Orthop 4B: 74-79, 1995. 81. Iilarnett LS: Little league shoulder syndrome: proximal humeral epiphyseolysis. J Bone Joint Surg Am 67: 495-496, 1985. 82. Jawish R, Rlgault P, Padovani JP, et al: Osteochondritis dissecans of the humeral capitellum in children. Eur] Pediatr Surg 3: 97-100, 1993. 83. ~:han D. Aldridge M], Maffulli N, Davies AM: Chronic stress injuries of the elbow in young gymnasts. Br J Radiol 64: 1113-1118, 1991. 84. IMaffulJi N, Chan D, Aldridge M]: Derangement of the articular surfaces of the elbow in young gymnasts. J Pediatr Orthop 12: 344-350, 1992. 85. Andrish]T, Bergfeld ]A, Walheim J: A prospective study on the management of shin splints. J Bone Joint Surg Am 56: 1697-1700, 1974. 86. Michael RH, Holder LE: The soleus syndrome: a caUSe of medial tibial stress (shin splints). Am J Sports Med 13: 87-94, 1985. 87. Sheon RP, Moskowitz RW, Goldberg VM: Soft tissue rheumatic pain: recognition, management and prevention. Philadelphia, Lea & Febiger, 1982. 88. IJrower AC: The osteochondroses. Orthop Clin North Am 14: 99-117, 1983. 89. Resnick D: The osteochondroses. In Resnick D, Niwayama G (eds): Diagnosis of Bone and Joint Disorders. Philadelphia, WB Saunders, 1981, p 2874. 90. Legg AT: An obscure affection of the hip joint. Boston Med SurgJ 162: 202,190. 91. Calve]: Sur une forme particuliere de coxalgie greffee sur des deformations caracteristiques de l'extremite superieure du femur. Rev Chir Orthop 42: 54. 1910.• 92. Peltier LF: The classic: concerning arthritis deformans juvenilis. Professor (,eorg C. Perthes. Clin Orthop 158: 5-9, 1981. 93. Weinstein SL: Natural history and treatment outcomes of childhood hip disorders [reviewl. Clin Orthop (344): 227-242, 1997. 94. Eckerwall G, Wingstrand H, Hagglund G, Karlberg J: Growth in 110 children with Legg-Calve-Perthes' disease: a longitudinal infancy childhood puberty growth model study. ] Pediatr Orthop 5B: 181-184, 1996. 95. Glueck CJ, Brandt G, Gruppo R, et al: Resistance to activated protein C and Legg-Perthes disease. Clin Orthop (38): 139-152, 1997. 96. Posan E, Szepesi K, Gaspar L, et al: Thrombotic and fibrinolytic alterations in the aseptic necrosis of femoral head. Blood Coagul Fibrinolysis 14: 243--248, 2003. 97. Hresko MT, McDougall PA, Gorlin ]B, et al: Prospective reevaluation of the association between thrombotic diathesis and Legg-Perthes disease. ] Bone Joint Surg Am 84: 1613-1618, 2002. 98. Dilley A, Hooper WC, Austin H, et al: The beta fibrinogen gene G-455-A polymorphism is a risk factor for Legg-Perthes disease. ] Thromb Haemost 1: 2317-2321, 2003.
695
99. Gaughan DM, Mofenson LM, Hughes MD, et al: Osteonecrosis of the hip (Legg-Calve-Perthes disease) in human immunodeficiency virus-infected children. PediatriCs 109: E74, 2002. 100. Lecuire F: The long-term outcome of primary osteochondritis of the hip (Legg-Calve-Perthes' disease). ] Bone Joint Surg Br 84: 636--640, 2002. 101. Lahdes-Vasama TT, Marttinen E], Merikanto ]E: Outcome of Perthes' disease in unselected patients after femoral varus osteotomy and splintage. J Pediatri Orthop 6B: 229-234, 1997. 102. Wang L, Bowen ]R, Puniak MA, et al: An evaluation of various methods of treatment for Legg-Calve-Perthes disease. Clin Orthop (314): 225-233, 1995. 103. Joseph B, Varghese G, Mulpuri K, et al: Natural evolution of Perthes disease: a study of 610 children under 12 years of age at disease onset. ] Pediatr Orthop 23: 590--600, 2003. 104. Sorenson KH: Scheuermann's Juvenile Kyphosis. Copenhagen, Munksgaard, 1964. 105. Tachdjian MO. Pediatric Orthopedics. Philadelphia, WB Saunders, 1972. 106. Moe ]H, Winter RB, Bradford DS, et al. Juvenile Kyphosis, Scoliosis and Other Spinal Deformities. Philadelphia, WB Saunders, 1978. 107. Poolman RW, Been HD, Ubags LH. Clinical outcome and radiographic results after operative treatment of Scheuermann's disease. Eur Spine] 11: 561-569, 2002. 108. Soo CL, Noble PC, Esses SI: Scheuermann kyphosis: long-term follow-up. Spine] 2: 49-56, 2002. 109. Giannestras NJ: Other problems of the forepart of the foot. In Giannestras N] (ed): Foot Disorders. Philadelphia, Lea & Febiger, 1973, p 410. 110. Borges ]L, Guille ]T, Bowen ]R: Kohler's bone disease of the tarsal navicular. J Pediatr Orthop 15: 596--598, 1995. 111. Braddock GTF: Experimental epiphyseal injury and Freiberg'S disease. ] Bone Joint Surg Br 41: 154, 1959. 112. Stanley D, Betts RP, Rowley 01, Smith TW: Assessment of etiologic factors in the development of Freiberg's disease.] Foot Surg 29: 444-447, 1990. 113. Molloy MG, Hamilton EB: Thiemann's disease. Rheumatol Rehabil 17: 179-180, 1978. 114. Gewanter H, BaumJ: Thiemann's disease.] Rheumatol 12:150-153, 1985. 115. Allison AC, Blumberg BS: Familial osteoarthropathy of the fingers. ] Bone Joint Surg Br 40: 538, 1958. 116. Stougaard J: Familial occurrence of osteochondritis dissecans. ] Bone Joint Surg Br 46: 542, 1964. 117. Robinson RP, Franck WA, Carey E], Goldberg EB: Familial polyarticular osteochondritis dissecans masquerading as juvenile rheumatoid arthritis. ] Rheumatol 5: 190-194, 1978. 118. Bohndorf K: Osteochondritis (osteochondrosis) dissecans: a review and new MRl classification [reviewl. Eur Radiol 8: 103--112, 1998. 119. Koch S, Kampen WU, Laprell H: Cartilage and bone morphology in osteochondritis dissecans. Knee Surg, Sports Traumatol, Arthrosc 5: 42-45, 1997. 120. Yoshida S, Ikata T, Takai H, et al: Osteochondritis dissecans of the femoral condyle in the growth stage. Clin Orthop (346): 162-170, 1998. 121. Nelson DW, DiPaola], Colville M, Schmidgall ]:. Osteochondritis dissecans of the talus and knee: prospective comparison of MR and arthroscopic classifications. ] Comput Assist Tomogr 14: 804-808, 1990. 122. De Smet AA, Ilahi OA, Graf BK:. Untreated osteochondritis dissecans of the femoral condyles: prediction of patient outcome using radiographic and MR findings. Skeletal Radiol 26: 463-467, 1997. 123. Poussa M, Schlenzka D, Yrjonen T:. Body mass index and slipped capital femoral epiphysis.] Pediatr Orthop B 12: 369-371, 2003. 124. Stott S, Bidwell T:. Epidemiology of slipped capital femoral epiphysis in a population with a high proportion of New Zealand Maori and Pacific children. N Z MedJ 116: U647, 2003. 125. Loder RT, Mehbod AA, Meyer C, Meisterling M:. Acetabular depth and race in young adults: a potential explanation of the differences in the prevalence of slipped capital femoral epiphysis between different racial groups? ] Pediatr Orthop 2003; 23: 699-702, 2003. 126. Lalaji A, Umans H, Schneider R, et al: MRl features of confirmed "pre-slip" capital femoral epiphysis: a report of two cases. Skeletal Radiol31: 362-365, 2002. 127. Loder RT:. Slipped capital femoral epiphysis in children. Curr Opin Pediatr 7: 95-97, 1995. 128. Mandell GA, Keret D, Harcke HT, Bowen ]R:. Chondrolysis: detection by bone scintigraphy. ] Pediatr Orthop 1992; 12: 8Q--85, 1992. 129. Spero CR, Masciale JP, Tornetta P3d, et al: Slipped capital femoral epiphysis in black children: incidence of chondrolysis. J Pediatr Orthop 12: 444-448, 1992. 130. Carney BT, Weinstein SL: Natural history of untreated chronic slipped capital femoral epiphysis. Clin Orthop (322): 43-47, 1996. 131. Duncan JW, Nasca R, Schrantz J: Idiopathic chondrolysis of the hip. J Bone Joint Surg Am 61: 1024-1028, 1979. 132. Bleck EE: Idiopathic chondrolysis of the hip. ] Bone Joint Surg Am 65: 1266--1275, 1983. 133. del Couz GA, Fernandez PL, Gonzalez MP, et al: Idiopathic chondrolysis of the hip: long-term evolution. J Pediatr Orthop 19: 449-454, 1999. 134. Koot MF, Berendsen HA, vad der Hoeven H, et al: Three adolescenl~ with hip pain caused by idiopathic chondrolysis. Ned Tijdschr Geneeskd 137: 86--90, 1993. 135. Brown], Melinkovich P: Schonlein-Henoch purpura misdiagnosed as suspected child abuse: a case report and literature review.]AMA 256: 617--618. 1986. 136. Carrera GF, Kozin F, McCarty DJ: Arthritis after frostbite injury in children. Arthritis Rl1eum 22: 1082-1087, 1979.
696
C HAP T E R
36
NONINFLAMMATORY MUSCULOSKELETAL PAIN CONDITIONS
137. Dreyfuss ]R, Glimcher MJ Epiphyseal injury following frostbite. N Engl ] Med 253: 1065, 1955. 138. Brown FE, Spiegel PK, Boyle WE]: Digital deformity: an effect of frostbite in children. Pediatrics 71: 955-959, 1983. 139. Derwin KA, Glover RA, Wojtys EM: Nociceptive role of substance-P in the knee joint of a patient with congenital insensitivity to pain. ,T Pediatr Orthop 14: 258-262, 1994. 140. Indo Y, Tsuruta M, Hayashida Y, et al: Mutations in the TRKAINGF receptor gene in patients with congenital insensitivity to pain with anhidrosis. Nat Genet 13: 485-488, 1996. 141. Nellhaus G: Neurogenic arthropathies (Charcot's joints) in children: description of a case traced to occult spinal dysraphism. Clin Pediatr 14: 647-653, 1975. 142. Leboeuf-Yde C, Kyvik KO: At what age does low back pain become a common problem? A study of 29,424 individuals aged 12-41 years. Spine 23: 228-234, 1998. 143. Balague F, Skovron ML, Nordin M, et al: Low back pain in school children: a study of familial and psychological factors. Spine 20: 1265-1270, 1995. 144. Kristjansdottir G: Prevalence of self-reported back pain in school children: a study of sociodemographic differences. Eur] Pediatr 155: 984--986, 1996. 145. Kujala UM, Taimela S, Erkintalo M, et al: Low-back pain in adolescent athletes. Med Sci Sports Exerc 28: 165-170, 1996. 146. Ramirez N, Johnston CE, Browne RH: The prevalence of back pain in children who have idiopathic scoliosis.] Bone Joint Surg Am 79: 364-368, 1997. 147. Volinn E: The epidemiology of low back pain in the rest of the world: areview of surveys in low- and middle-income countries. Spine 22: 1747-1754, 1997. 148. Turner PG, Green ]H, Galasko CS: Back pain in childhood. Spine 14: 812-814, 1989. 149. Malleson P, Clinch]: Pain syndromes in children. Curr Opin Rheumatol 15: 572-580, 2003. 150. Hayden lA, Mior SA, Verhoef M]: Evaluation of chiropractic management of pediatric patients with low back pain: a prospective cohort study. ] Manipulative Physiol Ther 26: 1-8, 2003. 151. Szpalski M, Gunzburg R, Balague F, et al: A 2-year prospective longitudinal study on low back pain in primary school children. Eur Spine] 11: 45~64, 2002. 152. Cardon GM, De Clercq DL, De Bourdeaudhuij 1M: Back education efficacy in elementary schoolchildren: a I-year follow-up study. Spine 27: 299-305, 2002. 153. Rowe BH, Dulberg CS, Peterson RG, et al: Characteristics of children presenting with chest pain to a pediatric emergency department. Can Med Assoc] 143: 388-394, 1990. 154. Gumbiner CH: Precordial catch syndrome. South Med] 96: 38-41, 2003. 155. Morita T, 1kata T, Katoh S, Miyake R: Lumbar spondylolysis in children and adolescents.] Bone Joint Surg Br 77: 620-625, 1995. 156. Beutler WI, Fredrickson BE, Murtland A, et al: The natural history of spondylolysis and spondylolisthesis: 45-year follow-up evaluation. Spine 28: 1027-1035, 2003. 157. Sponseller PD: Evaluating the child with back pain. Am Fam Physician 54: 1933-1941, 1996. 158. Payne WK3, OgilVie JW: Back pain in children and adolescents. Pediatr Clin North Am 3: 899-917, 1996. 159. Ralston S, Weir M: Suspecting lumbar spondylolysis in adolescent low back pain. Clin Pediatr 37: 287-293, 1998. 160. Muschik M, Hahnel H, Robinson PN, et al: Competitive sports and the progression of spondylolisthesis. ] Pediatr Orthop 16: 364-369, 1996. 161. Debnath UK, Freeman B], Gregory P, et al: Clinical outcome and return to sport after the surgical treatment of spondylolysis in young atWetes. ] Bone Joint Surg Br 85: 244-249, 2003. 162. Matsui H, Terahata N, Tsuji H, et al: Familial predisposition and clustering for juvenile lumbar disc herniation. Spine 17: 1323-1328, 1992. 163. Bruske-Hohlfeld I, Merritt ]L, Onofrio BM, et al. Incidence of lumbar disc surgery: a population-based study in Olmsted County, Minnesota. Spine 1990; 15: 31-35, 1990. 164. Villarejo-ortega F], Torres Campa-Santamarina 1M, Bencosme-Abinader ]A, et al: [Lumbar disc disease in adolescents,] Rev Neurol 36: 514-517, 2003. 165. Martinez-Lage ]F, Fernandez CV, Lopez F, Poza M: Lumbar disc herniation in early childhood: case report and literature review. Childs Nerv Syst 19: 258-260, 2003. 166. Nelson CL, ]anecki C], Gildenberg PL, Saya G: Disk protrusions in the young. Clin Orthop 88: 142-150, 1972. 167. Ishihara H, Matsui H, Hirano N, Tsuji H: Lumbar intervertebral disc herniation in children less than 16 years of age: long-term follow-up study of surgically managed cases. Spine 22: 2044-2049, 1997. 168. Papagelopoulos P], Shaughnessy WJ, Ebersold M], et al: Long-term outcome of lumbar discectomy in children and adolescents sixteen years of age or younger. Spine 22: 2044-2049, 2004. 169. Lee ,TV, Ernestus RI, Schroder R, K1ug N: Histological study of lumbar intervertebral disc herniation in adolescents. Acta Neurochir (Wien) 142: 1107-1110,2000.
170. ParL.ini P, Di Silvestre M, Greggi T, et al: Lumbar disc excision in children and adolescents. Spine 26: 1997-2000, 2001. 171. Taubman B, Vetter VL: Slipping rib syndrome as a cause of chest pain in children. Clin Pediatr 35: 403-405, 1996. 172. Porter GE: Slipping rib syndrome: an infrequently recognized entity in children. A report of three cases and review of the literature. Pediatrics 76: 810-813, 1985. 173. Brown RT: Costochondritis in adolescents.] Adolesc Health Care 1: 198-201. 1981. 174. Calabro ]], Marshesano ]M: Tietze's syndrome: report of a case with juvenile onset. ] Pediatr 68: 985, 1966. 175. Bolthauser E: Differential diagnosis of torticollis in childhood. Schweiz Med Wochenschr 106: 1261, 1976. 176. Hicazi A, Acaroglu E, Alanay A, et al: Atlantoaxial rotatory fixation-subluxation revisited: a computed tomographic analysis of acute torticollis in pediatric patients. Spine 27: 2771-2775, 2002. 177. Smith DL, DeMario MC: Spasmodic torticollis: a case report and review of therapies.] Am Board Fam Pract 9: 435-441, 1996. 178. Lew MF, Adornato BT, Duane DO, et al: Botulinum toxin type B: a doubleblind, placebo-controlled, safety and efficacy study in cervical dystonia. neurology 49: 701-707, 1997. 179. Peterson H: Growing pains. Pediatr Clin North Am 33: 1365-1372, 1986. 180. Brady M, Grey M: Growing pains: a myth or reality. I Pediatr Health Care .~: 219-220, 1989. 181. Oster], Nielsen A: Growing pains: a clinical investigation of a school population. Acta Pediatr Scand 61: 329-334, 1972. 182. Apley]: One child. In Apley], Ounsted C (edsl: One Child. Philadelphia, ]B Lippincott, 1982, pp 23-47. 183. Baxter MP, Dulberg C: "Growing pains" in childhood: a proposal for treatment.] Pediatr Olthop 8: 402-406, 1988. 184. Goette DK: Chilblains (perniosis). J Am Acad Dermatol 23: 257-262, 1990. 185. Crowson AN, Magro CM: Idiopathic perniosis and its mimics: a clinical and hIstological study of 38 cases. Hum Patho! 28: 478-484, 1997. 186. Cribier S, Djeridi N, Peltre B, Grosshans E: A histologic and immunohistochemical study of chilblains. I Am Acad Dermatol 45: 924-929, 2001. 187. Lamberti PM, Light TR: Carpal tunnel syndrome in children. Hand Clinics 18: 331-337, 2002. 188. Gellman H, Gelberman RH, Tan AM, Botte M]: Carpal tunnel syndrome: an evaluation of the provocative diagnostic tests. ] Bone ,Toint Surg Am 68: 735-737, 1986. 189. Rubin 01: Neuralgic amyotrophy: clinical features and diagnostic evaluation. Neurology 7: 350-356, 2001. 190. Lane R], Dewar ]A: Bilateral aneuralgic amyotrophy. Br Med] 6117: 895, 1978. 191. Dunn HG, Daube ]R, Gomez MR: Heredofamilial brachial plexus neuropathy (hereditary neuralgic amyotrophy with brachial predilection) in childhood. Dev Med Child Neurol 20: 28-46, 1978. 192. Shaywitz BA: Brachial plexus neuropathy in childhood. ] Pediatr 86: 913-915, 1975. 193. Bale IF ]r, Thompson lA, Petajan IH, Ziter FA: Childhood brachial plexus neuropathy.] Pediatr 95: 741-742, 1979. 194. Zeharia A, Mukamel M, Frishberg Y, et al: Benign plexus neuropathy in children. I Pediatr 116: 276-278, 1990. 195. Cruz-Martinez A, Barrio M, Arpa]: Neuralgic amyotrophy: variable expression in 40 patients. ] Peripher Nerv Syst 7: 198-204, 2002. 196. van A1fen N, van Engelen BG: Lumbosacral plexus neuropathy: a case report and review of the literature. Clin Neurol Neurosurg 99: 138-141, 1997. 197. Kurzrock R, Cohen PR: Erythromelalgia: review of clinical characteristics and pathophysioiogy. Am] Med 91: 416-422, 1991. 198. Drenth ]P, Michials]]: Three types of erythromelalgia. Br Med] 301: 454-455, 1990. 199. Finley WH, Lindsey]R], Fine ]0, et al: Autosomal dominant erythromelalgia. Am] Med Genet 42: 310-315, 1992. 200. Zheng ZM, Zhang,TH, Hu ]M, et al: Poxviruses isolated from epidemic erythromelalgia in China. Lancet 8580: 296, 1988. 201. Chervin RD, Archbold KH, Dillon ]E, et al: Associations between symptoms of inattention, hyperactivity, restless legs, and periodic leg movements. Sleep 25: 213-218, 2002. 202. Walters AS, Picchietti DL, Ehrenberg BL, Wagner ML: Restless legs syndrome in childhood and adolescence. Pediatr Neurol 11: 241-245, 1994. 203. Walters AS, Hickey K, Maltzrnan ], et al: A questionnaire study of 138 patients with restless legs syndrome: the "Night-Walkers" survey. Neurology 46: 92-95, 1996. 204. Walters AS: Is there a subpopulation of children with growing pains who really have restless legs syndrome? A review of the literature. Sleep Med 3: 93-98, 2002. 205. Desautels A, Turecki G, Montplaisir ], et al: Identification of a major susceptibility locus for restless legs syndrome on chromosome 12q. Am I Hum Genet 69: 1266-1270, 2001. 206. Lin S-C, Kaplan], Burger CD: Effect of paramipexole in treatment of resistant restless legs syndrome. Mayo Clin Proc 73: 497-500, 1994.
37
( HAP T E R
PAIN AND THE PAIN AMPLIFICATION SYNDROMES David D. Sherry and Peter N. Malleson
~ ANATOMY, PHYSIOLOGY, AND MEASUREMENT OF PAIN This chapter discusses a wide variety of musculoskeletal conditions that are not primarily caused by inflammation. Because pain is often the main symptom in these conditions, as it is for the so-called amplified pain syndromes, it is important to have some understanding of the nociceptor system and how pain is generated and perceived by the child. Pain has been defined by the Committee for Taxonomy of the International Association for the Snldy of Pain as "an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage."] Pain therefore is a subjective interpretation of a noxious or apparently noxious stimulus. For most acute pain, the noxious stimulus is ohvious; however, for many children with chronic pain syndromes, the origin of the pain is much less clear. What is certain is that pain, whether acute or chronic, is influenced by multiple modulating factors and occurs at all levels of the nervous system. To understand a child's pain, one must be aware of the me<,:hanisms underlying noxious transmission and modulation, in addition to those factors that influence how a child perceives pain. The child's and the family's concepts of pain affect pain behaviors and coping strategies. Different children react differently to pain with inherently different degrees of regulation of the focus of attention and arousal to pain. 1,3 Some of these differences are almost certainly genetically determined, but it is increasingly recognized that noxious events in the neonatal period and early infancy may play important roles in determining how individuals respond to pain later in life.',s An overview of the pain pathways from peripheral sensory nerves to the cerebral cortex is shown in Figure 37-1.
NOOCEPTIVE MECHANISMS Peripheral Nociceptor System (Nociceptors and Afferent Nerve Fibers) Nociceptors are free nerve endings. In the joints, they are present in the capsule, adipose tissues, ligaments,
menisci, and periosteum. They are not found in articular cartilage, and their presence in synovium is controversial. 6 In muscle, the nociceptors are typically located in the walls of arterioles and surrounding connective tissue. 7 Nociceptors have high thresholds that respond to noxious thermal, chemical, or mechanical forces. Some nociceptors are modality specific (specific to a single type of stimulus, such as mechanical or thermal); others are polymodal. Generally, nociceptors have no spontaneous activity; they are activated only by intense, potentially damaging stimuli. s Noxious impulses are transmitted either by thinly myelinated A8 fibers or by nonmyelinated, slow-conducting C fibers. The A8 fibers are responsible for the well-localized first pain sensation that occurs immediately after injury, whereas C fibers transmit the more poorly localized, diffuse, burning second pain. 9 These afferent nociceptive fibers have their cell bodies in the dorsal root ganglia and terminate over several spinal segments in the dorsal horn of the spinal cord6 (Fig. 37-2). A number of neurotransmitters, including the amino acids glutamate and aspartate and the neuropeptides substance P and calcitonin gene-related peptide, are involved in the synaptic transmission of nociceptive signals from the primary afferent pain fibers to the spinal cord neurons. lO At the same time, the neuropeptides, which are produced in the dorsal root ganglia, are also released into the joint and muscle and thereby modulate the afferent response (see later discussion).6.7.11
Spinal Cord and Central Nervous System The receptive fields of dorsal horn neurons receiving input from afferent fibers from the joint have converging input from adjacent structures such as muscle and skin, and sometimes input from skin distant to the joint12 (see Fig. 37-2). The facts that pain signals from the joint occur over several spinal segments and that the spinal neurons have receptive fields from sites other than joints help explain the relatively poorly localized quality of joint and muscle pain. Dorsal horn neurons responSible for transmitting noxious stimuli from the joint and muscles can be classified as either nociceptive/specific or wide-range/dynamic. The wide-range/dynamic neurons respond to both innocuous and noxious stimuli but fire at higher rates in response
697
698
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
~
/s:=E
1;-.'
•
Pit.
"Stress" system
L'\. /Aaph,,\
magnus
from large-diameter non-nociceptive afferents (AP) are inhibitory. The summation of excitatory and inhibitory impulses determines whether a nociceptive impulse is projected to the brain. Noxious stimuli ascend via the spinothalamic, spinoreticular, and spinomesencephalic tracts, which cross to the contralateral side of the spinal cord close to their origin and ascend to terminate within the thalamus, reticular formation, and periaqueductal gray area in the midbrain 13 (see Fig. 37-1). The sensory cortex is responsible for integrating the sensory message with ongoing cognitive functions and for organizing conscious behavior in response to the stimulus. If the thalamofrontal tracts are severed, patients can accurately detect and localize pain, but the pain loses its aversive and motivational components. Studies using positron emission tomography suggest that several specific areas of the sensory cortex have different functional roles in the perception and interpretation of pain. 14-16 Noxious impulses reach the locus ceruleus, a pontine nucleus that is one of the main autonomic (sympathetic) centers that regulates arousal. The locus ceruleus/norepinephrine system is closely related to the hypothalamicpituitary-adrenal axis; together, these can be considered part of a dedicated system responsible for reestablishing homeostasis after all forms of stress, including those associated with pain. 17
MODULATING (SENSITIZING AND INHIBITING) MECHANISMS • Figure 37-1 Diagram showing an overview of pain pathways from the peripheral sensory nerves to the cerebral cortex.
Peripheral Nervous System
to noxious inputs. There are also interneurons that are responsible for modulating the ascending progression of nociceptive impulses by the dorsal horn neurons. Interneurons receiving input from small-diameter nociceptive afferents are excitatory, whereas those receiving input
Inflammation in joints and other tissues can cause longlasting sensitization of the nociceptive units, leading to the clinical findings of hyperalgesia (increased sensitivity to noxious stimuli) and allodynia (non-noxious stimuli being appreciated as painful) (Fig. 37-3). The thresholds required to excite the nociceptors become reduced, so
• Figure 37-2 Diagram showing the pain pathway in the spinal cord and the so-called pain gate.
• Figure 37-3 Diagram showing central sensitization in the spinal cord after joint inflammation.
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
they are activated by movements that previously were nonactivating, and for any given stimulus they fire at increased frequency compared with the noninflamed stat¢. Some so-called silent nociceptors, which are normally nonresponsive to either innocuous or noxious stimuli, may become excitable, and some non-nociceptor neurons may change phenotype and produce the nociceptor transmitters substance P and brain-derived neurotrophic factor. 18 Sensitization and excitation in injured or inflamed joints is caused by many compounds acting directly, indirectly, or both, often synergistically. Release of substance P into the joint after afferent fiber stimulation produces vascular responses such as vasodilatation, thereby contributing to the inflammatory process as well as sensitizing nociceptive units to excitation by inflammatory mediators such as bradykinin, protons, serotonin, and histamine. 6.11 In addition, prostaglandins, leukotrienes produced by inflammation, and tissue damage directly sensitize the nociceptors. The release of adenosine may lead to sensitization or inhibition, depending on which receptors are activated. Various eytokines, norepinephrine, and nitric oxide also sensitize noCiceptive afferents indirectly. 11.19 Cytokines (tumor neerosis factor-a and interleukin-1~) cause the release of nerve growth factor, which appears to be a central molecule causing hyperalgesia in inflammation. 18 The sympathetic nervous system within the joint probably contributes to the potentiation of pain in arthritis. 20.21 Hyperesthesia and allodynia persisting long after an injory has resolved, as occurs in causalgia or algodystrophy, appear to be often sympathetically mediated (complex regional pain syndrome or reflex sympathetic dystrophy); however, the explanation for this phenomenon remains speculative. 22 In response to inflammation there is, in addition to the release of sensitizing molecules, the release of a number of molecules into the joint, including adenosine, the cannabinoids, and opioids, which have local analgesic and anti-inflammatory effects. 23.24
Spinal Cord The spinal cord plays a major role in modulating pain. After joint inflammation, the nociceptive input is amplified in the dorsal horn. This central sensitization occurs in several ways. Thresholds of nociceptive/specific neurons with receptive fields for the inflamed joint become lowered so that they can be activated by usually innocuous stimuli, and wide-range/dynamic neurons become more responsive to both innocuous and noxious signals. Spinal cord neurons manifest considerable neuroplasticity~ developing expanded receptive fields and thereby becoming responsive to stimuli from areas to which they were previously insensitive. There also appears to be an induction, an increase, or both of spontaneous, ongoing ne.uronal discharges.6.25.26 Repeated C-fiber afferent stimulation, as occurs in persistent inflammation, leads to a sequential increase in dorsal horn activity, which results in prolonged neuronal discharges lasting for many seconds after the stimulus has stopped-a phenomenon known as windup.27 Also, two more effects, each lasting many minutes, known as long-term potentiation and
699
long-term depression, are evoked in two subsets of dorsal horn cells; they are characterized by increased and decreased responsiveness, respectively.28 Phantom limb pain (persistence of pain in a limb that has been amputated) is a well-recognized phenomenon. 29 It is partly explained by neuroplasticity of dorsal horn neurons that leads to central sensitization,30 although more recent studies indicate that changes in the sensory cortex are also important,31 In 1995, Hogeweg and colleagues32.33 demonstrated (1) that children with juvenile chronic arthritis had reduced pain thresholds, not only at inflamed joints but also in noninflamed paraspinal areas, and (2) that children whose arthritis had resolved had persistently lower thresholds than healthy control subjects (although thresholds in this group were higher than in children with continuing joint inflammation). These findings appeared to indicate that children with chronic arthritis develop central sensitization. The pathophysiology of central sensitization is complex and beyond the scope of this chapter. IB•30.34 In brief, high levels of afferent input, as occurs in inflammation, cause the release of excitatory amino acids such as glutamate and aspartate, as well as neuropeptides such as substance P within the dorsal horn. Through actions on N-methylD-aspartate (NMDA) and non-NMDA neuronal receptors, these neurotransmitters cause intraneuronal changes that last minutes to hours. More long-term changes (hours to days) result from induction of the protooncogenes C-fos and Cjun, associated with an increase in the expression of both brain-derived neurotrophic factor receptor (trkB) and substance Preceptor (NK1), but not the glutamate receptors (NMDA and AMPA [a.-amino-3-hydroxy5-methyl-4-isoxazolepropionic acid]). IL-1~ produced in the central nervous system after inflammation has been demonstrated to induce cyclooxygenase-2 (COX-2) in the spinal cord, pons, midbrain, and thalamus; COX-2 in the central nervous system causes increased prostaglandin (PGE2) production and seems to be an important contributing factor in the development of hyperalgesia. 35 Recent investigations indicate that allodynia after nerve injury is, at least in part, caused by the accumulation of microglial cells in the dorsal horn that is subserved by the damaged peripheral nerve. The microglia express the adenosine triphosphate receptor P2X4, and blockade of this receptor diminishes the allodynia induced by the damaged, inflamed nerve.·16 Whether the microglial cells are also responsible for the previously mentioned production of IL-1~ is an unanswered question. In addition to these mechanisms, inhibition of positive feedback loops from supraspinal centers, mediated by serotonin and cholecystokinin receptors, occurs and also contributes to the sensitization of dorsal horn cells. These developments lead to enhanced excitability and expanded receptive fields. Another critical role of the spinal cord is pain inhibition. Nociceptive processes are modulated by several descending inhibitory systems, including a serotoninergic system, norepinephrine via a 2-adrenergic receptors, the opioid system, and the cannabinoids,u·37 These tonic descending inhibitory systems probably playa role in determining the size of the receptive fields of the spinal neurons and their excitation thresholds. Concentrations of these inhibitory
700
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
compounds are increased in experimental arthritis, presumably as an attempt to inhibit the fOlWard transmission of nociceptive signals from the inflamed joints.23,3H-40 Segmental activity in the cord by intraneurons using y-aminobutyric acid (GABA) and glycine neurotransmitters is another important inhibitory mechanism. There is also a form of inhibition known as heterotopic inhibition. Noxious stimuli applied to areas remote from the nociceptive receptive fields inhibit the initial pain. 41 Gentle stimulation of the contralateral inflamed ankle in rats can trigger inhibition of pain in the opposite inflamed ankleY These findings probably explain in part why counterirritants frequently provide some pain relief.
Different children respond very differently to painful medical procedures,56 and they demonstrate large differences in pain sensitivity and tolerance. 2,57 A study of 56 children with juvenile chronic arthritis indicated that only 50% of the pain variability could be explained by a model that included demographic data, medical status, and coping variables. 48 About 30% of the pain variance was explained by disease activity, and approximately 20% by coping strategies. The remaining 50% was not explained by the model. It is certain that many factors interact in a complex manner to modulate the extent to which noxious stimuli are perceived as pain even in conditions in which there are overt "structural" abnormalities to account for the pain.
Brain Although the spinal cord is now recognized as being important to pain modulation, it is the brain that determines the final outcome. The descending inhibitory system originates in several different areas, including the periaqueductal gray area, the rostral ventromedial medulla (which includes the nucleus raphe magnus), and the locus ceruleus. This system is mediated by fibers containing GABA, catecholamines, and serotonin. 18 The periaqueductal gray area contains large quantities of opioid peptides. Their analgesic effects are activated by signals from the frontal cortex, amygdala, and hypothalamus. Signals from the periaqueductal gray area then descend and synapse in the nucleus raphe magnus before proceeding down the dorsal lateral funiculus to the dorsal horns, where they synapse with inhibitory interneurons. Additionally, noradrenergic impulses from the locus ceruleus pass via the rostral ventromedial medulla. An electrical stimulus of the periaqueductal gray area or the rostral ventromedial medulla can produce analgesia without altering alertness. 43 ,44 Destructive lesions or local anesthetics injected into the rostral ventromedial medulla can abolish analgesia produced by stimulation of the periaqueduetal gray area. 45 There is some evidence that adults with fibromyalgia have decreased cerebrospinal fluid levels of enkephalins and serotonin and increased levels of substance p.46,47 Therefore, dysfunction of the central inhibitory systems may contribute to the development of some chronic noninflammatory pain syndromes. These findings confirm the importance of subcortical deep brain structures in modulating nociceptive inputs from the peripheral nervous system. Cortical structures modulate pain in both excitatory and inhibitory ways. Stressful situations perceived at the cortical level are transmitted to the locus ceruleus and hypothalamic-pituitary-adrenal axis and can lead to either increased pain perception or pain inhibitionY Other factors that are important in how pain is perceived by children include one's ability to regulate the focus of attention toward or away from pain, level of arousal in response to noxious stimuli,l.3 use of coping strategies,48.49 and self-esteem.50 These factors may be in part genetically determined, but they are significantly modified by exposure to environmental factors, including exposure to pain in early childhood,4.51 memories of previous painful episodes,52 parental responses to pain or painful situations,53.54 and the developmental age of the child. 55
THE CHILD IN PAIN Pediatric rheumatologists encounter children with a wide variety of musculoskeletal pains, including those with acute and chronic pain for which an "organic" cause can be found, or surmised, and those with a variety of conditions that are classified as pain amplification syndromes. In these disorders, there is frequently no overt primary cause for the pain, which, to the examining physician, seems excessive or amplified. Before focusing on these amplified pain conditions, it is necessary to discuss briefly the measurement of pain and the general approaches to its treatment.
EVALUATION AND MANAGEMENT OF MUSCULOSKELETAL PAIN Assessment of the Child with Musculoskeletal Pain Pain is the subjective expression of an unpleasant sensation associated with actual or perceived tissue damage. 58 It is difficult, if not impossible, for an observer to know with any certainty to what extent a child is in pain. Even experienced observers, such as nurses or parents, may differ in their assessments of the degree of pain the child is experiencing. Although parents and children tend to make similar assessments of the degree of pain, parents may overestimate or underestimate the child's pain in relation to the child's own self-report. An important premise in the evaluation and management of pain in a child is that the child's report of pain and its severity must be accepted per se. Malingering in childhood is exceedingly rare: "Pain is what the patient says it is, and exists when he says it does. "59 Effective management requires that the child know that he or she is believed. Many interacting issues determine whether a child's pain disturbs the child's and the family's functioning and whether or when medical help is sought (Fig. 37-4). The purpose of the remaining sections of this chapter is to provide a framework for assessment of pain, a logical approach to differential diagnosis, and an initial management approach for both acute and chronic pain.
C HAP T E R
Parent
I
"
l Society ~
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
IExternal Stressorsl
" "
Individual Traits
"
Personality, temperament, reactivity/sensitivity
Individual States Self-esteem, anxiety/depression, developmental level
"
I Pain Behavior I
•
IHealth Care Utilization I
• Flgure 17-4 Factors influencing pain expression.
701
pain-related symptoms, a complete past history and review of systems are essential to document the child's overall health status. Obtaining this information can be quite time-consuming, but is especially important in children with amplified pain, and it is the initial step in establishing a trusting relationship with the child and family, who often feel that their concerns have not been considered seriously. The answers elicited by the questions allow a directed clinical examination, enabling the physician to develop a focused differential diagnosis. The physician can then reasonably determine whether further investigations are indicated. A few key observations to be made during the physical examination are listed in Table 37-2.
Laboratory Examination History and Physical Examination In obtaining a history, answers to the following questions should be sought: • .. .. •
What is the character of the pain? Are there any other symptoms? Is there a family history of musculoskeletal disease? What are the family, social, emotional, and educational circumstances?
More detailed questions addressing these four general areas are listed in Table 37-1. In addition to a history of
1=..
TABLE 37-1
History of Musculoskeletal Pain
What is the character of the pain? Which body parts are painful? How long has it been present? Is the pain getting better, worse, or staying the same? What makes the pain better? What makes the pain worse? Is there diurnal variation in the severity of the pain? Is the pain present at night, and if so, does it awaken the patient? Does the pain interfere with function, and if so, what specifically? Is the pain sharp, aching, deep, boring, etc.? Does the pain radiate, migrate, or spread? Is the painful area tender to touch or clothing? Is the painful area either cold or hot to the touch? Does the painful part look abnormal or swollen? What is the child's or parent's assessment of the pain severity? Are there other symptoms? Fever? Rash? Change in gastrointestinal function? Weight loss? Upper or lower respiratory tract symptoms? Muscle weakness? Sleep disturbance? Depression? What is the family and social history? Ankylosing spondylitis, Reiter's syndrome, or intlammatory bowel disease? Back pain, heel pain, or acute iritis? Psoriasis? Fibromyalgia or other chronic pain condition? Is there an identifiable stressor in the family, school, or peer group?
It is important to have a clear rationale for undertaking any investigation. In many situations, no testing is required. A large number of investigations increases the likelihood of false-positive results that may confuse the issue. An evaluation of indices of inflammation (complete blood count, erythrocyte sedimentation rate, and C-reactive protein concentration) is appropriate. It is suspect to make a diagnosis of a noninflammatory condition in a child with an abnormal blood count or increased acute phase reactants, unless the abnormalities can be clearly ascribed to an intercurrent illness. Tests for antinuclear antibodies and rheumatoid factors are of little value in the absence of clinical evidence of inflammatory disease, and false-positive antinuclear antibody seropositivity in particular may lead to unnecessary investigations.6O--{,2 Imaging studies should be directed specifically to rule in or out a specific diagnosis (e.g., plain radiographs or bone scintigraphy for trauma or tumor, magnetic resonance imaging for spinal cord lesions).
Pain Assessment A number of methods are available to assess pain in children. Because pain is a complex state influenced by many factors, no single instrument can provide a complete evaluation. Nonetheless, application of one or more of the available instruments will aid in understanding
•.~~ TABLE 37-2 The Clinical Examination: Several Key Observations
Does the child look well or ill? Is the child's affect commensurate with the level of reported pain? Is there any joint swelling? Is there any muscle weakness or atrophy? Is there any tenderness to palpation, and if so, is it over joints, entheses, or muscles? Is there any body area of allodynia, and if so, is the area constant or does it vary over time? Are there any color, temperature, or perspiration changes? Are there any inconsistencies in the examination? Is there any neurologic dysfunction? Are there abnormal child-parent interactions, such as enmeshment, hostility, berating? Is there evidence of concurrent conversion symptoms?
702
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
pain in the individual child and in groups of children with similar diseases.
Physiologic Measures A number of physiologic measures may be useful to assess pain in very young children or in older children who have limited communication skills. These include heart rate, blood pressure, palmar sweating, transcutaneous oxygen tension, and cutaneous blood flow. However, most of these tests have little or no data to support their reliability, validity, sensitivity, specificity, or practicality.63
tapes, Jaworski and colleagues67 measured pain behaviors in 30 children with juvenile rheumatoid arthritis and concluded that this method was a reliable and provided a valid measure of pain. They also stated that measures of pain behavior might be particularly useful in outcome studies of treatment, because these behaviors are relatively independent of depression.
Self-Report Measures A number of both unidimensional and multidimensional self-report instruments for children were developed during the 1990s. 68
Behavioral Measures Physicians routinely interpret the child's behavior as an indicator of the severity of pain. However, perhaps because behavioral measurements are integrated into the assessment almost subconsciously, and because of inherent biases about what constitutes appropriate responses to pain, such behavioral assessments may be misleading. Nevertheless, it is important to be able to quantify pain behaviors, because they represent important indicators of how a child is dealing with or responding to noxious or perceived noxious stimuli. Furthermore, pain behaviors may themselves positively or negatively affect a child's and a family's pain-coping mechanisms, so an ability to measure these behaviors may help to understand and better manage the child's pain. 64 A number of different scales of pain behavior have been developed. These include various assessments of crying or other verbal responses, facial expressions, and limb movements. 64 The Children's Hospital of Eastern Ontario Pain Scale (CHEOPS)65 is validated and widely used. However, this instrument is not sensitive to change in children who have been in pain for several hours, presumably because pain behaviors habituate if the pain persists. In this situation, CHEOPS scores are generally low and correlate poorly with self-report measures. 66 Using a standardized observational method with video-
Unidimensional Pain Measures The most frequently used simple unidimensional pain scale is the visual analogue scale (VAS). For older children and adults, this is usually a 10-cm horizontal or vertical line anchored by phrases such as "No pain" at one end and "The worst imaginable pain" at the other. The patient puts a mark along the line to represent the level of pain. This mark is measured in millimeters from the "No pain" end to give a value that can be compared over time. For younger children, the VAS is usually anchored by a cartoon happy face at one end and a crying face at the other. Variants of the VAS for children include pain thermometers69 and pain ladders,70 Other unidimensional scales include Likert scales, in which the pain is rated on a four-, five-, or six-point verbal scale (e.g., No pain, Mild pain, Moderate pain, and Severe pain)71 or as a number from 1 to 10. A Faces Rating Scale72 (Fig. 37-5) is a nonverbal, non-numeric Likert scale for younger children.
Multidimensional Pain Measures Multidimensional instruments are questionnaires that collect information about a number of domains relevant to pain, including pain severity, psychologic well-being (anxiety, depression), coping strategies, and self-efficacy.
~
o
2
3
4
\EY 5
Instructions for Faces Rating Scale 1. Explain to the child that each face is for a person who feels happy because he or she feels no pain (hurt, or any word used by the child) or sad because he or she has some or a lot of pain. Face 0 = very happy, has no hurt Face 1 =hurts just a little bit Face 2 = hurts alittle more Face 3 = hurts even more Face 4 = hurts a whole lot Face 5 =hurts as much as you can imagine, although you don't have to be crying to feel this bad 2. Have the child choose the face that best describes how he or she feels. • Figure 37-5 Pain assessment in children: faces rating scale. (From Wong D, Whaley L: Clinical Handbook of Pediatric Nursing, 3rd ed. St. Louis, Mosby, 1990.)
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
The best known of these is the Varni/Thompson Pediatric Pain Questionnaire.73 Other questionnaires are designed to assess pain-coping strategies74 and competency.75 Often these self-report measures are used by health care providers or parents, rather than by the children themselves, to assess pain. The evidence that such observer evaluations are accurate proxy measures of a child's own assessment of pain is equivocal at best. 76 ,77
PAIN MANAGEMENT For 1'l10st of the 20th century, children's pain was generally ignored and undertreated. 78 Since about 1980, there has been a dramatic improvement with recognition that children, including those with chronic arthritis, do have pain, that they can describe it, and that it can be effectively and safely managed. 79-81
703
approach is insufficient, opioids (usually morphine) are introduced in a stepwise fashion. Children with joint pain resulting from end-stage arthritis require joint replacement to effectively control pain. 94 Pain management in some children requires a cognitive-behavioral approach in a multidisciplinary setting. 79,81,95 There are two strands to such treatment. 96 The cognitive strand is aimed at helping children develop more effective coping strategies to deal with pain. The behavioral strand is aimed at decreasing pain behaviors and increasing healthy behaviors. Each of these strands may have several components, depending on the needs of the individual child and the availability and skills of the therapists. The goal of this approach is not to cure the pain but to help the child and family discover ways of coping with pain and disability so that they are less disruptive to the child's life. There is increasing evidence that a cognitivebehavioral approach is effective in children and adolescents with chronic pain. 97
Management of Acute Pain Preventing procedural pain is preferable to treating it.82,83 The application of topical anesthetics before needle punctureS minimizes pain associated with these procedures. 84 The .addition of 2 mL of sodium bicarbonate to 10 mL of 1% lidocaine significantly decreases the stinging sensation associated with this local anesthetic,85 Painful procedures are also made more tolerable by careful attention to the environment in which the procedure is performed. 86,87 Ensuring that the room is quiet; that health care providers approach the child in an unhurried, calm manner; that the child is distracted (by reading or being read to, watching videos, listening to tapes, or playing hand-held computer games) helps the child to cope with pain associated with a procedure, Under most circumstances, the presence of a parent is reassuring to the child and therefore helps to minimize pain. Behavioral techniques and the early and regular use of analgesics are the mainstays of therapy for acute pain that lasts longer than that associated with minor medical procedures. There is convincing evidence that the use of local ane$thetics as adjunctive therapy during surgery under general anesthesia can decrease postoperative pain. 88 The routine use of postoperative analgesia, including morphine, has been demonstrated to decrease the time required for convalescence. It is now clear that children can tolerate intravenous morphine, with a low risk of respiratory depression and minimal risk of addiction. 89,90 Most chil~ren older than 5 years of age are able to use patientcontrolled analgesia effectively.91
Amplified Musculoskeletal Pain Many children with severe musculoskeletal pain do not have an identified inflammatory disease or mechanical derangement to cause their degree of pain and debility. These children have usually been seen by multiple physicians, and have undergone multiple investigations, before the correct diagnosis is made. Unfortunately, attempts to identify an increasingly rare and unlikely cause for the pain help to perpetuate it. An experienced physician can often recognize the condition promptly and, by halting further medical investigations and initiating appropriate therapy, provide the greatest service to the child and family.
HISTORICAL REVIEW Chronic musculoskeletal pain in children received virtually no attention until the latter half of the 20th century. In 1951, Naish and Apley98 published a study on pediatric limb pains due to nonarthritic causes. Since then, it has increasingly been recognized that a significant number of children suffer from both chronic and amplified musculoskeletal pain. 99 ,100 Reflex neurovascular dystrophy (complex regional pain syndrome, type 1) was first described in a child in 1971,101 as was fibromyalgia in 1985. 102 These two disorders are the subject of most studies; other, less clearly classified, or generally unclassifiable, amplified musculoskeletal pain conditions are less frequently reported. 103-105
Management of Chronic Pain The World Health Organization has outlined a progressive thempeutic stepladder for management of pain, beginning with simple analgesics. 92 An analgesic with adjunctive therapy is sufficient for most children with rheumatic diseases. Adjunctive therapy includes the behavioral techniques discussed previously as well as various physical modalities (massage, physical therapy, transcutaneous electrical nerve stimulation [TENS], acupuncture) and psychologic techniques (hypnotherapy, biofeedback).93 If this
DEFINITION AND CLASSIFICATION Current terms used to describe these conditions are inadequate and confusing, because many children have features that are shared among different subsets. 103,104 Various authors have separated these children depending on physical features (e.g., presence of overt autonomic signs,58 number of painful points with a variety of systemic symptoms 102,106) or on location (localized or diffuse), 104 The terminology used is, in one sense, moot
704
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
because evaluation and treatment are similar between subsets. 105.107 In this chapter, the term amplified musculoskeletal pain is used because it is descriptive, does not presume an etiology, and differentiates these children from adults with chronic pain. It is important to know what criteria in studies have been used to classify children with various forms of amplified musculoskeletal pain (Table 37-3). Discrete subsets exist in each of the groups. Specifically, children who fulfill criteria for fibromyalgia are included with those who have diffuse idiopathic musculoskeletal pain, and those with complex regional pain syndrome are included with those who have localized idiopathic musculoskeletal pain. There is a subset of children with intermittent amplified musculoskeletal pain in whom the criterion for duration is not satisfied; they are nevertheless included because of the severity of their pain and marked dysfunction reoccurring over many months. lOB
EPIDEMIOLOGY Incidence and Prevalence Population surveys of school-aged children confirm that musculoskeletal pain is common; the incidence of back pain is as high as 20%,109 and limb pain has been reported in 16%.99.110 The frequency of fibromyalgia has been reported in children and adolescents to be 2% to 6%.111.112 There are no specific data regarding the other amplified musculoskeletal pain syndromes, but 5% to 8% of new patients presenting to North American pediatric rheumatology centers most likely have a form of amplified musculoskeletal pain. I13,114 Although it may be a result of selection and referral bias, many pediatric rheumatologists believe that they are seeing increasing numbers of children with amplified musculoskeletal pain in the past two decades.
Age at Onset Amplified musculoskeletal pain has been described in patients as young as 2 years of age, but the majority of reports involve children in late childhood and adolescence, with a mean age at onset of 12.4 years.l02-104,115-124 Older adolescents may be underrepresented, presumably because they are referred to adult specialists.
Sex Ratio All large series agree that girls predominate over boys in a ratio of approximately 4:1. 102- 104,115-118 Because women seek medical advice more often than men, there may be a selection bias; however, given the disability involved, this is most likely not a major factor in children.
Geographic and Racial Distribution There have been no formal investigations regarding the relationship of amplified pain syndromes to ethnicity; however, a series from Philadelphia reported a disproportionate number of white patients (15 of 15),125 All
reports are from developed countries, and comparisons with developing nations are impossible.1 26
ETIOLOGY AND PATHOGENESIS The cause or causes of the various amplified musculoskeletal pain syndromes are unknown. No controlled studies exist, especially in children. Childhood pain syndromes may differ significantly from those occurring in adults; for example, there is a distinct difference between technetium bone scintigraphy in children and in adults with complex regional pain syndrome type 1. Children also respond more readily to exercise therapy,Hl8·115.118,125.127.1211 Nevertheless, in many children, these syndromes seem to be causally related to injury, illness, or psychologic distress, either singly or in combination. Injury, including surgery, frequently precedes complex regional pain syndrome in adults, and minor injury is commonly reported in children, Rarely, more overt trauma may be the inciting event,129-132 Six children in British Columbia developed complex regional pain syndrome type I after hepatitis B vaccination; this was thought to be caused either by the injection trauma or by the constituents of the vaccine. 133 Minor trauma may playa role in localizing the site of amplified musculoskeletal pain. Children who are hypermobile may be at increased risk of developing the fibromyalgia form of amplified musculoskeletal pain, perhaps because of chronic microtrauma; however, there is some uncertainty about this,12I.134-136 Illness such as myocardial infarction has been associated in adults with the complex regional pain syndrome, and arthritis may coexist with amplified pain in children. 137,138 Amplified musculoskeletal pain has also been observed in children with a variety of illnesses, including cerebral palsy, muscular dystrophy, new-onset diabetes, and leukemia. These associations may or may not be coincidental. Psychologic distress has been a recurring theme in multiple reports of children with amplified musculoskeletal pain syndromes, although controlled studies are substantially lacking. 98,103,115,1I7,120,121,124,139-145 Clearly, some children and their families are overtly psychologically dysfunctional, but whether this is the cause of, the effect of, or unrelated to the development of an amplified musculoskeletal pain syndrome is not known. Not all families with a painful, dysfunctional child are inappropriately distressed, however. The role of hormonal or environmental factors is uncertain. The observation that girls are more frequently affected may reflect the fact that girls have lower pain thresholds, increased levels of hypermobility, and increased frequency of sleep disorders than boys, as well as differences in coping and cultural expectations (especially in Western countries),146--148 The social history may document multiple recent major life events such as moving, changes in the nuclear family, family illness or death, or school stress. 103,l17 There is commonly a role model (usually a parent) for either chronic pain or disability. Interdependency, or enmeshment, between the patient and the parent (usually the mother) is striking in many families: even when the physician directly addresses a question to the child, the parent insists on answering. 103.1l7
C HAP T E R
f.
lABLE 37-3
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
705
Criteria for Various Subsets of Amplified Musculoskeletal Pain
ConIpIex Regional Pain Syndrome Type ICriteria 2-4 must be satisfied: 1. The presence of an initiating noxious event or a cause of immobilization 2. Continuing pain, allodynia, or hyperalgesia in which the pain is disproportionate to any inciting event 3. Evidence at some time of edema, changes in skin blood flow, or abnormal sudomotor activity in the region of the pain 4. This diagnosis is excluded by the existence of conditions that would otherwise account for the degree of pain and dysfunction
Complex Regional Pain Syndrome Type IIAll three criteria must be satisfied: 1. The presence of continuing pain, allodynia, or hyperalgesia after a nerve injury, not necessarily limited to the distribution of the injured nerve 2. Evidence at some time of edema, changes in skin blood flow, or abnormal sudomotor activity in the region of the pain 3. This diagnosis is excluded by the existence of conditions that would otherwise account for the degree of pain and dysfunction
1990 AmerIcan College of Rheumatology (ACR) Criteria for Flbromyalglat Both criteria must be satisfied: 1. Widespread pain (bilateral, above and below the waist, and axial pain) present for at least 3 rna 2. Pain (not tenderness) on digital palpation with 4 kg of pressure on 11 of the following 18 sites: a. Occiput: at insertion of suboccipital muscle b. Low cervical: at the anterior aspect of the intertransverse spaces of C5-C7 c. Trapezius: at the midpoint of the upper border d. Second rib: just lateral to the second costochondral junction at the upper rib border e. Lateral epicondyle: 2 cm distal to the epicondyle f. Gluteal: in the upper outer quadrant of the buttocks g. Greater trochanter: posterior to the trochanteric prominence h. Knees: at the medial fat pad proXimal to the joint mortise
Yu.,..s and Masl CrIteria for Childhood Flbromyalgla* All four major and three minor criteria, or the first three major criteria, four painful sites, and five minor criteria, must be satisfied: Major: 1. Generalized musculoskeletal aching at three or more sites for 3 or more months 2. Absence of underlying condition or cause 3. Normal laboratory test results 4. Five or more typical tender points (see sites listed in 1990 ACR criteria) Minor: 1. Chronic anxiety or tension 2. Fatigue 3. Poor sleep 4. Chronic headaches 5. Irritable bowel syndrome 6. Subjective soft tissue swelling 7. Numbness 8. Pain modulation by physical activities 9. Pain modulation by weather factors 10. Pain modulation by anxiety or stress
Diffuse idiopathic Paint Both criteria must be satisfied I. Generalized musculoskeletal aching at three or more sites for 3 or more months 2. Exclusion of disease that could reasonably explain the symptoms
localized idiopathic Paint All' three criteria must be satisfied: I. Pain localized to one limb, persisting 1 wk with medically directed treatment or 1 mo without medically directed treatment 2. Absence of prior trauma that could reasonably explain the symptoms 3. Exclusion of diseases that could reasonably explain the symptoms ·Mer,key OM. Bogduk N: Classitkation of Chronic Pain: Descriptions of Chronic Pain Syndromes and Definitions of Pain Terms. Seattle. IASP Press. 1994. 'Wolfe F. Smythe HA. YUmlS MB, et al: The American College of Rheumatology 1990 Criteria for the Classification of Fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis Rheum 33: 160-172. 1990. *Yunus MB, Masi AT: Juvenile primary fibromyalgia syndrome: a clinical study of thirty-three patients and matched normal controls. Arthritis Rheum 28: 138-145, 1985. IMalieson PN, al-Matar M, Petty RE: Idiopathic musculoskeletal pain syndromes in children. J Rheumatol 19: 1786--1789. 1992.
706
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
The pathophysiology of amplified musculoskeletal pain is unknown, but in some children, especially those with localized or regional pain, it may be related to either increased sympathetic nervous system activity or increased a-adrenoceptor responsiveness. 149-154 Diffuse amplified pain, particularly fibromyalgia, has been extensively studied in adults, and a wide variety of hypotheses have been suggested, including abnormal muscle anatomy and physiology, altered sleep pattern, abnormal serotonin metabolism, hypothalamic-pituitary-adrenal axis hypofunction, decreased cerebral blood flow, trauma, and psychologic distress. There is no convincing evidence that any of these factors is of primary importance. 1SS Two recent studies involving small numbers of adults with fibromyalgia suggest that they have a deranged sympathetic response to orthostatic stress. IS6 ,IS7
GENETIC BACKGROUND Amplified musculoskeletal pain syndromes have been reported in siblings,IS8,ls9 parentichild pairs,144,160,161 spouses, and multiple family members. 162 ,163 Buskila and colleagues l60 reported that 28% of 58 offspring from 20 mothers had fibromyalgia,160 and Pellegrino and colleagues l61 found that 52% of 50 subjects from 17 families had fibromyalgia, and another 22% had clinical evidence of abnormal muscle consistency, with a set of identical twins developing fibromyalgia within 6 months of each other. 161 Others have noted that not only fibromyalgia but other chronic pain conditions are more common in family members of children with fibromyalgia. 164 No particular gene has been implicated, although one small study of complex regional pain syndrome type I suggested that women with the human leukocyte antigen HLA-DR2 Is may be more resistant to treatment. 16S If genetic factors are important in amplified pain conditions, it is probable that multiple genetic polymorphisms are involved and predispose the individual to develop amplified pain conditions if inciting environmental or cultural factors and triggers are present.
CLINICAL MANIFESTATIONS Although there are differences between localized and diffuse amplified musculoskeletal pain syndromes, the medical history and physical examination in both are surprisingly consistent. In children with localized amplified musculoskeletal pain, minor trauma that might not be clearly recalled is common ("Someone must have stepped on my foot"). The pain and consequent disability increase over time regardless of medication. A cast or splint may minimize the pain while it is worn, but immobilization is an important factor in perpetuating the pain. Autonomic signs (edema, cyanosis, coolness, increased perspiration) may be persistent or transient, or they may not occur at all. Allodynia (pain generated by normally nonpainful stimuli) can be marked ("The breeze of someone walking by hurts") and can lead to significant impairment. This phenomenon, too, can be transient.
Any body part can be involved, and the child can have several areas of pain at presentation. The lower extremity is more commonly involved than the upper, and peripheral body parts are more commonly involved than central areas. Occasionally, only one small area is involved, such as a finger, the nose, or a tooth. l66 Localized pain is usually continuous. In diffuse amplified musculoskeletal pain, the onset is usually more gradual and can be vague in location and character. There is an absence of autonomic signs, but affected children complain of poor sleep and depression more often than do those with localized pain. ISS These children frequently report a multiplicity of symptoms. Pain is often centrally located, involving the back, chest, abdomen, and head as well as the extremities. Conversion symptoms are common.103.167.168 Numbness is frequently reported, but these children may also report paralysis, blindness, or a bizarre (histrionic) gait. Eating disorders may be present. 169 ,170 The child often has a markedly incongruent affect, smiling even when reporting severe pain (up to 10 out of 10), and la belle indifference about the pain and the dysfunction it causes. This disparity between affect and function is also frequently present in children with localized amplified pain. A few children demonstrate marked pain behaviors such as crying or screaming. This is more common in those with localized amplified musculoskeletal pain. Affected children often seem to be mature for their age, are accomplished in school and extracurricular activities, and are described by their parents as perfectionistic, empathetic, and pleasers. Notable points on physical examination include the absence of findings that would suggest an underlying disease, a normal neurologic examination, and the presence of allodynia. Careful sensory testing, with special attention to dermatomal and peripheral nerve innervation, is required. Allodynia is present if pain is reported in response to a light touch on the skin or gentle pinching of a fold of skin. There can be markedly different borders of allodynia on repeat testing. Signs of autonomic dysfunction, especially coolness and cyanosis, may be present after exercising the limb, or they may become apparent if the limb is held in a dependent position for a few minutes. The distribution of painful points is outlined in Table 37-3 and illustrated in Figure 37-6. Control points such as the forehead, shin, and thumbnail define how widespread the pain is. l7l Children may report that their entire body is painful, and some even have pain from gentle touching of their hair. A number of children with diffuse amplified pain do not have tender points, although they are otherwise indistinguishable from those fulfilling criteria for fibromyalgia. Prolonged back pain in childhood is often caused by a serious illness and should be carefully investigated. 172 However, there is a subset of children who have nonorganic back pain, usually in conjunction with diffuse amplified musculoskeletal pain. Distinguishing signs include the axial loading test, distracted straight leg raising, passive rotation test, overreaction, and allodynia (Table 37-4),173
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
707
~ 37-6 Diagram of painful points in fibromyalgia, as defined by the American College of Rheumatology,l06 (From Okifuji A,Turk DC, Sindair JK, et al: A standardized manual tender point survey: I, Development and determination of athreshold point for the identification of positive tender points in fibromyalgia syndrome. J Rheumatol24: 377-383, 1997.)
•
I:
II
'ABt [ 37-4
Test
Signs of Nonorganic Back Pain
DescrIption
A positive test occurs when back pain is reported while the examiner exerts downward pressure on the top of a standing patient's head. Neck pain may be elicited and is not a positive test. In a positive test, flexion of the hip causes Distracted straight back pain when the patient is supine but leg raising not when sitting. Passive rotation test A positive test occurs when the patient reports back pain with passive rotation at the ankles and knees, keeping the pelvis, back, and shoulders in the same plane. Overreaction Overreaction is defined as excessive wincing, muscle tremors, screaming, or collapsing with pain. "Excessive" is quite subjective and may vary based on age, mental status, cultural influences, or fear. Report of pain to light touch or a gentle Allodynia pinch of the skin, usually with a border that varies on repeat testing. Axial loading test
Data from Waddell G, McCulloch JA, Kummel E, Venner RM: Nonorganic physical silins in low-back pain. Spine 5: 117-125. 1980.
PATHOLOGY There is virtually no information concerning the histopathology of connective or nerve tissues from children with amplified musculoskeletal pain syndromes. Three children with complex regional pain syndrome type I had findings on biopsies of skin, muscle, and nerve consistent with ischemic injury.174 Endothelial swelling, basement membrane thickening and reduplication, and patchy fiber atrophy of muscle were observed. The authors have seen one patient with complex regional pain syndrome type I who had ischemic changes present in the synovium of her knee.
DIFFERENTIAL DIAGNOSIS A number of other painful conditions need to be considered in the evaluation of a child who may have an amplified musculoskeletal pain syndrome. Table 37-5 lists disorders confused with amplified pain syndromes, many of which are discussed in other chapters. One condition commonly misdiagnosed as an amplified pain syndrome is the seronegative enthesitis and arthritis
708
IfIi
C HAP T E R
fABLE 37 5
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
Differential Diagnoses in Children Presenting with ldioJMthit P,lin SyndrOlIH.'s
Diagnosis
Typical Age
Distinguishing Characteristics
Fabry's disease
Adolescents
Neoplasia Spinal cord tumors Erythromelalgia
Any Any Adolescents
Pernio (chilblains)
Any
Raynaud's disease Hypermobility Restless legs
Adolescents Younger Adolescents
Myofascial pain
Adolescents
Chronic recurrent multifocal osteomyelitis Chronic compartment syndrome Progressive diaphyseal dysplasia Peripheral mononeuropathy Transient migratory osteoporosis Vitamin D deficiency Thyroid disease
Any
Episodic, excruciating burning pain in the distal extremities; blue maculopapular, hyperkeratotic lesions clustered on the lower trunk and perineum; erythrocyte sedimentation rate is usually elevated Episodic or migratory pain or arthritis, generalized malaise, anorexia, and bone pain Abnormal neurologic examination, altered gait, or spinal curvature Pain with erythematous, warm, swollen hands or feet that is eased by cold to the point that patients refuse to remove ice or cold water from their affected limbs Burning pain with associated red to purple, swollen papules on exposed fingers or toes after cold injury Tricolor change (white, blue, red), associated with tingling; usually not very painful Intermittent nocturnal pains that may occur after certain activities Nocturnal discomfort in, and an inability to keep from moving, the legs; paresthesias, not pain per se, are common and rarely cause awakening Sustained contraction of part of a muscle, especially those about the head, jaw, and upper back; pain well localized and reproduced when that part of the muscle is palpated Specific point tenderness
Adolescents
Severe muscle pain (usually calO after exercising
Adolescents Adults
Severe leg pain, fatigue, headaches, weight loss, weakness, and an abnormal, waddling gait; radiographs show cortical thickening and sclerosis of the diaphysis of the long bones Post-traumatic mononeuropathy
Adolescents
Rapidly developing, painful osteoporosis
Adults Any
Hyperesthetic pain in debilitated patients with multiple reasons to be deficient in vitamin 0 Widespread musculoskeletal pain with either hypothyroidism or hyperthyroidism with associated symptoms of thyroid dysfunction
(SEA) syndrome, especially in children with back pain. The most common misdiagnoses for children who actually have an amplified pain syndrome are trauma, mechanical pain, and arthritis.
GENERAL ASSESSMENT OF PAIN There are two major independent variables to consider when assessing pain: the quality and quantity of the pain complaint itself and the amount of dysfunction incurred as a consequence of the pain. The report of pain is always valid because, by definition, pain is subjective. 58 Therefore, the most useful measurement of pain is the self-report on a verbal scale or VAS. The quality of pain can be assessed with the use of various instruments such as the McGill Pain Questionnaire or the Pediatric Pain Questionnaire.73,t75 The amount of reported pain does not directly correlate with the degree of incapacity, which can vary from almost none to being bedridden. An important observation is that function usually returns before the pain diminishes. Functional measurements vary depending on the location of the pain and the presence of coexisting conditions such as arthritis or, more commonly, conversion symptoms. Children with both amplified musculoskeletal pain and conversion paralysis are extremely dysfunctional. Children with amplified musculoskeletal pain do, in fact, suffer more than children with other musculoskeletal conditions, which, it may be speculated, may indicate
the degree to which the disorder is a manifestation of psychologic distress. In one comparison of the degree of well-being using a VAS, children with complex regional pain syndrome type I ranked themselves as more disabled than did those with juvenile rheumatoid arthritis. 1I7 Psychologic dysfunction is almost universally present by the time these children are identified. 103,1l5,1l7,12J,176,177 Even though it is not necessarily true that these syndromes are psychologic in origin, the psychologic toll on the child and family is often severe. The degree of psychosocial pathology is highly variable and may range from anxiety or poor coping to borderline personality disturbance; it may also involve siblings and other family members.
LABORATORY EXAMINATION Often, after a history has been taken and an examination performed, the diagnosis of an amplified pain syndrome is certain and no further investigations are required. Tests of blood and urine are normal. The most common "abnormal" test is a low-titer positive antinuclear antibody 0:40 to 1:160), which should be discounted. 61 Two reports have described normal or slightly slowed nerve conduction velocity in patients with complex regional pain syndrome type 1. 178 ,179 Any laboratory testing in these children should be done with caution, because the more tests that are performed, the more likely is the occurrence of false-positive results, leading to unjustified doubt about the diagnosis, anxiety concerning more serious illnesses, and delay in initiating treatment.
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
RADIOGRAPHIC EXAMINATION Radiographic findings are normal or demonstrate disuse osteoporosis depending on the duration and degree of disability; rarely do children have the spotty osteoporosis that occurs in adults with complex regional pain syndromes. 11S The technetium radionuclide bone scan is probably the most useful study if the diagnosis is in doubt.125.127 The most frequent abnormality is decreased radionuclide uptake in the affected limb. A normal study is evidence against an underlying bone disease. Magnetic resonance imaging in children with localized amplified pain documents regional bone marrow edema, with T1-weighted images of low signal intensity and T2-weighted images with high signal intensity; this test is reportedly more sensitive than scintigraphy.I80.181
TREATMENT The plethora of widely disparate treatments attests to the fact that there are no proven therapies demonstrated by well-controlled therapeutic trials in children with amplified musculoskeletal pain.143.167 Many reports describe a single child or a very small group of children. Therefore, treatment is based largely on clinical experience and must be considered as only possibly correct. Treatment should have two goals: restoration of function and relief of pain. Anything less is not ideal; however, because pain is subjective and not directly amenable to specific treatment, there are some patients in whom restoration to full function without total pain relief has to be accepted. Helping the child develop skills to cope with the pain is often effective in relieving distress ~tnd dysfunction, even if the pain persists. Mast publications deal with localized amplified musculoskeletal pain, specifically complex regional pain syndrome type 1. Of these, most report benefit, in order of success, with exercise therapy, TENS, and sympathetic blocks. 103,ll,\--II8.123.129,158.178.179.182-188 Most authors advocate aggressive exercise therapy aimed at reversing immobility and increasing function. Treatments used less commonly and with variable results include glucocorticoids,1I6,130 tricyclic. antidepressants (TCAs), anticonvulsants, opioids, sympathectomy, biofeedback, behavioral modification, and psychotherapy.1I6.130.189.190 Wilder and colleagues Jl6 advocMed a combination of multiple physical and medical approaches, including sympathetic blocks and sympathectomy, with pain resolution in fewer than half of their patients. Bernstein and associates 11 5successfully treated 23 children with exercise therapy alone and reported longterm follow-up on 20: 12 were without any pain, 5 had occasional discomfort without any physical signs, 2 had moderate discomfort with some swelling, and 1 had a recurrence of neurovascular dystrophy. Sherry and coworkers JlR treated 103 patients with an intense exercise program without medications (described later); 95 patients (92%) resolved all pain, and all patient,> regained function. They also report the longest follow-up data, just over 5 years, and 88% were symptom free. There are few reports of treatment of diffuse amplified musculoskeletal pain, mainly fibromyalgia, in children. Studies in adults have found combination therapy with
709
education, mild aerobic exercise, a low-dose TCA, and a nonsteroidal anti-inflammatory drug (NSAID) helpful but not curative.102.135.191-194 One report of 15 children judged cyclobenzaprine to be helpful in 11 (73%), but the durability of benefit was not reported. 193 However, in another study, only 3 of 33 children indicated that they would recommend cyclobenzaprine to other individuals with similar pain. In Psychologic support has also been advocated.142.176.194.195 Waleo and Ilowite,195 using progressive muscle relaxation and guided imagery, treated five girls with fibromyalgia with four to nine sessions, and four reported no pain an average of 10 months later. Gedalia and colleagues 134 reported that only two of five children with fibromyalgia found cognitive behavioral therapy to be of help. Formal psychotherapy based on an initial psychologic evaluation has been advocated by some.107.164.196 It is the authors' experience that children do benefit to a degree from cognitive-behavioral therapy, but it does not lead often to complete resolution of symptoms. Early results in the United Kingdom of a residential interdisciplinary cognitive-behavioral therapy program with parental involvement, for adolescents with chronic pain, are very promising. 197 More traditional psychotherapy may be beneficial for some children. Sleep disturbance is frequently mentioned as important in cause of childhood fibromyalgia; however, no data exist as to whether treating sleep disturbance per se is of benefit. Good sleep hygiene is always advocated, but the degree of help is not known. Low-dose TCA therapy has been recommended to facilitate sleep initiation.192.194 The authors have been successful with a team approach, with psychologic evaluation and therapy (if indicated) for children with both localized and diffuse amplified musculoskeletal pain that involves intense exercise therapy directed to the restoration of full function. 103,107,108,118,128,198 The exercise therapy is typically for 5 hours a day on weekdays for a mean of 2 to 3 weeks. The children are treated one-on-one, with the therapist encouraging both speed and quality of movement. Exercises are focused on normal function and aerobic training such as walking times, rope jumping, climbing stairs, dressing, and other activities of daily living. lOS ,107,1I8,198 Allodynia is treated with desensitization, using towel and lotion rubs and encouraging normal clothing and footwear. Most children are day patients and are expected to engage in normal family activities at night and on weekends while doing a home exercise program. Children who are very dysfunctional or have severe postexercise pain behaviors, are best treated as inpatients. Children with persistent poor sleep may be given a therapeutic trial of a low-dose TCA or a specific sleep medication such as zolpidem. If more pervasive sleep symptoms are present, a sleep study may be helpful, especially if sleep apnea is suspected. In the authors' experience, analgesics, physical therapy modalities such as TENS, sympathetic blocks, and corticosteroids are not required. Those patients with clinical depression need to be evaluated by a psychiatrist. Psychologic testing is helpful not only in ascertaining whether there are significant family or other environmental stress factors (e.g., learning disabilities) and whether the child is depressed, but also as a foundation for cognitive-behavioral therapy to teach pain-coping strategies.
710
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
Children with amplified musculoskeletal pain frequently use alternative treatments, but there are no data regarding benefit. Children have tried herbal therapy, massage, magnet therapy, homeopathy, reflexology, and aromatherapy, to name a few. At least for the authors' patients, these have been ineffective; none can be recommended.
COURSE AND PROGNOSIS No studies of the natural history are available, but some children have amplified pain that persists for years; however, those with self-limited involvement probably are not evaluated in tertiary care centers. There may be many children who have spontaneous remission of illness; in one report, 11 of 15 children incidentally diagnosed with fibromyalgia were asymptomatic after 30 months. l99 However, 920/0 of children diagnosed with fibromyalgia in a pediatric rheumatology center still had significant pain 15 to 60 months (mean, 33 months) later. 2oo Siegel and colleagues 192 reported that 94% of 33 patients (from an original 45) treated with low-dose TCAs, mild exercise, and NSAIDs still had diffuse pain and poor sleep after 1 year, although their overall well-being improved 1.8 points on a scale of 1 to 10. The authors have experienced much better short- and long-term outcomes. 107, 118 In general, children fulfilling criteria for complex regional pain syndrome type I do better than those children who have regional pain without signs of autonomic dysfunction; the latter group, in turn, do better than those with diffuse amplified musculoskeletal pain. After a mean of 5 years, 88% of children with complex regional pain syndrome type I were free of pain and fully functional. 118 Most (900/0) of those without autonomic signs were functional, but only 780/0 were without pain. 103 Initially, 90% of the group with fibromyalgia were pain free, but this success declined to 500/0 over 5 years of follow-up. However, 900/0 still remained fully active in school or employment. No difference in outcome was observed between girls and boys or between younger children and older adolescents. The frequency of relapses is rarely reported, but relapse occurs in all forms of amplified musculoskeletal pain. The clinical manifestation of the second episode may be different from that of the first, even changing between localized and diffuse disease. 103.104 In the authors' experience, children with relapses are more likely to have significant underlying psychopathology. In addition to recurrent episodes of amplified musculoskeletal pain, children may develop chronic pain involving other organ systems, especially headaches or abdominal pains, and other psychologic problems, including conversion disorders (blindness, paralysis), suicide attempts, panic attacks, or eating disorders. Controlled studies have not been done, so it is unclear whether these problems occur at a greater frequency than in the general population.
CONCLUSION Musculoskeletal pain is common in childhood, and it is important to accurately diagnose the cause and to develop a logical and consistent approach to its manage-
ment. A careful history and examination, combined with judicious laboratory or radiographic investigations, can usually allow an accurate diagnosis to be made. The use of a few simple measures of pain intensity, particularly self-report scales, can help the physician assess how the child and the parent perceive the pain and thereby enable institution of the most appropriate management. Reassurance, the use of simple analgesics or anti-inflammatory drugs, and physiotherapy are often sufficient. However, some children with amplified musculoskeletal pain need much more complex medical and psychologic management strategies involving a multidisciplinary team approach. It is challenging to care for these children, but with the use of an interdisciplinary, focused, team approach the outcomes appear to be encouraging. Further investigation is needed to design the optimal approach to management for children and adolescents with these complex pain syndromes.
REFERENCES 1. Merskey H: Pain terms: a list with definitions and notes on usage, Recommended by the International Association for the Study of Pain (IASP) Subcommittee on Taxonomy. Pain 6: 249-252, 1979. 2, LeBaron S, Zeltzer L, Fanurik D: An investigation of cold pressor pain in chil· dren: part I. Pain 37: 161-171, 1989. 3. Zeltzer L. Bursch B, Waleo G: Pain responsiveness and chronic pain: a psy· chobiological perspective. J Dev Behav Pediatr 18: 41}-422, 1997. 4. Taddio A, Katz J, Ilersich AL, Koren G: Effect of neonatal circumcision on pain response during subsequent routine vaccination, Lancet 349: 599-603, 1997. 5. Fitzgerald M: Development of the peripheral and spinal pain system, In Anand KJS, Stevens BJ, McGrath PJ (eds): Pain in Neonates. Amsterdam, Elsevier, 2000, p 9-22, 6. Schaible HG, Grubb BD: Afferent and spinal mechanisms of joint pain, Pain 55: 5-54, 1993. 7. Mense S: Nociception from skeletal muscle in relation to clinical muscle pain. Pain 54: 241-289, 1993. 8. Schaible HG, Schmidt RF: Effects of an experimental arthritis on the sen· sory properties of flne articular afferent units. J Neurophysiol 54: 1109-1122, 1985. 9. Dubner R: Neurophysiology of pain. Dent Clin North Am 22: 11-30, 1978. 10. Beaulieu P, Rice ASC: Applied physiology of nociception, In Rowbotham OJ. Macinryre PE (eds): Clinical Pain Management: Acute Pain. London, Arnold. 2003, PP 3-16, 11. Markenson JA: Mechanisms of chronic pain, Am J Med 101: 65-18S, 1996, 12, Schaible HG, Schmidt RF, Willis WO: Convergent inputs from articular, cuta· neous and muscle receptors onto ascending tract cells in the cat spinal cord. Exp Brain Res 66: 479-488, 1987. 13. Basbaum AI, Jessell TM: The perception of pain. In Kandel ER, Schwartz JH, Jessell TM (eds): Principles of Neural Science. New York, McGraw·Hill, 2000, pp 472-491. 14. Coghill RC, Sang CN, Maisog JM, Iadarola MJ: Pain intensity processing within the human brain: a bilateral, distributed mechanism, J Neurophysiol 82: 1934--1943, 1999, 15, Treede RD, Kenshalo DR, Gracely RH, Jones AK: The cortical representation of pain. Pain 79: 105-111, 1999. 16, Craig AD, Chen K, Bandy D, Reiman EM: Thermosensory activation of insular cortex. Nat Neurosci 3: 184--190, 2000, 17. Pillemer SR, Bradley LA, Crofford LJ, et al: The neuroscience and endocrinol· ogy of flbromyalgia. Arthritis Rheum 40: 1928-1939, 1997. 18. Nagy I, Rice AS: Applied physiology of inflammatory pain. In Rowbotham OJ, Macintyre PE (eds): Clinical Pain Management: Acute Pain. London, Arnold, 2003, pp 17-41. 19. Konttinen YT, Kemppinen P, Segerberg M, et al: Peripheral and spinal neu· ral mechanisms in arthritis, with particular reference to treatment of inflam· mation and pain. Arthritis Rheum 37: 965-982, 1994, 20, LevineJD, Dardick SJ, Roizen MF, et al: Contribution of sensory afferents and sympathetic efterents to joint injUry in experimental arthritL~. J Neurosci 6: 3423-3429, 1986, 21. Levine JD, Fye K, Heller P, et al: Clinical response to regional intravenous guanethidine in patients with rheumatoid arthritis. J Rheumatol 13: 1040--1043, 1986, 22, Roberts W): A hypothesis on the physiological basis for causalgia and related pains. Pain 24: 297-311, 1986.
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
23. Jaggar SI, Hasnie FS, Sellaturay S, Rice AS: The anti-hyperalgesic actions of the cannabinoid anandamide and the putative CB2 receptor agonist palmit(:lylethanolamide in visceral and somatic inflammatory pain. Pain 76: 189-199, 1998. 24. Stein C, Schafer M, Cabot PJ, et al: Peripheral opioid analgesia. Pain Rev 4: 173-187, 1997. 25. Grubb BD, Stiller RU, Schaible HG: Dynamic changes in the receptive field properties of spinal cord neurons with ankle input in rats with chronic unilateral inflammation in the ankle region. Exp Brain Res 92: 441-452, 1993. 26. Neugebauer V, Schaible HG: Evidence for a central component in the sensitization of spinal neurons with joint input during development of acute arthritis in eat's knee. J Neurophysiol 64: 299-311, 1990. 27. Mendell LM: Physiological properties of unmyelinated fiber projection to the spinal cord. Exp Neurol 16: 316--332, 1966. 28. Randic M, Jiang MC, Cerne R: Long-term potentiation and long-term depression of primary afferent neurotransmission in the rat spinal cord. J Neurosci 1:3: 5228-5241, 1993. 29. Melzack R: Phantom limb pain: implications for treatment of pathologic pain. A'nesthesiology 35: 409-419, 1971. 30. Coderre 1], Katz J, Vaccarino AL, Melzack R: Contribution of central neuroplasticity to pathological pain: review of clinical and experimental evidence. Pain 52: 259-285, 1993. 31. Harris AJ: Cortical origin of pathological pain. Lancet 354: 1464-1466, 1999. 32. Hogeweg JA, Kuis W, Huygen AC, et al: The pain threshold in juvenile chronic arthritis. Br J Rheumatol 34: 61-{i7, 1995. 33. J-Iogeweg JA, Kuis W, Oostendorp RA, Helders PJ: General and segmental reduced pain thresholds in juvenile chronic arthritis. Pain 62: 11-17, 1995. 34. Wilcox GL: Excitatory neurotransmitters and pain. In Bond MR, Charlton JE, Woolf CJ (eds): Pain Research and Clinical Management. Proceedings of Sixth World Congress on Pain. Amsterdam, Elsevier, 1991, pp 97-117. 35. Samad TA, Moore KA, Sapirstein A, et al: Interleukin-lbeta-mediated induction of cox-2 in the CNS contributes to inflammatory pain hypersensitivity. Nature 410: 471-475, 2001. 36. Tsuda M, Shigemoto-Mogami Y, Koizumi S, et al: P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 424: 778-783, 2003. 37. Ruda MA, Bennett GJ, Dubner R: Neurochemistry and neural circuitry in the dorsal horn. In Emson PC, Rossor MN, Fonyama M (ed,): Progress in Brain Research. Amsterdam, Elsvierp, 1986, pp 219-268. 38. qodefroy F, Weil-Fugazza J, Besson JM: Complex temporal changes in 5·hydroxytryptamine synthesis in the central nervous system induced by experimental polyarthritis in the rat. Pain 28: 223-238, 1987. 39. Hylden .IL, Thomas DA, Iadarola MJ, et al: Spinal opioid analgesic effects are enhanced in a model of unilateral inflammation/hyperalgesia: possible involvement of noradrenergic mechanisms. Eur .I Pharmacol 194: 135-143, Jf)91. 40. Iadarola M.I, Brady LS, Draisci G, Dubner R: Enhancement of dynorphin gene expression in spinal cord following experimental inflammation: stimulus specificity, behavioral parameters and opioid receptor binding. Pain 35: 313-326, 1988. 41. !.l' Bars D, Dickenson AH, Besson JM: Diffuse noxious inbibitory controls (DNIC): J. Effects on dorsal horn convergent neurones in the rat. Pain 6: 283-304, 1979. 42. Calvino B, Villanueva L, Le Bars D: Dorsal horn (convergent) neurones in the intact anaesthetized arthritic rat: J. Segmental excitatory influences. Pain 28: 81-98, 1987. 43. Mayer DJ, Liebeskind JC: Pain reduction by focal electrical stimulation of the brain: an anatomical and behavioral analysis. Brain Res 68: 73-93, 1974. 44. RI'ynolds DV: Surgery in the rat dUring electrical analgesia induced by focal brain stimulation. Science 164: 444-445, 1969. 45. Beitz AJ: TIle sites of origin brain stem neurotensin and serotonin projections to the rodent nucleus raphe magnus. .I Neurosci 2: 829-842, 1982. 46. Russell 1]: Neurochemical pathogenesis of fibromyalgia syndrome. J Musculoskeletal Pain 4: 61-92, 1996. 47. Russell 1], Orr MD, Littman B, et al: Elevated cerebrospinal fluid levels of s"bstance P in patients with the fibromyalgia syndrome. Arthritis Rheum 37: 1$93-1601, 1994. 48. Schanberg LE, Lefebvre JC, Keefe FJ, et al: Pain coping and the pain experience in children with juvenile chronic arthritis. Pain 73: 181-189, 1997. 49. Bedard GB, Reid GJ, McGrath PJ, Chambers CT: Coping and self-medication in a community sample of junior high school students. Pain Res Manage 2: 151-156, 1997. 50. Lin IF, Cuskey WR, Rosenberg A: Role of self-esteem and autonomy in determining medication compliance among adolescents with juvenile rheumatoid arthritis. Pediatrics 69: 15--17, 1982. 51. Gronau RV, Whitfield MF, Petrie JH, Fryer EL: Early pain experience, child amd family factors, as precursors of somatization: a prospective study of eJUremely premature and fullterm children. Pain 56: 353-359, 1994. 52. Siegel LJ, Smith KE: Children's strategies for coping with pain. Pediatrician 16: 110--118, 1989. 53. LUmley MA, Abeles LA, Melamed BG, et al: Coping outcomes in children umdergoing stressful medical procedures: the role of child-environment variables. Behav Assess 12: 223-238, 1990. 54. Chun DY, Turner JA, Romano JM: Children of chronic pain patients: risk factOIrS for maladjustment. Pain 52: 311-317, 1993.
711
55. Gaffney A, Dunne EA: Children's understanding of the causality of pain. Pain 29: 91-104, 1987. 56. Jay SM, Ozolins M, Elliott CH, Caldwell S: Assessment of children's distress during painful medical procedures. Health Psychol 2: 133-147, 1983. 57. Miller A, Barr RG, Young SN: The cold pressor test in children: methodological aspects and the analgesic effect of intraoral sucrose. Pain 56: 175-183, 1994. 58. Merskey DM, Bogduk N: Classification of Chronic Pain: Descriptions of Chronic Pain Syndromes and Definitions of Pain Terms. Seattle, IASP Press, 1994. 59. Meinhart NT, McCaffery M: Pain: A Nursing Approach to Assessment and Analysis. New York, Appleton-Century-Crofts, 1983. 60. Eichenfield AH, Athreya BH, Doughty RA, Cebul RD: Utility of rheumatoid factor in the diagnosis of juvenile rheumatoid arthritis. Pediatrics 78: 480-484, 1986. 61. Cabral DA, Petty RE, Fung M, Malieson PN: Persistent antinuclear antibodies in children without identifiable inflammatory rheumatic or autoimmune disease. Pediatrics 89: 441-444, 1992. 62. Malleson PN, Sailer M, Mackinnon MJ: Usefulness of antinuclear antibody testing to screen for rheumatic diseases. Arch Dis Child 77: 299-304, 1997. 63. Sweet SD, McGrath PJ: Physiological measures of pain. In Flnley GA, McGrath PJ (ed,); Measurement of Pain in Infants and Children: Progress in Pain Research and Management. Seattle, !ASP Press, 1998, pp 59-81. 64. McGrath PJ: Behavioral measures of pain. In Finley GA, McGrath PJ (eds): Measurement of Pain in Infants and Children: Progress in Pain Research and Management. Seattle, !ASP Press, 1998, pp 83-102. 65. McGrath PJ, Johnson G, Goodman JT, et al: CHEOPS: a behaviol"al scale for rating postoperative pain in children. In Fields HL, Dubner R, Cervero F (eds): Advances in Pain Research and Therapy, vol 9. Proceedings of the Fourth World Congress on Pain. New York, Raven, 1985, pp 395-402. 66. Beyer .IE, McGrath PJ, Berde CB: Discordance between self-report and behavioral pain measures in children aged 3-7 years after surgery. J Pain Symptom Manage 5: 350--356, 1990. 67. Jaworski TM, Bradley LA, Heck LW, et al: Development of an observation method for assessing pain behaviors in children with juvenile rheumatoid arthritis. Arthritis Rheum 38: 1142-1151, 1995. 68. Champion GD, Goodenough B, von Baeyer CL, Thomas W: Measurement of pain by self-report. In Finley GA, McGrath PJ (eds): Measurement of Pain in Infants and Children: Progress in Pain Research and Management. Seattle, !ASP Plress, 1998, pp 123-160. 69. Jay SM, Elliott CH, Ozolins M, et aI: Behavioral management of children's distress during painful medical procedures. Behav Res Ther 23: 513-520, 1985. 70. Hester NO, Foster R, Kristensen K: Measurement of pain in children: generalizability and validity of the Pain Ladder and the Pocker Chip Tool. In Tyler DC, Krane EJ (eds): Pediatric Pain: Advances in Pain Research and Therapy. New York, Raven, 1990, pp 79-84. 71. Savedra MC, Tesler MD: Assessing children's and adolescents' pain. Pediatrician 16 (1-2): 24-29, 1989. 72. Whaley L, Wong DL: Nursing Care of Infants and Children, 3rd ed. St. Louis, CV Mosby, 1987. 73. Varni JW, Thompson KL, Hanson V: The VarnUThompson Pediatric Pain Questionnaire: I. Chronic musculoskeletal pain in juvenile rheumatoid arthritis. Pain 28: 27-38, 1987. 74. Varni]W, Waldron SA, Gragg RA, et al: Development of the Waldron/Varni Pediatric Pain Coping Inventory. Pain 67: 141-150, 1996. 75. Smith MS, Wallston KA, Smith CA: The development and validation of the Perceived Health Competence Scale. Health Educ Res 10: 51-{)4, 1995. 76. Kelly AM, Powell CV, Williams A: Parent visual analogue scale ratings of children's pain do not reliably reflect pain reported by child. Pediatr Emerg Care 18: 159-162, 2002. 77. Singer A.I, Gulla.l, Thode HC Jr: Parents and practitioners are poor judges of young children's pain severity. Acad Emerg Med 9: 609-{i12, 2002. 78. Schechter NL: The undertreatment of pain in children: an overview. Pediatr Clin North Am 36: 781-794, 1989. 79. Anthony KK, Schanberg LE: Pain in children with arthritis: a review of the current literature. Arthritis Rheum 49: 272-279, 2003. 80. Jaworski TM: Juvenile rheumatoid arthritis: pain-related and psychosocial aspects and their relevance for assessment and treatment. Arthritis Care Res 6: 187-196, 1993. 81. Schanberg LE, Anthony KK, Gil KM, Maurin EC: Daily pain and symptoms in children with polyarticular arthritis. Arthritis Rheum 48: 1390--1397, 2003. 82. Jackson DL, Moore PA, Hargreaves KM: Preoperative nonsteroidal antiinflammatory medication for the prevention of postoperative dental pain. J Am Dental Assoc 119: 641-{i47, 1989. 83. Maunuksela E-L: Nonsteroidal anti-inflammatory drugs in pediatric pain management. In Schechter NL, Berde CB, Yaster M (eds): Pain in Infants, Children, and Adolescents. Baltimore, Williams & Wilkins, 1993, pp 135--143. 84. Halperin DL, Koren G, Attias D, et al: Topical skin anesthesia for venous, subcutaneous drug reservoir and lumbar punctures in children. Pediatrics 84: 281-284, 1989. 85. McKay W, Morris R, Mushlin P: Sodium bicarbonate attenuates pain on skin infiltration with lidocaine, with or without epinephrine. Anesth Analg 66: 572-574, 1987. 86. Zeltzer LK, Jay SM, Fisher DM: The management of pain associated with pediatric procedures. Pediatr Clin North Am 36: 941-964, 1989.
712
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
87. Gonzalez ]C, Routh OK, Saab PG, et al: Effects of parent presence on children's reactions to injections: behavioral, physiological, and subjective aspects.] Pediatr Psychol 14: 449-462, 1989. 8H. Schindler M, Swann M, Crawford M: A comparison of postoperative analgesia proVided by wound infiltration or caudal analgesia. Anaesth Intensive Care 19: 4H9, 1991. 89. Porter ], lick H: Addiction rare in patients treated with narcotics. N Engl ] Med 302: 123, 1980. 90. Lander]: Fallacies and phobias about addiction and pain. Br ] Addict 85: 803-809, 1990. 91. Gaukroger PB: Patient-controlled analgesia in children. Tn Schechter NL, Berde CB, Yaster M (eds): Pain in Infants, Children, and Adolescents. Baltimore, Williams & Wilkins, 1993, pp 203-211. 92. World Health Organization: Cancer pain relief and palliative care. Geneva, World Health Organization, 1990. 93. Lambert SA: Distraction, imagery, and hypnosis: techniques for management of children's pain. ] Child Fam Nurs 2: 5-15; quiz 16, 1999. 94. Haber 0, Goodman SB: Total hip arthroplasty in juvenile chronic arthritis: a consecutive series. ] Arthroplasty 13: 259-265, 1998. 95. Schanberg LE. Anthony KK, Gil KM, et al: Family pain history predicts child health status in children with chronic rheumatic disease. Pediatrics 108: E47, 2001. 96. Bursch B, Walco GA, Zeltzer L: Clinical assessment and management of chronic pain and pain-associated disability syndrome. ] Devel Behav Pediatr 19: 45-53, 1998. 97. Eccleston C, Morley S, Williams A, et al: Systematic review of mndomised controlled trials of psychological therapy for chronic pain in children and adolescents, with a subset meta-analysis of pain relief. Pain 99: 157-165, 2002. 9H. Nash ]M, Apley ]: "Growing pains": a clinical study of non-arthritic limb pains in children. Arch Dis Child 26: 134-140, 1951. 99. Abu-Arafeh I, Russell G: Recurrent limb pain in schoolchildren. Arch Dis Child 74: 336-339, 1996. 100. Taimela S, KUjala UM: Reaction times with reference to musculoskeletal complaints in adolescence. Percept Mot Skills 75: 1075-1082, 1992. 101. Matles AI: Reflex sympathetic dystrophy in a child: a case report. Bull Hosp ]t Dis 32: 193--197, 1971. 102. YllOUS MB, Masi AT: Juvenile primary fibromyalgia syndrome: a clinical study of thirty-three patients and matched normal controls. Arthritis Rheum 28: 138-145, 1985. 103. Sherry DO, McGuire T, Mellins E, et al: Psychosomatic musculoskeletal pain in childhood: clinical and psychological analyses of 100 children. Pediatrics 88: 1093-1099, 1991. 104. Malleson PN, al-Matar M, Petty RE: Idiopathic musculoskeletal pain syndromes in children. ] Rheumatol 19: 1786-1789, 1992. 105. Sherry DO: Art overview of amplified musculoskeletal pain syndromes. ] Rheumatol 27 (Suppl 58): 44-48, 2000. 106. Wolfe F, Smythe HA, Yunus MB, et al: The American College of Rheumatology 1990 Criteria for the Classification of Fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis Rheum 33: 160-172, 1990. 107. Sheny DO: Pain syndromes. In Miller ]]l (ed): Adolescent Rheumatology. London, Martin Duntz, 1998, pp 197-227. 108. Sherry DO: Musculoskeletal pain in children. Curr Opin Rheumatol 9: 465-470, 1997. 109. Payne WK 3rd, OgilVie ]W: Back pain in children and adolescents. Pediatr Clin North Am 43: H99-917, 1996, 110. Oster.J: Recurrent abdominal pain, headache and limb pains in children and adolescents. Pediatrics 50: 429-436, 1972. 111. Buskila 0, Press], Gedalia A, et al: Assessment of nonarticular tenderness and prevalence of fibromyalgia in children.] Rheumatol 20: 368-370, 1993. 112. Clark P, Burgos-Vargas R, Medina-Palma C, et al: Prevalence of fibromyalgia in children: a clinical study of Mexican children.] Rheumatol 25: 2009-2014, 1998, 113. Malleson PN, Fung MY, Rosenberg AM: The incidence of pediatric rheumatic diseases: results from the Canadian Pediatric Rheumatology Association Disease Registry. ] Rheumatol 23: 1981-1987, 1996. 114. Bowyer S, Roettcher P: Pediatric rheumatology clinic populations in the United States: results of a 3 year survey. Pediatric Rheumatology Database Research Group.] Rheumatol 23: 1968-1974, 1996. 115. Bernstein BH, Singsen BH, Kent ]T, et al: Reflex neurovascular dystrophy in childhood.] Pediatr 93: 211-215, 1978. 116, Wilder RT, Berde CB, Wolohan M, et al: Reflex sympathetic dystrophy in children: clinical characteristics and follow-up of seventy patients, ] Bone Joint Surg Am 74: 910-919, 1992. 117. Sherry DD, Weisman R: Psychologic aspects of childhood reflex neurovascular dystrophy, Pediatrics I'll: 572-578, 1988, 118. Sherry DD, Wallace CA, Kelley C, et al: Short- and long-term outcomes of children with complex regional pain syndrome type I treated with exercise therapy. Clin] Pain 15: 218-223, 1999. 119. Guler-Uysal F, Basaran S, Geertzen ]H, Goncu K: A 2 112-year-old girl with reflex sympathetic dystrophy syndrome (CRPS type I): case report. Clin Rehab 17: 224-227, 2003, 120, Flato B, Aasland A, Vandvik IH, Forre 0: Outcome and predictive factors in children with chronic idiopathic musculoskeletal pain. Clin Exp Rheumatol 15: 569-577, 1997.
121. Reid Gj, Lang BA, McGrath PJ: Primary juvenile fibromyalgia: psychological adjustment, family functioning, coping, and functional disability. Arthritis Rheum 40: 752-760, 1997. 122. Roizenblatt S, Tufik S, Goldenberg], et al: Juvenile fibromyalgia: clinical and polysomnographic aspects. ] Rheumatol 24: 579-585, 1997. 123. Stanton RP, Malcolm ]R, Wesdock KA, Singsen BH: Reflex sympathetic dystrophy in children: an orthopedic perspective. Orthopedics 16: 773-779: discussion 779-780, 1993, 124. Song KM, Morton AA, Koch KD, et al: Chronic musculoskeletal pain in childhood. ] Pediatr Orthop 18: 576-581, 199H. 125. Goldsmith OP, Vivino FB, Eichenfield AH, et al: Nuclear imaging and clinical features of childhood reflex neurovascular dystrophy: comparison with adulLs. Arthritis Rheum 32: 480-485, 1989, 126, VoHnn E: TIle epidemiology of low back pain in the rest of the world: a review of surveys in low- and middle-income countries. Spine 22: 1747-1754, 1997. 127. Laxer RM, Allen RC, Malleson PN, et al: Technetium 99m-methylene diphosphonate bone scans in children with reflex neurovascular dystrophy. ] Pediatr 106: 437-440, 1985. 128. Sherry DD, Wallace CA: Resolution of fibromyalgia with an intensive exercise program [abstract], Clin Exp Rheumatol 10: 196, 1992. 129. Richlin DM. Carron H, Rowlingson ]C, et al: Reflex sympathetic dystruphy: successful treatment by transcutaneous nerve stimulation. .I Pediatr 9.~: 84-86, 1978. 130. Kozin F, Haughton V, Ryan L: The reflex sympathetic dystrophy syndrome in a child.] Pediatr 90: 417-419, 1977. 131. Gangi A, Dietemann ]L, Gasser B, et al: Interstitial laser photocoagulation of osteoid osteomas with use of CT guidance. Radiology 203: H43-84H. 1997. 132, Wainapel SF: Reflex sympathetic dystrophy following traumatic myelopathy. Pain 18: 345-349, 1984. 133. ]astaniah WA. Dobson S, Lugsdin ]G, Petty RE: Complex regional pain syndrome after hepatitis B vaccine.] Pediatr 143: 802-804. 2003. 134. Gedalia A, Press], Klein M, Buskila D: Joint hypermobility and fibromyalgia in schoolchildren, Ann Rheum Dis 52: 494-496, 1993, 135. Gedalia A, Garcia CO, Molina ]F, et al: Fibromyalgia syndrome: experience in a pediatric rheumatology clinic. Clin Exp Rheumatol 18: 415-419, 2000. 136. Mikkelsson M, Salminen ]], Kautiainen H: Joint hypermobility is not a contributing factor to musculoskeletal pain in pre-adolescents. .I Rheumatol 23: 1963-1967, 1996. 137. Veldman PH, Reynen HM, Arntz IE, Goris R]: Signs and symptoms of re11ex sympathetic dystrophy: prospective study of 829 patients. Lancet 342: 1012-1016, 1993, 138. Sheny DO: Disproportional musculoskeletal pain in children with juvenile rheumatoid arthritis, ] Rheumatol 27 (Suppl 58!: 71. 2000, 139. Lynch ME: Psychological aspects of reflex sympathetic dystrophy: a review of the adult and paediatric literature. Pain 49: 337-347, 1992. 140. Aasland A, Flato E, Vandvik IH: Psychosocial factors in children with idiopathic musculoskeletal pain: a prospective, longitudinal study. Acta Paediatr H6: 740-746, 1997, 141. Jones GT, Silman A], Macfarlane G]: Predicting the onset of Widespread body pain among children. Arthritis Rheum 48: 2615-2621, 2003. 142. Mikke1sson M, Sourander A, Piha], Salminen ].J: Psychiatric symptoms in preadolescents with musculoskeletal pain and tlbromyalgia, Pediatrics !OO (2 Pt 1): 220-227. 1997, 143. White KP, Harth M: An analytical review of 24 controlled clinical trials for fibromyalgia syndrome (FMS). Pain 64: 211-219, 1996. 144. Balague F, Skovron ML, Nordin M, et al: Low back pain in schoolchildren: a study of familial and psychological factors. Spine 20: 1265-1270. 1995. 145. Bnlehl S, Carlson CR: Predisposing psychological factors in the development of reflex sympathetic dystrophy: a review of the empirical evidence. Clin .I Pain 8: 287-299, 1992. 146. Brazier OK, Venning HE: Conversion disorders in adolescents: :1 practical approach to rehabilitation. Br] Rheumatol 36: 594-598. 1997, 147, Cicuttini F. Littlejohn GO: Female adolescent rheumatological presentations: the importance of chronic pain syndromes. Aust Paediatr] 25: 21-24, 19H9. 148, Prazar G: Conversion reactions in adolescents. Pediatr Rev 8: 279-286, 1987. 149. Chelimsky TC, Low PA, Naessens ]M, et al: Value of autonomic testing in reflex sympathetic dystrophy, Mayo Clin Proc 70: 1029-1040, 1995. 150. Price DO, Long S, Huitt C: Sensory testing of pathophysiological mechanisms of pain in patients with reflex sympathetic dystrophy. Pain 49: 163-173, 1992. 151. Arnold ]M, Teasell RW, MacLeod AP, et al: Increased venous alpha-adrenoceptor responsiveness in patients with reflex sympathetic dystrophy. Ann Intern Med 118: 619-621, 1993, 152. Cronin KD, Kirsner RL, Fitzroy VP: Diagnosis of reflex sympathetiC dysfunction: use of the skin potentia] response. Anaesthesia 37: H48-H52. 19H2, 153. Herrick A, el-Hadidy K, Marsh 0, Jayson M: Abnormal thermoregulatory responses in patients with reflex sympathetic dystrophy syndrome. ] Rheumato] 21: 1319-1324. 1994. 154. Procacci P, Francini F. Maresca M, Zoppi M: Skin potential and EMG changes induced by cutaneous electrical stimulation: II. Subjects with reflex sympathetic dystrophies, Appl Neurophysiol 42: 125-134, 1979. 155, Simms RW: Fibromyalgia syndrome: current concepts in pathophysiology, clinical features, and management. Arthritis Care Res 9: 315-328. 1996.
C HAP T E R
37
PAIN AND THE PAIN AMPLIFICATION SYNDROMES
156. Bou-Holaigah 1, Calkins H, Flynn JA, et al: Provocation of hypotension and pain during upright tilt table testing in adults with fibromyalgia. Clin Exp ({hcumatol 15: 239-246, 1997. 157. Martinez-Lavin M, Hermosillo AG, Mendoza C, et al: Onhostatic sympathetic derangement in subjects with fibromyalgia. J Rheumatol 24: 714-718, 1997. 158. Rush PJ, Wilmot 0, Saunders N, et al: Severe rel1ex neurovascular dystrophy in childhood. Arthritis Rheum 28: 952-956, 1985. 159. Erdmann MW, Wynn-Jones CH: "Familial" rel1ex sympathetic dystrophy syn,Jrome and amputation. Injury 23: 13~138. 1992. 160. Buskila 0, Neumann L, Hazanov 1, Carmi R: Familial aggregation in the tihromyalgia syndrome. Semin Arthritis Rheum 26: 605-611, 1996. 161. Pellegrino MJ, Waylonis GW, Sommer A: Familial occurrence of primaly ~Ibromyalgia. Arch Phys Med Rehab 70: 61-63, 1989. 162. lluskila 0, Neumann L: Fibromyalgia syndrome (FM) and nonarticular tenderness in relatives of patients with FM. J Rheumatol 24: 941-944, 1997. 163. Mailis A, Furlong W, Taylor A: Chronic pain in a family of 6 in the context of litigation.. .I Rheumatol 27: 1315-1317,2000. 164. Schanherg LE, Keefe FJ, Lelebvre JC, et al: Social context of pain in children ,vith juvenile primary fibromyalgia syndrome: parental pain history and famIly environment. Clin J Pain 14: 107-115, 1998. 165. Mailis A, Wade J: Profile of Caucasian women with possihle genetic predisposition to rel1ex sympathetic dystrophy: a pilot study. Clin .1 Pain 10: 21<)-217, 1994. 166. tunter MH, van Albada-Kuipers GA, Heggelman BG: Rel1ex sympathetic dys,trophy syndrome of one finger. Clin Rheumatol 9: 542-544, 1990. 167. Sherry DO: Pain syndromes in children. Curl' Rheumatol Rep 2: 337-342, 2000. 168. Jaworowski S, Allen RC, Finkelstein E: Rel1ex sympathetic dystrophy in a 12year-old twin with comorbid conversion disorder in both twins. J Paediatr Child Health 34: 581-583, 1998. 169. Silher 1]: Anorexia nervosa and rel1ex sympathetic dystrophy syndrome. Psychosomatics 30: 108-111, 1989. 170. Silher 1]: Eating disorders and rel1ex sympathetic dystrophy syndrome: is there a common pathway? Med Hypoth 48: 197-200, 1997. 171. Okifuji A, Turk DC, Sinclair JD, et al: A standardized manual tender point survey: l. Development and determination of a threshold point for the identification of positive tender points in fibromyalgia syndrome. J Rheumatol 24: 377-383, 1997. 172. Hollingworth P: Back pain in children. Br,l Rheumatol 35: 1022-1028, 1996. 173. Waddell G, McCulloch .lA, Kummel E, Venner RM: Nonorganic physical signs in low-back pain. Spine 5: 117-125, 1980. 174. Nickeson R, Brewer E, Person 0: Early histologic and radionuclide scan changes in children with rel1ex sympathetic dystrophy syndrome (RSDS) [abstract]. Anhritis Rheum 28: S72, 1985. 175. Melzack R: The McGill Pain Questionnaire: major propenies and scoring methods. Pain 1: 277-299, 1975. 176. Schanberg LE, Keele FJ, Lefehvre jC, et al: Pain coping strategies in children with juvenile primary fibromyalgia syndrome: correlation with pain, physical function, and psychological distress. Anhritis Care Res 9: 89-96, 1996. 177. Apley J: One child. III ApleyJ, Ounsted C (eds): One Child. Philadelphia, JB Lippincott, 1982, pp 23-47. 178. Ashwal S, Tomasi L, Neumann M, Schneider S: Reflex sympathetic dystrophy syndrome in children. Pediatr Neurol 4: 38-42, 1988. 179. Lemahieu RA, Van Laere C, Verbruggen LA: Ret1ex sympathetic dystrophy: an underreponed syndrome in children? Eur.l Pediatr 147: 47-50, 1988.
713
180. Fialka V, Schimmerl S, Schurawitzki H. et al: Comparative clinical, roentgenologic, scintigraphic and nuclear magnetic resonance tomography studies in sympathetic ret1ex dystrophy. Wien Med Wochenschr 141: 383-388, 1991. 181. Masciocchi C, Fascetti E, Michelini 0, et al: Ret1ex sympathetic dystrophy syndrome: contribution of magnetic resonance. Radiol Med 74: 408-412, 1987 182. Ruggeri SB, Athreya BH, Doughty R, et 'II: Rel1ex sympathetic dystrophy in children. Clin Otthop (163): 225-230, 1982. 183. Silber 1], Majd M: Reflex sympathetic dystrophy syndrome in children and adolescents: report of 18 cases and review of the literature. Am J Dis Child 142: 1325-1330, 1988. 184. Stilz RJ, Carron H, Sanders DB: Ret1ex sympathetic dystrophy in a 6-year-old: successful treatment by transcutaneous nerve stimu lation. Anesth Analg 56: 438-443, 1977. 185. Lightman HI, Pochaczevsky R, Aprin H, I10wite NT: Thermography in childhood rel1ex sympathetic dystrophy. J Pediatr 111: 551-555, 1987. 186. Kesler RW, Saulsbury FT, Miller LT, Rowlingson .IC: Reflex sympathetic dystrophy in children: treatment With transcutaneous electric nerve stimulation. Pediatrics 82: 728-732, 1988. 187. Hood-White R, Gainor J: Ret1ex sympathetic dystrophy in an 8-year-old: successful treatment by physical therapy. Onhopedics 20: 73-74, 1997. 188. Lee BH, Scharff L, Sethna NF, et al: Physical therapy and cognitive-hehavioral treatment for complex regional pain syndromes. .1 Pediatr 141: 135-140, 2002. 189. Alioto .IT: Behavioral treatment of rel1ex sympathetic dystrophy. Psychosomatics 22: 539-540, 198!. 190. Schulman JL: Use of a coping approach in the management of children with conversion reactions. .1 Am Acad Child Adoles Psychiatry 27: 785-788 1988. 191. Russell1J: Fibromyalgia syndrome: approaches to management. Bull Rheum Dis 45: 1-4, 1996. 192. Siegel OM, Janeway 0, Baum,l: Fibromyalgia syndrome in children and adolescents: clinical features at presentation and status at follow-up. Pediatrics 101: 377-382, 1998. 193. Romano TJ: Fibromyalgia in children; diagnosis and treailllent. W V Med J 87: 112-114, 1991. 194. Anthony KK, Schanberg LE: Juvenile primary fibromyalgia syndrome. Curr Rheumatol Rep 3: 165-171, 200!. 195. Waleo GA, I10wite NT: Cognitive-behavioral intervention for juvenile primary fibromyalgia syndrome. J Rheumatol 19: 1617-1619, 1992. 196. Vandvik 1H, Forseth KO: A bio-psychosocial evaluation of ten adolescents with fibromyalgia. Acta Paediatr 83: 76~771. 1994. 197. Eccleston C, Malleson PN, Clinch J, et al: Chronic pain in adolescents: evaluation of a programme of interdisciplinary cognitive hehaviour therapy. Arch Dis Child 88: 881-885, 2003. 198. Sherry DO: Amplified Musculoskeletal Pain in Children: Diagnosis and Treatment. A Guide for Physical and Occupational Therapists. Wilmington, DE. Childhood RND Educational Foundation, 2002. 199. Buskila 0, Neumann L, Hershman E. et a1: Fibromyalgia syndrome in children: an outcome srudy. J Rheumatol 22: 525-528, 1995. 200. Rabinovich CE, Schanherg LE, Stein LD, Kredich OW: A follow up study of pediatric fibromyalgia patients [abstract!. Arthritis Rheum 33 (SuppD: S146, 1990. 201. Wong 0, Whaley L: Clinical Handhook of Pediatric Nursing, 3rd ed. St. Louis, Mosby, 1990.
( HAP T E R
38
SKELETAL MATURATION AND BONE MINERALIZATION IN THE PEDIATRIC RHEUMATIC DISEASES Rolando Clmaz and Fernanda Faldnl
~lJ!
NORMAL SKELETAL MATURATION An understanding of bone biology is essential to an appreciation of skeletal growth and adaptation to mechanical stresses throughout life. l -4 Bone can be divided into two primary types: cortical bone, which consists of compact bone in the long bones of the appendicular skeleton; and trabecular bone, which is the primary component of vertebral bodies and the flat bones of the skull and pelvis. s Trabecular bone has a much greater surface than cortical bone and is metabolically more active. Bones change remarkably in size, shape, and microstructure throughout the period of growth. 6•7 Bones of the appendicular and axial skeletons develop initially by ossification of preexisting cartilage from mesenchymal condensations in the embryo (enchondral ossification). In contrast, bones of the face, skull, and initially the mandible and clavicle develop by ossification of fibrocellular tissue (membranous ossification). All axial and appendicular bones also undergo secondary membranous ossification-the diaphyseal cortex is continuously modified by periosteal new bone apposition.
Anatomy of Bone Long bones consist of four parts. The diaphysis is the long tubular midportion of bone that ends in the metaphysis, the flared portion of bone that is separated from the epiphysis by the growth plate or physis. At birth, the diaphysis is relatively short. It grows in length by enchondral ossification. Cartilage cells proliferate toward the ends of bones; those closest to the middle of the bone ossify. Periosteal deposition of new bone increases the diameter of the diaphysis. The newborn diaphysis consists of laminar bone that lacks the Haversian canal system characteristic of mature bone; as the child ages, the intercellular matrix increases, porosity decreases, and the hardness of the bone increases. H All epiphyses, except that of the distal femur, are completely cartilaginous at birth. The cartilage is gradually replaced by bone
716
until only the articular cartilage remains unossified in the mature skeleton. The physis is a cellular zone in which mitoses are frequent and new cells are being formed. Factors influencing growth at the physis include thyroxine, growth hormone, and testosterone. Growth hormone and insulin-like growth factor I (IGF-I) act together to facilitate the achievement of peak bone mass during puberty. Testosterone stimulates the physis to undergo rapid cell division with resultant physeal widening during the growth spurt (the anabolic effect) but eventually slows growth (androgenic effect). Estrogens suppress the growth rate by increasing calcification of the matrix, a prerequisite to epiphyseal closure. The relative contributions of individual physes to limb length are summarized in Table 38-1. Growth of the appendicular skeleton ceases with completion of ossification of the iliac apophyses, although height of the vertebral bodies may continue to increase and contribute to overall height of the body until the third decade of life. 9 Skeletal bone age can be determined by radiographic identification of the onset of secondary ossification in the long bones and by physeal closure. In general, ossification centers appear earlier, and physes fuse earlier, in girls than in boys.
Bone Minerai Metabolism The complex structure and composition of bone is directly related to the two primary functions of the skeleton: to support the tissues of the body in order to permit locomotion and to provide a reserooir qf ions critical to metabolic functions. W-B Bone is composed of 70% mineral and 300/0 organic constituents. Hydroxyapatite, constituted primarily of calcium and phosphorus, accounts for 950/0 of the mineral content. Magnesium, present in smaller amounts, is also important in homeostasis. The organic component consists of 98% matrix which is predominantly type I collagen. Noncollagenous proteins, such as osteocalcin, fibronectin, osteonectin, and
C HAP T E R
C.
38
SKELETAL MATURATION AND BONE MANIFESTATION
IABLE 38-1 Relative Contributions of Individual Physes 10 I enyth of the Bone and limb
Contribution to Total Growth (%) Growth Area Humerus Radius/ulna Femur Tibia/fibula
Of Bone
Of Limb
80
40 10 10 40
Proximal Distal Proximal Distal Proximal Distal Proximal Distal
20 20
80
30
15
70
40 27 18
55 45
Data from Ogden JA: Skeletal Injury in the Child. Philadelphia, Lea & Febiger, 1982.
osteopontin, make up 5% of the matrix, Cells occupy the remaining 2% of the organic component of bone, and are responsible for formation, resorption, and maintenance of the remodeling cycle, Osteoclasts are derived from mononuclear cells and resorb bone, Osteoblasts form osteoid and osteoid matrix, Osteocytes differentiate from osteoblasts and maintain the integrity of bone through a network of canaliculi.
Biochemical Markers of Bone Formation and Resorption Bone turnover in a growing skeleton is a linked phenomenon of facilitating bone formation and limiting bone resorption in order for skeletal growth to occur. Studies of bone mineral metabolism generally assay a specific set of markers of bone formation and resorption in blood or urine. I 4-16 Table 38-2 summarizes the principal characteristics of commonly used biochemical markers of bone remodeling. However, there are many
'1
II
lABLE 38-2
confounding factors in using these measures (e.g. urinary acidity, medications, magnesium concentration, renal function), Moreover, additional difficulties are intrinsic in the interpretation of pediatric measurements, mainly because these markers reflect growth as well as remodeling. Therefore, geographic reference data for age, sex, and ethnicity are essential. Although these markers cannot be used for the diagnosis of osteoporosis, they are important in the study of bone turnover in pathologic conditions, and they can be useful in the follow-up of patients during antiosteoporotic treatment, for evaluation of compliance, and possibly of prognosis, as well. Measures of bone formation include the activity of bone-specific alkaline phosphatase, which is released during osteoblastic activity. Osteocalcin (also called bone-gla protein) is a vitamin K-dependent, y-carboxylated protein derived from osteoblasts. Its serum concentration reflects that portion of newly synthesized protein that does not bind to the mineral phase of bone and is released into the circulation. Serum carboxylterminal propeptide oftype Iprocollagen (PICP) is also marker of bone formation. 17 PICP is a globular protein that is cleaved by a specific peptidase at the C-terminal end of the procollagen triple helix. Its concentration in blood directly reflects the number of collagen fibrils that have been formed. Plasma tartrate-resistant acid phosphatase (TRAP) is a marker of bone resorption. 18 ,19 This labile enzyme is released during osteoclastic activity. The urinary concentration of the deoxypyridinoline cross-linked telopeptide of type I collagen represents hydroxylysyl and lysyl post-translational components of the cross-linkage of type I collagen that stabilize the molecule. It is measured in the urine in relation to the concentration of creatinine. These cross-links are reflective of mature collagen
a
Bio(hemical Markers of Bone Remodeling
Desalptlon
Marker ~ers
717
of Bone Fonnatlon
Alkaline phosphatase (ALP) Osteocalcin Prpcollagen type I propeptides
Enzyme secreted by osteoblasts, but also by other cells (e.g. liver, gut, kidneys). In children, about 80% of ALP is derived from bone. Bone-specific ALP is a constituent of osteoblast membrane and can be assayed in serum (no circadian variations). Small, noncollagenous protein synthesized by osteoblasts and chondrocytes and deposited in the extracellular bone matrix. A small amount enters the circulation and can be measured in serum. It is a sensitive and specific marker of bone formation N-terminal (PINP) and C-terminal (PICP) extension peptides are cleaved during the extracellular processing of type I collagen, before fibril formation, and can be measured in serum.
Markers of Bone Resorption T<\Itrate-resistant acid phosphatase (TRAP) Hydroxyproline Collagen cross-links
Collagen type IN-terminal (Ntx) and <:-terminal (Ctx or ICTP) telopeptides
Enzyme present in the osteoclast and released during osteoclastic activity. Serum TRAP is not bone specific. Amino acid found in collagenous proteins of bone and other soft connective tissues. A product of post-translational hydroxylation of proline in the procollagen chain. Can be measured in urine but is not specific (can be released by noncollagenous proteins and dietary proteins). Pyridinoline (Pyr) and deoxypyridinoline (DPyr) are generated from lysine and hydroxyiysine during post-translational modification of collagen. They are released during matrix resorption and excreted in urine but new assays are available for serum determination. Of the two, DPyr is more specific for bone. Derived from degradation of type I collagen. Ntx is more sensitive. Both can be measured in serum and Ntx also in urine.
718
C HAP T E R
38
SKELETAL MATURATION AND BONE MANIFESTATION
breakdown and are also a marker of bone resorption. 20 Deoxypyridinoline is found in large amounts only in type I collagen; therefore, its urinary excretion reflects the metabolic breakdown of that molecule. Urinary hydroxyproline has been used similarly. The urinary calcium/creatinine ratio is also a marker of bone resorption.
Calcium-Regulating Hormones Assessment of bone mineral metabolism includes assays for calcium-regulating hormones such as parathyroid hormone (pm), 25-hydroxyvitamin D] [25-(OH)D), and 1,25-dihydroxyvitamin D] [J,25-(OH)P}21 The primary function of PTH is to maintain the ionized calcium concentration of the blood within a narrow physiologic range. Hypocalcemia results in stimulation of PTH secretion, whereas hypercalcemia suppresses its secretion. PTH regulates calcium homeostasis by acting on the major calcium reservoir of the body, the skeleton. It stimulates osteoclastic activity and thereby bone resorption. It also stimulates the conversion of 25-(OH)D3 to 1,25(OH)P3' The principal source of 25-(OH)D3 is dietary vitamin D2. Ultraviolet light also endogenously stimulates the production of vitamin D) from 7-dehydrocholesterol in the skin. 25-(OH)D 3 is biologically inactive and is hydroxylated in the kidney to the 1,25-(OH)p hormone (see Fig 32-2). This hormone, calcitriol, stimufates intestinal absorption of calcium, thereby elevating the serum calcium concentration. Receptors for 1,25-(OH)2D3 are present on intestinal cells. Care must be taken in interpreting the results of measurement of the vitamin D hormones, because diet, malnutrition, the presence of diseases leading to malabsorption or a catabolic state, and geographic location and season of the year (sun exposure) influence the results.
The RANK, RANK-L, Osteoprotegerin System The osteoclast, a cell derived from the monocyte/ macrophage hematopoietic lineage, is one of the pivotal effectors in bone resorption. Discovery of the RANK signaling pathway in the osteoclast provided insight into the mechanisms of osteoclastogenesis and activation of bone resorption. 22-29 Osteoprotegerin (OPG), receptor activator of nuclear factor-KB (RANK), and the RANK ligand (RANKL) are part of a family of biologically related tumor necrosis factor receptor (TNFR)/TNF-like proteins that regulate osteoclast function. RANK, a transmembrane signaling receptor, is mainly expressed by monocytes and macrophages; it is essential for osteoclast differentiation and activation, and therefore for bone resorption. Its activation depends on binding with RANKL. OPG is a soluble protein that acts as a decoy receptor of RANKL, inhibiting osteoclast differentiation and activation, and thereby reduces bone resorption. To maintain bone homeostasis, balance in the RANKL, RANK, OPG system is required. The mature osteoclast, in response to activation of RANK by RANKL, undergoes internal structural changes that enables it to resorb bone.
Skeletal Maturation and Peak Bone Mass Bone mineralization during childhood and adolescence is highly correlated with anthropometric parameters such as age, weight, height, and Tanner stage. 3D--33 Rapid skeletal accretion occurs during intrauterine growth of the fetus and the early months of infancy. Thereafter, skeletal growth is linear throughout childhood. Before puberty, there is no substantial difference between boys and girls in bone mass of the axial or appendicular skeleton. Early puberty and adolescence are characterized by accelerated skeletal maturation and account for at least 40% of the total adult skeletal mass. During this period, sex differences in bone mass become expressed;3H6 skeletal growth correlates closely with sexual maturation, because epiphyseal closure is under hormonal control. In North America, girls reach puberty at approximately 11.15 ± 1.10 years of age. Menarche occurs soon after, at 13.5 ± 1.02 years, and epiphyseal closure is complete at approximately 16 years. Puberty in boys begins Y, year later, at 11.64 ± 1.07 years, and lasts approximately 1 year longer than in girls. Lumbar bone mineral density (BMD) increases 35% to 700/0 during adolescence in both boys and girls, but a more prolonged bone maturation period in males results in a larger increase in bone size and cortical thickness. 37-41 Several studies have documented the critical importance of puberty in achievement of peak bone mass, and evidence suggests that bone mass accrual in females essentially ceases within a few years after menarche. 42-46 Also, it has been reported that the age at onset of menstruation might predict BMD in the lumbar spine. 47
Peak bone mass is defined as the level of bone mass achieved at the end of skeletal maturation. The precise age in normal individuals at which peak bone mass is reached is still uncertain and may be depend on the site studied and the method used. 48 Studies by dual x-ray absorptiometry (DEX) indicate that peak bone mass is reached for the most part in late adolescence, within the second decade of life. At the lumbar spine, areal BMD progressively increases during childhood and adolescence, reaching a plateau at approXimately 15 and 17 years in girls and boys, respectively. At the femoral neck, areal BMD reaches a peak at approximately 14.5 years in girls and 16.5 in boys; thereafter, it remains stable or declines slightly. At the distal one third of the radius, BMD, assessed by single-photon absorptiometry, progressively increases up to 18 to 19 years in boys and 15 to 16 years in girls, but a slight additional increase may be evident until the third decade of life. 35 .49 The skull mass never peaks, increasing throughout life, as a result of continuous periosteal expansion. Total body BMD continues to increase modestly after epiphyseal closure in boys and plateaus in girls. 39 A recent studySo indicated that the age of attaining peak bone mass at the hip is younger than at the spine, and bone mineral content (BMC) and bone surface area at the spine might continue to increase throughout the early 30s in females. BMD was assessed by DEX in 300 healthy females aged 6 to 32 years. At the spine, femoral neck, greater trochanter, and Ward's triangle, the highest BMD level was observed at 23.0 ± 1.4, 18.5 ± 1.6, 14.2 ± 2.0, and 15.8 ± 2.1 years, respectively.
C HAP T E R
38
SKELETAL MATURATION AND BONE MANIFESTATION
Determinants of Bone Mass Determinants of peak bone mass include intrinsic factors, such as heredity, sex, and hormones,5l extrinsicfactors, such as nutrition (calcium, vitamins, calories, protein) and mechanical influences related to body weight and physical activity.52-54 In is estimated that heredity determines up to 75% to 85% of the skeletal mass.55-59 Environmental variables, incluaing those related to endocrine and nutritional influences, mechanical forces, and risk factors, account for the r~mainder. The genetic basis at the molecular level by whicn bone mass and strength are determined have yet to btl fully elucidated. Many genes may be involved, and sevetal polymorphisms have been implicated, including thos~ for interleukin-6 (lL-6), vitamin D receptors, calcitonm receptor, transforming growth factor-~ (TGF-~), estrogen receptor-a, osteocalcin, apolipoprotein E, OPG, androgen receptor, osteopontin, osteonectin, and type I coll~gen.59.60 Data suggest that genetic variations at multiple genetic loci are important in bone accrual, and that the combination of genotypes at several loci may have a majOr role in determining BMD and BMC. 60 Among extrinsic factors, an adequate intake of calcium and vitamin D is a relatively important factor in achievement of peak bone mass. 61-<>1 Calcium is, however, a threshold nutrient.65 AdcUtion of calcium citrate malate (approximately 1000 mg/day) to the diet over a 3-year period produced measurable increases in both appendicular and axial bone density in the supplemeQted twin of 70 pairs of monozygotic twins. 66 In the treatmeQt group, significant incremental bone formation at several mea~ured sites of cortical and trabecular bone was documented in 22 prepubertal children but did not occur in 23 postpubertal or pubertal children.
Vitamin D intake in peripubertal girls is also important in achievement of optimal peak bone mass,67 and polymorphisms in vitamin D receptor genes are important in the achievement of optimal BMD.68-70 Finally, skeletal loading associated with body weight is an additional crucial, variable that is an independent determinant of peak
BMD. n - 73
DIAGNOSTIC METHODS DeJlnltion of Osteoporosis and Osteopenla
an~ Measurement of Bone Density
Osteoporosis is a disorder characterized by parallel loss of matrix and BMC and microarchitectural deterioration of bone tissue. Osteopenia is a low bone mass for age or, more specifically, for skeletal age and stage of sexual maturation. Osteoporosis and osteopenia in adults are defined at levels relative to a "normal" young adult population. The World Health Organization (WHO) developed criteria for the diagnosis of osteoporosis in 1994. These criteria assess the risk of osteoporosis by determinin~ the number of standard deviations between normal peak bone mass and BMD measured with DEX at any
719
skeletal site. This "T score" defines normal bone density (T score between 0 and -1), osteopenia (between -1 and -2.5), and osteoporosis (less than -2.5),74 There are currently no accepted definitions for osteoporosis and osteopenia in childhood, because the WHO definition is based on the T score (obtained by comparison with young adults) and not on a Z score (obtained by comparison with age- and sex-matched geographic controls). In children and adolescents who have not yet achieved peak bone mass, BMD should be referred to as a Z score (see Chapter 6), which is calculated by the following formula: (BMD of the patient - mean BMD of the control group) + standard deviation ofthe control group. In contrast to adults, there are no studies that identify a fracture threshold in children for any specific Z score. Bone mass depends on both the size and density of skeletal bone; and osteoporosis is usually determined relative to bone density. However, with current diagnostic techniques such as DEX, it is not possible to measure true bone density, because this method measures only a cross-sectional area of the scan, not a true volume. To measure the true density, the depth (volume) of the bone would need to be known and taken into account.
Dual-Energy X-Ray Absorptlometry Absorptiometry is a means of assessing BMC or density of various areas of the skeleton. Single-photon absotptiometry uses an iodine 125 source and is restricted to measuring BMC at the one-third and one-tenth distal radial sites of the forearm. This technique is therefore used primarily for assessment of the appendicular skeleton. Although bone width can be calculated by single-photon absorptiometry, estimations of BMD are not accurate. Dual-photon absorptiometry uses a gadolinium source with photons of 44 and 100 KeV measured simultaneously in order to calculate BMC or BMD of any area of the skeleton by a computerized subtraction of tissue density from that of bone. Although this method is applicable to children and is accurate, measurement time is prolonged, at least 35 to 45 minutes for a complete skeletal survey, and therefore this technique is no longer in use. The technique that is most appropriate for children because of low radiation (= 3 mrem), speed, and accuracy is DEX. DEX employs two beams, of 70 and 140 KeV, to distinguish soft tissue from bone (Fig. 38-1). The introduction of DEX has materially increased knowledge of skeletal maturation and the pediatric diseases that affect bone metabolism. The ratio of cortical to trabecular bone differs in various parts of the skeleton; therefore DEX measures a composite of trabecular and cortical bone mass. Sites often measured clinically by absorptiometry include the one-third distal radius (95% cortical and 5% trabecular), the one-tenth distal radius (25% cortical and 75% trabecular); the lumbar vertebral bodies (5% cortical and 95% trabecular); the femoral neck (75% cortical and 25% trabecular); and the greater trochanteric area of the femur (50% cortical and 50% trabecular). DEX has been widely accepted as a noninvasive method for measurement of BMD. However, this diag-
720
C HAP T E R
38
SKELETAL MATURATION AND BONE MANIFESTATION
to adjust for bone size can therefore lead to erroneous interpretations ofDEX values. Moreover, with BMD measurements in children there are general problems that are not specific to the diagnostic technique. For example, when interpreting clinical results, one must consider that in growing children even a stable BMD value (i.e., a lack of an increase during a period of skeletal growth) represents an abnormality of bone metabolism. Also, areal BMD should be interpreted not only in relation to age-matched controls, but also with corrections for height and weight, as well as geographic matching. Unfortunately, published pediatric standards for BMD are limited, and the lack of normal control values often makes the interpretation of data difficult. Finally, pubertal status (Tanner stage), which frequently is delayed in children with chronic rheumatic diseases, is fundamental in the process of bone acquisition and should always be taken into account when comparing patients and controls.
Other Diagnostic Methods
• Figure i8-1 Dual energy x-ray absorptiometry (DEX) images of an 8-year-old girl with juvenile rheumatoid arthritis. A, Whole body. B, Radius. C, Vertebral bodies Ll through L4. BMC, bone mineral content; BM D, bone mineral density; UD, ultradistal.
nostic procedure has some limitations in childhood, especially because it measures only a two-dimensional computation of BMC (Le., divided by surface area): the areal BMD. A three-dimensional estimate of skeletal density can be obtained when both anteroposterior and lateral measurements are correlated for skeletal size (cubic centimeters), the volumetric BMD. The latter value is seldom available in published data. Areal BMD results as obtained from DEX are influenced by bone size. The growth of an individual can increase the areal BMD without any actual increase in true bone density; failure
111" ....
TABLE 38-1
Evaluation of mineralized bone mass using simple radiography is insensitive. Bone mass may have already diminished by 30% to 40% by the time osteoporosis is detectable on conventional x-ray films. Several additional noninvasive methods for measuring bone mass and mineml content have been developed. Table 38-3 includes the characteristics of the most commonly used methods of measuring bone density. Quantitative computed tomography is applicable to measurement of the axial skeleton with a radiation exposure comparable to that of plain radiography. Peripheral quantitative computed tomography has lower radiation exposure and permits analysis of volumetric bone density of appendicular cortical and trabecular sites. Quantitative high-frequency sonography (ultrasonography) is a new and noninvasive method of estimating bone quality. This radiation-free procedure measures the transmission of ultrasound waves through bones and has been proposed for the assessment of bone density. Two parameters can be simultaneously determined from the measured signal: speed of sound and broadband ultrasound attenuation (BUA). BUA measures the loss of
{olllpdrisoll of Methods of BOllI' Dellsity Medslllellwllt
Avel1lge nme for Method
Site
Dose (mrem)
Scanning (min)
SPA DPA DEX
Radius Lumbar spine. femur Lumbar spine, hip, radius. total body Lumbar spine Phalanges, heel, tibia
5-18 1-15
15
QCT US
1-3 (0.1 if peripheral, such as distal radius) 100-1000
o
20-45 <5 for lumbar spine 10-20 1-2
Comments Rarely used Rarely used Gold standard (best method available today) True volumetric density can be measured Advantages: ease of scan; fast; no radiation; inexpensive; portable. Disadvantages: operator dependent; needs standardization and more reference data
SPA, single-photon absorptiometry; DPA, dual-photon absorptiometry; DEX. dual-energy x-ray absorptiometry: QCf. quantitative computed tomography; US. ultrasonography.
C HAP T E R
38
SKELETAL MATURATION AND BONE MANIFESTATION
sound caused by bone as a function of frequency; in norm~l adults and children, it may be the parameter that demohstrates the highest correlation with BMD determined by DEX. In addition, sonographic measurements of bone may provide additional information about bone quality, such as stiffness and elasticity. Normative values for healthy children have been published.75.76 Also, pilot studies in children with rheumatic diseases have provided relatively good correlations between ultrasonographic bone density measured in the calcaneum77 or at the midtibia78 and BMD determined by DEX, supporting the clinical utility of ultrasound densitometry. This technique is a promising tool to assess bone mass and quality in children in view of the absence of radiation exposure, low cost, and portability of the equipment,79-83 BON~ MINERALIZATION IN THE RHEUMATIC DISEASES
Low: Bone Mass In Children with Chronic Arth,.tls Failure to develop adequate bone mineralization is common in children with chronic arthritis. Juxta-articular osteqpenia can be evident in plain radiographs even in early disease, whereas diffuse osteopenia or osteoporosis can develop later and lead to the risk of vertebral collapse and long-bone fractures after minimal trauma. Multiple risk factors are known to be associated with decreased bone mass (Table 38-4), and many studies have been published on this subject,84-103 Active arthritis has an osteopenic effect, both around affected joints and systemically, by means of a complex and still partly unknown network of pro-inflammatory cytokines. In particular, IL-6 is known to have a profound effect on bone metabolism. 104,105 Pepmueller and colleagues98 measured BMC and BMD in 41 children with juvenile rheumatoid arthritis (JRA) and 62 healthy geograPhic controls and analyzed serum markers of bone metabolism. Decreased BMD was found at all sites in the patient group with a negative correlation between measures of disease severity and bone mass. These researchers hypothesized that decreased mineralization rather than increased resorption was the primary pathophysiologic mechanism. However, the balance between bone formation and resorption is controlled by a variety of factors, and further studies on this subject have yielded cont1icting results. 96 •106 Although BMD may be decreased at all
f
II lABt E 38-4 Risk Factors for Osteoporosis in Children with ( Ilronic Arthritis
Active inflammatory disease Glucocorticoid treatment De{Teased mobility Protein/calorie malnutrition Inadequate calcium/vitamin D intake Derreased sun exposure Decreased height and weight Pubertal delay
721
sites in children with arthritis, the appendicular skeleton is predominantly affected. 107 Henderson and coworkers 102 evaluated predictors of BMD in prepubertal patients with JRA who had not been treated with glucocorticoids. Almost 30% of mild to moderately ill patients had low total body BMD. Parameters of disease severity (number of swollen joints, articular severity score, erythrocyte sedimentation rate) exerted a negative effect on bone mineralization. In another study of postpubertal females who had never received systemic glucocorticoids, approximately 30% of the subjects with mild to moderate disease demonstrated low bone mass. 103 A stepwise logistic regression model was used to identify contributing factors, and the only variable that significantly contributed to BMC was lean body mass, which accounted for the majority of the variance in total body BMC. This decreased lean body mass could be the result of altered body composition, which often occurs in chronic inflammatory arthritis.
Effects of Childhood Arthritis on Bone Mass In Adulthood Diseases occurring during childhood and adolescence that are associated with a loss of bone mass may predispose patients to premature osteoporosis and fractures during adulthood, if the predetermined peak bone mass is not established during skeletal maturation by late adolescence. Therefore, chronic inflammatory diseases can potentially have a detrimental effect on future BMD and fracture risk. Zak and colleagues J08 assessed BMD of the hip and spine in 65 young adults (mean age, 32.2 years) with a history of juvenile chronic arthritis. They found that BMD was significantly lower in these patients than in age-, sex-, height-, and weight-matched healthy controls. Moreover, significantly more patients than expected had osteopenia and osteoporosis. Factors associated with a lower BMD included active disease at the time of the study, baseline erosions, higher Steinbrocker functional class, polyarticular course, and chronic corticosteroid treatment. The presence of juvenile chronic arthritis by itself explained about 20% of BMD variation. French and coworkersJo'J also determined the extent of osteopenia in a population-based cohort of adults with a history of JRA. Forty-one percent of the patients had a T score of 1 or lower at either the lumbar spine or the femoral neck. In another study, lIO the impact of disease activity on peak bone mass was assessed in 229 young adults in their mid-20s with juvenile arthritis at a mean of 15 years after disease onset. Patients with persistent disease had a significantly lower BMD than did healthy subjects, whereas patients whose disease was in remission had overall a normal bone mass. However, even in women with only a history of arthritis in childhood, total body BMD was significantly lower, although this was not true for the lumbar spine or radius. In a later report,1II a large proportion (41%) of adolescents with early-onset disease were found to have a low bone mass more than 10 years after onset, and their low BMC was related to the duration of active disease, disease severity, measures of bone resorption, and anthropometric parameters such as height and weight.
Other Connective TIssue Diseases Children and adolescents with systemic lupus erythematosus (SLE), juvenile dermatomyositis (JDM), and the vasculitides are also at risk for the development of
722
C HAP T E R
38
SKELETAL MATURATION AND BONE MAN1FESTATION
osteopenia and osteoporosis, both from the disease itself and from its medical treatment. Glucocorticoid therapy, often in high doses and for prolonged periods, is the basis of treatment for most of these disorders. Avoidance of sun exposure and limited mobility in these patients can also contribute to decreased mineralization. A recent study evaluated 15 patients with JDM and found low BMD values in the majority and persistent or worsening osteopenia in patients with ongoing active disease. lJ2 In SLE, despite evidence that in adults osteopenia is common,113 scant pediatric data exist.114 In addition to the same risk factors that exist for chronic arthritis, renal impairment, reduced sun exposure, and endocrine dysfunction in SLE can all contribute to a low bone mass.
Effect of Drug Treatment on Bone Minerai Density The relationship between glucocorticoid therapy and vertebral fractures in the rheumatic diseases of childhood is well known,us Figure 38-2 shows a vertebral collapse in a young patient with SLE who had been treated with oral prednisone. In fact, steroids have profound effects on the skeleton and on bone formation, mainly through
a depletion of bone-forming cells and a decrease in their function. 116 Impaired osteoblastic-cell differentiation, associated with an increase in apoptosis, causes a decrease in bone-forming cells. Moreover, glucocorticoids shift the differentiation of stromal cells away from the osteoblastic and toward the adipocytic lineagei117 in addition to causing depletion of mature osteoblasts, they inhibit the function of differentiated cells. Histomorphometric analysis of bone biopsies from patients receiving glucocorticoids demonstrate increased bone resorption as wellya Significant bone loss occurs after the initial exposure to steroids, and even modest doses of these agents increase the risk of fractures. 119 However, it is important to note that patients taking glucocorticoids often have an underlying inflammatory disease that leads to bone resorption. Among other effects, glucocorticoids decrease gastrointestinal calcium absorption and increase its urinary excretion, leading to the development of secondary hyperparathyroidism. They also affect insulin-like growth factors (IGFs), mainly decreasing synthesis of IGF-I, and inhibit the synthesis of type I collagen, with a consequent decrease in bone matrix available for mineralization. 120 Recent research has provided new insights into the mechanisms of these glucocorticoid actions at the cellular and molecular levels.121-124 Another important effect on bone is mediated by RANKL and OPG. Glucocorticoids increase the expression by human osteoblastic and stromal cells in culture of RANKL and decrease that of OPG.125 The osteopenic effect of methotrexate (MTX) has been described in children with malignancies treated with high-dose protocols and confirmed by in vitro studies. 126-129 However, lower-dose MTX is not associated with this osteopenic effect.102.103.130 Most likely, the significant beneficial effect on arthritis counterbalances in vivo the demonstrated inhibitory effect on osteoblasts.
PRIMARY AND SECONDARY BONE MINERALIZATION DISORDERS Osteoporosis in an otherwise healthy child or adolescent is rare, although idiopathic osteoporosis is a well-known entity. Rather, pediatric osteoporosis is increasingly recognized in the setting of acute or, more often, chronic illness. Table 38-5 summarizes primary and secondary pediatric disorders associated with low bone mass.
PREVENTION
• Alllre 38-2 Vertebral crush fractures in a girl with systemic lupus erythematosus treated with oral prednisone for 3 years.
Prevention of osteopenia and osteoporosis during the critical developmental stages of skeletal maturation is fundamental in order to decrease osteoporosis and fracture risk later in life. Optimal dietary calcium intake is important, because it accounts for 5% to 10% of the variance in peak bone mass. An adequate calcium intake is recommended for all children, according to the published guidelines (800 mg/day from 1 to 5 years of age, 1200 mg/day from 6 to 10 years, and 1500 mg/day from
C HAP T E R
. : . U\HI f lX-5
38
SKELETAL MATURATION AND BONE MANIFESTATION
Ppdi"hi( Hbord('rs Assoddted with Low Bone
723
M.lS<,
Pathophysiology of Bone Mass Impairment
Idiopathic juvenile osteoporosis
Osteogenesis imperfecta
Connective tissue (e.g., chronic arthritis, SLR, ]DM, vasculitis) Gastrointestinal (inflammatory bowel disease, celiac disease, liver disorders) Endocrine (Turner's syndrome, diabetes mellitus, growth hormone deficiency, delayed puberty, Cushing's disease, hypothyroidism, hypopituitarism, hyperparathyroidism, hypogonadism) Hematologic (thalassemia, thrombophilia) Oncologic Met~bolic (glycogen-storage disease,
A demineralization disorder of unknown etiology with onset usually in early puberty. Clinical findings include pathologic fractures, bone pain, and difficulty walking. A diagnosis of exclusion. Spontaneous recovery within 3-4 yr (coinciding with sexual maturation). Heritable (autosomal dominant or recessive) disorders of connective tissue. Different types with different degrees of severity (from mild to lethal). Fragile bones, blue sclerae, deafness, short stature, abnormality of skin and teeth, and spinal deformity are all part of the spectrum. Treatment with pamidronate leads to substantial improvement. 131
Mainly pro-inflammatory cytokines, glucocorticoid use. Also nutritional defects, reduced mobility, and reduced sun exposure. As above, in addition to malabsorption (hence, calcium and vitamin D deficiency). Excess or deficiency of hormones associated with bone metabolism (e.g., T3, T4, PTH, GH, IGF-I, glucocorticoids, sex steroids). Also exercise-induced amenorrhea resulting in osteopenia in female athletes. In thalassemia, hypogonadism is present (from transfusion-induced iron overload). Anticoagulants such as heparin have a detrimental effect on bone mass. Neoplastic infiltration of endocrine organs, tumor cell production of humoral factors, toxicity of chemotherapy (e.g., high-dose methotrexate) and radiotherapy. Interference with type I coHagen fibrils, decreased mechanical load, organ infiltration.
ly~osomal-storage
disorders such as disease, homocystinuria) Retial (chronic renal failure) G~ucher's
Neurologic (cerebral palsy, paraplegia, epilepsy, myopathies) Cysftc fibrosis PsYchiatric (bulimia, anorexia nervosa)
Ast~ma
Nutritional deficiencies; vitamin D, PTH, and calciUm/phosphate metabolic abnormalities; growth failure. Immobilization, decreased mechanical load, nutritional defects, anticonvulsive medications. Pancreatic insufficiency (malabsorption), respiratory involvement (glucocorticoid use), hypogonadism. Low body mass index, malnutrition, low estrogen concentration. Secondary to steroid treatment. Even inhaled steroids reduce the acquisition of bone mineral in prepubertal children. 132
GH, wowth hormone; IGF-I, insulin-like growth factor I; ]OM, juvenile dermatomyositis; PTIf, parathyroid hormone; SLE, systemic lupus erythematosus; T3, triiodot1tyronine; T4, thyroxine.
11 ~o 24 years); during steroid treatment, supplemental daily doses might be required. It has been demonstrated that! calcium supplementation enhances bone mineral acq}iisition in postmenarcheal girls with low calcium int*e. 133 Likewise, vitamin D intake should be maximizFd to at least 400 U/day, and, if not contraindicated by the underlying disease, sun exposure should be recom~ended to facilitate vitamin D synthesis. Control of chr~nic illness, adequate general protein-calorie nutritio~, and appropriate weight-bearing activity are other determinants of bone accretion. Finally, smoking and excFssive alcohol consumption in adolescents should be avqided, because they are likely to be risk factors for a deqreased peak bone mass. The American College of Rheumatology has published revlsed gUidelines for the prevention and treatment of glu~ocorticoid-inducedosteoporosis in adult patients that may also be relevant to children and adolescents. 134 Recommendations include modification of lifestyle factors, weight-bearing physical exercise, calcium and vitamin D supplementation, avoidance or reduction of alcohol and tobacco consumption, and periodic mea-
surement of BMD. A baseline measurement of BMD at the lumbar spine is recommended at the initiation of long-term (longer than 6 months) glucocorticoid therapy, and longitudinal measurements may be repeated periodically during treatment to monitor bone status.
TREATMENT Studies of the medical treatment of osteoporosis in childhood are relatively new. It is difficult to perform controlled trials in uncommon diseases, and additional caution should be used when interpreting the results of published studies. For example, bone loss tends to taper or plateau after 6 to 12 months of steroid treatment, and during this period any given therapy might show benefit. Moreover, there are methodologic problems, some of which are specific to childhood (see earlier discussion of the limitations of diagnostic tools and interpretation of results). Calcium and vitamin D supplementation might have some benefit in mild disease, but severely affected
724
C HAP T E R
38
SKELETAL MATURATION AND BONE MANIFESTATION
patients need more potent interventions. There is a paucity of intervention studies on calcium/vitamin D supplementation in pediatric rheumatic diseases, and pharmacologic interventions with vitamin D in children with chronic arthritis have yielded conflicting results. 97 ,135 Among other candidates for treatment, growth hormone 136,137 and calcitoninB8,139 have both been tried with satisfactory results. Of the newer agents in clinical use, bisphosphonates seem to be the most promising. Bisphosphonates, which are analogues of pyrophosphate characterized by P-C-P bonds, were first studied in humans about 30 years ago. Several chemical features contribute to their biologic action: the P-C-P moiety facilitates the ability of these compounds to adsorb to hydroxyapatite, and therefore target to bone, while variations in their side chains determine the potency and spectmm of action of each individual compound (Fig. 38-3). Bisphosphonates are selectively concentrated in bone and inhibit bone resorption by interfering with the action of osteoclasts. Some of the biochemical mechanisms that account for these effects have been elucidated. !41}-144 Until recently, the use of bisphosphonates in pediatrics has been limited because of fear of adverse effects on a growing skeleton, because of potential risks to a fetus if administered to a girl approaching child-bearing age, and because the dmg is not appreciably eliminated in the short to medium term. More recently, bisphosphonates have been judged to be safe, at least in the short term, even in pediatric practice, and their use has been expanding. 13J ,14'i-148 Table 38-6 lists the observed and the possible adverse effects of pediatric use of bisphosphonates. Untoward effects in children have not been reported in greater frequency than in adults. With the newer, nitrogen-containing bisphosphonates, secondary osteomalacia is not a problem. Hypocalcemia and fever are infrequent and transient, and mild abdominal discomfort or dyspepsia are only occasional complaints. Radiologic abnormalities described in prepubertal patients include band-like metaphyseal sclerosis (Fig. 38-4) and concentric epiphyseal and apophyseal sclerosis. 149 After dmg discontinuation, these abnormalities tend to disappear. No adverse effects on growth have been noted, even after long follow-up. Bone biopsies have demonstrated no signs of mineralization defects and a normal bone stmcture. 1'iO However, many questions still remain unanswered, and, in particular, the long-term adverse effects in a growing skeleton are unknown at this time. Also, poten-
CIII
TABLE 3H-6
Adverse [ffe( Is of Bisphospholliltl'S
Observed Increase in body temperature after intravenous infusion, flu-like symptoms Nausea, dyspepsia, esophagitis, abdominal pain, diarrhea, constipation Hypocalcemia, hypophosphatemia, hypomagnesemia Transient lymphopenia Iritis, uveitis, scleritis, conjunctivitis Mineralization defects (with etidronate), transient skeletal pain, epiphyseal and metaphyseal radiologic sclerosis in groWing bones; mandibular osteonecrosis
Feared but not observed Irreversible and permanent effect on bone remodeling Impaired healing and non-union of fractures Damage to growth plates and impairment in linear growth Fetal abnormalities
tial risks for young women in their child-bearing years with respect to fetal toxicity cannot be disregarded. Therefore, at this time, bisphosphonates can be considered as valuable adjunctive therapy in pediatric patients only when treating severe osteoporotic disease or in the setting of experimental protocols. An open, multicenter, prospective study of safety and efficacy of alendronate in children with rheumatic diseases has been
1
OH R OH I
0
I
- p -c I
I2
I
p - 0 I
OH R OH • Figure 38-3 Chemical structure of bisphosphonates. Rl and R2 are side chains.
• Figure 38-4 Radiologic alterations (metaphyseal dense lines) observed after treatment with oral alendronate in a prepubertal child.This patient had previously received intravenous alendronate; the previous dense lines have migrated (annws).
C HAP T E R
38
SKELETAL MATURATION AND BONE MANIFESTATION
published.J'H To be included, patients had to be recelvmg chronic steroid treatment or had to have a low BMD determined by DEX. Forty-three patients (30 females and 13 males) with juvenile idiopathic arthritis (17 patients), SLE (12), JDM (7), or other connective tissue diseases (7) were studied. The mean age was 12.9 ±3.7 years; 14 patients were postpubertal. Alendronate was administered orally at a daily dosage of 5 or 10 mg (for children with body weight less than or greater than 20 kg, respectively). Baseline Z scores ranged from -1.3 to -5.3. Each patient also underwent serial clinical and laboratory evaluations. Lumbar spine BMD was measured at baseline and after 6 and 12 months by a standardized protocol and with cross-calibration of all instruments. Z scores were calculated by comparing results with age- and sex-matched controls. A substanti~l increase of bone mass was observed in all children with an average BMD increase of 14.2 ± 10% after 1 year. One third of the patients attained a normal BMD. Height increased by an average of 4.3 ± 3.7 em. In comparison, BMD had increased by only 1% in 16 patient~ who were monitored during the year immediately preceding alendronate therapy. Knee radiographs in the prepubertal children demonstrated an absence of rickets and the presence of metaphyseal lines. Alendronate was well tolerated except for occasional abdominal pain and one epis~de of esophagitis. Ina follow-up study, variations in parameters of bone metabolism and disease activity were evaluated. 152 Parameters of both bone resorption and bone formation significantly decreased during the first 12 months, whereas none of the disease activity indices changed significantly. Z-score variations in BMD did not correlate with variations of the inflammatory parameters (erythrocyte sedimentation rate, matrix metalloproteinase-3, IL6, C-reactive protein). It was concluded that the observed increase in BMD was not secondary to a change in disease activity but was most likely an effect of treatment with alendronate.
REFERENCES J. l~uckwalter ]A: Bone biology. ] Bone Joint Surg Am 77: 1256-1276, 1995. 2. I~uckwalter JA. Glimcher MJ. Cooper RR, et al: Bone biology. I: StnLcture, L\lood supply, cell, matrix, and mineralization. Instr Course Lect 45: 371-386, 1996. 3. 13uckwalter .lA, Glimcher M], Cooper RR, et al: Bone biology. II: Formation, fbml. modeling, remodeling, and regulation of cell function. Instr Course l,ect 45: 387-399, 1996. 4. Ng KW. Roms E, Donnan L. et al: Bone biology. Baillieres Clin Endocrinol Metab 11: 1-22, 1997. 5. l~aron N: Anatomy and ultrastructure of bone. In Favus .I (ed): Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism, 4th ed. l'hiladelphia, Lippincott Williams & Wilkins, 1999. pp 3-10. 6. Marks sq, Popoff SN: Bone cell biology: the regulation of development, $tructure, and function in the skeleton. Am J Anat 183: 1--44, 1988. 7. blsen BR: Bone morphogenesis and embryologic development. In Favus] kd): Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism, 4th ed. Philadelphia, Lippincott Williams & Wilkins, 1999. pp
11-13. 8. Robey PG: The biochemistry of bone. Endocrinol Metab Clin Nonh Am 18: 1l59, 1989. 9. Ogden ]A: Anatomy and physiology of skeletal development. In Odgen JA (ed): Skeletal Injury in the Child, 2nd ed. Philadelphia, WB Saunders, 1990, pp 23-<>3. 10. Owen M: The origin of bone cells in the postnatal organism. Arthritis Rheum 23: 1073-1080, 1980. 11. MacDonald BR, Gowen M: The cell biology of bone. Baillieres Clin Rheumatol 7: 421--443. 1993. 12. pan .lB. Stein GS. Canalis E, et al: Bone formation: osteoblast lineage cells. wowth factors. matrix proteins, and the mineralization process. In Favus J (ed): Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. ·ith ed. Philadelphia, Uppincnn Williams & Wilkins, 1999. pp 14-29. 13. Mundy GR: Bone remodeling. In Favus] (ed): Prinler on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. 4th ed. Philadelphia, Lippincon Williams & Wilkins, 1999, pp 30-38. 14. Woitge HW. Seibel MJ: Molecular markers of bone and cartilage metabolism. Curr Opin Rheumatol 11: 218-225, 1999.
725
15. Delmas PO: Biochemical assessment of bone turnover in osteoporosis. Tn DeLuca HG, Mazess R (eds): Osteoporosis: Physiologic Basis, Assessment and Treatment. New York, Elsevier, 1990, p 109. 16. Robins SP, New SA: Markers of bone turnover in relation to bone health. Proc Nutr Soc 56: 903-914. 1997. 17. Parfitt AM, Simon LS, Villanueva AR, et al: ProcolJagen type I carboxy-terminal extension peptide in serum as a marker of collagen biosynthesis in bone. Correlation with iliac bone formation rates and comparison with total alkaline phosphatase. .I Bone Miner Res 2: 427- 436, 1987. 18. Minkin C: Bone acid phosphatase: tartrate-resistant acid phosphatase as a marker of osteoclasts function. Calcif Tissue Int 34: 285-290, 1982. 19. DeCastro ]AS. Peakcock M, McClintock R: Plasma tartrate-resistant acid phosphatase (TRAP) as a marker of bone resorption. .J Bone Miner Res 4 (Suppl Il: 171, 1989. 20. Delmas PD. Schlemmer A, Gineyts E. et al: Urinary excretion of pyridoline crosslinks correlates with bone turnover measured on iliac crest biopsy in patients with vertebral osteoporosis. .I Bone Miner Res 6: 63~4. 1991. 21. Arnaud CD: The calcitropic hormones and metabolic bone disease. In Greenspan PS (ed): Basic and Clinical Endocrinology. Norwalk. CT, Appleton & Lange, 1991. p 247. 22. Hofbauer LC: Osteoprotegerin ligand and osteoprotegerin: novel implications for osteoclast hiology and bone metabolism. EurJ Endocrinol 141: 195-210, 1995. 23. Hofbauer LC, Heufelder AE: Role of receptor activator of nuclear factor-kB and osteoprotegerin in bone cell biology. .I Mol Med 79: 243-253, 2001. 24. Suda T, Takahashi N, Udagawa N, et al: Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev 20: 345-357, 1999. 25. Aubin JE, Bonnelye E: Osteoprotegerin and its ligand: a new paradigm for regulation of osteoclastogenesis and bone resorption. Osteoporosis Int 11: 905-913, 2000. 26. Hotbauer LC, Khosla S, Dunstan CR, et al: The roles of osteoprOlegerin and osteoprotegerin ligand in the paracrine regulation of bone resorption. .J Bone Miner Res 15: 2-12, 2000. 27. Bolon B, Shalhoub V, Kostenuik 1'], et aI: Osteoprotegerin: an endogenous antiosteoclast factor for protecting bone in rheumatoid arthritis. Arthritis Rheum 46: 3121-1335. 2002. 28. Kong YY, Yoshida H, Sarosi, et al: OPGL is a key regulator of osteoclastogenesis, Iymphoqte development and lymph-node organogenesis. Nature 397: 315-323, 1999. 29. Boyle WJ, scon Simonet W, Lacey DL: Osteoclast differentiation and activation. Nature 423: 337-342, 2003. 30. De Schepper ], Derde MP, Van der Broeck M, et al: Normative data for lumbar spine bone mineral content in children: influence of age, height, weight, and pubertal stage. ] Nucl Med 32: 216-220, 1991. 31. Katzman OK, Bachrach LK. Carter DR, et al: Clinical and anthropometric correlates of bone mineral acquisition in healthy adolescent girls. J Clin Endocrinol Metab 73: 1332-1339, 1991. 32. Miller JZ, Slemenda CW, Meaney F], et al: The relationship of bone mineral density and anthropometric variables in healthy male and female children. Bone Miner 14: 137-152, 1991. .~3. Gl~stre C, Braillon P, David L, et al: Measurement of bone mineral content of the lumbar spine by dual energy x-ray absorptiometry in nonnal children: correlations with growth parameters. .I Clin Endocrinol Metab 70: U30-1333, 1990. 34. DePriester ]A, Cole T], Bishop NJ: Bone growth and mineralisation in children aged 4 to 10 years. Bone Miner 12: 57-65, 1991. 35. Matkovic v, ]elic T, Wardlaw GM, et al: Timing of peak bone mass in Caucasian females and its implication for the prevention of osteoporosis. Interference ti-om a cross-sectional model.] Clin Invest 93: 799-808, 1994. 36. Rubin K, Schirduan V, Gendreau P, et al: Prellictors of axial and peripheral bone mineral density in healthy children and adolescents. with special anention to the role of puberty. .J Pediatr 123: 863-870, 1993. 37. Bonjour JP, Theintz G, Buchs B, et al: Critical years and stages of puberty for spinal and femoral bone mass accumulation during adolescence. ] Clin Endocrinol Metab 73: 555-563. 1991. 38. Gilsanz V, Gibbens DT, Carlson M, et al: Peak trabecular venebral density: a comparison of adolescent and adult females. Calcif Tissue Int 43: 260-262, 1988. 39. Lu PW. BriodyJN, Ogle GO. et al: Bone mineral density of total hody, spine, and femoral neck in children and young adults: a cross-sectional and longitudinal study. ] Bone Miner Res 9: 1451-1458. 1994. 40. Kroger H, Kotaniemi A. Kroger L, et al: Development of bone mass and bone density of the spine and femoral neck-a prospective study of 65 children and adolescents. Bone Miner 23: 171-182, 1993. 41. Lloyd T, Rollings N, Andon MB, et al: Deternlinants of bone denSity in young women. I: Relationships among pubertal development, total body bone mass, and total body bone density in premenarchal females. ] Clin Endocrinol Metab 75: 383-387, 1992. 42. Matkovic V, Fontana 0, Tominac C, et al: Factors that influence peak hone mass formation: a study of calcium balance and the inheritance of hone mass in adolescent females. Am] Clin Nutr 52: 878--888, 1990. 43. Cadogan]. Blumsohn A, Barker ME. Eastell R: A longitudinal study of bone gain in pubeltal girls: anthropometric and biochemical correlates. ] Bone Miner Res 13: 1602-1612, 1998.
726
C HAP T E R
38
SKELETAL MATURATION AND BONE MANIFESTATION
44. Sabatier JP, Guaydier-Souquieres G, Laroche D, et al: Bone mineral acquisition dUring adolescence and early adulthood: a study in 574 healthy females 10-24 years of age, Osteoporosis Int 6: 141-148, 1996. 45. Sabatier]p, Guaydier-Souquieres, Benmalek A, Marcelli C: Evolution of lumbar bone mineral content during adolescence and adulthood: a longitudinal study in 395 healthy females 10-24 years of age and 206 premenopausal women. Osteoporosis int 9: 476-482, 1999. 46. Thainz G, Buchs B, Rizzoli R, et al, Longitudinal monitoring of bone mass accumulation in healthy adolescents: evidence for a marked reduction after 16 years of age at the level of lumbar spine and femoral neck in female subjects. J Clin Endocrinol Metab 75: 1060-1065, 1992. 47. Lazcano-Ponce E, Tamayo J, Cruz-Valdez A, et al: Peak bone mineral area density and determinants among females aged 9 to 24 years in Mexico. Osteoporosis Int 14: 539-547, 2003. 48. Bonjour JP, Rizzoli R: Bone acquisition in adolescence. In Marcus R, Feldman D, Kelsey J (eds): Osteoporosis. San Diego, CA, Academic Press, 2001, pp 621--638. 49. Haapasalo H, Kamus P, Sievanen H, et al: Development of mass, density, and estimated mechanical characteristics of bones in Caucasian females. J Bone Miner Res 11: 1751-1760, 1996. 50. Lin YC, Lyle RM, Weaver CM, et al: Peak spine and femoral neck bone mass in young women. Bone 32: 546-553, 2003. 51. Rizzoli R, Bonjour JP: Hormones and bones. Lancet 349 (Suppl 1): SI20-5123, 1997. 52. Cassidy JT: Osteopenia and osteoporosis in children. Clin Exp Rheumatol 17: 245-250, 1999. 53. Slemenda CW, Reister TK, Hui SL, et al: Influences on skeletal mineralisation in children and adolescents: evidence for varying effects of sexual maturation and physical activity. J Pediatr 125: 201-207, 1994. 54. Ruiz JC, Mandel C, Garabedian M, et al: Influence of spontaneous calcium intake and physical exercise on dle vertebral and femoral bone mineral density of children and adolescents. J Bone Miner Res 10: 675--682, 1995. 55. Kelly PJ, Hopper JL, Macaskill GT, et al: Genetic factors in bone turnover. J Clin Endocrinol Metab 72: 808--813, 1991. 56. Pocock NA, Eisman JA, Hopper JL, et aI: Genetic determinants of bone mass in adults. A twin study. J Clin Invest 80: 706-710, 1987. 57. Slemenda CW, Christian JC, Williams C), et al: Genetic determinants of bone mass in adult women: a reevaluation of the twin model and the potential importance of gene interaction on heritability estimates. J Bone Miner Res 6: 561-567, 1991. 58. Ralston SH: What determines peak bone mass and bone loss? Best Pract Res Clin Rheumatol 11: 479-493, 1997. 59. Ralston SH: The genetics of osteoporosis. Bone 25: 85-86, 1999. 60. Willing MC, Tomer JC, Burns U: Gene polymorphisms, bone mineral density and bone mineral content in young children: the Iowa bone development study. Osteoporosis Int 14: 650-658, 2003. 61. Abrams SA, Stuff JE: Calcium metabolism in girls: current dietary intakes lead to low rates of calcium absorption and retention during puberty. Am J Clin Nutr 60: 739-743, 1994. 62. Lloyd T, Martel JK, Rollings N, et al: The effect of calcium supplementation and Tanner stage on bone density, content and area in teenage women. Osteoporos Int 6: 276-283, 19%. 63. Sentipal JM, Wardlaw GM, Mahan J, et al: Influence of calcium intake and growth indexes on vertebral bone mineral density in young females. Am J Clin Nutr 54: 425-428, 1991. 64. Wang MC, Moore EC, Crawford PB, et al: Influence of preadolescent diet on quantitative ultrasound measurements of the calcaneus in young adult women. Osteopor Int 9: 532-535, 1999. 65. Matkovic V, Heaney RP: Calcium balance during human growth: evidence for threshold behaviour. Am J Clin Nutr 55: 992-996, 1995. 66. Johnston CC], Miller JZ, Slemenda CW, et al: Calcium supplementation and increases in bone mineral density in children. N Eng!] Med 327: 82-87, 1992. 67. Lehtonen-Veromaa MK, MOllonen IT, Nuotio 10, et al: Vitamin D and attainment of peak bone mass among peripubertal Finnish girls: a 3-y prospective study. Am J Clin Nutr 76: 1446-1453, 2002. 68. Riggs BL, Nguyen TV, Melton LJ, et al: The contribution of vitamin D receptor gene alleles to the determination of bone mineral density in normal and osteoporotic women. J Bone Miner Res 10: 991-996, 1995. 69. Hustmyer FG, Peacock M, Hui S, et al: Bone mineral density in relation to polymorphism at the vitamin D receptor gene locus. J ClIn Invest 94: 2130-2134, 1994. 70. Masi L, Cinlaz R, Simonini G, et al: Association of low bone mass with vitamin D receptor gene and calcitonin receptor gene polymorphisms in juvenile idiopathic arthritis. J Rheumatol 29: 2225-2231, 2002. 71. Welten DC, Kemper HC, Post GB, et al: Weight-bearing activity during youth is a more inlportant factor for peak bone mass than calcium intake. J Bone Miner Res 9: 1089-1096, 1994. 72. Bouxsein ML, Marcus R: Overview of exercise and bone mass. Rheum Dis Clin North Am 20: 787-802, 1994. 73. Kannus P, Haasapalo H, Sankelo M, et al: Effect of starting age of physical activity on bone mass in the dOlninant arm of tennis and squash players. Ann Intern Med 123: 27-31, 1995. 74. Kanis JA, Melton LJ, Christiansen C, et aI: The diagnosis of osteoporosis. J Bone Miner Res 9: 1137-1140, 1994.
75. Baroncelli GI, Federico G, Bertelloni S, et al: Bone quality assessment by quantitative ultrasound of proximai phalanxes of the hand in healthy subjects aged 3-21 years. Pediatr Res 49: 713-718, 2001. 76. Barkmann R, Rohrscheider W, Vierling M, et al: German pediatric reference data for quantitative transverse transmission ultrasound of finger phalanges. Osteoporosis Int 13: 55--61, 2002. 77. Falcini F, Bindi G, Ermini M, et al: Comparison of quantitative calcaneal ultmsound and dual energy x-ray absorptiometry in the evaluation of osteoporotic risk in children with chronic rheumatic diseases. Calcif Tissue Int 67: 19-23, 2000. 78. Njeh CF, Shaw N, Gardner-Medwin JM, et al: Use of quantitative ultrasound to assess bone status in children with juvenile idiopathic arthritis: a pilot study. J Clin Densitom 3: 251-260, 2000. 79. Falcini F, Bindi G, Simonini G, et al: Bone status evaluation with calcaneal ultrasound in children with chronic rheumatic diseases. A one year followup study. J Rheumatol 30: 179-184, 2003. 80. Jaworski M, Lebiedowski M, Lorenc RS, et al: Ultmsound bone measurement in pediatric subject'. Calcif Tissue Int 56: 368--371, 1995. 81. Mughal MZ, Langton CM, Utretch G, et al: Comparison between broad-band ultrasound attenuation of the calcaneum and total body mineml density in children. Acta P",diatr 85: 663--665, 1996. 82. Baroncelli GI, Federico G, Bertelloni S, et al: Assessment of bone quality by quantitative ultrasound of proximal phalanxes of the hand and fracture in children and adolescents with bone and minerals disorders. Pediatr Res 54: 125-136, 2003. 83. Wuster C, Albanese C, De Aloysio D, et al: Phalangeal osteosonogmmmetry: age-related changes, diagnostic sensitivity, and discrimination power. The Phalangeal Osteosonogrammetry Study Group. J Bone Miner Res 15: 1603-1614, 2000. 84. Cimaz R, Biggioggero M: Osteoporosis. Curr Rheum Reports 3: 365-370, 2001. 85. Cassidy JT, Langman CB, Allen SH, et al: Bone mineral metabolism in children with juvenile rheumatoid arthritis. Ped Clin North Am 42: 1017-1033, 1995. 86. Cetin A, Celiker R, Dincer F, Ariyurek M: Bone mineral density in children with juvenile chronic arthritis. Clin Rheumatol 17: 551-553, 1998. 87. Falcini F, Ermini M, Bagnoll F, et al: Bone turnover is reduced in children with juvenile rheumatoid arthritis. J Endocrinol Invest 21: 31-36, 1998. 88. Havelka S, Vavrincova P, Stepan J: Metabolic bone status in young women with juvenile chronic arthritis. J Rheumatol 37: 14-16, 1993. 89. Henderson C], Lovell DJ: Bone mineral content in juvenile rheumatoid arthritis--pilot project resuits. J Rheumatol 18: 1-22, 1991. 90. Hillman L, Cassidy]T, Johnson L, et al: Vitamin D metabolism and bone mineralization in children with juvenile rheumatoid arthritis. J Pediatr 121: 910-916, 1994. 91. Hopp R, Degan J, Gallagher JC, Cassidy JT: Estimation of bone mineral density in children with juvenile rheumatoid arthritis. J Rheumatol 18: 1235-1239, 1991. 92. Kotaniemi A, Savolainen A, Kontiainen H, Kroger H: Estimation of central osteopenia in children with chronic polyarthritis treated with glucocorticoids. Pediatrics 91: 1127-1130, 1993. 93. Kotaniemi A: Growth retardation and bone ioss as determinants of axial osteopenia in juvenile chronic arthritis. Scand J Rheumatol26: 14-18, 1997. 94. Pereira RMR, Corrente JE, Chahade WH, Yoshinari NH: Evaluation by dual x-ray absorptiometry (DXA) of bone mineral density in children with juvenile chronic arthritis. Clin Exp Rheumatol 16: 495-501, 1998. 95. Polito C, Strano CG, Rea L, et al: Reduced bone mineral content and normal serum osteocalcin in non-steroid-treated patients with juvenile rheumatoid arthritis. Ann Rheum Dis 54: 193-196, 1995. 96. Rabinovich EC: Bone mineml status in juvenile rheumatoid arthritis. J Rheumatol 27 (Suppl 58): 34-37, 2000. 97. Warady BD, Lindsley CB, Robinson RG, Lukert BP: Effects of nutritional supplementation on bone mineral status of children with meumatic diseases receiving corticosteroid therapy. J Rheumatol 21: 530-535, 1994. 98. Pepmueller PH, Cassidy]T, Allen SH, Hillman LS: Bone minemlization and bone mineral metabolism in children with juvenile rheumatoid arthritis. Arthritis Rheum 39: 746-757, 1996. 99. Brik R, Keidar Z, Schapira D, Israel 0: Bone mineral density and turnover in children with systemic Juvenile chronic arthritis. J Rheumatol 25: 990-992, 1998. 100. Kotaniemi A, Savolainen A, Kroger H, et al: Development of bone mineml density at the lumbar spine and femoml neck in juvenile chronic arthritis. A prospective one year follow-up study. J Rheumatol 25: 2450-2455, 1998. 101. Kotaniemi A, Savolainen A, Kroger H, et al: Weight-bearing physical activity, calcium intake, systemic glucocorticoids, chronic inflammation, and body constitution as determinants of lumbar and femoml bone mineral in juvenile chronic arthritis. Scand J Rheumatol 28: 19-26, 1999. 102. Henderson CJ, Cawkwell GD, Specker BL, et al: Predictors of total body bone mineral density in non-corticosteroid-treated prepubertal children with juvenile rheumatoid arthritis. Arthritis Rheum 40: 1967-1975, 1997. 103. Henderson C], Specker BL, Sierm RI, et al: Total-body bone mineml content in non-corticosteroid-treated postpubertal females with juvenile rheumatoid arthritis. Frequency of osteopenia and contributing factors. Arthritis Rheum 43: 531-540, 2000.
C HAP T E R
38 SKELETAL MATURATION AND BONE MANIFESTATION
104. Malflolagas SC, ]ilka RL: Bone marrow, cytokines, and bone remodeling. Emerging insights into the pathophysiology of osteoporosis. N Eng! ] Med 332: 305--311, 1995. 105. ]ilkk RL, Hangoc G, Girasole G, et al: Increased osteoclast development after est~ogen loss: mediation by interleukin-6. Science 257: 88-91, 1992. 106. Pereira RMR, Falco V, Corrente ]E, et al: Abnormalities in the biochemical markers of bone turnover in children with juvenile chronic arthritis. elin Exp Rh¢umatol 17: 251-254, 1999. 107. Cassidy ]T, Hillman LS: Abnormalities in skeletal growth in children with juvenile rheumatoid arthritis. Rheum Dis Clin North Am 23: 499-522, 1997. 108. zak M, Hassager C, Lovell 0], et al: Assessment of bone mineral density in adults with a history of juvenile chronic arthritis: a cross-sectional long-term follow-up study. Arthritis Rheum 42: 790-798, 1999. 109. French AR, Mason T, Nelson AM, et al: Osteopenia in adults with a history of juvenile rheumatoid arthritis. A population based study. J Rheumatol 29: 1065-1070, 2002. 110. Ha~gen M, Lien G, Flato B, et al: Young adults with juvenile arthritis in remission attain normal peak bone mass at the lumbar spine and forearm. Arthritis Rheum 43: 1504-1510, 2000. 111. Lien G, Flato B, Haugen M, et al: Frequency of osteopenia in adolescents wilh early-onset juvenile idiopathic arthritis. A long-term study of one hundrtld flve patients. Arthritis Rheum 48: 2214-2223, 2003. 112. Stewart WA, Acott PO, Salisbury SR, et al: Bone mineral density in juvenile dermatomyositis: assessment using dual x-ray absorptiometry. Arthritis Rheum 48: 2294-2298, 2003. 113. Sirjigaglia L, Varenna M, Binelli L, et al: Bone mass in systemic lupus erythc!matosus. Clin Exp Rheumatol 18 (Suppl 21); S27-S34, 2000. 114. Trapani S, Civinini R, Ermini M, et al: Osteoporosis in juvenile systemic lupus erythematosus: a longitudinal study of the effect of steroids on bone mineral de/lsity. Rheumatol Int 18: 45--49, 1998. 115. Varamos S, Ansell BM, Reeve J: Vertebral collapse in juvenile chronic arthritis: its relationships with glucocorticoid therapy. Calcif Tissue Int 41: 75--78, 19/17. 116. CainalL. E: Mechanism of glucocorticoid action in bone: implications to gluco,orticoid-induced osteoporosis. J Clin Endocrinol Metab 81: 3441-3447, 19/)6. 117. Peireira RC, Delany AM, Canalis E: Effects of cortisol and bone morphogenetic protein-2 on stromal cell differentiation: correlation with CCAATenhancer binding protein expression. Bone 30: 685--691, 2002. 118. Carbonare ill, Arlot ME, Chavassieux PM. et al: Comparison of trabecular b<jne microarchitecture and remodeling in glucocorticoid-induced and postmenopausal osteoporosis.] Bone Miner Res 16: 97-103, 2001. 119. Van Staa TP, Leufkens HGM, Abenhaim L, et al: Use of oral corticosteroids and risk of fractures.] Bone Miner Res 15: 993-1000. 2000. 120. D¢lany AM, Gabbitas BY, Canalis E: Cortisol down regulates osteoblast alpha 1 II) procollagen mRNA by transcriptional and post-transcriptional mechanisms. J Cell Biochem 57: 488-494, 1995. 121. Weinstein FS, Chen JR, Powers CC, et al: Promotion of osteoclast survival arid antagonism of bisphosphonate-induced osteoclast apoptosis by glucocorticoids. ] Clin Invest 109: 1041-1048, 2002. 122. Pereira RMR, Delany AM, Canalis E: Cortisol inhibits the differentiation and apoptosis of osteoblasts in culture. Bone 28: 484-490, 2001. 123. ~instein RS, Jilka RL, Parfitt AM, et al: Inhibition of osteoblastogenesis and p~omotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. J Clin Invest 102: 274-282, 1998. 124. C~JOper MS, Rabbitt EH, Goddard PE, et al: Autocrine activation of glucocc;rticoids in osteoblasts increase with age and glucocorticoid exposure. ] eone Miner Res 17: 979-986, 2002. 125. Hpfbauer LC, Gori F, Riggs BL, et al: Stimulation of osteoprotegerin ligand amd inhibition of osteoprotegerin production by glucocorticoids in human osteoblastic lineage ceUs: potential paracrine mechanisms of glucocorticoidiIlduced osteoporosis. Endocrinology 140: 4382-4389, 1999. 126. Bplogna C, Edno L, Anaya JM, et al: Methotrexate concentrations in synovial membrane and trabecular and cortical bone in rheumatoid arthritis patients. Arthritis Rheum 37: 1770-1773, 1994. 127. ~ay KP, Mercill 0, McDermott MT, West SG: The effect of methotrexate on njouse bone ceUs in culture. Arthritis Rheum 39: 489-494, 1996. 128. ~ay KP, West SG, McDermott MT, Huffer WE: The effect of low-dose methotrexate on bone metabolism and histomorphometry in rats. Arthritis Rheum 37: 201-206, 1994.
727
129. Scheven BA, van der Veen MJ, Oamen CA, et al: Effects of methotrexate on human osteoblasts in vitro: modulation by 1,25-dihydroxyvitamin Dr J Bone Miner Res 10: 874-880, 1995. 130. Bianchi ML, Cimaz R, Galbiati E, et al: Bone mass change during methotrexate treatment in patients with juvenile rheumatoid arthritis. Osteoporosis 1nt 10: 20-25, 1999. 131. Glorieux FH, Bishop NJ, Plotkin H, et al: Cyclic administration of pamidronate in children with severe osteogenesis imperfecta. N Engl J Med 339: 947-952, 1998. 132. Allen HO, Thong IG, Clifton-Bligh P, et al: Effects of high-dose inhaled corticosteroids on bone metabolism in prepubertal children with asthma. Pediatr Pulmonol 29: 188--193, 2000. 133. Rozen GS, Rennert G, Dodiuk-Gad RP, et al: Calcium supplementation provides an extended window of opportunity for bone mass accretion after menarche. Am J Clin Nutr 78: 993-998, 2003. 134. American CoUege of Rheumatology ad hoc Committee on glucocorticoidinduced osteoporosis: Recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis-200! update. Arthritis Rheum 44: 1496-1503, 2001. 135. Reed A, Haugen M, Pachman L, Langman CB: 25-Hydroxyvitamin 0 therapy in children with active juvenile rheumatoid arthritis. Short-term effects on serum osteocalcin levels and bone mineral density. J Pediatr 119: 657-660, 1991. 136. Rooney M, Davies UM, Reeve J, et al: Bone mineral content and bone minerai metabolism: changes after growth hormone treatment in juvenile chronic arthritis. J Rheumatol 27: 1073-1081, 2000. 137. Simon 0, Lucidarme N, Prieur AM, et al: Effects on growth and body composition of growth hormone treatment in children with juvenile idiopathic arthritis requiring steroid therapy. J Rheumatol 30: 2492-2499, 2003. 138. Nishioka T, Kurayama H, Yasuda T, et al : Nasal administration of salmon calcitonin for prevention of glucocorticoid-induced osteoporosis in children with nephrosis. ] Pediatr 118: 703-707, 1991. 139. Siamopoulou A, Challa A, Kapoglou P, et al: Effects of intranasal salmon calcitonin in juvenile idiopathic arthritis: an observational study. Calcif Tissue Int 69: 25--30, 2001. 140. Russell RGG, Rogers M]: Bisphosphonates: from the laboratory to the clinic and back again. Bone 25: 97-106, 1999. 141. Breuil V, Cosman F, Stein L. et al: Osteoclast formation and activity in vitro: effects of alendronate. J Bone Miner Res 13: 1721-1729, 1998. 142. Fisher GE, Rogers MJ, Halasy]M, et al: Alendronate mechanism of action: geranylgeraniol, an intermediate in the mevalonate pathway, prevents inhibition of osteoclast formation, bone resorption, and kinase activation in vitro. Proc Natl Acad Sci USA 96: 133-138, 1999, 143. Van Beek E, Lowik C, Van Oer P!uijm G, Papapoulos S: The role of geranylgeranylation in bone resorption and its suppression by bisphosphonates in fetal bone explants in vitro. A clue to the mechanism of action of nitrogen-containing bisphosphonates. J Bone Miner Res 14: 722-729, 1999. 144. Grove]E, Brown R], Walts OJ: The intracellular target for the antiresorptive aminobisphosphonate drugs in Dictyostelium discoideum is the enzyme farnesyl diphosphate synthase. J Bone Miner Res 15: 971-981, 2000. 145. Falcini F, Trapani S, Errnini M, Brandi ML: Intravenous administration of alendronate counteracts the in vivo effects of glucocorticoids on bone remodelling. Calcif Tissue Int 58: 166-169, 1996. 146. Shaw N], Boivin CM, Crabtree NJ: Intravenous pamidronate in juvenile osteoporosis. Arch Dis Child 83: 143-145, 2000. 147. Shoemaker LR: Expanding role of bisphosphonate therapy in children. J Pediatr 134: 264-267, 1999. 148. Srivastava T, Alon US: Bisphosphonates: from grandparents to grandchildren. Clin Pediatr 38: 687-702, 1999. 149. Van Persijn van Meerten EL, Kroon HM, Papapoulos SE: Epi- and metaphyseal changes in children caused by administration of bisphosphonates. Radiology 184: 249-254, 1992. 150. Brumsen C, Hamdy NA, Papopoulos SE: Long-term effects of bisphosphonates on the growing skeleton: studies of young patients with severe osteoporosis, Medicine (Baltimore) 76: 266-283, 1997. 151. Bianchi ML, Cimaz R, Bardare M, et al: Efficacy and safery of alendronate for the treatment of osteoporosis in diffuse connective tissue diseases in children. Arthritis Rheum 43: 1960-1966, 2000. 152. Cimaz R, Gattorno M, Sormani MP, et al: Changes in markers of bone turnover and inflammatory parameters during alendronate therapy in pediatric patients with rheumatic diseases. J Rheumatol 29: 1786-1792, 2002.
( HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS James T. Cassidy and Ross E. Petty
l_~
Occasionally, a child in whom arthritis or a musculoskeletal pain syndrome has been erroneously diagnosed is discovered to have a bone tumor. 1 Although this unfortunate occurrence is rare, physicians must consider this possibility when examining any child with musculoskeletal pain. Musculoskeletal manifestations of malignancy in childhood may take one of four forms: 1. Primary benign or malignant tumors of bone, cartilage, fibrous or soft tissue, or miscellaneous origin 2. Metastatic bone tumors 3. Malignant infiltration of bone marrow: leukemia 4. Secondary effects of malignancy Table 39-1 provides a classification of common bone tumors of childhood. This list is not comprehensive. 2-5 Overall, approximately one half of the bone tumors occurring in the first 2 decades of life are malignant. 6 This proportion contrasts sharply with the ratio in adulthood, when malignant tumors are much more frequent. Except for parosteogenic sarcoma, which is more frequent in girls, malignant bone tumors occur more frequently in boys, with a ratio of approximately 1.5:1; the male-tofemale ratio is approximately 3: 1 for osteoid osteomas and osteoblastomas. 2,3,7 Bone tumors may be completely asymptomatic until a mass is detected by the patient or parent. They usually present with the insidious onset of tenderness, swelling, and localized pain that is accentuated at night or by weight bearing (Table 39-2). Local tenderness or bony swelling in the absence of trauma suggests the diagnosis. Systemic symptoms such as fever and weight loss are nonspecific but support diagnosis of a malignant rather than a benign tumor, A plain radiograph of the affected area is the best initial diagnostic evaluation. H,9 It is important to notice if the lesion is osteolytic or osteogenic and whether there is a soft tissue reaction, Each tumor occurs in characteristic bones and locations (Tables 39-3 and 39-4). Most benign or malignant tumors of a long bone arise in the metaphysis. Malignant tumors of the epiphysis are rare; Ewing's sarcoma is the only malignant tumor commonly arising in the diaphysis. The extent of the lesion can be estimated by computed tomography (CT). Magnetic resonance imaging (MRl) delineates the soft tissue extent of early lesions, including
728
involvement of the bone marrow. W- 12 Ultrasonography is valuable in assessing the extraosseous extension of a tumor. 13 Radionuclide scanning is useful in localiZing a tumor or tumors, rather than in assisting with a specific diagnosis. Definitive diagnosis of a bone tumor rests on the histologic evaluation of an open or fine-needle biopsy. The laboratory otherwise provides little assistance because anemia, leukocytosis, elevation of the erythrocyte sedimentation rate (ESR), and other indicators of inflammation are nonspecific and may be absent even in the presence of osseous malignancy. Serum alkaline phosphatase levels may be elevated beyond those associated with growth of the child.
PRIMARY TUMORS OF BONE, CARTILAGE, FIBROUS OR SOFT TISSUE, OR MISCELLANEOUS ORIGIN Benign Tumors of Bone Of the benign tumors, osteoid osteomas, osteochondromas, and chondromas are most common in the first 2 decades of life. 14 ,15 Approximately one half of these lesions occur before the age of 20 years (Table 39-5). Surgical treatment is often necessaryY'
n:L~
TABLE 39 I of Childhood
ClassifiCiltioll of Common Bone Tumors
Histologic type
Benign
Malignant
Osteogenic
Osteoid osteoma Osteoblastoma Osteochondroma Chondroma Chondroblastoma Chondromyxoid fibroma Fibrous defect Giant cell tumor
Osteosarcoma Parosteal osteosarcoma Chondrosarcoma
Chondrogenic
Fibrogenic Stromal Neuroectodermal Hematopoietic
Fibrosarcoma Ewing's Sarcoma Reticulum cell sarcoma
C HAP T E R
tilT
ABLE 39- 2
General Clinical Characteristics of Bone Tumors
Characteristic
Comments
Pain
May be absent Usually steady, "boring" ache Often worse at night Overlying soft tissues and affected bone are tender May be localized, palpable, or visible swelling in large tumors Absent in patients with benign tumors If fever, weight loss, or pallor occur, suspect malignant tumor Hypertrophic osteoarthropathy suggests pleuropulmonary metastases Results all normal for benign tumors Increased ESR, abnormal CBC for malignant tumors Often diagnostic at initial presentation
Tenderness Swelling Systemic symptoms
Laboratory studies
Radiographs
CBC. complete blood count; ESR. erythrocyte sedimentation rate.
Osteoid Osteoma Osteoid osteomas are benign tumors that are most common between the ages of 10 and 20 years, although they can also occur in younger children (Table 39-6).17,18 The most common sites are the proximal femur, often in the neck and greater trochanter; the proximal tibia; and the pedicles, facets, and spinous processes of the vertebrae. 19 Pain is the typical presentation, It is described as a deep and penetrating ache that is usually worse at night and may be dramatically responsive to low-dose aspirin or other nonsteroidal anti-inflammatol)' drugs (NSAIDs), The site of the lesion is tender, and there may be marked muscle atrophy, weakness of that limb, or a limp. Lesions in the vertebrae may be associated with scoliosis with the concavity toward the site of the tumor. Occasionally, a synovial effusion is present if the tumor is adjacent to the hip or knee within the confines of the capsule. Signs of systemic illness are absent. There are no abnormalities on laboratol)' evaluation; the diagnosis is based principally on radiologic findings. On a plain radiograph, the typical lesion is a nidus of increased density within a ring of decreased denSity, which
•
TABLE 39-3
Most Common Types ofTumors in Specific Bones'
Bolle
Benign
Malignant
Felmlr Tibia
Osteochondroma Osteoid osteoma Osteochondroma Giant cell tumor Osteochondroma Osteochondroma Osteoid osteoma Osteochondroma Chondroma Giant cell tumor
Osteosarcoma Osteosarcoma
Innominate Humerus Vertebra Rib Hand Radius
729
39 SKELETAL MALIGNANCIES AND RELATED DISORDERS
fill
TABI E 39-4 Bones
'List¢d in order of decreasing frequency. Data from Dahlin DC. Unni KK: Bone Tumors: General Aspects and Data on 8,542 Cases, 4th ed. Springfield, IL, Charles C Thomas, 1986.
Malignant
location
Benign
Epiphysis
Chondroblastoma Giant cell tumor Fibrous defect Osteochondroma Chondromyxoid tibroma Chondroma Osteoid osteoma Osteoblastoma
Metaphysis
Diaphysis
Osteosarcoma Parosteal osteosarcoma Chondrosarcoma Ewing's sarcoma
is surrounded by bone of increased density (Fig. 39-1). Histologically, the nidus is a mixture of osteoid, bone, and blood vessels surrounded by a fibrovascular layer that separates it from the surrounding sclerotic bone. This lesion is often difficult to identify by standard radiographic examination, especially in nondiaphyseal sites; other imaging techniques may be required for identification. 18 If an osteoid osteoma is suspected, even if it is not demonstrable on plain radiographs, technetium 99m bone scintigraphy is the diagnostic procedure of choice (Fig. 39-2). CT documents the precise extent of the lesion and its characteristic structure and is invaluable in planning a surgical approach. MRI may identify the nature of a soft tissue mass that is associated with an early lesion. 2()-22 The course of untreated osteoid osteoma varies. This tumor is not malignant, does not metastasize or cause death, and may spontaneously heal radiographically without surgical or other treatment. 17.23 However, the debility induced by the associated pain usually requires removal of the lesion. Excision is curative, and the recurrence rate is low. Aspirin or another NSAID can provide symptomatic relief and is occasionally justified on a longterm basis if the lesion is surgically inaccessible.
Osteoblastoma The osteoblastoma is usually regarded as an osteoid osteoma larger than 1 to 2 em in diameter. 23 It occurs much more often in boys and is more frequent in ado-
IE'.
fABLE 39-5 of Childhood
Aye Distribution of Beniyn Bone Tumors
Percentage of Tumors of this type In: Tumor
Osteosarcoma Osteosarcoma Reticulum cell sarcoma Chondrosarcoma Chondrosarcoma Osteosarcoma
Typical Anatomif Sites of Tumors in lony
Osteoid osteoma Osteoblastoma Giant cell tumor Osteochondroma Chondroma Chondroblastoma Chondromyxoid fibroma Fibrous defect
In 1st Decade
InZnd Decade
18 3
17
2
44 15 1 3
8 46 8 6 13
11
8
4
Percentage of Tumors of this type In: In 1st Decade
12 8 1
InZnd Decade
13
54 44 15 49 26 55 25
19
67
10 10 2
730 PI' L,.
C HAP T E R
TABLE 39 6
39 SKELETAL MALIGNANCIES AND RELATED DISORDERS
Osteoid Osteoma
Chuaderlstlc
Description
Age at onset Sex ratio Symptoms
Childhood to young adulthood Boys> girls Localized aching or boring pain, mild at first, increasing in severity Worse at night, with rest, or with elevation Limpness, stiffness, or weakness Long history Joint effusion occasionally Localized swelling or tenderness Solitary, lucent nucleus surrounded by sclerosis on radiographs Positive bone scan Pain is dramatically relieved by aspirin or nonsteroidal anti-inflammatory drugs Excision
Signs Investigations Treatment
often asymptomatic and presents as a painless exostotic mass that, by virtue of location and size, may induce local functional changes or result in pain because of pressure on neurovascular structures. An osteochondroma usually affects the metaphysis of a long bone and often arises at the site of a tendon insertion (most commonly around the knee in the distal femur or proximal tibia) or in the distal humerus. The osteochondroma extends away from the epiphysis as a bony outgrowth capped with cartilage up to 1 Col thick (Fig. 39-3). Some children with multiple lesions exhibit an autosomal dominant pattern of inheritance (i.e., multiple hereditary exostoses) (Fig. 39-4).21 Treatment consists of surgical excision, Malignant change may occur in solitary or multiple osteochondromas.
Chondroma lescence than childhood. Pain is the presenting symptom. This tumor is most common in a vertebral arch and has an aggressive radiographic appearance with circumscribed erosion of the cortex. Malignant transformation has been reported. Surgical resection may be difficult because of the site or size of the lesion.
Benign Tumors of Cartilaginous Origin Osteochondroma Osteochondroma is the most common benign bone tumor between the ages of 5 and 15 years. 2,3 It occurs with equal frequency in boys and girls, This tumor is
Chondromas (Le., enchondromas) make up about 10% of benign bone tumors. They occur with equal frequency in boys and girls and may affect young children as well as those in the second decade of life. 2 .3 This cartilaginous tumor, which may represent overgrowth of normal epiphyseal hyaline cartilage, occurs most commonly in the small tubular bones of the hand and foot, although other sites may be involved (Fig, 39-5). The radiographic appearance of a chondroma is that of a well-demarcated metaphyseal lesion of central destruction that may protrude from the surface of the bone or be confined within the medullary canal (Le., enchondroma), usually with linear or speckled calcification. It often presents as a solitary, asymptomatic mass with a pathologic fracture or is discovered as an incidental radiologic finding. Prophylactic resection after biopsy confirmation is not
• Fllure J9-1 A, Osteoid osteoma at a radiolucent area in the femoral neck (atrow). 8, Radiographically more apparent osteoid osteoma (atrow) in atypical location in femoral neck.
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
731
• FIgare 31-Z A, Increased uptake of technetium 99m in the region of an osteoid osteoma of the spine (al7llw). 8. Computed tomographic scan of the fourth lumbar vertebra delineates charaderistics of the osteoid osteoma of the vertebral arch. Notice the sclerotic central nidus, surrounded by radiolucent granulation tissue (al7llw), which is surrounded by sderotic reactive bone.
usually considered necessary, but careful follow-up is warranted, because there is a low rate of malignant transformation to chondrosarcoma. Cytogenetic abnormalities, not randomly distributed, are common and of value in tumor typing. 2s Multiple enchondromatosis, or Ollier's disease, commonly affects the hands and feet (see Chapter 40). Joint range of motion may be impaired, and the clinical presentation may be mistaken for arthritis. Growth deformities are common and require surgical management. Endochondromatosis in the presence of multiple cavernous hemangiomas is called Maffucci's syndrome
(Fig. 39-6). The frequency of malignant sarcomatous change is high (50%) in patients with Oilier's or Maffucci's syndrome. 3•26
Periosteal Chondroma Periosteal chondroma arises from the cortical surface and is most common in the proximal humerus and other long bones. Although the lesion is nontender, pain is often the presenting symptom. Wide excision is the treatment of choice.
Chondroblastoma Chondroblastoma is an uncommon epiphyseal cartilaginous tumor of childhood; it is most common in the second and third decades of life and occurs in the hip, shoulder, or knee (Fig. 39-7).27 The histopathology consists of polyhedral and giant cells with areas of fine calcifications. Foci of osteoid and bone may resemble a chondromyxoid fibroma. Most lesions are cured by excision and bone graft, but recurrences are a major concern. Growth disturbances and loss of function occur but are not common.
Chondromyxoid Fibroma A chondromyxoid fibroma arises usually in the metaphyseal area with pain and tenderness as the most common presenting symptoms. It is an uncommon lesion and begins to occur in children at about 10 years of age. 28 The radiographic appearance is one of an eccentric, sharply circumscribed zone of rarefaction, often with expansion of surrounding bone.
Benign Tumors of Fibrous nssue Fibrous Cortical Defect (Nonossifying Fibroma)
• Flture 31-3
Osteochondroma of the distal femoral metaphysis.The tumor is directed away from the joint.
Fibromas are common between the ages of 4 and 8 years, occur more frequently in boys, and may affect up to 40% of children. They are significant in that they may be
732
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
rare inherited disorder characterized by deposition of hyaline material in skin with the formation of nodules, in the musculoskeletal system with development of progressive joint contractures, and in viscera. 29 Death occurs by 2 years of age. The basic defect is in mutations in the capillary morphogenesis protein 2. 30 .31 Arabs, Japanese, and those living in the Indian subcontinent are particularly affected, thought by some to be a result of the frequency of consanguineous marriage among these ethnic groupS.32 Another subtype of the disorder is juvenile hyalinefibromatosis. 33 Both varieties are histologically similar and may represent a comparable or identical pathogenesis, Other names have included molluscum fibrosum and mesenchymal dysplasia. 34 The early onset of this disorder is in some ways similar to that of neonatal-onset multisystem inflammatory disorder. Further discussion of this topic is in Chapter 21.
Fibrous Dysplasia Fibrous dysplasia is difficult to classify and probably represents a developmental abnormality or a benign neoplastic fibrous tissue lesion. 3s It is a relatively common disorder with a variable presentation min1icking that of almost any bone lesion. Monostotic disease, most commonly in a rib, occurs in approximately 85%
• AlIIIre 39-4 Radiograph of the tibia and fibula with multiple osteochondromas (a17llws).
mistaken for more serious disease. There may be two or more fibromas, and they are usually an incidental finding (most often in the distal femur and proximal tibia) on radiographic evaluation. 8,9 The radiologic appearance is virtually diagnostic, with a sharply marginated eccentric lucency in the metaphyseal cortex (Fig. 39-8). These lesions disappear with increasing age, leaving no significant residual defect. They rarely cause symptoms, and treatment is conservative; occasionally, pathologic fractures occur.
Juvenile Fibromatosis The disorders known as the juvenile fibromatoses are rare and include juvenile aponeurotic fibroma, extraabdominal desmoid tumor, and diffuse infantile fibromatosis. Lipomas and neurofibromas are occasionally associated with musculoskeletal symptoms, often resulting in the development of scoliosis. A variety of other forms of fibromatous soft tissue lesions have been described in children. Infantile systemic hyalinosis is a
• AlIIIre 3fJ-5 Radiograph of the hand of a child with enchondromas of the middle and proximal phalanges of the second finger (a17llws).
C HAP T E R
39 SKELETAL MALIGNANCIES AND RELATED DISORDERS
733
• FItPa'e 39-6
A, Characteristic changes of Maffucci's syndrome. Notice the enchondromas of the proximal and middle phalanges of the second finger. The fusiform swelling of the third finger represents the soft tissue swelling of the hemangioma. 8, Radiograph of the humerus with the multiple enchondromas (alTOws) of Maffucci's syndrome.
of patients. Polyostotic fibrous dysplasia may be limited to two or three sites or result in extensive skeletal abnormalities. It is also part of the MCCune-Albright syndrome in association with multiple endocrine abnormalities. 36 The classic radiologic appearance of fibrous dysplasia is an intramedullary diaphyseal lesion with a thinned, sometimes bulging cortex. An angular deformity of the
• fIture 39-7
Chondroblastoma presenting as pain and effusion of the right knee of a 9-year-old boy. A, Anteroposterior view of a welldefined, oval, lytic lesion (alTOw). 8, lateral radiograph indicates that the lesion has eroded through the epiphysis (alTOw) with small foci of calcification.
bone is often present at the site of the lesion and may require surgical intervention, depending on its severity and the potential for pathologic fracture. Most monostotic lesions present no problem to the child, and resection is not indicated. Even with curettage, the recurrence rate is high. Bisphosphonates may be therapeutically indicatedY-w Osteofibrous dysplasia is a rare lesion of childhood that resembles monostotic disease.'o
734
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
has aided in diagnostic evaluation. A characteristic finding is low signal density on Tl- and T2-weighted studies.'" Treatment often requires surgical excision; this is difficult with the nodular variety because of extension of the lesion into tendon sheaths. NSAIDs are useful in suppressing the inflanunatory disease, and intra-articular glucocorticoids may have a role in management. Malignant PVS is rare, and its precise classification is controversial.'IH
Synovial Hemangioma
• Figure 39-8 Benign fibrous cortical defect of the metaphysis of the tibia (alTOw). This lesion was detected as an incidental finding in a radiograph taken for evaluation of trauma.
Ossifying Fibroma Ossifying fibroma, or osteofibrous dysplasia, almost always involves the mandible or tibiaY Although usually benign, it may be locally aggressive. Symptoms are often absent, and bony deformity prompts the consultation. Curettage is often unsuccessful, and observation alone is usually recommended after careful histopathologic review. 42
Benign Tumors of Soft nssue Pigmented Villonodular Synovitis Pigmented villonodular synovitis (PVS) may represent a benign neoplasia, be caused by an infectious process, or be associated with repeated episodes of intra-articular hemorrhage. This condition is rare in childhood, and is most common between 20 and 40 years of age. PVS affects males and females equally. It may present as recurrent swelling in a knee, ankle, or tendon sheath, and be nodular or diffuse.43---4'i Nodular disease affects joints, bursae, or tendon sheaths; in diffuse PVS, a monarthritis of the knee, ankle, or less frequently, the hip is most common. Only rarely is the upper extremity affected. The disorder is characterized by recurrent painless effusions and a slowly progressive destruction of cartilage and erosion of bone. A boggy fullness about the joint is present on clinical examination. The most striking feature is the presence of blood-stained, dark brown synovial fluid on joint aspiration. The synovium is dark and characterized by nodular areas of hypertrophy and hemosiderin-laden macrophages. There are proliferating synovial cells and fibroblasts, masses of stromal cells with frequent mitoses, and multinucleated giant cells. 46 MRI
Synovial hemangioma is most common in the knee but is an infrequent lesion. It may present as intermittent hemarthrosis simulating monarticular juvenile rheumatoid arthritis (IRA). Synovial hemangioma may be associated with contiguous cutaneous hemangiomas, varicose veins, and bone and soft tissue hypertrophy (Le., KlippelTrenaunay-Weber syndrome)49 or with capillary hemangiomas, thrombocytopenia, and depressed coagulation components (Le., Kasabach-Merritt syndrome).'iO Bleeding from the hemangioma results in the sudden onset of painful joint swelling with effusion, often after minor local trauma. Recurrences are common and may result in chronic inflammatory synovitis and joint damage. Aspiration of synovial fluid at the onset of an effusion produces frank blood; later, the fluid may be xanthochromic and have a high bilirubin content. Radiographs demonstrate soft tissue swelling or a mass and, occasionally, phleboliths. The extent of the lesion is more accurately judged by MRI (Fig. 39-9).
Synovial Chondromatosis In this unusual condition, synovial cartilaginous and osteocartilaginous bodies develop in the synovium and then are released free into the synovial fluid. Synovial chondromatosis is more common in boys and usually affects the knee. Presenting symptoms are usually those of a loose body with intermittent pain, swelling, locking, and giving way. In early disease, radiographic studies are normal, but as the lesions calcify, they may be identified as
• Figure 39-9 Magnetic resonance imaging study of the left knee of a 7-year-old boy with recurrent effusion and widespread cutaneous hemangiomata confirms the presence of a vascular mass just proximal to the knee joint (alTOw).
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
discrete areas of stippled calcification. Treatment requires surgical excision. Malignant transformation is rare. 51 ,52
Benign Tumors of Miscellaneous Origin Unicameral (Solitary) Bone Cyst Unicameral bone cyst is rare before 3 years of age, occurs most commonly between the ages of 6 and 10 years, and is more frequently diagnosed in boys. 53 It usually arises in the metaphysis of a long bone and often is asymptomatic or causes only localized pain. 54 Because of its size, it may result in localized swelling, a pathologic fracture, or growth disturbance of the Iimb. 5s Radiography demonstrates a lucent lesion that adjoins but does not cross the physis or a fusiform widening of the bone (Fig. 39-10). These cysts almost always require curettage and insertion of bone chips, although injection of the lesion with glucocorticoid has been effective.53
735
Onset may be insidious and prolonged over weeks to a few years. Radiographically, the lesion appears as a "bubble" with a circumscribed zone of rarefaction and surrounding destruction of the metaphyseal bone (Fig. 39-11). Differentiation from a unicameral cyst may be aided by MRJ.S8 Treatment requires surgical curettage, sometimes with resection or local irradiation or sclerotherapy.53,59,60
Giant Cell Tumor Pain is usually the presenting symptom of a giant cell tumor of bone. This tumor is most common in the second to third decades of life and occurs more frequently in girls. On radiographs, an expanding zone of eccentric radiolucency in the epiphysis of a long bone is characteristic. 2,3 Tumor size, location, and aggressiveness determine the outcome of the neoplastic process, if present,61
Eosinophilic Granuloma Aneurysmal Bone Cyst Anellrysmal bone cyst is less common than the unicameral cyst, occurs more frequently in girls, and has its peak frequency in adolescence to early adulthoodY' It presents most often as a reactive lesion with pain and swelling, commonly occurring in the metaphysis of a long bone or in a posterior element of the spine. 57
• Apre 39-10 Radiograph of a pathologic fracture (anvw) through the wall of a unicameral cyst of the tibia.
Eosinophilic granuloma, a lesion that is classified as a form of localized Langerhans cell histiocytosis (previously designated histiocytosis X), occurs predominantly
• Rgure 39-11 Radiograph of an aneurysmal bone cyst of the proximal fibula. Notice the benign cortical defect in the distal femur (anvw). The two abnormalities are unrelated.
736
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
L. 011'
TABLE 39-7 of Childhood
Age Distribution of Malignant Bone TUlllors
Percentage of TumOR of this type In: Tumor
In 1st Decade
Osteosarcoma 38 ParosteaI osteosarcoma 0 Ewing's sarcoma 45 Chondrosarcoma 2 Fibrosarcoma 2 Reticulum cell sarcoma 8
Percentage of Tumon of this type In:
In 2nd Decade
In 1st Decade
In 2nd Decade
60
s o
52
1
23 4
3 5
18 1 2
3
20 57 5 14
11
disposition, especially in Ewing's sarcoma and rhabdomyosarcoma. 68
Osteosarcoma
• Rgure 31-1Z Radiograph of the femur with changes (arrow) of Langerhans cell histiocytosis (Le., eosinophilic granuloma). In addition to the lytic lesion, there is marked periosteal new-bone apposition (arrowheads).
in young children between the ages of 5 and 10 years and most commonly affects boys. Characteristic findings at onset are localized pain and swelling over a solitary mass. 62 The bones of the skull, spine, and pelvis and the diaphysis of the femur are most commonly affected. A radiograph demonstrates a discrete lytic lesion with cortical erosion and a periosteal reaction (Fig. 39-12). Percutaneous needle biopsy is indicated to establish the diagnosis. 63 Treatment requires surgical curettage, sometimes with low-dose radiation. It has been suggested that intralesional glucocorticoid may be a useful adjunct to therapy.64 Other types of histiocytosis include HandSchitller-Cbristian disease and Letterer-Siwe disease.
MALIGNANT TUMORS Malignant Tumors of Bone Malignant musculoskeletal tumors of childhood account for 5% to 10% of malignant neoplasms; the most common of these lesions are osteogenic sarcoma, rhabdomyosarcoma, and Ewing's sarcoma. 7.65 Osteogenic sarcoma is rare in the first decade of life but is the most common malignant bone tumor in the second decade. The mean age of children with Ewing's sarcoma is younger than that for any other primary tumor of bone (Table 39-7). Survival rates (60% to 70%) have continued to improve modestly for these tumors. 66.67 There has also been progress in understanding their biology and genetic pre-
Osteosarcoma (Le., osteogenic sarcoma) accounts for 60% of all bone tumors in children. 69-73 It is most common at the time of maximal growth velocity in the second decade of life, with 75% of cases occurring between the ages of 8 and 25 years, especially in taller children. The annual incidence of osteosarcoma is approximately 0.6 to 0.7 per 100,000 children. 74 •75 It occasionally occurs in siblings.76 Approximately 3% of these tumors develop in a field that has received previous irradiation. Osteosarcomas are also associated with certain acquired or genetic disorders such as retinoblastoma, enchondromatosis, hereditary multiple exostoses, and fibrous dysplasia. An increased risk of osteosarcoma is associated with the Rotbmund-7bomson syndrome, a rare disorder characterized by short stature, t~langiectases, small hands and feet, and hypoplastic thumbs. 77 •7H This tumor usually arises in the medullary canal of the bone, has a metaphyseal location, and occurs most often (60%) around the knee (e.g., distal femur, proximal tibia) and in the proximal humerus. Osteogenic sarcomas are highly malignant and metastasize early by hematogenous spread to many organs, especially the lungs, where they are a significant cause of secondary hypertrophic osteoarthropathy. 79 Pain is the most common presenting symptom. Stl Swelling over the involved bone occurs a few weeks to months later. Secondary signs include local intlammation, involvement of regional lymph nodes, and loss of function of the limb. The presence of systemic signs such as weight loss, fever, or secondary hypertrophic osteoarthropathy suggests that skeletal and pulmonary metastases have already occurred. Radiologic investigations provide the most meaningful diagnostic information. HI On plain radiographs, the lesion has a moth-eaten appearance with cortical destruction, periosteal elevation O.e., Codman's triangle), and a soft tissue mass (Fig. 39-13). The differential diagnosis may include osteomyelitis. Occasionally, two or more tumor sites are present, representing a multifocal origin or metastases to bone. Bone scintigraphy, CT, or MRI may be indicated to delineate the extent of the lesion. The diagnosis of osteogenic sarcoma is confirmed by the
C HAP T E R
39 SKELETAL MALIGNANCIES AND RELATED DISORDERS
737
• Agure 39-13 A, Radiograph of an osteosarcoma of the distal femoral metaphysis in ayoung boy. B, Technetium 99m bone scan of the femurs documents increased uptake of isotope by the osteosarcoma. C, Computed tomographic section through the thigh shows the extracortical bony densities (alTOws) characteristic of osteosarcoma. D, Magnetic resonance imaging study demonstrates the tumor in the right thigh (alTOw).
histologic appearance on biopsy of marked cellular pleomorphism with spindle-shaped cells, chondrocytes, and osteoid. Osteosarcoma is commonly divided into five principal types: osteoblastic, chondroblastic, fibroblastic, telangiectatic, and a small-cell type that has features overlapping those of Ewing's sarcoma. The treatment of this tumor, with a 5-year survival rate of only 21%, has until recently included amputation of the limb. 66 Modification of the surgical approach, including segmental resection of the primary tumor 82 and the addition of adjuvant chemotherapy (including doxorubicin, cisplatinum, and high-dose methotrexate with leu-
covorin rescue), has led to a disease-free survival of more than 50% at 5 years. 74 .7 'i,83. 84 Prognostic features include the histologic grade of the tumor, size of the initial lesion, and its response to preoperative treatment. Metastases at diagnosis are associated with a poor outcome (less than 20% survival). The rare multifocal sclerosing osteosarcoma has a poor prognosis. Surface osteosarcomas do not involve the medullary cavity and often encircle the entire shaft of the bone. s5 They have been divided into two types: parosteal (or juxtacortical) and periosteal. These tumors behave differently from osteogenic sarcoma. The parosteal
738
C HAP T E R
39
SKELETAL MALlGNANCIES AND RELATED DISORDERS
variety is more common in girls and occurs most often on the posterior surface of the distal femur. Malignant potential is low, it tends to metastasize late, and it has a much better prognosis, The periosteal type occurs more commonly in the tibia and is associated with an abundant proliferation of cartilage.
Malignant Tumors of Cartilage: Chondrosarcoma Chondrosarcoma is a rare tumor (less than 5%) that develops in children from malignant transformation of a preexisting enchondroma or, later in life, in a patient with multiple heritable exostoses. 86 The initial symptoms are localized swelling or pain. 87 Radiographs are usually diagnostic with destruction of bone combined with mottled densities of calcification and ossification (Le., "popcorn" appearance). Treatment consists of wide resection, adjuvant chemotherapy, and irradiation. 811
Malignant Tumors of Fibrous nssue: Fibrosarcoma Fibrosarcoma is an uncommon tumor that occurs most often on a distal extremity as a soft, infiltrative mass with areas of hemorrhage or necrosis,89,90 Histopathology confirms fibroblastic or myofibroblastic differentiation of all degrees. 9l There are two major patterns of presentation: congenital fibrosarcoma is most common in boys younger than 2 years; postpubertal fibrosarcoma is more aggressive. 89,92 Onset is characterized in most instances by painful swelling, No diagnostic radiographic features distinguish this tumor from osteosarcoma. In children younger than 2 to 5 years, congenital fibrosarcomas undergo rapid growth and extensive local invasion despite a relative lack of distant metastasis (7%), In children older than 10 years, the metastatic rate approaches 50%, Treatment involves surgical resection; approaches to treatment of the postpubertal type combine radical excision, postoperative irradiation, and adjuvant chemotherapy,Y3 The overall combined survival rate is approximately 40% at 5 years and 30% at 10 years. Prognosis is obviously best for the more superficial and differentiated tumors, Fluorodeoxyglucose positron-emission tomography (FDG-PET) may have a role in identification and grading of these sarcomas of bone and soft tissue. 94
Malignant Tumors of Soft nssue Rhabdomyosarcoma Rhabdomyosarcomas are the most common soft tissue sarcomas in children and account for one half of all soft tissue neoplasms in patients younger than 15 years. 87,95-98 They occur most often in children between the ages of 2 and 6 years or during adolescence with an annual incidence of 0.4 to 0.9 per 100,000 children,75 and are rare in older age groups. The tumor presents as a localized, painless, soft tissue mass. These tumors are most com-
mon in the head and neck but can arise in any striated muscle. Approximately 20% occur in the extremities, especially during adolescence. Rhabdomyosarcoma metastasizes early to lung, bone, and bone marrow. It may erode into adjacent bone and produce a radiographic appearance of a soft tissue mass with an underlying periosteal reaction. 99 Treatment includes surgical excision, irradiation, and chemotherapy.lOIH02 The 5-year survival rate is approximately 70%,75,103,104
Synovial Cell Sarcoma Although rare in childhood, synovial cell sarcoma is the most common soft tissue sarcoma (6% to 10%) after the rhabdomyosarcomas.I05-IOB This tumor rarely occurs within a joint; it usually develops in the periarticular soft tissue. Lower extremity involvement is most common, especially around the knee or foot or in the hand. The tumor may be associated with calcifications that suggest the diagnosis on radiographs. Tendon sheaths may also be involved.109 Surgical excision, irradiation, and chemotherapy result in a 7-year survival rate of 60% to 70%. Prognosis is related to tumor size and ease of resectionYO-114 Most synovial sarcomas express a specific chromosomal abnormality: t(X;18) (pl1.2;ql1.2).1l5
Malignant Tumors of Miscellaneous Origin: Ewing's Sarcoma The Ewing sarcoma family of tumors probably arises from primitive multipotential mesenchymal or neural crest cell lineages in the medulla of bone or occasionally in soft tissue.72.116,l17 It is the most malignant of bone tumors and the second most common cancer of bone in childrenY8 It accounts for 7% to 15% of all malignant bone tumors in childhood with an annual incidence 0.2 to 0.3 per 100,000 children. 75 ,119 This lesion is most common in white boys in the second decade of life (male to female ratio of 1.5:1), although the mean age at onset is somewhat younger than that for osteogenic sarcoma. It is uncommon in children of Asian or African descent. Ewing's sarcoma often occurs in the diaphysis of the long bones (Le" femur, humerus, or tibia) or in the innominate, but it can develop in any bone, including those of the axial skeleton, or even as an extraskeletal lesion. 120 The tumor presents with pain and local swelling systemic signs, including fever, are common, often occurring with abnormalities of laboratory indices of inflammation. so The radiographic appearance is characteristically that of an aggressive, elongated, lytic lesion filling the medullary cavity, disrupting the cortex, and causing a roughening of the periosteum described as having an onion-skin or sunburst appearance (Fig. 39-14). This reaction may be easily confused with osteomyelitis. Ewing's sarcoma eventually involves the entire shaft of the long bone and metastasizes to other bones and lungs. The histologic appearance is that of sheets of small round cells that are positive on staining with periodic acid-Schiff reagent, sometimes with a perivascular pseudorosette appearance. Neural markers, chimeric fusion products, or a specific chromosomal translocation (t [11;22] [q24;q12D
C HAP T E R
39 SKELETAL MALIGNANCIES AND RELATED DISORDERS
739
• Figure 39-14 A, Radiograph of Ewing's sarcoma in the diaphysis of the femur.The central lytic lesion is accompanied by periosteal reaction (arrowheads). 8, Technetium 99m bone scan of the femurs of the same child with shows localized increased uptake of the isotope in the tumor.
in more than 95% of cases can be identified to differentiate this tumor from lymphoma, rhabdomyosarcoma, or neuroblastoma. Treatment combines surgical resection with irradiation and adjuvant chemotherapy.93,12l,J22 The 5-year survival rate varies from 50% to 80%, depending on the site of the primary tumor. W,124 Metastases to lung or skeleton reduce the survival rate to 20% to 30%. m,126 Allogenic or autologous stem cell transplantation has not improved survivorship.127
METASTATIC BONE TUMORS In childhood, metastatic bone tumors are uncommon except for neuroblastoma. In a retrospective review of metastatic skeletal disease in childhood over a 38-year period, Leeson and coworkers l28 described 39 patients ranging in age from 18 months to 20 years. The tumors most commonly producing skeletal metastases were neuroblastoma (41%), rhabdomyosarcoma (18%), teratoma-carcinoma (l (010) , Wilms' tumor (8%), and retinoblastoma (5%). Neuroblastoma, a tumor of the sympathetic nervous system, has an incidence of 1.6 cases per 100,000 persons, primarily young children. 75 Bone metastases usually occur early, and may be accompanied by fever, but little else in the way of localizing abnormalities. The development of bone pain related to bony metastases or bone marrow infiltration soon follows. Neuroblastoma most commonly metastasizes to the spine (81%), skull (69%), femur (50%), ribs (44%), and pelvis (31%). Multiple bony metastases are the rule.
The radiographic appearance is that of a lytic lesion arising from the marrow cavity (Fig. 39-15). Scintigraphy is the most sensitive technique for determining the site and number of metastases and may document abnormal findings before radiographic changes on plain film are evident. Treatment includes surgical removal of the tumor together with irradiation and chemotherapy. 129 The survival rate at 5 years is approximately 55%.75 Wilms' tumor is the most common retroperitoneal malignant tumor of childhood with an incidence of 2 cases per 100,000 children. 75 It occurs most commonly in infants and young children (less than 4 years old) in whom it usually presents as an abdominal mass. Tumors with a sarcomatous histology are particularly likely to metastasize to bone and may be associated with bone pain. Wilms' tumor is frequently associated with several congenital anomalies, including sporadic aniridia, hemihypertrophy, genitourinary anomalies, and a deletion in chromosome 11. 130 Treatment includes nephrectomy, irradiation, and chemotherapy. The 5-year survival rate is approximately 80%.75
MALIGNANT INRLTRATION OF BONE MARROW: LEUKEMIA Leukemia is the most common childhood malignancy that results in musculoskeletal pain and arthritis. Bl-133 In most instances, the pain is diffusely localized to one area of the body, particularly over the metaphyses of the long bones (Table 39-8). Sometimes, a joint effusion occurs.
740
C HAP T E R
39
SKELETAL MALIGNANCIES AND RE LATED DISORDERS
• Figure 31-15 A, Radiograph of the humerus illustrates the moth-eaten appearance of extensive metastatic neuroblastoma in the proximal humerus. 8, Technetium 99m bone scan shows increased uptake in the right humerus of the same child (alTOw).
Large joints, especially the knees, are most commonly affected, although small joints of the hands may be involved. The number of affected joints is usually relatively few. Associated periarticular disease is common. The frequency of musculoskeletal signs or symptoms in childhood leukemia was evaluated in a 2-year prospective study of 28 children. l3l Objective joint findings, often mild, were present in 50% of the children, most often at or near onset of the disease. In one half of the children, a single joint was involved, most often the knee; in the remainder, 2 or 3 joints, rarely more, were affected.
!'.
TABLE 39--8 in Children
Clinical Characleristics of Acute Leukemia
CharaderlsUc
DesalpUon
Presentation Pain
Low-grade fever, fatigue, pallor, weight loss Often disproportionate to objective findings Diffuse musculoskeletal aching or pain and tenderness of metaphyses of long bones May be migratory joint pain or periarticular swelling or joint effusion May be normal or with increased or decreased WBC count or platelet count, sometimes with blast cells in peripheral smear Increased ESR and lactate dehydrogenase and urate levels Metaphyseal rarefaction, periosteal new-bone apposition
Hematologic parameters
Radiographs
ESR.
erythrocyte sedimentation rate; WEC. white blood cell.
Retrospective studies have usually indicated a lower prevalence of joint disease of 11% to 12%.132,134 A high index of suspicion is needed to confirm the diagnosis of leukemia in a child presenting with musculoskeletal pain. 13S The most distinctive diagnostic features of leukemia are the degree and location of the pain. In leukemia, pain is much more severe than in JRA and is characteristically metaphyseal in location rather than directly over a joint. Abnormally elevated levels of acute phase indices such as the ESR are out of proportion to the small number of affected joints. Dissociation of the inflammatory indices (e,g., an elevated ESR with a normal or low platelet count), a low white blood cell count, or a striking increase in the serum level of lactic dehydrogenase or urate should also alert the physician to the possibility of leukemia. The hematologic findings (i.e" complete blood count, white blood cell differential count, and platelet count) may be normal for weeks or months after the onset of symptoms, and repeated evaluations are essential. Blast cells in the peripheral blood may not be present at onset of the musculoskeletal symptoms, in which case diagnostic bone marrow aspiration and biopsy are indicated. Antinuclear antibodies are occasionally detected in children with leukemia, and their presence should not be interpreted as an indication of inflammatory arthritis. 132 Radiographic changes may be of assistance in the diagnosis. In addition to localized metaphyseal rarefaction (Fig. 39-16), there may be subperiosteal elevation or an elongated osteolytic reaction, Such changes, however, may be absent even in the presence of
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
• figure 39-16
Radiograph of metaphyseal rarefaction in a patient with acute lymphoblastic leukemia (atrows).
severe symptoms. Scintigraphy with technetium 99m documents an increased uptake in marrow, and in metaphyseal and periosteal areas. Differentiation from osteomyelitis or neoplasm (e.g., Ewing's sarcoma) is paramount. Primary bone lymphoma, although rare, is best evaluated by MRI because plain x-ray films are often normal or near normal. 136
SECONDARY EFFECTS OF MALIGNANCY Secondary Hypertrophic Osteoarthropathy The syndrome of secondary hypertrophic osteoarthropathy (SHO) consists of terminal clubbing, painful swelling of distal joints and soft tissues, profuse sweating, and radiographic evidence of periosteal new-bone formation. affecting the hands, feet, and distal limbs (see Table 39-7). Although periosteal new-bone apposition can also be a radiologic characteristic of ]RA or juvenile psoriatic arthritis, it is not associated in these children with the severe pain and tenderness of SHOo Most cases of SHO in childhood are related to chronic pulmonary disease,129 congenital heart disease, or occasionally, the antiphospholipid antibody syndrome. 137 Associations with biliary atresia and regional enteritis have been reported. The pathogenesis of SHO is unknown, although hypoxic, endocrine, and neurogenic mechanisms have been suggested. SHO also occurs in the POEMS syndrome (i.e., polyneuropathy, organomegaly, endocrinopathy, M protein, skin changes) in adults. 137 An important cause of SHO in childhood is pulmonary malignancy, most often caused by metastases from
741
• Figure 39-17 Secondary hypertrophic osteoarthropathy in a child with pulmonary metastases from osteosarcoma. Notice the marked periosteal newbone apposition along the phalanges, metacarpals, and distal radius and ulna (atrows).
osteogenic sarcoma. 79 ,138 The typical clubbing and diffuse swelling of the joints and soft tissues of the hands, together with marked periosteal new-bone apposition along the proximal phalanges, metacarpals, and distal radius and ulna, are depicted in Figure 39-17. Scintigraphy with technetium 99m documents increased uptake in areas of involvement (see Fig. 15-3). Children with symptomatic SHO are usually profoundly ill. The pain is severe, predominantly distal in location, and symmetric in distribution. It is present during the daytime and may awaken the child at night. Medical treatment is usually unsatisfactory, although NSAlDs are sometimes temporarily effective. Resection of the pulmonary or pleural tumor may result in dramatic resolution of all signs and symptoms.
Acanthosis Nlgrlcans Acanthosis nigricans is a rare disorder associated with a number of conditions, including malignancy, diabetes, obesity, various genetic syndromes, connective tissue diseases such as dermatomyositis, and glucocorticoid administration.139.14o It may also occur in children with lipodystrophy or be familial. 141 In juvenile dermatomyositis, it is associated with premature thelarche, lipodystrophy, and insulin resistance (see Chapter 18).
ACKNOWLEDGMENT We are grateful for the expert assistance of Betty Wood, M.D., Department of Radiology, British Columbia's Children's Hospital, Vancouver, for selected radiographs.
742
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
REFERENCES 1. Tnteworthy RC, Templeton K]: Malignant bone tumors presenting as mus-
culoskeletal pain. Pediatr Ann 31: 355-359, 2002. 2. Mirra ]M, Picd P, Gold RH: Bone Tumors: Clinical, Radiologic, and Pathologic Correlations. Philadelphia, Lea & Febiger, 1989. 3. Dahlin DC, Unni KK: Dahlin's Bone Tumors: General Aspects and Data on 11,087 Cases, 5th ed. Philadelphia, Uppincott-Raven, 1996. 4. Herrera ]M, Krebs A, Harris P, et al: Childhood tumors. Surg Clin North Am 80: 747-760, xii, 2000, 5. Springfield DS, Gebhardt MC: Bone and soft tissue tumors. In Morrissy RT, Weinstein SL (eds): Lovell and Winter's Pediatric Orthapaedics, 6th ed. Philadelphia, Lippincott Williams & Wilkins, 2001, pp 507-562. 6. Arndt CA, Crist WM: Common musculoskeletal rumors of childhood and adolescence. N Eng! ] Med 341: 342-352, 1999. 7, Parkin DM, Stiller CA, Nectoux]: International variations in the incidence of childhood bone tumours. Int] Cancer 53: 371-376, 1993. 8. Silvennan FN, Kuhn ]P, Bender TM, et al: Essentials of Caffey's Pediatric xray Diagnosis. Chicago, Year Book Medical Publishers, 1990, 9. Resnick D, Greenway GD, Madewell ]E, et al: Diagnosis of Bone and Joint Disorders, 3rd ed. Philadelphia, WB Saunders, 1995, chapters 82--85. 10. Moore SG, Bisset GS Ill, Siegel M], et aJ: Pediatric musculoskeletal MR imaging, Radiology 179: 345-360, 1991. 11, Miller SL, Hoffer FA: Malignant and benign bone tumors. Radiol Oin North Am 39: 673-699, 2001. 12. Azouz EM: Magnetic resonance imaging of benign bone lesions: cysts and tumors, Top Magn Reson Imaging 13: 219-229, 2002. 13. Saifuddin A, Burnett S], Mitchell R: Pictorial review: ultrasonography of primary bone tumours. Clin Radiol 53: 239-246, 1998. 14. Copley L, Dormans ]P: Benign pediatric bone tumors, Evaluation and treatment. Pediatr Clin North Am 43: 949-966, 1996. 15. Biermann ]S: Common benign lesions of bone in children and adolescents, ] Pediatr Orthop 22: 268-273, 2002. 16. Aboulafia A], Temple HT, Scully SP: Surgical treatment of benign bone tumors, lnstr Course Lect 51: 441-450, 2002, 17. Cohen MD, Harrington TM, Ginsburg WW: Osteoid osteoma: 95 cases and a review of the literature, Semin Arthritis Rheum 12: 265-281, 1983. 18. Davidson RS, Mahhoubi S, Heyman S, et al: Nondiaphyseal osteoid osteomas in the pediatric patient. Clin Orthop 243: 230-234, 1989. 19, Graham GN, Browne H: Primary bony tumors of the pediatric spine. Yale] Bioi Med 74: 1--8, 2001. 20. Woods ER, Martel W, Mandell SH, et al: Reactive soli-tissue mass associated with osteoid osteoma: correlation of MR imaging features with pathologic nndings. Radiology 186: 221-225, 1993. 21. Biebuyck ]C, Katz ill, McCauley T: Soft tissue edema in osteoid osteoma. Skeletal Radiol 22: 37-41, 1993. 22. Drevelegas A, Chourmouzi D, Boulogianni G, et al: Imaging of primary bone tumors of the spine. Eur Radiol 13: 1859-1871, 2003. 23. Frassica F], Waltrip RL, Sponseller PD, et al: Clinicopathologic features and treatment of osteoid osteoma and osteoblastoma in children and adolescents. Orthop clin North Am 27: 559-574, 1996. 24, Porter DE, Emerton ME, Villanueva-Lopez F, et al: Clinical and radiographic analysis of osteochondromas and growth disturbance in hereditary multiple exostoses. ] Pediatr Orthop 20: 246-250, 2000. 25. Tallini G, Dorfman H, Brys P, et al: Correlation between clinicopathological featll1'es and karyotype in 100 cartilaginous and chordoid tumours. A report from the Chromosomes and Morphology (CHAMP) Collaborative Study Group. .I PathoI 196: 194--203. 2002. 26. Pierz KA, Womer RH, Dormans fP: Pediatric bone tumors: osteosarcoma ewing's sarcoma, and chondrosa~coma associated with multiple hereditary osteochondromatosis.] Pediatr Orthop 21: 412-418, 2001. 27, Schuppers HA, van del' Eijken JW: Chondroblastoma during the growing age, .I Pediatr Orthop B 7: 293-297, 1998, 28. Durr HR, Lienemann A, Nerlich A, et al: Chondromyxoid fibroma of bone. Arch Orthop Trauma Surg 120: 42-47, 2000. 29, Fayad MN, Yacoub A, Salman S, et al: Juvenile hyaline fibromatosis: two new patients and review of the literature. Am] Med Genet 26: 123-131, 1987, 30. Hanks S, Adams S, Douglas], et al: Mutations in the gene encoding capillary morphogenesis protein 2 cause juvenile hyaline fibromatosis and infantile systemic hyalinosis. Am .I Hum Genet 73: 791--800, 2003. 31. Dowling 0, Difeo A, Ramirez MC, et al: Mutations in capillary morphogenesis gene-2 result in the allelic disorders juvenile hyaline fIbromatosis and infantile systemic hyalinosis. Am .I Hum Genet 73: 957-966, 2003. 32. el Hazmi MA, al Swailem AR, Warsy AS, et al: Consanguinity among the Saudi Arabian population. .I Med Genet 32: 623-626, 1995. 33. Landing BH, Nadorra R: Infantile systemic hyalinosis: report of four cases of a disease, fatal in infancy, apparently different from juvenilt' systemic hyalinosis, Pediatr Pathol 6: 55-79, 1986. 34. Puretic S, Puretic B, Fiser-Herman M, et al: A unique form of mesenchymal dysplasia. BrJ Dermatol 74: 8--19, 1962. 35. Stanton RP, Hobson GM, Montgomery BE, et al: GIlIcocorticoids decrease interleukin-6 levels and induce mineralization of cultured osteogenic cells from children with fibrous dysplasia.] Bone Miner Res 14: 1104--1114, 1999. 36. Lumbroso S, Paris F, Sultan C: McCune-Albright syndrome: molecular genetics. .I Pediatr Endocrinol Metab 15 (Suppl 3): 875--882, 2002.
37, Devogelaer JP: Treatment of bone diseases with bisphosphonates, excluding osteoporosis. Curr Opin Rheumatol 12: 331-335, 2000. 38. Chapllrlat RD, Meunier P]: Fibrous dysplasia of bone, Baillieres Best Pmct Res Clio Rheumatol 14: 385-398, 2000. 39, Schoenau E, Rauch F: Fibrous dysplasia. Horm Res 57 (Suppl 2): 79--82. 2002. 40. McCaffrey M, Lells M, Carpenter B, et al: Osteofibrolls dysplasia: a review of the literature and presentation of an additional 3 cases. Am ] Orthop 32: 479-486, 2003, 41. Williams HK, Mangham C, Speight PM: Juvenile ossifying fibroma. An analysis of eight cases and a comparison with other Ilbro-osseous lesions. ] Oral Pathol Med 29: 13-18, 2000. 42, Folpe AL, Weiss SW: Ossifying fibromyxoid tumor of soft parts: a clinicopathologic study of 70 cases with emphasis on atypical and malignant variants. Am] Surg Pathol 27: 421-431, 2003. 43. Bravo SM, Winalski CS, Weissman BN: Pigmented villonodular synovitis. Radial Clin North Am 34: 311-326, 1996. 44, de Visser E, Veth RP, Pruszczynski M, et al: Diffuse and localized pigmented villonodular synovitis: evaluation of treatment of 38 patients. Arch Orthop Trauma Surg 119: 401-404, 1999. 45. Somerhausen NS, Fletcher CD: Diffuse-type giant cell tumor: clinicopathologic and immunohistochemical analysis of 50 cases with extraarticular disease. Am .I Surg Pathol 24: 479-492, 2000, 46. Schumacher HR, Lotke P, Athreya B, et al: Pigmented villonodular synovitis: light and electron microscopic studies, Semin Arthritis Rheum 12: 32-43, 1982. 47. Frassica FJ, Khanna ]A, McCarthy EF: The role of MR imaging in soft tissue tumor evaluation: perspective of the orthopedic oncologist and musculoskeletal pathologist. Magn Reson Imaging Clin North Am 8: 915-927, 2000. 48. Bertoni F, Unni KK, Beabout JW, et aJ: Malignant giant cell tumor of the tendon sheaths and joints (malignant pigmented villonodular synovitis). Am] Surg Pathol 21: 153-163, 1997. 49. Jones KL: Klippel-Trenaunay-Weber syndrome. In Jones KL (ed): Smith's Recognizable Pallerns of Human Malformation, 5th ed, Philadelphia, WB Saunders, 1997, pp 512-513, 50, Resnick D, Oliphant M: Hemophilia-like arthropathy of the knee associated with cutaneous and synOVial hemangiomas. Report of 3 cases and review of the literature. Radiology 114: 323-326, 1975. 51. Wuisman PI, Noorda RJ, Julle PC: Chondrosarcoma secondary to synovial chondromatosls. Report of two cases and a review of the literature. Arch Orthop Trauma Surg 116: 307-311, 1997. 52, Sciot R, Dal Cin P, Bellemans .I, et al: Synovial chondromatosis: clonal chromosome changes provide further evidence for a neoplastic disorder, Vlrchows Arch 433: 189-191, 1998, 53. Campanacci M, Capanna R, Picci P: Unicameral and aneurysmal bone cysts. Clin Orthop 204: 25-36, 1986, 54. Lokiec F, Wientroub S: Simple bone cyst: etiology, classillcation, pathology, and treatment modalities. .I Pediatr Orthop B 7: 262-273, 1998. 55. Clayer M, Boatright C, Conrad E: Growth disturbances associated with untreated benign bone cysts. Aust N ZJ Surg 67: 872--873, 1997. 56, Leithner A, Windhager R, Lang S, et al: Aneurysmal bone cyst. A population hased epidemiologic study and literature review, Clin Orthop 363: 176-179, 1999. 57. Rizzo M, Dellaero DT, Harrelson .1M, et al: ]uxtaphyseal aneurysmal bone cysts. Clin Orthop 364: 205-212, 1999. 58, Sullivan R], Meyer ]S, Dormans .II', et al: Diagnosing aneurysmal and unicameral bone cysts with magnetic resonance imaging. Clin Orthop 366: 186-190, 1999. 59. Ozaki T, Halm H, Hillmann A, et al: Aneurysmal bone cysts of the spine, Arch Orthop Trauma Surg 119: 159-162, 1999, 60, Dubois], Chigot V, GrimaI'd G, et al: Sclerotherapy in aneurysmal bone cysts in children: a review of 17 cases. Pediatr Radial 33: 365-372, 2003. 61. Boutou-Bredaki S, Agapios P, Papachristou G: Prognosis of giant cell tumor of bone, Histopathological analysis of 15 cases and review of the literature. Adv Clin Pathol 5: 71-78, 2001. 62. Burgdorf WH, Zeiger B: The non-Langerhans' cell histiocytoses in childhood. Cutis 58: 201-207, 1996. 63. Yasko AW, Fanning CV, Ayala AG, et al: Percutaneous techniques for the diagnosis and treatment of localized Langerhans-cell histiocytosis (eosinophilic granuloma of bone). ] Bone Joint Surg Am 80: 219-228, 1998. 64. Cohen M, Zornoza .I, Cangir A, et al: Direct injection of methylprednisolone sodium succinate in the treatment of solitary eosinophilic granuIom'l of bone: a report of 9 cases, Radiology 136: 289-293, 1980. 65. Hoffer FA: Primary skeletal neoplasms: osteosarcoma and ewing sarcoma. Top Magn Reson Imaging 13: 231-239, 2002. 66, Mercuri M, Capanna R, Manfrini M, et al: The management of malignant bone tumors in children and adolescen~s. Clin Orthop 264: 156-168, 1991. 67. Himelstein BP, Dormans ]P: Malignant bone tumors of childhood. Pediatr Clin North Am 43: 967-984. 1996. 68. Van den BH: Biology and therapy of malignant solid tumors in childhood. Cancer Chemother Bioi Response Modif 20: 605-625, 2002. 69. Jaffe N: Osteosarcoma, Pediatr Rev 12: 333-343, 1991. 70. Link MP, Eilber F: Osteosarcoma. In Pizzo PA, Poplack DG (eds): Principles and Practice of Pediatric Oncology, 3rd ed. Philadelphia, Lippincott-Raven, 1997, pp 889-920. 71. Whelan .IS: Osteosarcoma. EurJ Cancer 33: 1611-1618, 1997,
C HAP T E R
39
SKELETAL MALIGNANCIES AND RELATED DISORDERS
72. Yaw KM: Pediatric bone tumors. Semin Surg Oncol 16: 173-183, 1999. 73. Rytling M, Pearson P, Raymond AK, et al: Osteosarcoma in preadolescent '. patients. Clin Orthop 373: 39-50, 2000. 74. Goorin AM, Abelson HT, Frei E 111: Osteosarcoma: ftfteen years later. N Engl .1 Med 313: 1637-1643, 1985. 75. Crist WM, Kun LE: Common solid tumors of childhood, N Engl J Med 324: 461-471, 1991. 76. Colyer RA: Osteogenic sarcoma in siblings. Johns Hopkins Med .1 145: B1-135. 1979. 77. Leonard A. Craft AW, Moss C, et al: Osteogenic sarcoma in the RothmundThomson syndrome, Med Pediatr Oncol 26: 249-253. 1996. 78. EI Khoury JM, Haddad SN, Atallah NG: Osteosarcomatosis with RothmundThomson syndrome. BrJ Radiol 70: 215-218, 1997, 79, Petty RE, Cassidy.!T, Heyn R, et al: Secondary hypertrophic osteoarthropathy. An unusual cause of arthritis in childhood. Arthritis Rheum 19: 902-906, 1976. 80, Widhe B, Widhe 1': 1nitial symptoms and clinical features in osteosarcoma and Ewing sarcoma, J Bone Joint Surg Am 82: 667-674, 2000. 81. Fletcher BD: Imaging pediatric bone sarcomas, Diagnosis and treatrnentrelated issues. Radiol Clin North Am 35: 1477-1494, 1997. 82. San Julian M, Aquerreta JD, Benito A, et al: 1ndications for epiphyseal preservation in metaphyseal malignant bone tumors of children: relationship between image methods and histological findings. J Pediatr Orthop 19: 543-548, 1999. 83. Lane JM, Hurson B, Boland PJ, et a1: Osteogenic sarcoma, Clin Orthop 204: 93-110, 1986. 84, Ferguson WS, Goorin AM: Current treatment of osteosarcoma, Cancer Invest 19: 292-315, 2001. 85. Vander Griend RA: Osteosarcoma and its variants. Orthop Clin North Am 27: 575-581, 1996. 86. Kivioja A, Ervasti H, Kinnunen J, et al: Chondrosarcoma in a family with multiple hereditary exostoses. J Bone Joint Surg Br 82: 261-266, 2000, 87. Carli M, Gugliemi G, Sotti G, et al: Soft tissue sarcomas. In Plowman PN, Pinkerton CR (eds): Paediatric Oncology: Clinical Practice and Controversies, 1st ed. London, Chapman & Hall, 1992, pp 291-324. 88. Lee FY, Mankin HJ, Fondren G, et al: Chondrosarcoma of hone: an assessment of outcome. .1 Bone Joint Surg Am 81: 326-338, 1999, 89. Soule EH, Pritchard OJ: Fibrosarcoma in infants and children: a review of 110 cases. Cancer 40: 1711-1721, 1977. 90. Neville H, Corpron C, Blakely ML, et al: Pediatric neurofibrosarcoma. .1 Pediatr Surg 38: 343--346, 2003, 91. Ward WG, Savage P, Boles CA, et al: Fine-needle aspiration biopsy of sarcomas and related tumors. Cancer Control 8: 232-238, 2001. 92. Trobs R, Meier 1', Bennek J, et al: Fibrosarcoma in infants and children: a retrospective analysis--{)verdiagnosis in earlier years, Pediatr Surg Int 15: 123--128, 1999, 93, Kennedy JG, Frelinghuysen P, Hoang BH: Ewing sarcoma: current concepts in diagnosis and treatment. Curr Opin Pediatr 15: 53--57, 2003, 94. Bastiaannet E, Groen H, Jager PL, et al: The value of FOG-PET in the detection, grading and response to therapy of soft tissue and bone sarcomas: a systematic review and meta-analysis, Cancer Treat Rev 30: 83-101, 2004. 95. Merlino G, Helman LJ: Rhabdomyosarcoma-working out the pathways, Oncogene 18: 5340--5348, 1999. 96. Dagher R, Helman L: Rhabdomyosarcoma: an overview. Oncologist 4: 34-44, 1999. 97, Ruymann FB, Grovas AC: Progress in the diagnosis and treatment of rhabdomyosarcoma and related soft tissue sarcomas. Cancer Invest 18: 223--241, 2000. 98. Raney RB: Soft-tissue sarcoma in childhood and adolescence, Curr Oncol Rep 4: 291-298, 2002, 99. McCarville MB. Spunt SL, Pappo AS: Rhabdomyosarcoma in pediatric patients: the good, the bad, and the unusual. AJR Am .1 Roentgenol 176: 1563--1569, 2001. 100, Pappo AS, Shapiro ON. Crist WM: Rhabdomyosarcoma, Biology and treatment, Pediatr Clin North Am 44: 953--972, 1997. 101. Andrassy RJ: Rhabdomyosarcoma, Semin Pediatr Surg 6: 17-23, 1997, 102, Schalow EL, Broecker BH: Role of surgery in children with rhabdomyosarcoma. Med Pediatr Oncol 41: 1-6, 2003. 103. Raney RB, Anderson JR, Barr FG, et al: Rhabdomyosarcoma and undifferentiated sarcoma in the first two decades of life: a selective review of intergroup rhabdomyosarcoma study group experience and rationale for Intergroup Rhabdomyosarcoma Study V. J Pediatr Hematol Oncol 23: 215-220, 2001. 104, McDowell HP: Update on childhood rhabdomyosarcoma. Arch Dis Child 8H: 354-357, 2003. 105. Schmidt D, Thum P, Harms 0, et al: Synovial sarcoma in children and adolescents. A report from the Kiel Pediatric Tumor Registry, Cancer 67: 1667-1672, 1991. 106. Ferrari A, Casanova M, Massimino M, et al: Synovial sarcoma: report of a series of 25 consecutive children from a single institution. Med Pediatr Oncol 32: 32-37, 1999, 107. Skytting B: SynOVial sarcoma. A Scandinavian Sarcoma Group project. Acta Orthop Scand (Suppl 291): 1-28. 2000, lOR Andrassy RJ, Okcu MF, Despa S, et al: Synovial sarcoma in children: surgical lessons from a single institution and review of the literature. J Am Coli Surg 192: 305-313, 2001.
743
109. Hajdu SI, Shiu MH, Fortner JG: Tendosynovial sarcoma: a clinicopathological study of 136 cases, Cancer 39: 1201-1217, 1977, 110. Bergh P, Meis-Kindblom JM, Gherlinzoni F, et al: Synovial sarcoma: identitlcation of low and high risk groups. Cancer 85: 259l'>-2607, 1999. 111. Skytting BT, Bauer HC. Perfekt R, et al: Clinical course in synovial sarcoma: a Scandinavian sarcoma group study of 104 patients. Acta Orthop Scand 70: 53l'>-542, 1999. 112. Skytting B, Meis-Kindblom JM. Larsson 0, et al: Synovial sarcoma-identification of favorable and unfavorable histologiC types: a Scandinavian sarcoma group study of 104 cases. Alta Orthop Scand 70: 543--554, 1999. 113. Thompson RC Jr, Garg A, Goswitz J, et al: Synovial sarcoma. Large size predicts poor outcome. Clin Orthop 373: 18-24, 2000. 114, LewLs .1.1, Antonescu CR, Leung DH, et al: SynOVial sarcoma: a multivariate analysis of prognostic factors in 112 patients with primary localized tumors of the extremity. .1 Clin Oneal 18: 2087-2094, 2000. 115. Fisher C: Synovial sarcoma. Ann Diagn Pathol 2: 401-421. 1998, 116, Horowitz ME, Malewer MM, Woo SY, et al: Ewing's sarcoma family of tumors: Ewing's sarcoma of bone and soft tissue and the peripheral primitive neuroectodermal tumors. In Pizzo PA, Poplack DG (eds): Principles and Practice of Pediatric Oncology, 3rd ed. Philadelphia, JB Lippincott-Raven, 1997, pp 831-864, 117, West DC: Ewing sarcoma family of tumors. Curr Opin Oncol 12: 323--329, 2000, 118. Vlasak R, Sim FH: Ewing's sarcom.~. Orthop Clin North Am 27: 591-603, 1996. 119. Miser JS, Triche Tj, Kinsella 1'.1. et al: Other soft tissue sarcomas of childhood. In Pizzo PA, Poplack DG (eds): Principles and Practice of Pediatric Oncology, 3rd ed. Philadelphia, JB Lippincott-Raven, 1997, pp 865-888. 120. Ahmad R, Mayol BR, Davis M. et al: Extraskeletal Ewing's sarcoma. Cancer 85: 725-731, 1999. 121. Shankar AG, Pinkerton CR, Atra A, et al: Local therapy and other factors intluencing site of relapse in patients with localbed Ewing's sarcoma. United Kingdom Children's Cancer Study Group WKCCSG). Eur j Cancer 35: 1698-1704, 1999. 122. Pinkerton CR, Bataillard A, Guillo S, et al: Treatment strategies for metastatic Ewing's sarcoma. Eur J Cancer 37: 1338-1344, 2001. 123, Rosito P, Mancini AF, Rondelli R, et al: Italian Cooperative Study for the treatment of children and young adults with localized EWing sarcoma of bone: a preliminary report of 6 years of experience. Cancer 86: 421-428, 1999. 124, Mclean TW, Hertel C, Young ML, et al: Late events in pediatric patients with Ewing sarcoma/primitive neuroectodermal tumor of bone: the Dana-Farber Cancer Institute/Children's Hospital experience, J Pediatr Hematol Oncol 21: 486-493, 1999. 125. Mameghan H, Fisher RJ, O'Gorman-Hughes 0, et al: Ewing's sarcoma: longterm follow-up in 49 patients treated from 1967 to 1989. Int J Radiat Oncol Bioi Phys 25: 431-438, 1993. 126. Cardenas-Cardos R, Rivera-Luna R, Lopez-Facundo NA, et al: Ewing's sarcoma: prognosis and survival in Mexican children from a single institution. Pediatr Hematol Oncol 16: 519-523, 1999, 127. Burdach S, van Kaick B, Laws HJ. et a1: Allogeneic and autologous stemcell transplantation in advanced Ewing tumors. An update after long-term follow-up from two centers of the European Intergroup study EICESS. Stem-Cell Transplant Programs at Dusseldorf University Medical Center, Germany and St. Anna Kinderspital, Vienna, Austria. Ann Oncol 11: 1451-1462, 2000. 128. Leeson Me. MaldeyJT, Carter JR: Metastatic skeletal disease in the pediatric population, J Pediatr Orthop 5: 261-267, 1985. 129. Losty P, Quinn F, Breatnach F. et al: Neuroblastoma-a surgical perspective. Eur.J Surg Oncol 19: 33-36, 1993. 130, Breslow NE, Beckwith JB: Epidemiological features of Wilms' tumor: results of the National Wilms' Tumor Study. .1 Natl Cancer Imt 68: 429-436, 1982. 131. Costello PB, Brecher ML, Starr .11, et al: A prospective analysis of the frequency, course, and possible prognostic significance of the joint manifestations of childhood leukemia. J Rheumatol 10: 753--757, 1983. 132. Saulsbury FT, Sahio H: Acute leukemia presenting as arthritis in children. Clin Pediatr (Phila) 24: 625-628. 1985, 133. Jean-Baptiste G, De Ceulaer K: Osteoarticular disorders of haematological origin. Baillieres Best Pract Res Clin Rheumatol 14: 307-323, 2000, 134. Fink CWo Windmiller J, Sartain P: Arthritis as the presenting feature of childhood leukemia. Arthritis Rheum 15: 347-349, 1972. 135. Gallagher OJ, Phillips OJ, Heinrich SO: OrthopediC manifestations of acute pediatric leukemia, Orthop Clin North Am 27: 635-644, 1996. 136. Krishnan A, Shirkhoda A, Tehranzadeh J, et al: Primary bone lymphoma: radiographic-MR imaging correlation. Radiographics 23: 1371-1383. 2003. 137. Martinez-Lavin M: Hypertrophic osteoarthropathy. Curr Opin Rheumatol 9: 83-86, 1997, 138. Roebuck OJ; Skeletal complications in pediatric oncology patients. Radiographies 19: 873-885. 1999. 139. Stuart CA, Driscoll MS, Lundquist KF, et al: Acantbosis nigricans. J Basic Clin Physiol Pharmacol 9: 407-418, 1998, 140. Baird JS, Johnson JL, Elliott-Mills D. et al: Systemic lupus erythematosus with acanthosis nigricans, hyperpigmentation, and insulin receptor antibody, Lupus 6: 275-278, 1997. 141. Dhar S, Dawn G, Kanwar AJ, et al: Familial acanthosis nigricans, 1nt J Dermatol 35: 126-127, 1996,
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES Carol B. Lindsley, Ross E. Petty and Judith G. Hall -UD
L~
A variety of uncommon primary disorders of bone or connective tissue may manifest in childhood with symptoms that suggest a diagnosis of one of the inflammatory arthritides, although they do not have an inflammatory cause. They are genetically determined disorders with swelling and prominence of joints, .hypermobility, or joint contractures, and they are often associated with disproportionately short or tall stature. This chapter is not intended to be comprehensive but provides an approach to the identification and differential diagnosis of the conditions most likely to manifest in childhood with musculoskeletal pain or degenerative joint disease. Many excellent references include differential diagnoses. l -6 An international working group frequently updates a classification of osteochondrodysplasias. 7 These disorders are identified numerically (McKusick numbers). The latest primary references are also listed in Online Mendelian Inheritance in Man (OMIM), to which there is access through the Internet,s
ESTABLISHING A DIAGNOSIS Although the specific diagnosis of many of the primary disorders of bones and joints lies in the realm of genetics, the pediatric rheumatologist must be able to differentiate such disorders from diseases of inflammatory pathogenesis. In so doing, consideration should be given to a family history of similar complaints, the pattern of heredity, age at onset of signs or symptoms, natural history of the disorder, distribution of affected bones and joints, radiographic abnormalities, other organ involvement, and pathologic changes on examination of tissues. Because specific genes responsible for some of these diseases have been identified, mutational analysis may also contribute to diagnosis. In an excellent review, Chalom and associates9 suggested that the clinical characteristics in Table 40-1 should alert the physician to the possible diagnosis of a congenital or familial arthropathy, rather than inflammatory arthritis.
744
Family History In reviewing the family history, careful questioning about consanguinity (suggesting an autosomal recessive disorder) and about advanced paternal age (suggesting a new dominant mutation) should be undertaken. Frequently, the disease in question may have a known genetic basis, but there may be no family history of the disorder.
Age at Onset The presence of disease at birth helps distinguish diseases such as spondyloepiphyseal dysplasia congenita and diastrophic dysplasia from those with onset later in childhood such as Stickler's syndrome or progressive pseudorheumatoid arthropathy of childhood. The age at which abnormalities first become evident is characteristic of each disorder (e.g., Hurler's syndrome in the first year, pseudoachondroplasia in the second year). The age at which abnormalities first appear and the order in which various body parts become involved are characteristic of individual syndromes.
Distribution of Involvement The distribution of affected bones, JOints, or other connective tissues is often characteristic and can usually be determined by physical examination. The examiner should check for abnormalities of joint range of motion (Le., hypermobility or contractures), disproportion between the trunk and the limb, and disproportion within segments of the limb. Relative shortness of the most proXimal segment of the limb is called rhizomelic shortening, when the middle segment is involved, it is called mesomelic shortening, and when the most distal segment is affected, acromelic shortening is present.
Radiographic Abnormalities A radiographic approach to the differential diagnosis is used to determine what part of the bone (e.g., epiphysis,
C HAP T E R
t. r
ABLE 40-1
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
Characteristics That Suggest Congenital
or ramilial Arthropathy
Presence of alleged juvenile rheumatoid arthritis in more than one family member Presence of two or more dysmorphic features Absence of rheumatoid factor and antinuclear antibody Absence of evidence of systemic or synovial inflanunation (normal erythrocyte sedimentation rate, normal synovial fluid cell count) From Chalom EC, Ross], Athreya BH: Syndromes and arthritis. Rheum Dis Clin North Am 23: 709, 1997.
metaphysis, diaphysis) is involved and whether the spine is affected. Often, the radiographic changes are not present at birth, They may become much more prominent with age or, as in pseudoachondroplasia, may disappear with time, Many of the bony dysplasias are named according to the area of bone affected (e,g., multiple epiphyseal dysplasias, spondyloepiphyseal dysplasias).
Involvement of Other Organs Some of the primary disorders of bones and connective tissues have specific associated congenital anomalies, such as structural heart abnormalities in Ellis-van Creveld syndrome or cleft palate in spondyloepiphyseal dysplasia congenita and diastrophic dysplasia, Patients with others disorders develop complications such as retinal detachment, deafness, or storage disorders with time,
MOLECULAR MECHANISMS OF DISEASE Pathologic Changes and Pathogenic Mechanisms The pathologic changes in tissues have helped to define the metabolic pathways involved in many of these disorders. In general, they can be separated into four categories: abnormalities of connective tissues (i.e" fibers or matrix changes), abnormalities in growth factors and their receptors, abnormalities of transmembrane regulation, and abnormalities of transcription factors.
Mutations Most types of primary bone and connective tissue disorders have a genetic basis and represent somatic mutations, new dominant mutations, dominantly inherited conditions, autosomal recessively inherited disorders, or X-linked disorders (recessive or dominant). Different disorders can arise from different mutations in a single gene, In osteogenesis imperfecta, almost every new mutation has its own particular site along the type I collagen gene. However, in some genes, there are "hot spots" where more than one new mutation occurs. For instance, in achondroplasia, more than 95% of all new mutations occur at exactly the same nucleotide within the transmembrane part of the fibroblast growth factor-3 receptor gene, The rate of mutation at this site is almost 1000 times that at other sites.
745
Mosaicism With newer molecular techniques, it is possible to distinguish whether a mutation is present in all cells within an individual or in only a proportion of cells (i,e., mosaicism). A person may have patchy involvement and be relatively mildly affected, whereas in another, every cell in the body is involved, and the individual is much more severely affected, This phenomenon has occurred in mutations of type II collagen in which the parent is described clinically as having Stickler's syndrome and the child as having Kniest's syndrome, It is therefore important to examine both parents to identify minor changes suggesting mosaicism. 10
Parent of Origin Parent-of-origin effects (i.e" genomic imprinting) occur when the severity or other characteristics of a disease depend on whether the disease-associated gene is inherited from the mother or from the father, For instance, in Albright's hereditary osteodystrophy, children are more severely affected when they inherit the condition from their motherY
COMMON CLINICAL CONCERNS Arthritis children with a wide range of primary disorders of connective tissue may present with arthritis or what appears to be arthritis (Table 40-2).12-17 Others develop arthritis as a result of degenerative changes in the joints,6 The development of "wear and tear" degenerative joint disease is related to abnormal alignment, irregular joint surfaces, ligamentous laxity, and abnormal cartilage. In conditions such as achondroplasia, asymmetric growth (Le., overgrowth of the fibula) leads to bowing of the fibula, as a result of which weight Of unevenly distributed across the knee joint) leads to wear and tear on the cartilaginous surface of the tibia, Biochemical abnormalities of the cartilage or supporting structures may limit their ability to withstand even normal weight bearing and movement. If the bones of the skull or face are affected, malalignment of teeth may result, and there may be abnormal stresses on the temporomandibular joints, Some patients with disproportionately short stature tend to be obese, which further contributes to the risk of degenerative joint disease (especially in achondroplaSia), Joint contractures may suggest the presence of inflammatory arthritis, but there is generally no joint swelling, although juxta-articular muscle atrophy may make the joints appear disproportionately large, Contractures are characteristic of a number of primary disorders of connective tissues (Table 40-3) .1&-S6 A more detailed account of these disorders has been provided in a review by Chalom and colleagues,9
Effects of Ligamentous Laxity Many children with ligamentous laxity have hypermobility of joints of the extremities and complain of muscu-
746 ~
I
C HAP T E R
I ABLE 40-l
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
Primary Risorders of (oruw
.IS
Arllll ills
Disorder
Joint AlmcHmallUes
Other Features
CACP syndrome 12
Polyarthritis with contractures, camptodactyly at birth
Arthritis with scoliosis 13 Hereditary osteolysis14 Nail-patella syndrome15
Polyarthritis; finger flexion contractures "Arthritis" in wrists, ankles, elbows, metacarpophalangeal, interphalangeal joints; limitation of elbow range, ulnar deviation of wrist; painless deformity of wrists and feet Polyarthritis; restricted elbow range
Pericarditis Coxa vara Thoracic scoliosis Marfan-like syndrome
Stickler's syndrome J6
Symmetric polyarthritis
Trichorhinophalangeal dysplasia J7
Enlarged proXimal interphalangeal joints, short fourth and fifth metacarpals, progressive hip arthritis
Hypoplastic patellae and thumbnails, radial head dislocations, iliac horns, clubfeet Flat facies, long philtrum, epicanthal folds, myopia, hypermobility, kyphosis, cleft palate Bulbous nose, thin hair, large ears, micrognathia
CACP, camptodactyly, arthropathy, coxa Yam, pericarditis,
loskeletal pain, Recurrent subluxations of joints may occur, Children with many types of spondyloepiphyseal dysplasias have poor odontoid development and, because of loose ligaments, may have subluxation of C1 on C2 with spinal cord compression, Congenital and familial disorders characterized by joint hypermobility are shown in Table 40-4,18,57-71
Surgical and Anesthetic Concerns Many persons with primary disorders of bones and joints require surgery, and particular attention must be paid to anesthetic risks, As is the case in children with juvenile idiopathic arthritis (JIA), preanesthetic radiographic evaluation of the cervical spine and protection of the neck during surgery help to avoid neurologic damage resulting from cervical instability. Similarly, intubation may be difficult because of restriction of spinal movement or micrognathia. In conditions such as diastrophic dysplasia, pressure on the tracheal cartilages can cause swelling and obstruction of the airway. Spinal anesthesia may be difficult in children with spondyloepiphyseal dysplasias, and great care should be taken when using spinal anesthesia in any patient with disproportionately short stature. Intravenous access may be difficult in children with incomplete extension of the elbows.
OSTEOCHONDRODYSPLASIAS Definition and Oasslficatlon The osteochondrodysplasias are developmental disorders of chondral and osseous tissue that are often accompanied by short stature. 72 They include defects of tubular and flat bones or of the axial skeleton, disorganized development of cartilaginous and fibrous components of the skeleton, and idiopathic osteolyses. As genetic abnormalities responsible for these disorders are identified, the traditional classification shown in Table 40-5 will be modified. A number of osteochondrodysplasias have autosomal dominant inheritance patterns, but most occur as the result of new mutations. For example, 85% to 90% of cases of achondroplasia and pseudoachondroplasia result from new mutations, Some disorders in this group
have a nongenetic basis. Warfarin-induced embryopathy, maternal lupus embryopathy, and congenital rubella syndrome are associated with abnormal bone growth and chondrodysplasia punctata.
Achondroplasia Family Achondroplasia and hypochondroplasia are the similar and related disorders of the achondroplasia family. They can manifest as possible inflammatory arthritis because of degenerative joint changes in weight-bearing joints and limitation of range of motion, but the disproportionately short stature is striking, and the diagnosis is unlikely to be missed after the newborn period. Members of the achondroplasia family have point mutations of the fibroblast growth factor receptor-3 gene (FGFRjJ on chromosome 4p16,3. 73
Classic Achondroplasia Classic achondroplasia CMcKusick 100800) is the most common form of viable disproportionate short stature. It occurs with a frequency of 1 case in 20,000 births, The distinguishing features are present at birth, although the diagnosis is not always made at that time. The head is large, with frontal bossing, midface hypoplasia, and prognathism. There is rhizomelic shortening of arms and legs, increased lumbar lordosis, bowing of the legs, overgrowth of the fibula, and a trident-shaped hand with short, broad phalanges. Kyphosis at 11 occurs because of hypotonia of the trunk in the first few years of life. Radiographs show a very small sacrosciatic notch of the pelVis, chevron changes of the epiphyses of the distal femur, and constriction at the base of the skull. The pedicles of the vertebrae are short, leading to symptomatic spinal stenosis in many affected persons. Prenatal diagnosis is possible through DNA testing and through sequential ultrasound measurement of the growth of the long bones. The natural history and optimal medical supervision of patients with achondroplasia have been well defined by the American Academy of Pediatrics. 74 Adult life is shortened when spinal stenosis causes spinal cord compression, but otherwise, most of these persons live a healthy, productive, and independent life.
C HAP T E R
1'1 "'"
1ABLE 40- 3
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
747
Congenital dnd Fdnlllial Disorders with ContrMtures or SliII Joints
~'_.
Charaderlstlc Joint Abnormalities
Syndrome Aarskog's syndrome
1s
Achondroplasia 19 Antley-Bixler syndrome 20 Apert's syndrome" Arthrogryposis'" 23 Beals' syndrome" Gangliosidosis 1'; Chromosome 2p deletion 26 Cockayne's syndrome 27 Conradi-Hunermann28 Farber's disease 29 ,30 Multicentric osteolysis l4 Cornelia de Lange syndrome 3l Diastrophic dwarfism 32 Ectrodactyly-ectodermal dysplasia syndrome33 Fabry's disease (angiokeratoma corporis diffusum)34 Pena-Shokier syndrome3; GEMSS syndrome (glaucoma, lens ectopia, microspherophakia, short stature)36 Homocystinuria 37 Hypochondroplasia 38 Jansen's dysostosis (Jansen's metaphyseal chondrodysplasia)39 Leri pleonosteosis40 Leri-Weill dyschondrosteosis41 Marden-Walker syndrome42 Metatrophic dwarfism43 Mucolipidoses (MLSS)44 !-cell disease (MLS II) Pseudo-Hurler's syndrome CMLS III) Mucopolysaccharidoses (MPSs)45 Hunter's syndrome (MPS II) Hurler's syndrome (MPS I-H) Maroteaux-Lamy syndrome (MPS VI) San Filippo syndrome (MPS III) Scheie's syndrome (MPS I-S) Riley-Day syndrome (familial dysautonomia)46 Schmid's syndrome (metaphyseal chondrodysplasia syndrome)47 Schwartz-Jampel syndrome 48 Seckel's syndrome49 Spondyloepiphyseal dysplasia" Spondylometaphyseal dysplasia (Kozlowski's syndrome)5l Trisomy 5q52 Trisomy 853 Weaver syndrome 54 Weill-Marchesani syndrome ss Winchester's syndrome 56 Zellweger's syndrome S7
Hyperextensible proximal interphalangeal (PIP) joints with distal joint restriction; short fifth finger with clinodactyly Incomplete extension of the elbow Congenital contractures of wrist, finger, hip, knee, ankle Ankylosis of elbow, shoulder, hip Stiffness, multiple contractures of large and small joints Contractures of knee, elbow, hand; arachnodactyly Contractures of elbow, knee, claw hands Contractures of hip, knee, ankle Mild contractures of knee, elbow, ankle Variable contractures; punctate bone mineralization Symmetric polyarthritis "Arthritis" of wrist, ankles, elbows, metacarpophalangeal and interphalangeal joints; limited elbow motion, ulnar deviation of involved hand; painless deformity of wrist and feet Flexion contractures of elbows Limited flexion of PIP joints and elbow; clubfoot Limited extension of elbows; abnormal third digit Bony swelling, distal interphalangeal joints, flexion deformity Multiple joint contractures, camptodactyly Reduced mobility of large joints Enlarged joints with reduced mobility Mild reduction of elbow mobility Contractures of hips and knees Limited mobility; flexion contractures of digits, broad thumb Limitation of mobility at wrist and elbow Multiple congenital contractures, micrognathia Limited movement at knee, hip; hypermobile fingers Limited hip flexion; hips dislocated Limited motion of hands, elbows, knees, shoulders, claw hand Joint contractures, claw hand Limitation of extension, claw hand Limited motion of knee, hip, elbow Mild joint stiffness Limitation of motion in hands and feet; claw hand Neuropathic joint (knee) Mild decrease in finger, wrist, and elbow extension; genu varum Limited motion at hip, wrist, fingers, toes, spine Limited motion at elbows; dislocated hips Contractures at elbows, knees, hips; pain, soft tissue swelling at PIP joints, hipS, elbows Limited mobility of hips, elbows Contractures at hips, elbows Limited elbow supination Limited extension of elbow and knee; camptodactyly Progressive joint stiffness Symmetric flexion contractures with pain at fingers, elbows, hips, knees, ankles Variable contractures especially of knees, fingers; ulnar deviation of hand
Adapted from Chalom EC, Ross], Athreya BH: Syndromes and arthritis, Rheum Dis Clin North Am 23: 709, 1997,
Hypochondroplasia Hypochondroplasia (McKusick 146000) is a milder form of disproportionate short stature involving several different mutations of the FGFR3 gene. Affected persons are often slightly taller and have less enlargement of the head than those with achondroplasia. 38
gen synthesis is steadily increasing. They include most of the Ehlers-Danlos syndromes (EDSs) and a number of others (Table 40-6). At least five different types of collagen are found in articular cartilage (types II, IV, IX, X, and XI), and genetically determined defects in these molecules might be expected to be associated with joint disease,75
Collagen Disorders
Ehlers-Danlos Syndromes
The number of primary disorders of bones and connective tissues that are associated with identified defects in colla-
The EDSs are disorders of connective tissue characterized by hypermobility, easy bruising, and hyperextensibility
748 ~
*'..
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
TABI E 40-{1 (ongenital and F,lIl1ilidl Syndromes Asso(lated wilh Hypermobilily
I!; II lABLE 40 -0 Collagen 'JYpe
Aarskog's syndrome l8 Coffin-Lowry syndrome s8 Coffin-Siris syndromes9 Cohen's syndrome60 Costello's syndrome6l Cutis laxa-growth deficiency syndrome62 Dubowitz's syndrome") Ehlers-Danlos syndrome rype II"" Hajdu-Cheney syndrome (aero-osteolysis, arthrodentodysplasia)h5 Kabuki makeup syndrome66 Larsen's syndrome6' Marfan syndrome"" Cartilage-hair hypoplasia"9 Morquio's syndrome (MPS IV)'O Velo-facio-skeletal syndrome 71 From ChaJom EC, Ross], Athreya BH: Syndromes and althritis. Rheum Dis Clin North Am 23: 709, 1997.
of the skin and sometimes by atrophic scars, molluscoid pseudotumors, and epicanthal folds (Table 40-7; see also Fig, 40-5) (McKusick 120215, 120160, 120180, 225410, 225320, 130000-90).62,72,73,76 The nine types of EDS have traditionally been considered as a group, but as their genetic and biochemical abnormalities are elucidated, it is more appropriate to consider them in terms of the spe-
II:III
TABLE 40 5 Intemalional (Idssili(ation of Ostcochondrodysplasias
Defects of Tubular and Flat Bones and/or AxIal Skeleton Achondroplasia group Achondrogenesis Spondylodysplastic group Metatropic dysplasia group Short-rib dysplasia group Atelosteogenesis/diastrophic dysplasia group Kniest-Stickler dysplasia group Spondyloepiphyseal dysplasia congenita group Other spondyloepiphyseal and metaphyseal dysplasias Dysostosis multiplex group Spondylometa physeal dysplasias Epiphyseal dysplasias Chondrodysplasia punctata group Metaphyseal dysplasias Brachyrachia Mesomelic dysplasias Acromelic/acromesomelic dysplasias Dysplasias with significant membranous bone involvement Bent-bone dysplasia group Multiple dislocations with dysplasias Osteodysplastic primordial dwarfism group Dysplasias with decreased bone density Dysplasias with defective mineralization Dysplasias with increased bone density
Disorganized Development of Cartilaginous and FIbrous Components of the Skeleton Idiopathic osteolysis Predominantly phalangeal Predominantly carpal/tarsal Multicentric Other Fmm Spl"dnger E]p, for the International Working Group on Constitutional Disease of Bone: International classification of osteochondrodysplasias. Eur] Pedi
II
III
IV V VI IX X XI
C1assifi(ation of Genetic Disorders of (ollagen
Disorder Osteogenesis imperfecta Idiopathic juvenile osteoporosis Ehlers-Danlos type VII Stickler's syndrome Kniesl's syndrome Spondyloepiphyseal dysplasia congenita Spondyloepiphyseal dysplasia tarda Ehlers-Danlos type III Ehlers-Danlos type IV Ehlers-Danlos type VIII Ehlers-Danlos type VI Ehlers-DanIos type I Ehlers-Danlos type II Cutis laxa Fairbank's multiple epiphyseal dysplasia Schmid's type metaphyseal dysplasia Stickler's syndrome type II Otospondylomegaepiphyseal dysplasias
cific biochemical abnormalities of collagen that they represent. Two others are no longer included in this disease category; one EDS previously designated as type IX (occipital horn syndrome) (McKusick 304150) is now grouped with disorders of copper metabolism (Menkes' syndrome),and the former type XI has been grouped with the benign hypermobiIity syndromes (McKusick 130020), A classification of disorders of collagen based on their known biochemical abnormalities in shown in Table 40-6.
Disorders ofType I Collagen Mutations of the type I collagen genes (McKusick 120150 and 120160) cause a spectrum of abnormalities, from familial osteoarthritis and osteoporosis to severe osteogenesis imperfecta, Each family appears to have its own mutation. Severely affected persons have short stature, blue sclerae, dentinogenesis imperfecta, and osteoporosis with breakable bones. In these patients, joint laxity, cardiac valve regurgitation, myopia, and degenerative joint changes may occur with age, Type I collagen mutations should be considered when any of these features are present.
Osteogenesis Imperfecta Osteogenesis imperfecta, one of the most common heritable disorders of connective tissue (I case in 20,000 births), is inherited in an autosomal recessive or an autosomal dominant manner, depending on the disease subtype (Table 40-8),77 The various subtypes of osteogenesis imperfecta are related to the site of the mutation that affects the chains of the collagen triple helix. 77 The severity of the disease ranges from a slight increase in susceptibility to fractures of bones with improvement in adolescence (type I) to forms that are lethal in utero or in the neonatal period (type 11), In type III, fractures occur throughout life (Fig. 40-1). In some children, blue sclerae, otosclerosis, and dentinogenesis imperfecta occur, The skin is excessively thin, with abnormal scar formation. The joints are often hyperextensible. In addi-
C HAP T E R
t.
TABLE 40-7
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
749
Ehlers·Danlos Syndromes
Olnlcal Characteristics Hyperextenslblllty Type
GenetIcs
Biochemical Defect
Skin
Joints
[ (gravis)
AD
Type V collagen
+++
+++
II (mitis) III (benign) IV (arterial)
AD
Type V collagen Type III collagen Type III collagen
++
AD
+
++ +++ +
Lysyl oxidase deficiency Type IV collagen
++ +++
+ +++
Defective conversion of type I procollagen to collagen Type III collagen Fibronectin functional deficiency
++
+++
Severe disease, fragile vasculature, molluscoid pseudommors Mild disease Benign hypermobility Ecchymoses, rupmre of vessels, GI tract, "china doll" appearance Ecchymoses, atrophic scars Retinal detachment joint dislocations, scoliosis, hypotonia Congenital hip dislocations, short stamre
++ +
++ +++
Periodontitis Ecchymoses, scarring mitral valve prolapse
V VI (ocular)
AD, AR, sporadic XLR AR
VII
AD, AR
VIII X
AR
AD
±
Other Features
AD. "utosomal domimnt; AR, "utosoma] recessive; Gl, gastrointestinal; XLR, X-linked recessive; ±, equivocal; +, mild; ++, moderate, +++, severe.
tion to multiple fractures and progressive osteoporosis, radiographs reveal a large skull with wormian bones in the suture lines, A number of disorders resemble osteogenesis imperfecta7S (Table 40-9), Increased bone turnover and resorption are characteristic. Plasma osteocalcin levels are elevated, although serum calcium, phosphorus, and alkaline phosphatase concentrations are normal,79 Treatment with calcitoninSo ,81 or bisphosphonates82 has been suggested, Short- and intermediate-term results of treatment with bisphosphonates indicate that bone pain and the frequency of fractures are reduced, but long-term implications are not yet known,78 Long-term management requires expert physical therapy and orthopediC surgery.83.84
Idiopathic Juvenile Osteoporosis There are many forms of osteoporosis (McKusick 114130, 166710, 120160, 259750), some of which are associated with type I collagen mutations. Idiopathic juvenile osteoporosis manifests late in childhood or at puberty with pain in the joints, usually the ankles or knees, and is characterized by the development of metaphyseal fractures resulting from osteopenia,85-87 Back pain may result from vertebral fractures. Severely affected children develop protrusio acetabuli. All biochemical investigations are normal except for the presence of hypercakiuria. The metabolic abnormalities disappear after completion of growth. 88 Minor fractures around joints may simulate arthritis.
Ehlers-Danlos SyndromeType VII -il • r ABLE 40-8 Classification of Osteogenesis Imperfeda Type Inheritance Sclera AD II
III
IV
AD
AD
AD
Blue
Deafness
Skeletal Abnormalities
Normal stature, little or no deformity Blue No Congenital fracmres, marked long bone abnormalities, platyspondyly, beaded ribs; lethal in perinatal period Variable Common Progressive deformity of bones, usually with moderate deformity at birth; very short stature; dentinogenesis imperfecta Normal Uncommon Variable short stature, mild to moderate bone deformity; dentinogenesis imperfecta 50%
AD, mllosom"l dominant; AR, autosomal recessive. Adapted from Sillence DO, Senn A, Banks OM: Genetic heterogeneity in osteogenesis imperfecta. J Med Genet 16: 101, 1979.
EDS type VII (i.e., arthrochalasis multiplex congenita) is characterized by extreme laxity of the joints that results in early dislocations of the hips, short stature, and mandibular hypoplasia, together with the characteristics of EDS type I. The disorder results from a splicing defect that causes a deletion of a peptidase cleavage site on the a l or the a/I) genes of type I collagen, preventing the normal processing of type I procollagen (McKusick 130060, 225410).89,90
en
Disorders ofType II Collagen Type II collagen is found primarily in hyaline cartilage, the nucleus pulposus, and the vitreous of the eye. Mutations of the type II collagen gene lead to osteoarthritis and abnormal growth and often affect the eye and the spine. Diversity within the spectrum seems to be related to mutations in different domains of the gene (i.e., particular domains relate to degrees of severity). Because of hyperextensibility and abnormal cartilage, degenerative changes in the joints often occur at an early age.
750
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
• Figure 40-1 Radiographs of the legs of an 8-month-old boy with osteogenesis imperfecta who has had many fractures. A, The arrow indicates area of periosteal new bone along the tibia. 8, The arrow indicates the location of a supracondylar fracture.
Occasionally, a parent is affected in a mosaic form with much milder involvement. The child in whom all tissues are involved is likely to be more severely affected.
pattern of inheritance. Children with this syndrome may have a marfanoid body habitus, with hyperextensibility of the jointsY' Enlargement of the knees, ankles, and wrists---often congenital-can be misdiagnosed as ]IA or the chronic infantile neurocutaneous and articular syndrome CClNCA). Associated but variable features include deafness, myopia, cataracts, congenital glaucoma, and retinal degeneration and detachment. 92 Midfacial
Stickler's Syndrome Stickler's syndrome CMcKusick 184840) is a heterogeneous group of disorders with an autosomal dominant
-'I
[ . r ABLE
4()··9
Skeletal Disorders that Resemble Osteogenesis Imperfecta
Disorder
Bone Fragility
Charaderlstlcs
Inheritance
Gene Defed
Bruck syndrome
Moderate/ severe Moderate
Congenital joint contraclUres
AR
telopeptide Iysyl hydroxylase deficiency
Congenital blindness
AR
LRP5
Severe
Cystic or ground-glass lesions in all hones, phosphatase levels, wide diaphyses Raised alkaline phosphatase levels, wide diaphyses, thick calvareum Low alkaline phosphatase level Craniosynostosis/proptosis No extraskeletal abnormalities
Somatic mutation
GNAS
AR
7NFRSFllB
AR. AD
ALPL
Unknown None
Unknown Unknown
Osteoporosis/ pseudoglioma Panostotic fibrous dysplasia Idiopathic hyperphosphatasia
Severe
Hypophosphatasia
Mild to severe
Cole-Carpenter syndrome Idiopathic juvenile osteoporosis
Severe Mild to moderate
Adapted from Rauch F. Glorieux FH: Osteogenesis imperfecta. Lancet
363:1377-13A~.
2004.
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
751
Kniest's Syndrome
• figure 40-2
Fragmentation of the femoral epiphysis in a child with
Stickler's syndrome.
The Kniest, or Swiss-cheese cartilage, syndrome (McKusick 156550), inherited as an autosomal dominant disorder of type II collagen, is characterized by congenitally short limbs and trunk and by macrocephaly with a round face and a depressed nasal bridgeY'i Progressive stiffness of the fingers, dislocation of the hips, and kyphoscoliosis develop. A cleft palate, hypertelorism, myopia, retinal detachment, deafness, and hernias are characteristic. Later, enlargement of the joints and severe contractures interfere significantly with mobility and are associated with pain. Radiographs reveal platyspondyly, flared metaphyses, and epiphyseal deformity in the tubular bones (Fig. 40-3). The cartilaginous growth plate is abnormal, with poorly calcified matrix (i.e., Swiss-cheese cartilage). The differences between the Stickler and the Kniest syndromes represent effects of mosaicism. In a severe form of Kniest's dysplasia, structurally abnormal type II collagen was caused by a dominant mutation that caused skipping of exon 24. 96
Spondyloepiphyseal Dysplasias hypoplasia, a cleft palate, and micrognathia are common. Intelligence is normal. Radiographs in the newborn show characteristic dumbbell-shaped long bones with enlarged epiphyses and metaphyses. With increasing age, the epiphyses become fragmented, and degenerative arthritis develops (Fig. 40-2), There may be mild platyspondyly and steeply sloping ribs. Scoliosis and kyphosis are common in older childrenY3 In Stickler's syndrome type I, the defects in the COL2Al gene for type II collagen have been localized to the long arm of chromosome 12 02q 13.1-qI3.3).94 A and B, Radiographs of a knee of a 9-year-old boy with Kniest syndrome show irregular caldfication of cartilage, giving a Swisscheese appearance. The metaphyses are flared, and the epiphyses are deformed. (A and 8, Courtesy of Dr. R. Cairns.) • FI..... 40-3
The spondyloepiphyseal dysplasias (SEDs) constitute a group of closely related disorders characterized by short stature and a disproportionately short trunk (Table 40-10). Infants with the most severe forms of SEDs within the type II collagen spectrum are obviously dwarfed at birth. Many other, less severely affected patients may not present until later in life, and their symptoms can easily be confused with those of osteoarthritis. They present with short stature and symmetric joint pain caused by epiphyseal changes. The possibility of an inborn error of connective tissue metabolism should be considered when there is
752 ~
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
I
III
TABLE 40- I 0
Type
Onset
Inheritance
AssocIated Features
Congenita
Birth
AD
Tarda
Late childhood
XLR
Short limbs, equinovarus feet, myopia, retinal detachment Moderately short stature, platyspondyly, onset of pain in hips and back at about 10 years old; progressive deformity
Spondyloepiphyseal Dysplasias
AD, autosomal dominant; XLR, X-linked recessive.
symmetric involvement or epiphyseal fragmentation, or both, associated with arthritis. In SED congenita (McKusick 183900), the limbs are short, and equinovarus deformities of the feet are common. Radiographs show dysplastic and late-developing femoral heads, marked platyspondyly, and characteristic abnormalities of the pelvis and vertebral bodies, especially in the cervical and lumbosacral regions. 50,97 This autosomal dominant condition reflects mutations in the COL2A 1 gene located on the long arm of chromosome 12 (l2qI3.1-q13.3)' A specific RNA-splicing mutation (G+51VS20) has been described,98 and an associated immunodeficiency has been reported in some patients. In the X-linked recessive form of SED tarda (McKusick 313400), there is moderate shortness of stature, principally from platyspondyly, with onset of hip or back pain and stiffness by 5 to 10 years of age,99,IOO Radiographic changes include a characteristic posterior hump on the vertebral bodies, which eventually become fused to each other, and mild fragmentation of the epiphyses of the hips and shoulders (Fig. 40-4). The milder SEDs with coxa vara are often characterized by an autosomal dominant inheritance. A progressive "pseudorheumatoid" arthritis has been described in some children with SED tarda,loJ.to2 and pseudogout has
been observed. 103 Hypoplasia of the odontoid predisposes the child to cervical cord injury from mild trauma.
Disorders ofType III Collagen Ehlers-Danlos Syndrome Type III EDS type III (McKusick 225350) is inherited as an autosomal dominant trait or results from a new mutation. The distinguishing features are minimal, and the child often has only hypermobility, although some patients have hyperelasticity of the skin and cutaneous striae over the lower back. Affected children may have aortic root dilatation, early-onset periodontitis, and fetal growth retardation. I04-106 A mutation in COLjAl has been demonstrated in a patient with EDS type III. 107
Ehlers-Danlos Syndrome Type IV The distinguishing features of EDS type IV (McKusick 130050, 225350), of which there are at least four subtypes, are the marked vascular fragility and aneurysms that mainly affect the aorta, medium-sized muscular arteries, and cerebral arteries. Other characteristics include prematurely aging and thinning of skin over the dorsa of the hands (Le., acrogeria) (types IVA and IVB), sparse scalp hair, large eyes (Le., china doll appearance), and lobeless ears. Ruptures of pleura, peritoneum, or intestinal diverticula may occur. Musculoskeletal abnormalities include aero-osteolysis of the fingertips, bilateral clubfeet, and dislocated hips. A variety of mutations of the COLjAl gene on the long arm of chromosome 2 cause abnormalities of type III collagen in patients with EDS type IV. 108 In some families, mosaicism is present. 109 Histologic examination of the skin shows characteristic collagen depletion and elastin proliferation. Eighty percent have at least one complication by age 40, and median survival of a reported cohort was 48 years. tlO • Figure 40-4 Spondyloepiphyseal dysplasia. A, Radiograph of the spine demonstrates the characteristic "humps" on the vertebral bodies. 8, Flattening and sderosis of the femoral capital epiphysis. (A and 8, Courtesy of Dr. R. Cairns.)
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
Ehlers-Danlos SyndromeType VIII The outstanding characteristics of EDS type VIII (McKusick 130080) include periodontal disease, with alveolar bone resorption and early loss of teeth, and thickened scars in the pretibial areas. It is an autosomal dominant defect, possibly involving the synthesis of type III collagen. I 11
Disorders of Type IV Collagen An autosomal recessive disorder, III EDS type VI (McKusick 130050, 225350) is characterized by neonatal hypotonia and, later, by retinal detachment, thin sclerae, scleral herniation, ligamentous laxity, tendency to late aortic rupture, short stature, kyphoscoliosis, and features of EDS type I. Cystic malformation of the meninges may occur. I II It is associated with abnormalities of lysyl hydroxylase that result in a defiCiency of hydroxylysine in type IV collagen. 114 Gene transfer studies in rodents have restored normal enzyme activity. m
753
Disorders ofType V Collagen EDS types I and II (McKusick 305200), the most common of the EDS variants, closely resemble each other but vary in severity. These disorders are inherited as autosomal dominant traits or occur as new mutations. A defect in type V collagen, a mutation in the COL5Al gene encoding the pro-a.] (V) fibrillar collagen chain, has been identified in type I and type II EDS.116 In EDS type I, the hyperextensible skin tends to split over bony prominences, with tissue paper scars occurring over the shins (Fig. 40-5). The hands, face, and feet are usually broad. Pectus excavatum is not present, and the palate is normal. Shoulder dislocations can occur. Mitral valve prolapse and, rarely, aortic rupture occur. M •1l7 Fibrous nodules (i.e., molluscoid tumors) occur over the elbows, knees, and heels. EDS type II has all of the manifestations of type I but to a much milder degree.
• Figure 40-5 Achild with Ehlers-Danlos type I shows hypermobility of the thumb (A), hyperextensibility of the skin of the forearm (8), and a fishmouth type of scar over the knee (C). (A-e, Courtesy of Dr. 1. Prendiville.)
754
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
Disorders ofType VI Collagen Cutis laxa (McKusick 123700) is characterized by hyperextensibility of the skin. It is a genetically heterogeneous group and can be divided into subtypes. Excessive production of type VI collagen and increased expression of mRNA for type VI collagen has been demonstrated in fibroblasts from patients with this rare disorder. 11A
chromosome 18q21.1. 12h Affected newborns always present with some limitation of movement. Patients are often shott, have an exaggerated lumbar lordosis and sternal prominence, and have progressive mental retardation. They develop c1awhand deformities. Radiographs show platyspondyly epiphyseal dysplasia, irregular metaphyses, and a lacy appearance of the iliac crests. Biopsy shows widened cisternae of rough endoplasmic reticulum in chondrocytes. 127 Dyggve-Melchior-Clausen syndrome and Smith McCott dysplasia (similar but without mental retardation) are allelic disorders.12H.129
Disorders ofType IX Collagen Persons with Fairbank's multiple epiphyseal c(ysplasia (McKusick 120260) often appear normal until they experience joint pain and symmetric epiphyseal changes are detected radiographically. These persons may be of normal height and have progressive degenerative joint changes that may require joint replacement. This disease may result from abnormalities in the cattilage oligometric matrix protein gene (COMP).119
Progressive Pseudorheumatoid Arthropathy Infants and young children with this autosomal recessive disorder (McKusick 208230) are often thought to be completely normal, but with time, they develop swollen joints with progressive stiffness and kyphoscoliosis.l,;o Radiographs show platyspondyly (i,e., narrowing of joint spaces) and widening of the metaphyses (Fig. 40-6). This disorder mimics JIA but has
Disorders ofType X Collagen In the 5chmid ~ype afmetaphyseal dysplasia (McKusick 120110), an autosomal dominant disorder in which children appear normal at bitth, there is progressive enlargement of joints, with a waddling gait and limitation of joint movement. The limbs are relatively shortened. Osteoarthritis is progressive.
Disorders ofType XI Collagen Mutations in three genes coding for type XI collagen result in syndromes that resemble Stickler's syndrome type L120
Ehlers-Danlos Syndromes Types V and X EDS type V (McKusick 153455) resembles EDS types II and III and has been associated with deficiency of Iysyl oxidase, although the relation of this biochemical defect is not certain. 121 Affected persons have joint hyperlaxity, fragility of skin and blood vessels, and a risk for uterine rupture. 122 In addition to joint hypermobility and hyperextensibility of skin, as occurs in patients with EDS type II, patients with the rare EDS type X have abnormal platelet aggregation associated with a fibronectin abnormality.123 A variety of other abnormalities of hemostasis have been reported in patients with several types of EDS. 124
Disorders Characterized by Stiff Joints Diastrophic Dysplasias Diastrophic dysplasia (McKusick 222600, 600972) and its variants are characterized by enlarged joints (patticularly the knees), clubbing of the feet, and limitation of finger movement. Features include progressive fragmentation and calcification of the cattilage with swelling and eventual fusion of the joints, patticularly the small joints of the phalanges. Pressure on the cattilage leads to cell death with calcification of the cartilage in many parts of the body, including the ears, trachea, and costochondral junctions. Diastrophic dysplasia is an autosomal recessive disorder caused by a mutation in the sulfate transporter gene. 12)
Dyggve-Melchior-Clausen Dysplasia Dyggve-Melchior-Clausen dysplasia (McKusick 223800) is a rare autosomal recessive disorder. The gene for the disorder is on
• Hlure 40-& Widening of the metaphyses in a child with platyspondyly and progressive pseudorheumatoid arthropathy.
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
none of the laboratory abnormalities of that disease. The specific gene has not been determined, although a locus on chromosome 6q22 has been suggested. 13 ! At least six genes involved in collagen synthesis are also located in this region.
Wolcott-Rallison Dysplasia In Wolcott-Rallison dysplasia, an autosomal recessive disorder (McKusick 226980), there is stiffness and pain in the joints, leading to difficulty walking. The spine may also be affected. Infancyonset diabetes mellitus is usually present; with time, affected persons develop renal insufficiency and hepatomegaly.13Z Collagen fibrils in cartilage appear abnonnal. Mutations of the gene encoding translation factor-2a kinase (EIF2AK3) have been identified. 133
Disorders Characterized by a Short Trunk A short trunk can be a part of any condition with platyspondyly and is often accompanied by back pain. Brachyolmias of the Hobaek type (McKusick 271530) and Maroteaux type (McKusick 217530) are characterized by severe shortening of the trunk caused by platyspondyly that becomes evident during childhood. These autosomal recessive disorders manifest with back pain, sometimes with scoliosis or spinal stenosis. Radiographs reveal characteristic lateral extension of the vertebral bodies. 134
Mesomellc Dysplaslas Mesomelic dysplasias are characterized by shortening of the middle segments of the limbs. The most common one is LeriWeill dysostosis (McKusick 127300) (Fig. 40-7). It is present at birth and, particularly in girls, is characterized by a Madelung deformity (i.e., hypoplasia and dorsal subluxation of the distal ulna with shortening and bowing of the radius), which can be quite painful. It was thought to be an autosomal dominant disorder, but the gene has been found to be X-linked, and specific mutations in the short stature homeobox gene (SHOX) have been described. 13s A patient with Leri-Wei! dysostosis and systemic lupus erythematosus has been reported. 136
• FI..... 40-7 Radiographs of the forearm of ayoung boy with Leri-Weill dyschondrosteosis show the typical Madelung deformity.
755
Acromellc Dysplaslas A number of acromelic and acromesomelic dysplasias, including various types of brachydactyly, are associated with short stature. All patients have disproportionate shortening of the hands and feet. Many have enlargement of the joints associated with limitation of movement and sometimes with pain. Trichorhinophalangeal dysplaSia, an autosomal dominant disorder (McKusick 190350), is characterized by a bulbous nose and hypoplastic nares, short stature, sparse hair, and enlarged interphalangeal joints with broad fingers (Fig. 40-8). Cone-shaped epiphyses, short metacarpals and metatarsals, and small, flat, fragmented capital femoral epiphyses suggestive of Legg-Calve-Perthes disease are characteristic radiologic findings. 17. B7 There are several forms of this disorder; type II, the Langer-Giedion syndrome (McKusick 150230), is also associated with mental retardation and multiple exostoses. The exostoses may cause pain when they occur around joints and are often associated with prominence of the joint. The condition involves a deletion of multiple genes (e.g., EXTl, TRPS1) at 8q24.11-q13.
Metaphyseal Dysplaslas The metaphyseal dysplasias include those associated with adenosine deaminase deficiency138 .139 and the Schwachman-Diamond syndrome (i.e., pancreatic insufficiency and neutropenia).140 They are characterized by generalized changes (e.g., flaring, irregular ossification) in the metaphyses of tubular bone. Children with adenosine deaminase deficiency have deficient Band T lymphocyte function and low levels of immunoglobulins. Metaphyseal dysplasia primarily affects the ribs and long bones. The bony and immunologic abnormalities have been reversed by bone marrow transplantation or enzyme replacement. 141 Children with SchwachmanDiamond syndrome have low birth weights, malabsorption because of deficiency of enzymes produced by the exocrine pancreas, and frequent bacterial infections as a result of neutropenia and defects in neutrophil chemo-
756
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
• figure 40-1 Trichorhinophalangeal dysplasia. A,The right hand of a 12-year-old girl with trichorhinophalangeal dysplasia, shown beside a plaster imprint made of her hand when she was in kindergarten, illustrates the progressive nature of this deformity. B, Radiograph of the hand of a 10-year-old girl with trichorhinophalangeal dysplasia who complained of pain in her finger joints but had no moming stiffness. Epiphyseal abnormalities are visible at all proximal interphalangeal joints.
taxis. There are irregularities in ossification, most commonly noticed in the ribs, femurs, and tibias. Mutations in the SBDS gene have been implicated in SchwachmanDiamond syndrome. 142
MARFAN SYNDROME AND SIMILAR DISORDERS Several disorders characterized by tall stature and contractures can be clinically differentiated,
Epiphyseal Dysplaslas The epiphyseal dysplasias have in common the progressive emergence of abnormalities of the epiphyses of the axial or peripheral skeleton, resulting in joint pain, stiffness, and usually marked degenerative joint disease. Most cases are not evident at birth but become clinically apparent in infancy and childhood. They are classified according to the pattern of epiphyseal involvement. Multiple epiphyseal dysplasia is one of the most common skeletal dysplasias and is inherited as an autosomal dominant trait. Causative mutations have been recognized in six different genes, one of which is the gene encoding matrilin-3, an extracellular matrix protein. 143 The disorder is characterized by short stature and short limbs; it manifests in childhood with pain and stiffness in affected joints, leading to joint contractures and occasionally to scoliosis. 144 ,145 There are progressive irregularities of the end plates of the midthoracic vertebral bodies on radiographs, short metacarpals and terminal phalanges, and flattening, sclerosis, and fragmentation of the epiphyses of the hips, knees, and other joints.
Marfan Syndrome In the Marfan syndrome68 ,146 (McKusick 154700), clinical abnormalities are most prominent in the skeletal, ocular, cardiovascular, and cutaneous systems, In the presence of congenital contractures of digits and elbows, the diagnosis may be made in infancy 147; the syndrome was first described by Marfan in a 5-year-old girl. Joint hypermobility, pain and effusion, and arachnodactyly become increasingly obvious by the second decade of life/>H although many patients are only mildly affected and the diagnosis can be difficult. Children with the Marfan syndrome are tall; their arm span exceeds their height, and the pubis-to-heel measurement is greater than the crown-to-pubis distance. The palate is high and arched, and there may be other skeletal abnormalities such as moderate to severe kyphoscoliosis, pectus carinatum, slipped capital femoral epiphysis, and talipes equinovarus. Muscular hypotonia is common. Skin lesions include striae distensae and elastosis perforans serpiginosa. Ectopia lentis, with upward dislocation of the lens and iridodonesis, and cardiovascular abnormalities occur in about one third of patients.
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
Cardiovascular involvement includes aortic root dilation and aneurysm formation, mitral valve prolapse, and conduction defects. Affected patients often die unexpectedly from cardiac complications, and the value of pharmacologic approaches to the prevention of such developments is uncertain. 148 Criteria for the diagnosis of Marfan syndrome have been proposed. 149 A definite diagnosis requires the presence of an abnormality in at least three of the categories listed in Table 40-11. Collagen turnover is increased, and a cross-linking defect has been proposed. The defective type 1 fibrillin gene has been localized to chromosome 15, and the disease is inherited in an autosomal dominant manner. A number of variants of the Marfan syndrome have been associated with different mutations in the fibrillin gene (FBN1J,l50 An excellent overview of the Marfan syndrome is provided by Pyeritz.I'i1 Objective measurements that are of some use in the diagnosis of the Marfan syndrome include the ratio of the upper segment (US, vertex to pubis) to the lower segment (LS, top of the pubis to floor) and the II
II
I ABLE 4()- 11
Diagnosis of Marfan Syndrome
757
metacarpal index. The US-to-LS ratio is usually increased to greater than 0.85 in children with the Marfan syndrome, but normal age-related changes and complications such as kyphoscoliosis make it difficult to apply, and the overall excessive height is a more important indicator. 152 The metacarpal index, a radiographic measurement of arachnodactyly, is the ratio of the length to the width of the midshaft of metacarpals (2-5). The normal value ranges from 5.4 to 7.9. In children with the Marfan syndrome, the ratio ranges from 8.4 to 10.4. 153
Congenital Contradural Arachnodactyly Congenital contractural arachnodactyly (McKusick 121020) may be confused with the Marfan syndrome. 154 There are congenital contractures of the knees, elbows, and proximal interphalangeal joints, but the hands and the feet are long as in the Marfan syndrome, and the head is elongated. Linear growth is accelerated. Early and progressive kyphoscoliosis may develop. The helix of the ear is abnormal. The defect is associated with a defect in the type 2 fibrillin gene (FBN2) localized to 5q23-q31, and the disorder in inherited as an autosomal dominant trait. ISS The contractures tend to improve as the child grows older.
Abnormalities of Skeletal and Connective nssue
Homocystinuria
Tall stature Long limbs (dolichostenomelia) Long fingers (arachnodactyly) High arched palate Joint laxity Congenital contractures (digits and elbows) Pectus deformity (carinatum or excavatum) Scoliosis Pes planus
Patients with type I homocystinuria resemble those with the Marfan syndrome. They are tall, have long limbs, and may have a high arched palate, arachnodactyly, myopia, peripheral retinal degeneration, and inferior (rather than superior) displacement of the lensY Affected children are light skinned and fair haired. Cutaneous ulcerations and livedo reticularis are common. Hypotonia is present, but the joints are usually stiff rather than hyperextensible. Progressive severe osteoporosis and mental retardation are characteristic. The basic biochemical defect, a deficiency of cystathionine synthetase, is inherited as an autosomal recessive trait. There is an accumulation in tissues of the sulfur-containing amino acids homocystine, homocysteine, serine, and methionine. There may also be a defect in collagen cross-linking. Demonstration of homocystine in the urine clearly differentiates children with homocystinuria from those with the Marfan syndrome. Treatment includes methionine restriction and pyridoxine supplementation. 156 Without therapy, arterial or venous thromboses lead to premature death. Types II and III homocystinuria differ from type I homocystinuria in the nature of the biochemical defect and in the absence of skeletal abnormalities and occlusive arterial disease. 157
OCular AbnormallUes Myopia Upward subluxation of the lens Flat cornea Retinal detachment
cardiovascular Abnonnalltles Aortic root dilatation Mitral valve prolapse Mitral valve regurgitation Aortic valve regurgitation Aortic dissection
Abnormalities of Skin and Integument Striae distensae Inguinal hernia Pneumothorax
Centnl Nervous System Abnonnalltles Dural ectasia Sacral meningocele Dilated cisterna magna
Family History Marfan syndrome in a first-degree relative Data from Pyeritz RE. McKusick VA: The Marfan syndrome: diagnosis and management. N Eng! J Med 300: 772, 1979.
DYSOSTOSIS MULTIPLEX The dysostosis multiplex group of disorders includes the mucopolysaccharidoses, the mucolipidoses, mannosidosis, fucosidosis, gangliosidosis, sialidosis, sialic storage disease, galactosialidosis, and mucosulfatidosis. Deficiency of a lysosomal degradative enzyme leads to an accumulation of its substrate within the lysosomes of
758
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
~. TABLE flO 12 MU((lPoIYSd((h,uidoses Type
Name
Inheritance
MPS
Enzyme Defect
Clinical Features
IH
Hurler
AR
OS, HS
a-L-iduronidase
IS
Scheie"
AR
OS, HS
a-L-iduronidase
II IlIA
Hunter Sanfilippo
XR AR
OS, HS HS
IIID IVA
Morquio
AR
KS
Iduronate sulfatase Heparan-N-sulfatase N-acetyl-a-n-glucosaminidase Acetyl-CoA-glucosaminidase acetyltransferase N-acetyl-glucosamine-6-sulfatase N-acetylgalactosoamine-6-sulfatase
Corneal clouding, dysostosis multiplex, heart disease, severe mental retardation, death in childhood Milder skeletal disease, normal intelligence, normal life span (?) Milder than type I; no corneal clouding Mild skeletal, severe CNS abnormalities
IVB VI
Maroteaux-Lamy
AR
OS
N-acetylgalactosoamine-4-sulfatase
VII
Sly
AR
OS, HS
~-Glucuronidase
IIIB IIIC
Severe skeletal changes; corneal clouding; normal intelligence
~-Galactosidase
Severe skeletal changes, corneal clouding, heart disease, normal intelligence Dysostosis multiplex, variable intelligence, hepatosplenomegaly, white blood cell inclusions
'Formerly classified as Type V. DS, dermatan sulfate; GS, heparan sulfate; KS, keratan sulfate; MPS, mucopolysaccharide found in urine. Modified from Beighton P: McKusick's Heritable Disorders of Connective Tissue, 5th ed. St. Louis, Mosby-Year Book, 1993.
the cell. 45 The phenotype of the specific disease depends on the tissue distribution of the enzyme deficiency. The resulting multisystem degenerative disorders are progressive and unremitting, Skeletal changes include dwarfism, joint contractures, and dysostosis multiplex. Although these disorders are classified as osteochondrodysplasias, they differ from all other members of that group in that they are storage disorders.
In Scheie's syndrome (MPS type I S) CMcKusick 252800), intelligence is normal, and stature is preserved. However, there is progressive stiffening of the joints of the hands, elbows, and knees without swelling or pain. Corneal clouding occurs. All acute phase reactants are normal, but urinary excretion of dermatan sulfate is increased.
Mucollpldoses Mucopolysaccharldoses The mucopolysaccharidoses are genetically determined deficiencies of enzymes involved in the metabolism of glycosaminoglycans (Table 40-12).45 Progressive skeletal dysplasia particularly affects the vertebrae, hips, and hands. 45 ,J58 In the more severe types, such as Hurler's syndrome (i.e., mucopolysaccharidosis [MPS] type I H), dwarfism and marked coarsening of the facial features are present. Deposition of mucopolysaccharide leads to mental retardation and corneal clouding, A claw-hand deformity is often the first clue to the diagnosis. Two of these storage diseases particularly mimic inflammatory arthritis. The comparatively mild dysostosis but severe dwarfing of Morquio's syndrome (MPS type IV) CMcKusick 253000, 252300, 230500) may suggest ]IA. Children with this syndrome, who have normal intelligence, may present with an effusion of a large joint (particularly the knee) or with progressive musculoskeletal stiffness, usually by 3 or 4 years of age. The small joint,> of the hands become enlarged and stiff, a valgus deformity of the knees develops, and the gait becomes stiff and waddling. The joints are not always stiff, however, and some joints (such as the wrists), although enlarged, may be hypermobile. A pectus deformity and barrel chest are usual features. Characteristic radiographic flndings include platyspondyly and odontoid hypoplasia and should help differentiate this disorder from the various forms of spondyloepiphyseal dysplasia. J59 Urinary excretion of keratan sulfate is increased.
The term mucolipidosis (ML) is applied to a group of four disorders that are characterized by the intracellular accumulation of glycosaminoglycans and sphingolipids but without excess urinary glycosaminoglycan excretion. Progressive neurologic and ocular abnormalities occur in all of these autosomal recessive disorders (Table 40-13).44 ML type I (McKusick 256550), an isolated neuraminidase (sialidase) deficiency, causes a Hurler-like syndrome with joint contractures, short trunk and stature, and dysostosis multiplex (i.e., vertebral anomalies, hypoplastic odontoid and ilia, coxa valga, and deformed capital femoral epiphyses). Urinary excretion of sialated
II Type
TABL E 40 13
Mucolipidoses
Name
Enzyme Defect
Sialidase deficiency
Sialidase deficiency
II
I-cell disease
Phosphotransferase deficiency
III
Pseudo-Hurler's polydystrophy
Phosphotransferase deficiency
IV
Sialolipidosis
Uncertain
Musculoskeletal Features Contractures, short stature, dysostosis multiplex Progressive limitation of range of motion Progressive limitation of range of motion; dysostosis multiplex No characteristic skeletal changes
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
urinary oligosaccharides (bound sialic acid) is markedly elevated. I-cell disease (i.e., ML type II) also causes a Hurler-like syndrome with progressive limitation of joint range of motion; the name is derived from the presence of prominent intracytoplasmic CD inclusions in cultured fibroblasts. The biochemical defect is not clearly understood but appears to involve an abnormality in the cellular localization of acid hydrolases. Pseudo-Hurter's polydystrophy is a term applied to ML type III (McKusick 252500). Restriction of joint mobility becomes apparent by 2 years of age, but there is no inflammatory arthritis. Radiologic findings are those of dysostosis multiplex. By 6 years of age, features of Hurler's syndrome dominate the clinical picture. Prognosis for life is good. Inclusions are also found in cultured fibroblasts from some patients with this disease; they probably represent a milder form of ML type II. In ML type IV (McKusick 252600), there are no characteristic skeletal abnormalities.
759
Chondrodysplasia punctata is a peroxisomal disorder. A milder form (McKusick 302960) is associated with deletion of the distal short arm of the X chromosome and manifests as an X-linked recessive disorder. In the X-linked dominant ConradiHunermann disease (McKusick 302960), the limbs are characteristically affected asymmetrically, and contraetures or deformities of the feet may be present. Radiologic abnormalities include stippling of the vertebrae and epiphyses representing disturbed epiphyseal maturation that leads to asymmetric growth. The skin often has patches of ichthyotic change. Only females are affected; it appears to be lethal in males. Genetically determined abnormalities in sterol metabolism may be pathogenically related. 165
Chondrodysplasia punctata has been observed in one infant with neonatal lupus. 166.167 The disorder is also associated with the maternal use of warfarin 168 and maternal vitamin K deficiency.169
Osteopetrosis
A great many disorders of cartilage and bone defy classification. Some appear to have a genetic basis; others have an inflammatory component as well. Those likely to be of interest to the pediatric rheumatologist are briefly summarized in this section.
Osteopetrosis (McKusick 166600, 259700) is a rare disorder that may manifest at birth with frontal bossing, hypertelorism, exophthalmos, nasal obstruction, and cranial nerve palsies. It progresses in severity during infancy and early childhood with repeated fractures, abnormal bleeding, seizures, and early death. The density of all bones is increased, and the metaphyses are flared. Bone marrow transplantation offers hope of effective treatment. 170 Albers-Schonberg disease is a form of osteopetrosis with late onset that is milder in degree than the early-onset form. It is inherited as an autosomal dominant trait. 171
Pseudopseudohypoparathyroidism
Melorheostosis
Pseudopseudohypoparathyroidism, also known as Albright's hereditary osteodysplasia, is characterized by short fourth metacarpals, short stature, and sometimes by mental retardation, a round face, and subcutaneous calcifications. Hypocalcemia and hyperphosphatemia may occur. The condition results from defects in the ~ subunit of the G protein. 160 There is an unusual pattern of inheritance in that children who inherited the gene from the mother are more severely affected than those who inherited the gene from the father (i.e., genomic imprinting).161
Patchy disorders such as melorheostosis (McKusick 155950) and McCune-Albright syndrome (McKusick 174800) appear to result from somatic mutations. Melorheostosis develops after the neonatal period and commonly affects only one limb. Clinically, there may be intermittent swelling and pain around joints, with loss of range of motion and development of contractures at the wrists, elbows, hips, and knees. 172 Skin changes may precede contractures and include tense, red, shiny skin with edema of the subcutaneous tissues. Melorheostosis may occur together with other radiographic abnormalities such as osteopoikilosis (Fig. 40-9).173,174 Melorheostosis of the iliac bone has been described in a boy with linear scleroderma. m Radiographs show cortical hyperostosis in a pattern resembling dripping candle wax, sometimes with endosteal hyperostosis and prominent soft tissue calcification.
OTHER DISORDERS
Pseudoachondroplasia Pseudoachondroplasia, a relatively common form of disproportionate short stature (McKusick 177170), is not present at birth but becomes obvious between the ages of 1 and 2 years. There is mild shortening of the trunk, shortening of limbs with hyperextensible joints, but normal face and skull. The spectrum of short stature is very broad, and it has been suggested that mosaicism plays a role in determining severity.162 Radiographs show platyspondyly and delay in maturation of epiphyses. Because of ligamentous laxity, degenerative arthritis develops, and joint replacement may become necessary. Pseudoachondroplasia is caused by mutations in the cartilage oligomeric matrix protein. 163
Engelmann's Syndrome Engelmann's syndrome, or progressive diaphyseal dysplasia (McKusick 131300), manifests early in childhood with leg pain, abnormalities of gait, muscle weakness, and pain. Radiographs reveal thickening and sclerosis of the cortex of long bones (Fig. 40-10). It is an autosomal dominant disorder, although the gene is not known. Glucocorticoid therapy was reported to be successful in one patient. 176 The abnormalities may resolve spontaneously in adolescence. 177
Chondrodysplasia Punctata The term chondrodysplasia punctata describes a radiologic appearance rather than a specific disease and occurs in a number of conditions. In the autosomal recessive rhizomelic type of chondrodysplasia punctata (McKusick 215100), there are joint contractures, large head, cataracts, and ichthyosis-like skin changes. Most infants with this syndrome die within the first year of life. l64
Osteolyses There are several disorders in which bone dissolves or disappears (McKusick 166300, 259600). The idiopathic osteolyses are grouped according to the area predominantly affected: phalangeal, carpal/tarsal, or multicentric. Familial aero-osteolysis is
760
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
• Figure 40-10 Radiograph illustrates the thickening and sderosis of the diaphyseal cortex of the femurs of a child with diaphyseal dysplasia (i.e., Camurati-Engelmann syndrome). (Courtesy of Dr. R. Cairns.) • Figure 40-9 Radiograph of the hand of a 15-year-old girl with a history of fractures in her toes and diffuse musculoskeletal pain. All bones except the skull were affected by osteopoikilosis, melorheostosis, and fibrous dysplasia.These abnormalities may occur in isolation, with cate-au-Iait spots and sexual precocity (fibrous dysplasia in McCune-Albright syndrome), or as multiple sclerosing bone dysplasia, as in this patient. (Courtesy of Dr. R. Cairns.)
inherited as an autosomal dominant trait and becomes apparent at about 3 years of age (Fig. 40-11).178,179 The carpus or tarsus alone may be affected, or the bones of the hands, feet, elhows, and knees may be involved. The onset can mimic JIA in that affected areas are swollen and warm. Eventually, radiographs show progressive bone lysis and destruction of the involved joints. Spontaneous remissions during the young adult years are characteristic. 1Ho ,lHI The osteolysis is often associated with pain or neuropathic changes and may lead to skin ulcerations that overlie the bony abnormalities. Hereditary distal osteolysis, inherited as an autosomal dominant trait, involves the phalanges and metacarpal or metatarsal bones, and it produces recurrent ulcerations at the affected sites, It usually manifests during late childhood. Spontaneous remission is usual, but unfortunately does not occur before there is loss of digits. IH2
Disorders such as Winchester's syndrome (McKusick 277950), a form of multicentric osteolysis, begin at about 6 weeks of age with restricted joint mobility, swelling, and pain of the proximal interphalangeal joints and enlargement of the wrists. 56 ,1 83,1H4 Later, corneal clouding, coarsening of the face, and joint contractures occur. Osteoporosis, bone erosion, and atlantoaxial subluxation are characteristic radiographic findings. Phantom bone disease (Le., Gorham's disease) occurs between the ages of 5 and 10 years and is not hereditary.18S A carpal/tarsal osteolysis is usual. Carpal/tarsal osteolysis associated with nephropathy has also been described.IHO,IHI
• Figure 40-11 Radiograph of the hand of a 3-year-old child with familial aero-osteolysis shows swelling of the wrist, dissolution of the carpal ossification centers, and generalized osteoporosis of the hand. The radius has overgrown the ulna.This child died later, probably from associated nephropathy.
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
Arthrogryposls Arthrogryposis is the term used to refer to a number of disorders characterized by the presence of multiple congenital contractures. They all appear to result from decreased movement in utero and often produce apparent enlargement of joints, which is actually caused by loss of connective tissue and muscle. 22•23 These disorders tend to be nonprogressive and are not usually associated with pain.
Larsen's Syndrome Larsen's syndrome (McKusick 150250, 145600) is characterized by multiple dislocations of large joints, dysplasias of the spine, and midface hypoplasia. Dislocations cause pain and, if recurrent, may lead to degenerative joint disease. It is inherited as an autosomal dominant condition. 67 Analysis of one family suggests that the responsible gene is located on 3p21.1-pI4.1, which is close to, but distinct from, the collagen type VII (Xl chain gene. l l!6
Menkes' Syndrome The skeletal abnormalities of Menkes' syndrome include osteoporosis, repeated fractures, metaphyseal spurring, and wormian bones in the sutures of the skulL These features reflect an abnormality of copper metabolism, with low serum levels of ceruloplasmin and copper but high tissue levels of copper. 187 The OCCipital horn syndrome, formerly classified as EDS type IX, is also thought to be an abnormality of copper metaboIism. l88 It is characterized by bony occipiral horns, cutaneous hyperextensibility, joint hypermobility, and (in many patients) chronic diarrhea.
761
Cortical Hyperostosis Pachydermoperlostosls Pachydermoperiostosis is a rare autosomal dominant disorder characterized by onset (usually in adolescent boys) of a spadelike enlargement of the hands and feet, sometimes accompanied by pain along the distal long bones. 191- 193 In addition to the cylindrical enlargement of the digits, forearms, and lower legs, there may be minimal joint effusions, coarsening of the facial features, excessive oiliness of the skin, and occasionally gynecomastia, female hair distribution, striae, and acne.
Familial Infantile Cortical Hyperostosis (Caffey's Disease) Caffey's disease is a rare disorder that manifests before 4 months of age with fever, irritability, abnormal acute phase indices, and swelling, tenderness, erythema, or altered contour of the mandible, shoulder girdles, and long bones (Fig. 40-12).194-196 Bony involvement tends to be asymmetric. The calvaria is never affected. The ribs and clavicles are often involved by marked cortical thickening with altered bone shape. The cause is unknown, although the condition appears to be inflammatory and may be triggered by an infection. It usually has a self-limited course of weeks to months, after which it subsides without sequelae. Short-term treatment with glucocorticoids may be considered for the infant with severe disease and marked systemic symptoms. There appears to be a familial but nongenetic basis for this disorder (see also Chapter 28).
Camptodactyly The term camptodactyly refers to the presence of congenital or acquired flexion contractures of the proximal interphalangeal joints, resulting from soft tissue tightening without limitation of flexion. 189 It is most common in the fifth finger but can occur in all digits of the hand except the thumb. The cause is not certain but appears to be related to fibrotic changes in the subcutaneous tissue of the palmar aspect of the joint. Radiographs reveal neither bony nor articular abnormalities. Camptodactyly may occur with diseases such as the Marfan syndrome and has been reported in association with familial arthritis by Malleson and associates. 19O Three children in one family had iridocyclitis, and one boy died suddenly at the age of 4 years. Postmortem examination revealed chronic synovitis and granulomatous arteritis affecting the aorta, coronary arteries, myocardium, and pericardium. There have been other reports of familial camptodactyly, and a syndrome of camptodactyly, arthritis, coxa vara, and pericarditis has been described. The camptodactyly, arthritis, coxa vara, and pericarditis syndrome is inherited as an autosomal recessive disorder caused by a defective gene on chromosome lq25-q31Y It is characterized by congenital camptodactyly and childhood onset of noninflammatory synovial hyperplasia. Some patients have pericarditis; others have coxa vara. r'
• Figure 40-12 Marked hyperostosis of the radius in a 5-month-old boy with Caffey's disease.
762
C HAP T E R
40 PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
Disordered Development of Cartilage Trevor's Disease Patients with Trevor's disease have overgrowth of one of the tarsal or carpal bones or of an epiphysis, often at the knee or ankle. The abnormality is more common in boys than in girls, and it is self-limited. 197
Diaphyseal Aclasis Diaphyseal aclasis is characterized by cartilaginous and bony outgrowths from the metaphyses of long bones, ribs, pelvis, and scapulae. These may interfere with joint function. The gene causing this autosomal dominant condition has been localized to the long arm of chromosome 8 (8q23-q24.l).5
Oilier's Disease Multiple enchondromatosis, or Ollier's disease, becomes evident during childhood, with multiple juxta-articular outgrowths or fractures. Radiographs demonstrate the radiolucent cartilaginous areas in the metaphyseal regions of the tubular and flat bones. Multiple enchondromatosis with hemangiomas is called Maffucci's syndrome (see also Chapter 39).
Fetal Alcohol Syndrome Children with fetal alcohol syndrome have a characteristic facial appearance (i.e., flattening of the midface, short palpebral fissures, and smooth, elongated upper lip) but may also have flexion contractures of the elbows, restricted motion of the metacarpophalangeal joints, camptodactyly, and clinodactyly. Developmental delay, impaired linear growth, and cardiac septal defects are associated problems. 198
REFERENCES 1. Rimoin DL, Connor JM, Pyeritz RE: Emery and Rimoin's Principles and Pra~'tlce of Medical Genetics. New York, Churchill Livingstone, 2002. 2. Kozlowski K, Beighton P: Gamut Index of Skeletal Dysplasias: An Aid to Radiodiagnosis. London, Springer-Verlag, 1995. 3. Taybi H, Lachman RS: Radiology of Syndromes, Metabolic Disorders and Skeletal Dysplasia. St. Louis, Mosby-Year Book, 1996. 4. Gorlin RJ, Cohen MM, Hennekam RCM: Syndromes of the Head and Neck. New York, Oxford University Press, 2001. 5. Grahame R: Heritable disorders of connective tissue. Baillieres Clin Rheum 14: 345-361, 2000. 6. Balint G, Szebenyi B: Hereditary disorders mimicking and/or causing premature osteoarthritis. Baillieres Clin Rheumatol 14: 219--250, 2000. 7. Spranger E]p, for the International Working Group on Constitutional Diseases of Bone: International nomenclature and classification of the osteochondrodysplasias (1997). Am J Med Genet 79: 376, 1998. 8. McKusick V (ed): Mendelian Inheritance in Man: A Catalog of Human Genes and Genetic Disorders. Baltimore, Johns Hopkins University Press, 1998. Online Mendelian Inheritance in Man (OMIM), Available at: www.ncbi.nlm.nih.gov/omim. 9. Chalom EC, Ross J, Athreya BH: Syndromes and arthritis. Rheum Dis Clin North Am 23: 709--727, 1997. 10. Hall JG, Lopez-Rangel E: Bone dysplasias, nontraditional mechanisms of inheritance and monozygotic twins. Pediatr Radiol 27: 422-427, 1997. 11. Hall JG: Genomic imprinting: nature and clinical relevance. Annu Rev Med 48: 35-44, 1997. 12. Bahabri SA, Suwairi WM, Laxer RM, et aI: The camptodactyly-arthropathycoxa vara-pericarditis syndrome: clinical features and genetic mapping to human chromosome I. Arthritis Rheum 41: 730-735, 1998. 13. Rogers JG, McKusick VA: Dominant familial arthritis with scoliosis, In Bergsma 0 (ed): Disorders of Connective Tissue. New York, Stratton Intercontinental Medical, 1975, pp 75-80. 14. Pai GS, Macpherson RI: Idiopathic multicentric osteolysis: report of two new cases and a review of the literature. Am J Med Genet 29: 929--936, 1988. 15. Guidera KG, Satterwhite Y, Ogden JA, et al: Nail patella syndrome: a review of 44 orthopaedic patients, J Pediatr Orthop 11: 737-742, 1991.
16. Stickler GB, Belau PG, Farrell FJ, et al: Hereditary progressive arthro-ophthalmopathy. Mayo Clin Proc 40: 433-455, 1965. 17. Cope R. Beais RK, Bennett RM: The trichorhinophalangeal syndrome: report of eight kindreds, with emphasis on hip complications, late presentations and premature osteoarthritis. J Pediatr Orthop 6: 133-138, 1986. 18. Teebi AS, Rucquoi]K, Meyn MS: Aarskog syndrome: a report of a family with review and discussion of nosology. Am J Med Genet 46: 501-509, 1993. 19. Hecht]T, Butler U: Neurologic morbidity associated with achondroplasia. J Child Neurol 5: 84--97, 1990. 20. Rumball KM, Pang E, Letts RM: Musculoskeletal manifestations of the AntleyBixler syndrome. J Pediatr Orthop B 8: 139--143, 1999. 21. Cohen MM Jr, Kreiborg S: Skeletal abnormalities in the Apert syndrome. Am J Med Genet 47: 624--632, 1993. 22, Gordon N: Arthrogryposis multiplex congenita. Brain Dev 20: 507-511, 1998. 23. Hall JG: Arthrogryposis multiplex congenita: etiology, genetics, classification, diagnostic approach and general aspects. J Pediatr Orthop B 6: 159--166, 1997. 24. Viljoen D: Congenital contractural arachnodactyly (Beals syndrome), J Med Genet 31: 640-643, 1994. 25. O'Brien J: Generalized gangliosidosis. J Pediatr 75: 167-186, 1969. 26. Neidich J, Zackai E, Aronson M, et al: Deletion of 2p: a cytogenetic and ciinical update. AmJ Med Genet 27: 707-710,1987. 27. Nance MA, Berry SA: Cockayne syndrome: review of 140 cases, Am J Med Genet 42: 68--84, 1992. 28. Happle R: X-linked dominant chondrodysplasia punctata: review of the literature and report of a case, Hum Genet 53: 65--73, 1979. 29. Jameson RA, Holt PJ, KeenJH: Farber's disease. Ann Rheum Dis 46: 559--561, 1987. 30. Fujiwaki T, Hamanaka S, Koga M, et al: A case of Farber's disease. Acta Paediatr Jpn 34: 72-79, 1992. 31. Kousseff BG, Newkirk P, Root AW: Brachmann-de Lange syndrome: 1994 update. Arch Pediatr Adolesc Med 148: 749--755, 1994. 32. Ryoppy S, Poussa M, Merikanto J, et al: Foot deformities in diastrophic dysplasia: an analysis of 102 patients. J Bone Joint Surg Br 74: 441-444, 1992. 33, Akita S, Kuratomi H, Abe K, et al: EC syndrome in a girl with paracentric inversion (7)(q22.1;q36.3). Clin Dysmorphol 2: 62-67, 1993. 34. Wallace HJ: Anderson-Fabry disease. Br J Dermatol 88: 1-23, 1973. 35. Perlman JM, Burns OK, Twickler OM, Weinberg AG: Fetal dyskinesia syndrome in the monochorionic pair of a triplet pregnancy secondary to severe disruptive cerebral injury, Pediatrics 96: 521-523, 1995. 36, Verloes A, Hermia JP, Galand A, et al: Glaucoma-Jens-ectopia-microspherophakia-stiffness-shortness syndrome: a dominant disease with manifestations of Weill-Marchesani syndromes. Am J Med Genet 44: 48--51, 1992. 37. Mudd SH, Skovby F, Levy HL, et al: The natural history of homocystinuria due to cystathionine beta synthetase deficiency. AmJ Hum Genet 37: 1-31, 1985. 38. Lemyre E, Azouz EM, Teebi AS, et al: Bone dysplasia series. Achondroplasia. hypochondroplasia and thanatophoric dysplasia: review and update, Can Assoc RadiolJ 50: 185-197, 1999. 39, Gordon SL, Varano LA, Alandete A, Maisels MJ: Jansen's metaphyseal dysostosis. Pediatrics 58: 556-560, 1976. 40. Hilton RC, Wentzel J: Leri pleonosteosis. Q J Med 49: 419-429, 1980. 41. Felman AH, Kirkpatrick JA: Dyschondrosteoses: mesomelic dwarfL~m of Leri and Weill. AmJ Dis Child 120: 329--331, 1970. 42. Williams MS, Josephson KD, Wargowski DS: Marden-Walker syndrome: a case report and a critical review of the literature, Clin Dysmorphol 2: 211-219, 1993. 43. Beck M, Roubicek M, Rogers JG, et al: Heterogeneity of metatropic dysplasia. Eur J Pediatr 140: 231-237, 1983. 44. Gilbert-Barness EF, Barness LA: The mucolipidoses. Perspect Pediatr Pathol 17: 148--184, 1993. 45. Wraith JE: The mucopoiysaccharidoses: a clinical review and guide to management. Arch Dis Child 72: 263-267, 1995, 46. Brunt PW, McKusick VA: Familial dysautonomia: a report of genetic and clinical studies with a review of the literature. Medicine (Baltimore) 49: 343-374, 1970. 47. Patel AC, McAlister WH. Whyte MP: Spondyloepimetaphyseal dysplasia: clinical and radiologic investigation of a large kindred manifesting autosomal dominant inheritance, and a review of the literature. Medicine (Baltimore) 72: 326-342, 1993. 48. Pascuzzi RM: Schwartz-Jampel syndrome. Semin Neurol 11: 267-273, 1991. 49. Harpet RG, Orti E, Baker RK: Bird-headed dwarfs (Seckel's syndrome): a familial pattern of developmental, dental, skeletal, genital and centml nervous system anomalies. J Pediatr 70: 799--804, 1967. 50. Wynne-Davies R, Hall C: The clinical variants of spondyloepiphyseal dysplasia. J Bone Joint Surg Br 64: 435-441, 1982. 51. Nores JM, Dizien 0, Remy JM, Maroteaux P: Two cases of spondylometaphyseal dysplasia: literature review and discussion of the genetic inheritance of the disease. J Rheumatol 20: 170-172, 1993. 52. Elias-Jones AC, Habibi P, Larcher VF, et al: The trisomy (5)(q31qter) syndrome: study of a family with a [(5:14) translocation. Arch Dis Child 63: 427-431, 1988. 53. Caspersson T, Lindsten J, Zech L, et al: Four patients with trisomy 8 identified by the fluorescence and Giemsa-banding techniques. J Med Genet 9: 1-7, 1972.
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
54. Weaver DD, Graham CB, Thomas IT, et al: A new overgrowth syndrome with accelerated skeletal maturation, unusual facies, and camptodaetyly. J Pediatr 84: 547-552, 1974. 55. Halk GM, Terrell WL III, Haik GMJr: The Weill-Marchesani syndrome: report of two cases and a review. J La State Med Soc 142: 25--28, 30--32,1990. 56. Hollister DW, Rimoin DL, Lachman RS, et al: The Winchester syndrome: a nonlysosomal connective tissue disease. J Pediatr 84: 701-709, 1974. 57. Zellweger H, Maertens P, Superneau D, Wertelecki W: History of the cerebrohepatorenal syndrome of Zellweger and other peroxisomal disorders. South Med J 81: 357-364, 1988. 58. Procopis PG, Turner B: Mental retardation, abnormal fmgers and skeletal anomalies: Coffin's syndrome. Am J Dis Child 124: 258--261, 1972. 59. Levy P, Baraitser M: Coffin-Siris syndrome. J Med Genet 28: 338--341, 1991. 60. Cohen MM Jr, Hall BD, Smith DW, et al: A new syndrome with hypotonia, obesity, mental deficiency and facial, oral, ocular, and limb anomalies. J Pediatr 83: 28Q--284, 1973. 61. Teebi AS, Shaabani IS: Further delineation of Costello syndrome. Am J Med Genet 47: 166--168, 1993. 62. Reisner SH, Seelenfreund M, Ben-Bassat M: Cutis laxa associated with severe intrauterine growth retardation and congenital dislocation of the hip. Acta Pediatr Scand 60: 357-360, 1971. 63. Hansen KE, Kirkpatrick SJ, Laxova R: Dubowitz syndrome: long-term followup of an original patient. Am J Med Genet 55: 161-164, 1995. 64. Pope FM: Ehlers-Danlos syndrome. Baillieres Clin Rheumatol 5: 321-349, 1991. 65. Crifasi PA, Patterson MC, Bonde D, Michels VV: Severe HajdU-Cheney syndrome with upper airway obstruction. Am J Med Genet 70: 261-266, 1997. 66. Ikegawa S, Sakaguchi R, Kimizuka M, et al: Recurrent dislocation of the patella in Kabuki make-up syndrome. J Pediatr Orthop 13: 265-267, 1993. 67. Larsen LJ, Schottstaedt ER, Bost FC: Multiple congenital dislocations associated with characteristic facial abnormalities. J Pediatr 37: 574-581, 1950. 68. Gray JR, Davies SJ: Marfan syndrome. J Med Genet 33: 403--408, 1996. 69. Makitie 0, Kaitila I: Cartilage-hair hypoplasia-clinical manifestations in 108 Finnish patients. Eur J Pediatr 152: 211-217, 1993. 70. Mikles M, Stanton RP: A review of Morquio syndrome. Am J Orthop 26: 533-540, 1997. 71. Teebi AAS, Qumsiyeh MB, Meyers-Seifer CH, et al: Velo-facia-skeletal syndrome in a mother and daughter. Am J Med Genet 58: 8--12, 1995. 72. Bassett GS, Scott CI Jr: The osteochondrodysplasias. In Morrisey RT (ed): Pediatric Orthopaedics, 3rd ed. Philadelphia, JB Lippincott, 1990, p 91. 73. Seiko Y, Morawakw T, Tanaka H et al: Molecular defects in achondroplasia and the effects of growth hormone treatment. Acta Pediatr 88 (Suppl 488): 118--120, 1999. 74. American Academy of Pediatrics Committee on Genetics: Health supervision for children with achondroplasia. Pediatrics 95: 443--451, 1995. 75. Eyre DR: The collagens of articular cartilage. Semin Arthritis Rheum 21: 2-11, 1991. 76. Beighton P: The Ehlers-Danlos Syndromes. London, William Heinemann Medical, 1970. 77. Sillence DO, Senn A, Danks DM: Genetic heterogeneity in osteogenesis imperfeeta. J Med Genet 16: 101-116, 1979. 78. Rauch F, Glorieux FH: Osteogenesis imperfecta. Lancet 363: 1377-1385,2004. 79. Castells S, Yasumura S, Fusi MA, et al: Plasma osteocalcin levels in patients with osteogenesis imperfecta. J Pediatr 109: 88--91, 1986. 80. castells S, Colbert C, Chakrabarti C, et aI: Therapy of osteogenesis imperfecta with synthetic salmon calcitonin. J Pediatr 95: 807--811, 1979. 81. Nishi Y, Hamamoto K, Kajiyama M, et al: Effect of long-term calcitonin therapy by injection and nasal spray on the incidence of fractures in osteogenesis imperfecta. J Pediatr 1121: 477-480, 1992. 82. Glorieux FH, Bishop NJ, Plotkin H, et al: Cyclic administration of pamidronate in children with severe osteogenesis imperfeeta. N Engl J Med 339: 947-952, 1998. 83. Engelbert RH, Pruijs HE, Beemer FA, Helders PJ: Osteogenesis imperfecta in childhood: treatment strategies. Arch Phys Med Rehabil 79: 159Q--1594, 1998. 84. Kocher MS, Shapiro F: Osteogenesis imperfecta. J Am Acad Orthop Surg 6: 225--236, 1998. 85. Brenton DP, Dent CF: Idiopathic juvenile osteoporosis. In Bickel H, Stern J (eds): Inborn Errors of Calcium and Bone Metabolism. Baltimore, University Park Press, 1976, p 222. 86. Smith R: Idiopathic juvenile osteoporosis: experience of twenty-one patients. Br J Rheumatol 34: 68--77, 1995. 87. Chlebna-Sok61 D, Loba-Jakubowska E, Sikora E: Clinical evaluation of patients with idiopathic juvenile osteoprosis. J Pediatr Orthop 10: 259-263, 2001. 88. Dent CE, Friedman M: Idiopathic juvenile osteoporosis. Q J Med 34: 177-210, 1965. 89. Halila R, Steinmann B, PeItonen L: Processing types I and III procollagen in Ehlers-Danlos syndrome type vrr. Am J Hum Genet 39: 222-231, 1986. 90. Fujimoto A, Wilcox WR, Cohn DH: Clinical, morphological, and biochemical phenotype of a new case of Ehlers-Danlos syndrome type VIIC. Am J Med Genet 68: 25--28, 1997. 91. Lewkonia RM: The arthropathy of hereditary arthroophthalmopathy (Stickler syndrome). j Rheumatol 19: 1271-1275, 1992. 92. Ziakas NG, Ramsay AS, Lynch SA, Clarke MP: Stickler's syndrome associated with congenital glaucoma. Ophthalmic Genet 19: 55-58, 1998.
763
93. Letts M, Kabir A, Davidson D: The spinal manifestations of Stickler's syndrome. Spine 24: 1260--1264, 1999. 94. Ahmad NN, A1a-Kokko L, Knowlton RG, et aI: A stop codon in the procolIagen II gene (COL2A!) in a family with the Stickler syndrome (arthra-ophthalmopathy). Proc Nat! Acad Sci USA 88: 6624-6627, 1991. 95. Sconyers SM, Rimoin DL, Lachman RS, et aI: A distinct chondrodysplasia resembling Kniest dysplasia: clinical, roentgenographic. histologic and ultrastructural findings. J Pediatr 12: 898--904, 1985. 96. Weis MA, Wilkin DJ, Kim HJ, et al: Structurally abnormal type II collagen in a severe form of Kniest dysplasia caused by an exon 24 skipping mutation. J BioI Chern 273: 4761-4768, 1998. 97. Spranger]W, Langer La: Spondyloepiphyseal dysplasia congenita. Radiology 94: 313-322, 1979. 98. Tiller GE, Weis MA, Polumbo PA, et al: An RNA-splicing mutation (G+51VS20) in the type II collagen gene (COL2A!) in a family with spondyloepiphyseal dysplasia congenita. Am J Hum Genet 56: 388--395, 1995. 99. Iceton JA, Home G: Spondyloepiphyseal dysplasia tarda: the X-linked variety in three brothers. J Bone Joint Surg Br 68: 61~19, 1986. 100. Whyte MP, Gottesman GS, Eddy MC, McAlister WH: X-linked recessive spondyloepiphyseal dysplasia tarda: clinical and radiographic evolution in a 6-generation kindred and review of the literature. Medicine (Baltimore) 78: 9-25, 1999. 101. Kozlowski K, Lewis IC, Kennedy J, et al: Progressive pseudorheumatoid arthritis. Paediatr Indones 25: 237-244, 1985. 102. Wynne-Davis R, Hall C, Ansell BM: Spondyla-epiphyseal dysplasia tarda with progressive arthropathy. J Bone Joint Surg Br 64: 442-445, 1982. 103. Bradley JD: Pseudoseptic pseudogout in progressive pseudorheumatoid arthritis of childhood. Ann Rheum Dis 46: 709-712, 1987. 104. Tiller GE, Cassidy SB, Wensel C, Wenstrup RJ: Aortic root dilatation in EhlersDanlos syndrome types 1, II and III: a report of five cases. Clin Genet 53: 460--465, 1998. 105. Reichert S, Riemann D, Plaschka B, Machulla HK: Early-onset periodontitis in a patient with Ehlers-Danlos syndrome type III. Quintessence Int 30: 785--790, 1999. 106. Ploeckinger B, Ulm MR, Chalubinski K: Ehlers-Danlos syndrome type II in pregnancy. Am J Perinatol 14: 99-101, 1997. 107. Narcisi P, Richards AJ, Ferguson SD, Pope FM: A family with Ehlers-Danlos syndrome type III/articular hypermobility syndrome has a glycine 637 to serine substitution in type III collagen. Hum Mol Genet 3: 1617-1620, 1994. 108. Pyeritz RE, Stolle A, Parfrey NA, Myers JC: Ehlers-Danlos syndrome is due to a novel defect in type III procollagen. Am J Med Genet 19: 607--622, 1984. 109. Palmeri S, Mari F, Meloni I, et al: Neurological presentation of Ehlers-Danlos syndrome type IV in a family with parental mosaicism. Clin Genet 63: 510--515, 2003. 110. Pepin M, Schwarze U, Superti-Furga A, Byers PH: Clinical and genetic features of Ehlers-Danlos syndrome type IV, the vascular type. N Engl J Med 342: 673-680, 2000. 111. Hartsfield ]K, Kousseff BG: Phenotypic overlap of EWers-Danlos types IV and VIII. AmJ Med Genet 37: 465-470,1990. 112. Pinnell SR, Krane SM, Kenzora JLE, Glimcher MJ: A heritable disorder of connective tissue-hydroxylysine deficient collagen. N Engl J Med 286: 1013-1020, 1972. 113. Brunk I, Stover B, Ikonomidou C, et al: Ehlers-Danlos syndrome type VI with cystic malformations of the meninges in a 7-year-old girl. Eur J Pediatr 163: 214-217, 2004. 114. Wenstrup J, Murad S, Pinnell SR: Ehlers-Danlos syndrome type VI: clinical manifestations of collagen type IV hydroxylase deficiency. j Pediatr 115: 405-409, 1989. 115. Rauma 1', Kumpumaki S, Anderson R, et al: Adenoviral gene transfer restores lysyl hydroxylase activity in type VI Ehlers-Danlos syndrome. J Invest Dermatol 116: 602--605, 2001. 116. DePaepe A, Nuytinck L, Hausser I, et al: Mutations in the COL5Al gene are causal in the Ehlers-Danlos syndromes 1 and II. Am J Hum Genet 60: 547-554, 1997. 117. Beighton P, Price A, Lord J, Diskson E: Variants of the Ehlers-Danlos syndrome: clinical, biochemical, haematological features of 100 patients. Ann Rheum Dis 28: 228-245, 1969. 118. Hatamochi A, Arakawa M, Mori K, et al: Increased expression of type VI collagen genes in cutis laxa fibroblasts. J Dermatol Sci 11: 97-103. 1996. 119. Susic S, McGrory J, Ahier j, Cole WG: Multiple epiphyseal dysplasia and pseudoachondroplasia due to novel mutations in the calmodulin-like repeats of cartilage oligomeric matrix protein. Clin Genet 51: 219-224, 1997. 120. Spranger J: The type Xl collagenopathies. Pediatr Radiol 28: 745--750, 1998. 121. Siegel RC, Black CM, Bailey AJ; Cross-linking of collagen in the X-linked Ehlers-Danlos type V syndrome. Biochem Biophys Res Commun 88: 281-287, 1979. 122. Bruno PA, Napolitano V, Votino F, et al: Pregnancy and delivery in EhlersDanlos syndrome type V. Clin Exp Obstet Gynecol 24: 152-153, 1997. 123. Arneson MA, Hammerschmidt DE, Furcht 1', King RA: A new form of EhlersDanlos syndrome. JAMA 244: 144-147, 1980. 124. Anstey A, Mayne K, Winter M, et al: Platelet and coagulation studies in EWers-Danlos syndrome. Br J Dermatol 125: 155--163, 1991. 125. Hastbacka J, de la Chapell A, Mahtani MM, et al: The diastrophic gene encodes a novel sulfate transporter: positional cloning by fine-structure linkage disequilibrium mapping. Cell 78: 1073-1087, 1994.
764
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
126. El Ghouzzi V, Dagoneau N, Kinning E, et al: Mutations in a novel gene Dymeclin (FLJ2007l) are responsible for Dyggve-Melchior-Clausen syndrome. Hum Mol Genet 12: 357-364, 2003. 127. Beighton P: Dyggve-Melchior-Clausen syndrome. J Med Genet 27: 512-515, 1990. 128. Thauvin-Robinet C, EI Ghouzzi V, Chemaitilly W, et al: Homozygosity mapping of a Dyggve-Melchior-Clausen syndrome gene to chromosome 18q21.1. J Med Genet 39: 714-717, 2002. 129. Ehtesham N, Cantor RM, King LM, et al: Evidence that Smith-McCort dysplasia and Dyggve-Melchior-Clausen dysplasia are allelic disorders that result from mutation in a gene on chromosome 18q12. Am J Hum Genet 71: 947-951, 2002. 130. eI Shanti HE, Omari HZ, Qubain HI: Progressive pseudorheumatoid dysplasia: report of a family and review. ./ Med Genet 34: 559-563, 1997. 131. Fischer ./, Urtizberea ./A, Pavek S, et al: Genetic linkage of progressive pseudorheumatoid dysplasia to a 3-cM interval of chromosome 6q22. Hum Genet 103: 60-64, 1998. 132. Stoss H, Pesch H./, Pontz B, et al: Wolcott-RaJlison syndrome: diabetes mellitus and spondyloepiphyseal dysplasia. Eur./ Pediatr 138: 120--129, 1982. 133. Senee V, Vattem KM, Delepine M, et al: Wolcott-Rallison syndrome: clinical, genetic, and functional study of EIF2AK3 mutations and suggestion of genetic heterogeneity. Diabetes 53: 1876--1883, 2004. 134. Horton WA, Langer LO, Collins DL, Dwyer C: Brachyolmia, recessive type lHobaek): a clinical, radiographic, and histochemical study. Am./ Med Genet 16: 201-211, 1983. 135. Belin V, Cusin V, Viot G, et al: SHOX mutations in dyschondrosteosis (LeriWeill syndrome). Nat Genet 19: 67-69, 1998. 136. Laborde H, Rodrigue S, Catoggio PM: Mycobacterium/anu/tum in systemic lupus erythematosus. Clin Exp Rheumatol 7: 291-293, 1989. 137. Carrington PR, Chen H, Altick ./A: Trichorhinophalangeal syndrome, type I. ./ Am Acad Dermatol 31: 331-336, 1994. 138. Cederbaum SC, Kaitila 1, Rimoin DL, Stiehm ER: The chondra-osseous dysplasia of adenosine deaminase deficiency with severe combined immunodeficiency. ./ Pediatr 89: 737-742, 1976. 139. MacDermot KD, Winter RM, Wigglesworth ./S, Strobel S: Short stature/short limb skeletal dysplasia with severe combined immunodeficiency and bowing of the femom: report of two patients and review../ Med Genet 28: 10--17, 1991. 140. McLennan TW, Steinbach HL: Schwachman's syndrome: the broad spectrum of hony abnormalities. Radiology 112: 167-173, 1974. 141. Yulish BS, Stern RC, Polmar SH: Partial resolution of bone lesions: a child with severe combined immunodeficiency disease and adenosine deaminase deficiency after enzyme-replacement therapy. Am ./ Dis Child 134: 61-63, 1980. 142. Makitie 0, Ellis L, Durie PR, et al: Skeletal phenotype in patients with Shwachman-Diamond syndrome and mutations in SBDS. Clin Genet: 65: 101-112, 2004. 143. Makitie 0, Mortier GR, Czarny-Ratajczak M, et al: Clinical and radiographic findings in multiple epiphyseal dysplasia caused by MATN3 mutarions: description of 12 patients. Am./ Med Genet 125A: 278--284, 2004. 144. Patrone NA, Kredich DW: Arthritis in children with multiple epiphyseal dysplasia. ./ Rheumatol 12: 145-149, 1985. 145. Shapiro F: Epiphyseal disorders. N Engl.J Med 317: 1702-1710, 1987. 146. Marfan AB: Un cas de deformation congenitale des quatre membres, plus prononcee aux extremites charaterisee par I'allongement des os, avec un certain degre d'amancissement. Bull Mem Soc Med Hop (Paris) 13: 220, 1896. 147. Morse RPP, Rockenmacher S, Pyeritz RE, et al: Diagnosis and management of infantile Marfan syndrome. Pediatrics 86: 888--895, 1990. 148. Reed CM, Fox ME, Alpert BS: Aortic biomechanical properties in pediarric patients with Marfan syndrome, and the effects of atenolo!. Am./ Cardiol 71: 606-608, 1993. 149. Pyeritz RE, McKusick VA: The Marfan syndrome: diagnosis and management. N Engl./ Med 300: 772-777, 1979. 150. Montgomery RA, Geraghty MT, Bull E, et al: Multiple molecular mechanisms underlying subdiagnostic variants of Marfan syndrome. Am./ Hum Genet 63: 1703-1711, 1998. 151. Pyeritz RE: The Marfan syndrome. Am Fam Physician 34: 83-94, 1986. 152. Pyerirz RE, Murphy EA, Lin S./, Rosell EM: Growth and anthropometrics in the Marfan syndrome. Prog Clin Bioi Res 200: 355-366, 1985. 153. Sinclair RTG, Kitchin AH, Turner RWD: The Marfan syndrome. Q./ Med 53: 19, 1960. 154. Mirise RT, Shear S: Congenital contractural amchnodactyly: description of a new kindred. Arthritis Rheum 22: 542-546, 1979. ISS. Park ES, Putnam EA, Chitayat D, et al: Clustering of FBN2 mutations in patients with congenital contractural arachnodactyly indicates an important role of the domains encoded by exons 24 through 34 during human development. Am./ Med Genet 78: 350--355, 1998. 156. Boers GH./, Smals AGH, Trijbels F,/M, et al: Heterogeneity for homocystinuria in premature peripheral and cerebml occlusive arterial disease. N Engl./ Med 313: 709-715, 1985. 157. Mudd SH, Skovby F, Levy HL, et al: The natural hestory of homocystinuria due to cystathionine beta synthase deficiency. Am./ Hum Genet 37: 1-31, 1985. 158. Fisher RC, Horner RI, Wood VE: The hand in mucopolysaccharide disorders. Clin Orthop 104: 191-199, 1974.
159. Mikles M, Stanton RP: A review of Morquio syndrome. Am ./ Orthop 26: 533-540, 1997. 160. Ringel MD, Schwindinger WF, Levine MA: Clinical implications of genetic defects in G proteins: the molecular basis of McCune-Albright syndrome and Albright hereditary osteodystrophy. Medicine (Baltimore) 75: 171, 1996. 161. Davies S./, Hughes HE: Imprinting in Albright's hereditary osteodystrophy. ./ Med Genet 30: 101-103, 1993. 162. Ferguson HL, Deere M, Evans R, et al: Mosaicism in pseudoachondroplasia. Am./ Med Gent 70: 287-291,1997. 163. Deere M, Sanford T, Ferguson HL: Identification of twelve mutations in cartilage oligomeric matrix protein (COMP) in patients with pseudoachondroplasia. Am./ Med Genet 80: 510--513, 1998. 164. Spranger./, Opitz ./M, Bidder U: Heterogeneity of chondrodysplasia punctata. Humangenetik 11: 190--212, 1971. 165. Kelley RI, Wilcox WG, Smith M, et al: Abnormal sterol metabolism in patients with Conradi-Hunermann-Happle syndrome and spomdic lethal chondrodysplasia punctata. Am./ Med Genet 83: 213-219, 1999. 166. Kelly TE, Alford BA, Greer KM: Chondrodysplasia punctata stemming from maternal lupus erythematosus. Am./ Med Genet 83: 397-401, 1999. 167. Austin-Ward E, Castillo S, Cuchacovich M, et al: Neonatal lupus syndrome: a case with chondrodysplasia punctata and other unusual manifestations. ./ Med Genet 35: 695-698, 1998. 168. Savarirayan R: Common phenotype and etiology in warfarin embryopathy and X-Linked chondrodysplasia punctata. Pediatr Radiol 29: 322, 1999. 169. Menger H, Lin AE, Toriello HV, et al: Vitamin K deficiency embryopathy: a phenocopy of the warfarin embryopathy due to a disorder of embryonic vitamin K metabolism. Am./ Med Genet 72: 129-134, 1997. 170. Eapen M, Davies SM, Ramsay NK, Orchard P./: Hematopoietic stem cell tmnsplantation for infantile osteopetrosis. Bone Marrow Transplant 22: 941-946. 1998. 171. ./ohnston CC, Lavy N, Lord T, et al: Osteopetrosis: a clinical, genetic, metabolic, and morphologic study of the dominantly inherited benign form. Medicine (Baltimore) 47: 149-167, 1968. 172. Beauvais P, Faure C, Montagne JP, et al: Len's melorheostosis: three pediatric cases and a review of the literature. Pediatr Radiol 6: 153-159, 1977. 173. Nevin NC, Thomas PS, Davis RI, Cowie GH: Melorheostosis in a family with autosomal dominant osteopoikilosis. Am./ Med Genet 82: 409-414, 1999. 174. Foeldvari 1, Cairns RA, Petty RE, Cabral DA: An unusual case of mixed sclerosing bone dystrophy presenting with morning stiffness and joint swelling in childhood: a case report. Clin Exp Rheumatol 13: 525-528, 1995. 175. Moreno Alvarez M./, Lazaro MA, Espada G. et al: Linear scleroderma and melorheostosis: case presentation and literature review. Clin Rheumatol 15: 389-393, 1996. 176. Bourantas K, Tsiara S, Drosos AA: Successful treatment with corticosteroid in a patient with progressive diaphyseal dysplasia. Clin Exp Rheumatol 14: 485-486, 1995. 177. Fallon MD, Whyte NP, Murphy WA: Progressive diaphyseal dysplasia (Engelmann's disease). ./ Bone Joint Surg Am 62: 465-472, 1980. 178. Gluck ./, Miller .J.J II1: Familial osteolysis of the carpal and tarsal bones. ./ Pediatr 81: 506--510, 1972. 179. Erickson CM Hirschberger M, Stickler GB: Carpai-tarsal osteolysis. ./ Pediatr 93: 779-782, 1978. 180. Brown DN, Bradford DS, Godin R.I, et al: The acroosteolysis syndrome: morphologic and biochemical studies. ./ Pediatr 88: 573-580, 1976. 181. Beals RK, Bird CB: Carpal and tarsal osteolysis: a case report and review of the literature. J Bone ./oint Surg Am 57: 681-686, 1975. 182. Elias AN, Pinals RS, Anderson He, et al: Hereditary osteodysplasia with acroosteolysis (the HajdU-Cheney syndrome). Am./ Med 65: 627--{)36, 1978. 183. Winchester PH. Grossman H, Lim WN, Danes BS: A new acid mucopolysaccharidosis with skeletal deformities simulating rheumatoid arthritis. A./R Am ./ Roentgenol 106: 121-128, 1969. 184. Costa MM, Santos H, Santos MJ, et al: Idiopathic multicentric osteolysis: a rare disease mimicking juvenile chronic arthritis. Clin Rheumatol 15: 97-98, 1996. 185. Gorham LW, Stout AP: Massive osteolysis (acute spontaneous absorption of bone, phantom bone, disappearing hone): its relationship to hemangiomatosis. ./ Bone ./oint Surg Am 37: 985-1004, 1955. 186. Vujic M, Hallstensson K, Wahlstrom./, et al: Localization of a gene for autosomal dominant Larsen syndrome to chromosome region 3p21.1-14.1 in the vicinity of, but distinct from, the COL7Al locus. Am ./ Hum Genet 57: 1104-1113, 1995. 187. Scriver CR, Beaudet AL, Sly WS, Valle D (eds): The Metabolic Basis of Inherited Disease, 6th ed. New York, McGraw-Hill, 1989. 188. Kuivaniemi H, Peltonen I, Kivirikko K1: Type IX Ehlers-Danios syndrome and Menkes syndrome: the decrease in Iysyl oxidase activity is associated with a corresponding deficiency in the enzyme protein. Am./ Hum Genet 37: 798--808, 1985. 189. Welch JP, Temtamy SA: Hereditary contractures of the fingers (camptodactyly). J Med Genet 3: 104-113, 1%6. 190. Malleson P, Schaller JG, Dega F, et al: Familial arthritis and camptodactyly. Arthritis Rheum 24: 1199-1204, 1981. 191. Vogl A, Goldfisher S: Pachydermoperiostosis: primary or idiopathic hypertrophic osteoarthropathy. Am./ Med 33: 166--187, 1962.
C HAP T E R
40
PRIMARY DISORDERS OF BONE AND CONNECTIVE TISSUES
192. Rimoin DL: Pachydennoperiostosis (idiopathic clubbing and periostosis): genetics and physiologic considerations. N Engl] Med 272: 923-931, 1965. 193. Calabro ]E, Marchesano JM, Abruzzo ]L: Idiopathic hypertrophic osteoarthropathy Cpachydermoperiostosis): onset before puberty. Arthritis Rheum 9: 496, 1966. 194. Caffey], Silverman WA: Infantile cortical hyperostosis: preliminary report of a new syndrome. A]R Am] Roentgenol 54: I, 1945. 195. Caffey J: Infantile cortical hyperostosis. ] Pediatr 29: 541, 1946.
765
196. Wilson AK: Infantile cortical hyperostosis: review of the literature and report of a case without mandibular involvement. Clin Orthop 62: 209-217, 1969. 197. Azouz EM, Siomic AM, Marton D, et al: The variable manifestations of epiphysealis hemimelica. Pediatr Radiol 15: 44-49, 1985. 198. Smith DF, Sandor GG, MacLeod PM, et al: Intrinsic defects in the fetal alcohol syndrome: studies on 76 cases from British Columbia and Yukon Territory. Neurobehav ToxicoI Teratol 3: 145-152, 1981.
I
A A77-1726, 98 Aarskog's syndrome, 747t Absolute risk, 146t Abuse, 688-689 Academic research organizations, 155 Acanthosis nigricans, 741 Accelerated nodulosis, 95, 234 Acetylsalicylic acid administration of, 87 contraindications, 88 description of, 77-78 dosing of, 811 drug interactions, 88 glucocorticoids' effect on, 88 hypersensitivity to, 86 mechanism of action, 80, 87 pharmacology of, 87 stnJcture of, 77f toxicities associated with, 83t Achondroplasia, 746 Acne, 579 Acrocyanosis, 449 Acrodermatitis chronic atrophicans, 591, 593--594 Acromelic dysplasias, 755 Acromelic shortening, 744 Acro-osteolysis, 457 Actin, 16f Actin filaments, 15 Activation-induced cell death, 44, 45f, 46 Activities of daily living, 196 Actomyosin, 15 Actuarial method, 169 Acute adrenal insufficiency, 110-111 Acute chondrolysis of hip, 688 Acute interstitial nephritis with nephrotic syndrome, 85 Acute lupus pneumonitis, 355 Acute pain, 703 Acute rheumatic fever age at onset, 614 alleles with, 618t antistreptolysin 0 test, 616, 622 aortic insufficiency in, 619 arthritis in, 618, 623 carditis in, 618--619, 621-622 classification of, 614, 621-622 clinical manifestations of, 618--620, 619f course of, 624 definition of, 614 description of, 51 diagnosis of, 621-622 differential diagnosis, 622 endocarditis and, 619, 621 epidemiology of, 614-615 erythema marginatum associated with, 620, 620f etiology of, 615-616 gender ratio for, 614 genetic background of, 617-618 geographic distribution of, 614-615
group A streptococcus, 615-616 HLA antigens, 618 incidence of, 614 mitral regurgitation in, 619 morbidity in, 624 myocarditis in, 619 pathogenesis of, 615f pathologic findings in, 620-621 prevalence of, 614 prognosis for, 624 racial distribution of, 614-615 rheumatic heart disease prophylaxis, 623-624 skin manifestations of, 620, 620f streptococcal antibody tests for, 616-617 differential diagnosis, 622 pyrogenic exotoxins, 616 treatment of, 622, 623t subcutaneous nodules in, 620-621 Sydenham's chorea associated with, 620-623 tissue injury mechanisms, 617 treatment of, 622-623 Adalimumab, 123 Adaptive immune response, 28 Adaptive immunity characteristics of, 26, 28 definition of, 19 disorders of description of, 643t rheumatic disease associated with, 646-653 interleukin-6's role in, 57 Adaptive randomization, 153 Adenosine deaminase deficiency, 647t Adherence, 186, 186t Adolescent transition to adult care, 187-188 Adult-onset Still's disease, 297-298 Adverse drug effect, 150 Adverse event, 150 Affinity maturation, 30 Aggrecan, 11 Aldolase, 426, 477-478 A1endronate, 724f Alkaline phosphatase, 717, 717t Alkylating agents chlorambucil, 117, 117f cyclophosphamide. See Cyclophosphamide fertility effects of, 116 Allergic granulomatosis, 494t, 547f A1lodynia, 698, 706, 707t Allogeneic bone marrow transplantation with high-dose immunotherapy, 126 A1lotypes, 30 Alopecia, 352 Alphaviruses, 582-583 Altered peptide ligands, 37 Alternative hypothesis, 144 American College of Rheumatology description of, 2 founding of, 4
Page numbers followed by f refer to figures; page numbers followed by I refer to tables.
N D E
x
juvenile rheumatoid arthritis criteria, 206, 207t liver toxicity monitoring gUidelines of, 92 systemic lupus erythematosus criteria, 342, 343t American Juvenile Arthritis Organization, 7 t-Aminobutyric acid, 700 5-Aminoimidazole-4-carboxamide ribonucleotide, 88-89, 97 Amphiarthroses, 9 Amyloidosis juvenile psoriatic arthritis and, 331 juvenile rheumatoid arthritis and, 244 systemic arthritis and, 296-297 Amyopathic dermatomyositis, 413 Anakinra, 124, 238 Analysis of covariance definition of, 168 multivariate, 170 Analysis of variance multivariate, 170 multi-way, 167 nonparametric, 167 one-way, 167 Anaphylactoid purpura, 496 Anchor residues, 33 ANCOVA. See Analysis of covariance Androgenic hormone deficiency, 348 Anemia sickle cell, 635 in systemic lupus erythematosus, 367-368, 368f Anergy, 31, 119 Aneurysmal bone cyst, 735 Angiogenesis definition of, 57 juvenile psoriatic arthritis pathogenesis and, 326 Angiokeratoma corporis diffusum universale, 639 Ankle, 229 Ankle exercises, 194f Ankylosing spondylitis frequency of, 6t genetics of, 308-309, 309t New York diagnostic criteria for, 305t, 306 ANOVA. See Analysis of variance Anterior knee pain, 678 Anti-annexin XI, 425 Antibiotics description of, 118 Lyme disease treated with, 598t, 598-599 septic arthritis treated with, 573, 573t Antibodies diversity of, 30-31 to extractable nuclear antigens, 370-371 heavy chains, 28 light chains, 28 myositis-associated, 424-425 mYOSitis-specific, 424-425 structure of, 28 Antibody-dependent cellular cytotoxicity, 23,42 Anticardiolipin antibodies, 357-358, 368t
767
768
INDEX
Anti-CD40 ligand, 120--121 Anti-dsDNA antibodies, 369t, 369-370 Anti-a-fodrin antibodies, 394 Antigen(s) definition of, 28 human leukocyte. See Human leukocyte antigens oral administration of, 47-48 TO,39 TI,39 "Antigen" arrays, 73 Antigenic determinant, 30 Antigenic mimicry, 617 Antigen-presenting cells activation of, 48 B cells. See B cells dendritic cells, 36 description of, 28, 32 receptors, 36 T-cell interactions with, 34 T-cell recognition of, 41 types of, 36 Antiglobulins, 371 Antihistone antibodies, 371, 371t Anti-IL6 receptor antibody, 124-125 Anti-laminin autoantibodies, 394 Anti-La/SS-B antibodies in neonatal lupus erythematosus, 392-393 in systemic lupus erythematosus, 370, 370t Antimalarials description of, 96-97 juvenile rheumatoid arthritis treated with, 239 Antineutrophil cytoplasmic antibodies description of, 337 in Kawasaki disease, 522, 530 in polyarteritis nodosa, 515 small-vessel vasculitis, 504 in Wegener's granulomatosis, 541, 542f, 542t Antinuclear antibodies in diffuse cutaneous systemic scleroderma, 456 juvenile rheumatoid arthritis findings, 224-225 in morphea, 477 in polyarthritis, 268 in systemic lupus erythematosus, 369-371, 383 tests for, 224-225 in uveitis, 281-282, 328 Antiphospholipid antibodies, 368-369 Antiphospholipid syndrome catastrophic, 357-358 characteristics of, 357 description of, 346, 506 diagnostic criteria for, 357t management of, 380 systemic lupus erythematosus and, 357-358 Anti-Ro/SS-A antibodies in neonatal lupus erythematosus, 392 in systemic lupus erythematosus, 370, 370t Anti-Sm antibodies, 370--371, 371t Antistreptolysin 0 test, 616, 622 Anti-synthetase antibodies, 425 Anti-synthetase syndrome, 425 Anti-Ul RNP antibodies, 371, 371t Anti-UlRNP antibodies, 425 Antley-Bixler syndrome, 747t Aortic insufficienc.y, 619 Apert's syndrome, 747t Aphthous stomatitis description of, 352, 564 periodic fever with, 666-667 Apophyseal joints description of, 217 juvenile ankylosing spondylitis findings, 311
Apophysitis, pelvic, 681 Apoptosis activation-induced, 44 B cell, 42 definition of, 125 description of, 642 frequency of, 44 passive cell death, 44 pathways of, 44, 45f, 642 in systemic lupus erythematosus, 347 T-cell, 46 Arachidonic acid, 105 Area under the time-concentration curve, 150 Arteritis Takayasu's, 337, 548-550, 551f temporal, 493t-494t, 550--551 Arthralgia, 2 Arthritis. See also speeific arthritis acne and, 579 aseptic, 652 chronic,5 fungal, 584 gonococcal, 574 in immunocompromised patients, 575-576 infectious agents that cause, 214 ]acoub type of, 352 in juvenile ankylosing spondylitis, 310 Lyme, 597, 597t pancreatitis with, 638 postinfectious, 568, 585 sacroiliac in inflammatory bowel disease, 335 in juvenile ankylosing spondylitis, 315-316 in juvenile rheumatoid arthritis, 230--231 with scoliosis, 746t in systemic lupus erythematosus, 352 traumatic, 687-688 tuberculous, 574, 574f-575f word origin of, 2 Arthritis-dermatitis syndrome, 585 Arthrogryposis, 747t, 761 Arthropathy frostbite, 689, 689f gouty, 633 hemophilic, 636f psoriatic, 4t rubella virus-associated, 581-582 Articular cartilage anatomy of, 11£ chondrocytes of, 10--11 description of, 9-10 zones of, 10 Aschoff body, 621 Aseptic arthritis, 652 Aseptic meningitis, 673 Aseptic necrosis, 352, 353f Aspartic proteinases, 13, 14t Aspirin dosing of, 233-234 juvenile rheumatoid arthritis treated with, 233-234 Kawasaki disease treated with, 531 Assay sensitivity, 154 Ataxia telangiectasia, 647t Atheromata, 355 Atlantoaxial subluxation, 230, 231f Atrioventricular block, congenital animal models of, 400 description of, 393-394 endomyocardial fibroelastosis in, 395 epidemiology of, 395 history of, 394-395 long-term outcome for, 396 pathologic findings, 395 treatment of, 395-396 Atrophoderma of Pasini and Pierini, 472
Anributable risk, 146t Auranofin description of, 99-100 juvenile rheumatoid arthritis treated with, 239-240 Aurothioglucose, 100, lOOt Aurothiomalate description of, 99-100, lOOt juvenile rheumatoid arthritis treated with, 239 Autoantibodies in juvenile dermatomyositis, 424-426 in juvenile rheumatoid arthritis, 212 in neonatal lupus erythematosus, 393-394 in oligoarthritis, 277 in systemic arthritis, 292 Autoantigens, 48 Autoimmune diseases common variable immunodeficiency and, 652 development of, 48 environmental factors associated with, 49-50 gene mutations associated with, 49 genetic predisposition to, 48-49 juvenile rheumatoid arthritis and, 71, 216 major histocompatibility complexes and, 49 multifactorial origin of, 48 pathogenic effects in, 48, 49f tissue injury in leukocytes involved in, 52, 54-55 mechanisms of, 50--59 type I hypersensitivity, 50 Autoimmune lymphoproliferative syndrome, 45,642 Autoimmune polyendocrinopathy candidiasisectodermal dystrophy, 46 Autoimmune regulator, 46 Autoimmunity diffuse cutaneous systemic scleroderma and, 443-444 infection and, 49-50 Autologous bone marrow transplantation, 380 Autologous stem cell transplantation description of, 126 diffuse cutaneous systemic scleroderma treated with, 459-460 juvenile dermatomyositis treated with, 431-432 juvenile rheumatoid arthritis treated with, 240 severe combined immunodeficiency treated with,647 systemic arthritis treated with, 300 Autorecognition, 48 Avascular necrosis glucocorticoid-related, 107 Legg-Calve-Perthes disease, 684, 685f of femoral head, 684 Axial loading test, 707t Axial skeleton, 313-314 Azathioprine description of, 113f, 113-114, 114t systemic lupus erythematosus treated with, 378,378t Azurophilic granules, 19-20
B B7-1, 35, 43 B7-2, 35, 43 B cell(s) abnormalities of primary, 646-648 in systemic lupus erythematosus, 346
INDEX
activation of cytokines involved in, 41-42 signals for, 39 T helper-dependent mechanism of, 41-42 thymus-dependent, 40, 40f thymus-independent, 39-40, 40f apoptosis of, 42 development of, 31 immature, 31 immunodeficiencies of, 647t memory, 43 selection of, 31 T-cell interactions with, 40-42 B cell receptor complex, 40, 41£ B cell-activating factor of TNF, 42 Bl cells, 31 Back exercises, 193f Back pain diskitis, 689-690 epidemiology of, 689 nonorganic, 707t treatment of, 690 Baker's cyst, 217, 218f Band keratopathy, 280, 282f, 283 Bartonella henselae, 575 Basilar interstitial pneumonitis, 365-366 Bayesian approach, 161 Beaded filament-forming collagens, 12t Beals' contractural arachnodactyly syndrome, 6791, 747t Beh~et's syndrome central nervous system disease in, 563 classification of, 561 clinical manifestations of, 562-564 course of, 565 definition of, 561 description of, 102, 505 diagnosis of, 5611, 562t, 564 epidemiology of, 561 etiology of, 562 fever associated with, 658 gastrointestinal disease in, 564 genetic background for, 561-562 genital ulcers in, 565 history of, 561 HLA-B51 and, 562 imaging studies for, 564 laboratory examination for, 564 methylprednisolone for, 565 mucocutaneous disease associated with, 562-563 musculoskeletal disease and, 563 ocular disease in, 563, 565 oral ulcerations in, 562, 563f, 565 pathogenesis of, 562 pathologic findings of, 564 prednisone for, 565 prognosis for, 565 renal disease in, 564 skin manifestations of, 562-563 treatment of, 565 ulcerations in, 562, 563f, 565 uveitis in, 563 vascular disease in, 563-564 Benign angiitis of the central nervous system, 547 Benign hypermobility syndrome, 676 Benign rheumatoid nodules, 264, 264f Benign tumors chondroma, 730-731, 732f-733f eosinophilic granuloma, 735-736 fibrous cortical defect, 731-732, 734f fibrous dysplasia, 732-733 giant cell tumor, 735
juvenile fibromatosis, 732 of bone, 728-730 of cartilage, 730-731 of fibrous tissue, 731-734 of soft tissue, 734-735 osteochondroma, 730, 731£-732f pigmented villonodular synovitis, 734 synovial chondromatosis, 734-735 synovial hemangioma, 734, 734f Bias confounding, 159 description of, 158-159 interview, 159 measurement, 159 recall, 159 selection, 159 Bibasilar pulmonary fibrosis, 457, 459f Bimodal distribution, 160 Binomial test of proportions, 164 Bioavailability, 76 Bioinformatics, 66-67 Biologic therapies adalimumab, 123 anakinra, 124 etanercept, 121-122, 237 infliximab, 122-123 intravenous immunoglobulin, 118-119, 119t juvenile dermatomyositis treated with, 431-432 juvenile rheumatoid arthritis treated with, 237-238 tolerance induction, 119-120 Biotechnology, 66-67 Bisphosphonates adverse effects of, 724t chemical structure of, 724f osteogenesis imperfecta treated with, 749 osteoporosis treated with, 110, 724 Bleomycin, 454-455 Blind assessor, 152-153 Blinded withdrawal design, 154 Blinding, 152-153 Blocked randomization, 153 Bloom's syndrome, 349 Bone anatomy of, 716 biochemical markers associated with, 717-718 calcium-regulating hormones in, 718 composition of, 716-717 cortical, 716 diaphysis of, 716 endochondral ossification of, 716 epiphysis of, 716 glucocorticoid-related loss of, 107, 722 membranous ossification of, 716 metaphysis of, 716 remodeling of, 717t resorption of, 717-718 testosterone effects on, 716 trabecular, 716 turnover of, 717 Bone cysts, 735, 735f Bone disorders age at onset, 744 diagnosis of, 744-745 distribution of involvement, 744 molecular mechanisms of, 745 radiographic abnormalities associated with, 744-745 Bone marrow transplantation, 126, 380 Bone mass childhood arthritis effects on, 721 determinants ot~ 719 heredity factors, 719
769
low description of, 721 pediatric disorders associated with, 723t peak, 718-719 sex differences in, 718 Bone mineral(s) calcium-regulating hormones, 718 1,25-dihydroxyvitamin 03, 718 metabolism of, 716-717 osteoprotegerin, 718 receptor activator of nuclear factor-KB, 718 receptor activator of nuclear factor-leB ligand, 718 types of, 716-717 Bone mineral content, 718 Bone mineral density areal,720 description of, 718 drug treatment effects on, 722 measurement of dual x-ray absorptiometry,719-720 dual-energy x-ray absorptiometry, 719-720, 720f methods for, 720t quantitative computed tomography, 720, 720t quantitative high-frequency sonography, 720t, 720-721 volumetric, 720 Bone morphogenetic proteins, 9 Bone tumors benign, 728-730 characteristics of, 729t classification of, 728, 728t clinical presentation of, 728 malignant, 736-738 metastatic, 739 osteoblastoma, 729-730 osteoid osteoma, 729, 730f-731f, 730t osteosarcoma, 736-738, 737f Bonferroni correction, 162 Borrelia burgdorferi description of, 475, 592 laboratory detection of, 595-597 Brachial neuritis, 693 Brachial synovial cyst, 217, 217f Brachydactyly, 219, 219f, 227, 2291' Brain pain modulation by, 700 periaqueductal gray area of, 700 Brain-derived neurotrophic factor, 699 British Isles Lupus Assessment Group Activity Index, 372, 374t Broadband ultrasound attenuation, 720-721 Brodie's abscess, 576-577, 577t, 5781' Brucellosis arthritis associated with, 575 fever caused by, 657 Budd-Chiari ~)'ndrome, 367 Bullous morphea, 472 Bursae function of, 14 subscapular, 14 Bystander activation hypothesis, 50 Bystander immunosuppression, 48
c Cl deficiency of, 645-646 immune complex binding of, 645 inhibitors of, 25 C2 deficiency, 645, 646
770
INOEX
C3 deficiency of, 644, 646 description of, 24 juvenile rheumatoid arthritis levels, 225 C4 deficiency, 646 C5 deficiency, 646 C fibers, 697 Cadherins, 54t Caffey's disease, 578, 761 Calcineurin, 34, 117 Calcinosis in diffuse cutaneous systemic scleroderma, 447, 449f in juvenile dermatomyositis course of, 433 description of, 413, 416f management of, 432 Calcinosis circumscripta, 416f Calcinosis universalis, 417f Calcitriol, 110, 718 Calcium hormones that regulate, 718 osteoporosis prevention and, 722-723 Calcium pyrophosphate deposition disease, 635 Calreticulin, 393 Camptodactyly, 266, 761 Camptodactyly-arthropathy-coxa varapericarditis syndrome, 11, 746t, 761 Candidate genes, 68, 71 Canonical correlation, 170-171 Capacity-limited kinetics, 77 Carboxylterrninal propeptide of type I procollagen, 717, 717t Cardiac disease in diffuse cutaneous systemic scleroderma, 451 in juvenile rheumatoid arthritis, 221 in polyarthritis, 264-265 Cardiac tamponade juvenile rheumatoid arthritis and, 221 systemic arthritis and, 294 Cardiovascular system glucocorticoids effect on, 109 Kawasaki disease and, 526-527 Carpal tunnel syndrome, 693 Cartilage benign tumors of, 730-731 disordered development of, 762 malignant tumors of, 738 Cartilage oligometric matrix protein gene, 754 Cartilage-derived morphogenetic protein 1, 9 Cartilage-hair hypoplasia, 647t, 648 Cartilaginous joints, 9, lOt Cartilaginous tumors, 730-731 Case exposure rate, 146t Case-control classroom peers, 146 Case-controlled retrospective study, 145-146 Caspase-1, 56-57 Caspases, 44 Cataracts, 109 Catastrophic antiphospholipid syndrome, 357-358 Categorical variables, 157 Cathepsin B, 14t Cathepsin 0, 14t Cathepsin G, 14t Cathepsin L, 14t CCR7, 41, 43 CD!,29t C02,29t C03 deficiency of, 647t description of, 29t, 32 C04, 29t, 32
C05,29t C08 deficiency of, 647t description of, 32 COlla,29t C018,29t C019,29t C021,29t C027,29t C028, 29t, 35 C034,29t C040, 29t, 41 C040 ligand, 35, 38, 41 C044, 29t, 43, 444 C045, 29t, 43 C080,29t C086,29t C095, 29t, 44 C0152, 29t C0154, 29t, 120 cONA molecules, 65 Ceiling effect, 175t Celecoxib gastrointestinal effects of, 84 structure of, 78f Cell adhesion molecules description of, 54t in juvenile psoriatic arthritiS, 329 Cell cycle, 113 Cell-mediated immune reactions, 108 Central nervous system Beh~et's syndrome and, 563 benign angiitis of, 547 diffuse cutaneous systemic scleroderma effects, 451 glucocorticoids effect on, 108-109 juvenile ankylosing spondylitis findings, 311 juvenile rheumatoid arthritis findings, 222 Kawasaki disease effects, 527 methotrexate effects on, 95 nociceptive pathways, 697-698 nonsteroidal anti-inflammatory drug-related toxicities, 85 primary angiitis of, 547-548 systemic lupus erythematosus manifestations, 353-354, 354t, 366-367, 380 Central sensitization, 698f Central tolerance, 45-46 Cervical spine arthritis of, 311 radiographs of, 318f Charcot's joint, 689 Chediak-Higashi syndrome, 644 ChemokineCs) definition of, 54 description of, 52 inflammatory, 54 lymphoid, 54 subfamilies of, 54 types of, SSt Chemokine receptors, 26t Chest pain, 690-691, 69lt Chilblains, 449, 693, 708t Child Health Questionnaire, 177t, 180 Childhood Arthritis Health Profile, 177t, 179 Childhood Arthritis Impact Measurement Scales, 177 Childhood Health Assessment Questionnaire, 177t, 177-178,242-243 Childhood Myositis Assessment Scale, 180 Children's Hospital of Eastern Ontario Pain Scale, 702 Chi-square test errors with, 166
goodness-of-fit, 164 Mantel-Haenszel, 159, 166 McNemar, 166, 166t with more than one degree of freedom, 167 with one degree of freedom, 165, 165t Chlorambucil description of, 117, 117f juvenile rheumatoid arthritis treated with, 238 Choline magnesium trisalicylate, 77f, 234 Chondroblastoma, 731, 733f Chondrocalcinosis, 635 Chondrocytes, 10-11 Chondrodysplasia punctata, 399, 759 Chondroitin sulfate, 11 Chondroma, 730-731, 732f-733f Chondromalacia patellae, 680 Chondromyxoid fibroma, 731 Chondrosarcoma, 738 Chorea, 354 Chromosome maps, 65 Chronic arthritis. See also Juvenile chronic arthritis; Juvenile idiopathic arthritis; Juvenile rheumatoid arthritis American College of Rheumatology criteria for, 206, 207t classification of, 206 clinical manifestations of, 216-220 definition of, 206 European League Against Rheumatism classification criteria for, 206-207, 207t family history of, 213 historical review of, 208-209 International League of Associations for Rheumatology classification criteria for, 207-208 juvenile psoriatic arthritis as cause of, 324 osteoporosis risk factors in, 721t pain associated with, 216-217 selective IgA deficiency and, 649, 650f signs and symptoms of, 216 Chronic graft-versus-host disease, 453-454, 454t Chronic granulomatous disease, 643-644 Chronic infantile neurologic cutaneous and articular syndrome characteristics of, 67If clinical manifestations of, 660t, 673f--674f, 673-674 genetics of, 672 ocular findings in, 674f skin manifestations of, 673, 673f Stickler's syndrome vs., 750 treatment of, 674 Chronic pain, 192, 194, 703 Chronic recurrent multifocal osteomyelitis, 577-578, 580f, 708t Churg-Strauss syndrome, 493t, 545-546 Circinate balanitis, 607, 608f Circulus articularis vasculosus, 14 Clearance, 76-77 Clinical epidemiology, 144 Clinical equipoise, ISS Clinical research definition of, 143 obstacles to, 142 translational blocks in, 142 view of, 142 Clinical research organizations, 155 Clinical response claims, 156 Clinical studies bias in, 158-159 case-controlled retrospective, 145-146 concepts associated with, 143 confidence intervals, 161
INDEX experimental, 143 hypothesis-generating, 143 hypothesis-testing, 143 measures of central tendency, 159-160 meta-analysis, 143 non-normal distributions, 161 objectives of, 143 observational, 143 prospective, 143 prospective cohort study, 146 report of, 171 retrospective, 143 standard deviation, 160 standard error of the mean, 161 terminology associated with, 143 variables categorical, 157 concepts related to measurement of, 158-159 continuous, 157-158 ordinal, 157, 157f variance, 160 Z scores, 160-161 Clonal anergy, 46 Cluster of differentiation, 28, 29t Coagulation, 86 Cochrane Collaboration, 142 Cockayne's syndrome, 349, 747t Coefficient of concordance, 169 Cogan's syndrome, 518 Cohen's kappa, 169 Colchicine characteristics of, 101-102 diffuse cutaneous systemic scleroderma treated with, 460 dosing of, 102 familial Mediterranean fever treated with, 661 structure of, 102f Cold therapy, 197-198 Collagen beaded filament-forming, 12t biosynthesis of, 12, 13f characteristics of, 11-12 diffuse cutaneous systemic scleroderma and,445 fibril-associated collagens with interrupted triple-helices, 12t fibril-forming, 12t nerwork-forming, 12t of anchoring fibrils, 12t proteinases for, 13 with transmembrane domain, 12t type II, 749-750 types of, 11-12, 12t Collagen disorders classification of, 748t description of, 747 Ehlers-Danlos syndromes, 747-748, 749t Kniest's syndrome, 745, 751, 751£ osteogenesis imperfecta, 748-749, 749t-750t, 750f spondyloepiphyseal dysplasias, 751-752, 752f,752t Stickler's syndrome, 677t, 745, 746t, 750-751 type I, 748-749 type II, 749-752 type III, 752-753 type IV, 753 type IX, 754 type V, 753 type VI, 754 type X, 754
Collagenase, 14t Collectin, 26 Combined immunodeficiency, 648 Common variable immunodeficiency, 649, 651-653 Comparative clinical trials, 152, 154-155 Complement cascade, 25, 50 Complement deficiencies description of, 25 rheumatic diseases associated with, 644-646 in systemic lupus erythematosus, 344-345, 371 Complement receptors, 24-25, 25t Complement system activation of, 24 alternative pathway of, 24, 25f biologic activities of, 24-25 classic pathway of, 24, 25f description of, 24 fragments, 24-25 lectin pathway of, 24, 25f Complex genetic traits description of, 67-68 pediatric rheumatic illnesses as, 69-71 Complex regional pain syndrome, 704, 705t Compliance, 186, 186t Confidence intervals for clinical studies, 161 for statistical tests, 164 Confounding bias, 159 Congenital atrioventricular block animal models of, 400 description of, 393-394 endomyocardial fibroelastosis in, 395 epidemiology of, 395 history of, 394-395 long-term outcome for, 396 pathologic findings, 395 treatment of, 395-396 Congenital contractural arachnodactyly, 757 Conjunctivitis, 524, 607 Connective tissue collagens of, 11-12, 12t composition of, 12-13 entheses, 15 fasciae, 15 ligaments, 15 tendons, 15 Connective tissue diseases and disorders arthritis associated with, 745, 746t diagnosis of, 744-745 frequency of, 4t ligamentous laxity, 745-746 mixed anti-UI-70Kd antibodies in, 485 characteristics of, 483t classification of, 482 clinical manifestations of, 482-483 course of, 485-486 definition of, 482 diagnostic criteria for, 483-484, 484t differential diagnosis, 483-484 epidemiology of, 482 immunogenetic background of, 482 laboratory examination for, 484-485 pathologic findings in, 483 prognosis for, 485-486 treatment of, 485 molecular mechanisms of, 745 mosaicism associated with, 745 osteopenia and, 721-722 osteoporosis and, 721-722 types of, 746t Connective tissue growth factor, 444 Conradi-Hiinermann syndrome, 747t, 759
771
Conserved sequence elements, 64 Consolidated Standards of Reporting Trials, 171 Content validity, 175t Continuity correction of Yates, 165-166 Continuous variables, 157-158 Control exposure rate, 146t Convergent validity, 175t Coombs' test-positive hemolytic anemia, 358 Coordinating center, 155 Cornelia de Lange syndrome, 747t Coronary artery aneurysms, 526, 527f Coronary artery disease description of, 109 systemic lupus erythem:llosus and, 354-355 Correlation canonical, 170-171 intraclass, 169 Pearson product-moment, 167-168 Spearman rank, 168 Cortical bone, 716 Cortical hyperostosis, 761 Costimulatory molecules, 35 Costochondritis, 692 Cotton-wool spots, 358, 359f, 366 Counseling description of, 186 family, of juvenile rheumatoid arthritis patients, 242 Covariates, 168 CRl,25t CR2,25t CR3,25t CR4,25t CRZ-CD19-CD81 complex, 39-40 C-reactive protein acute rheumatic fever and, 622 description of, 26 juvenile rheumatoid arthritis and, 223 septic arthritis and, 571 Creatine kinase description of, 15 in juvenile dermatomyositis, 426 Creatine/creatinine ratio, 424 CREST syndrome, 442 Criterion validity, 175t CRMO syndrome, 329 Crohn's disease. See also Inflammatory bowel disease antineutrophil cytoplasmic antibody in, 337 gastrointestinal manifestations, 335 laboratory examination of, 337 methotrexate for, 338 Cronkhite-Canada syndrome, 506 Crossover design, 153 Cryoglobulins, 371 Cryopyrin-associated periodic syndromes mutations in, 672f overview of, 671-672 Cryopyrinopathies, 662 Cryptic epitopes, 47 Cryptic self, 47 C-terminal telopeptides, 717t CTLA-4, 43, 46, 120 Cumulative percentage of drug recovered, 150 Cushing'S syndrome clinical features of, 106-107 glucocorticoids and, 106t, 106--107, 236, 430 iatrogenic, 107 Cutaneous leukocytoclastic angiitis, 493t Cutaneous neonatal lupus erythematosus dermatitis of, 397 description of, 393 differential diagnosis of, 398
772
INDEX
Cutaneous neonatal lupus erythematosus (Continued) epidemiology of, 396 history of, 396 lesions of, 397 pathologic findings in, 397-398 rash associated with, 396 subacute, 396, 397f-398f telangiectasias in, 397 treatment of, 398 Cutaneous polyarteritis, 517-518, 518f Cutis laxa, 754 CYRR,643 Cyclic citrullinated peptides, 268 Cyclic hematopoiesis, 66~66 Cyclooxygenase-l, 58,78 Cyclooxygenase-2 description of, 58, 78-79 inflammation-induced, 699 physiologic function of, 79 Cyclooxygenase-2 inhibitors effectiveness of, 79 juvenile psoriatic arthritis treated with, 330 juvenile rheumatoid arthritis treated with, 233 mechanism of action, 78-79 structure of, 78f Cyclophosphamide administration of, 116, 116t adverse effects of, 116 Cogan's syndrome treated with, 518 diffuse cutaneous systemic scleroderma treated with, 459 dosing of, 115t fertility effects of, 116 guidelines for, 115t intravenous pulse, 379 malignancies associated with, 116-117 pharmacokinetics of, 115 polyarteritis nodosa treated with, 515 structure of, 115f syndrome of inappropriate antidiuretic hormone secretion and, 116 systemic lupus erythematosus treated with, 378t, 379 lOxicities associated with, 116 Wegener's granulomatosis treated with, 542, 544t Cyclosporine description of, 117f, 117-118, 118t diffuse cutaneous systemic scleroderma treated with, 459 juvenile dermatomyositis treated with, 431 juvenile rheumatoid arthritis treated with, 238 systemic arthritis treated with, 299 systemic lupus erythematosus treated with, 379 uveitis treated with, 292 Cysteine, 13, 14t Cystic fibrosis, 637-{)38 Cystinosis, 633f Cytokine(s) antibody response functions of, 41-42 cell activation mechanisms, 56f classification of, 26, 27t definition of, 26 description of, 55-57 in juvenile psoriatic arthritiS pathogenesis, 326 in juvenile rheumatoid arthritis pathogenesis, 211-212 methotrexate effects on, 89 monocyte-derived, 121 of adaptive immunity, 27t of innate immunity, 26, 27t
in polyarthritis pathogenesis, 261-262 proinflammatory, 23-24 in systemic arthritis, 291-292 in systemic lupus erythematosus, 345 Thl cell production of, 38 Th2 cell production of, 38 Cytokine receptors, 26, 26t Cytotoxic agents azathioprine, 113f, 113-114, 114t description of, 113 juvenile rheumatoid arthritis treated with, 238 mycophenolate mofetil, 114f, 114-115, 115t Cytotoxic lymphocyte-associated antigen 4, 35,36f Cytotoxic T-cells apoptosis induced by, 39 description of, 31-32 effector functions of, 39 function of, 28
D Dactylitis, 266, 327f, 574 Data analysis plan analysis sets, 157 description of, 155 primary response variables, 156 response variables, 156-157 secondary response variables, 156 Data collation, 155 de Quervain's disease, 683 Death domain, 44, 45f Decision analysis, 148-149 Deep morphea, 473-474, 476 Deflazacort, 109-110 Degree of freedom, chi-square test with, 165, 165t, 167 Dehydroepiandrosterone, 212-213 Delayed-type hypersensitivity, 52 Dendritic cells description of, 19, 23, 36 follicular, 42 in systemic lupus erythematosus, 347 Dependent variable, 161 Dermatan sulfate, 11 Dermatitis, 397 Dermatomyositis sine myositis, 413 Dermatomyositis-like syndrome, 652-653, 653t Descriptive epidemiology, 144 Descriptive statistics, 159 Dexamethasone dosing of, 103t structure of, 103f Diabetes cheiroarthropathy, 636, 637f Diabetes mellitus hemochromatosis and, 637 juvenile rheumatoid arthritis and, 216 musculoskeletal complications of, 636-{)37 Diabetes osteopathy, 637 Diabetic cheiroarthropathy, 456 Diagnosis accuracy in, 185 description of, 146-147 Diagnostic test validity, 147, 147t Diapedesis, 52 Diaphyseal aclasis, 762 Diarthrodial joints, 9 Diarthroses, 9 Diastrophic dwarfism, 747t Diastrophic dysplasia, 754
Diclofenac dosing of, 82t mechanism of action, 79 structure of, 77f Differential misclassification bias, 159 Diffuse cutaneous systemic scleroderma age at onset, 442-443 antinuclear antibodies in, 456 autoimmunity associated with, 443-444 bibasilar pulmonary fibrosis in, 457, 459f calcinosis in, 447, 449f cardiac disease in, 451 cardiac function and, 456 CD44 and, 444 central nervous system disease in, 451 chemokines involved in, 444 chronic graft-versus-host disease vs., 453-454, 454t clinical manifestations of, 445-452 collagen abnormalities in, 445 connective tissue growth factor and, 444 course of, 462-463 cytokines involved in, 444 deaths caused by, 462-463 definition of, 442 diabetic cheiroarthropathy vs., 456 differential diagnosis, 453-456 disease activity and severity assessments, 457,459t endothelial cell factors associated with, 445 epidemiology of, 442-443 etiology of, 443-445 familial patterns of, 445 fibroses differentiated from, 455 gastrointestinal disease in, 450-451, 461 genetic background of, 445 histopathologic findings in, 452-453 immunologic factors in, 444 incidence of, 442 laboratory examination in, 456-457 mortality rates for, 462-463 musculoskeletal disease in, 449-450 myocardial fibrosis in, 452, 453f nail fold abnormalities in, 448, 448f nephrogenic fibrosing dermopathy vs., 454 pathogenesis of, 443-445 pathologic findings in, 452-453 phenylketonUria vs., 455 porphyria cutanea tarda vs., 456 progeria vs., 455 prognosis for, 462-463 pseudosclerodermas vs., 455 pulmonary disease in, 451, 461 pulmonary function and, 456-457 pulmonary hypertension in, 461 radiologic examination in, 457 Raynaud's phenomenon in description of, 447-449, 453 treatment of, 460, 461t renal disease in, 451,460-461 renal function and, 457 scleredema vs., 455-456 sclerosis associated with, 446 Sicca syndrome and, 451-452 signs and symptoms of, 445, 446t skin manifestations of, 445-447 telangiectases in, 446, 447f-448f treatment of colchicine, 460 cyclophosphamide, 459 cyclosporine, 459 description of, 457-458 glucocorticoids, 459 hematopoietic stem cell transplantation, 459-460
INDEX immunomodulatory therapy, 459-460 interferons, 460 methotrexate, 459 mycophenolate mofetil, 459 D-penicillamine, 460 recombinant human relaxin, 460 skin care, 458 supportive measures, 458 tumor necrosis factor inhibitors, 459 vascular factors associated with, 444-445 Werner's syndrome vs., 455 Diffuse proliferative glomerulonephritis, 363, 363f-364f, 380 Diffusing capacity in the lung, 356 DiGeorge syndrome, 647t, 648 Dihydrofolate reductase, 88 1,25-Dihydroxyvitamin D3, 718 Dimers,30 Disability claim, 156 Disabling pansclerotic morphea of children, 474 Discoid lupus erythematosus, 643 Discriminant function analysis, 170 Discriminant instrument, 175t Disease diagnosis of, 146-147 etiology of, 145 prognosis of, 147-148 Disease-modifying antirheumatic drugs antimalarials and, 96 combination therapies using, 125 definition of, 88 juvenile rheumatoid arthritis treated with, 238-239 Disease-specific susceptibility, 68 Disk herniation, 691 Diskitis, 579-580, 581f, 689-690 Distal interphalangeal joint polyarthritis, 263 Distracted straight leg raising, 707t Domains, 175t Double-blind clinical trials, 152 Double-dummy design, 153 Down syndrome, 216, 677t Dressing, 196, 197f Drug absorption, 76 Drug biotransformation, 77 Drug safety, 150 Drug tolerability, 150 Drug-induced systemic lupus erythematosus, 348, 348t, 382-383 Dual x-ray absorptiomerry, 718 Dual-energy x-ray absorptiometry, 719-720, 720f Dual-photon absorptiometry, 719 Dupuytren's contracture, 455, 637 Dutch-type periodic fever, 665 Dyggve-Melchior-Clausen dysplasia, 754 Dynamic randomization, 153 Dynamic splints, 198, 198f Dyslipidemia, 109 Dyslipoproteinemia, 225, 381 Dysostosis multiplex description of, 757-758 mucolipidoses, 758t, 758-759 mucopolysaccharidoses, 758, 758t
E Ectrodactyly-ectodermal dysplasia syndrome, 747t Ehlers-Danlos syndromes clinical manifestations of, 677t, 747-748, 749t description of, 747-748 type I, 749t, 753, 753f
type II, 749t, 753 type III, 749t, 752 type IV, 749t, 752 type V, 749t, 754 type VI, 749t, 753 type VII, 749, 749t type VIII, 749t, 753 type X, 749t, 754 Eicosanoids, 58, 58f Eigenvalue, 170 Elastase, 14t Elastin, 12 Elbow exercises, 190f Ellis-van Creveld syndrome, 745 Embryopathy, 102 En coup de sabre scleroderma, 472, 474f, 476 Enchondromatosis, 731, 762 Endemic osteoarthritis, 631-633 Endocarditis acute rheumatic fever and, 619, 621 juvenile rheumatoid arthritis and, 221 Libman-Sacks, 355, 366, 366f, 622 prophylaxis for, 624 systemic arthritis and, 294 Endochondral ossification, 716 Endomyocardial fibroelastosis, 395 Endomysium, 15, 16f Endotenon, 15 Endothelial cells antigen antibodies in juvenile dermatomyositis, 425-426 in Kawasaki disease, 530 diffuse cutaneous systemic scleroderma and, 445 Endothelin-l, 107 Engelmann's syndrome, 759, 760f Entheses description of, 15, 309 examination of, 312, 312f radiographic evaluation of, 317, 318f Enthesitis arthritis caused by, 265 differential diagnosis, 311 in juvenile ankylosing spondylitis, 309-311, 319 in juvenile psoriatic arthritis, 328 management of, 319 in reactive arthritis, 606 Enzyme-linked immunosorbent assay, 224 Eosinophil(s), 38 Eosinophilia-myalgia syndrome, 475 Eosinophilic fasciitis, 474 Eosinophilic granuloma, 735-736 Epidemiology clinical, 144 descriptive, 144 frequency of disease occurrence, 144-145 Koch's postulates, 145 Epimysium, 15, 16f Epiphyseal dysplasias, 756 Epiphyseal plate, 14 Epiphysis, 14-15 Epitenon, 15 Epitope, 30 Epitope spreading, 49 Epstein-Barr virus-associated lymphoma, 95 EqUivalence margins, 154 Errors Chi-square test, 166 nonrandom, 158 random, 158 statistical, 162-163 type I, 162 type II, 162
773
Erysipeloid erythema, 660, 660f Erythema elevatum diutinum, 507 Erythema marginatum, 620, 620f Erythema migrans, 594, 594f, 598-599 Erythema nodosum, 336, 336f Erythrocyte sedimentation rate, 223 Erythromelalgia, 693, 708t E-selectin, 52, 54t, 329, 445 Essential cryoglobulinemic vasculitis, 493t Etanercept description of, 121-122 juvenile ankylosing spondylitis treated with, 319 juvenile dermatomyositis treated with, 429t juvenile rheumatoid arthritis treated with, 237 polyarthritis treated With, 269 tumor necrosis factor receptor-associated periodic syndrome treated with, 663 Etiology of disease, 145. See also specific disease European League Against Rheumatism, 2, 206-207, 207t European Spondyloarthropathy Study Group, 305 Evans' syndrome, 368 Evidence-based medicine, 142 Ewing's sarcoma, 738-739, 739f Exercises play-related, 196t range of motion, 189, 189f-194f stretching, 195, 195f therapeutic, 195-196 water, 198-199 Experimental myositis, 410 Experimental studies, 143 Exploratory studies, 143 Expressed sequence tags, 64-65 External validity, 158 Extracellular matrix composition of, 11 description of, 9-11 proteoglycans, 11
F F ratio, 167 Fabry's disease, 639, 639t, 708t, 747t Face validity, 175t Faces Rating Scale, 702, 702f Factor analYSiS, 170 Factor loading, 170 Factor loading matrix, 170 Factorial design, 153 Fairbank's multiple epiphyseal dysplasia, 754 False pOSitive, 147 Familial aero-osteolysis, 759-760 Familial chondrocalcinosis, 635 Familial cold autoinflammatory syndrome characteristics of, 671f clinical manifestations of, 660t, 672-673 description of, 662 genetics of, 672 Familial fibrosing serositis, 679t Familial Hihernian fever, 659 Familial hypertrophic synovitis, 266 Familial infantile cortical hyperostosis, 761 Familial lipochrome histiocytosis, 644 Familial Mediterranean fever amyloidosis associated with, 661 clinical manifestations of, 659-661, 660t colchicine for, 661 cutaneous manifestations of, 660 description of, 101, 505 diagnosis of, 661 genetics of, 659
774
INDEX
Familial Mediterranean fever (Continued) joints affected by, 660 laboratory studies of, 661 outcome of, 661--662 pathogenesis of, 659 prognosis for, 661--662 skin manifestations of, 660 treatment of, 661 vasculitis and, 518 Farber's disease, 638--639, 6391, 747t Fas gene mutations, 642-643 Fas-associated death domain, 45f, 642 Fasciae, 15 Fast muscle fibers, 16, 16t Fatigue, 191 Fc receptors description of, 38, 42, 43t polymorphisms of, in systemic lupus erythematosus, 345 Felty syndrome, 264 Femoral head osteonecrosis, 230 Fenoprofen dosing of, 81t structure of, 77f toxicities associated with, 83t Fetal alcohol syndrome, 762 Fever description of, 657 Kawasaki disease and, 523-524 periodic syndromes. See Periodic fever syndromes rat-bite, 657--658 relapsing, 658 repeated episodes of, 657 rheumatic. See Acute rheumatic fever in systemic arthritis, 293, 297 Fever of unknown origin, 657 Fibril-associated collagens with interrupted triple-helices, 12t Fibril-forming collagens, 12t Fibroblasts, 23-24 Fibrodysplasia ossificans progressiva, 424 Fibroma nonossifying, 731-732, 734f ossifying, 734 Fibromatosis collJ, 455 Fibromatosis hyallnica multiplex, 455 Fibromyalgia, 705t, 709 Fibronectin, 12 Fibrosarcoma, 738 Fibrous cortical defect, 731-732, 734f Fibrous dysplasia, 732-733 Fibrous joints, 9, lOt Fibrous tissue benign tumors of, 731-734 malignant tumors of, 738 Finger exercises, 192f First-order kinetics, 77 First-pass effect, 77 Fisher's exact test, 166 FK506,34 Flat foot, 679, 679f Floor effects, 175t Fluorosis, 631 Focal segmental proliferative glomerulitis, 362f-363~ 362-363 Folate supplementation, with methotrexate, 96, 234 Follicular dendritic cells, 42 Foot juvenile rheumatoid arthritis of, 229 pes cavus, 678--679 pes planus, 678--679 FoxP3,47
Freiberg's disease, 685--686, 686f Frequency distribution, 159
Frostbite arthropathy, 689, 689f Full-analysis set, 157 Functional asplenia, 360 Functional genomics description of, 71-72 in pediatric rheumatic diseases, 73-74 Functional map, 66 Functional splints, 198 Functional status definition of, 174 measurement of description of, 175 Instruments for, 177-180 Fungal arthritis, 584
G G-actin, 15 Gangliosidosis, 747t Gardner-Diamond syndrome, 358f Gardner's syndrome, 455 Gastrointestinal system azathioprine effects on, 114 Beh~et's syndrome manifestations, 564 celecoxib effects on, 84 diffuse cutaneous systemic scleroderma manifestations of, 450-451 Henoch-Schonlein purpura manifestations, 497 inflammatory bowel disease symptoms, 335-336 juvenile rheumatoid arthritis findings, 221 methotrexate effects on, 92 nonsteroidal anti-inflammatory drugs effect on, 80, 82-84 rofecoxib effects on, 84 Gastropathy, nonsteroidal anti-inflammatory drug-associated, 83 GATA-3,37 Gaucher's disease, 639, 639t Gaussian distribution, 159, 160f Gelatinase, 13, 14t GEMSS syndrome, 747t Gene therapy, 125-126 Generalizability, 158, 175t Generalized morphea, 472, 476 Genetic map, 65--66 Genome definition of, 64 polymorphic elements of, 65 polymorphisms, 65 structure of, 64--65 Genome maps, 65 Genome screen, 68--69 Genomics definition of, 64 functional, 71-72 Genu recurvatum, 679 Giant cell arteritis clinicopathologic characteristics of, 494t definition of, 493t, 548 Takayasu's arteritis, 337, 493t-494t, 548-550, 551f temporal arteritiS, 493t-494t, 550-551 Giant cell tumor, 735 Glaucoma, 109 Glomerular sclerosis, 364-365 Glomerulonephritis. See also Lupus nephritis diffuse proliferative, 363, 363f-364f, 380 Henoch-Schonlein purpura and, 497-498 membranous, 363-364, 364f, 377t systemic lupus erythematosus and, 361-365 Glucocorticoid(s) acetylsalicylic acid excretion affected by, 88
acute adrenal insufficiency caused by, 110-111 adverse effects of avascular necrosis, 107 bone loss, 107, 722 cardiovascular system, 109 cataracts, 109 cell-mediated immune reactions, 108 central nervous system, 108-109 coronary artery disease, 109 Cushing's syndrome, 106t, 106-107, 236, 430 dyslipidemia, 109 glaucoma, 109 growth suppression, 107, 218, 236, 240 hematologic system, 108 immunity, 108 infection, 108 muscle wasting, 109 myopathy, 109, 430 osteoporosis, 107, 108f, 723 overview of, 106t psychoses, 108-109 anti-inflammatory actions of, 105 behavioral effects of, 111-112 carbohydrate metabolism and, 104 carditis treated with, 623 Cogan's syndrome treated With, 518 description of, 102-103 diffuse cutaneous systemic scleroderma treated with, 459 dose-effect relationships of, 104t dosing of description of, 103t reductions In, 110 tapering of, 110, 377-378 high-dose intravenous, 111-112 hypothalamic-pituitary-adrenal axis suppression caused by, III immunizations and, 200 immunosuppressive actions of, 105 indications for, 105-106 intra-articular adverse effects of, 112-113 description of, 112 dosage of, 112 frequency of, 112 juvenile rheumatoid arthritis treated with, 236-237 oligoarthritis treated With, 285 triamcinolone hexacetonide, 112 types of, 112 intravenous, 236 juvenile ankylosing spondylitis treated with, 319 juvenile dermatomyositis treated with, 429-430 juvenile psoriatic arthritis treated with, 330 juvenile rheumatoid arthritis treated with, 235-237 Kawasaki disease treated with, 533-534 lipid metabolism and, 104 maintenance of, 378 pharmacologic effects of, 103-104 pharmacology of, 103, 103f physiologic effects of, 103-104, lOSt polyarteritis nodosa treated With, 515 polyarthritis treated with, 270 presurgical supplementation of, III production of, 59--60 protein metabolism and, 104 reactive arthritis treated with, 611 sarcoidosis treated with, 555 short-acting, 110
INDEX structure of, 103f systemic, 110t, 235-236 systemic lupus erythematosus treated with, 376-378 T-cells affected by, 105 Takayasu's arteritis treated with, 550 toxicity caused by, 109-110 uveitis treated with, 292 Wegener's granulomatosis treated with, 542-543 Glucocorticoid-responsive elements, 104 Glucose-6-phosphatase deficiency, 635 Glycolipids, 24 Glycosaminoglycans description of, 11 synovial fluid levels of, 225-226 Gold compounds adverse effects of, 100 description of, 99 intramuscular administration of, 100 juvenile rheumatoid arthritis treated with, 239-240 oral administration of, 101 pharmacology of, 99-100 structure of, 100f toxicity of, 101 Goldbloom syndrome, 638 Golfer's elbow, 682 Gonadal immaturity,S Gonococcal arthritis, 574 Goodness-of-fit chi-square test, 164 Goodpasture's syndrome, 51, 504-505 Gorham's disease, 760 Gottron's papules, 412, 423 Gout, 633, 634t Gouty arthropathy, 633 Gowers' sign, 411 gp130, 57, 292 Graft-versus-host disease autoimmune hematologic phenomena secondary to, 648 chronic, 453-454 diffuse cutaneous systemic scleroderma vs., 453-454, 454t Granzymes, 39 Graves' disease, 51 Griseofulvin, 460 Group A streptococcus, 615-616 Group sequential designs, 153--154 Growing pains, 692--693 Growth chronic arthritis effects on, 218--219 glucocorticoid-related suppression of, 107, 218,240 juvenile rheumatoid arthritis effects on, 218--219, 227~ 227-228 Growth factors, 57 Growth hormone, for juvenile rheumatoid arthritis, 240 Growth-regulated oncogene-a, 444 Guanethidine, 461t Guttate morphea, 472
H Haemophilus influenzae, 569 Half-life, 76, 150 Haplotypes, 66 Haptens,40 Hashimoto's thyroiditis, 360, 637 Haversian canals, 716 Health-related quality of life, 174 Heat and cold therapy, 197-198 Heat-shock proteins
description of, 22, 33 in juvenile rheumatoid arthritis, 213 in oligoarthritis, 277 Heliotrope discoloration, 41Of, 412, 423 Helper T-cells description of, 31 differentiation of, 37-38 effector functions of, 37 function of, 28 Th1 cytokines produced by, 38 description of, 31, 37, 119 development of, 38f effector functions of, 38--39 interleukin-4 effects on, 37 interleukin-10 effects on, 37 macrophage recruitment by, 38 Th2 anti-inflammatory cytokines released by, 39 cytokines produced by, 38 description of, 31, 37 development of, 38f effector functions of, 38--39 reactive, 119 Hemarthrosis, 636 Hematologic system methotrexate-related tOXicities, 94 neonatal lupus erythematosus manifestations in, 398--399 Hematophagocytic lymphohistiocytosis, 295 Hematoxylin bodies, 360 Hematuria, 222 Hemifacial atrophy, 472--473 Hemochromatosis, 637 Hemoglobinopathies, 635 Hemophilia, 635-636 Henoch-Schonlein purpura in adults, 500 anaphylactoid purpura, 496 arthritis associated with, 217, 498--499 classification of, 496 clinical manifestations of, 497--499 clinicopathologic characteristics of, 494t course of, 501 defmition of, 493t, 496 diagnostic criteria for, 500, SOOt differential diagnosis, 500, SOOt epidemiology of, 495, 495t, 496 in familial Mediterranean fever, 505 features of, 498t gastrointestinal disease in, 497 genetic background of, 496--497 glomerulonephritis in, 497-498 histopathologic fmdings, 499f, 499-500 19A-containing immune complexes in, 500 laboratory examination in, 500 leukocytoclastic vasculitis in, 499f pathologic findings, 499f, 499-500 prognosis for, 501 radiologic examination in, SOD-SOl renal disease in, 497--498, 501 skin manifestations of, 497, 498f--499f treatment of, 501 Hepatitis, 84 Hepatitis B arthritis-dermatitis syndrome, 582 Hepatomegaly juvenile rheumatoid arthritis and, 221-222 progressive, 296 systemic arthritis and, 296 systemic lupus erythematosus and, 360 Hereditary angioneurotic edema, 25 Hereditary osteolysis, 746t Herpesviruses, 583
775
Heterotopic inhibition, 700 High endothelium venules, 40-41, 41£ High mobility group box chromosomal protein 1, 57 High-dose immunotherapy with transplantation, 126 High-dose intravenous glucocorticoids, 111-112 Hip exercises, 193f Hip joint acute chondrolysis of, 688 juvenile rheumatoid arthritis effects, 230 septic arthritis of, 572f, 573--574 transient synOVitis of, 583, 583t Histocompatibility antigens, 344 HLA-B27 description of, 64, 68, 215 juvenile ankylosing spondylitis and, 308, 314-315 juvenile psoriatic arthritis and, 326 oligoarthritis and, 275 post-streptococcal reactive arthritis and, 625 reactive arthritis and, 604--606 HLA-B51, 562 Homocystinuria, 677t, 747t, 757 Hotelling's T' test, 170 Howell-Jolly bodies, 360 Human Genome Project, 64, 66 Human immunodeficiency virus, 583 Human leukocyte antigens description of, 32 juvenile dermatomyositis and, 410-411 juvenile psoriatic arthritis and, 326 neonatal lupus erythematosus and, 394 oligoarthritis and, 275-276, 276t polyarthritis and, 262 Human pharmacology study, 151t Hunter's syndrome, 747t Hurler's syndrome, 747t, 758t Hyaline cartilage anatomy of, 9-10 composition of, 9-10 Hyaluronan, 11 Hyaluronic acid, 14 Hydrocortisone dosing of, 103t during stress, 111 structure of, 103f Hydroxyapatite, 716 Hydroxychloroquine sulfate description of, 96, 96f juvenile dermatomyositis treated with, 429t, 430-431 juvenile rheumatoid arthritis treated with, 239 systemic lupus erythematosus treated with, 376 7-Hydroxymethotrexate, 90 Hydroxyproline, 717t Hyperalgesia, 698 Hypergammaglobulinemia, 223 Hyperimmunoglobulinemia D with periodic fever syndrome clinical manifestations of, 660t, 663 description of, 659, 663 diagnosis of, 665 erythematous macules associated with, 663 genetics of, 663 laboratory studies, 663 maculopapular rash associated With, 663, 665f pathogenesis of, 663 treatment of, 665
776
INDEX
Hyperlipoproteinemia, 635 Hypermobility in athletes, 678 benign hypermobility syndrome, 676 conditions associated with, 677t, 748t diagnostic criteria for, 678t genu recurvatum, 679 ligamentous laxity and, 745-746 osteoarthritis associated with, 678 pain associated with, 676--680 pes cavus, 678-679 pes planus, 678-679 recurrent patellar dislocation secondary to, 679 Hyperostosis cortical, 761 description of, 638 Hyperparathyroidism, 637 Hyperpyrexia, 293 Hypersensitivity delayed-type, 52 immediate, 50 nonsteroidal anti-inflammatory drug-related, 86-87 type I, 50, 51t type II, 50-51, 51£, 51t type III, 51, 51t type IV, 51t, 52 Hypersensitivity angiitis, 494t, 501-503 Hyperthyroidism, 637 Hypertrophic osteoarthropathy description of, 335, 638 secondary, 741 Hyperuricemia, 633-635, 634t Hypervitaminosis A, 631 Hypochondroplasia, 747, 747t Hypocomplementemic urticarial vasculitis, 503-504 Hypogammaglobulinemia characteristics of, 651 t dermatomyositis-like syndrome associated with, 652-653, 653t description of, 94, 650 X-linked agammaglobulinemia, 650-651 Hypomobility, 679t, 679-680 Hypophosphatasia, 631 Hypophosphatemic vitamin D-resistant rickets, 630 Hypothalamic-pituitary-adrenal axis description of, 59 glucocorticoids effect on, 111 Hypotheses, 143-144 Hypothesis-generating studies, 143 Hypothesis-testing studies, 143 Hypothyroidism, 637 Hypoxanthine-guanine phosphoribosyltransferase, 634
I Ibuprofen dosing of, 81t indications for, 81 juvenile psoriatic arthritis treated with, 330 juvenile rheumatoid arthritis treated with, 233 structure of, 77f toxicities associated with, 83t I-cell disease, 747t, 758t, 759 Idiotopes, 30 Idiotype, 30 ling, 37 Immediate hypersensitivity, 50
Immune complex vasculitis illustration of, 361f juvenile dermatomyositis and, 409 Immune complexes C1 binding to, 645 in Henoch-Sch6nlein purpura, 500 19A-containing, 500 juvenile rheumatoid arthritis and, 211, 225 in systemic lupus erythematosus, 346, 371 Immune response adaptive, 28 effectors of, 20, 20f Immune system genetic disorders of, 642 innate, 211 juvenile rheumatoid arthritis pathogenesis and, 211 Immunity adaptive. See Adaptive immunity antibody-mediated, 42 glucocorticoids effect on, 108 innate. See Innate immunity Immunizations, 199-200 Immunodeficiency disorders classification of, 643t common variable immunodeficiency, 649, 651-653 hypogammaglobulinemia characteristics of, 651t description of, 650 X-linked agammaglobulinemia, 650-651 primary humoral, 648-653 severe combined immunodeficiency, 647-648 T-cells, 648 Wiskott-Aldrich syndrome, 647t, 648 Immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome, 47 Immunogens, 45 Immunoglobulin(s) antigen binding to, 31 Fc portion of, 42 functions of, 28 heavy chains, 28 hypervariable regions of, 30 in juvenile rheumatoid arthritis, 223-224 light chains, 28 membrane, 30 structure of, 28, 29f surface, 39--40 types of, 28, 30t Immunoglobulin G subclass deficiencies, 653 Immunoglobulin M, 211 Immunologic memory, 42-43 Immunologic tolerance, 45 Immunomodulators diffuse cutaneous systemic scleroderma treated with, 459--460 juvenile rheumatoid arthritis treated with, 238 sarcoidosis treated with, 555-556 Immunoreceptor tyrosine activation motifs, 30 Immunosuppression bystander, 48 immunization considerations in children undergoing, 200 Immunosuppressive drugs juvenile dermatomyositis treated with, 429t, 431 juvenile rheumatoid arthritis treated with, 238 systemic lupus erythematosus treated With, 378-379 Immunotherapy high-dose, 126 transplantation and, 126
Incidence, 145, 146t Independent variable, 161 Individualized education plans, 187 Indomethacin bronchospasm induced by, 86 central nervous system toxicities, 85 dosing of, 82t indications for, 81 juvenile psoriatic arthritis treated with, 330 juvenile rheumatoid arthritis treated with, 233 mechanism of action, 79 structure of, 77f toxicities associated With, 83t Inducible costimulator, 34 Inducible nitric oxide synthase, 22, 22f, 59 Infantile acute hemorrhagic edema, 500 Infantile myofibromatosis, 455 Infantile polyarteritis nodosa, 521 Infantile systemic hyalinosis, 732 Infections autoimmunity and, 49-50 glucocorticoid-related, 108 juvenile psoriatic arthritis and, 325 juvenile rheumatoid arthritis pathogenesis and, 213 methotrexate and, 94 musculoskeletal manifestations of, 585 myositis after, 421 periodic fever syndromes caused by, 657-658 systemic lupus erythematosus and, 348, 376, 381 Infectious arthritis Bartonella henselae, 575 brucellosis, 575 frequency of, 4t gonococcal arthritis, 574 Mycoplasma pneumoniae, 575 septic arthritis. See Septic arthritis tuberculous arthritis, 574, 574f-575f Infective endocarditis, 585 Inflammation cyclooxygenase-2 released secondary to, 699 cyrokines involved in, 23-24 endocrine mediators in, 59-60 growth factors secreted in, 57 hyperalgesia in, 699 hypothalamic-pituitary-adrenal axis activation in, 59 inducible nitric oxide synthase in, 59 markers of, 267, 367 in polyarthritis, 267 sacroiliac joint, 316 in systemic lupus erythematosus, 367 tumor necrosis factor-u's role in, 56 Inflammatory bowel disease. See also Crohn's disease; Ulcerative colitis age at onset, 334 classification of, 334 clinical manifestations of arthritis, 335, 335t erythema nodosum, 336, 336f gastrointestinal disease, 335-336 hypertrophic osteoarthropathy, 335 mucocutaneous lesions, 336 osteoporosis, 335 pyoderma gangrenosum, 336, 337f sacroiliac arthritiS, 335 skin, 336-337 uveitis, 337 vasculitis, 337 course of, 338 definition of, 334
INDEX di~gnosis of, 337 epidemiology of, 334 etiology of, 334 geJllder ratio of, 334 geJlletic background of, 334-335 histopathologic findings, 337 HLA.-B27-associated spondylitis of, 338 inddence of, 334 joint involvement in, 334 la~ratory examination of, 337 pathogenesis of, 334 pathologic findings, 337 prevalence of, 334 pro~nosis for, 338 radiplogic examination of, 337, 338f treatment of, 337-338 Inflammatory chemokines, 54 Inflixirtlab desqription of, 122-123 juvenile dermatomyositis treated with, 431-432 juvellile rheumatoid arthritis treated with, 237-238 Inhibit~ry regulatory T-eells, 46 Innate Immunity cells involved in dendritic, 23 fibroblasts, 23-24 natural killer, 23, 24f phagocytes. See Innate immunity, J!>hagocytes in circulating proteins involved in, 26 complement system, 24-26 eytok,nes of, 26, 27t definition of, 19 disorders of, 643t phagOCytes in acti~ation of, 20-22 alternative activation of, 22-23 description of, 19 effector functions, 20-22 maqrophages, 19, 20f microbe binding to, 22 m0'locytes, 19, 20f neutrophils, 19-20 sUrf~ce receptors, 20 Toll1like receptors, 21-22, 22t Insulin-li\<e growth factors, 722 Integrins, 54t, 445 Interferom-a, 74 Interferon-j3, 74 Interferon-y description of, 23, 38, 74 recomqinant, for juvenile rheumatoid arthritis, 238 Interferons, 27t Interleukih-l biologiC agents against, 7 functiot:ls of, 56 glucocqrticoids effect on, 105 isoform$ of, 56 in phagPcytosis, 23 sulfasalazine effects on, 97 Interleukil)-1 receptor antagonist, 124 Interleukil)-l receptor-associated kinase, 57 Interleukirl-1a description of, 27t, 56 in polya/thritis, 261 Interleukine1~ description of, 27t, 56-57 oligoarthil'itis and, 276 Interleukin.lA2, 215 Interleukin.1Ra, 23
InterJeukin-2 characteristics of, 27t description of, 44 T-cell activation effects on, 35 InterJeukin-4, 27t Interleukin-5, 27t, 38 Interleukin-6 in adaptive immunity, 57 anti-IL6 receptor antibody, 124-125, 299-300 description of, 27t, 42 in polyarthritis, 261-262 pro-inflammatory functions of, 57 soluble receptor for, 291-292 in systemic arthritiS, 291 Interleukin-IO description of, 23, 27t, 37, 47 in oligoarthritis, 276 Interleukin-12, 27t, 37 InterJeukin-13, 27t Interleukin-15, 27t Interleukin-18, 27t Interleukin-23, 27t, 37 Internal validity, 158 International League of Associations for Rheumatology juvenile idiopathic arthritis classification criteria, 207-208, 274 oligoarthritis classification, 275t polyarthritis defined by, 261 Internet resources, 186 Interquartile range, 161 Inter-rater agreement, 169 Interstitial nephritis, 365 Intervertebral disk herniation, 691 Interview bias, 159 Intra-articular glucocorticoids adverse effects of, 112-113 description of, 112 dosage of, 112 frequency of, 112 triamcinolone hexacetonide, 112 types of, 112 Intraclass correlation, 169 Intra-rater agreement, 169 Intravenous immunoglobulin description of, 118-119, 119t juvenile dermatomyositis treated with, 429t, 431 juvenile rheumatoid arthritis treated with, 238 Kawasaki disease treated with, 531-533, 533t polyarteritis nodosa treated with, 515-516 systemic arthritis treated With, 299 systemic lupus erythematosus treated With, 379-380 Investigational new drug, 149 Iritis, 310 Isotype switch, 31 Ixodes scapularis, 592, 593f
J Jaccoud's arthritiS, 624 Jansen's dysostosis, 747t Jarisch-Herxheimer reaction, 599 JAS. See Juvenile ankylosing spondylitis Job's syndrome, 644 JointCs) apophyseal, 217 cartilaginous, 9, lOt classification of, 9, lOt diarthrodial, 9
777
fibrous, 9, lOt inflammation of, 217 polyarthritis manifestations, 262-263, 263f stiff, 754-755 synovial, 9, lOt vascular supply to, 14-15 Joint contractures, 745 JPsA. See Juvenile psoriatic arthritis JRA. See Juvenile rheumatoid arthritis Juvenile ankylosing spondylitis age at onset, 308 antiphospholipid antibodies in, 314 characteristics of, 306t classification of, 304 clinical manifestations of apophyseal joints, 311 arthritis, 310 cardiopulmonary disease, 310-311 central nervous system, 311 description of, 309 enthesitis, 309-311, 319 iritiS, 310 pleuropulmonary disease, 310-311 renal disease, 311 course of, 319-320 definition of, 304 diagnostic criteria for, 304-306, 305t differential diagnosis, 311-312 entheses in, 317, 318f epidemiology of, 307-308 etiology of, 308 gender ratio for, 308 genetics of, 308-309 geographic distribution of, 308 histopathologic findings in, 311 HLA-B27 and, 308, 309t, 314-315 incidence of, 307t, 307-308 juvenile rheumatoid arthritis vs., 313t laboratory examination in, 314-315 musculoskeletal examination in axial skeleton, 313-314 entheses, 312, 312f peripheral joints, 312-313, 317, 318f musculoskeletal signs and symptoms in, 3lOt occupational therapy for, 319 outcome measures for, 320 pathogenesis of, 308 pathologic findings in, 311 peripheral arthropathy in, 312 physical therapy for, 319 prevalence of, 307-308 prognosis for, 320 racial distribution of, 308 radiologic characteristics of, 315f-317f, 315-317 range of motion limitations secondary to, 319-320 selective IgA deficiency and, 314 seronegative enthesitis and arthritis syndrome and, 304-306, 307t surgery for, 319 treatment of etanercept, 319 general approach to, 317-318 glucocorticoids, 319 individualized approach to, 318 methotrexate, 319 naproxen, 319 nonsteroidal anti-inflammatory drugs, 318-319 sulfasalazine, 319 Juvenile Arthritis Functional Assessment Scale and Report, 177t, 178
788
INDEX
Juvenile Arthritis Quality of Life Questionnaire, 177t, 179, 181 .Juvenile Arthritis Self-Report Index, 177t, 178-179 Juvenile chronic arthritis. See also Chronic arthritis diagnostic criteria for, 206-207, 207t European League Against Rheumatism classification criteria for, 206-207, 207t pauciarticular, 286 polyarticular, 263, 271 Juvenile dermatomyositis age at onset, 407-408, 409f arteries in, 419 arthritis in, 412 blood vessels in, 418-419 calcinosis in course of, 433 description of, 413, 416f treatment of, 413, 416f capillaries in, 419 cardiac findings, 420 cardiopulmonary disease in, 413-414 classification of, 407 clinical manifestations of, 411-414 course of, 432-434 deaths from, 434 definition of, 407 diagnostic criteria for, 407, 407t differential diagnosis, 420-424 dual-energy x-ray absorptiometry evaluations, 428 dystrophic calcification in, 41St electromyography of, 427, 427t endocrinopathies vs., 422 environmental factors associated with, 410 epidemiology of, 407-408 erythema associated With, 349 etiology of, 408-410, 409t familial patterns of, 410 features of, 432t frequency of, 6t functional disability in, 433 gastrointestinal tract findings, 420 gender ratio for, 407-408 genetic background of, 410-411 geographic distribution of, 408 Gottron's papules and, 412, 423 heliotrope discoloration associated with, 410f, 412, 423 historical review of, 407 human leukocyte antigens and, 410-411 immune complex-mediated vasculitis associated with, 409 immunoglobulins in, 416, 418 incidence of, 407, 408t interleukin-1 expression in, 409 juvenile polymyositis vs., 420-421 laboratory examination for aldolase, 426 anti-UlRNP, 425 autoantibodies, 424-426 creatine kinase, 426 creatine/creatinine ratio, 424 endothelial cell antigen antibodies, 425-426 general findings, 424 lactate dehydrogenase, 426-427 muscle enzymes, 426-427 myositis-associated antibodies, 424-425 myositis-specific antibodies, 424-425 transaminases, 426 lipodystrophy in description of, 414, 418f management of, 432
magnetic resonance imaging of, 427-428 maternal cell chimerism in, 409 mortality from, 434 mucocutaneous disease associated With, 412-413 muscle biopsy findings, 427 muscle histopathology findings in, 418t muscular dystrophy vs., 421 musculoskeletal disease associated with, 411-412 myoadenylate deaminase deficiency vs., 421-422 neuromuscular diseases and, 421-422 osteoporosis in, 427, 428f pathogenesis of, 408-410, 409t pathologic findings in, 414-420 patient counseling for, 429 periungual erythema in, 412, 412f-413f, 423 phases of, 432t polyarthritis and, 265 postinfectious myositis vs., 421 prognosis for, 432-433, 433t psychosocial outcome of, 433-434 racial distribution of, 408 radiologic examination in, 427-428 Raynaud's phenomenon in, 414 renal findings, 420 signs and symptoms of, 411 skeletal muscle findings in, 415-416, 418, 418f skin manifestations of, 412-413, 419 Toxoplasmagondii and, 410 treatment of biologic agents, 429t, 431-432 cyclosporine, 431 etanercept, 429t general supportive care, 429 glucocorticoids, 429-430 hydrochloroquine, 429t, 430-431 immunosuppressive drugs, 429t, 431 infliximab, 431-432 intravenous immunoglobulin, 429t, 431 methotrexate, 431 overview of, 428 physiotherapy, 432 plasmapheresis, 431 prednisone, 429 stem cell transplantation, 431-432 tumor necrosis factor-a inhibitors, 431 ultrasound findings in, 428f vasculitis in, 413 veins in, 419 Juvenile fibromatosis, 732 Juvenile gastrointestinal polyposis, 506 Juvenile hyaline fibromatosis, 732 Juvenile idiopathic arthritis combination therapies for, 125t diagnostic criteria for, 207t, 207-208 illustration of, 106f infliximab for, 123t International League of Associations for Rheumatology classification criteria for, 207-208, 274 juvenile psoriatic arthritis vs., 327 oligoarticular, 278 systemic. See Systemic arthritis Juvenile polymyositis, 420-421 Juvenile psoriatic arthritis age at onset, 324 amyloidosis and, 331 angiogenesis in, 326 CD8+ T-cells and, 325-326 chronic arthritis caused by, 324 classification of, 324
clinical manifestations of arthritis, 327 enthesitis, 328 extra-articular, 328-329 nail changes, 328, 328f rash, 328, 328f skin disease, 328, 328f uveitis, 328-329 course of, 331 cyclooxygenase-2 inhibitors for, 330 cytokines' role in, 326 dactylitis in, 327f definition of, 324 diagnostic criteria for, 324, 325t epidemiology of, 324--325, 325t etiology of, 325-326 gender ratio for, 324--325 genetics of, 326-327 glucocorticoids for, 330 histopathologic findings in, 329 HLA-B27 and, 326 incidence of, 324 joints affected in, 327t juvenile idiopathic arthritis vs., 327 juvenile rheumatoid arthritis vs., 327 laboratory examination for, 329 methotrexate for, 330-331 naproxen, 330 nonsteroidal anti-inflammatory drugs for, 329-330 pathogenesis of, 325-326 pathologic findings in, 329 prevalence of, 324 prognosis for, 331 radiologic features of, 329, 330f studies of, 326t treatment of, 329-331 viral infections associated with, 325 Juvenile rheumatoid arthritis age at onset, 210 American College of Rheumatology criteria for, 206, 207t amyloidosis and, 244 animal models of, 214 ankle abnormalities, 229 arthrography evaluations, 226 atlantoaxial subluxation associated With, 230, 231£ autoimmune diseases associated with, 71, 216 cardiac disease associated with, 221 cartilage destruction associated With, 228, 228f chromosomal abnormalities associated with, 216 classification of, 206 clinical manifestations of, 216-220 complex genetic traits, 67-68 counseling, 242 course of, 242-244 criteria for, 206, 207t deaths caused by, 244 description of, 206 diabetes mellitus and, 216 diagnostic criteria for, 206, 207t Down's syndrome and, 216 in elbow, 228 endocarditis and, 221 epidemiology of, 209-211 etiology of, 211 extra-articular manifestations of, 218-220 facial morphology abnormalities associated with, 219, 219f family counseling, 242
INDEX
fanilly history of, 64, 213 foot abnormalities, 229 footwear for, 241 frequency of, 4t functional disability secondary to, 242-244 functional status measurement instruments, 177-180 ga$trointestinal tract manifestations of, 221 gehder rates for, 210 genetics of family background, 213 human leukocyte antigen relationships, 214-215 geographic distribution of, 210-211 gr(Jwth and development abnormalities associated with, 218-219, 227f, 227-228 m hands, 228, 228f hepatic disease associated with, 221-222 hip abnormalities, 230 histopathologic findings, 222f, 222-223 historical review of, 208-209 hypergammaglobulinemia and, 223-224 incidence of, 2091, 209-210, 307t influenza virus A2H2N2 and, 213 insulin-dependent diabetes mellitus and, 216 joints affected, 217 apophyseal, 217 inflammation of, 217 pain m, 216, 241 subluxation of, 227 ju,venile ankylosmg spondylitis vs., 313t jl.lvenile psoriatic arthritis vs., 327 knee abnormalities, 229 laboratory studies for antinuclear antibodies, 224-225 blood mdices, 223 C3,225 complement, 225 C-reactive protein, 223 description of, 223 erythrocyte sedimentation rate, 223 immune complexes, 225 immunoglobulins, 223-224 lipids, 225 plasma lipids, 225 rheumatoid factors, 224 synovial flUid analysis, 225-226 leg-length mequality secondary to, 219 leukocytosis associated with, 223 lymphadenopathy associated with, 221 lymphedema associated with, 220 macrophage inhibitory factor in, 211 magnetic resonance imagmg of, 226 micrognathia associated with, 219, 219f mortality caused by, 244 muscle disease associated with, 220-221 myocarditis and, 221 neurologic disease associated with, 222 riutrition for, 240 occupational therapy for, 240-241 oligoarticular characteristics of, 207t description of, 206 hip jomt arthritis associated with, 278 human leukocyte antigens associated with,276 onset of, 206 osteopenia and, 219-220 osteoporosis associated with, 226 0utcomes of, 242-244, 243t pain associated with, 216-217 pathogenesis of
autoantibodies, 212 cytokines, 211-212 description of, 211 hormones, 212-213 immune complexes, 211 irnmunopathogenic mechanisms, 211 infections, 213 peripheral blood mononuclear cells, 212 psychologic factors, 213 studies regarding, 213 T-cells, 211, 222 T-cell receptor polymorphism, 212 trauma, 213 pathologic findings, 222f, 222-223 pauciarticular description of, 69, 74, 274 joint erosion associated with, 286 pericarditis and, 221 physical therapy for, 240-241 pleuropulmonary disease and, 221 polyarticular cervical spine arthritis vs., 311 characteristics of, 207t description of, 206 outcome for, 243t radiographic fmdings, 234f soft tissue swelling associated with, 228 postvaccination, 213 prevalence of, 209t, 210 prognosis for, 242-244 psychosocial outcome of, 242-244 racial distribution of, 210-211 radiologic examination in imaging techniques for, 226 overview of, 226-228 subluxation findings, 227 radionuclide imaging in, 226 reconstructive surgery for, 242 renal disease associated with, 222 m shoulder, 228-229 signs and symptoms of, 216 skin manifestations of, 220, 220f slipped capital femoral epiphysis vs., 278 soft tissue surgery for, 242 spmal abnormalities, 230f, 230-231 splenomegaly associated with, 221 subcutaneous tissue manifestations of, 220, 220f subtalar jomt abnormalities, 229 synovial cysts associated with, 217, 217f-218f systemic characteristics of, 207t clinical features of, 208f description of, 206 technetium 99m imaging of, 226 temporomandibular jomt in, 219, 229, 229f tenosynovitis associated with, 218 transaminases in, 84 treatment of anakinra, 238 analgesics, 234 antimalarials, 239 approaches to, 231-232 aspirin, 233-234 autologous stem cell transplantation, 240 azathioprine, 114 basic program for, 232-233 biologic response modifiers, 237-238 chlorambucil, 238 COX-2 mhibitors, 233 cyclosporine, 118, 238 cytotoxic drugs, 238
779
disease-modifying antirheumatic drugs, 88, 238-239 etanercept, 237 glucocorticoids, 235-237 goals of, 231-232 gold compounds, 239-240 growth hormone, 240 hydroxychloroqume, 239 ibuprofen, 233 immune modulators, 238 immunosuppressive drugs, 238 indomethacin, 233 infliximab, 237-238 intravenous immunoglobulin, 238 leflunomide, 991 methotrexate, 89-92, 911, 232, 234-235 methylprednisolone, 236 naproxen, 233 nonsteroidal anti-inflammatory drugs, 83, 232-234 objectives of, 231t, 231-232 orthopedic surgery, 241-242 D-penicillamine, 240 prednisone, 235-236 recombinant interferon-'t, 238 slow-acting antirheumatic drugs, 238-239 sulfasalazine, 239 synovectomy, 241 tolmetin sodium, 233 triamcinolone hexacetonide, 236 tumor necrosis factor-a, 237 urinary tract abnormalities associated with, 222 uveitis associated with antinuclear antibodies in, 281-282 band keratopathy, 280, 282f, 283 clinical manifestations of, 279-280 course of, 283 description of, 278 differential diagnosis, 281 epidemiology of, 279, 279t etiology of, 279 genetics of, 279 glucocorticoids for, 282 histopathology of, 280-281 laboratory examination of, 281-282 management of, 282-283 monitoring of, 2811 pathogenesis of, 279 pathology of, 280-281 prognosis of, 283 slit-lamp biomicroscopy of, 280, 281£ vasculitis associated with, 220, 220f in wrist, 228 Juvenile temporal arteritis, 551
K Kaplan-Meier survival analysis, 169 Kappa test ratio, 169 Kashin-Beck disease, 632, 633t Kawasaki disease antineutrophil cytoplasmic antibodies in, 522,530 atypical, 525 cardiac status monitoring in, 534-536 cardiovascular disease and, 526-527 central nervous system complications, 527 cerebrospmal fluid analysis in, 530 clinicopathologic characteristics of, 494t conjunctivitis and, 524 coronary artery aneurysms and, 526, 527f course of, 523, 524f, 536
780
INDEX
Kawasaki disease (Continued) cutaneous manifestations of, 524-525, 525f definition of, 493t, 521 diagnostic criteria for, 521, 522t differential diagnosis, 529, 529t discovery of, 3 epidemiology of, 495, 495t, 521-522 etiology of, 522-523 extremiry changes, 525, 526f fever and, 523-524 follow-up for, 535t gastrointestinal tract disease in, 528 genetic background of, 523 genitourinary tract involvement in, 528-529 giant aneurysms and, 535 histopathologic findings, 529 historical background of, 521 incidence of, 521-522 intravenous immunoglobulin for, 119 laboratory examination for, 529-530 lymphadenopathy and, 525 mucosal changes associated with, 524 musculoskeletal disease and, 528 ocular disease associated with, 528, 528t pathogenesis of, 522-523 pathologic findings, 529 phases of, 523, 524f prevalence of, 521-522 prognosis for, 536 relapses of, 534 respiratory tract disease in, 528 salicylates for, 87 treatment of algorithm for, 532f aspirin, 531 general approach to, 530, 530t glucocorticoids, 533-534 goals for, 530--531 intravenous immunoglobulin, 531-533, 533t methylprednisolone, 534 pentoxifylline, 534 prednisone, 533 uveitis and, 528 Kendall's W, 169 Keratoconjunctivitis sicca, 451 Keratocytes, 400 Keratoderma blennorrhagicum, 607, 608f, 611 Ketoprofen dosing of, 81t structure of, 77f Kidneys aCllte interstitial nephritis with nephrotic syndrome, 85 juvenile ankylosing spondylitis findings, 311 nonsteroidal anti-inflammatory drug-related toxicities, 84-85 papillary necrosis of, 85 Kikuchi's disease, 296, 360 Klinefelter's syndrome,S Knee exercises for, 193f sagittal section of, lOf Knee pain, 678 Kniest's syndrome, 745, 751, 751f Koch's postulates, 145 Koebner phenomenon, 294 Kohler's disease, 685, 686f Kohlmeier-Degos syndrome, 506 Kozlowski's syndrome, 747t K-sample tests, 167
Kupffer cells, 19 Kurtosis, 160 Kveim-Siltzbach skin test, 555
L La protein, 392 Laboratory research, 143 Lactate dehydrogenase, 426--427 Laminin, 12 Langer-Gledion syndrome, 755 Langerhans cell histiocytosis, 735-736 Larsen's syndrome, 761 Last-observation-carried-forward approach, 157 Lectin pathway, 24, 25f Leflunomide description of, 98-99 polyarthritis treated with, 269 Leg exercises, 194f Legg-Calve-Perthes disease, 684, 685f Leg-length inequality in juvenile rheumatoid arthritis, 219 management of, 285 in oligoarthritis, 285 Leptin,432 Leri pleonosteosis, 747t Leri-Weill dyschondrosteosis, 747t Leri-Weill dysostosis, 755, 755f Lesch-Nyhan syndrome, 633-634, 634t Leukemia, 739-741 Leukocyte(s) in autoimmune diseases, 52, 54-55 inflammatory site recruitment of, 53f migration of, 52 Leukocyte adhesion deficiency type 1, 52 type 2, 52 Leukocyte function-associated antigen-1 description of, 52 in Lyme disease, 593 Leukocytoclastic vasculitis conditions associated with, 497t definition of, 496 in Henoch-Schonlein purpura. See HenochSchonlein purpura histopathologic findings in, 496, 497f historical descriptions of, 3 Leukotriene(s) A4,58 B4, 58 definition of, 58 synthesis of, 78f Libman-Sacks endocarditis, 355, 366, 366f, 622 Ligamentous laxity, 745-746 Ligaments, 15 Limited cutaneous systemic scleroderma, 463-464, 464f-465f Linear morphea, 475-476 Linear scleroderma, 472-473, 473f Lipocortins, 105 Lipodystrophy description of, 414, 418f management of, 432 Lipogranulomatosis subcutanea of RothmannMakai,455 Lipoid dermatoarthritis, 639 Lipopolysaccharide-binding protein, 26 Lipoxins, 58 5-Lipoxygenases, 58 Little league elbow, 682 Little league shoulder, 682 Livedo reticularis, 350--351, 517
Liver cirrhosis of, 92 methotrexate-related toxicity, 92-94 neonatal lupus erythematosus-related dysfunction of, 398 LJP-394, 120 Localized scleroderma bullous morphea, 472 characteristics Of, 472 clinical manifestations of, 475-476 course of, 478-479 deep morphea, 473-474 differential diagnosis, 477 epidemiology of, 474-475 etiology of, 475 generalized morphea, 472 immunologic abnormalities in, 478t incidence of, 474-475 laboratory examination of, 477-478 linear scleroderma, 472-473, 473f pathogenesis of, 475 pathologic findings in, 476--477 plaque morphea, 472, 473f prognosis for, 478-479 treatment of, 478 Locus ceruleus, 698 Long-term depression, 699 Long-term potentiation, 699 Lower leg exercises, 194f L-selectin, 43, 52, 54t Lubricin, 11 Lupoid hepatitis, 360 Lupus Activity Index, 372, 373t Lupus anticoagulants, 368 Lupus crisis, 358 Lupus erythematosus discoid, 643 neonatal. See Neonatal lupus erythematosus systemic. See Systemic lupus erythematosus Lupus erythematosus cell, 369, 369f Lupus nephritis. See also Glomerulonephritis characteristics of, 352-353, 353t, 361t, 372 end-stage, 38lt treatment of, 377t, 380--381 Luteinizing hormone-releasing hormone, 116 Lyme disease in acrodermatitis chronic atrophicans, 593-594 age at onset, 591 antibiotics for, 598t, 598-599 arthritis associated with, 597, 597t
Borrelia bur,gdorferi avoidance of, 599--600 description of, 475, 592 immunizations against, 600 laboratory detection of, 595-597 classification of, 591 clinical manifestations of, 593-595, 594t course of, 600 cutaneous manifestations of, 594 definition of, 591 description of, 584 diagnosis of, 597-598 epidemiology of, 591 erythema mlgrans associated with, 594, 594f, 598-599 etiology of, 592 gender ratio for, 591 genetic background of, 592 geographic distribution of, 591 history of, 591 immunizations against, 600 immunoglobulin G responses in, 596
INDEX ~munoglobulin M responses in, 596
in¢idence of, 591 lxlJdes scapulaTis and, 592, 593f laboratory examination for, 595-597 lel1k ocyte function-associated antigen-1 in, 593 microbiologic findings in, 592 musculoskeletal disease associated with, 594-595 n¢uroborreliosis in, 600 ocular involvement in, 595 p~thogenesis of, 592-593 pathologic findings of, 595 Prevalence of, 591 ptevention of, 599--600 ptognosis for, 600 s~rologic tests for, 597 skin manifestations of, 594 synovial fluid analysis in, 598 treatment of, 598-599 Lymphadenopathy juvenile rheumatoid arthritis and, 221 Kpwasaki disease and, 525 sarcoidosis and, 552 systemic arthritis and, 295 Lymphadenosis cutis benigna, 594 Lymphedema, 220 Lymphocytes attivation of, 43-44 See B cell(s) defective control of, 642-643 morphologic features of, 28 naive, 28 self-antigens and, 45 T. See T-cell(s) Lymphoid chemokines, 54 Lymphoma, Epstein-Barr virus-associated, 95 LYlJ1phomatoid granulomatosis, 546-547 Lymphotoxin, 27t
a.
M M proteins, 615 MaCrophage(s)
secondary effects of, 741 systemic lupus erythematosus and, 360 vasculitis associated with, 506 Malignant tumors chondrosarcoma, 738 Ewing's sarcoma, 738-739, 739f fibrosarcoma, 738 of bone, 736-738 of cartilage, 738 of fibrous tissue, 738 of soft tissue, 738 osteosarcoma, 736-738, 737f rhabdomyosarcoma, 738 synOVial cell sarcoma, 738 Mannose receptor, 26 Mannose-binding lectins, 26, 646 Mantel-Haenszel chi-square test, 159, 166 Map chromosome, 65 functional, 66 genetic, 65-66 physical, 66 Marden-Walker syndrome, 747t Marenostrin, 659 Marfan's syndrome, 677t, 756-757, 757t Maroteaux-Lamy syndrome, 747t, 758t Marshall's syndrome, 666-667 Mass spectroscopy peptide fingerprinting, 72-73 protein identification by, 72-73 Matrix metalloproteinases definition of, 59 function of, 59 Matrix-assisted laser desorption ionization-time of flight, 72 MBL2,646 McCune-Albright syndrome, 733, 759 McNemar chi-square test, 166, 166t Mean, 159 Measurement bias, 159 Measures of central tendency, 159-160 Meclofenamate sodium mechanism of action, 79 structure of, 77f toxicities associated With, 83t Median, 159 Mediopatellar plica syndrome, 680, 680t Melorheostosis, 759 Meloxicam, 82t Membrane immunoglobulins, 30 Membranous cells, 48 Membranous glomerulonephritis, 363-364, 364f,377t Membranous ossification, 716 Memory cells B,43 description of, 28 T, 43 Mendelian inheritance, 67 Meningoencephalitis, 595 Menkes' syndrome, 761 Mesangial nephritis, 362, 362f, 377t Mesenchymal stem cells, 23 Mesomelic dysplasias, 755 Mesomelic shortening, 744 Meta-analysis, 143 Metabolic diseases calcium pyrophosphate deposition disease, 635 glucose-6-phosphatase deficiency, 635 gout, 633, 634t hyperlipoproteinemia, 635 Lesch-Nyhan syndrome, 633-634, 634t ochronosis, 635
781
phosphoribosyl pyrophosphate synthetase superactivity, 634 Metacarpophalangeal joints nodules of, 263f polyarthritis of, 263 Metalloproteinases activation of, 13 description of, 13 tissue inhibitor of, 13 Metaphyseal dysplasias, 755-756 Metaphyseal rarefaction, 740, 741f Metaphysis, 14-15 Metastatic bone tumors, 739 Methotrexate accelerated nodulosis associated with, 95, 234 administration of, 91-92 adverse effects of central nervous system, 95 osteopathy, 95 overview of, 93t, 234-235, 331 teratogenicity, 95-96 toxicity. See Methotrexate, toxicities caused by anakinra and, 124 Cogan's syndrome treated with, 518 combination therapies using, 125 Crohn's disease treated with, 338 cyclosporine and, 118 description of, 88 diffuse cutaneous systemic scleroderma treated with, 459 dosage of, 91, 235 duration of, 92 efficacy of, 90-91 etanercept and, 122 folate supplementation with, 96, 234 hypogammaglobulinemia associated with, 94 infection risks, 94 juvenile ankylosing spondylitis treated with, 319 juvenile dermatomyositis treated with, 431 juvenile psoriatic arthritis treated with, 330-331 juvenile rheumatoid arthritis treated with, 89-92, 91t, 232, 234-235 malignancies secondary to, 94-95 mechanism of action, 88-89 metabolism of, 90 nonsteroidal anti-inflammatory drug interactions with, 80, 90 osteopenic effect of, 722 pancytopenia caused by, 94 pharmacology of, 89-90 polyarthritis treated with, 270 safety of, 92 structure of, 88f sulfasalazine and, 98 systemic arthritis treated with, 300 systemic lupus erythematosus treated with, 379 toxicities caused by folate supplementation for, 96 gastrointestinal, 92 hematologic, 94 hepatotoxicity, 92-94 liver, 92-94 pulmonary, 95 Wegener's granulomatosis treated with, 543 Methyldopa, 461t Methylprednisolone Beh~et's disease treated with, 565 intravenous administration of, 111t
782
INDEX
Methylprednisolone (Continued) juvenile rheumatoid arthritis treated with, 236 Kawasaki disease treated with, 534 stmcture of, 103f systemic arthritis treated with, 299 systemic lupus erythematosus treated with, 376-377, 377t uveitis treated with, 292 Wegener's granulomatosis treated with, 542 Mevalonate pathway, 664f Mi-2, 425 Microarrays, 73 Microchimerism, 445 ~2-Microglobulin, 32 Micrognathia, 219, 219f, 227 Microsatellites, 65 Microscopic polyangiitis, 493t Mineralocorticoid receptors, 103-104 Minimum detectable difference, 163-164 Misoprostol, 84 Mitral regurgitation, 619 Mixed connective tissue disease anti-Ul-70Kd antibcx:lies in, 485 characteristics of, 483t classification of, 482 clinical manifestations of, 482-483 course of, 485-486 definition of, 482 diagnostic criteria for, 483-484, 484t differential diagnosis, 483-484 epidemiology of, 482 immunogenetic background of, 482 laboratory examination for, 484-485 pathologic findings in, 483 prognosis for, 485-486 treatment of, 485 Mixed cryoglobulinemia, 504 Mode, 159-160 Model, 175t Molecular mimicry, 50 Monoclonal antibodies anti-CD7, 120 systemic lupus erythematosus treated with, 380 Monocytes cytokines derived from, 121 description of, 19, 20f Morphea antinuclear antibodies in, 477 Borrelia bU1lJdorferi, 475 bullous, 472 classification of, 473t deep, 473-474, 476 definition of, 4 generalized, 472, 476 guttate, 472 linear, 475-476 plaque, 472, 473f subcutaneous, 474 Morphea en plaque, 472, 473f, 478 Morphea profunda, 474, 477 Morquio's syndrome, 758, 758t Mosaicism, 745 Moyamoya disease, 548, 549f Mseleni joint disease, 632-633, 633t, 634f Mucha-Habermann disease, 505 Muckle-Wells syndrome characteristics of, 6711 clinical manifestations of, 660t, 673 genetics of, 672 history of, 659 skin manifestations of, 662f Mucocutaneous disease in Behc;et's syndrome, 562-563
in juvenile dermatomyositis, 412-413 in reactive arthritis, 607, 608f Mucolipidoses, 747t, 758t, 758-759 Mucopolysaccharidoses, 758, 758t Multicentric osteolysis, 747t Multicentric reticulohistiocylosis, 639, 639f Multifocal sclerosing osteosarcoma, 737 Multimodal distribution, 160 Multiple enchondromatosis, 731, 762 Multiple epiphyseal dysplasia, 756 Multiple hypothesis testing, 162 Multiple linear regression, 168 Multiple logistic regression, 168 Multiplexing, 68 Multivariate analyses, 169-171 Multivariate analysis of covariance, 170 Multivariate analysis of variance, 170 Mumps vims arthritis, 583 Munchausen's syndrome by proxy, 688 Muscle contraction of, 15-16 skeletal, 15-16, 16f Muscle fibers, 16t, 16-17 Muscular dystrophy, 421 Musculoskeletal abnormalities and diseases in Behc;et's syndrome, 563 cystic fibrosis, 637-638 diabetes mellitus, 636-637 in diffuse cutaneous systemic scleroderma, 449-450 endemic osteoarthritis, 631-633 fluorosis, 631 hemoglobinopathies, 635 hemophilia, 635-636 hyperostosis, 638 hypervitaminosis A, 631 in Lyme disease, 594-595 overview of, 630 in reactive arthritis, 606-607 rickets, 630-631, 631£, 631t in sarcoidosis, 553-554 scurvy, 631 in systemic arthritis, 293 in systemic lupus erythematosus, 352, 353f Musculoskeletal pain age at onset, 704 amplified, 703-710, 705t assessment of, 700 classification of, 703-704 clinical manifestations of, 706, 707f definition of, 703-704 differential diagnosis, 707-708 epidemiology of, 704 gender ratio for, 704 geographic distribution of, 704 historical review of, 703 history-taking for, 701, 7011 laboratory examination for, 701 physical examination for, 701, 701t prevalence of, 704 psychologic distress in, 704 racial distribution of, 704 summary of, 710 Mycophenolate mofetil description of, 114f, 114-115, 115t diffuse cutaneous systemic scleroderma treated With, 459 systemic lupus erythematosus treated with, 379
Mycoplasma pneumoniae, 575 Myelin basic protein, 49 Myeloperoxidase, 59 Myoadenylate deaminase deficiency, 421-422
Myocarditis acute rheumatic fever and, 619 juvenile rheumatoid arthritis and, 221 systemic arthritis and, 294 systemic lupus erythematosus and, 354 Myofascial pain, 708t Myofibrils, 15 Myofilaments, 15 Myoglobin, 424 Myosin,16f Myositis postinfectious, 421 in systemic scleroderma, 423 Myositis ossificans progressiva, 266 Myositis-associated antibodies, 424-425 Myositis-specific antibodies, 424-425
N N of 1 design, 154 Nabumetone, 82t N-Acetylgalactosamine, 11 Nail-patella syndrome, 746t Nails juvenile psoriatic arthritis-related findings, 328, 328f pitting of, 328 ridging of, 328 NALp, 672 Naproxen dosing of, 81t juvenile ankylosing spondylitis treated with, 319 juvenile psoriatic arthritis treated with, 330 juvenile rheumatoid arthritis treated with, 233 pharmacokinetics of, 80 stmcture of, 77f toxicities associated with, 83t Narcolepsy, 354 Natural killer cells, 23, 24f Neck exercises, 189f Necrotizing angiitis, 365 Necrotizing sarcoid granulomatosis, 555 Negative skewing, 160
Neisseriagonorrhoeae, 574 Neonatal lupus erythematosus animal models of, 400-401 autoantibodies in, 393-394 autoantigens associated with, 392-393 calreticulin in, 393 chondrodysplasia punctata associated with, 399 congenital atrioventricular block in animal models of, 400 description of, 393-394 epidemiology of, 395 history of, 394-395 long-term outcome for, 396 pathologic findings, 395 treatment of, 395-396 course of, 399 cutaneous dermatitis of, 397 description of, 393 differential diagnosis of, 398 epidemiology of, 396 history of, 396 lesions of, 397 pathologic findings in, 397-398 rash associated with, 396 subacute, 396, 397f-398f
INDEX
telangiectasias in, 397 ,treatment of, 398 d¢finition of, 392 description of, 5 etiiology of, 392-394 genetic background of, 394 hematologic abnormalities in, 398-399 hepatic dysfunction in, 398 human leukocyte antigen genes associated with,394 ill vitro experiments of, 400-401 ill vivo experiments of, 401 4t protein in, 392 ymgendorff experiments in, 400-401 li~er disease in, 398 ith. mothers, 399-400 ~eurologic manifestations of, 399 pathogenesis of, 392-394 Ro protein in, 393 thrombocytopenia in, 398-399 Ne(lfiatal septic arthritis, 573 NeOpterin, 424 Nel'hritis. See also Glomerulonephritis focal, 377t iJlterstitial, 365 lupus characteristics of, 352-353, 353t, 361t, 372 end-stage, 381t treatment of, 377t, 380-381 tlnembranous, 377t mesangial, 362, 362f, 377t Nephrogenic fibrosing dermopathy, 454 Network-forming collagens, 12t Neljralgic amyotrophy, 693 N~uroblastoma, 739 Neuroborreliosis, 600 Neuromuscular diseases classification of, 422t-423t juvenile dermatomyositis and, 421-422 Neuropeptide Y, 59 N(lutropenia, 368 Neutrophils IIbnormalities of, 347 IIctivation of, 20 description of, 19-20 glucocorticoids effect on, 105 New drug application, 149 Nicotinamide adenine dinucleotide phosphate oxidase, 59, 643 Nilric OXide, 59 Nilric oxide synthase, 22, 22f, 59 Mmethyl-D-aspartate, 699 Nqx:iception central nervous system, 697-698 peripheral, 697 spinal cord, 697-698 Nociceptors description of, 697-698 silent, 699 Nominal variables, 157 Nbncomparative clinical trial, 152 Nondifferential measurement bias, 159 Noninflammatory musculoskeletal pain syndromes, 3 Nbnossifying fibroma, 731-732, 734f Nonparametric ttest, 166-167 Nonrandom error, 158 Nonsteroidal anti-inflammatory drugs clearance of, 80 description of, 77 dosing of, 79 initiation of use, 81--82 juvenile ankylosing spondylitis treated with, 318-319
juvenile psoriatic arthritis treated with, 329-330 juvenile rheumatoid arthritis treated with, 83, 232-234 mechanism of action, 78-79 methotrexate interactions with, 80, 90 oligoarthritis treated with, 285 patient response to, 81 pharmacokinetics of, 80 plasma protein binding of, 80 polyarthritis treated with, 269 post-streptococcal reactive arthritis treated with,626 principles of use, 80--82 processes influenced by, 79t reactive arthritis treated with, 610-61.1 structure of, 77f systemic lupus erythematosus treated with, 376 toxicities associated with central nervous system, 85 coagulation, 86 cutaneous, 85--86 description of, 82, 83t gastrointestinal, 80, 82--84 hepatotoxicity, 84 hypersensitivity, 86--87 misoprostol for, 84 renal, 84--85 Reye's syndrome, 86 ulcers, 82--83 types of, 81t--82t Norepinephrine, 59 Nuclear activation factor of T-cells, 34, 117 Nuclear factor-lCB cyclosporine effects on, 118 tumor necrosis factor-a activation of, 326 Null hypothesis, 144 Nutritional abnormalities endemic osteoarthritis, 631-633 fluorosis, 631 hypervitaminosis A, 631 overview of, 630 rickets, 630-631, 631f, 631t scurvy, 631
o Observational studies, 143 Occipital horn syndrome, 761 Occupational therapy activities of daily living, 196 exercise play, 196t range of motion, 189, 189f-194f therapeutic, 195-196 fatigue, 191 goals of, 189 juvenile ankylosing spondylitis treated With, 319 juvenile rheumatoid arthritis managed using, 240-241 modalities description of, 196 heat and cold, 197-198 shoe modifications, 199 water exercises, 198-199 pain chronic, 192, 194 description of, 191-192 polyarthritis treated with, 270-271 posture/positioning, 194-195
783
principles of, 189t referral for, 189, 190t rest, 190-191 Ochronosis, 635 Odds ratio, 145, 146t Oligoarthritis age at onset, 274, 275f autoantibodies in, 277 bone overgrowth associated With, 283, 284f-285f clinical manifestations of, 277 course of, 286 definition of, 274 description of,S diagnosis of, 277-278 differential diagnosis, 277, 278t early-onset, 275 epidemiology of, 274-275 etiology of, 277 extended, 286 gender ratio of, 275 genetics of, 275-276 human leukocyte antigen genes associated with, 275-276, 276t incidence of, 274 International League of Associations for Rheumatology classification of, 275t intra-articular steroid use for, 7 laboratory evaluation of, 283 leg-length inequalities associated with, 285-286 management of, 284-285 monarthritis vs., 277, 278t non-human leukocyte antigen genes associated With, 276 nonsteroidal anti-inflamntatory drugs for, 285 pathogenesis of, 277 persistent, 286 prevalence of, 274 prognosis of, 285-286 radiographic evaluation of, 283--284, 284f-285f uveitis associated with antinuclear antibodies in, 281-282 band keratopathy, 280, 282f, 283 clinical manifestations of, 279-280 course of, 283 description of, 278 differential diagnosis, 281 epidemiology of, 279, 279t etiology of, 279 genetics of, 279 glucocorticoids for, 282 histopathology of, 280-281 laboratory examination of, 281-282 management of, 282-283 monitoring of, 28lt pathogenesis of, 279 pathology of, 280-281 prognosis of, 283 slit-lamp biomicroscopy of, 280, 281£ Oilier's disease, 731, 762 Omenn's syndrome, 647t One-tailed hypothesis, 144 Onion-skin lesions, 360-361, 361£ Open label, 152 Open sUldies, 152 Opsonin,24 Oral tolerization, 47-48 Ordinal variables, 157, 157f Osgood-Schlatter disease, 681, 681t, 682f Osseous sarcoid, 554 Ossifying fibroma, 734
784
INDEX
Osteoarthritis endemic, 631--{i33 hypermobility and, 678 Osteoblastoma, 729--730 Osteocalcin, 717t Osteochondritis dissecans, 686--687, 687f Osteochondrodysplasias achondroplasia, 746 classification of, 746, 748t definition of, 746 description of, 638 hypochondroplasia, 747, 747t International classification of, 748t Osteochondroma, 730, 731£-732f Osteochondroses definition of, 683 Freiberg's disease, 685-686, 686f Kohler's disease, 685, 686f Legg-Calve-Perthes disease, 684, 685f Scheuermann's disease, 684-685, 689 types of, 683t Osteoclast, 718 Osteofibrous dysplasia, 733 Osteogenesis imperfecta, 677t, 748-749, 749t-750t, 750f Osteogenic lesion, 728, 729t Osteoid osteoma, 729, 730f-731£, 730t Osteolyses, 759--760 Osteolytic lesion, 728, 729t Osteomyelitis Brodie's abscess in, 576-577, 577t, 578f chronic recurrent multifocal, 577-578, 580f, 708t classification of, 576 clinical manifestations of, 576-577 course of, 577 definition of, 576 diagnosis of, 577, 579f diskitis vs., 579--580, 581£ epidemiology of, 576 etiology of, 576 frequency of, 4t in metaphyses, 576 microorganisms associated with, 576 pathogenesis of, 576 prognosis for, 577 SAPHO syndrome vs., 329, 578-579 treatment of, 577 Whipple's disease vs., 580 Osteonecrosis, 381 Osteopathy, methotrexate-related, 95 Osteopenia in connective tissue diseases, 721-722 definition of, 719 juvenile rheumatoid arthritis and, 219--220 prevention of, 722-723 reactive arthritis and, 609 Osteopetrosis, 759 Osteopoikilosis, 759, 760f Osteoporosis bisphosphonates for, 110, 724 calcium intake to prevent, 722-723 in chronic arthritis, 721 t in connective tissue diseases, 721-722 corticosteroid-associated, 110 definition of, 719 glucocortiCOid-related, 107, 108f, 110, 723 idiopathic juvenile, 749 inflammatory bowel disease and, 335 in juvenile dermatomyositis, 427, 428f in juvenile psoriatic arthritis, 329 in juvenile rheumatoid arthritiS, 226 periarticular, 329 prevention of, 722-723 risk factors for, 721 t
transient migratory, 708t treatment of, 723-725 vitamin D for, 110 Osteoprotegerin, 718 Osteosarcoma, 736-738, 737f Outcomes research decision analysis, 148-149 description of, 147-148 Overreaction, 707t
p P values, 162-163 Pachydermoperiostosis, 638, 761 Pain acute, 703 analgesics for, 703 anatomy of, 697 assessment of behavioral measures for, 702 Children's Hospital of Eastern Ontario Pain Scale, 702 Faces Rating Scale, 702, 702f general approach to, 708 multidimensional measures for, 702-703 overview of, 701-702 physiologic measures for, 702 self-report measures for, 702-703 visual analogue scale for, 702 back diskitis, 689-690 epidemiology of, 689 nonorganic, 707t treatment of, 690 chest, 690-691, 691t chroniC, 192, 194, 703 congenital indifference to, 689, 690f course of, 710 definition of, 697 diffuse idiopathic, 705t etiology of, 704, 706 genetic background of, 706 growing-related, 692-693 hypermobility-related, 676-680 idiopathic, 705t inhibition of, 699--700 intervertebral disk herniation, 691 juvenile rheumatoid arthritis-related, 216-217, 241 . knee, 678 laboratory examination of, 708 localized idiopathic, 705t management of, 703 measurement of, 697 modulation of brain's role in, 700 spinal cord's role in, 699--700 musculoskeletal age at onset, 704 amplified, 703-710, 705t assessment of, 700 classification of, 703-704 clinical manifestations of, 706, 707f definition of, 703-704 differential diagnosis, 707-708 epidemiology of, 704 gender ratio for, 704 geographic distribution of, 704 historical review of, 703 history-taking for, 701, 701t laboratory examination for, 701 physical examination for, 701, 70It prevalence of, 704
psychologic distress in, 704 racial distribution of, 704 summary of, 710 myofascial, 708t osteosarcoma-related, 736 outcomes for, 710 patellofemoral, 680 pathogenesis of, 704, 706 pathology of, 707 pathways for, 697, 698f phantom limb, 699 physical and occupational therapy for, 191-192 physiology of, 697 plica syndromes and, 680, 680t procedural, 703 prognosis for, 710 radiographic examination of, 709 sensitivity to, 700 sleep disturbances and, 709 stress fractures and, 680-681, 681£ summary of, 710 team-based approach to, 709 tolerance to, 700 treatment of, 709--710 undertreatment of, 216-217 vocalization by child, 216 Palmoplantar pustulosis, 578 Pancreatitis, 359--360, 638 Pancytopenia, methotrexate-related, 94 Pannus, 222 Papillary necrosis, 85 Papillitis, 278 Parallel group design, 153 Parasites, 585 Parathyroid hormone, 718 Parenchymal pulmonary disease, 221 Parosteal tumors, 737-738 Paroxysmal nocturnal hemoglobinuria, 25-26 Parvovirus, 582 Passive cell death, 44 Passive rotation test, 707t Patellar dislocation, recurrent, 679 Patellofemoral pain, 680 Pathogen-associated molecular patterns description of, 21, 2It Toll-like receptors, 21-22 Patient(s) adherence by, 186, 186t compliance by, 186, 186t counseling of, 186 positioning of, 194--195 siblings of, 186-187, 187t transition to adult care, 187-188 Patient preference, 175t Patient-oriented research clinical trials blinded comparative, 152-153 blinded withdrawal design, 154 classification of, 149, 151, 151t comparative, 152, 154--155 conducting of, 155 crossover designs, 153 data analysis plan. See Data analysis plan data collation, 155 data collection, 155 design of, 152-157 double-dummy design of, 153 factorial designs, 153 group sequential designs, 153-154 guidelines for, 149 initiator of, 149 N of 1 design, 154 noncomparative, 152
INDEX
objectives of, 151-152 open, 152 parallel group design of, 153 phase I, 149-150 phase II, 150-151 phase III, 151 phase IV, 151 population in, 152 randomization, 153, 153t deScription of, 149 Patt~rn-recognition receptors dejscription of, 21, 2lt signal transduction pathways activated by, 22 Pau9iarticular juvenile rheumatoid arthritis, 69, 74 Peak bone mass, 718-719 PeaJt concentration, 150 Pearson product-moment correlation, 167-168 Ped,atric Quality of Life Inventory, 177t, 180 Pedjatric rheumatology advances in, &-7 description of, 3 elnergence of, 4 historical studies of, 3-4 monogenic diseases in, 67, 67t iIi United States, 4 Pelvic apophysitis, 681 Pena-Shokeir syndrome, 747t D-P~nicillamine
description of, 101, lOlt, 460 jllVenile rheumatoid arthritis treated with, 240 Pen:tamers, 30 PeQtoxifylline, 534 Pentraxins, 26 Peptidoglycan, 615 Perforin, 23, 39 Perlaqueductal gray area, 700 Pericarditis j\.lvenile rheumatoid arthritis and, 221 s;ystemic arthritis and, 294 s;ystemic lupus erythematosus and, 354, 366 Perimysium, 15, 16f Period prevalence, 145 Pe~iodic fever syndromes with aphthous stomatitis, phar;yngitis, and adenitis, 66&-667 IiJrucellosis, 657 ¢Yclic hematopoiesis as cause of, 665--666 dlescription of, 657 liJereditary description of, 658--659 history of, 659 inheritance patterns of, 658t, 658--659 hyperimmunoglobulinemia D with clinical manifestations of, 660t, 663 description of, 659, 663 diagnosis of, 665 erythematous macules associated with, 663 genetics of, 663 laboratory studies, 663 maculopapular rash associated with, 663, 665f pathogenesis of, 663 treatment of, 665 infectious causes of, 657-658 .rat-bite fever, 657-658 relapsing fever, 658 rheumatic diseases associated with, 658 Periosteal chondroma, 731 Periostitis, 337, 338f Peripheral afferent neurons, 59
Peripheral blood mononuclear cells, 212 Peripheral joints, in juvenile ankylosing spondylitis, 312-313, 317, 318f Peripheral mononeuropathy, 708t Peripheral tolerance, 46, 47f Peritenon, 15 Periungual erythema in juvenile dermatomyositis, 412, 412f-413f in systemic lupus erythematosus, 350-351, 351f Perniosis, 449, 693, 708t Peroxisome proliferator-activated receptor y" 104, 107 Per-protocol set, 157 Pes cavus, 199, 678--679 Pes planovalgus, 199 Pes planus, 678--679 Peyer's patches, 48 Phagocytosis, 22f, 23 Phalen maneuver, 693 Phantom bone disease, 760 Phantom limb pain, 699 Pharmacodynamics, 150 Pharmacogenetics, 147-148 Pharmacokinetics studies, 150 Pharmacology bioavailability, 76 clearance, 7&-77 drug absorption, 76 drug biotransformation, 77 half-life, 76 volume of distribution, 76 Phase I clinical trials, 149-150 Phase II clinical trials, 150-151 Phase III clinical trial, 151 Phase IV clinical trial, 151 Phenox;ybenzamine, 460, 46lt Phenylketonuria, 455 Phosphatidylserine, 46 Phospholipase A" 105 Phosphoribosyl pyrophosphate synthetase superactivity, 634 Physical abuse, 688-689 Physical map, 66 Physical therapy activities of daily living, 196 exercise play, 196t range of motion, 189, 189f-194f therapeutic, 195-196 fatigue, 191 goals of, 189 juvenile ankylosing spondylitis treated with, 319 juvenile rheumatoid arthritis managed using, 240-241 juvenile rheumatoid arthritis treated with, 240-241 modalities description of, 196 heat and cold, 197-198 shoe modifications, 199 water exercises, 198-199 pain chronic, 192, 194 description of, 191-192 polyarthritis treated with, 270-271 posture/positioning, 194-195 principles of, 189t reactive arthritis treated with, 611 referral for, 189, 190t rest, 190-191 Physician, 185t Pigmented villonodular synovitis, 734
785
Piroxicam dosing of, 82t juvenile psoriatic arthritis treated with, 330 mechanism of action, 79 structure of, 77f Pityriasis lichenoides et varioliformis acuta, 220,505 Plant thorn synovitis, 584, 585t Plaque morphea, 472, 473f, 475 Plasma cells affinity maturation of, 42 generation of, 42 Plasmacytoid dendritic cell, 23 Plasmapheresis juvenile dermatomyositis treated with,431 thrombotic thrombocytopenia purpura treated with, 380 Plasmin, 14t Plasminogen activator, 14t Platelet-activating factor, 225 Platelet-derived growth factor, 57 Play exercises, 196t Pleural effusions, 355 Pleuropulmonary disease diffuse cutaneous systemic scleroderma and, 451 juvenile ankylosing spondylitis and, 310-311 juvenile rheumatoid arthritis and, 221 systemic arthritis and, 294-295 systemic lupus erythematosus and, 355t-356t, 355-356 Plica syndromes, 680, 680t Pneumocystis carinii pneumonia, 108, 356 POEMS syndrome, 741 Point prevalence, 145 Polyangiitis overlap syndrome, 492 Polyarteritis nodosa age at onset, 512 American College of Rheumatology criteria for, 512, 513t animal models of, 513 antineutrophil cytoplasmic antibodies in, 515 central nervous system involvement in, 514 classification of, 512, 513t clinical manifestations of, 513-514, 514t clinicopathologic characteristics of, 494t course of, 51&-517 cutaneous involvement in, 514, 514f-515f cyclophosphamide for, 515 definition of, 493t, 512 differential diagnosis, 514-515 epidemiology of, 512 etiology of, 512-513 frequency of, 6t genetic background of, 513 glucocorticoids for, 515 incidence of, 512 intravenous immunoglobulin for, 515-516 laboratory examination in, 515, 51St pathogenesis of, 512-513 pathology of, 514, 516f prednisone for, 515 prevalence of, 512 prognosis for, 516-517 radiologic examination in, 515, 517f skin manifestations of, 514, 514f-515f treatment of, 515-516 Polyarthritis age at onset, 261, 262f clinical manifestations of cardiac disease, 264-265 Felty syndrome, 264
786
INDEX
Polyarthritis (Continued) joint disease, 262-263, 263f nodules, 263-264, 264f, 266-267 pulmonary disease, 265 systemic, 263 vasculitis, 264 course of, 271 cytokines in, 261-262 definition of, 261 dermatomyositis and, 265 diagnosis of, 265t, 265-266 differential diagnosis, 265t enthesitis-related arthritis vs., 265 epidemiology of, 261 etiology of, 261 gender ratio for, 261 genetics of, 262 geographic distribution, 261 histopathologic findings, 266-267 human leukocyte antigens in, 262 incidence of, 261 International League of Associations for Rheumatology classification of, 261, 262t laboratory findings antinuclear antibodies, 268 autoantibodies, 267-268 inflammatory markers, 267 rheumatoid factors, 267-268 medical management of, 269-270 occupational therapy for, 270-271 onset of, 265 pathogenesis of, 261-262 pathologic findings, 266-267 physical therapy for, 270-271 polyarticular, 262 prevalence of, 261 prognosis of, 271 racial distribution, 261 radiologic examination, 268, 268f-269f, 270t scleroderma and, 265 septic, 265 sickle cell anemia vs., 266 systemic lupus erythematosus vs., 265 treatment of anti-tumor necrosis factor agents, 268--269 etanercept, 268 glucocorticoids, 270 leflunomide, 268 methotrexate, 270 nonsteroidal anti-inflammatory drugs, 269 overview of, 268--269 type II collagen and, 214 Polyarticular juvenile rheumatoid arthritis, 99 Polymerase chain reaction Borrelia but;gdorferi diagnosis by, 596 description of, 66-67 Polymorphisms genome-wide, 65 Single-nucleotide, 65 for transmission disequilibrium testing, 69 Polymyositis, 6t Polyvinyl chloride, 454 Popliteal, 217, 218f Porphyria cutanea tarda, 456 Positive skewing, 160 Post hoc power analysis description of, 163 minimum detectable difference, 163-164 Posterior probability distribution, 161 Postganglionic sympathetic neurons, 59 Postinfectious arthritis, 568, 585 Postinfectious myositis, 421 Post-streptococcal reactive arthritis, 624-626 Postsurgical therapy, 199 Posture, 194-195
Power, 162-163 Prazosin, 461t Precordial catch syndrome, 690-691 Predictive validity, 175t Prednisolone dosing of, 103t reactive arthritis treated with, 611 structure of, 103f Prednisone Beh~et's syndrome treated with, 565 dosing of, 103t juvenile dermatomyositis treated with, 429 juvenile rheumatoid arthritis treated with, 235-236 Kawasaki disease treated With, 533 polyarteritis nodosa treated with, 515 reactive arthritis treated with, 611 structure of, 103f systemic lupus erythematosus treated with, 376-377, 377t Prevalence definition of, 144-145 period, 145 point, 145 Primary angiitis of the central nervous system, 547-548 Principal component analysis, 170 Prior probability distribution, 161 Procaspase-8, 642 Procedural pain, 703 ProcoHagen type I propeptides, 717t Progeria, 455 Prognosis of disease, 147-148 Programmed death-l description of, 34, 49 polymorphisms of, in systemic lupus erythematosus, 345-346 Progressive diaphyseal dysplasia, 708t Progressive hemifacial atrophy, 472-473 Progressive pseudorheumatoid arthropathy, 754f, 754-755 Prolactin, 60, 213, 348 Promiscuous peptide theory, 605 Prospective cohort study, 146 Prospective studies, 143 Prostacyclin, 87 Prostaglandins characteristics of, 58 definition of, 58 E,,58 synthesis of, 78f Protein arrays, 73 Protein stains, n Proteinases, 13 Protein-losing enteropathy, 360 Proteinuria, 222 Proteoglycans description of, 11 heparin sulfate-containing, 54 Proteolytic enzymes, 59 Proteome, 71 Proteomics definition of, 72 mass spectrometry applications, 72-73 microarray-based methods, 73 Protrusio acetabuli, 230, 269f Proximal interphalangeal joints, 263 Proxy reporter, 175t P-selectin, 52, 54t, 445 P-selectin glycoprotein ligand 1, 52 Pseudoachondroplasia, 759 Pseudogenes, 66 Pseudo-Hurler's syndrome, 747t, 758t, 759 Pseudohypoparathyroidism, 637 Pseudopseudohypoparathyroidism, 759
Pseudorheumatoid nodules, 220 Pseudosclerodermas, 455 Pseudotumor cerebri, 110 Psoriatic arthritis. See Juvenile psoriatic arthritis Psoriatic arthritis sine psoriasis, 324 Psoriatic arthropathy, 4t Pulmonary hemorrhage, 355-356 Pulmonary hypertension, 461 Pulmonary toxicity, methotrexate-induced, 95 Pulseless disease, 548 Purine nucleoside phosphorylase deficiency, 647t, 648 Pyoderma gangrenosum, 336, 337f Pyrin, 659
Q Quality of life definition of, 174 health-related, 174 hierarchy of outcomes, 175-176 Juvenile Arthritis Quality of Life Questionnaire, 177t, 179 measuring of instruments for, 176 reasons for, 174-176 Pediatric Quality of Life Inventory, 177t, 180 Quality of My Life Questionnaire, 177t, 179-180 terminology associated with, 175t Quality of My Life Questionnaire, 177t, 179-180 Quantitative computed tomography, 720, 720t Quantitative high-frequency sonography, nOt, 720-721 Quantitative trait locus, 68
R Random error, 158 Randomization, 153, 153t Range of motion exercises, 189, 189f-194f Rash in hyperimmunoglobulinemia D with periodic fever syndrome, 663, 665f in juvenile psoriatic arthritiS, 328, 328f in neonatal lupus erythematosus, 396 in systemic arthritis, 293f, 293-294 Rat-bite fever, 657-658 Rate, 159 Raynaud's disease, 447-448, 708t Raynaud's phenomenon acrocyanosis vs., 449 clinical findings of, 448-449 differential diagnosis, 449 in diffuse cutaneous systemic scleroderma, 447-449, 453 in juvenile dermatomyositis, 414 renovascular, 451 in systemic lupus erythematosus, 358 treatment of, 460, 4611 Reactive arthritis arthritogenic bacteria associated with, 605-606 bacteria that cause, 605 Campylobacter infection and, 606, 609 Chlamydia infection and, 606, 609 circinate balanitis in, 607, 608f classifkation of, 604 clinical manifestations of, 606-607, 608f course of, 611-Q12 description of, 568, 604
INDEX diagnostic criteria for, 604, 605t differential diagnosis, 610 enthesitis in, 606 epidemiology of, 604 etiology of, 605-606 frequency of, 4t g~netics of, 605 glucocorticoids for, 611 HiLA-B27 and, 604-606 iJifectious causes of, 213, 605t, 605-606 k~ratoderma blennorrhagicum in, 607, 608f, 611 l~boratory examination of, 609 nfIucocutaneous disease in, 607, 608f nfIusculoskeletal disease in, 606--607 I1onsteroidal anti-inflammatory drugs for, 610-611 qrthopedic surgery in, 611 osteopenia in, 609 pathogenesis of, 605--606 physical therapy and rehabilitation for, 611 post-streptococcal, 624-626 prednisolone for, 611 prednisone for, 611 prognosis for, 611-612 promiscuous peptide theory of, 605 radiologic examination of, 609, 6091:...6lOf $acroiliac joint involvement in, 609, 610f Salmonella infection and, 606, 612 Shigella enteritis and, 606 $igns and symptoms of, 607 $ulfasalazine for, 611 synovial fluid effusion in, 607 treatment of, 610-611 Yersinia infection and, 606, 609 Reactive hematophagoeytic lymphohistiocytosis, 295 Reactive oxygen products, 59 Rea<.1:ive sclerosis, 315, 316f Recall bias, 159 Receiver operating characteristic, 147, 148f Receptor activator of nuclear factor-KB, 718 Rll'ceptor activator of nuclear factor-KB ligand, 718 R¢ceptor editing, 31 Receptor-associated tyrosine kinases, 39 R~gression, 168 Reiter's syndrome conjunctivitis in, 607 defmition of, 604 Relapsing fever, 658 Relapsing polychondritis, 505-506, 506f, 658 Relative carpal length, 228 RHative risk, 145, 146t Reliability, 175t R¢mission, 156 Remodeling of bone, 717t Renal disease Beh\;et's syndrome and, 564 diffuse cutaneous systemic scleroderma and, 451 Henoch-Schonlein purpura and, 497-498, 501 juvenile ankylosing spondylitis and, 311 juvenile rheumatoid arthritis and, 222 treatment of, 501 Jteserpine, 461t Respective pattern-recognition receptors, 21t Response variables, 156-157 Responsiveness, 175t Rest, 190-191 ~sting splints, 194-195, 195f, 198 Restless legs syndrome, 694, 708t Reticular dysgenesis, 647t
Reticulin, 12-13 Retina antimalarial toxicity, 97 vasculitis of, 358, 359f Retrospective studies, 143 Reverse placebo effect, 154 Reye's syndrome, 86, 222, 531 Rhabdomyolysis, 424 Rhabdomyosarcoma, 738 Rheumatic diseases. See also specific disease age-related manifestations of, 5 definition of, 2, 184 disability associated with, 6 frequency of, 6t gonadal immaturity and, 5 management of adherence, 186, 186t components of, 185t counseling, 186 family-centered, community-based care, 185 financial, 188-189 Internet resources, 186 overview of, 184-185 patient education, 185-186 physicians' role in, 185t school issues, 187, 187t-188t sibling issues, 186-187 team-based approach, 185f transition to adult care, 187-188 modifying factors for, 4-5 of childhood, 3-6 overview of, 184-185 problems associated with, 199t skeletal maturity and, 5 terminology associated with, 2-3 unconventional remedies for, 189 Rheumatic fever, 297 Rheumatic heart disease prophylaxis, 623-624 Rheumatism, 2-3 Rheumatoid arthritis frequency of, 6t juvenile. See Juvenile rheumatoid arthritis word origin of, 2 Rheumatoid factors in juvenile rheumatoid arthritis, 224 in oligoarthritis, 283 in polyarthritis, 267-268 in systemic lupus erythematosus, 371 Rheumatoid nodules in juvenile rheumatoid arthritis, 220, 223 in polyarthritis, 263-264, 264f, 266-267 zones of, 266-267 Rheumatoid vasculitis in juvenile rheumatoid arthritis, 220, 220f in polyarthritis, 264 Rhizomelic shortening, 744 Rice bodies, 266, 267f Rickets, 630-631, 631£, 631t Riley-Day syndrome, 747t Risk absolute, 146t attributable, 146t definition of, 145 relative, 145, 146t Risk factors, 146t Risk ratio, 145 Rituximab, 121 RMRP, 648 Ro protein, 393 Rofecoxib gastrointestinal effects of, 84 structure of, 78f RoRNP,393 Rosenbloom syndrome, 216
787
Rothmund-Thomson syndrome, 349, 736 Rubella virus-associated arthropathy, 581-582
s Sacroiliac joints arthritis of in inflammatory bowel disease, 335 in juvenile ankylosing spondylitis, 315-316 in juvenile rheumatoid arthritis, 230-231 computed tomography of, 316, 316f inflammation of, 316 magnetic resonance imaging of, 316 radiographic examination of, 315-316 in reactive arthritis, 609, 610f widening of, 315, 315f Saddle-nose deformity, 541£ Salicylates administration of, 87 contraindications, 88 definition of, 87 drug interactions, 88 mechanism of action, 87 pharmacology of, 87 Reye's syndrome and, 86 toxicity of, 87t, 87--88 Salicylism, 87-88 Salmonella infection, 606, 612 Sample size, 163 Sanfilippo syndrome, 747t, 758t SAPHO syndrome, 329, 578-579 Sarcoid arthritis, 555f-556f Sarcoidosis clinical manifestations of, 552-554 course of, 556 definition of, 551 differential diagnosis, 554-555 epidemiology of, 552 gastrointestinal tract involvement in, 554 genetic background of, 552 glucocorticoids for, 555 hepatosplenomegaly in, 552 immunomodulating drugs for, 555-556 laboratory examination in, 555 lymphadenopathy in, 552 musculoskeletal disease in, 553--554 neurologic abnormalities in, 553 ocular abnormalities in, 552-553 prednisolone for, 555 prevalence of, 552 prognosis for, 556 pulmonary disease in, 553 pulmonary function measurements in, 555 radiologic examination in, 555, 555f-556f skin manifestations of, 552 treatment of, 555-556 vasculitides associated with, 554 Sarcoma chondrosarcoma, 738 Ewing's, 738-739, 739f fibrosarcoma, 738 osteosarcoma, 736--738, 737f rhabdomyosarcoma, 738 synovial cell, 738 Scheie's syndrome, 747t, 758, 758t Scheuermann's disease, 684-685, 689 Schmid type of metaphyseal dysplasia, 754 Schmid's syndrome, 747t Schober test, 314, 314f Schooling, 187, 187t-188t Schwachman-Diamond syndrome, 755-756 Schwartz-Jampe! syndrome, 747t Scl-70, 456
788
INDEX
Scleredema, 455-456 Scleroderma chemically induced, 454-455 definition of, 442 diffuse cutaneous. See Diffuse cutaneous systemic scleroderma frequency of, 6t historical review of, 442 linear, 472-473, 473f polyarthritis and, 265 proximal, 453 systemic. See Systemic scleroderma Scleromyxedema, 455 Scoliosis, arthritis with, 746t Scurvy, 631 Seckel's syndrome, 747t Secondary hypertrophic osteoarthropathy, 741 Secondary null hypothesis, 144 Seizures, 354 Selectins, 52, 54t Selection bias, 159 Selective 19A deficiency chronic arthritis and, 649, 650f diseases associated with, 649-650 epidemiology of, 648 etiology of, 648--649 juvenile ankylosing spondylitis and, 314 juvenile rheumatoid arthritis and, 216, 649 prevalence of, 648 systemic lupus erythematosus and, 649 Self-antigens cryptic epitopes, 47 description of, 45 Self-tolerance, 28 Sepsis, 382 Septic arthritis age at onset, 568 antibiotics for, 573, 573t blood studies for, 571 clinical manifestations of, 569-570, 570t course of, 576 description of, 568, 652 diagnosis of, 570-572, 572f epidemiology of, 568 etiology of, 569 familial clustering of, 569 gender ratio for, 568 geographic clustering of, 569 gonococcal arthritis and, 574 hip joint, 572f, 573-574 joints affected by aspiration and drainage of, 573 description of, 569-570, 570t, 572f hip, 572f, 573-574 magnetic resonance imaging of, 571-572 microorganisms involved in, 569, 569t, 652 neonatal, 573 pathogenesis of, 569 prognosis for, 576 radiologic examination of, 571, 572f radionuclide scans for, 571 synovial fluid analysis in, 225, 571 treatment of, 572-573 tuberculous arthritis, 574, 574f-575f ultrasonography of, 571 SeptiC polyarthritis, 265 Serine proteinases, 13 Serious adverse event, 150 Seronegative enthesitis and arthritis syndrome, 304-306, 306£, 307t, 313t Seronegative spondylarthritides, 304 Serum amyloid A protein, 26, 244, 661 Semm sickness, 51, 502 Severe combined immunodeficiency, 647--648 Sever's disease, 682
Sharpey's fibers, 15 Shigella enteritiS, 606 Shin splints, 683 Shoe modifications, 199 Short trunk, 755 Shoulder exercises for, 190f juvenile rheumatoid arthritis of, 228-229 Shrinking lung, 356 Sialidase deficiency, 758t Sialolipidosis, 758t Siblings, 186-187, 187t Sicca syndrome, 451-452 Sickle cell anemia dactylitis caused by, 266 description of, 635 polyarthritis vs., 266 Signal recognition peptide, 425 Silent nociceptors, 699 Sinding-Larsen-]ohansson disease, 681--682, 682f Single-blind clinical trials, 152 Single-nucleotide polymorphisms, 65 Site management organizations, 155 Sitosterolemia, 635 Sjogren's syndrome classification of, 486-487 clinical manifestations of, 487, 487f definition of, 486 description of, 359, 359t laboratory examination in, 487-488 pathologic findings in, 487, 487f primary, 486 secondary, 486 treatment of, 488 Skeletal muscle anatomy of, 15, 16f contraction of, 15-16 juvenile dermatomyositis findings, 415-416, 418, 418f Skeleton axial, 313-314 functions of, 716 growth of, 716 maturation of, 718-719 Skewing, 160 Skin acute rheumatic fever manifestations, 620, 620f Beh\;et's syndrome manifestations, 562-563 chronic infantile neurologic cutaneous and articular syndrome manifestations, 673, 673f diffuse cutaneous systemic scleroderma manifestations, 445-447 familial Mediterranean fever manifestations, 660 Henoch-Schonlein purpura manifestations, 497, 498f-499f hypersensitiVity angiitis manifestations, 503f inflammatory bowel disease findings, 336-337 juvenile dermatomyositis findings, 412-413, 419 juvenile psoriatic arthritis findings, 328, 328f juvenile rheumatoid arthritis findings, 220, 220f Kawasaki disease manifestations, 524-525, 525f Lyme disease manifestations of, 594 Muckle-Wells syndrome manifestations, 662f nodules of, 220, 223 nonsteroidal anti-inflammatory drug-related toxicities, 85-86 polyarteritis nodosa manifestations, 514, 514f-515f
sarcoidosis manifestations, 552 systemic lupus erythematosus manifestations, 349-352, 350f-351£, 365,366£ tumor necrosis factor receptor-associated periodic syndrome manifestations, 662 Sleep disturbances, 709 Slipped capital femoral epiphysis, 688 Slipping rib syndrome, 692 Slow muscle fibers, 16, 16t Slow-acting antirheumatic dmgs definition of, 88 juvenile rheumatoid arthritis treated with, 238-239 Sly syndrome, 758t Small-bowel bypass, arthritis-dermatitis syndrome associated with, 585 Soft tissue benign tumors of, 734-735 malignant tumors of, 738 stress injuries, 682--683 Soluble intercellular adhesion molecule-I, 262 Soluble interleukin-6 receptor, 291-292 Somatic hypermutation, 30 Somatic recombination, 30 Soter's syndrome, 518, 518t Spearman rank correlation, 168 Sphingolipidoses, 638--639, 6391 Spinal cord central sensitization, 698f, 699 nociceptive pathways in, 697--698 pain modulation by, 699-700 Spine fusion of, 230, 230f juvenile rheumatoid arthritis effects on, 230f, 230-231 Spirochetes, 584-585 Splenomegaly juvenile rheumatoid arthritis and, 221 systemic arthritis and, 295 systemic lupus erythematosus and, 360 Splints dynamic, 198, 198f functional, 198 purposes of, 198 resting, 194-195, 195f, 198 wrist, 198, 198f, 241 Spondyloarthritis, 64, 304 Spondyloarthropathy frequency of, 4t historical review of, 304-305 Spondyloepiphyseal dysplasias, 747t, 751-752, 752f, 752t Spondylolisthesis, 691 Spondylolysis, 691 Spondylometaphyseal dysplasia, 747t Sporothrix schenckii, 584, 584f Sporotrichosis, 584, 584f Sports, 196 Standard deviation, 160 Standard error of the mean, 161 Standardized response mean, 156 Staphylococcal pyomyositis, 421
Staphylococcus aureus, 569 Statistical error, 162-163 Statistical tests analysis of covariance, 168 analysis of variance, 167 bayesian approaches, 161 binomial test of proportions, 164 clinical significance of, 164 confidence intervals for, 164 correlation, 167-168 Fisher's exact test, 166
INDEX frequentist methods, 161 gQodness-of-fit chi-square test, 164 K4sample tests, 167 multivariate analyses, 169-171 one-sample, 164-165 overview of, 161 p'values, 162-163 pOst hoc power analysis, 163 pOwer of, 162-163 r;lJter agreement measures, 169 regression, 168 s'lmple size for, 163 s\i1rvival analysis, 168-169 t test, 164-167 two-sample, 165-167 variables in, 161-162 Stem cell transplantation description of, 126 diffuse cutaneous systemic scleroderma treated with, 45H60 jllvenile dermatomyositis treated with, 431-432 j\ilvenile rheumatoid arthritis treated with, 240 s~vere combined immunodeficiency treated with,647 systemic arthritis treated with, 300 Steroid myopathy, 430 Steroid pseudorheumatism, 110, 236 Stevens-Johnson syndrome, 98, 506-507 Stickler's syndrome, 677t, 745, 746t, 750-751 Stiff-hand syndrome, 636, 637f Stiff-skin syndrome, 455 Still's disease, 294, 297-298 Streptococcal toxic shock syndrome, 615 Streptococcus '1ntibody tests, 616-617 <:lescription of, 615 JilOst-streptococcal reactive arthritis, 624-626 pyrogenic exotoxins, 616
StJieptococcus pneumoniRe, 569 Stress fractures, 680-681 Str~ss injuries ~f lower limbs, 680-682, 681£ Of upper limbs, 682 $oftt~sue, 682-683 Str~tching exercises, 195, 195f Stmmelysin, 13, 14t Stljdent's t test, 164, 166 Subcutaneous granuloma annulare, 220, 264 Sulx:utaneous morphea, 474 Subcutaneous tissue, 220, 220f Subscapular bursa, 14 Substance P, 699 Suptalar joints, 229 Sulfasalazine description of, 97-98, 98t juvenile ankylosing spondylitis treated with, 319 juvenile rheumatoid arthritis treated with, 239 reactive arthritis treated with, 611 Sulindac central nervous system toxicities, 85 dosing of, 82t structure of, 77f toxicities associated with, 83t Superantigens, 37, 50 Surrogate, 175t Stjrrogate end points, 156 Survival analysis, 168-169 Sweet's syndrome, 506 Sydenham's chorea, 620-622 Syndrome of inappropriate antidiuretic hormone, 116
Synovectomy, for juvenile rheumatoid arthritis, 241 SynOVial A cells, 13 Synovial B cells, 13 Synovial cell sarcoma, 738 Synovial chondromatosis, 734-735 SynOVial fluid description of, 14, 14t glycosaminoglycans in, 225-226 juvenile rheumatoid arthritis findings, 225-226 Lyme disease findings, 598 septic arthritis findings, 571 systemic arthritis findings, 298 Synovial hemangioma, 734, 734f Synovial joints anatomy of, 9, 10f characteristics of, lOt Synovial membrane, 13 Synoviocytes, 13 Synovitis, 53f Synteny,66 Syphilis, 584-585 Systemic arthritis age at onset, 291 amyloidosis associated with, 296-297 cardiac disease associated with, 294 classification of, 291 clinical manifestations of, 292-297 course of, 300 cytokine profiles in, 291-292 definition of, 291 diagnostic criteria for, 291, 292t differential diagnosis, 297, 297t epidemiology of, 291 etiology of, 291 extra-articular manifestations of, 292t, 292-297 fever and, 293, 297 genetics of, 292 hepatic disease and, 296 hepatomegaly and, 296 histopathologic findings, 298 laboratory examination of, 298 lymphadenopathy and, 295 lymphocytes in, 292 macrophage activation syndrome and, 295f, 295-296 mortality rate for, 300, 382 musculoskeletal disease findings, 293 myocarditis associated with, 294 pathogenes~ of, 291-292 pathologic findings, 298 pericarditis associated with, 294, 366 pleuropulmonary disease associated with, 294-295 prognosis of, 300 radiologic examination of, 298, 298t rash associated with, 293f, 293-294 splenomegaly and, 295 synovial fluid findings in, 298 tenosynovitis associated with, 293 treatment of anti interleukin-6 receptor, 299-300 anti-tumor necrosis factor agents, 299 approach to, 298 autologous stem cell transplantation, 300 cyclosporine, 299 intravenous immunoglobulin, 299 methotrexate, 300 methylprednisolone, 299 pharmacologic, 298-299 tumor necrosis factor-a levels in, 292 Systemic Lupus Activity Measure, 372, 374t
789
Systemic lupus erythematosus acute hemolytic anemia associated with, 380 acute lupus pneumonitis in, 355 age at onset, 343 alopecia associated with, 352 American College of Rheumatology criteria for, 342, 343t androgenic hormone deficiency in, 348 animal models of, 348-349 antiphospholipid syndrome in, 357-358, 380 aphthous stomatitis in, 352 apoptosis in, 347 arthritis in, 352 aseptic necrosis in, 352, 353f atheromata risks in, 355 atherosclerosis and, 382 autoimmune endocrinopathies and, 360 autologous stem cell transplantation for, 126 B cell abnormalities in, 346 cardiac manifestations, 354-355 central nervous system disease associated with description of, 353-354, 354t laboratory examination for, 366-367 management of, 380 chemicals and, 348 chorea in, 354 clinical manifestations of, 349-360 clinical presentation of, 349 coagulation abnormalities in, 368-369 cognitive impairments in, 354 complement deficiencies and, 25 coronary artery disease and, 354-355 cotton-wool spots in, 358, 358f, 366 course of, 381-382 cutaneous manifestations of, 349-352, 350f-351£ definition of, 342 dendritic cells in, 347 diagnostic criteria of, 342, 343t dietary considerations, 376 diffuse proliferative glomerulonephritis and, 363, 363f-364f, 364t discoid lesions in, 351, 351£ drug-induced, 348, 348t, 382-383 dyslipoproteinemia associated with, 381 environmental factors associated with, 348 epidemiology of, 343-344 etiology of, 346-349 features of, 342, 349t focal segmental proliferative glomerulitis, 362f-363~ 362-363 frequency of, 6t gastrointestinal manifestations of, 359-360 gender ratio, 5, 343 general assessments of, 372, 373t-375t genetic background of complement deficiencies, 344-345 cytokine polymorphisms, 345 description of, 344 family studies, 344 Fc receptor polymorphisms, 345 histocompatibility antigens, 344 PD-1 polymorphisms, 345-346 T-cell receptor polymorphisms, 345 twin studies, 344 geographic influences, 343-344 glomerular sclerosis and, 364-365 glomerulonephritis and, 361-365 headaches in, 354 hematologic abnormalities in anemia, 367-368, 368f description of, 358
790
INDEX
Systemic lupus erythematosus (Continued) laboratory examination of, 367-368 leukocytes, 368 platelets, 368 hepatic disease and, 360 hepatomegaly and, 360 historical review of, 342 hormonal factors, 347-348 immune complexes in, 346 immune dysregulation associated with, 346-347 incidence of, 343 infections and, 348, 376, 381 interstitial nephritis and, 365 laboratory examination for antibodies to extractable nuclear antigens, 370-371 anticardiolipin antibodies, 368t anti-dsDNA antibodies, 369t, 369-370 antiglobulins, 371 antihistone antibodies, 371, 371t anti-La/SS-B antibodies, 370, 370t antinuclear antibodies, 369-371 antiphospholipid antibodies, 368-369 anti-Ro/SS-A antibodies, 370, 370t anti-Sm antibodies, 370-371, 371t anti-VI RNP, 371, 371t complement, 371 immune complexes, 371 inflammatory fluids, 372 inflammatory markers, 367 leukocytes, 368 lupus anticoagulants, 368 rheumatoid factors, 371 synovial fluid, 372 urinalysis, 372 Libman-Sacks endocarditis in, 355, 366,366f livedo reticularis, 350-351 lungs and, 365-366 lupus nephritis associated with, 352-353, 353t, 361t, 372, 380-381 lymphatic tissue involvement in, 360 maculopapular rashes in, 350 malar erythema associated with, 350f malignancy and, 360 membranous glomerulonephritis and, 363--364, 364f morbidity of, 381-382 mucosal involvement in, 352, 352f murine models of, 348-349, 349t musculoskeletal disease in, 352, 353f myocarditis in, 354 narcolepsy in, 354 necrotizing angiitis and, 365 neuropsychiatric manifestations of, 353, 353t neutrophil abnormalities in, 347 ocular disease in, 358-359 osteonecrosis in, 381 pancreatitis and, 359-360 pathogenesis of, 346-349 pathologic findings, 360-361 pericarditis in, 354 periungual erythema associated with, 350-351, 35lf pleural effusions in, 355 pleuropulmonary disease in, 355t-356t, 355-356 polyarthritis vs., 265 prevalence of, 343 prognosis for, 381-382 prolactin levels and, 213, 348 pulmonary hemorrhage in, 355-356 racial influences, 343--344 rash associated with, 349-350, 350f
Raynaud's phenomenon in, 358 renal lesions, 361-365 seizures in, 354 selective IgA deficiency in, 649-650 sepsis in, 382 sex hormones associated with, 347-348 Sjogren's syndrome in, 359, 359t skin manifestations of, 349-352, 350f-351f, 365,366f splenomegaly and, 360 sun exposure and, 376 T-cell abnormalities in, 347 thrombotic thrombocytopenia purpura in, 358,380 treatment of autologous bone marrow transplantation, 380 azathioprine, 378, 378t biological agents, 379-380 cyclophosphamide, 378t, 379 cyclosporine, 379 general measures for, 372, 374-376, 375t glucocorticoids, 376-378 hydroxychloroquine, 376 immunosuppressive agents, 378-379 intravenous immunoglobulin, 379-380 methotrexate, 379 methylprednisolone, 376-377, 377t monoclonal antibodies, 380 mycophenolate mofetil, 379 nonsteroidal anti-inflammatory drugs, 376 pharmacologic, 376-380 prednisone, 376-377, 377t ultraviolet irradiation effects, 348 valvulitis in, 355 vasculitis in, 350, 358, 505 viral infections and, 348 Systemic Lupus Erythematosus Disease Activity Index, 181, 372, 373t-374t Systemic Lupus International Collaborating Clinics Damage Index, 372, 375t Systemic scleroderma classification of, 442, 443t, 453 diffuse cutaneous. See Diffuse cutaneous systemic scleroderma limited cutaneous, 463--464, 464f-465f myositis in, 423 Systemic sclerosis sine scleroderma, 442
T T-cellCs) abnormalities of primary, 646-648 in systemic lupus erythematosus, 347 activation of activation-induced cell death effects on, 44,45f description of, 34-36 infection-induced, 49 regulation of, 43--44 anergic, 49, 119 antigen recognition, 32-33 antigen-presenting cell interactions with, 34 antigen-specific, 47 apoptosis of, 46 autoreactive, 119 B cell interactions with, 40-42 Bcl-2 expression on, 347 CD4+, 31, 33, 36, 46, 212 CD8+
description of, 33, 40f, 212 juvenile psoriatic arthritis and, 325-326 CD40 ligand on, 34
central tolerance of, 46 cytotoxic apoptosis induced by, 39 description of, 28, 31-32 effector functions of, 39 function of, 28 description of, 28 development of, 33--34 effector, 43 Fas expression by, 44 FasL expression by, 44 glucocorticoids effect on, 105 helper. See Helper T-cells homeostasis of, 43--44 immunodeficiencies of, 647t, 648 inducible costimulator on, 34 inhibitory regulatory, 46 interleukin-2 production and, 34 in juvenile rheumatoid arthritis pathogenesis, 211 memory, 43 peripheral tolerance of, 46-47, 47f regulatory, 647 selection of, 33-34 self-tolerance, 31 with suppressor activity, 28 synovial fluid, 212 in systemic arthritis, 292 T-cell receptor aggregation of, 34 altered peptide ligands, 37 degeneracy, 36-37 description of, 28, 31-32 polymorphisms of description of, 212 in systemic lupus erythematosus, 345 structure of, 32f T-cell activation by, 34 vaccines, 120 T score, 719 t test nonparametric, 166-167 one-sample, 164-165 Student's, 164, 166 Tag single-nucleotide polymorphisrns, 66 Takayasu's arteritis, 337, 493t-494t, 548-550, 55lf TAP2B,215 Tartrate-resistant acid phosphatase, 717, 717t Telangiectasias in cutaneous neonatal lupus erythematosus, 397 in diffuse cutaneous systemic scleroderma, 446, 447f-448f Temporal arteritis, 493t-494t, 550-551 Temporomandibular joint description of, 9 juvenile rheumatoid arthritis of, 229, 229f Tendons, 15 Tennis elbow, 682 Tenosynovitis description of, 683 in juvenile rheumatoid arthritis, 218 in systemic arthritis, 293 Teratogenicity of leflunomide, 99 of methotrexate, 95-96 of thalidomide, 102 Testosterone, 716 Th1 cells, 31, 37 Th2 cells, 31, 37 Thalassemia minor, 635 Thalidomide, 102, 645 Therapeutic confirmatory study, 151t
INDEX The~peutic exercises, 19S-196 Ther3fpeutic exploratory studies, 150, 15lt Therapeutic use study, 15lt Thib*rge-Weissenbach syndrome, 463 Thigh exercises, 193f ThioJ1lurine methyltransferase, 114 Throlnbocytopenia, 398-399 Thrombotic thrombocytopenia purpura de$Cription of, 358 plasmapheresis for, 380 Thy~ocytes, 33, 35f Thymopentin, 238 Thyrpid acropachy, 637 T1 aI).tigens, 39 Tide1Tlark, 10 Tie~e's syndrome, 692 Timlj to peak concentration, 150 Tinel's sign, 693 Tissue inhibitor of metalloproteinase, 13 Tola~oline, 461t Tolerance central, 4>-46 peripheral, 46 Tolerogens, 45 Toll-like receptors, 21-22, 22t Tolmetin sodium dosing of, 82t ju!venile rheumatoid arthritis treated with, 233 structure of, 77f tqxicities associated with, 83t Torticollis, 692 Toxic epidermal necrolysis, 506, 507f Toxic-oil syndrome, 455
Toxi?plRsmagondii,41O Tral!>ecular bone, 716 Transcriptome, 71 Tramscutaneous electrical nerve stimulation, 192 Translational research, 143 Transmission disequilibrium testing, 67, 69 Transplantation bone marrow, 126, 380 high-dose immunotherapy with, 126 stem cell description of, 126 diffuse cutaneous systemic scleroderma treated with, 459-460 juvenile dermatomyositis treated with, 431-432 juvenile rheumatoid arthritis treated with, 240 severe combined immunodeficiency treated with, 647 systemic arthritis treated with, 300 Tr::juma acute chondrolysis of hip, 688 arthritis caused by, 687--688 frostbite arthropathy, 689, 689f juvenile rheumatoid arthritis and, 213 osteochondritis dissecans, 686--687, 687f physical abuse, 688-689 $lipped capital femoral epiphysis, 688 Tr¢atrnent effect, 156 Trendelenburg sign, 411 Treponema pallidum, 584 Trevor's disease, 762 Triamcinolone hexacetonide description of, 112 juvenile rheumatoid arthritis treated with, 236 Trichinella spiralis, 421 Trichinosis, 421 Trlchorhinophalangeal dysplasia, 755
Trimethoprim-sulfamethoxazole, for Wegener's granulomatosis, 544t Trisomy 8, 747t Trisomy 5q, 747t Tropomyosin B, 15 Troponins, 15 True negative, 147 True positive, 147 Tuberculous arthritis, 574, 574f-575f Tumor necrosis factor-ex, 27t biologic agents against, 7, 123-124 description of, 55-56 etanercept effects on, 122 inflammation role of, 56 synthesis of, 56 in systemic arthritiS, 292 systemic lupus erythematosus and, 345 Tumor necrosis factor-ex inhibitors diffuse cutaneous systemic scleroderma treated with, 459 juvenile dermatomyositis treated with, 431 juvenile psoriatic arthritis treated with, 326 juvenile rheumatoid arthritis treated with, 237 Tumor necrosis factor-a, 27t Tumor necrosis factor receptor-associated death domain, 56 Tumor necrosis factor receptor-associated factors, 41, 56 Tumor necrosis factor receptor-associated periodic syndrome clinical manifestations of, 660t, 662--663 description of, 659 diagnosis of, 663 etanercept for, 663 genetics of, 662 laboratory studies for, 663 pathogenesis of, 662 skin manifestations of, 662 treatment of, 663 Tumor necrosis factor receptors, 56 Turner's syndrome, 216 Two-tailed hypothesis, 144 Type 1 error, 162 Type I hypersensitivity, 50, 5lt Type II error, 162 Type 1I hypersensitivity, 50-51, 51f, 5lt Type III hypersensitivity, 51, 5lt Type IV hypersensitivity, 5lt, 52
u Ulcerative colitis. See also Inflammatory bowel disease antineutrophil cytoplasmic antibody in, 337 gastrointestinal manifestations, 335 genetic background of, 334 incidence of, 334 laboratory examination of, 337 Ultraviolet irradiation, 348 Unacceptableness bias, 159 Unicameral bone cyst, 735 Uniparental disomy, 67
Ureaplasma urealyticum, 576 Uveitis antinuclear antibodies in, 281-282, 328 band keratopathy, 280, 282f, 283 in Beh\;et's syndrome, 563 clinical manifestations of, 279-280 course of, 283 description of, 278
791
differential diagnosis, 281 epidemiology of, 279, 279t etiology of, 279 genetics of, 279 glucocorticoids for, 282 histopathology of, 280-281 in inflammatory bowel disease, 337 in juvenile psoriatic arthritis, 328-329 Kawasaki disease and, 528 laboratory examination of, 281-282 management of, 282-283 monitoring of, 281t pathogenesis of, 279 pathology of, 280-281 prognosis of, 283 slit-lamp biomicroscopy of, 280, 281f
v Validity content, 175t convergent, 175t criterion, 175t external, 158 face, 175t internal, 158 predictive, 175t Valvulitis, 355 Variables categorical, 157 continuous, 157-158 dependent, 161 independent, 161 nominal, 157 ordinal, 157 statistical, 161-162 Variance, 160 Varicella, 200 Varicella-zoster infection, 583 Vascular adhesion molecule-I, 52 Vascular endothelial growth factor, 57 Vasculitis antineutrophil cytoplasmic antibodyassociated small-vessel, 504 in Beh\;et's syndrome, 563 classification of, 492 clinical aspects of, 492 cryoglobulinemic, 504 definition of, 492 epidemiology of, 492, 495 in familial Mediterranean fever, 505 familial Mediterranean fever and, 518 hypocomplementemic urticarial, 503-504 immune complex, 361f incidence of, 492, 495 in inflammatory bowel disease, 337 in juvenile dermatomyositis, 413 large-vessel, 493t malignancy and, 506 medium-vessel, 493t Mucha-Habermann disease, 505 prevalence of, 492, 495 relapsing polychondritis, 50S-506, 506f retinal, 358, 359f rheumatoid. See Rheumatoid vasculitis small-vessel antineutrophil cytoplasmic antibodyassociated, 504 classification of, 493t in systemic lupus erythematosus, 350, 358, 505 Velocardiofacial syndrome, 216 Very numerous tandem repeats, 65, 67, 69
792
INDEX
Viral arthritis alphaviruses, 582-583 hepatitis B arthritis-dermatitis syndrome, 582 herpesviruses, 583 human immunodeficiency virus, 583 mumps virus, 583 overview of, 58(}-581 parvovirus, 582 rubella virus, 581-582 syndromes associated with, 583-584 viruses that cause, 581t Visual analogue scale, for pain assessment, 702 Vitamin D deficiency of, 630, 708t description of, 11 0 metabolism of, 632f in peripubertal girls, 719 rickets caused by deficiency of, 630 Volume of distribution, 76 von Willebrand's disease, 636
w Water exercises, 198-199 Weaver syndrome, 747t Wegener's granulomatosis antineutrophil cytoplasmic antibodies in, 541, 542f, 542t
classification of, 539, 540t clinical manifestations of, 540, 541t clinicopathologic characteristics of, 494t course of, 543-545 cyclophosphamide for, 542, 544t definition of, 493t diagnosis of, 54(}-541 epidemiology of, 539 etiology of, 539--540 familial occurrence of, 539--540 genetic factors in, 539--540 glucocorticoids for, 542-543 incidence of, 539 laboratory examination in, 541£ methotrexate for, 543 methylprednisolone for, 542 pathologic findings in, 541, 543f prevalence of, 539 prognosis for, 543-545 radiologic examination for, 541-542, 543f saddle-nose deformity associated with, 541£ treatment of, 542-543 trimethoprim-sulfamethoxazole for, 544t Weighted kappa test, 169 Weill-Marchesani syndrome, 747t Werner's syndrome, 455 Whipple's disease, 580
Wide-range dynamic neurons, 697-{i98 Williams' syndrome, 677t Wilms' tumor, 739 Winchester's syndrome, 679t, 747t, 760 Windup, 699 Wiskott-A1drich syndrome, 647t, 648 Wolcott-Rallison dysplasia, 755 Wrist exercises, 191£ Wrist splints, 198, 198f, 241
x Xerostomia, 451 X-linked agammaglobulinemia, 65Q-{i51
y Yates continuity correction, 165-166 Yeast two-hybrid screens, 73 Yersinia infection, 606
z Z scores, 16(}-161, 719 Zellweger's syndrome, 747t Zero-order kinetics, 77