Source: GUIPCAR Group. In: Clinical practice guideline for the management of rheumatoid arthritis. Madrid: Spanish Society of Rheumatology; 2001. 146 p. GUIPCAR CLINICAL PRACTICE GUIDELINE FOR THE MANAGEMENT OF RHEUMATOID ARTHRITIS - Translated from the Spanish Contents * PARTICIPATING INSTITUTIONS * HOW TO CITE THIS GUIDELINE * GUIPCAR GROUP * ACKNOWLEDGEMENTS * SUMMARY * PREFACE * BACKGROUND * GUIDELINE OBJECTIVES * METHODOLOGY * HOW TO USE THIS GUIDELINE * CONTENTS AND LIMITATIONS OF THE GUIDELINE * CHAPTER 1. DIAGNOSING RHEUMATOID ARTHRITIS * CHAPTER 2. INITIAL EVALUATION * CHAPTER 3. CLASSIFYING RHEUMATOID ARTHRITIS * CHAPTER 4. MEDICAL TREATMENT OF RHEUMATOID ARTHRITIS * CHAPTER 5. CRITERIA FOR RESPONSE TO TREATMENT * CHAPTER 6. ADVERSE EFFECTS OF DRUGS USED IN THE TREATMENT OF RHEUMATOID ARTHRITIS * CHAPTER 7. OTHER TREATMENTS * CHAPTER 8. EXTRA-ARTICULAR COMPLICATIONS OF RHEUMATOID ARTHRITIS * ACRONYMS * APPENDICES * TABLES * REFERENCES * QUICK REFERENCE GUIDE Rheumatoid arthritis (RA) is a highly prevalent disease in Spain, and its management consumes a large amount of health and social resources. The disease often has serious repercussions on the quality of life of those affected, provoking chronic pain, limitations in the activities of daily living, and work disability. Although there is currently no cure for the disease, its symptoms can be controlled by early detection and the initiation of appropriate treatment. In recent years, however, the appearance of new drugs, new forms of combined treatment, and a plethora of studies on the efficacy of different therapeutic modalities means that the clinician is not always sure which treatment strategy to follow for a particular patient. Faced with this explosion of new information, it is not surprising that there is large variability in the treatment of patients with RA, which cannot always be explained by characteristics of the disease. Thus, the Spanish Society of Rheumatology (SER), aware of the difficulties involved in making decisions about the management of RA, decided to develop this Clinical Practice Guideline for the Management of Rheumatoid Arthritis in Spain (Guía de Práctica Clínica para el manejo de Artritis Reumatoide en España - GUIPCAR). The aim of the guideline is to help clinicians make decisions about their patients. The guideline recommendations are based on the best available scientific evidence and, when this is lacking, on the work of an expert panel that developed a series of detailed recommendations on the diagnosis, management, and treatment of patients with RA. The recommendations on disease-modifying antirheumatic drugs (DMARDs) are the result of an exhaustive review and synthesis of the published randomized clinical trials meeting strict methodological criteria for these types of studies. Clinical practice guidelines are one of the most appropriate tools for improving the quality of care and decreasing unnecessary variability in clinical practice. Their objective is to offer systematically developed recommendations to aid the physician and patient in decision making. Although these recommendations aim to include most patients who would present to a physician, there may be particular circumstances in which they do not apply. With this work, the SER hopes to offer the clinician a practical tool that will prove useful in daily clinical practice. We hope the information included here will be of assistance to physicians, will help improve the quality of care, and, above all, will help patients who suffer from RA. Should this be so, we will have achieved an important objective for the year 2001, designated by the Society as the "Year of Rheumatoid Arthritis." Armando Laffón Roca Emilio Martín Mola PARTICIPATING INSTITUTIONS This guideline is an initiative of the Spanish Society of Rheumatology. A number of institutions have collaborated in carrying out this initiative: Spanish Society of Rheumatology (Sociedad Española de Reumatología - SER). The SER promoted the idea for this guideline, chose the research group to develop it, helped select the panel of experts, sponsored its development, and presented the project to the financing organization. Health Services Research Unit (Unidad de Investigación en Servicios de Salud - UISS). When the SER decided to produce the guideline in late 1998, the Society proposed that it be developed by the UISS. At that time the UISS was a research unit within the Carlos III Health Institute (ISCIII). The UISS began to develop the guideline, but organizational changes in the ISCIII took place at the end of 2000. The UISS then became a private company with the name of TAISS, part of whose research staff came from the UISS. Ignacio de Mercado Foundation (Fundación Ignacio de Mercado - FIdeM) for research and education in the health services. The FIdeM contracted project personnel who were not on the staff of the UISS. Advanced Research Techniques in the Health Services (Técnicas Avanzadas de Investigación en Servicios de Salud, S.L. - TAISS). TAISS is a company devoted to producing knowledge to improve decision making in the health sector at the macro (policy) level, as well as at the meso (management) and micro (physician-patient) levels. Its research staff came from the UISS. All the investigators who participated in the project at its inception have continued to work on it. Novartis. Novartis is the organization that financed the development of this guideline. It also oversaw each project activity and, together with the SER, monitored project tasks to ensure they were carried out in a correct and timely fashion. Abbott. This English-language version of GUIPCAR was made possible thanks to Abbott, which provided financing for the translation. HOW TO CITE THIS GUIDELINE The following format is suggested for citation of this guideline: GUIPCAR Group. Clinical Practice Guideline for the Management of Rheumatoid Arthritis in Spain. Spanish Society of Rheumatology. Madrid, 2001. GUIPCAR GROUP The following persons are members of the GUIPCAR Group, which authored this guideline: 1. Principal Investigator Pablo Lázaro y de Mercado, Director of TAISS. Founder and director of the Health Services Research Unit (UISS) of the Carlos III Health Institute (1993-2000). Vice-Director General of Health and Technological Evaluation of the Ministry of Health and Consumer Affairs (1997-98). University graduate in Medicine (Complutense University of Madrid, 1973). Doctor of Medicine (Autonomous University of Madrid, 1989). Specialist in Internal Medicine and in Respiratory Diseases (1977). Residency in Respiratory Diseases at the 12 de Octubre Hospital in Madrid (1974-77), and associate in Respiratory Diseases at the Ramón y Cajal Hospital in Madrid (1978-86). Master of Business Administration from the IESE (1989). Post-doctoral studies in health policy analysis at the RAND/UCLA Center for Health Policy Analysis, Santa Monica, California (USA), where he helped develop clinical practice guidelines (1990-93). His areas of expertise include research in the health services, socioeconomic evaluation, medical technology evaluation, development of clinical practice guidelines, and development of appropriateness criteria for clinical procedures. 2. Expert Panel The experts who developed the recommendations in this guideline, listed in alphabetical order, are: José Luis Andréu Sánchez, rheumatologist, Clínica Puerta de Hierro, Madrid. University graduate in Medicine and Surgery (Autonomous University of Madrid, 1983), specialist in Rheumatology via the MIR (Puerta de Hierro Clinic, Madrid, 1984-87) and Doctor of Medicine with special honors (Autonomous University of Madrid, 1990). Has participated in different research projects related with Immunology at the Center of Molecular Biology (Professor Martínez-Alonso) and has been a member of various expert groups (e.g., the Strategic Plan of the Spanish Society of Rheumatology and the Consensus Conference on the use of biological therapies in rheumatoid arthritis) sponsored by the Spanish Society of Rheumatology, of which he was Secretary (1998-2000). Formerly or currently serving on the editorial boards of various clinical journals such as Annals of the Rheumatic Diseases, Revista Española de Reumatología and Revista Clínica Española. Has published over 100 book chapters, 54 articles in national scientific journals, and 30 articles in the most prestigious international journals such as Nature, Journal of Experimental Medicine, and Arthritis and Rheumatism. Editor of 8 books. Has given over 40 post-graduate seminars on different subjects related with his speciality. Currently works as a rheumatologist at the Puerta de Hierro Clinic in Madrid, is honorary professor of Rheumatology at the Faculty of Medicine of the Autonomous University of Madrid and the Hospital for Rheumatic Diseases of Barcelona, and is President of the Society of Rheumatology of the Community of Madrid. Enrique Batlle Gualda, rheumatologist, University Hospital General of Alicante, Alicante. University graduate in Medicine and Surgery by the Autonomous University of Barcelona (1978), specialist in Rheumatology via the MIR (1984), Doctor of Medicine (University of Alicante, 1992), and University Specialist in Health Economics and Management of Health and Social Services (University of Alicante, 1993). Trained in epidemiology and statistics. Experienced as a teacher, evaluator, and advisor for different agencies, organizations, and research groups. Research experience in brucelar arthritis and clinical epidemiology in the evaluation of health-related quality of life, clinical methodology, and clinical trials. Currently associate in the Rheumatology Section and Secretary of the Clinical Research Ethics Committee of the General University Hospital of Alicante, and Associate Professor of the Miguel Hernández University. Loreto Carmona Ortells, rheumatologist and epidemiologist, Spanish Society of Rheumatology, Madrid. University graduate in Medicine and Surgery (Autonomous University of Madrid, 1990) and specialist in Rheumatology via the MIR (La Princesa Hospital, Madrid, 1991-94). Carried out the project "Clinical Epidemiology and Application to the Study of Rheumatic Diseases" (FIS BAE grant 96/5485 and 97/5090) in the Arthritis Research Group (directed by Edward Yelin) and the Departments of Rheumatology and Epidemiology and Biostatistics of the University of California in San Francisco. Has worked in the Spanish Medicines Agency (grant FC1 1999- 2000) as technical evaluator for various recently developed drugs in rheumatology. Coordinator since 1998 for various epidemiological projects of the Spanish Society of Rheumatology. Currently contracted as investigator of the Health Research Fund in the Research Unit of La Princesa Hospital to develop a line of Research in Rheumatic Diseases and coordinate the installation of a Cochrane Centre in Madrid for the diffusion of Evidence-Based Medicine. Federico Díaz González, rheumatologist, University Hospital of the Canary Islands, Santa Cruz de Tenerife. University graduate in Medicine and Surgery (La Laguna University, 1986), specialist in Rheumatology via the MIR (La Princesa Hospital, 1991), and Doctor of Medicine from the Autonomous University of Madrid (1994). Post-doctoral training under grants from the Ministry of Health and the Spanish Ministry of Education and Science at the Laboratory of Dr. Mark H. Ginsberg in the Department of Vascular Biology of the Scripps Research Institute, La Jolla, California. Has published 28 articles in Spanish and international journals, has authored 2 book chapters and has wide experience in research. Formerly, specialist in the Rheumatology Department of La Princesa Hospital of Madrid, and currently occupies the same position in the University Hospital of the Canary Islands. Juan José Domínguez Reboiras, orthopedist, La Paz University Hospital, Madrid. University graduate in Medicine and Surgery by the Santiago de Compostela University (1968); Doctor of Medicine and Surgery from the Autonomous University of Madrid (1974); specialist in Orthopedic Surgery and Traumatology, in Physical Education and Sports Medicine and in Occupational Medicine, and Advanced Degree in the Management of Health Institutions. Tenured Professor in Anatomy and Physiology in the La Paz University School of Nursing. Formerly, Chief of the Orthopedic Surgery and Traumatology Department in the Xeral Cies Hospital of Vigo. Currently, Chief of the Shoulder and Elbow Diseases Unit in the La Paz University Hospital of Madrid. Ángel Elena Ibáñez, rheumatologist, San Millán-San Pedro Hospital, Logroño. University graduate in Medicine and Surgery (Autonomous University of Madrid 1979), specialist in Rheumatology via the MIR (Ramón y Cajal Hospital, Madrid, 1980-1984), and Doctor in Medicine and Surgery (Autonomous University of Madrid, 1994). Area specialist of the Rheumatology Section, San Millán-San Pedro Hospital of La Rioja, since 1987. Degree in Clinical Research (Autonomous University of Madrid, 1997 and National School of Health, 1999). Formerly responsible for the Clinical-Epidemiological Research Unit of La Rioja from 1995 to 1999. Currently working in the Rheumatology Section of the San Millán-San Pedro Hospital of La Rioja, Logroño. Juan Gómez-Reino Carnota, rheumatologist, University Clinical Hospital, Santiago de Compostela. Residency in Medicine and Fellowship in Rheumatology at the State University of New York at Stony Brook (USA). Previously Chief of Section of Rheumatology and Director of the Research Centre of the 12 de Octubre University Hospital of Madrid. Formerly responsible for the Area of Biomedicine at the National Agency of Planning and Evaluation (ANEP) and member of evaluation committees for national and international research. Has published numerous articles in speciality journals in the area of autoimmune diseases and inflammation. Currently, Chief of the Rheumatology Department in the Santiago University Clinical Hospital. Agustún Gómez de la Cámara, epidemiologist, Research Unit, 12 de Octubre Hospital, Madrid. Doctor in Medicine and Surgery from the Autonomous University of Madrid. Currently, Chief of the Clinical Epidemiology Unit of the Research Unit in the 12 de Octubre Hospital. His training in clinical epidemiology was received, among other centres, at the Houston Medical Center, University of Texas (USA). Has worked mainly in teaching and research in the Health Research Fund, Carlos III Health Institute, and 12 de Octubre Hospital in diagnostic, prognostic, and therapeutic research areas from the perspective of clinical epidemiology. Actively promotes the use and development of Evidence-Based Medicine and has translated its basic texts. Technical advisor to the Health Research Fund and ANEP, and member of the Spanish Medicines Agency as advisor to the Committee on Drug Safety. Nuria Guañabens Gay, rheumatologist, Clinical and Provincial Hospital, Barcelona. University graduate in Medicine and Surgery (1978), specialist in Rheumatology (Clinical Hospital, Barcelona, 1979-1984), and Doctor of Medicine (University of Barcelona, 1987). Responsible for the Bone Metabolic Disease Team of the Agust Pi I Sunyer Biomedical Research Institute (IDIBAPS), Clinical Hospital, Barcelona. Currently, medical rheumatology adviser at the Clinical Hospital and Associate Professor of Medicine of the University of Barcelona. César Hernández García, rheumatologist, San Carlos Clinical Hospital, Madrid. University graduate in Medicine and Surgery (Complutense University of Madrid, 1987), Doctor of Medicine (Complutense University of Madrid, 1996, special honors), and specialist in Rheumatology via the MIR (San Carlos Clinical Hospital, Madrid, 1988-91). Visiting Fellow at the Division of Arthritis and Rheumatism, Oregon Health Sciences University. FIS fellow (1992-94) at the San Carlos Clinical Hospital, Madrid, and Area Specialist in the Rheumatology Department of that hospital since 1994. Current major research area is rheumatoid arthritis, involving both clinical research and health services projects. M^a Victoria Irigoyen Oyarzábal, rheumatologist, Carlos Haya General Hospital, Málaga. University graduate in Medicine and Surgery (University of Malaga, 1979) and specialist in Rheumatology via the MIR (Ramón y Cajal Hospital, Madrid 1981-1984). One year of specialization via the MIR in Anesthesiology (Carlos Haya Hospital, Malaga 1980). Area specialist in Rheumatology (Hospital of Navarra, Pamplona 1987-1992) and area specialist in Rheumatology (Carlos Haya Hospital, Malaga, Connective Tissue Diseases Unit, from 1992 to present). José Luis Marenco de la Fuente, rheumatologist, Hospital Universitario de Valme, Sevilla. University graduate in Medicine and Surgery (University of Sevilla,1974-80), specialist in Rheumatology via the MIR (Puerto de Hierro Clinic, Autonomous University of Madrid, 1981-84). Doctorate cum laude in 1986 for the thesis "Synoviorthesis with yttrium-90 in knees. Long-term results" (directed by Professor Noguera Hernando). Responsible for the creation of the Rheumatology Section in the Hospitals of Jerez (1987-91) and Valme of Sevilla (1992) and Chief of Rheumatology Section in the latter hospital since 1994. Associate Professor of Rheumatology in the Faculty of Medicine of Sevilla. Víctor Manuel Martínez Taboada, rheumatologist, Marqués de Valdecilla University Hospital, Santander. University graduate in Medicine and Surgery (University of Zaragoza, 1988) and specialist in Rheumatology via the MIR (Marqués de Valdecilla University Hospital, Santander, 1992). Completed clinical and research training in the Lupus Research Unit (The Rayne Institute, St. Thomas Hospital) in London (UK) and in the Rheumatology Research Unit (Mayo Clinic) of Rochester, Minnesota (USA). Doctor of Medicine from the University of Cantabria (1998). Since 1994, associate physician of the Rheumatology Department at the Marqués de Valdecilla University Hospital in Santander. Cristina Riera Riezu, rehabilitation therapist, Germans Trias i Pujol Hospital of Badalona, Barcelona. University graduate in Medicine and Surgery (Central University of Barcelona, 1983), specialist in Physical Therapy and Rehabilitation via the MIR (Germans Trias i Pujol University Hospital, 1989), and Doctor of Medicine (Autonomous University of Barcelona, 2000). Member of the Medical Commission of the International Para-Olympics Committee and of the Catalan and Spanish Sports Federations for the Physically Handicapped. Member of the Catalan Society of Rheumatology; received first prize from the Catalan Society of Rehabilitation Therapy for her work "Rehabilitation therapy in rheumatoid arthritis: protocol for action." Has published 12 articles in national journals and 8 in international journals, in addition to 93 conference contributions. Worked as associate physician for the Department of Rehabilitation Therapy at the General Hospital of Cataluña (1990-91), and is currently associate physician for the Department of Rehabilitation Therapy at the Germans Trias i Pujol University Hospital, where she is responsible for the Rheumatological Rehabilitation Unit. José María Salazar Vallinas, rheumatologist, Infanta Cristina Hospital, Badajoz. University graduate in Medicine and Surgery (Autonomous University of Madrid,1977), specialist in Rheumatology via the MIR (Ramón y Cajal Hospital, Madrid 1980-83), and in Family and Community Medicine (Ministry of Education and Science, 1987). Master of Public Health (National School of Health, Madrid, 1986). Formerly, President and Founding Member of the Rheumatological Association of Extremadura and spokesperson for the Spanish Society of Rheumatology, and responsible for residency training in Family and Community Medicine and member of the Teaching Commission (1988-95) and the Clinical Chart Commission (1990-1996) of the Infanta Cristina de Badajoz University Regional Hospital. Also Associate Professor of the Health Sciences in the Department of Human Clinical Pathology, Area of Medicine/Rheumatology, of the University of Extremadura. 3. Investigators The research team, originally from the UISS (most of whom currently work at TAISS), that was responsible for designing the study methodology, coordinating the work of the expert panel, carrying out the synthesis of the scientific evidence, and producing the final guideline document, was made up of the following persons: Setefilla Luengo Matos (UISS). Design of the synthesis of the evidence. Doctor of Medicine (Autonomous University of Madrid, 1990) and specialist in Preventive Medicine and Public Health (La Paz Hospital, 1985). Master of Public Health (National School of Health). Area Chief at the Agency for Health Technology Evaluation, Carlos III Health Institute, Madrid. José Manuel Estrada Lorenzo (TAISS). Bibliographic search. University graduate in Geography and History from the Complutense University of Madrid (1986), specializing in documentation in the Health Sciences. Santiago Alonso Corral (UISS). Database design. University graduate in Medicine and Surgery (University of Salamanca, 1987) and specialist in Preventive Medicine and Public Health (San Carlos Clinical Hospital, Madrid, 2001). Master of Public Health (National School of Health, 1997). Specialist in Health Information Systems (Complutense University of Madrid, 1994) and Master in Applied Electronics (School of Industrial Organisation, 1992). Miguel Ángel Abad Hernández (FIdeM). Analysis of the scientific evidence. University graduate in Medicine (Complutense University of Madrid, 1991) and specialist in Rheumatology via the MIR (Gregorio Marañón University General Hospital of Madrid, 1997). Hildegarda Godoy Tundidor (FIdeM). Analysis of the scientific evidence. University graduate in Medicine (Complutense University of Madrid 1994) and specialist in Rheumatology via the MIR (Gregorio Marañón University General Hospital of Madrid, 1998). Sandra García Armesto (UISS). Analysis of the scientific evidence. University graduate in Medicine (Autonomous University of Madrid) and Master of Public Health, specializing in Health Services Management and Evaluation, from the University Center of Public Health (CUSP) in Madrid. Yesenia Tordecillas Echenique (FIdeM). Analysis of the scientific evidence. University graduate in Medicine (University of Cartagena, Colombia, 1997). Has participated in various research projects in Colombian hospitals. Trained in the methodology of scientific research and the critical review of medical literature. Yira Tordecillas Echenique (FIdeM). Analysis of the scientific evidence. University graduate in Medicine (University of Cartagena, Colombia, 1996). Has participated in various research projects in Colombian hospitals. Trained in the methodology of scientific research and the critical review of medical literature. Milena Gobbo Montoya (TAISS). General coordinator and editor of GUIPCAR. University graduate in Psychology (Autonomous University of Madrid, 1984). Previous training and experience related with the practice of clinical psychology and the administration and management of hospital units. ACKNOWLEDGEMENTS This guideline is the fruit of collaboration among a number of institutions and persons, all of whom have helped to make it possible. The SER, for its initiative in promoting the development of this guideline, monitoring activities, and providing the meeting ground for strategic discussions. Novartis should receive special thanks, not only for its sensitivity in sponsoring the guideline, but also for not influencing its content or development except to ensure its timely completion. The investigators and panelists can thus confirm that their freedom of opinion and criteria was preserved so that this guideline is free of financing bias. The ISCIII made a significant contribution to making this guideline possible, both by carrying out research activities in the UISS, and for generously allowing the use of its installations for various research meetings, including those of the panel. TAISS, which assumed responsibility from the UISS for producing the guideline, contracted the UISS investigators to continue its development, and provided the site for various research meetings, including the last meeting of the panel. FIdeM, for handling the contracting of non-UISS staff. The panelists, who so generously dedicated their time, energy, and knowledge to producing this guideline, are responsible for developing the key parts of the guideline. The investigators, who overcame difficult moments and maintained their enthusiasm throughout all phases of the project, from its initial design, to the synthesis of the evidence, and the final assemblage of the pieces that make up this guideline. María Dolores Aguilar and Kathy Fitch, who provided suggestions in previous versions of the guideline, and Ignacio Lázaro, for his silent miracles in solving computer-related mysteries. The librarians at the National Library of the Health Sciences and at the libraries of the 12 de Octubre and the Ramón y Cajal Hospitals, who helped locate the documents necessary for the synthesis of the evidence. SUMMARY Rheumatoid arthritis (RA) affects some 200,000 persons in Spain, with 20,000 new cases emerging each year. RA reduces quality of life and functional capacity, produces work disability, and increases mortality. It has been estimated that the annual social cost of RA in Spain exceeds 200 billion pesetas (US$1.04 billion), of which 65 billion pesetas ($338.5 million) are devoted to health expenditures. There is evidence of large variability in the management of RA in Spain; this variability depends not only on patient or disease characteristics, but also on characteristics of the hospital, department, or physician providing patient care. These facts suggest that some diagnostic or therapeutic procedures are overused, while others may be underused. For these reasons, in 1998 the Spanish Society of Rheumatology (SER) decided to develop a clinical practice guideline for the management of rheumatoid arthritis (GUIPCAR) to help physicians make decisions about the diagnosis and treatment of patients with RA. The objective of GUIPCAR is to develop standards of quality for the treatment of RA and to reduce the variability that does not depend on patient characteristics. This guideline, aimed at rheumatologists, describes the diagnostic and management strategies for the evaluation and treatment of patients with RA. It focuses on RA in adults (excluding juvenile RA) and includes diagnosis, evaluation, prognosis, and treatments such as drugs, rehabilitation, and surgery. It does not cover other treatments such as acupuncture, and only briefly treats extra-articular complications of RA such as amyloidosis, anemia, or Sjögren's syndrome. The guideline begins with a description of its origin and justification in the preface and an explanation of its objectives. The methodology followed to develop the recommendations is then described. The recommendations for treatment with disease-modifying antirheumatic drugs (DMARDs) in this guideline are based on a synthesis of the best available scientific evidence, after making a systematic review of the clinical trials and meta-analyses that have been published. A total of 2,281 articles was identified, 103 of which met the inclusion criteria for this review. The rest of the recommendations or considerations are based on scientific evidence obtained without a systematic literature review, or on the opinions of the expert panel. The experts were chosen by the SER, applying a series of criteria designed to support their validity at the national level and to avoid conflicts of interest. The guideline describes how it can be used, its contents, and its limitations. Chapter 1 focuses on the diagnosis of RA, chapter 2 on the initial evaluation, based on which, in chapter 3, patients are classified according to the number of swollen joints and the presence of erosions. The classification is further broken down by acute phase reactants, the health assessment questionnaire (HAQ), and rheumatoid factor (RF). The recommendations on medical treatment are presented in chapter 4, the criteria for response to treatment in chapter 5, and the adverse effects of medical treatment in chapter 6. Surgical, rehabilitative, and local treatments are covered in chapter 7. Chapter 8 describes the extra-articular complications of RA. The guideline includes an appendix with instruments to facilitate the clinician's data collection for the initial classification and follow-up of patients with RA, and another appendix with the complete patient classification. To make it easier to follow the guideline recommendations, readers may wish to make use of a simplified algorithm or the other, complete algorithm. The key features of the guideline are contained in a summary version, the "rapid reference guide." The bibliography has been divided into two sections, one listing all the references provided by the panelists or the investigators without a systematic search, and the other listing all the references used in the systematic review. The Health Services Research Unit (Unidad de Investigación en Servicios de Salud - UISS) of the Carlos III Health Institute was commissioned by the SER to develop GUIPCAR. The project took 2 years to complete. During this time, the UISS became a private entity under the name of Advanced Research Techniques in the Health Services (Técnicas Avanzadas de Investigación en Servicios de Salud -TAISS). Novartis financed this project. The investigators established the necessary mechanisms to ensure that the guidelines would have no financing bias and that the persons involved in its development would have no conflicts of interest. PREFACE In 1998 the Spanish Society of Rheumatology (SER) undertook the development of a Clinical Practice Guideline (CPG) to help physicians make decisions about the diagnosis and treatment of patients with rheumatoid arthritis (RA). This initiative of the SER was in response to a phenomenon frequently seen in clinical practice: the large variability in the use of diagnostic, therapeutic, and rehabilitative procedures. RA is a disease with a relatively high prevalence, affecting from 0.2 to 0.8% of the population in Spain. The different specialties and levels of care involved in its treatment are not always well coordinated. Furthermore, recent years have seen rapid innovation in medical treatment, with the appearance of new drugs, new forms of combined treatment, and numerous studies on the effects of the drugs utilized. The large amount of information produced as a result of the ever-growing growing number of studies, their variable methodological quality, and the complexity of comparing the results of different studies constitutes a major obstacle for clinicians in keeping up to date on important knowledge in their field. Thus came about the project to produce a CPG for the management of RA (GUIPCAR), which was undertaken by the Health Services Research Unit (UISS) of the Carlos III Health Institute. This project took 2 years to complete. During this time, the UISS became a private company with the name of Advanced Research Techniques in the Health Services (TAISS). CPGs should be based on scientific evidence, but when such evidence is absent or contradictory, they may be complemented by unbiased techniques of handling expert opinion. This guideline uses both methods, and is based on three central tasks: 1) a synthesis of the evidence on the efficacy of disease-modifying antirheumatic drugs (DMARDs); 2) specific recommendations on the diagnosis, evaluation, and treatment of patients with RA made by a panel of experts, generally rheumatologists with scientific prestige proposed by the SER, following a methodology designed to produce CPG recommendations; and 3) assignment of the level of scientific evidence that supports each recommendation for treatment with DMARDs. The ultimate objective of this guideline is to aid clinical decision making by offering recommendations for the therapeutic management of RA that are based on the best available evidence. We have tried to present the recommendations in a clear and agreeable manner, so that it is easy to find the information sought. We hope it will prove useful to those who face the difficult task of making decisions about different courses of treatment and that, in the final analysis, it will result in better outcomes for patients with RA. BACKGROUND RA is a systemic disease of unknown etiology, which is characterized by chronic inflammation of the diarthrodial joints. It is frequently associated with severe morbidity, functional abnormalities involving work disability, reduced quality of life, and increased mortality. RA affects all populations, with an estimated prevalence of about 0.8% and an incidence of approximately 0.5/1,000 population per year [Silman, 1993]. In Spain, RA affects about 5 of every 1,000 adults [Villaverde, 2000] and causes 5% of all permanent work disability [Tornero, 1998]. The economic impact of this disease is reflected in direct costs (physician visits, diagnostic tests, drugs, hospitalization) and indirect costs (loss of income due to work disability). In one health district of Madrid it has been estimated that the mean annual cost of a patient with RA is 1.11 million pesetas (about US $5,700) Some 70% of these costs are direct costs - 32% for patient care and 38% not for patient care - and 30% are indirect costs, that is, lost hours of work. If these results are extrapolated to the whole Spanish population, based on the estimated prevalence of RA in this country, the annual social costs of this disease would reach more than 200 billion pesetas ($1.04 billion), of which 65 billion ($338.5 million) would be for health costs. As with most diseases, there is enormous variability in the management of RA - among countries, among institutions, among physicians, and even within the same physician - and in the use of diagnostic procedures, follow-up methods, and therapeutic and rehabilitative measures. At times it may be thought that health resources are used inappropriately, or that a variety of different recommendations are followed due to the large amount of information about the disease that appears in different publications or other channels of communication. Sometimes modifications in disease management are introduced even though they do not have a clear advantage over existing strategies, while at other times effective strategies may be ignored. Health professionals caring for RA patients are faced not only with a disease whose etiology, management, and diagnosis are complicated, but also with an enormous amount of information that is not always easy to interpret. CPGs are one of the most appropriate instruments to decrease unnecessary variability and improve clinical practice. A CPG is defined as a set of "systematically defined statements to assist practitioner and patient decisions about appropriate health care for specific clinical circumstances." A good CPG should have the attributes of validity, reliability, reproducibility, clinical applicability, flexibility, clarity, multidisciplinarity, and periodic review [IOM, 1990a; IOM, 1992]. CPGs are part of a cultural change which consists of moving from unsubstantiated confidence in the opinions of professionals to a more structured and well-founded support for clinical decisions. The US Institute of Medicine (IOM) has suggested various criteria for selecting the contents of a CPG: first, the guideline should be applied to a clinical condition for which there is the potential to improve health for a significant number of persons; second, it should reduce clinically significant variations in services and procedures; third, it should reduce clinically significant variations in the outcomes of health care; and fourth, policies for guideline development should reflect the needs and priorities of the health system. Following these criteria, the SER considers that RA is a clinical condition that would benefit from the development of a CPG. GUIDELINE OBJECTIVES The objectives of this guideline are to improve the quality of care, to reduce variability in the management of RA, to move toward the integral management of RA, and to align clinical practice more closely with the best available scientific evidence. Improving the Quality of Care The quality of care can be seen, defined, and measured in many ways [Donabedian, 1980; OTA, 1988; Lohr, 1990; Rubenstein, 1990; Chassin, 1991; Brook, 1991; Keeler, 1992; Kassirer, 1993; Burstin, 1993; Gates, 1993; Miller, 1993; Hayward, 1993]. The IOM defines it as "the degree to which health services for individuals and populations increase the likelihood of desired health outcomes and are consistent with current professional knowledge" [IOM, 1990b]. According to the characteristics implicit in this definition, quality refers both to the physician-patient (individual) level and to the social (population) level; it can be measured in probabilistic terms (quantification of uncertainty); and it is based on scientific knowledge. Donabedian, who conceives quality of care as based on three dimensions (structure, process, and result), emphasizes the degree to which technology is used well in different centers or places [Donabedian, 1988]. Evaluating the quality of care is a critical component of the correct functioning of health systems. Quality of care can be evaluated and can dramatically improve medical practice [Brook, 1990]. It is rarely evaluated, however, even though sufficient methodological instruments are available. Measuring outcomes is critically important in evaluating the quality of care. Two classic measures of the outcomes of clinical practice are efficacy and effectiveness. Efficacy refers to the patient outcome produced under ideal conditions (e.g., in a clinical trial), while effectiveness is the outcome produced under real conditions (e.g., in daily practice). The difference between efficacy and effectiveness can be produced by random error, by systematic error or bias (for example, the inventors of a drug or technology, consciously or subconsciously, may make it appear better than it really is), by variability in the quality of care, or by personal health characteristics that may or may not be controlled by the individual [Brook, 1990]. For this reason, the degree to which effectiveness (the effect achieved) approximates efficacy (the maximum achievable effect) can be considered as an indicator of the quality of care. This guideline will deal with both concepts. Implementing quality measures involves constructive actions to establish a relation between the process of care and patient outcomes [Brook, 1990]. For example, statistically significant and clinically important differences have been shown in clinical outcomes depending on whether procedures are applied with a good or poor process of care [Kahn, 1990]. The evidence showing that improving the process of care leads to better outcomes and that CPGs are one of the instruments designed to improve the process of care is one of the reasons behind the development of this guideline. Reducing Variability in the Management of Rheumatoid Arthritis There is evidence that decisions made in identical clinical situations may differ depending on individual, institutional, or geographic factors that are unrelated with patient characteristics [IOM, 1990a; IOM, 1992]. This does not mean that variability in clinical practice is unacceptable. It may be acceptable when there is uncertainty about the advantages of one technique over another, when patient characteristics or preferences are different, when the characteristics of the center or its resources are different, or because of changing science or social and individual values. Such variability is not acceptable, however, when it is due to inadequate medical skill, poor institutional organization, ignorance, or the deliberate decision not to use procedures for which there is proven evidence of their superiority. Unacceptable clinical practice does not benefit patients; it may harm them, and it consumes resources unnecessarily. In Spain there is large inter- and intra-hospital variability in the use of health resources, diagnostic procedures, and therapeutic procedures in patients with RA. For example, in one comparison among centers, the mean number of visits per person over a 2-year period ranged from a low of 5.1 (range 1-11) to a high of 10.2 (range 2-31). These differences remained after adjusting for disease severity and functional class, according to the preliminary results of the "Study of RA Management" (emAR). In the same study, the percentage of patients who underwent some orthopedic surgical procedure during the course of their disease ranged from 0 to 71%, depending on the center. Even though most patients were being treated with nonsteroidal anti-inflammatory drugs (NSAIDs), corticosteroids, and DMARDs, the use of specific drugs varied significantly, independently of the patient's clinical situation. That is, the variability observed cannot be explained solely by differences related with the disease itself, but is also associated with variables related with the hospital center, department, or physician responsible for the patient. This variability in RA management that cannot be explained by patient characteristics justifies the development of standards of quality for the treatment of RA. Moving Toward Integral Management of Rheumatoid Arthritis At any given time, a variety of specialists may collaborate in managing specific disease aspects, from diagnosis to treatment to follow-up. Primary care physicians, rheumatologists, radiologists, ophthalmologists, orthopedic surgeons, rehabilitation specialists, orthopedists, psychologists or psychiatrists, and even other, non-medical health personnel, should be aware of the need to unify criteria and to integrate care in accordance with the best available scientific evidence. One way to avoid RA patients' receiving contradictory messages from different specialists is the development of CPGs that are easy to understand, and with a strong enough scientific basis to earn the support of all specialists who help manage patients with this disease. Aligning Clinical Practice with the Best Available Scientific Evidence Clinical practice in RA treatment has varied considerably in recent years, despite the fact that therapeutic resources have remained almost constant. There is growing use of early and aggressive treatment, which is supported by evidence that such measures may delay disease progression. The controlled clinical trials showing the short-term efficacy of a treatment intervention in selected populations initially led to disparate treatment practices and options, which have not subsequently been supported by studies of effectiveness in persons with RA. For example, in clinical trials of short-term efficacy, the efficacy of all the available DMARDs is higher than that of placebo (PCB). In comparative studies of a similar duration, it is difficult to find evidence that one DMARD is better than others. In studies evaluating compliance with long-term treatment, the number of patients who continue taking a specific DMARD after 5 years is significantly higher for methotrexate. This type of evidence has led to the generalized use of methotrexate as the drug of choice in most centers that care for these patients. There is insufficient scientific to support certain clinical decisions, however, and not all the scientific evidence is within easy reach of clinicians. Furthermore, the evidence available is of varying methodological quality, thus its scientific validity may be more or less solid. The use of more highly developed instruments to analyze information may facilitate changes in clinical practice as important as that of early intervention, based on the best available scientific evidence. Thus, this guideline is an attempt to base the process of care on the best available scientific evidence and, where this does not exist or is contradictory, applies the collective judgments of an expert panel selected according to specific criteria and using a scientific methodology developed to avoid biases. METHODOLOGY 1. Synthesis of the Evidence 1.1 Objective The synthesis of the evidence in the development of CPGs has three objectives: 1) to base the recommendations on the best available scientific evidence; 2) to allow users to obtain the information on which the recommendations are based; and 3) to make it possible to evaluate the quality of the guideline. One problem in carrying out a synthesis of the evidence is that the amount of information may be so extensive as to be impossible to cover. There are more than 35,000 medical journals in the world, which publish over 3 million articles every year. In addition, these journals are published in many different languages, the designs of the studies may be very heterogeneous, the methodological quality is highly variable, the patients selected may be very different, the measure of results may refer to different outcomes, and different subjects may be treated for the same intervention (e.g., whereas one study focuses on clinical outcomes, others may focus on complications, utilization, physiopathological mechanisms, or costs). This makes it necessary to limit the search for scientific evidence. Limiting such searches is justified not only by real limitations on resources and time, but also because a search aimed specifically at key aspects of disease management may constitute one of the strengths of a CPG. This naturally depends on achieving an intelligent balance among the topics searched, the quality of the review process, and the available resources. Thus, once the large number of RA publications had been explored, it was agreed that, for this guideline, the synthesis of the evidence would focus on the efficacy of DMARDs. 1.2 Drugs included in the synthesis of the evidence After making an initial estimate of the volume of literature to be reviewed, the project investigators and the members of the expert panel agreed to focus the synthesis of the evidence on studies evaluating the efficacy of treatment with DMARDs (clinical trials) by means of a systematic review. Table 1 shows the DMARDs included in the review. Table 1. Disease modifying antirheumatic drugs included in the systematic literature review * Antimalarials: Chloroquine (CLQ) and Hydroxychloroquine (HCQ) * Azathioprine (AZA) * Cyclophosphamide (CPA) * Cyclosporin A (CSA) * D-penicillamine (DP) * Anti-tumor necrosis factor (anti-TNF) agents: infliximab (IFM) and etanercept (ETN) * Leflunomide (LEF) * Methotrexate (MTX) * Gold salts: Oral gold (OG) and injectable gold (IG) * Sulphasalazine (SSZ) 1.3 Search strategy for identifying the scientific evidence The literature search for the synthesis of the evidence on the efficacy of DMARDs in RA was made in four databases, for the time periods noted below: * MEDLINE: 1966-2000 * EMBASE (Drugs and Pharmacology section): 1984-2000 * Spanish Medical Index (Indice Médico Español - IME): 1971-2000 * Cochrane Library: year 2000 version. The articles for the bibliographic search had to meet the following inclusion criteria: 1) controlled clinical trial, meta-analysis, or systematic review; 2) study referring to one or more of the selected DMARDs; 3a) comparison of the efficacy of a drug or combined therapy vs. another drug or combined therapy including at least one of the drugs listed in Table 1, or comparison of LEF or TNF with placebo; 3b) if a systematic review or meta-analysis, comparison of a drug or combined therapy vs. another drug or combined therapy including at least one of the drugs listed in table 1, or with placebo; 4) study carried out in patients with RA; 5) trial carried out in humans; and 6) published in English or Spanish. The descriptors used were those specified in each database with regard to the research methodology (e.g., RANDOMIZED-CONTROLLED-TRIAL), rheumatoid arthritis (e.g., RHEUMATOID ARTHRITIS), and specific drugs (e.g., METHOTREXATE ). 1.4 Selection of articles We first identified all existing systematic reviews on DMARDs. Nine systematic reviews were located in the Cochrane Library that compared placebo with the following drugs: methotrexate, sulphasalazine, cyclosporin, oral gold salts, injectable gold salts, cyclophosphamide, d-penicillamine, azathioprine, and anti-malarials. Using the search strategy described, we then located 982 bibliographic records in MEDLINE, 968 in EMBASE, 268 in the Cochrane Library, and 63 in IME, for a total of 2,281 records; of these, 666 were found in more than one database. Thus, the final number of records selected was 1,615. All the bibliographic records selected were read independently by two reviewers (a rheumatologist and a physician with training in epidemiology), both of whom had received training in how to carry out a synthesis of the evidence. The objective of reading the title of the bibliographic record was to verify that the article met the inclusion criteria. A 4-phase strategy was used to screen the records: 1. Review of the title. Resulting in the following two possibilities: a. The record was rejected because it did not meet the inclusion criteria. b. The record was accepted either because it met the criteria described or because it was not possible to decide whether or not it met the inclusion criteria based on the data provided. Of the 1,615 records selected, 289 were rejected during the first phase; thus, 1,326 bibliographic records went on to the second phase. 2. Review of the abstract. In those cases in which the record passed the first phase and had an abstract, the abstract was read, which led to two options: a. It was rejected because it did not meet the inclusion criteria. b. It was accepted, either because it met the criteria described or because it was not possible to decide whether or not it met the inclusion criteria based on the data provided. All the articles passing the first phase that did not have an abstract passed directly to the third stage. Of the 1,326 articles that passed to the second phase, 1,225 had an abstract and 101 did not. After reading the abstract, 1,039 were eliminated and 186 were accepted and passed to the third phase, together with the 101 records with no abstract. That is, 287 articles passed to the "request for articles" stage. In the first two stages of screening the records, each rheumatologist-methodologist team compared the records read and, in case of discrepancy, the two reviewers discussed the article. If no agreement was reached, the record was read by another pair of reviewers. Discrepancies between reviewers occurred in 4% of records, and agreement was reached in 100% of cases. 3. Request for articles. In this phase the articles that had not been rejected after phases 1 and 2 were requested. The requests were directed first to the National Library of the Health Sciences (Biblioteca Nacional de Ciencias de la Salud - BNCS) of the ISCIII, to which the UISS also belonged. If the article was not in the BNCS, it was requested from the library of a Madrid hospital, generally the 12 de Octubre Hospital or the Ramón y Cajal Hospital. If the article could not be found in any Madrid library, the Loan Service of the BNCS requested it through a biomedical library within Spain, and if it could not be found in any Spanish library of the health sciences, it was requested from the British Library or from the Netherlands Institute for Scientific Information Services (Nederlands Instituut voor Wetenschappelijke Informatiediensten - NIWI). 4. Data collection and evaluation of the quality of the evidence. The relevant information for each article was collected using a form designed with four objectives in mind: 1) to identify the bibliographic information included in the review; 2) to verify that the article, in its complete text, met the inclusion criteria for the review; 3) to collect the relevant clinical information for each article accepted; and 4) to evaluate the methodological quality and the level of evidence of the trials. Objective 1: To identify the bibliographic information for each article included in the review. General information was obtained on the relevant bibliographic data of the article (e.g., first author, year of publication, journal). Each article was coded to facilitate its location in the database. Objective 2: To verify that the article, in its complete text, met the inclusion criteria for the review. This verification process resulted in four possibilities: 1. All inclusion criteria were met, in which case the article was included. 2. At least one of the inclusion criteria was not met, in which case it was excluded. 3. The inclusion criteria were met, but the article was redundant. Articles were considered to be redundant if they were published on successive occasions, generally in a different journal from the first publication, but referred to the same clinical trial and did not provide additional information in comparison to the first publication. In this case, one of the publications was selected to complete the form with the study data, and the redundant one was excluded. 4. The inclusion criteria were met but the article was a complement to a previous one. Articles were considered to be complementary if, in comparison with another previous publication of the same trial, they included different outcome measures or, if they included the same outcome measures, these were collected for a different period of time (e.g., clinical variables in one journal and radiographic variables in another; or radiographic variables up to 6 months in one journal and up to 3 years in another). In these cases, the results of both publications were combined; the complete information from one of the articles was used to fill out the form, adding the new and complementary information provided by the other article on the same form. Redundant or complementary articles were identified a posteriori after reading the articles, by verifying that the study was written by the same authors, used the same guidelines for the treatments compared, and used the same study design. Objective 3: To collect the relevant clinical information from each article accepted. If the article met the inclusion criteria, data were collected on the number and characteristics of the participants (gender, age, mean time of disease progression, previous use of NSAIDs and DMARDs), interventions, outcome measures evaluated, persons lost to follow-up and withdrawals from the study, reliability of the measurement instruments used, follow-up of the groups, and statistical analysis. Objective 4: To assess the methodological quality and level of evidence of the trials. The methodological quality of the trials was assessed using the Jadad scale for rating the quality of clinical trials (Table 2 [Jadad, 1996a]. The level of evidence was assessed in accordance with the Hadorn scale designed to evaluate the quality of the evidence of publications used to develop CPGs (Table 3) [Hadorn, 1996]. Table 2. Jadad scale for rating the quality of evidence from clinical trials ------------------------------------------------------------------------ To rate the quality of a clinical trial, three questions are posed: 1. Was the study described as randomized? 2. Was the study described as double blind? 3. Was there a description of withdrawals and drop outs? ------------------------------------------------------------------------ One point is given for each "yes" and 0 points for each "no". There are no intermediate scores. An additional point is given in question 1 if the randomization method is described and is appropriate, and an additional point is given in question 2 if the method for making the study double blind is described and is appropriate. One point is subtracted in question 1 if the randomization method is described but is inappropriate, and one point is subtracted in question 2 if the study is described as double blind, but the blinding method is inappropriate. An article can receive a score of 0 to 5 points. An article is considered to be of good quality if the score is 3 or higher, and of poor quality if the score is less than 3. Table 3. Hadorn scale for rating the quality of scientific evidence from articles for CPGs 1. Well-conducted multicenter randomized controlled trials including 100 or more patients Level of evidence * A 2. Well-conducted randomized controlled trials with fewer than 100 patients, in one or more institutions Level of evidence * A 3. Well-conducted cohort studies * A 4. Well-conducted case-control studies Level of evidence * B 5. Poorly controlled or uncontrolled studies Level of evidence * B 6. Conflicting evidence in favor of the recommendation * B 7. Expert opinion * C Levels 1, 2, and 3 refer to a high level of evidence (A); levels 4, 5, and 6 refer to a level of evidence with potential biases that could invalidate the results (B); and level 7 is the evidence most vulnerable to potential biases (C). Since only clinical trials were evaluated in the synthesis of the evidence for this guideline, the levels of evidence assigned are A1 (1 on the Hadorn scale ), A2 (2 on the Hadorn scale), and B (5 on the Hadorn scale). Data collection form. A form was designed to collect the bibliographic data for each article, information about the study methodology, clinical data, the quality of the methodology, and the level of evidence. The first version of the form was evaluated by three reviewers (two rheumatologists and one methodologist) who applied it to 10 articles. After introducing the appropriate modifications, the reviewers began to use the form. It was necessary to change the form for later articles, however, in order to adapt it to the peculiarities of each trial (for example, the number of interventions compared, outcomes measured, and so on) and to be able to create homogeneous and unbiased evidence tables. After 13 successive versions, the definitive form was obtained, which was sufficiently valid to permit inclusion and categorization of the relevant information from each article. This form is available to interested readers (milena@taiss.com). Evaluation of the articles. Five reviewers read and evaluated the articles: two specialists in rheumatology and three in research methodology. All had been trained in techniques for the critical review of the scientific literature. Each article was read independently by two reviewers, one physician with training in epidemiology and one rheumatologist. Each reviewer completed one form for each article. Each epidemiologist-rheumatologist team then compared the individual forms and, if they agreed, completed the final form. The two reviewers discussed any discrepancies in an attempt to reach consensus. If they did not agree, the article was read and discussed by all the reviewers until agreement was reached, at which point the final form was completed. Each form made up one record in the database designed for the synthesis of the evidence. The form describes a comparison between two different treatment interventions. This means that a trial assessing more than two treatment interventions, making different comparisons among them, would give rise to more than one form. Thus, the number of forms is larger than the number of clinical trials included in the synthesis of the evidence. Of the 287 articles requested, 162 did not meet at least one of the inclusion criteria, 13 were redundant and 9 were articles comparing drugs with placebo that had already been included in the systematic reviews of the Cochrane Library. Consequently, a total of 103 articles was included in the review, 91 of which were different clinical trials (12 were complementary articles), allowing 140 comparisons between different treatment strategies (single drugs or combinations thereof). Thus, there are 140 records in the database for the efficacy of DMARDs (see Table 4). (Table 4 of the GUIPCAR CLINICAL PRACTICE GUIDELINE FOR THE MANAGEMENT OF RHEUMATOID ARTHRITIS guideline is available in the original guideline document.) These 91 trials and the 140 resulting comparisons are grouped by the Jadad and Hadorn scales for classifying the quality of the evidence as shown in Tables 5 and 6, respectively. Table 5. Clinical trials and comparisons by the Jadad scale for rating the quality of the evidence LEVEL OF EVIDENCE (Jadad Scale) * 1 CLINICAL TRIALS * 7 (8%) COMPARISONS * 7 (5%) LEVEL OF EVIDENCE (Jadad Scale) * 2 CLINICAL TRIALS * 19 (21%) COMPARISONS * 24 (17%) LEVEL OF EVIDENCE (Jadad Scale) * 3 CLINICAL TRIALS * 18 (20%) COMPARISONS * 31 (22%) LEVEL OF EVIDENCE (Jadad Scale) * 4 CLINICAL TRIALS * 32 (35%) COMPARISONS * 53 (38%) LEVEL OF EVIDENCE (Jadad Scale) * 5 CLINICAL TRIALS * 15 (16%) COMPARISONS * 25 (18%) LEVEL OF EVIDENCE (Jadad Scale) * TOTAL CLINICAL TRIALS * 91 (100%) COMPARISONS * 140 (100%) Table 6. Clinical trials and comparisons by the Hadorn scale for rating the quality of the evidence LEVEL OF EVIDENCE (Hadorn Scale) * A1 CLINICAL TRIALS * 13 (14%) COMPARISONS * 25 (18%) LEVEL OF EVIDENCE (Hadorn Scale) * A2 CLINICAL TRIALS * 13 (14%) COMPARISONS * 20 (14%) LEVEL OF EVIDENCE (Hadorn Scale) * B CLINICAL TRIALS * 65 (72%) COMPARISONS * 95 (68%) LEVEL OF EVIDENCE (Hadorn Scale) * TOTAL CLINICAL TRIALS * 91 (100%) COMPARISONS * 140 (100%) Thirty-eight of the 140 comparisons assessed a DMARD vs. certain drugs not included in the systematic review (e.g., collagen II, tiopronin, or pyrithinol). These 38 comparisons were excluded because the expert panel believed they were not relevant. Thus, the final 102 comparisons refer only to the DMARDs listed in Table 1. For the purposes of the synthesis of the evidence, the comparisons were grouped by drug to make it easier to find the clinical trials comparing a specific drug (alone or in combination) with any of the other drugs in table 1 (also alone or in combination). For each possible comparison we identified the number of existing studies, their level of evidence, and the intervention favored by the outcome measures collected in each study. (The tables synthesizing the outcome measures collected in each study for the comparisons are available to interested readers by contacting milena@taiss.com). The panel was consulted when a) different studies comparing the same drugs included different outcome measures, some (e.g., clinical effect) in favor of one treatment intervention and others (e.g., radiographic results) in favor of the other intervention; and b) to assess the clinical relevance of some outcome measures with statistically significant differences when the differences were not significant for many other outcomes. In these divergent cases, greater weight was given to outcome measures in agreement with the American College of Rheumatology (ACR) criteria, especially the number of swollen joints and radiographic damage. For example, a study with A2 level evidence comparing chloroquine with oral gold salts shows significant differences in favor of chloroquine with regard to strength of grip and morning stiffness; however, functional status is significantly better in the group with oral salts, and no significant differences are found between the two groups with regard to the number of swollen joints, pain, or acute phase reactants. In this case it was concluded that there were no important clinical differences in the efficacy of chloroquine and oral gold salts. Evidence tables The results of the synthesis of the evidence are shown in Tables 7 and 8. Table 7 includes the comparisons of DMARDs used in monotherapy. Table 8 includes the comparisons of single or combined drugs vs. combinations of drugs. For those comparisons for which evidence exists, there are three lines in the corresponding box with the following data: Line 1. Number of studies and level of evidence of these studies (e.g., "3-A1; 2-B" means there are three studies with an A1 level of evidence and two with level B). Line 2. The identification numbers for these articles (used as the bibliographic reference). Numbers separated by a dash (e.g., 7-9) mean that all articles in the interval are included (7, 8, and 9). Numbers separated by a comma (e.g., 7, 9) represent references only to those specific articles (7 and 9). Line 3. One of the following possibilities is shown in bold print: - The drug or combination of drugs (COMB) which the evidence favors (using the abbreviations shown in table 1.) - NS: if the differences are not significant. Table 7. Comparisons of DMARDs used only in monotherapy CPA PCB * AUR * AZA * 1-B 1 CPA CPA * CLQ * CSA * DP * ETN * HCQ * IFM * LEF * MTX * IG * CLQ PCB * AUR * 1-A2 2 NS AZA * 1-B 3 NS CPA * CLQ * CSA * DP * ETN * HCQ * IFM * LEF * MTX * IG * CSA PCB * AUR * AZA * 1-B 4 NS CPA * CLQ * 1-A2 5 NS CSA * DP * ETN * HCQ * IFM * LEF * MTX * IG * DP PCB * AUR * 1-B 6 DP AZA * 3-B 7-9 NS CPA * CLQ * 1-B 10 NS CSA * 1-A2 11 NS DP * ETN * HCQ * IFM * LEF * MTX * IG * ETN PCB * 2A1;1B 12-14 ETN AUR * AZA * CPA * CLQ * CSA * DP * ETN * 2-A1 15,72 NS HCQ * IFM * LEF * MTX * IG * HCQ PCB * AUR * 1-B 16 NS AZA * CPA * CLQ * CSA * DP * 2-B 17-18 NS ETN * HCQ * IFM * LEF * MTX * IG * IFM PCB * 2-A2 19,20 IFM AUR * AZA * CPA * CLQ * CSA * DP * ETN * HCQ * IFM * 1-A2 21 DOSE=10 LEF * MTX * IG * LEF PCB * 2-A1;3-B 22-26 LEF AUR * AZA * CPA * CLQ * CSA * DP * ETN * HCQ * IFM * LEF * MTX * IG * MTX PCB * AUR * 1-A1;1-B 27, 28 MTX AZA * 2-A2;2-B 29-32 MTX CPA * CLQ * 1-B 33 NS CSA * DP * ETN * 2-A1 72 NS HCQ * IFM * 3-B 34-36 NS LEF * 1-A1;2-B 37-39 NS MTX * IG * IG PCB * AUR * 1-A1;1-A2; 8-B 40-49 NS AZA * 2-B 50,51 AZA CPA * 1-B 52 CPA CLQ * 1-B 53 NS CSA * 1-A2 54 NS DP * 3-B 55-57 NS ETN * HCQ * IFM * LEF * MTX * 1-A2;4-B 58-62 NS IG * SSZ PCB * AUR * 1-B 63 NS AZA * CPA * CLQ * CSA * DP * 2-B 64, 65 NS ETN * HCQ * 2-A2 66, 67 SSZ IFM * LEF * 1-A1 68 NS MTX * 1-A1;1-A2 69, 70 NS IG * 1-B 71 NS * Compares doses of 1 mg/kg vs. 10 mg/kg Table 8. Comparisons of single or combined DMARDs vs. drug combinations AUR AUR+MTX * 1-B 73 NS AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * AZA AUR+MTX * AZA+MTX * 1-B 74 COMB CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * CPA AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * CLQ AUR+MTX * AZA+MTX * CLQ+DP * 1-B 75 NS MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * CSA AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * DP AUR+MTX * AZA+MTX * CLQ+DP * 1-B 76 NS MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * 1-B 77 NS MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * ETN AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * HCQ AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * 1-B 78 NS MTX+ETN * HCQ+IG * SSZ+HCQ * 1-A2 79 COMB HCQ+MTX * 1-B 80 COMB MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * IFM AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * LEF AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * MTX AUR+MTX * 1-B 81 NS AZA+MTX * 1-B 82 COMB CLQ+DP * MTX+CLQ * 1-A2 83 COMB CSA+IG * CSA+MTX * 1-A1 84 COMB HCQ+DP * MTX+ETN * 1-B 85 COMB HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * 1-A2 86 COMB IFM+MTX * 7-B 87-93 COMB MTX+SSZ * 1-A1:1-A2 94,95 NS IG AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * 1-B 96 NS CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * 1-A1;1-B 97,98 NS SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * SSZ AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * 1-A2 99 NS HCQ+MTX * MTX+SSZ+HCQ * IFM+MTX * MTX+SSZ * 1-A1:1-A2 100, 101 NS SSZ+HCQ AUR+MTX * AZA+MTX * CLQ+DP * MTX+CLQ * CSA+IG * CSA+MTX * HCQ+DP * MTX+ETN * HCQ+IG * SSZ+HCQ * HCQ+MTX * MTX+SSZ+HCQ * 1-A2 102 COMB. TRIP. IFM+MTX * MTX+SSZ * Tables 9a and 9b show the mean time of disease progression, previous use of DMARDs (if the box is blank, this data was not included in the article), duration of treatment in the trial (if the box is blank, this data was not included in the article), the level of evidence, the bibliographic reference for each comparison (cited in references 2.2, 2.3, and 2.4), and the identification number for each article (used to locate it in Tables 7 and 8). Table 9a. Mean time of RA progression, previous DMARD use, duration of treatment in the trial, level of evidence, bibliographic reference, and ID number in the comparisons of DMARDs used in monotherapy included in the synthesis of the evidence DMARDs compared (used in monotherapy) * Azathioprine * Cyclophosphamide Mean time of RA progression (in months) * 60 Previous DMARD use * Duration of treatment (in weeks) * 72 Level of evidence * B Bibliographic reference * 1, R1 ID N° * 1 DMARDs compared (used in monotherapy) * Oral gold salts * Chloroquine Mean time of RA progression (in months) * 39.4 Previous DMARD use * Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 2 ID N° * 2 DMARDs compared (used in monotherapy) * Azathioprine * Chloroquine Mean time of RA progression (in months) * 23 Previous DMARD use * Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 3 ID N° * 3 DMARDs compared (used in monotherapy) * Cyclosporin * Azathioprine Mean time of RA progression (in months) * 79.2 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 4 ID N° * 4 DMARDs compared (used in monotherapy) * Cyclosporin * Chloroquine Mean time of RA progression (in months) * 78 Previous DMARD use * NO Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 5 ID N° * 5 DMARDs compared (used in monotherapy) * Oral gold salts * D-penicillamine Mean time of RA progression (in months) * 83 Previous DMARD use * YES Duration of treatment (in weeks) * 52 Level of evidence * B Bibliographic reference * 6 ID N° * 6 DMARDs compared (used in monotherapy) * Azathioprine * D-penicillamine Mean time of RA progression (in months) * 113.4 Previous DMARD use * Duration of treatment (in weeks) * 52 Level of evidence * B Bibliographic reference * 7, R2 ID N° * 7 DMARDs compared (used in monotherapy) * Azathioprine * D-penicillamine Mean time of RA progression (in months) * 134 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 8 ID N° * 8 DMARDs compared (used in monotherapy) * Azathioprine * D-penicillamine Mean time of RA progression (in months) * 132 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 9 ID N° * 9 DMARDs compared (used in monotherapy) * D-penicillamine * Chloroquine Mean time of RA progression (in months) * 30 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 10 ID N° * 10 DMARDs compared (used in monotherapy) * Cyclosporin * D-penicillamine Mean time of RA progression (in months) * 86.4 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 11 ID N° * 11 DMARDs compared (used in monotherapy) * Etanercept * Placebo Mean time of RA progression (in months) * 150 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * A1 Bibliographic reference * 12 ID N° * 12 DMARDs compared (used in monotherapy) * Etanercept * Placebo Mean time of RA progression (in months) * 138 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * A1 Bibliographic reference * 12 ID N° * 13 DMARDs compared (used in monotherapy) * Etanercept * Placebo Mean time of RA progression (in months) * Previous DMARD use * YES Duration of treatment (in weeks) * 12 Level of evidence * B Bibliographic reference * 13 ID N° * 14 DMARDs compared (used in monotherapy) * Etanercept * Etanercept Mean time of RA progression (in months) * 144 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * A1 Bibliographic reference * 12 ID N° * 15 DMARDs compared (used in monotherapy) * Oral gold salts * Hydroxychloroquine Mean time of RA progression (in months) * 124.5 Previous DMARD use * Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 14 ID N° * 16 DMARDs compared (used in monotherapy) * Hydroxychloroquine * D-penicillamine Mean time of RA progression (in months) * 71.4 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 15 ID N° * 17 DMARDs compared (used in monotherapy) * Hydroxychloroquine * D-penicillamine Mean time of RA progression (in months) * Previous DMARD use * NO Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 16 ID N° * 18 DMARDs compared (used in monotherapy) * Infliximab * Placebo Mean time of RA progression (in months) * 99 Previous DMARD use * YES Duration of treatment (in weeks) * 4 Level of evidence * A2 Bibliographic reference * 17 ID N° * 19 DMARDs compared (used in monotherapy) * Infliximab * Placebo Mean time of RA progression (in months) * 97.8 Previous DMARD use * YES Duration of treatment (in weeks) * 4 Level of evidence * A2 Bibliographic reference * 17 ID N° * 20 DMARDs compared (used in monotherapy) * Infliximab * Infliximab Mean time of RA progression (in months) * 88.8 Previous DMARD use * YES Duration of treatment (in weeks) * 4 Level of evidence * A2 Bibliographic reference * 17 ID N° * 21 DMARDs compared (used in monotherapy) * Leflunomide * Placebo Mean time of RA progression (in months) * 79.8 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A1 Bibliographic reference * 18 ID N° * 22 DMARDs compared (used in monotherapy) * Leflunomide * Placebo Mean time of RA progression (in months) * 83.4 Previous DMARD use * YES Duration of treatment (in weeks) * 52 Level of evidence * A1 Bibliographic reference * 19, R3 ID N° * 23 DMARDs compared (used in monotherapy) * Leflunomide * Placebo Mean time of RA progression (in months) * 96 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 20 ID N° * 24 DMARDs compared (used in monotherapy) * Leflunomide * Placebo Mean time of RA progression (in months) * 100.8 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 20 ID N° * 25 DMARDs compared (used in monotherapy) * Leflunomide * Placebo Mean time of RA progression (in months) * 102.6 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 20 ID N° * 26 DMARDs compared (used in monotherapy) * Methotrexate * Oral gold salts Mean time of RA progression (in months) * 70.3 Previous DMARD use * YES Duration of treatment (in weeks) * 36 Level of evidence * A1 Bibliographic reference * 21, C1 ID N° * 27 DMARDs compared (used in monotherapy) * Methotrexate * Oral gold salts Mean time of RA progression (in months) * 59.5 Previous DMARD use * NO Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 22, C2 ID N° * 28 DMARDs compared (used in monotherapy) * Methotrexate * Azathioprine Mean time of RA progression (in months) * 104.4 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 23 ID N° * 29 DMARDs compared (used in monotherapy) * Methotrexate * Azathioprine Mean time of RA progression (in months) * 133.2 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 24, R4, R5 ID N° * 30 DMARDs compared (used in monotherapy) * Methotrexate * Azathioprine Mean time of RA progression (in months) * 96 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 25, R6, R7 ID N° * 31 DMARDs compared (used in monotherapy) * Methotrexate * Azathioprine Mean time of RA progression (in months) * 156 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 26 ID N° * 32 DMARDs compared (used in monotherapy) * Cyclosporin * Methotrexate Mean time of RA progression (in months) * 25.8 Previous DMARD use * NO Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 27 ID N° * 33 DMARDs compared (used in monotherapy) * Infliximab * Methotrexate Mean time of RA progression (in months) * 91.2 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 28 ID N° * 34 DMARDs compared (used in monotherapy) * Infliximab * Methotrexate Mean time of RA progression (in months) * 92.4 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 28 ID N° * 35 DMARDs compared (used in monotherapy) * Infliximab * Methotrexate Mean time of RA progression (in months) * 103.8 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 28 ID N° * 36 DMARDs compared (used in monotherapy) * Leflunomide * Methotrexate Mean time of RA progression (in months) * 81 Previous DMARD use * YES Duration of treatment (in weeks) * 52 Level of evidence * A1 Bibliographic reference * 19 ID N° * 37 DMARDs compared (used in monotherapy) * Leflunomide * Methotrexate Mean time of RA progression (in months) * 45 Previous DMARD use * YES Duration of treatment (in weeks) * 52 Level of evidence * B Bibliographic reference * 29 ID N° * 38 DMARDs compared (used in monotherapy) * Leflunomide * Methotrexate Mean time of RA progression (in months) * 43.8 Previous DMARD use * YES Duration of treatment (in weeks) * 104 Level of evidence * B Bibliographic reference * 29 ID N° * 39 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 76 Previous DMARD use * Duration of treatment (in weeks) * 21 Level of evidence * A1 Bibliographic reference * 30, C3, R8, R9 ID N° * 40 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 115.8 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 31 ID N° * 41 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 83.4 Previous DMARD use * Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 32 ID N° * 42 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 61.8 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 33, C4 ID N° * 43 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 144 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * A2 Bibliographic reference * 34 ID N° * 44 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 63.6 Previous DMARD use * NO Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 35 ID N° * 45 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 109.2 Previous DMARD use * NO Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 36 ID N° * 46 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 45.6 Previous DMARD use * YES Duration of treatment (in weeks) * 52 Level of evidence * B Bibliographic reference * 37, C5 ID N° * 47 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 63 Previous DMARD use * Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 38 ID N° * 48 DMARDs compared (used in monotherapy) * Oral gold salts * Injectable gold Mean time of RA progression (in months) * 24 Previous DMARD use * Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 39, C6, R10 ID N° * 49 DMARDs compared (used in monotherapy) * Azathioprine * Injectable gold Mean time of RA progression (in months) * 60 Previous DMARD use * Duration of treatment (in weeks) * 72 Level of evidence * B Bibliographic reference * 1 ID N° * 50 DMARDs compared (used in monotherapy) * Azathioprine * Injectable gold Mean time of RA progression (in months) * 25 Previous DMARD use * Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 3 ID N° * 51 DMARDs compared (used in monotherapy) * Cyclophosphamide * Injectable gold Mean time of RA progression (in months) * 48 Previous DMARD use * Duration of treatment (in weeks) * 72 Level of evidence * B Bibliographic reference * 1 ID N° * 52 DMARDs compared (used in monotherapy) * Injectable gold * Chloroquine Mean time of RA progression (in months) * 20 Previous DMARD use * Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 3 ID N° * 53 DMARDs compared (used in monotherapy) * Cyclosporin * Injectable gold Mean time of RA progression (in months) * 11.76 Previous DMARD use * YES Duration of treatment (in weeks) * 72 Level of evidence * B Bibliographic reference * 40 ID N° * 54 DMARDs compared (used in monotherapy) * Injectable gold * D-penicillamine Mean time of RA progression (in months) * 66 Previous DMARD use * Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 41, R11 ID N° * 55 DMARDs compared (used in monotherapy) * Injectable gold * D-penicillamine Mean time of RA progression (in months) * 14.8 Previous DMARD use * Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 42 ID N° * 56 DMARDs compared (used in monotherapy) * Injectable gold * D-penicillamine Mean time of RA progression (in months) * Previous DMARD use * Duration of treatment (in weeks) * 21 Level of evidence * B Bibliographic reference * 43 ID N° * 57 DMARDs compared (used in monotherapy) * Injectable gold * Methotrexate Mean time of RA progression (in months) * Previous DMARD use * NO Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 44 ID N° * 58 DMARDs compared (used in monotherapy) * Injectable gold * Methotrexate Mean time of RA progression (in months) * 14 Previous DMARD use * Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 45 ID N° * 59 DMARDs compared (used in monotherapy) * Injectable gold * Methotrexate Mean time of RA progression (in months) * 17.5 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 46 ID N° * 60 DMARDs compared (used in monotherapy) * Injectable gold * Methotrexate Mean time of RA progression (in months) * 23.9 Previous DMARD use * YES Duration of treatment (in weeks) * 144 Level of evidence * B Bibliographic reference * 47, C7, C8 ID N° * 61 DMARDs compared (used in monotherapy) * Injectable gold * Methotrexate Mean time of RA progression (in months) * 68.4 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * A2 Bibliographic reference * 48 ID N° * 62 DMARDs compared (used in monotherapy) * Sulphasalazine * Oral gold salts Mean time of RA progression (in months) * 114 Previous DMARD use * YES Duration of treatment (in weeks) * 240 Level of evidence * B Bibliographic reference * 49, C9 ID N° * 63 DMARDs compared (used in monotherapy) * Sulphasalazine * D-penicillamine Mean time of RA progression (in months) * 84 Previous DMARD use * Duration of treatment (in weeks) * 576 Level of evidence * B Bibliographic reference * 50, C10 ID N° * 64 DMARDs compared (used in monotherapy) * Sulphasalazine * D-penicillamine Mean time of RA progression (in months) * 105 Previous DMARD use * Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 51 ID N° * 65 DMARDs compared (used in monotherapy) * Hydroxychloroquine * Sulphasalazine Mean time of RA progression (in months) * 12.8 Previous DMARD use * NO Duration of treatment (in weeks) * 48 Level of evidence * A2 Bibliographic reference * 52, C11, C12, R12 ID N° * 66 DMARDs compared (used in monotherapy) * Hydroxychloroquine * Sulphasalazine Mean time of RA progression (in months) * 75.6 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 53 ID N° * 67 DMARDs compared (used in monotherapy) * Leflunomide * Sulphasalazine Mean time of RA progression (in months) * 90 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A1 Bibliographic reference * 18 ID N° * 68 DMARDs compared (used in monotherapy) * Sulphasalazine * Methotrexate Mean time of RA progression (in months) * 3.05 Previous DMARD use * NO Duration of treatment (in weeks) * 52 Level of evidence * A2 Bibliographic reference * 54 ID N° * 69 DMARDs compared (used in monotherapy) * Sulphasalazine * Methotrexate Mean time of RA progression (in months) * 14.6 Previous DMARD use * NO Duration of treatment (in weeks) * 52 Level of evidence * A1 Bibliographic reference * 55 ID N° * 70 DMARDs compared (used in monotherapy) * Injectable gold * Sulphasalazine Mean time of RA progression (in months) * 68 Previous DMARD use * NO Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 56 ID N° * 71 DMARDs compared (used in monotherapy) * Etanercept * Methotrexate Mean time of RA progression (in months) * 11.5 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * A1 Bibliographic reference * 57 ID N° * 72 Table 9b. Mean time of RA progression, previous DMARD use, duration of treatment in the trial, level of evidence, bibliographic referenc,e and ID number in the comparisons of DMARDs used in monotherapy or combined therapy vs. drug combinations included in the synthesis of the evidence DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Oral gold salts * Methotrexate * Oral gold salts Mean time of RA progression (in months) * 64.5 Previous DMARD use * NO Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 22 ID N° * 73 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Azathioprine * Methotrexate * Azathioprine Mean time of RA progression (in months) * 96 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 25, R6, R7 ID N° * 74 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Chloroquine * Chloroquine * D-penicillamine Mean time of RA progression (in months) * 24 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 10 ID N° * 75 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * D-penicillamine * Chloroquine * D-penicillamine Mean time of RA progression (in months) * 18 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 10 ID N° * 76 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * D-penicillamine * Hydroxychloroquine * D-penicillamine Mean time of RA progression (in months) * 72.6 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 15 ID N° * 77 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Hydroxychloroquine * Hydroxychloroquine * D-penicillamine Mean time of RA progression (in months) * 70.8 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * B Bibliographic reference * 15 ID N° * 78 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Hydroxychloroquine * Sulphasalazine * Hydroxychloroquine Mean time of RA progression (in months) * 75.6 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 53 ID N° * 79 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Hydroxychloroquine * Hydroxychloroquine * Methotrexate Mean time of RA progression (in months) * Previous DMARD use * Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 58 ID N° * 80 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Oral gold salts Mean time of RA progression (in months) * 69 Previous DMARD use * NO Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 22 ID N° * 81 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Azathioprine Mean time of RA progression (in months) * 96 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * B Bibliographic reference * 25 ID N° * 82 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Chloroquine Mean time of RA progression (in months) * 92.58 Previous DMARD use * Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 59 ID N° * 83 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Cyclosporin Mean time of RA progression (in months) * 122.4 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A1 Bibliographic reference * 60 ID N° * 84 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Etanercept Mean time of RA progression (in months) * 156 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 61 ID N° * 85 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Sulphasalazine * Hydroxychloroquine Mean time of RA progression (in months) * 120 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * A2 Bibliographic reference * 62 ID N° * 86 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Infliximab * Methotrexate Mean time of RA progression (in months) * 103.8 Previous DMARD use * YES Duration of treatment (in weeks) * 54 Level of evidence * B Bibliographic reference * 63, R13 ID N° * 87 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Infliximab * Methotrexate Mean time of RA progression (in months) * 96.6 Previous DMARD use * YES Duration of treatment (in weeks) * 54 Level of evidence * B Bibliographic reference * 63, R13 ID N° * 88 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Infliximab * Methotrexate Mean time of RA progression (in months) * 107.4 Previous DMARD use * YES Duration of treatment (in weeks) * 54 Level of evidence * B Bibliographic reference * 63, R13 ID N° * 89 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Infliximab * Methotrexate Mean time of RA progression (in months) * 105.6 Previous DMARD use * YES Duration of treatment (in weeks) * 54 Level of evidence * B Bibliographic reference * 63, R13 ID N° * 90 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Infliximab * Methotrexate Mean time of RA progression (in months) * 131.4 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 28 ID N° * 91 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Infliximab * Methotrexate Mean time of RA progression (in months) * 118.2 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 28 ID N° * 92 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Infliximab * Methotrexate Mean time of RA progression (in months) * 112.2 Previous DMARD use * YES Duration of treatment (in weeks) * 26 Level of evidence * B Bibliographic reference * 28 ID N° * 93 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Sulphasalazine Mean time of RA progression (in months) * 14.5 Previous DMARD use * NO Duration of treatment (in weeks) * 52 Level of evidence * A1 Bibliographic reference * 55 ID N° * 94 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Methotrexate * Methotrexate * Sulphasalazine Mean time of RA progression (in months) * 2.8 Previous DMARD use * NO Duration of treatment (in weeks) * 52 Level of evidence * A2 Bibliographic reference * 54 ID N° * 95 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Injectable gold * Injectable gold * Cyclosporin Mean time of RA progression (in months) * 133.2 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 64 ID N° * 96 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Injectable gold * Hydroxychloroquine * Injectable gold Mean time of RA progression (in months) * 24 Previous DMARD use * YES Duration of treatment (in weeks) * 48 Level of evidence * A1 Bibliographic reference * 65 ID N° * 97 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Injectable gold * Hydroxychloroquine * Injectable gold Mean time of RA progression (in months) * 78 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * B Bibliographic reference * 66 ID N° * 98 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Sulphasalazine * Sulphasalazine * Hydroxychloroquine Mean time of RA progression (in months) * 75.6 Previous DMARD use * YES Duration of treatment (in weeks) * 24 Level of evidence * A2 Bibliographic reference * 53 ID N° * 99 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Sulphasalazine * Methotrexate * Sulphasalazine Mean time of RA progression (in months) * 75.6 Previous DMARD use * NO Duration of treatment (in weeks) * 52 Level of evidence * A1 Bibliographic reference * 55 ID N° * 100 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Sulphasalazine * Methotrexate * Sulphasalazine Mean time of RA progression (in months) * 2.9 Previous DMARD use * NO Duration of treatment (in weeks) * 52 Level of evidence * A2 Bibliographic reference * 54 ID N° * 101 DMARDs compared (used in monotherapy or combined therapy vs. drug combinations) * Sulphasalazine * Hydroxychloroquine * Methotrexate * Sulphasalazine * Hydroxychloroquine Mean time of RA progression (in months) * 96 Previous DMARD use * YES Duration of treatment (in weeks) * 96 Level of evidence * A2 Bibliographic reference * 62 ID N° * 102 Evaluation of the methodological quality of meta-analyses The quality of the Cochrane Library meta-analyses was rated using the index of the quality of review articles, as revised by Oxman and Guyatt [Oxman, 1991a; Oxman, 1991b; Jadad, 1996b]. This index is based on answers to the following questions: 1. Were the search methods used to find evidence on the primary question stated? No Partially Yes 2. Was the search for evidence reasonably comprehensive? No Can't tell Yes 3. Were the criteria used for deciding which studies to include in the review reported? No Partially Yes 4. Was bias in the selection of studies avoided? No Can't tell Yes 5. Were the criteria used for assessing the validity of the included studies reported? No Partially Yes 6. Was the validity of all of the studies referred to in the text assessed using appropriate criteria (either in selecting studies for inclusion or in analyzing the studies that are cited)? No Can't tell Yes 7. Were the methods used to combine the findings of the studies reported? No Partially Yes 8. Were the findings of the relevant studies combined appropriately relative to the primary question the overview addresses? No Can't tell Yes 9. Were the conclusions made by the author(s) supported by the data and/or analysis reported in the overview? No Can't tell Yes 10. How would you rate the scientific quality of this overview? Extensive flaws | Major flaws | Minor flaws | Minimal flaws - 1 - - 2 - - 3 - - 4 - - 5 - - 6 - - 7 - The score for question 10 (overall scientific quality) is based on the replies to the previous 9 questions. If the answer to one or more questions is "Can't tell" the overview probably has, at the very least, minor flaws, and it is difficult to exclude major flaws (score of 4 or less). If the answer to questions 2, 4, 6, or 8 is "No," the overview probably has major flaws (score of 3 or less), depending on the number and severity of the flaws). The score for the quality of the overview can range from 1 to 7, as shown in the figure for question 10. In accordance with the previously described methodology, the quality of the scientific evidence for the nine meta-analyses found is shown in Table 10. Table 10. Evaluation of the quality of the meta-analyses of DMARDs DMARD [Systematic review citation]* Questions in meta-analysis ratings * 1 * 2 * 3 * 4 * 5 * 6 * 7 * 8 * 9 * 10 * Auranofin [RS1] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * Can't tell * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 4 * Azathioprine [RS2] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * Can't tell * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 4 * Cyclophosphamide [RS3] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * Can't tell * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 4 * Cyclosporin [RS4] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * No * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 3 * D-penicillamine [RS5] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * Can't tell * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 4 * Hydroxychloroquine [RS6] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * Can't tell * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 4 * Methotrexate [RS7] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * Can't tell * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 4 * Injectable gold [RS8] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * Can't tell * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 4 * Sulphasalazine [RS9] * 1 * Yes * 2 * Yes * 3 * Yes * 4 * No * 5 * Yes * 6 * Yes * 7 * Yes * 8 * Yes * 9 * Yes * 10 * 3 * The citation for each systematic review is shown in reference list 2.1. Table 11 shows the conclusions of the systematic reviews of the Cochrane Library and the quality of these reviews. Table 11. Conclusions of the Cochrane Library meta-analyses and their level of evidence DMARD [Systematic review citation]* * Auranofin [RS1] Conclusions * Auranofin appears to be efficacious in the short-term treatment of patients with RA. It has a small clinically and statistically significant benefit on disease activity. It may be more appropriate for patients with early and mild disease, who probably respond to less aggressive treatments. Level of Evidence (1-7) * 4 DMARD [Systematic review citation]* * Azathioprine [RS2] Conclusions * Appears to be beneficial in the short term in the treatment of patients with RA. It cannot be considered more efficacious than other DMARDs, however, and its toxicity is greater. This suggests that other DMARDs should be used before considering azathioprine. Level of Evidence (1-7) * 4 DMARD [Systematic review citation]* * Cyclophosphamide [RS3] Conclusions * Cyclophosphamide appears to be an effective drug in treating patients with RA, but its use is limited due to its toxicity. Since its efficacy appears to be similar to that of other, less toxic antirheumatic drugs, its use should be limited to patients who do not respond to other treatments. Level of Evidence (1-7) * 4 DMARD [Systematic review citation]* * Cyclosporin [RS4] Conclusions * Cyclosporin has an important clinical benefit in the short-term treatment of patients with progressive RA. Level of Evidence (1-7) * 3 DMARD [Systematic review citation]* * D-penicillamine [RS5] Conclusions * D-penicillamine appears to be efficacious for the short term treatment of patients with RA. It has a clinically and statistically significant benefit on disease activity. Its effects on long-term functional status and radiographic progression are not clear at this time. There does not appear to be any clear advantage to using doses higher than 500 mg/day. Level of Evidence (1-7) * 4 DMARD [Systematic review citation]* * Hydroxychloroquine [RS6] Conclusions * Hydroxychloroquine appears to be efficacious in the treatment of RA. Its total effect appears to be moderate, but its low toxicity profile should be considered in treating RA patients. Level of Evidence (1-7) * 4 DMARD [Systematic review citation]* * Methotrexate [RS7] Conclusions * Methotrexate has a clinical and statistically significant benefit in the short-term treatment of patients with RA. Level of Evidence (1-7) * 4 DMARD [Systematic review citation]* * Injectable gold [RS8] Conclusions * Although its use may be limited due to the incidence of serious toxicity, injectable gold has a clinical and statistically significant benefit in the short-term treatment of patients with RA. Level of Evidence (1-7) * 4 DMARD [Systematic review citation]* * Sulphasalazine [RS9] Conclusions * Sulphasalazine appears to be efficacious in the short-term treatment of patients with RA. It has a clinically and statistically significant benefit on disease activity. Its effects on functional status and radiographic progression are not clear at this time, but they appear to be modest. Level of Evidence (1-7) * 3 * The citation for each systematic review is shown in reference list 2.1. 2. Development of the Recommendations Several of the tasks involved in making this guideline, including the development of the recommendations, were carried out by the expert panel. 2.1 Selection of the expert panel Panel nominations were provided by the SER, by independent rheumatologists, and by investigators familiar with the methodology for producing CPG recommendations. To help make the guideline more valid, the principal investigator proposed to those responsible for nominations that the panel should meet the following criteria: 1) multidisciplinarity, that is, the panel should include not only rheumatologists, but also other specialists whose opinions would help improve RA management or improve the methodology for developing the recommendations (e.g., rehabilitation specialists, orthopaedists, or epidemiologists); 2) professional prestige, that is, that by virtue of their professional trajectory the opinions of the expert panel would be respected by the scientific community; 3) geographic diversity, that is, if the guideline is to be applied at the national level, an attempt should be made to have a reasonable representation of Spain's different autonomous communities; 4) diversity in levels of care, so that the recommendations would make sense in both hospital and outpatient settings; 5) diversity of academic level, so as to represent the viewpoints of teaching centers and highly developed academic and research centers, as well as the perspectives of professionals who manage patients in less academic centers; and 6) representation of both men and women on the panel. Although it was not easy to incorporate all these criteria, a list of nominees was produced. Panelists were contacted to request their participation, and a panel of 15 members was formed: 12 rheumatologists, one rehabilitation specialist, one orthopaedist, and two epidemiologists (one of the rheumatologists is also an epidemiologist). The autonomous communities from which there was at least one panel member were: Andalucía, Canary Islands, Cantabria, Cataluña, Extremadura, Galicia, Madrid, La Rioja, and Comunidad Valenciana. Finally, there were 4 women and 11 men on the panel. The most important individual criterion for selection was the explicit absence of conflicts of interest. Each panelist signed a document assuring that he or she was not affected by any of the conflicts described in a comprehensive list of possible conflicts of interest. Nominees with conflicts of interest were excluded. Another individual criterion for acceptance was availability during the time needed to carry out the tasks. Panelists received no remuneration for their work. 2.2 Panel tasks The panel's tasks were: 1) to define the contents of the guideline; 2) to develop the recommendations; 3) to write the definitions; 4) to review and contribute to the synthesis of the evidence; 5) to write parts of the guideline that had not originally been foreseen (for example, the most frequent extra-articular complications); and 6) to review the text written by the investigators. The panelists were asked to write recommendations that would provide practical and specific advice on the subjects covered. They were explicitly told that their recommendations should be based on the risk-benefit balance for the patient and that costs were not to be taken into account. That is, the recommendation should be made considering what would be best for the patient, consistent with the objective of improving the quality of care. 2.3 Formulation of the recommendations The 15 panelists formed 10 working groups of 3 persons each; thus, each panelists could serve on more than one group The working groups were responsible for writing different chapters of the guideline. The draft recommendations made by each group were sent to the investigators, who edited and circulated them to the rest of the panelists for suggestions, and in subsequent interactions the pre-definitive version of the recommendation was written. A joint document was written based on all this information, which was submitted to all the panel members for discussion and correction. Panelists interacted by telephone, email, regular mail, and small group meetings; the project investigators held four joint meetings with the whole panel. Thus, although each group wrote the initial version of a specific part of the guideline, all the panelists had the opportunity to contribute their knowledge and opinions in the rest of the guideline recommendations. There was large inter-panelist variability with relation to patient classification and the treatment approach in managing RA. Part of this variability may have been due to the fact that one panelist was thinking of a patient with certain characteristics while another was thinking of a patient with different characteristics. To classify patients by disease characteristics and to group them in a clinically meaningful way, various clinical variables were used. The variables considered for patient classification were: a) receipt of NSAIDs and/or corticosteroids in the previous 3 months (2 categories); b) number of swollen joints (less than 6, 6-10 or greater than 10) (3 categories); c) presence of erosions (0, 1-3 or greater than 3 erosions) (3 categories); d) presence of elevated acute-phase reactants (2 categories); Health Assessment Questionnaire (HAQ) score (less than 1 or greater than or equal to 1) (2 categories); and f) rheumatoid factor (less than 40, 40-100 or greater than 100 UI/mL) (3 categories). Combining the categories of these clinical variables yielded 144 different patient scenarios that might call for different treatment. The panelists provided anonymous and independent recommendations about the initial treatment for each of these 144 clinical scenarios. If the recommendations differed, panelists were free to express their arguments and the evidence supporting their opinion to try to convince the rest of the panel, but they were not obliged to reach consensus. That is, this guideline is based not on consensus, but rather on the best available scientific evidence and, when this was absent or contradictory, on the judgment of an expert panel that was not forced to reach a consensus. In other words, since part of the variability in clinical practice may be due to the fact that there is insufficient scientific evidence on the efficacy of the different DMARDs, this guideline recognizes that fact and considers that it is admissible for different professionals to have different opinions. The panel members chose the best treatment for each of the 144 different patient scenarios. Their recommendations were analyzed mathematically, especially the proportion of panelists who suggested each treatment recommendation. Since the recommended treatment was similar for many of the 144 different patient scenarios, these were grouped into 52 scenarios in which the treatment decisions were the same. Reducing the number of options seems more reasonable for guideline users, and these are the classification options reflected in the decision algorithm for the initial treatment of RA that is included at the end of this guideline. (Both the panel votes and the mathematical analysis are available to interested readers by contacting TAISS at milena@taiss.com.) The treatment decisions are further simplified in the text of the recommendations, where the use of corticosteroids and/or NSAIDs is treated separately from DMARD treatment, since the former are used only in particular, very specific situations. Thus, it is necessary to consider only the two objective parameters (number of swollen joints and presence of erosions) that have been shown to be the most important in disease classification due to their treatment implications. The reduced scenario classification was used to establish the alternative treatment in case of toxicity or unsatisfactory response to initial treatment. The panelists again voted individually, and the alternative treatment options with the most votes were chosen, ordered by preference of use. The systematic scientific review of the literature on DMARDs made it possible to identify the level of evidence supporting the panel recommendations for medical treatment. In the other chapters, each panelist or group provided the evidence on which they had based their recommendations. In other words, support for the recommendations based on a systematic review and evaluation of the scientific evidence in this guideline is limited to management with DMARDs. The rest of the recommendations are based on a non-systematic review of the scientific evidence, which is cited as the bibliographic reference, or on expert opinion. HOW TO USE THIS GUIDELINE This guideline is organized by chapters. Each chapter shows the recommendations in bold print, followed by a more detailed justification or explanation of the recommendations. We have tried to make the recommendations clear and practical, without losing flexibility. Thus, at times the choice between various possibilities considered equally valid by the panel is left to the judgment of the rheumatologist (e.g., various methods to evaluate RA severity). One of the attributes of this guideline is its flexibility, allowing it to be applied to the real world where situations may vary, for example with regard to local characteristics of the center (availability of certain technologies, training, etc.), personal preferences of the patient, or other situations that may affect clinical decision making in patients with RA. Rheumatologists using this guideline for the first time may find it useful to read the chapters in order, since this is the natural order of a medical evaluation: diagnosis, initial evaluation, classification, treatment, criteria for response to treatment, adverse effects of drug treatment, and possible extra-articular complications. Several documents are included with this guideline that may be of use to readers in evaluating and monitoring patients: data collection forms for initial patient evaluation and follow-up (Appendix 1) and an algorithm to aid treatment choices (Appendix 2). This algorithm allows the user to follow a logical decision-making process in patient management. The user of this guideline can consult the quality of the scientific evidence supporting the use of DMARDs and identify the articles from which the evidence was obtained. Also included is a "Rapid Reference Guide," which is a brief version of the guideline recommendations and includes the simplified algorithm. CONTENTS AND LIMITATIONS OF THE GUIDELINE RA is a disease that can affect different patient subgroups, can be managed in different settings, can be treated with a variety of therapeutic approaches, and can affect various organs or systems. To cover all these aspects in a guideline, besides being an endless task, would be impractical. Thus, the SER, together with the expert panel and the investigators, agreed to limit its contents as follows: * It is intended for rheumatologists. * It focuses on RA in adults (juvenile RA is excluded). * It includes diagnosis, evaluation, prognosis, treatments (drugs, rehabilitation, etc.), and extra-articular complications (amyloidosis, anemia, Sjögren's syndrome, etc.). * The synthesis of the scientific evidence based on a systematic review of the literature focuses on the efficacy of DMARDs. * The remaining recommendations or considerations are based on scientific evidence obtained without systematic review or on expert opinion. CHAPTER 1. DIAGNOSING RHEUMATOID ARTHRITIS The diagnosis of RA is essentially a clinical task. RA should be suspected in patients over 16 years of age who have joint inflammation or effusion of more than 6 weeks duration in three or more joints, preferably of the hands and feet. To date, the only universally accepted and used diagnostic criteria for RA are those proposed for classification of the disease by the American College of Rheumatology (ACR) in 1987 [Arnett, 1988]. According to the ACR, the diagnosis of RA requires confirmation of at least four of the following criteria: 1. Morning stiffness lasting at least one hour before maximal improvement, for at least 6 consecutive weeks. 2. Soft tissue swelling or fluid, observed by a physician, in at least three of the following joint areas (right or left): proximal interphalangeal (PIP), metacarpophalangeal (MCP), wrist, elbow, knee, ankle, or metatarsophalangeal (MTP) joints, for at least 6 consecutive weeks. 3. Swelling or fluid, observed by a physician, in the proximal interphalangeal, metacarpophalangeal, or wrist joints, for at least 6 consecutive weeks. 4. Symmetrical (right and left sides) swelling or fluid in the joints mentioned in point 2, observed by a physician, for at least 6 consecutive weeks. 5. Subcutaneous nodules over bony prominences or extensor surfaces, or in juxta-articular regions, observed by a physician. 6. Demonstration of serum rheumatoid factor (RF) detected by any method that has been positive in less than 5% of control subjects. 7. Radiographic evidence in the hands or wrists of articular erosions or osteopenia in or around the affected joints. These criteria have a sensitivity of 91% and a specificity of 89%. However, their predictive value varies depending on the prevalence of RA in the clinical or population setting, as can be seen in Table 12 [Balsa, 1996; Villaverde, 2000]. Table 12. Predictive values (%) for RA diagnostic criteria in different settings Population * General RA prevalence * 0.5 Positive predictive value * 4.7 Negative predictive value * 99.9 Population * Primary care medical consultation for osteomuscular pain RA prevalence * 19 Positive predictive value * 69.9 Negative predictive value * 97.2 Population * Specialized consultation for polyarthritis RA prevalence * 67 Positive predictive value * 95.3 Negative predictive value * 80.3 Thus, if a patient in the general population does not meet the RA criteria, for all practical purposes the disease can be ruled out (probability of error 1/1000). If the criteria are positive, however, the patient is unlikely to have the disease, which makes it necessary to confirm or rule out the diagnosis using additional techniques. In a consultation for polyarthritis, on the other hand, patients meeting the criteria are highly likely to have RA (95%), and if they do not meet the criteria, the probability of having RA is 20%. CHAPTER 2. INITIAL EVALUATION 2.1 Patients with RA should be evaluated and treated by physicians who are familiar with the clinical management and treatment of the disease. RA is a chronic disease, with an estimated prevalence of 0.5% (0.2-0.8%) in the Spanish adult population [Villaverde, 2000]. Its evaluation and treatment are somewhat complex, especially when the disease is clinically active. For this reason it is recommended that the diagnosis, monitoring, and treatment of RA be carried out by physicians who can identify patients in the early stages of the disease, evaluate the disease stage correctly, plan appropriate treatment for each stage of disease evolution, and measure the response to treatment. The recommendations in the following sections should always be followed in the initial evaluation and monitoring of patients. This systematic evaluation of patients is needed to plan appropriate treatment in accordance with disease characteristics and to measure the response to the treatment undertaken. 2.2 The initial evaluation of a patient with RA should include a clinical history and physical examination. The same as in any other clinical process, the first evaluation should include a clinical history and physical examination. The history should include a personal and family history (of diseases, surgical interventions, allergies), with special emphasis on processes that were life threatening or required medical treatment or hospitalization. The history should review life styles with regard to exercise, nutrition, smoking, and alcohol use. Gynecological history and date of first menstruation should also be recorded. The sociodemographic data to be collected include age, gender, educational level, main occupation, and work setting, because of their importance as prognostic factors in RA. In the initial evaluation of a patient with RA, the rheumatologist should keep in mind the previous history of the disease. Some patients will have RA of short evolution, for which hardly any medical treatment has been received, while others may visit the physician following a more or less prolonged period of arthritis, with a clinical and therapeutic history that must be considered. In these cases, the clinical characteristics of the disease should be obtained by interviewing the patient and reviewing reports and other documents provided by the patient, such as radiographs and certain laboratory tests. To understand the evolution of RA, it is critically important to identify any previous or concurrent treatment, especially with analgesics, NSAIDs, corticosteroids, and DMARDs. A detailed history should be taken of DMARDs received to date, the dosage, duration, and reasons for discontinuing the medication. The same information should be obtained for corticosteroid use. With regard to NSAIDs, the physician should ask about their observed tolerance and side effects, with special regard to digestive problems. In the physical examination, in addition to the signs that would be observed for any patient, careful note should be taken of the presence of pain, joint inflammation, deformities, and subcutaneous nodules. 2.3 The evaluation and monitoring of RA should be based on a systematic evaluation of a minimum set of parameters including joint pain and inflammation (2.3.1), the patient's global assessment of pain (2.3.2), global assessment of disease (2.3.3), functional disability (2.3.4), acute phase reactants (2.3.5), and radiologic evidence of damage (2.3.6). The clinical evaluation of RA has been the subject of interesting debate. At the first conference of OMERACT (Outcome Measures in Rheumatoid Arthritis Clinical Trials), held in Maastricht in 1992, North Americans [Felson, 1993a; Felson 1993b] and Europeans [Scott, 1992] reached a historic consensus, with experts from different countries agreeing on a minimum set of parameters to be used in evaluating RA patients included in clinical trials [OMERACT, 1993]. These recommendations were subsequently ratified by the ACR [Felson, 1993a], the World Health Organization (WHO), the European Leagues Against Rheumatism (EULAR), and the International Leagues Against Rheumatism (ILAR) [Boers, 1994]. The parameters were chosen by consensus after examining the reliability, validity, and sensitivity of those most frequently used in the clinical evaluation of RA, with the aim of obtaining a set of parameters that would allow evaluation of all relevant aspects of the disease, without redundancy. This core set of parameters, which was originally selected for use in clinical trials, has been shown to be useful in daily clinical practice. The core set of parameters recommended by the OMERACT conference includes: 1) number of painful joints, 2) number of swollen joints, 3) pain, 4) the patient's overall assessment of disease, 5) the physician's overall assessment of disease, 6) physical functional capacity, and 7) acute phase reactants. Another parameter recommended in this conference for studies lasting at least 1 year was radiologic evaluation of damage, although more recent investigations have shown that radiographs of the hands and feet can detect changes in periods of 6 months [Sharp, 2000]. In any case, radiologic evidence of damage is not a parameter that can be clinically evaluated very often, therefore it should not be systematically assessed at all visits, but only once a year (see section 2.3.6.). There are clear advantages to using these parameters to monitor patients [Pincus, 1996; Wolfe, 1999a]. Nevertheless, rheumatologists do not systematically monitor patients with RA, and their use of different parameters for disease evaluation varies greatly [Bellamy, 1998; Bellamy, 1999]. According to the emAR study [Hernández-García, 2001], the use of quantitative measures of disease activity or functional damage is very limited in Spain. For example, joint counts were made at all or most visits in only 23% of 1,379 patients over a 2-year period; the figures for pain assessment or global assessment of disease status by visual analogue scale or the use of functional capacity questionnaires were still lower. The infrequent use of the OMERACT parameters and part of this variability could be explained by the fact that the application of standardized clinical instruments in daily practice involves additional time and effort in a high pressure setting. However, variability in the results of different treatments and the emergence of new, more costly and efficacious therapies with uncertain adverse events in the medium term will require closer patient monitoring if we aim to make more effective decisions at a socially acceptable cost. 2.3.1 Validated methods should be used to assess the number of painful joints and the number of swollen joints. For individual assessment of disease activity the use of a complete index is recommended, based on the examination of 68 joints for pain and 66 for swelling (excluding the hip). Articular indices assess the degree of pain and swelling by counting the number of painful joints and the number of swollen joints. Three different methods have been described, varying in the number of joints evaluated, although only four are in widespread use: * ACR Count. The ACR (previously the ARA - American Rheumatism Association) count can be defined as the most complete index [Deandrade, 1965; Williams, 1983; Ward, 1983; Paulus, 1984]. It is the US standard. It includes evaluation of tenderness in 68 joints and swelling in 66 joints (excluding both hips). The following joints are assessed: distal interphalangeal, proximal interphalangeal, metacarpophalangeal, wrist, elbow, shoulder, acromioclavicular, sternoclavicular, temporomandibular, hip (only for pain), knee, ankle, subtalar-midtarsal, metatarsophalangeal, and proximal interphalangeal joints. * Ritchie Index. This is the European index most commonly used. It includes assessment of pain alone in 53 joints and is calculated based on 26 joints, since some joints are considered together [Ritchie, 1968]. The following joints or groups of joints are evaluated: right and left proximal interphalangeal,(2), right and left metacarpophalangeal (2), wrist (2), elbow (2), shoulder (2), cervical spine (1), acromioclavicular (1), sternoclavicular (1), temporomandibular (1), hip (2), knee (2), ankle (2), subtalar (2), midtarsal (2), and right and left metatarsophalangeal (2) joints. This method quantifies joint tenderness or pain on motion only in the case of the cervical spine, hip, subtalar and midtarsal joints. Pain is scored on a 4-level scale: 0 = no pain; 1 = pain; 2 = pain and wincing; 3 = pain, wincing, and withdrawal (maximum score: 78). In the case of joint groups, the highest score assigned to any of the joints in the group is assigned to the whole group. * 44-Joint Index. Swelling is evaluated in the following 44 joints: proximal interphalangeal, metacarpophalangeal, wrist, elbow, shoulder, acromioclavicular, sternoclavicular, knee, ankle, and metatarsophalangeal joints. The fact that swollen joints are included in this index makes it a complement to the Ritchie index. * 28-Joint Index. Fuchs et al. [Fuchs, 1989] observed that a simple evaluation of tenderness and swelling in 28 joints provided the same sensitivity to change in clinical trials as more complex indices [Fuchs, 1994]. The index includes the following joints: proximal interphalangeal, metacarpophalangeal, wrist, elbow, shoulder, and knee joints. When counting joints, one can either make a simple count of the number of painful and swollen joints (present/absent) or semi-quantify the degree of pain and swelling in each joint using a 4-level ordinal scale (0-3). This guideline recommends counting painful and swollen joints without adding any type of quantification. The advantages obtained by quantifying are lost in the increased variability of the measurements. The ACR recommends the use of complete counts of 68 joints, although it later accepted the use of counts based on 28 joints in clinical trials. The same committee emphasized, however, that indices based on 28 joints exclude those in the foot and ankle, which are affected in over 50% of patients, therefore they provide less information at the individual level in daily clinical practice [OMERACT, 1994]. The use of a reduced index does not mean that these joints should not be examined. Thus, this guideline recommends the use of the ACR index with 68 joints. 2.3.2 Pain should be assessed by the patient him or herself. It is recommended that pain be measured using a horizontal visual analog scale, 10 cm in length, divided by vertical marks into ten equal 1-cm segments. The measurements should be accompanied by numeric descriptors from 0 to 10, with indicators at each end showing no pain (0) and worst pain (10). Pain is a subjective experience, therefore it should be quantified by the individual patient. Among the different methods used to measure pain, those that stand out due to their simplicity and ease of use are visual scales, or Likert-type scales, which usually have five or seven levels. The visual analog scale (VAS), with a horizontal 10 cm line and descriptors at each end, is the most widespread method. The ACR/OMERACT recommendations advise the use of either of the two methods, although there is a clear preference in studies for the VAS. Most patients are able to complete this scale. It is first necessary to devote some time to explaining the scale and giving a specific example; patients then answer quickly and with confidence. Some modifications, such as the use of numeric descriptors, may improve reliability in persons with a low educational level [Ferraz, 1990]. The VAS for pain shows a good correlation with the Likert scale, and both are sensitive to clinically important changes, with the VAS showing certain advantages [Langley, 1984; Anderson, 1993; Buchbinder, 1995]. 2.3.3 A global assessment of disease should be made from the medical point of view and another one from the patient's point of view. For this measurement, the use of a 10 cm horizontal visual analog scale is recommended, with vertical marks dividing it into 10 equal 1-cm segments. The measurements should be accompanied by numeric descriptors from 0 to 10, indicating at each end "very good" (0) and "very poor" (10). Global disease assessments by both the physician and the patient are useful because their evaluations may be quite different. The global assessment is very sensitive to clinical changes [Anderson 1989; Buchbinder, 1995]. 2.3.4 Self-perceived functional disability attributed to the disease should be evaluated using specific, previously validated questionnaires such as the Health Assessment Questionnaire (HAQ) (see Appendix 1). There are various ways to estimate functional capacity based on joint mobility or the ability to perform certain tasks as evaluated by an observer. The most widespread methods currently used consist of specific questionnaires for rheumatic disease such as the HAQ, the Modified Health Assessment Questionnaire (MHAQ) (shortened version of the HAQ), or the Arthritis Impact Measurement Scales (AIMS). They are based on the patient's own opinion about his or her disease. These questionnaires are standardized instruments, which have been shown to be valid and reliable. They evaluate those health dimensions that are most affected by RA, particularly disability, especially in relation to physical function, and pain. The HAQ is a 20-item, self-administered questionnaire that evaluates self-perceived physical disability to perform certain basic activities of daily living, grouped into eight areas: dressing and grooming, arising, eating, walking, hygiene, reach, grip, and other activities [Fries, 1980]. A Spanish version of the questionnaire has been validated [Esteve-Vives, 1993]. The MHAQ is a shortened version of the HAQ, with only eight items. Its main advantage is its simplicity, which makes it possible to use it in routine patient monitoring [Pincus, 1983]. The Spanish version of the MHAQ can be self administered by most patients with RA [Esteve-Vives, 1994]. This guideline recommends the use of the HAQ as a standardized instrument to evaluate disability because of its widespread dissemination, acceptance, and proven metric characteristics. Persons interested in evaluating broader aspects of health-related quality of life can also use the so-called generic questionnaires, such as the Short-Form 36 (SF-36), the Nottingham Health Profile (NHP), the Sickness Impact Profile (SIP), or the EuroQoL-5D. These questionnaires provide an estimate of self-perceived physical, psychological, and social health status based on questions about activities, feelings, and emotions covering a large number of daily life situations. The generic questionnaires provide complementary information and make it possible to compare the level of health with other diseases. 2.3.5 Laboratory tests should include two acute phase reactants (APRs): erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP). The behavior of these two APRs is closely related with the inflammatory activity of the disease. Laboratory tests should consist of a complete blood count, acute phase reactants (ESR, CRP), rheumatoid factor (RF), liver function (GOT, GPT, GGT, alkaline phosphate, albumin), kidney function (creatinine), calcium, and urinalysis. The presence of hepatitis B and C virus should be evaluated (in relation to the hepatotoxicity of some of the drugs used in treatment). These basic tests will facilitate RA monitoring and early detection of disease complications and side effects of treatment (see chapter 6). Measurement of APRs is very helpful in monitoring inflammatory processes in general, and of articular inflammation in particular. Their levels are associated with the intensity of underlying inflammation. There are several acute phase reactants but, in practice, the ESR and CRP are the most widely used. Both were included in the ACR recommendations and have been shown to be about equally useful in the evaluation of inflammatory activity [Paulus, 1999]. The advantage of the ESR is that it is inexpensive and available in any laboratory, and its disadvantage is its low specificity, since its levels can be modified by factors that are independent of articular inflammation. The advantage of the CRP is that its levels are virtually non-existent in the absence of inflammation, and its synthesis is closely related with inflammatory activity, with a very short half life. The techniques for measuring CRP are currently available in most laboratories. Persistent elevation of acute phase reactants with respect to normal reference values, especially for CRP, has been associated with poorer disease outcome [Dawes, 1986; van Leeuwen 1993; van Leeuwen, 1997]. Whether to include other complementary tests is left to the judgment of the individual physician, who should evaluate each case in terms of history, age, associated treatments, the possibility of using preventive interventions (e.g., cholesterol, glucose control), and associated co-morbidity. 2.3.6 Radiographs of the hands, feet, and chest are recommended at the initial evaluation. Radiographs of the feet and hands should be repeated annually for the first 3 years of disease evolution, and thereafter as deemed appropriate. One of the findings to be looked for in the radiographs are bony erosions, which are more frequent at the onset of disease. About 70% of patients have erosions in the hands or feet by the end of the first 2 or 3 years [Van der Heijde, 1995b; Hulsmans, 2000]. Their presence and the speed of onset are associated with poorer outcome. Radiographic changes are moderately associated with physical disability [Drossaers-Bakker, 1999; Scott, 2000]. Numerous methods have been described to quantify radiographic changes in the joints. Almost all of them are based on radiograph readings of the hands, although some authors have pointed out the importance of including a systematic evaluation of the feet [Van der Heijde, 1992b]. Most of these methods are based on the Larsen method [Larsen, 1977; Larsen, 1995; Edmonds, 1999] or on the Sharp method [Sharp, 1971; Sharp, 1985; Kaye, 1987; van der Heijde, 1992b; Sharp, 1995]. There is no clear preference for either one [Pincus, 1995], although the van der Heijde method [Van der Heijde, 1992b], which includes hands and feet, appears to have some advantage. The disadvantage of all these methods is that they are very time-consuming to apply, therefore they are generally reserved for use in research. This guideline recommends a simple qualitative evaluation to identify the presence of new erosions or their progression. Radiographs of both hands and feet are justified by the fact that asymmetrical erosions (right or left) may appear, and by the observation that in the first 2-3 years of the disease, erosions appear only on the feet, without clinical symptoms, in up to 23-36% of patients [Brook, 1977; Paimela, 1992; van der Heijde, 1999]. Other imaging techniques, such as magnetic resonance and sonography, have shown promising results in the early detection of inflammatory changes and structural damage like bony erosions [Wakefield, 2000]. These techniques involve the use of advanced technology that is not available in the vast majority of clinical centers. In addition, there is not enough evidence to date to make recommendations in this respect, thus their use remains reserved, for the moment, to the field of experimentation [Grassi, 2001]. With regard to the chest X-ray, a baseline radiograph is recommended, both to have knowledge of the baseline situation and to identify possible problems as the disease evolves and is treated. Table 13. Summary of instruments for the measurement of parameters in RA evaluation PARAMETER * Joint inflammation and pain VALID OPTIONS * ACR count * Ritchie index * 44-joint index * 28-joint index RECOMMENDATION * ACR count PARAMETER * Global assessment of pain VALID OPTIONS * VAS for pain rated by the patient * Likert scale RECOMMENDATION * VAS for pain rated by the patient PARAMETER * Global assessment of disease by the patient VALID OPTIONS * VAS * Likert scales for severity and/or activity RECOMMENDATION * VAS for disease rated by the patient PARAMETER * Global assessment of disease by the physician VALID OPTIONS * VAS * Likert scales for severity and/or activity RECOMMENDATION * VAS for disease rated by the physician PARAMETER * Functional capacity VALID OPTIONS * HAQ * MHAQ * AIMS RECOMMENDATION * HAQ PARAMETER * Laboratory tests VALID OPTIONS * ESR * CRP RECOMMENDATION * ESR and CRP PARAMETER * Radiographic damage VALID OPTIONS * Presence of absence of erosions * Sharp index * Larsen index RECOMMENDATION * Presence or absence of erosions evaluated qualitatively by radiography 2.4 An alternative procedure that is useful and valid in assessing disease activity is the use of composite indices summarizing the information for various parameters in a single indicator. If this method is preferred, this guideline recommends the use of the Disease Activity Score (DAS). Different composite indices have been published, and their validity has been reviewed in the framework of the OMERACT conference [OMERACT, 1993]. Some good examples are the Pooled index, the Mallya and Mace index, the Stoke index, the Scott index, and the DAS. These indices differ in the number of parameters included as well as the methods used for their calculation. Their advantages in comparison to conventional evaluation using single parameters are that they avoid duplicate measurements and are more sensitive to change. Their disadvantages are a certain degree of complexity in the calculations, difficulty of interpretation, and some problems related with how they are constructed. The DAS deserves particular mention [Van der Heijde, 1990; van der Heijde, 1992a]. It includes the Ritchie index (RI), number of swollen joints out of 44 joints (NSJ44), erythrocyte sedimentation rate (ESR), and the patient's global assessment of health (PGA) on a visual analog scale (0 cm "very good" - 10 cm "very poor"). The patient's global assessment of disease can be substituted for the global assessment of health, using the same scale. The DAS is calculated using the following formula: DAS=0.54 (RI) + 0.065 (NSJ44) + 0.33 (In ESR) +0.0072 (PGA) There is a modified DAS based on the number of painful joints (NPJ28) and swollen joints (NSJ28) out of 28 joints [Prevoo, 1995]: DAS28=0.56(NPJ28)+0.28(NSJ28)+0.70(In ESR)+0.014(PGA) The score for the complete DAS and the DAS28 can range from 0 to 10. The DAS is of particular interest because it is the basis for the EULAR improvement criteria [Van Gestel, 1996]. 2.5 The initial and subsequent evaluation of patients with RA should include a continual estimate of disease prognosis. The outcome of RA varies considerably among patients. Some treatment strategies, more aggressive and therefore more toxic, improve RA outcome when used early in patients with a high risk of serious disease, understood as functional disability, structural damage, and/or mortality. The decision of whether or not to use these aggressive strategies should be based on the prognosis in each individual patient. Since most radiographic changes and, to a lesser extent, loss of functional capacity, occur in the first 2 or 3 years of evolution, the earlier a disease prognosis is formulated, the sooner an informed decision can be made on the most appropriate treatment strategy. The factors that predict serious disease can be classified into three types: sociodemographic, disease-dependent and treatment-dependent. No single parameter, by itself, will permit estimation of RA outcome, therefore a combination of several parameters should be used. Furthermore, it is difficult to separate the individual effect of a particular risk factor from its interrelation with other factors associated with poor outcome. The following factors are considered to be predictive of functional disability, radiographic erosions, and/or mortality, and therefore of poor outcome: Sociodemographic factors: Female gender. Being a woman is associated with presentation of functional disability 4 years after disease onset (Odds Ratio=3.01) [Pease, 1999]. Not all cohort studies have reproduced this finding. Female gender is probably related with other prognostic factors. Age at disease onset. This is a controversial prognostic factor. Different groups have shown poorer, better, or equal outcome in elderly patients. Low educational level. This is associated with increased mortality. Less than secondary level education is associated with over 50% decrease in functional status or with mortality at 9 years (OR=7.5) [Pincus, 1985]. In Mexican patients with RA, fewer than 6 years of formal education is associated with severe forms of RA (OR=3.52) [Glave Testino, 1994]. Genetic markers. These have not been confirmed in all populations. They are limited to experimental use. Disease-dependent factors: Positive RF. Positive RF from 1/80 or = 60 IU by nephelometry is associated with the development of erosions (OR: 4.2 - 12) [Van der Heijde, 1995a]. The persistence of elevated RF is associated with erosions at 6 years follow-up. At 3 years from symptom onset, the presence of positive RF IgA is associated with more erosions, worse HAQ score, and more painful and swollen joints. Large number of swollen joints. A large number of swollen joints (greater than 20 at disease onset) is predictive of future activity, including mortality [Van Zeben, 1992]. Cumulative joint inflammation is associated with progression to radiographic damage in 1 year (OR=2) [Pincus, 1985]. Elevated acute phase reactants. A CRP of twice the normal value at the patient's initial evaluation is associated with the development of erosions in 4 years (OR=1.81) [Glave Testino, 1994]. A continuous ESR higher than 60 mm in the first hour is associated with the presence of disability at 18 years (OR=4.88) [Furst, 1994a]. Elevated HAQ at initial visit (Greater than or equal to 1 out of 3). This is associated with disability at 4 years (OR=3.02) [Pincus, 1985]. For each HAQ unit over 0 at the baseline visit, the OR for disability increases by 1.60 to 2.94 [Wolfe, 1998]. In patients with a baseline HAQ of at least 2.5, the relative risk for developing disability is 2.15 [Wolfe, 1991]. Early involvement of large joints (Greater than or equal to 2) is associated with the presence of erosions at 1 year (OR=2.03) [Brennan, 1996]. Rapid appearance of erosions (Greater than or equal to 2/year). The speed with which erosions appear is associated with poorer outcome. Presence of extra-articular manifestations (rheumatoid nodules, vasculitis, scleritis, or others). In general, these manifestations are associated with seropositive RF, therefore their prognostic value by themselves is unclear. The presence of extra-articular manifestations is associated above all with increased mortality [Gordon, 1973]. Treatment-dependent factors: Length of treatment. Longer treatment time with DMARDs is associated with better long-term functional outcome. For example, the difference between patients treated with DMARDs throughout the course of their disease and those who never received treatment is 0.53 HAQ units. [Fries, 1996]. Early treatment with DMARDs. Patients who delay beginning DMARD treatment have poorer functional outcome than those who begin treatment early. The longer the delay in beginning treatment, the smaller the probability of reaching a satisfactory response (OR=5.57), and this in turn implies poorer functional outcome (with a mean increase of 0.12 HAQ units for each visit in which a 50% improvement is not reached) [Tsakonas, 2000]. 2.6 Consideration should be given to the psychological and social effects on the patient, since these factors are involved in pain assessment and the development of disability. In addition to the evaluation of structural damage, disability, and health-related quality of life, certain important psychological and social factors should be evaluated. Psychological disturbances (depression and anxiety) are very frequent in RA from the time of disease onset [Van der Heijde, 1994], due to the impact of confronting its diagnosis and evolution. There is a close relation between depression, anxiety, and chronic pain. This can make the assessment (VAS rating by the patient and physician) more difficult and should be taken into account when planning treatment. In addition, anxiety and depression appear to play an important role in the development of disability [Escalante, 1999]. It is currently thought that some psychological characteristics of the patient (level of perceived helplessness, coping ability, level of self-management) play an important role as factors predictive of disability and health status. A high level of helplessness makes for a poorer outcome, whereas a greater coping ability and level of self-management improves it [Scharloo, 1999]. With regard to social factors, one third of patients lose their job during the first year of the disease [Jantti, 1999], in close association with their inflammatory activity [Wolfe, 1998; Reisine, 1998]. The reduced income associated with job loss affects all members of the family unit [Wolfe, 1998]. Patients who receive substantial social support from family and friends, especially from their spouses, have better outcomes and less disability [Fitzpatrick, 1991, Kraaimaat, 1995]. Some clinical manifestations, such as pain or fatigue, are more frequent in persons who lack social support [Riemsma, 1998]. 2.7 A detailed evaluation should be made to rule out latent tuberculosis infection before beginning treatment with immunosuppresants, anti-TNF agents, or corticosteroids in doses higher than 15 mg/day. If latent tuberculosis infection is present, prophylactic treatment with isoniazide is recommended. Treatment with anti-TNF agents or corticosteroids is a risk factor for tuberculosis (TB) in persons who have a primary infection with the Koch bacillus. A chest X-ray and tuberculin test should be included in the initial evaluation, with a repeat test (booster) in a week if the result is negative. A positive result is considered to be an induration larger than 10 mm, or larger than 5mm if the patient has a history of TB, or radiographic signs on a simple chest X-ray. If the tuberculin test is negative, the patient should be monitored for TB. If it is positive, the eradication treatment to use is isoniazide for a minimum of 4 months, and preferably 6. Prophylaxis can be given concomitantly with RA treatment if there is no evidence of liver toxicity. CHAPTER 3. CLASSIFYING RHEUMATOID ARTHRITIS 3.1 General classification The two characteristics that have the most influence on the initial classification between serious and mild disease, and therefore on the treatment decision, are the presence or absence of erosions and the number of swollen joints. The classification may be made more precise if other factors, such as APR, HAQ, and RF, are taken into account. The subsequent classification will depend on the response to treatment. Two types of RA with special characteristics are not included in this classification: "burnt-out" RA and pseudopolymyalgic RA (sections 3.2 and 3.3). RA cannot be neatly classified into different categories. In this guideline, the classification of patients is based on two principles: first, classifying RA is useful for making treatment decisions and estimating patient outcome; second, the classification should help the physician in actual practice. In accordance with these two principles, RA is classified based on the two parameters that, in the panel's opinion, have the most influence on the treatment decision and on outcome: the presence of erosions and the number of swollen joints. The use of two categories for the presence of erosions (yes/no) and two categories for the number of swollen joints (less than 6/greater than or equal to 6), gives four types of RA which, on a scale of increasing severity, would go from non-erosive disease with few swollen joints to erosive disease with swelling of multiple joints at clinical presentation. This simple alternative makes it possible to classify patients rapidly and is proposed in the recommendation for initial treatment in this guideline. While this guideline was being developed, we found that panelist disagreement about the initial treatment proposed was often due, not to a true difference of opinion, but to the fact that there are many different forms of RA. The differences arose because panelists were thinking about different types of patients within the same category. Thus, it was decided to include more variables in the categorization to be able to discriminate more precisely among different types of patients. Other factors were taken into account, such as APRs, HAQ, RF, and previous treatment with corticosteroids and/or NSAIDs. The combination of these factors resulted in 144 different scenarios describing different types of patients, from the mildest clinical presentation (no erosions, less than 6 swollen joints, normal APRs, HAQ less than 1 and negative RF) to the most severe form of disease (erosions present, greater than 10 swollen joints, elevated APRs, HAQ greater than or equal to 1 and high titers of positive RF). Each patient, according to the initial disease characteristics, should begin a specific treatment option (see chapter 4). If the response to initial treatment is unsatisfactory, the disease is considered to have a poor prognosis and thus should be considered as severe. The parameters that the physician should consider when classifying a patient within the continuous spectrum of disease severity are probably more extensive than those proposed in this guideline. The inclusion of characteristics such as age and time of evolution, among others, or the inclusion of additional categories for the parameters used would be feasible, however it would lead to an exponential increase in the number of types of RA. It is also debatable which parameters are most useful in classifying RA and which cut-off points best discriminate the milder forms of disease from those that are more severe. Since the evaluation and follow-up of RA patients should be based on longitudinal monitoring of measures of disease activity, joint damage, and health status, any classification should be made as a function of these parameters. Based on these parameters, we propose the two classifications of RA (simpler and more complex) included in this guideline. Nevertheless, new research may lead to a more precise classification of RA with implications for the clinical management of these patients. 3.2 "Burnt-out" or end-stage rheumatoid arthritis In "burnt-out" or end-stage RA there is no inflammatory activity and there is complete or practically complete destruction of the patient's joints. It is characterized clinically by joint pain at rest or with minimal exertion, joint deformities, significant muscular atrophy, extreme functional disability, and radiographic evidence of major joint destruction (erosions, subluxations, and ankylosis) [Schur, 1999]. The evaluation should rule out the possible presence of the extra-articular complications or manifestations of RA that most frequently appear at this stage of the disease, for example, skin ulcers, vasculitis, and amyloidosis [Gordon, 1973; Thurtle, 1983; Vollertsen, 1986; Breedveld, 1989]. Their management will be based on symptomatic treatment and repair of function. Special considerations should be taken into account for elderly patients (section 4.7). 3.3 Pseudopolymyalgic rheumatoid arthritis Pseudopolymyalgic rheumatoid arthritis is a disease that affects patients over 60 years of age and is characterized by the sudden onset of symptoms, mainly affecting the proximal joints (shoulders and hips) as well as the knees and carpal joints. It is accompanied by considerable morning stiffness, negative RF, and a marked increase in acute phase reactants. Erosions do not usually develop, and the prognosis is generally good. The disease may remit spontaneously in 6-24 months [Healey, 1986]. The main problem with pseudopolymyalgic RA is the difficulty of making a differential diagnosis since it is very similar to polymyalgia rheumatica. Cases diagnosed as pseudopolymyalgia may include cases of polymyalgia rheumatica and RS3PE (remitting seronegative symmetrical synovitis with pitting edema), and perhaps some atypical forms of other arthropathies that are clinically indistinguishable from RA, a fact that may partially explain the good evolution in this subgroup. It is usually managed effectively only with corticosteroids (section 4.5). If a satisfactory response is not obtained, it should be treated the same as RA in general, taking special considerations into account for elderly patients (section 4.7). CHAPTER 4. MEDICAL TREATMENT OF RHEUMATOID ARTHRITIS 4.1 Initial treatment of rheumatoid arthritis 4.1.1 In general, all patients with RA should be treated with a DMARD as soon as the disease is diagnosed. The aim of RA treatment is to induce complete disease remission or, alternatively, to achieve the best possible response. Treatment with DMARDs is the only way to ensure the most favorable evolution for the patient and to improve the quality of life. The function of each drug prescribed should be explained to patients so that they know to discontinue or reduce symptom-modifying drugs (NSAIDs or analgesics) if pain and inflammation subside, but that a DMARD should never be discontinued without medical consultation, and that instructions for the use of corticosteroids should be followed closely. All patients with RA should initiate DMARD treatment as soon as possible during the course of the disease. An attempt may be made to treat only with NSAIDs and/or corticosteroids for a maximum of 3 months, and only in patients who have not used these drugs during the 3 months before the disease was diagnosed, who have fewer than 6 swollen joints, no erosions, negative RF, and normal APRs. Many patients in Spain experience an unacceptably long delay before the first DMARD treatment. According to the emAR study, the time from first symptoms to first treatment with any DMARD is high, ranging from 7 to 48 months, with a median of 16 months [Hernández-García, 2001]. Furthermore, this interval increases with increasing time between first symptoms and the patient's first visit for specialized care. [Hernández-García, 2000]. The time between first symptoms and initiation of DMARD treatment is one of the few variables that the physician can modify. Early commencement of treatment is associated with a higher probability of favorable response [Anderson, 2000; Villaverde, 2000] and a lower probability of functional and radiologic deterioration [Fries, 1996; Abu-Shakra, 1998]. Thus, it is essential not only that all patients with RA be treated with DMARDs, but that this treatment should be initiated as early as possible in the course of the disease. All the DMARDs in Table 1 have been shown to be more efficacious than placebo (Table 11). However, not all possible drug combinations in monotherapy or combined therapy have been compared in clinical trials (Tables 7 and 8). For years DMARDs were used as monotherapy. If the response to one DMARD was not complete, it was discontinued and a second DMARD was initiated. This strategy often caused a reactivation of disease between the time one drug was withdrawn and the other begun. The fact that many RA patients are refractory to treatment led to the first use of combinations of two or more DMARDs [Ehrlich, 1982; Bitter, 1984; Gibson, 1987; Taggart, 1987; Scott, 1989; Williams, 1992; McCarty, 1995; Willkens, 1995]. The use of combined treatments slowed down, however, partly due to the results of a meta-analysis of five initial studies which concluded that combined therapy was no more efficacious and furthermore was more toxic than monotherapy [Felson, 1994]. The publication in the last decade of various clinical trials with sound methodology and a sufficient number of patients has encouraged renewed use of combined therapies in RA treatment [Tugwell, 1995; O'Dell, 1996; Stein, 1997; Boers, 1997; Haagsma, 1997]. The good results obtained using combined treatment in patients refractory to monotherapy have led some authors to propose they be used as sustained initial treatment or by sequentially discontinuing some of the drugs [Wilske, 1989; Boers, 1997]. The corticosteroids have also gained renewed acceptance as part of this "aggressive" strategy, especially while waiting for the DMARDs to take effect [Kirwan, 1995]. The introduction of new DMARDs opens the range of possibilities in RA treatment even wider. Some of these strategies are moving into daily practice. For example, in Spain almost 28% of patients included in the emAR study [Hernández-García, 2001] used some combination of two or more DMARDs throughout the study period. Another 72% followed some kind of treatment with corticosteroids. The impact of new treatments on the treatment strategy for RA is unknown. Whatever the case may be, with the limitations derived from our imperfect knowledge, RA patients have recourse to a wide range of strategies aimed at controlling their disease, which can be used pending the results of new comparative studies. 4.1.2 In addition to DMARD treatment, all RA patients should be treated with optimum doses of steroidal or nonsteroidal anti-inflammatory agents and/or analgesics if symptoms (pain and swelling) persist (sections 4.4, 4.5 and 4.6). Regardless of DMARD treatment, all patients also require treatment with symptom-modifying drugs (NSAIDs and/or corticosteroids and/or analgesics) so long as there are persistent symptoms of joint pain and/or swelling. 4.1.3 Because of its efficacy and toxicity profile, methotrexate is the recommended initial treatment in all patients who have not previously received DMARD treatment. Nevertheless, initial treatment with other drugs is considered acceptable, depending on the clinical classification of disease (Table 14 or Appendix 2). The advantages of methotrexate over other DMARDs with similar short-term efficacy are a well-known toxicity profile, easy management, and a lower rate of withdrawal from treatment in the medium and long term. For all these reasons, it is recommended as the drug of choice in this guideline. The administration of 5 to 10 mg/wk of folic acid is recommended for RA patients treated with MTX [American College of Rheumatology, 1996]. Despite this recommendation, only slightly more than half of RA patients who receive MTX in Spain are treated with folic acid or folinic acid [Hernández-García, 2001]. Due to the clinical complexity of RA, the panel considers that the use of other DMARDs or DMARD combinations of proven efficacy is valid as initial treatment in some clinical situations, in accordance with the treatment scheme in Appendix 2 or Table 14. Although the vast majority (93%) of RA patients who receive specialized care are treated with DMARDs, there is wide variability in their use in Spain. According to data from the emAR study, during a 2-year period and considering both monotherapy and combined therapy, 46% of patients had taken MTX, 21% anti-malarials, 14% IG, 8% SSZ, and 6% CSA. Among the factors associated with the use of each DMARD are patient age; some disease characteristics, such as functional class, rheumatoid factor, or disease activity; and the existence of associated disease. However, there is also significant variability that is associated, not with patient characteristics, but with physician specialty or characteristics, or with geographic region. Thus, for the same patient, the probability of receiving a particular DMARD depends on whether or not the prescribing physician is a specialist in rheumatology, his or her age, or the autonomous community of residence. About one-forth of patients treated with DMARDs use a combination of two or more drugs, although this percentage varies considerably between and among autonomous communities. The most frequent combinations include MTX and anti-malarials, followed by MTX with SSZ, MTX with CSA, and MTX with IG [Hernández-García, 2001]. Table 14. Initial treatment by simplified clinical classification of RA SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * less than 6 swollen joints Recommended initial treatment, by order of preference (supporting evidence) * Methotrexate (1) * Sulphasalazine (2) * Chloroquine (3) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * Greater than or equal to 6 swollen joints Recommended initial treatment, by order of preference (supporting evidence) * Methotrexate (1) * Injectable gold (4) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * less than 6 swollen joints Recommended initial treatment, by order of preference (supporting evidence) * Methotrexate (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * Greater than or equal to 6 swollen joints Recommended initial treatment, by order of preference (supporting evidence) * Methotrexate (1) * Leflunomide (5) * Methotrexate + injectable gold (6) 1. Methotrexate is more efficacious than oral gold (A1 evidence ) or azathioprine (A2 evidence). No significant differences have been found in the efficacy of methotrexate compared with etanercept, leflunomide, sulphasalazine (A1 evidence), injectable gold (A2 evidence), cyclosporin, or infliximab (B evidence). 2. Sulphasalazine is more efficacious than hydroxychloroquine (A2 evidence), and no significant differences have been found in the efficacy of sulphasalazine compared with leflunomide, methotrexate (A1 evidence), oral or injectable gold, and D-penicillamine (B evidence). 3. Chloroquine is not significantly different in efficacy from cyclosporin, oral gold (A2 evidence), azathioprine, injectable gold and D-penicillamine (B evidence). 4. Injectable gold is not significantly different in efficacy from oral gold (A1evidence), cyclosporin and methotrexate (A2 evidence), or chloroquine, D-penicillamine and sulphasalazine (B evidence). It is less efficacious than azathioprine and cyclophosphamide (B evidence). 5. Leflunomide (A1 evidence) shows no differences in efficacy as compared to methotrexate and sulphasalazine (A1 evidence). 6. No clinical trials have evaluated the efficacy of treatment with methotrexate+injectable gold (C evidence). 4.2 Changes in treatment After initiating any treatment, it is necessary to evaluate the response (chapter 5) and to monitor its toxicity (chapter 6). Treatment failure or toxicity should be evaluated within a maximum of 3 months, and a consequent change in treatment should be considered. Whatever initial treatment is chosen, the patient should be monitored closely. If a satisfactory response is not obtained within 3 months, or if there is evidence of DMARD-related toxicity, consideration should be given to the possibility of a change in treatment by adding a new drug or modifying the dosage. It is critically important that a patient with RA who has not responded to a particular treatment with single or combined DMARDs have the option to try other treatments of proven efficacy in the shortest possible period of time. 4.2.1 If serious adverse effects appear, an alternative treatment should be substituted for the treatment of first choice, in accordance with Table 15. Table 15. Alternative treatment in case of severe toxicity to initial treatment SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * less than 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Leflunomide (1) * Injectable gold (2) * Sulphasalazine (4) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * less than 6 swollen joints First-choice treatment used * Sulphasalazine Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Methotrexate (3) * Injectable gold (2) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * less than 6 swollen joints First-choice treatment used * Chloroquine Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Methotrexate (3) * Injectable gold (2) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * Greater than or equal to 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Leflunomide (1) * Injectable gold (2) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * Greater than or equal to 6 swollen joints First-choice treatment used * Injectable gold Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Methotrexate (3) * Leflunomide (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * less than 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Leflunomide (1) * Injectable gold (2) * Sulphasalazine (4) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * Greater than or equal to 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Leflunomide (1) * Injectable gold (2) * Sulphasalazine (4) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * Greater than or equal to 6 swollen joints First-choice treatment used * Leflunomide Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Methotrexate (3) * Anti-TNF (5) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * Greater than or equal to 6 swollen joints First-choice treatment used * Methotrexate+injectable gold Alternative treatment in case of toxicity, in order of preference (supporting evidence) * Leflunomide (1) * Anti-TNF (5) The evidence supporting the alternative treatments proposed in case of toxicity or unsatisfactory response is as follows: 1. Leflunomide (A1 evidence) shows no differences in efficacy as compared to methotrexate and sulphasalazine (A1 evidence). 2. Injectable gold has not been shown to have significant differences in efficacy as compared to oral gold (A1 evidence), cyclosporin and methotrexate (A2 evidence), or chloroquine, D-penicillamine and sulphasalazine (B evidence). It is less efficacious than azathioprine and cyclophosphamide (B evidence). 3. Methotrexate is more efficacious than oral gold (A1 evidence) or azathioprine (A2 evidence). No significant differences in the efficacy of methotrexate have been found in comparison with etanercept, leflunomide, sulphasalazine (A1 evidence), injectable gold (A2 evidence), cyclosporin, or infliximab (B evidence). 4. Sulphasalazine is more efficacious than hydroxychloroquine (A2 evidence) and no significant differences have been found in the efficacy of sulphasalazine compared with leflunomide, methotrexate (A1 evidence), oral or injectable gold, and D-penicillamine (B evidence). 5. Anti-TNF agents (infliximab and etanercept) have been shown to be efficacious in the treatment of RA (A1 evidence), and they show no significant differences in efficacy with respect to methotrexate (B evidence for infliximab and A1 for etanercept). 4.2.2 If the treatment shows no toxicity but the response is unsatisfactory, even after using the maximum dosage (see 4.3), an alternative treatment should be substituted for the treatment of first choice, in accordance with Table 16. Table 16. Alternative treatment in case of unsatisfactory response to initial treatment SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * less than 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Leflunomide (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * less than 6 swollen joints First-choice treatment used * Sulphasalazine Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Methotrexate (2) * Leflunomide (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * less than 6 swollen joints First-choice treatment used * Chloroquine Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Methotrexate (2) * Leflunomide (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * Greater than or equal to 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Leflunomide (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * No erosions * Greater than or equal to 6 swollen joints First-choice treatment used * Injectable gold Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Methotrexate (2) * Leflunomide (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * less than 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Leflunomide (1) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * Greater than or equal to 6 swollen joints First-choice treatment used * Methotrexate Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Leflunomide (1) * Anti-TNF (3) * Methotrexate+ anti-TNF (4) * Methotrexate+chloroquine+sulphasalazine (5) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * Greater than or equal to 6 swollen joints First-choice treatment used * Leflunomide Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Methotrexate (2) * Anti-TNF (3) * Methotrexate+ anti-TNF (4) SIMPLIFIED CLINICAL CLASSIFICATION OF RA * Erosions present * Greater than or equal to 6 swollen joints First-choice treatment used * Methotrexate+injectable gold Alternative treatment in unsatisfactory response, in order of preference (supporting evidence) * Leflunomide (1) * Anti-TNF (3) 1. Leflunomide (A1 evidence) has not shown differences in efficacy compared with methotrexate and sulphasalazine (A1 evidence). 2. Methotrexate is more efficacious than oral gold (A1 evidence) or azathioprine (A2 evidence). No significant differences in efficacy have been found in methotrexate as compared to etanercept, leflunomide, sulphasalazine (A1 evidence), injectable gold (A2 evidence), cyclosporin, or infliximab (B evidence). 3. Anti-TNF agents (infliximab and etanercept) have been shown to be efficacious in the treatment of RA (A1 evidence) in comparison with placebo, and they show no significant differences in efficacy as compared to methotrexate (B evidence for infliximab and A1 for etanercept). 4. The combination of methotrexate+anti-TNF agents (infliximab or etanercept) has been shown to be more efficacious than methotrexate alone (B evidence). 5. The combination of methotrexate+chloroquine+sulphasalazine has been shown to be more efficacious than methotrexate alone or chloroquine+sulphasalazine (A2 evidence). In addition to the panel's recommendations, there is scientific evidence regarding the efficacy of several drug combinations in case of failure of treatment with methotrexate or the antimalarials. In case of failure with methotrexate, the following combinations have been shown to be more efficacious: * Methotrexate+cyclosporin (A1 evidence) * Methotrexate+chloroquine (A2 evidence) * Methotrexate+azathioprine (B evidence) In case of failure with the antimalarials, the following combinations have been shown to be more efficacious: * Sulphasalazine+hydroxychloroquine (A2 evidence) * Methotrexate+hydroxychloroquine (B evidence) 4.2.3 In patients for whom the preceding guidelines are not useful, due to lack of efficacy, toxicity, or other causes, this guideline recommends the use of any DMARD or DMARD combination of proven efficacy; if these fail, experimental treatments are recommended. The natural history of RA is long and no treatment has been shown to cure all patients. Thus, regardless of the recommendations in the preceding sections, it is admissible to introduce alternative treatments whose efficacy has been shown in clinical trials (Tables 7 and 8.) If disease control is not achieved with any of the proposed treatments, experimental treatments may be explored (new drugs or new combinations of existing drugs) so that the patient is never without some type of disease-modifying treatment. 4.3 Dosage for disease-modifying antirheumatic drugs Table 17. Recommended doses and commercial names of DMARDs DRUG * AZATHIOPRINE DOSE * 1.5 - 2.5 mg/kg/day, by mouth * Begin with low dose of around 1 mg/kg/day and increase in 4-6 weeks to maintenance dose of 100-150 mg/day. COMMERCIAL NAMES * IMURAN® Tab. 50 mg * IMURAN®, Freeze-dried vial, 50 DRUG * CYCLOPHOSPHAMIDE DOSE * 1.5 - 2.5 mg/kg/day, by mouth * Begin with 50 mg/day and increase dose every 4-6 weeks until a response is obtained, without exceeding 2.5 mg/kg/day. COMMERCIAL NAMES * GENOXAL® Amp. IV 1000 mg * GENOXAL® Amp. IV 200 mg * GENOXAL® Tab. 50 mg DRUG * CHLOROQUINE DOSE * 250 mg/day, by mouth * Do not exceed 4 mg/kg/day. COMMERCIAL NAMES * RESOCHIN® Tab. 250 mg DRUG * CYCLOSPORIN DOSE * 2.5 - 5.0 mg/kg/day, by mouth * The initial dose can be increased by 0.5mg/kg/day every 2 weeks up to 5 mg/kg/day. COMMERCIAL NAMES * SANDIMMUNE NEORAL® 100 mg * SANDIMMUNE NEORAL® 50 mg * SANDIMMUNE NEORAL® 25 mg * SANDIMMUNE NEORAL® Oral sol. 100mg/ml DRUG * D-PENICILLAMINE DOSE * 125 - 500 mg/day, by mouth * Begin treatment with 125-250 mg/day and if there is no improvement, increase dose at 8 weeks by 125 mg/day. Dose can be increased gradually every 8 weeks up to 500-750 mg/day. Should be administered 2 hrs before the main meal. COMMERCIAL NAMES * CUPRIMENE® Caps .250 mg * CUPRIMENE® Caps .125 mg * CUPRIMENE® Tab.50 mg * SUFORTANON® Tab. 250 mg DRUG * ETANERCEPT DOSE * 25 mg, reconstituted in 1 ml water, in subcutaneous injection, twice a week at intervals of 72-96 hours * One dose of 25 mg administered once a week offers a slower response and may be less effective. COMMERCIAL NAMES * ENBREL® Vial 25 mg DRUG * HYDROXYCHLOROQUINE DOSE * 400 mg/day, by mouth * Do not exceed 6.5 mg/kg/day COMMERCIAL NAMES * PLAQUENIL® Tab. 200 mg DRUG * INFLIXIMAB DOSE * 3 mg/kg intravenous perfusion for 2 hours * Then administer additional doses of 3 mg/kg in perfusion at weeks 2 and 6 following the first week, and one dose every 8 weeks thereafter. Dose may be increased to 5 mg/kg if ineffective or in case of relapse. Some patients require a shorter interval of infusion every 4-6 weeks, instead of the recommended 8 weeks for maintenance. Infliximab should be administered together with methotrexate. COMMERCIAL NAMES * REMICADE® Freeze-dried vial 100 mg DRUG * LEFLUNOMIDE DOSE * 20 mg/day, by mouth * Begin with 100 mg/day for 3 days and then 20 mg/day continuously. COMMERCIAL NAMES * ARAVA® Tab.100 mg * ARAVA® Tab.20 mg * ARAVA® Tab.10 mg DRUG * METHOTREXATE DOSE * 7.5-10 mg/week, by mouth for 4 weeks, 15 mg/week for the next 4 weeks, and then increase up to 20 mg/week. In case of inefficacy or gastrointestinal toxicity, consider parenteral administration. * Administer folic acid (5-10 mg/week) 24 hours after methotrexate. COMMERCIAL NAMES * METHOTREXATE ALMIRALL® Inj. sol. Vial 50 mg , A.D.1000 mg, 5000 mg, and 500 mg * METHOTREXATE LEDERLE® Tab. 2,5 mg; Inj. sol. 25 mg/ml (2, 20, 40 and 200 ml); Freeze-dried vial 50 and 500 mg * METHOTREXATE WASSERMANN® Inj. sol. 25 mg/ml (2 and 20 ml) * EMTHEXATE® Vial 50 and 500mg/2ml DRUG * ORAL GOLD DOSE * 6 mg/day by mouth 2 tablets per day. COMMERCIAL NAMES * RIDAURA® Tab. 3 mg * CRISINOR® Tab. 3 mg DRUG * INJECTABLE GOLD DOSE * 50 mg/week in intramuscular injections * Increasing doses of 10, 25 and 50 mg/week, maintaining the dose (from 6 to 24 months) or adjusting it depending on clinical response or adverse effects. COMMERCIAL NAMES * MIOCRIN® Inj. sol. IM 10 mg * MIOCRIN® Inj. sol. IM 25 mg * MIOCRIN® Inj. sol. IM 50 mg DRUG * SULPHASALAZINE DOSE * 2-3 g/day, by mouth COMMERCIAL NAMES * SALAZOPYRIN® Tab. 500 mg 4.4 Treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) 4.4.1 The NSAIDs are used to modify the symptoms of RA. The use of NSAIDs is recommended at disease onset, when a new DMARD is introduced, and when uncontrolled isolated symptoms persist despite good response to a DMARD. The NSAIDs are used to modify the symptoms of RA. They have not been shown to have any additional effect on disease outcome. The use of NSAIDs is recommended: * At disease onset. If the disease is low risk (less than 6 swollen joints, no erosions, negative RF, and normal APRs), they can be used alone or in combination with corticosteroids for a maximum of 3 months. * When a new DMARD is introduced. NSAIDs should be used until the disease and its symptoms can be controlled by the DMARD alone. NSAIDs should be used for 2-12 weeks, depending on the time needed for the DMARD to reach effective therapeutic levels. The period of combined use may sometimes be prolonged until the DMARD dose is adjusted. * When uncontrolled symptoms persist. NSAIDs should be used when, despite treatment with a DMARD, some isolated symptom (painful inflammation or swelling or morning stiffness) is not sufficiently controlled, and there is no evidence of inflammatory activity that justifies increasing the DMARD dose or changing to a new treatment. NSAIDs should not be used without first trying other analgesics such as acetaminophen for mechanical pain (pain that worsens with exercise and improves with rest, becomes worse during the day, with no joint stiffness after rest). It is important to weigh the benefit-risk relation for the patient whenever an NSAID is used. The side effects and interactions of the NSAIDs used should be known. 4.4.2 All NSAIDs should be used at the full dose for at least 1 week before considering the treatment to have failed. Once symptoms have been controlled, the minimum effective dose should be used. When the NSAIDs are withdrawn after prolonged use (over 3 months), they should be discontinued gradually to avoid the effects of rebound pain. No guidelines for withdrawal have been shown to be more effective than others. Length of treatment with NSAIDs is a risk factor for gastric erosion. Table 18. Usual dosage and frequency of administration of NSAIDs DRUG * AAS TOTAL DOSE (mg/24 h) * 3,000 - 6,000 FREQUENCY OF ADMINISTRATION * 6-8 h. DRUG * Ibuprofen TOTAL DOSE (mg/24 h) * 1,200 - 2,400 FREQUENCY OF ADMINISTRATION * 8 h. DRUG * Flurbiprofen * Flurbiprofen Retard TOTAL DOSE (mg/24 h) * 200 - 300 * 200 FREQUENCY OF ADMINISTRATION * 12 h. * 24 h. DRUG * Mefenamic acid TOTAL DOSE (mg/24 h) * 750 - 1,500 FREQUENCY OF ADMINISTRATION * 8 h. DRUG * Meclofenamic acid TOTAL DOSE (mg/24 h) * 200 - 400 FREQUENCY OF ADMINISTRATION * 8 h. DRUG * Diflunisal TOTAL DOSE (mg/24 h) * 500 - 1,000 FREQUENCY OF ADMINISTRATION * 12 h. DRUG * Sodium Tolmetin TOTAL DOSE (mg/24 h) * 800 - 1,200 FREQUENCY OF ADMINISTRATION * 6-8 h. DRUG * Naproxen TOTAL DOSE (mg/24 h) * 500 - 1,000 FREQUENCY OF ADMINISTRATION * 12 h. DRUG * Ketoprofen * Ketoprofen Retard TOTAL DOSE (mg/24 h) * 200 * 200 FREQUENCY OF ADMINISTRATION * 8-12 h. * 24 h. DRUG * Aceclofenac TOTAL DOSE (mg/24 h) * 200 FREQUENCY OF ADMINISTRATION * 12 h. DRUG * Diclofenac * Diclofenac Retard TOTAL DOSE (mg/24 h) * 150 - 200 * 100 FREQUENCY OF ADMINISTRATION * 8-12 h. * 24 h. DRUG * Phenylbutazone TOTAL DOSE (mg/24 h) * 200 - 400 FREQUENCY OF ADMINISTRATION * 12-24 h. DRUG * Indomethacin TOTAL DOSE (mg/24 h) * 75 - 150 FREQUENCY OF ADMINISTRATION * 8 h. DRUG * Sulindac TOTAL DOSE (mg/24 h) * 200 - 400 FREQUENCY OF ADMINISTRATION * 12 h. DRUG * Piroxicam TOTAL DOSE (mg/24 h) * 20 FREQUENCY OF ADMINISTRATION * 24 h. DRUG * Tenoxicam TOTAL DOSE (mg/24 h) * 20 FREQUENCY OF ADMINISTRATION * 24 h. DRUG * Droxicam TOTAL DOSE (mg/24 h) * 20 FREQUENCY OF ADMINISTRATION * 24 h. DRUG * Meloxicam TOTAL DOSE (mg/24 h) * 7.5 - 15 FREQUENCY OF ADMINISTRATION * 24 h. DRUG * Nimesulide TOTAL DOSE (mg/24 h) * 200 FREQUENCY OF ADMINISTRATION * 12 h. DRUG * Nabumetone TOTAL DOSE (mg/24 h) * 1,000-2,000 FREQUENCY OF ADMINISTRATION * 12-24 h. DRUG * Rofecoxib TOTAL DOSE (mg/24 h) * 12.5 - 25 FREQUENCY OF ADMINISTRATION * 24 h DRUG * Celecoxib TOTAL DOSE (mg/24 h) * 200 - 400 FREQUENCY OF ADMINISTRATION * 12-24 h 4.4.3 There is no evidence that some NSAIDs are better than others, therefore the one that best fits the patient characteristics should be used. There is no evidence that the efficacy of combined NSAIDs is greater than each NSAID alone. In a recent review of different meta-analyses and trials comparing the efficacy of the NSAIDs, it was not possible to show that some NSAIDs are more efficacious than others, although it was shown that they have different safety profiles, in favor of ibuprofen [Gotzsche, 2000a]. No controlled clinical trial of sufficient size has compared the efficacy of the NSAIDs to acetaminophen. There are no convincing studies showing that specific patients benefit more from some NSAIDs than from others. Generally, different NSAIDs are tried until symptom control is reached. A large number of different NSAIDs is available in Spain, thus it is important to know them all, especially their different pharmacokinetic profiles, in order to adapt them to the patient's needs. Some NSAIDs, such as naproxen or acetylsalicylic acid (ASA) have more rapid uptake (about 20 minutes), and thus would be indicated for acute pain. Others with delayed uptake and prolonged action (retard forms) can be administered at night so they will act when the patient wakes up. Selective cyclooxigenase inhibitors of the COX-2 isoenzyme, or coxibs (refecoxib, celecoxib, and others under investigation), have not been shown to have a safety profile significantly better than other NSAIDs, except in the gastrointestinal system [Schnitzer, 1999; Simon, 1999; Emery, 1999; Langman, 1999]. They should be used in patients at risk for gastroduodenal ulcers. Patients with cardiovascular disease can benefit from the platelet-inhibitory action of the NSAIDs, which is not shared by the coxibs. The SER (Spanish Society of Rheumatology) guidelines for the rational use of coxibs are recommended [SER, 2000a]. 4.4.4 The need for co-treatment with gastric protectors should be evaluated on an individual basis. Since the NSAIDs are associated with a high frequency of gastrointestinal adverse effects and are often used for prolonged periods, the need for a gastric protector should be evaluated in accordance with other existing risk factors for gastroduodenal ulcers. 4.5 Treatment with corticosteroids 4.5.1 The use of oral corticosteroids at low doses is recommended in patients in whom NSAIDs are not effective or are contraindicated for any reason. They can be used instead of NSAIDs or in association with them. The corticosteroids should never replace treatment with DMARDs, unless their possible role as a disease modifying agent should be shown. They are indicated as the treatment of choice only in the case of pseudopolymyalgic RA (section 3.3). Corticosteroids should be used: * When NSAIDs are contraindicated or have a high risk of adverse effects (the elderly, associated morbidity). * As bridge therapy until the onset of DMARD action. * When NSAIDs do not effectively control inflammation (generally, by adding corticosteroids to the NSAID treatment). * In the treatment of pseudopolymyalgic RA. It is not clear what role the corticosteroids should play in RA treatment. More studies are needed to analyze their efficacy in comparison with the NSAIDs. However, it is doubtful that corticosteroids alone should constitute an alternative to NSAIDs if the latter treatment fails and cannot be considered as first-line treatment when rapid control of inflammation is needed. Some studies that have used a high initial dose, especially in very aggressive RA [Boers, 1997], indicate that treatment with methylprednisolone pulses may constitute an alternative for the initial control of disease activity (always in association with a DMARD and pending the onset of its effect). 4.5.2 Low-doses of oral corticosteroids (less than 15 mg/day of prednisone or the equivalent) are an effective anti-inflammatory treatment in RA. Doses not exceeding 15 mg/day of prednisone should be used during the shortest possible period of time, and maintenance doses should not exceed 10 mg/day of prednisone or the equivalent. The use of corticosteroids in RA treatment is controversial, but they are used frequently, especially in the long term. The corticosteroids are better than placebo and similar to or better than NSAIDs or chloroquine in controlling RA activity [Saag, 1997; Criswell, 2000; Gotzsche, 2000b]. Several authors have studied the role of the corticosteroids in RA management from different perspectives: as disease modifiers [Kirwan, 1995; Hickling, 1998; Weiss, 1989] and as "bridge" therapy while waiting for the DMARDs to take effect [Harris, 1983; Caldwell, 1991; Van Gestel, 1995]. The use of corticosteroids has been associated with increased mortality, and their chronic use, at low doses, is related with increased morbidity. It is difficult, however, to separate the effect of corticosteroid use from the fact that patients who usually take them have more severe disease that cannot be controlled with NSAIDs alone. 4.5.3 No corticosteroid preparation has been shown to be superior to any other, thus any of them can be used at equivalent doses. There is currently no evidence that the most commonly used preparations (prednisone, prednisolone, methylprednisolone, and deflazacort) are significantly different in efficacy or adverse effects when used at equivalent doses. The dosage of corticosteroids always depends on the underlying disease for which they are prescribed, and on its clinical and biological activity. Whenever possible, a single daily dose should be recommended at the beginning of the day. The dose should be progressively reduced (changing fractionated doses to a single dose before decreasing the dose). Table 19. Classification of the corticosteroids by duration of action Short-acting corticosteroids * Hydrocortisone, prednisone, and prednisolone Intermediate-acting corticosteroids * Methylprednisolone, paramethasone, triamcinolone, and deflazacort Long-acting corticosteroids * Betamethasone and dexamethasone 4.5.4 Given the association between corticosteroid use and rapid loss of bone mass, the use of vitamin D plus calcium is recommended as a minimum measure; if treatment is expected to exceed 3 months, other preventive osteoporosis treatments (section 8.4) should be evaluated. 4.6 Treatment for pain 4.6.1 Analgesics are indicated to control pain. If there is no response, surgical treatment can be considered, especially to restore function and mobility. Pain control treatment should be instituted if pain persists despite the adoption of previous disease-control measures. Simple analgesics (e.g., acetaminophen, ASA) should be used first, increasing to the maximum dose of 3-4 g/day in the case of acetaminophen and up to 4gr/day for ASA. If pain persists, dipyridamole, NSAIDs, or codeine may be used. If pain is due to neuropathy, tricyclic antidepressants (amitryptiline) and some anticonvulsants (gabapentine or carbamazepine) may be used. When pain is very localized, local analgesics such as capsaicin cream may be used. The ideal dose is 0.75 mg of cream (not available in Spain). Surgical treatment should be considered when pain does not respond to pharmacological treatments and is due to joint destruction, producing changes in the patient's functional capacity [Dunbar, 1998]. If pain is intense, there is no response to previous analgesic treatments, and surgery is not an option, opiate analgesics may be administered. [Hazes, 1994; Schur, 1999]. 4.7 Special considerations in the treatment of elderly patients 4.7.1 Kidney and liver function should be monitored in elderly patients, and the dosage intervals of the drugs eliminated by these routes should be adapted accordingly. Aging may be accompanied by changes in various organs, especially those responsible for metabolizing and excreting different drugs. This means that the pharmacokinetic and pharmacodynamic properties of a large number of drugs used in elderly patients may be different than in younger individuals [Morgan, 1986; Bird, 1990]. Optimal pharmacological treatment in a particular patient depends on a variety of factors, which are frequently not well known or are difficult to determine; this fact may contribute to the large variability among different individuals in the response to the same drug, a phenomenon that is especially notable in the elderly [Bird, 1990]. The dosage of drugs eliminated by the renal route should be adjusted so that it is similar to what is used in patients with renal failure (decreasing the dose and/or lengthening the intervals between doses). Even in the absence of kidney disease, renal clearance in elderly individuals is decreased by 35-50%. The elderly, and especially those who suffer RA, have reduced muscular mass, which produces a decline in the production of creatinine. Thus, an elderly individual may have a normal creatinine value even though creatinine clearance is altered. [Oates, 1998]. Aging may also produce alternations in hepatic function, thus the metabolization of drugs that are broken down in the liver may also be reduced [Morgan, 1986]. 4.7.2 The possible appearance of adverse effects and drug interactions should be monitored in elderly patients. Adverse drug effects have traditionally been considered more frequent in elderly individuals [Hurwitz, 1969; Dahl, 1990], although little information is available about most drugs in this age group, including those used in RA patients. The lack of data is due to the frequent exclusion of extreme age groups in clinical trials. For this reason, unexpected side effects are not uncommon in individuals with late onset RA, once the drugs have come into generalized use [Morgan, 1986; Dahl, 1990]. In general, elderly patients have more than one disease and need treatment with multiple drugs. This means there is an increased probability of drug interactions and contributes to a larger number of side effects [Buchan, 1991]. The use of multiple drugs in elderly patients is often accompanied by a lack of treatment compliance, which has been estimated at 10% [Bird, 1990]. The DMARDs and the immunosuppressors have a similar efficacy and safety profile in young and old individuals, although for the reasons mentioned above, toxicity should be monitored more closely in the elderly [O´Callaghan, 1986]. 4.8 Special considerations in the treatment of rheumatoid arthritis during pregnancy 4.8.1 Women of childbearing age should be informed of the possible effects of RA on pregnancy, in particular, because of the implications for treatment. There is no evidence that RA has a negative effect on pregnancy outcome. However, treatment with DMARDs can have negative consequences on pregnancy, the fetus, and breastfeeding. Thus, women of childbearing age should know the risk so they can act accordingly. The symptoms of RA disappear during pregnancy in 70% of cases, to reappear early in the postpartum period [Nicholas, 1988]. When there is improvement, this usually occurs in the first trimester. Nevertheless, the disease commonly fluctuates and, at the very least, cycles of analgesics will be required. The disease almost always recurs early in the postpartum period, and this does not seem to depend either on breastfeeding or on the return of menstruation. Most patients need full doses of NSAIDs in the postpartum period. Children of mothers with Sjögren's syndrome with Ro antibodies have an increased risk of neonatal lupus. 4.8.2 The use of NSAIDs during pregnancy and breastfeeding should be avoided insofar as possible. Corticosteroids can be used under controlled conditions. DMARDs should be managed on an individual basis, and should preferably be continued during pregnancy. Teratogenic effects in the early weeks of pregnancy have been observed in animals receiving larger than pharmacological doses of NSAIDs. In both humans and animals, premature closure of the ductus arteriosus has also been observed in the last trimester. NSAIDs are not recommended near the time of delivery due to their inhibitor effects on platelets and the uterine musculature. All NSAIDs are transmitted, in greater or lesser measure, to the mother's milk. For these reasons, the NSAIDs should be avoided in the first and last trimester and during breastfeeding. If necessary, NSAIDs with a short half-life (ibuprofen or ketoprofen) should be used. During breastfeeding, NSAIDs should be taken while the baby is feeding to avoid elevated concentrations in the milk. There is no evidence that the corticosteroids produce serious adverse effects at average doses during pregnancy, except for promoting glucose intolerance, fluid retention, and hypertension. Consequently, they should be administered under controlled conditions. Table 20 shows the considerations to be taken into account with regard to DMARD use during pregnancy and breastfeeding. The decision to withdraw continuous treatment during pregnancy should be made on an individual basis. If the disease is aggressive, it is preferable not to withdraw the DMARD (unless it has been shown to affect the embryo, fetus, or infant) and to leave it at the minimum effective dose. Total withdrawal of the drug could provoke a recurrence of disease during pregnancy and a poorer outcome. Thus, for women of childbearing age, treatment involving the least risk for the fetus should be proposed to avoid drastic, last-moment decisions. Table 20. Use of DMARDs during pregnancy and breast-feeding Drug * Auranofin FDA classification* * C Transmitted through the placenta * Yes Effects on the mother * Not studied Effects on the fetus * Insufficient data Breastfeeding * Compatible** Drug * Aurothiomalate FDA classification* * C Transmitted through the placenta * Yes Effects on the mother * Not studied Effects on the fetus * Complex malformation of the central nervous system has been described in animals. Breastfeeding * Compatible** Drug * Azathioprine FDA classification* * D Transmitted through the placenta * Yes Effects on the mother * Contraindicated during pregnancy Effects on the fetus * Intrauterine growth retardation, premature birth. Transitory immunosuppression in the neonate. Potential alternation of the germ cells. Breastfeeding * Contraindicated due to potential immunosuppression Drug * Cyclophosphamide FDA classification* * D Transmitted through the placenta * Not studied Effects on the mother * Not studied Effects on the fetus * Severe malformations Breastfeeding * Contraindicated due to bone marrow depression Drug * Cyclosporin A FDA classification* * C Transmitted through the placenta * Yes Effects on the mother * Not studied Effects on the fetus * Intrauterine growth retardation, premature birth Breastfeeding * Contraindicated due to potential immunosuppression Drug * Antimalarials: Chloroquine, Hydroxychloroquine FDA classification* * C Transmitted through the placenta * Yes Effects on the mother * Rare Effects on the fetus * Rare Breastfeeding * Compatible, with precautions*** Drug * D-penicillamine FDA classification* * D Transmitted through the placenta * Yes Effects on the mother * Not studied Effects on the fetus * Connective tissue abnormalities, cutis laxa. Breastfeeding * Not studied Drug * Etanercept FDA classification* * B Transmitted through the placenta * Not studied Effects on the mother * Not studied Effects on the fetus * Unknown Breastfeeding * Not studied Drug * Infliximab FDA classification* * C Transmitted through the placenta * Not studied Effects on the mother * Not studied Effects on the fetus * Unknown Breastfeeding * Not studied Drug * Leflunomide FDA classification* * X Transmitted through the placenta * Yes Effects on the mother * Contraindicated in pregnancy Effects on the fetus * Unknown Breastfeeding * Not studied Drug * Methotrexate FDA classification* * X Transmitted through the placenta * Yes Effects on the mother * Spontaneous abortion Effects on the fetus * Fetal abnormalities (including cleft palate and hydrocephaly) Breastfeeding * Contraindicated. Potential accumulation in the infant's tissues Drug * Sulphasalazine FDA classification* * B and D near time of delivery Transmitted through the placenta * Yes Effects on the mother * Not studied Effects on the fetus * Kernicterus if administered near delivery Breastfeeding * Compatible, with precautions*** * FDA classification of drug teratogenicity: Category A: Appropriate and well controlled trials have not shown risk for the fetus in the first trimester of pregnancy, and there is no evidence of risk in subsequent trimesters. Category B: Indicates one of the following: a) No evidence of teratogenic effects in animal studies, but this has not been confirmed in humans. b) Some evidence of teratogenic effects in animal studies, but this has not been confirmed in humans. Category C: Indicates one of the following: a) Teratogenic effects have been detected in animal studies, but no studies in humans have been carried out. b) No studies have been carried out (either in animals or in humans). Category D: Studies have shown teratogenic effects on the human fetus, but at times the benefit obtained with the use of these drugs may exceed the expected risk (use in extreme situations of possible maternal death). Category X: Drugs that have unquestionably been shown to have clear teratogenic effects and whose risks greatly exceed the possible benefit obtained. ** 20 % of the dose administered is excreted in the milk. Rashes, hepatitis, and blood disorders have been described in infants. *** 7% of the dose is secreted in the milk. There is evidence of accumulation in the infant if renal excretion is reduced. **** Between 40% and 60% of the dose administered is secreted in the milk. Bloody diarrhea in the infant has been described. CHAPTER 5. CRITERIA FOR REPONSE TO TREATMENT 5.1 The objective of RA treatment is to induce complete disease remission or, alternatively, to achieve the best possible response. RA patients who have spontaneous or drug-induced remissions in the course of their disease have a better medium-term outcome than those who have persistent clinical activity [Eberhardt, 1998]. However, the rates of complete remission with DMARDs and/or corticosteroids are low (18-25%) [Wolfe, 1985; Prevoo, 1996; Harrison, 1996; Eberhardt, 1998] and are rarely prolonged. Consequently, criteria to evaluate the patient's clinical improvement are needed to aid the clinician in making treatment decisions. Complete disease remission, or at least attainment of the lowest possible level of inflammatory activity, is the only way to improve disease outcome and to assure the most favorable evolution for the patient. Two basic approaches to defining clinical remission in RA have been described: the ACR criteria and the EULAR criteria. 5.1.1 ACR criteria for clinical remission * Morning stiffness absent or not exceeding 15 minutes * No fatigue * No joint pain (by clinical history) * No joint tenderness * No soft tissue swelling in joints or tendon sheaths * Normal erythrocyte sedimentation rate The presence of five or more of these criteria for at least 2 months is sufficient to classify a patient as in complete remission, with a sensitivity of 72-80% and a specificity of 96-100% [Pinals, 1981; Wolfe, 1985]. The predictive values of these criteria may vary in different populations [Alarcón, 1987]. Their main disadvantages are the lack of guidelines on how to measure them, the fact that they are dichotomous (meaning that small changes in disease activity may change the classification), and that two of the criteria (fatigue and morning stiffness) are not included in the parameters recommended for the evaluation of RA patients [van Riel, 1992; Tugwell, 1993; Felson, 1993b; Boers, 1994; Wolfe, 1999b]. 5.1.2 EULAR criteria for clinical remission The EULAR criteria use the DAS as a continuous variable of disease activity [Van der Heijde, 1990]. A cut-off point below 1.6 on the DAS is consistent with the ACR definition of remission [Prevoo, 1996]. Since the measurement scale is continuous, the cut-off point recommended by the EULAR may vary depending on future investigations. 5.2 Patients with RA should be clinically monitored for an indefinite period of time. Patients in complete disease remission should be seen every 6 months or 1 year, and patients with recent disease onset, frequent flare-ups, or persistent activity should be seen "on demand" ( in general, every 1 or 2 months), depending on the treatment used and disease activity, until control is achieved. No treatment has been shown to cure RA, thus all patients who suffer this disease should be monitored clinically for an indefinite period. Patients in complete disease remission should be seen every 6 or 12 months. To avoid an overload of patients, they can be seen in primary care during the periods between rheumatologist appointments so as to assure clinical and laboratory monitoring and permit rapid referral to the specialist in case of disease reactivation and/or adverse effects. Patients with recent disease onset, frequent flare-ups, or persistent activity should be seen "on demand," depending on treatment used and disease activity, until the best possible control is achieved. These patients should be monitored every 1 or 2 months, at the same time as laboratory tests are done. 5.3 Follow-up of patients with RA should be based on longitudinal monitoring of the parameters described in the initial evaluation: joint pain and inflammation, global pain assessment by the patient, global disease assessment, functional disability, acute phase reactants, and radiologic damage. No studies have established the core set of parameters to be used in the routine clinical monitoring of patients with RA. The quantitative measurement of disease characteristics (e.g., the Ritchie index) offers several advantages: it provides information about disease severity that is valid, reliable, and of proven sensitivity; it can be used to classify the patient within the continuous spectrum of disease; and it can be used to guide the decisions that must be made by the physician, patient, and/or third parties. One way to improve the quality of care for patients is to apply the treatment response criteria designed for use in clinical trials to daily clinical practice. Thus, it is proposed that the same parameters assessed at the initial evaluation be used to monitor patients and evaluate their response to treatment: pain and joint inflammation, global pain assessed by the patient, global disease activity assessed by the patient and by the physician, functional disability, and acute phase reactants. The same instruments used in the initial evaluation should be used in follow-up. 5.4 The criteria for treatment response applied to individual patients should take into account: a) changes in disease activity and b) current level of activity. The clinician should evaluate the response to treatment, classifying it as satisfactory (complete remission of disease or sufficient even if not complete remission) or unsatisfactory (complete or almost complete lack of improvement). The evaluation can be made in accordance with any of the response criteria proposed in sections 5.4.1, 5.4.2, 5.4.3, and 5.4.4. Many approaches to defining clinical improvement in RA have been described, most of them focusing on the application to clinical trials. There is no published clinical experience in daily practice with any of the response indices developed for clinical trials. Although there is no scientific evidence to support a uniform recommendation, in the near future it is likely that these types of studies will be carried out, new indices will appear, or existing ones will be modified for application in daily practice. The treatment response criteria used throughout this guideline are based on two categories: satisfactory response, meaning complete remission of disease or a "sufficient" response, even though complete remission is not achieved, and unsatisfactory response, meaning complete or almost complete lack of improvement. The clinician can apply different response criteria to classify a patient in one of these categories. The following sections describe four approaches that have been explored in the literature on rheumatology: the ACR criteria for improvement [Felson, 1995], the EULAR definition of response [Boers, 1994], the simplified Scott index [Scott, 1993], and the modified Paulus criteria [Paulus, 1990]. 5.4.1 ACR response criteria [Felson, 1995] The ACR response criteria define a dichotomous result (response/no response) according to the following criteria: * At least 20% improvement in the painful joint count and in the swollen joint count; and * At least 20% improvement in at least three of the following parameters: ESR or CRP, physician's global assessment of disease activity, patient's global assessment of disease activity, patient's assessment of pain, and physical disability. These criteria are known as the ACR20, reflecting the need for a 20% improvement in each parameter, which is considered the clinically relevant cut-off point. Some authors have proposed that this requirement be raised to 50% (ACR50) or 70% (ACR70). The ACR improvement criteria use the core variables proposed by the ACR itself, which can be applied without major problems in daily practice [Felson, 1993b]. The fact that the criteria do not consider the current activity level, however, limits their application in daily clinical practice unless they are adapted to take this factor into account. The ACR improvement criteria are likely to be modified in the near future; in the meantime, it is proposed that they be adapted as follows [SER, 2000b]: * Satisfactory response: Meeting the ACR20 criteria, fewer than 6 swollen joints, and no impairment of any joint that, for the particular patient, produces intolerable loss of functional capacity in the opinion of the patient or physician. * Unsatisfactory response: Not meeting the criteria for satisfactory response. 5.4.2 EULAR response criteria The EULAR response criteria use the Disease Activity Scale (DAS), which combines different clinical information into a single index that can be used to classify patients in different categories. Although the proliferation of modified DAS scales [Van der Heijde, 1998] has caused some confusion, two basic formulas have been validated [Van Gestel, 1998] and can be applied to the EULAR improvement criteria: the original DAS [Van der Heijde, 1990; van der Heijde, 1992a], which uses the Ritchie articular index [Ritchie, 1968] and a count of the number of swollen joints out of 44 joints, and the DAS28, which uses a non-graduated count of 28 joints (Appendix 1). Unlike the ACR criteria, the EULAR definition takes into account both the patient's current status and the amount of improvement. It has been shown to have comparable validity to the ACR response criteria in clinical trials of RA patients [Van Gestel, 1999]. The definitions of satisfactory and unsatisfactory response as a function of the original DAS and the DAS28 are shown in Table 21. Table 21. EULAR definition of response EULAR DEFINITION OF RESPONSE (DAS) Current DAS * less than 2.4 DAS decrease * greater than 1.2 * Satisfactory * 0.6 - 1.2 * Unsatisfactory * less than 0.6 * Unsatisfactory Current DAS * 2.4 - 3.7 DAS decrease * greater than 1.2 * Unsatisfactory * 0.6 - 1.2 * Unsatisfactory * less than 0.6 * Unsatisfactory Current DAS * greater than 3.7 DAS decrease * greater than 1.2 * Unsatisfactory * 0.6 - 1.2 * Unsatisfactory * less than 0.6 * Unsatisfactory EULAR DEFINITION OF RESPONSE (DAS28) Current DAS28 * less than 3.2 DAS28 decrease * greater than 1.2 * Satisfactory * 0.6 - 1.2 * Unsatisfactory * less than 0.6 * Unsatisfactory Current DAS28 * 3.2 - 5.1 DAS28 decrease * greater than 1.2 * Unsatisfactory * 0.6 - 1.2 * Unsatisfactory * less than 0.6 * Unsatisfactory Current DAS28 * greater than 5.1 DAS28 decrease * greater than 1.2 * Unsatisfactory * 0.6 - 1.2 * Unsatisfactory * less than 0.6 * Unsatisfactory 5.4.3 Simplified Scott index [Scott, 1993] The simplified Scott index takes into account the change in disease activity and the current activity level, which makes it possible to apply it to individual patients. It is easy to apply, which facilitates its use during physician visits, but it requires further validation before it can be recommended for routine use. Its scoring system from 0 to 6 (a total of 7 categories) may be too wide for classification of treatment response. It is based on the following criteria: * Morning stiffness absent or lasting no more than 15 minutes * At least 50% reduction in pain * No more than 3 painful joints * No more than 3 swollen joints * ESR less than or equal to 30 * Functional improvement greater than or equal to 40%. For each category, the patient receives 1 point if the response criterion is met after 6 months of treatment; this gives a range of possibilities from 0 (no improvement) to 6 (complete improvement). 5.4.4 Modified Paulus criteria [Paulus, 1990] These criteria have been used in recent clinical trials [O´Dell, 1996]. They define a dichotomous result (response/no response) in accordance with the following criteria: * Morning stiffness absent or reduced by 50% * 50% decrease in the number of tender joints * 50% decrease in the number of swollen joints * ESR less than 30 mm/hr for women and less than 20 mm/hr for men. These criteria combine reduction in disease activity with the patient's current status. 5.4.5 Physician's subjective assessment of disease activity This is the most commonly used clinical criterion in daily practice. It is not recommended for use as the only criterion of response. If the physician's assessment of disease activity is the only criterion used to judge response to treatment, this assessment needs to be adapted to the treatment objectives (complete remission of disease or achieving the best possible response). It should be a combination of objective and quantifiable parameters evaluating disease activity, joint pain, and health status, with the final results classified into the categories of satisfactory or unsatisfactory response. CHAPTER 6. ADVERSE EFFECTS OF DRUGS USED IN THE TREATMENT OF RHEUMATOID ARTHRITIS 6.1 Antimalarials 6.1.1 The antimalarials are relatively safe drugs if they are used at the recommended doses. The most frequent side effects are gastrointestinal toxicity, skin rashes, and constitutional symptoms. Most of these side effects are reversible and do not require discontinuation of treatment [Maksymowych, 1987). 6.1.2 Monitoring: A baseline opthalmological examination should be made in patients over 40 years of age andor with a family history of ocular disease [ACR Committee, 1996]. All patients should receive an opthalmological examination including funduscopic and visual field evaluation every 6-12 months. Patients with kidney failure or those who take the drug for more than 10 years should be monitored more frequently. 6.1.3 Contraindications: Allergy to 4-aminoquinoline derivatives. Retinopathy or visual field deterioration. Hydroxychloroquine is generally better tolerated and less toxic than chloroquine [Finbloom, 1985]. Toxicity is more closely related with daily dose than with cumulative dose or duration of treatment [Rynes, 1983]. The most frequent contraindication is gastrointestinal toxicity with nausea, vomiting, pain, and bloated abdomen. In these cases, if the patient was taking chloroquine, the dosage can be reduced by half or the patient can be switched to hydroxychloroquine. Since its bioavailability is not reduced by taking it with food, it can be administered with meals, which improves tolerance. Skin toxicity rarely leads to withdrawal of the medication. Side effects include maculopapular, scaly, or morbilliform rashes, urticaria and pruritus; loss and graying of hair; lichenoid reaction; and exfoliative dermatitis. In cases of yellowish hyperpigmentation of the skin and mucosa, which generally appears after periods of prolonged treatment, especially in patients treated with quinacrine, it may be useful to reduce the dose by half. Ocular toxicity is very infrequent if the recommended doses are not exceeded. It can be detected early if periodic ophathalmological (funduscopic and visual field) examinations are performed. There are two types of ocular toxicity: * Deposits on the corneas or the ciliary bodies, accompanied by blurry vision, photophobia, visual halos, or difficulty in focusing, which generally disappear when treatment is discontinued. * Retinopathy, which may lead to lasting loss of vision and may progress despite withdrawal of treatment. Other rare side effects, such as neuromyopathy (less than 1%), usually with symptoms of myopathy predominating, resolve by discontinuing treatment. In patients with a deficiency of glucose 6-phosphase dehydrogenase (G6PD), antimalarials, and especially chloroquine, can lead to hemolytic anemia. Cases of aplastic anemia in patients treated with quinacrine have also been reported. 6.2 Anti-TNF agents 6.2.1 The safety profiles of the two commercial preparations (etanercept and infliximab) are not sufficiently well known. Their potential adverse effects, however, may be related with increased risk of infection, tumors, hematologic changes, and multiple sclerosis. 6.2.2 Monitoring: Rule out acute or chronic infection before initiating treatment. Rule out the presence of active or latent tuberculosis. Do not use live vaccines. Monitor the appearance of neoplasms. 6.2.3 Contraindications: Sepsis or clinically manifest infections and/or abscesses. Past medical history of hypersensitivity to infliximab or other murine proteins. Infections are infrequent [CCOHTA, 1999]. Although the use of anti-TNF agents exacerbates the outcome of sepsis [Fisher, 1996], there is no evidence to confirm a higher incidence of serious infections in patients treated with these drugs [Moreland, 1997; Maini, 1998; Maini, 1999; Moreland, 1999; Weinblatt, 1999a]. To date, 28 cases of tuberculosis infection have been reported, both miliary and extrapulmonary (9 in the United States and 19 in Europe), one of which had a fatal outcome. In most cases, the infection presents shortly after the second or third infusion. Given the severity of these complications, the Spanish Medicines Agency, in agreement with the European Agency for the Evaluation of Medicinal Products, recommends evaluation for active or latent (inactive) tuberculosis before beginning treatment with infliximab. The evaluation should include previous history of and/or contact with the disease, chest X-ray, and tuberculin test. Tuberculin tests in severely ill or immunodepressed patients may have a false negative result. If latent (inactive) tuberculosis is diagnosed, preventive measures should be taken to avoid activating the tuberculosis and to evaluate the benefit-risk relation before beginning treatment with infliximab. The patient should be instructed to inform the physician of the appearance of signs and/or symptoms that suggest tuberculosis, for example, persistent cough, weakness/weight loss, and low-grade fever. If active tuberculosis is suspected, infliximab treatment should be discontinued until the diagnosis is ruled out or the infection has been treated according to accepted guidelines. For both preparations (infliximab and etanercept), upper respiratory infection is the most frequently reported side effect; this is usually mild and does not require discontinuation of treatment. Since the potential immunosuppressive effect of anti-TNF agents may favor infection, vaccination with live vaccines while they are being used is not recommended. (Although this guideline focuses on RA in adults, children should be up-to-date on all their vaccinations before beginning treatment with etanercept [CCOHTA, 1999].) Some authors have mentioned the development of neoplasia as an adverse effect [Koopman, 1998; Camussi, 1998], although there is no evidence of a higher incidence of tumors in patients treated with anti-TNF agents. Pending the results of long-term studies on the use of anti-TNF agents and their association with tumors, it is essential to monitor the patient clinically for possible neoplasms. In pharmacological surveillance studies following the commercialization of etanercept, 10 cases of blood disorders have been reported in patients treated with this drug (of an estimated total of 5,500 patients, 3 aplastic anemias and 7 pancytopenias), with 5 deaths attributed to this cause (Pharmacological surveillance report from Wyeth). Nine cases treated with etanercept have presented with abnormalities, detected by magnetic resonance imaging, compatible with demyelinating disease of the central nervous system. Five of these patients were diagnosed with multiple sclerosis and one with possible multiple sclerosis. It is difficult at present to establish a causal relation between the drug and multiple sclerosis, although anti-TNF agents are known to exacerbate the clinical course of this disease [LMSSG, 1999]. Local reactions at the injection site (erythema, localized pain, edema) occurred in 49% of patients treated with 25 mg of etanercept [Moreland, 1999] and in 42% of those who received etanercept plus methotrexate [Weinblatt, 1999a]. These complications are generally self-limiting, lasting 3-5 days; they tend to appear in the first month of treatment and do not require its interruption. Five percent of patients treated with infliximab have had non-specific symptoms during intravenous administration such as fever, chills, chest pain, hypertension or hypotension, pruritus/urticaria, headache, sinusitis, rhinitis, and non-specific cardiorespiratory symptoms. In 16% of this 5% of patients, the infusion had to be discontinued [Remicade, 1999]. The appearance of antinuclear and anti-DNA antibodies has been described for both preparations more frequently than in control groups; in the case of etanercept, this was not associated with autoimmune disease. 6.3 Azathioprine 6.3.1 The most frequent side effects of azathioprine are gastrointestinal intolerance, blood disorders, and infections. 6.3.2 Monitoring: Baseline laboratory tests should be performed, including a complete blood count (leukocytes, hemoglobin, and platelets), creatinine, and liver function tests. Thereafter, a blood count should be made every 1-2 weeks while the dosage is being adjusted, and every 1-3 months upon reaching a stable dose [ACR Committee, 1996]. Liver function tests are recommended every 6-8 weeks. The dose should be reduced in patients with renal failure. Extreme precaution should be taken if it is used concurrently with allopurinol or with the angiotensin-converting enzyme inhibitors. 6.3.3 Contraindications: Known neoplastic disease. The most frequent gastrointestinal symptoms (20%) are anorexia, nausea, and vomiting. Less frequent are diarrhea (less than 1%) or elevated liver enzymes (5%). Although these side effects may make it necessary to discontinue the drug (10%), they usually improve or resolve by reducing the dose. Blood disorders are dose-dependent; the most frequent are leukopenia (25%) and thrombocytopenia (5%), although cases of pure red-cell aplasia have been described. Mild blood disorders may resolve by reducing the dose. A xanthine oxidase deficiency increases side effects in general and hematologic ones in particular [Black, 1998]. The use of allopurinol (a xanthine oxidase inhibitor) should be avoided; if it must be used, the AZA dose should be reduced (50-75%), and the leukocyte count should be monitored more frequently. Infections appear in about 10% of patients treated with AZA. Viral infections are the most frequent, especially herpes zoster. Bacterial infections usually develop in patients with neutropenia [Singh, 1989]. A hypersensitive reaction has been described in the first weeks of treatment, which includes fever, general malaise, arthralgias/myalgias, skin lesions, leukocytosis, elevated liver enzymes, and even hypotension and shock [Blanco, 1996]. The risk of neoplasia appears to be increased in RA patients (relative risk 2.2-8.7), mainly skin cancers and hematologic neoplasias [Silman, 1988]. 6.4 Cyclophosphamide 6.4.1 Cyclophosphamide has frequent adverse effects, which vary in relation with dose and route of administration [Decker, 1973;Williams, 1980]. Intravenous administration is recommended. Most side effects are reversible by discontinuing the drug. The most frequent side effects are gonadal, urologic, and bone marrow toxicity, neoplasms, and infections. Other frequent but less important effects are alopecia, nausea, and vomiting. 6.4.2 Monitoring: Complete blood count every 1-2 weeks during the first 2-3 months of treatment, then every 2-4 weeks once the dosage has been stabilized [Clements, 1986]. In patients with pulses of intravenous therapy, a blood count should be obtained before each infusion of cyclophosphamide. Monthly tests should be obtained for liver enzymes, urinalysis, and urinary sediment. If microscopic hematuria is detected, other, more specific studies are indicated, such as cystoscopy and urinary cytology. 6.4.3 Contraindications: Pregnancy, chronic or active infection, liver disease, or history of neoplasia. Renal failure is a relative contraindication that requires adjustment of the dosage. CPA is a useful drug in the treatment of serious RA complications. Administration by intravenous pulses is recommended, since they are as effective as oral administration and have less serious side effects. Gonadal toxicity from CPA is produced in women at the level of the primordial and antral follicles, giving rise to oligomenorrhea and amenorrhea [Warne, 1973]. In men it is produced in the epithelial germ layer of the seminal vesicles, causing azoospermia or oligospermia, and testicular atrophy or reduction in size [Fairley, 1972; Schilsky, 1980]. Libido and sexual function are normally not affected [Sherins, 1973; Schilsky, 1980]. Accepted risk factors for infertility are age over 25 years, concomitant radiation therapy, and prolonged treatment [Roubenoff, 1988]. Table 22. CPA dosage producing toxicity Gender * Men [Kovarsky,1983] Age (years) * CPA dosage producing toxicity * Greater than or equal to 18 g Gender * Women [Damewood, 1986] Age (years) * 20-29 CPA dosage producing toxicity * 20.4 g Gender * Women [Damewood, 1986] Age (years) * 30-39 CPA dosage producing toxicity * 9.3 g Gender * Women [Damewood, 1986] Age (years) * Greater than or equal to 40 CPA dosage producing toxicity * 5.2 g Intravenous treatment with CPA entails a risk of sustained amenorrhea (understood as lasting over 1 year), which is associated with patient age, dose administered, and duration of treatment [Schilsky, 1980; Damewood, 1986; Boumpas, 1993; Ramsey-Goldman, 1997]. Women who do not develop amenorrhea may experience early menopause [Fosdick, 1968; Miller, 1971; Roubenoff, 1988]. Oral CPA administration may provoke amenorrhea in up to 36% of cases, and is irreversible in 27% [Wang, 1995]. Table 23. Percentage of women treated with CPA who develop amenorrhea Age * Less than or equal to 25 years % of women with amenorrhea * 12 Age * 26-30 years % of women with amenorrhea * 27 Age * Greater than or equal to 31 years % of women with amenorrhea * 62 Duration of treatment * Short cycles (less than 7 bolus injections) % of women with amenorrhea * 12 Duration of treatment * Long cycles (greater than 15 bolus injections) % of women with amenorrhea * 39 Although recovery of spermatogenesis has been described after suspending treatment in young patients treated with CPA for short periods [Fairley, 1972; Trompeter, 1981], recovery of ovarian function or spermatogenesis is uncertain, and irreversible sterility may result [Fosdick, 1968; Miller, 1971; Fairley, 1972] Thus, freezing of ova or sperm is recommended before beginning treatment with CPA. The risk of infertility in women may be reduced by treatment with gonadotropin inhibitors [Langevitz, 1992; Ataya, 1993] or by using oral contraceptives [Chapman, 1981], and in men by the use of testosterone [Masala, 1997]. The urologic toxicity of CPA consists basically of the development of hemorrhagic cystitis [Hutter, 1969; Decker, 1973; Aptekar, 1973; Townes, 1976; Plotz, 1979; Fauci, 1983; Ansher, 1983; Hansen, 1983], vesicle fibrosis [Johnson, 1971], and squamous cell and transitional cell bladder cancer [Worth, 1971; Wall, 1975; Ansell, 1975; Plotz, 1979; Baker, 1987; Pedersen-Bjergaard, 1988; Thrasher; 1990]. Hemorrhagic cystitis is present in 15-30% of patients treated with CPA [Fauci, 1983], and its appearance, the same as with vesicle fibrosis, is directly related with oral administration. Patients treated intravenously do not develop hemorrhagic cystitis or neoplasms [Plotz, 1979; Austin, 1986; Balow, 1987; Klippel, 1987a]. There is a high risk of developing malignant vesicle neoplasm with total doses exceeding 80 g. Tumors may appear early or several years after initiation of treatment. The risk remains even years after discontinuing treatment [Baker, 1987; Hoffman, 1992; Radis, 1995]. The development of carcinoma of the bladder does not appear to be related to pre-existing hemorrhagic cystitis [Pedersen-Bjergaard, 1988]. Abundant oral (2-3 liters in 24 hours) or intravenous hydration and frequent urination are recommended to decrease vesicle toxicity [Ahmed, 1984; Balow, 1987; Klippel, 1987b]. The use of acetyl cysteine [Steinberg, 1981] or 2-mercaptoethane sodium sulfonate together with intravenous CPA also reduces vesicle toxicity [Bryant, 1980; Brock; 1982; Hows, 1984; Ehrlich, 1984; Nashel, 1985]. If the patient shows signs of reduced vesicular volume (e.g., polakiuria), CPA should be discontinued and cystometry performed. If there is hematuria suggesting the presence of incipient hemorrhagic cystitis or other urological complications, treatment should be discontinued and cystoscopy and urinary cytology should be performed. The inhibition of DNA replication produces bone marrow toxicity, which is cumulative. Its hematologic effects are: * Neutropenia. 32% of patients treated with high doses (150 mg) and 6% of those treated with low doses (75 mg). * Leukopenia [Weinstein, 1985]. Dose-dependent. Maximum suppression occurs 8-12 days after intravenous administration [Klippel, 1998]. Avoid leukocyte count dropping below 3,000/mm^3 and neutrophils below 1,000/mm^3 , adjusting the dosage until the desired levels are regained. Avoid concomitant treatment with allopurinol due to the increased risk of leukopenia [Clements, 1986]. * Anemia and thrombopenia are less frequent, and aplasia, if it occurs, is transitory. Several authors have studied tumor development in association with the use of CPA. Whereas some have found a high incidence of neoplasms [Baltus, 1983], others have not found significant increases [Kirsner, 1982]. There have been reports in the literature of leukemias and malignant lymphomas [Love, 1975; Grunwald, 1979; Penn, 1981; Wheeler, 1981; Baltus, 1983; Baker, 1987], sarcomas [Steinberg, 1981], and skin cancer [Baltus, 1983; Baker, 1987; Radis, 1995]. The factors that appear to be related with the development of carcinomas are total CPA dose [Greene, 1986; Baker, 1987] and duration of treatment [Pedersen-Bjergaard, 1985; Baker, 1987]. Upper respiratory tract infections are related with the use of CPA, as are bacterial, fungal, and viral infections [Bradley, 1989; Kattwinkel, 1991], especially with herpes zoster (incidence from 21 to 33%) [Moutsopoulos, 1978; Balow, 1987; Klippel, 1987a; Klippel, 1998]. Risk factors are considered to be the involvement of multiple organs, concomitant treatment with high-dose steroids, and leukocyte counts under 3,000 cells/mm^3 [Pryor, 1996]. Patients being treated with CPA and high-dose steroids should receive prophylaxis for Pneumocystis carinii with trimethoprim-sulphamethoxazol [Ognibene, 1995]. Other adverse effects that have been observed are gastrointestinal toxicity (mainly nausea and vomiting) [Belmonte, 1988; Fox, 1994; Klippel, 1998], pulmonary toxicity [Patel, 1976; Medrano, 1986; Klippel, 1998], cardiac toxicity [Appelbaum, 1976; Klippel, 1998], hepatic toxicity [Bacon, 1982; Goldberg, 1985; Fox, 1994 Klippel, 1998], ophthalmological toxicity [Jack, 1981], hypersensitive reactions [Klippel, 1988; Knisack, 1994], hypogammaglobulinemia [Fauci, 1989], alopecia [Kovarsky, 1983; Belmonte, 1988; Fauci, 1989], nail abnormalities [Klippel, 1998], and inadequate secretion of the antidiuretic hormone [De Fronzo, 1973; Steinberg, 1981; Bressler, 1985; Klippel, 1998]. These effects are dose-dependent, occurring more frequently at high drug doses. 6.5 Cyclosporin A 6.5.1 The most serious and relatively frequent adverse effects are nephrotoxicity and hypertension. Both are dose-dependent and constitute the most important limitation to their use. 6.5.2 Monitoring: Before beginning treatment, the following tests should be obtained: blood pressure (two measurements), complete blood count, liver and kidney biochemistry (with special attention to serum urea and creatinine), and urinalysis with sediment. Blood pressure, renal function, and K^+ and Mg^++ electrolytes should be monitored every 2 weeks during the first trimester and monthly thereafter. If the dose is changed or if there is an increase in creatinine levels or blood pressure, the patient should be monitored weekly until stabilization. If the levels of serum creatinine increase by more than 30% with respect to baseline, the dose should be reduced by 25-50%. If renal function does not improve in 1 month, cyclosporin should be discontinued; it may be resumed if creatinine returns to levels within 10% of the pre-treatment value. If hypertension is detected, treatment with angiotensin converting enzyme inhibitors or beta-blockers may be initiated. The drug of choice is nifedipine (which does not increase the levels of cyclosporinemia). 6.5.3 Contraindications: co-existing cancer (except non-melanoma skin cancer), uncontrolled hypertension, renal dysfunction, uncontrolled infections, and primary or secondary immunodeficiency [Cush, 1999]. The nephrotoxicity produced by CSA may provoke: * Reversible functional changes: tubular dysfunction (reduced reabsorption of Mg^+ , reduced secretion of K^+ , and reduced excretion of uric acid with resulting hypomagnesemia, hypercalcemia, and hyperuricemia) and vascular dysfunction (vasoconstriction, reduced renal perfusion and filtration with elevated serum urea and creatinine). * Structural changes: reversible proximal tubular disease and vessel disease (affecting the afferent arterioles that condition glomerulosclerosis; ischemia, tubular atrophy, and interstitial fibrosis. Reversible changes are frequent, while persistent structural changes are rare and usually present with elevated serum levels of CSA and associated risk factors such as concurrent treatment with nephrotoxic drugs, pre-existing nephropathy, and hypertension [Grant, 1987; Land, 1988; Morris, 1988; Mihatsch, 1989; Landewé, 1996; Rodríguez, 1996]. At the usual doses (2.5 to 5 mg/kg/day), renal dysfunction is not severe and is rapidly reversible [Dougados, 1989; Tugwell, 1990; Boers, 1990; Van Rijthoven, 1991; Landewé, 1994]. High doses (from 10 mg/kg/day at initiation of treatment) produce more severe and less reversible changes in renal function (Boers, 1988; Berg, 1989]. To prevent structural nephropathy the following recommendations are made: * Exclusion of patients with potential risk factors such as renal dysfunction * Limitation of the maximum dose to 5 mg/kg/day * Use of the smallest possible maintenance dose, depending on the level of serum creatinine * Frequent and careful monitoring of renal function * Routine clinical examination and laboratory tests [Panayi, 1993; Panayi, 1994; Tugwell, 1995a]. The frequency of hypertension is 10-20% [Cohen, 1992]. The patient should have normal blood pressure before beginning therapy. If diastolic BP is higher than 95 mmHg or systolic BP higher than 160, the dose should not be increased. If hypertension is present (diastolic BP greater than 105 mmHg or sustained at more than 95 mmHg) at two consecutive measurements, antihypertensive treatment should be initiated or the CSA dose reduced [Panayi, 1993; Panayi, 1994]. Other non-renal adverse effects related with the use of CSA are gastrointestinal effects (dyspepsia, nausea, vomiting, abdominal pain, and diarrhea), hypertrichosis, gingival hypertrophy, and paresthesias. These are usually dose-dependent and are reversible by reducing the dose of the drug. Less frequent manifestations are fatigue, leukopenia, sinusitis, arthralgias/myalgias, flushing, ocular pain, gynecological symptoms (menorrhagia, breast hypertrophy or pain), hyperlipidemia, hepatic abnormalities (hyperbilirubinemia and hypertransaminemia), and neurological effects (headache, tremor). Other rare adverse reactions, present in 2% or less of patients are: conjunctivitis, fever, allergic reactions (exanthema, angioedema with reduced C1-esterase inhibitor), anemia, thrombocytopenia, anorexia, confusion, convulsions, weak and broken nails, and peptic ulcer [Klintmalm, 1981; Maddux 1986; Grant, 1987; Tindall, 1987; Lin, 1989; Cairns, 1992; Arellano, 1993; Fauci, 1993; Forre, 1994; Furst, 1994b; Tugwell, 1995b; Klippel, 1998; Rodríguez, 1998]. 6.6 D-penicillamine 6.6.1 The most frequent side effects of D-penicillamine are skin lesions, gastrointestinal symptoms, and renal involvement. 6.6.2 Monitoring: Baseline tests should be performed, including a complete blood count, creatinine, and urinalysis (including sediment). These tests should be repeated every 2 weeks until a stable dose is reached and every 1-3 months thereafter (ACR Committee, 1996]. 6.6.3 Contraindications: Kidney disease, blood disorders (leukopenia, thrombocytopenia). All types of skin lesions may appear (25-50%), from morbilliform and pruritic rashes to pemphigus-like lesions [Willemsen, 1990]. These generally disappear when medication is withdrawn [Munro, 1997]. Muscosal lesions, especially mouth ulcers, are less frequent. About 30% of patients have gastrointestinal symptoms (nausea, anorexia, abdominal pain, and diarrhea) during the first months of treatment. These symptoms usually disappear even though DP is continued, although it is sometimes necessary to suspend it [Munro, 1997]. About one fourth of patients report dysgeusia (altered sense of taste) during the first months of treatment. This symptom usually disappears spontaneously despite continued treatment, or it may improve following the administration of zinc [Jaffe, 1977]. Some 30% of RA patients treated with DP have some type of renal involvement. This most frequently takes the form of proteinuria accompanied by microscopic hematuria [Stein, 1980]. About 7% of patients develop a nephrotic syndrome secondary to membranous glomerulonephritis which disappears completely in a variable period of time after discontinuing treatment [Hall, 1988a]. Much less frequent is the development of acute renal failure secondary to a rapidly progressive "half-moon" glomerulonephritis [Ntoso, 1986]. Other side effects are blood disorders (thrombocytopenia 8-10% and leukopenia), pulmonary toxicity (bronchiolitis obliterans less than 1%), breast hyperplasia [Taylor, 1981], development of autoimmune processes, systemic lupus erythematosus [Chalmers, 1982], inflammatory myopathies [Lund, 1983], myasthenia gravis [Andonopoulos, 1994], and Goodpasture syndrome [Munro, 1997]. 6.7 Leflunomide 6.7.1 The most frequent adverse effects in published clinical trials are gastrointestinal and respiratory. These effects are generally mild, are not dose-dependent, and do not require discontinuation of treatment. 6.7.2 Monitoring: Liver enzymes should be monitored, initially every 4 weeks until a stable treatment dose is reached. If they are elevated to over twice the maximum reference value, the dose should be reduced to 10 mg/day. If a reduction to 1.2 times the maximum reference value is not obtained, leflunomide should be discontinued and cholysteramine or charcoal administered. If the transaminases remain elevated, a liver biopsy should be performed [Weinblatt, 1999b; Arava, 1999]. Periodic monitoring for possible anemia and leukopenia is recommended. 6.7.3 Contraindications: Serious immunodeficiency, dysplasias, uncontrolled infection (due to the theoretical possibility of immunosuppression), moderate or severe renal failure (there is no experience in this group of patients), liver function disorder, significant bone marrow disorder, severe hypoproteinemia. The most frequent adverse effects in published clinical trials are gastrointestinal (diarrhea 17%, nausea 9%, and abdominal pain 6%) and respiratory (upper tract infections 15% and bronchitis 7%). These effects are generally mild, are not dose-dependent, and do not require that treatment be discontinued. [Mladenovic, 1995; Weinblatt, 1999b; Arava, 1999; Smolen, 1999; Strand, 1999;]. Elevated transaminases have been described in about 6% of RA patients treated with 25 mg/day, and are reversible on discontinuing treatment. Generally these elevations do not exceed twice the normal maximum value and they tend to remit over time [Mladenovic, 1995; Smolen, 1999; Weinblatt, 1999b; Strand, 1999]. Cases of severe liver disease have been reported, some resulting in death, most of which occurred during the first 6 months of treatment. Other less frequent effects are hypertension (10%), headache (7%), vertigo (4%), weight loss (4%), and reversible alopecia (1% with a dose of 10 mg/day and 7% with 25 mg/day) [Mladenovic, 1995; Furst, 1995; Strand, 1999]. One case of anaphylaxis has been reported. In experimental models, treatment with leflunomide has been associated with anemia and leukopenia [Yuh, 1995]. This toxic effect has not been produced in clinical trials in humans, but until long-term pharmacological surveillance data are available, the patient should be monitored periodically for the possible appearance of anemia and leukopenia. In animal models, leflunomide has severe teratogenic effects and increases the risk of fetal death [Arava, 1999]. As its safety in humans is unknown, contraceptive measures are recommended before beginning treatment, not only in women of childbearing age but also in men, due to the possibility of teratogenic effects caused by the paternal route. If pregnancy occurs or if a man wishes to have children, it is recommended that the drug be immediately discontinued, and the patient should be treated with 8 g of cholestyramine, three times a day, for 11 days. Because it is potentially immunosuppressive, it is assumed to promote or exacerbate infection, but so far no serious infections during clinical use have been reported. Due to its potential immunosuppressive effect, and in the absence of safety and efficacy studies on the concomitant use of leflunomide and live vaccines, vaccination is not recommended during treatment with this drug. 6.8 Methotrexate 6.8.1 The most important side effects of MTX are pulmonary, hepatic, and hematologic toxicity. Other adverse effects are carcinogenesis, infections, nodules, gastrointestinal toxicity, and neurological toxicity. 6.8.2 Monitoring: Before beginning treatment a complete blood count, liver and kidney biochemistry, and serum albumin should be obtained, together with hepatitis A, B, and C serology. If pre-existing liver disease or exposure to liver toxins is suspected, a liver biopsy should be performed before beginning treatment. If the patient has a history of pleuropulmonary disease, a chest X-ray, as well as folic acid and vitamin B12 levels should be obtained. Follow-up tests should be obtained every 4 to 8 weeks to monitor blood count and liver and kidney biochemistry. A follow-up liver biopsy should be performed if there are persistent abnormalities in liver biochemistry that can not be attributed to concomitant treatment with nonsteroidal anti-inflammatories. Other non-routine studies are indicated if symptoms suggestive of specific complications appear (e.g., blood gas analysis and chest X-ray if pneumonia is suspected). 6.8.3 Contraindications: Pregnancy, alcohol abuse, hepatitis B or C, and cirrhosis of any origin are considered to be absolute contraindications. Relative contraindications are renal failure, chronic pulmonary disease, and active infection not associated with Felty's syndrome. Six MTX-induced clinical syndromes of pulmonary toxicity are known: acute interstitial pneumonitis, interstitial fibrosis, pulmonary nodules, non-cardiogenic pulmonary edema, pleuritis, and pleural effusion [Cannon, 1997]. The first three have been described in RA patients and the latter three in patients with neoplasms. The most frequent type of pulmonary toxicity (from 2 to 6%) is acute interstitial pneumonitis or hypersensitivity pneumonitis [Furst, 1994b]. This is a potentially lethal complication [Furst, 1990] by itself or as a result of pulmonary superinfection [Searles, 1987]. It is not associated with either duration of treatment, weekly dose, or cumulative dose received [White, 1989; Hargreaves, 1992; Alarcón, 1995; Kremer, 1995; Cannon, 1997]. Pre-existing pulmonary disease is a risk factor [Golden, 1995]. Treatment consists basically of withdrawing MTX, administering corticosteroids, and managing the respiratory insufficiency. The hepatic toxicity of MTX manifests as fibrosis and cirrhosis, especially in patients who consume excessive amounts of alcohol (more than 100 g/week) or have pre-existing liver disease [Farell, 1998]. Nonetheless, although transaminases are frequently elevated [Songsiridej, 1990], fibrosis rarely progresses to cirrhosis [Furst, 1990], even after cumulative MTX doses exceeding 5 g [West, 1997]. The main risk factors are diabetes mellitus [Erickson, 1995; Bass, 1996; Farrell, 1998], alcohol abuse, obesity, fatty liver, underlying liver disease (chronic liver disease due to hepatitis B, C, or other viruses), age over 60 years, renal failure, cumulative doses of MTX exceeding 1.5-2 g, concomitant treatment with NSAIDs, and related systemic disease [Walker, 1993; O´Dell, 1997; Farrell, 1998]. It has been suggested that patients with a deficiency of alpha-1 antitrypsin are more susceptible to MTX hepatotoxicity [Hilsden, 1995; O´Dell, 1997]. Hepatotoxicity is reduced by using low doses and following recommended guidelines for weekly administration of the drug [Sznol, 1987]. Pre-treatment biopsy is indicated only in patients with a history of excessive alcohol consumption, persistent transaminase elevation, or pre-existing liver disease [Kremer, 1992; Kremer, 1994]. If liver biopsy shows acute fibrosis or cirrhosis (Roegnick stages class III-b or IV), the treatment is contraindicated [Kremer, 1994]. It should also be discontinued in patients who refuse biopsy if they have persistent liver test abnormalities (5 of 9 GOT measurements above the normal range in a 12-month period, or 6 of 12 if tested monthly) or hypoalbuminemia [Kremer, 1994]. Hemotologic toxicity (5-25%), consists of leukopenia, thrombocytopenia, megaloblastic anemia, and pancytopenia (1-2%) [Gutiérrez Ureña, 1996]. It is usually mild or moderate and remits when the dosage is reduced. Pancytopenia responds to folic acid and supportive treatment (steroids, transfusions, antibiotics, and hematopoietic stimulating factors) [McKendrey, 1997]. Probable risk factors are considered to be folate deficiency and macrocytosis [Al-Awadhi, 1993], concomitant treatment with other antifolate drugs such as sulphasalazine [Morgan, 1993] or trimethoprim-sulphanotoxazol, concurrent viral infections [Naides, 1995], advanced age, and renal failure [Al-Awadhi, 1993, RACTAG, 1995]. Increased risk of cancer has not been shown in numerous studies of patients with diseases other than RA who were treated with MTX. Tumors of the liver [Ruymann, 1977] and lymphomas [Ellman, 1991; Shiroky, 1991; Kingsmore, 1992; Kamel, 1993; Bachman, 1996], however, have been observed in RA patients. Studies of lung cancer incidence in RA patients treated with MTX have yielded contradictory results, with increases shown in some cases [McKendry, 1993] and normal incidence in others [Alarcón, 1994]. It does not appear to increase the risk of hematologic malignancies [Moder, 1995]. MTX treatment may increase the risk of bacterial, viral, herpes zoster, or opportunistic infections in patients with RA. The risk is higher with combined steroid treatment [Kanik, 1997]. Some 60% of patients treated with MTX have gastrointestinal toxicity (stomatitis, nausea, vomiting, dyspepsia, abdominal pain, indigestion, diarrhea, anorexia, or weight loss) [McKendry, 1997]. These effects are generally reversible by reducing the drug dosage or changing from the oral to the parenteral route of administration [O´Dell, 1997]. They can be prevented and treated with folic acid supplements. Exceptionally, hemorrhagic enteritis may occur. Neurotoxicity occurs at high doses (more than 1 g/m^2 ) in 15% of patients. It may manifest as depression, confusion, memory loss, drowsiness, headache, fatigue, or discomfort; it most frequently occurs within 24 hours of drug administration [McKendry, 1997]. Exceptionally, hallucinations or leukoencephalopathy may appear [Worthley, 1995]. Nodules are histologically indistinguishable from rheumatoid nodules; they regress when treatment is discontinued and reappear when it is renewed [Kerstens, 1992]. They may be associated with vasculitis [Segal, 1988], pleural effusion, and pericardial tamponade [Abu-Shakra, 1994]. Other toxic manifestations are rashes [Neiman, 1985; Bannwarth, 1994], alopecia, osteopathy [McKendry, 1997], arthralgias/myalgias, general malaise on the first or second day of treatment, or even fever unrelated with infection. These "post-dose" effects are the second most common cause of MTX withdrawal [Halla, 1994]. 6.9 Gold salts 6.9.1 The most important clinical side effects are hematologic and renal toxicity. Both are more frequent with intramuscular treatment. They require careful clinical monitoring and the immediate discontinuation of treatment to prevent irreversible sequelae. 6.9.2 Monitoring: Periodic testing for proteinuria by routine urinalysis (qualitative or semiquantitative) every 4 weeks during the first 6 months and every 3 months thereafter. If proteinuria is detected, a 24-hour urine quantification should be obtained. If the proteinuria exceeds 500 mg/24 h, treatment should be discontinued until it disappears or drops below 200 mg/24 h, after which it may be renewed. If the proteinuria is severe (above 1 g/24 h), treatment should be discontinued permanently. 6.9.3 Contraindications: Severe liver or kidney disease and blood or bone marrow disorders. The most frequent hematologic effects are related with the hematopoietic system: eosinophilia and thrombocytopenia (1-3%), followed by neutropenia [Lockie, 1985; Ridaura, 1995; Myochrysine, 1997]. Aplastic anemia and pure red-cell aplasia have also been reported. Of all these effects, the only one that does not require discontinuation of treatment is eosinophilia, which some authors consider a warning sign for the presentation of other toxic effects [Davis, 1974; Bretza, 1983]. Thrombocytopenia may appear suddenly or progressively; treatment should be discontinued if platelet counts drop below 100,000 platelets/mm^3 [Ridaura, 1995]. The most frequent renal toxicity is membranous glomerulonephritis, usually preceded by proteinuria and/or hematuria, followed in frequency by nephrotic syndrome and acute renal failure, which may be secondary to acute tubular necrosis [Robbins, 1980; Hall, 1988b]. Other adverse effects are skin toxicity (dermatitis, pruritus) [Ridaura, 1995]; ulcers of the mouth, gums, or tongue, including the upper palate and pharynx; abnormal taste sensations (dysgeusia, metallic taste); hepatic toxicity (jaundice with or without intrahepatic cholestasis); pulmonary toxicity (hypersensitivity pneumonitis, bronchiolitis obliterans); gastrointestinal toxicity (diarrhea, especially with auranofin - 47%, toxic enterocolitis); neurologic toxicity (peripheral neuropathy, cranial neuropathy, Guillan-Barré syndrome, encephalopathy); vaso-vagal hypotension; conjunctivitis (auranofin); and corneal ulcers and gold deposits (sodium aurothiomalate). 6.10 Sulphasalazine 6.10.1 The most frequent sites of adverse reactions (33%) to sulphasalazine are the central nervous system and gastrointestinal tract. These are usually mild and do not require discontinuation of treatment. Other less frequent adverse effects are hematologic and hepatic toxicity. 6.10.2 Monitoring: Blood cell count every 4 weeks during the first 3 months and every 3 months thereafter. 6.10.3 Contraindications: Allergy to salicylates or sulfonamides. The adverse effects on the central nervous system (headache, vertigo) and gastrointestinal tract (anorexia, nausea, vomiting, abdominal pain) are generally mild and do not require discontinuation of treatment [Amos, 1986; Farr, 1986; Williams, 1988]. Hematologic toxicity occurs particularly in the hematopoietic system: macrocytosis (9%), leukopenia (3.7%), neutropenia (2%) and megaloblastic anemia (less than 1%). Isolated episodes of aplastic anemia, agranulocytosis, thrombocytopenia, and leukocytosis have been reported. Hematologic toxicity may present at any time during treatment, although it usually appears early (between the 5th and 12th week), except for macrocytosis and megaloblastic anemia, which may present after prolonged periods of treatment [Drugex, 1999]. These effects are reversible if the drug is discontinued and treatment is initiated [Guillemin, 1989; Canvin, 1993]. Folic acid treatment (5-10 mg/day) should be used in the case of megaloblastic anemia. A deficiency of glucose 6-phosphase dehydrogenase (G6PD) may produce hemolytic anemia [ACR Committee, 1996]. Hepatic toxicity manifests as acute, febrile episodes, with pruritic skin lesions, lymphadenopathy, hepatomegaly, lymphocytosis, eosinophilia, and elevated transaminases [Williams, 1979; Losek, 1981; Boyer, 1989; Marinos, 1992; Vyse, 1992]. This situation is serious and can lead to death [Pears, 1989; Marinos, 1992]. Discontinuing medication is not sufficient to prevent the patient's deterioration, and corticosteroids are needed. Continued treatment with sulphasalazine has been associated with male infertility (spermiogram abnormalities in 86% and oligospermia in 72%) [Birnie, 1981]. These abnormalities are reversible after discontinuing treatment for 3 months [Toovey, 1981]. There have been isolated reports of cases of altered taste (ageusia and metallic taste), skin abnormalities (drug-induced exanthema, pruritic maculopapular rashes, Stevens-Johnson syndrome, toxic epidermic necrolysis), pulmonary disorders (eosinophilic pneumonia, fibrosing alveolitis, subacute hypersensitivity pneumonitis), neurologic disorders (motor and sensory neuropathy, aseptic meningitis), muscular disorders (myopathy), and renal disorders (hemolytic-uremic syndrome, nephrotic syndrome, bilateral kidney stones). Table 24. Adverse effects of DMARDs Drug * Antimalarials Toxicity * Gastrointestinal, retina, skin Initial evaluation * Funduscopic examination in persons over 40 years of age, visual field. Clinical effects * Visual disturbances, skin lesions. Tests * Funduscopic examination every 6-12 months. Most common reasons for suspending treatment * Corneal deposits, retinopathy. Drug * Anti-TNF agents Toxicity * Hematologic, central nervous system, immune system (toxicity assumed). Initial evaluation * Rule out acute or chronic infection (particularly TB). Rule out multiple sclerosis. Clinical effects * Local or anaphylactic reactions. Signs or symptoms of infection or neoplasm, myelosuppression or demyelinating disease. Tests * CBC on administration of infliximab, and every 4-6 weeks with etanercept treatment. Most common reasons for suspending treatment * Systemic infection. Drug * Azathioprine Toxicity * Hematologic, gastrointestinal. Initial evaluation * Baseline CBC, creatinine, liver function. Clinical effects * Signs or symptoms of infection or neoplasm. Tests * CBC every 1-2 weeks until dosage stable, then every 1-3 months. Liver function tests every 6-8 weeks. Most common reasons for suspending treatment * Leukopenia (less than 3,000 leukocytes and/or less than 1,000 neutrophils). Thrombocytopenia (less than 150,000 /mm^3 ). Systemic infection. Drug * Cyclophosphamide Toxicity * Gonadal, urological, bone marrow. Initial evaluation * Rule out pregnancy, infections, neoplasms, or re-existing liver disease. Clinical effects * Signs or symptoms of infection or neoplasm, alopecia, nausea, vomiting. Tests * CBC every 1-2 weeks until dosage stable, then every 2-4 weeks. Liver enzymes, urinalysis, urinary sediment, monthly. Most common reasons for suspending treatment * Leukopenia (less than 3,000 leukocytes and/or less than 1,000 neutrophils). Thrombocytopenia (less than 150,000 /mm^3 ). Systemic infection, hemorrhagic cystitis. Drug * Cyclosporin A Toxicity * Kidney, hypertension. Initial evaluation * Rule out neoplasms or premalignant lesions. Two BP tests for hypertension. CBC, hepato-renal biochemistry, routine urinalysis with sediment. Clinical effects * Signs or symptoms of infection or neoplasm. Tests * Monitor for hypertension, renal and electrolyte function every 2 weeks the first 3 months, and then monthly. Most common reasons for suspending treatment * Greater than or equal to 30% increase in creatinine not reversed after halving dose. Development of uncontrolled hypertension. Drug * D-penicillamine Toxicity * Skin, gastrointestinal, kidney. Initial evaluation * CBC, creatinine, urinalysis. Clinical effects * CBC, creatinine, urinalysis every 2 weeks until dosage stable, and then every 1-3 months. Tests * Skin lesions, anaphylactic reactions. Most common reasons for suspending treatment * Proteinuria greater than 500 g/24 h. Leukopenia (less than 3,000 leukocytes and/or less than 1,000 neutrophils). Thrombocytopenia (less than 150,000 /mm^3 ). Drug * Leflunomide Toxicity * Pulmonary, gastrointestinal, hepatic. Initial evaluation * Begin contraceptive use. Rule out active infection, liver disease, and renal failure. Clinical effects * Skin lesions, hair loss, weight loss, hypertension. Tests * CBC and GOT, GPT every 4 weeks until dosage stable. Most common reasons for suspending treatment * Liver enzymes persistently elevated to more than twice the reference values. Drug * Methotrexate Toxicity * Pulmonary, hepatic, hematologic, gastrointestinal, and neurological. Initial evaluation * CBC, liver and kidney biochemistry, serum albumin, hepatitis A, B, C serology. If there is history of pleuropulmonary disease: chest X-ray, folic acid and vitamin B12 levels. If excessive alcohol consumption or previous liver disease, liver biopsy. Clinical effects * Dry cough, shortness of breath, fever, hemorrhage, nausea, vomiting, diarrhea, nodules, adenopathy. Tests * CBC and kidney and liver biochemistry every 4-8 weeks. If liver function abnormalities persist: follow-up liver biopsy. Most common reasons for suspending treatment * Pregnancy, pulmonary toxicity, liver enzymes persistently elevated to more than twice the reference values. Leukopenia (less than 3,000 leukocytes and/or less than 1,000 neutrophils). Thrombocytopenia (less than 150,000 mm^3 ). Drug * Gold salts Toxicity * Hematologic and renal. Initial evaluation * Baseline Clinical effects * Skin lesions, mouth ulcers, edema, diarrhea, signs or symptoms of myelosuppression. Tests * CBC and proteinuria every 4 weeks for the first 6 months, and then every 3 months. Most common reasons for suspending treatment * Proteinuria greater than 500mg/24 h. Leukopenia (less than 3,000 leukocytes and/or less than 1,000 neutrophils). Thrombocytopenia (less than 150,000 /mm^3 ). Drug * Sulphasalazine Toxicity * Central nervous system, gastrointestinal, hematologic. Initial evaluation * Hypersensitivy to sulfonamides. History compatible with G6PD deficiency. Clinical effects * Skin lesions, signs or symptoms of myelosuppression. Tests * CBC every 4 weeks for 3 months, and then every 3 months. Most common reasons for suspending treatment * Hypersensitivity syndrome. Leukopenia (less than 3,000 leukocytes and/or less than 1,000 neutrophils). Thrombocytopenia (less than 150,000 /mm^3 ). * Because there has been little experience with its use, it is not possible to recommend a specific period of time for monitoring adverse effects once a stable dose has been reached (3 months would seem to be prudent). CHAPTER 7. OTHER TREATMENTS 7.1 Surgical treatment 7.1.1 The rheumatologist should consider surgical treatment when articular function does not improve or is notably worse, when incapacitating pain persists, or when there are potentially serious or limiting neurological complications [Dreyer, 1999; Grob, 1999]. Appropriate medical treatment will reduce the indications for surgery and will improve the likelihood of surgical success. Consultation with an orthopedic surgeon should not always be an indication for surgery, but the exchange of opinions and clinical evaluation will help improve the patient's clinical and functional status. A number of factors should be considered before surgical treatment: bone quality, the patient's level of motivation and preferences, the probability that surgery will modify the course of the disease, and an estimate of the extent to which surgical treatment can reconstruct articular function and make the patient more independent [Bogoch, 1999]. In deciding on surgical intervention, clinical and functional evaluation should predominate over simple radiographic change in the disease. RA patients who consult the orthopedic surgeon often have various joints needing surgical evaluation, therefore priorities need to be established. The joint that the patient finds most incapacitating will usually be treated first. Patients who cannot walk due to lower limb pain or deformity need to have functional upper limbs to facilitate the post-surgical period. When the upper limbs are affected (pain, deformity, or stiffness) to the point that they would impede the use of walking aids, the upper limbs should be reconstructed first. If the joints are affected with different degrees of severity, the one with the best prognosis should be reconstructed first. 7.1.2 The joint prosthesis is the most efficient surgical means to arrest progressive loss of functional capacity. Prosthetic implant in any joint should be performed before irreparable deformities occur (e.g., flexus or axial deviations and instabilities) because these will limit the success of arthroplasty [Waldman, 1998; Creighton, 1998; Hargreaves, 1999]. Synovectomy may produce slight improvement in the synovectomized joints, but this effect is not maintained at 3 years. Arthrodesis is a good control measure but is more limited from the functional point of view. It remains a commonly used technique in RA to palliate damage from joint destruction, especially in the interphalangeal joints of the hand, the metacarpophalangeal joint of the thumb, the wrist, ankle, and hindfoot [Bogoch, 1999]. Arthrodesis of other joints is less acceptable. Surgical success or complications in RA are associated with the surgeon's experience, the patient's previous status, and post-operative care, especially rehabilitation and occupational therapy. The latter two factors are an important aid in establishing optimal joint function, especially after arthroplasy of the knee or shoulder and hand surgery. The incidence of infection in orthopedic surgery may increase during the perioperative period, although this has not been conclusively established. A reasonable course of action is to omit the weekly dose of MTX in the week before and after surgery, which reduces the small possibility of perisurgical complications, at the expense of the also small risk of reactivating the disease [Bridges, 1991; Carpenter, 1996]. 7.2 Rehabilitative therapy 7.2.1 The objective of a rehabilitation program in RA patients is to improve pain relief, joint mobility, and the performance of the activities of daily living. This is intended to prevent disability and maintain maximum personal independence [Sutej, 1991]. Rehabilitative techniques that may be used in treating RA patients are thermotherapy, physical exercise, prescription of splints, and occupational therapy. Although rehabilitation is prescribed relatively frequently in RA patients, few studies with sound methodology have demonstrated its benefits with respect to control groups. Several studies show that patients who undergo a rehabilitation program have a 25 to 40% improvement in function. Other benefits that have also been attributed to rehabilitation are improvement in "disease perception" as well as in indicators of disease "activity" [Conaty, 1971; Spiegel, 1986]. Rehabilitation techniques that may be used in treating RA patients are thermotherapy, physical exercise, prescription of splints, and occupational therapy. 7.2.2 Thermotherapy (the use of heat or cold as an analgesic method in joints with symptoms of inflammation) is useful in the treatment of RA. Superficial heat is indicated in RA, whereas deep heat is formally contraindicated. The local application of cold increases the pain threshold in the inflamed joints. Heat can be applied superficially (hot compresses, paraffin, infrared rays) or deeply (microwaves, ultrasound). Deep heat raises the temperature of the inflamed joints, while superficial heat, paradoxically, decreases it and thus reduces pain. Consequently, superficial heat is indicated in RA, while deep heat is formally contraindicated. Superficial heat increases the blood flow to the inflamed joints, producing a cooling effect, whereas deep heat does not increase blood flow [Lehmann, 1966; Harris, 1974; Feibel, 1976]. Cold should only be applied superficially in the form of cold compresses, ice massage, or cold spray. The local application of cold increases the pain threshold in inflamed joints [Benson, 1974]. The application of cold is contraindicated in the case of Raynaud's phenomenon, hypersensitivity to cold, cryoglobulinemia, or paroxysmal nocturnal hemoglobinuria [Olson, 1972]. 7.2.3 The ideal type of physical exercise in patients with RA is static or isometric active exercise, which involves minimal increase in intra-articular pressure or bone destruction. Resistance active exercises (lifting weights) are formally contraindicated because they increase joint inflammation, pain, muscular fatigue, temperature, and intra-articular pressure. In passive exercise there is no voluntary muscular contraction on the part of the patient. The physiotherapist moves the joints. Its main objective is to maintain articular balance in joints at risk of developing ankylosis or contractures. Its efficacy in RA patients, however, is uncertain. If used, it should never cause pain [Merritt, 1983]. In active exercise there is voluntary muscular contraction by the patient. There are three types: * Static or isometric active exercise: limited muscular contraction without joint motion but with maximal muscular tension. * Dynamic or isotonic active exercise: muscular contraction associated with joint motion that may act with or against the force of gravity. * Resistance active exercise: exercise performed against the force of gravity and with added weight. The ideal type of physical exercise for RA patients is static or isometric active exercise, which involves a minimal increase in intra-articular pressure or bone destruction [Castillo, 1965; Jaison, 1970]. Isometric exercise of the quadriceps has been shown to be beneficial for the knee joint [Ekblom, 1975a]. Resistance active exercises (lifting of weights) are formally contraindicated in RA patients because they increase joint inflammation, pain, muscular fatigue, temperature, and intra-articular pressure. Patients without active disease who have developed sufficient muscular strength through isometric exercise may benefit from repetitive isotonic active exercises with minimal resistance, so long as they are performed with a reduced range of joint motion. Some ways of practicing these exercises would be swimming, stationary bicycling, or gardening, with the stipulation that fatigue should be avoided [Ekblom, 1975b; Minor, 1989]. 7.3 Local therapy 7.3.1 Local therapy in RA is indicated in joints with persistent disease activity despite adequate systemic control of the disease. The smaller the radiographic damage in a joint and the less systemic inflammatory activity of RA, the higher the probability that local treatment will have good results. Local treatment in RA is understood as therapy applied to an individual joint. During the course of RA it is frequently found that, although the disease's systemic inflammatory activity is controlled with the use of DMARDs and anti-inflammatories (steroidal or nonsteroidal), some joints still have active inflammation characterized by the presence of pain at rest, tenderness, pain on motion, swelling, and localized warmth. In these situations the systemic medication that is controlling the disease should be maintained, adding a local treatment to reduce inflammation in the joint where it is not controlled. 7.3.2 Intra-articular steroid injection is the procedure of choice. The most effective corticosteroid preparation is triamcinolone hexacetonide due to its prolonged intra-articular duration. If corticosteroid infiltration fails, radioisotopic synoviolisis and chemical synoviolisis with osmic acid may be useful procedures before referring the patient for arthroscopic or surgical synovectomy. The first step is to obtain synovial liquid for testing. The presence of infection should be ruled out by Gram stain and appropriate cultures. The administration of intra-articular corticosteroids is the local therapy of choice for an out-of-phase joint when infection has been ruled out. A number of drugs exist, including methylprednisolone acetate, paramethasone acetonide, triamcinolone acetonide, and triamcinolone hexacetonide. This guideline recommends the use of triamcinolone hexacetonide because of its prolonged action, lasting several months [Bain, 1972; Bird, 1979; Blyth, 1994]. After administering the corticosteroid, it is a good idea to prescribe joint rest for at least 24 hours [Chakravarty, 1994]. Radioisotopic synovectomy consists of the intra-articular administration of a radioactive drug that emits high energy beta particles. The synovial membrane receives local radiation, destroying the synovial membrane cells and reducing their thickness, which improves the symptoms of inflammation in the medium- and long term. The most commonly used drugs in large joints such as the knee are dysprosium-165, 90-yttrium, and samarium-153. For medium-sized joints such as the elbows, carpals, or ankles, rhenium-186 has been used. For small joints such as the metacarpophalangeal or proximal interphalangeal joints, erbium-169 has been used [Muller, 1975; Gumpel, 1979; Sledge, 1984; Sledge, 1986; Sledge 1987; Jones, 1993; Barnes, 1994; Clunie, 1996; Asavatanabodee, 1997; Jahangier, 1997; O´Duffy, 1999]. Chemical synovectomy refers to the intra-articular administration of a chemical agent capable of destroying the synovial membrane by metabolic or abrasive action on the cells or extracellular material. Osmic acid is the most commonly used agent [Bontoux, 1978]. CHAPTER 8. EXTRA-ARTICULAR COMPLICATIONS OF RHEUMATOID ARTHRITIS 8.1 Amyloidosis 8.1.1 Secondary amyloidosis should be suspected in RA patients who develop proteinuria, renal failure, gastrointestinal symptoms, myocardiopathy and/or hepatomegaly, and in those having elevated phase reactants concurrent with little clinical activity. 8.1.2 The recommended treatment is chlorambucil at a dosage of 0.1-0.2 mg/kg/day [Berglund, 1993; David, 1993]. Background: Amyloidosis is a syndrome characterized by the presence of insoluble deposits of normal serum proteins in the extracellular material of one or more organs. Secondary amyloidosis, produced by the serum amyloid A, a phase reactant, is the most frequent form of amyloidosis. It occurs in association with a large number of chronic inflammatory diseases such as RA. The inflammatory response tends to increase the production of serum amyloid A in the liver, which is broken down in the circulating macrophages into smaller fragments that are deposited in the tissues [Glenner, 1980]. Amyloidosis increases RA mortality by causing organ dysfunction. When to suspect: Secondary amyloidosis should be suspected in RA patients who develop proteinuria, renal failure, gastrointestinal symptoms, myocardiopathy and/or hepatomegaly [Okuda, 1994; Hazenberg, 1994], as well as in those having elevated phase reactants concurrent with little clinical activity. Clinical description: The prevalence of amyloidosis in RA in post-mortem studies ranges between 10 and 25% [Husby, 1985; Pai, 1993]; it becomes symptomatic in only 2-10 %. It is more frequent in patients with severe long-standing disease [Okuda, 1994]. One of the most frequent indicators of amyloid deposits in RA patients is the presence of proteinuria, which may progress to nephrotic syndrome. Other organs that may be affected are the heart, liver, spleen, gut, and skin. Diagnosis: The diagnosis of amyloidosis requires documentation of an extracellular fibrillar deposit with green birefringence under polarized light in biopsies stained with Congo red [Glenner, 1980]. It is recommended that biopsies be made of abdominal fat [Duston, 1989] or of rectal or gingival mucosa [Kyle, 1983], which is safe and accessible. It has recently been suggested that scintigraphy with serum amyloid P component (a phase reactant that coprecipitates in all forms of amyloidosis) marked with iodine-123 may be a reliable alternative to biopsy in the diagnosis of secondary amyloidosis [Tan, 1995]. Treatment: Treatment should always aim to suppress RA activity [Gerts, 1991; Berglund, 1993; David, 1993; Lovat, 1997]. Chlorambucil at a dosage of 0.1-0.2 mg/kg/day has been shown to play a beneficial role in the treatment of secondary amyloidosis in patients with chronic arthritis [Berglund, 1993; David 1993]. 8.2 Anemia 8.2.1 Anemia in RA is usually asymptomatic, therefore periodic blood cell counts should be obtained including erythrocyte, leukocyte, and platelet counts; calculation of the mean corpuscular volume (MCV); reticulocyte count; and general liver and kidney function tests. 8.2.2 There is no specific treatment for anemia in RA. It should be considered for possible changes in RA treatment guidelines. Background: Anemia is one of the hematologic disorders that can appear during the course of RA. It may be related with the disease's inflammatory character and chronic nature, with associated iron-deficiency, or it may be an undesired effect of treatment. The severity depends on its intensity. If the anemia is severe (hemoglobin less than 80 g/l), not only iron deficiency, but also other, infrequent causes of anemia should be considered, such as autoimmune hemolytic anemia and drug-induced aregenerative anemia. In the absence of blood loss, the diagnosis of hemolytic anemia is based on reticulocytosis, and the diagnosis of aregenerative anemia is based on the drug used in treatment and its association with leukopenia and thrombocytopenia. When to suspect: The anemia is usually moderate and asymptomatic, and is detected in routine tests. Typical symptoms may also develop at times, such as fatigue, increasing difficulty in normal physical exertion, palpitations, and/or pale skin and mucosa. The range of symptoms depends on the intensity of the anemia and on the organism's possible development of adaptive mechanisms. Diagnosis: A blood count should be obtained, including erythrocyte, leukocyte, and platelet counts; calculation of the mean corpuscular volume (MCV); reticulocyte count; and general liver and kidney function tests. If anemia exists, a second round of tests may be obtained to diagnose its etiology. These tests may include: * Determination of the serum levels of ferritin and transferrin and their saturation index, indicated to evaluate associated iron deficiency (Table 25). Iron deficiency is also accompanied by elevated transferrin levels, with a low saturation index (less than 16%). * Myelogram. Indicated only in cases where there is unconfirmed suspicion of coexisting iron-deficiency anemia. * Bone marrow biopsy. Only if aregenerative anemia is suspected. Table 25. Definition and characteristics of anemia in RA DEFINITION * Hemoglobin * less than 130 g/l (men); less than 120 g/l (women) CHARACTERISTICS * Normocytic * MCV: 83-97 fl CHARACTERISTICS * Normochromic * MCH: 27-33 pg CHARACTERISTICS * Transferrinemia * Slightly reduced saturation index CHARACTERISTICS * Reticulocytes * Normal or low CHARACTERISTICS * Ferritin (serum values) * greater than 60 ng/ml less than 12 ng/ml (iron deficiency anemia) 12-60 ng/ml (anemia associated with iron deficiency) Treatment: Transfusions of concentrated red blood cells are contraindicated, except in exceptional cases of severe anemia with high cardiocirculatory risk. Iron (oral ferrous salt compounds) should only be administered when there is evidence of coexisting iron-deficiency anemia. The cause of blood loss should also be investigated. The usefulness of administering erythropoietin has not been defined, although it could be indicated prior to surgery, particularly before joint prosthesis. The treatment required for pure red cell aplasia is complex and should be carried out in a hematology unit. 8.3 Cardiological complications 8.3.1 Cardiac involvement should be suspected if there is pericardial pain, heart failure, or conduction abnormalities. The two most frequent complications are pericarditis and myocarditis. 8.3.2 Pericarditis should be treated with full doses of NSAIDs (150 mg/day of indomethacin). If this treatment is not effective, prednisone (1 mg/kg/day) is useful to control symptoms. Myocarditis is treated with high-dose steroids, diuretics, digitalis, vasodilatador agents, and antiarrhythmia drugs. Background: The heart and adjacent structures (pericardium and vessels) may be affected by nodular lesions or fibrosis in RA patients. In these patients, however, heart involvement is likely to be due to causes other than rheumatoid disease itself. The cardiac lesions derived from RA have little clinical expression, and are usually mild alterations not requiring treatment. When to suspect: Pericardial-type pain, heart failure, or conduction abnormalities should be a warning of possible cardiac involvement in an RA patient. The two types of cardiac involvement in RA patients are pericarditis and myocarditis. Pericarditis Clinical description: This is the most frequent type, with an annual incidence of 0.3-0.4% [Jurik, 1986]. Echocardiographic studies show asymptomatic pericardial effusion in over 30% of RA patients. In some exceptional cases of massive effusions or rapid onset, cardiac tamponade manifesting as shock is produced. Most RA patients with symptomatic pericarditis are males with positive RF. Diagnosis: Pericardial effusion seen on echography. Treatment: Full doses of NSAIDs (150 mg/day of indomethacin). If this is not effective, prednisone (1 mg/kg/day) is useful to control symptoms. Cases of cardiac tamponade should be treated with evacuation by pericardiocentesis. Myocarditis Clinical description: Myocarditis may be interstitial or granulomatous and is rare in RA [Lebowitz, 1963]. The granulomatous variety is highly specific in RA, while the interstitial variety is more frequent in lupus. Myocarditis presents as progressive shortness of breath. The physical examination reveals tachycardia, reduced differential blood pressure, and frequently a third heart sound. The coronary valves may be affected by granulomas leading to valve insufficiency and affecting, in this order, the mitral, aortic, tricuspid, and pulmonary valves. The coronary arteries may be affected in RA, especially the small intramyocardial vessels. Coronary arteritis is not usually expressed clinically, thus the presence of symptoms of ischemia in RA patients is practically always due to associated coronary arteriosclerosis. Diagnosis: Reduced contractility shown on echocardiography. Biopsy of the right ventricle may be indicated in these patients. Treatment: High-dose steroids, combined with diuretics, digitalis, vasodilators, and antiarrhythmia drugs. 8.4 Osteoporosis 8.4.1 Osteoporosis should be suspected in the presence of vertebral or peripheral fractures not due to trauma. When RA is first diagnosed, all patients should be evaluated for the main risk factors for fracture and loss of bone mass; this analysis should include both RA-associated and independent risk factors (Tables 26 and 27). 8.4.2 First-line agents for the specific treatment of osteoporosis are alendronate, risedronate, or hormone replacement therapy or, alternatively, cyclical etidronate or calcitonine. Background: A large percentage of RA patients have low bone mass in the axial and peripheral skeleton. These patients are considered to have twice the risk of the general population for developing fractures of the vertebra and femur, with a relative risk of 2.1 for fracture of the vertebra and 1.5-2.1 for fracture of the femur, rising to 4.4 in patients with marked alternation of functional capacity. Many risk factors are involved in the development of osteoporosis: age, post-menopause (in women), disease activity, functional capacity, immobility, and the influence of drugs used in treatment, especially the corticosteroids. Major bone mass loss has been described even in the initial phases of RA. Despite this evidence, no CPG for osteoporosis treatment in RA has been developed to date. When to suspect: Vertebral or peripheral fractures, excluding those caused by trauma. Diagnosis: At the initial examination an analysis should be made of the main risk factors for fracture and loss of bone mass, both risk factors that are associated with RA and those that are independent of the disease (Tables 26 and 27). If one or more of these factors is present, a bone densitometry of the lumbar spine and femur is indicated, according to the recommendations of the National Osteoporosis Foundation (US), the International Committee for Osteoporosis Clinical Guidelines in post-menopausal women, the ACR (US), and the National Osteoporosis Society (Great Britain) for the prevention and treatment of corticosteroid-induced osteoporosis. Since a large percentage of vertebral fractures are asymptomatic, a lateral radiograph should be made of the dorsal and lumbar spine to evaluate the existence of vertebral fractures in accordance with the following criterion for fracture: a 20% or greater reduction of the anterior, mid, or posterior height of the vertebral body. Laboratory tests will usually also be obtained to rule out associated processes that may be causing the osteoporosis. Table 26. RA-independent risk factors for osteoporosis * Age over 65 years * History of fracture due to bone fragility after 40 years of age * Body weight under 58 kg * Fractures due to bone fragility in nuclear family members * Smoking * Early menopause * Prolonged amenorrhea * Hypogonadism in males * Other predisposing diseases Table 27. RA-associated risk factors for osteoporosis * Active disease * HAQ greater than 1.25 * Treatment with corticosteroids greater than 7.5 mg/day for over 6 months, or cumulative doses over 30 g * Treatment with methotrexate In accordance with the WHO criteria for the diagnosis of osteoporosis in women, osteopenia or osteoporosis is considered to exist when the T-scale value is between -1 and -2.5, or is less than -2.5, respectively. Although there is no official consensus, these diagnostic criteria appear to be valid in men. Treatment: Since all RA patients are at risk for osteoporosis, the following recommendations for preventive treatment are made for all such patients: * Stop smoking and excessive alcohol consumption. * Maintain physical activity. * Take necessary precautions to avoid falls. * Calcium supplements sufficient to reach a daily intake, including diet, of 1,500 mg, plus 400-800 IU of vitamin D3. * If hypercalciuria is present, thiazides should be administered. Begin specific treatment of osteoporosis if: * There is a history of fracture of the vertebra or femur. * Bone mineral density is less than -1.5 on the T scale. * The patient is over 65 years of age. * The dose of corticosteroids is higher than 7.5 mg/day of prednisone for more than 6 months, and there are other risk factors (Tables 26 and 27). Treatment with alendronate, residronate, or hormone replacement therapy (estrogens either alone or in association with progestogens - depending on whether or not the uterus is intact - in postmenopausal women, or testosterone in hypogonadal men). If contraindicated, cyclical etidronate or calcitonine can be used as an alternative. At the time this guideline was written, no information was available on the efficacy of raloxifene in secondary osteoporosis. Monitoring: Patients who have a baseline densitometry should repeat the test one year later, and every 1-3 years thereafter. If annual bone loss is more than 4% in the lumbar spine or 7% in the femur, treatment should be initiated or modified. 8.5 Lung complications 8.5.1 The appearance of pleuritic pain, shortness of breath (either progressive or of recent onset), or hemoptysis suggests pulmonary disease in RA patients. Lung complications may include pleural disease, rheumatoid nodules, interstitial fibrosis, or bronchiolitis obliterans with organizing pneumonia (BOOP). 8.5.2 Treatment for pleural disease includes thoracocentesis to obtain an exudate and rule out other pathologies (infection or neoplasm), NSAIDs at full doses, or steroids at half doses (10-20 mg/day of prednisone). Rheumatoid nodules do not require treatment in the absence of complications. Recent onset (acute) interstitial involvement is treated with prednisone (1-1.5 mg/kg/day). If there is no response, patients may be treated with cyclophosphamide or azathioprine. BOOP is treated with prednisone (1.5 mg/kg/day). Background: It is not clear how pulmonary disease is associated with RA [Kelly, 1993]. Pleuritis (with or without effusion), pulmonary nodules (with or without cavitation), rheumatoid pneumoconiosis (Caplan's syndrome), and BOOP are accepted rheumatoid manifestations. The association of RA with bronchiectasis or with diffuse interstitial fibrosis, however, is questionable [McMahon, 1993; Shadick, 1994]. In any case, the appearance of pulmonary lesions in a patient with RA makes it necessary to rule out the presence of a neoplastic process, infection, or a drug side effect (pneumonitis due to gold salts or methotrexate). When to suspect: Pulmonary disease should be suspected in the event of pleuritic pain, shortness of breath, either progressive or of recent onset, or hemoptysis. Pleural disease: Clinical description: The incidence of pleuritis in RA patients is nearly 20% [Jurik, 1982]. It is usually asymptomatic and especially affects male patients with seropositive and nodular RA. When it is symptomatic, it manifests as pleuritic pain and pleural effusion. Diagnosis: Low cell count in the pleural fluid (less than 5,000 leukocytes/mm^3), low concentration of glucose (less than 40 mg/dl) and complement, together with a high total protein content. Treatment: Treatment includes thoracocentesis to obtain an exudate and rule out other pathologies (infection or neoplasm), full doses of NSAIDs, and half-doses of steroids (10-20 mg/day of prednisone). Rheumatoid nodules: Clinical description: Intrapulmonary rheumatoid nodules usually remain asymptomatic, except when they cavitate, in which case they may become superinfected or produce hemoptysis. The presence of pulmonary nodules has most frequently been described in RA patients exposed to inorganic dust (Caplan's syndrome in coal miners). All pulmonary nodules should be biopsied to rule out neoplasia. Diagnosis: The only certain diagnosis of rheumatoid nodules is histological. Treatment: The nodules have not been shown to respond to pharmacological treatment, nor do they usually require surgical treatment unless complications occur (pneumothorax). Interstitial fibrosis: Clinical description: Diffuse interstitial fibrosis affects 10% of patients with RA [Jurik, 1982]. Its clinical presentation is no different from the idiopathic syndrome, and it presents as a slowly progressing shortness of breath. Diagnosis: In all RA patients with pulmonary findings on the physical examination (fine basal crepitations) or on the radiography (reticular or reticulo-nodular pattern), the physician should request blood gas analysis and respiratory function tests (diffusion test). High resolution tomography, although it does not detect pulmonary inflammation, is a highly specific and sensitive technique for the diagnosis of pulmonary interstitial fibrosis. Treatment: Interstitial disease of recent onset (acute) is an indication for prednisone therapy (1-1.5 mg/kg/day). Patients who do not respond can be treated with cyclophosphamide or azathioprine, although there is no evidence that these agents reduce the progression of fibrosis. Bronchiolitis obliterans with organizing pneumonia (BOOP): Clinical description: BOOP is a rare pulmonary disease that has been related with RA [Rees, 1991]. It presents with cough and severe shortness of breath of recent onset. Diagnosis: Chest X-ray shows bilateral opacities of the pulmonary parenchyma without loss of volume. High resolution tomography reveals patchy areas in the pulmonary parenchyma, which are usually peripheral. A definitive diagnosis requires thoracoscopic lung biopsy, with the observation of intraluminal plugs in the bronchioles. Treatment: Oral prednisone (1.5 mg/kg/day) in a single daily dose for 4-6 weeks, then slowly tapering off until discontinuing the drug in 4-6 months. 8.6 Felty's syndrome 8.6.1 Felty's syndrome is characterized by the presence of splenomegaly, leukopenia (less than 3,500/mm^3), and neutropenia (less than 2,000/mm^3) in patients meeting RA criteria. 8.6.2 The use of filgastrim is recommended when the absolute neutrophil count is less than 1,000/mm^3 and the patient has a history of associated severe infection. Background and diagnosis: Felty's syndrome is an infrequent but serious extra-articular manifestation of RA. Patients with this syndrome meet the criteria for RA and in addition have splenomegaly, leukopenia (less than 3,500/mm^3), and neutropenia (less than 2,000/mm^3). Its prognosis is largely governed by the higher incidence of systemic manifestations, mainly bacterial infections. This higher incidence of infection is due both to neutropenia and to defective functioning of the neutrophils. Treatment: There are no controlled studies showing the efficacy of any specific treatment for Felty's syndrome, thus the patient should be managed the same as for RA, together with prophylactic measures against infection and empirical treatment for fever the same as those used in patients with secondary neutropenia due to the use of cytotoxic drugs. These recommendations will vary in each center depending on the frequency of isolation of particular infectious agents, thus each center should use its own guidelines. Filgastrim is recommended when the absolute neutrophil count is lower than 1,000/mm^3 and the patient has a history of severe infection associated with the course of Felty's syndrome. A bone marrow biopsy should be performed before using filgastrim, since myeloid processes may simulate Felty's syndrome. In refractory cases surgical splenectomy or embolization of the splenic artery is indicated. 8.7 Secondary Sjögren's syndrome 8.7.1 A patient with RA is considered to have secondary Sjögren's syndrome (SSS) if there are signs and symptoms indicative of xerophthalmia and xerostomia. 8.7.2 There are no specific recommendations for modifying the course of SSS in RA. This guideline recommends symptomatic treatment of xerophthalmia and xerostomia. Dental and ophthalmological examinations are recommended every 6 months. Background and diagnosis: An RA patient is considered to have SSS if there are objective signs and symptoms of xerophthalmia together with objective signs and symptoms of xerostomia. Objective signs of xerophthalmia are considered to be an altered Schirmer test, together with a diagnosis of keratoconjunctivitis sicca by rose Bengal staining or fluorescein. Objective signs of xerostomia are considered to be reduced production of saliva measured by Lashley cup or other methods, together with a positive minor salivary gland biopsy and a lymphoid foci count of 2 or higher, measured as the average of four salivary gland lobules. A differential diagnosis should be made in patients with sarcoidosis, lymphoma, AIDS, hepatitis, autonomous neuropathy, and salivary gland hypertrophy. Treatment: There are no specific recommendations for modifying the course of SSS in RA. Dryness of the eyes should be treated with: * Withdrawal, if possible, of drugs that produce ocular dryness, such as drugs for hypertension, diuretics, and psychotropic drugs * Use of artificial tears * Avoidance of dry areas, those that are excessively warm, or contain irritating gases, including tobacco smoke * Temporary or permanent surgical occlusion of the tear duct. Dryness of the mouth should be treated with: * Withdrawal, if possible, of drugs that produce mouth dryness, such as drugs for hypertension, diuretics, and psychotropic drugs * Use of artificial saliva * Use of sugar-free lemon drops * Use of oral pilocarpine (5 mg/6 h). Multidisciplinary teams should be created consisting of a rheumatologist, a dentist, and an ophthalmologist. Dental and opthalmological examinations are recommended every 6 months. 8.8 Vasculitis 8.8.1 Rheumatoid vasculitis is understood to be a set of vascular processes (periungual splinter hemorrhages, palpable purpura, polyarteritis nodosa) with variable prognosis and treatment. 8.8.2 Palpable purpura should be treated with full doses of NSAIDs and medium to low doses of prednisone (15-30 mg/day). Polyarteritis nodosa is treated initially with high- dose steroids (40-120 mg/day of prednisone). If there is no response, cyclophosphamide can be added, either 2-3 mg/kg/day orally or 0.5-1 g/m^2 in intravenous pulses of 2 to 4 weeks. Background: Rheumatoid vasculitis is an infrequent extra-articular manifestation of RA. It appears in RA of long evolution, often with little or no joint inflammation. Risk factors for rheumatoid vasculitis are male gender, positive RF, the presence of other extra-articular manifestation of RA, and time of disease evolution. Palpable purpura: Diagnosis: Diagnosed clinically. Systematic skin punch biopsy is not recommended for histopathological confirmation, unless a vascular process other than small vessel leukocytoclastic vasculitis is suspected. Recently prescribed drugs should be reviewed to identify a possible pharmacological cause of the palpable purpura. Treatment: Generally disappears spontaneously. The most important factor in treatment is rest. If it does not disappear, palpable purpura should be treated with full doses of NSAIDs and medium to low doses of prednisone, beginning with 15 to 30 mg/day and progressively reducing the dosage depending on disease evolution. Polyarteritis nodosa-type rheumatoid vasculitis: Diagnosis: This is the most severe form of rheumatoid vasculitis and is life threatening in many patients. Histopathological confirmation is recommended whenever possible, since treatment of this form of vasculitis is frequently accompanied by severe adverse effects. Nevertheless, the physician can initiate treatment without histopathological confirmation in the most common and typical clinical presentations such as distal necrosis, skin ulceration, or multiple mononeuritis. Depending on the clinical manifestations, various complementary studies should be made, such as liver and kidney tests, arteriography, electromyogram-electroneurogram, skin biopsy, subcutaneous tissue biopsy, or biopsy of the sural nerve. Treatment: Initial treatment is with high-dose steroids: from 40 to 120 mg of prednisone or its equivalent, in single or divided doses. The dosage selected for a particular patient will depend on the severity of the process and the threat to life. If the clinical manifestations are not controlled with high-dose prednisone or if they reappear when trying to reduce the dosage, cyclophosphamide can be added, either 2-3 mg/kg/day orally or 0.5 to 1 g/m^2 in intravenous pulses every 2 to 4 week. Periungual splinter hemorrhages: Although periungual splinter hemorrhages are traditionally included in the vascular manifestations associated with RA, they are not histologically related with vasculitis. Diagnosis: They are diagnosed clinically and do not require complementary examinations. Treatment: No specific treatment is required. Close clinical monitoring is recommended for the early identification and treatment of vascular phenomena that may develop in the future. ACRONYMS * ACR * American College of Rheumatology * AIMS * Arthritis Impact Measurement Scales * APR * Acute phase reactants * ARA * American Rheumatism Association * ASA * Acetylsalicylic acid * AUR * Auranofin * AZA * Azathioprine * BNCS * Biblioteca Nacional de Ciencias de la Salud (National Library of the Health Sciences) * BOOP * Bronchiolitis obliterans with organizing pneumonia * BP * Blood pressure * CLQ * Chloroquine * CPA * Cyclophosphamide * CPG * Clinical practice guideline * CRP * C-reactive protein * CS * Corticosteroids * CSA * Cyclosporin A * DAS * Disease activity score * DAS 28 * Disease activity score counting 28 joints * DP * D-penicillamine * ESR * Erythrocyte sedimentation rate * ETN * Etanercept * EULAR * European Leagues Against Rheumatism * FDA * Food and Drug Administration * FIdeM * Fundación Ignacio de Mercado (Ignacio de Mercado Foundation) * GUIPCAR * Guía de Práctica Clínica para el manejo de Artritis Reumatoide en España (Clinical Practice Guideline for the Mangement of Rheumatoid Arthritis in Spain) * HAQ * Health Assessment Questionnaire * HCQ * Hydroxychloroquine * IFM * Infliximab * IG * Injectable gold * ILAR * International Leagues Against Rheumatism * IOM * Institute of Medicine (National Academy of Sciences) * ISCIII * Instituto de Salud Carlos III (Carlos III Health Institute) * LEF * Leflunomide * MCH * Mean corpuscular hemoglobin * MCP * Metacarpophalangeal joint * MCV * Mean corpuscular volume * MHAQ * Modified Health Assessment Questionnaire * MTP * Metatarsophalangeal joint * MTX * Methotrexate * NIWI * Nederlands Instituut voor Wetenschappelijke Informatiediensten (Netherlands Institute for Scientific Information Services) * NPJ * Number of painful joints * NPJ28 * Number of painful joints out of 28 joints * NPJ44 * Number of painful joints out of 44 joints * NSAIDs * Nonsteroidal anti-inflammatory drugs * NSJ * Number of swollen joints * NSJ28 * Number of swollen joints out of 28 joints * NSJ44 * Number of swollen joints out of 44 joints * OMERACT * Outcome Measures in Rheumatoid Arthritis Clinical Trials * OR * Odds ratio * PCB * Placebo * PGA * Patient's global assessment (of disease or health) * PIP * Proximal interphalangeal joint * RA * Rheumatoid arthritis * RF * Rheumatoid factor * RI * Ritchie index * SER * Sociedad Española de Reumatología (Spanish Society of Rheumatology) * SSS * Secondary Sjögren's syndrome * SSZ * Sulphasalazine * TAISS * Técnicas Avanzadas de Investigación en Servicios de Salud (Advanced Research Techniques in the Health Services) * TB * Tuberculosis * TNF * Tumor necrosis factor * UISS * Unidad de Investigación en Servicios de Salud (Health Services Research Unit) * VAS * Visual analog scale * WHO * World Health Organization APPENDICES Appendix 1. Data collection instruments for parameters used in initial evaluation and monitoring of RA patients This appendix contains a model data collection sheet for the evaluation and monitoring of RA patients. This model can be adapted to each specialist's needs in accordance with the way the particular hospital or clinical practice is run, and can be added to the patient's usual clinical record. First, there are three scales that the patient should fill out with reference to the previous week: change with respect to the last visit, pain, and global assessment of disease. The bottom half is for the physician. It is useful to mark the swollen joints with a dot (*) and the painful joints with an X. The usual procedure is to give this sheet to the patient at the end of the visit, and ask the patient to fill it out at home the day before returning for the next appointment. The physician should emphasize that this is not to be done any sooner, and that the patient should fill out the form thinking only of the previous 7 days. The bottom half is for the physician's evaluation. The HAQ, also included in this appendix, should be printed on the back of the same sheet. Finally, this appendix also includes instructions on how to correct the HAQ, the most commonly used joint indices, and different ways of calculating the DAS. Patient Home Worksheet Complete this section only Clinical History |__|__|__|__|__| Date ________________ Please answer the following questions one day before your appointment with the rheumatologist. 1. How is your arthritis in comparison with your last visit? Much better Somewhat better The same Somewhat worse Much worse 2. How much pain have you noted during this past week? No pain Worse pain 1 2 3 4 5 6 7 8 9 10 3. In general, how has your arthritis been during this past week? Very good Very bad PGA (0-100) 1 2 3 4 5 6 7 8 9 10 ------------------------------------------------------------------------ Physician Physician's global assessment of disease. Very good Very bad 1 2 3 4 5 6 7 8 9 10 * NSJ: * NPJ: * RI: * ESR: /CPR: DAS=0.54(R1)+0.065(NSJ)+0.33(In ESR)+0.0072(PGA) DAS28=0.56(NPJ28)+0.28(NSJ28)+0.70(In ESR)+0.014(PGA) NOTE: Original Spanish evaluation sheet provided by Enrique Batlle Gualda. University General Hospital of Alicante. Health Assessment Questionnaire (HAQ) Name ________________________________________ Date ________________ We are interested in learning how your illness affects your ability to function in daily life. Please feel free to add any comments at the end of this form. Please check the response which best describes your usual abilities OVER THE PAST WEEK: Without ANY difficulty Score = 0 With SOME difficulty Score = 1 With MUCH difficulty Score = 2 UNABLE to do Score = 3 DRESSING AND GROOMING - Are you able to: - Dress yourself, including tying shoelaces and doing buttons? - Shampoo your hair? ARISING - Are you able to: - Stand up from a straight chair? - Get in and out of bed? EATING - Are you able to: - Cut your meat? - Lift a full cup or glass to your mouth? - Open a new milk carton? WALKING - Are you able to: - Walk outdoors on flat ground? - Climb up five steps? Please check any AIDS OR DEVICES that you usually use for any of these activities: * Cane * Devices used for dressing (buttonhook, zipper pull, long handled shoe horn, etc.) * Walker * Built up or special utensils * Crutches * Special or built up chair * Wheelchair * Other (Specify: __________________) Please check any categories for which you usually need HELP FROM ANOTHER PERSON: * Dressing and grooming * Eating * Arising * Walking Without ANY difficulty Score = 0 With SOME difficulty Score = 1 With MUCH difficulty Score = 2 UNABLE to do Score = 3 HYGIENE - Are you able to: - Wash and dry your entire body? - Take a tub bath? - Get on and off the toilet? REACH - Are you able to: - Reach and get down a 5-pound object (such as a bag of sugar) from just above your head? - Bend down to pick up clothing from the floor? GRIP - Are you able to: - Open car doors? - Open jars which have been previously opened? - Turn faucets on and off? ACTIVITIES - Are you able to: - Run errands and shop? - Get in and out of a car? - Do chores such as vacuuming or yardwork? Please check any AIDS OR DEVICES that you usually use for any of these activities: * Raised toilet seat * Bathtub bar * Bathtub seat * Long-handled appliances for reach * Jar opener (for jars previously opened) * Long-handled appliances in bathroom * Other (Specify: __________________) Please check any categories for which you usually need HELP FROM ANOTHER PERSON: * Hygiene * Gripping and opening things * Reach * Errands and chores SCORING OF HAQ Add the maximum score for each of the 8 sections and divide by 8 to give a score between 0-3. If aid/device or help is needed the score for that activity automatically = 2 (unless 3 has already been ticked). Normal function = 0 Most affected function = 3 Indices for the evaluation of joint inflammation and pain Cervical spine ACR (66/68) * - Ritchie (53) * +*m NSJ (44) * - Fuchs (28) * - Temporomandibular ACR (66/68) * + Ritchie (53) * +* NSJ (44) * - Fuchs (28) * - Sternoclavicular ACR (66/68) * + Ritchie (53) * +* NSJ (44) * + Fuchs (28) * - Acromioclavicular ACR (66/68) * + Ritchie (53) * +* NSJ (44) * + Fuchs (28) * - Shoulder ACR (66/68) * + Ritchie (53) * + NSJ (44) * + Fuchs (28) * + Elbow ACR (66/68) * + Ritchie (53) * + NSJ (44) * + Fuchs (28) * + Wrist ACR (66/68) * + Ritchie (53) * + NSJ (44) * + Fuchs (28) * + Metacarpophalangeal ACR (66/68) * + Ritchie (53) * +* NSJ (44) * + Fuchs (28) * + Proximal interphalangeal ACR (66/68) * + Ritchie (53) * +* NSJ (44) * + Fuchs (28) * + Distal interphalangeal ACR (66/68) * + Ritchie (53) * - NSJ (44) * - Fuchs (28) * - Hip ACR (66/68) * + Ritchie (53) * +m NSJ (44) * - Fuchs (28) * - Knee ACR (66/68) * + Ritchie (53) * + NSJ (44) * + Fuchs (28) * + Ankle ACR (66/68) * + Ritchie (53) * + NSJ (44) * + Fuchs (28) * - Subtalar ACR (66/68) * + Ritchie (53) * +m NSJ (44) * - Fuchs (28) * - Midtarsal ACR (66/68) * + Ritchie (53) * +*m NSJ (44) * - Fuchs (28) * - Metatarsophalangeal ACR (66/68) * + Ritchie (53) * +* NSJ (44) * + Fuchs (28) * - Interphalangeal (foot) ACR (66/68) * + Ritchie (53) * - NSJ (44) * - Fuchs (28) * - - The joints marked with an asterisk (*) are counted as a single joint. - In the ARA/ACR index the subtalar and midtarsal joints are counted as a single joint. - The Ritchie index counts (0-3) the presence of tenderness or pain on motion (*m*) Disease Activity Score Ranges from 0 (no disease activity) to 10 (maximum disease activity) DAS28 with four variables: DAS28=0.56(NPJ28)+0.28(NSJ28)+0.70(In ESR)+0.014(PGA) DAS28 with three variables: DAS28=0.56 (NPJ28) + 0.28 (NSJ28) + 0.70 (In ESR) + 1.08 + 0.16 Formula to transform original DAS to DAS28: DAS28 = 1.072 (DAS) + 0.938 Original DAS with four variables: DAS=0.54(RI) +0.065(NSJ44) + 0.33(In ESR) + 0.0072(PGA) Original DAS with three variables: DAS = 0.54 (RI) + 0.065 (NSJ44) + 0.33 (In ESR) + 0.224 ------------------------------------------------------------------------ NPJ28: Number of painful joints based on a count of 28 joints NSJ28: Number of swollen joints based on a count of 28 joints ln: Natural logarithm ESR: Erythrocyte sedimentation rate PGA: Patient's global assessment of health or disease on a VAS from 0 (very good) to 100 (very poor). Either of the two scales produces the same results, although the latter one is preferable. RI: Ritchie index NSJ44: Number of swollen joints based on a count of 44 joints Appendix 2. Initial RA treatment with DMARDs, according to the complete classification scheme (144 patient scenarios) The following list shows how the 144 different patient scenarios were classified as a function of disease characteristics, together with the recommended treatment/s in each case. I. PATIENT WITH less than 6 SWOLLEN JOINTS A.1. No erosions and has NOT taken NSAIDs and/or corticosteroids during the last 3 months 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 NSAIDs and/or corticosteroids - RF 40-100 NSAIDs and/or corticosteroids or chloroquine - RF greater than 100 NSAIDs and/or corticosteroids or chloroquine or sulphasalazine b. HAQ greater than or equal to 1 - RF less than 40 NSAIDs and/or corticosteroids - RF 40-100 NSAIDs and/or corticosteroids or chloroquine or sulphasalazine - RF greater than 100 NSAIDs and/or corticosteroids or chloroquine or sulphasalazine 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 NSAIDs and/or corticosteroids or chloroquine or sulphasalazine - RF 40-100 NSAIDs and/or corticosteroids or chloroquine or sulphasalazine - RF greater than 100 NSAIDs and/or corticosteroids or methotrexate b. HAQ greater than or equal to 1 - RF less than 40 NSAIDs and/or corticosteroids or methotrexate or chloroquine - RF 40-100 NSAIDs and/or corticosteroids or methotrexate - RF greater than 100 NSAIDs and/or corticosteroids or methotrexate A.2. No erosions and HAS taken NSAIDs and/or corticosteroids during the last 3 months 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Chloroquine - RF 40-100 Chloroquine or methotrexate - RF greater than 100 Chloroquine or methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate or sulphasalazine or chloroquine - RF 40-100 Methotrexate or sulphasalazine - RF greater than 100 Methotrexate or sulphasalazine 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Chloroquine or sulphasalazine or methotrexate - RF 40-100 Methotrexate or sulphasalazine - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate or sulphasalazine - RF 40-100 Methotrexate - RF greater than 100 Methotrexate B. From 1 to 3 erosions 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate C. More than 3 erosions 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate II. PATIENT WITH 6-10 SWOLLEN JOINTS A.1. No erosions and has NOT taken NSAIDs and/or corticosteroids during the last 3 months 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 NSAIDs and/or corticosteroids or methotrexate - RF 40-100 NSAIDs and/or corticosteroids or methotrexate - RF greater than 100 NSAIDs and/or corticosteroids or methotrexate or injectable gold b. HAQ greater than or equal to 1 - RF less than 40 NSAIDs and/or corticosteroids or methotrexate - RF 40-100 NSAIDs and/or corticosteroids or methotrexate - RF greater than 100 NSAIDs and/or corticosteroids or methotrexate or injectable gold 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 NSAIDs and/or corticosteroids or methotrexate - RF 40-100 NSAIDs and/or corticosteroids or methotrexate or injectable gold - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate A.2. No erosions and HAS taken NSAIDs and/or corticosteroids during the last 3 months 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate or injectable gold b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate or injectable gold - RF greater than 100 Methotrexate or injectable gold 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate B. From 1 to 3 erosions 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate C. More than 3 erosions 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate III. PATIENT WITH greater than 10 SWOLLEN JOINTS A.1 No erosions and has NOT taken NSAIDs and/or corticosteroids during the last 3 months 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 NSAIDs and/or corticosteroids or methotrexate or injectable gold - RF 40-100 NSAIDs and/or corticosteroids or methotrexate or injectable gold - RF greater than 100 NSAIDs and/or corticosteroids or methotrexate or injectable gold b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 NSAIDs and/or corticosteroids or methotrexate or injectable gold - RF 40-100 NSAIDs and/or corticosteroids or methotrexate or injectable gold - RF greater than 100 NSAIDs and/or corticosteroids or methotrexate or injectable gold b. HAQ greater than or equal to 1 - RF less than 40 NSAIDs and/or corticosteroids or methotrexate or injectable gold - RF 40-100 Methotrexate or injectable gold - RF greater than 100 Methotrexate or injectable gold A.2 No erosions and HAS taken NSAIDs and/or corticosteroids during the last 3 months 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate B. From 1 to 3 erosions 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate or injectable gold b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate or methotrexate + injectable gold - RF greater than 100 Methotrexate or methotrexate + injectable gold 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate or leflunomide - RF 40-100 Methotrexate or leflunomide - RF greater than 100 Methotrexate or leflunomide b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate or methotrexate + injectable gold - RF greater than 100 Methotrexate or methotrexate + injectable gold C. More than 3 erosions 1. Normal acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate - RF greater than 100 Methotrexate or injectable gold or leflunomide b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate - RF 40-100 Methotrexate or methotrexate + injectable gold - RF greater than 100 Methotrexate or methotrexate + injectable gold 2. Elevated acute phase reactants a. HAQ less than 1 - RF less than 40 Methotrexate or methotrexate + sulphasalazine - RF 40-100 Methotrexate or leflunomide - RF greater than 100 Methotrexate or leflunomide b. HAQ greater than or equal to 1 - RF less than 40 Methotrexate or injectable gold - RF 40-100 Methotrexate or methotrexate + injectable gold - RF greater than 100 Methotrexate or methotrexate + injectable gold TABLES Table 1. Disease modifying antirheumatic drugs included in the systematic literature review Table 2. Jadad scale for rating the quality of evidence from clinical trials Table 3. Hadorn scale for rating the quality of scientific evidence from articles for CPGs Table 4. Outline of article selection process Table 5. Clinical trials and comparisons by the Jadad scale for rating the quality of the evidence Table 6. Clinical trials and comparisons by the Hadorn scale for rating the quality of the evidence Table 7. Comparisons of DMARDs used only in monotherapy Table 8. Comparisons of single or combined DMARDs vs. drug combinations Table 9a. Mean time of RA progression, previous DMARD use, duration of treatment in the trial, level of evidence, bibliographic reference, and ID number in the comparisons of DMARDs used in monotherapy included in the synthesis of the evidence Table 9b. Mean time of RA progression, previous DMARD use, duration of treatment in the trial, level of evidence, bibliographic reference, and ID number in the comparisons of DMARDs used in monotherapy or combined therapy vs. drug combinations included in the synthesis of the evidence Table 10. Evaluation of the quality of the meta-analyses of DMARDs Table 11. Conclusions of the Cochrane Library meta-analyses and their level of evidence Table 12. Predictive values (%) for RA diagnostic criteria in different settings Table 13. Summary of instruments for the measurement of parameters in RA evaluation Table 14. Initial treatment by simplified clinical classification of RA Table 15. Alternative treatment in case of severe toxicity to initial treatment Table 16. Alternative treatment in case of unsatisfactory response to initial treatment Table 17. Recommended doses and commercial names of DMARDs Table 18. Usual dosage and frequency of administration of NSAIDs Table 19. Classification of the corticosteroids by duration of action Table 20. Use of DMARDs during pregnancy and breast-feeding Table 21. EULAR definition of response Table 22. CPA dosage producing toxicity Table 23. Percentage of women treated with CPA who develop amenorrhea Table 24. Adverse effects of DMARDs Table 25. Definition and characteristics of anemia in RA Table 26. RA-independent risk factors for osteoporosis Table 27. RA-associated risk factors for osteoporosis REFERENCES 1. References used in the guideline Abu-Shakra, 1994 Abu Shakra M, Nicol P, Urowitz MB. Accelerated nodulosis, pleural effusion, and pericardial tamponade during methotrexate therapy. J Rheumatol 1994; 21(5): 934-7. ACR Committee, 1996 American College of Rheumatology Ad Hoc Committee on Clinical Guidelines. Guidelines for monitoring drug therapy in rheumatoid arthritis. Arthritis Rheum 1996; 39(5): 723-31. Ahmed, 1984 Ahmed AR, Hombal SM. Cyclophosphamide (Cytoxan): a review on relevant pharmacology and clinical uses. J Am Acad Dermatol 1984; 11(6): 1115-26. Alarcón, 1987 Alarcon GS, Blackburn WD Jr, Calvo A, Castaneda O. Evaluation of the American Rheumatism Association preliminary criteria for remission in rheumatoid arthritis: a prospective study. J Rheumatol 1987; 14(1): 93-6. Alarcón, 1994 Alarcón GS, Macaluso M, Singh K, et al. Excess mortality in MTX-treated RA patients is not due to cancer or cardiovascular diseases. Arthritis Rheum 1994; 37: S421. Alarcón, 1995 Alarcón GS, Kremer JM, Macaluso M, et al. MTX-lung toxicity in RA: a multicenter case control study. Arthritis Rheum 1995; 38: S205. Al-Awadhi, 1993 Al-Awadhi A, Dale P, McKendry RJ. Pancytopenia associated with low dose methotrexate therapy: a regional survey. J Rheumatol 1993; 20(7): 1121-5. Amos, 1986 Amos RS, Pullar T, Bax DE, Situnayake D, Capell HA, McConkey B, et al. Sulphasalazine for rheumatoid arthritis: toxicity in 774 patients monitored for one to 11 years. Br Med J Clin Res Ed 1986; 293(6544): 420-3. Anderson, 1989 Anderson JJ, Felson DT, Meenan RF, Williams HJ. Which traditional measures should be used in rheumatoid arthritis clinical trials? Arthritis Rheum 1989; 32(9): 1093-9. Anderson, 1993 Anderson JJ, Chernoff MC. Sensitivity to change of rheumatoid arthritis clinical trial outcome measures. J Rheumatol 1993; 20(3): 535-7. Anderson, 2000 Anderson JJ, Wells G, Verhoeven AC, Felson DT. Factors predicting response to treatment in rheumatoid arthritis: the importance of disease duration. Arthritis Rheum 2000; 43(1): 22-9. Andonopoulos, 1994 Andonopoulos AP, Terzis E, Tsibri E, Papasteriades CA, Papapetropoulos T. D-Penicillamine induced myasthenia gravis in rheumatoid arthritis: an unpredictable common occurrence? Clin Rheumatol 1994; 13(4): 586-8. Ansell, 1975 Ansell ID, Castro JE. Carcinoma of the bladder complicating cyclophosphamide therapy. Br J Urol 1975; 47(4): 413-8. Ansher, 1983 Ansher AF, Melton JW 3d, Sliwinski AJ. Bladder malignancy in a patient receiving low dose cyclophosphamide for treatment of rheumatoid arthritis. Arthritis Rheum 1983; 26(6): 804-5. Appelbaum, 1976 Appelbaum F, Strauchen JA, Graw RG Jr, Savage DD, Kent KM, Ferrans VJ, et al. Acute lethal carditis caused by high-dose combination chemotherapy: a unique clinical and pathological entity. Lancet 1976; 1(7950): 58-62. Aptekar, 1973 Aptekar RG, Atkinson JP, Decker JL, Wolff SM, Chu EW. Bladder toxicity with chronic oral cyclophosphamide therapy in nonmalignant disease. Arthritis Rheum 1973; 16(4): 461-7. Arava, 1999 Prod Inf Arava (TM) 1999. Arellano, 1993 Arellano F, Krupp P. Malignancies in rheumatoid arthritis patients treated with cyclosporin A. Br J Rheumatol 1993; 32(Suppl 1): 72-5. Arnett, 1988 Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 1988; 31(3):315-24. Asavatanabodee, 1997 Asavatanabodee P, Sholter D, Davis P.Yttrium-90 radiochemical synovectomy in chronic knee synovitis: a one year retrospective review of 133 treatment interventions. J Rheumatol 1997; 24(4): 639-42. Ataya, 1993 Ataya R, Ramahi Ataya A. Reproductive performance of female rats treated with cyclophosphamide and/or LHRH agonist. Reprod Toxicol 1993; 7(3): 229-35. Austin, 1986 Austin HA 3d, Klippel JH, Balow JE, Le Riche NG, Steinberg AD, Plotz PH, et al. Therapy of lupus nephritis: controlled trial of prednisone and cytotoxic drugs. N Engl J Med 1986; 314(10): 614-9. Bacon, 1982 Bacon AM, Rosenberg SA. Cyclophosphamide hepatotoxicity in a patient with systemic lupus erythematosus. Ann Intern Med 1982; 97(1): 61-3. Bachman, 1996 Bachman TR, Sawitzke AD, Perkins SL, Ward JH, Cannon GW. Methotrexate-associated lymphoma in patients with rheumatoid arthritis: report of two cases. Arthritis Rheum 1996; 39(2): 325-9. Bain, 1972 Bain LS, Balch HW, Wetherly JM, Yeadon A. Intraarticular triamcinolone hexacetonide: double-blind comparison with methylprednisolone. Br J Clin Pract 1972; 26(12): 559-61. Baker, 1987 Baker GL, Kahl LE, Zee BC, Stolzer BL, Agarwal AK, Medsger TA Jr. Malignancy following treatment of rheumatoid arthritis with cyclophosphamide: long-term case-control follow-up study. Am J Med 1987; 83(1): 1-9. Balow, 1987 Balow JE, Austin HA 3d, Tsokos GC, Antonovych TT, Steinberg AD, Klippel JH. NIH conference. Lupus nephritis. Ann Intern Med 1987; 106(1): 79-94. Balsa, 1996 Balsa A, Richi P, Cabezas J, Usón J, Martín Mola E, Gijón J. Estudio descriptivo de los 100 primeros pacientes atendidos en una consulta de artritis de reciente comienzo. Rev Esp Reumatol, 1996; 23: 400-6. Baltus, 1983 Baltus JA, 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 1983; 42(4): 368-73. Bannwarth, 1994 Bannwarth B, Labat L, Moride Y, Schaeverbeke T. Methotrexate in rheumatoid arthritis: an update. Drugs 1994; 47(1): 25-50. Barnes, 1994 Barnes CL, Shortkroff S, Wilson M, Sledge CB. Intra-articular radiation treatment of rheumatoid synovitis of the ankle with dysprosium-165 ferric hydroxide macroaggregates. Foot Ankle Int 1994; 15(6): 306-10. Bass, 1996 Bass NM, Ockner RK. Drug-induced liver disease. In: Zakim D, Boyer TD, editors. Hepathology: A Textbook of Liver Disease. 3 ed. Philadelphia: WB Saunders; 1996. p. 962-1017. Bellamy, 1998 Bellamy N, Kaloni S, Pope J, Coulter K, Campbell J. Quantitative rheumatology: a survey of outcome measurement procedures in routine rheumatology outpatient practice in Canada. J Rheumatol 1998; 25(5): 852-8. Bellamy, 1999 Bellamy N, Muirden KD, Brooks PM, Barraclough D, Tellus MM, Campbell J. A survey of outcome measurement procedures in routine rheumatology outpatient practice in Australia. J Rheumatol 1999; 26(7): 1593-9. Belmonte, 1988 Belmonte Serrano MA, Roig Escofet D. Inmunomoduladores. Tratamiento (V). In: Roig Escofet D., editor. Artritis reumatoide. Barcelona: Doyma; 1988. p. 226-34. Benson, 1974 Benson TB, Copp EP. The effects of therapeutic forms of heat and ice on the pain threshold of the normal shoulder. Rheumatol Rehabil 1974; 13(2): 101-4. Berg, 1989 Berg KJ, Forre O, Djoseland O, Mikkelsen M, Narverud J, Rugstad HE. Renal side effects of high and low cyclosporin A doses in patients with rheumatoid arthritis. Clin Nephrol 1989; 31(5): 232-8. Berglund, 1993 Berglund K, Thysell H, Keller C. Results, principles and pitfalls in the management of renal AA-amyloidosis: a 10-21 year followup of 16 patients with rheumatic disease treated with alkylating cytostatics. J Rheumatol 1993; 20(12): 2051-7. Bird, 1979 Bird HA, Ring EF, Bacon PA. A thermographic and clinical comparison of three intra-articular steroid preparations in rheumatoid arthritis. Ann Rheum Dis 1979; 38(1): 36-9. Bird, 1990 Bird HA. Drugs and the elderly. Ann Rheum Dis 1990; 49(12): 1021-4. Birnie, 1981 Birnie GG, McLeod TI, Watkinson G. Incidence of sulphasalazine-induced male infertility. Gut 1981; 22(6): 452-5. Bitter, 1984 Bitter, T. Combined disease-modifying chemotherapy for intractable rheumatoid arthritis. Clin Rheum Dis 1984;10:417-28 Black, 1998 Black AJ, McLeod HL, Capell HA, Powrie RH, Matowe LK, Pritchard SC, et al. Thiopurine methyltransferase genotype predicts therapy-limiting severe toxicity from azathioprine. Ann Intern Med 1998; 129(1): 716-8. Blanco, 1996 Blanco R, Martínez Taboada VM, González Gay MA, Armona J, Fernández Sueiro JL, Gonzalez Vela MC, et al. Acute febrile toxic reaction in patients with refractory rheumatoid arthritis who are receiving combined therapy with methotrexate and azathioprine. Arthritis Rheum 1996; 39(6): 1016-20. Blyth, 1994 Blyth T, Hunter JA, Stirling A. Pain relief in the rheumatoid knee after steroid injection: a single-blind comparison of hydrocortisone succinate, and triamcinolone acetonide or hexacetonide. Br J Rheumatol 1994; 335): 461-3. Boers, 1988 Boers M, van Rijthoven AW, Goei The HS, Dijkmans BA, Cats A. Serum creatinine levels two years later: follow-up of a placebo-controlled trial of cyclosporin in rheumatoid patients. Transplant Proc 1988; 20(3 Suppl 4): 371-5. Boers, 1990 Boers M, Dijkmans BA, van Rijthoven AW, Goei The HS, Cats A. Reversible nephrotoxicity of cyclosporin in rheumatoid arthritis. J Rheumatol 1990: 17(1): 38-42. Boers, 1994 Boers M, Tugwell P, Felson DT, van Riel PL, Kirwan JR, Edmonds JP, et al. World Health Organization and International League of Associations for Rheumatology core endpoints for symptom modifying antirheumatic drugs in rheumatoid arthritis clinical trials. J Rheumatol 1994; 21(Suppl 41): 86-9. Boers, 1997 Boers M,Verhoeven AC, Markusse HM, van de Laar MA, Westhovens R, van Denderen JC, van Zeben C, et al. Randomised comparison of combined step-down prednisolone, methotrexate and sulphasalazine with sulphasalazine alone in early rheumatoid arthritis. Lancet 1997; 350(9074): 309-18. Bogoch, 1999. Bogoch ER, Moran EL. Bone abnormalities in the surgical treatment of patients with rheumatoid arthritis. Clin Orthop 1999; (366): 8-21. Bontoux, 1978 Bontoux D, Alcalay M, Reboux JF, Puthon A. Synoviortheses du genou par l'acide osmique: a propos de 61 injections. Rev Rhum Mal Osteoartic 1978; 45(2): 101-5. Boumpas, 1993 Boumpas DT, Austin HA 3d, Vaughan EM, Yarboro CH, Klippel JH, Balow-JE. Risk for sustained amenorrhea in patients with systemic lupus erythematosus receiving intermittent pulse cyclophosphamide therapy. Ann Intern Med 1993; 119(5): 366-9. Boyer, 1989. Boyer DL, Li BU, Fyda JN, Friedman RA. Sulphasalazine-induced hepatotoxicity in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 1989; 8(4): 528-32. Bradley, 1989 Bradley JD, Brandt KD, Katz BP. Infectious complications of cyclophosphamide treatment for vasculitis. Arthritis Rheum 1989; 32(1): 45-53. Breedveld, 1989 Breedveld FC, Markusse HM, MacFarlane JD. Subcutaneous fat biopsy in the diagnosis of amyloidosis secondary to chronic arthritis. Clin Exp Rheumatol 1989; 7(4): 407-10. Brennan, 1996 Brennan P, Harrison B, Barrett E, Chakravarty K, Scott D, Silman A, et al. A simple algorithm to predict the development of radiological erosions in patients with early rheumatoid arthritis: prospective cohort study. BMJ 1996; 313(7055): 471-6. Bressler, 1985 Bressler RB, Huston DP. Water intoxication following moderate-dose intravenous cyclophosphamide. Arch Intern Med 1985; 145(3): 548-9. Bretza, 1983 Bretza J, Wells I, Novey HS. Association of IgE antibodies to sodium aurothiomalate and adverse reactions to chrysotherapy for rheumatoid arthritis. Am J Med 1983; 74(6): 945-50. Bridges, 1991 Bridges SL Jr, Lopez Mendez A, Han KH, Tracy IC, Alarcon GS. Should methotrexate be discontinued before elective orthopedic surgery in patients with rheumatoid arthritis? J Rheumatol 1991; 18(7): 984-8. Brock, 1982 Brock N, Pohl J, Stekar J, Scheef W. Studies on the urotoxicity of oxazaphosphorine cytostatics and its prevention. III. Profile of action of sodium 2-mercaptoethane sulfonate (mesna). Eur J Cancer Clin Oncol 1982; 18(12): 1377-87. Brook, 1977 Brook A, Fleming A, Corbett M. Relationship of radiological change to clinical outcome in rheumatoid arthritis. Ann Rheum Dis 1977; 36(3): 274-5. Brook, 1990 Brook RH. Quality assessment and technology assessment: critical linkages. In: Lohr KN, Rettig RA, editors. Qualtity of care and technology assessment. Washington, D.C.: National Academy Press; 1990. p. 21-8. Brook, 1991 Brook RH. Quality of care: do we care? Ann Intern Med 1991; 115(6): 486-90. Bryant, 1980 Bryant BM, Jarman M, Ford HT, Smith IE. Prevention of isophosphamide-induced urothelial toxicity with 2-mercaptoethane sulphonate sodium (mesnum) in patients with advanced carcinoma. Lancet 1980; 2(8196): 657-9. Buchan, 1991 Buchan IE, Bird HA. Drug interactions in arthritic patients. Ann Rheum Dis 1991; 50(10): 680-1. Buchbinder, 1995 Buchbinder R, Bombardier C, Yeung M, Tugwell P. Which outcome measures should be used in rheumatoid arthritis clinical trials?: clinical and quality-of-life measures' responsiveness to treatment in randomized controlled trial. Arthritis Rheum 1995; 38(11): 1568-80. Burstin, 1993 Burstin HR, Lipsitz SR, Udvarhelyi IS, Brennan TA. The effects of hospital financial characteristics on quality of care. JAMA 1993; 270(7): 845-9. Cairns, 1992 Cairns HS, Neild G. Cyclosporin nephrotoxicity. In: Cameron S, Davison AM, Grünfeld JP, et al., editors. Oxford Textbook of Clinical Nephrology. Oxford: Oxford University Press; 1992. p. 1560-9. Caldwell, 1991 Caldwell JR, Furst DE. The efficacy and safety of low-dose corticosteroids for rheumatoid arthritis. Semin Arthritis Rheum 1991; 21(1): 1-11. Camussi, 1998 Camussi G, Lupia E. The future role of anti-tumour necrosis factor (TNF) products in the treatment of rheumatoid arthritis. Drugs 1998; 55(5): 613-20. Cannon, 1997 Cannon GW. Methotrexate pulmonary toxicity. Rheum Dis Clin North Am 1997; 23(4): 917-37. Canvin, 1993 Canvin JM, El Gabalawy HS, Chalmers MI. Fatal agranulocytosis with sulfasalazine therapy in rheumatoid arthritis. J Rheumatol 1993; 20(5): 909-10. Carpenter, 1996 Carpenter MT, West SG, Vogelgesang SA, Casey Jones DE. Postoperative joint infections in rheumatoid arthritis patients on methotrexate therapy. Ortthopedics 1996; 19(3): 207-10. Castillo, 1965 Castillo BA, el-Sallab RA, Scott JT. Physical activity, cystic erosion, and osteoporosis in rheumatoid arthritis. Ann Rheum Dis 1965; 24(6): 522-7. CCOHTA, 1999 The Canadian Coordinating Office for Health Technology Assessment. Etanercept: anti-tumor necrosis factor therapy for rheumatoid arthritis. Issues in Emerging Health Technologies 1999; Issue 8, Oct. Chakravarty, 1994 Chakravarty K, Pharoah PD, Scott DG. A randomized controlled study of post-injection rest following intra-articular steroid therapy for knee synovitis. Br J Rheumatol 1994; 33(5): 464-8. Chalmers, 1982 Chalmers A, Thompson D, Stein HE, Reid G, Patterson AC. Systemic lupus erythematosus during penicillamine therapy for rheumatoid arthritis. Ann Intern Med 1982; 97(5): 659-63. Chapman, 1981 Chapman RM, Sutcliffe SB. Protection of ovarian function by oral contraceptives in women receiving chemotherapy for Hodgkin´s disease. Blood. 1981; 58(4): 849-51. Chassin, 1991 Chassin MR. Quality of care. Time to act. JAMA 1991; 266(24): 3472-3. Clements, 1986 Clements PJ, Davis J. Cytotoxic drugs: their clinical application to the rheumatic diseases. Semin Arthritis Rheum 1986; 15(4): 231-54. Clunie, 1996 Clunie G, Lui D, Cullum I, Ell PJ, Edwards JC. Clinical outcome after one year following samarium-153 particulate hydroxyapatite radiation synovectomy. Scand J Rheumatol 1996; 25(6): 360-6. Cohen, 1992 Cohen DJ, Appel GB. Cyclosporin: nephrotoxic effects and guidelines for safe use in patients with rheumatoid arthritis. Semin Arthritis Rheum 1992; 21(6 Suppl 3): 43-8. Conaty, 1971 Conaty JP, Nikel VL. Functional incapacitation in rheumatoid arthritis: a rehabilitation challenge: a correlative study of function before and after hospital treatment. J Bone Joint Surg Am 1971; 53(4): 624-37. Creighton, 1998 Creighton MG, Callaghan JJ, Olegniczak JP, Johnson RC. Total hip arthroplasty with cement in patients who have rheumatoid arthritis: a minimum ten-year follow-up study. J Bone Joint Surg 1998; 80(10): 1439-46. Criswell, 2000 Criswell LA, Saag KG, Sems KM, Welch V, Shea B, Wells G, Suarez-Almazor ME Moderate-term, low-dose corticosteroids for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD001158. Cush, 1999 Cush JJ, Tugwell P, Weimblatt M, Yocum D. US consensus guidelines for the use of cyclosporin A in rheumatoid arthritis. J Rheumatol 1999; 26(5): 1176-86. Dahl, 1990 Dahl SL, Samuelson CO, Williams HJ, Ward JR, Karg M. Second-line antirheumatic drugs in the elderly with rheumatoid arthritis: a post hoc analysis of three controlled trials. Pharmacotherapy 1990; 10(2): 79-84. Damewood, 1986 Damewood MD, Grochow LB. Prospects for fertility after chemotherapy or radiation for neoplastic disease. Fertil Steril 1986, 45(4): 443-59. David, 1993 David J, Vouyiouka O, Ansell BM, Hall A, Woo P. Amyloidosis in juvenile chronic arthritis: a morbidity and mortality study. Clin Exp Rheumatol 1993; 11(1): 85-90. Davis, 1974 Davis P, Hughes GR. Significance of eosinophilia during gold therapy. Arthritis Rheum 1974; 17(6): 964-8. Dawes, 1986 Dawes PT, Fowler PD, Clarke S, Fisher J, Lawton A, Shadforth MF. Rheumatoid arthritis: treatment which controls the C- reactive protein and erythrocyte sedimentation rate reduces radiologic progression. Br J Rheumatol 1986; 25(1): 44-9. De Fronzo, 1973 DeFronzo RA, Braine H, Colvin M, Davis PJ. Water intoxication in man after cyclophosphamide therapy. Time course and relation to drug activation. Ann Intern Med 1973; 78(6): 861-9. Deandrade, 1965 Deandrade JR, Casagrande PA. A seven-day variability study of 499 patients with peripheral rheumatoid arthritis. Arthritis Rheum 1965; 19: 302-34. Decker, 1973 Decker JL. Toxicity of immunosuppressive drugs in man. Arthritis Rheum 1973; 16(1): 89-91. Donabedian, 1980 Donabedian A. Explorations in quality assessment and monitoring. Ann Arbor, MI: Health Administration Press; 1980. Donabedian, 1988 Donabedian A. The assessment of technology and quality: a comparative study of certainties and ambiguities. Int J Technol Assess Health Care 1988; 4(4): 487-96. Dougados, 1989 Dougados M, Duchesne L, Awada H, Amor B. Assessment of efficacy and acceptability of low dose cyclosporin in patients with rheumatoid arthritis. Ann Rheum Dis 1989; 48(7): 550-6. Dreyer, 1999 Dreyer SJ, Boden SD. Natural history of rheumatoid arthritis of the cervical spine. Clin Orthop1999; (366): 98-106. Drossaers-Bakker, 1999 Drossaers-Bakker KW, de Buck M, van Zeben D, Zwinderman AH, Breedveld FC, Hazes JM. Long-term course and outcome of functional capacity in rheumatoid arthritis: The effect of disease activity and radiologic damage over time. Arthritis Rheum 1999; 42(9): 1854-60. Drugex, 1999 Drugex® Ed. Staff. Drugex Information System. Micromedex Inc. Denver, Colorado. Vol. 101, 1999: Sulphasalazine. Dunbar, 1998 Dunbar RP, Alexiades MM. Decision making in rheumatoid arthritis. Determining surgical priorities. Rheum Dis Clin North Am 1998; 24(1): 35-54. Duston, 1989 Duston MA, Skinner M, Meenan RF, Cohen AS. Sensitivity, specificity, and predictive value of abdominal fat aspiration for the diagnosis of amyloidosis. Arthritis Rheum 1989; 32(1): 82-5. Eberhardt, 1998 Eberhardt K, Fex E. Clinical course and remission rate in patients with early rheumatoid arthritis: relationship to outcome after 5 years. Br J Rheumatol 1998; 37(12): 1324-9. Edmonds, 1999 Edmonds J, Saudan A, Lassere M, Scott D. Introduction to reading radiographs by the Scott modification of the Larsen method. J Rheumatol 1999; 26(3): 740-2. Ehrlich, 1982 Ehrlich GE. Heroic treatment for nonmalignant diseases. J Am Med Assoc 1982;248:1743-4. Ehrlich, 1984 Ehrlich RM, Freedman A, Goldsobel AB, Stiehm ER. The use of sodium-2-mercaptoethane sulfonate to prevent cyclophosphamide cystitis. J Urol 1984; 131(5): 960-2. Ekblom, 1975a Ekblom B, Lovgren O, Alderin M, Fridstrom M, Satterstrom G. Effect of short-term physical training on patients with rheumatoid arthritis I. Scand J Rheumatol 1975; 4(2): 80-6. Ekblom, 1975b Ekblom B, Lovgren O, Alderin M, Fridstrom M, Satterstrom G. Effect of short-term physical training on patients with rheumatoid arthritis: a six-month follow-up study. Scand J Rheumatol 1975; 4(2): 87-91. Ellman, 1991 Ellman MH, Hurwitz H, Thomas C, Kozloff M. Lymphoma developing in a patient with rheumatoid arthritis taking low dose weekly methotrexate. J Rheumatol 1991; 18(11): 1741-3. Emery, 1999 Emery P, Zeidler H, Kvien TK, Guslandi M, Naudin R, Stead H, et al. Celecoxib versus diclofenac in long-term management of rheumatoid arthritis: randomised double-blind comparison. Lancet 1999; 354(9196): 2106-11. Erickson, 1995 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 1995; 38(8): 1115-9. Escalante, 1999 Escalante A, Rincon I. How much disability in rheumatoid arthritis is explained by rheumatoid arthritis? Arthritis Rheum 1999; 42(8): 1712-21. Esteve- Vives, 1993 Esteve Vives J, Batlle Gualda E, Reig A. Spanish version of the Health Assessment Questionnaire (HAQ): reliability, validity and transcultural equivalency. Grupo para la Adaptación del HAQ a la Población Española. J Rheumatol 1993; 20(12): 2116-22. Esteve-Vives, 1994 Esteve-Vives J, Batlle-Gualda E, Tornero J, Tenorio M, Boquet D, Martínez-Sanchís A. Adaptación del Modified Health Assessment Questionnaire (MHAQ) a la población española. Rev Esp Reumatol 1994; 21: 165. Fairley, 1972 Fairley KF, Barrie JU, Johnson W. Sterility and testicular atrophy related to cyclophosphamide therapy. Lancet 1972; 1(7750): 568-9. Farr, 1986 Farr M, Scott DG, Bacon PA. Side effect profile of 200 patients whit inflammatory arthritis treated with sulphasalazine. Drugs 1986; 32 (Suppl 1): 49-53. Farrell, 1998 Farrell GC. Liver disease caused by drugs, anesthetics, and toxins. In: Feldman M, Scharschmidt BF, Sleisenger MH, editors. Gastrointestinal and Liver Disease. 6 ed. Philadelphia: WB Saunders; 1998. p. 1221-53. Fauci, 1983 Fauci AS, Haynes BF, Katz P, Wolf SM. Wegener´s granulomatosis: prospective clinical and therapeutic experience with 85 patients over 21 years. Ann Intern Med 1983; 98(1): 76-85. Fauci, 1989 Fauci AS, Young KR Jr. Immunoregulatory agents. In: Kelley WN, Harris ED, Ruddy S, Sledge CB, editors. Textbook of Rheumatology. 3 ed. Philadelphia: WB Saunders; 1989. p. 862-88. Fauci, 1993 Fauci AS, Randall KY. Immunoregulatory agents. In: Kelley W, Harris E, Ruddy S, Sledge CB, editors. Textbook of Rheumatology. Philadelphia: WB Saunders; 1993. p. 797-821. Feibel, 1976 Feibel A, Fast A. Deep heating of joints: a reconsideration. Arch Phys Med Rehab 1976; 57(11): 513-4. Felson, 1993a Felson DT. Choosing a core set of disease activity measures for rheumatoid arthritis clinical trials. J Rheumatol 1993; 20(3): 531-4. Felson, 1993b Felson DT, Anderson JJ, Boers M, Bombardier C, Chernoff M, Fried B, et al. The American College of Rheumatology preliminary core set of disease activity measures for rheumatoid arthritis clinical trials. The Committee on Outcome Measures in Rheumatoid Arthritis Clinical Trials. Arthritis Rheum 1993; 36(6): 729-40. Felson, 1994 Felson DT, Anderson JJ, Meenan RF. The efficacy and toxicity of combination therapy in rheumatoid arthritis. A meta-analysis. Arthritis Rheum 1994; 37(10): 1487-91. Felson, 1995 Felson DT, Anderson JJ, Boers M, Bombardier C, Furst D, Goldsmith C, et al. American College of Rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum 1995; 38(6): 727-35. Ferraz, 1990 Ferraz MB, Quaresma MR, Aquino LR, Atra E, Tugwell, Goldsmith CH. Reliability of pain scales in the assessment of literate and illiterate patients with rheumatoid arthritis. J Rheumatol 1990; 17(8): 1022-4. Finbloom, 1985 Finbloom D, Silver K, Newsome DA, Gunkel R. Comparison of hydroxychloroquine and chloroquine use and the development of retinal toxicity. J Rheumatol 1985; 12(4): 692-4. Fisher, 1996 Fisher CJ Jr, Agostini JM, Opal SM, Lowry SF, Blak RA, Sadoff JC, et al. Treatment of septic shock with tumor necrosis factor receptor: Fc fusion protein. N Engl J Med 1996; 334(26): 1697-702. Fizpatrick, 1991 Fitzpatrick R, Newman S, Archer R, Shipley M. Social support, disability and depression: a longitudinal study of rheumatoid arthritis. Soc Sci Med 1991; 33(5): 605-11. Forre, 1994 Forre O. Radiologic evidence of disease modification in rheumatoid arthritis patients treated with cyclosporine. Results of a 48-week multicenter study comparing low-dose cyclosporine with placebo. Arthritis Rheum 1994; 37(10): 1506-12. Fosdick, 1968 Fosdick WM, Parsons JL, Hill DF. Long-term cyclophosphamide therapy in rheumatoid arthritis. Arthritis Rheum 1968; 11(2): 151-61. Fox, 1994 Fox DA, McCune WJ. Immunosuppressive drug therapy of systemic lupus erythematosus. Rheum Dis Clin North Am. 1994; 20(1): 265-99. Fries, 1980 Fries JF, Spitz P, Kraines RG, Holman HR. Measurement of patient outcome in arthritis. Arthritis Rheum 1980; 23(2): 137-45. Fries, 1996 Fries JF, Williams CA, Morfeld D, Singh G, Sibley J Reduction in long-term disability in patients with rheumatoid arthritis by disease-modifying antirheumatic drug-based treatment strategies. Arthritis Rheum, 1996; 39(4): 616-22. Fuchs, 1989 Fuchs HA, Brooks RH, Callahan LF, Pincus T. A simplified twenty-eight-joint quantitative articular index in rheumatoid arthritis. Arthritis Rheum 1989; 32(5): 531-7. Fuchs, 1994 Fuchs HA, Pincus T. Reduced joint counts in rheumatoid arthritis clinical trials. Arthritis Rheum 1994; 37(4): 470-5. Furst, 1990 Furst DE. Proposition: methotrexate should not be the first second-line agent to be used in rheumatoid arthritis if NSAIDs fail. Semin Arthritis Rheum 1990; 20(2): 69-75. Furst, 1994a Furst DE. Predictors of worsening clinical variables and outcomes in rheumatoid arthritis. Rheum Dis Clin North Am 1994; 20(2): 309-19. Furst, 1994b Furst DE, Clements PJ. SAARDs. II. Pharmacologic approaches. In: Klippel JH, Dieppe PA, editors. Rheumatology. London: Mosby; 1994. p. 8: 13.1-13.8. Furst, 1995 Furst DE. Innovative treatment approaches for rheumatoid arthritis: cyclosporin, leflunomide and nitrogen mustard. Baillieres Clin Rheumatol 1995; 9(4): 711-29. Gates, 1993 Gates PE. Clinical quality improvement: getting physicians involved. QRB Qual Rev Bull 1993; 19(2): 56-61. Gertz, 1991 Gertz MA, Kyle RA. Secondary systemic amyloidosis: response and survival in 64 patients. Medicine (Baltimore) 1991; 70(4): 246-56. Gibson, 1987 Gibson T, Emery P, Armstrong RD, Crisp AJ, Panayi GS. Combined D-penicillamine and chloroquine treatment of rheumatoid arthritis -a comparative study. Br J Rheumatol 1987; 26:279-84. Glave Testino, 1994 Glave Testino C, Cardiel M, Arce Salinas A, Alarcon Segovia D. Factors associated with disease severity in Mexican patients with rheumatoid arthritis. Clin Exp Rheumatol 1994; 12(6): 589-94. Glenner, 1980 Glenner GG. Amyloid deposits and amyloidosis: the beta-fibrilloses (first of two parts). N Engl J Med 1980; 302(23): 1283-92. Goldberg, 1985 Goldberg JW, Lidsky MD. Cyclophosphamide-associated hepatotoxicity. South Med J 1985; 78(2): 222-3. Golden, 1995 Golden MR, Katz RS, Balk RA, Golden HE. The relationship of preexisting lung disease to the development of methotrexate pneumonitis in patients with rheumatoid arthritis. J Rheumatol 1995; 22(6): 1043-7. Gordon, 1973 Gordon DA, Stein JL, Broder I. The extra-articular features of rheumatoid arthritis: a systematic analysis of 127 cases. Am J Med 1973; 54(4): 445-52. Gotzsche, 2000a Gotzsche PC. Non-steroidal anti-inflammatory drugs. BMJ 2000; 320(7241): 1058-61. Gotzsche, 2000b Gotzsche PC, Johansen HK Short-term low-dose corticosteroids vs placebo and nonsteroidal antiinflammatory drugs in rheumatoid arthritis Cochrane Database Syst Rev 2000; (2): CD000189. Grant, 1987 Grant D, Wall W, Duff J, Stiller C, Ghent C, Keown P. Adverse effects of cyclosporin therapy following liver transplantation. Transplant Proc 1987; 19(4): 3463-5. Grassi, 2001 Grassi W, Filippucci E, Farina A, Salaffi F, Cervini C. Ultrasonography in the evaluation of bone erosions. Ann Rheum Dis 2001; 60(2): 98-104. Greene, 1986 Greene MH, Harris EL, Gershenson DM, Malkasian GD Jr, Melton LJ 3d, Dembo AJ, et al. Melphalan may be a more potent leukemogen than cyclophosphamide. Ann Intern Med 1986; 105(3): 360-7. Grob, 1999 Grob D, Schutz V, Plotz G. Occipitocervical fusion in patients with rheumatoid arthritis. Clin Orthop 1999; (366): 46-53. Grunwald, 1979 Grunwald HW, Rosner F. Acute leukemia and immunosuppressive drug use: a review of patients undergoing immunosuppressive therapy for non-neoplastic diseases. Arch Intern Med 1979; 139(4): 461-6. Guillemin, 1989 Guillemin F, Aussedat R, Guerci A, Lederlin P, Trechot P, Pourel J. Fatal agranulocytosis in sulphasalazine treated rheumatoid arthritis. J Rheumatol 1989; 16(8): 1166-7. Gulley, 1979 Gulley RM, Mirza A, Kelly CE. Hepatotoxicity of salicylazosulphapyridine: a case report and review of the literature. Am J Gastroenterol 1979; 72(5): 561-4. Gumpel, 1979 Gumpel JM, Matthews SA, Fisher M. Synoviorthesis with erbium-169: a double-blind controlled comparison of erbium-169 with corticosteroid. Ann Rheum Dis 1979; 38(4): 341-3. Gutiérrez-Ureña, 1996 Gutierrez Ureña S, Molina JF, García CO, Cuellar ML, Espinoza LR. Pancytopenia secondary to methotrexate therapy in rheumatoid arthritis. Arthritis Rheum 1996; 39(2): 272-6. Haagsma, 1997 Haagsma CJ, van Riel, de Jong AJ, van de Putte LB. Combination of sulphasalazine and methotrexate versus the single components in early rheumatoid arthritis: a randomized, controlled, double-blind, 52 week clinical trial. Br J Rheumatol 1997; 36:1082-8. Hadorn, 1996 Hadorn DC, Baker D, Hodges JS, Hicks N. Rating the quality of evidence for clinical practice guidelines. J Clin Epidemiol 1996; 49(7): 749-54. Hall, 1988a Hall CL, Jawad S, Harrison PR, MacKenzie JC, Bacon PA, Klouda PT, et al. Natural course of penicillamine nephropathy: a long term study of 33 patients. Br Med J 1988; 296(6629): 1083-6. Hall, 1988b Hall CL. Gold nephropathy. Nephron 1988; 50(4): 267-72. Halla, 1994 Halla JT, Hardin JG. Underrecognized postdosing reactions to methotrexate in patients with rheumatoid arthritis. J Rheumatol 1994; 21(7): 1224-6. Hansen, 1983 Hansen SE. Carcinoma of the bladder in a patient treated with cyclophosphamide for rheumatoid arthritis. Scand J Rheumatol 1983; 12(2): 73-4. Hargreaves, 1992 Hargreaves MR, Mowat AG, Benson MK. Acute pneumonitis associated with low dose methotrexate treatment for rheumatoid arthritis: report of five cases and review of published reports. Thorax 1992; 47(8): 628-33. Hargreaves, 1999 Hargreaves D, Emery R. Total elbow replacement in the treatment of rheumatoid disease. Clin Orthop 1999; (366): 61-71. Harris, 1974 Harris ED Jr, McCroskery PA. The influence of temperature and fibril stability on degradation of cartilage collagen by rheumatoid synovial colagenase. N Engl J Med 1974; 290(1): 1-6. Harris, 1983 Harris ED, Emkey RD, Nichols JE, Newberg A. Low dose prednisone therapy in rheumatoid arthritis: a double blind study. J Rheumatol 1983: 10(5): 713-21. Harrison, 1996 Harrison BJ, Symmons DP, Brennan P, Barrett EM, Silman AJ. Natural remission in inflammatory polyarthritis: issues of definition and prediction. Br J Rheumatol 1996; 35(11): 1096-100. Hayward, 1993 Hayward RA, Bernard AM, Rosevear JS, Anderson JE, McMahon LF. An evaluation of generic screens for poor quality of hospital care on a general medicine service. Med Care 1993; 31(5): 394-402. Hazenberg, 1994 Hazenberg BP, van Rijswijk MH. Clinical and therapeutic aspects of AA amyloidosis. Baillieres Clin Rheumatol 1994; 8(3): 661-90. Hazes, 1994 Hazes JMW, Cats A. In: Klippel JH, Dieppe PA, editors. Rheumatology. London: Mosby; 1994. p. 3: 14.1-14.8. Healey, 1986 Healey LA. Rheumatoid arthritis in the elderly. Clin Rheum Dis 1986; 12(1): 173-9. Hernández-García, 2000 Hernández-García C, Vargas E, Abasolo L, Lajas C, Bellarjdell B, Morado IC, Macarrón P, Pato E, Fernández-Gutiérrez B, Bañares A, Jover JA. Long-time between a set of symptoms and access to rheumatology care and DMARDs therapy in a cohort of patients with rheumatoid arthritis. J Rheumatol 2000; 27: 2323-28. Hernández-García, 2001 Hernández-García C, González-Alvaro I, Villaverde V, Vargas E, Jover JA, Lázaro P y el Grupo de Estudio emAR. Estudio sobre el manejo de la rheumatoid arthritis en España. Sociedad Española de Reumatología. Madrid, 2001. Hickling, 1998 Hickling P, Jacoby RK, Kirwan JR. Joint destruction after glucocorticoids are withdrawn in early rheumatoid arthritis. Arthritis and Rheumatism Council Low Dose Glucocorticoid Study Group. Br J Rheumatol 1998; 37(9): 930-6. Hilsden, 1995 Hilsden RJ, Urbanski SJ, Swain MG. End-stage liver disease developing with the use of methotrexate in heterozygous alfa1-antitrypsin deficiency. Arthritis Rheum 1995; 38(7): 1014-8. Hoffman, 1992 Hoffman GS, Kerr GS, Leavitt RY, Hallahan CW, Lebovics RS, Travis WD, et al. Wegener granulomatosis: an analysis of 158 patients. Ann Intern Med 1992; 116(6): 488-98. Howard, 1986 Howard Lock HE, Lock CJ, Mewa A, Kean WF. D-penicillamine: chemistry and clinical use in rheumatic disease. Semin Arthritis Rheum 1986; 15(4): 261-81. Hows, 1984 Hows JM, Mehta A, Ward L, Woods K, Perez R, Gordon MY, et al. Comparison of mesna with forced diuresis to prevent cyclophosphamide induced haemorrhagic cystitis in marrow transplantation: a prospective randomised study. Br J Cancer 1984; 50(6): 753-6. Hulsmans, 2000 Hulsmans HM, Jacobs JW, van der Heijde DM, van Albada-Kuipers GA, Schenk Y, Bijlsma JW. The course of radiologic damage during the first six years of rheumatoid arthritis. Arthritis Rheum 2000; 43(9): 1927-40. Hurwitz, 1969 Hurwitz N. Predisposing factors in adverse reactions to drugs. Br Med J 1969; 1(643): 536-9. Husby, 1985 Husby G. Amyloidosis and rheumatoid arthritis. Clin Exp Rheumatol 1985; 3(2): 173-80. Hutter, 1969 Hutter AM Jr, Bauman AW, Frank IN. Cyclophosphamide and severe hemorrhagic cystitis. N Y State J Med 1969; 69(2): 305-9. IOM, 1990a Field MJ, Lohr KN, editors. Clinical Practice Guidelines: Directions for a New Program. Washington, D.C.: National Academy Press; 1990. IOM, 1990b Lohr KN, editor. Medicare: A strategy for quality assurance. Washington, D.C.: National Academy Press; 1990. IOM, 1992 Field MJ, Lohr KN, editors. Guidelines for Clinical Practice: from Development to Use. Washington, D.C.: National Academy Press; 1992. Jack, 1981 Jack MK, Hicks JD. Ocular complications in high-dose chemotherapy and marrow transplantation. Ann Ophtalmol 1981; 13(6): 709-11. Jadad, 1996a Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, McQuay HJ. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials 1996;17(1):1-12. Jadad, 1996b Jadad AR, McQuay HJ. Meta-analyses to evaluate analgesic interventions: a systematic qualitative review of their methodology. Clin Epidemil 1996; 49(2): 235-43. Jaffe, 1977 Jaffe IA. D-penicillamine. Bull Rheum Dis 1977-78; 28(6): 948-52. Jahangier, 1997 Jahangier ZN, Jacobs JW, van Isselt JW, Bijlsma JW. Persistent synovitis treated with radiation synovectomy using yttrium-90: a retrospective evaluation of 83 procedures for 45 patients. Br J Rheumatol 1997; 36(8): 861-9. Jaison, 1970 Jaison MI, Rubenstein D, Dixon AS. Intra-articular pressure and rheumatoid geodes (bone 'cysts'). Ann Rheum Dis 1970; 2985): 496-502. Jantti, 1999 Jantti J, Aho K, Kaarela K, Kautiainen H. Work disability in an inception cohort of patients with seropositive rheumatoid arthritis: a 20 year study. Rheumatology 1999; 38(11): 1138-41. Johnson, 1971 Johnson WW, Meadows DC. Urinary-bladder fibrosis and telangiectasia associated with long-term cyclophosphamide therapy. N Engl J Med 1971; 284(6): 290-4. Jones, 1993 Jones G. Yttrium synovectomy: a meta-analysis of the literature. Aust N Z J Med 1993; 23(3): 272-5. Jurik, 1982 Jurik AG, Davidsen D, Graudal H. Prevalence of pulmonary involvement in rheumatoid arthritis and its relationship to some characteristics of the patients: a radiological and clinical study. Scand J Rheumatol 1982; 11(4): 217-24. Jurik, 1986 Jurik AG, Graudal H. Pericarditis in rheumatoid arthritis: a clinical and radiological study. Rheumatol Int 1986; 6(1): 37-42. Kahn, 1990 Kahn KL, Rogers WH, Rubenstein LV, Sherwood MJ, Reinish EJ, Keeler EB, et al. Measuring quality of care with explicit process criteria before and after implementation of the DRG-based prospective payment system. JAMA 1990; 264(15): 1969-73. Kamel, 1993 Kamel OW, van de Rijn M, Weiss LM, Del Zoppo GJ, Hench PK, Robbins BA, et al. Brief report: reversible lymphomas associated with Epstein-Barr virus occurring during methotrexate therapy for rheumatoid arthritis and dermatomyositis. N Engl J Med 1993; 328(18): 1317-21. Kanik, 1997 Kanik KS, Cash JM. Does methotrexate increase the risk of infection or malignancy? Rheum Dis Clin North Am 1997; 23(4): 955-67. Kaplinsky, 1978 Kaplinsky N, Frankl O. Salicylazosulphapyridine-induced Heinz body anemia. Acta Haematol 1978; 59(5): 310-4. Karam, 1994 Karam NE, Roger L, Hankins LL, Reveille JD. Rheumatoid nodulosis of the meninges. J Rheumatol 1994; 21(10): 1960-3. Kassirer, 1993 Kassirer JP. The quality of care and the quality of measuring it. N Engl J Med 1993; 329(17): 1263-5. Kattwinkel, 1991 Kattwinkel N, Cook L, Agnello V. Overwhelming fatal infection in a young woman after intravenous cyclophosphamide therapy for lupus nephritis. J Rheumatol 1991; 18(1): 79-81. Kaye, 1987 Kaye JJ, Callahan LF, Nance EP Jr, Brooks RH, Pincus T. Rheumatoid arthritis: explanatory power of specific radiographic findings for patient clinical status. Radiology 1987; 165(3): 753-8. Keeler, 1992 Keeler EB, Rubenstein LV, Kahn KL, Draper D, Harrison ER, McGinty MJ, et al. Hospital characteristics and quality of care. JAMA 1992; 268(13): 1709-14. Kelly, 1993 Kelly CA. Rheumatoid arthritis: other rheumatoid lung problems. Baillieres Clin Rheumatol 1993; 7(1): 17-29. Kende, 1979 Kende G, Sirkin S, Thomas PR, Freeman AI. Blurring of vision: a previously undescribed complication of cyclophosphamide therapy. Cancer 1979; 44(1): 69-71. Kerstens, 1992 Kerstens PJ, Boerbooms AMT, Jeurissen ME, Fast JH, Assmann KJ, van de Putte LB. Accelerated nodulosis during low dose methotrexate therapy for rheumatoid arthritis: an analysis of ten cases. J Rheumatol 1992; 19(6): 867-71. Kingsmore, 1992 Kingsmore SF, Hall BD, Allen NB, Rice JR, Caldwell DS. Association of methotrexate, rheumatoid arthritis and lymphoma: report of two cases and literature review. J Rheumatol 1992; 19(9): 1462-5. Kirsner, 1982 Kirsner AB, Farber SJ, Sheon RP, Finkel RI. The incidence malignant disease in patients receiving cytotoxic therapy for rheumatoid arthritis. Ann Rheum Dis 1982; 41(Suppl 1): 32-3. Kirwan, 1995 Kirwan R. The effect of glucocorticoids on joint destruction in rheumatoid arthritis. The Arthritis and Rheumatism Council Low-Dose Glucocorticoid Study Group. N Engl J Med 1995; 333(3): 142-6. Klintmalm, 1981 Klintmalm GB, Iwatsuki S, Starzl TE. Cyclosporin A hepatotoxicity in 66 renal allograft recipients. Transplantation 1981; 32(6): 488-9. Klippel, 1987a Klippel JH, Austin HA 3d, Balow JE, LeRiche NG, Steinberg AD, Plotz PH, et al. Studies of immunosuppressive drugs in the treatment of lupus nephritis. Rheum Dis Clin North Am 1987; 13(1): 47-56. Klippel, 1987b Klippel JH. Immunosuppressive therapy. In: Lahita RG, editor. Systemic lupus erythematosus. New York: John Wiley & Sons; 1987. p. 923-45. Klippel, 1998 Klippel JH. Indications for, and use of, cytotoxic agents in SLE. Baillieres Clin Rheumatol 1998; 12(3): 511-27. Knysak, 1994 Knysak DJ, McLean JA, Solomon WR, Fox DA, McCune WJ. Immediate hypersensitivity reaction to cyclophosphamide. Arthritis Rheum 1994; 37(7): 1101-4. Koopman, 1998 Koopman WJ, Moreland LW. Rheumatoid arthritis: anticytokine therapies on the horizon. Ann Int Med 1998; 128(3): 231-3. Kovarsky, 1983 Kovarsky J. Clinical pharmacology and toxicology of cyclophosphamide: emphasis on use in rheumatic diseases. Semin Arthritis Rheum 1983; 12(4): 359-72. Kraaimaat, 1995 Kraaimaat FW, Van Dam-Baggen RM, Bijlsma JW. Association of social support and the spouse's reaction with psychological distress in male and female patients with rheumatoid arthritis. J Rheumatol 1995; 22(4): 644-8. Kremer, 1992 Kremer JM, Koff R. A debate: should patients with rheumatoid arthritis on methotrexate undergo lever biopsies? Semin Arthritis Rheum 1992; 21(6): 376-86. Kremer, 1994 Kremer JM, Alarcon GS, Lightfoot RW Jr, Willkens RF, Furst DE, Williams HJ, et al. Methotrexate for rheumatoid arthritis. Suggested guidelines for monitoring liver toxicity. Arthritis Rheum 1994; 37(3): 316-28. Kremer, 1995 Kremer JM, Alarcon GS, Alexander RW, Cannon GW, Weinblatt ME, et al. Methotrexate (MTX)-lung toxicity: clinical features of thirty-seven cases from seven rheumatology practices. Arthritis Rheum 1995; 38: S205. Kyle, 1983. Kyle RA, Greipp PR. Amyloidosis (AL): clinical and laboratory features in 229 cases. Mayo Clin Proc 1983; 58(10): 665-83. Lajas, 2000 Lajas C, Bellajdel B, Abásolo L, Salido M, Hernández García C, Fernández B, et al. El coste anual de la artritis reumatoide. Rev Esp Reumatol 2000; 27(5): 206. Land, 1988 Land W. Cyclosporin in cadaveric renal transplantation: five year follow-up results of the European Multicentre Trial. Transplant Proc 1988; 20(3 Suppl 3): 73-7. Landewé, 1994 Landewé RBM, Goei The HS, van Rijthoven AWAN, Breedveld FC, Dijkmans BA. A randomized, double blind, 24 week controlled study of low-dose cyclosporin versus chloroquine for early rheumatoid arthritis. Arthritis Rheum 1994; 37(5): 637-43. Landewé, 1996 Landewé RB, Dijkmans BA, van der Woude FJ, Breedveld FC, Mihatsch MJ, Bruijn JA. Longterm low-dose cyclosporin in patients with rheumatoid arthritis: renal function loss without structural nephropathy. J Rheumatol 1996; 23(1): 61-4. Langevitz, 1992 Langevitz P, Klein L, Pras M, Many A. The effect of cyclophosphamide pulses on fertility in patients with lupus nephritis. Am J Reprod Immunol 1992; 28(3-4): 157-8. Langley, 1984 Langley GB, Sheppeard H. Problems associated with pain measurement in arthritis: comparison of the visual analogue and verbal rating scales. Clin Exp Rheumatol 1984; 2(3): 231-4. Langman, 1999 Langman MJ, Jensen DM, Watson DJ, Harper SE, Zhao PL, Quan H, et al. Adverse upper gastrointestinal effects of rofecoxib compared to other NSAIDs. JAMA 1999; 282(20): 1929-33. Larsen, 1977 Larsen A, Dale K, Eek M. Radiographic evaluation of rheumatoid arthritis and related conditions by standard reference films. Acta Radiol Diagn Stockh 1977; 18(4): 481-91. Larsen, 1995 Larsen A. How to apply Larsen score in evaluating radiographs of rheumatoid arthritis in longterm studies. J Rheumatol 1995; 22(10): 1974-5. Lebowitz, 1963 Lebowitz WB. The heart in rheumatoid arthritis (rheumatoid disease): a clinical and pathological study of sixty-two cases. Ann Intern Med 1963; 58: 102-23. Lehmann, 1966 Lehmann JF, Silverman DR, Baum BA, Kirk NL, Johnston VC. Temperature distributions in the human thigh, produced by infrared, hot pack and microwave applications. Arch Phys Med Rehab 1966; 47(5): 291-9. Lin, 1989 Lin HY, Rocher LL, McQuillan MA, Schmaltz S; Palella TD, Fox IH. Cyclosporin-induced hyperuricemia and gout. N Engl J Med 1989; 321(5): 287-92. Livneh, 1994 Livneh A, Zemer D, Langevitz P, Laor A, Sohar E, Pras M. Colchicine treatment of AA amyloidosis of familial Mediterranean fever: an analysis of factors affecting outcome. Arthritis Rheum 1994; 37(12): 1804-11. LMSSG, 1999 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 1999; 53(3): 457-65. Lockie, 1985 Lockie LM, Smith DM. Forty-seven years experience with gold therapy in 1019 rheumatoid arthritis patients. Semin Arthritis Rheum 1985; 14(4): 238-46. Lohr, 1990 Lohr KN, Rettig RA. Quality of care and technology assessment: report of a forum of the Council on Health Care Technology. Institute of Medicine. Washington, DC: National Academy Press, 1990. Losek, 1981 Losek JD, Werlin SL. Sulphasalazine hepatotoxicity. Am J Dis Child 1981; 135(11): 1070-2. Lovat, 1997 Lovat LB, Madhoo S, Pepys MB, Hawkins PN. Long-term survival in systemic amyloid A amyloidosis complicating Crohn's disease. Gastroenterology 1997; 112(4): 1362-5. Love, 1975 Love RR, Sowa JM. Myelomonocytic leukemia following cyclophosphamide therapy of rheumatoid disease. Ann Rheum Dis 1975; 34(6): 534-5. Lund, 1983 Lund HI, Nielsen M. Penicillamine-induced dermatomyositis: a case history. Scand J Rheumatol 1983; 12(4): 350-2. Maddux, 1986 Maddux MS, Frigo LC, Veremis SA, Pollak R, Mozes MF. Cyclosporin hepatotoxicity following renal transplantation. Clin Pharmacol Ther 1986; 39: 208. Maini, 1998 Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, McFarlane JD, et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum 1998; 41(9): 1552-63. Maini, 1999 Maini R, St Clair EW, Breedveld F, Furst D, Kalden J, Weisman M, et al. Infliximab (chimeric anti-tumour necrosis factor alpha monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial. Lancet 1999; 354(9194): 1932-9. Maksymowych, 1987 Maksymowych W, Russell AS. Antimalarials in rheumatology: efficacy and safety. Semin Arthritis Rheum 1987; 16(3): 206-21. Marinos, 1992 Marinos G, Riley J, Painter DM, McCaughan GW. Sulphasalazine-induced fulminant hepatic failure. J Clin Gastroenterol 1992; 14(2): 132-5. Masala, 1997 Masala A, Faedda R, Alagna S, Satta A, Chiarelli G, Rovasio PP, et al. Use of testosterone to prevent cyclophosphamide-induced azoospermia. Ann Intern Med 1997; 126(4): 292-5. McCarty, 1995 McCarty DJ, Harman JG, Grassanovich JL, Qian C, Klein JP. Combination drug therapy of seropositive rheumatoid arthritis. J Rheumatol 1995; 22:1636-45. McKendry, 1993 McKendry RJ, Dale P. Adverse effects of low dose methotrexate therapy in rheumatoid arthritis. J Rheumatol 1993; 20(11): 1850-6. McKendry, 1997 McKendry RJ. The remarkable spectrum of methotrexate toxicities. Rheum Dis Clin North Am 1997; 23(4): 939-54. McMahon, 1993 McMahon MJ, Swinson DR, Shettar S, Wolstenholme R, Chattopadhyay C, Smith P, et al. Bronchiectasis and rheumatoid arthritis: a clinical study. Ann Rheum Dis 1993; 52(11): 776-9. Medrano, 1986 Medrano San Ildefonso M, Mauri Llerda JA, Calabozo Raluy M, Barrio J, Orte Martínez J, Beltrán Gutiérrez J. Panarteritis nodosa: fibrosis pulmonar después del tratamiento con ciclofosfamida. Rev Esp Reumatol. 1986; 13(4): 130-2. Merritt, 1983 Merritt JL, Hunder GG. Passive range of motion, not isometric exercise, amplifies acute urate synovitis. Arch Phys Med Rehab 1983; 64(3): 130-1. Mihats, 1989 Mihats MJ, Thiel G, Ryffel, B Cyclosporin A: action and side effects. Toxico Lett. 1989; 46: 125-139. Miller, 1971 Miller JJ 3d, Williams GF, Leissring JC. Multiple late complications of therapy with cyclophosphamide, including ovarian destruction. Am J Med 1971; 50(4): 530-5. Miller, 1993 Miller ST, Flanagan E. The transition from quality assurance to continuous quality improvement in ambulatory care. QRB Qual Rev Bull 1993; 19(2): 62-5. Minor, 1989 Minor MA, Hewett JE, Webel RR, Anderson SK, Kay DR. Efficacy of physical conditioning in patients with rheumatoid arthritis and osteoarthritis. Arthritis Rheum 1989; 32(11): 1396-405. Mladenovic, 1995 Mladenovic V, Domljan Z, Rozman B, Jajic I, Mihajlovic D, Dordevic J, et al. Safety and effectiveness of leflunomide in the treatment of patients with active rheumatoid arthritis: results of a randomized, placebo-controlled, phase II study. Arthritis Rheum 1995; 38(11): 1595-603. Moder, 1995 Moder KG, Tefferi A, Cohen MD, Menke DM, Luthra HS. Hematologic malignancies and the use of methotrexate in rheumatoid arthritis: a retrospective study. Am J Med 1995; 99(3): 276-81. Moreland, 1997 Moreland LW, Baumgartner SW, Schiff MH, Tindall EA, Fliischmann RM, Weaver AL, et al. Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)-Fc fusion protein. N Engl J Med 1997; 337(3): 141-7. Moreland, 1999 Moreland LW, Schiff MH, Baumgartner SW, Tindall EA, Fleischmann RM, Bulpitt KJ, et al. Etanercept therapy in rheumatoid arthritis: a randomized, controlled trial. Ann Intern Med 1999; 130(6): 478-86. Morgan, 1986 Morgan J, Furst DE. Implications of drug therapy in the elderly. Clin Rheum Dis 1986; 12(1): 227-44. Morgan, 1993 Morgan SL, Baggott JE, Alarcon GS. Methotrexate and sulfasalazine combination therapy: is it worth the risk? Arthritis Rheum 1993; 36(2): 281-2. Morris, 1988 Morris PJ. Cyclosporin. In: Morris PJ, editor. Kidney Transplantation: Principles and Practice. (3 ed). Philadelphia: Harcourt Brace Jovanovich; 1988. p. 285-317. Moutsopoulos, 1978 Moutsopoulos HM, Gallagher JD, Decker JL, Steinberg AD. Herpes zoster in patients with systemic lupus erythematosus Arthritis Rheum 1978; 21(7): 789-802. Muller, 1975 Muller W, Pavelka K, Fridrich R. Treatment of chronic articular effusions with 90-yttrium (90Y). Scand J Rheumatol 1975; 4(4): 216-20. Munro, 1997 Munro R, Capell HA. Penicillamine. Br J Rheumatol 1997; 36(1): 104-9. Myochrysine, 1997 Prod Info Myochrysine® 1997. Naides, 1995 Naides SJ. Acute parvovirus B19-induced pancytopenia in the setting of methotrexate therapy for rheumatoid arthritis. Arthritis Rheum 1995; 38(7): 1023. Nashel, 1985 Nashel DJ. Mechanisms of action and clinical applications of cytotoxic drugs in rheumatic disorders. Med Clin North Am 1985; 69(4): 817-40. Neiman, 1985 Neiman RA, Fye KH. Methotrexate induced false photosensitivity reaction. J Rheumatol 1985; 12(2): 354-5. Nicholas, 1988 Nicholas NS, Panayi GS. Rheumatoid arthritis and pregnancy. Clin Exp Rheumatol 1988; 6(2): 179-82. Ntoso, 1986 Ntoso KA, Tomaszewski JE, Jimenez SA, Neilson EG. Penicillamine-induced rapidly progressive glomerulonephritis in patients with progressive systemic sclerosis: successful treatment of two patients and a review of the literature. Am J Kidney Dis 1986; 8(3): 159-63. O´Callaghan, 1986 O'Callaghan JW, Brooks PM. Disease-modifying agents and immunosuppressive drugs in the elderly. Clin Rheum Dis 1986; 12(1): 275-89. O´Dell, 1996 O'Dell JR, Haire CE, Erikson N, Drymalski W, Palmer W, Eckhoff PJ, et al. Treatment of rheumatoid arthritis with methotrexate alone, sulfasalazine and hydroxychloroquine, or a combination of all three medications. N Engl J Med 1996; 334(20): 1287-91. O´Dell, 1997 O'Dell JR. Methotrexate use in rheumatoid arthritis. Rheum Dis Clin North Am 1997; 23(4): 779-96. O´Duffy, 1999 O'Duffy EK, Clunie GP, Lui D, Edwards JC, Ell PJ. Double blind glucocorticoid controlled trial of samarium-153 particulate hydroxyapatite radiation synovectomy for chronic knee synovitis. Ann Rheum Dis 1999; 58(9): 554-8. Oates, 1998 Oates JA, Wilkinson GR. Principles of drug therapy. In: Harrison's Principles of Internal Medicine (14th ed.). New York: McGraw-Hill; 1998. p. 411-21. Ognibene, 1995 Ognibene FP, Shelhamer JH, Hoffman GS, Kerr GS, Reda D, Fauci AS, et al. Pneumocystis carinii pneumonia: a major complication of immunosuppressive therapy in patients with Wegener's granulomatosis. Am J Resp Critical Care Med 1995; 151(3 Pt 1): 795-9. Okuda, 1994 Okuda Y, Takasugi K, Oyama T, Onuma M, Oyama H. Amyloidosis in rheumatoid arthritis clinical study of 124 histologically proven cases. Ryumachi 1994; 34(6): 939-46. Olson, 1972 Olson JE, Stravino VD. A review of cryotherapy. Phys Ther 1972; 52(8): 840-53. OMERACT, 1993 OMERACT, Conference on Outcome Measures in Rheumatoid Arthritis Clinical Trials. Proceedings. Maastricht, The Netherlands, April 29-May 3, 1992. J Rheumatol 1993; 20(3): 527-91. OMERACT, 1994 Reduced joint counts in rheumatoid arthritis clinical trials. American College of Rheumatology Committee on Outcome Measures in Rheumatoid Arthritis Clinical Trials. Arthritis Rheum 1994; 37(4): 463-4. Onrust, 1998 Onrust SV, Lamb HM. Infliximab: a review of its use in Crohn's disease and rheumatoid arthritis. BioDrugs 1998; 10(5): 397-422. OTA, 1988 Office of Technology Assessment. The Quality of Medical Care: Information for Consumers. Washington, DC: U.S. Government Printing Office; 1988 June. Report Nº: OTA-H-386. Oxman, 1991a Oxman AD, Guyatt GH. Validation of an index of the quality of review articles. J Clin Epidemiol 1991; 44: 1271-8. Oxman, 1991b Oxman AD, Guyatt GH, Singer J, Goldsmith CH, Hutchison BG, Milner RA, Streiner DL. Agreement among reviewers of review articles. J Clin Epidemiol 1991; 44: 91-8. Pai, 1993 Pai S, Helin H, Isomaki H. Frequency of amyloidosis in Estonian patients with rheumatoid arthritis. Scand J Rheumatol 1993; 22(5): 248-9. Paimela, 1992 Paimela L. The radiographic criterion in the 1987 revised criteria for rheumatoid arthritis: reassessment in a prospective study of early disease. Arthritis Rheum 1992; 35(3): 255-8. Panayi, 1993 Panayi Gs, Tugwell P. An international consensus report: the use of cyclosporin A in rheumatoid arthritis. Br J Rheumatol 1993; 32(Suppl 1): 1-3. Panayi, 1994 Panayi GS, Tugwell P. The use of cyclosporin A in rheumatoid arthritis: conclusions of an international review. Br J Rheumatol 1994; 33(10): 967-9. Patel, 1976 Patel AR, Shah PC, Rhee HL, Sassoon H, Rao KP. Cyclophosphamide therapy and interstitial pulmonary fibrosis. Cancer 1976; 38(4): 1542-9. Paulus, 1984 Paulus HE, Williams HJ, Ward JR, Reading JC, Egger MJ, Coleman ML, et al. Azathioprine versus D-Penicillamine in rheumatoid arthritis patients who have been treated unsuccessfully with gold. Arthritis Rheum 1984; 27(7): 721-7. Paulus, 1990 Paulus HE, Egger MJ, Ward JR, Williams HJ. Analysis of improvement in individual rheumatoid arthritis patients treated with disease-modifying antirheumatic drugs, based on the findings in patients treated with placebo. The Cooperative Systematic Studies of Rheumatic Diseases Group. Arthritis Rheum 1990; 33(4): 477-84. Paulus, 1999 Paulus HE, Ramos B, Wong WK, Ahmed A, Bulpitt K, Park G, et al. Equivalence of the acute phase reactants C-reactive protein, plasma viscosity, and Westergren erythrocyte sedimentation rate when used to calculate American College of Rheumatology 20% improvement criteria or the disease activity score in patients with early rheumatoid arthritis. J Rheumatol 1999; 26(11): 2324-31. Pears, 1989 Pears JS, Morley KD. Fatal hypersensitivity reaction to sulphasalazine. Br J Rheumatol 1989; 28(3): 274-5. Pease, 1999 Pease CT, Bhakta BB, Devlin J, Emery P. Does the age of onset of rheumatoid arthritis influence phenotype?: a prospective study of outcome and prognostic factors. Rheumatology 1999; 38(3): 228-34. Pedersen-Bjergaard, 1985 Pedersen-Bjergaard J, Ersboll J, Sorensen HM, Keiding N, Larsen SO, Philip P, et al. Risk of acute nonlymphocytic leukemia and preleukemia in patients treated with cyclophosphamide for non-Hodgkin's lymphomas: comparison with results obtained in patients treated for Hodgkin's disease and ovarian carcinoma with other alkylating agents. Ann Intern Med 1985; 103(2): 195-200. Pedersen-Bjergaard, 1988 Pedersen Bjergaard J, Ersboll J, Hansen VL, Sorensen BL, Christoffersen K, Hou Jensen K, et al. Carcinoma of the urinary bladder after treatment with cyclophosphamide for non-Hodgkin´s lymphoma. N Engl J Med 1988; 318(16): 1028-32. Penn, 1981 Penn I. Depressed immunity and the development of cancer. Clin Exp Immunol 1981; 46(3): 459-74. Pinals, 1981 Pinals RS, Masi AT, Larsen RA. Preliminary criteria for clinical remission in rheumatoid arthritis. Arthritis Rheum 1981; 24(10): 1308-15. Pincus, 1983 Pincus T, Summey JA, Soraci SA Jr, Wallston KA, Hummon NP. Assessment of patient satisfaction in activities of daily living using a modified Stanford Health Assessment Questionnaire. Arthritis Rheum 1983; 26(11): 1346- 53. Pincus, 1985 Pincus T, Callahan LF. Formal education as a marker for increased mortality and morbidity in rheumatoid arthritis. J Chronic Dis 1985; 38(12): 973-84. Pincus, 1995 Pincus T, Callahan LF, Fuch HA, Larsen A, Kaye J. Quantitative analysis of hand radiographs in rheumatoid arthritis: time course of radiographic changes, relation to joint examination measures, and comparison of different scoring methods. J Rheumatol 1995; 22(10): 1983-9. Pincus, 1996 Pincus T. Documenting quality management in rheumatic disease: are patient questionnaires the best (and only) method? Arthritis Care Res 1996; 9(5): 339-48. Plotz, 1979 Plotz PH, Klippel JH, Decker JL, Grauman D, Wolff B, Brown BC, et al. Bladder complications in patients receiving cyclophosphamide for systemic lupus erythematosus or rheumatoid arthritis. Ann Intern Med 1979; 91(2): 221-3. Prevoo, 1995 Prevoo ML, van't Hof MA, Kuper HH, van Leeuwen MA, van de Putte BA, van Riel PL. Modified disease activity scores that include twenty-eight-joint counts: development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum 1995; 38(1): 44-8. Prevoo, 1996 Prevoo ML, van Gestel AM, van't Hof MA, van Rijswijk MH, van de Putte LB, van Riel PL. Remission in a prospective study of patients with rheumatoid arthritis. American Rheumatism Association preliminary remission criteria in relation to the disease activity score. Br J Rheumatol 1996; 35(11): 1101-5. Pryor, 1996 Pryor BD, Bologna SG, Kahl LE. Risk factors for serious infection during treatment with cyclophosphamide and high-dose corticosteroids for systemic lupus erythematosus. Arthritis Rheum 1996; 39(9): 1475-82. RACTAG, 1995 The effect of age and renal function on the efficacy and toxicity of methotrexate in rheumatoid arthritis. Rheumatoid Arthritis Clinical Trial Archive Group. J Rheumatol 1995; 22(2): 218-23. Radis, 1995 Radis CD, Kahl LE, Baker GL, Wasko MC, Cash JM, Gallatin A, et al. Effects of cyclophosphamide on the development of malignancy and on long-term survival of patients with rheumatoid arthritis: a 20-years follow-up study. Arthritis Rheum 1995; 38(8): 1120-7. Ramsey-Goldman, 1997 Ramsey-Goldman R, Schilling E. Immunosuppressive drug use during pregnancy. Rheum Dis Clin North Am 1997; 23(1): 149-67. Rees, 1991 Rees JH, Woodhead MA, Sheppard MN, Du Bois RM. Rheumatoid arthritis and cryptogenic organising pneumonitis. Respir Med 1991; 85(3): 243-6. Reisine, 1998 Reisine S, Fifield J, Winkelman DK. Employment patterns and their effect on health outcomes among women with rheumatoid arthritis followed for 7 years. J Rheumatol 1998; 25(10): 1908-16. Remicade, 1999M Prod Inf Remicade (TM) 1999. Información sobre el producto que ofrece el propio laboratorio en una página disponible en Internet. Ridaura, 1995 Prod Info Ridaura® 1995. Riemsma, 1998 Riemsma RP, Rasker JJ, Taal E, Griep EN, Wouters JM, Wiegman O. Fatigue in rheumatoid arthritis: the role of self-efficacy and problematic social support. Br J Rheumatol 1998; 37(10): 1042-6. Ritchie, 1968 Ritchie DM, Boyle JA, McInnes JM, Jasani MK, Dalakos TG, Grieveson P, et al. Clinical studies with an articular index for the assessment of joint tenderness in patients with rheumatoid arthritis. Q J Med 1968; 37(147): 393-406. Robbins, 1980 Robbins G, McIllmurray MB. Acute renal failure due to gold. Postgrad Med J 1980 56(655): 366-7. Rodríguez, 1996 Rodríguez F, Krayenbuhl JC, Harrison WB, Forre O, Dijkmans BA, Tugwell P, et al. Renal biopsy findings and follow-up of renal function in rheumatoid arthritis patients treated with cyclosporin A. An update from the International Kidney Biopsy Registry. Arthritis Rheum 1996; 39(9): 1491-8. Rodríguez, 1998 Rodríguez de la Serna A. Fármacos inmunosupresores e inmunoreguladores. In: Pascual Gómez E, Rodríguez Valverde V, Carbonell Abelló J, Gómez-Reino Carnota JJ, editores. Tratado de Reumatología. Madrid: Arán Ediciones; 1998. p. 2393-2403. Roubenoff, 1988 Roubenoff R, Hoyt J, Petri M, Hochberg MC, Hellmann DB. Effects of antiinflammatory and immunosuppressive drugs on pregnancy and fertility. Semin Arthritis Rheum 1988; 18(2): 88-110. Rubenstein, 1990 Rubenstein LV, Kahn KL, Reinisch EJ, Sherwood MJ, Rogers WH, Kamberg C, et al. Changes in quality of care for five diseases measured by implicit review, 1981 to 1986. JAMA 1990; 264(15): 1974-9. Ruymann, 1977 Ruymann FB, Mosijczuk AD, Sayers RJ. Hepatoma in a child with methotrexate-induced hepatic fibrosis. JAMA 1977; 238(24): 2631-3. Rynes, 1983 Rynes RI. Ophthalmologic safety of long-term hydroxychloroquine sulfate treatment. Am J Med 1983; 75(Suppl 1A): 35-9. Saag, 1997 Saag KG. Low-dose corticosteroid therapy in rheumatoid arthritis: balancing the evidence. Am J Med 1997; 103(6A): 31S-39S. Scott, 1989 Scott DL, Dawes PT, Tunn E, et al. Combination therapy with gold and hydroxychloroquine in rheumatoid arthritis: a prospective, randomized, placebo-controlled study. Br J Rheumatol 1989; 28:128-33. Scott, 1992 Scott DL, Panayi GS, van Riel PL, Smolen J, van de Putte LB. Disease activity in rheumatoid arthritis: preliminary report of the Consensus Study Group of the European Workshop for Rheumatology Research. Clin Exp Rheumatol 1992; 10(5): 521-5. Scott, 1993 Scott DL. A simple index to assess disease activity in rheumatoid arthritis. J Rheumatol 1993; 20(3): 582-4. Scott, 2000 Scott DL, Pugner K, Kaarela K, Doyle DV, Woolf A, Holmes J, et al. The links between joint damage and disability in rheumatoid arthritis. Rheumatology 2000; 39(2): 122-32. Scharloo, 1999 Scharloo M, Kaptein AA, Weinman JA, Hazes JM, Breedveld FC, Rooijmans HG. Predicting functional status in patients with rheumatoid arthritis. J Rheumatol 1999; 26(8): 1686-93. Schilsky, 1980 Schilsky RL, Lewis BJ, Sherins RJ, Young RC. Gonadal dysfunction in patients receiving chemotherapy for cancer. Ann Intern Med 1980; 93(1): 109-14. Schnitzer, 1999 Schnitzer TJ, Truitt K, Fleischmann R, Dalgin P, Block J, Zeng Q, et al. The safety profile, tolerability, and effective dose range of rofecoxib in the treatment of rheumatoid arthritis: Phase II Rofecoxib Rheumatoid Arthritis Study Group. Clin Ther 1999; 21(10): 1688-702. Schur, 1999 Schur PH, Helfgott SM. Evaluation and medical management of end-stage rheumatoid arthritis. UpToDate 1999 [CD-ROM]. version 7.3. Segal, 1988 Segal R, Caspi D, Tishler M, Fishel B, Yaron M. Accelerated nodulosis and vasculitis during methotrexate therapy for rheumatoid arthritis. Arthritis Rheum 1988; 31(9): 1182-5. SER, 2000a Sociedad Española de Reumatología, Sociedad Española de Medicina Familiar y Comunitaria, Instituto de Salud Carlos III, Dirección General de Farmacia y Productos Sanitarios. Uso racional de coxibs. Rev Esp Reumatol 2000; 27: 398-400. SER, 2000b Comité de Expertos de la Sociedad Española de Reumatología. Consenso de la Sociedad Española de Reumatología sobre la terapia con inhibidores del TNF y otros fármacos inductores de remisión en la artritis reumatoide. Rev Esp Reumatol 2000; 27(8): 352-4. Searles, 1987 Searles G, McKendry RJ. Methotrexate pneumonitis in rheumatoid arthritis: potential risk factors: four case reports and a review of the literature. J Rheumatol 1987; 14(6): 1164-71. Shadick. 1994 Shadick NA, Fanta CH, Weinblatt ME, O'Donnell W, Coblyn JS. Bronchiectasis: a late feature of severe rheumatoid arthritis. Medicine (Baltimore) 1994; 73(3): 161-70. Sharp, 1971 Sharp JT, Lidsky MD, Collins LC, Moreland J. Methods of scoring the progression of radiologic changes in rheumatoid arthritis: correlation of radiologic, clinical and laboratory abnormalities. Arthritis Rheum 1971; 14(6): 706-20. Sharp, 1985 Sharp JT, Young DY, Bluhm GB, Brook A, Brower AC, Corbett M, et al. How many joints in the hands and wrists should be included in a score of radiologic abnormalities used to assess rheumatoid arthritis? Arthritis Rheum 1985; 28(12): 1326-35. Sharp, 1995 Sharp JT. Assessment of radiographic abnormalities in rheumatoid arthritis: what have we accomplished and where should we go from here? J Rheumatol 1995; 22(9): 1787-91. Sharp, 2000 Sharp JT, Strand V, Leung H, Hurley F, Loew-Friedrich I. Treatment with leflunomide slows radiographic progression of rheumatoid arthritis: results from three randomized controlled trials of leflunomide in patients with active rheumatoid arthritis. Leflunomide Rheumatoid Arthritis Investigators Group. Arthritis Rheum 2000; 43(3): 495-505. Sherins, 1973 Sherins RJ, De Vita VT. Effect of drug treatment for lymphoma on male reproductive capacity: studies of men in remission after therapy. Ann Intern Med. 1973; 79(2): 216-20. Shiroky, 1991 Shiroky JB, Frost A, Skelton JD, Haegert DG, Newkirk MM, Neville C. Complications of immunosuppression associated with weekly low-dose methotrexate. J Rheumatol 1991; 18(8): 1172-5. Silman, 1988 Silman AJ, Petrie J, Hazleman B, Evans SJ. Lymphoproliferative cancer and other malignancy in patients with rheumatoid arthritis treated with azathioprine: a 20 year follow-up study. Ann Rheum Dis 1988; 47(12): 988-92. Silman, 1993 Silman AJ. Rheumatoid arthritis. In: Silman AJ, Hochberg MC, editors. Epidemiology of the rheumatic diseases. Oxford: Oxford University Press; 1993. Simon, 1999 Simon LS, Weaver AL, Graham DY, Kivitz AJ, Lipsky PE, Hubbard RC, et al. Anti-inflammatory and upper gastrointestinal effects of celecoxib in rheumatoid arthritis: a randomized controlled trial. JAMA 1999; 282(20): 1921-8. Singh, 1989 Singh G, Fries JF, Spitz P, Williams CA. Toxic effects of azathioprine in rheumatoid arthritis. A national post-marketing perspective. Arthritis Rheum 1989; 32(7): 837-43. Sledge, 1984 Sledge CB, Atcher RW, Shortkroff S, Anderson RJ, Bloomer WD, Hurson BJ. Intra-articular radiation synovectomy. Clin Orthop 1984; (182): 37-40. Sledge, 1986 Sledge CB, Zuckerman JD, Zalutsky MR, Atcher RW, Shortkroff S, Lionberger DR, et al. Treatment of rheumatoid synovitis of the knee with intraarticular injection of dysprosium 165-ferric hydroxide macroaggregates. Arthritis Rheum 1986; 29(2): 153-9. Sledge, 1987 Sledge CB, Zuckerman JD, Shortkroff S, Zalutsky MR, Venkatesan P, Snyder MA, et al. Synovectomy of the rheumatoid knee using intra-articular injection of dysprosium-165-ferric hydroxide macroaggregates. J Bone Joint Surg Am 1987; 69(7): 970-5. Smolen, 1999 Smolen JS, Kalden JR, Scott DL, Rozman B, Kvien TK, Larsen A, et al. Efficacy and safety of leflunomide compared with placebo and sulphasalazine in active rheumatoid arthritis: a double-blind, randomized, multicentre trial. Lancet 1999; 353(9149): 259-66. Songsiridej, 1990 Songsiridej N, Furst DE. Methotrexate: the rapidly acting drug. Baillieres Clin Rheumatol 1990; 4(3): 575-93. Spiegel, 1986 Spiegel JS, Spiegel TM, Ward NB, Paulus HE, Leake B, Kane RL. Rehabilitation in rheumatoid arthritis patients: a controlled trial. Arthritis Rheum 1986; 29(5): 628-37. Stein, 1997 Stein CM, Pincus T, Yocum D. Combination treatment of severe rheumatoid arthritis with cyclosporin and methotrexate for forty-eight weeks: an open-label extension study. The Methotrexate-Cyclosporin Combination Study Group. Arthritis Rheum 1997; 40:1843-51. Stein, 1980 Stein HB, Patterson AC, Offer RC, Atkins CJ, Teufel A, Robinson HS, et al. Adverse effects of D-penicillamine in rheumatoid arthritis. Ann Intern Med 1980; 92(1): 24-9. Steinberg, 1981 Steinberg AD. Immunoregulatory agents. In: Kelley WN, Harris ED, Ruddy S and Sledge CB, editors. Textbook of rheumatology. Philadelphia: Saunders WB; 1981. p. 841-61. Strand, 1999 Strand V, Cohen S, Schiff M, Weaver A, Fleischmann R, Cannon G, et al. Treatment of active rheumatoid arthritis with leflunomide compared with placebo and methotrexate. Arch Intern Med 1999; 159(21): 2542-50. Sutej, 1991 Sutej PG, Hadler NM. Current principles of rehabilitation for patients with rheumatoid arthritis. Clin Orthop.1991; (265): 116-24. Sznol, 1987 Sznol M, Ohnuma T, Holland JF. Hepatotoxicity of drugs used for hematologic neoplasia. Semin Liver Dis 1987; 7(3): 237-56. Taggart, 1987 Taggart AJ, Hill J, Astbury C, Dixon JS, Bird HA, Wright V. Sulphasalazine alone or in combination with D-penicillamine in rheumatoid arthritis. Br J Rheumatol 1987; 26:32-6. Tan, 1995 Tan SY, Pepys MB, Hawkins PN. Treatment of amyloidosis. Am J Kidney Dis 1995; 26(2): 267-85. Taylor, 1981 Taylor PG, Cumming DC, Coreenblum B. Successful treatment of D-penicillamine-induced breast gigantism with danazol. Br Med J 1981; 282(6261): 362-3. Thrasher, 1990. Thrasher JB, Miller GJ, Wettlaufer JN. Bladder leiomyosarcoma following cyclophosphamide therapy for lupus nephritis. J Urol 1990; 143(1): 119-21. Thurtle, 1983 Thurtle OA, Cawley MI. The frequency of leg ulceration in rheumatoid arthritis: a survey. J Rheumatol 1983; 10(3): 507-9. Tindall, 1987 Tindall RS, Rollins JA, Phillips JT, Greenlee RG, Wells L, Belendiuk G. Preliminary results of a double-blind, randomized, placebo-controlled trial of cyclosporin in myasthenia gravis. N Engl J Med 1987; 316(12): 719-24. Toovey, 1981 Toovey S, Hudson E, Hendry WF, Levi AJ. Sulphasalazine and male infertility: reversibility and possible mechanism. Gut 1981; 22(6): 445-51. Tornero, 1998 Tornero Molina J, Fernández Echevarría JA, Vidal Fuentes J. Repercusión socioeconómica de la invalidez permanente por enfermedad reumática en la provincia de Guadalajara. Rev Esp Reumatol 1998; 25(9): 346-51. Townes, 1976 Townes AS, Sowa JM, Shulman LE. Controlled trial of cyclophosphamide in rheumatoid arthritis. Arthritis Rheum 1976; 19(3): 563-73. Trompeter, 1981 Trompeter RS, Evans PR, Barratt TM. Gonadal function in boys with steroid-responsive nephrotic syndrome treated with cyclophosphamide for short periods. Lancet 1981; 1(8231): 1177-9. Tsakonas, 2000 Tsakonas E, Fitzgerald AA, Fitzcharles MA, Cividino A, Thorne JC, M'Seffar A, et al. Consequences of delayed therapy with second-line agents in rheumatoid arthritis: a 3 year follow-up on the hydroxychloroquine in early rheumatoid arthritis (HERA) study. J Rheumatol 2000; 27(3): 623-9. Tugwell, 1990 Tugwell P, Bombardier C, Gent M, Ho M, Iwatsuki S, Griffith BP. Low-dose cyclosporin versus placebo in patients with rheumatoid arthritis. Lancet 1990; 335(8697): 1051-5. Tugwell, 1993 Tugwell P, Boers M. Developing consensus on preliminary core efficacy endpoints for rheumatoid arthritis clinical trials. OMERACT Committee. J Rheumatol 1993; 20(3): 555-6. Tugwell, 1995a Tugwell P. International consensus recommendations on cyclosporin use in rheumatoid arthritis. Drugs 1995; 50(Suppl 1): 48-56. Tugwell, 1995b Tugwell P, Pincus T, Yocum D, Stein M, Gluck O, Kraag G, et al. Combination therapy with cyclosporin and methotrexate in severe rheumatoid arthritis. N Engl J Med 1995; 333(3): 137-41. Van der Heijde, 1990 Van der Heijde DM, van't Hof MA, van Riel PL, Theunisse LA, Lubberts EW, van Leeuwen MA, et al., Judging disease activity in clinical practice in rheumatoid arthritis: first step in the development of a disease activity score. Ann Rheum Dis 1990; 49(11): 916-20. Van der Heijde, 1992a Van der Heijde DM, van't Hof MA, van Riel PL, van Leeuwen MA, van Rijswijk MH, van de Putte LB. Validity of single variables and composite indices for measuring disease activity in rheumatoid arthritis. Ann Rheum Dis 1992; 51(2): 177-81. Van der Heijde, 1992b Van der Heijde DMFM, van Leeuwen MA, van Riel PL, Koster AM, van't Hof MA, van Rijswijk MH, et al. Biannual radiographic assessments of hands and feet in a three-year prospective followup of patients with early rheumatoid arthritis. Arthritis Rheum 1992; 35(1): 26-34. Van der Heijde, 1994 Van der Heijde A, Jacobs JW, van Albada-Kuipers GA, Kraaimaat FW, Geenen R, Bijlsma JW. Physical disability and psychological well being in recent onset rheumatoid arthritis. J Rheumatol 1994; 21(1): 28-32. Van der Heijde, 1995a Van der Heijde A, Remme CA, Hofman DM, Jacobs JWG, Bijlsma JWJ. Prediction of progression of radiologic damage in newly diagnosed rheumatoid arthritis. Arthritis Rheum 1995; 38(10): 1466-74. Van der Heijde, 1995b Van der Heijde DM. Joint erosions and patients with early rheumatoid arthritis. Br J Rheumatol 1995; 34(Suppl 2): 74-8. Van der Heijde, 1998 Van der Heijde DM, Jacobs JW. The original "DAS" and the "DAS28" are not interchangeable: comment on the articles by Prevoo et al. Arthritis Rheum 1998; 41(5): 942-5. Van der Heijde, 1999 Van der Heijde D. Boers M, Lassere M. Methodological issues in radiographic scoring methods in rheumatoid arthritis. J Rheumatol 1999; 26(3): 726-30. Van Gestel, 1995 Van Gestel AM, Laan RF, Haagsma CJ, van de Putte LBA, van Riel PLCM. Oral steroids as bridge therapy in rheumatoid arthritis patients starting with parenteral gold: a randomized double-blind placebo-controlled trial. Br J Rheumatol 1995; 34(4): 347-51. Van Gestel, 1996 Van Gestel AM, Prevoo ML, van't Hof MA, van Rijswijk MH, van de Putte LB, van Riel PL. Development and validation of the European League Against Rheumatism response criteria for rheumatoid arthritis. Comparison with the preliminary American College of Rheumatology and the World Health Organization/International League Against Rheumatism Criteria. Arthritis Rheum 1996; 39(1): 34-40. Van Gestel, 1998 Van Gestel AM, Haagsma CJ, van Riel PLCM. Validation of rheumatoid arthritis improvement criteria that included simplified joint counts. Arthritis Rheum 1998; 41(10): 1845-50. Van Gestel, 1999 Van Gestel AM, Anderson JJ, van Riel PL, Boers M, Haagsma CJ, Rich B, et al. ACR and EULAR improvement criteria have comparable validity in rheumatoid arthritis trials. J Rheumatol 1999; 26(3): 705-11. Van Leeuwen, 1993 Van Leeuwen MA, van Rijswijk MH, van der Heijde DM, Te Meerman GJ, van Riel PL, Houtman PM, et al. The acute-phase response in relation to radiographic progression in early rheumatoid arthritis. A prospective study during the first three years of the disease. Br J Rheumatol 1993; 32(Suppl 3): 9-13. Van Leeuwen, 1997 Van Leeuwen MA, van Rijswijk MH, Sluiter WJ, van Riel PL, Kuper IH, van de Putte LB et al. Individual relationship between progression of radiological damage and the acute phase response in early rheumatoid arthritis: towards development of a decision support system. J Rheumatol 1997; 24(1): 20-7. Van Riel, 1992 Van Riel PL. Provisional guidelines for measuring disease activity in clinical trials on rheumatoid arthritis. Br J Rheumatol 1992; 31(12): 793-4. Van Rijthoven, 1991 Van Rijthoven AW, Dijkmans BAC, Goei The HS, Meijers KA, Montnor Beckers ZL, Moolenburgh JD, et al. Comparison of cyclosporin and D-penicillamine for rheumatoid arthritis: a randomized, double blind, multicentre study. J Rheumatol 1991; 18(6): 815-20. Van Zeben, 1992 Van Zeben D, Hazes JM, Zwinderman AH, Cats A, van der Voort EA, Breedveld FC. Clinical significance of rheumatoid factors in early rheumatoid arthritis: results of a follow-up study. Ann Rheum Dis 1992; 51(9): 1029-35. Villaverde, 2000 Villaverde V, Hernández C, Fernández Sueiro JL, Hernández A, Graña J, Carmona L. Prevalencia de la artritis reumatoide en la población española. Rev Esp Reumatol 2000; 27(5): 157. Vollertsen, 1986 Vollertsen RS, Conn DL, Ballard DJ, Ilstrup DM, Kazmar RE, Silverfield JC. Rheumatoid vasculitis: survival and associated risk factors. Medicine (Baltimore) 1986; 65(6): 365-75. Vyse, 1992 Vyse T, So AK Sulphasalazine induced autoimmune syndrome. Br J Rheumatol 1992; 31(2): 115-6. Wakefield, 2000 Wakefield RJ, Gibbon WW, Conaghan PG, O'Connor P, McGonagle D, Pease C, et al. The value of sonography in the detection of bone erosions in patients with rheumatoid arthritis: a comparison with conventional radiography. Arthritis Rheum 2000; 43(12): 2762-70. Waldman, 1998 Waldman BJ, Figgie MP. Indications, technique, and results of total shoulder arthroplasty in rheumatoid arthritis. Orthop Clin North Am 1988; 29(3): 435-44. Walker, 1993 Walker AM, Funch D, Dreyer NA, Tolman KG, Kremer JM, Alarcon GS, et al. Determinants of serious liver disease among patients receiving low-dose methotrexate for rheumatoid arthritis. Arthritis Rheum 1993; 36(3): 329-35. Wall, 1975 Wall RL, Clausen KP. Carcinoma of the urinary bladder in patients receiving cyclophosphamide. N Engl J Med 1975; 293(6): 271-3. Warne, 1973 Warne GL, Fairley KF, Hobbs JB, Martin FI. Cyclophosphamide-induced ovarian failure. N Engl J Med 1973; 289(22): 1159-62. Wang, 1995 Wang CL, Wang F, Bosco JJ. Ovarian failure in oral cyclophosphamide treatment for systemic lupus erythematosus. Lupus 1995; 4(1): 11-4. Ward, 1983 Ward JR, Williams HJ, Egger MJ, Reading JC, Boyce E, Altz Smith M, et al. Comparison of auranofin, gold sodium thiomalate, and placebo in the treatment of rheumatoid arthritis: a controlled clinical trial. Arthritis Rheum 1983; 26(11): 1303-15. Weinblatt, 1999a Weinblatt ME, Kremer JM, Bankhurst AD, Bulpitt KJ, Fleischmann RM, Fox IR, 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 1999; 340(4): 253-9. Weinblatt, 1999b Weinblatt ME, Kremer JM, Coblyn JS, Maier AL, Helfgott SM, Morrell M, et al. Pharmacokinetics, safety, and efficacy of treatment with methotrexate and leflunomide in patients with active rheumatoid arthritis. Arthritis Rheum 1999; 42(7): 1322-8. Weinstein, 1985 Weinstein A, Utsinger PD. Immunoregulatory drugs In: Utsinger PD, Zvaifler NJ, Ehrlich GE, editors. Rheumatoid Arthritis. Philadelphia: Lippincott JB Company; 1985. p. 635-44. Weiss, 1989 Weiss MM: Corticosteroids in rheumatoid arthritis. Semin Arthritis Rheum 1989;19(1): 9-21. West, 1997 West SG. Methotrexate hepatotoxicity. Rheum Dis Clin North Am 1997; 23(4): 883-915. Wheeler, 1981 Wheeler GE. Cyclophosphamide-associated leukemia in Wegener's granulomatosis. Ann Intern Med 1981; 94(3): 361-2. White, 1989 White DA, Rankin JA, Stover DE, Gellene RA, Gupta S. Methotrexate pneumonitis: bronchoalveolar lavage findings suggest an immunologic disorder. Am Rev Resp Dis 1989; 139(1): 18-21. Willemsen, 1990 Willemsen MJ, De Coninck AL, De Raeve LE, Roseeuw DI. Penicillamine-induced pemphigus erythematosus. Int J Dermatol 1990; 29(3): 193-7. Williams, 1980 Williams HJ, Reading JC, Ward JR, O'Brien WM. Comparison of high and low dose cyclophosphamide therapy in rheumatoid arthritis. Arthritis Rheum 1980; 23(5): 521-7. Williams, 1983 Williams HJ, Ward JR, Reading JC, Egger MJ, Grandone JT, Samuelson CO, et al. Low-dose D-penicillamine therapy in rheumatoid arthritis: a controlled, double-blind clinical trial. Arthritis Rheum 1983; 26(5): 581-92. Williams, 1988 Williams HJ, Ward JR, Dahl SL, Clegg DO, Willkens RF, Oglesby T, et al. A controlled trial comparing sulfasalazine, gold sodium thiomalate, and placebo in rheumatoid arthritis. Arthritis Rheum 1988; 31(6): 702-13. Williams, 1992 Williams HJ, Ward JR, Reading JC, Brooks RH, Clegg DO, Skosey JL, et al. Comparison of auranofin, methotrexate, and the combination of both in the treatment of rheumatoid arthritis. A controlled clinical trial. Arthritis Rheum 1992; 35:259-69. Willkens, 1995 Willkens RF, Sharp JT, Stablein D, Marks C, Wortmann R. Comparison of azathioprine, methotrexate, and the combination of the two in the treatment of rheumatoid arthritis. A forty-eight-week controlled clinical trial with radiologic outcome assessment. Arthritis Rheum 1995; 38:1799-806. Wilske, 1989 Wilske KR, Healey LA. Remodeling the pyramid - a concept whose time has come. J Rheumatol 1989; 16:565-7. Wolfe, 1985 Wolfe F, Hawley DJ. Remission in rheumatoid arthritis. J Rheumatol 1985;12(2): 245-52. Wolfe, 1991 Wolfe F, Cathey MA. The assessment and prediction of functional disability in rheumatoid arthritis. J Rheumatol 1991; 18(9): 1298-306. Wolfe, 1998 Wolfe F, Hawley DJ. The longterm outcomes of rheumatoid arthritis: Work disability. A prospective 18 year study of 823 patients. J Rheumatol 1998; 25(11): 2108-17. Wolfe, 1999a Wolfe F, Pincus T. Listening to the patient: a practical guide to self-report questionnaires in clinical care. Arthritis Rheum 1999; 42(9): 1797-808. Wolfe, 1999b Wolfe F, Lassere M, van der Heijde D, Stucki G, Suarez Almazor M, Pincus T, et al. Preliminary core set of domains and reporting requirements for longitudinal observational studies in rheumatology. J Rheumatol 1999; 26(2): 484-9. Worth, 1971 Worth PH. Cyclophosphamide and the bladder. Br Med J 1971; 3(767): 182. Worthley, 1995 Worthley SG, McNeil JD. Leukoencephalopathy in a patient taking low dose oral methotrexate therapy for rheumatoid arthritis. J Rheumatol 1995; 22(2): 335-7. Yuh, 1995 Yuh DD, Gandy KL, Morris RE, Hoyt G, Gutierrez J, Reitz BA, et al. Leflunomide prolongs pulmonary allograft and xenograft survival. J Heart Lung Transplant 1995; 14(6 Pt 1): 1136-44. Zemer, 1986 Zemer D, Pras M, Sohar E, Modan M, Cabili S, Gafni J. Colchicine in the prevention and treatment of the amyloidosis of familial Mediterranean fever. N Engl J Med 1986; 314(16): 1001-5. 2. References used in the studies included in the synthesis of the evidence 2.1. Systematic reviews (SR) SR1. Suarez Almazor ME, Spooner CH, Belseck E, Shea B. Auranofin versus placebo in rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD002048. SR2. Suarez Almazor ME, Spooner C, Belseck E. Azathioprine for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD001461. SR3. Suarez Almazor ME, Belseck E, Shea B, Wells G, Tugwell P. Cyclophosphamide for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD001157. SR4. Wells G, Haguenauer D, Shea B, Suarez Almazor ME, Welch VA, Tugwell P. Cyclosporin for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD001083. SR5. Suarez Almazor ME, Spooner C, Belseck E. Penicillamine for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD001460. SR6. Suarez Almazor ME, Belseck E, Shea B, Homik J, Wells G, Tugwell P. Antimalarials for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD000959. SR7. Suarez Almazor ME, Belseck E, Shea B, Wells G, Tugwell P. Methotrexate for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD000957. SR8. Clark P, Tugwell P, Bennet K, Bombardier C, Shea B, Wells G, Suarez Almazor ME. Injectable gold for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD000520. SR9. Suarez Almazor ME, Belseck E, Shea B, Wells G, Tugwell P. Sulfasalazine for rheumatoid arthritis. Cochrane Database Syst Rev 2000; (2): CD000958. 2.2. Articles on clinical trials referenced in the evidence tables (Tables 7, 8, and 9) 1. Currey HL, Harris J, Mason RM, Woodland J, Beveridge T, Roberts CJ, et al. Comparison of azathioprine, cyclophosphamide, and gold in treatment of rheumatoid arthritis. BMJ 1974; 3(5934): 763-6. 2. Rao URK, Naidu MUR, Kumar TR, Shobha U, Askar MA, Ahmed N, et al. Comparison of phenytoin with auranofin and chloroquine in rheumatoid arthritis: a double blind study. J Rheumatol 1995; 22(7): 1235-40. 3. Dwosh IL, Stein HB, Urowitz MB, Smythe HA, Hunter T, Ogryzlo MA. Azathioprine in early rheumatoid arthritis: comparison with gold and chloroquine. Arthritis Rheum 1977; 20(2): 685-92. 4. Forre O, Bjerkhoel F, Salvesen CF, Berg KJ, Rugstad HE, Saelid G, et al. An open, controlled, randomized comparison of cyclosporin and azathioprine in the treatment of rheumatoid arthritis: a preliminary report. Arthritis Rheum 1987; 30(1): 88-92. 5. Landewé RBM, Goei-The HS, Rijthoven AWAM, Breedveld FC, Dijkmans BAC. A randomized, double-blind, 24-week controlled study of low-dose cyclosporin versus chloroquine for early rheumatoid arthritis. Arthritis Rheum 1994; 37(5): 637-43. 6. Hochberg MC. Auranofin or D-penicillamine in the treatment of rheumatoid arthritis. Ann Intern Med 1986; 105(4): 528-35. 7. Berry H, Liyanage SP, Durance RA, Barnes CG, Berger LA, Evans S. Azathioprine and penicillamine in treatment of rheumatoid arthritis: a controlled trial. BMJ 1976; 1(6017): 1052-4. 8. Paulus HE, Williams HJ, Ward JR, Reading JC, Egger MJ. Coleman ML, et al. Azathioprine versus D-penicillamine in rheumatoid arthritis patients who have been treated unsuccessfully with gold. Arthritis Rheum 1984; 27(7): 721-7. 9. Halberg P, Bentzon MW, Crohn O, Gad I, Halskov O, Heyn J, et al. Double-blind trial of levamisole, penicillamine and azathioprine in rheumatoid arthritis: clinical, biochemical, radiological and scintigraphic studies. Dan Med Bull 1984; 31(5): 403-9. 10. Gibson T, Emery P, Armstrong RD, Crisip AJ, Panayi GS. Combined D-penicillamine and chloroquine treatment of rheumatoid arthritis: a comparative study. Br J Rheumatol 1987; 26(4): 279-84. 11. Rijthoven AWAM, Dijkmans BAC, Goei Thé HS, Meijers KAE, Montnor-Beckers ZLBM, Moolenburgh JD, et al. Comparison of cyclosporin and D-penicillamine for rheumatoid arthritis: a randomized, double blind, multicenter study. J Rheumatol 1991; 18(6): 815-20. 12. Moreland LW, Schiff MH, Baumgartner SW, Tindall EA, Fleischmann RM, Bulpitt KJ, et al. Etanercept therapy in rheumatoid arthritis: a randomized, controlled trial. Ann Intern Med 1999; 130: 478-86. 13. Moreland LW, Baumgartner SW, Schiff MH, Tindall EA, Fleischmann RM, Weaver AL, et al. Treatment of rheumatoid arthritis with a recombinant human tumor necrosis factor receptor (p75)-Fc fusion protein. N Engl J Med 1997; 337(3): 41-7. 14. Bird HA, Le Gallez P, Dixon JS, Surrall KE, Cole DS, Goldman MH, et al. A single-blind comparative study of auranofin and hydroxychloroquine in patients with rheumatoid arthritis. Clin Rheumatol 1984; 3(suppl. 1): 57-66. 15. Bunch TW, O'Duffy JD, Tompkins RB, O'Fallon WM. Controlled trial of hydroxychloroquine and D-penicillamine singly and in combination in the treatment of rheumatoid arthritis. Arthritis Rheum 1984; 27(3): 267-76. 16. Scott DL, Greenwood A, Davies J, Maddison PJ, Maddison MC, Hall ND. Radiological progression in rheumatoid arthritis: do d-penicillamine and hydroxychloroquine have different effects? Br J Rheumatol 1990; 29(2): 126-7. 17. Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C, Smolen JS, et al. Ramdomized double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor alfa (cA2) versus placebo in rheumatoid arthritis. Lancet 1994; 34: 1105-10. 18. Smolen JS. Efficacy and safety of leflunomide compared with placebo and sulphasalazine in active rheumatoid arthritis: a double-blind, randomized, multicentre trial: European Leflunomide Study Group. Lancet 1999; 353(9149): 259-66. 19. Strand V, Cohen S, Schiff M, Weaver A, Fleischmann R, Cannon G, et al. Treatment of active rheumatoid arthritis with leflunomide compared with placebo and methotrexate. Arch Intern Med 1999; 159(21): 2542-50. 20. Mladenovic V, Domljan Z, Rozman B, Jajic I, Mihajlovic D, Dordevic J, et al. Safety and effectiveness of leflunomide in the treatment of patients with active rheumatoid arthritis: results of a randomized, placebo-controlled, phase II study. Arthritis Rheum 1995; 38(11): 1595-1603. 21. Weinblatt ME, Kaplan H, Germain BF, Merriman RC, Solomon SD, et al. Low-dose methotrexate compared with auranofin in adult rheumatoid arthritis: a thirty-six-week, double-blind trial. Arthritis Rheum 1990; 33(3): 330-8. 22. Williams HJ, Ward JR, Reading JC, Brooks RH, Clegg DO, Skosey JL, et al. Comparison of auranofin, methotrexate, and the combination of both in the treatment of rheumatoid arthritis: a controlled clinical trial. Arthritis Rheum 1992; 35(3): 259-69. 23. Hamdy H, McKendry RJR, Mierins E, Liver JA. Low-dose methotrexate compared with azathioprine in the treatment of rheumatoid arthritis: a twenty-four-week controlled clinical trial. Arthritis Rheum 1987; 30(4): 361-8. 24. Jeurissen MEC, Boerbooms AMT, Van de Putte LBA, Doesburg WH, Mulder J, Rasker JJ, et al. Methotrexate versus azathioprine in the treatment of rheumatoid arthritis: a forty-eight-week randomized, double-blind trial. Arthritis Rheum 1991; 34(8): 961-72. 25. Willkens RF, Sharp JT, Stablein D, Marks C, Wortmann R. Comparison of azathioprine, methotrexate, and the combination of the two in the treatment of rheumatoid arthritis: a forty-eight-week controlled clinical trial with radiologic outcome assessment. Arthritis Rheum 1995; 38(12): 1799-1806. 26. Arnold MH, O'Callaghan J, McCredie M, Beller EM, Kelly DE, Brooks PM. Comparative controlled trial of low-dose weekly methotrexate versus azathioprine in rheumatoid arthritis: 3-year prospective study. Br J Rheumatol 1990; 29: 120-5. 27. Drosos AA, Voulgari PV, Papadopoulos IA, Politi EN, Georgiou PE, Zikou AK. Cyclosporin A in the treatment of early rheumatoid arthritis: a prospective, randomized 24-month study. Clin Exp Rheumatol 1998; 16(6): 695-701. 28. Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, Macfarlane JD, et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis actor alfa monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum 1998; 41(9): 1552-63. 29. Emery P, Breedveld FC, Lemmel EM, Kaltwasser JP, Dawes PT, Gömör B, et al. A comparison of the efficacy and safety of leflunomide and methotrexate for the treatment of rheumatoid arthritis. Rheumatology 2000; 39: 655-65. 30. Ward JR, Williams HJ, Egger MJ, Reading JC, Boyce E, Altz-Smith M, et al. Comparison of auranofin, gold sodium thiomalate, and placebo in the treatment of rheumatoid arthritis: a controlled clinical trial. Arthritis Rheum 1983; 26(11): 1303-15. 31. Prete PE, Zane J, Krailo M, Bulanowski M. Randomized trial of switching rheumatoid arthritis patients in remission with injectable gold to auranofin. Clin Rheumatol 1994; 13(1): 60-9. 32. Smith PR, Brown GMM, Meyers OL. An open comparative study of auranofin versus gold sodium thiomalate. J Rheumatol Suppl 1982; 8: 190-6. 33. Harth M, Davis P, Thompson JM, Menard H, Beaudet F. Comparison between sodium aurothiomalate and auranofin in rheumatoid arthritis: results of a two-year open randomized study. Scand J Rheumatol 1987; 16(3): 177-84. 34. Hull RG, Morgan SH, Parke AL, Childs L, Goldman M, Hughes GRV. A double-blind study comparing sodium aurothiomalate and auranofin in patients with rheumatoid arthritis previously stabilized on sodium aurothiomalate. Int J Clin Pharmacol Res 1984; 4(6): 395-401. 35. Rau R, Schattenkirchner M, Müller-Fass-Bender H, Kaik B, Zeidelr H. A three year comparative multicenter study of auranofin (AF) and gold sodium thiomalate (GST) in the treatment of rheumatoid arthritis (RA). Clin Rheumatol 1987; 6(suppl. 2): 43-52. 36. Yamamoto S, Mitomo T, Saito T, Inoue K, Uchida S, Namiki O, et al. Randomized comparative study in early rheumatoid arthritis with gold sodium thiomalate versus auranofin. Jpn J Rheumatol 1992; 4(1): 33-53. 37. Van Riel PL, Van de Putte LBA, Gribnau FWJ, Macrae KD. Comparison of auranofin and aurothioglucose in the treatment of rheumatoid arthritis: a single blind study. Clin Rheumatol 1984; 3(suppl. 1): 51-6. 38. Lewis D, Capell HA. Oral gold: a comparison with placebo and with intramuscular sodium aurothiomalate. Clin Rheumatol 1984; 3(suppl. 1): 83-96. 39. Rau R, Schattenkirchner M, Muller-Fassbender H, Kaik B, Zeidler H, Missler B. A three year comparative study of auranofin and gold sodium thiomalate in rheumatoid arthritis. Clin Rheumatol 1990; 9(4): 461-74. 40. Zeidler HK, Kvien TK, Hannonen P, Wollheim FA, Forre O, Geidel H, et al. Progression of joint damage in early active severe rheumatoid arthritis during 8 months of treatment: comparison of low-dose cyclosporin and parenteral gold. Br J Rheumatol 1998; 37: 874-82. 41. Huskisson EC, Gibson TJ, Balme HW, Berry H, Burry HC, Grahame R, et al. Trial comparing D-penicillamine and gold in rheumatoid arthritis: preliminary report. Ann Rheum Dis 1974; 33(6): 532-5. 42. Mäkisara P, Nissilä M, Kajander A, Martio J, Essen R, Anttila P, et al. Comparison of penicillamine and gold treatment in early rheumatoid arthritis. Scand J Rheumatol 1978; 7(3): 166-70. 43. Thomas MH, Rothermich NO, Philips VK, Bergen W, Hedrick SW. Gold versus D-penicillamine double-blind study and followup. J Rheumatol 1984; 11(6): 764-7. 44. Suarez Almazor ME, Fitzgerald A, Grace M, Russell AS. A randomized controlled trial of parenteral methotrexate compared with sodium aurothiomalate (Myochrysine registered) in the treatment of rheumatoid arthritis. J Rheumatol 1988; 15(5): 753-6. 45. Rau R, Herborn G, Karger T, Menninger H, Elhardt D, Schmitt J. A double-blind comparison of parenteral methotrexate and parenteral gold in the treatment of early erosive rheumatoid arthritis: an interim report on 102 patients after 12 months. Semin Arthritis Rheum 1991; 21(2 Suppl 1): 13-20. 46. Rau R, Herborn G, Karger T, Menninger H, Elhardt D, Schmitt J. A double blind randomized parallel trial of intramuscular methotrexate and gold sodium thiomalate in early erosive rheumatoid arthritis. J Rheumatol 1991; 18(3): 328-33. 47. Menninger H, Herborn G, Sander O, Blechschmidt J, Rau R. A 36 month comparative trial of methotrexate and gold sodium thiomalate in the treatment of early active and erosive rheumatoid arthritis. Br J Rheumatol 1998; 37: 1060-8. 48. Morassut P, Goldstein R, Cyr M, Karsh J, McKendry RJ. Gold sodium thiomalate compared to low dose methotrexate in the treatment of rheumatoid arthritis: a randomized, double blind 26-week trial. J Rheumatol 1989; 16(3): 302-6. 49. McEntegart A, Porter D, Capell HA, Thomson EA. Sulphasalazine has a better efficacy/toxicity profile than auranofin: evidence from a 5 year prospective, randomized trial. J Rheumatol 1996; 23(11): 1887-90. 50. Capell HA, Maiden N, Madhok R, Hampson R, Thomson EA. Intention to treat analysis of 200 patients with rheumatoid arthritis 12 years after random allocation to either sulfasalazine or penicillamine. J Rheumatol 1998; 25(10): 1880-6. 51. Carroll GJ, Will RK, Breidahl PD, Tinsley LM. Sulphasalazine versus penicillamine in the treatment of rheumatoid arthritis. Rheumatol Int 1989; 8(6): 251-5. 52. Nuver-Zwart IH, Van Riel PLCM, Van de Putte LBA, Grignau FWJ. A double blind comparative study of sulphasalazine and hydroxychloroquine in rheumatoid arthritis: evidence of an earlier effect of sulphasalazine. Ann Rheum Dis 1989; 48(5): 389-95. 53. Faarvang KL, Egsmose Ch, Kryger P, Podenphant J, Ingeman-Nielsen M, Hansen TM. Hydroxychloroquine and sulphasalazine alone and in combination in rheumatoid arthritis: a randomized double blind trial. Ann Rheum Dis 1993; 52(10): 711-5. 54. Haagsma CJ, Van Riel PLCM, De Jong AJL, Van de Putte LBA. Combination of sulphasalazine and methotrexate versus the single components in early rheumatoid arthritis: a randomized, controlled, double-blind 52 week clinical trial. Br J Rheumatol 1997; 36: 1082-8. 55. Dougados M, Combe B, Cantagrel A, Goupille P, Olive P, Schattenkirchner M, et al. Combination therapy in early rheumatoid arthritis: a randomized, controlled, double-blind 52 week clinical trial of sulphasalazine and methotrexate compared with the single components. Ann Rheum Dis 1999; 58: 220-5. 56. Peltomaa R, Paimela L, Helve T, Leirisalo-Repo M. Comparison of intramuscular gold and sulphasalazine in the treatment of early rheumatoid arthritis: a one year prospective study. Scand J Rheumatol 1995; 24(6): 330-5. 57. Bathon JM, Martin RW, Fleischmann RM, Tesser JR, Schiff MH, Keystone EC, et al. A comparison of etanercept and methotrexate in patients with early rheumatoid arthritis. N Engl J Med 1994; 343(22): 1586-93. 58. Trnavsky K, Gatterová J, Linduskova M, Pelisková Z. Combination therapy with hydroxychloroquine and methotrexate in rheumatoid arthritis. Z Rheumatol 1993; 52(5): 292-6. 59. Ferraz MB, Pinheiro GRC, Helfenstein M, Albuquerque E, Rezende C, Roimicher L, et al. Combination therapy with methotrexate and chloroquine in rheumatoid arthritis: a multicenter randomized placebo-controlled trials. Scand J Rheumatol 1994; 23: 231-6. 60. Tugwell P, Pincus T, Yocum D, Stein M, Gluck O, Kraag G, et al. Combination therapy with cyclosporin and methotrexate in severe rheumatoid arthritis. N Engl J Med 1995; 333(3): 137-41. 61. Weinblatt ME, Kremer JM, Bankhurst AD, Bulpitt KJ, Fleischmann RM, Fox RI, 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 1999; 340(4): 253-9. 62. O'Dell JR, Haire CE, Erikson N, Drymalski W, Palmer W, Eckhoff PJ, et al. Treatment of rheumatoid arthritis with methotrexate alone, sulfasalazine and hydroxychloroquine, or a combination of all three medications. N Engl J Med 1996; 334(20): 1287-91. 63. Lipsky PE, Van der Heijde DMFM, St. Clair W, Furst DE, Breedveld FC, Kalden JR. Infliximab and methotrexate in the treatment of rheumatoid arthritis. N Engl J Med 1994; 343(22): 1594-1602. 64. Bendix G, Bjelle A. Adding low-dose cyclosporin A to parenteral gold therapy in rheumatoid arthritis: a double-blind placebo-controlled study. Br J Rheumatol 1996; 35: 1142-9. 65. Scott DL, Dawes PT, Tunn E, Fowler PD, Shadforth MF, Fisher J, et al. Combination therapy with gold and hydroxychloroquine in rheumatoid arthritis: a prospective, randomized, placebo-controlled study. Br J Rheumatol 1989; 28(2): 128-33. 66. Porter DR, Capell HA, Hunter J. Combination therapy in rheumatoid arthritis: no benefit of addition of hydroxychloroquine to patients with a suboptimal response to intramuscular gold therapy. J Rheumatol 1993; 20(4): 645-9. 2.3. Articles that complement one of the articles on the clinical trials (Tables 7, 8, and 9) Complement of 21 C1. Weinblatt ME, Polisson R, Blotner SD, Sosman JL, Aliabadi P, Baker N, et al. The effects of drug therapy on radiographic progression of rheumatoid arthritis: results of a 36-week randomized trial comparing methotrexate and auranofin. Arthritis Rheum 1993; 36(5): 613-9. Complement of 22 C2. Lopez-Mendez A, Daniel WW, Reading JC, Ward JR, Alarcon GS. Radiographic assessment of disease progression in rheumatoid arthritis patients enrolled in the cooperative systematic studies of the rheumatic diseases program randomized clinical trial of methotrexate, auranofin, or a combination of the two. Arthritis Rheum 1993; 36(10): 1364-9. Complement of 30 C3. Ward JR, Williams HJ, Boyce E, Egger MJ, Reading JC, Samuelson CO. Comparison of auranofin, gold sodium thiomalate, and placebo in the treatment of rheumatoid arthritis: subsets of responses. Am J Med 1983; 75(6A): 133-7. Complement of 33 C4. Davis P, Menard H, Thompson J, Harth M, Beaudet F. One-year comparative study of gold sodium thiomalate and auranofin in the treatment of rheumatoid arthritis. J Rheumatol 1985; 12(1): 60-7. Complement of 37 C5. Van Riel PLCM, Larsen A, Van de Putte LBA, Gribnau FWJ. Effects of aurothioglucose and auranofin on radiographic progression in rheumatoid arthritis. Clin Rheumatol 1986; 5(3): 359-64. Complement of 39 C6. Schattenkirchner M, Kaik B, Muller-Fassbender H, Rau R, Zeidler H. Auranofin and sodium aurothiomalate in the treatment of rheumatoid arthritis: a double-blind, comparative multicenter study. J Rheumatol Suppl 1982; 8: 184-9. Complement of 47 C7. Rau R, Herborn G, Menninger H, Sangha O. Progression in early erosive rheumatoid arthritis: 12 month results from a randomized controlled trial comparing methotrexate and gold sodium thiomalate. Br J Rheumatol 1998; 37(11): 1220-6. C8. Rau R, Herborn G, Menninger H, Blechschmidt J. Comparison of intramuscular methotrexate and gold sodium thiomalate in the treatment of early erosive rheumatoid arthritis: 12 month data of a double-blind parallel study of 174 patients. Br J Rheumatol 1997; 36(3): 342-52. Complement of 49 C9. Porter D, Madhok R, Hunter JA, Capell HA. Prospective trial comparing the use of sulphasalazine and auranofin as second line drugs in patients with rheumatoid arthritis. Ann Rheum Dis 1992; 51(4): 461-4. Complement of 50 C10. Capell HA, Marabani M, Madhok R, Torley H, Hunter JA. Degree and extent of response to sulphasalazine or penicillamine therapy for rheumatoid arthritis: results from a routine clinical environment over a two-year period. Q J Med 1990; 75(276): 335-44. Complement of 52 C.11. Heijde DM, Riel PL, Nuver-Zwart IH, Gribnau FW, Putte LB. Effects of hydroxychloroquine and sulphasalazine on progression of joint damage in rheumatoid arthritis. Lancet 1989; 1(8646): 1036-8. C12. Van der Heijde DM, Van Riel PLCM, Van de Putte LBA. Sensitivity of a Dutch Health Assessment Questionnaire in a trial comparing hydroxychloroquine vs sulphasalazine. Scand J Rheumatol 1990; 19(6): 407-12. 2.4. Articles that are redundant with one of the articles on clinical trials (Tables 7, 8, and 9) Redundant with 1 R1. Woodland J, Mason RM, Harris J, Dixon AS, Currey HL, Brownjohn AM, et al. Trial of azathioprine, cyclophosphamide, and gold in rheumatoid arthritis. Ann Rheum Dis 1974; 33(4): 399-401. Redundant with 7 R2. Berry H, Liyanage S, Durance R, Barnes CG, Berger L. Trial comparing azathioprine and penicillamine in treatment of rheumatoid arthritis. Ann Rheum Dis 1976; 35(6): 542-3. Redundant with 19 R3. Strand V, Tugwell P, Bombardier C, Maetzel A, Crawford B, Dorrier C, et al. Function and health-related quality of life: results from a randomized controlled trial of leflunomide versus methotrexate or placebo in patients with active rheumatoid arthritis. Arthritis Rheum 1999; 42(9): 1870-8. Redundant with 24 R4. Jeurissen MEC, Boerbooms AMT, Van de Putte LBA, Doesburg WH, Lemmens AM. Influence of methotrexate and azathioprine on radiologic progression in rheumatoid arthritis: a randomized, double-blind study. Arch Intern Med 1991; 114: 999-1004. R5. Kerstens PJSM, Boerbooms AMT, Jeurissen MEC, Westgeest TAA, Van Erp A, Mulder J, et al. Antiperinuclear factor and disease activity in rheumatoid arthritis: longitudinal evaluation during methotrexate and azathioprine therapy. J Rheumatol 1994; 21(12): 2190-4. Redundant with 25 R6. Willkens RF, Stablein D. Combination treatment of rheumatoid arthritis using azathioprine and methotrexate: a 48-week controlled clinical trial. J Rheumatol 1996; 23(Suppl 44): 64-8. R7. Willkens RF, Urowitz MB, Stablein DM, McKendry RJR, Berger RG, Box JH, et al. Comparison of azathioprine, methotrexate, and the combination of both in the treatment of rheumatoid arthritis: a controlled clinical trial. Arthritis Rheum 1992; 35(8): 849-56. Redundant with 30 R8. Williams HJ, Ward JR, Egger MJ, Reading JC, Samuelson CO, Altz-Smith M, et al. Auranofin, gold sodium thiomalate, and placebo in the treatment of rheumatoid arthritis: cooperative systematic studies of rheumatic diseases. Clin Rheumatol 1984; 3(suppl.1): 39-50. R9. Williams HJ, Ward JR. Comparison of oral and parenteral gold therapy and placebo in the treatment of rheumatoid arthritis. Scand J Rheumatol Suppl 1983; 51: 92-9. Redundant with 39 R10. Schattenkirchner M, Bröll H, Kaik B, M¨ller-Fassbender H, Rau R, Zeidler H. Auranofin and gold sodium thiomalate in the treatment of rheumatoid arthritis: a one-year, double-blind, comparative multicenter study. Klin Wochenschr 1988; 66(4): 167-74. Redundant with 41 R11. Huskisson EC, Gibson TJ, Wykeham Balme H, Berry H, Burry HC, Grahame R, et al. Penicillamine or gold for rheumatoid arthritis?: multicentre trial using 'blind' observers: the first six months. Ann Rheum Dis 1974; 33(4): 399. Redundant with 52 R12. Van der Heijde DMFM, Van Riel PLCM, Nuwer-Zwart IH, Van de Putte LBA. Alternative methods for analysis of radiographic damage in a randomized, double blind, parallel group clinical trial comparing hydroxychloroquine and sulfasalazine. J Rheumatol 2000; 27(2): 535-8. Redundant with 63 R13. Maini R, St Clair EW, Breedveld F, Furst D, Kalden J, Weisman M, et al. Infliximab (chimeric anti-tumour necrosis factor alpha monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial. ATTRACT Study Group. Lancet 1999; 354(9194): 1932-9. Copyright: All copyrights reserved by the Spanish Society of Rheumatology.