1
|
Perniola S, Alivernini S, Gremese E, Landolfi G, Carrara G, Iagnocco A, Scirè CA. A multiparametric risk table for loss of clinical remission status in patients with rheumatoid arthritis: a STARTER study post-hoc analysis. Rheumatology (Oxford) 2025; 64:526-532. [PMID: 38364299 DOI: 10.1093/rheumatology/keae094] [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: 10/04/2023] [Revised: 12/01/2023] [Accepted: 01/06/2024] [Indexed: 02/18/2024] Open
Abstract
OBJECTIVE This post-hoc analysis was carried out on data acquired in the longitudinal Sonographic Tenosynovitis Assessment in RheumaToid arthritis patiEnts in Remission (STARTER) study. Our primary aim was to determine the predictive clinical and musculoskeletal ultrasonographic (MSUS) features associated with disease flare in RA patients in clinical remission, while our secondary aim was to evaluate the probability of disease flare based on clinical and MSUS features. METHODS We analysed data for a total of 389 RA patients in DAS28-defined remission. All patients underwent a MSUS examination according to the OMERACT guidelines. Logistic regression and results, presented as odds ratio and 95% CI, were used for the evaluation of the association between selected variables and disease flare. Significant clinical and MSUS features were incorporated into a risk table for predicting disease flare within at least 12 months of follow-up in patients with RA remission. RESULTS Within 12 months, 137 (35%) RA patients experienced a disease flare. RA patients who experienced a flare disease differed from those with persistent remission in terms of ACPA positivity (75.9% vs 62.3%, respectively; P = 0.007), percentage of sustained clinical remission at baseline (44.1% vs 68.5%, respectively; P = 0.001) and synovium power Doppler signal presence (58.4% vs 33.3%, respectively; P < 0.001). Based on these results, these three features were considered in a predictive model of disease flare with an adjusted odds ratio of 3.064 (95% CI 1.728-5.432). Finally, a risk table was constructed including the three significant predictive factors of disease flare occurring within 12 months from the enrolment. CONCLUSION An adaptive flare-prediction model tool, based on data available in outpatient settings, was developed as a multiparametric risk table. If confirmed by external validation, this tool might support the defining of therapeutic strategies in RA patients in DAS28-defined remission status.
Collapse
Affiliation(s)
- Simone Perniola
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP) - Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Clinical Immunology Division, Department of Ageing, Neurosciences, Head-neck and Orthopaedics Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefano Alivernini
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP) - Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Rheumatology Division, Department of Ageing, Neurosciences, Head-neck and Orthopaedics Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elisa Gremese
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP) - Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Clinical Immunology Division, Department of Ageing, Neurosciences, Head-neck and Orthopaedics Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Greta Carrara
- Epidemiology Unit, Italian Society for Rheumatology (SIR), Milan, Italy
| | - Annamaria Iagnocco
- Academic Rheumatology Center, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carlo Alberto Scirè
- Epidemiology Unit, Italian Society for Rheumatology (SIR), Milan, Italy
- School of Medicine, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
2
|
Zhang R, Lin X, Lin R, Chen Z, Miao C, Wang Y, Deng X, Lin J, Lin S, Weng S, Chen M. Effectively alleviate rheumatoid arthritis via maintaining redox balance, inducing macrophage repolarization and restoring homeostasis of fibroblast-like synoviocytes by metformin-derived carbon dots. J Nanobiotechnology 2025; 23:58. [PMID: 39881361 PMCID: PMC11776225 DOI: 10.1186/s12951-025-03159-7] [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: 09/17/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Overproduction of reactive oxygen species (ROS), elevated synovial inflammation, synovial hyperplasia and fibrosis are the main characteristic of microenvironment in rheumatoid arthritis (RA). Macrophages and fibroblast-like synoviocytes (FLSs) play crucial roles in the progression of RA. Hence, synergistic combination of ROS scavenging, macrophage polarization from pro-inflammatory M1 phenotype towards M2 anti-inflammatory phenotype, and restoring homeostasis of FLSs will provide a promising therapeutic strategy for RA. In this study, we successfully synthesized metformin-derived carbon dots (MCDs), and investigated the antirheumatic effect in vivo and in vitro. Designed MCDs could target inflamed cells and accumulate at the inflammatory joints of collagen-induced arthritis (CIA) rats. In vivo therapeutic investigation suggested that MCDs reduced synovial inflammation and hyperplasia, ultimately prevented cartilage destruction, bone erosion, and synovial fibrosis in CIA rats. In addition, MCDs eliminated the cellular ROS in M1 phenotype macrophages in RA microenvironment through the enzyme-like catalytic activity as well as inhibiting NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome signaling pathway, effectively polarizing them into the M2 phenotype to realize the anti-inflammatory effect. Furthermore, MCDs could inhibit the proliferation, migration, and fibrosis of inflamed FLSs. Mechanistically, MCDs restored the homeostasis of FLSs while reducing the level of synovial inflammation by blocking IL-6/gp130 signaling pathway. Combined with preferable biocompatibility, MCDs offer a prospective treatment approach for RA.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xingyu Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Zhenbin Chen
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Chenfang Miao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Yao Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Xiaoqin Deng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Jianlong Lin
- Department of Orthopedic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Shishui Lin
- Department of Orthopedic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| | - Shaohuang Weng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Min Chen
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
3
|
Manzo A, Bozzalla Cassione E, Montecucco C, Sakellariou G, Xoxi B, Luvaro T, Sammali Y, De Stefano L, Alpini C, Klersy C, Bugatti S. Prediction of long-term drug-free outcomes in ACPA-positive and ACPA-negative rheumatoid arthritis by combined clinical and ultrasound assessment of residual disease: a 5-year prospective study. RMD Open 2025; 11:e005079. [PMID: 39880410 PMCID: PMC11781140 DOI: 10.1136/rmdopen-2024-005079] [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: 10/02/2024] [Accepted: 01/01/2025] [Indexed: 01/31/2025] Open
Abstract
OBJECTIVE To delineate, within the framework of current clinical practice and criteria, the sustainability of first-line immuno-suppressive treatment discontinuation in rheumatoid arthritis (RA) and the impact of residual disease in remission on long-term drug-free (DF) outcomes. METHODS RA patients, referring to the Pavia early arthritis clinic (EAC) between 2009 and 2021 and achieving remission after Disease Activity Score-driven methotrexate (MTX) monotherapy, were recruited. Eligible patients underwent DF follow-up at 3-month intervals over 5 years after MTX discontinuation. Pre-selected clinical, serological and ultrasound (US) exposure variables at MTX withdrawal were analysed using multivariable Cox regression to predict time-to-flare. RESULTS Of 761 EAC patients with RA, 132 started DF follow-up (person-months: 3678). 62 experienced a flare after a median (range) of 9 (3-60) months, resulting in a progressive decline in flare-free survival throughout the observation period. Whole-cohort multivariate Cox regression identified anti-citrullinated protein antibody (ACPA) positivity (HR: 4.20, 95% CI 2.37 to 7.44) and hands' joints with grey scale (US-GS) alterations (GS>1; HR: 2.18, 95% CI 1.20 to 3.93) as independent predictors. ACPA-positive patients in Simplified Disease Activity Index (SDAI) remission displayed a flare-free survival estimate at 5 years of 6.4% (95% CI 1.2 to 35.7) versus 78.2% (95% CI 67.4 to 90.8) for ACPA-negative patients in SDAI remission without residual US-GS alteration in hands' joints (n=59); the latter group showing no evidence of radiographic progression and functional deterioration. CONCLUSIONS Long-term DF remission is attainable in a niche subset of ACPA-negative RA. Examining clinical and subclinical residual synovial abnormalities during remission allows for effective preemptive identification of this subset in real life.
Collapse
Affiliation(s)
- Antonio Manzo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Emanuele Bozzalla Cassione
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Carlomaurizio Montecucco
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Garifallia Sakellariou
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Istituti Clinici Scientifici Maugeri SpA IRCCS Pavia, Pavia, Italy
| | - Blerina Xoxi
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Terenzj Luvaro
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ylenia Sammali
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ludovico De Stefano
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Claudia Alpini
- Laboratory of Biochemical-Clinical Analyses, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Catherine Klersy
- Biostatistics and Clinical Trial Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Serena Bugatti
- Rheumatology and Translational Immunology Research Laboratories (LaRIT), Department of Internal Medicine and Therapeutics, Universita di Pavia, Pavia, Italy
- Arthritis Research Clinic (ARC), Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
4
|
Putri KG, Sampath S, Richardson CL, McCloskey A, Rathbone AP. A qualitative study exploring experiences of treatment in paediatric rheumatology - children's, young people's, parents' and carers' perspectives. Pediatr Rheumatol Online J 2025; 23:7. [PMID: 39849516 PMCID: PMC11756134 DOI: 10.1186/s12969-025-01063-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 01/18/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND There is limited literature in paediatric rheumatology describing holistic lived experiences of medical treatment from perspectives of children and young people (CYP) and their parents or carers (PC). This is important as it could have implications for adherence. This study aimed to explore treatment experiences of CYP and PC in a paediatric rheumatology service. METHODS Participants were recruited at a day-case unit for intravenous infusions at a tertiary paediatric rheumatology centre. Joint qualitative semi-structured interviews with CYP and PC were used to collect data. Data were transcribed, quality checked and thematically analysed using NVivo 12.4 to identify findings. RESULTS Thirty-two participants (15 CYP between the ages of 6 and 16 years, 17 PC) took part in interviews lasting 41 min and 43 s, on average. Participants described experiences using infliximab, followed by tocilizumab and abatacept. Participants experienced a wave, oscillating between positive and negative trajectories. Experiences of medical treatments were described as temporary, eventually changing and leading to treatment changes or cessation. Behaviours were influenced through somatic factors (pain, function), social factors (advice from health professionals, encouragement from friends, family and teachers, practicality of using treatment in relation to school, work and finance) and cognitive factors (fear of needles, fear of specific medications, beliefs about necessity). CONCLUSIONS Collectively, findings demonstrate experiences of medical treatment reflect the nature of many paediatric rheumatology conditions, oscillating between periods of positive and negative trajectories. Somatic, social and cognitive experiences can be positive, when treatment is considered 'successful'. Negative somatic, social or cognitive experiences led to behaviours such as treatment non-adherence. A limitation of the study is interviews were conducted jointly with CYP and PC, which may have influenced what participants were willing to say in front of one another however this does mean findings relate to both CYP and PC and so could be suitable targets for interventions to improve adherence.
Collapse
Affiliation(s)
- Kassie Gracella Putri
- School of Pharmacy, Faculty of Medical Sciences, Newcastle University, King George VI Building, Queen Victoria Road, Newcastle-upon-Tyne, NE2 4RU, UK
| | - Sunil Sampath
- Great North Children's Hospital, Newcastle upon Tyne NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | - Charlotte Lucy Richardson
- School of Pharmacy, Faculty of Medical Sciences, Newcastle University, King George VI Building, Queen Victoria Road, Newcastle-upon-Tyne, NE2 4RU, UK
| | - Alice McCloskey
- Faculty of Science, Liverpool John Moore's University, Liverpool, UK
| | - Adam Pattison Rathbone
- School of Pharmacy, Faculty of Medical Sciences, Newcastle University, King George VI Building, Queen Victoria Road, Newcastle-upon-Tyne, NE2 4RU, UK.
- Great North Children's Hospital, Newcastle upon Tyne NHS Foundation Trust, Newcastle-upon-Tyne, UK.
| |
Collapse
|
5
|
Bergstra SA, van Ouwerkerk L, Nevins IS, van der Pol JA, Helmich GS, Hest I, van Veen A, Bos R, Goekoop-Ruiterman YPM, Vonkeman HE, Bijsterbosch J, de Jong PHP, Güler-Yüksel M, Böhringer S, Huizinga TWJ, van Gaalen FA. Induction of Cure in Early Arthritis (I CEA): study protocol for an investigator-initiated randomized single-blind clinical trial with open-label extension to compare three treatment strategies in patients with newly diagnosed undifferentiated arthritis. Trials 2024; 25:758. [PMID: 39533416 PMCID: PMC11558825 DOI: 10.1186/s13063-024-08609-5] [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: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Undifferentiated arthritis (UA) is a term used to describe patients with inflammatory arthritis that has not differentiated into a specific rheumatic disease. UA may be a pre-stage of rheumatoid arthritis (RA) or another inflammatory disease or remain undifferentiated, but a substantial proportion of patients may also achieve spontaneous remission. As UA may be an early presentation of RA, rheumatologists often start methotrexate (or another csDMARD) as early as possible. There are however very little data on the potential benefits of early DMARD treatment, and longitudinal data suggests that long-term outcomes such as physical functioning hardly improved in these patients in the past decades. In the I CEA trial, we investigate if it is beneficial to start early treatment with MTX or baricitinib, a more rapidly acting drug with a broader mechanism of action, compared to waiting for spontaneous remission with symptomatic therapy in patients with UA. METHODS The I CEA is a multicenter single-blind (independent assessor) randomized clinical trial with a 3-month interventional and 9-month observational follow-up period. The study includes patients with early (< 1 year symptom duration) DMARD-naïve undifferentiated arthritis. Patients with an increased risk of AEs with baricitinib treatment are excluded. Participants are randomized 1:1:1 to (1) symptom relief with NSAIDs and a single injection of glucocorticoids and (waiting for spontaneous remission); (2) methotrexate and a single injection of glucocorticoids, and NSAIDs are optional; and (3) baricitinib and a single injection of glucocorticoids and NSAIDs are optional. During the observational follow-up period, patients are treated in shared decision with their rheumatologist. The primary outcome will be the change in DAS at 3 months. Secondary outcomes include radiographic progression, physical functioning, patient-reported outcomes, cost utilities, safety, progression to classifiable RA, and disease activity over time. DISCUSSION The 12-month I CEA trial studies early treatment of patients with UA with methotrexate, baricitinib, or NSAIDs. The study initially had a more complex design. Emerging safety warnings about baricitinib necessitated adjustment of the trial protocol including more extensive exclusion criteria. The number of included patients was lower than initially planned, supported by an updated sample size calculation. TRIAL REGISTRATION Dutch trial register NL73202.058.20, EudraCT 2019-004359-35, registered 10-06-2020. Protocol version 1Q, 06-05-2024.
Collapse
Affiliation(s)
- S A Bergstra
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands.
| | - L van Ouwerkerk
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - I S Nevins
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - J A van der Pol
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - G S Helmich
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - I Hest
- Patient Partner Panel, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - A van Veen
- Patient Partner Panel, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - R Bos
- Department of Rheumatology, Medical Center Leeuwarden, Leeuwarden, the Netherlands and Department of Rheumatology, Nij Smellinghe Hospital, Drachten, the Netherlands
| | | | - H E Vonkeman
- Department of Rheumatology and Clinical Immunology, Medisch Spectrum Twente, Enschede, the Netherlands and Department of Psychology, Health and Technology, University of Twente, Enschede, the Netherlands
| | - J Bijsterbosch
- Department of Rheumatology, Amphia Hospital, Breda, the Netherlands
| | - P H P de Jong
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - M Güler-Yüksel
- Department of Rheumatology and Immunology, Maasstad Hospital, Rotterdam, the Netherlands
| | - S Böhringer
- Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands and Department of Pharmacology and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - T W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - F A van Gaalen
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
6
|
Bui VD, Jeon J, Duong VH, Shin S, Lee J, Ghahari F, Kim CH, Jo YJ, Jung WK, Um W, Park JH. Chondroitin sulfate-based microneedles for transdermal delivery of stem cell-derived extracellular vesicles to treat rheumatoid arthritis. J Control Release 2024; 375:105-115. [PMID: 39218160 DOI: 10.1016/j.jconrel.2024.08.050] [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: 06/09/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
For the non-invasive treatment of rheumatoid arthritis (RA), a chondroitin sulfate C (CSC)-based dissolving microneedles (cMN) was prepared to deliver human adipose stem cell-derived extracellular vesicles (hASC-EV) into inflamed joints. Owing to their anti-inflammatory function, the hASC-EV-bearing cMN (EV@cMN) significantly suppressed activated fibroblast-like synoviocytes (aFLS) and M1 macrophages (M1), which are responsible for the progression of RA. In addition, EV@cMN facilitated the chondrogenic differentiation of bone marrow-derived stem cells. In mice with collagen-induced arthritis, EV@cMN efficiently delivered both hASC-EV and CSC to inflamed joints. Interestingly, pro-inflammatory cytokines in the inflamed joints were remarkably downregulated by the synergistic effect of CSC and hASC-EV. Consequently, as judged from the overall clinical score and joint swelling, EV@cMN showed an outstanding therapeutic effect, even comparable to the wild-type mice, without significant adverse effects. Overall, EV@cMN might have therapeutic potential for RA by efficiently delivering CSC and hASC-EV into the inflamed joints in a non-invasive manner.
Collapse
Affiliation(s)
- Van Dat Bui
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jueun Jeon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Van Hieu Duong
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Sol Shin
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jungmi Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Farrokhroo Ghahari
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Yu Jin Jo
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
7
|
Álvaro-Gracia Álvaro JM, Díaz Del Campo Fontecha P, Andréu Sánchez JL, Balsa Criado A, Cáliz Cáliz R, Castrejón Fernández I, Corominas H, Gómez Puerta JA, Manrique Arija S, Mena Vázquez N, Ortiz García A, Plasencia Rodríguez C, Silva Fernández L, Tornero Molina J. Update of the Consensus Statement of the Spanish Society of Rheumatology on the use of biological and synthetic targeted therapies in rheumatoid arthritis. REUMATOLOGIA CLINICA 2024; 20:423-439. [PMID: 39341701 DOI: 10.1016/j.reumae.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVE To update the consensus document of the Spanish Society of Rheumatology (SER) regarding the use of targeted biological and synthetic therapies in rheumatoid arthritis (RA) with the aim of assisting clinicians in their therapeutic decisions. METHODS A panel of 13 experts was assembled through an open call by SER. We employed a mixed adaptation-elaboration-update methodology starting from the 2015 Consensus Document of the Spanish Society of Rheumatology on the use of biological therapies in RA. Starting with systematic reviews (SR) of recommendations from EULAR 2019, American College of Rheumatology 2021, and GUIPCAR 2017, we updated the search strategies for the PICO questions of GUIPCAR. An additional SR was conducted on demyelinating disease in relation to targeted biological and synthetic therapies. Following the analysis of evidence by different panelists, consensus on the wording and level of agreement for each recommendation was reached in a face-to-face meeting. RESULTS The panel established 5 general principles and 15 recommendations on the management of RA. These encompassed crucial aspects such as the importance of early treatment, therapeutic goals in RA, monitoring frequency, the use of glucocorticoids, the application of conventional synthetic disease-modifying antirheumatic drugs (csDMARDs), biological DMARDs (bDMARDs), and targeted synthetic DMARDs. Additionally, recommendations on dose reduction of these drugs in stable patients were included. This update also features recommendations on the use of bDMARDs and Janus Kinase inhibitors in some specific clinical situations, such as patients with lung disease, a history of cancer, heart failure, or demyelinating disease. CONCLUSIONS This update provides recommendations on key aspects in the management of RA using targeted biological and synthetic therapies.
Collapse
Affiliation(s)
- José María Álvaro-Gracia Álvaro
- Servicio de Reumatología, Hospital General Universitario Gregorio Marañón, IiSGM, Universidad Complutense Madrid, Madrid, Spain.
| | | | - José Luis Andréu Sánchez
- Servicio de Reumatología, H.U. Puerta de Hierro Majadahonda, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | - Isabel Castrejón Fernández
- Servicio de Reumatología, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital Gregorio Marañón, Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Hèctor Corominas
- Servicio de Reumatología, Hospital Universitari de la Santa Creu i Sant Pau & Hospital Dos de Maig, Barcelona, Spain
| | | | - Sara Manrique Arija
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma Bionand, UGC de Reumatología, Hospital Regional Universitario de Málaga, Departamento de Medicina, Universidad de Málaga, Málaga, Spain
| | - Natalia Mena Vázquez
- UGC de Reumatología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma Bionand, Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Ana Ortiz García
- Servicio de Reumatología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Lucía Silva Fernández
- Servicio de Reumatología, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Jesús Tornero Molina
- Servicio de Reumatología, Hospital Universitario de Guadalajara, Departamento de Medicina, Universidad de Alcalá de Henares, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
8
|
Heutz JW, Looijen AEM, van der Helm-van Mil AHM, de Jong PHP, van Mulligen E. Tapering csDMARD or TNFi first: is the risk of flares different for ACPA-positive or ACPA-negative rheumatoid arthritis? RMD Open 2024; 10:e004258. [PMID: 39032947 PMCID: PMC11261664 DOI: 10.1136/rmdopen-2024-004258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 07/23/2024] Open
Affiliation(s)
- Judith W Heutz
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Agnes E M Looijen
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Annette H M van der Helm-van Mil
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Pascal H P de Jong
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Elise van Mulligen
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, Netherlands
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
9
|
Gul H, Di Matteo A, Anioke I, Shuweidhi F, Mankia K, Ponchel F, Emery P. Predicting Flare in Patients With Rheumatoid Arthritis in Biologic Induced Remission, on Tapering, and on Stable Therapy. ACR Open Rheumatol 2024; 6:294-303. [PMID: 38411023 PMCID: PMC11089437 DOI: 10.1002/acr2.11656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 02/28/2024] Open
Abstract
OBJECTIVE The tapering of biologic disease-modifying antirheumatic drug (b-DMARD) therapy for patients with rheumatoid arthritis (RA) in stable remission is frequently undertaken, but specific guidance on how to successfully taper is lacking. The objective of this study is to identify predictors of flare in patients in stable b-DMARD-induced clinical remission, who did or did not follow structured b-DMARD tapering. METHODS Patients with RA receiving b-DMARD treatment who had achieved sustained remission according to a Disease Activity Score in 28 joints using the C-reactive protein level (DAS28-CRP) <2.6 for ≥6 months were offered tapering. Clinical, ultrasound (US) (total power Doppler [PD]/grayscale abnormalities), CD4+ T cell subsets, and patient-reported outcomes (PROs) were collected at inclusion. The primary endpoint was the occurrence of flare (loss of DAS28-CRP remission) over 12 months. Logistic regression analyses identified predictors of flare. Dichotomization into high/low-risk groups was based on 80% specificity using the area under the receiving operator curve (AUROC). RESULTS Of 63 patients choosing tapering, 23 (37%) flared compared with 12 of 60 (20%) on stable treatment (P = 0.043). All patients who flared regained remission upon reinstating treatment. In the tapering group, flare was associated with lower regulatory T cell (Treg) (P < 0.0001) and higher CRP levels (P < 0.0001), erythrocyte sedimentation rate (P < 0.035), and inflammation-related cells (IRCs) (P = 0.054); stepwise modeling selected Tregs (odds ratio [OR] = 0.350, P = 0.004), IRCs (OR = 1.871, P = 0.007), and CRP level (OR = 1.577, P = 0.004) with 81.7% accuracy and AUROC = 0.890. In the continued therapy group, modeling retained the tender joint count, total PD, and visual analog scale pain score, with 82.1% accuracy and AUROC = 0.899. Most patients in the study were considered low risk of flare (80 of 123 patients [65%]). Only 5 of 37 (13.5%) of the low-risk patients who tapered flared, which was notable compared with the continued therapy group (20% flare). CONCLUSION Flare on tapering b-DMARDs was predicted by lower Tregs and elevated inflammation biomarkers (IRCs/CRP level); flare on continued b-DMARDs was associated with raised pain parameters and US inflammation. Knowledge of these biomarkers should improve outcomes by targeted selection for tapering, and by increased monitoring of those on continued therapy predicted to flare.
Collapse
Affiliation(s)
| | - Andrea Di Matteo
- University of Leeds and National Institute for Health and Care Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS TrustLeedsUK
| | | | | | - Kulveer Mankia
- University of Leeds and National Institute for Health and Care Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS TrustLeedsUK
| | | | - Paul Emery
- University of Leeds and National Institute for Health and Care Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS TrustLeedsUK
| |
Collapse
|
10
|
Brown P, Pratt AG, Hyrich KL. Therapeutic advances in rheumatoid arthritis. BMJ 2024; 384:e070856. [PMID: 38233032 DOI: 10.1136/bmj-2022-070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Rheumatoid arthritis (RA) is one of the most common immune mediated inflammatory diseases. People with rheumatoid arthritis present with pain, swelling, and stiffness that typically affects symmetrically distributed small and large joints. Without effective treatment, significant joint damage, disability, and work loss develop, owing to chronic inflammation of the joint lining (synovium). Over the past 25 years, the management of this condition has been revolutionized, resulting in substantially higher levels of disease remission and better long term outcomes. This improvement reflects a paradigm shift towards early and aggressive pharmacological intervention coupled with a proliferation in treatment choice, in turn related to enhanced pathobiological understanding and the advent of new drugs for rheumatoid arthritis. Following an overview of these developments from a historical perspective, and with a general audience in mind, this review focuses on newer, targeted treatments in an ever evolving landscape. The review highlights ongoing areas of debate and unmet need, including the proportion of patients with persistent, difficult-to-treat disease, despite recent advances. Also discussed are personalized, strategic approaches to individual patients, the role for imaging in clinical decision making, and the goal of sustained, drug free remission and disease prevention in the future.
Collapse
Affiliation(s)
- Philip Brown
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- National Institute for Health and Care Research Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne Hospitals and Cumbria, Northumberland; and Tyne and Wear NHS Foundation Trusts, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- National Institute for Health and Care Research Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne Hospitals and Cumbria, Northumberland; and Tyne and Wear NHS Foundation Trusts, Newcastle upon Tyne, UK
| | - Kimme L Hyrich
- Centre for Musculoskeletal Research, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
- National Institute for Health and Care Research Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
11
|
Bhat SS, Ahmed S, Reji R, Mehta P, Paul A, Mohanan M, Babu S, Vinayak B, Vijayan A, Nalianda KK, Joseph S, Narayanan K, Padmaja R, Alex G, Shenoy P. Efficacy and safety of curcumin in maintaining remission during disease-modifying antirheumatic drug withdrawal in rheumatoid arthritis at 52 weeks: a phase III double-blind, randomized placebo-controlled trial. Rheumatol Int 2023; 43:2193-2200. [PMID: 37650921 DOI: 10.1007/s00296-023-05417-z] [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: 06/19/2023] [Accepted: 08/01/2023] [Indexed: 09/01/2023]
Abstract
Curcumin has anti-inflammatory properties but current evidence is limited to advocate its use in rheumatoid arthritis (RA). We explored whether curcumin could maintain remission in patients with RA while tapering conventional synthetic Disease-Modifying Antirheumatic Drugs (csDMARD). In this patient-and investigator-blinded trial, adults with RA in sustained remission for more than six months were randomized to oral curcumin (1 g) with piperine (5 mg) twice daily or matching placebo. Patients who had received biological DMARDs or curcumin supplements in the last 6 months were excluded. csDMARD were tapered and stopped sequentially as per a fixed protocol. The primary outcome was flare-free survival at 52 weeks. The secondary outcomes were flare rate, correlation of serum curcuminoid levels with flares and safety. 200 patients (100 per arm) entered the trial with comparable baseline characteristics. Per protocol analysis included 92 and 93 participants in the curcumin and the placebo group, respectively. Flare-free survival at week 52 was similar between both groups (60% versus 64%; p = 0.76). The median time to flare was similar [Curcumin: 219 days (IQR: 123) versus placebo: 214 days (95.8); p = 0.067]. Cox proportionate regression modelling showed that the flare-free survival was independent of serum curcuminoid levels [adjusted HR = 0.99 (95% CI: 0.97-1.0)]. The model showed that flare-free survival was not associated with age, gender, seropositivity, or csDMARD used at baseline. No serious adverse effects were noted. Curcumin did not impact the flare-free survival in patients with RA in remission during the tapering of csDMARDs despite achieving adequate serum levels.Trial registration: CTRI/2018/04/013279.
Collapse
Affiliation(s)
- Sreeja S Bhat
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Sakir Ahmed
- Kalinga Institute of Medical Sciences, Bhubaneswar, India
| | - Reshma Reji
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Pankti Mehta
- King George's Medical University, Lucknow, India
| | - Aby Paul
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Manju Mohanan
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Sageer Babu
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Biju Vinayak
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Anuroopa Vijayan
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Kaveri K Nalianda
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Sanjana Joseph
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - K Narayanan
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - R Padmaja
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Glaxon Alex
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India
| | - Padmanabha Shenoy
- Centre for Arthritis and Rheumatism Excellence (CARE), Dr Shenoys Care, Nettor, Kochi, Kerala, 682040, India.
| |
Collapse
|
12
|
Toghi M, Bitarafan S, Ghafouri-Fard S. Pathogenic Th17 cells in autoimmunity with regard to rheumatoid arthritis. Pathol Res Pract 2023; 250:154818. [PMID: 37729783 DOI: 10.1016/j.prp.2023.154818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Th17 cells contribute the pathobiology of autoimmune diseases, including rheumatoid arthritis (RA). However, it was shown that differentiated Th17 cells display a high degree of plasticity under the influence of inflammatory conditions. In some autoimmune diseases, the majority of Th17 cells, especially at sites of inflammation, have a phenotype that is intermediate between Th17 and Th1. These cells, which are described as Th17.1 or exTh17 cells, are hypothesized to be more pathogenic than classical Th17 cells. In this review, the involvement of Th17.1 lymphocytes in RA, and potential features that might render these cells to be more pathogenic are discussed.
Collapse
Affiliation(s)
- Mehdi Toghi
- Department of Immune and Infectious Diseases, Université Laval, Quebec City, Quebec, Canada
| | - Sara Bitarafan
- Department of Molecular Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Jeljeli MM, Adamopoulos IE. Innate immune memory in inflammatory arthritis. Nat Rev Rheumatol 2023; 19:627-639. [PMID: 37674048 PMCID: PMC10721491 DOI: 10.1038/s41584-023-01009-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 09/08/2023]
Abstract
The concept of immunological memory was demonstrated in antiquity when protection against re-exposure to pathogens was observed during the plague of Athens. Immunological memory has been linked with the adaptive features of T and B cells; however, in the past decade, evidence has demonstrated that innate immune cells can exhibit memory, a phenomenon called 'innate immune memory' or 'trained immunity'. Innate immune memory is currently being defined and is transforming our understanding of chronic inflammation and autoimmunity. In this Review, we provide an up-to-date overview of the memory-like features of innate immune cells in inflammatory arthritis and the crosstalk between chronic inflammatory milieu and cell reprogramming. Aberrant pro-inflammatory signalling, including cytokines, regulates the metabolic and epigenetic reprogramming of haematopoietic progenitors, leading to exacerbated inflammatory responses and osteoclast differentiation, in turn leading to bone destruction. Moreover, imprinted memory on mature cells including terminally differentiated osteoclasts alters responsiveness to therapies and modifies disease outcomes, commonly manifested by persistent inflammatory flares and relapse following medication withdrawal.
Collapse
Affiliation(s)
- Maxime M Jeljeli
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Iannis E Adamopoulos
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Østergaard M, van Vollenhoven RF, Rudin A, Hetland ML, Heiberg MS, Nordström DC, Nurmohamed MT, Gudbjornsson B, Ørnbjerg LM, Bøyesen P, Lend K, Hørslev-Petersen K, Uhlig T, Sokka T, Grondal G, Krabbe S, Lindqvist J, Gjertsson I, Glinatsi D, Kapetanovic MC, Aga AB, Faustini F, Parmanne P, Lorenzen T, Giovanni C, Back J, Hendricks O, Vedder D, Rannio T, Grenholm E, Ljoså MK, Brodin E, Lindegaard H, Söderbergh A, Rizk M, Kastbom A, Larsson P, Uhrenholt L, Just SA, Stevens DJ, Bay Laurbjerg T, Bakland G, Olsen IC, Haavardsholm EA, Lampa J. Certolizumab pegol, abatacept, tocilizumab or active conventional treatment in early rheumatoid arthritis: 48-week clinical and radiographic results of the investigator-initiated randomised controlled NORD-STAR trial. Ann Rheum Dis 2023; 82:1286-1295. [PMID: 37423647 DOI: 10.1136/ard-2023-224116] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND The optimal first-line treatment in early rheumatoid arthritis (RA) is debated. We compared clinical and radiographic outcomes of active conventional therapy with each of three biological treatments with different modes of action. METHODS Investigator-initiated, randomised, blinded-assessor study. Patients with treatment-naïve early RA with moderate-severe disease activity were randomised 1:1:1:1 to methotrexate combined with (1) active conventional therapy: oral prednisolone (tapered quickly, discontinued at week 36) or sulfasalazine, hydroxychloroquine and intra-articular glucocorticoid injections in swollen joints; (2) certolizumab pegol; (3) abatacept or (4) tocilizumab. Coprimary endpoints were week 48 Clinical Disease Activity Index (CDAI) remission (CDAI ≤2.8) and change in radiographic van der Heijde-modified Sharp Score, estimated using logistic regression and analysis of covariance, adjusted for sex, anticitrullinated protein antibody status and country. Bonferroni's and Dunnet's procedures adjusted for multiple testing (significance level: 0.025). RESULTS Eight hundred and twelve patients were randomised. Adjusted CDAI remission rates at week 48 were: 59.3% (abatacept), 52.3% (certolizumab), 51.9% (tocilizumab) and 39.2% (active conventional therapy). Compared with active conventional therapy, CDAI remission rates were significantly higher for abatacept (adjusted difference +20.1%, p<0.001) and certolizumab (+13.1%, p=0.021), but not for tocilizumab (+12.7%, p=0.030). Key secondary clinical outcomes were consistently better in biological groups. Radiographic progression was low, without group differences.The proportions of patients with serious adverse events were abatacept, 8.3%; certolizumab, 12.4%; tocilizumab, 9.2%; and active conventional therapy, 10.7%. CONCLUSIONS Compared with active conventional therapy, clinical remission rates were superior for abatacept and certolizumab pegol, but not for tocilizumab. Radiographic progression was low and similar between treatments. TRIAL REGISTRATION NUMBER NCT01491815.
Collapse
Affiliation(s)
- Mikkel Østergaard
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ronald F van Vollenhoven
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Rheumatology Clinic, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Merete Lund Hetland
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Marte Schrumpf Heiberg
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Dan C Nordström
- Division of Internal Medicine and Rheumatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Michael T Nurmohamed
- Location VUmc, Reade and Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Bjorn Gudbjornsson
- Landspitali University Hospital, University of Iceland, Reykjavik, Iceland
- Department of Rheumatology, Centre for Rheumatology Research, Reykjavik, Iceland
| | - Lykke Midtbøll Ørnbjerg
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Copenhagen, Denmark
| | - Pernille Bøyesen
- Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | - Kristina Lend
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam Rheumatology Center, Amsterdam, Netherlands
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Kim Hørslev-Petersen
- Department of Rheumatology, Danish Hospital for Rheumatic Diseases, Sønderborg, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Till Uhlig
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Tuulikki Sokka
- Jyväskylä Central Hospital, University of Eastern Finland, Jyväskylä, Finland
| | - Gerdur Grondal
- Landspitali University Hospital, University of Iceland, Reykjavik, Iceland
| | - Simon Krabbe
- Department of Radiology, Herlev-Gentofte University Hospital, Herlev, Denmark
| | - Joakim Lindqvist
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Inger Gjertsson
- Rheumatology Clinic, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| | - Daniel Glinatsi
- Copenhagen Center for Arthritis Research (COPECARE), Center for Rheumatology and Spine Diseases, Centre of Head and Orthopaedics, Rigshospitalet, Rigshospitalet, Glostrup, Denmark
- Department of Rheumatology, Skaraborg Hospital, Skövde, Sweden
| | | | | | - Francesca Faustini
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Pinja Parmanne
- Division of Internal Medicine and Rheumatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tove Lorenzen
- Department of Rheumatology, Silkeborg University Hospital, Silkeborg, Denmark
| | - Cagnotto Giovanni
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Rheumatology, Skåne University Hospital, Lund, Sweden
| | - Johan Back
- Department of Medical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Oliver Hendricks
- Department of Rheumatology, Danish Hospital for Rheumatic Diseases, Sønderborg, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Daisy Vedder
- Department of Rheumatology, Reade, Amsterdam, Netherlands
| | - Tuomas Rannio
- Jyväskylä Central Hospital, University of Eastern Finland, Jyväskylä, Finland
| | | | | | - Eli Brodin
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - Hanne Lindegaard
- Rheumatology Research Unit, Odense University Hospital, Odense, Denmark
| | - Annika Söderbergh
- Department of Rheumatology, Örebro University Hospital, Orebro, Sweden
| | - Milad Rizk
- Department of Rheumatology, Västmanlands Hospital Västerås, Västerås, Sweden
| | - Alf Kastbom
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Rheumatology in Östergötland, Linköping, Sweden
| | - Per Larsson
- Academic Specialist Center, Stockholm, Sweden
| | - Line Uhrenholt
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark
| | - Søren Andreas Just
- Department of Rheumatology, Odense Universitetshospital, Odense, Denmark
- Section of Rheumatology, Department of Medicine, Svendborg Hospital, Svendborg, Denmark
| | - David J Stevens
- Department of Rheumatology, St. Olavs Hospital, University Hospital of Trondheim, Trondheim, Norway
| | | | - Gunnstein Bakland
- Department of Rheumatology, University Hospital of Northern Norway, Tromsø, Norway
- Department Rheumatology, University of Tromsø, Tromsø, Norway
| | - Inge Christoffer Olsen
- Department of Research Support for Clinical Trials, Oslo University Hospital, Oslo, Norway
| | - Espen A Haavardsholm
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Jon Lampa
- Department of Medicine, Rheumatology Unit, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
van Esveld L, Cox JM, Kuijper TM, Bosch TM, Weel-Koenders AE. Cost-utility analysis of tapering strategies of biologicals in rheumatoid arthritis patients in the Netherlands. Ann Rheum Dis 2023; 82:1296-1306. [PMID: 37423648 DOI: 10.1136/ard-2023-224190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/21/2023] [Indexed: 07/11/2023]
Abstract
OBJECTIVES Current guidelines recommend tapering biological disease-modifying antirheumatoid drugs (bDMARDs) in rheumatoid arthritis (RA) if the disease is under control. However, guidelines on tapering are lacking. Assessing cost-effectiveness of different tapering strategies might provide broader input for creating guidelines on how to taper bDMARDs in patients with RA. The aim of this study is to evaluate the long-term cost-effectiveness from a societal perspective of bDMARD tapering strategies in Dutch patients with RA, namely 50% dose reduction (tapering), discontinuation and a 50% dose reduction followed by discontinuation (de-escalation). METHODS Using a societal perspective, a Markov model with a life-time horizon of 30 years was used to simulate 3-monthly transitions between Disease Activity 28 (DAS28)-defined health states of remission (<2.6), low disease activity (2.63.2). Transition probabilities were estimated through literature search and random effects pooling. Incremental costs, incremental quality-adjusted life-years (QALYs), incremental cost-effectiveness ratios (ICERs) and incremental net monetary benefits for each tapering strategy were compared with continuation. Deterministic, probabilistic sensitivity analyses and multiple scenario analyses were performed. RESULTS After 30 years, the ICERs were €115 157/QALY lost, €74 226/QALY lost and €67 137/QALY lost for tapering, de-escalation and discontinuation, respectively; mainly driven by bDMARD cost savings and a 72.8% probability of a loss in quality of life. This corresponds to a 76.1%, 64.3% and 60.1% probability of tapering, de-escalation and discontinuation being cost-effective, provided a willingness-to-accept threshold of €50 000/QALY lost. CONCLUSIONS Based on these analyses, the 50% tapering approach saved the highest cost per QALY lost.
Collapse
Affiliation(s)
| | - Juul M Cox
- Hospital Pharmacy, Maasstad Hospital, Rotterdam, The Netherlands
- Clinical Pharmacology and Toxicology, MaasstadLab Maasstad Hospital, Rotterdam, The Netherlands
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | | | - Tessa M Bosch
- Hospital Pharmacy, Maasstad Hospital, Rotterdam, The Netherlands
- Clinical Pharmacology and Toxicology, MaasstadLab Maasstad Hospital, Rotterdam, The Netherlands
| | - Angelique Eam Weel-Koenders
- Rheumatology, Maasstad Hospital, Rotterdam, The Netherlands
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
16
|
Singh V, Naldi A, Soliman S, Niarakis A. A large-scale Boolean model of the rheumatoid arthritis fibroblast-like synoviocytes predicts drug synergies in the arthritic joint. NPJ Syst Biol Appl 2023; 9:33. [PMID: 37454172 DOI: 10.1038/s41540-023-00294-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune disease with an unknown aetiology. However, rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) play a significant role in initiating and perpetuating destructive joint inflammation by expressing immuno-modulating cytokines, adhesion molecules, and matrix remodelling enzymes. In addition, RA-FLS are primary drivers of inflammation, displaying high proliferative rates and an apoptosis-resistant phenotype. Thus, RA-FLS-directed therapies could become a complementary approach to immune-directed therapies by predicting the optimal conditions that would favour RA-FLS apoptosis, limit inflammation, slow the proliferation rate and minimise bone erosion and cartilage destruction. In this paper, we present a large-scale Boolean model for RA-FLS that consists of five submodels focusing on apoptosis, cell proliferation, matrix degradation, bone erosion and inflammation. The five-phenotype-specific submodels can be simulated independently or as a global model. In silico simulations and perturbations reproduced the expected biological behaviour of the system under defined initial conditions and input values. The model was then used to mimic the effect of mono or combined therapeutic treatments and predict novel targets and drug candidates through drug repurposing analysis.
Collapse
Affiliation(s)
- Vidisha Singh
- Université Paris-Saclay, Laboratoire Européen de Recherche pour la Polyarthrite rhumatoïde-Genhotel, Univ Evry, Evry, France
| | - Aurelien Naldi
- Lifeware Group, Inria, Saclay-île de France, 91120, Palaiseau, France
| | - Sylvain Soliman
- Lifeware Group, Inria, Saclay-île de France, 91120, Palaiseau, France
| | - Anna Niarakis
- Université Paris-Saclay, Laboratoire Européen de Recherche pour la Polyarthrite rhumatoïde-Genhotel, Univ Evry, Evry, France.
- Lifeware Group, Inria, Saclay-île de France, 91120, Palaiseau, France.
| |
Collapse
|
17
|
Chen SF, Yeh FC, Chen CY, Chang HY. Tailored therapeutic decision of rheumatoid arthritis using proteomic strategies: how to start and when to stop? Clin Proteomics 2023; 20:22. [PMID: 37301840 DOI: 10.1186/s12014-023-09411-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Unpredictable treatment responses have been an obstacle for the successful management of rheumatoid arthritis. Although numerous serum proteins have been proposed, there is a lack of integrative survey to compare their relevance in predicting treatment outcomes in rheumatoid arthritis. Also, little is known about their applications in various treatment stages, such as dose modification, drug switching or withdrawal. Here we present an in-depth exploration of the potential usefulness of serum proteins in clinical decision-making and unveil the spectrum of immunopathology underlying responders to different drugs. Patients with robust autoimmunity and inflammation are more responsive to biological treatments and prone to relapse during treatment de-escalation. Moreover, the concentration changes of serum proteins at the beginning of the treatments possibly assist early recognition of treatment responders. With a better understanding of the relationship between the serum proteome and treatment responses, personalized medicine in rheumatoid arthritis will be more achievable in the near future.
Collapse
Affiliation(s)
- Shuo-Fu Chen
- Department of Heavy Particles & Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Fu-Chiang Yeh
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Yun Chen
- Department of Biomedical Sciences and Engineering, Institute of Biomedical Engineering and Nanomedicine, National Central University, Taoyuan, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Hui-Yin Chang
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, No. 300, Zhongda Rd., Zhongli District, Taoyuan, 320317, Taiwan.
| |
Collapse
|
18
|
Stephan M, Tascilar K, Yalcin-Mutlu M, Hagen M, Haschka J, Reiser M, Hartmann F, Kleyer A, Hueber AJ, Manger B, Figueiredo C, Cobra JF, Tony HP, Finzel S, Kleinert S, Wendler J, Schuch F, Ronneberger M, Feuchtenberger M, Fleck M, Manger K, Ochs W, Schmitt-Haendle M, Lorenz HM, Nüsslein H, Alten R, Henes J, Krüger K, Schett G, Rech J. Physical Function of RA patients Tapering Treatment-A Post Hoc Analysis of the Randomized Controlled RETRO Trial. J Clin Med 2023; 12:3723. [PMID: 37297917 PMCID: PMC10253840 DOI: 10.3390/jcm12113723] [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: 04/24/2023] [Revised: 05/15/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Several studies have shown that tapering or stopping disease-modifying anti-rheumatic drugs (DMARDs) in rheumatoid arthritis (RA) patients in sustained remission is feasible. However, tapering/stopping bears the risk of decline in physical function as some patients may relapse and face increased disease activity. Here, we analyzed the impact of tapering or stopping DMARD treatment on the physical function of RA patients. The study was a post hoc analysis of physical functional worsening for 282 patients with RA in sustained remission tapering and stopping DMARD treatment in the prospective randomized RETRO study. HAQ and DAS-28 scores were determined in baseline samples of patients continuing DMARD (arm 1), tapering their dose by 50% (arm 2), or stopping after tapering (arm 3). Patients were followed over 1 year, and HAQ and DAS-28 scores were evaluated every 3 months. The effect of treatment reduction strategy on functional worsening was assessed in a recurrent-event Cox regression model with a study-group (control, taper, and taper/stop) as the predictor. Two-hundred and eighty-two patients were analyzed. In 58 patients, functional worsening was observed. The incidences suggest a higher probability of functional worsening in patients tapering and/or stopping DMARDs, which is likely due to higher relapse rates in these individuals. At the end of the study, however, functional worsening was similar among the groups. Point estimates and survival curves show that the decline in functionality according to HAQ after tapering or discontinuation of DMARDs in RA patients with stable remission is associated with recurrence, but not with an overall functional decline.
Collapse
Affiliation(s)
- Marlene Stephan
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Koray Tascilar
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Melek Yalcin-Mutlu
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Melanie Hagen
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Judith Haschka
- Karl Landsteiner Institute for Gastroenterology and Rheumatology, 1100 Vienna, Austria;
- Ludwig Boltzmann Institute of Osteology, I Medical Department, Hanusch Hospital Vienna, 1140 Vienna, Austria
| | - Michaela Reiser
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Fabian Hartmann
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Axel J. Hueber
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Bernhard Manger
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Camille Figueiredo
- Institutio de Rheumatologia, Sao Paolo 01317-001, Brazil; (C.F.); (J.F.C.)
| | | | - Hans-Peter Tony
- Rheumatology/Clinical Immunology, Department of Internal Medicine II, University of Würzburg, 97080 Würzburg, Germany;
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Stefan Kleinert
- Rheumatology Clinical Practice Erlangen, 91054 Erlangen, Germany (J.W.); (F.S.); (M.R.)
| | - Jörg Wendler
- Rheumatology Clinical Practice Erlangen, 91054 Erlangen, Germany (J.W.); (F.S.); (M.R.)
| | - Florian Schuch
- Rheumatology Clinical Practice Erlangen, 91054 Erlangen, Germany (J.W.); (F.S.); (M.R.)
| | - Monika Ronneberger
- Rheumatology Clinical Practice Erlangen, 91054 Erlangen, Germany (J.W.); (F.S.); (M.R.)
| | - Martin Feuchtenberger
- Rheumatology Practice and Department of Internal Medicine 2, Clinic Burghausen, 84489 Burghausen, Germany;
| | - Martin Fleck
- Asklepios Medical Center, Department of Rheumatology and Clinical Immunology, 93077 Bad Abbach, Germany
| | - Karin Manger
- Rheumatology Practice Bamberg, 96047 Bamberg, Germany
| | - Wolfgang Ochs
- Rheumatology Practice Bayreuth, 95444 Bayreuth, Germany (M.S.-H.)
| | | | - Hannes Martin Lorenz
- Department of Medicine V, Center for Rheumatic Diseases Baden-Baden, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | | | - Rieke Alten
- Schlosspark Klinik, Internal Medicine/Rheumatology, 14059 Berlin, Germany
| | - Joerg Henes
- Centre for Interdisciplinary Clinical Immunology, University of Tübingen, 72076 Tübingen, Germany
| | - Klaus Krüger
- Praxiszentrum St. Bonifatius, 81541 Munich, Germany;
| | - Georg Schett
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Jürgen Rech
- Department of Internal Medicine 3, FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.S.); (M.Y.-M.); (F.H.); (A.K.); (A.J.H.); (B.M.); (G.S.)
- Deutsches Zentrum fuer Immuntherapie (DZI), FAU Erlangen-Nuremberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
19
|
Emery P, Tanaka Y, Bykerk VP, Bingham CO, Huizinga TWJ, Citera G, Huang KHG, Wu C, Connolly SE, Elbez Y, Wong R, Lozenski K, Fleischmann R. The trajectory of clinical responses in patients with early rheumatoid arthritis who achieve sustained remission in response to abatacept: subanalysis of AVERT-2, a randomized phase IIIb study. Arthritis Res Ther 2023; 25:67. [PMID: 37087459 PMCID: PMC10122306 DOI: 10.1186/s13075-023-03038-2] [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: 09/02/2022] [Accepted: 03/27/2023] [Indexed: 04/24/2023] Open
Abstract
BACKGROUND AVERT-2 (a phase IIIb, two-stage study) evaluated abatacept + methotrexate versus methotrexate alone, in methotrexate-naive, anti-citrullinated protein antibody-positive patients with early (≤ 6 months), active RA. This subanalysis investigated whether individual patients who achieved the week 24 Simplified Disease Activity Index (SDAI) remission primary endpoint could sustain remission to 1 year and then maintain it following changes in therapy. METHODS During the 56-week induction period (IP), patients were randomized to weekly subcutaneous abatacept 125 mg + methotrexate or abatacept placebo + methotrexate. Patients completing the IP who achieved SDAI remission (≤ 3.3) at weeks 40 and 52 entered a 48-week de-escalation (DE) period. Patients treated with abatacept + methotrexate were re-randomized to continue weekly abatacept + methotrexate, or de-escalate and then withdraw abatacept (after 24 weeks), or receive abatacept monotherapy. Proportions of patients achieving sustained SDAI and Boolean remission, and Disease Activity Score in 28 joints using C-reactive protein (DAS28 [CRP]) < 2.6, were assessed. For patients achieving early sustained SDAI remission at weeks 24/40/52, flow between disease activity categories and individual trajectories was evaluated; flow was also evaluated for later remitters (weeks 40/52 but not week 24). RESULTS Among patients treated with abatacept + methotrexate (n/N = 451/752) at IP week 24, 22% achieved SDAI remission, 17% achieved Boolean remission, and 42% achieved DAS28 (CRP) < 2.6; of these, 56%, 58%, and 74%, respectively, sustained a response throughout IP weeks 40/52. Among patients with a sustained response at IP weeks 24/40/52, 82% (14/17) on weekly abatacept + methotrexate, 81% (13/16) on abatacept monotherapy, 63% (12/19) who de-escalated/withdrew abatacept, and 65% (11/17) on abatacept placebo + methotrexate were in SDAI remission at end of the DE period; rates were higher than for later remitters in all arms except abatacept placebo + methotrexate. CONCLUSIONS A high proportion of individual patients achieving clinical endpoints at IP week 24 with abatacept + methotrexate sustained their responses through week 52. Of patients achieving early and sustained SDAI remission through 52 weeks, numerically more maintained remission during the DE period if weekly abatacept treatment continued. TRIAL REGISTRATION NCT02504268 (ClinicalTrials.gov), registered July 21, 2015.
Collapse
Affiliation(s)
- Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and Leeds NIHR Biomedical Research Centre, Leeds, UK.
| | - Yoshiya Tanaka
- University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | | | | - Gustavo Citera
- Instituto de Rehabilitación Psicofísica, Buenos Aires, Argentina
| | | | - Chun Wu
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | - Roy Fleischmann
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
20
|
Kitamura N, Kobayashi H, Nagasawa Y, Sugiyama K, Tsuzuki H, Tanikawa Y, Ikumi N, Okada Y, Takahashi Y, Asai S, Tamura N, Ogasawara M, Kawamoto T, Kuwatsuru R, Tamaki H, Kidoguchi G, Tateishi M, Kimura M, Mochida Y, Harigane K, Shimazaki T, Koike T, Tanimura K, Kataoka H, Amano K, Yasuoka H, Takei M. Risk factors associated with relapse after methotrexate dose reduction in patients with rheumatoid arthritis receiving golimumab and methotrexate combination therapy. Int J Rheum Dis 2023. [PMID: 37058849 DOI: 10.1111/1756-185x.14695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/06/2023] [Accepted: 03/25/2023] [Indexed: 04/16/2023]
Abstract
AIM To identify risk factors for relapse after methotrexate (MTX) dose reduction in rheumatoid arthritis (RA) patients receiving golimumab (GLM)/MTX combination therapy. METHOD Data on RA patients ≥20 years old receiving GLM (50 mg) + MTX for ≥6 months were retrospectively collected. MTX dose reduction was defined as a reduction of ≥12 mg from the total dose within 12 weeks of the maximum dose (≥1 mg/wk average). Relapse was defined as Disease Activity Score in 28 joints using C-reactive protein level (DAS28-CRP) score ≥3.2 or sustained (≥ twice) increase of ≥0.6 from baseline. RESULTS A total of 304 eligible patients were included. Among the MTX-reduction group (n = 125), 16.8% of patients relapsed. Age, duration from diagnosis to the initiation of GLM, baseline MTX dose, and DAS28-CRP were comparable between relapse and no-relapse groups. The adjusted odds ratio (aOR) of relapse after MTX reduction was 4.37 (95% CI 1.16-16.38, P = 0.03) for prior use of non-steroidal anti-inflammatory drugs (NSAIDs), and the aORs for cardiovascular disease (CVD), gastrointestinal disease and liver disease were 2.36, 2.28, and 3.03, respectively. Compared to the non-reduction group, the MTX-reduction group had a higher proportion of patients with CVD (17.6% vs 7.3%, P = 0.02) and a lower proportion of prior use of biologic disease-modifying antirheumatic drugs (11.2% vs. 24.0%, P = 0.0076). CONCLUSION Attention should be given to RA patients with history of CVD, gastrointestinal disease, liver disease, or prior NSAIDs-use when considering MTX dose reduction to ensure benefits outweigh the risks of relapse.
Collapse
Affiliation(s)
- Noboru Kitamura
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Hitomi Kobayashi
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Yosuke Nagasawa
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Kaita Sugiyama
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Hiroshi Tsuzuki
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Yutaka Tanikawa
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| | - Natsumi Ikumi
- Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Yuito Okada
- Clinical Trials Research Center, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuo Takahashi
- Clinical Trials Research Center, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Asai
- Department of Pharmacology and Biofunction Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Naoto Tamura
- Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Michihiro Ogasawara
- Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshio Kawamoto
- Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Ryohei Kuwatsuru
- Department of Radiology & Center for Promotion of Data Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiromichi Tamaki
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Genki Kidoguchi
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Mutsuto Tateishi
- Department of Rheumatology, Tokyo Metropolitan Ohtsuka Hospital, Tokyo, Japan
| | - Makiko Kimura
- Department of Rheumatology, Tokyo Metropolitan Ohtsuka Hospital, Tokyo, Japan
| | - Yuichi Mochida
- Center for Rheumatic Diseases, Yokohama City University Medical Center, Yokohama, Japan
| | - Kengo Harigane
- Center for Rheumatic Diseases, Yokohama City University Medical Center, Yokohama, Japan
| | - Takayuki Shimazaki
- Center for Rheumatic Diseases, Yokohama City University Medical Center, Yokohama, Japan
| | - Takao Koike
- Hokkaido Medical Center for Rheumatic Diseases, Sapporo, Japan
| | | | - Hiroshi Kataoka
- Department of Rheumatology and Clinical Immunology, Sapporo City General Hospital, Sapporo, Japan
| | - Koichi Amano
- Department of Rheumatology and Clinical Immunology, Saitama Medical Center Saitama Medical University, Saitama, Japan
| | - Hidekata Yasuoka
- Department of Internal Medicine, Division of Rheumatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masami Takei
- Department of Medicine, Division of Hematology and Rheumatology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Perniola S, Chimenti MS, Spinelli FR, Frediani B, Foti R, Ferrigno S, Garufi C, Cassone G, Venerito V, Atzeni F, Caporali R, Conti F, Favalli EG, Iannone F, Sebastiani M, Ferraccioli GF, Lapadula G, Gremese E. Rheumatoid Arthritis from Easy to Complex Disease: From the "2022 GISEA International Symposium". J Clin Med 2023; 12:jcm12082781. [PMID: 37109118 PMCID: PMC10143148 DOI: 10.3390/jcm12082781] [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: 02/20/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
Rheumatoid Arthritis (RA) is a systemic disease with many different clinical phenotypes. RA could be classified according to disease duration, seropositivity for rheumatoid factor (RF) and/or anti-citrullinated protein antibodies (ACPA), joint subtype, clinical behaviourbehavior and many other subgroups. In this review, we summarize and discuss the multifaceted aspects of RA, focusing on the relationship between autoimmunity status and clinical outcome, achievement of remission and influence on treatment response, from the 2022 International GISEA/OEG Symposium.
Collapse
Affiliation(s)
- Simone Perniola
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS-Rome, 00168 Rome, Italy
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore-Rome, 00168 Rome, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of "Medicina dei Sistemi", University of Rome Tor Vergata, 00133 Rome, Italy
| | - Francesca Romana Spinelli
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza Università di Roma, 00185 Rome, Italy
| | - Bruno Frediani
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Rosario Foti
- Rheumatology Unit, San Marco Hospital, Policlinico University of Catania, 95124 Catania, Italy
| | - Sara Ferrigno
- Rheumatology, Allergology and Clinical Immunology, Department of "Medicina dei Sistemi", University of Rome Tor Vergata, 00133 Rome, Italy
| | - Cristina Garufi
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza Università di Roma, 00185 Rome, Italy
| | - Giulia Cassone
- Rheumatology Unit, Azienda Ospedaliera Policlinico di Modena, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Vincenzo Venerito
- Department of Emergency and Organ Transplantation, Rheumatology Unit, University of Bari, 70121 Bari, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, 98122 Messina, Italy
| | - Roberto Caporali
- Department of Rheumatology and Clinical Sciences, ASST Gaetano Pini-CTO, 20126 Milan, Italy
| | - Fabrizio Conti
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza Università di Roma, 00185 Rome, Italy
| | - Ennio Giulio Favalli
- Department of Rheumatology and Clinical Sciences, ASST Gaetano Pini-CTO, 20126 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Florenzo Iannone
- Department of Emergency and Organ Transplantation, Rheumatology Unit, University of Bari, 70121 Bari, Italy
| | - Marco Sebastiani
- Rheumatology Unit, Azienda Ospedaliera Policlinico di Modena, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Gian Franco Ferraccioli
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS-Rome, 00168 Rome, Italy
| | - Giovanni Lapadula
- Department of Emergency and Organ Transplantation, Rheumatology Unit, University of Bari, 70121 Bari, Italy
| | - Elisa Gremese
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS-Rome, 00168 Rome, Italy
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore-Rome, 00168 Rome, Italy
| |
Collapse
|
22
|
Lillegraven S, Paulshus Sundlisæter N, Aga AB, Sexton J, Solomon DH, van der Heijde D, Haavardsholm EA. Discontinuation of Conventional Synthetic Disease-Modifying Antirheumatic Drugs in Patients With Rheumatoid Arthritis and Excellent Disease Control. JAMA 2023; 329:1024-1026. [PMID: 36976288 PMCID: PMC10051094 DOI: 10.1001/jama.2023.0492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/13/2023] [Indexed: 03/29/2023]
Abstract
This open-label randomized clinical trial assessed the 12-month risk of disease activity flares after discontinuation of conventional synthetic DMARDs (csDMARDs) compared with continuing half-dose csDMARDs in adult Norwegian patients with rheumatoid arthritis and excellent disease control.
Collapse
Affiliation(s)
- Siri Lillegraven
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Nina Paulshus Sundlisæter
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Anna-Birgitte Aga
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Joseph Sexton
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Daniel H. Solomon
- Division of Rheumatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Espen A. Haavardsholm
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
23
|
Brewer RC, Lanz TV, Hale CR, Sepich-Poore GD, Martino C, Swafford AD, Carroll TS, Kongpachith S, Blum LK, Elliott SE, Blachere NE, Parveen S, Fak J, Yao V, Troyanskaya O, Frank MO, Bloom MS, Jahanbani S, Gomez AM, Iyer R, Ramadoss NS, Sharpe O, Chandrasekaran S, Kelmenson LB, Wang Q, Wong H, Torres HL, Wiesen M, Graves DT, Deane KD, Holers VM, Knight R, Darnell RB, Robinson WH, Orange DE. Oral mucosal breaks trigger anti-citrullinated bacterial and human protein antibody responses in rheumatoid arthritis. Sci Transl Med 2023; 15:eabq8476. [PMID: 36812347 PMCID: PMC10496947 DOI: 10.1126/scitranslmed.abq8476] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 02/02/2023] [Indexed: 02/24/2023]
Abstract
Periodontal disease is more common in individuals with rheumatoid arthritis (RA) who have detectable anti-citrullinated protein antibodies (ACPAs), implicating oral mucosal inflammation in RA pathogenesis. Here, we performed paired analysis of human and bacterial transcriptomics in longitudinal blood samples from RA patients. We found that patients with RA and periodontal disease experienced repeated oral bacteremias associated with transcriptional signatures of ISG15+HLADRhi and CD48highS100A2pos monocytes, recently identified in inflamed RA synovia and blood of those with RA flares. The oral bacteria observed transiently in blood were broadly citrullinated in the mouth, and their in situ citrullinated epitopes were targeted by extensively somatically hypermutated ACPAs encoded by RA blood plasmablasts. Together, these results suggest that (i) periodontal disease results in repeated breaches of the oral mucosa that release citrullinated oral bacteria into circulation, which (ii) activate inflammatory monocyte subsets that are observed in inflamed RA synovia and blood of RA patients with flares and (iii) activate ACPA B cells, thereby promoting affinity maturation and epitope spreading to citrullinated human antigens.
Collapse
Affiliation(s)
- R. Camille Brewer
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Tobias V. Lanz
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Caryn R. Hale
- Rockefeller University, New York City, NY 10065, USA
| | | | - Cameron Martino
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Austin D. Swafford
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Thomas S. Carroll
- Bioinformatics Resource Center, Rockefeller University, 1230 York Ave., New York, NY 10065, USA
| | - Sarah Kongpachith
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Lisa K. Blum
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Serra E. Elliott
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Nathalie E. Blachere
- Rockefeller University, New York City, NY 10065, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | - John Fak
- Rockefeller University, New York City, NY 10065, USA
| | - Vicky Yao
- Department of Computer Science, Rice University, Houston, TX 77005, USA
- Department of Computer Science, Princeton University, Princeton, NJ, 08544, USA
| | - Olga Troyanskaya
- Department of Computer Science, Princeton University, Princeton, NJ, 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
- Flatiron Institute, Simons Foundation, New York, NY, 10010, USA
| | - Mayu O. Frank
- Rockefeller University, New York City, NY 10065, USA
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Alejandro M. Gomez
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Radhika Iyer
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Nitya S. Ramadoss
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Orr Sharpe
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | | | - Lindsay B. Kelmenson
- Division of Rheumatology, University of Colorado - Denver, Aurora, CO, 80045, USA
| | - Qian Wang
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Heidi Wong
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | | | - Mark Wiesen
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Dana T. Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kevin D. Deane
- Division of Rheumatology, University of Colorado - Denver, Aurora, CO, 80045, USA
| | - V. Michael Holers
- Division of Rheumatology, University of Colorado - Denver, Aurora, CO, 80045, USA
| | - Rob Knight
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Robert B. Darnell
- Rockefeller University, New York City, NY 10065, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Dana E. Orange
- Rockefeller University, New York City, NY 10065, USA
- Hospital for Special Surgery, New York City, NY 10075, USA
| |
Collapse
|
24
|
Labinsky H, Ukalovic D, Hartmann F, Runft V, Wichmann A, Jakubcik J, Gambel K, Otani K, Morf H, Taubmann J, Fagni F, Kleyer A, Simon D, Schett G, Reichert M, Knitza J. An AI-Powered Clinical Decision Support System to Predict Flares in Rheumatoid Arthritis: A Pilot Study. Diagnostics (Basel) 2023; 13:148. [PMID: 36611439 PMCID: PMC9818406 DOI: 10.3390/diagnostics13010148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/11/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Treat-to-target (T2T) is a main therapeutic strategy in rheumatology; however, patients and rheumatologists currently have little support in making the best treatment decision. Clinical decision support systems (CDSSs) could offer this support. The aim of this study was to investigate the accuracy, effectiveness, usability, and acceptance of such a CDSS-Rheuma Care Manager (RCM)-including an artificial intelligence (AI)-powered flare risk prediction tool to support the management of rheumatoid arthritis (RA). Longitudinal clinical routine data of RA patients were used to develop and test the RCM. Based on ten real-world patient vignettes, five physicians were asked to assess patients' flare risk, provide a treatment decision, and assess their decision confidence without and with access to the RCM for predicting flare risk. RCM usability and acceptance were assessed using the system usability scale (SUS) and net promoter score (NPS). The flare prediction tool reached a sensitivity of 72%, a specificity of 76%, and an AUROC of 0.80. Perceived flare risk and treatment decisions varied largely between physicians. Having access to the flare risk prediction feature numerically increased decision confidence (3.5/5 to 3.7/5), reduced deviations between physicians and the prediction tool (20% to 12% for half dosage flare prediction), and resulted in more treatment reductions (42% to 50% vs. 20%). RCM usability (SUS) was rated as good (82/100) and was well accepted (mean NPS score 7/10). CDSS usage could support physicians by decreasing assessment deviations and increasing treatment decision confidence.
Collapse
Affiliation(s)
- Hannah Labinsky
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | | | - Fabian Hartmann
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | | | | | | | - Kira Gambel
- Siemens Healthineers, 91502 Erlangen, Germany
| | | | - Harriet Morf
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Jule Taubmann
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - David Simon
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | | | - Johannes Knitza
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
25
|
Mazzoni M, Pistorio A, Magnaguagno F, Viola S, Urru A, Magnano GM, Ravelli A, Malattia C. Predictive Value of Magnetic Resonance Imaging in Patients With Juvenile Idiopathic Arthritis in Clinical Remission. Arthritis Care Res (Hoboken) 2023; 75:198-205. [PMID: 34286915 PMCID: PMC10087925 DOI: 10.1002/acr.24757] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/14/2020] [Accepted: 07/15/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To define the prevalence of subclinical synovitis on magnetic resonance imaging (MRI) in a large cohort of patients with juvenile idiopathic arthritis (JIA) in clinical remission and to evaluate its predictive value in terms of disease flare and joint deterioration. METHODS Ninety patients with clinically inactive JIA who underwent a contrast-enhanced (CE)-MRI of a previously affected joint were retrospectively included. Each joint was evaluated for synovitis, tenosynovitis, and bone marrow edema. Baseline and follow-up radiographs were assessed to evaluate structural damage progression. RESULTS CE-MRI was acquired in 45 wrists, 30 hips, 13 ankles, and 2 knees. Subclinical synovitis was detected in 59 (65.5%) of 90 patients and bone marrow edema in 42 (46.7%) of 90 patients. Fifty-seven of 90 (63.3%) patients experienced a disease flare during follow-up. Forty-four of 59 (74.6%) patients with subclinical synovitis experienced a disease flare versus 13 (41.9%) of 31 patients with no residual synovitis on MRI (P = 0.002). The presence of subclinical synovitis was the best predictor of disease flare on multivariable regression analysis (hazard ratio [HR] 2.45, P = 0.003). Baseline and follow-up radiographs were available for 54 patients, and 17 (31.5%) of 54 patients experienced radiographic damage progression. The presence of bone marrow edema (HR 4.40, P = 0.045) and being >17 years old (HR 3.51, P = 0.04) were strong predictors of joint damage progression in the multivariable analysis. CONCLUSION MRI-detected subclinical inflammation was present in a large proportion of patients with JIA despite clinical remission. Subclinical synovitis and bone marrow edema have been shown to play a role in predicting the risk of disease relapse and joint deterioration, with potential implications for patients' management of the disease.
Collapse
Affiliation(s)
- Marta Mazzoni
- Università degli Studi di Genova, Dipartimento di Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Genova, Italy
| | - Angela Pistorio
- Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Francesca Magnaguagno
- IRCCS Istituto Giannina Gaslini, UOC di Radiologia, Istituto Giannina Gaslini, Genova, Italy
| | - Stefania Viola
- IRCCS Istituto Giannina Gaslini, Clinica Pediatrica e Reumatologia, Genova, Italy
| | - Alessia Urru
- IRCCS Istituto Giannina Gaslini, Clinica Pediatrica e Reumatologia, Genova, Italy
| | - Gian Michele Magnano
- IRCCS Istituto Giannina Gaslini, UOC di Radiologia, Istituto Giannina Gaslini, Genova, Italy
| | - Angelo Ravelli
- Università degli Studi di Genova, Dipartimento di Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Clara Malattia
- Università degli Studi di Genova, Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Genova, Italy and Clinica Pediatrica e Reumatologia, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
26
|
Terslev L, Ostergaard M, Georgiadis S, Brahe CH, Ellegaard K, Dohn UM, Fana V, Møller T, Juul L, Huynh TK, Krabbe S, Ornbjerg LM, Glinatsi D, Røgind H, Hansen A, Nørregaard J, Jacobsen S, Jensen DV, Manilo N, Asmussen K, Boesen M, Rastiemadabadi Z, Morsel-Carlsen L, Møller JM, Krogh NS, Hetland ML. Flare during tapering of biological DMARDs in patients with rheumatoid arthritis in routine care: characteristics and predictors. RMD Open 2022; 8:rmdopen-2022-002796. [PMID: 36549857 PMCID: PMC9791469 DOI: 10.1136/rmdopen-2022-002796] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To identify predictors of flare in a 2-year follow-up study of patients with rheumatoid arthritis (RA) in sustained clinical remission tapering towards withdrawal of biological disease-modifying anti-rheumatic drugs (bDMARDs). METHODS Sustained clinical remission was defined as Disease Activity Score for 28 joints (DAS28)-C reactive protein (CRP) ≤2.6 without radiographic progression for >1 year. bDMARDs were tapered according to a mandatory clinical guideline to two-thirds of standard dose at baseline, half of dose at week 16 and discontinuation at week 32. Prospective assessments for 2 years included clinical evaluation, conventional radiography, ultrasound and MRI for signs of inflammation and bone changes. Flare was defined as DAS28-CRP ≥2.6 with ∆DAS28-CRP ≥1.2 from baseline. Baseline predictors of flare were assessed by logistic regression analyses. RESULTS Of 142 included patients, 121 (85%) flared during follow-up of which 86% regained remission within 24 weeks after flare. Patients that flared were more often rheumatoid factor positive, had tried more bDMARDs and had higher baseline ultrasound synovitis sum scores than those not flaring. For patients on standard dose, predictors of flare within 16 weeks after reduction to two-thirds of standard dose were baseline MRI-osteitis (OR 1.16; 95% CI 1.03 to 1.33; p=0.014), gender (female) (OR 6.71; 95% CI 1.68 to 46.12; p=0.005) and disease duration (OR 1.06; 95% CI 1.01 to 1.11; p=0.020). Baseline predictors for flare within 2 years were ultrasound grey scale synovitis sum score (OR 1.19; 95% CI 1.02 to 1.44; p=0.020) and number of previous bDMARDs (OR 4.07; 95% CI 1.35 to 24.72; p=0.007). CONCLUSION The majority of real-world patients with RA tapering bDMARDs flared during tapering, with the majority regaining remission after stepwise dose increase. Demographic and imaging parameters (MR-osteitis/ultrasound greyscale synovitis) were independent predictors of immediate flare and flare overall and may be of importance for clinical decision-making in patients eligible for tapering.
Collapse
Affiliation(s)
- L Terslev
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Ostergaard
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stylianos Georgiadis
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Cecilie Heegaard Brahe
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Karen Ellegaard
- The Parker Institute, Bispebjerg og Frederiksberg Hospital, Frederiksberg, Denmark
| | - UM Dohn
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Viktoria Fana
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Torsten Møller
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Lars Juul
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Tuan Khai Huynh
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark,Holte Rheumatology Clinic, Capital Region, Holte, Denmark
| | - Simon Krabbe
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - L M Ornbjerg
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Daniel Glinatsi
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark,Department of Rheumatology, Skaraborg Hospital Skövde, Skovde, Sweden
| | - Henrik Røgind
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Annette Hansen
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Jesper Nørregaard
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Søren Jacobsen
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dorte V Jensen
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark,Center for Rheumatology and Spine Diseases, Center of Head and Orthopedics, Danbio Registry, Glostrup, Denmark
| | - Natalia Manilo
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Karsten Asmussen
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Mikael Boesen
- Department of Radiology, Bispebjerg and Frederiksberg University Hospitals, Copenhagen, Denmark
| | | | - Lone Morsel-Carlsen
- Department of Radiology, Bispebjerg and Frederiksberg University Hospitals, Copenhagen, Denmark
| | | | | | - Merete Lund Hetland
- Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet Glostrup, Glostrup, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Alivernini S, Firestein GS, McInnes IB. The pathogenesis of rheumatoid arthritis. Immunity 2022; 55:2255-2270. [PMID: 36516818 DOI: 10.1016/j.immuni.2022.11.009] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/20/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022]
Abstract
Significant recent progress in understanding rheumatoid arthritis (RA) pathogenesis has led to improved treatment and quality of life. The introduction of targeted-biologic and -synthetic disease modifying anti-rheumatic drugs (DMARDs) has also transformed clinical outcomes. Despite this, RA remains a life-long disease without a cure. Unmet needs include partial response and non-response to treatment in many patients, failure to achieve immune homeostasis or drug free remission, and inability to repair damaged tissues. RA is now recognized as the end of a multi-year prodromal phase in which systemic immune dysregulation, likely beginning in mucosal surfaces, is followed by a symptomatic clinical phase. Inflammation and immune reactivity are primarily localized to the synovium leading to pain and articular damage, but is also associated with a broader series of comorbidities. Here, we review recently described immunologic mechanisms that drive breach of tolerance, chronic synovitis, and remission.
Collapse
Affiliation(s)
- Stefano Alivernini
- Immunology Research Core Facility, Gemelli Science and Technology Park, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Division of Rheumatology - Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gary S Firestein
- Division of Rheumatology, Allergy, and Immunology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| | | |
Collapse
|
28
|
Shin K, Kwon HM, Kim MJ, Yoon MJ, Chai HG, Kang SW, Park W, Park SH, Suh CH, Kim HA, Lee SG, Lee CK, Bae SC, Park YB, Song YW. Two-year clinical outcomes after discontinuation of long-term golimumab therapy in Korean patients with rheumatoid arthritis. Korean J Intern Med 2022; 37:1061-1069. [PMID: 34883551 PMCID: PMC9449209 DOI: 10.3904/kjim.2021.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/15/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS The aim of this study was to investigate long-term post-discontinuation outcomes in patients with rheumatoid arthritis (RA) who had been treated with tumor necrosis factor-α inhibitors (TNF-αi) which was then discontinued. METHODS Sixty Korean patients with RA who participated in a 5-year GO-BEFORE and GO-FORWARD extension trials were included in this retrospective study. Golimumab was deliberately discontinued after the extension study (baseline). Patients were then followed by their rheumatologists. We reviewed their medical records for 2 years (max 28 months) following golimumab discontinuation. Patients were divided into a maintained benefit (MB) group and a loss-of-benefit (LB) group based on treatment pattern after golimumab discontinuation. The LB group included patients whose conventional disease-modifying antirheumatic drug(s) were stepped-up or added/switched (SC) and those who restarted biologic therapy (RB). RESULTS The mean age of patients at baseline was 56.5 years and 55 (91.7%) were females. At the end of follow-up, 23 (38.3%) patients remained in the MB group. In the LB group, 75.7% and 24.3% were assigned into SC and RB subgroups, respectively. Fifty percent of patients lost MB after 23.3 months. Demographics and clinical variables at baseline were comparable between MB and LB groups except for age, C-reactive protein level, and corticosteroid use. Restarting biologic therapy was associated with swollen joint count (adjusted hazard ratio [HR], 1.90; 95% confidence interval [CI], 1.01 to 3.55) and disease duration (adjusted HR, 1.12; 95% CI, 1.02 to 1.23) at baseline. CONCLUSION Treatment strategies after discontinuing TNF-αi are needed to better maintain disease control and quality of life of patients with RA.
Collapse
Affiliation(s)
- Kichul Shin
- Division of Rheumatology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul,
Korea
| | - Hyun Mi Kwon
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
| | - Min Jung Kim
- Division of Rheumatology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul,
Korea
| | - Myung Jae Yoon
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
| | - Hyun Gyung Chai
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
| | - Seong-Wook Kang
- Division of Rheumatology, Chungnam National University Hospital, Daejeon,
Korea
| | - Won Park
- Division of Rheumatology, Inha University Hospital, Incheon,
Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| | - Chang Hee Suh
- Division of Rheumatology, Ajou University Hospital, Suwon,
Korea
| | - Hyun Ah Kim
- Division of Rheumatology, Hallym University Sacred Heart Hospital, Anyang,
Korea
| | - Seung-Geun Lee
- Division of Rheumatology, Pusan National University Hospital, Busan,
Korea
| | - Choong Ki Lee
- Division of Rheumatology, Yeungnam University Medical Center, Daegu,
Korea
| | - Sang-Cheol Bae
- Division of Rheumatology, Hanyang University Seoul Hospital, Seoul,
Korea
| | - Yong-Beom Park
- Division of Rheumatology, Severance Hospital, Yonsei University College of Medicine, Seoul,
Korea
| | - Yeong Wook Song
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine, Medical Research Center, Seoul National University, Seoul,
Korea
| |
Collapse
|
29
|
Birkner B, Rech J, Edelmann E, Verheyen F, Schett G, Stargardt T. Patient-individual tapering of DMARDs in rheumatoid arthritis patients in a real-world setting. Rheumatology (Oxford) 2022; 62:1476-1484. [PMID: 35980267 DOI: 10.1093/rheumatology/keac472] [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: 03/06/2022] [Revised: 07/17/2022] [Accepted: 07/17/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE We aim to provide real-world evidence on the effectiveness of patient-individual tapering of DMARDs for patients with rheumatoid arthritis (RA) in daily clinical practice using medical records and claims data. METHODS We utilize data obtained through a controlled prospective cohort study in Germany conducted from July 2018 to March 2021. Participants consist of RA patients in sustained remission (>6 months) who were eligible for tapering at enrolment. Patients treated with individual tapering based on shared decision making (n = 200) are compared with patients without any dose-reduction (n = 237). The risk of loss of remission and the risk of flare is assessed with risk-adjusted Kaplan-Meier estimators and Cox-Regressions. We evaluate differences in costs one year before and after baseline based on claims data for the subgroup of patients insured at one major sickness fund in Germany (n = 76). RESULTS The risk of flare (HR 0.88 95%-CI: 0.59-1.30) or loss of remission (HR 1.04 95%-CI: 0.73, 1.49) was not statistically different between the individual tapering group and the continuation group. Minor increases of disease activity and decreases of quality of life were observed 12 months after baseline, again with no statistically significant difference. Drug costs decreased by 1,017€in the individual tapering group while increased by 1,151€in the continuation group (p< 0.01). CONCLUSION Individual tapering of DMARDs does not increase the average risk of experiencing flares or loss of remission. Encouraging rheumatologists and patients to apply tapering in shared decision may be a feasible approach to allow individualisation of treatment in RA.
Collapse
Affiliation(s)
- Benjamin Birkner
- Hamburg Center for Health Economics (HCHE), Universität Hamburg, Hamburg, Germany
| | - Jürgen Rech
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | | | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tom Stargardt
- Hamburg Center for Health Economics (HCHE), Universität Hamburg, Hamburg, Germany
| |
Collapse
|
30
|
Chaiamnuay S, Jiemjit S, Songdechaphipat W, Narongroeknawin P, Pakchotanon R, Asavatanabodee P. Predictors of flare in rheumatoid arthritis patients with persistent clinical remission/low disease activity: Data from the TARAC cohort. Medicine (Baltimore) 2022; 101:e29974. [PMID: 35960097 PMCID: PMC9371488 DOI: 10.1097/md.0000000000029974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
To identify predictors of rheumatoid arthritis (RA) disease activity flare in RA patients who achieved low disease activity (LDA) or persistent remission from the observational Thai Army Rheumatoid Arthritis Cohort study. RA patients with persistent clinical remission, defined by disease activity score 28 (DAS28) < 2.6 and LDA defined by DAS28 ≤ 3.2 for 3 consecutive months, were recruited and followed-up for at least 2 years. The flare was defined by an escalation of DAS28 ≥ 1.2 plus their physicians' decision to enhance RA treatment. Differences between sustained remission/LDA and flare groups were analyzed, by Chi-square test and unpaired Student t test. Multivariate Cox proportional hazard regression analysis was conducted to determine flare predictors. From 199 RA patients, female were 82.9%. Anticitrullinated peptide antibodies (ACPA) or Rheumatoid factor (RF) were found in 69.8% of patients. Flares occurred in 69 patients (34.9%). Multivariate analysis found that the timescale from symptoms emergence to DMARD commencement, the timescale from DMARD commencement to when RA patients showed remission/LDA, the occurrence of RF or ACPA, LDA (in contrast to remission) and the increased DAS28 score when remission/LDA was achieved and tapering DMARDs promptly when persistent remission/LDA was achieved were predictors of RA flares with hazard ratios of (95% confidence interval [CI]) of 1.017 (1.003-1.030), 1.037 (1.015-1.059), 1.949 (1.035-3.676), 1.926 (0.811-4.566), 2.589 (1.355-4.947), and 2.497 (1.458-4.276), respectively. These data demonstrated that early and aggressive DMARDs treatment approach could maintain remission espcially in seropositive patients. Tapering should be applied minimally 6 months after reaching remission.
Collapse
Affiliation(s)
- Sumapa Chaiamnuay
- Rheumatic Disease Unit, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
- * Correspondence: Sumapa Chaiamnuay, MD, Rheumatic Disease Unit, Department of Internal Medicine, Phramongkutklao Hospital & Phramongkutklao College of Medicine, 315 Ratchawithi Road Ratchathewi district, Bangkok 10400, Thailand (e-mail: )
| | - Srisakul Jiemjit
- Rheumatic Disease Unit, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | | | | | - Rattapol Pakchotanon
- Rheumatic Disease Unit, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - Paijit Asavatanabodee
- Rheumatic Disease Unit, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| |
Collapse
|
31
|
Lei S, Li Z, Zhang X, Zhou S. Efficacy and safety of progressively reducing biologic disease-modifying antirheumatic drugs in patients with rheumatoid arthritis in persistent remission: a study protocol for a non-inferiority randomized, controlled, single-blind trial. Trials 2022; 23:600. [PMID: 35897052 PMCID: PMC9327307 DOI: 10.1186/s13063-022-06543-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Background To compare the effects of two biologic disease-modifying antirheumatic drug (bDMARD) administration strategies on the maintenance effect and safety of patients with rheumatoid arthritis (RA) in remission, to analyze the effects of gradual drug reduction and dose maintenance treatment on clinical outcomes in patients who have achieved remission with different types of bDMARDs, to search and screen out people who may benefit from drug reduction strategies, and to provide references for drug reduction strategies and treatment options for patients with RA in remission, so as to help improve the safety of the treatment and reduce the economic burden. Methods The study will be a 24-month non-inferiority randomized, controlled, single-blind trial and is planned to be launched in our hospital from September 2021 to August 2023. Patients will be randomized in a ratio of 2:1 to two groups: maintenance or injection spacing by 50%/gradual reduction of dosage every 3 months up to complete stop. When the patient relapses, return to the last effective dose. If the remission can be maintained, the medication of bDMARDs can be stopped 9 months after enrollment. The primary outcome will be the persistent flare rate. Discussion Our study may provide a reference for the selection of drug reduction strategies and treatment options for patients with RA in remission, so as to help improve the safety of the treatment and reduce the economic burden. Trial registration Chinese Clinical Trial Registry ChiCTR2100044751. Registered on 26 March 2021
Collapse
Affiliation(s)
- Shangwen Lei
- Department of Rheumatology, Gansu Provincial People's Hospital, 204 Donggang West Road, Lanzhou, 730000, People's Republic of China.
| | - Zijia Li
- Department of Rheumatology, Gansu Provincial People's Hospital, 204 Donggang West Road, Lanzhou, 730000, People's Republic of China
| | - Xiaoli Zhang
- Department of Thoracic surgery, Gansu Provincial People's Hospital, Lanzhou, 730000, People's Republic of China
| | - Shuhong Zhou
- Department of Rheumatology, Gansu Provincial People's Hospital, 204 Donggang West Road, Lanzhou, 730000, People's Republic of China
| |
Collapse
|
32
|
Kurowska-Stolarska M, Alivernini S. Synovial tissue macrophages in joint homeostasis, rheumatoid arthritis and disease remission. Nat Rev Rheumatol 2022; 18:384-397. [PMID: 35672464 DOI: 10.1038/s41584-022-00790-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Synovial tissue macrophages (STMs) were principally recognized as having a pro-inflammatory role in rheumatoid arthritis (RA), serving as the main producers of pathogenic tumour necrosis factor (TNF). Recent advances in single-cell omics have facilitated the discovery of distinct STM populations, providing an atlas of discrete phenotypic clusters in the context of healthy and inflamed joints. Interrogation of the functions of distinct STM populations, via ex vivo and experimental mouse models, has re-defined our understanding of STM biology, opening up new opportunities to better understand the pathology of the arthritic joint. These works have identified STM subpopulations that form a protective lining barrier within the synovial membrane and actively participate in the remission of RA. We discuss how distinct functions of STM clusters shape the synovial tissue environment in health, during inflammation and in disease remission, as well as how an increased understanding of STM heterogeneity might aid the prediction of clinical outcomes and inform novel treatments for RA.
Collapse
Affiliation(s)
- Mariola Kurowska-Stolarska
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK.
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | - Stefano Alivernini
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK.
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Rome, Italy.
| |
Collapse
|
33
|
Luo P, Wang P, Xu J, Hou W, Xu P, Xu K, Liu L. Immunomodulatory role of T helper cells in rheumatoid arthritis : a comprehensive research review. Bone Joint Res 2022; 11:426-438. [PMID: 35775145 PMCID: PMC9350707 DOI: 10.1302/2046-3758.117.bjr-2021-0594.r1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that involves T and B cells and their reciprocal immune interactions with proinflammatory cytokines. T cells, an essential part of the immune system, play an important role in RA. T helper 1 (Th1) cells induce interferon-γ (IFN-γ), tumour necrosis factor-α (TNF-α), and interleukin (IL)-2, which are proinflammatory cytokines, leading to cartilage destruction and bone erosion. Th2 cells primarily secrete IL-4, IL-5, and IL-13, which exert anti-inflammatory and anti-osteoclastogenic effects in inflammatory arthritis models. IL-22 secreted by Th17 cells promotes the proliferation of synovial fibroblasts through induction of the chemokine C-C chemokine ligand 2 (CCL2). T follicular helper (Tfh) cells produce IL-21, which is key for B cell stimulation by the C-X-C chemokine receptor 5 (CXCR5) and coexpression with programmed cell death-1 (PD-1) and/or inducible T cell costimulator (ICOS). PD-1 inhibits T cell proliferation and cytokine production. In addition, there are many immunomodulatory agents that promote or inhibit the immunomodulatory role of T helper cells in RA to alleviate disease progression. These findings help to elucidate the aetiology and treatment of RA and point us toward the next steps. Cite this article: Bone Joint Res 2022;11(7):426–438.
Collapse
Affiliation(s)
- Pan Luo
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Peixu Wang
- Department of Orthopedics, China-Japan Friendship Hospital, China-Japan Friendship Institute of Clinical Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Graduate School of Peking Union Medical College, Beijing, China
| | - Jiawen Xu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Weikun Hou
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lin Liu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
34
|
Duong SQ, Crowson CS, Athreya A, Atkinson EJ, Davis JM, Warrington KJ, Matteson EL, Weinshilboum R, Wang L, Myasoedova E. Clinical predictors of response to methotrexate in patients with rheumatoid arthritis: a machine learning approach using clinical trial data. Arthritis Res Ther 2022; 24:162. [PMID: 35778714 PMCID: PMC9248180 DOI: 10.1186/s13075-022-02851-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Methotrexate is the preferred initial disease-modifying antirheumatic drug (DMARD) for rheumatoid arthritis (RA). However, clinically useful tools for individualized prediction of response to methotrexate treatment in patients with RA are lacking. We aimed to identify clinical predictors of response to methotrexate in patients with rheumatoid arthritis (RA) using machine learning methods. METHODS Randomized clinical trials (RCT) of patients with RA who were DMARD-naïve and randomized to placebo plus methotrexate were identified and accessed through the Clinical Study Data Request Consortium and Vivli Center for Global Clinical Research Data. Studies with available Disease Activity Score with 28-joint count and erythrocyte sedimentation rate (DAS28-ESR) at baseline and 12 and 24 weeks were included. Latent class modeling of methotrexate response was performed. The least absolute shrinkage and selection operator (LASSO) and random forests methods were used to identify predictors of response. RESULTS A total of 775 patients from 4 RCTs were included (mean age 50 years, 80% female). Two distinct classes of patients were identified based on DAS28-ESR change over 24 weeks: "good responders" and "poor responders." Baseline DAS28-ESR, anti-citrullinated protein antibody (ACPA), and Health Assessment Questionnaire (HAQ) score were the top predictors of good response using LASSO (area under the curve [AUC] 0.79) and random forests (AUC 0.68) in the external validation set. DAS28-ESR ≤ 7.4, ACPA positive, and HAQ ≤ 2 provided the highest likelihood of response. Among patients with 12-week DAS28-ESR > 3.2, ≥ 1 point improvement in DAS28-ESR baseline-to-12-week was predictive of achieving DAS28-ESR ≤ 3.2 at 24 weeks. CONCLUSIONS We have developed and externally validated a prediction model for response to methotrexate within 24 weeks in DMARD-naïve patients with RA, providing variably weighted clinical features and defined cutoffs for clinical decision-making.
Collapse
Affiliation(s)
- Stephanie Q Duong
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Cynthia S Crowson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.,Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Arjun Athreya
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | | - John M Davis
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kenneth J Warrington
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Eric L Matteson
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Elena Myasoedova
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA. .,Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Kim SH, Kim H, Jeong SH, Jang SY, Park EC. Impact of continuity of care on risk for major osteoporotic fracture in patients with new onset rheumatoid arthritis. Sci Rep 2022; 12:10189. [PMID: 35715560 PMCID: PMC9205920 DOI: 10.1038/s41598-022-14368-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/17/2022] [Indexed: 11/24/2022] Open
Abstract
There is a clear relationship between rheumatoid arthritis (RA) and major osteoporotic fracture (MOF), although there is limited evidence on the effect of continuity of care (COC) on MOF in these patients. We investigated the association between COC and risk of MOF, including fractures of the lumbar spine and pelvis, forearm, and hip, among newly diagnosed RA patients aged ≥ 60 years. A total of 8715 incident RA patients from 2004 to 2010 were included from the Korean National Health Insurance Service-Senior cohort database. Participants were categorized into a good and bad COC group according to the COC index. The cumulative incidence of MOF was higher in RA patients with bad than in those with good COC (p < 0.001). The incidence rates of MOF were 4439 and 3275 cases per 100,000 person-years in patients with bad and good COC, respectively. RA patients with bad COC had an increased incidence of overall MOF (adjusted hazard ratio, 1.32; 95% confidence interval, 1.14–1.53), with the highest increase in risk being that of forearm fracture. An increased MOF risk in patients with bad COC was predominantly observed in females. This study suggested that interventions that can improve COC in patients with RA should be considered.
Collapse
Affiliation(s)
- Seung Hoon Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea.,Department of Public Health, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Hyunkyu Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea.,Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Hoon Jeong
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea.,Department of Public Health, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Suk-Yong Jang
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea.,Department of Healthcare Management, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Eun-Cheol Park
- Department of Preventive Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea. .,Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Kashiwado Y, Kiyohara C, Kimoto Y, Nagano S, Sawabe T, Oryoji K, Mizuki S, Nishizaka H, Yoshizawa S, Yoshizawa S, Tsuru T, Inoue Y, Ueda N, Ota SI, Suenaga Y, Miyamura T, Tada Y, Niiro H, Akashi K, Horiuchi T. Clinical course of patients with rheumatoid arthritis who continue or discontinue biologic therapy after hospitalization for infection: a retrospective observational study. Arthritis Res Ther 2022; 24:131. [PMID: 35650638 PMCID: PMC9158270 DOI: 10.1186/s13075-022-02820-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 05/21/2022] [Indexed: 05/31/2023] Open
Abstract
Background To analyse the subsequent clinical course of patients with rheumatoid arthritis (RA) who either continued or discontinued biologic agents after hospitalization for infections. Methods We retrospectively reviewed the clinical records of 230 RA patients with 307 hospitalizations for infections under biologic therapy between September 2008 and May 2014 in 15 institutions for up to 18 months after discharge. The risks of RA flares and subsequent hospitalizations for infections from 61 days to 18 months after discharge were evaluated. Results Survival analyses indicated that patients who continued biologic therapy had a significantly lower risk of RA flares (31.4% vs. 60.6%, P < 0.01) and a slightly lower risk of subsequent infections (28.7% vs. 34.5%, P = 0.37). Multivariate analysis showed that discontinuation of biologic therapy, diabetes, and a history of hospitalization for infection under biologic therapy were associated with RA flares. Oral steroid therapy equivalent to prednisolone 5 mg/day or more and chronic renal dysfunction were independent risk factors for subsequent hospitalizations for infections. Conclusions Discontinuation of biologic therapy after hospitalization for infections may result in RA flares. Continuation of biologic therapy is preferable, particularly in patients without immunodeficiency. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02820-y.
Collapse
Affiliation(s)
- Yusuke Kashiwado
- Department of Internal Medicine, Kyushu University Beppu Hospital, 4546 Tsurumibaru, Beppu, Oita, 874-0838, Japan
| | - Chikako Kiyohara
- Department of Preventive Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasutaka Kimoto
- Department of Internal Medicine, Kyushu University Beppu Hospital, 4546 Tsurumibaru, Beppu, Oita, 874-0838, Japan
| | - Shuji Nagano
- Department of Rheumatology, Aso Iizuka Hospital, Iizuka, Japan
| | - Takuya Sawabe
- Department of Rheumatology, Hiroshima Red Cross Hospital & Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Kensuke Oryoji
- The Center for Rheumatic Diseases, Matsuyama Red Cross Hospital, Matsuyama, Japan
| | - Shinichi Mizuki
- The Center for Rheumatic Diseases, Matsuyama Red Cross Hospital, Matsuyama, Japan
| | - Hiroaki Nishizaka
- Department of Rheumatology, Kitakyushu Municipal Medical Center, Kitakyushu, Japan
| | - Seiji Yoshizawa
- Department of Rheumatology, Hamanomachi Hospital, Fukuoka, Japan
| | - Shigeru Yoshizawa
- Department of Rheumatology, National Hospital Organization Fukuoka Hospital, Fukuoka, Japan
| | - Tomomi Tsuru
- Department of Rheumatology, Med.Co. LTA PS Clinic, Fukuoka, Japan
| | - Yasushi Inoue
- Department of Rheumatology, Japanese Red Cross Fukuoka Hospital, Fukuoka, Japan
| | - Naoyasu Ueda
- Department of Rheumatology and Infection, Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Shun-Ichiro Ota
- Department of Rheumatology, Internal medicine and connective tissue disorders, Shimonoseki City Hospital, Shimonoseki, Japan
| | - Yasuo Suenaga
- Department of Rheumatology, Beppu Medical Center, NHO, Beppu, Japan
| | - Tomoya Miyamura
- Department of Internal Medicine and Rheumatology, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Yoshifumi Tada
- Department of Rheumatology, Saga University Hospital, Saga, Japan
| | - Hiroaki Niiro
- Department of Medical Education, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, 4546 Tsurumibaru, Beppu, Oita, 874-0838, Japan.
| |
Collapse
|
37
|
Ahn SS, Pyo JY, Song JJ, Park YB, Lee SW. Anti-Citrullinated Peptide Antibody Expression and Its Association with Clinical Features and Outcomes in Patients with Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58040558. [PMID: 35454396 PMCID: PMC9025032 DOI: 10.3390/medicina58040558] [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] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 11/22/2022]
Abstract
Background and objectives: Anti-citrullinated peptide antibody (ACPA), a characteristic antibody detected in rheumatoid arthritis, could be linked to antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) via the formation of neutrophil extracellular traps. We investigated the rate of ACPA positivity in patients with AAV and evaluated the association of ACPAs with their clinical features and outcomes. Materials and Methods: A total of 168 AAV patients with both ACPA and ANCA results at diagnosis were identified. Clinical and laboratory variables, including the disease-specific indices of Birmingham Vasculitis Activity Score (BVAS) and Five-Factor Score (FFS), were investigated. All-cause mortality, relapse, and end-stage renal disease, as well as interstitial lung disease (ILD) were evaluated as outcomes of the patients, and the Kaplan–Meier survival analysis was used to compare the event-free survival rates of the groups. Results: Fifteen (8.9%) and 135 (80.4%) patients were positive for ACPA and ANCA, respectively. There were no significant differences in the baseline variables of ACPA-negative and ACPA-positive patients. The absolute titre of ACPAs also did not significantly correlate with BVAS, FFS, erythrocyte sedimentation rate, or C-reactive protein. In addition, there was no difference noted regarding overall, relapse-free, and ESRD-free survival rates between ACPA-negative and ACPA-positive AAV patients. However, when the patients were divided into four groups according to ACPA and ANCA status, differences were present in the outcomes, and the ACPA-positive ANCA-positive group exhibited the lowest cumulative relapse-free survival rate, while no significant difference was present in the relapse between the ANCA-positive ANCA-positive, ACPA-positive ANCA-negative, and ACPA-negative ANCA-positive groups. Finally, the cumulative ILD-free survival rates were comparable between ACPA-positive and ACPA-negative AAV patients. Conclusions: The detection of ACPA expression is not uncommon in AAV. However, the presence of ACPA did not influence patients’ basal characteristics and outcomes, suggesting that further exploration of the role of this antibody is needed in patients with AAV.
Collapse
Affiliation(s)
- Sung Soo Ahn
- Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Korea;
| | - Jung Yoon Pyo
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea; (J.Y.P.); (J.J.S.); (Y.-B.P.)
| | - Jasong Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea; (J.Y.P.); (J.J.S.); (Y.-B.P.)
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea; (J.Y.P.); (J.J.S.); (Y.-B.P.)
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea; (J.Y.P.); (J.J.S.); (Y.-B.P.)
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence:
| |
Collapse
|
38
|
Defining the Optimal Strategies for Achieving Drug-Free Remission in Rheumatoid Arthritis: A Narrative Review. Healthcare (Basel) 2021; 9:healthcare9121726. [PMID: 34946453 PMCID: PMC8701994 DOI: 10.3390/healthcare9121726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022] Open
Abstract
Background: It is now accepted that the optimum treatment goal for rheumatoid arthritis (RA) is sustained remission, as this has been shown to be associated with the best patient outcomes. There is little guidance on how to manage patients once remission is achieved; however, it is recommended that patients can taper therapy, with a view to discontinuing and achieving drug-free remission if treatment goals are maintained. This narrative review aims to present the current literature on drug-free remission in rheumatoid arthritis, with a view to identifying which strategies are best for disease-modifying anti-rheumatic drug (DMARD) tapering and to highlight areas of unmet clinical need. Methods: We performed a narrative review of the literature, which included research articles, meta-analyses and review papers. The key search terms included were rheumatoid arthritis, remission, drug-free remission, b-DMARDS/biologics, cs-DMARDS and tapering. The databases that were searched included PubMed and Google Scholar. For each article, the reference section of the paper was reviewed to find additional relevant articles. Results: It has been demonstrated that DFR is possible in a proportion of RA patients achieving clinically defined remission (both on cs and b-DMARDS). Immunological, imaging and clinical associations with/predictors of DFR have all been identified, including the presence of autoantibodies, absence of Power Doppler (PD) signal on ultrasound (US), lower disease activity according to composite scores of disease activity and lower patient-reported outcome scores (PROs) at treatment cessation. Conclusions: DFR in RA may be an achievable goal in certain patients. This carries importance in reducing medication-induced side-effects and potential toxicity, the burden of taking treatment if not required and cost effectiveness, specifically for biologic therapy. Prospective studies of objective biomarkers will help facilitate the prediction of successful treatment discontinuation.
Collapse
|
39
|
Burkard T, Williams RD, Vallejo-Yagüe E, Hügle T, Finckh A, Kyburz D, Burden AM. Prediction of sustained biologic and targeted synthetic DMARD-free remission in rheumatoid arthritis patients. Rheumatol Adv Pract 2021; 5:rkab087. [PMID: 34888435 PMCID: PMC8651222 DOI: 10.1093/rap/rkab087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023] Open
Abstract
Objectives The aim was to develop a prediction model of sustained remission after cessation of biologic or targeted synthetic DMARD (b/tsDMARD) in RA. Methods We conducted an explorative cohort study among b/tsDMARD RA treatment episode courses stopped owing to remission in the Swiss Clinical Quality Management registry (SCQM; 2008-2019). The outcome was sustained b/tsDMARD-free remission of ≥12 months. We applied logistic regression model selection algorithms using stepwise, forward selection, backward selection and penalized regression to identify patient characteristics predictive of sustained b/tsDMARD-free remission. We compared c-statistics corrected for optimism between models. The three models with the highest c-statistics were validated in new SCQM data until 2020 (validation dataset). Results We identified 302 eligible episodes, of which 177 episodes (59%) achieved sustained b/tsDMARD-free remission. Two backward and one forward selection model, with eight, four and seven variables, respectively, obtained the highest c-statistics corrected for optimism of c = 0.72, c = 0.70 and c = 0.69, respectively. In the validation dataset (47 eligible episodes), the models performed with c = 0.99, c = 0.80 and c = 0.74, respectively, and excellent calibration. The best model included the following eight variables (measured at b/tsDMARD stop): RA duration, b/tsDMARD duration, other pain/anti-inflammatory drug use, quality of life (EuroQol), DAS28-ESR score, HAQ score, education, and interactions of RA duration and other pain/anti-inflammatory drug use and of b/tsDMARD duration and HAQ score. Conclusion Our results suggest that models with up to eight unique variables may predict sustained b/tsDMARD-free remission with good efficiency. External validation is warranted.
Collapse
Affiliation(s)
- Theresa Burkard
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Ross D Williams
- Department of Medical Informatics, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Thomas Hügle
- Department of Rheumatology, Lausanne University Hospital, University of Lausanne, Lausanne
| | - Axel Finckh
- Department of Rheumatology, University Hospitals Geneva, Geneva
| | - Diego Kyburz
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - Andrea M Burden
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
O'Neil LJ, Hu P, Liu Q, Islam MM, Spicer V, Rech J, Hueber A, Anaparti V, Smolik I, El-Gabalawy HS, Schett G, Wilkins JA. Proteomic Approaches to Defining Remission and the Risk of Relapse in Rheumatoid Arthritis. Front Immunol 2021; 12:729681. [PMID: 34867950 PMCID: PMC8636686 DOI: 10.3389/fimmu.2021.729681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022] Open
Abstract
Objectives Patients with Rheumatoid Arthritis (RA) are increasingly achieving stable disease remission, yet the mechanisms that govern ongoing clinical disease and subsequent risk of future flare are not well understood. We sought to identify serum proteomic alterations that dictate clinically important features of stable RA, and couple broad-based proteomics with machine learning to predict future flare. Methods We studied baseline serum samples from a cohort of stable RA patients (RETRO, n = 130) in clinical remission (DAS28<2.6) and quantified 1307 serum proteins using the SOMAscan platform. Unsupervised hierarchical clustering and supervised classification were applied to identify proteomic-driven clusters and model biomarkers that were associated with future disease flare after 12 months of follow-up and RA medication withdrawal. Network analysis was used to define pathways that were enriched in proteomic datasets. Results We defined 4 proteomic clusters, with one cluster (Cluster 4) displaying a lower mean DAS28 score (p = 0.03), with DAS28 associating with humoral immune responses and complement activation. Clustering did not clearly predict future risk of flare, however an XGboost machine learning algorithm classified patients who relapsed with an AUC (area under the receiver operating characteristic curve) of 0.80 using only baseline serum proteomics. Conclusions The serum proteome provides a rich dataset to understand stable RA and its clinical heterogeneity. Combining proteomics and machine learning may enable prediction of future RA disease flare in patients with RA who aim to withdrawal therapy.
Collapse
Affiliation(s)
- Liam J O'Neil
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Department of Computer Science, University of Manitoba, Winnipeg, MB, Canada
| | - Qian Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Department of Computer Science, University of Manitoba, Winnipeg, MB, Canada
| | - Md Mohaiminul Islam
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.,Department of Computer Science, University of Manitoba, Winnipeg, MB, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Juergen Rech
- Department of Medicine, Friedrich-Alexander University Erlangen-Nuernberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Axel Hueber
- Department of Medicine, Friedrich-Alexander University Erlangen-Nuernberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - Vidyanand Anaparti
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Irene Smolik
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Hani S El-Gabalawy
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| | - Georg Schett
- Department of Medicine, Friedrich-Alexander University Erlangen-Nuernberg and Universitaetsklinikum Erlangen, Erlangen, Germany
| | - John A Wilkins
- Section of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, Winnipeg, MB, Canada
| |
Collapse
|
41
|
Uhrenholt L, Christensen R, Dinesen WKH, Liboriussen CH, Andersen SS, Dreyer L, Schlemmer A, Hauge EM, Skrubbeltrang C, Taylor PC, Kristensen S. Risk of flare after tapering or withdrawal of b-/tsDMARDs in patients with RA or axSpA: A systematic review and meta-analysis. Rheumatology (Oxford) 2021; 61:3107-3122. [PMID: 34864896 DOI: 10.1093/rheumatology/keab902] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE To evaluate flare risk when tapering or withdrawing biological or targeted synthetic disease-modifying antirheumatic drugs (b-/tsDMARDs) compared to continuation in patients with inflammatory arthritis (IA) in sustained remission or low disease activity. METHODS Articles were identified in Cochrane Library, PubMed, EMBASE and Web of Science. Eligible trials were randomised, controlled trials comparing tapering and/or withdrawal of b- and/or tsDMARDs with standard dose in IA. Random-effects meta-analysis was performed with risk ratio (RR), or Peto's Odds Ratio (POR) for sparse events, and 95% confidence intervals (95%CI). RESULTS The meta-analysis comprised 22 trials: 11 assessed tapering and 7 addressed withdrawal (4 assessed both). Only trials with a rheumatoid arthritis (RA) or axial spondyloarthritis (axSpA) population were identified. An increased flare risk was demonstrated when b-/tsDMARD tapering was compared to continuation, RR = 1.45 (95%CI: 1.19 to 1.77, I2 = 42.5%), and potentially increased for persistent flare, POR = 1.56 (95%CI: 0.97 to 2.52, I2 = 0%). Comparing tumour necrosis factor inhibitor (TNFi) withdrawal to continuation, a highly increased flare risk (RR = 2.28, 95%CI: 1.78 to 2.93, I2 = 78%) and increased odds of persistent flare (POR = 3.41, 95%CI: 1.91 to 6.09, I2 = 49%) was observed. No clear difference in flare risk between RA or axSpA was observed. CONCLUSION A high risk for flare and persistent flare was demonstrated for TNFi withdrawal whereas an increased risk for flare but not for persistent flare was observed for b-/tsDMARD tapering. Thus, tapering seems to be the more favourable approach. REGISTRATION PROSPERO (CRD42019136905).
Collapse
Affiliation(s)
- Line Uhrenholt
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Section for Biostatistics and Evidence-Based Research, Bispebjerg and Frederiksberg Hospital, The Parker Institute, Copenhagen, Denmark
| | - Robin Christensen
- Section for Biostatistics and Evidence-Based Research, Bispebjerg and Frederiksberg Hospital, The Parker Institute, Copenhagen, Denmark.,Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | | | | | - Stine S Andersen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Lene Dreyer
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Annette Schlemmer
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Rheumatology, Randers Regional Hospital, Randers, Denmark
| | - Ellen-Margrethe Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Conni Skrubbeltrang
- Department of Medical Library, Aalborg University Hospital, Aalborg, Denmark
| | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Salome Kristensen
- Department of Rheumatology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
42
|
Tu AB, Lewis JS. Biomaterial-based immunotherapeutic strategies for rheumatoid arthritis. Drug Deliv Transl Res 2021; 11:2371-2393. [PMID: 34414564 PMCID: PMC8376117 DOI: 10.1007/s13346-021-01038-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 02/08/2023]
Abstract
Rheumatoid arthritis (RA) is an extremely painful autoimmune disease characterized by chronic joint inflammation leading to the erosion of adjacent cartilage and bone. Rheumatoid arthritis pathology is primarily driven by inappropriate infiltration and activation of immune cells within the synovium of the joint. There is no cure for RA. As such, manifestation of symptoms entails lifelong management via various therapies that aim to generally dampen the immune system or impede the function of immune mediators. However, these treatment strategies lead to adverse effects such as toxicity, general immunosuppression, and increased risk of infection. In pursuit of safer and more efficacious therapies, many emerging biomaterial-based strategies are being developed to improve payload delivery, specific targeting, and dose efficacy, and to mitigate adverse reactions and toxicity. In this review, we highlight biomaterial-based approaches that are currently under investigation to circumvent the limitations of conventional RA treatments.
Collapse
Affiliation(s)
- Allen B Tu
- Department of Biomedical Engineering, University of California, 1 Shields Ave, Davis , CA, 95616, USA
| | - Jamal S Lewis
- Department of Biomedical Engineering, University of California, 1 Shields Ave, Davis , CA, 95616, USA.
| |
Collapse
|
43
|
Tascilar K, Hagen M, Kleyer A, Simon D, Reiser M, Hueber AJ, Manger B, Englbrecht M, Finzel S, Tony HP, Schuch F, Kleinert S, Wendler J, Ronneberger M, Figueiredo CP, Cobra JF, Feuchtenberger M, Fleck M, Manger K, Ochs W, Schmitt-Haendle M, Lorenz HM, Nuesslein H, Alten R, Kruger K, Henes J, Schett G, Rech J. Treatment tapering and stopping in patients with rheumatoid arthritis in stable remission (RETRO): a multicentre, randomised, controlled, open-label, phase 3 trial. THE LANCET. RHEUMATOLOGY 2021; 3:e767-e777. [PMID: 38297524 DOI: 10.1016/s2665-9913(21)00220-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 02/02/2024]
Abstract
BACKGROUND Owing to increasing remission rates, the management of patients with rheumatoid arthritis in sustained remission is of growing interest. The Rheumatoid Arthritis in Ongoing Remission (RETRO) study investigated tapering and withdrawal of disease-modifying antirheumatic drugs (DMARDs) in patients with rheumatoid arthritis in stable remission to test whether remission could be retained without the need to take DMARD therapy despite an absence of symptoms. METHODS RETRO was an investigator-initiated, multicentre, prospective, randomised, controlled, open-label, parallel-group phase 3 trial in patients aged at least 18 years with rheumatoid arthritis for at least 12 months before randomisation who were in sustained Disease Activity Score using 28 joints with erythrocyte sedimentation rate (ESR) remission (score <2·6 units). Eligible patients were recruited consecutively from 14 German hospitals or rheumatology practices and randomly assigned (1:1:1) without stratification and regardless of baseline treatment, using a sequence that was computer-generated by the study statistician, to continue 100% dose DMARD (continue group), taper to 50% dose DMARD (taper group), or 50% dose DMARD for 6 months before stopping DMARDs (stop group). Neither patients nor investigators were masked to the treatment assignment. Patients were assessed every 3 months and screened for disease activity and relapse. The primary endpoint was the proportion of patients in sustained DAS28-ESR remission without relapse at 12 months, analysed using a log-rank test of trend and Cox regression. Analysis by a trained statistician of the primary outcome and safety was done in a modified intention-to-treat population that included participants with non-missing baseline data. This study is completed and closed to new participants and is registered with ClinicalTrials.gov (NCT02779114). FINDINGS Between May 26, 2010, and May 29, 2018, 303 patients were enrolled and allocated to continue (n=100), taper (n=102), or stop DMARDs (n=101). 282 (93%) of 303 patients were analysed (93 [93%] of 100 for continue, 93 [91%] of 102 for taper, and 96 [95%] of 101 for stop). Remission was maintained at 12 months by 81·2% (95% CI 73·3-90·0) in the continue group, 58·6% (49·2-70·0) in the taper group, and 43·3% (34·6-55·5) in the stop group (p=0·0005 with log-rank test for trend). Hazard ratios for relapse were 3·02 (1·69-5·40; p=0.0003) for the taper group and 4·34 (2·48-7·60; p<0.0001)) for the stop group, in comparison with the continue group. The majority of patients who relapsed regained remission after reintroduction of 100% dose DMARDs. Serious adverse events occurred in ten of 93 (11%) patients in the continue group, seven of 93 (8%) patients in taper group, and 13 of 96 (14%) patients in the stop group. None were considered to be related to the intervention. The most frequent type of serious adverse event was injuries or procedural complications (n=9). INTERPRETATION Reducing antirheumatic drugs in patients with rheumatoid arthritis in stable remission is feasible, with maintenance of remission occurring in about half of the patients. Because relapse rates were significantly higher in patients who tapered or stopped antirheumatic drugs than in patients who continued with a 100% dose, such approaches will require tight monitoring of disease activity. However, remission was regained after reintroduction of antirheumatic treatments in most of those who relapsed in this study. These results might help to prevent overtreatment in a substantial number of patients with rheumatoid arthritis. FUNDING None.
Collapse
Affiliation(s)
- Koray Tascilar
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - Melanie Hagen
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - David Simon
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - Michaela Reiser
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - Axel J Hueber
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany; Rheumatology Section, Sozialstiftung Bamberg, Bamberg, Germany
| | - Bernhard Manger
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - Matthias Englbrecht
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, University of Freiburg, Freiburg, Germany
| | - Hans-Peter Tony
- Department of Internal Medicine 2, University of Wuerzburg, Wuerzburg, Germany
| | | | - Stefan Kleinert
- Department of Internal Medicine 2, University of Wuerzburg, Wuerzburg, Germany; Rheumatology Practice, Erlangen, Germany
| | | | | | - Camille P Figueiredo
- Division of Rheumatology, Faculty of Medicine, Sao Paulo University, Sao Paulo, Brazil
| | | | - Martin Feuchtenberger
- Rheumatology Practice and Department of Internal Medicine 2, Clinic Burghausen, Burghausen, Germany
| | - Martin Fleck
- Department of Internal Medicine I, University of Regensburg, Regensburg, Germany; Asklepios Medical Center Bad Abbach, Bad Abbach, Germany
| | | | | | | | - Hanns-Martin Lorenz
- Department of Internal Medicine V, Division of Rheumatology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | - Joerg Henes
- Department of Internal Medicine 2, University of Tubingen, Tubingen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany.
| | - Juergen Rech
- Department of Internal Medicine 3 and Deutsches Zentrum für Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| |
Collapse
|
44
|
A novel multi-biomarker combination predicting relapse from long-term remission after discontinuation of biological drugs in rheumatoid arthritis. Sci Rep 2021; 11:20771. [PMID: 34675298 PMCID: PMC8531387 DOI: 10.1038/s41598-021-00357-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Biological disease modifying anti-rheumatic drugs (bDMARDs) show dramatic treatment efficacy in rheumatoid arthritis (RA). Long-term use of bDMARDs, however, has disadvantages such as high costs and infection risk. Therefore, a methodology is needed to predict any future RA relapse. Herein, we report a novel multi-biomarker combination which predicts relapse after bDMARDs-withdrawal in patients in remission. Forty patients with RA in remission for more than 12 months were enrolled. bDMARDs were withdrawn and they were followed monthly for the next 24 months. Fourteen patients (35%) of 40 in the cohort remained in remission at 24 months, whereas 26 (65%) relapsed at various time-points. Serum samples obtained longitudinally from patients in remission were assessed for the relapse-prediction biomarkers and index from 73 cytokines by the exploratory multivariate ROC analysis. The relapse-prediction index calculated from the 5 cytokines, IL-34, CCL1, IL-1β, IL-2 and IL-19, strongly discriminated between patients who relapsed and those who stayed in remission. These findings could contribute to clinical decision-making as to the timing of when to discontinue bDMARDs in RA treatment.
Collapse
|
45
|
Sokolova MV, Hagen M, Bang H, Schett G, Rech J, Steffen U. IgA anti-citrullinated protein antibodies (IgA ACPA) are associated with flares during DMARD tapering in rheumatoid arthritis. Rheumatology (Oxford) 2021; 61:2124-2131. [PMID: 34508547 DOI: 10.1093/rheumatology/keab585] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/09/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES A substantial proportion of rheumatoid arthritis (RA) patients flare upon withdrawal of disease modifying anti-rheumatic drugs (DMARDs), thus the definition of prognostic markers is crucial. Anti-citrullinated protein antibody (ACPA)-positivity has been identified as a risk factor for flare. However, only the role of IgG is established in this context, while the role of IgA ACPA is poorly defined. We thus aimed to investigate the role of IgA ACPA in flare of RA. METHODS Serum levels of IgA1 and IgA2 ACPA at baseline and after 12 months were measured in 108 patients from the randomized controlled RETRO study. RA patients in stable remission for at least 6 months at study recruitment were assigned to either one of the DMARD tapering arms or to continuation of DMARDs. RESULTS In patients remaining in remission but not in the ones who flared, IgA2 ACPA levels and proportion of IgA2 in ACPA (IgA2%ACPA) significantly declined (median of 17.5%; p< 0.0001). This seemed to be independent of the treatment choice, as there was no difference in IgA2 ACPA dynamics between the study arms. IgA2% ACPA was associated with disease activity (DAS28) at flare (r = 0.36; p= 0.046). IgA and IgG ACPA showed a tendency towards independent contribution to the risk of flare with the highest risk if a patient had both antibody classes. CONCLUSION In this study, IgA ACPA was identified as a risk factor for flare in combination with IgG ACPA. IgA2 ACPA levels were associated with flare severity and declined in patients in stable remission.
Collapse
Affiliation(s)
- Maria V Sokolova
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Melanie Hagen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Juergen Rech
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ulrike Steffen
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | |
Collapse
|
46
|
Maassen JM, van Ouwerkerk L, Allaart CF. Tapering of disease-modifying antirheumatic drugs: an overview for daily practice. THE LANCET. RHEUMATOLOGY 2021; 3:e659-e670. [PMID: 38287612 DOI: 10.1016/s2665-9913(21)00224-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/25/2021] [Accepted: 07/08/2021] [Indexed: 01/31/2024]
Abstract
In this Review, we discuss the possibility of drug tapering in patients with rheumatoid arthritis in remission or low disease activity, for glucocorticoids and disease-modifying antirheumatic drugs. We review international guidelines and recommendations, as well as remaining uncertainties, and provide an overview of the current literature. Three strategies of tapering are discussed: (1) tapering by discontinuation of one of the drugs in combination therapy regimens, (2) tapering by reducing the dose of one of the drugs in combination therapy regimens, and (3) tapering by dose reduction of monotherapy with disease-modifying antirheumatic drugs. We discuss the outcomes and robustness of evidence of trials and observational cohorts, and we give a trajectory for further research and drug tapering in daily practice.
Collapse
Affiliation(s)
| | - Lotte van Ouwerkerk
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | | |
Collapse
|
47
|
Vorsicht beim Ausschleichen aus der RA-Therapie. Dtsch Med Wochenschr 2021. [DOI: 10.1055/a-1519-0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
48
|
Rayner F, Anderson AE, Baker KF, Buckley CD, Dyke B, Fenton S, Filer A, Goodyear CS, Hilkens CMU, Hiu S, Kerrigan S, Kurowska-Stolarska M, Matthews F, McInnes I, Ng WF, Pratt AG, Prichard J, Raza K, Siebert S, Stocken D, Teare MD, Young S, Isaacs JD. BIOlogical Factors that Limit sustAined Remission in rhEumatoid arthritis (the BIO-FLARE study): protocol for a non-randomised longitudinal cohort study. BMC Rheumatol 2021; 5:22. [PMID: 34275488 PMCID: PMC8286860 DOI: 10.1186/s41927-021-00194-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/09/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Our knowledge of immune-mediated inflammatory disease (IMID) aetiology and pathogenesis has improved greatly over recent years, however, very little is known of the factors that trigger disease relapses (flares), converting diseases from inactive to active states. Focussing on rheumatoid arthritis (RA), the challenge that we will address is why IMIDs remit and relapse. Extrapolating from pathogenetic factors involved in disease initiation, new episodes of inflammation could be triggered by recurrent systemic immune dysregulation or locally by factors within the joint, either of which could be endorsed by overarching epigenetic factors or changes in systemic or localised metabolism. METHODS The BIO-FLARE study is a non-randomised longitudinal cohort study that aims to enrol 150 patients with RA in remission on a stable dose of non-biologic disease-modifying anti-rheumatic drugs (DMARDs), who consent to discontinue treatment. Participants stop their DMARDs at time 0 and are offered an optional ultrasound-guided synovial biopsy. They are studied intensively, with blood sampling and clinical evaluation at weeks 0, 2, 5, 8, 12 and 24. It is anticipated that 50% of participants will have a disease flare, whilst 50% remain in drug-free remission for the study duration (24 weeks). Flaring participants undergo an ultrasound-guided synovial biopsy before reinstatement of previous treatment. Blood samples will be used to investigate immune cell subsets, their activation status and their cytokine profile, autoantibody profiles and epigenetic profiles. Synovial biopsies will be examined to profile cell lineages and subtypes present at flare. Blood, urine and synovium will be examined to determine metabolic profiles. Taking into account all generated data, multivariate statistical techniques will be employed to develop a model to predict impending flare in RA, highlighting therapeutic pathways and informative biomarkers. Despite initial recruitment to time and target, the SARS-CoV-2 pandemic has impacted significantly, and a decision was taken to close recruitment at 118 participants with complete data. DISCUSSION This study aims to investigate the pathogenesis of flare in rheumatoid arthritis, which is a significant knowledge gap in our understanding, addressing a major unmet patient need. TRIAL REGISTRATION The study was retrospectively registered on 27/06/2019 in the ISRCTN registry 16371380 .
Collapse
Affiliation(s)
- Fiona Rayner
- Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Amy E Anderson
- Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Kenneth F Baker
- Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Christopher D Buckley
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Bernard Dyke
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Sally Fenton
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Andrew Filer
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Catharien M U Hilkens
- Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Shaun Hiu
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sean Kerrigan
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | | - Fiona Matthews
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Wan-Fai Ng
- Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Jonathan Prichard
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Karim Raza
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Department of Rheumatology, Sandwell and West Birmingham NHS Trust, Birmingham, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Deborah Stocken
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - M Dawn Teare
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Stephen Young
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
49
|
Jung SY, Koh JH, Kim KJ, Park YW, Yang HI, Choi SJ, Lee J, Choi CB, Kim WU. Switching from TNFα inhibitor to tacrolimus as maintenance therapy in rheumatoid arthritis after achieving low disease activity with TNFα inhibitors and methotrexate: 24-week result from a non-randomized, prospective, active-controlled trial. Arthritis Res Ther 2021; 23:182. [PMID: 34233727 PMCID: PMC8265052 DOI: 10.1186/s13075-021-02566-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022] Open
Abstract
Background Tapering or stopping biological disease-modifying anti-rheumatic drugs has been proposed for patients with rheumatoid arthritis (RA) in remission, but it frequently results in high rates of recurrence. This study evaluates the efficacy and safety of tacrolimus (TAC) as maintenance therapy in patients with established RA in remission after receiving combination therapy with tumor necrosis factor inhibitor (TNFi) and methotrexate (MTX). Methods This 24-week, prospective, open-label trial included patients who received TNFi and MTX at stable doses for ≥24 weeks and had low disease activity (LDA), measured by Disease Activity Score-28 for ≥12 weeks. Patients selected one of two arms: maintenance (TNFi plus MTX) or switched (TAC plus MTX). The primary outcome was the difference in the proportion of patients maintaining LDA at week 24, which was assessed using a logistic regression model. Adverse events were monitored throughout the study period. Results In efficacy analysis, 80 and 34 patients were included in the maintenance and switched arms, respectively. At week 24, LDA was maintained in 99% and 91% of patients in the maintenance and switched arms, respectively (odds ratio, 0.14; 95% confidence interval, 0.01–1.59). Drug-related adverse effects tended to be more common in the switched arm than in the maintenance arm (20.9% versus 7.1%, respectively) but were well-tolerated. Conclusion This controlled study tested a novel treatment strategy of switching from TNFi to TAC in RA patients with sustained LDA, and the findings suggested that TNFi can be replaced with TAC in most patients without the patients experiencing flare-ups for at least 24 weeks. Trial registration Korea CDC CRIS, KCT0005868. Registered 4 February 2021—retrospectively registered Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02566-z.
Collapse
Affiliation(s)
- Sang Youn Jung
- Division of Rheumatology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jung Hee Koh
- Division of Rheumatology, Department of Internal Medicine, Bucheon St. Mary's Hospital, the Catholic University of Korea, Seoul, South Korea
| | - Ki-Jo Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent Hospital, the Catholic University of Korea, Seoul, South Korea
| | - Yong-Wook Park
- Division of Rheumatology, Department of Internal Medicine, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Hyung-In Yang
- College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, South Korea
| | - Sung Jae Choi
- Division of Rheumatology, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, South Korea
| | - Jisoo Lee
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, South Korea
| | - Chan-Bum Choi
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St Mary's Hospital, Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, the Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.
| |
Collapse
|
50
|
Terslev L, Ostergaard M. Rheumatoid Arthritis Relapse and Remission - Advancing Our Predictive Capability Using Modern Imaging. J Inflamm Res 2021; 14:2547-2555. [PMID: 34163211 PMCID: PMC8215903 DOI: 10.2147/jir.s284405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/27/2021] [Indexed: 01/18/2023] Open
Abstract
Clinical remission has become an achievable target for the majority of patients with rheumatoid arthritis, but subclinical inflammation as assessed by ultrasound and magnetic resonance imaging (MRI) has been demonstrated to be frequent in patients in clinical remission. Subclinical synovitis has been shown to be linked to both subsequent structural damage progression and a risk of flare, demonstrating that subclinical synovitis represents incomplete suppression of inflammation and questions whether it is appropriate only to use clinical composite scores as treatment target in clinical practice. Maintaining a state of remission has proven important as sustained clinical remission impacts long-term outcome regarding joint damage progression, physical function and quality of life. Treating subclinical inflammation has been attempted and has led to more frequent strict clinical remission and better physical function, but also to more adverse events. Thus, an overall benefit of incorporating imaging goals in treat-to-target strategies has not been documented. However, in patients in clinical remission on biological disease-modifying anti-rheumatic drugs, both ultrasound and MRI may aid in the clinical decision regarding whether drug tapering or even discontinuation should be attempted.
Collapse
Affiliation(s)
- Lene Terslev
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Ostergaard
- Copenhagen Center for Arthritis Research, Center for Rheumatology and Spine Diseases, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|