1
|
Nakayama Y, Nagata W, Takeuchi Y, Fukui S, Fujita Y, Hosokawa Y, Ueno M, Ono K, Sumitomo S, Tabuchi Y, Nakanishi Y, Saito S, Ikeuchi H, Kawamori K, Sofue H, Doi G, Minami R, Hirota T, Minegishi K, Maeshima K, Motoyama R, Nakamura S, Suzuki S, Nishioka N, Wada TT, Onishi A, Nishimura K, Watanabe R, Yanai R, Kida T, Nishiwaki H, Yajima N, Kaneko Y, Tanaka E, Kawahito Y, Harigai M. Systematic review and meta-analysis for the 2024 update of the Japan College of Rheumatology clinical practice guidelines for the management of rheumatoid arthritis. Mod Rheumatol 2024; 34:1079-1094. [PMID: 38814660 DOI: 10.1093/mr/roae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVES The aim of this article is to update evidence on the efficacy and safety of disease-modifying antirheumatic drugs (DMARDs) and provide information to the taskforce for the 2024 update of the Japan College of Rheumatology clinical practice guidelines for the management of rheumatoid arthritis (RA). METHODS We searched various databases for randomised controlled trials on RA published until June 2022, with no language restriction. For each of the 15 clinical questions, two independent reviewers screened the articles, evaluated the core outcomes, and performed meta-analyses. RESULTS Subcutaneous injection of methotrexate (MTX) showed similar efficacy to oral MTX in MTX-naïve RA patients. Ozoralizumab combined with MTX improved drug efficacy compared to the placebo in RA patients with inadequate response (IR) to conventional synthetic DMARD (csDMARD). Rituximab with and without concomitant csDMARDs showed similar efficacy to other biological DMARDs (bDMARDs) in bDMARD-IR RA patients. Combined Janus kinase inhibitors and MTX achieved similar clinical responses and equal safety during a 4-year period compared to tumour necrosis factor inhibitors in MTX-IR RA patients. Biosimilars showed efficacy equivalent to that of the original bDMARDs in csDMARD-IR and bDMARD-IR RA patients. CONCLUSIONS This systematic review provides latest evidence for the 2024 update of the Japan College of Rheumatology clinical practice guidelines for RA management.
Collapse
Affiliation(s)
- Yoichi Nakayama
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Nagata
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Japan
| | - Yoichi Takeuchi
- Department of Rheumatology and Nephrology, Japanese Red Cross Maebashi Hospital, Maebashi, Japan
| | - Sho Fukui
- Department of Emergency and General Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Yuya Fujita
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yohei Hosokawa
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Masanobu Ueno
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kumiko Ono
- Department of Joint Surgery, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuji Sumitomo
- Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yuya Tabuchi
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichiro Nakanishi
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shuntaro Saito
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroko Ikeuchi
- Department of Preventive Services, School of Public Health, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Kazutaka Kawamori
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hideaki Sofue
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Goro Doi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | - Runa Minami
- Department of Orthopaedic Surgery and Rheumatology, Otokoyama Hospital, Kyoto, Japan
| | - Tomoya Hirota
- Department of Infection and Rheumatology, University of Fukui Hospital, Fukui, Japan
| | - Kaoru Minegishi
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Ryo Motoyama
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Shohei Nakamura
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Shotaro Suzuki
- Division of Rheumatology and Allergology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Norihiro Nishioka
- Department of Preventive Services, School of Public Health, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Takuma Tsuzuki Wada
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Akira Onishi
- Department of Advanced Medicine of Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenichi Nishimura
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryu Watanabe
- Department of Clinical Immunology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Ryo Yanai
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takashi Kida
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Nishiwaki
- Division of Nephrology, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Nobuyuki Yajima
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Eiichi Tanaka
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Yutaka Kawahito
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Kay J, Nikolov NP, Weisman MH. American College of Rheumatology and Food and Drug Administration Summit: Summary of the Meeting, May 17-18, 2022. Arthritis Rheumatol 2024; 76:1323-1333. [PMID: 38622107 DOI: 10.1002/art.42864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/30/2024] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
The American College of Rheumatology and the US Food and Drug Administration co-sponsored a public meeting in May 2022 about challenges in the clinical development of drugs for rheumatoid arthritis (RA) and psoriatic arthritis (PsA), focusing on innovative clinical trial designs, outcome measures, and data collection methods. Recommendations include early dose-ranging studies and use of active comparators. Challenges and opportunities in assessing long-term safety by leveraging real-world data from electronic health records (EHRs) and claims data are discussed, along with insights from European registries and the evolving role of real-world evidence and artificial intelligence in regulatory evaluations. Endpoints for assessing disease activity and outcome measures used in RA and PsA trials are explored, emphasizing challenges in defining remission, assessing clinical response, and evaluating structural progression. The need for outcome measures that better reflect treatment targets and the potential of advanced imaging in future trials are highlighted. Challenges with placebo-controlled trials in RA are discussed and use of non-inferiority clinical trial design, in which new drugs are evaluated with active comparators, is proposed. Pragmatic trials in RA and PsA, employing decentralized approaches, are highlighted for their real-world relevance and administrative efficiencies. Strategies for identifying at-risk populations for RA and the challenges of using EHRs and insurance claims data in drug development are discussed. Registry data and digital health technologies show promise in bridging the gap between clinical trials and real-world effectiveness.
Collapse
Affiliation(s)
- Jonathan Kay
- UMass Chan Medical School and UMass Memorial Medical Center, Worcester, Massachusetts
| | - Nikolay P Nikolov
- Food and Drug Administration Center for Drug Evaluation and Research, Silver Spring, Maryland
| | | |
Collapse
|
3
|
Su QY, Luo J, Zhang Y, Li Q, Jiang ZQ, Wen ZR, Wang YY, Shi MR, Zhang SX. Efficacy and safety of current therapies for difficult-to-treat rheumatoid arthritis: a systematic review and network meta-analysis. J Transl Med 2024; 22:795. [PMID: 39198829 PMCID: PMC11351028 DOI: 10.1186/s12967-024-05569-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/04/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Difficult-to-treat Rheumatoid arthritis (D2T RA) is primarily characterised by failure of at least two different mechanism of action biologic/targeted synthetic disease-modifying antirheumatic drug (DMARDs) with evidence of active/progressive disease. While a variety of drugs have been used in previous studies to treat D2T RA, there has been no systematic summary of these drugs. This study conducted a systematic review of randomized controlled trials aimed at analyzing the efficacy and safety of individual therapeutic agents for the treatment of D2T RA and recommending the optimal therapeutic dose. METHODS The English databases were searched for studies on the treatment of D2T RA published between the date of the database's establishment and March, 2024. This study uses R 3.1.2 for data analysis, and the rjags package runs JAGS 3.4.0.20. The study fitted a stochastic effects Bayesian network meta-analysis for each outcome measure. RESULT A total of 42 studies were included in this study. Compared with placebo, the improvement of Disease Activity Score of 28 Joints (DAS28) score is ranked from high to low as tocilizumab, baricitinib and opinercept. The improvement of American College of Rheumatology 50 response (ACR50) score in patients with drug use was ranked from good to poor as follows: olokizumab, tocilizumab, adalimumab, baricitinib, and upadacitinib, and 8 mg/4w tocilizumab demonstrated the best efficacy. Notably, rituximab is generally the safest drug. Janus kinase (JAK) inhibitors and T cell costimulation modulators are effective in D2T RA refractory to biologic DMARDs, while JAK inhibitors and interleukin-6 (IL-6) inhibitors show effectiveness in D2T RA refractory to csDMARDs. CONCLUSION Tocilizumab and rituximab have better efficacy and safety in the treatment of D2T RA, and the 8 mg/4w dose of tocilizumab may be the first choice for achieving disease remission.
Collapse
Affiliation(s)
- Qin-Yi Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Jing Luo
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Yan Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Qian Li
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Zhong-Qing Jiang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Zi-Rong Wen
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Yu-Ying Wang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Mo-Ran Shi
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, The Shanxi Medical University, Taiyuan, Shanxi, China.
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, China.
| |
Collapse
|
4
|
Schlereth M, Mutlu MY, Utz J, Bayat S, Heimann T, Qiu J, Ehring C, Liu C, Uder M, Kleyer A, Simon D, Roemer F, Schett G, Breininger K, Fagni F. Deep learning-based classification of erosion, synovitis and osteitis in hand MRI of patients with inflammatory arthritis. RMD Open 2024; 10:e004273. [PMID: 38886001 PMCID: PMC11184189 DOI: 10.1136/rmdopen-2024-004273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVES To train, test and validate the performance of a convolutional neural network (CNN)-based approach for the automated assessment of bone erosions, osteitis and synovitis in hand MRI of patients with inflammatory arthritis. METHODS Hand MRIs (coronal T1-weighted, T2-weighted fat-suppressed, T1-weighted fat-suppressed contrast-enhanced) of rheumatoid arthritis (RA) and psoriatic arthritis (PsA) patients from the rheumatology department of the Erlangen University Hospital were assessed by two expert rheumatologists using the Outcome Measures in Rheumatology-validated RA MRI Scoring System and PsA MRI Scoring System scores and were used to train, validate and test CNNs to automatically score erosions, osteitis and synovitis. Scoring performance was compared with human annotations in terms of macro-area under the receiver operating characteristic curve (AUC) and balanced accuracy using fivefold cross-validation. Validation was performed on an independent dataset of MRIs from a second patient cohort. RESULTS In total, 211 MRIs from 112 patients (14 906 region of interests (ROIs)) were included for training/internal validation using cross-validation and 220 MRIs from 75 patients (11 040 ROIs) for external validation of the networks. The networks achieved high mean (SD) macro-AUC of 92%±1% for erosions, 91%±2% for osteitis and 85%±2% for synovitis. Compared with human annotation, CNNs achieved a high mean Spearman correlation for erosions (90±2%), osteitis (78±8%) and synovitis (69±7%), which remained consistent in the validation dataset. CONCLUSIONS We developed a CNN-based automated scoring system that allowed a rapid grading of erosions, osteitis and synovitis with good diagnostic accuracy and using less MRI sequences compared with conventional scoring. This CNN-based approach may help develop standardised cost-efficient and time-efficient assessments of hand MRIs for patients with arthritis.
Collapse
Affiliation(s)
- Maja Schlereth
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Melek Yalcin Mutlu
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jonas Utz
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sara Bayat
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Heimann
- Digital Technology and Innovation, Siemens Healthcare GmbH, Erlangen, Germany
| | - Jingna Qiu
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Chris Ehring
- Institute of Radiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Chang Liu
- Department of Computer Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David Simon
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Roemer
- Institute of Radiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Radiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katharina Breininger
- Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
5
|
Rivellese F, Nerviani A, Giorli G, Warren L, Jaworska E, Bombardieri M, Lewis MJ, Humby F, Pratt AG, Filer A, Gendi N, Cauli A, Choy E, McInnes I, Durez P, Edwards CJ, Buch MH, Gremese E, Taylor PC, Ng N, Cañete JD, Raizada S, McKay ND, Jadon D, Sainaghi PP, Stratton R, Ehrenstein MR, Ho P, Pereira JP, Dasgupta B, Gorman C, Galloway J, Chinoy H, van der Heijde D, Sasieni P, Barton A, Pitzalis C. Stratification of biological therapies by pathobiology in biologic-naive patients with rheumatoid arthritis (STRAP and STRAP-EU): two parallel, open-label, biopsy-driven, randomised trials. THE LANCET. RHEUMATOLOGY 2023; 5:e648-e659. [PMID: 38251532 DOI: 10.1016/s2665-9913(23)00241-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Despite highly effective targeted therapies for rheumatoid arthritis, about 40% of patients respond poorly, and predictive biomarkers for treatment choices are lacking. We did a biopsy-driven trial to compare the response to rituximab, etanercept, and tocilizumab in biologic-naive patients with rheumatoid arthritis stratified for synovial B cell status. METHODS STRAP and STRAP-EU were two parallel, open-label, biopsy-driven, stratified, randomised, phase 3 trials done across 26 university centres in the UK and Europe. Biologic-naive patients aged 18 years or older with rheumatoid arthritis based on American College of Rheumatology (ACR)-European League Against Rheumatism classification criteria and an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (DMARDs) were included. Following ultrasound-guided synovial biopsy, patients were classified as B cell poor or B cell rich according to synovial B cell signatures and randomly assigned (1:1:1) to intravenous rituximab (1000 mg at week 0 and week 2), subcutaneous tocilizumab (162 mg per week), or subcutaneous etanercept (50 mg per week). The primary outcome was the 16-week ACR20 response in the B cell-poor, intention-to-treat population (defined as all randomly assigned patients), with data pooled from the two trials, comparing etanercept and tocilizumab (grouped) versus rituximab. Safety was assessed in all patients who received at least one dose of study drug. These trials are registered with the EU Clinical Trials Register, 2014-003529-16 (STRAP) and 2017-004079-30 (STRAP-EU). FINDINGS Between June 8, 2015, and July 4, 2019, 226 patients were randomly assigned to etanercept (n=73), tocilizumab (n=74), and rituximab (n=79). Three patients (one in each group) were excluded after randomisation because they received parenteral steroids in the 4 weeks before recruitment. 168 (75%) of 223 patients in the intention-to-treat population were women and 170 (76%) were White. In the B cell-poor population, ACR20 response at 16 weeks (primary endpoint) showed no significant differences between etanercept and tocilizumab grouped together and rituximab (46 [60%] of 77 patients vs 26 [59%] of 44; odds ratio 1·02 [95% CI 0·47-2·17], p=0·97). No differences were observed for adverse events, including serious adverse events, which occurred in six (6%) of 102 patients in the rituximab group, nine (6%) of 108 patients in the etanercept group, and three (4%) of 73 patients in the tocilizumab group (p=0·53). INTERPRETATION In this biologic-naive population of patients with rheumatoid arthrtitis, the dichotomic classification into synovial B cell poor versus rich did not predict treatment response to B cell depletion with rituximab compared with alternative treatment strategies. However, the lack of response to rituximab in patients with a pauci-immune pathotype and the higher risk of structural damage progression in B cell-rich patients treated with rituximab warrant further investigations into the ability of synovial tissue analyses to inform disease pathogenesis and treatment response. FUNDING UK Medical Research Council and Versus Arthritis.
Collapse
Affiliation(s)
- Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Giovanni Giorli
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Louise Warren
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Edyta Jaworska
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK
| | - Frances Humby
- Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK; Rheumatology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Directorate of Musculoskeletal Services, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Andrew Filer
- Rheumatology Research Group, Institute for Inflammation and Ageing, NIHR Birmingham Biomedical Research Centre and Clinical Research Facility, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Nagui Gendi
- Basildon University Hospital, Basildon and Thurrock University NHS Hospitals Foundation Trust, Basildon, UK
| | - Alberto Cauli
- Rheumatology Unit, AOU and University of Cagliari, Monserrato, Italy; UOC of Radiology, Ospedale SS Trinità, ATS Cagliari, Italy
| | - Ernest Choy
- CREATE Centre, Cardiff University, Cardiff, UK; Department of Rheumatology, University Hospital of Wales, Cardiff, UK
| | - Iain McInnes
- Glasgow Clinical Research Facility, Glasgow Royal Infirmary, Glasgow, UK
| | - Patrick Durez
- Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium; Department of Rheumatology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christopher J Edwards
- NIHR Southampton Clinical Research Facility, University Hospital Southampton, Southampton, UK; Faculty of Medicine, University of Southampton, Southampton, UK
| | - Maya H Buch
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester, UK
| | - Elisa Gremese
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Nora Ng
- Rheumatology Department, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Juan D Cañete
- Rheumatology Department, Hospital Clínic de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pí I Sunyer, Barcelona, Spain
| | - Sabrina Raizada
- New Cross Hospital and Cannock Chase Hospital, Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Neil D McKay
- Edinburgh Rheumatology Research Group and Rheumatic Diseases Unit, NHS Lothian, Edinburgh, UK
| | - Deepak Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Pier Paolo Sainaghi
- Department of Rheumatology, University Eastern Piedmont and Maggiore della Carita Hospital, Novara, Italy
| | - Richard Stratton
- Royal Free Hospital, Royal Free London NHS Foundation Trust, London, UK
| | | | - Pauline Ho
- The Kellgren Centre for Rheumatology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Joaquim P Pereira
- Rheumatology Department, Hospital De Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal; Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Bhaskar Dasgupta
- Rheumatology Department, Mid and South Essex University Hospitals NHS Foundation Trust, Southend University Hospital, Westcliff-on-Sea, UK
| | - Claire Gorman
- Department of Rheumatology, Homerton University Hospital, Homerton Healthcare NHS Foundation Trust, London, UK
| | - James Galloway
- King's College Hospital, King's College Hospital NHS Foundation Trust, London, UK
| | - Hector Chinoy
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, UK
| | | | - Peter Sasieni
- King's Clinical Trials Unit, Kings College London, London, UK
| | - Anne Barton
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK; Barts Health NHS Trust and Barts Biomedical Research Centre, National Institute for Health and Care Research (NIHR), London, UK; IRCCS Humanitas Research Hospital, Milan, Italy.
| |
Collapse
|
6
|
Wu CY, Yang HY, Lai JH. Potential therapeutic targets beyond cytokines and Janus kinases for autoimmune arthritis. Biochem Pharmacol 2023; 213:115622. [PMID: 37230194 DOI: 10.1016/j.bcp.2023.115622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Synovial inflammation and destruction of articular cartilage and bone are hallmarks of autoimmune arthritis. Although current efforts to inhibit proinflammatory cytokines (biologics) or block Janus kinases (JAK) appear to be promising in many patients with autoimmune arthritis, adequate disease control is still lacking in a significant proportion of autoimmune arthritis patients. The possible adverse events from taking biologics and JAK inhibitors, such as infection, remain a major concern. Recent advances showing the effects of a loss of balance between regulatory T cells and T helper-17 cells as well as how the imbalance between osteoblastic and osteoclastic activities of bone cells exaggerates joint inflammation, bony destruction and systemic osteoporosis highlight an interesting area to explore in the search for better therapeutics. The recognition of the heterogenicity of synovial fibroblasts in osteoclastogenesis and their crosstalk with immune and bone cells provides an opportunity for identifying novel therapeutic targets for autoimmune arthritis. In this commentary, we comprehensively review the current knowledge regarding the interactions among heterogenic synovial fibroblasts, bone cells and immune cells and how they contribute to the immunopathogenesis of autoimmune arthritis, as well as the search for novel therapeutic targets not targeted by current biologics and JAK inhibitors.
Collapse
Affiliation(s)
- Chao-Yi Wu
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Huang-Yu Yang
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan; Graduate Institute of Medical Science, National Defense Medical Center, Taipei 114, Taiwan.
| |
Collapse
|
7
|
Proof-of-Concept Double-Blind Placebo-Controlled Trial Measuring Cartilage Composition in Early Rheumatoid Arthritis under TNF-α-Inhibitor Therapy. J Clin Med 2023; 12:jcm12062306. [PMID: 36983306 PMCID: PMC10051673 DOI: 10.3390/jcm12062306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Low levels of delayed gadolinium-enhanced magnetic resonance imaging of cartilage (dGEMRIC) values are indicative of cartilage degeneration. Patients with early rheumatoid arthritis are known to have low dGEMRIC values due to inflammatory activity. The additional effect of biological disease-modifying antirheumatic drug (bDMARD) and conventional synthetic disease-modifying antirheumatic drug (csDMARD) treatment on cartilage status is still unclear. In this prospective, double-blinded, randomized proof-of-concept clinical trial, patients with early rheumatoid arthritis (disease duration less than 12 months from symptoms onset) were treated with methotrexate + adalimumab (10 patients: 6/4 (f/m)). A control group with methotrexate alone (four patients: 2/2 (f/m)) was used. Cartilage integrity in the metacarpophalangeal joints was compared using dGEMRIC at baseline, 12, and 24 weeks after treatment initiation. A statistically significant increase in dGEMRIC levels was found in the adalimumab group considering the results after 12 and 24 weeks of therapy (p < 0.05) but not in the control group (p: non-significant). After 24 weeks, a tendency towards increased dGEMRIC values under combination therapy was observed, whereas methotrexate alone showed a slight decrease without meeting the criteria of significance (dGEMRIC mean change: +85.8 ms [−156.2–+346.5 ms] vs. 30.75 ms [−273.0–+131.0 ms]; p: non-significant). After 24 weeks of treatment with a combination of methotrexate and adalimumab, a trend indicating improvement in cartilage composition is seen in patients with early rheumatoid arthritis. However, treatment with methotrexate alone showed no change in cartilage composition, as observed in dGEMRIC sequences of metacarpophalangeal joints.
Collapse
|
8
|
Sharma S, Basu S, Goyal RK, Sahoo PK, Mathur R. Rituximab, a Safer Option for Rheumatoid Arthritis: A Comparison of the Reported Adverse Events of Approved Monoclonal Antibodies. J Pharmacol Pharmacother 2023. [DOI: 10.1177/0976500x231154743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Background & Objectives Monoclonal antibodies (mAbs), which are commonly used to treat rheumatoid arthritis (RA), have been linked to a variety of adverse events (AEs). The objective of the study was to compare the safety profiles of six FDA-approved mAbs (sarilumab, tocilizumab, adalimumab, golimumab, infliximab, and rituximab) marketed for the treatment of RA. Methods A systematic review of the literature was conducted using the databases PubMed, Cochrane Library, and Science Direct. The manuscript comprised a total of 23 clinical studies. The percentage of patients who had AEs was calculated and presented using box-whisker and forest plots. Results Infections and infestations were found to be the most common AEs in RA patients treated with mAbs. Raised alanine aminotransferase (ALT), aspartate aminotransferase (AST), upper respiratory tract infection (URTI), and nasopharyngitis were frequently reported. The most common AEs were reported with adalimumab. The highest percentage of patients reporting AEs was associated with golimumab (52%), while rituximab had the fewest AEs (4.9%). Conclusion In conclusion, rituximab appears to be a safer treatment option for RA as it is found to be associated with a lower risk of AEs, particularly respiratory infections.
Collapse
Affiliation(s)
- Sweety Sharma
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, PushpVihar, New Delhi, India
| | - Somnath Basu
- Central Drug Standard Control Organisation, Directorate General of Health Services, Ministry of Health & Family Welfare, Govt. of India, India
| | - Ramesh K. Goyal
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, PushpVihar, New Delhi, India
| | - Parbhat K. Sahoo
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, India
| | - Rajani Mathur
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, Delhi Pharmaceutical Sciences and Research University, PushpVihar, New Delhi, India
| |
Collapse
|
9
|
Fonseca Peixoto R, Ewerton Maia Rodrigues C, Henrique de Sousa Palmeira P, Cézar Comberlang Queiroz Davis Dos Santos F, Keesen de Souza Lima T, de Sousa Braz A. Immune hallmarks of rheumatoid arthritis management: A brief review. Cytokine 2022; 158:156007. [PMID: 35985174 DOI: 10.1016/j.cyto.2022.156007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
The purpose of this review was to examine current evidence on immunomodulation mediated by conventional drugs and the use of novel biological agents for the treatment of rheumatoid arthritis (RA). Currently, treatment is focused on maximizing quality of life through sustained clinical remission and/or attenuating disease activity. To do so, disease-modifying antirheumatic drugs, especially methotrexate, are used alone or in combination with other drugs, including leflunomide, biological disease-modifying antirheumatic drugs (bDMARDs) and targeted synthetic disease-modifying antirheumatic drugs (tsDMARDs). The most recent strategies modulate the immune response of the individual RA patient using tsDMARDs such as JAK inhibitors and bDMARDs such as ig-CTLA-4, anti- IL6R, anti-TNF-α and anti-CD20. To better understand current immunopharmacological interventions, we also looked at documented mechanisms of RA-mediated immunomodulation, highlighting perspectives potentially boosting RA treatment.
Collapse
Affiliation(s)
- Rephany Fonseca Peixoto
- Laboratory of Immunology of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | - Carlos Ewerton Maia Rodrigues
- Post‑Graduate Program in Medical Sciences, Medical School, University of Fortaleza (Unifor), Fortaleza, Brazil; Department of Internal Medicine, Federal University of Ceará, Brazil.
| | - Pedro Henrique de Sousa Palmeira
- Laboratory of Immunology of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | | | - Tatjana Keesen de Souza Lima
- Laboratory of Immunology of Infectious Diseases, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa, Paraíba 58051-900, Brazil
| | | |
Collapse
|
10
|
Somatic Mutations and Autoimmunity. Cells 2021; 10:cells10082056. [PMID: 34440825 PMCID: PMC8394445 DOI: 10.3390/cells10082056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022] Open
Abstract
Autoimmune diseases are among the most common chronic illness caused by a dysregulated immune response against self-antigens. Close to 5% of the general population in Western countries develops some form of autoimmunity, yet its underlying causes, although intensively studied, are still not fully known, and no curative therapies exist. It is well established that autoimmune diseases have common mechanisms and are caused by both genetic and non-genetic risk factors. One novel risk factor that can contribute to autoimmunity is somatic mutations, in a role parallel to their role in cancer. Somatic mutations are stochastic, de novo, non-inherited mutations. In this hypothesis, the persistent proliferation of self-reactive lymphocytes (that is usually hindered by a series of checkpoints) is permitted, due to somatic mutations in these expanding cells, allowing them to bypass multiple regulatory checkpoints, causing autoimmunity. This novel concept of the contribution of these mutations in non-malignant diseases has recently started to be explored. It proposes a novel paradigm for autoimmunity etiology and could be the missing piece of the autoimmunity puzzle.
Collapse
|
11
|
Genovese MC, Berkowitz M, Conaghan PG, Peterfy C, Davy K, Fisheleva E, Gupta A, Inman D, Janiczek R, Layton M, Mitchell N, Patel J, Roberts A, Saurigny D, Smith JE, Williamson R, Tak PP. MRI of the joint and evaluation of the granulocyte-macrophage colony-stimulating factor-CCL17 axis in patients with rheumatoid arthritis receiving otilimab: a phase 2a randomised mechanistic study. THE LANCET. RHEUMATOLOGY 2020; 2:e666-e676. [PMID: 38279363 DOI: 10.1016/s2665-9913(20)30224-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/29/2020] [Accepted: 06/23/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Otilimab is a human monoclonal antibody that inhibits granulocyte-macrophage colony-stimulating factor (GM-CSF), a driver in many immune-mediated inflammatory conditions. We evaluated the effect of otilimab on the GM-CSF-chemokine (C-C motif) ligand 17 (CCL17) axis and synovitis in patients with rheumatoid arthritis. METHODS This phase 2a, randomised, double-blind, multicentre, placebo-controlled, parallel-group study was done at nine sites across the USA, Poland, and Germany. Patients aged 18 years or older with rheumatoid arthritis per American College of Rheumatology-European League Against Rheumatism 2010 criteria and receiving stable methotrexate were randomly assigned (3:1) by an interactive response technology system to either subcutaneous otilimab 180 mg or placebo once weekly for 5 weeks, then every other week until week 10 (within a 12-week treatment period), followed by a 10-week safety follow-up. Randomisation was stratified by early rheumatoid arthritis (≤2 years since diagnosis) and established rheumatoid arthritis (>2 years since diagnosis). Patients and study personnel (except for an unblinded coordinator or nurse who prepared and administered the study drug) were blinded to treatment assignment; the syringe was shielded during administration. Patients were enrolled by study investigators and allocated to a treatment by central randomisation on the basis of a schedule generated by the sponsor. The primary endpoint was change over time (assessed at baseline and weeks 1, 2, 4, 6, 8, 12, and 22 of follow-up) in 112 biomarkers, including target engagement biomarkers and those that may be indicative of rheumatoid arthritis disease activity and response to otilimab. Secondary endpoints were change from baseline in synovitis, osteitis and erosion assessed by rheumatoid arthritis MRI scoring system (RAMRIS) and rheumatoid arthritis MRI quantitative score (RAMRIQ), and safety evaluation. The primary, secondary, and safety endpoints were assessed in the intention-to-treat population. Biomarker and MRI endpoints were analysed for differences between treatment groups using a repeated measures model. This study is registered with ClinicalTrials.gov, NCT02799472. FINDINGS Between Aug 9, 2016, and Oct 30, 2017, 39 patients were randomly assigned and included in the analysis (otilimab n=28; placebo n=11). In the otilimab group, mean serum concentrations of GM-CSF-otilimab complex peaked at week 4 (138·4 ng/L, 95% CI 90·0-212·9) but decreased from week 6-12. CCL17 concentrations decreased from baseline to week 1, remained stable to week 8, and returned to baseline at week 12; least-squares mean ratio to baseline was 0·65 (95% CI 0·49-0·86; coefficient of variation 13·60) at week 2, 0·68 (0·53-0·88; 12·51) at week 4, 0·78 (0·60-1·00; 12·48) at week 6, and 0·68 (0·54-0·85; 11·21) at week 8. No meaningful change in CCL17 concentrations was observed with placebo. In the otilimab group, the least-squares mean ratio to baseline in MMP-degraded type I collagen was 0·86-0·91 over weeks 1-8, returning to baseline at week 12; concentrations remained above baseline at all timepoints in the placebo group. There were no observable differences between otilimab and placebo for all other biomarkers. At week 12, least-squares mean change in RAMRIS synovitis score from baseline was -1·3 (standard error [SE] 0·6) in the otilimab group and 0·8 (1·2) with placebo; RAMRIQ synovitis score showed a least-squares mean change from baseline of -1417·0 μl (671·5) in the otilimab group and -912·3 μl (1405·8) with placebo. Compared with placebo, otilimab did not show significant reductions from baseline to week 12 in RAMRIS synovitis, osteitis and bone erosion, or in RAMRIQ synovitis and erosion damage. Adverse events were reported in 11 (39%) of 28 otilimab-treated and four (36%) of 11 placebo-treated patients, most commonly cough in the otilimab group (2 [7%] of 28; not reported in placebo group), and pain in extremity (four [36%] of 11) and rheumatoid arthritis (two [18%] of 11) in the placebo group (not reported in otilimab group). There were no serious adverse events or deaths. INTERPRETATION Serum concentrations of GM-CSF-otilimab complex indicated that target engagement was achieved with initial weekly dosing, but not sustained with every other week dosing. CCL17 might be a pharmacodynamic biomarker for otilimab activity in future studies. Otilimab was well tolerated and, despite suboptimal exposure, showed some evidence for improved synovitis over 12 weeks in patients with active rheumatoid arthritis. FUNDING GlaxoSmithKline.
Collapse
Affiliation(s)
- Mark C Genovese
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, CA, USA.
| | | | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; National Institute for Health Research Leeds Biomedical Research Centre, Leeds, UK
| | | | - Katherine Davy
- Statistics, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Elena Fisheleva
- ImmunoInflammation, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Anubha Gupta
- Clinical Pharmacology Modeling and Simulation, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - David Inman
- Statistics, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Robert Janiczek
- Experimental Medicine Imaging, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Mark Layton
- ImmunoInflammation, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Nina Mitchell
- ImmunoInflammation, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Jatin Patel
- ImmunoInflammation, GlaxoSmithKline, Stockley Park, Uxbridge, Middlesex, UK
| | - Alexandra Roberts
- Experimental Medicine Imaging, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Didier Saurigny
- ImmunoInflammation, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Julia E Smith
- ImmunoInflammation, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Russell Williamson
- ImmunoInflammation, GlaxoSmithKline, Stockley Park, Uxbridge, Middlesex, UK
| | - Paul P Tak
- Research and Development, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
12
|
Genovese MC, Gaylis NB, Sikes D, Kivitz A, Lewis Horowitz D, Peterfy C, Glass EV, Levine YA, Chernoff D. Safety and efficacy of neurostimulation with a miniaturised vagus nerve stimulation device in patients with multidrug-refractory rheumatoid arthritis: a two-stage multicentre, randomised pilot study. THE LANCET. RHEUMATOLOGY 2020; 2:e527-e538. [PMID: 38273617 DOI: 10.1016/s2665-9913(20)30172-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/02/2023]
Abstract
Background The inflammatory reflex plays a role in regulating innate and adaptive immunity by modulating cellular and molecular inflammatory pathways. The vagus nerve is a major constituent of the inflammatory reflex and studies have shown that the reflex can be activated by electrical stimulation of the vagus nerve. In this first in-human pilot study, we assessed the safety and efficacy of a novel miniaturised vagus nerve stimulation (VNS) device for the treatment of multidrug-refractory rheumatoid arthritis. METHODS Participants with moderately to severely active rheumatoid arthritis and prior insufficient response to two or more biological disease-modifying anti-rheumatic drugs or Janus kinase inhibitors with at least two different modes of action were enrolled in a two-stage study done at five clinical research sites in the USA. Stage 1 was open label; participants were implanted with a miniaturised VNS device, which was activated for 1 min once a day. In stage 2, participants were randomly assigned (1:1:1) to receive active stimulation (1 min once a day or 1 min four times a day) or sham stimulation (device implanted but not activated), with the sites and participants masked to treatment assignment. The primary outcome was incidence of treatment-emergent adverse events. Clinical efficacy was assessed as a key secondary outcome. The study was registered with ClinicalTrials.gov, NCT03437473. FINDINGS 14 patients were enrolled between March 13 and Aug 8, 2018. Three patients received stimulation in stage 1 and, following safety review board approval, the remaining 11 patients were implanted during stage 2 and randomly assigned to receive 1 min of stimulation once daily (n=3), 1 min of stimulation four times daily (n=4), or no stimulation (n=4) for 12 weeks. There were no device-related or treatment-related serious adverse events. Surgery-related adverse events were Horner's syndrome and vocal cord paralysis (in one patient each), which resolved without clinically significant sequelae. No deaths were recorded. INTERPRETATION VNS with a miniaturised neurostimulator was safe and well tolerated and reduced signs and symptoms of rheumatoid arthritis in patients with multidrug-refractory disease. These results support further evaluation in a larger randomised sham-controlled study. FUNDING SetPoint Medical.
Collapse
Affiliation(s)
- Mark C Genovese
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, CA, USA.
| | - Norman B Gaylis
- Arthritis and Rheumatic Disease Specialties, Aventura, FL, USA
| | - David Sikes
- Department of Rheumatology, Florida Medical Clinic, Zephyrhills, FL, USA
| | - Alan Kivitz
- Altoona Center for Clinical Research, Duncansville, PA, USA
| | | | | | | | | | | |
Collapse
|
13
|
Caporali R, Fakhouri WKH, Nicolay C, Longley HJ, Losi S, Rogai V. New Rheumatoid Arthritis Treatments for 'Old' Patients: Results of a Systematic Review. Adv Ther 2020; 37:3676-3691. [PMID: 32705531 PMCID: PMC7444401 DOI: 10.1007/s12325-020-01435-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION In the last 20 years, biologic and targeted synthetic disease-modifying antirheumatic drugs (DMARDs) have become available for treating rheumatoid arthritis (RA), and a treat-to-target strategy has been introduced. We hypothesise that these advances should have resulted in changes to the characteristics of patients with RA participating in clinical trials of the newest therapies. This study determined whether the baseline characteristics of patients with RA enrolled in clinical trials have changed in the past decade versus patients participating in earlier RA studies. METHODS This secondary analysis was based on randomised controlled trials (RCTs) identified in a systematic literature review. Baseline characteristics of patients with RA with inadequate response to conventional synthetic DMARDs were compared between RCTs published in 1999-2009 and those published in 2010-2017 using random-effects meta-analyses. RESULTS Forty RCTs were analysed: 22 from 1999-2009 and 18 from 2010-2017. No significant difference between the two timeframes and no obvious trend over time were observed for age, gender, disease duration, rheumatoid factor status, tender and swollen joint counts, physician and patient global assessments of disease activity, and pain scores. Variability between RCTs was high. Similar results were observed for Disease Activity Scores and Health Assessment Questionnaire-Disability Index scores, but with low variability between RCTs. CONCLUSION The baseline characteristics of patients with RA participating in RCTs do not appear to have changed in the last decade despite the availability of new treatments and a different treatment approach. Further research should determine the impact of baseline patient characteristics on patients' response to RA treatments.
Collapse
Affiliation(s)
- Roberto Caporali
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
- Rheumatology Unit, ASST Gaetano Pini-CTO Hospital, Milan, Italy.
| | | | | | | | - Serena Losi
- Eli Lilly Italy S.p.A., Sesto Fiorentino, Italy
| | | |
Collapse
|
14
|
Park EH, Yoon CH, Kang EH, Baek HJ. Utility of Magnetic Resonance Imaging and Positron Emission Tomography in Rheumatic Diseases. JOURNAL OF RHEUMATIC DISEASES 2020. [DOI: 10.4078/jrd.2020.27.3.136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Eun Hye Park
- Division of Rheumatology, Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Korea
| | - Chong-Hyeon Yoon
- Division of Rheumatology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Eun Ha Kang
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Han Joo Baek
- Division of Rheumatology, Department of Internal Medicine, Gachon University College of Medicine Gil Medical Center, Incheon, Korea
| |
Collapse
|
15
|
Fakhouri W, Wang X, de La Torre I, Nicolay C. A Network Meta-Analysis to Compare Effectiveness of Baricitinib and Other Treatments in Rheumatoid Arthritis Patients with Inadequate Response to Methotrexate. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2020; 7:10-23. [PMID: 32685594 PMCID: PMC7299462 DOI: 10.36469/jheor.2020.12273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND/OBJECTIVES This article compares the effectiveness of baricitinib (BARI) 4 mg (oral, Janus kinase [JAK] 1/2 inhibitor) versus other targeted synthetic/biologic disease-modifying antirheumatic drugs, in combination with methotrexate (MTX), in moderate-to-severe rheumatoid arthritis patients with inadequate response (IR) to MTX. METHODS A systematic literature review was conducted to identify randomized controlled trials (RCTs) of the interventions of interest. Bayesian network meta-analyses (NMA) were used to compare American College of Rheumatology (ACR) responses at 24 weeks. A series of prespecified sensitivity analyses addressed the potential impact of, among others, baseline risk, treatment effect modifiers, and trial design on treatment response. RESULTS Nineteen RCTs were included in the NMA (primary analysis). For ACR20, BARI 4 mg + MTX was found to be more effective than adalimumab (ADA) 40 mg + MTX (Odds Ratio [OR] 1.33), abatacept (ABA) 10 mg + MTX (IV/4 weeks) (OR 1.45), infliximab (IFX) 3 mg + MTX (IV/8 wks) (OR 1.63), and rituximab (RTX) 1000 mg + MTX (OR 1.63). No differences were found on ACR50. For ACR70, BARI 4 mg + MTX was more effective than ADA 40 mg + MTX (OR 1.37), ABA 10 mg + MTX (OR 1.86), and RTX 1000 mg + MTX (OR 2.26). Sensitivity analysis including 10 additional RCTs with up to 20% of patients with prior biologic use showed BARI 4 mg + MTX to be more effective than tocilizumab (TCZ) 8 mg + MTX on ACR20 (OR 1.44). Results for all sensitivity analyses were consistent with the direction and magnitude of the primary results. Key limitations include the time span in which trials were conducted (1999-2017), during which patient characteristics and treatment approaches might have changed. CONCLUSION This NMA suggests that BARI 4 mg + MTX is an efficacious treatment option in the MTX-IR population as evidenced by the robustness of results.
Collapse
|
16
|
Ørnbjerg LM, Østergaard M. Assessment of structural damage progression in established rheumatoid arthritis by conventional radiography, computed tomography, and magnetic resonance imaging. Best Pract Res Clin Rheumatol 2020; 33:101481. [PMID: 32001166 DOI: 10.1016/j.berh.2019.101481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Structural damage progression in patients with established rheumatoid arthritis (RA) has traditionally been assessed by conventional radiography (CR), which has proven its value in clinical practice and clinical trials over the past decades. The most prominent abnormalities visualized by CR in RA patients are erosions as a consequence of bone destruction and joint space narrowing (JSN) as a consequence of cartilage damage. Several validated scoring systems to quantify the structural joint damage and progression herein are available. Computed tomography and magnetic resonance imaging are newer, more sensitive methods for detection and monitoring of structural joint damage. A validated scoring system for magnetic resonance imaging of the hands and wrists exists, while no consensus has been reached on a scoring system for computed tomography. Structural damage identified by either CR or magnetic resonance imaging predicts a poorer disease course in patients with both early and established rheumatoid arthritis.
Collapse
Affiliation(s)
- Lykke Midtbøll Ørnbjerg
- Copenhagen Center for Arthritis Research, COPECARE, Center for Rheumatology and Spine Diseases, Centre of Head and Orthopedics, Rigshospitalet, Valdemar Hansens Vej 17, 2600, Glostrup, Denmark.
| | - Mikkel Østergaard
- Copenhagen Center for Arthritis Research, COPECARE, Center for Rheumatology and Spine Diseases, Centre of Head and Orthopedics, Rigshospitalet, Valdemar Hansens Vej 17, 2600, Glostrup, Denmark.
| |
Collapse
|
17
|
Nasonov EL, Beketova TV, Ananyeva LP, Vasilyev VI, Solovyev SK, Avdeeva AS. PROSPECTS FOR ANTI-B-CELL THERAPY IN IMMUNO-INFLAMMATORY RHEUMATIC DISEASES. RHEUMATOLOGY SCIENCE AND PRACTICE 2019. [DOI: 10.14412/1995-4484-2019-3-40] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- E L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology; I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| | | | | | | | | | | |
Collapse
|
18
|
de Groot M, Patel N, Manavaki R, Janiczek RL, Bergstrom M, Östör A, Gerlag D, Roberts A, Graves MJ, Karkera Y, Fernando D, Mistry P, Walker A, Wisniacki N, Fryer TD, Jimenez-Royo P. Quantifying disease activity in rheumatoid arthritis with the TSPO PET ligand 18F-GE-180 and comparison with 18F-FDG and DCE-MRI. EJNMMI Res 2019; 9:113. [PMID: 31858293 PMCID: PMC6923307 DOI: 10.1186/s13550-019-0576-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/15/2019] [Indexed: 02/14/2023] Open
Abstract
Purpose While the aetiology of rheumatoid arthritis (RA) remains unclear, many of the inflammatory components are well characterised. For diagnosis and therapy evaluation, in vivo insight into these processes would be valuable. Various imaging probes have shown value including dynamic contrast-enhanced (DCE) MRI and PET/CT using 18F-fluorodeoxyglucose (18F-FDG) or tracers targeting the translocator protein (TSPO). To evaluate 18F-GE-180, a novel TSPO PET tracer, for detecting and quantifying disease activity in RA, we compared 18F-GE-180 uptake with that of 18F-FDG and DCE-MRI measures of inflammation. Methods Eight RA patients with moderate-to-high, stable disease activity and active disease in at least one wrist were included in this study (NCT02350426). Participants underwent PET/CT examinations with 18F-GE-180 and 18F-FDG on separate visits, covering the shoulders and from the pelvis to the feet, including hands and wrists. DCE-MRI was performed on one affected hand. Uptake was compared visually between tracers as judged by an experienced radiologist and quantitatively using the maximum standardised uptake value (SUVmax). Uptake for both tracers was correlated with DCE-MRI parameters of inflammation, including the volume transfer coefficient Ktrans using Pearson correlation (r). Results PET/CT imaging with 18F-GE-180 in RA patients showed marked extra-synovial uptake around the affected joints. Overall sensitivity for detecting clinically affected joints was low (14%). 18F-GE-180 uptake did not or only weakly correlate with DCE-MRI parameters in the wrist (r = 0.09–0.31). 18F-FDG showed higher sensitivity for detecting symptomatic joints (34%), as well as strong positive correlation with DCE-MRI parameters (SUVmax vs. Ktrans: r = 0.92 for wrist; r = 0.68 for metacarpophalangeal joints). Conclusions The correlations between DCE-MRI parameters and 18F-FDG uptake support use of this PET tracer for quantification of inflammatory burden in RA. The TSPO tracer 18F-GE-180, however, has shown limited use for the investigation of RA due to its poor sensitivity and ability to quantify disease activity in RA.
Collapse
Affiliation(s)
- Marius de Groot
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, UK.,Department of Radiology, University of Cambridge, Cambridge, UK
| | - Neel Patel
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, UK
| | - Roido Manavaki
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Robert L Janiczek
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, UK
| | - Mats Bergstrom
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, UK
| | - Andrew Östör
- Monash University, Cabrini Medical Centre, Melbourne, Australia
| | | | - Alexandra Roberts
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, UK
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, UK
| | | | - Disala Fernando
- Clinical Unit Cambridge (CUC), GlaxoSmithKline R&D, Addenbrooke's Hospital, Cambridge, UK
| | | | - Adam Walker
- Clinical Unit Cambridge (CUC), GlaxoSmithKline R&D, Addenbrooke's Hospital, Cambridge, UK
| | - Nicolas Wisniacki
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Pilar Jimenez-Royo
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, UK.
| |
Collapse
|
19
|
Kaegi C, Wuest B, Schreiner J, Steiner UC, Vultaggio A, Matucci A, Crowley C, Boyman O. Systematic Review of Safety and Efficacy of Rituximab in Treating Immune-Mediated Disorders. Front Immunol 2019; 10:1990. [PMID: 31555262 PMCID: PMC6743223 DOI: 10.3389/fimmu.2019.01990] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
Background: During the past years biologic agents (also termed biologicals or biologics) have become a crucial treatment option in immunological diseases. Numerous articles have been published on biologicals, which complicates the decision making process on the use of the most appropriate biologic for a given immune-mediated disease. This systematic review is the first of a series of articles assessing the safety and efficacy of B cell-targeting biologics for the treatment of immune-mediated diseases. Objective: To evaluate rituximab's safety and efficacy for the treatment of immune-mediated disorders compared to placebo, conventional treatment, or other biologics. Methods: The PRISMA checklist guided the reporting of the data. We searched the PubMed database between 4 October 2016 and 26 July 2018 concentrating on immune-mediated disorders. Results: The literature search identified 19,665 articles. After screening titles and abstracts against the inclusion and exclusion criteria and assessing full texts, 105 articles were finally included in a narrative synthesis. Conclusions: Rituximab is both safe and effective for the treatment of acquired angioedema with C1-inhibitor deficiency, ANCA-associated vasculitis, autoimmune hemolytic anemia, Behçet's disease, bullous pemphigoid, Castleman's disease, cryoglobulinemia, Goodpasture's disease, IgG4-related disease, immune thrombocytopenia, juvenile idiopathic arthritis, relapsing-remitting multiple sclerosis, myasthenia gravis, nephrotic syndrome, neuromyelitis optica, pemphigus, rheumatoid arthritis, spondyloarthropathy, and systemic sclerosis. Conversely, rituximab failed to show an effect for antiphospholipid syndrome, autoimmune hepatitis, IgA nephropathy, inflammatory myositis, primary-progressive multiple sclerosis, systemic lupus erythematosus, and ulcerative colitis. Finally, mixed results were reported for membranous nephropathy, primary Sjögren's syndrome and Graves' disease, therefore warranting better quality trials with larger patient numbers.
Collapse
Affiliation(s)
- Celine Kaegi
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Benjamin Wuest
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Jens Schreiner
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Urs C Steiner
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Alessandra Vultaggio
- Department of Biomedicine, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Andrea Matucci
- Department of Biomedicine, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Catherine Crowley
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Peterfy C, Kremer J, Rigby W, Singer N, Birchwood C, Gill D, Reiss W, Pei J, Michalska M. Magnetic Resonance Imaging (MRI) Results Following Discontinuation of Methotrexate in Rheumatoid Arthritis Treated with Subcutaneous Tocilizumab: The COMP-ACT MRI Substudy. J Rheumatol 2019; 47:325-332. [PMID: 31154414 DOI: 10.3899/jrheum.180953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess differences in joint damage and inflammation using magnetic resonance imaging (MRI) between patients with rheumatoid arthritis (RA) who achieved low disease activity with tocilizumab (TCZ) + methotrexate (MTX) and subsequently continued or discontinued MTX. METHODS In the COMP-ACT trial, US patients with RA received subcutaneous TCZ 162 mg + MTX. Those who achieved 28-joint count Disease Activity Score calculated with erythrocyte sedimentation rate (DAS28-ESR) ≤ 3.2 at Week 24 were randomized 1:1 (double-blind) to discontinue MTX (TCZ monotherapy; mono) or continue TCZ + MTX until Week 52. In a subset of patients, 1.5-Tesla MRI was used to obtain images of bilateral hands and wrists at weeks 24 and 40. Outcomes included changes in MRI-assessed synovitis, osteitis, erosion, and cartilage loss from Week 24 to Week 40, and in the proportion of patients with progression of each score. RESULTS Of 296 patients who achieved DAS28-ESR ≤ 3.2 at Week 24, 79 were enrolled in the pilot MRI substudy and randomized to TCZ mono (n = 38) or TCZ + MTX (n = 41). Treatment with either TCZ mono or TCZ + MTX suppressed erosion progression, synovitis, osteitis, and cartilage loss. The proportion of patients with no progression in each outcome measure was similar between groups (range, TCZ mono: 84.8-97.0%; TCZ + MTX: 92.3-100%). CONCLUSION In a subset of patients who achieved low disease activity with TCZ + MTX, MRI changes were minimal in intraarticular inflammation and damage measures in patients who discontinued MTX versus those who continued TCZ + MTX.
Collapse
Affiliation(s)
- Charles Peterfy
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA. .,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc.
| | - Joel Kremer
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| | - William Rigby
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| | - Nora Singer
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| | - Christine Birchwood
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| | - Darcy Gill
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| | - William Reiss
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| | - Jinglan Pei
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| | - Margaret Michalska
- From Spire Sciences Inc., Boca Raton, Florida; Albany Medical College and The Center for Rheumatology, Albany, New York; Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire; Case Western Reserve University and MetroHealth System, Cleveland, Ohio; Genentech Inc., South San Francisco, California, USA.,C. Peterfy, MD, Spire Sciences Inc.; J. Kremer, MD, Albany Medical College and The Center for Rheumatology; W. Rigby, MD, Geisel School of Medicine, Dartmouth College; N. Singer, MD, Case Western Reserve University and MetroHealth System; C. Birchwood, PhD, Genentech Inc.; D. Gill, BS, Genentech Inc.; W. Reiss, PharmD, Genentech Inc.; J. Pei, BS, Genentech Inc.; M. Michalska, MD, Genentech Inc
| |
Collapse
|
21
|
Østergaard M, Boesen M. Imaging in rheumatoid arthritis: the role of magnetic resonance imaging and computed tomography. Radiol Med 2019; 124:1128-1141. [DOI: 10.1007/s11547-019-01014-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/21/2019] [Indexed: 10/27/2022]
|
22
|
Peterfy C, DiCarlo J, Emery P, Genovese MC, Keystone EC, Taylor PC, Schlichting DE, Beattie SD, Luchi M, Macias W. MRI and Dose Selection in a Phase II Trial of Baricitinib with Conventional Synthetic Disease-modifying Antirheumatic Drugs in Rheumatoid Arthritis. J Rheumatol 2019; 46:887-895. [PMID: 30647190 DOI: 10.3899/jrheum.171469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Magnetic resonance imaging (MRI) was used in a phase IIb study of baricitinib in patients with RA to support dose selection for the phase III program. METHODS Three hundred one patients with active RA who were taking stable methotrexate were randomized 2:1:1:1:1 to placebo or once-daily baricitinib (1, 2, 4, or 8 mg) for up to 24 weeks. One hundred fifty-four patients with definitive radiographic erosion had MRI of the hand/wrist at baseline and at weeks 12 and 24. Two expert radiologists, blinded to treatment and visit order, scored images for synovitis, osteitis, bone erosion, and cartilage loss. Combined inflammation (osteitis + 3× synovitis score) and total joint damage (erosion + 2.5× cartilage loss score) scores were calculated. Treatment groups were compared using ANCOVA adjusting for baseline scores. RESULTS Mean changes from baseline to Week 12 for synovitis were -0.10, -1.50, and -1.60 for patients treated with placebo, baricitinib 4 mg, and baricitinib 8 mg, respectively (p = 0.003 vs placebo for baricitinib 4 and 8 mg). Mean changes for osteitis were 0.00, -3.20, and -2.10 (p = 0.001 vs placebo for baricitinib 4 mg and p = 0.037 for 8 mg), respectively. Mean changes for bone erosion were 0.90, 0.10, and 0.40 (p = 0.089 for 4 mg and p = 0.275 for 8 mg), respectively, in these treatment groups. CONCLUSION MRI findings in this subgroup of patients suggest suppression of synovitis, osteitis, and combined inflammation by baricitinib 4 and 8 mg. This corroborates previously demonstrated clinical efficacy of baricitinib and increases confidence that baricitinib 4 mg could reduce the radiographic progression in phase III studies. [Clinical trial registration number (www.ClinicalTrials.gov): NCT01185353].
Collapse
Affiliation(s)
- Charles Peterfy
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada. .,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co.
| | - Julie DiCarlo
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Paul Emery
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Mark C Genovese
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Edward C Keystone
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Peter C Taylor
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Doug E Schlichting
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Scott D Beattie
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Monica Luchi
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - William Macias
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| |
Collapse
|
23
|
Lambert RG, Østergaard M, Jaremko JL. Magnetic Resonance Imaging in Rheumatology. Magn Reson Imaging Clin N Am 2018; 26:599-613. [DOI: 10.1016/j.mric.2018.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
24
|
Murray E, Ellis A, Butylkova Y, Skup M, Kalabic J, Garg V. Systematic review and network meta-analysis: effect of biologics on radiographic progression in rheumatoid arthritis. J Comp Eff Res 2018; 7:959-974. [PMID: 30129776 DOI: 10.2217/cer-2017-0106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To evaluate the comparative effectiveness of biologics in inhibiting radiographic progression among rheumatoid arthritis (RA) patients. MATERIALS & METHODS Bayesian network meta-analysis of published trials investigating the USA FDA approved biologics treatment in RA patients, using methotrexate (MTX) as the reference comparator. RESULTS Nine trials met the inclusion criteria for base case analysis. Compared with MTX, most biologics (except golimumab) + MTX had significantly lower rates of radiographic progression at 1 year. Mean difference in radiographic progression rates between MTX monotherapy and biologics + MTX was highest for adalimumab + MTX (-3.8) and lowest for tocilizumab + MTX (-0.7). Inhibition of radiographic progression was sustained. CONCLUSION Biologics inhibit radiographic progression in patients with RA at 1 year; however, published evidence beyond 1 year is limited.
Collapse
Affiliation(s)
- Erin Murray
- Doctor Evidence, Santa Monica, CA, 90401, USA
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Dual-energy computed tomography (DECT) enables material decomposition and virtual monochromatic images by acquiring 2 different energy X-ray data sets. DECT can detect musculoskeletal pathologic conditions that CT alone cannot, and that would otherwise require MR imaging. In this review, the authors discuss several useful techniques and applications of DECT in musculoskeletal research: virtual monochromatic images, virtual noncalcium images, gout, iodine map, and tendons.
Collapse
Affiliation(s)
- Takeshi Fukuda
- Department of Radiology, The Jikei University School of Medicine, 3-19-18, Nishi-Shimbashi, Minato-ku, Tokyo 105-8471, Japan; Department of Radiology, Stony Brook Medicine, HSC Level 4, Room 120, Stony Brook, NY 11794, USA.
| | - Kunihiko Fukuda
- Centre for International Affairs, The Jikei University, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
26
|
Zollars ES, Hyer M, Wolf B, Chapin R. Measuring lupus arthritis activity using contrasted high-field MRI. Associations with clinical measures of disease activity and novel patterns of disease. Lupus Sci Med 2018; 5:e000264. [PMID: 30094039 PMCID: PMC6069922 DOI: 10.1136/lupus-2018-000264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 11/03/2022]
Abstract
Objective Arthritis in SLE is poorly described, and there is no objective measure for quantification of arthritis. In this pilot study, we aim to assess the utility of the Rheumatoid Arthritis MRI Scoring System (RAMRIS) for the quantification of lupus arthritis. Methods Patients were eligible for entry into the study if they were evaluated at the Medical University of South Carolina Lupus Center and determined by their treating rheumatologist to have active hand arthritis due to SLE. Standard of care lupus activity measures were collected, along with a detailed physical exam. MRIs were obtained using standard musculoskeletal sequences with gadolinium contrast. Semiquantitative scoring of the images used the Outcome Measures in Rheumatology Clinical Trials RAMRIS system. Results RAMRIS demonstrates large amounts of synovitis, tenosynovitis, bone marrow oedema and erosive disease in only a minority of patients. Some patients were not scored as having any synovitis or tenosynovitis. We describe potential features of lupus arthritis that are not captured in the RAMRIS scores and may be contributing to symptoms. Conclusion Lupus arthritis is an entity separate from rheumatoid arthritis and requires the development of new quantitative methods to describe and quantitate it. MRI findings suggest the inadequacy of a typical lupus musculoskeletal measure including swollen/tender joint counts to assess the level of disease activity.
Collapse
Affiliation(s)
- Eric S Zollars
- Division of Rheumatology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Madison Hyer
- Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bethany Wolf
- Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Russell Chapin
- Division of Musculoskeletal Radiology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
27
|
Jayashree S, Nirekshana K, Guha G, Bhakta-Guha D. Cancer chemotherapeutics in rheumatoid arthritis: A convoluted connection. Biomed Pharmacother 2018; 102:894-911. [PMID: 29710545 DOI: 10.1016/j.biopha.2018.03.123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy is one of the most popular therapeutic strategies to treat cancer. However, cancer chemotherapeutics have often been associated with impairment of the immune system, which might consequently lead to an augmented risk of autoimmune disorders, such as rheumatoid arthritis. Though the accurate mechanistic facets of rheumatoid arthritis induction have not been interpreted yet, a conglomeration of genetic and environmental factors might promote its etiology. What makes the scenario more challenging is that patients with rheumatoid arthritis are at a significantly elevated risk of developing various types of cancer. It is intriguing to note that diverse cancer chemotherapy drugs are also commonly used to treat symptoms of rheumatoid arthritis. However, a colossal multitude of such cancer therapeutics has demonstrated highly varied results in rheumatoid arthritis patients, including both beneficial and adverse effects. Herein, we attempt to present a holistic account of the variegated modalities of this complex tripartite cross-talk between cancer, rheumatoid arthritis and chemotherapy drugs in order to decode the sinuous correlation between these two appalling pathological conditions.
Collapse
Affiliation(s)
- S Jayashree
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India
| | - K Nirekshana
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India
| | - Gunjan Guha
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India.
| | - Dipita Bhakta-Guha
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India.
| |
Collapse
|
28
|
Yu X, Zhang L, Wang L, Lu W, Sun F, Xu P, Lan G. MRI assessment of erosion repair in patients with long-standing rheumatoid arthritis receiving double-filtration plasmapheresis in addition to leflunomide and methotrexate: a randomized controlled trial. Clin Rheumatol 2018; 37:917-925. [DOI: 10.1007/s10067-017-3956-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/24/2017] [Accepted: 12/12/2017] [Indexed: 12/31/2022]
|
29
|
Hunt L, Eugénio G, Grainger AJ. Magnetic resonance imaging in individuals at risk of rheumatoid arthritis. Best Pract Res Clin Rheumatol 2017; 31:80-89. [PMID: 29221601 DOI: 10.1016/j.berh.2017.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/06/2017] [Accepted: 08/18/2017] [Indexed: 01/07/2023]
Abstract
Individuals with rheumatoid arthritis (RA) benefit from early diagnosis and initiation of therapy. There can be delays in both due to diagnostic uncertainties. Imaging modalities, including magnetic resonance imaging (MRI), can detect inflammation earlier than clinical examination alone in early RA patients. Furthermore, the predictive role of MRI for the future development of RA has recently been explored in 'at-risk' individuals. This review details the use of MRI in early and undifferentiated arthritis and summarises the studies to date in individuals at risk of RA.
Collapse
Affiliation(s)
- Laura Hunt
- Leeds Biomedical Research Centre, Chapel Allerton Hospital Leeds, LS7 4SA, UK.
| | - Gisela Eugénio
- Rheumatology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
| | - Andrew J Grainger
- Leeds Biomedical Research Centre, Chapel Allerton Hospital Leeds, LS7 4SA, UK
| |
Collapse
|
30
|
Kondo Y, Kaneko Y, Sugiura H, Matsumoto S, Nishina N, Kuwana M, Jinzaki M, Takeuchi T. Pre-treatment interleukin-6 levels strongly affect bone erosion progression and repair detected by magnetic resonance imaging in rheumatoid arthritis patients. Rheumatology (Oxford) 2017; 56:1089-1094. [PMID: 28340008 DOI: 10.1093/rheumatology/kex046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Indexed: 11/14/2022] Open
Abstract
Objective To examine the relationship between MRI structural damage and repair and plasma inflammatory cytokines in patients with RA. Methods A total of 88 newly diagnosed, untreated RA patients were enrolled. Contrast MRI of the dominant hand and X-rays of the hands and feet were performed at baseline and 1 year later. MR images were evaluated using RA MRI scoring, and X-ray. Results Progression of bone erosion and repair were observed more frequently in MRI than in X-rays (erosion, 52% vs 26%, P < 0.001; repair, 26% vs 15%, P = 0.003, respectively). Baseline IL-6 levels and seropositivity were independent relevant factors for MRI erosion progression, with IL-6 having stronger effect than seropositivity. A receiver operating characteristic curve identified the baseline IL-6 level of 7.6 pg/ml for predicting erosion progression during 1 year, with an area under the curve of 0.82; higher IL-6 levels resulted in more erosion progression. Baseline low IL-6 was also an independent predictor for MRI erosion repair. Conclusion In newly diagnosed, untreated RA patients, baseline plasma IL-6 levels are responsible for 1-year MRI bone erosion progression and repair.
Collapse
Affiliation(s)
- Yasushi Kondo
- Division of Rheumatology, Department of Internal Medicine
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine
| | - Hiroaki Sugiura
- Department of Diagnostic Radiology, Keio University School of Medicine
| | | | - Naoshi Nishina
- Division of Rheumatology, Department of Internal Medicine
| | - Masataka Kuwana
- Division of Rheumatology, Department of Internal Medicine.,Division of Allergy and Rheumatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Masahiro Jinzaki
- Department of Diagnostic Radiology, Keio University School of Medicine
| | | |
Collapse
|
31
|
Nasonov EL, Mazurov VI, Zonova EV, Knyazeva LA, Marusenko IM, Nesmeyanova OB, Plaksina TV, Shapovalova YS, Ilivanova EP, Krechikova DG, Petrochenkova NA, Reshetko OV, Denisov LN, Gordeev IG, Davydova AF, Eremina NA, Zemerova EV, Ivanova TB, Kastanayan AA, Pokrovskaya TG, Smakotina SA, Smolyarchuk EA, Artemyeva AV, Ivanov RA, Usacheva YV, Chernyaeva EV. THE EFFICACY AND SAFETY OF RITUXIMAB BIOSIMILAR (ACELLBIA®) IN RHEUMATOID ARTHRITIS AS THE FIRST BIOLOGICAL AGENT: RESULTS OF PHASE III (ALTERRA) CLINICAL TRIAL. RHEUMATOLOGY SCIENCE AND PRACTICE 2017. [DOI: 10.14412/1995-4484-2017-351-359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The Russian biotechnological company «BIOCAD» has designed a chimeric monoclonal antibody against CD20 (BCD-020, Acellbia®) that is a biosimilar of rituximab (RTM; MabThera®, F. Hoffmann-La Roche Ltd., Switzerland). In recent years, there has been evidence that RTM can be used at lower doses than those given in the standard recommendations and instructions for the use of this drug. This serves as the basis for the BCD-020-4/ALTERRA (ALTErnative Rituximab regimen in Rheumatoid Arthritis) trial, the objective of which was to investigate the efficiency and safety of using Acellbia® (at a dose of 600 mg twice at a 2-week interval) as the first biological agent (BA) for methotrexate (MTX)-resistant active rheumatoid arthritis (RA). The investigation enrolled 159 patients aged 18 to 80 years with active RA. After 24 weeks 65.7 and 29.4% of patients achieved 20% improvement by the American College of Rheumatology (ACR) criteria in the Acellbia® + MTX and placebo (PL) + MTX groups, respectively (p<0.0001). The differences in the ACR20 response rate in the two groups were 36.3% (95% CI, 19.27–53.28%). There were significant differences between the groups in the ACR50 response rates: 28.4% and 5.9% (p=0.001) and in the ACR70 ones: 12.8% and only 2.0%, respectively (p=0.036). Analysis of all recorded adverse events (AE) frequency showed no significant differences between the patients in the study and control groups and demonstrates its equivalence with that of RTM (MabThera®); all the AE were expectable. It is noted that antibodies to RTM with binding and neutralizing activities had no impact on the efficiency and safety of therapy.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology
I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| | - V. I. Mazurov
- I.I. Mechnikov North-Western State Medical University, Ministry of Health of Russia
| | | | | | - I. M. Marusenko
- V.A. Baranov Republican Hospital, Ministry of Health of the Republic of Karelia
| | | | - T. V. Plaksina
- N.A. Semashko Nizhny Novgorod Regional Clinical Hospital
| | | | | | | | | | | | | | | | - A. F. Davydova
- Professor S.V. Ochapovsky Territorial Clinical Hospital One
| | - N. A. Eremina
- Railway Clinical Hospital at the Gorky Station, OAO «RZhD»
| | | | | | | | | | - S. A. Smakotina
- Kemerovo State Medical Academy, Ministry of Health of Russia
| | - E. A. Smolyarchuk
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| | | | | | | | | |
Collapse
|
32
|
Østergaard M, Peterfy CG, Bird P, Gandjbakhch F, Glinatsi D, Eshed I, Haavardsholm EA, Lillegraven S, Bøyesen P, Ejbjerg B, Foltz V, Emery P, Genant HK, Conaghan PG. The OMERACT Rheumatoid Arthritis Magnetic Resonance Imaging (MRI) Scoring System: Updated Recommendations by the OMERACT MRI in Arthritis Working Group. J Rheumatol 2017; 44:1706-1712. [DOI: 10.3899/jrheum.161433] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2017] [Indexed: 02/08/2023]
Abstract
Objective.The Outcome Measures in Rheumatology (OMERACT) Rheumatoid Arthritis (RA) Magnetic Resonance Imaging (MRI) scoring system (RAMRIS), evaluating bone erosion, bone marrow edema/osteitis, and synovitis, was introduced in 2002, and is now the standard method of objectively quantifying inflammation and damage by MRI in RA trials. The objective of this paper was to identify subsequent advances and based on them, to provide updated recommendations for the RAMRIS.Methods.MRI studies relevant for RAMRIS and technical and scientific advances were analyzed by the OMERACT MRI in Arthritis Working Group, which used these data to provide updated considerations on image acquisition, RAMRIS definitions, and scoring systems for the original and new RA pathologies. Further, a research agenda was outlined.Results.Since 2002, longitudinal studies and clinical trials have documented RAMRIS variables to have face, construct, and criterion validity; high reliability and sensitivity to change; and the ability to discriminate between therapies. This has enabled RAMRIS to demonstrate inhibition of structural damage progression with fewer patients and shorter followup times than has been possible with conventional radiography. Technical improvements, including higher field strengths and improved pulse sequences, allow higher image resolution and contrast-to-noise ratio. These have facilitated development and validation of scoring methods of new pathologies: joint space narrowing and tenosynovitis. These have high reproducibility and moderate sensitivity to change, and can be added to RAMRIS. Combined scores of inflammation or joint damage may increase sensitivity to change and discriminative power. However, this requires further research.Conclusion.Updated 2016 RAMRIS recommendations and a research agenda were developed.
Collapse
|
33
|
Fukuda T, Umezawa Y, Asahina A, Nakagawa H, Furuya K, Fukuda K. Dual energy CT iodine map for delineating inflammation of inflammatory arthritis. Eur Radiol 2017; 27:5034-5040. [PMID: 28674965 DOI: 10.1007/s00330-017-4931-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/10/2017] [Accepted: 06/07/2017] [Indexed: 01/08/2023]
Abstract
Iodine mapping is an image-processing technique used with dual-energy computed tomography (DECT) to improve iodine contrast resolution. CT, because of its high spatial resolution and thin slice reconstruction, is well suited to the evaluation of the peripheral joints. Recent developments in the treatment of inflammatory arthritis that require early diagnosis and precise therapeutic assessment encourage radiological evaluation. To facilitate such assessment, we describe DECT iodine mapping as a novel modality for evaluating rheumatoid arthritis and psoriatic arthritis of the hands and feet. KEY POINTS • Dual-energy CT iodine mapping can delineate inflammation of peripheral inflammatory arthritis. • DECT iodine mapping has high spatial resolution compared with MRI. • DECT iodine mapping has a high iodine contrast resolution. • DECT iodine mapping may reflect therapeutic effects.
Collapse
Affiliation(s)
- Takeshi Fukuda
- Department of Radiology, The Jikei University School of Medicine, 3-19-18, Nishi-shimbashi, Minato-ku, Tokyo, 1058471, Japan.
| | - Yoshinori Umezawa
- Department of Dermatology, The Jikei University School of Medicine, 3-19-18, Nishi-shimbashi, Minato-ku, Tokyo, 1058471, Japan
| | - Akihiko Asahina
- Department of Dermatology, The Jikei University School of Medicine, 3-19-18, Nishi-shimbashi, Minato-ku, Tokyo, 1058471, Japan
| | - Hidemi Nakagawa
- Department of Dermatology, The Jikei University School of Medicine, 3-19-18, Nishi-shimbashi, Minato-ku, Tokyo, 1058471, Japan
| | - Kazuhiro Furuya
- Division of Rheumatology Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18, Nishi-shimbashi, Minato-ku, Tokyo, 1058471, Japan
| | - Kunihiko Fukuda
- Department of Radiology, The Jikei University School of Medicine, 3-19-18, Nishi-shimbashi, Minato-ku, Tokyo, 1058471, Japan
| |
Collapse
|
34
|
Woodworth TG, Morgacheva O, Pimienta OL, Troum OM, Ranganath VK, Furst DE. Examining the validity of the rheumatoid arthritis magnetic resonance imaging score according to the OMERACT filter-a systematic literature review. Rheumatology (Oxford) 2017; 56:1177-1188. [PMID: 28398508 PMCID: PMC5850856 DOI: 10.1093/rheumatology/kew445] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 11/01/2016] [Indexed: 02/05/2023] Open
Abstract
Objective To examine whether the RA MRI score (RAMRIS) for RA of the wrist/hand meets the OMERACT filter criteria-truth (validity), discrimination and feasibility. Methods We conducted a systematic literature review in PubMed and Scopus, from 1970 through June 2014, focused on MRI measures of synovitis, osteitis/bone marrow oedema, erosions and/or joint space narrowing in RA randomized controlled trials and observational studies with cohort size ⩾10. Strength of evidence was assessed using the Cochrane Handbook criteria. Results Of 634 MRI titles/abstracts, 202 met the review criteria, with 92 providing at least 1 type of validity. Four articles provided criterion validity, and 26 articles utilized RAMRIS to assess 1.5 T MRI images. Histopathology data showed inflammation corresponding to MRI of synovitis and osteitis. MRI erosions corresponded to those identified with CT. Content and construct validity for RAMRIS synovitis, osteitis and erosions were documented by correlations with clinical, laboratory and/or radiographic data. Each measure was sensitive to change and responsive to therapy. RAMRIS synovitis and osteitis were able to discriminate between the efficacy of treatments vs placebo in 12-week studies, whereas RAMRIS erosions required studies of ⩾24 weeks. Conclusion RAMRIS synovitis, osteitis and erosions imaged with 1.5 T MRI are valid and useful for evaluating joint inflammation and damage for RA of the wrist/hand, according to the OMERACT filter.
Collapse
Affiliation(s)
- Thasia G. Woodworth
- Department of Medicine, Division of Rheumatology, David Geffen School of
Medicine, UCLA, Los Angeles
| | - Olga Morgacheva
- Department of Medicine, Division of Rheumatology, David Geffen School of
Medicine, UCLA, Los Angeles
| | - Olga L. Pimienta
- Keck School of Medicine, University of Southern California, Santa Monica,
CA, USA
| | - Orrin M. Troum
- Keck School of Medicine, University of Southern California, Santa Monica,
CA, USA
| | - Veena K. Ranganath
- Department of Medicine, Division of Rheumatology, David Geffen School of
Medicine, UCLA, Los Angeles
| | - Daniel E. Furst
- Department of Medicine, Division of Rheumatology, David Geffen School of
Medicine, UCLA, Los Angeles
| |
Collapse
|
35
|
Møller-Bisgaard S, Ejbjerg BJ, Eshed I, Hørslev-Petersen K, Hetland ML, Jurik AG, Thomsen H, Torfing T, Stengaard-Pedersen K, Junker P, Krogh NS, Lottenburger T, Ellingsen T, Andersen LS, Skjødt H, Svendsen AJ, Tarp U, Hansen IT, Pødenphant J, Pedersen JK, Lindegaard H, Hanson LG, Vestergaard A, Glinatsi D, Østergaard M. Effect of a treat-to-target strategy based on methotrexate and intra-articular betamethasone with or without additional cyclosporin on MRI-assessed synovitis, osteitis, tenosynovitis, bone erosion, and joint space narrowing in early rheumatoid arthritis: results from a 2-year randomized double-blind placebo-controlled trial (CIMESTRA). Scand J Rheumatol 2016; 46:335-345. [PMID: 27775461 DOI: 10.1080/03009742.2016.1209550] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVES To investigate whether a treat-to-target strategy based on methotrexate (MTX) and intra-articular (IA) betamethasone suppresses magnetic resonance imaging (MRI)-determined measures of disease activity and reduces joint destruction in early rheumatoid arthritis (eRA) patients, and to investigate whether concomitant cyclosporin A (CyA) provides an additional effect. METHOD In the 2-year randomized, double-blind, treat-to-target trial CIMESTRA, 160 patients with eRA (< 6 months) were randomized to MTX, intra-articular betamethasone and CyA, or placebo CyA. A total of 129 patients participated in the MRI substudy, and had contrast-enhanced MR images of the non-dominant hand at months 0, 6, 12, and 24. MR images were evaluated for osteitis, synovitis, tenosynovitis, bone erosion, and joint space narrowing (JSN), using validated scoring methods. RESULTS Significant reductions were seen at 6 months in all inflammatory parameters [synovitis, mean change -1.6 (p < 0.001, Wilcoxon), tenosynovitis, -3.5 (p < 0.001), and osteitis, -1.3 (p < 0.05)] and at 12/24 months in synovitis and tenosynovitis [-1.6/-2.2 and -3.6/-3.8, respectively; all p < 0.001]. MRI signs of inflammation were not fully eliminated, and increases in erosion and JSN scores were observed at 6 months [0.4 (p < 0.01)/0.1 (p < 0.05)], 12 months [0.8 (p < 0.001)/0.3 (p < 0.01)], and 24 months [1.0 (p < 0.001)/0.4 (p < 0.001)]. Clinical measures decreased significantly (p < 0.001) at all time points. There were no consistent statistically significant differences between treatment groups. CONCLUSIONS In this eRA treat-to-target trial, MTX and intra-articular glucocorticoids markedly reduced, but did not eliminate, MRI osteitis, synovitis, and tenosynovitis. Accordingly, minimal but statistically significant increases in bone erosion and JSN were observed. No additional effect of CyA was demonstrated.
Collapse
Affiliation(s)
- S Møller-Bisgaard
- a Copenhagen Centre for Arthritis Research, Centre for Rheumatology and Spine Diseases , Rigshospitalet , Glostrup , Denmark.,b Department of Rheumatology , Slagelse Hospital , Slagelse , Denmark
| | - B J Ejbjerg
- b Department of Rheumatology , Slagelse Hospital , Slagelse , Denmark
| | - I Eshed
- c Department of Radiology , Sheba Medical Centre , Tel Hashomer , Israel
| | - K Hørslev-Petersen
- d King Christian X's Hospital for Rheumatic Diseases , Graasten , Denmark
| | - M L Hetland
- a Copenhagen Centre for Arthritis Research, Centre for Rheumatology and Spine Diseases , Rigshospitalet , Glostrup , Denmark.,e The DANBIO Registry, Centre for Rheumatology and Spine Diseases , Rigshospitalet , Glostrup , Denmark
| | - A G Jurik
- f Department of Radiology , Aarhus University Hospital , Aarhus , Denmark
| | - H Thomsen
- g Department of Radiology , Herlev-Gentofte University Hospital , Herlev , Denmark
| | - T Torfing
- h Department of Radiology , Odense University Hospital , Odense , Denmark
| | | | - P Junker
- j Department of Rheumatology , Odense University Hospital , Odense , Denmark
| | - N S Krogh
- k Zitelab ApS , Copenhagen , Denmark
| | - T Lottenburger
- l Department of Rheumatology , Vejle Hospital , Vejle , Denmark
| | - T Ellingsen
- j Department of Rheumatology , Odense University Hospital , Odense , Denmark
| | - L S Andersen
- m Department of Rheumatology , Herlev-Gentofte University Hospital , Gentofte , Denmark
| | - H Skjødt
- a Copenhagen Centre for Arthritis Research, Centre for Rheumatology and Spine Diseases , Rigshospitalet , Glostrup , Denmark
| | - A J Svendsen
- n Department of Epidemiology , Institute of Public Health, University of Southern Denmark , Odense , Denmark
| | - U Tarp
- i Department of Rheumatology , Aarhus University Hospital , Aarhus , Denmark
| | - I T Hansen
- i Department of Rheumatology , Aarhus University Hospital , Aarhus , Denmark
| | - J Pødenphant
- m Department of Rheumatology , Herlev-Gentofte University Hospital , Gentofte , Denmark
| | - J K Pedersen
- j Department of Rheumatology , Odense University Hospital , Odense , Denmark
| | - H Lindegaard
- j Department of Rheumatology , Odense University Hospital , Odense , Denmark
| | - L G Hanson
- o Centre for Functional and Diagnostic Imaging and Research , Danish Research Centre for Magnetic Resonance (DRCMR), Copenhagen University Hospital Hvidovre , Hvidovre , Denmark.,p Biomedical Engineering, Elektro , Technical University of Denmark (DTU) , Lyngby , Denmark
| | - A Vestergaard
- q Department of Radiology , Copenhagen University Hospita Hvidovrel , Hvidovre , Denmark
| | - D Glinatsi
- a Copenhagen Centre for Arthritis Research, Centre for Rheumatology and Spine Diseases , Rigshospitalet , Glostrup , Denmark
| | - M Østergaard
- a Copenhagen Centre for Arthritis Research, Centre for Rheumatology and Spine Diseases , Rigshospitalet , Glostrup , Denmark
| |
Collapse
|
36
|
Ku E, Pedoia V, Tanaka M, Heilmeier U, Imboden J, Graf J, Link T, Li X. Evaluating radiocarpal cartilage matrix changes 3-months after anti-TNF treatment for rheumatoid arthritis using MR T1ρ imaging. J Magn Reson Imaging 2016; 45:1514-1522. [PMID: 27661002 DOI: 10.1002/jmri.25448] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/10/2016] [Indexed: 12/26/2022] Open
Abstract
PURPOSE To evaluate the feasibility of MR T1ρ in assessing radiocarpal cartilage matrix changes following rheumatoid arthritis (RA) treatment. MATERIALS AND METHODS Five healthy controls and nine RA patients were studied: three RA patients with low disease activity that were treated with methotrexate (MTX) alone and six with active disease despite MTX treatment who were additionally treated with certolizumab pegol, an anti-tumor necrosis factor biologic. Wrist 3 Tesla MRI were acquired at baseline and 3-month follow-up. T1ρ were quantified for lunar, radius, and scaphoid cartilage. Reproducibility was evaluated using coefficients of variation (CV). Longitudinal changes were evaluated with t-test and relationships between T1ρ with clinical, MRI, and patient-reported outcomes were evaluated with Spearman's rho. RESULTS Scan/re-scan CVs of T1ρ values were all <5%, and intra- and inter-reader CVs were all < 2.0%. Baseline scaphoid T1ρ values were significantly higher in RA patients compared with healthy controls (P = 0.032). Changes in T1ρ (baseline, 3-month) were correlated with EULAR treatment response criteria: -2.26 ± 0.75 ms, 1.08 ± 0.52 ms, and 2.18 ± 0.45 ms for good, moderate, and nonresponders, respectively. Significant correlations were found between changes in global T1ρ values and changes in DAS28-CRP (rs = 0.683; P = 0.042), MHQ (rs = -0.783; P = 0.013), and HAQ (rs = 0.833; P = 0.010). CONCLUSION Despite the limited sample size and follow-up time points, there were significant correlations between changes in radiocarpal T1ρ and changes in disease activity as assessed by clinical and patient-reported outcomes. Our findings encourage further research into MR T1ρ assessment of RA disease activity and treatment response. LEVEL OF EVIDENCE 1 J. MAGN. RESON. IMAGING 2017;45:1514-1522.
Collapse
Affiliation(s)
- Eric Ku
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Valentina Pedoia
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Matthew Tanaka
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Ursula Heilmeier
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - John Imboden
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jonathan Graf
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Thomas Link
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Xiaojuan Li
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
37
|
Abstract
Rituximab is a monoclonal antibody that depletes B cells from the circulation. It was originally used to treat lymphoma but is increasingly used for the treatment of autoimmune diseases. Rituximab was found to be effective in randomised controlled trials for rheumatoid arthritis, granulomatosis with polyangiitis and other antineutrophil cytoplasmic antibody-associated vasculitides. However, evidence of efficacy is very limited for many other autoimmune conditions. Before starting rituximab, it is important to check the patient's baseline immunoglobulins and immunisation status. Patients should also be screened for latent infections and other contraindications.
Collapse
|
38
|
|
39
|
Genovese MC, Yang F, Østergaard M, Kinnman N. Efficacy of VX-509 (decernotinib) in combination with a disease-modifying antirheumatic drug in patients with rheumatoid arthritis: clinical and MRI findings. Ann Rheum Dis 2016; 75:1979-1983. [PMID: 27084959 DOI: 10.1136/annrheumdis-2015-208901] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/23/2016] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To assess early effects on joint structures of VX-509 in combination with stable disease-modifying antirheumatic drug (DMARD) therapy using MRI in adults with rheumatoid arthritis (RA). METHODS This phase II, placebo-controlled, double-blind, dose-ranging study randomised patients with RA and inadequate DMARD response to VX-509 100 mg (n=11), 200 mg (n=10) or 300 mg (n=10) or placebo (n=12) once daily for 12 weeks. Outcome measures included American College of Rheumatology score (ACR20; improvement of ≥20%) and disease activity score (DAS28) using C reactive protein (CRP), and the RA MRI scoring (RAMRIS) system. RESULTS ACR20 response at week 12 was 63.6%, 60.0% and 60.0% in the VX-509 100-mg, 200-mg and 300-mg groups, respectively, compared with 25.0% in the placebo group. DAS28-CRP scores decreased in a dose-dependent manner with increasing VX-509 doses. Decreases in RAMRIS synovitis scores were significantly different from placebo for all VX-509 doses (p<0.01) and for RAMRIS osteitis scores (p<0.01) for VX-509 300 mg. Treatment was generally well tolerated. CONCLUSIONS VX-509 plus a DMARD reduced the signs and symptoms of RA in patients with an inadequate response to a DMARD alone. MRI responses were detected at week 12. Treatment was generally well tolerated. TRIAL REGISTRATION NUMBER NCT01754935; results.
Collapse
Affiliation(s)
- Mark C Genovese
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, California, USA
| | - Fang Yang
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| | - Mikkel Østergaard
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Rigshospitalet-Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Nils Kinnman
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Peterfy C, Burmester GR, Bykerk VP, Combe BG, DiCarlo JC, Furst DE, Huizinga TWJ, Wong DA, Conaghan PG, Emery P. Sustained improvements in MRI outcomes with abatacept following the withdrawal of all treatments in patients with early, progressive rheumatoid arthritis. Ann Rheum Dis 2016; 75:1501-5. [PMID: 26865601 PMCID: PMC4975847 DOI: 10.1136/annrheumdis-2015-208258] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/16/2016] [Indexed: 12/25/2022]
Abstract
Objectives To assess structural damage progression with subcutaneous abatacept (ABA) in the Assessing Very Early Rheumatoid arthritis Treatment (AVERT) trial following abrupt withdrawal of all rheumatoid arthritis (RA) medication in patients achieving Disease Activity Score (DAS)-defined remission or low disease activity. Methods Patients with early, active RA were randomised to ABA plus methotrexate (ABA/MTX) 125 mg/week, ABA 125 mg/week or MTX for 12 months. All RA treatments were withdrawn after 12 months in patients with DAS28 (C reactive protein (CRP)) <3.2. Adjusted mean changes from baseline in MRI-based synovitis, osteitis and erosion were calculated for the intention-to-treat population. Results 351 patients were randomised and treated: ABA/MTX (n=119), ABA (n=116) or MTX (n=116). Synovitis and osteitis improved, and progression of erosion was statistically less with ABA/MTX versus MTX at month 12 (−2.35 vs −0.68, −2.58 vs −0.68, 0.19 vs 1.53, respectively; p<0.01 for each) and month 18 (−1.34 vs −0.49 −2.03 vs 0.34, 0.13 vs 2.0, respectively; p<0.01 for erosion); ABA benefits were numerically intermediate to those for ABA/MTX and MTX. Conclusions Structural benefits with ABA/MTX or ABA may be maintained 6 months after withdrawal of all treatments in patients who have achieved remission or low disease activity. Trial registration number NCT01142726; Results.
Collapse
Affiliation(s)
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | - Vivian P Bykerk
- Department of Rheumatology, Hospital for Special Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Bernard G Combe
- Department of Rheumatology, Service d'Immuno-Rheumatologie, Montpellier, France
| | | | - Daniel E Furst
- Department of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Philip G Conaghan
- 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
| |
Collapse
|
41
|
Svetlicky N, Kivity S, Odeh Q, Shovman O, Gertel S, Amital H, Gendelman O, Volkov A, Barshack I, Bar-Meir E, Blank M, Shoenfeld Y. Anti-citrullinated-protein-antibody-specific intravenous immunoglobulin attenuates collagen-induced arthritis in mice. Clin Exp Immunol 2015; 182:241-50. [PMID: 26132809 DOI: 10.1111/cei.12673] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2015] [Indexed: 02/02/2023] Open
Abstract
Administration of intravenous immunoglobulin (IVIg) is a recognized safe and efficient immunomodulation therapy for many autoimmune diseases. Anti-idiotypic antibody binding to pathogenic autoantibodies was proposed as one of the mechanisms attributed to the protective activity of IVIg in autoimmunity. The aim of this study was to fractionate the anti-anti-citrullinated protein anti-idiotypic-antibodies (anti-ACPA) from an IVIg preparation and to test it as a treatment for collagen-induced arthritis in mice. IVIg was loaded onto an ACPA column. The eluted fraction was defined as ACPA-specific-IVIg (ACPA-sIVIg). Collagen-induced-arthritis (CIA) was induced in mice. Mice were treated weekly with ACPA-sIVIg, low-dose-IVIg, high-dose-IVIg and phosphate-buffered saline (PBS). Sera-ACPA titres, anti-collagen anitbodies and cytokine levels were analysed by enzyme-linked immunosorbent assay (ELISA); antibody-forming-cell activity by enzyme-linked imunospot (ELISPOT) assay; and expansion of regulatory T cell (Treg ) population by fluorescence activated cell sorter (FACS). ACPA-sIVIg inhibited ACPA binding to citrullinated-peptides (CCP) in vitro 100 times more efficiently than the IVIg compound. ACPA-sIVIg was significantly more effective than the IVIg-preparation in attenuating the development of collagen-induced arthritis. Splenocytes from CIA mice treated with ACPA-sIVIg reduced the ACPA and anti-collagen-antibody titres, including the number of anti-collagen and ACPA antibody-forming cells. In parallel, splenocytes from ACPA-sIVIg treated mice secreted higher levels of anti-inflammatory cytokines and lower proinflammatory cytokines. The ACPA-sIVIg inhibitory potential was accompanied with expansion of the Treg population. Low-dose IVIg did not affect the humoral and cellular response in the CIA mice in comparison to the PBS-treated mice. Based on our results, IVIg may be considered as a safe compound for treating patients with rheumatoid arthritis by neutralizing pathogenic autoantibodies, reducing proinflammatory cytokines and expanding the Treg population.
Collapse
Affiliation(s)
- N Svetlicky
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| | - S Kivity
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv.,Internal Medicine B, Sheba Medical Center, Tel-Hashomer
| | - Q Odeh
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| | - O Shovman
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| | - S Gertel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| | - H Amital
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv.,Internal Medicine B, Sheba Medical Center, Tel-Hashomer
| | - O Gendelman
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv.,Internal Medicine B, Sheba Medical Center, Tel-Hashomer
| | - A Volkov
- Institute of Pathology, Sheba Medical Center, affiliated with the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| | - I Barshack
- Institute of Pathology, Sheba Medical Center, affiliated with the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| | - E Bar-Meir
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv.,Poria Medical Center affiliated to Faculty of Medicine in the Galilee Bar-ilan University, Poria Israel
| | - M Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| | - Y Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv
| |
Collapse
|
42
|
Baker JF, Conaghan PG, Emery P, Baker DG, Østergaard M. Validity of early MRI structural damage end points and potential impact on clinical trial design in rheumatoid arthritis. Ann Rheum Dis 2015; 75:1114-9. [PMID: 26091907 DOI: 10.1136/annrheumdis-2014-206934] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 06/06/2015] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To evaluate the construct validity of the rheumatoid arthritis MRI score (RAMRIS) erosion evaluation as structural damage end point and to assess the potential impact of incorporation in clinical trials. METHODS In a randomised trial of early methotrexate-naïve RA (GO-BEFORE), RAMRIS scores were determined from MRIs and van der Heijde-Sharp (vdHS) scores from radiographs, at baseline, week 12, week 24 and week 52. Progression in damage scores was defined as change >0.5. Associations of X-ray and MRI outcomes with clinical features were evaluated for convergent validity. Iterative Wilcoxon rank sum tests and tests of proportion estimated the sample size required to detect differences between combination therapy (methotrexate+golimumab) and methotrexate-monotherapy arms in (A) change in damage score and (B) proportion of patients progressing. RESULTS Patients with early MRI progression had higher DAS28, C reactive protein (CRP) and vdHS at baseline, and higher 2-year HAQ. Associations were similar to those with 1-year vdHS progression. Differences in change in structural damage between treatment arms achieved significance with fewer subjects when 12-week or 24-week MRI erosion score was the outcome (150 patients; 100 among an enriched sample with baseline-synovitis >5) compared with the 52-week vdHS (275 patients). Differences in the proportion progressing could be detected in 234 total subjects with 12-week MRI in an enriched sample whereas 1-year X-ray required between 468 and 1160 subjects. CONCLUSIONS Early MRI erosion progression is a valid measure of structural damage that could substantially decrease sample size and study duration if used as structural damage end point in RA clinical trials.
Collapse
Affiliation(s)
- Joshua F Baker
- Philadelphia Veterans' Affairs Medical Center, Philadelphia, Pennsylvania, USA University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania, USA Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Daniel G Baker
- Janssen Research & Development, LLC., Horsham, Pennsylvania, USA
| | - Mikkel Østergaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Glostrup Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Bredemeier M, Campos GG, de Oliveira FK. Updated systematic review and meta-analysis of randomized controlled trials comparing low- versus high-dose rituximab for rheumatoid arthritis. Clin Rheumatol 2015; 34:1801-5. [PMID: 26070536 DOI: 10.1007/s10067-015-2977-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 05/08/2015] [Accepted: 05/15/2015] [Indexed: 01/18/2023]
Abstract
The purpose of this study is to update a systematic review and meta-analysis comparing low- (2 × 500 or 1 × 1000 mg) and high-dose (2 × 1000 mg) rituximab (RTX) for the treatment of rheumatoid arthritis (RA), considering the recent emergence of new evidence. The systematic literature review searching for randomized controlled trials (RCTs) was updated to November 6, 2014 using the PubMed, EMBASE, Cochrane Library, Web of Science databases, and hand searching. The primary outcomes were the American College of Rheumatology (ACR) criteria for 20 % improvement (ACR20), ACR50, and ACR70 responses and the Disease Activity Score in 28 joints (DAS28) at 24 and 48/52 weeks. The secondary outcomes were change in Health Assessment Questionnaire (HAQ) score, change in the radiographic modified Total Sharp Score (mTSS), levels of immunoglobulin G (IgG), and adverse events. In total, seven RCTs were identified, including two new full publication versions and one abstract of RCTs. There were no significant differences in the primary outcomes and change in HAQ, although the mean change in mTSS was 0.25 units (95 % CI, 0.01 to 0.49; P = 0.04) higher in low-dose group at week 52. Two RCTs did not demonstrate difference between the RTX regimens for maintaining clinical response (obtained initially using high-dose RTX) in anti-TNF-experienced patients. IgG levels were significantly higher (P ≤ 0.02), and first infusion reactions were less frequent in the low-dose group (P = 0.02). Our updated results further support the similar efficacy of both RTX regimens in different subsets of RA patients, demonstrating a better clinical and laboratory safety profile of the low-dose scheme.
Collapse
Affiliation(s)
- Markus Bredemeier
- Rheumatology Service, Hospital Nossa Senhora da Conceição - Grupo Hospitalar Conceição, Av. Francisco Trein, 596, Bloco H, sala 2048, Porto Alegre, 91350-200, RS, Brazil.
| | - Guilherme G Campos
- Rheumatology Service, Hospital Nossa Senhora da Conceição - Grupo Hospitalar Conceição, Av. Francisco Trein, 596, Bloco H, sala 2048, Porto Alegre, 91350-200, RS, Brazil
| | - Fernando K de Oliveira
- Rheumatology Service, Hospital Nossa Senhora da Conceição - Grupo Hospitalar Conceição, Av. Francisco Trein, 596, Bloco H, sala 2048, Porto Alegre, 91350-200, RS, Brazil
| |
Collapse
|