1
|
Ejaz S, Gurugubelli S, Prathi SK, Palou Martinez Y, Arrey Agbor DB, Panday P, Yu AK. The Role of Etanercept in Controlling Clinical and Radiological Progression in Rheumatoid Arthritis: A Systematic Review. Cureus 2024; 16:e58112. [PMID: 38738082 PMCID: PMC11088797 DOI: 10.7759/cureus.58112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Etanercept (ETN) is a disease-modifying anti-rheumatic drug (DMARD) used in the treatment of rheumatoid arthritis (RA) that works as a tumor necrosis factor inhibitor (TNF inhibitor) by blocking the effects of naturally occurring TNF. This review will evaluate the effect of ETN as a monotherapy or combination therapy with methotrexate (MTX) in the treatment of RA. This systematic review was carried out in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) 2020 guidelines. A systematic search was done on PubMed and Google Scholar from 1999 to 2023. Predefined eligibility criteria were set for selected studies, which include: free full-text articles published; randomized control trials (RCTs); systematic reviews and meta-analyses; and observational studies in a patient with RA treated with ETN as initial therapy or as an add-on to conventional disease-modified therapy. Hence, the data had been extracted, and a quality assessment of each study was done by two individual authors. When comparing patients who received 15-25 mg of MTX with those who also received 25 mg of ETN in combination, 71% achieved American College of Rheumatology 20 (ACR20) by 24 weeks, compared to 27% in the MTX and placebo groups (p<0.001), and 39% achieved American College of Rheumatology 50 (ACR50), compared to 3% in the placebo + MTX group (p<0.001). Low disease activity (DAS 28) was more common in patients who had both MTX and ETN (64.5% with DAS <2.4 and 56.3% with DAS 28 <3.2) compared to patients who received only one medication (44.4% with DAS <2.4 and 33.2% with DAS 28 <3.2 for ETN and 38.6% with DAS <2.4 and 28.5% with DAS 28 <3.2 for MTX, with P<0.01). ETN demonstrated smaller changes from baseline in the modified Sharp score (TSS) and erosion scores (ES) at 12 months and two years, as well as a decreased change in the ES score at one year (with a trend of P value = 0.06 for the TSS score), in comparison to those receiving DMARD. Reactions at the injection site (42% vs. 7%, P<0.001) were the only events that occurred significantly more frequently in the ETN plus-MTX group. Combining ETN and MTX appears to help control RA symptoms by decreasing the American College of Rheumatology (ACR) response and DAS score, as well as halting the disease's progression on X-rays. The most common adverse effects were reactions to ETN administered alone at the injection site, likely because of patient awareness of the treatment received. There was also concern about tuberculosis and malignancy, but no recent data is available. Therefore, a larger clinical trial with longer follow-up is required to ascertain long-term safety and benefits.
Collapse
Affiliation(s)
- Samrah Ejaz
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Simhachalam Gurugubelli
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Internal Medicine, Memorial Healthcare, Gulfport, USA
| | - Suviksh K Prathi
- General Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- General Medicine, St. George's University School of Medicine, St. Georges, GRD
| | - Yaneisi Palou Martinez
- Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Divine Besong Arrey Agbor
- Clinical Research and Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Internal Medicine, Richmond University Medical Center, New York City, USA
| | - Priyanka Panday
- Research, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Ann Kashmer Yu
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
2
|
Comparison of Biological Agent Monotherapy and Associations Including Disease-Modifying Antirheumatic Drugs for Rheumatoid Arthritis: Literature Review and Meta-Analysis of Randomized Trials. J Clin Med 2022; 12:jcm12010286. [PMID: 36615086 PMCID: PMC9821556 DOI: 10.3390/jcm12010286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Update the available evidence comparing biologic disease-modifying antirheumatic drugs (bDMARDs) in combination with conventional synthetic disease-modifying antirheumatic drugs (CsDMARDs) to bDMARDs in monotherapy in patients with rheumatoid arthritis. METHODS Research was limited to randomized controlled trials. Major outcome: ACR 20 response criteria at 24 weeks. SECONDARY OUTCOMES clinical and radiographic criteria at week 24, 52 and 104. RESULTS 23 trials (6358 patients), including seven bDMARDs and one other molecule: Anbainuo (anti-TNF-R). No study satisfied our search criteria for anakinra, certolizumab and infliximab. Compared to bDMARD monotherapy, combination therapy gives a better ACR 20 at 24 weeks (RR: 0.88 (0.84-0.94)) in fixed and random effect models, and this result is sustained at 52 and 104 weeks. The results were mostly similar for all other outcomes without increasing the risk of adverse effects. CONCLUSION This meta-analysis confirms the superiority of combination therapy over monotherapy in rheumatoid arthritis, in accordance to the usual guidelines.
Collapse
|
3
|
He B, Li Y, Luo WW, Cheng X, Xiang HR, Zhang QZ, He J, Peng WX. The Risk of Adverse Effects of TNF-α Inhibitors in Patients With Rheumatoid Arthritis: A Network Meta-Analysis. Front Immunol 2022; 13:814429. [PMID: 35250992 PMCID: PMC8888889 DOI: 10.3389/fimmu.2022.814429] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/24/2022] [Indexed: 12/31/2022] Open
Abstract
Objectives To evaluate the safety of each anti-TNF therapy for patients with rheumatoid arthritis (RA) and then make the best choice in clinical practice. Methods We searched PUBMED, EMBASE, and the Cochrane Library. The deadline for retrieval is August 2021. The ORs, Confidence Intervals (CIs), and p values were calculated by STATA.16.0 software for assessment. Result 72 RCTs involving 28332 subjects were included. AEs were more common with adalimumab combined disease-modifying anti-rheumatic drugs (DMARDs) compared with placebo (OR = 1.60, 95% CI: 1.06, 2.42), DMARDs (1.28, 95% CI: 1.08, 1.52), etanercept combined DMARDs (1.32, 95% CI: 1.03, 1.67); certolizumab combined DMARDs compared with placebo (1.63, 95% CI: 1.07, 2.46), DMARDs (1.30, 95% CI: 1.10, 1.54), etanercept combined DMARDs (1.34, 95% CI: 1.05, 1.70). In SAEs, comparisons between treatments showed adalimumab (0.20, 95% CI: 0.07, 0.59), etanercept combined DMARDs (0.39, 95% CI: 0.15, 0.96), golimumab (0.19, 95% CI: 0.05, 0.77), infliximab (0.15, 95% CI: 0.03,0.71) decreased the risk of SAEs compared with golimumab combined DMARDs. In infections, comparisons between treatments showed adalimumab combined DMARDs (0.59, 95% CI: 0.37, 0.95), etanercept (0.49, 95% CI: 0.28, 0.88), etanercept combined DMARDs (0.56, 95% CI: 0.35, 0.91), golimumab combined DMARDs (0.51, 95% CI: 0.31, 0.83) decreased the risk of infections compared with infliximab combined DMARDs. No evidence indicated that the use of TNF-α inhibitors influenced the risk of serious infections, malignant tumors. Conclusion In conclusion, we regard etanercept monotherapy as the optimal choice for RA patients in clinical practice when the efficacy is similar. Conversely, certolizumab + DMARDs therapy is not recommended. Systematic Review Registration identifier PROSPERO CRD42021276176.
Collapse
Affiliation(s)
- Bei He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Wen Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huai-Rong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qi-Zhi Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Xing Peng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
4
|
Sellam J, Morel J, Tournadre A, Bouhnik Y, Cornec D, Devauchelle-Pensec V, Dieudé P, Goupille P, Jullien D, Kluger N, Lazaro E, Le Goff B, de Lédinghen V, Lequerré T, Nocturne G, Seror R, Truchetet ME, Verhoeven F, Pham T, Richez C. PRACTICAL MANAGEMENT of patients on anti-TNF therapy: Practical guidelines drawn up by the Club Rhumatismes et Inflammation (CRI). Joint Bone Spine 2021; 88:105174. [PMID: 33992225 DOI: 10.1016/j.jbspin.2021.105174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Jérémie Sellam
- Service de Rhumatologie, CHU Saint-Antoine, Paris, France
| | - Jacques Morel
- Service de Rhumatologie, CHU Montpellier, Montpellier, France
| | - Anne Tournadre
- Service de Rhumatologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Yoram Bouhnik
- Service de Gastro-entérologie, CHU Hôpital Beaujon, Clichy, France
| | - Divi Cornec
- Service de Rhumatologie, CHRU La Cavale Blanche, Brest, France
| | | | - Philippe Dieudé
- Service de Rhumatologie, CHU Bichat-Claude Bernard, Paris, France
| | | | | | - Nicolas Kluger
- Dpt Dermatology, Helsinki, Finland; Service de Dermatologie, CHU Bichat-Claude Bernard, Paris, France
| | - Estibaliz Lazaro
- Service de Médecine interne, Hôpital Haut-Lévêque, CHU Bordeaux, Pessac, France
| | | | - Victor de Lédinghen
- Unité d'Hépatologie et transplantation hépatique, Hôpital Haut-Lévêque, CHU Bordeaux, Pessac, France
| | | | | | - Raphaèle Seror
- Service de Rhumatologie, Bicêtre, Le Kremlin-Bicêtre, France
| | | | | | - Thao Pham
- Service de Rhumatologie, CHU Sainte-Marguerite, Marseille, France
| | | |
Collapse
|
5
|
Wu M, Tao M, Wang Q, Lu X, Yuan H. Fusion proteins of biologic agents in the treatment of rheumatoid arthritis (RA): A network meta-analysis. Medicine (Baltimore) 2021; 100:e26350. [PMID: 34128886 PMCID: PMC8213327 DOI: 10.1097/md.0000000000026350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/01/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND To evaluate the efficacy of fusion proteins biologics (Etanercept (ETN), Anakinra (ANA), and Abatacept) combinations in the treatment of rheumatoid arthritis (RA) using network meta-analysis to rank those according to their performance medicines. The performance of these processes is ranked according to the results of the analysis and an explanatory study of the possible results is carried out. METHODS Multiple databases including PubMed, EMBASE, and Cochrane Library were used to identify applicable articles and collect relevant data to analyze using STATA (14.0) software. The literature included in this study was divided into a combination of a placebo, methotrexate (MTX), and an observation group (1 of the 3 drugs). The last search date was December 12, 2019. RESULTS A total of 19 eligible randomized controlled trials of fusion proteins biologics were identified, a total of 1109 papers were included, and the results showed that the ETN + MTX had the highest probability of being the most clinically efficacious intervention, with a surface under the cumulative ranking curve of 91.6, was significantly superior (P < .05). Patients who had received ETN or ETN + MTX or ANA had effective compared with patients who had received placebo (95% CI 1.28%-8.47%; 1.92%-19.18%; 1.06%-10.45%). CONCLUSIONS 1. The combination of ETN and MTX had the highest probability of optimal treatment compared to other drugs and 2. ENT, ENT + MTX, and ANA were effective in the treatment of RA compared to placebo.
Collapse
Affiliation(s)
- Mingcai Wu
- Department of Biochemistry and Molecular Biology
| | - Mengjun Tao
- Department of Epidemiology and Biostatistics
| | - Quanhai Wang
- Department of Biochemistry and Molecular Biology
| | - Xiaohua Lu
- Functional experiment and training center, School of Public Health, Wannan Medical College, Wuhu, China
| | - Hui Yuan
- Department of Epidemiology and Biostatistics
| |
Collapse
|
6
|
Hoffmann JHO, Knoop C, Schäkel K, Enk AH, Hadaschik EN. Long-term safety of combination treatment with methotrexate and tumor necrosis factor (TNF)-α antagonists versus TNF-α antagonists alone in psoriatic patients. J Dermatol 2021; 48:835-843. [PMID: 33470023 DOI: 10.1111/1346-8138.15754] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/08/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
Methotrexate, a folic acid analog, is the conventional systemic anti-psoriatic agent most commonly chosen for combination with biologics in the treatment of psoriasis. Real-world long-term safety data of this combination versus biologic treatment alone in dermatological practice are sparse. Here, we present results of a comparative retrospective study of laboratory dynamics and adverse events in psoriatic patients receiving a tumor necrosis factor (TNF)-α antagonist (adalimumab or etanercept) with and without concomitant methotrexate (176 treatment courses, mean duration of 629 days). Co-treatment with methotrexate significantly (P < 0.05) correlated with a decrease of leukocyte, neutrophil and erythrocyte counts and an increase of glutamate pyruvate transaminase (GPT) (Pearson correlation, n > 148). The relative risk for a Common Terminology Criteria for Adverse Events (CTCAE) grade 1-2 laboratory adverse event was significantly elevated to 1.11 for anemia and 1.16 for a GPT increase if the patients received concomitant methotrexate at the time the laboratory test was performed. Combination treatment was given for equal or more than 30% of the time (MTX≥30% ) during 12% of the treatment courses. During these treatment courses, dynamics of leukocyte (-8.1%), neutrophil (-8.1%), erythrocyte (-3.2%) counts and GPT (+16.9%) from baseline to average under treatment were significantly more pronounced. CTCAE grade 3-4 laboratory adverse events occurred in 9.5% and 5.2% of treatment courses with and without MTX≥30% , respectively (p = 0.70), and affected transaminases in 90% of the cases. Methotrexate was discontinued due to CTCAE grade 3-4 laboratory adverse events in 4.25% of the treatment courses with MTX of 30% or more. Elevated baseline γ-glutamyl transferase levels significantly predicted the occurrence of CTCAE grade 3-4 laboratory adverse events and should trigger investigations for pre-existing liver disease or alcohol abuse. In conclusion, our comparative data supplement previous short-term studies and support a tolerable long-term safety profile of the combination treatment. However, given the additional toxicities and low evidence for benefits, alternative options such as biologic monotherapy or switching to a different biologic should be considered in a dermatological setting.
Collapse
Affiliation(s)
| | - Christian Knoop
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Kunt Schäkel
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Alexander H Enk
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Eva N Hadaschik
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
7
|
Lee SH, Yoo YS, Oh HS. The Effect of Adalimumab on Refractory Uveitis. JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2020. [DOI: 10.3341/jkos.2020.61.7.746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. Infectious Complications of Biological and Small Molecule Targeted Immunomodulatory Therapies. Clin Microbiol Rev 2020; 33:e00035-19. [PMID: 32522746 PMCID: PMC7289788 DOI: 10.1128/cmr.00035-19] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The past 2 decades have seen a revolution in our approach to therapeutic immunosuppression. We have moved from relying on broadly active traditional medications, such as prednisolone or methotrexate, toward more specific agents that often target a single receptor, cytokine, or cell type, using monoclonal antibodies, fusion proteins, or targeted small molecules. This change has transformed the treatment of many conditions, including rheumatoid arthritis, cancers, asthma, and inflammatory bowel disease, but along with the benefits have come risks. Contrary to the hope that these more specific agents would have minimal and predictable infectious sequelae, infectious complications have emerged as a major stumbling block for many of these agents. Furthermore, the growing number and complexity of available biologic agents makes it difficult for clinicians to maintain current knowledge, and most review articles focus on a particular target disease or class of agent. In this article, we review the current state of knowledge about infectious complications of biologic and small molecule immunomodulatory agents, aiming to create a single resource relevant to a broad range of clinicians and researchers. For each of 19 classes of agent, we discuss the mechanism of action, the risk and types of infectious complications, and recommendations for prevention of infection.
Collapse
Affiliation(s)
- Joshua S Davis
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - David Ferreira
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Emma Paige
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Craig Gedye
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Oncology, Calvary Mater Hospital, Newcastle, NSW, Australia
| | - Michael Boyle
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
9
|
Wu KC, Zhong Y, Maher J. Predicting Human Infection Risk: Do Rodent Host Resistance Models Add Value? Toxicol Sci 2019; 170:260-272. [DOI: 10.1093/toxsci/kfz116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AbstractUse of genetically engineered rodents is often considered a valuable exercise to assess potential safety concerns associated with the inhibition of a target pathway. When there are potential immunomodulatory risks associated with the target, these genetically modified animals are often challenged with various pathogens in an acute setting to determine the risk to humans. However, the applicability of the results from infection models is seldom assessed when significant retrospective human data become available. Thus, the purpose of the current review is to compare the outcomes of infectious pathogen challenge in mice with genetic deficiencies in TNF-α, IL17, IL23, or Janus kinase pathways with infectious outcomes caused by inhibitors of these pathways in humans. In general, mouse infection challenge models had modest utility for hazard identification and were generally only able to predict overall trends in infection risk. These models did not demonstrate significant value in evaluating specific types of pathogens that are either prevalent (ie rhinoviruses) or of significant concern (ie herpes zoster). Similarly, outcomes in mouse models tended to overestimate the severity of infection risk in human patients. Thus, there is an emerging need for more human-relevant models that have better predictive value. Large meta-analyses of multiple clinical trials or post-marketing evaluations remains the gold-standard for characterizing the true infection risk to patients.
Collapse
Affiliation(s)
- Kai Connie Wu
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| | - Yu Zhong
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| | - Jonathan Maher
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
10
|
Daien C, Hua C, Gaujoux-Viala C, Cantagrel A, Dubremetz M, Dougados M, Fautrel B, Mariette X, Nayral N, Richez C, Saraux A, Thibaud G, Wendling D, Gossec L, Combe B. Update of French society for rheumatology recommendations for managing rheumatoid arthritis. Joint Bone Spine 2019; 86:135-150. [DOI: 10.1016/j.jbspin.2018.10.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2018] [Indexed: 02/07/2023]
|
11
|
Turkish League Against Rheumatism (TLAR) Recommendations for the Pharmacological Management of Rheumatoid Arthritis: 2018 Update Under Guidance of Current Recommendations. Arch Rheumatol 2019; 33:251-271. [PMID: 30632540 DOI: 10.5606/archrheumatol.2018.6911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 05/08/2018] [Indexed: 01/15/2023] Open
Abstract
Objectives This study aims to report the assessment of the Turkish League Against Rheumatism (TLAR) expert panel on the compliance and adaptation of the European League Against Rheumatism (EULAR) 2016 recommendations for the management of rheumatoid arthritis (RA) in Turkey. Patients and methods The EULAR 2016 recommendations for the treatment of RA were voted by 27 specialists experienced in this field with regard to participation rate for each recommendation and significance of items. Afterwards, each recommendation was brought forward for discussion and any alteration gaining ≥70% approval was accepted. Also, Turkish version of each item was rearranged. Last version of the recommendations was then revoted to determine the level of agreement. Levels of agreement of the two voting rounds were compared with Wilcoxon signed-rank test. In case of significant difference, the item with higher level of agreement was accepted. In case of no difference, the changed item was selected. Results Four overarching principles and 12 recommendations were assessed among which three overarching principles and one recommendation were changed. The changed overarching principles emphasized the importance of physical medicine and rehabilitation specialists as well as rheumatologists for the care of RA patients in Turkey. An alteration was made in the eighth recommendation on treatment of active RA patients with unfavorable prognostic indicators after failure of three conventional disease modifying anti-rheumatic drugs. Remaining principles were accepted as the same although some alterations were suggested but could not find adequate support to reach significance. Conclusion Expert opinion of the TLAR for the treatment of RA was composed for practices in Turkish rheumatology and/or physical medicine and rehabilitation clinics.
Collapse
|
12
|
Ma K, Li L, Liu C, Zhou L, Zhou X. Efficacy and safety of various anti-rheumatic treatments for patients with rheumatoid arthritis: a network meta-analysis. Arch Med Sci 2019; 15:33-54. [PMID: 30697252 PMCID: PMC6348345 DOI: 10.5114/aoms.2018.73714] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/22/2017] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Biologics and traditional disease-modifying anti-rheumatic drugs (DMARDs) are generally used in treating patients with rheumatoid arthritis (RA). Previous studies have presented abundant data and information about the efficacy of such treatments, but the results were incomplete and inconclusive. This network meta-analysis was conducted to compare and assess the efficacy and safety of 15 therapies employing biologics and DMARDs for RA patients. MATERIAL AND METHODS Six outcomes (American College of Rheumatology 20% response rate (ACR20), ACR50, ACR70, remission, adverse events (AEs) and serious adverse events (SAEs)) were used to evaluate the efficacy and safety of different treatments. The node-splitting method was used to assess the inconsistency, and the rank probabilities of the therapies were estimated by surface under the cumulative ranking curve. Besides, Jadad scale was used to evaluate the methodological quality of eligible studies. RESULTS A total of 67 randomized controlled trials with 20,898 patients met the inclusion criteria. Most of the therapies presented better performance than conventional DMARDs (cDMARDs) and placebo in ACR20, ACR50 and ACR70. Conversely, the safety of cDMARDs and placebo seemed to be superior in AEs and SAEs. Also, tocilizumab (TCZ) and TCZ + methotrexate (MTX) showed better remission in pain compared to other treatments. Overall, certolizumab pegol (CZP) + MTX and TCZ + MTX had higher probability than the other treatments in efficacy outcomes. CONCLUSIONS We recommend CZP + MTX as the optimal drug therapy because it has the highest ranking in efficacy outcomes and relatively low risk of adverse events. TCZ + MTX is recommended as an alternative. Abatacept (ABT) and cDMARDs are not recommended due to their low efficacy.
Collapse
Affiliation(s)
- Kexun Ma
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ling Li
- Department of Rheumatology, Taizhou Hospital of TCM, Taizhou, Jiangsu, China
| | - Chunhui Liu
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lingling Zhou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xueping Zhou
- The First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Claxton L, Taylor M, Gerber RA, Gruben D, Moynagh D, Singh A, Wallenstein GV. Modelling the cost-effectiveness of tofacitinib for the treatment of rheumatoid arthritis in the United States. Curr Med Res Opin 2018; 34:1991-2000. [PMID: 29976110 DOI: 10.1080/03007995.2018.1497957] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND AND OBJECTIVES Rheumatoid arthritis (RA) is a chronic, debilitating disease affecting an estimated 1.5 million patients in the US. The condition is associated with a substantial health and economic burden. An economic model was developed to evaluate the cost-effectiveness of tofacitinib (a novel oral Janus kinase inhibitor) versus biologic therapies commonly prescribed in the US for the treatment of RA. METHODS A cost-utility model was developed whereby sequences of treatments were evaluated. Response to treatment was modeled by HAQ change, and informed by a network meta-analysis. Mortality, resource use and quality of life were captured in the model using published regression analyses based on HAQ score. Treatment discontinuation was linked to response to treatment and to adverse events. Patients were modeled as having had an inadequate response to methotrexate (MTX-IR), or to a first biologic therapy (TNFi-IR). RESULTS The tofacitinib strategy was associated with cost savings compared with alternative treatment sequences across all modeled scenarios (i.e. in both the MTX-IR and TNFi-IR scenarios), with lifetime cost savings per patient ranging from $65,205 to $93,959 (2015 costs). Cost savings arose due to improved functioning and the resulting savings in healthcare expenditure, and lower drug and administration costs. The tofacitinib strategies all resulted in an increase in quality-adjusted life years (QALYs), with additional QALYs per patient ranging from 0.01 to 0.22. CONCLUSIONS Tofacitinib as a second-line therapy following methotrexate failure and as a third-line therapy following a biologic failure produces lower costs and improved quality of life compared with the current pathway of care.
Collapse
Affiliation(s)
- Lindsay Claxton
- a York Health Economics Consortium , University of York , UK
| | - Matthew Taylor
- a York Health Economics Consortium , University of York , UK
| | - Robert A Gerber
- b Pfizer Incorporated, Global Innovative Products , Groton , CT , USA
| | - David Gruben
- b Pfizer Incorporated, Global Innovative Products , Groton , CT , USA
| | - Dermot Moynagh
- c Pfizer Incorporated, Global Innovative Products , Collegeville , PA , USA
| | - Amitabh Singh
- c Pfizer Incorporated, Global Innovative Products , Collegeville , PA , USA
| | | |
Collapse
|
14
|
Tarp S, Jørgensen TS, Furst DE, Dossing A, Taylor PC, Choy EH, Suarez-Almazor ME, Lyddiatt A, Kristensen LE, Bliddal H, Christensen R. Added value of combining methotrexate with a biological agent compared to biological monotherapy in rheumatoid arthritis patients: A systematic review and meta-analysis of randomised trials. Semin Arthritis Rheum 2018; 48:958-966. [PMID: 30396592 DOI: 10.1016/j.semarthrit.2018.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 01/21/2023]
Abstract
OBJECTIVES To assess the efficacy and safety of methotrexate (MTX) in combination with an approved biological agent compared to biological monotherapy, in the management of patients with rheumatoid arthritis (RA). METHODS MEDLINE, EMBASE, CENTRAL and other sources were searched for randomised trials evaluating a biological agent plus MTX versus the same biological agent in monotherapy. Co-primary outcomes were ACR50 and the number of patients who discontinued due to adverse events (AEs). Random-effects models were applied for meta-analyses with risk ratio and 95% confidence intervals and the GRADE approach was used to assess confidence in the estimates. RESULTS The analysis comprised 16 trials (4965 patients), including all biological agents approved for RA except anakinra and certolizumab. The overall likelihood of responding to therapy (i.e. ACR50) after 6 months was 32% better when MTX was given concomitantly with biological agents (1.32 [1.20-1.45]; P < 0.001) corresponding to 11 more out of 100 patients (7-16 more); Moderate Quality Evidence. Discontinuing due to AEs from concomitant use of MTX was potentially 20% increased (1.21 [0.97-1.50]; P = 0.09) compared to biological monotherapy corresponding to 1 more out of 100 patients (0-3 more); Moderate Quality Evidence. CONCLUSIONS Randomised trials provide Moderate Quality Evidence for a favourable benefit-harm balance supporting concomitant use of MTX rather than monotherapy when prescribing a biological agent in patients with RA although in absolute terms only 7-16 more out of 100 patients will achieve an ACR50 response after 6 months of this combination therapy.
Collapse
Affiliation(s)
- Simon Tarp
- Musculoskeletal Statistics Unit, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Copenhagen, Denmark.
| | - Tanja S Jørgensen
- Musculoskeletal Statistics Unit, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Copenhagen, Denmark
| | - Daniel E Furst
- Division of Rheumatology, Department of Medicine, David Geffen School of Medicine, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA
| | - Anna Dossing
- Musculoskeletal Statistics Unit, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Copenhagen, Denmark
| | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Headington, Oxford, UK
| | - Ernest H Choy
- Section of Rheumatology, Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK; CREATE Centre, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Maria E Suarez-Almazor
- Section of Rheumatology and Clinical Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anne Lyddiatt
- Musculoskeletal Group, Cochrane Collaboration, Ottawa, ON K1H 8L6, Canada
| | - Lars E Kristensen
- Musculoskeletal Statistics Unit, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Copenhagen, Denmark
| | - Henning Bliddal
- Musculoskeletal Statistics Unit, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Copenhagen, Denmark
| | - Robin Christensen
- Musculoskeletal Statistics Unit, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Copenhagen, Denmark
| |
Collapse
|
15
|
Mahmood NMA, Hussain SA, Mirza RR. Azilsartan improves the effects of etanercept in patients with active rheumatoid arthritis: a pilot study. Ther Clin Risk Manag 2018; 14:1379-1385. [PMID: 30122937 PMCID: PMC6086094 DOI: 10.2147/tcrm.s174693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background and aim Much evidence has emerged documenting the involvement of the renin-angiotensin system (RAS) in inflammatory processes. The objective of this study was to evaluate the effects of blocking RAS with azilsartan (Azil) on the clinical efficacy of etanercept (Etan) in patients with active rheumatoid arthritis (RA). Patients and methods Forty-two patients diagnosed with active RA and poorly responding to methotrexate were enrolled in this pilot clinical study. They were randomly allocated into two groups, and treated with either Etan (50 mg/week) and placebo or the same dose of Etan with Azil (20 mg/day) for 90 days. The clinical outcome was evaluated using the Disease Activity Score-28 joint (DAS-28), simplified disease activity index (SDAI), clinical disease activity index (CDAI) and the health assessment questionnaire disease index (HAQ-DI). Blood samples were obtained for the assessment of C-reactive protein and erythrocyte sedimentation rate at baseline and after 90 days. Results The markers of pain and disease activity, C-reactive protein and erythrocyte sedimentation rate were significantly improved when Azil was used, as an adjuvant with Etan, compared with the use of Etan and placebo. Conclusion Blocking RAS with azilsartan may improve the effects of etanercept on the clinical markers of pain and disease severity of patients with active RA not responding to methotrexate.
Collapse
Affiliation(s)
- Naza Mohammed Ali Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Sulaimani, Kurdistan Region, Iraq
| | - Saad Abdulrahman Hussain
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Rafidain University College, Baghdad, Iraq,
| | - Raouf Rahim Mirza
- Department of Medicine, College of Medicine, University of Sulaimani, Kurdistan Region, Iraq
| |
Collapse
|
16
|
Stevenson M, Archer R, Tosh J, Simpson E, Everson-Hock E, Stevens J, Hernandez-Alava M, Paisley S, Dickinson K, Scott D, Young A, Wailoo A. Adalimumab, etanercept, infliximab, certolizumab pegol, golimumab, tocilizumab and abatacept for the treatment of rheumatoid arthritis not previously treated with disease-modifying antirheumatic drugs and after the failure of conventional disease-modifying antirheumatic drugs only: systematic review and economic evaluation. Health Technol Assess 2018; 20:1-610. [PMID: 27140438 DOI: 10.3310/hta20350] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic inflammatory disease associated with increasing disability, reduced quality of life and substantial costs (as a result of both intervention acquisition and hospitalisation). The objective was to assess the clinical effectiveness and cost-effectiveness of seven biologic disease-modifying antirheumatic drugs (bDMARDs) compared with each other and conventional disease-modifying antirheumatic drugs (cDMARDs). The decision problem was divided into those patients who were cDMARD naive and those who were cDMARD experienced; whether a patient had severe or moderate to severe disease; and whether or not an individual could tolerate methotrexate (MTX). DATA SOURCES The following databases were searched: MEDLINE from 1948 to July 2013; EMBASE from 1980 to July 2013; Cochrane Database of Systematic Reviews from 1996 to May 2013; Cochrane Central Register of Controlled Trials from 1898 to May 2013; Health Technology Assessment Database from 1995 to May 2013; Database of Abstracts of Reviews of Effects from 1995 to May 2013; Cumulative Index to Nursing and Allied Health Literature from 1982 to April 2013; and TOXLINE from 1840 to July 2013. Studies were eligible for inclusion if they evaluated the impact of a bDMARD used within licensed indications on an outcome of interest compared against an appropriate comparator in one of the stated population subgroups within a randomised controlled trial (RCT). Outcomes of interest included American College of Rheumatology (ACR) scores and European League Against Rheumatism (EULAR) response. Interrogation of Early Rheumatoid Arthritis Study (ERAS) data was undertaken to assess the Health Assessment Questionnaire (HAQ) progression while on cDMARDs. METHODS Network meta-analyses (NMAs) were undertaken for patients who were cDMARD naive and for those who were cDMARD experienced. These were undertaken separately for EULAR and ACR data. Sensitivity analyses were undertaken to explore the impact of including RCTs with a small proportion of bDMARD experienced patients and where MTX exposure was deemed insufficient. A mathematical model was constructed to simulate the experiences of hypothetical patients. The model was based on EULAR response as this is commonly used in clinical practice in England. Observational databases, published literature and NMA results were used to populate the model. The outcome measure was cost per quality-adjusted life-year (QALY) gained. RESULTS Sixty RCTs met the review inclusion criteria for clinical effectiveness, 38 of these trials provided ACR and/or EULAR response data for the NMA. Fourteen additional trials contributed data to sensitivity analyses. There was uncertainty in the relative effectiveness of the interventions. It was not clear whether or not formal ranking of interventions would result in clinically meaningful differences. Results from the analysis of ERAS data indicated that historical assumptions regarding HAQ progression had been pessimistic. The typical incremental cost per QALY of bDMARDs compared with cDMARDs alone for those with severe RA is > £40,000. This increases for those who cannot tolerate MTX (£50,000) and is > £60,000 per QALY when bDMARDs were used prior to cDMARDs. Values for individuals with moderate to severe RA were higher than those with severe RA. Results produced using EULAR and ACR data were similar. The key parameter that affected the results is the assumed HAQ progression while on cDMARDs. When historic assumptions were used typical incremental cost per QALY values fell to £38,000 for those with severe disease who could tolerate MTX. CONCLUSIONS bDMARDs appear to have cost per QALY values greater than the thresholds stated by the National Institute for Health and Care Excellence for interventions to be cost-effective. Future research priorities include: the evaluation of the long-term HAQ trajectory while on cDMARDs; the relationship between HAQ direct medical costs; and whether or not bDMARDs could be stopped once a patient has achieved a stated target (e.g. remission). STUDY REGISTRATION This study is registered as PROSPERO CRD42012003386. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Matt Stevenson
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Rachel Archer
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Jon Tosh
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Emma Simpson
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Emma Everson-Hock
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - John Stevens
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | | | - Suzy Paisley
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Kath Dickinson
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - David Scott
- Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK
| | - Adam Young
- Department of Rheumatology, West Hertfordshire Hospitals NHS Trust, Hertfordshire, UK
| | - Allan Wailoo
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| |
Collapse
|
17
|
Gohar F, Anink J, Moncrieffe H, Van Suijlekom-Smit LWA, Prince FHM, van Rossum MAJ, Dolman KM, Hoppenreijs EPAH, Ten Cate R, Ursu S, Wedderburn LR, Horneff G, Frosch M, Foell D, Holzinger D. S100A12 Is Associated with Response to Therapy in Juvenile Idiopathic Arthritis. J Rheumatol 2018; 45:547-554. [PMID: 29335345 DOI: 10.3899/jrheum.170438] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Around one-third of patients with juvenile idiopathic arthritis (JIA) fail to respond to first-line methotrexate (MTX) or anti-tumor necrosis factor (TNF) therapy, with even fewer achieving ≥ American College of Rheumatology Pediatric 70% criteria for response (ACRpedi70), though individual responses cannot yet be accurately predicted. Because change in serum S100-protein myeloid-related protein complex 8/14 (MRP8/14) is associated with therapeutic response, we tested granulocyte-specific S100-protein S100A12 as a potential biomarker for treatment response. METHODS S100A12 serum concentration was determined by ELISA in patients treated with MTX (n = 75) and anti-TNF (n = 88) at baseline and followup. Treatment response (≥ ACRpedi50 score), achievement of inactive disease, and improvement in Juvenile Arthritis Disease Activity Score (JADAS)-10 score were recorded. RESULTS Baseline S100A12 concentration was measured in patients treated with anti-TNF [etanercept n = 81, adalimumab n = 7; median 200, interquartile range (IQR) 133-440 ng/ml] and MTX (median 220, IQR 100-440 ng/ml). Of the patients in the anti-TNF therapy group, 74 (84%) were also receiving MTX. Responders to MTX (n = 57/75) and anti-TNF (n = 66/88) therapy had higher baseline S100A12 concentration compared to nonresponders: median 240 (IQR 125-615) ng/ml versus 150 (IQR 87-233) ng/ml, p = 0.021 for MTX, and median 308 (IQR 150-624) ng/ml versus 151 (IQR 83-201) ng/ml, p = 0.002, for anti-TNF therapy. Followup S100A12 could be measured in 44/75 MTX-treated patients (34/44 responders) and 39/88 anti-TNF-treated patients (26/39 responders). Responders had significantly reduced S100A12 concentration (MTX: p = 0.031, anti-TNF: p < 0.001) at followup versus baseline. Baseline serum S100A12 in both univariate and multivariate regression models for anti-TNF therapy and univariate analysis alone for MTX therapy was significantly associated with change in JADAS-10. CONCLUSION Responders to MTX or anti-TNF treatment can be identified by higher pretreatment S100A12 serum concentration levels.
Collapse
Affiliation(s)
- Faekah Gohar
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Janneke Anink
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Halima Moncrieffe
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Lisette W A Van Suijlekom-Smit
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Femke H M Prince
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Marion A J van Rossum
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Koert M Dolman
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Esther P A H Hoppenreijs
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Rebecca Ten Cate
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Simona Ursu
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Lucy R Wedderburn
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Gerd Horneff
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Michael Frosch
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Dirk Foell
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany.,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen
| | - Dirk Holzinger
- From the Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster, Münster, Germany; Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam, Rotterdam, the Netherlands; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA; Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis, Amsterdam; Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre, Nijmegen; Leiden University Medical Centre, Leiden, the Netherlands; School of Biological Sciences, Royal Holloway, University of London; Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street (GOS) Institute of Child Health; UK National Institute for Health Research (NIHR) GOS Hospital Biomedical Research Centre (BRC); Arthritis Research UK Centre for Adolescent Rheumatology at UCL, London, UK; Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin, Sankt Augustin; German Pediatric Pain Centre, Children's and Adolescents' Hospital, Datteln; Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen, Essen, Germany. .,F. Gohar, MD, Department of Pediatric Rheumatology and Immunology, University Children's Hospital Münster; J. Anink, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; H. Moncrieffe, PhD, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati; L.W. Van Suijlekom-Smit, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; F.H. Prince, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Erasmus MC Sophia Children's Hospital Rotterdam; M.A. van Rossum, MD, PhD, Emma Children's Hospital, Academic Medical Centre and Amsterdam Rheumatology and Immunology Centre, Reade location, Jan van Breemen Institute; K.M. Dolman, MD, PhD, Department of Pediatrics/Pediatric Rheumatology, Onze Lieve Vrouwe Gasthuis; E.P. Hoppenreijs, MD, Department of Pediatrics/Pediatric Rheumatology, St. Maartenskliniek and Radboud University Medical Centre; R. ten Cate, MD, PhD, Leiden University Medical Centre; S. Ursu, PhD, School of Biological Sciences, Royal Holloway, University of London; L.R. Wedderburn, MD, PhD, Infection, Immunity, Inflammation Programme, UCL GOS Institute of Child Health, UCL; G. Horneff, MD, Centre of Pediatric Rheumatology, Department of General Pediatrics, Asklepios Clinic Sankt Augustin; M. Frosch, MD, German Pediatric Pain Centre, Children's and Adolescents' Hospital; D. Foell, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster; D. Holzinger, MD, Department of Paediatric Rheumatology and Immunology, University Children's Hospital Münster, and Klinik für Kinderheilkunde III, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Essen.
| |
Collapse
|
18
|
Rheumatoid arthritis patients treated in trial and real world settings: comparison of randomized trials with registries. Rheumatology (Oxford) 2017; 57:354-369. [DOI: 10.1093/rheumatology/kex394] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Indexed: 12/18/2022] Open
|
19
|
Etanercept is effective as monotherapy or in combination with methotrexate in rheumatoid arthritis: subanalysis of an observational study. Clin Rheumatol 2017; 36:1989-1996. [DOI: 10.1007/s10067-017-3757-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/04/2017] [Accepted: 07/07/2017] [Indexed: 01/25/2023]
|
20
|
Singh JA, Hossain A, Tanjong Ghogomu E, Mudano AS, Maxwell LJ, Buchbinder R, Lopez‐Olivo MA, Suarez‐Almazor ME, Tugwell P, Wells GA. Biologics or tofacitinib for people with rheumatoid arthritis unsuccessfully treated with biologics: a systematic review and network meta-analysis. Cochrane Database Syst Rev 2017; 3:CD012591. [PMID: 28282491 PMCID: PMC6472522 DOI: 10.1002/14651858.cd012591] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Biologic disease-modifying anti-rheumatic drugs (DMARDs: referred to as biologics) are effective in treating rheumatoid arthritis (RA), however there are few head-to-head comparison studies. Our systematic review, standard meta-analysis and network meta-analysis (NMA) updates the 2009 Cochrane overview, 'Biologics for rheumatoid arthritis (RA)' and adds new data. This review is focused on biologic or tofacitinib therapy in people with RA who had previously been treated unsuccessfully with biologics. OBJECTIVES To compare the benefits and harms of biologics (abatacept, adalimumab, anakinra, certolizumab pegol, etanercept, golimumab, infliximab, rituximab, tocilizumab) and small molecule tofacitinib versus comparator (placebo or methotrexate (MTX)/other DMARDs) in people with RA, previously unsuccessfully treated with biologics. METHODS On 22 June 2015 we searched for randomized controlled trials (RCTs) in CENTRAL, MEDLINE, and Embase; and trials registries (WHO trials register, Clinicaltrials.gov). We carried out article selection, data extraction, and risk of bias and GRADE assessments in duplicate. We calculated direct estimates with 95% confidence intervals (CI) using standard meta-analysis. We used a Bayesian mixed treatment comparison (MTC) approach for NMA estimates with 95% credible intervals (CrI). We converted odds ratios (OR) to risk ratios (RR) for ease of understanding. We have also presented results in absolute measures as risk difference (RD) and number needed to treat for an additional beneficial outcome (NNTB). Outcomes measured included four benefits (ACR50, function measured by Health Assessment Questionnaire (HAQ) score, remission defined as DAS < 1.6 or DAS28 < 2.6, slowing of radiographic progression) and three harms (withdrawals due to adverse events, serious adverse events, and cancer). MAIN RESULTS This update includes nine new RCTs for a total of 12 RCTs that included 3364 participants. The comparator was placebo only in three RCTs (548 participants), MTX or other traditional DMARD in six RCTs (2468 participants), and another biologic in three RCTs (348 participants). Data were available for four tumor necrosis factor (TNF)-biologics: (certolizumab pegol (1 study; 37 participants), etanercept (3 studies; 348 participants), golimumab (1 study; 461 participants), infliximab (1 study; 27 participants)), three non-TNF biologics (abatacept (3 studies; 632 participants), rituximab (2 studies; 1019 participants), and tocilizumab (2 studies; 589 participants)); there was only one study for tofacitinib (399 participants). The majority of the trials (10/12) lasted less than 12 months.We judged 33% of the studies at low risk of bias for allocation sequence generation, allocation concealment and blinding, 25% had low risk of bias for attrition, 92% were at unclear risk for selective reporting; and 92% had low risk of bias for major baseline imbalance. We downgraded the quality of the evidence for most outcomes to moderate or low due to study limitations, heterogeneity, or rarity of direct comparator trials. Biologic monotherapy versus placeboCompared to placebo, biologics were associated with clinically meaningful and statistically significant improvement in RA as demonstrated by higher ACR50 and RA remission rates. RR was 4.10 for ACR50 (95% CI 1.97 to 8.55; moderate-quality evidence); absolute benefit RD 14% (95% CI 6% to 21%); and NNTB = 8 (95% CI 4 to 23). RR for RA remission was 13.51 (95% CI 1.85 to 98.45, one study available; moderate-quality evidence); absolute benefit RD 9% (95% CI 5% to 13%); and NNTB = 11 (95% CI 3 to 136). Results for withdrawals due to adverse events and serious adverse events did not show any statistically significant or clinically meaningful differences. There were no studies available for analysis for function measured by HAQ, radiographic progression, or cancer outcomes. There were not enough data for any of the outcomes to look at subgroups. Biologic + MTX versus active comparator (MTX/other traditional DMARDs)Compared to MTX/other traditional DMARDs, biologic + MTX was associated with a clinically meaningful and statistically significant improvement in ACR50, function measured by HAQ, and RA remission rates in direct comparisons. RR for ACR50 was 4.07 (95% CI 2.76 to 5.99; high-quality evidence); absolute benefit RD 16% (10% to 21%); NNTB = 7 (95% CI 5 to 11). HAQ scores showed an improvement with a mean difference (MD) of 0.29 (95% CI 0.21 to 0.36; high-quality evidence); absolute benefit RD 9.7% improvement (95% CI 7% to 12%); and NNTB = 5 (95% CI 4 to 7). Remission rates showed an improved RR of 20.73 (95% CI 4.13 to 104.16; moderate-quality evidence); absolute benefit RD 10% (95% CI 8% to 13%); and NNTB = 17 (95% CI 4 to 96), among the biologic + MTX group compared to MTX/other DMARDs. There were no studies for radiographic progression. Results were not clinically meaningful or statistically significantly different for withdrawals due to adverse events or serious adverse events, and were inconclusive for cancer. Tofacitinib monotherapy versus placeboThere were no published data. Tofacitinib + MTX versus active comparator (MTX)In one study, compared to MTX, tofacitinib + MTX was associated with a clinically meaningful and statistically significant improvement in ACR50 (RR 3.24; 95% CI 1.78 to 5.89; absolute benefit RD 19% (95% CI 12% to 26%); NNTB = 6 (95% CI 3 to 14); moderate-quality evidence), and function measured by HAQ, MD 0.27 improvement (95% CI 0.14 to 0.39); absolute benefit RD 9% (95% CI 4.7% to 13%), NNTB = 5 (95% CI 4 to 10); high-quality evidence). RA remission rates were not statistically significantly different but the observed difference may be clinically meaningful (RR 15.44 (95% CI 0.93 to 256.1; high-quality evidence); absolute benefit RD 6% (95% CI 3% to 9%); NNTB could not be calculated. There were no studies for radiographic progression. There were no statistically significant or clinically meaningful differences for withdrawals due to adverse events and serious adverse events, and results were inconclusive for cancer. AUTHORS' CONCLUSIONS Biologic (with or without MTX) or tofacitinib (with MTX) use was associated with clinically meaningful and statistically significant benefits (ACR50, HAQ, remission) compared to placebo or an active comparator (MTX/other traditional DMARDs) among people with RA previously unsuccessfully treated with biologics.No studies examined radiographic progression. Results were not clinically meaningful or statistically significant for withdrawals due to adverse events and serious adverse events, and were inconclusive for cancer.
Collapse
Affiliation(s)
- Jasvinder A Singh
- Birmingham VA Medical CenterDepartment of MedicineFaculty Office Tower 805B510 20th Street SouthBirminghamALUSA35294
| | - Alomgir Hossain
- University of Ottawa Heart InstituteCardiovascular Research Methods Centre40 Ruskin StreetRoom H‐2265OttawaONCanadaK1Y 4W7
| | | | - Amy S Mudano
- University of Alabama at BirminghamDepartment of Medicine ‐ RheumatologyBirminghamUSA
| | - Lara J Maxwell
- Ottawa Hospital Research Institute (OHRI), The Ottawa Hospital ‐ General CampusCentre for Practice‐Changing Research (CPCR)501 Smyth Road, Box 711OttawaONCanadaK1H 8L6
| | - Rachelle Buchbinder
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash UniversityMonash Department of Clinical Epidemiology, Cabrini HospitalSuite 41, Cabrini Medical Centre183 Wattletree RoadMalvernVictoriaAustralia3144
| | - Maria Angeles Lopez‐Olivo
- The University of Texas, M.D. Anderson Cancer CenterDepartment of General Internal Medicine1515 Holcombe BlvdUnit 1465HoustonTexasUSA77030
| | - Maria E Suarez‐Almazor
- The University of Texas, M.D. Anderson Cancer CenterDepartment of General Internal Medicine1515 Holcombe BlvdUnit 1465HoustonTexasUSA77030
| | - Peter Tugwell
- Faculty of Medicine, University of OttawaDepartment of MedicineOttawaONCanadaK1H 8M5
| | - George A Wells
- University of OttawaDepartment of Epidemiology and Community MedicineRoom H128140 Ruskin StreetOttawaONCanadaK1Y 4W7
| | | |
Collapse
|
21
|
Tofacitinib versus Biologic Treatments in Moderate-to-Severe Rheumatoid Arthritis Patients Who Have Had an Inadequate Response to Nonbiologic DMARDs: Systematic Literature Review and Network Meta-Analysis. Int J Rheumatol 2017; 2017:8417249. [PMID: 28377787 PMCID: PMC5362710 DOI: 10.1155/2017/8417249] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/24/2016] [Indexed: 12/16/2022] Open
Abstract
Objective. To compare the efficacy and tolerability of tofacitinib, an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis (RA), as monotherapy and combined with disease-modifying antirheumatic drugs (DMARDs) versus biological DMARDs (bDMARDs) and other novel DMARDs for second-line moderate-to-severe rheumatoid arthritis (RA) patients by means of a systematic literature review (SLR) and network meta-analysis (NMA). Methods. MEDLINE®, EMBASE®, and Cochrane Central Register of Controlled Trials were searched to identify randomized clinical trials (RCTs) published between 1990 and March 2015. Efficacy data based on American College of Rheumatology (ACR) response criteria, improvements in the Health Assessment Questionnaire Disability Index (HAQ-DI) at 6 months, and discontinuation rates due to adverse events were analyzed by means of Bayesian NMAs. Results. 45 RCTs were identified, the majority of which demonstrated a low risk of bias. Tofacitinib 5 mg twice daily (BID) and 10 mg BID monotherapy exhibited comparable efficacy and discontinuation rates due to adverse events versus other monotherapies. Tofacitinib 5 mg BID and 10 mg BID + DMARDs or methotrexate (MTX) were mostly comparable to other combination therapies in terms of efficacy and discontinuation due to adverse events. Conclusion. In most cases, tofacitinib had similar efficacy and discontinuation rates due to adverse events compared to biologic DMARDs.
Collapse
|
22
|
Baradat C, Degboé Y, Constantin A, Cantagrel A, Ruyssen-Witrand A. No impact of concomitant methotrexate use on serious adverse event and serious infection risk in patients with rheumatoid arthritis treated with bDMARDs: a systematic literature review and meta-analysis. RMD Open 2017; 3:e000352. [PMID: 28270933 PMCID: PMC5337718 DOI: 10.1136/rmdopen-2016-000352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVES To compare the risk of serious adverse events, serious infections and death caused by methotrexate and biological disease-modifying antirheumatic drug (bDMARD) combination therapy versus a bDMARD prescribed as monotherapy in rheumatoid arthritis (RA). METHODS A systematic literature review was conducted until February 2016 in PubMed, Embase and Cochrane Library databases by selecting randomised controlled trials comparing methotrexate and bDMARD combination therapy to bDMARD monotherapy in RA. The meta-analysis compared the occurrence of (1) serious adverse events, (2) serious infections and (3) death among these groups by the Mantel-Haenszel method. RESULTS The literature review selected 16 controlled trials comparing methotrexate and bDMARD combination therapy to bDMARD monotherapy. After meta-analysis comparing patients under monotherapy to those under combination therapy: (1) the risk of occurrence of serious adverse events was comparable in 12 trials: RR (95% CI) 0.92 (0.78 to 1.08). (2) No significant difference was observed in the risk of occurrence of serious infections in 13 trials: RR (95% CI) 1.15 (0.84 to 1.58). We noted a trend, although insignificant, towards a high risk of the occurrence of tuberculosis in 10 studies: RR (95% CI) 1.78 (0.63 to 4.99). (3) The risk of death was comparable in 12 trials: RR (95% CI) 0.73 (0.40 to 1.35). CONCLUSIONS The results showed no significant difference between the two groups, confirming that the use of methotrexate and bDMARD combination therapy in RA does not cause an increased risk of serious adverse events or serious infections or death compared with bDMARD monotherapy.
Collapse
Affiliation(s)
- Claire Baradat
- Rheumatology Center, Purpan Teaching Hospital, CHU of Toulouse, Toulouse, France; Paul Sabatier University, Toulouse, France
| | - Yannick Degboé
- Rheumatology Center, Purpan Teaching Hospital, CHU of Toulouse, Toulouse, France; Paul Sabatier University, Toulouse, France; Inserm, UMR 1043, Toulouse, France
| | - Arnaud Constantin
- Rheumatology Center, Purpan Teaching Hospital, CHU of Toulouse, Toulouse, France; Paul Sabatier University, Toulouse, France; Inserm, UMR 1043, Toulouse, France
| | - Alain Cantagrel
- Rheumatology Center, Purpan Teaching Hospital, CHU of Toulouse, Toulouse, France; Paul Sabatier University, Toulouse, France; Inserm, UMR 1043, Toulouse, France
| | - Adeline Ruyssen-Witrand
- Rheumatology Center, Purpan Teaching Hospital, CHU of Toulouse, Toulouse, France; Paul Sabatier University, Toulouse, France; Inserm, UMR 1027, Toulouse, France
| |
Collapse
|
23
|
Stevenson MD, Wailoo AJ, Tosh JC, Hernandez-Alava M, Gibson LA, Stevens JW, Archer RJ, Simpson EL, Hock ES, Young A, Scott DL. The Cost-effectiveness of Sequences of Biological Disease-modifying Antirheumatic Drug Treatment in England for Patients with Rheumatoid Arthritis Who Can Tolerate Methotrexate. J Rheumatol 2017; 44:973-980. [PMID: 28202743 DOI: 10.3899/jrheum.160941] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To ascertain whether strategies of treatment with a biological disease-modifying antirheumatic drug (bDMARD) are cost-effective in an English setting. Results are presented for those patients with moderate to severe rheumatoid arthritis (RA) and those with severe RA. METHODS An economic model to assess the cost-effectiveness of 7 bDMARD was developed. A systematic literature review and network metaanalysis was undertaken to establish relative clinical effectiveness. The results were used to populate the model, together with estimates of Health Assessment Questionnaire (HAQ) score following European League Against Rheumatism response; annual costs, and utility, per HAQ band; trajectory of HAQ for patients taking bDMARD; and trajectory of HAQ for patients using nonbiologic therapy (NBT). Results were presented as those associated with the strategy with the median cost-effectiveness. Supplementary analyses were undertaken assessing the change in cost-effectiveness when only patients with the most severe prognoses taking NBT were provided with bDMARD treatment. The costs per quality-adjusted life-year (QALY) values were compared with reported thresholds from the UK National Institute for Health and Care Excellence of £20,000 to £30,000 (US$24,700 to US$37,000). RESULTS In the primary analyses, the cost per QALY of a bDMARD strategy was £41,600 for patients with severe RA and £51,100 for those with moderate to severe RA. Under the supplementary analyses, the cost per QALY fell to £25,300 for those with severe RA and to £28,500 for those with moderate to severe RA. CONCLUSION The cost-effectiveness of bDMARD in RA in England is questionable and only meets current accepted levels in subsets of patients with the worst prognoses.
Collapse
Affiliation(s)
- Matt D Stevenson
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK. .,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust.
| | - Allan J Wailoo
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - Jonathan C Tosh
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - Monica Hernandez-Alava
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - Laura A Gibson
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - John W Stevens
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - Rachel J Archer
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - Emma L Simpson
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - Emma S Hock
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - Adam Young
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| | - David L Scott
- From the School of Health and Related Research, University of Sheffield, Sheffield; Department of Rheumatology, West Hertfordshire Hospitals National Health Service (NHS) Trust, Hertfordshire; Department of Rheumatology, King's College Hospital NHS Foundation Trust, London, UK.,M.D. Stevenson, PhD, Professor of Health Technology Assessment, School of Health and Related Research, University of Sheffield; A.J. Wailoo, PhD, Professor of Health Economics, School of Health and Related Research, University of Sheffield; J.C. Tosh, PhD, Research Fellow in Health Economics, School of Health and Related Research, University of Sheffield; M. Hernandez-Alava, PhD, Senior Research Fellow in Econometrics, School of Health and Related Research, University of Sheffield; L.A. Gibson, PhD, Research Associate in Econometrics, School of Health and Related Research, University of Sheffield; J.W. Stevens, PhD, Reader in Decision Science, School of Health and Related Research, University of Sheffield; R.J. Archer, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; E.L. Simpson, PhD, Research Fellow, School of Health and Related Research, University of Sheffield, E.S. Hock, PhD, Research Fellow, School of Health and Related Research, University of Sheffield; A. Young, FRCP, Professor of Clinical Rheumatology, Centre for Lifespan and Chronic Illness Research, University of Hertfordshire; D.L. Scott, MD, FRCP, Professor of Clinical Rheumatology, Department of Rheumatology, King's College Hospital NHS Foundation Trust
| |
Collapse
|
24
|
Jørgensen TS, Turesson C, Kapetanovic M, Englund M, Turkiewicz A, Christensen R, Bliddal H, Geborek P, Kristensen LE. EQ-5D utility, response and drug survival in rheumatoid arthritis patients on biologic monotherapy: A prospective observational study of patients registered in the south Swedish SSATG registry. PLoS One 2017; 12:e0169946. [PMID: 28151971 PMCID: PMC5289416 DOI: 10.1371/journal.pone.0169946] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/24/2016] [Indexed: 01/24/2023] Open
Abstract
Objectives Biologic agents have dramatically changed treatment of rheumatoid arthritis (RA). To date only scarce head-to-head data exist especially when the biological therapies are given as monotherapy without concomitant disease modifying drugs (DMARDs). Thus the objective of the current study is to evaluate treatment response of all available biological therapies with special focus on utility (EQ-5D-3L) and drug survival of biologic DMARDs (bDMARDs) prescribed as monotherapy in RA patients in southern Sweden. Materials and methods All RA patients registered in a regional database as initiating bDMARD as monotherapy, i.e. without concomitant conventional synthetic DMARDs (csDMARDs), from 1st of January 2006 through 31st of December 2012, were included. Patients were followed from initiation of the first dose of bDMARD monotherapy treatment until withdrawal from treatment, loss of follow-up or 31st of December 2012. Descriptive statistics for utility (EQ-5D-3L), effectiveness, and drug survival of bDMARD monotherapy were calculated. Results During the study period, a total of 554 patients were registered in SSATG as initiating bDMARD monotherapy. Most of the patients were women (81%), with a mean age of 57 years. The average disease duration was more than 12 years, and on average the patients had previously been treated with approximately four different csDMARDs. Fifty-five percent of the patients were initiating their first bDMARD, 26% their second, and 19% their third or more. At baseline the average EQ-5D-3L was 0.34. Most patients had moderate to high disease activity, with a mean DAS28 of 5.0, and were substantially disabled, with an average HAQ score of 1.4. At 6 months´ follow-up, the EQ-5D-3L in patients still on the biologic drug had increased by mean 0.23 (SD 0.4) with no differences between type of bDMARD (p = 0.49). The mean change in EQ-5D-3L ranged from 0.11 (rituximab and infliximab) to 0.42 (tocilizumab). Although the changes were numerically different, no distinct pattern favored any particular bDMARD for EQ-5D-3L (p = 0.49) or other clinical outcomes. Overall, DAS28 defined remission and low disease activity were achieved in 20% and 43% of patients, respectively. Drug survival rates were statistically significantly different between bDMARDs (p = 0.01), with the highest rates observed for rituximab, followed by etanercept. After failing first course of anti-TNF, patients switching to another mode of action had significantly higher drug survival than those switching to a second course of anti-TNF therapy (p = 0.02). Conclusions Utility (EQ-5D-3L) increased after 6 months of all bDMARD treatments in monotherapy, indicating improvement of patients’ quality of life. After failure of anti-TNF treatment in monotherapy, switching to another mode of action may be associated with better drug survival than starting a second TNF-inhibitor.
Collapse
Affiliation(s)
- Tanja Schjødt Jørgensen
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Copenhagen, Denmark
- * E-mail:
| | - Carl Turesson
- Rheumatology, Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
- Department of Clinical Sciences, Lund, Section of Rheumatology Lund University and Skåne University Hospital, Lund, Sweden
| | - Meliha Kapetanovic
- Department of Clinical Sciences, Lund, Section of Rheumatology Lund University and Skåne University Hospital, Lund, Sweden
- Rheumatology, Department of Clinical Science, Lund University, Lund, Sweden
| | - Martin Englund
- Clinical Epidemiology Unit, Orthopaedics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Aleksandra Turkiewicz
- Clinical Epidemiology Unit, Orthopaedics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Robin Christensen
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Henning Bliddal
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Pierre Geborek
- Department of Clinical Sciences, Lund, Section of Rheumatology Lund University and Skåne University Hospital, Lund, Sweden
- Rheumatology, Department of Clinical Science, Lund University, Lund, Sweden
| | - Lars Erik Kristensen
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Rheumatology, Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
25
|
Fleischmann R, Tongbram V, van Vollenhoven R, Tang DH, Chung J, Collier D, Urs S, Ndirangu K, Wells G, Pope J. Systematic review and network meta-analysis of the efficacy and safety of tumour necrosis factor inhibitor-methotrexate combination therapy versus triple therapy in rheumatoid arthritis. RMD Open 2017; 3:e000371. [PMID: 28123782 PMCID: PMC5237767 DOI: 10.1136/rmdopen-2016-000371] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/18/2016] [Accepted: 11/28/2016] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Clinical trials have not consistently demonstrated differences between tumour necrosis factor inhibitor (TNFi) plus methotrexate and triple therapy (methotrexate plus hydroxychloroquine plus sulfasalazine) in rheumatoid arthritis (RA). The study objective was to estimate the efficacy, radiographic benefits, safety and patient-reported outcomes of TNFi-methotrexate versus triple therapy in patients with RA. METHODS A systematic review and network meta-analysis (NMA) of randomised controlled trials of TNFi-methotrexate or triple therapy as one of the treatment arms in patients with an inadequate response to or who were naive to methotrexate was conducted. American College of Rheumatology 70% response criteria (ACR70) at 6 months was the prespecified primary endpoint to evaluate depth of response. Data from direct and indirect comparisons between TNFi-methotrexate and triple therapy were pooled and quantitatively analysed using fixed-effects and random-effects Bayesian models. RESULTS We analysed 33 studies in patients with inadequate response to methotrexate and 19 in patients naive to methotrexate. In inadequate responders, triple therapy was associated with lower odds of achieving ACR70 at 6 months compared with TNFi-methotrexate (OR 0.35, 95% credible interval (CrI) 0.19 to 0.64). Most secondary endpoints tended to favour TNFi-methotrexate in terms of OR direction; however, no clear increased likelihood of achieving these endpoints was observed for either therapy. The odds of infection were lower with triple therapy than with TNFi-methotrexate (OR 0.08, 95% CrI 0.00 to 0.57). There were no differences observed between the two regimens in patients naive to methotrexate. CONCLUSIONS In this NMA, triple therapy was associated with 65% lower odds of achieving ACR70 at 6 months compared with TNFi-methotrexate in patients with inadequate response to methotrexate. Although secondary endpoints numerically favoured TNFi-methotrexate, no clear differences were observed. The odds of infection were greater with TNFi-methotrexate. No differences were observed for patients naive to methotrexate. These results may help inform care of patients who fail methotrexate first-line therapy.
Collapse
Affiliation(s)
- Roy Fleischmann
- Department of Internal Medicine, University of Texas Southwestern Medical Center and Metroplex Clinical Research Center, Dallas, Texas, USA
| | | | | | - Derek H Tang
- Amgen Inc., Thousand Oaks, California, USA
- Novartis Pharmaceuticals, East Hanover, New Jersey, USA
| | | | | | - Shilpa Urs
- Oxford Outcomes, ICON plc, Morristown, New Jersey, USA
- Doctors’ Hospital of Michigan, Pontiac, Michigan, USA
| | | | - George Wells
- Cardiovascular Research Methods Centre, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Janet Pope
- Department of Rheumatology, St. Joseph's Health Care, London, Ontario, Canada
| |
Collapse
|
26
|
Singh JA, Hossain A, Tanjong Ghogomu E, Mudano AS, Tugwell P, Wells GA. Biologic or tofacitinib monotherapy for rheumatoid arthritis in people with traditional disease-modifying anti-rheumatic drug (DMARD) failure: a Cochrane Systematic Review and network meta-analysis (NMA). Cochrane Database Syst Rev 2016; 11:CD012437. [PMID: 27855242 PMCID: PMC6469573 DOI: 10.1002/14651858.cd012437] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND We performed a systematic review, a standard meta-analysis and network meta-analysis (NMA), which updates the 2009 Cochrane Overview, 'Biologics for rheumatoid arthritis (RA)'. This review is focused on biologic monotherapy in people with RA in whom treatment with traditional disease-modifying anti-rheumatic drugs (DMARDs) including methotrexate (MTX) had failed (MTX/other DMARD-experienced). OBJECTIVES To assess the benefits and harms of biologic monotherapy (includes anti-tumor necrosis factor (TNF) (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab) or non-TNF (abatacept, anakinra, rituximab, tocilizumab)) or tofacitinib monotherapy (oral small molecule) versus comparator (placebo or MTX/other DMARDs) in adults with RA who were MTX/other DMARD-experienced. METHODS We searched for randomized controlled trials (RCTs) in the Cochrane Central Register of Controlled Trials (CENTRAL; The Cochrane Library 2015, Issue 6, June), MEDLINE (via OVID 1946 to June 2015), and Embase (via OVID 1947 to June 2015). Article selection, data extraction and risk of bias and GRADE assessments were done in duplicate. We calculated direct estimates with 95% confidence intervals (CI) using standard meta-analysis. We used a Bayesian mixed treatment comparisons (MTC) approach for NMA estimates with 95% credible intervals (CrI). We converted odds ratios (OR) to risk ratios (RR) for ease of understanding. We calculated absolute measures as risk difference (RD) and number needed to treat for an additional beneficial outcome (NNTB). MAIN RESULTS This update includes 40 new RCTs for a total of 46 RCTs, of which 41 studies with 14,049 participants provided data. The comparator was placebo in 16 RCTs (4,532 patients), MTX or other DMARD in 13 RCTs (5,602 patients), and another biologic in 12 RCTs (3,915 patients). Monotherapy versus placeboBased on moderate-quality direct evidence, biologic monotherapy (without concurrent MTX/other DMARDs) was associated with a clinically meaningful and statistically significant improvement in American College of Rheumatology score (ACR50) and physical function, as measured by the Health Assessment Questionnaire (HAQ) versus placebo. RR was 4.68 for ACR50 (95% CI, 2.93 to 7.48); absolute benefit RD 23% (95% CI, 18% to 29%); and NNTB = 5 (95% CI, 3 to 8). The mean difference (MD) was -0.32 for HAQ (95% CI, -0.42 to -0.23; a negative sign represents greater HAQ improvement); absolute benefit of -10.7% (95% CI, -14% to -7.7%); and NNTB = 4 (95% CI, 3 to 5). Direct and NMA estimates for TNF biologic, non-TNF biologic or tofacitinib monotherapy showed similar results for ACR50 , downgraded to moderate-quality evidence. Direct and NMA estimates for TNF biologic, anakinra or tofacitinib monotherapy showed a similar results for HAQ versus placebo with mostly moderate quality evidence.Based on moderate-quality direct evidence, biologic monotherapy was associated with a clinically meaningful and statistically significant greater proportion of disease remission versus placebo with RR 1.12 (95% CI 1.03 to 1.22); absolute benefit 10% (95% CI, 3% to 17%; NNTB = 10 (95% CI, 8 to 21)).Based on low-quality direct evidence, results for biologic monotherapy for withdrawals due to adverse events and serious adverse events were inconclusive, with wide confidence intervals encompassing the null effect and evidence of an important increase. The direct estimate for TNF monotherapy for withdrawals due to adverse events showed a clinically meaningful and statistically significant result with RR 2.02 (95% CI, 1.08 to 3.78), absolute benefit RD 3% (95% CI,1% to 4%), based on moderate-quality evidence. The NMA estimates for TNF biologic, non-TNF biologic, anakinra, or tofacitinib monotherapy for withdrawals due to adverse events and for serious adverse events were all inconclusive and downgraded to low-quality evidence. Monotherapy versus active comparator (MTX/other DMARDs)Based on direct evidence of moderate quality, biologic monotherapy (without concurrent MTX/other DMARDs) was associated with a clinically meaningful and statistically significant improvement in ACR50 and HAQ scores versus MTX/other DMARDs with a RR of 1.54 (95% CI, 1.14 to 2.08); absolute benefit 13% (95% CI, 2% to 23%), NNTB = 7 (95% CI, 4 to 26) and a mean difference in HAQ of -0.27 (95% CI, -0.40 to -0.14); absolute benefit of -9% (95% CI, -13.3% to -4.7%), NNTB = 2 (95% CI, 2 to 4). Direct and NMA estimates for TNF monotherapy and NMA estimate for non-TNF biologic monotherapy for ACR50 showed similar results, based on moderate-quality evidence. Direct and NMA estimates for non-TNF biologic monotherapy, but not TNF monotherapy, showed similar HAQ improvements , based on mostly moderate-quality evidence.There were no statistically significant or clinically meaningful differences for direct estimates of biologic monotherapy versus active comparator for RA disease remission. NMA estimates showed a statistically significant and clinically meaningful difference versus active comparator for TNF monotherapy (absolute improvement 7% (95% CI, 2% to 14%)) and non-TNF monotherapy (absolute improvement 19% (95% CrI, 7% to 36%)), both downgraded to moderate quality.Based on moderate-quality direct evidence from a single study, radiographic progression (scale 0 to 448) was statistically significantly reduced in those on biologic monotherapy versus active comparator, MD -4.34 (95% CI, -7.56 to -1.12), though the absolute reduction was small, -0.97% (95% CI, -1.69% to -0.25%). We are not sure of the clinical relevance of this reduction.Direct and NMA evidence (downgraded to low quality), showed inconclusive results for withdrawals due to adverse events, serious adverse events and cancer, with wide confidence intervals encompassing the null effect and evidence of an important increase. AUTHORS' CONCLUSIONS Based mostly on RCTs of six to 12-month duration in people with RA who had previously experienced and failed treatment with MTX/other DMARDs, biologic monotherapy improved ACR50, function and RA remission rates compared to placebo or MTX/other DMARDs.Radiographic progression was reduced versus active comparator, although the clinical significance was unclear.Results were inconclusive for whether biologic monotherapy was associated with an increased risk of withdrawals due to adverse events, serious adverse events or cancer, versus placebo (no data on cancer) or MTX/other DMARDs.
Collapse
Affiliation(s)
- Jasvinder A Singh
- Birmingham VA Medical CenterDepartment of MedicineFaculty Office Tower 805B510 20th Street SouthBirminghamALUSA35294
| | - Alomgir Hossain
- University of Ottawa Heart InstituteCardiovascular Research Methods Centre40 Ruskin StreetRoom H‐2265OttawaONCanadaK1Y 4W7
| | | | - Amy S Mudano
- University of Alabama at BirminghamDepartment of Medicine ‐ RheumatologyBirminghamUSA
| | - Peter Tugwell
- Faculty of Medicine, University of OttawaDepartment of MedicineOttawaONCanadaK1H 8M5
| | - George A Wells
- University of OttawaDepartment of Epidemiology and Community MedicineRoom H128140 Ruskin StreetOttawaONCanadaK1Y 4W7
| | | |
Collapse
|
27
|
Recommendations by the Spanish Society of Rheumatology for the management of patients diagnosed with rheumatoid arthritis who cannot be treated with methotrexate. ACTA ACUST UNITED AC 2016; 13:127-138. [PMID: 27825791 DOI: 10.1016/j.reuma.2016.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/23/2016] [Accepted: 10/01/2016] [Indexed: 01/08/2023]
Abstract
To establish a set of recommendations for the management of patients diagnosed with rheumatoid arthritis (RA) who cannot be treated with methotrexate (MTX) due to contraindications, drug toxicity or lack of adherence, and to establish therapeutic strategies more effective and safer in these RA patients. A qualitative analysis of the scientific evidence available to June 2015. The 2-round Delphi technique of consensus was used to collect and establish expert opinion based on the participants' clinical experience when only low quality evidence was available. A total of eighteen recommendations were developed for the management of this patient profile. Fourteen of these recommendations were related to drug safety aspects. Recommendations on contraindication and toxicity of MTX have been updated. The experts recommend the use of biological monotherapy, a preferred treatment option, in patients whose profiles reveal a contraindication, intolerance or circumstances that prevent us against the use of MTX. There is some high-quality scientific evidence that supports contraindication and establishes certain conditions of MTX use in RA patients with specific clinical profiles.
Collapse
|
28
|
Combe B, Furst DE, Keystone EC, van der Heijde D, Luijtens K, Ionescu L, Goel N, Emery P. Certolizumab Pegol Efficacy Across Methotrexate Regimens: A Pre-Specified Analysis of Two Phase III Trials. Arthritis Care Res (Hoboken) 2016; 68:299-307. [PMID: 26238672 PMCID: PMC5067694 DOI: 10.1002/acr.22676] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 06/18/2015] [Accepted: 07/21/2015] [Indexed: 11/07/2022]
Abstract
Objective Anti–tumor necrosis factor (anti‐TNF) agents are frequently used in combination with methotrexate (MTX) to treat rheumatoid arthritis (RA). We investigated the effect of a background MTX dose, in combination with anti‐TNF certolizumab pegol (CZP), on treatment efficacy and safety in RA patients. Methods A pre‐specified subgroup analysis comparing 2 MTX dosage categories (<15 mg/week and ≥15 mg/week) was carried out using data pooled from phase III clinical trials, Rheumatoid Arthritis Prevention of Structural Damage 1 (RAPID 1) and RAPID 2, according to treatment group: CZP 200 mg, CZP 400 mg, or placebo, every 2 weeks. Inclusion criteria required MTX dosage ≥10 mg/week. Efficacy end points included week 24 American College of Rheumatology criteria for 20%, 50%, and 70% improvement (ACR20/50/70) responses analyzed by logistic regression, and changes from baseline in the Disease Activity Score in 28 joints using the erythrocyte sedimentation rate (DAS28‐ESR) and the modified Sharp/van der Heijde score (SHS) were analyzed by analysis of covariance. Incidence rates of treatment‐emergent adverse events (TEAEs) were categorized by baseline MTX dose. Post hoc sensitivity analysis investigated 3 MTX dose categories: ≤10 mg/week, >10 and ≤15 mg/week, and >15 mg/week. Results A total of 638, 635, and 325 patients received CZP 200 mg, CZP 400 mg, and placebo, respectively. At week 24, treatment responses in both CZP groups were uninfluenced by baseline MTX dose category, and were superior to the placebo group for all investigated end points: ACR20/50/70, DAS28‐ESR, and SHS. TEAE incidence rates were higher in patients receiving MTX ≥15 mg/week for most TEAE types across treatment groups. Conclusion CZP efficacy was not affected by background MTX dose category. It can be hypothesized that to minimize TEAEs, background MTX doses could be tailored to individual patient tolerance without affecting CZP efficacy.
Collapse
Affiliation(s)
- Bernard Combe
- Lapeyronie University Hospital, Montpellier I University Hospital, Montpellier, France
| | | | | | | | | | | | | | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and National Institute for Health Research Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals National Health Service Trust, Leeds, UK
| |
Collapse
|
29
|
Singh JA, Hossain A, Tanjong Ghogomu E, Kotb A, Christensen R, Mudano AS, Maxwell LJ, Shah NP, Tugwell P, Wells GA. Biologics or tofacitinib for rheumatoid arthritis in incomplete responders to methotrexate or other traditional disease-modifying anti-rheumatic drugs: a systematic review and network meta-analysis. Cochrane Database Syst Rev 2016; 2016:CD012183. [PMID: 27175934 PMCID: PMC7068903 DOI: 10.1002/14651858.cd012183] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND This is an update of the 2009 Cochrane overview and network meta-analysis (NMA) of biologics for rheumatoid arthritis (RA). OBJECTIVES To assess the benefits and harms of nine biologics (abatacept, adalimumab, anakinra, certolizumab pegol, etanercept, golimumab, infliximab, rituximab, tocilizumab) and small molecule tofacitinib, versus comparator (MTX, DMARD, placebo (PL), or a combination) in adults with rheumatoid arthritis who have failed to respond to methotrexate (MTX) or other disease-modifying anti-rheumatic drugs (DMARDs), i.e., MTX/DMARD incomplete responders (MTX/DMARD-IR). METHODS We searched for randomized controlled trials (RCTs) in the Cochrane Central Register of Controlled Trials (CENTRAL) (via The Cochrane Library Issue 6, June 2015), MEDLINE (via OVID 1946 to June 2015), and EMBASE (via OVID 1947 to June 2015). Data extraction, risk of bias and GRADE assessments were done in duplicate. We calculated both direct estimates using standard meta-analysis and used Bayesian mixed treatment comparisons approach for NMA estimates to calculate odds ratios (OR) and 95% credible intervals (CrI). We converted OR to risk ratios (RR) which are reported in the abstract for the ease of interpretation. MAIN RESULTS This update included 73 new RCTs for a total of 90 RCTs; 79 RCTs with 32,874 participants provided usable data. Few trials were at high risk of bias for blinding of assessors/participants (13% to 21%), selective reporting (4%) or major baseline imbalance (8%); a large number had unclear risk of bias for random sequence generation (68%) or allocation concealment (74%).Based on direct evidence of moderate quality (downgraded for inconsistency), biologic+MTX/DMARD was associated with a statistically significant and clinically meaningful improvement in ACR50 versus comparator (RR 2.71 (95% confidence interval (CI) 2.36 to 3.10); absolute benefit 24% more patients (95% CI 19% to 29%), number needed to treat for an additional beneficial outcome (NNTB) = 5 (4 to 6). NMA estimates for ACR50 in tumor necrosis factor (TNF) biologic+MTX/DMARD (RR 3.23 (95% credible interval (Crl) 2.75 to 3.79), non-TNF biologic+MTX/DMARD (RR 2.99; 95% Crl 2.36 to 3.74), and anakinra + MTX/DMARD (RR 2.37 (95% Crl 1.00 to 4.70) were similar to the direct estimates.Based on direct evidence of moderate quality (downgraded for inconsistency), biologic+MTX/DMARD was associated with a clinically and statistically important improvement in function measured by the Health Assessment Questionnaire (0 to 3 scale, higher = worse function) with a mean difference (MD) based on direct evidence of -0.25 (95% CI -0.28 to -0.22); absolute benefit of -8.3% (95% CI -9.3% to -7.3%), NNTB = 3 (95% CI 2 to 4). NMA estimates for TNF biologic+MTX/DMARD (absolute benefit, -10.3% (95% Crl -14% to -6.7%) and non-TNF biologic+MTX/DMARD (absolute benefit, -7.3% (95% Crl -13.6% to -0.67%) were similar to respective direct estimates.Based on direct evidence of moderate quality (downgraded for inconsistency), biologic+MTX/DMARD was associated with clinically and statistically significantly greater proportion of participants achieving remission in RA (defined by disease activity score DAS < 1.6 or DAS28 < 2.6) versus comparator (RR 2.81 (95% CI, 2.23 to 3.53); absolute benefit 18% more patients (95% CI 12% to 25%), NNTB = 6 (4 to 9)). NMA estimates for TNF biologic+MTX/DMARD (absolute improvement 17% (95% Crl 11% to 23%)) and non-TNF biologic+MTX/DMARD (absolute improvement 19% (95% Crl 12% to 28%) were similar to respective direct estimates.Based on direct evidence of moderate quality (downgraded for inconsistency), radiographic progression (scale 0 to 448) was statistically significantly reduced in those on biologics + MTX/DMARDs versus comparator, MD -2.61 (95% CI -4.08 to -1.14). The absolute reduction was small, -0.58% (95% CI -0.91% to -0.25%) and we are unsure of the clinical relevance of this reduction. NMA estimates of TNF biologic+MTX/DMARD (absolute reduction -0.67% (95% Crl -1.4% to -0.12%) and non-TNF biologic+MTX/DMARD (absolute reduction, -0.68% (95% Crl -2.36% to 0.92%)) were similar to respective direct estimates.Based on direct evidence of moderate quality (downgraded for imprecision), results for withdrawals due to adverse events were inconclusive, with wide confidence intervals encompassing the null effect and evidence of an important increase in withdrawals, RR 1.11 (95% CI 0.96 to 1.30). The NMA estimates of TNF biologic+MTX/DMARD (RR 1.24 (95% Crl 0.99 to 1.57)) and non-TNF biologic+MTX/DMARD (RR 1.20 (95% Crl 0.87 to 1.67)) were similarly inconclusive and downgraded to low for both imprecision and indirectness.Based on direct evidence of high quality, biologic+MTX/DMARD was associated with clinically significantly increased risk (statistically borderline significant) of serious adverse events on biologic+MTX/DMARD (Peto OR [can be interpreted as RR due to low event rate] 1.12 (95% CI 0.99 to 1.27); absolute risk 1% (0% to 2%), As well, the NMA estimate for TNF biologic+MTX/DMARD (Peto OR 1.20 (95% Crl 1.01 to 1.43)) showed moderate quality evidence of an increase in the risk of serious adverse events. The other two NMA estimates were downgraded to low quality due to imprecision and indirectness and had wide confidence intervals resulting in uncertainty around the estimates: non-TNF biologics + MTX/DMARD: 1.07 (95% Crl 0.89 to 1.29) and anakinra: RR 1.06 (95% Crl 0.65 to 1.75).Based on direct evidence of low quality (downgraded for serious imprecision), results were inconclusive for cancer (Peto OR 1.07 (95% CI 0.68 to 1.68) for all biologic+MTX/DMARD combinations. The NMA estimates of TNF biologic+MTX/DMARD (Peto OR 1.21 (95% Crl 0.63 to 2.38) and non-TNF biologic+MTX/DMARD (Peto OR 0.99 (95% Crl 0.58 to 1.78)) were similarly inconclusive and downgraded to low quality for both imprecision and indirectness.Main results text shows the results for tofacitinib and differences between medications. AUTHORS' CONCLUSIONS Based primarily on RCTs of 6 months' to 12 months' duration, there is moderate quality evidence that the use of biologic+MTX/DMARD in people with rheumatoid arthritis who have failed to respond to MTX or other DMARDs results in clinically important improvement in function and higher ACR50 and remission rates, and increased risk of serious adverse events than the comparator (MTX/DMARD/PL; high quality evidence). Radiographic progression is slowed but its clinical relevance is uncertain. Results were inconclusive for whether biologics + MTX/DMARDs are associated with an increased risk of cancer or withdrawals due to adverse events.
Collapse
Affiliation(s)
- Jasvinder A Singh
- Birmingham VA Medical CenterDepartment of MedicineFaculty Office Tower 805B510 20th Street SouthBirminghamALUSA35294
| | - Alomgir Hossain
- University of Ottawa Heart InstituteCardiovascular Research Methods Centre40 Ruskin StreetRoom H‐2265OttawaONCanadaK1Y 4W7
| | | | - Ahmed Kotb
- University of Ottawa Heart InstituteCardiovascular Research Methods Centre40 Ruskin StreetRoom H‐2265OttawaONCanadaK1Y 4W7
| | - Robin Christensen
- Copenhagen University Hospital, Bispebjerg og FrederiksbergMusculoskeletal Statistics Unit, The Parker InstituteNordre Fasanvej 57CopenhagenDenmarkDK‐2000
| | - Amy S Mudano
- University of Alabama at BirminghamDepartment of Medicine ‐ RheumatologyBirminghamUSA
| | - Lara J Maxwell
- Ottawa Hospital Research Institute (OHRI), The Ottawa Hospital ‐ General CampusCentre for Practice‐Changing Research (CPCR)501 Smyth Road, Box 711OttawaONCanadaK1H 8L6
| | - Nipam P Shah
- University of Alabama at BirminghamDepartment of Clinical Immunology and RheumatologyFaculty Office Tower, Suite 805, 510 20th Street SouthBirminghamALUSA35294
| | - Peter Tugwell
- Faculty of Medicine, University of OttawaDepartment of MedicineOttawaONCanadaK1H 8M5
| | - George A Wells
- University of OttawaDepartment of Epidemiology and Community MedicineRoom H128140 Ruskin StreetOttawaONCanadaK1Y 4W7
| | | |
Collapse
|
30
|
Yun H, Xie F, Delzell E, Levitan EB, Chen L, Lewis JD, Saag KG, Beukelman T, Winthrop KL, Baddley JW, Curtis JR. Comparative Risk of Hospitalized Infection Associated With Biologic Agents in Rheumatoid Arthritis Patients Enrolled in Medicare. Arthritis Rheumatol 2016; 68:56-66. [PMID: 26315675 DOI: 10.1002/art.39399] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/18/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The risks of hospitalized infection associated with biologic agents used to treat rheumatoid arthritis (RA) are unclear. The aim of this study was to determine whether the associated risk of hospitalized infections differed between specific biologic agents used to treat RA. METHODS In a retrospective cohort study using Medicare data from 2006-2011 for all enrolled patients with RA, new episodes of treatment with etanercept, adalimumab, certolizumab, golimumab, infliximab, abatacept, rituximab, and tocilizumab were identified. Patients were required to have received another biologic agent previously and to have been continuously enrolled in Medicare medical and pharmacy plans during the baseline period and throughout followup. Followup started on the date of initiation of treatment with the new biologic agent (after previous treatment with a different biologic agent) and ended on the date of the earliest hospitalized infection, at 12 months, after an exposure gap of >30 days, or at the time of death or loss of Medicare coverage. Cox regression analysis was used to calculate the adjusted hazard ratio (HR) for hospitalized infection, adjusting for an infection risk score and other confounders. RESULTS Of 31,801 new biologic treatment episodes in patients who had previously received another biologic agent, 12.0% were with etanercept, 15.2% with adalimumab, 5.9% with certolizumab, 4.4% with golimumab, 12.4% with infliximab, 28.9% with abatacept, 14.8% with rituximab, and 6.3% with tocilizumab. During followup, we identified 2,530 hospitalized infections; incidence rates ranged from 13.1 per 100 person-years (abatacept) to 18.7 per 100 person-years (rituximab). After adjustment, etanercept (HR 1.24, 95% confidence interval [95% CI] 1.07-1.45), infliximab (HR 1.39, 95% CI 1.21-1.60), and rituximab (HR 1.36, 95% CI 1.21-1.53) had significantly higher HRs for hospitalized infection compared with abatacept. CONCLUSION In RA patients with prior exposure to a biologic agent, exposure to etanercept, infliximab, or rituximab was associated with a greater 1-year risk of hospitalized infection compared with the risk associated with exposure to abatacept.
Collapse
|
31
|
Gallo G, Brock F, Kerkmann U, Kola B, Huizinga TWJ. Efficacy of etanercept in combination with methotrexate in moderate-to-severe rheumatoid arthritis is not dependent on methotrexate dosage. RMD Open 2016; 2:e000186. [PMID: 27175292 PMCID: PMC4860865 DOI: 10.1136/rmdopen-2015-000186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 02/02/2016] [Accepted: 02/11/2016] [Indexed: 12/29/2022] Open
Abstract
Objective To evaluate the impact of methotrexate (MTX) dosage on clinical, functional and quality of life outcomes in patients with rheumatoid arthritis (RA) from two previous etanercept (ETN) trials after 24 months of treatment. Methods Patients with active RA in the ETN+MTX combination treatment arms of the Trial of Etanercept and Methotrexate with Radiographic Patient Outcomes (TEMPO) and COmbination of Methotrexate and ETanercept in Active Early Rheumatoid Arthritis (COMET) studies were pooled in this post hoc analysis and stratified by MTX dosage at 24 months, having MTX monotherapy groups as control: low dose, <10.0 mg/week; medium dose, 10.0–17.5 mg/week; and high dose, >17.5 mg/week. Data from these patient subgroups were included in descriptive summaries of demographic and disease characteristics at baseline. The following outcomes at 24 months were also evaluated for each subgroup: Disease Activity Score in 28 joints (DAS28) low disease activity (LDA) and remission; American College of Rheumatology 20%, 50% and 70% improvement criteria (ACR20, 50 and 70) responses; and changes from baseline in DAS28, Health Assessment Questionnaire Disease Index (HAQ-DI) and EuroQol 5-dimensions visual analogue scale (EQ-5D VAS). Results Baseline demographics were similar between the low, medium and high MTX dose groups in the ETN+MTX combination and MTX monotherapy arms, with the exception of disease duration (ETN+MTX low 5.5; medium 5.1; high 0.8 years vs MTX low 8.3; medium 4.7; high 0.8 years). Responses to ETN+MTX combination therapy at 24 months were consistently high across MTX dosage groups, with very similar rates of DAS28 LDA/remission and ACR20/50/70. Improvements in DAS28, HAQ-DI and EQ-5D VAS were also not dependent on MTX dosage in the combination treatment arm. Conclusions Patients with RA in the TEMPO and COMET trials who received ETN+MTX showed similar efficacy outcomes at 24 months, regardless of MTX dosage. Trial registration numbers NCT00195494 (COMET) and NCT00393471 (TEMPO).
Collapse
Affiliation(s)
- G Gallo
- Pfizer Europe , Rome , Italy
| | - F Brock
- Statistical Consultancy, Quanticate Ltd. , Hitchin , UK
| | | | - B Kola
- Pfizer Europe , Rome , Italy
| | - T W J Huizinga
- Leiden University Medical Center , Leiden , The Netherlands
| |
Collapse
|
32
|
Dumitru RB, Horton S, Hodgson R, Wakefield RJ, Hensor EMA, Emery P, Buch MH. A prospective, single-centre, randomised study evaluating the clinical, imaging and immunological depth of remission achieved by very early versus delayed Etanercept in patients with Rheumatoid Arthritis (VEDERA). BMC Musculoskelet Disord 2016; 17:61. [PMID: 26847108 PMCID: PMC4743173 DOI: 10.1186/s12891-016-0915-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 02/02/2016] [Indexed: 12/05/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic inflammatory arthritis, with significant impact on quality of life and functional status. Whilst biologic disease modifying anti-rheumatic drugs (bDMARD) such as tumour necrosis factor-inhibitor (TNFi) agents have revolutionised outcomes in RA, early diagnosis with immediate conventional therapy, titrated in a treat to target approach is also associated with high remission rates. The main aim of the VEDERA study (Very Early versus Delayed Etanercept in Rheumatoid Arthritis) is to assess the depth of remission, sustainability of remission and immunological normalisation induced by very early TNFi with etanercept (ETN) or standard of care +/- delayed ETN. Methods/Design VEDERA is a pragmatic, phase IV single-centre open-label randomised superiority trial of 120 patients with early, treatment-naive RA. Patients will be randomised 1:1 to first-line ETN and methotrexate (MTX) or MTX with additional synthetic disease modifying anti-rheumatic drugs (sDMARDs) according to a treat to target (TT) protocol with further step up to ETN and MTX after 24 weeks if remission is not achieved. Participants will have regular disease activity assessments and imaging evaluation including musculoskeletal ultrasound and MRI. The main objective of this study is to assess the proportion of patients with early RA that achieve clinical remission at 48 weeks, following either treatment strategy. In addition, the participants are invited to take part in a cardio-vascular sub-study (Coronary Artery Disease in RA, CADERA), which aims to identify the incidence of cardiovascular abnormalities in early RA. Discussion The hypothesis underlining this study is that very early treatment with first-line ETN increases the proportion of patients with rheumatoid arthritis achieving clinical remission, in comparison to conventional therapy. Trial registration NCT02433184, 23/04/2015
Collapse
Affiliation(s)
- Raluca B Dumitru
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sarah Horton
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Richard Hodgson
- University of Manchester Centre for Imaging Sciences, Manchester, UK
| | - Richard J Wakefield
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Elizabeth M A Hensor
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Maya H Buch
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK. .,NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
33
|
Singh JA, Saag KG, Bridges SL, Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, Curtis JR, Furst DE, Parks D, Kavanaugh A, O'Dell J, King C, Leong A, Matteson EL, Schousboe JT, Drevlow B, Ginsberg S, Grober J, St Clair EW, Tindall E, Miller AS, McAlindon T. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol 2015; 68:1-26. [PMID: 26545940 DOI: 10.1002/art.39480] [Citation(s) in RCA: 1310] [Impact Index Per Article: 145.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/14/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To develop a new evidence-based, pharmacologic treatment guideline for rheumatoid arthritis (RA). METHODS We conducted systematic reviews to synthesize the evidence for the benefits and harms of various treatment options. We used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) methodology to rate the quality of evidence. We employed a group consensus process to grade the strength of recommendations (either strong or conditional). A strong recommendation indicates that clinicians are certain that the benefits of an intervention far outweigh the harms (or vice versa). A conditional recommendation denotes uncertainty over the balance of benefits and harms and/or more significant variability in patient values and preferences. RESULTS The guideline covers the use of traditional disease-modifying antirheumatic drugs (DMARDs), biologic agents, tofacitinib, and glucocorticoids in early (<6 months) and established (≥6 months) RA. In addition, it provides recommendations on using a treat-to-target approach, tapering and discontinuing medications, and the use of biologic agents and DMARDs in patients with hepatitis, congestive heart failure, malignancy, and serious infections. The guideline addresses the use of vaccines in patients starting/receiving DMARDs or biologic agents, screening for tuberculosis in patients starting/receiving biologic agents or tofacitinib, and laboratory monitoring for traditional DMARDs. The guideline includes 74 recommendations: 23% are strong and 77% are conditional. CONCLUSION This RA guideline should serve as a tool for clinicians and patients (our two target audiences) for pharmacologic treatment decisions in commonly encountered clinical situations. These recommendations are not prescriptive, and the treatment decisions should be made by physicians and patients through a shared decision-making process taking into account patients' values, preferences, and comorbidities. These recommendations should not be used to limit or deny access to therapies.
Collapse
Affiliation(s)
| | | | | | - Elie A Akl
- American University of Beirut, Beirut, Lebanon, and McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | - Deborah Parks
- Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | - Amye Leong
- Healthy Motivation, Santa Barbara, California
| | | | - John T Schousboe
- University of Minnesota and Park Nicollet Clinic, St. Louis Park
| | | | - Seth Ginsberg
- Global Healthy Living Foundation, New York, New York
| | - James Grober
- NorthShore University Health System, Evanston, Illinois
| | | | | | - Amy S Miller
- American College of Rheumatology, Atlanta, Georgia
| | | |
Collapse
|
34
|
Singh JA, Saag KG, Bridges SL, Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, Curtis JR, Furst DE, Parks D, Kavanaugh A, O'Dell J, King C, Leong A, Matteson EL, Schousboe JT, Drevlow B, Ginsberg S, Grober J, St.Clair EW, Tindall E, Miller AS, McAlindon T. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Care Res (Hoboken) 2015; 68:1-25. [DOI: 10.1002/acr.22783] [Citation(s) in RCA: 794] [Impact Index Per Article: 88.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/14/2015] [Indexed: 12/11/2022]
Affiliation(s)
| | | | | | - Elie A. Akl
- American University of Beirut, Beirut, Lebanon, and McMaster University; Hamilton Ontario Canada
| | | | | | | | | | | | | | | | | | - Deborah Parks
- Washington University School of Medicine; St. Louis Missouri
| | | | | | | | - Amye Leong
- Healthy Motivation; Santa Barbara California
| | | | | | | | | | - James Grober
- NorthShore University Health System; Evanston Illinois
| | | | | | | | | |
Collapse
|
35
|
Downey C. Serious infection during etanercept, infliximab and adalimumab therapy for rheumatoid arthritis: A literature review. Int J Rheum Dis 2015. [DOI: 10.1111/1756-185x.12659] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Colum Downey
- Graduate Entry Medical School; University of Limerick; Limerick Ireland
| |
Collapse
|
36
|
Sanmartí R, García-Rodríguez S, Álvaro-Gracia JM, Andreu JL, Balsa A, Cáliz R, Fernández-Nebro A, Ferraz-Amaro I, Gómez-Reino JJ, González-Álvaro I, Martín-Mola E, Martínez-Taboada VM, Ortiz AM, Tornero J, Marsal S, Moreno-Muelas JV. 2014 update of the Consensus Statement of the Spanish Society of Rheumatology on the use of biological therapies in rheumatoid arthritis. ACTA ACUST UNITED AC 2015; 11:279-94. [PMID: 26051464 DOI: 10.1016/j.reuma.2015.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To establish recommendations for the management of patients with rheumatoid arthritis (RA) to serve as a reference for all health professionals involved in the care of these patients, and focusing on the role of available synthetic and biologic disease-modifying antirheumatic drugs (DMARDs). METHODS Consensual recommendations were agreed on by a panel of 14 experts selected by the Spanish Society of Rheumatology (SER). The available scientific evidence was collected by updating three systematic reviews (SR) used for the EULAR 2013 recommendations. A new SR was added to answer an additional question. The literature review of the scientific evidence was made by the SER reviewer's group. The level of evidence and the degree of recommendation was classified according to the Oxford Centre for Evidence-Based Medicine system. A Delphi panel was used to evaluate the level of agreement between panellists (strength of recommendation). RESULTS Thirteen recommendations for the management of adult RA were emitted. The therapeutic objective should be to treat patients in the early phases of the disease with the aim of achieving clinical remission, with methotrexate playing a central role in the therapeutic strategy of RA as the reference synthetic DMARD. Indications for biologic DMARDs were updated and the concept of the optimization of biologicals was introduced. CONCLUSIONS We present the fifth update of the SER recommendations for the management of RA with synthetic and biologic DMARDs.
Collapse
Affiliation(s)
- Raimon Sanmartí
- Servicio de Reumatología, Hospital Clínic de Barcelona, Barcelona, España.
| | | | | | - José Luis Andreu
- Servicio de Reumatología, Hospital Universitario Puerta de Hierro, Madrid, España
| | - Alejandro Balsa
- Servicio de Reumatología, Hospital Universitario La Paz, Madrid, España
| | - Rafael Cáliz
- Servicio de Reumatología, Hospital Universitario Virgen de las Nieves, Granada, España
| | - Antonio Fernández-Nebro
- Unidad de Gestión Clínica de Reumatología, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, España
| | - Iván Ferraz-Amaro
- Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife, España
| | - Juan Jesús Gómez-Reino
- Servicio de Reumatología, Hospital Clínico Universitario de Santiago, Santiago de Compostela, A Coruña, España
| | | | | | | | - Ana M Ortiz
- Servicio de Reumatología, Hospital Universitario de la Princesa, Madrid, España
| | - Jesús Tornero
- Servicio de Reumatología, Hospital Universitario de Guadalajara, Guadalajara, España
| | - Sara Marsal
- Servicio de Reumatología, Hospital Universitario Vall d́Hebron, Barcelona, España
| | - José Vicente Moreno-Muelas
- Servicio de Reumatología, Hospital Universitario Vall d́Hebron, Barcelona, España; Sociedad Española de Reumatología, Madrid, España
| |
Collapse
|
37
|
Abstract
With its approval more than 15 years ago, subcutaneous etanercept (Enbrel(®)) was the first biological disease-modifying antirheumatic drug (bDMARD) and the first tumour necrosis factor inhibitor to be approved for use in rheumatic diseases. Etanercept remains an important cost-effective treatment option in adult patients with rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis or plaque psoriasis, and in paediatric patients with juvenile idiopathic arthritis or plaque psoriasis. In all of these populations, etanercept (with or without methotrexate) effectively reduced signs and symptoms, disease activity and disability, and improved health-related quality of life, with these benefits sustained during long-term treatment. The safety profile of etanercept during short- and long-term treatment was consistent with the approved product labelling, with adverse events being of a predictable and manageable nature. The introduction of etanercept and other bDMARDs as therapeutic options for patients with autoimmune rheumatic diseases and spondyloarthropathies revolutionized disease management and these agents continue to have a central role in treatment strategies. This article reviews the extensive clinical experience with etanercept in these patient populations.
Collapse
Affiliation(s)
- Lesley J Scott
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand,
| |
Collapse
|
38
|
Abstract
Biologics, possibly in combination with a conventional disease-modifying antirheumatic drug (DMARD) - preferably methotrexate (MTX), are used in accordance with the recommendations of the international rheumatological societies. However, in clinical practice, this recommendation is often problematic, as many rheumatologists know from personal experience. The quality of life of the patient is affected mainly by drug-induced intolerances (eg, MTX). Thus, the acceptance of the patient to treatment is often so inadequate that a discontinuation of the drug is necessary. In daily practice, approximately 30% of patients with biological therapy receive no concomitant DMARD according to the register data.
Collapse
Affiliation(s)
- Jacqueline Detert
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Pascal Klaus
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
39
|
Gabay C, Riek M, Scherer A, Finckh A. Effectiveness of biologic DMARDs in monotherapy versus in combination with synthetic DMARDs in rheumatoid arthritis: data from the Swiss Clinical Quality Management Registry. Rheumatology (Oxford) 2015; 54:1664-72. [PMID: 25922549 DOI: 10.1093/rheumatology/kev019] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES To determine the frequency of use of biologic DMARDs (bDMARDs) in monotherapy, to describe the baseline characteristics of patients treated with bDMARDs in monotherapy and to compare the effectiveness of bDMARDs in monotherapy with that of bDMARDs in combination with synthetic DMARDs (sDMARDs). METHODS Using data from the Swiss RA (SCQM-RA) registry, bDMARD treatment courses (TCs) were classified either as monotherapy or as combination therapy, depending on the presence of concomitant sDMARDs. Prescription of bDMARD monotherapy was analysed using logistic regression. bDMARD retention was analysed using Kaplan-Meier and Cox models with the addition of time-varying covariate effects. Evolution of the DAS28 over time was analysed with mixed-effects models for longitudinal data. RESULTS A total of 4218 TCs on bDMARDs from 3111 patients were included, of which 1136 TCs (27%) were initiated as monotherapy. bDMARD monotherapy was preferentially prescribed to older, co-morbid patients with longer disease duration, lower BMI, more active disease and more previous bDMARDs. After adjusting for potential confounding factors, drug retention was significantly lower in monotherapy [hazard ratio 1.15 (95% CI: 1.03, 1.30)]. Other factors such as type of bDMARD and calendar year of prescription were associated with a stronger effect on drug retention. Response to treatment in terms of DAS28 evolution was also slightly but significantly less favourable in monotherapy (P = 0.04). CONCLUSION Our data suggest that bDMARD monotherapy is prescribed to more complex cases and is significantly less effective than bDMARD therapy in combination with sDMARDs, but to an extent that is clinically only marginally relevant.
Collapse
Affiliation(s)
- Cem Gabay
- Division of Rheumatology, Department of Medical Specialties, University Hospitals of Geneva, Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva and
| | - Myriam Riek
- Swiss Clinical Quality Management Foundation, Zurich, Switzerland
| | - Almut Scherer
- Swiss Clinical Quality Management Foundation, Zurich, Switzerland
| | - Axel Finckh
- Division of Rheumatology, Department of Medical Specialties, University Hospitals of Geneva
| | | |
Collapse
|
40
|
Michaud TL, Rho YH, Shamliyan T, Kuntz KM, Choi HK. The comparative safety of tumor necrosis factor inhibitors in rheumatoid arthritis: a meta-analysis update of 44 trials. Am J Med 2014; 127:1208-32. [PMID: 24950486 DOI: 10.1016/j.amjmed.2014.06.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/22/2014] [Accepted: 06/09/2014] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The study objective was to evaluate and update the safety data from randomized controlled trials of tumor necrosis factor inhibitors in patients treated for rheumatoid arthritis. METHODS A systematic literature search was conducted from 1990 to May 2013. All studies included were randomized, double-blind, controlled trials of patients with rheumatoid arthritis that evaluated adalimumab, certolizumab pegol, etanercept, golimumab, or infliximab treatment. The serious adverse events and discontinuation rates were abstracted, and risk estimates were calculated by Peto odds ratios (ORs). RESULTS Forty-four randomized controlled trials involving 11,700 subjects receiving tumor necrosis factor inhibitors and 5901 subjects receiving placebo or traditional disease-modifying antirheumatic drugs were included. Tumor necrosis factor inhibitor treatment as a group was associated with a higher risk of serious infection (OR, 1.42; 95% confidence interval [CI], 1.13-1.78) and treatment discontinuation due to adverse events (OR, 1.23; 95% CI, 1.06-1.43) compared with placebo and traditional disease-modifying antirheumatic drug treatments. Specifically, patients taking adalimumab, certolizumab pegol, and infliximab had an increased risk of serious infection (OR, 1.69, 1.98, and 1.63, respectively) and showed an increased risk of discontinuation due to adverse events (OR, 1.38, 1.67, and 2.04, respectively). In contrast, patients taking etanercept had a decreased risk of discontinuation due to adverse events (OR, 0.72; 95% CI, 0.55-0.93). Although ORs for malignancy varied across the different tumor necrosis factor inhibitors, none reached statistical significance. CONCLUSIONS These meta-analysis updates of the comparative safety of tumor necrosis factor inhibitors suggest a higher risk of serious infection associated with adalimumab, certolizumab pegol, and infliximab, which seems to contribute to higher rates of discontinuation. In contrast, etanercept use showed a lower rate of discontinuation. These data may help guide clinical comparative decision making in the management of rheumatoid arthritis.
Collapse
Affiliation(s)
- Tzeyu L Michaud
- Division of Health Policy and Management, School of Public Health, University of Minnesota, Minneapolis
| | - Young Hee Rho
- Section of Rheumatology and the Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Mass
| | - Tatyana Shamliyan
- Evidence-Based Medicine Quality Assurance Elsevier, Clinical Solutions, Philadelphia, PA
| | - Karen M Kuntz
- Division of Health Policy and Management, School of Public Health, University of Minnesota, Minneapolis
| | - Hyon K Choi
- Section of Rheumatology and the Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Mass.
| |
Collapse
|
41
|
Chiang YC, Kuo LN, Yen YH, Tang CH, Chen HY. Infection risk in patients with rheumatoid arthritis treated with etanercept or adalimumab. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2014; 116:319-327. [PMID: 25022467 DOI: 10.1016/j.cmpb.2014.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/23/2014] [Accepted: 06/13/2014] [Indexed: 06/03/2023]
Abstract
OBJECTIVES To compare the risk of infection for rheumatoid arthritis (RA) patients who took etanercept or adalimumab medication in a nationwide population. METHODS RA patients who took etanercept or adalimumab were identified in the Taiwan's National Health Insurance Research Database. The composite outcome of serious infections, including hospitalization for infection, reception of an antimicrobial injection, and tuberculosis were followed for 365 days. A Kaplan-Meier survival curve with a log-rank test and Cox proportional hazards regression were used to compare risks of infection between the two cohorts of tumor necrosis factor (TNF)-α antagonists users. Hazard ratios (HRs) were obtained and adjusted with propensity scores and clinical factors. Sensitivity analyses and subgroup analyses were also performed. RESULTS In total, 1660 incident etanercept users and 484 incident adalimumab users were eligible for the analysis. The unadjusted HR for infection of the etanercept users was significantly higher than that of the adalimumab users (HR: 1.93; 95% confidence interval (CI): 1.09-3.42; p=0.024). The HRs were 2.04 (95% CI: 1.14-3.65; p=0.016) and 2.02 (95% CI: 1.13-3.61; p=0.018) after adjusting for propensity scores and for propensity scores in addition to clinical factors, respectively. The subgroup analyses revealed that HRs for composite infection was significantly higher in patient subgroups of older age, female, as well as patients who did not have DM, COPD, and hospitalization history at the baseline. CONCLUSION In this head-to-head cohort study involving a nationwide population of patients with RA, etanercept users demonstrated a higher risk of infection than adalimumab users. Results of this study suggest the possible existence of an intra-class difference in infection risk among TNF-α antagonists.
Collapse
Affiliation(s)
- Yi-Chun Chiang
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Li-Na Kuo
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsuan Yen
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chao-Hsiun Tang
- School of Health Care Administration, Taipei Medical University, Taipei, Taiwan
| | - Hsiang-Yin Chen
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
42
|
Treatment comparison in rheumatoid arthritis: head-to-head trials and innovative study designs. BIOMED RESEARCH INTERNATIONAL 2014; 2014:831603. [PMID: 24839607 PMCID: PMC4009266 DOI: 10.1155/2014/831603] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/15/2014] [Indexed: 02/01/2023]
Abstract
Over the last decades, the increasing knowledge in the area of rheumatoid arthritis has progressively expanded the arsenal of available drugs, especially with the introduction of novel targeted therapies such as biological disease modifying antirheumatic drugs (DMARDs). In this situation, rheumatologists are offered a wide range of treatment options, but on the other side the need for comparisons between available drugs becomes more and more crucial in order to better define the strategies for the choice and the optimal sequencing. Indirect comparisons or meta-analyses of data coming from different randomised controlled trials (RCTs) are not immune to conceptual and technical challenges and often provide inconsistent results. In this review we examine some of the possible evolutions of traditional RCTs, such as the inclusion of active comparators, aimed at individualising treatments in real-life conditions. Although head-to-head RCTs may be considered the best tool to directly compare the efficacy and safety of two different DMARDs, surprisingly only 20 studies with such design have been published in the last 25 years. Given the recent advent of the first RCTs truly comparing biological DMARDs, we also review the state of the art of head-to-head trials in RA.
Collapse
|
43
|
Curtis EM, Marks JL. Optimal dose of etanercept in the treatment of rheumatoid arthritis. Open Access Rheumatol 2014; 6:27-38. [PMID: 27790032 PMCID: PMC5045112 DOI: 10.2147/oarrr.s41409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Etanercept (ETN) is one of a number of biological therapies targeting the proinflammatory cytokine tumor necrosis factor-alpha that have demonstrated efficacy in the management of rheumatoid arthritis (RA). As experience has grown, a number of different treatment strategies have been investigated to ascertain the optimal conditions for use of ETN in RA and maximize the clinical gains from therapy. These have included the use of higher- and lower-dose treatment regimens, ETN as a monotherapy or in combination with other nonbiologic disease-modifying antirheumatic drugs, the use of ETN in very early clinical disease, and intraarticular ETN administration for resistant synovitis. Recent trials have focused on phased dose reduction or withdrawal of ETN in patients achieving low disease activity states or clinical remission. This review summarizes existing data regarding the optimal timing of ETN initiation and dosing regimens and also evaluates more recent evidence regarding dose-reduction strategies that offer the possibility of biologic-free remission in RA.
Collapse
Affiliation(s)
- Elizabeth Mary Curtis
- Department of Rheumatology, University Hospital Southampton, Southampton, Hampshire, UK
| | - Jonathan Lewis Marks
- Department of Rheumatology, University Hospital Southampton, Southampton, Hampshire, UK
| |
Collapse
|
44
|
Smolen JS, Landewé R, Breedveld FC, Buch M, Burmester G, Dougados M, Emery P, Gaujoux-Viala C, Gossec L, Nam J, Ramiro S, Winthrop K, de Wit M, Aletaha D, Betteridge N, Bijlsma JWJ, Boers M, Buttgereit F, Combe B, Cutolo M, Damjanov N, Hazes JMW, Kouloumas M, Kvien TK, Mariette X, Pavelka K, van Riel PLCM, Rubbert-Roth A, Scholte-Voshaar M, Scott DL, Sokka-Isler T, Wong JB, van der Heijde D. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis 2014; 73:492-509. [PMID: 24161836 PMCID: PMC3933074 DOI: 10.1136/annrheumdis-2013-204573] [Citation(s) in RCA: 1439] [Impact Index Per Article: 143.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 10/05/2013] [Accepted: 10/11/2013] [Indexed: 02/07/2023]
Abstract
In this article, the 2010 European League against Rheumatism (EULAR) recommendations for the management of rheumatoid arthritis (RA) with synthetic and biological disease-modifying antirheumatic drugs (sDMARDs and bDMARDs, respectively) have been updated. The 2013 update has been developed by an international task force, which based its decisions mostly on evidence from three systematic literature reviews (one each on sDMARDs, including glucocorticoids, bDMARDs and safety aspects of DMARD therapy); treatment strategies were also covered by the searches. The evidence presented was discussed and summarised by the experts in the course of a consensus finding and voting process. Levels of evidence and grades of recommendations were derived and levels of agreement (strengths of recommendations) were determined. Fourteen recommendations were developed (instead of 15 in 2010). Some of the 2010 recommendations were deleted, and others were amended or split. The recommendations cover general aspects, such as attainment of remission or low disease activity using a treat-to-target approach, and the need for shared decision-making between rheumatologists and patients. The more specific items relate to starting DMARD therapy using a conventional sDMARD (csDMARD) strategy in combination with glucocorticoids, followed by the addition of a bDMARD or another csDMARD strategy (after stratification by presence or absence of adverse risk factors) if the treatment target is not reached within 6 months (or improvement not seen at 3 months). Tumour necrosis factor inhibitors (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab, biosimilars), abatacept, tocilizumab and, under certain circumstances, rituximab are essentially considered to have similar efficacy and safety. If the first bDMARD strategy fails, any other bDMARD may be used. The recommendations also address tofacitinib as a targeted sDMARD (tsDMARD), which is recommended, where licensed, after use of at least one bDMARD. Biosimilars are also addressed. These recommendations are intended to inform rheumatologists, patients, national rheumatology societies and other stakeholders about EULAR's most recent consensus on the management of RA with sDMARDs, glucocorticoids and bDMARDs. They are based on evidence and expert opinion and intended to improve outcome in patients with RA.
Collapse
Affiliation(s)
- Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
- 2nd Department of Medicine, Hietzing Hospital Vienna, Vienna, Austria
| | - Robert Landewé
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Atrium Medical Center, Heerlen, The Netherlands
| | - Ferdinand C Breedveld
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maya Buch
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Gerd Burmester
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine, Free University and Humboldt University, Berlin, Germany
- Clinical Immunology Free University and Humboldt University, Berlin, Germany
| | - Maxime Dougados
- Department of Rheumatology B, Cochin Hospital, René Descartes University, Paris, France
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Cécile Gaujoux-Viala
- Department of Rheumatology, Nîmes University Hospital, Montpellier I University, Nimes, France
| | - Laure Gossec
- Rheumatology Department, Paris 06 UPMC University, AP-HP, Pite-Salpetriere Hospital, Paris, France
| | - Jackie Nam
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sofia Ramiro
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Hospital Garcia de Orta, Almada, Portugal
| | - Kevin Winthrop
- Oregon Health and Science University, Portland, Oregon, USA
| | - Maarten de Wit
- EULAR Standing Committee of People with Arthritis/Rheumatism in Europe (PARE), Zurich, Switzerland
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Neil Betteridge
- EULAR Standing Committee of People with Arthritis/Rheumatism in Europe (PARE), Zurich, Switzerland
| | - Johannes W J Bijlsma
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten Boers
- VU University Medical Center, Amsterdam, The Netherlands
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine, Free University and Humboldt University, Berlin, Germany
- Clinical Immunology Free University and Humboldt University, Berlin, Germany
| | - Bernard Combe
- Service d'Immuno-Rhumatologie, Montpellier University, Lapeyronie Hospital, Montpellier, France
| | - Maurizio Cutolo
- Academic Clinical Unit of Rheumatology, Department of Internal Medicine, University of Genova, Genova, Italy
| | - Nemanja Damjanov
- 2nd Hospital Department, Institute of Rheumatology, University of Belgrade Medical School, Belgrade, Serbia
| | - Johanna M W Hazes
- Department of Rheumatology, Erasmus MC, University Medical Center, Dr Molewaterplein, Rotterdam, The Netherlands
| | - Marios Kouloumas
- EULAR Standing Committee of People with Arthritis/Rheumatism in Europe (PARE), Zurich, Switzerland
| | - Tore K Kvien
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Xavier Mariette
- Hopitaux Universitaires Paris Sud, AP-HP, and Université Paris-Sud, Le Kremlin Bicetre, France
| | - Karel Pavelka
- Institute of Rheumatology and Clinic of Rheumatology, Charles University, Prague, Czech Republic
| | - Piet L C M van Riel
- Department of Rheumatology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | - Marieke Scholte-Voshaar
- EULAR Standing Committee of People with Arthritis/Rheumatism in Europe (PARE), Zurich, Switzerland
| | - David L Scott
- King's College School of Medicine, Weston Education Centre, London, UK
| | | | - John B Wong
- Division of Clinical Decision Making, Informatics and Telemedicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
45
|
Atzeni F, Sarzi-Puttini P. Twelve years’ experience with etanercept in the treatment of rheumatoid arthritis: how it has changed clinical practice. Expert Rev Clin Immunol 2014; 8:213-22. [DOI: 10.1586/eci.12.6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
46
|
Nam JL, Ramiro S, Gaujoux-Viala C, Takase K, Leon-Garcia M, Emery P, Gossec L, Landewe R, Smolen JS, Buch MH. Efficacy of biological disease-modifying antirheumatic drugs: a systematic literature review informing the 2013 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2014; 73:516-28. [PMID: 24399231 DOI: 10.1136/annrheumdis-2013-204577] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To update the evidence for the efficacy of biological disease-modifying antirheumatic drugs (bDMARD) in patients with rheumatoid arthritis (RA) to inform the European League Against Rheumatism(EULAR) Task Force treatment recommendations. METHODS Medline, Embase and Cochrane databases were searched for articles published between January 2009 and February 2013 on infliximab, etanercept, adalimumab, certolizumab-pegol, golimumab, anakinra, abatacept, rituximab, tocilizumab and biosimilar DMARDs (bsDMARDs) in phase 3 development. Abstracts from 2011 to 2012 American College of Rheumatology (ACR) and 2011-2013 EULAR conferences were obtained. RESULTS Fifty-one full papers, and 57 abstracts were identified. The randomised controlled trials (RCT) confirmed the efficacy of bDMARD+conventional synthetic DMARDs (csDMARDs) versus csDMARDs alone (level 1B evidence). There was some additional evidence for the use of bDMARD monotherapy, however bDMARD and MTX combination therapy for all bDMARD classes was more efficacious (1B). Clinical and radiographic responses were high with treat-to-target strategies. Earlier improvement in signs and symptoms were seen with more intensive initial treatment strategies, but outcomes were similar upon addition of bDMARDs in patients with insufficient response to MTX. In general, radiographic progression was lower with bDMARD use, mainly due to initial treatment effects. Although patients may achieve bDMARD- and drug-free remission, maintenance of clinical responses was higher with bDMARD continuation (1B), but bDMARD dose reduction could be applied (1B). There was still no RCT data for bDMARD switching. CONCLUSIONS The systematic literature review confirms efficacy of biological DMARDs in RA. It addresses different treatment strategies with the potential for reduction in therapy, particularly with early disease control, and highlights emerging therapies.
Collapse
Affiliation(s)
- Jackie L Nam
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, and NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, , Leeds, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nakajima A, Inoue E, Shidara K, Hoshi D, Sato E, Seto Y, Tanaka E, Taniguchi A, Momohara S, Yamanaka H. Standard treatment in daily clinical practice for early rheumatoid arthritis improved disease activity from 2001 to 2006. Mod Rheumatol 2014. [DOI: 10.3109/s10165-011-0457-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Kameda H, Ueki Y, Saito K, Nagaoka S, Hidaka T, Atsumi T, Tsukano M, Kasama T, Shiozawa S, Tanaka Y, Takeuchi T. Etanercept (ETN) with methotrexate (MTX) is better than ETN monotherapy in patients with active rheumatoid arthritis despite MTX therapy: a randomized trial. Mod Rheumatol 2014. [DOI: 10.3109/s10165-010-0324-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Koike T, Harigai M, Inokuma S, Ishiguro N, Ryu J, Takeuchi T, Tanaka Y, Yamanaka H, Fujii K, Yoshinaga T, Freundlich B, Suzukawa M. Postmarketing surveillance of safety and effectiveness of etanercept in Japanese patients with rheumatoid arthritis. Mod Rheumatol 2014. [DOI: 10.3109/s10165-010-0406-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Practice guidelines for the use of subcutaneous abatacept. ACTA ACUST UNITED AC 2014; 10:218-26. [PMID: 24387951 DOI: 10.1016/j.reuma.2013.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 10/21/2013] [Accepted: 11/06/2013] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To review the clinical evidence on subcutaneous (sc) abatacept and to formulate recommendations in order to clear up points related to its use in rheumatology. METHOD An expert panel of rheumatologists objectively summarized the evidence on the mechanism of action, practicality, effectiveness, and safety of abatacept sc and formulated recommendations after a literature review. RESULTS The efficacy and safety of abatacept sc was studied in 7 clinical trials, 3 double-blind, 3 open, and one mixed, with the following endpoints: comparison against abatacept iv, impact on immunogenicity, effect of replacing iv by sc, abatacept sc in monotherapy, and non-inferiority to adalimumab. No significant differences were found between sc and iv abatacept on efficacy or safety. The development of sc abatacept has allowed a complementary study to the iv, formulation, thus making the abatacept profile better defined. CONCLUSIONS This is a practical document to supplement the summary of product characteristics. In summary, abatacept sc is presented as an effective and safe drug and, therefore, as an alternative for use within the broad armamentarium the rheumatologist has to treat RA. It also has the advantage of being the only biological agent that can be administered iv and sc which can facilitate its use in certain patients.
Collapse
|