51
|
Smolen JS, van Vollenhoven RF, Florentinus S, Chen S, Suboticki JL, Kavanaugh A. Predictors of disease activity and structural progression after treatment with adalimumab plus methotrexate or continued methotrexate monotherapy in patients with early rheumatoid arthritis and suboptimal response to methotrexate. Ann Rheum Dis 2018; 77:1566-1572. [PMID: 30076156 PMCID: PMC6225797 DOI: 10.1136/annrheumdis-2018-213502] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/03/2018] [Accepted: 07/05/2018] [Indexed: 01/21/2023]
Abstract
Objectives Methotrexate is considered to be first-line therapy for rheumatoid arthritis (RA). However, a substantial proportion of treated patients do not achieve the desired goals of therapy. This analysis aimed to identify predictors of insufficient response to methotrexate in patients with early RA. Methods The Optimal Protocol for Treatment Initiation with Methotrexate and Adalimumab (OPTIMA) and PREMIER studies in patients with RA for <1 and <3 years, respectively, examined the efficacy of methotrexate and adalimumab in methotrexate-naive patients. This post hoc analysis included patients for whom initial methotrexate monotherapy was not successful after 6 months. Candidate predictors of insufficient response and clinically relevant radiographic progression (CRRP) included demographics, baseline disease characteristics and time-averaged disease variables over a 12-week interval. In OPTIMA, adalimumab was added to therapy after insufficient treatment response; in PREMIER, initial methotrexate therapy was continued; clinical, functional and radiologic outcomes were assessed after 1 year. Results Baseline 28-joint Disease Activity Score based on C-reactive protein (DAS28(CRP)) and time-averaged DAS28(CRP) over 4, 8 and 12 weeks were the strongest predictors of insufficient response to methotrexate and CRRP. Addition of adalimumab to methotrexate therapy was associated with better clinical, functional and radiographic outcomes after 1 year compared with continuing on methotrexate monotherapy. Conclusions In patients with early RA, baseline disease characteristics and early disease activity can predict response to methotrexate treatment and radiographic progression at 6 months. The addition of adalimumab at 6 months after methotrexate failure is associated with improved outcomes. These results support treatment-to-target strategies and timely adaptation of therapy in patients with early RA. Trial registration number NCT00420927, NCT00195663; Post-results.
Collapse
Affiliation(s)
- Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Ronald F van Vollenhoven
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Center (ARC), Amsterdam, The Netherlands
| | | | - Su Chen
- Data and Statistical Sciences, AbbVie Inc., North Chicago, Illinois, USA
| | | | - Arthur Kavanaugh
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
52
|
Alemao E, Litman HJ, Connolly SE, Kelly S, Hua W, Rosenblatt L, Rebello S, Kremer JM, Harrold LR. Do Poor Prognostic Factors in Rheumatoid Arthritis Affect Treatment Choices and Outcomes? Analysis of a US Rheumatoid Arthritis Registry. J Rheumatol 2018; 45:1353-1360. [PMID: 29961696 DOI: 10.3899/jrheum.171050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To characterize patients with rheumatoid arthritis (RA) by number of poor prognostic factors (PPF: functional limitation, extraarticular disease, seropositivity, erosions) and evaluate treatment acceleration, clinical outcomes, and work status over 12 months by number of PPF. METHODS Using the Corrona RA registry (January 2005-December 2015), biologic-naive patients with diagnosed RA having 12-month (± 3 mos) followup were identified and categorized by PPF (0-1, 2, ≥ 3). Changes in medication, Clinical Disease Activity Index (CDAI), and work status (baseline-12 mos) were evaluated using linear and logistic regression models. RESULTS There were 3458 patients who met the selection criteria: 1489 (43.1%), 1214 (35.1%), and 755 (21.8%) had 0-1, 2, or ≥ 3 PPF, respectively. At baseline, patients with ≥ 3 PPF were older, and had longer RA duration and higher CDAI versus those with 0-1 PPF. In 0-1, 2, and ≥ 3 PPF groups, respectively, 20.9%, 23.2%, and 26.5% of patients received ≥ 1 biologic (p = 0.011). Biologic/targeted synthetic disease-modifying antirheumatic drug (tsDMARD) use was similar in patients with/without PPF (p = 0.57). After adjusting for baseline CDAI, mean (standard error) change in CDAI was -4.95 (0.24), -4.53 (0.27), and -2.52 (0.34) for 0-1, 2, and ≥ 3 PPF groups, respectively. More patients were working at baseline but not at 12-month followup in 2 (13.9%) and ≥ 3 (12.5%) versus 0-1 (7.3%) PPF group. CONCLUSION Despite high disease activity and worse clinical outcomes, number of PPF did not significantly predict biologic/tsDMARD use. This may warrant reconsideration of the importance of PPF in treat-to-target approaches.
Collapse
Affiliation(s)
- Evo Alemao
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA. .,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School.
| | - Heather J Litman
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| | - Sean E Connolly
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| | - Sheila Kelly
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| | - Winnie Hua
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| | - Lisa Rosenblatt
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| | - Sabrina Rebello
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| | - Joel M Kremer
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| | - Leslie R Harrold
- From Bristol-Myers Squibb, Princeton, New Jersey; Corrona LLC, Southborough, Massachusetts; Albany Medical College and The Center for Rheumatology, Albany, New York; University of Massachusetts Medical School, Worcester, Massachusetts, USA.,E. Alemao, MS, RPh, Bristol-Myers Squibb; H.J. Litman, PhD, Corrona LLC; S.E. Connolly, PhD, Bristol-Myers Squibb; S. Kelly, MD, Bristol-Myers Squibb; W. Hua, MS, Corrona LLC; L. Rosenblatt, MD, MPH, Bristol-Myers Squibb; S. Rebello, MPH, Corrona LLC; J.M. Kremer, MD, Corrona LLC, and Albany Medical College and The Center for Rheumatology; L.R. Harrold, MD, MPH, Corrona LLC, and the University of Massachusetts Medical School
| |
Collapse
|
53
|
Mueller RB, Kaegi T, Haile SR, Schulze-Koops H, Schiff M, von Kempis J. Clinical and radiographic course of early undifferentiated arthritis under treatment is not dependent on the number of joints with erosions at diagnosis: results from the Swiss prospective observational cohort. RMD Open 2018; 4:e000673. [PMID: 29955385 PMCID: PMC6018869 DOI: 10.1136/rmdopen-2018-000673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 11/08/2022] Open
Abstract
Objective To analyse whether early arthritis patients who do not fulfil the American College of Rheumatology/European League Against Rheumatism (ACR/EULAR) 2010 classification criteria for rheumatoid arthritis (RA) have a different course of the disease dependent on whether they can or cannot be classified as RA because of radiographic disease (EULAR task force) at diagnosis. Methods For this observational study within the Swiss RA cohort SCQM, we included patients with early undifferentiated arthritis (disease duration ≤1 year), who had not received any previous disease-modifying antirheumatic drugs (DMARDs). 2010 ACR/EULAR criteria negative patients were separated into two groups (radiographic vs non-radiographic arthritis) depending on whether or not they had radiographic changes defined as erosive disease by a EULAR task force (≥3 joints with erosions). The primary outcome measure was the radiographic progression detected employing the Ratingen erosion score. Health Assessment Questionnaire (HAQ) and DAS-28 were used as secondary outcome measures. The average observation period was 4 years. Results A total of 592 patients were analysed. 240 were not classifiable as RA by application of the 2010 ACR/EULAR criteria at baseline. In 57 patients, radiographs at the first visit were not available. 133 patients had radiographic arthritis and 50 non-radiographic arthritis. Treatment was initiated in all patients with DMARDs, mostly methotrexate. No differences in DAS-28 and HAQ scores were found during follow-up. The average erosion scores were higher among patients with initially radiographic arthritis throughout the study. The progression of erosion scores over time, however, was higher in patients with initially non-radiographic arthritis with less subsequent radiological progression (3.3 erosions/year vs 0.4, respectively, p<0.0001). Conclusions The clinical and radiographic course of early undifferentiated arthritis under treatment was not dependent on the presence of erosions in three or more joints (ie, the definition of radiographic disease by the EULAR task force) at diagnosis in our cohort.
Collapse
Affiliation(s)
- Ruediger B Mueller
- Division of Rheumatology and Immunology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Division of Rheumatology and Clinical Immunology, Department of Internal Medicine IV, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Toni Kaegi
- Division of Rheumatology and Immunology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Sarah R Haile
- Clinical Trials Unit, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Division of Biostatistics, Institute for Social and Preventive Medicine, University of Zürich, Zürich, Switzerland
| | - Hendrik Schulze-Koops
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine IV, Ludwig-Maximilian-University Munich, Munich, Germany
| | | | - Johannes von Kempis
- Division of Rheumatology and Immunology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
54
|
Louveau B, De Rycke Y, Lafourcade A, Saraux A, Guillemin F, Tubach F, Fautrel B, Hajage D. Effect of cumulative exposure to corticosteroid and DMARD on radiographic progression in rheumatoid arthritis: results from the ESPOIR cohort. Rheumatology (Oxford) 2018; 57:1563-1573. [DOI: 10.1093/rheumatology/key122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Indexed: 11/14/2022] Open
Affiliation(s)
- Baptiste Louveau
- APHP, Centre de Pharmacoépidémiologie (Cephepi), Département de Biostatistique Santé Publique et Information Médicale, CIC 1421, Hôpital Pitié Salpêtrière, Paris, France
- INSERM, UMR 1123, ECEVE, CIC 1421, Paris, France
| | - Yann De Rycke
- APHP, Centre de Pharmacoépidémiologie (Cephepi), Département de Biostatistique Santé Publique et Information Médicale, CIC 1421, Hôpital Pitié Salpêtrière, Paris, France
- INSERM, UMR 1123, ECEVE, CIC 1421, Paris, France
| | - Alexandre Lafourcade
- APHP, Centre de Pharmacoépidémiologie (Cephepi), Département de Biostatistique Santé Publique et Information Médicale, CIC 1421, Hôpital Pitié Salpêtrière, Paris, France
| | - Alain Saraux
- Département de Rhumatologie, CHU Brest, INSERM 1227, UBO, Brest, France
| | - Francis Guillemin
- Université de Lorraine, Université Paris Descartes, EA 4360 APEMAC, Nancy, France
| | - Florence Tubach
- APHP, Centre de Pharmacoépidémiologie (Cephepi), Département de Biostatistique Santé Publique et Information Médicale, CIC 1421, Hôpital Pitié Salpêtrière, Paris, France
- INSERM, UMR 1123, ECEVE, CIC 1421, Paris, France
- Sorbonne Université, Faculté de médecine Sorbonne Université, Paris, France
| | - Bruno Fautrel
- Sorbonne Université, GRC-08 (EEMOIS), Paris, France
- Département de Rhumatologie, APHP, Hôpital Pitié Salpêtrière, Paris, France
| | - David Hajage
- APHP, Centre de Pharmacoépidémiologie (Cephepi), Département de Biostatistique Santé Publique et Information Médicale, CIC 1421, Hôpital Pitié Salpêtrière, Paris, France
- INSERM, UMR 1123, ECEVE, CIC 1421, Paris, France
- Sorbonne Université, Faculté de médecine Sorbonne Université, Paris, France
| |
Collapse
|
55
|
Effect of treat-to-target strategies on bone erosion progression in early rheumatoid arthritis: An HR-pQCT study. Semin Arthritis Rheum 2018; 48:374-383. [PMID: 29858113 DOI: 10.1016/j.semarthrit.2018.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 03/12/2018] [Accepted: 05/01/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVES To investigate the efficacy of two tight-control treatment strategies aimed at simplified disease activity score [SDAI] remission (SDAI ≤ 3.3) compared to DAS28 remission (DAS28 < 2.6) on progression of bone erosions in early rheumatoid arthritis (ERA) patients using high-resolution peripheral quantitative computed tomography (HR-pQCT). METHODS This was an open-label study in which 80 early RA patients were randomized to receive 1-year of tight-control treatment. Group 1 (n = 37) aimed at SDAI ≤ 3.3 and group 2 (n = 43) aimed at DAS28-CRP < 2.6. The number and size of bone erosions, as well as the bone mineral density (BMD) surrounding bone erosion at the second metacarpophalangeal joint (MCP2), were measured at baseline and 12 months. RESULTS After 12 months, images were analyzed in 63 patients. Changes in clinical parameters, number and size of bone erosions as well as the BMD surrounding bone erosion between the two treatment groups were similar. Therefore, a post-hoc analysis including all 63 patients was performed to elucidate the independent predictors of erosion progression and repair. Multivariate analysis revealed that not achieving sustained SDAI remission at month 6, 9 and 12 (p = 0.034) and rheumatoid factor >16U (p = 0.021) were independent predictors associated with an increase in erosion volume. Logistic regression analysis showed that achieving sustained SDAI remission (p = 0.043) was associated with partial erosion repair. CONCLUSIONS Although more stringent treatment target did not notably affect clinical treatment outcome and erosion progression at 1 year, achieving sustained SDAI remission was found to be associated with partial erosion repair.
Collapse
|
56
|
Rydell E, Forslind K, Nilsson JÅ, Jacobsson LTH, Turesson C. Smoking, body mass index, disease activity, and the risk of rapid radiographic progression in patients with early rheumatoid arthritis. Arthritis Res Ther 2018; 20:82. [PMID: 29720260 PMCID: PMC5932864 DOI: 10.1186/s13075-018-1575-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/22/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Identification of risk factors for rapid joint destruction in early rheumatoid arthritis (RA) can be helpful for optimizing treatment, and improving our understanding of destructive arthritis and its mechanisms. The objective of this study was to investigate the relationship between early RA patient characteristics and subsequent rapid radiographic progression (RRP). METHODS An inception cohort of patients with early RA (symptom duration < 12 months), recruited during 1995-2005 from a defined area (Malmö, Sweden), was investigated. Radiographs of the hands and feet were scored in chronological order according to the modified Sharp-van der Heijde score (SHS), by a trained reader. RRP was defined as an increase of ≥ 5 points in SHS per year. RESULTS Two hundred and thirty-three patients were included. Radiographs were available from 216 patients at baseline, 206 patients at 1 year, and 171 patients at 5 years. Thirty-six patients (22%) had RRP up to 5 years. In logistic regression models, rheumatoid factor (RF) and anti-cyclic citrullinated peptides (anti-CCP), and increased erythrocyte sedimentation rate (ESR) or C-reactive protein (CRP) at baseline, predicted RRP over 5 years. Patients identified as overweight or obese had a significantly reduced risk of RRP up to 5 years (odds ratio (OR) 0.26; 95% confidence interval (CI) 0.11-0.63; adjusted for RF, baseline erosions, and ESR). Similar point estimates were obtained when stratifying for antibody status, and in models adjusted for smoking. A history of ever smoking was associated with a significantly increased risk of RRP up to 5 years, independent of body mass index (BMI) (OR 3.17; 95% CI 1.22-8.28; adjusted for BMI). At the 1-year follow-up, erosive changes, Disease Activity Score of 28 joints, Health Assessment Questionnaire, swollen joint count, and patient's global assessment of disease activity and pain were also significantly associated with RRP up to 5 years. CONCLUSIONS A history of smoking, presence of RF and/or anti-CCP and early erosions, high initial disease activity and active disease at 1 year, all increase the risk of RRP. Patients with a high BMI may have a reduced risk of severe joint damage. This pattern was not explained by differences in disease activity or antibody status. The results of this study suggest independent effects of smoking and BMI on the risk of RRP.
Collapse
Affiliation(s)
- Emil Rydell
- Rheumatology, Department of Clinical Sciences, Malmö, Lund University, Jan Waldenströms gata 35, SE-202 13, Malmö, Sweden. .,Department of Rheumatology, Skåne University Hospital, Inga Marie Nilssons gata 32, SE-214 28, Malmö, Sweden.
| | - Kristina Forslind
- Department of Research and Education, Helsingborg Hospital, Charlotte Yhlens gata 10, SE-251 87, Helsingborg, Sweden.,Rheumatology, Department of Clinical Sciences, Helsingborg, Lund University, Svartbrödragränden 3-5, SE-251 87, Helsingborg, Sweden
| | - Jan-Åke Nilsson
- Rheumatology, Department of Clinical Sciences, Malmö, Lund University, Jan Waldenströms gata 35, SE-202 13, Malmö, Sweden.,Department of Rheumatology, Skåne University Hospital, Inga Marie Nilssons gata 32, SE-214 28, Malmö, Sweden
| | - Lennart T H Jacobsson
- Rheumatology, Department of Clinical Sciences, Malmö, Lund University, Jan Waldenströms gata 35, SE-202 13, Malmö, Sweden.,Department of Rheumatology and Inflammation Research, Sahlgrenska Academy at Gothenburg University, Guldhedsgatan 10 A, SE-405 30, Göteborg, Sweden
| | - Carl Turesson
- Rheumatology, Department of Clinical Sciences, Malmö, Lund University, Jan Waldenströms gata 35, SE-202 13, Malmö, Sweden.,Department of Rheumatology, Skåne University Hospital, Inga Marie Nilssons gata 32, SE-214 28, Malmö, Sweden
| |
Collapse
|
57
|
Association of anti-cyclic citrullinated protein antibodies, erosions, and rheumatoid factor with disease activity and work productivity: A patient registry study. Semin Arthritis Rheum 2018; 47:630-638. [DOI: 10.1016/j.semarthrit.2017.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 02/04/2023]
|
58
|
Fedele AL, Petricca L, Tolusso B, Alivernini S, Canestri S, Di Mario C, Bosello SL, Ferraccioli G, Gremese E. Interleukin-6 and IgA-rheumatoid factor are crucial for baseline erosiveness, and anti-citrullinated peptide antibodies for radiographic progression in early rheumatoid arthritis treated according to a treat-to-target strategy. Scand J Rheumatol 2018. [PMID: 29542372 DOI: 10.1080/03009742.2017.1416668] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To define baseline clinical and immunological characteristics [anti-citrullinated peptide antibodies (ACPAs), immunoglobulin M (IgM)- and IgA-rheumatoid factor (RF), and interleukin-6 (IL-6) levels] involved in determining baseline erosiveness, outcome, and radiographic progression among seropositive and seronegative early rheumatoid arthritis (ERA) patients. METHOD The 408 ERA patients enrolled in the study were monitored every 3 months according to the treat-to-target strategy. At baseline and after 12 months, hand and foot radiographs were evaluated using the Sharp/van der Heijde erosion score. RESULTS At diagnosis, seronegative patients were older and had higher Disease Activity Scores (DASs) than seropositive patients. A higher risk of erosiveness at baseline was conferred by IgA-RF positivity and IL-6 plasma levels ≥7.6 pg/mL, particularly when simultaneously present. In multivariate analysis, disease duration and IL-6 plasma levels ≥7.6 pg/mL arose as independent variables associated with presence of erosions at onset. Radiographic progression at 1 year follow-up, which occurred in 11.1% of ERA patients, was predicted by ACPA positivity, together with higher age at diagnosis. Despite similar percentages of good European League Against Rheumatism response, DAS and Boolean remission being observed over time among seropositive and seronegative patients and between erosive and non-erosive subjects, ERA patients who were erosive at onset, IgA-RF seropositive, and simultaneously having high baseline IL-6 plasma levels (≥7.6 pg/mL) were treated to a greater extent with tumour necrosis factor blockers after 12 months. CONCLUSION IgA-RF positivity and IL-6 plasma levels are crucial for baseline erosiveness, while ACPA positivity represents the strongest risk factor for developing radiographic progression in ERA.
Collapse
Affiliation(s)
- A L Fedele
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - L Petricca
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - B Tolusso
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - S Alivernini
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - S Canestri
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - C Di Mario
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - S L Bosello
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - G Ferraccioli
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| | - E Gremese
- a Division of Rheumatology , Catholic University of the Sacred Heart, Agostino Gemelli University Polyclinic , Rome , Italy
| |
Collapse
|
59
|
Scherer HU, Huizinga TWJ, Krönke G, Schett G, Toes REM. The B cell response to citrullinated antigens in the development of rheumatoid arthritis. Nat Rev Rheumatol 2018; 14:157-169. [DOI: 10.1038/nrrheum.2018.10] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
60
|
|
61
|
Dextromethorphan Exhibits Anti-inflammatory and Immunomodulatory Effects in a Murine Model of Collagen-Induced Arthritis and in Human Rheumatoid Arthritis. Sci Rep 2017; 7:11353. [PMID: 28900117 PMCID: PMC5595833 DOI: 10.1038/s41598-017-11378-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 08/23/2017] [Indexed: 01/04/2023] Open
Abstract
Dextromethorphan (d-3-methoxy-17-methylmorphinan, DXM) is a commonly used antitussive with a favorable safety profile. Previous studies have demonstrated that DXM has anti-inflammatory and immunomodulatory properties; however, the effect of DXM in rheumatoid arthritis (RA) remains unknown. Herein, we found that DXM treatment attenuated arthritis severity and proinflammatory cytokine expression levels, including TNF-α, IL-6, and IL-17A, in paw tissues of CIA mice. DXM treatment also reduced serum TNF-α, IL-6, and IL-17A levels of CIA mice and patients with RA. DXM further decreased the production of anti-CII IgG, IFN-γ, and IL-17A in collagen-reactive CD4+ T cells extracted from the lymph nodes of CIA mice. In vitro incubation of bone marrow–derived dendritic cells with DXM limited CD4+ T-cell proliferation and inflammatory cytokine secretion. In conclusion, our results showed that DXM attenuated arthritis symptoms in CIA mice and significantly reduced proinflammatory cytokines in patients with RA, suggesting that it can be used as an anti-arthritic agent.
Collapse
|
62
|
Knevel R, Huizinga TW, Kurreeman F. Genomic Influences on Susceptibility and Severity of Rheumatoid Arthritis. Rheum Dis Clin North Am 2017; 43:347-361. [DOI: 10.1016/j.rdc.2017.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
63
|
Asai S, Fujibayashi T, Oguchi T, Hanabayashi M, Hayashi M, Matsubara H, Ito T, Yabe Y, Watanabe T, Hirano Y, Kanayama Y, Kaneko A, Kato T, Takagi H, Takahashi N, Funahashi K, Takemoto T, Asai N, Watanabe T, Ishiguro N, Kojima T. Predictors of biologic discontinuation due to insufficient response in patients with rheumatoid arthritis who achieved clinical remission with biologic treatment: A multicenter observational cohort study. Mod Rheumatol 2017; 28:221-226. [DOI: 10.1080/14397595.2017.1332558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shuji Asai
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | | | - Takeshi Oguchi
- Department of Orthopedic Surgery, Anjo Kosei Hospital, Anjo, Aichi, Japan
| | - Masahiro Hanabayashi
- Department of Orthopedic Surgery, Ichinomiya Municipal Hospital, Ichinomiya, Aichi, Japan
| | - Masatoshi Hayashi
- Department of Rheumatology, Nagano Red Cross Hospital, Nagano, Japan
| | | | | | - Yuichiro Yabe
- Department of Rheumatology, Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Tsuyoshi Watanabe
- Department of Orthopedic Surgery, Kariya-Toyota General Hospital, Kariya, Aichi, Japan
| | - Yuji Hirano
- Department of Rheumatology, Toyohashi Municipal Hospital, Toyohashi, Aichi, Japan
| | - Yasuhide Kanayama
- Department of Rheumatology, Toyota Kosei Hospital, Toyota, Aichi, Japan
| | - Atsushi Kaneko
- Department of Orthopedic Surgery and Rheumatology, Nagoya Medical Center, Nagoya, Aichi, Japan
| | | | - Hideki Takagi
- Department of Orthopedic Surgery, Nagoya Central Hospital, Nagoya, Aichi, Japan
| | - Nobunori Takahashi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Koji Funahashi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toki Takemoto
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Nobuyuki Asai
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tatsuo Watanabe
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Naoki Ishiguro
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshihisa Kojima
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
64
|
Sirotti S, Generali E, Ceribelli A, Isailovic N, De Santis M, Selmi C. Personalized medicine in rheumatology: the paradigm of serum autoantibodies. AUTOIMMUNITY HIGHLIGHTS 2017; 8:10. [PMID: 28702930 PMCID: PMC5507804 DOI: 10.1007/s13317-017-0098-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/04/2017] [Indexed: 02/08/2023]
Abstract
The sequencing of the human genome is now well recognized as the starting point of personalized medicine. Nonetheless, everyone is unique and can develop different phenotypes of the same disease, despite identical genotypes, as well illustrated by discordant monozygotic twins. To recognize these differences, one of the easiest and most familiar examples of biomarkers capable of identifying and predicting the outcome of patients is represented by serum autoantibodies. In this review, we will describe the concept of personalized medicine and discuss the predictive, prognostic and preventive role of antinuclear antibodies (ANA), anti-citrullinated peptide antibodies (ACPA), rare autoantibodies and anti-drug antibodies (ADA), to evaluate how these can help to identify different disease immune phenotypes and to choose the best option for treating and monitoring rheumatic patients in everyday practice. The importance of ANA resides in the prediction of clinical manifestations in systemic sclerosis and systemic lupus erythematosus and their association with malignancies. ACPA have a predictive role in rheumatoid arthritis, they are associated with the development of a more aggressive disease, extra-articular manifestations and premature mortality in RA patients; moreover, they are capable of predicting therapeutic response. Rare autoantibodies are associated with different disease manifestations and also with a greater incidence of cancer. The determination of ADA levels may be useful in patients where the clinical efficacy of TNF-α inhibitor has dropped, for the assessment of a right management. The resulting scenario supports serum autoantibodies as the cornerstone of personalized medicine in autoimmune diseases.
Collapse
Affiliation(s)
- Silvia Sirotti
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Elena Generali
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Angela Ceribelli
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Natasa Isailovic
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Via A. Manzoni 56, Rozzano, 20089, Milan, Italy. .,BIOMETRA Department, University of Milan, Milan, Italy.
| |
Collapse
|
65
|
Koga T, Okada A, Fukuda T, Hidaka T, Ishii T, Ueki Y, Kodera T, Nakashima M, Takahashi Y, Honda S, Horai Y, Watanabe R, Okuno H, Aramaki T, Izumiyama T, Takai O, Miyashita T, Sato S, Kawashiri SY, Iwamoto N, Ichinose K, Tamai M, Origuchi T, Nakamura H, Aoyagi K, Eguchi K, Kawakami A. Anti-citrullinated peptide antibodies are the strongest predictor of clinically relevant radiographic progression in rheumatoid arthritis patients achieving remission or low disease activity: A post hoc analysis of a nationwide cohort in Japan. PLoS One 2017; 12:e0175281. [PMID: 28505163 PMCID: PMC5432072 DOI: 10.1371/journal.pone.0175281] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/23/2017] [Indexed: 11/18/2022] Open
Abstract
Objectives To determine prognostic factors of clinically relevant radiographic progression (CRRP) in patients with rheumatoid arthritis (RA) achieving remission or low disease activity (LDA) in clinical practice. Methods Using data from a nationwide, multicenter, prospective study in Japan, we evaluated 198 biological disease-modifying antirheumatic drug (bDMARD)-naïve RA patients who were in remission or had LDA at study entry after being treated with conventional synthetic DMARDs (csDMARDs). CRRP was defined as the yearly progression of modified total Sharp score (mTSS) >3.0 U. We performed a multiple logistic regression analysis to explore the factors to predict CRRP at 1 year. We used receiver operating characteristic (ROC) curve to estimate the performance of relevant variables for predicting CRRP. Results The mean Disease Activity Score in 28 joints-erythrocyte sedimentation rate (DAS28-ESR) was 2.32 ± 0.58 at study entry. During the 1-year observation, remission or LDA persisted in 72% of the patients. CRRP was observed in 7.6% of the patients. The multiple logistic regression analysis revealed that the independent variables to predict the development of CRRP were: anti-citrullinated peptide antibodies (ACPA) positivity at baseline (OR = 15.2, 95%CI 2.64–299), time-integrated DAS28-ESR during the 1 year post-baseline (7.85-unit increase, OR = 1.83, 95%CI 1.03–3.45), and the mTSS at baseline (13-unit increase, OR = 1.22, 95%CI 1.06–1.42). Conclusions ACPA positivity was the strongest independent predictor of CRRP in patients with RA in remission or LDA. Physicians should recognize ACPA as a poor-prognosis factor regarding the radiographic outcome of RA, even among patients showing a clinically favorable response to DMARDs.
Collapse
Affiliation(s)
- Tomohiro Koga
- Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
- * E-mail:
| | - Akitomo Okada
- Japanese Red Cross Nagasaki Genbaku Hospital, Department of Rheumatology, Mori-machi 3–15, Nagasaki, Japan
| | - Takaaki Fukuda
- Kurume University Medical Center, Department of Rheumatology, Kokubun 155–1, Kurume, Japan
| | - Toshihiko Hidaka
- Zenjinkai Shimin-no-Mori Hospital, Shioji 2783–37, Miyazaki, Japan
| | - Tomonori Ishii
- Tohoku University Hospital, Department of Hematology and Rheumatology, Aoba-ku, seiryo 1–1, Sendai, Japan
| | - Yukitaka Ueki
- Sasebo Chuo Hospital, Rheumatic and Collagen Disease Center, Yamato 15, Sasebo, Japan
| | - Takao Kodera
- Tohoku Medical and Pharmaceutical University Hospital, Aoba-ku, komatsujima 4-4-1, Sendai, Japan
| | - Munetoshi Nakashima
- Japanese Red Cross Nagasaki Genbaku Hospital, Department of Rheumatology, Mori-machi 3–15, Nagasaki, Japan
- Kurume University Medical Center, Department of Rheumatology, Kokubun 155–1, Kurume, Japan
| | | | - Seiyo Honda
- Kurume University School of Medicine, asahi-machi 67, Kurume, Japan
| | - Yoshiro Horai
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Ryu Watanabe
- Tohoku University Hospital, Department of Hematology and Rheumatology, Aoba-ku, seiryo 1–1, Sendai, Japan
| | - Hiroshi Okuno
- Tohoku University Hospital, Department of Orthopaedic Surgery, Hirase 9–3, Sasebo, Japan
| | - Toshiyuki Aramaki
- Japanese Red Cross Nagasaki Genbaku Hospital, Department of Rheumatology, Mori-machi 3–15, Nagasaki, Japan
- Sasebo Chuo Hospital, Rheumatic and Collagen Disease Center, Yamato 15, Sasebo, Japan
| | - Tomomasa Izumiyama
- East Sendai Rheumatism and internal medicine Clinic, Miyagino, Nittahigashi 1-17-5, Sendai, Japan
| | - Osamu Takai
- Osaki Citizen Hospital, Furukawa-honami 3-8-1, Osaki, Japan
| | | | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, Sakamoto 1-7-1, Nagasaki, Japan
| | - Shin-ya Kawashiri
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-12-4, Nagasaki, Japan
| | - Naoki Iwamoto
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Kunihiro Ichinose
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Mami Tamai
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Tomoki Origuchi
- Department of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-12-4, Nagasaki, Japan
| | - Hideki Nakamura
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Kiyoshi Aoyagi
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-12-4, Nagasaki, Japan
| | | | - Atsushi Kawakami
- Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | | |
Collapse
|
66
|
Humby FC, Al Balushi F, Lliso G, Cauli A, Pitzalis C. Can Synovial Pathobiology Integrate with Current Clinical and Imaging Prediction Models to Achieve Personalized Health Care in Rheumatoid Arthritis? Front Med (Lausanne) 2017; 4:41. [PMID: 28516086 PMCID: PMC5413506 DOI: 10.3389/fmed.2017.00041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/27/2017] [Indexed: 11/13/2022] Open
Abstract
Although great progress has been made in the past decade toward understanding the pathogenesis of rheumatoid arthritis (RA), clinicians remain some distance from a goal of personalized health care. The capacity to diagnose RA early, predict prognosis, and moreover predict response to biologic therapies has been a research focus for many years. How currently available clinical prediction models can facilitate such goals is reviewed in this article. In addition, the role of current imaging techniques in this regard is also discussed. Finally, the authors review the current literature regarding synovial biomarkers and consider whether integration of synovial pathobiology into clinical prediction algorithms may enhance their predictive value.
Collapse
Affiliation(s)
- Frances Claire Humby
- Department of Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Gloria Lliso
- Department of Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Alberto Cauli
- Dipartimento di Scienze Mediche, Facoltà di Medicina e Chirurgia, Università degli Studi di Cagliari, Cagliari, Italy
| | - Costantino Pitzalis
- Department of Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
67
|
Wie häufig sind prognostisch ungünstige Faktoren bei Patienten mit rheumatoider Arthritis? Z Rheumatol 2017; 76:434-442. [DOI: 10.1007/s00393-017-0306-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
68
|
Albrecht K, Zink A. Poor prognostic factors guiding treatment decisions in rheumatoid arthritis patients: a review of data from randomized clinical trials and cohort studies. Arthritis Res Ther 2017; 19:68. [PMID: 28335797 PMCID: PMC5364634 DOI: 10.1186/s13075-017-1266-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Prognostic factors are used for treatment decisions in rheumatoid arthritis (RA). High disease activity, the early presence of erosions, and autoantibody positivity are the most frequently used poor prognostic factors but other features, such as functional disability, extraarticular disease, or multibiomarkers, are also assessed. Prognostic factors are incorporated in current treatment recommendations for the management of RA and are used as inclusion criteria in randomized controlled trials. They are defined heterogeneously and the relevance of a single or combined presence of poor prognostic factors remains unclear. This review summarizes the current definitions of poor prognostic factors and their use in clinical research. Perspectives on future research are also outlined.
Collapse
Affiliation(s)
- Katinka Albrecht
- German Rheumatism Research Centre, Epidemiology Unit, Charitéplatz 1, 10117 Berlin, Germany
| | - Angela Zink
- German Rheumatism Research Centre, Epidemiology Unit, Charitéplatz 1, 10117 Berlin, Germany
- Rheumatology and Clinical Immunology, Charité University Medicine, Berlin, Germany
| |
Collapse
|
69
|
Smolen JS, Landewé R, Bijlsma J, Burmester G, Chatzidionysiou K, Dougados M, Nam J, Ramiro S, Voshaar M, van Vollenhoven R, Aletaha D, Aringer M, Boers M, Buckley CD, Buttgereit F, Bykerk V, Cardiel M, Combe B, Cutolo M, van Eijk-Hustings Y, Emery P, Finckh A, Gabay C, Gomez-Reino J, Gossec L, Gottenberg JE, Hazes JMW, Huizinga T, Jani M, Karateev D, Kouloumas M, Kvien T, Li Z, Mariette X, McInnes I, Mysler E, Nash P, Pavelka K, Poór G, Richez C, van Riel P, Rubbert-Roth A, Saag K, da Silva J, Stamm T, Takeuchi T, Westhovens R, de Wit M, van der Heijde D. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis 2017; 76:960-977. [PMID: 28264816 DOI: 10.1136/annrheumdis-2016-210715] [Citation(s) in RCA: 1765] [Impact Index Per Article: 252.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 01/05/2017] [Accepted: 02/09/2017] [Indexed: 02/07/2023]
Abstract
Recent insights in rheumatoid arthritis (RA) necessitated updating the European League Against Rheumatism (EULAR) RA management recommendations. A large international Task Force based decisions on evidence from 3 systematic literature reviews, developing 4 overarching principles and 12 recommendations (vs 3 and 14, respectively, in 2013). The recommendations address conventional synthetic (cs) disease-modifying antirheumatic drugs (DMARDs) (methotrexate (MTX), leflunomide, sulfasalazine); glucocorticoids (GC); biological (b) DMARDs (tumour necrosis factor (TNF)-inhibitors (adalimumab, certolizumab pegol, etanercept, golimumab, infliximab), abatacept, rituximab, tocilizumab, clazakizumab, sarilumab and sirukumab and biosimilar (bs) DMARDs) and targeted synthetic (ts) DMARDs (Janus kinase (Jak) inhibitors tofacitinib, baricitinib). Monotherapy, combination therapy, treatment strategies (treat-to-target) and the targets of sustained clinical remission (as defined by the American College of Rheumatology-(ACR)-EULAR Boolean or index criteria) or low disease activity are discussed. Cost aspects were taken into consideration. As first strategy, the Task Force recommends MTX (rapid escalation to 25 mg/week) plus short-term GC, aiming at >50% improvement within 3 and target attainment within 6 months. If this fails stratification is recommended. Without unfavourable prognostic markers, switching to-or adding-another csDMARDs (plus short-term GC) is suggested. In the presence of unfavourable prognostic markers (autoantibodies, high disease activity, early erosions, failure of 2 csDMARDs), any bDMARD (current practice) or Jak-inhibitor should be added to the csDMARD. If this fails, any other bDMARD or tsDMARD is recommended. If a patient is in sustained remission, bDMARDs can be tapered. For each recommendation, levels of evidence and Task Force agreement are provided, both mostly very high. These recommendations intend informing rheumatologists, patients, national rheumatology societies, hospital officials, social security agencies and regulators about EULAR's most recent consensus on the management of RA, aimed at attaining best outcomes with current therapies.
Collapse
Affiliation(s)
- Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria.,2nd Department of Medicine, Hietzing Hospital, Vienna, Austria
| | - Robert Landewé
- Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands.,Zuyderland Medical Center, Heerlen, The Netherlands
| | - Johannes Bijlsma
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerd Burmester
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Free University and Humboldt University Berlin, Berlin, Germany
| | | | | | - Jackie Nam
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust and Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Sofia Ramiro
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke Voshaar
- Department of Psychology, Health and Technology, University of Twente, Enschede, The Netherlands
| | - Ronald van Vollenhoven
- Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands.,Zuyderland Medical Center, Heerlen, The Netherlands
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Martin Aringer
- Division of Rheumatology, Medizinische Klinik und Poliklinik III, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Maarten Boers
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Chris D Buckley
- Birmingham NIHR Wellcome Trust Clinical Research Facility, Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Free University and Humboldt University Berlin, Berlin, Germany
| | - Vivian Bykerk
- Department of Rheumatology, Hospital for Special Surgery, Weill Cornell Medical College, New York, New York, USA.,Rebecca McDonald Center for Arthritis & Autoimmune Disease, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mario Cardiel
- Centro de Investigación Clínica de Morelia SC, Michoacán, México
| | - Bernard Combe
- Rheumatology Department, Lapeyronie Hospital, Montpellier University, UMR 5535, Montpellier, France
| | - Maurizio Cutolo
- Research Laboratory and Division of Clinical Rheumatology, University of Genoa, Genoa, Italy
| | - Yvonne van Eijk-Hustings
- Department of Patient & Care and Department of Rheumatology, University of Maastricht, Maastricht, The Netherlands
| | - Paul Emery
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Axel Finckh
- Division of Rheumatology, University Hospitals of Geneva, Geneva, Switzerland
| | - Cem Gabay
- Division of Rheumatology, University Hospitals of Geneva, Geneva, Switzerland
| | - Juan Gomez-Reino
- Fundación Ramón Dominguez, Hospital Clinico Universitario, Santiago, Spain
| | - Laure Gossec
- Department of Rheumatology, Sorbonne Universités, Pitié Salpêtrière Hospital, Paris, France
| | - Jacques-Eric Gottenberg
- Institut de Biologie Moléculaire et Cellulaire, Immunopathologie, et Chimie Thérapeutique, Strasbourg University Hospital and University of Strasbourg, CNRS, Strasbourg, France
| | - Johanna M W Hazes
- Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tom Huizinga
- Department of Psychology, Health and Technology, University of Twente, Enschede, The Netherlands
| | - Meghna Jani
- Arthritis Research UK Centre for Epidemiology, Centre for Musculoskeletal Research, University of Manchester, Manchester, UK
| | - Dmitry Karateev
- V.A. Nasonova Research Institute of Rheumatology, Moscow, Russian Federation
| | - Marios Kouloumas
- European League Against Rheumatism, Zurich, Switzerland.,Cyprus League against Rheumatism, Nicosia, Cyprus
| | - Tore Kvien
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Beijing University People's Hospital, Beijing, China
| | - Xavier Mariette
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Université Paris-Sud, INSERM U1184, Center for Immunology of viral Infections and Autoimmune Diseases (IMVA), Le Kremlin Bicêtre, France
| | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Eduardo Mysler
- Organización Médica de Investigación, Buenos Aires, Argentina
| | - Peter Nash
- Department of Medicine, University of Queensland, Queensland, Australia
| | - Karel Pavelka
- Institute of Rheumatology and Clinic of Rheumatology, Charles University, Prague, Czech Republic
| | - Gyula Poór
- National Institute of Rheumatology and Physiotherapy, Semmelweis University, Budapest, Hungary
| | - Christophe Richez
- Rheumatology Department, FHU ACRONIM, Pellegrin Hospital and UMR CNRS 5164, Bordeaux University, Bordeaux, France
| | - Piet van Riel
- Department of Rheumatology, Bernhoven, Uden, The Netherlands
| | | | - Kenneth Saag
- Division of Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jose da Silva
- Serviço de Reumatologia, Centro Hospitalar e Universitário de Coimbra Praceta Mota Pinto, Coimbra, Portugal
| | - Tanja Stamm
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Tsutomu Takeuchi
- Keio University School of Medicine, Keio University Hospital, Tokyo, Japan
| | - René Westhovens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Department of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Maarten de Wit
- Department Medical Humanities, VU Medical Centre, Amsterdam, The Netherlands
| | | |
Collapse
|
70
|
Joo YB, Bang SY, Ryu JA, Lee S, Lee HS, Bae SC. Predictors of severe radiographic progression in patients with early rheumatoid arthritis: A Prospective observational cohort study. Int J Rheum Dis 2017; 20:1437-1446. [DOI: 10.1111/1756-185x.13054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Young Bin Joo
- Department of Rheumatology; St. Vincent's Hospital; The Catholic University of Korea; Suwon Korea
| | - So-Young Bang
- Department of Rheumatology; Hanyang University Hospital for Rheumatic Diseases; Seoul Korea
| | - Jeong Ah Ryu
- Department of Radiology; Hanyang University Hospital; Guri Korea
| | - Seunghun Lee
- Department of Radiology; Hanyang University Hospital; Seoul Korea
| | - Hye-Soon Lee
- Department of Rheumatology; Hanyang University Hospital for Rheumatic Diseases; Seoul Korea
| | - Sang-Cheol Bae
- Department of Rheumatology; Hanyang University Hospital for Rheumatic Diseases; Seoul Korea
| |
Collapse
|
71
|
Bird P, Nicholls D, Barrett R, de Jager J, Griffiths H, Roberts L, Tymms K, McCloud P, Littlejohn G. Longitudinal study of clinical prognostic factors in patients with early rheumatoid arthritis: the PREDICT study. Int J Rheum Dis 2017; 20:460-468. [PMID: 28205333 DOI: 10.1111/1756-185x.13036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
AIM To assess the association between baseline clinical prognostic factors and subsequent Disease Activity Score of 28 joints (DAS28) remission in early rheumatoid arthritis (RA). METHODS Data were collected using point of care clinical software from participating rheumatology centres. Patients aged 18 years or over whose date of clinical onset of RA was within the previous 12-24 months, who had at least 6 months of follow-up data and a DAS28-ESR (erythrocyte sedimentation rate) score recorded between 12 and 24 months from first being seen for RA were included. Data collected included baseline demographics, mode of disease onset, pattern of joint involvement at onset, smoking status, DAS28, rheumatoid factor (RF), anti-citrullinated peptide antibodies (ACPA), time from symptom onset to presentation and disease activity at baseline. Univariate and multivariate logistic regression of DAS28-ESR remission between 12 and 24 months after first assessment were performed. RESULTS Data from 1017 patients were analyzed: 70% female; mean age 60 years (SD: 14.7); 70% RF-positive, 58% ACPA-positive. The strongest age and sex adjusted baseline predictors of DAS28-ESR remission at 12-24 months were remission at baseline (odds ratio [OR]: 4.49, 95% CI: 2.17-9.29, P < 0.001), being male (OR: 2.42, 95% CI: 1.46-4.01, P < 0.001), abstaining from alcohol (P < 0.001) and being lower weight (OR: 0.98, 95% CI: 0.97-1.00, P = 0.015). There was no statistically significant association between joint onset patterns, mode of onset, RF, ACPA or smoking status. CONCLUSION In this observational study, patients with early RA at risk of not achieving remission include those with high disease activity at baseline, women, those who drink alcohol and those with higher body weight.
Collapse
Affiliation(s)
- Paul Bird
- University of New South Wales and Combined Rheumatology Practice, Kogarah, New South Wales, Australia
| | - Dave Nicholls
- Coast Joint Care, Maroochydore, Queensland, Australia
| | - Rina Barrett
- Roche Products, Pty. Limited, Sydney, New South Wales, Australia
| | | | | | | | - Kathleen Tymms
- Canberra Rheumatology, Canberra, Australian Capital Territory, Australia
| | - Philip McCloud
- McCloud Consulting Group, Sydney, New South Wales, Australia
| | - Geoffrey Littlejohn
- Monash Medical Centre, Melbourne, Victoria, Australia.,Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
72
|
Landewé RBM, Connell CA, Bradley JD, Wilkinson B, Gruben D, Strengholt S, van der Heijde D. Is radiographic progression in modern rheumatoid arthritis trials still a robust outcome? Experience from tofacitinib clinical trials. Arthritis Res Ther 2016; 18:212. [PMID: 27663201 PMCID: PMC5034418 DOI: 10.1186/s13075-016-1106-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/01/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The detection of statistically significant reductions in radiographic progression during clinical studies in patients with rheumatoid arthritis (RA) has become increasingly difficult over the past decade due to early-escape study designs and declining rates of progression in control-group patients. We investigated the impact of extremes of radiographic data (outliers) and baseline prognostic factors on detection of treatment effects, to provide guidance on future analysis of joint structural data in RA clinical trials. METHODS Data were from two, phase 3, randomized, double-blind, placebo-controlled trials of tofacitinib in adult patients with moderate to severe RA: ORAL Scan (NCT00847613) and ORAL Start (NCT01039688). These studies detected significant reductions in radiographic progression with tofacitinib 10 mg twice daily (BID) plus background methotrexate (ORAL Scan), and with tofacitinib 5 or 10 mg BID as monotherapy (ORAL Start). We evaluated mean changes from baseline in van der Heijde modified total Sharp score (mTSS) at month 6 and month 12, using analysis of covariance (ANCOVA). A trimmed analysis was used to deal with extremes of data. The impact of baseline prognostic factors on radiographic progression was evaluated using ANCOVA to analyze the mean change from baseline in mTSS for each factor in turn. RESULTS The analysis included data from 720 patients from ORAL Scan and 880 patients from ORAL Start. Trimmed analyses were unbiased for the true mean estimate and enabled us to remove the effect of influential extreme observations in the data set. Almost all patients had at least one poor prognostic factor at baseline (e.g., high level of disease activity, or positive for rheumatoid factor). The strongest predictor of treatment effect was the severity of radiographic damage at baseline. CONCLUSIONS A trimmed analysis can establish whether any significant inhibition of structural damage is being driven by extremes of data, and should be one of the sensitivity analyses of choice for structural data in RA clinical trials. Furthermore, analysis of radiographic data based on baseline prognostic factors may reveal increased treatment effects. Application of these methods to analysis of radiographic data from clinical trials in patients with RA, allows a more complete interpretation of data. TRIAL REGISTRATION Clinicaltrials.gov NCT00847613 (registered 17 February 2009) and NCT01039688 (registered 23 December 2009).
Collapse
Affiliation(s)
- Robert B M Landewé
- Amsterdam Rheumatology Center, Amsterdam, The Netherlands.,Atrium Medical Center, Heerlen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
73
|
Kawashiri SY, Nishino A, Shimizu T, Umeda M, Fukui S, Nakashima Y, Suzuki T, Koga T, Iwamoto N, Ichinose K, Tamai M, Nakamura H, Origuchi T, Aoyagi K, Kawakami A. Ultrasound disease activity of bilateral wrist and finger joints at three months reflects the clinical response at six months of patients with rheumatoid arthritis treated with biologic disease-modifying anti-rheumatic drugs. Mod Rheumatol 2016; 27:252-256. [DOI: 10.1080/14397595.2016.1221874] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Shin-Ya Kawashiri
- Departments of Public Health,
- Departments of Immunology and Rheumatology, and
| | | | | | | | | | | | | | | | | | | | - Mami Tamai
- Departments of Immunology and Rheumatology, and
| | | | - Tomoki Origuchi
- Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | |
Collapse
|
74
|
Figueiredo CP, Simon D, Englbrecht M, Haschka J, Kleyer A, Bayat S, Hueber A, Pereira RMR, Rech J, Schett G. Quantification and Impact of Secondary Osteoarthritis in Patients With Anti-Citrullinated Protein Antibody-Positive Rheumatoid Arthritis. Arthritis Rheumatol 2016; 68:2114-21. [DOI: 10.1002/art.39698] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/24/2016] [Indexed: 02/02/2023]
Affiliation(s)
- Camille P. Figueiredo
- University of Erlangen-Nuremberg, Erlangen, Germany, and Universidade de São Paulo; São Paulo Brazil
| | - David Simon
- University of Erlangen-Nuremberg; Erlangen Germany
| | | | - Judith Haschka
- University of Erlangen-Nuremberg, Erlangen, Germany, and St. Vincent Hospital; Vienna Austria
| | - Arnd Kleyer
- University of Erlangen-Nuremberg; Erlangen Germany
| | - Sara Bayat
- University of Erlangen-Nuremberg; Erlangen Germany
| | - Axel Hueber
- University of Erlangen-Nuremberg; Erlangen Germany
| | | | - Juergen Rech
- University of Erlangen-Nuremberg; Erlangen Germany
| | - Georg Schett
- University of Erlangen-Nuremberg; Erlangen Germany
| |
Collapse
|
75
|
Katchamart W, Narongroeknawin P, Chevaisrakul P, Dechanuwong P, Mahakkanukrauh A, Kasitanon N, Pakchotanon R, Sumethkul K, Ueareewongsa P, Ukritchon S, Bhurihirun T, Duangkum K, Intapiboon P, Intongkam S, Jangsombatsiri W, Jatuworapruk K, Kositpesat N, Leungroongroj P, Lomarat W, Petcharat C, Sittivutworapant S, Suebmee P, Tantayakom P, Tipsing W, Asavatanabodee P, Chiowchanwisawakit P, Foocharoen C, Koolvisoot A, Louthrenoo W, Siripaitoon B, Totemchokchyakarn K, Kitumnuaypong T. Evidence-based recommendations for the diagnosis and management of rheumatoid arthritis for non-rheumatologists: Integrating systematic literature research and expert opinion of the Thai Rheumatism Association. Int J Rheum Dis 2016; 20:1142-1165. [DOI: 10.1111/1756-185x.12905] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Wanruchada Katchamart
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj hospital; Mahidol University; Bangkok Thailand
| | - Pongthorn Narongroeknawin
- Division of Rheumatology; Department of Medicine; Pramongkutklao and College of Medicine; Bangkok Thailand
| | - Parawee Chevaisrakul
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Ramathibodi Hospital; Mahidol; Bangkok Thailand
| | - Pornchai Dechanuwong
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Vajira Hospital; Navamindradhiraj University; Bangkok Thailand
| | - Ajanee Mahakkanukrauh
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - Nuntana Kasitanon
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | - Rattapol Pakchotanon
- Division of Rheumatology; Department of Medicine; Pramongkutklao and College of Medicine; Bangkok Thailand
| | - Kittiwan Sumethkul
- Rheumatology Unit; Department of Internal Medicine; Rajavithi Hospital; Bangkok Thailand
| | - Parichat Ueareewongsa
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Prince of Songkla University; Songkla Thailand
| | - Sittichai Ukritchon
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Chulalongkorn University; Bangkok Thailand
| | - Thitirat Bhurihirun
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj hospital; Mahidol University; Bangkok Thailand
| | - Kittikorn Duangkum
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - Porntip Intapiboon
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Prince of Songkla University; Songkla Thailand
| | - Samanan Intongkam
- Division of Rheumatology; Department of Medicine; Pramongkutklao and College of Medicine; Bangkok Thailand
| | - Wimol Jangsombatsiri
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Ramathibodi Hospital; Mahidol; Bangkok Thailand
| | - Kanon Jatuworapruk
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | - Naravadee Kositpesat
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Chulalongkorn University; Bangkok Thailand
| | - Pawinee Leungroongroj
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Ramathibodi Hospital; Mahidol; Bangkok Thailand
| | - Wiyanoot Lomarat
- Division of Rheumatology; Department of Medicine; Pramongkutklao and College of Medicine; Bangkok Thailand
| | - Chonachan Petcharat
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj hospital; Mahidol University; Bangkok Thailand
| | | | - Patcharawan Suebmee
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - Pongchirat Tantayakom
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj hospital; Mahidol University; Bangkok Thailand
| | - Worakan Tipsing
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Vajira Hospital; Navamindradhiraj University; Bangkok Thailand
| | - Paijit Asavatanabodee
- Division of Rheumatology; Department of Medicine; Pramongkutklao and College of Medicine; Bangkok Thailand
| | - Praveena Chiowchanwisawakit
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj hospital; Mahidol University; Bangkok Thailand
| | - Chingching Foocharoen
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Khon Kaen University; Khon Kaen Thailand
| | - Ajchara Koolvisoot
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Siriraj hospital; Mahidol University; Bangkok Thailand
| | - Worawit Louthrenoo
- Division of Rheumatology; Department of Internal Medicine; Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | - Boonjing Siripaitoon
- Division of Rheumatology; Department of Medicine; Faculty of Medicine; Prince of Songkla University; Songkla Thailand
| | - Kitti Totemchokchyakarn
- Division of Allergy, Immunology and Rheumatology; Department of Medicine; Faculty of Medicine; Ramathibodi Hospital; Mahidol; Bangkok Thailand
| | - Tasanee Kitumnuaypong
- Rheumatology Unit; Department of Internal Medicine; Rajavithi Hospital; Bangkok Thailand
| | | |
Collapse
|
76
|
Ke Y, Fu B, Zhang W. Semi-varying coefficient multinomial logistic regression for disease progression risk prediction. Stat Med 2016; 35:4764-4778. [DOI: 10.1002/sim.7034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 05/27/2016] [Accepted: 06/13/2016] [Indexed: 02/03/2023]
Affiliation(s)
- Yuan Ke
- Department of Operational Research and Financial Engineering; Princeton University; Princeton 08540 NJ U.S.A
| | - Bo Fu
- Administrative Data Research Centre for England and Institute of Child Health; University College London; London NW1 2DA U.K
- Centre for Biostatistics and Arthritis Research UK Epidemiology Unit; The University of Manchester; Manchester M13 9PL U.K
| | - Wenyang Zhang
- Department of Mathematics; The University of York; York YO10 5DD U.K
| |
Collapse
|
77
|
Detert J, Burmester GR. [Treat to target and personalized medicine (precision medicine)]. Z Rheumatol 2016; 75:624-32. [PMID: 27365026 DOI: 10.1007/s00393-016-0137-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- J Detert
- Klinik m.S. Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland.
| | - G R Burmester
- Klinik m.S. Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland
| |
Collapse
|
78
|
Granger B, Combe B, Le Loet X, Saraux A, Guillemin F, Fautrel B. Performance of matrices developed to identify patients with early rheumatoid arthritis with rapid radiographic progression despite methotrexate therapy: an external validation study based on the ESPOIR cohort data. RMD Open 2016; 2:e000245. [PMID: 27252898 PMCID: PMC4879338 DOI: 10.1136/rmdopen-2016-000245] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/07/2016] [Accepted: 04/16/2016] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Use of prediction matrices of risk or rapid radiographic progression (RRP) for early rheumatoid arthritis (RA) in clinical practice could help to better rationalise the first line of treatment. Before use, they must be validated in populations that have not participated in their construction. The main objective is to use the ESPOIR cohort to validate the performance of 3 matrices (ASPIRE, BEST and SONORA) to predict patients at high risk of RRP at 1 year of disease despite initial treatment with methotrexate (MTX). METHODS We selected from the ESPOIR cohort 370 patients receiving MTX or leflunomide (LEF) for ≥3 months within the first year of follow-up. Patients were assessed clinically every 6 months, and structural damage progression seen on radiography was measured by the van der Heijde-modified Sharp score (vSHS) at 1 year. RRP was defined as an increase in the vSHS≥5 points during the first year. RESULTS At 1 year, the mean vSHS score was 1.7±5.0 and 46 patients had RRP. The ASPIRE matrix had only moderate validity in the ESPOIR population, with area under the receiver operating characteristic curve (AUC) <0.7. The AUC for the BEST and SONORA matrices were 0.73 and 0.76. Presence of rheumatoid factor (RF)-or anti-citrullinated protein antibodies (ACPAs) and initial structural damage were always predictive of RRP at 1 year. Disease Activity Score in 28 joints (DAS28) and C reactive protein (ASPIRE threshold) were not associated with RRP. CONCLUSIONS Matrices to identify patients at risk of RRP tested in the ESPOIR cohort seem to perform moderately. There is no matrix that shows clearly superior performance.
Collapse
Affiliation(s)
- Benjamin Granger
- Université Pierre et Marie Curie (UPMC)—Paris 6, GRC 08, Pierre Louis Institute of Epidemiology and Public Health, Paris, France
- Department of Biostatistics, Public Health and Medical Information, AP-HP Pitié Salpêtrière Hospital, Paris, France
| | - Bernard Combe
- Department of Rheumatology, Montpellier I University; Lapeyronie Hospital, Montpellier, France
| | - Xavier Le Loet
- Rheumatology Department, Rouen University Hospital & INSERM U905, Institute for Research and Innovation in Biomedicine, Rouen University, Rouen, France
| | - Alain Saraux
- Department of Rheumatology, Brest University, La Cavale Blanche University Hospital, Brest, France
- INSERM ESPRI, ERI29 Université Bretagne Occidentale, Brest, France
| | - Francis Guillemin
- EA 4360 APEMAC, Lorraine University, Paris-Descartes University, Nancy, France
- Faculty of Medicine, CS 50184, 54505 Vandoeuvre-lès-Nancy & Inserm, CIC-EC, CHU de Brabois, 54505 Vandoeuvre-lès-Nancy, Nancy, France
| | - Bruno Fautrel
- Université Pierre et Marie Curie (UPMC)—Paris 6, GRC 08, Pierre Louis Institute of Epidemiology and Public Health, Paris, France
- Department of Rheumatology, Pitié Salpêtrière Hospital, Paris, France
| |
Collapse
|
79
|
Nakashima Y, Tamai M, Kita J, Michitsuji T, Shimizu T, Fukui S, Umeda M, Nishino A, Suzuki T, Horai Y, Okada A, Nishimura T, Koga T, Kawashiri SY, Iwamoto N, Ichinose K, Hirai Y, Arima K, Yamasaki S, Nakamura H, Origuchi T, Takao S, Uetani M, Aoyagi K, Eguchi K, Kawakami A. Magnetic Resonance Imaging Bone Edema at Enrollment Predicts Rapid Radiographic Progression in Patients with Early RA: Results from the Nagasaki University Early Arthritis Cohort. J Rheumatol 2016; 43:1278-84. [PMID: 27134251 DOI: 10.3899/jrheum.150988] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To clarify whether magnetic resonance imaging (MRI) bone edema predicts the development of rapid radiographic progression (RRP) in the Nagasaki University Early Arthritis Cohort of patients with early-stage rheumatoid arthritis (RA). METHODS Patients with early-stage RA (n = 76) were enrolled and underwent 1.5-T MRI of both wrists and finger joints. Synovitis, bone edema, and bone erosion were evaluated using the Rheumatoid Arthritis Magnetic Resonance Imaging Scoring (RAMRIS). RRP was defined as an annual increment > 3 at 1 year by the Genant-modified Sharp score of plain radiographs. A multivariate logistic regression analysis was performed to establish the risk factors for RRP. RESULTS Median disease duration at enrollment was 3 months. RRP was found in 12 of the 76 patients at 1 year. A univariate analysis revealed that matrix metalloprotease-3, RAMRIS bone edema score, and RAMRIS bone erosion score were associated with RRP. Multivariate logistic regression analyses demonstrated that the RAMRIS bone edema score at enrollment (5-point increase, OR 2.18, 95% CI 1.32-3.59, p = 0.002) was the only independent predictor of the development of RRP at 1 year. A receiver-operating characteristic analysis identified the best cutoff value for RAMRIS bone edema score as 5. RRP was significantly rare among the patients with a RAMRIS bone edema score < 5 at enrollment (2 from 50 patients). CONCLUSION Our findings suggest that MRI bone edema is closely associated with the development of RRP in patients with early-stage RA. Physicians should carefully control the disease activity when MRI bone edema is observed in patients with early RA.
Collapse
Affiliation(s)
- Yoshikazu Nakashima
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Mami Tamai
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Junko Kita
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Toru Michitsuji
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Toshimasa Shimizu
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Shoichi Fukui
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Masataka Umeda
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Ayako Nishino
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Takahisa Suzuki
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Yoshiro Horai
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Akitomo Okada
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Takayuki Nishimura
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Tomohiro Koga
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Shin-Ya Kawashiri
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Naoki Iwamoto
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Kunihiro Ichinose
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Yasuko Hirai
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Kazuhiko Arima
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Satoshi Yamasaki
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Hideki Nakamura
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Tomoki Origuchi
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Shoichiro Takao
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Masataka Uetani
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Kiyoshi Aoyagi
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Katsumi Eguchi
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| | - Atsushi Kawakami
- From the Department of Immunology and Rheumatology, and Department of Public Health, and Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences; Japanese Red Cross Nagasaki Genbaku Hospital; Department of Radiology and Radiation Research, Nagasaki University Hospital, Nagasaki; Department of Clinical Immunology and Rheumatology, Hiroshima University, Hiroshima; Department of Radiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima; Sasebo Chuo Hospital, Sasebo, Japan.Y. Nakashima, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Tamai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; J. Kita, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Michitsuji, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Shimizu, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Fukui, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; M. Umeda, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Nishino, MD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; T. Suzuki, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; Y. Horai, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; A. Okada, MD, PhD, Japanese Red Cross Nagasaki Genbaku Hospital; T. Nishimura, PhD, Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences; T. Koga, MD, PhD, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences; S. Kawashiri, MD
| |
Collapse
|
80
|
Ajeganova S, van Steenbergen HW, Verheul MK, Forslind K, Hafström I, Toes REM, Huizinga TWJ, Svensson B, Trouw LA, van der Helm-van Mil AHM. The association between anti-carbamylated protein (anti-CarP) antibodies and radiographic progression in early rheumatoid arthritis: a study exploring replication and the added value to ACPA and rheumatoid factor. Ann Rheum Dis 2016; 76:112-118. [DOI: 10.1136/annrheumdis-2015-208870] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/31/2016] [Accepted: 04/09/2016] [Indexed: 11/04/2022]
Abstract
ObjectiveAnti-carbamylated protein (anti-CarP) antibodies are reported to associate with more radiographic progression within the total rheumatoid arthritis (RA) population and anti-citrullinated peptide antibody (ACPA)-negative subgroup. We explored the association of anti-CarP with radiographic progression in RA and aimed to replicate the association and evaluate the added value of anti-CarP antibodies in relation to ACPA and rheumatoid factor (RF).Methods576 Swedish and 628 Dutch patients with RA (2394 and 3247 sets of radiographs, respectively) were longitudinally studied. Replication was restricted to the Swedish patients. In both cohorts, the association of anti-CarP with radiographic progression was determined in strata of patients with similar ACPA and RF status; results of both cohorts were combined in fixed-effect meta-analyses. The net percentage of patients for whom the radiographic progression in 5 years was additionally correctly classified when adding anti-CarP to a model including ACPA and RF was evaluated.ResultsAnti-CarP associated with radiographic progression in the total Swedish RA population (beta=1.11 per year, p=8.75×10−13) and in the ACPA-negative subgroup (beta=1.14 per year, p=0.034). Anti-CarP associated with more radiographic progression in the strata of ACPA-positive/RF-negative, ACPA-negative/RF-positive and ACPA-positive/RF-positive patients with RA (respective p values 0.014, 0.019 and 0.0056). A model including ACPA and RF correctly classified 54% and 57% of the patients; adding anti-CarP to this model did not increase these percentages (54% and 56% were correctly classified).ConclusionsAnti-CarP antibodies associated with more severe radiographic progression in the total and ACPA-negative RA population. Anti-CarP-positivity had a statistically significant additive value to ACPA and RF, but did not improve correct classification of patients.
Collapse
|
81
|
Koga T, Okada A, Fukuda T, Hidaka T, Ishii T, Ueki Y, Kodera T, Nakashima M, Takahashi Y, Honda S, Horai Y, Watanabe R, Okuno H, Aramaki T, Izumiyama T, Takai O, Miyashita T, Sato S, Kawashiri SY, Iwamoto N, Ichinose K, Tamai M, Origuchi T, Nakamura H, Aoyagi K, Eguchi K, Kawakami A. Prognostic Factors Toward Clinically Relevant Radiographic Progression in Patients With Rheumatoid Arthritis in Clinical Practice: A Japanese Multicenter, Prospective Longitudinal Cohort Study for Achieving a Treat-to-Target Strategy. Medicine (Baltimore) 2016; 95:e3476. [PMID: 27124044 PMCID: PMC4998707 DOI: 10.1097/md.0000000000003476] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
To determine prognostic factors of clinically relevant radiographic progression (CRRP) in patients with rheumatoid arthritis (RA) in clinical practice.We performed a multicenter prospective study in Japan of biological disease-modifying antirheumatic drug (bDMARD)-naive RA patients with moderate to high disease activity treated with conventional synthetic DMARDs (csDMARDs) at study entry. We longitudinally observed 408 patients for 1 year and assessed disease activity every 3 months. CRRP was defined as yearly progression of modified total Sharp score (mTSS) > 3.0 U. We also divided the cohort into 2 groups based on disease duration (<3 vs ≥3 years) and performed a subgroup analysis.CRRP was found in 10.3% of the patients. A multiple logistic regression analysis revealed that the independent variables to predict the development of CRRP were: CRP at baseline (0.30 mg/dL increase, 95% confidence interval [CI] 1.01-1.11), time-integrated Disease Activity Score in 28 joints-erythrocyte sedimentation rate (DAS28-ESR) during the 1 year postbaseline (12.4-unit increase, 95%CI 1.17-2.59), RA typical erosion at baseline (95%CI 1.56-21.1), and the introduction of bDMARDs (95%CI 0.06-0.38). The subgroup analysis revealed that time-integrated DAS28-ESR is not a predictor whereas the introduction of bDMARDs is a significant protective factor for CRRP in RA patients with disease duration <3 years.We identified factors that could be used to predict the development of CRRP in RA patients treated with DMARDs. These variables appear to be different based on the RA patients' disease durations.
Collapse
Affiliation(s)
- Tomohiro Koga
- From the Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences (TK,YH, S-YK, NI, KI, MT, HN, AK); Japanese Red Cross Nagasaki Genbaku Hospital, Department of Rheumatology, Nagasaki (AO, MN, TA); Department of Rheumatology, Kurume University Medical Center, Kurume (TF, MN); Zenjinkai Shimin-no-Mori Hospital, Miyazaki (TH); Department of Hematology and Rheumatology, Tohoku University Hospital, Sendai (TI, RW); Rheumatic and Collagen Disease Center, Sasebo Chuo Hospital, Sasebo (YU); Tohoku Pharmaceutical University Hospital, Sendai (TK); Yu Family Clinic, Miyagi (YT); Kurume University School of Medicine, Kurume (SH); Department of Orthopaedic Surgery, Tohoku University Hospital (HO); East Sendai Rheumatism and Internal Medicine Clinic, Sendai (TI); Osaki Citizen Hospital, Osaki (OT); NHO Nagasaki Medical Center, Omura (TM); Clinical Research Center, Nagasaki University Hospital (SS); Department of Public Health (S-YK, KA); Department of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki (TO); and Sasebo City General Hospital, Sasebo (KE), Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Genetic data: The new challenge of personalized medicine, insights for rheumatoid arthritis patients. Gene 2016; 583:90-101. [PMID: 26869316 DOI: 10.1016/j.gene.2016.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/18/2016] [Accepted: 02/05/2016] [Indexed: 01/15/2023]
Abstract
Rapid advances in genotyping technology, analytical methods, and the establishment of large cohorts for population genetic studies have resulted in a large new body of information about the genetic basis of human rheumatoid arthritis (RA). Improved understanding of the root pathogenesis of the disease holds the promise of improved diagnostic and prognostic tools based upon this information. In this review, we summarize the nature of new genetic findings in human RA, including susceptibility loci and gene-gene and gene-environment interactions, as well as genetic loci associated with sub-groups of patients and those associated with response to therapy. Possible uses of these data are discussed, such as prediction of disease risk as well as personalized therapy and prediction of therapeutic response and risk of adverse events. While these applications are largely not refined to the point of clinical utility in RA, it seems likely that multi-parameter datasets including genetic, clinical, and biomarker data will be employed in the future care of RA patients.
Collapse
|
83
|
Sugihara T, Harigai M. Targeting Low Disease Activity in Elderly-Onset Rheumatoid Arthritis: Current and Future Roles of Biological Disease-Modifying Antirheumatic Drugs. Drugs Aging 2016; 33:97-107. [PMID: 26833350 PMCID: PMC4756046 DOI: 10.1007/s40266-015-0341-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Elderly rheumatoid arthritis (RA) is classified into two clinical subsets, elderly-onset RA (EORA) and younger-onset elderly RA. With the improvement of life expectancy in the general population and advent of the super-aging society, the number of patients with EORA is anticipated to increase. Both large and small joints are affected initially at onset, and individuals with early EORA have higher scores of disease activity and levels of acute-phase reactants than those with early younger-onset RA. EORA is a progressive disease similar to younger-onset RA. Tumor necrosis factor (TNF) inhibitors are equally or slightly less effective in elderly patients than in younger patients with RA, and disease duration may have a greater impact on disease outcomes than age. Evidence of non-TNF biological disease-modifying antirheumatic drug use in EORA is limited. TNF inhibitors may not increase the risk for infection in elderly patients any more than methotrexate; however, increasing age is an independent and strong risk factor for serious infections in patients with RA. Treatment choice in patients with EORA is strongly influenced by comorbidities, especially cardiovascular disease, chronic lung disease, and frailty. To prevent progression to irreversible geriatric syndromes, non-frail patients with EORA, who are aging successfully should undergo intensive treatment using the treat-to-target strategy, and pre-frail and frail patients with EORA should be treated with the aim of returning to a non-frail or pre-frail stage, respectively. An appropriate treatment strategy for EORA and younger-onset elderly RA should be developed in the next decade using a multi-disciplinary approach.
Collapse
Affiliation(s)
- Takahiko Sugihara
- Department of Medicine and Rheumatology, Tokyo Metropolitan Geriatric Hospital, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Masayoshi Harigai
- Department of Epidemiology and Pharmacoepidemiology, Institute of Rheumatology, Tokyo Women's Medical University, 10-22 Kawada-cho, Shinjuku-ku, Tokyo, 162-0054, Japan.
| |
Collapse
|
84
|
Erosions in the foot at baseline are predictive of orthopaedic shoe use after 10 years of treat to target therapy in patients with recent onset rheumatoid arthritis. Clin Rheumatol 2015; 35:2101-2107. [PMID: 26694056 PMCID: PMC4960271 DOI: 10.1007/s10067-015-3145-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/25/2015] [Accepted: 12/06/2015] [Indexed: 11/17/2022]
Abstract
The objective of this study is to investigate if foot joint damage due to rheumatoid arthritis (RA) can predict whether patients will start wearing orthopaedic shoes (OS) within 10 years after treatment start. Data from recent onset RA patients with 10 years follow-up from the BeSt (Dutch acronym for treatment strategies) study were used. Treatment was tightly controlled, targeted at disease activity score (DAS) ≤2.4, according to 1 of 4 pre-specified treatment strategies. After 10 years of follow-up, orthopaedic shoe use was recorded in 285/508 patients (responders to questionnaires at 10 years). Between-group differences for orthopaedic shoe users and non-users were calculated at baseline, after 10 years, and change scores over the 10-year period were calculated. Predictors for orthopaedic shoe use were identified by univariable and multivariable logistic regression analyses. Orthopaedic shoe use was reported by 57/285 patients after 10 years. Orthopaedic shoe users had more joint damage, joint swelling and pain in the feet already at baseline and after 10 years. At both time points, DAS of orthopaedic shoe users and non-users was similar. Multivariable logistic regression showed that dichotomized foot erosions score (cut-off ≥1 erosion) (OR 2.42), anti-citrullinated protein antibodies (ACPA) (OR 4.64) and DAS (OR 1.77) were independent predictors of orthopaedic shoe use. Despite intensive targeted treatment, 57/285 recent onset RA patients started using orthopaedic shoes over 10 year of follow-up. Presence of foot erosions at treatment start predicts orthopaedic shoe use after 10 years. The risk of orthopedic shoe use increased for ACPA-positive patients and for those with higher baseline disease activity.
Collapse
|
85
|
Markusse IM, Lems WF, Huizinga TW, Allaart CF. Response to ‘Feasibility of tailored treatment based on risk stratification in patients with early rheumatoid arthritis’ – authors’ reply. Arthritis Res Ther 2015; 17:171. [PMID: 26135890 PMCID: PMC4488129 DOI: 10.1186/s13075-015-0681-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
86
|
Vastesaeger N, Fautrel B, Smolen J. Response to ‘Feasibility of tailored treatment based on risk stratification in patients with early rheumatoid arthritis’. Arthritis Res Ther 2015; 17:166. [PMID: 26091916 PMCID: PMC4475331 DOI: 10.1186/s13075-015-0680-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/10/2015] [Indexed: 11/27/2022] Open
|
87
|
Fautrel B, Nab HW, Brault Y, Gallo G. Identifying patients with rheumatoid arthritis with moderate disease activity at risk of significant radiographic progression despite methotrexate treatment. RMD Open 2015; 1:e000018. [PMID: 26509051 PMCID: PMC4613151 DOI: 10.1136/rmdopen-2014-000018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 06/05/2015] [Accepted: 06/08/2015] [Indexed: 01/18/2023] Open
Abstract
Objectives To determine the baseline factors predictive of significant radiographic progression (SRP) in patients with moderately active rheumatoid arthritis (RA) despite receiving methotrexate (MTX). Methods Patients from the MTX arm of the Trial of Etanercept and Methotrexate with Radiographic Patient Outcomes (TEMPO) trial with sustained moderate RA (defined as ≥3.2 mean disease activity score in 28 joints ≤5.1 during the last 6 months of the first year) were analysed for SRP (mTSS >3.0 overall) after 2 and 3 years. Baseline predictors for SRP were identified by univariate and multivariate analyses. All variables shown to be significantly associated with SRP were categorised based on clinically relevant cut-offs and tertiles and were included in a matrix risk model. Results 228 patients were assigned MTX treatment, 210 patients were in the radiographic intention-to-treat population, and 96 of these had sustained moderate RA. SRP occurred in 25 (26%) and 33 (34%) patients after 2 and 3 years of MTX treatment, respectively. Univariate and multivariate analyses found that C reactive protein (CRP) and rheumatoid factor (RF) positivity at baseline were predictive of SRP after 2 and 3 years (p<0.05 for all). The matrix risk model showed that RF positivity and CRP levels >40 mg/L at baseline were significantly associated with SRP after 2 (p<0.05 for both; R2=0.24) and 3 years (p<0.05 for both; R2=0.22). The baseline erosion score was not found to be predictive of SRP. Conclusions Patients with sustained moderate RA despite receiving MTX treatment are at risk of SRP, with both RF positivity and high CRP levels shown to be predictive of this.
Collapse
Affiliation(s)
- B Fautrel
- UPMC-GRC 08 (EEMOIS) , Paris , France ; AP-HP, Pitie Salpetriere Hospital, Dept of Rheumatology , Paris , France
| | | | | | | |
Collapse
|
88
|
Tanaka Y, Harigai M, Takeuchi T, Yamanaka H, Ishiguro N, Yamamoto K, Ishii Y, Nakajima H, Baker D, Miyasaka N, Koike T. Prevention of joint destruction in patients with high disease activity or high C-reactive protein levels: Post hoc analysis of the GO-FORTH study. Mod Rheumatol 2015; 26:323-30. [PMID: 26471830 DOI: 10.3109/14397595.2015.1086041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES To assess the influence of golimumab dosage and disease activity on joint destruction in patients with active rheumatoid arthritis (RA) in the GO-FORTH study. METHODS Efficacy was compared among groups given basal methotrexate plus placebo, golimumab (50 mg), or golimumab (100 mg) with stratification by high (HDA) or moderate (MDA) baseline disease activity and by high or low baseline C-reactive protein (CRP). RESULTS Among HDA or high CRP patients, the mean change of the total Sharp score was 3.48 and 3.41 in the placebo group, 1.94 and 2.71 in the 50 mg group, and 0.39 and 1.15 in the 100 mg group, respectively. The percentage of progression-free patients with HDA or high CRP was 40.4% and 40.0%, 43.1% and 38.2%, and 69.8% and 61.5%, respectively. Among MDA or low CRP patients, both golimumab doses showed similar prevention of joint destruction. Among HDA or high CRP patients, a shorter disease duration and higher TSS/disease duration ratio were associated with joint destruction. CONCLUSION Both doses of golimumab (50 or 100 mg) prevented joint destruction in MDA or low CRP patients, but 100 mg was better for HDA or high CRP patients with a shorter disease duration or higher TSS/disease duration ratio.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- a The First Department of Internal Medicine, University of Occupational and Environmental Health Japan , Kitakyushu , Fukuoka , Japan
| | - Masayoshi Harigai
- b Department of Medicine and Rheumatology, Tokyo Medical and Dental University , Bunkyo-Ku , Tokyo , Japan
| | - Tsutomu Takeuchi
- c Department of Internal Medicine, Keio University , Shinjuku-Ku , Tokyo , Japan
| | - Hisashi Yamanaka
- d Institute of Rheumatology, Tokyo Women's Medical University , Shinjuku-Ku , Tokyo , Japan
| | - Naoki Ishiguro
- e Department of Orthopedic Surgery, Nagoya University, Graduate School and Faculty of Medicine , Nagoya , Japan
| | - Kazuhiko Yamamoto
- f Department of Allergy and Rheumatology, The University of Tokyo , Bunkyo-Ku , Tokyo , Japan
| | - Yutaka Ishii
- g Janssen Pharmaceutical K.K. , Chiyoda-Ku , Tokyo , Japan
| | | | - Daniel Baker
- h Janssen Research and Development, LLC , Spring House , PA , USA
| | - Nobuyuki Miyasaka
- i Department of Medicine and Rheumatology, Tokyo Medical and Dental University , Bunkyo-Ku , Tokyo , Japan , and
| | - Takao Koike
- j Sapporo Medical Center NTT EC , Sapporo , Japan
| |
Collapse
|
89
|
Li W, Sasso EH, van der Helm-van Mil AHM, Huizinga TWJ. Relationship of multi-biomarker disease activity score and other risk factors with radiographic progression in an observational study of patients with rheumatoid arthritis. Rheumatology (Oxford) 2015; 55:357-66. [PMID: 26385370 PMCID: PMC4710803 DOI: 10.1093/rheumatology/kev341] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Indexed: 12/12/2022] Open
Abstract
Objectives. To evaluate the multi-biomarker disease activity (MBDA) score as a predictor of radiographic progression and compare it with other risk factors among patients with established RA receiving non-biologic DMARDs. Methods. For 163 patients with RA, we assessed 271 visits for MBDA score (scale of 1−100), clinical data and subsequent 1-year radiographic progression (change in Sharp−van der Heijde score [SHS]). Scatter plot and non-parametric quantile regression curves evaluated the relationship between the MBDA score and change in SHS. Changes in joint space narrowing and erosions were compared among MBDA categories with Wilcoxon rank-sum tests. The ability of the MBDA score to independently predict progression was determined by multivariate models and cross-classification of MBDA score with other risk factors. Generalized estimating equation methodology was used in model estimations to adjust for same-patient visits, always ≥1 year apart. Results. Patient characteristics included 67% female, 66%/67% RF+/anti-CCP+; mean age 55 years, MBDA score 43 (moderate = 30−44); median disease duration 4.6 years, SHS 23. Radiographic progression was infrequent for low MBDA scores. Relative risk for progression increased continuously as the MBDA score increased, reaching 17.4 for change in SHS >5 with MBDA scores ≥60. Joint space narrowing and erosion progression were associated with MBDA score. MBDA score was associated with radiographic progression after adjustments for other risk factors. MBDA score significantly differentiated risk for progression when swollen joint count, CRP or DAS28–CRP was low, and among seropositive patients. Conclusion. MBDA score enhanced the ability of conventional risk factors to predict radiographic progression in patients with established RA receiving non-biologic DMARDs.
Collapse
Affiliation(s)
| | - Eric H Sasso
- Medical and Scientific Affairs, Crescendo Bioscience, South San Francisco, USA and
| | | | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
90
|
Audo R, Daien C, Papon L, Lukas C, Vittecoq O, Hahne M, Combe B, Morel J. Osteoprotegerin and tumor necrosis factor-related apoptosis-inducing ligand as prognostic factors in rheumatoid arthritis: results from the ESPOIR cohort. Arthritis Res Ther 2015. [PMID: 26220665 PMCID: PMC4518710 DOI: 10.1186/s13075-015-0705-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Introduction We previously reported that low ratio of osteoprotegerin (OPG) to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) was associated with Disease Activity Score in 28 joints (DAS28) remission at 6 months in patients with early rheumatoid arthritis (RA). Here, we aimed to evaluate the value of baseline OPG/TRAIL ratio in predicting clinical and radiological outcomes in patients with early RA in the ESPOIR cohort. Methods OPG and TRAIL serum concentrations were assessed in the ESPOIR cohort patients. Patients with definite RA were included in this study. Patients were excluded if they had high erosion score at baseline (>90th percentile) or received biological therapy during the first 2 years of follow-up. Data were analyzed by univariate analysis and multivariate logistic regression to predict 1-year DAS28 remission and 2-year radiographic disease progression. Results On univariate analysis of 399 patients, OPG/TRAIL ratio at baseline was significantly lower in patients with than without remission at 1 year (p = 0.015). On multivariate logistic regression including age, gender, body mass index and DAS28, low OPG/TRAIL ratio was independently associated with remission at 1 year (odds ratio 1.68 [95 % confidence interval 1.01–2.79]). On univariate analysis, high OPG/TRAIL ratio at baseline was associated with rapid progression of erosion at 2 years (p = 0.041), and on multivariate logistic regression including age, anti-citrullinated protein antibody positivity and C-reactive protein level, OPG/TRAIL ratio independently predicted rapid progression of erosion at 2 years. Conclusions OPG/TRAIL ratio at baseline was an independent predictor of 1-year remission and 2-year rapid progression of erosion for patients with early rheumatoid arthritis. Thus, OPG/TRAIL ratio could be included in matrix prediction scores to predict rapid radiographic progression. Further confirmation in an independent cohort is warranted.
Collapse
Affiliation(s)
- Rachel Audo
- Department of Rheumatology, Lapeyronie Hospital, Montpellier University, 371 avenue doyen Giraud, 34295, Montpellier, France. .,Montpellier University, 163 rue Auguste Broussonnet, 34000, Montpellier, France. .,Institut de Génétique Moléculaire de Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293, Montpellier, France.
| | - Claire Daien
- Department of Rheumatology, Lapeyronie Hospital, Montpellier University, 371 avenue doyen Giraud, 34295, Montpellier, France. .,Montpellier University, 163 rue Auguste Broussonnet, 34000, Montpellier, France. .,Institut de Génétique Moléculaire de Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293, Montpellier, France.
| | - Laura Papon
- Montpellier University, 163 rue Auguste Broussonnet, 34000, Montpellier, France. .,Institut de Génétique Moléculaire de Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293, Montpellier, France.
| | - Cédric Lukas
- Department of Rheumatology, Lapeyronie Hospital, Montpellier University, 371 avenue doyen Giraud, 34295, Montpellier, France. .,Montpellier University, 163 rue Auguste Broussonnet, 34000, Montpellier, France.
| | - Olivier Vittecoq
- Department of Rheumatology and CIC/CRB 1404, Rouen University Hospital, Inserm U 905, Institute for Research and Innovation in Biomedicine, 1, rue de Germont, 76031 Rouen, France.
| | - Michael Hahne
- Montpellier University, 163 rue Auguste Broussonnet, 34000, Montpellier, France. .,Institut de Génétique Moléculaire de Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293, Montpellier, France. .,Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Bernard Combe
- Department of Rheumatology, Lapeyronie Hospital, Montpellier University, 371 avenue doyen Giraud, 34295, Montpellier, France. .,Montpellier University, 163 rue Auguste Broussonnet, 34000, Montpellier, France. .,Institut de Génétique Moléculaire de Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293, Montpellier, France.
| | - Jacques Morel
- Department of Rheumatology, Lapeyronie Hospital, Montpellier University, 371 avenue doyen Giraud, 34295, Montpellier, France. .,Montpellier University, 163 rue Auguste Broussonnet, 34000, Montpellier, France. .,Institut de Génétique Moléculaire de Montpellier, CNRS-UMR 5535, 1919 Route de Mende, 34293, Montpellier, France.
| |
Collapse
|
91
|
Araujo GR, Vaz ER, Fujimura PT, Fonseca JE, de Lima LM, Canhão H, Venturini G, Cardozo KHM, Carvalho VM, Napimoga MH, Goulart LR, Gonçalves J, Ueira-Vieira C. Improved serological detection of rheumatoid arthritis: a highly antigenic mimotope of carbonic anhydrase III selected in a murine model by phage display. Arthritis Res Ther 2015; 17:168. [PMID: 26099944 PMCID: PMC4493817 DOI: 10.1186/s13075-015-0685-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 06/12/2015] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects around 1% of the human population worldwide. RA diagnosis can be difficult as there is no definitive test for its detection. Therefore, the aim of this study was to identify biomarkers that could be used for RA diagnosis. METHODS Sera from a collagen-induced arthritis mouse model were used to select potential biomarkers for RA diagnosis by phage display technology. In silico and in vitro analyses were performed to characterize and validate the selected peptides. Samples were classified into three groups: RA; two other immune-mediated rheumatic diseases (systemic lupus erythematosus (SLE) and ankylosing spondylitis (AS)); and healthy controls (HC). Enzyme-linked immunosorbent assay (ELISA) was carried out to determine antibody levels, and diagnostic parameters were determined by constructing receiver operating characteristic curves. Mass spectrometry and Western blot were performed to identify the putative autoantigen that was mimicked by a highly reactive mimotope. RESULTS After three rounds of selection, 14 clones were obtained and tested for immunoreactivity analysis against sera from RA and HC groups. The phage-fused peptide with the highest immunoreactivity (M12) was synthesized, and was able to efficiently discriminate RA patients from SLE, AS and HCs (p < 0.0001) by ELISA. The specificity and sensitivity of anti-M12 antibodies for RA diagnosis were 91 % and 84.3 %, respectively. The M12 peptide was identified as one that mimics a predicted antigenic site of the carbonic anhydrase III (CAIII) protein, a ubiquitous biomarker that has been identified in patients with other diseases. CONCLUSION M12 is the first peptide associated with the CAIII protein that may be used as an antigen for antibody detection to aid in RA diagnosis with high sensitivity and specificity.
Collapse
Affiliation(s)
- Galber Rodrigues Araujo
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, MG, Brazil.
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal.
| | - Emília Rezende Vaz
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Patricia Tiemi Fujimura
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - João Eurico Fonseca
- Rheumatology Research Unit, Institute of Molecular Medicine, Lisbon, Portugal.
- Rheumatology Department, Santa Maria Hospital, Lisbon Academic Medical Center, Lisbon, Portugal.
| | - Lucélia Maria de Lima
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Helena Canhão
- Rheumatology Research Unit, Institute of Molecular Medicine, Lisbon, Portugal.
- Rheumatology Department, Santa Maria Hospital, Lisbon Academic Medical Center, Lisbon, Portugal.
| | - Gabriela Venturini
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil.
| | | | | | - Marcelo Henrique Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Campinas, SP, Brazil.
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, MG, Brazil.
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA.
| | - João Gonçalves
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal.
- IMM - Institute of Molecular Medicine, Lisbon, Portugal.
| | - Carlos Ueira-Vieira
- Laboratory of Nanobiotechnology, Institute of Genetics and Biochemistry, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
92
|
Sanmartí R, García-Rodríguez S, Álvaro-Gracia JM, Andreu JL, Balsa A, Cáliz R, Fernández-Nebro A, Ferraz-Amaro I, Gómez-Reino JJ, González-Álvaro I, Martín-Mola E, Martínez-Taboada VM, Ortiz AM, Tornero J, Marsal S, Moreno-Muelas JV. 2014 update of the Consensus Statement of the Spanish Society of Rheumatology on the use of biological therapies in rheumatoid arthritis. ACTA ACUST UNITED AC 2015; 11:279-94. [PMID: 26051464 DOI: 10.1016/j.reuma.2015.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To establish recommendations for the management of patients with rheumatoid arthritis (RA) to serve as a reference for all health professionals involved in the care of these patients, and focusing on the role of available synthetic and biologic disease-modifying antirheumatic drugs (DMARDs). METHODS Consensual recommendations were agreed on by a panel of 14 experts selected by the Spanish Society of Rheumatology (SER). The available scientific evidence was collected by updating three systematic reviews (SR) used for the EULAR 2013 recommendations. A new SR was added to answer an additional question. The literature review of the scientific evidence was made by the SER reviewer's group. The level of evidence and the degree of recommendation was classified according to the Oxford Centre for Evidence-Based Medicine system. A Delphi panel was used to evaluate the level of agreement between panellists (strength of recommendation). RESULTS Thirteen recommendations for the management of adult RA were emitted. The therapeutic objective should be to treat patients in the early phases of the disease with the aim of achieving clinical remission, with methotrexate playing a central role in the therapeutic strategy of RA as the reference synthetic DMARD. Indications for biologic DMARDs were updated and the concept of the optimization of biologicals was introduced. CONCLUSIONS We present the fifth update of the SER recommendations for the management of RA with synthetic and biologic DMARDs.
Collapse
Affiliation(s)
- Raimon Sanmartí
- Servicio de Reumatología, Hospital Clínic de Barcelona, Barcelona, España.
| | | | | | - José Luis Andreu
- Servicio de Reumatología, Hospital Universitario Puerta de Hierro, Madrid, España
| | - Alejandro Balsa
- Servicio de Reumatología, Hospital Universitario La Paz, Madrid, España
| | - Rafael Cáliz
- Servicio de Reumatología, Hospital Universitario Virgen de las Nieves, Granada, España
| | - Antonio Fernández-Nebro
- Unidad de Gestión Clínica de Reumatología, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, España
| | - Iván Ferraz-Amaro
- Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife, España
| | - Juan Jesús Gómez-Reino
- Servicio de Reumatología, Hospital Clínico Universitario de Santiago, Santiago de Compostela, A Coruña, España
| | | | | | | | - Ana M Ortiz
- Servicio de Reumatología, Hospital Universitario de la Princesa, Madrid, España
| | - Jesús Tornero
- Servicio de Reumatología, Hospital Universitario de Guadalajara, Guadalajara, España
| | - Sara Marsal
- Servicio de Reumatología, Hospital Universitario Vall d́Hebron, Barcelona, España
| | - José Vicente Moreno-Muelas
- Servicio de Reumatología, Hospital Universitario Vall d́Hebron, Barcelona, España; Sociedad Española de Reumatología, Madrid, España
| |
Collapse
|
93
|
Kirino Y, Hama M, Takase-Minegishi K, Kunishita Y, Kishimoto D, Yoshimi R, Asami Y, Ihata A, Oba MS, Tsunoda S, Ohno S, Ueda A, Takeno M, Ishigatsubo Y. Predicting joint destruction in rheumatoid arthritis with power Doppler, anti-citrullinated peptide antibody, and joint swelling. Mod Rheumatol 2015; 25:842-8. [PMID: 25736364 DOI: 10.3109/14397595.2015.1026025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To determine combined evaluation of musculoskeletal ultrasonography (MSUS) and power Doppler (PD) signals, anti-citrullinated peptide antibody (ACPA), and other clinical findings improve the prediction of joint destruction in rheumatoid arthritis (RA). METHODS We performed a retrospective study of 331 RA patients (female n = 280 and male n = 51, mean age: 57.9 ± 13.2 years) who underwent MSUS from 2002 to 2012. Correlations with progression of joint destructions in 1,308 2nd and 3rd metacarpophalangeal (MCP) joints and various factors including PD signals of the same joints, clinical findings, age, disease duration at the study entry, gender, observation period, radiographic bone scores according to modified Sharp-van der Heijde methods, ACPA, and rheumatoid factor (RF) were analyzed in patient- and joint-based fashions, using univariate and multivariate logistic regression analyses and generalized linear mixed model. RESULTS Patients' characteristics were as follows: mean disease duration: 5.7 ± 7.5 years, observation period: 4.6 ± 2.6 years, RF positivity: 79.9%, and ACPA positivity: 77.5%. PD-positive 2nd and 3rd joints showed higher rate of joint destruction, especially in ACPA-positive patients. Moreover, PD-positive joints in ACPA-positive patients showed joint destruction even in joints without swelling. Multivariate analysis determined PD, swollen joint (SJ), observation period, basal radiographic bone scores, and ACPA as independent risks for joint destruction. CONCLUSION PD, SJ, basal radiographic bone scores, and ACPA are independent predictors for the joint destruction of 2nd and 3rd MCPs in RA; thus, considering these factors would be useful in daily practice.
Collapse
Affiliation(s)
- Yohei Kirino
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Maasa Hama
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Kaoru Takase-Minegishi
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Yosuke Kunishita
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Daiga Kishimoto
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Ryusuke Yoshimi
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Yukiko Asami
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Atsushi Ihata
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan.,b Department of Rheumatology and Infectious disease , Minami Kyosai Hospital, Yokohama Minami Kyosai Hospital , Yokohama, Kanagawa , Japan
| | - Mari S Oba
- c Department of Biostatistics and Epidemiology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Shinichiro Tsunoda
- d Division of Rheumatology, Department of Internal Medicine , Hyogo College of Medicine , Nishinomiya, Hyogo , Japan
| | - Shigeru Ohno
- e Center for Rheumatic diseases, Yokohama City University Medical Center , Yokohama, Kanagawa , Japan
| | - Atsuhisa Ueda
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Mitsuhiro Takeno
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| | - Yoshiaki Ishigatsubo
- a Department of Internal Medicine and Clinical Immunology , Yokohama City University Graduate School of Medicine , Yokohama, Kanagawa , Japan
| |
Collapse
|
94
|
de Punder YM, van Riel PL, Fransen J. A Simplified Baseline Prediction Model for Joint Damage Progression in Rheumatoid Arthritis: A Step toward Personalized Medicine. J Rheumatol 2015; 42:391-7. [DOI: 10.3899/jrheum.140327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Objective.To compare the performance of an extended model and a simplified prognostic model for joint damage in rheumatoid arthritis (RA) based on 3 baseline risk factors: anticyclic citrullinated peptide antibodies (anti-CCP), erosions, and acute-phase reaction.Methods.Data were used from the Nijmegen early RA cohort. An extended model and a simplified baseline prediction model were developed to predict joint damage progression between 0 and 3 years. Joint damage progression was assessed using the Ratingen score. In the extended model, prediction factors were positivity for anti-CCP and/or rheumatoid factor, the level of erythrocyte sedimentation rate, and the quantity of erosions. The prediction score was calculated as the sum of the regression coefficients. In the simplified model, the prediction factors were dichotomized and the number of risk factors was counted. Performances of both models were compared using discrimination and calibration. The models were internally validated using bootstrapping.Results.The extended model resulted in a prediction score between 0 and 5.6 with an area under the receiver-operation characteristic (ROC) curve of 0.77 (95% CI 0.72–0.81). The simplified model resulted in a prediction score between 0 and 3. This model had an area under the ROC curve of 0.75 (95% CI 0.70–0.80). In internal validation, the 2 models showed reasonably well the agreement between observed and predicted probabilities for joint damage progression (Hosmer-Lemeshow test p > 0.05 and calibration slope near 1.0).Conclusion.A simple prediction model for joint damage progression in early RA, by only counting the number of risk factors, has adequate performance. This facilitates the translation of the theoretical prognostic models to daily clinical practice.
Collapse
|
95
|
de Punder YM, Jansen TL, van Ede AE, den Broeder AA, van Riel PL, Fransen J. Personalizing Treatment Targets in Rheumatoid Arthritis by Using a Simple Prediction Model. J Rheumatol 2015; 42:398-404. [DOI: 10.3899/jrheum.140085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective.To develop a personalized treatment target approach in patients with rheumatoid arthritis (RA) based on baseline risk factors for joint damage progression in combination with disease activity over time.Methods.Data were used from the Nijmegen early RA cohort. Presence or absence of anticyclic citrullinated peptide antibodies (anti-CCP), high erythrocyte sedimentation rate, and erosions were translated into 4 risk profiles: 0, 1, 2, and 3. Joint damage progression was assessed with the Ratingen score, and disease activity with the original Disease Activity Score (DAS) over 3 years. The probability for joint damage progression was calculated for each risk profile and each DAS category using logistic regression models. The probabilities were translated into personalized disease activity treatment targets.Results.More risk factors at baseline as well as a higher DAS level resulted in a higher probability for joint damage progression in a dose-dependent way. Low DAS corresponded with a probability of 0.0, 0.08, 0.20, and 0.58 in patients with 0, 1, 2, and 3 risk factors, respectively. Moderate DAS corresponded with a probability of 0.06 in patients with 0 risk factors and 0.35 with 1 risk factor. High DAS resulted in a probability of 0.50 with no risk factors present at baseline.Conclusion.Presence of anti-CCP, acute-phase response, and erosions at baseline can be used to set individual treatment targets in RA. In patients without these risk factors, a moderate DAS as a target is sufficient, while for patients with all 3 risk factors, a low DAS is not strict enough to limit the risk for joint damage.
Collapse
|
96
|
Bykerk VP. Radiographic progression in rheumatoid arthritis: does it still happen and does it matter? J Rheumatol 2014; 41:2337-9. [PMID: 25452176 DOI: 10.3899/jrheum.141133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Vivian P Bykerk
- Department of Rheumatology, Hospital for Special Surgery, 535 East 70th St., New York, New York 10021, USA
| |
Collapse
|
97
|
Michaud TL, Rho YH, Shamliyan T, Kuntz KM, Choi HK. The comparative safety of tumor necrosis factor inhibitors in rheumatoid arthritis: a meta-analysis update of 44 trials. Am J Med 2014; 127:1208-32. [PMID: 24950486 DOI: 10.1016/j.amjmed.2014.06.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/22/2014] [Accepted: 06/09/2014] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The study objective was to evaluate and update the safety data from randomized controlled trials of tumor necrosis factor inhibitors in patients treated for rheumatoid arthritis. METHODS A systematic literature search was conducted from 1990 to May 2013. All studies included were randomized, double-blind, controlled trials of patients with rheumatoid arthritis that evaluated adalimumab, certolizumab pegol, etanercept, golimumab, or infliximab treatment. The serious adverse events and discontinuation rates were abstracted, and risk estimates were calculated by Peto odds ratios (ORs). RESULTS Forty-four randomized controlled trials involving 11,700 subjects receiving tumor necrosis factor inhibitors and 5901 subjects receiving placebo or traditional disease-modifying antirheumatic drugs were included. Tumor necrosis factor inhibitor treatment as a group was associated with a higher risk of serious infection (OR, 1.42; 95% confidence interval [CI], 1.13-1.78) and treatment discontinuation due to adverse events (OR, 1.23; 95% CI, 1.06-1.43) compared with placebo and traditional disease-modifying antirheumatic drug treatments. Specifically, patients taking adalimumab, certolizumab pegol, and infliximab had an increased risk of serious infection (OR, 1.69, 1.98, and 1.63, respectively) and showed an increased risk of discontinuation due to adverse events (OR, 1.38, 1.67, and 2.04, respectively). In contrast, patients taking etanercept had a decreased risk of discontinuation due to adverse events (OR, 0.72; 95% CI, 0.55-0.93). Although ORs for malignancy varied across the different tumor necrosis factor inhibitors, none reached statistical significance. CONCLUSIONS These meta-analysis updates of the comparative safety of tumor necrosis factor inhibitors suggest a higher risk of serious infection associated with adalimumab, certolizumab pegol, and infliximab, which seems to contribute to higher rates of discontinuation. In contrast, etanercept use showed a lower rate of discontinuation. These data may help guide clinical comparative decision making in the management of rheumatoid arthritis.
Collapse
Affiliation(s)
- Tzeyu L Michaud
- Division of Health Policy and Management, School of Public Health, University of Minnesota, Minneapolis
| | - Young Hee Rho
- Section of Rheumatology and the Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Mass
| | - Tatyana Shamliyan
- Evidence-Based Medicine Quality Assurance Elsevier, Clinical Solutions, Philadelphia, PA
| | - Karen M Kuntz
- Division of Health Policy and Management, School of Public Health, University of Minnesota, Minneapolis
| | - Hyon K Choi
- Section of Rheumatology and the Clinical Epidemiology Unit, Boston University School of Medicine, Boston, Mass.
| |
Collapse
|
98
|
Cuchacovich M, Bueno D, Carvajal R, Bravo N, Aguillón JC, Catalán D, Soto L. Clinical parameters and biomarkers for anti-TNF treatment prognosis in rheumatoid arthritis patients. Clin Rheumatol 2014; 33:1707-14. [PMID: 25085274 DOI: 10.1007/s10067-014-2756-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/20/2014] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor (TNF) plays a pivotal role in the pathogenesis of rheumatoid arthritis (RA). This finding has led to the development of TNF blockers for RA treatment. However, response to these therapies is heterogeneous with success in only two thirds of patient. Some clinical aspects useful in the attempt to predict the response to TNF inhibitors is the promptness and the magnitude of the response at the first weeks and a low basal disease activity, while comorbidities, tobacco, glucocorticoids treatment, and high basal radiological score correlate with a poorer response. The role of TNF promoter polymorphisms in clinical response to anti-TNF therapies is controversial. A correlation between the presence of high baseline titers of rheumatoid factor (RF) and decreased response to anti-TNF treatment has been reported. Most studies show decreased RF titers during anti-TNF treatment mainly in patients who responded to treatment. There is no consensus about the usefulness of basal anti-citrullinated protein antibodies (ACPA) levels, and a decrease in ACPA titers as predictor of clinical response to anti-TNF therapy. Despite some promising markers identified to fulfill this role, currently the predictive value of single markers seems not strong enough to predict treatment response in an individual RA patient.
Collapse
Affiliation(s)
- Miguel Cuchacovich
- Department of Medicine, Clinical Hospital University of Chile, San Pío X, N 2460, Off 607, Providencia, Santiago, Chile,
| | | | | | | | | | | | | |
Collapse
|
99
|
Can we prevent rapid radiological progression in patients with early rheumatoid arthritis? Clin Rheumatol 2014; 34:163-6. [PMID: 25431327 DOI: 10.1007/s10067-014-2815-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 08/31/2014] [Accepted: 10/23/2014] [Indexed: 10/24/2022]
Abstract
The aim of this study is to test the performance of a matrix model to predict rapid radiological progression (RRP) in a study population of early rheumatoid arthritis (RA) or undifferentiated arthritis (UA) patients. A matrix model using baseline CRP, erosion score, autoantibody status, and initial treatment choice to predict RRP (increase ≥5 points in Sharp-van der Heijde score (SHS) in 1 year) was derived from the BeSt study where patients with active RA (1987-criteria) were treated with initial monotherapy or combination therapy, aiming at low disease activity. In the IMPROVED study, patients with early RA (2010 criteria) and UA were initially treated with methotrexate and prednisone aiming at remission. A receiver operating characteristics (ROC) curve was used to assess the discriminative value of the model to predict damage progression in the IMPROVED population. Four hundred thirty-one out of 479 patients with RA and 106/122 with UA could be categorized as high, intermediate, low, or very low risk for RRP. One patient, with a very low risk profile, showed RRP. Thirty-two other patients (5 %) showed radiological progression ≥0.5 point SHS; none had a high risk profile and 22 had a very low risk profile. The area under the curve (AUC) of the ROC curve was 0.56 (95% CI 0.45; 0.68). A matrix model predicting RRP based on risk factors identified in recent onset active RA according to the 1987-criteria performed poorly in recent onset RA (2010 criteria) and UA. It appears that known risk factors for damage progression lose their impact with early remission steered treatment, so that RRP might be considered a phenomenon of the past.
Collapse
|
100
|
Navarro-Compán V, Gherghe AM, Smolen JS, Aletaha D, Landewé R, van der Heijde D. Relationship between disease activity indices and their individual components and radiographic progression in RA: a systematic literature review. Rheumatology (Oxford) 2014; 54:994-1007. [PMID: 25416711 DOI: 10.1093/rheumatology/keu413] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the relationship between different disease activity indices (DAIs) and their individual components and radiographic progression in patients with RA. METHODS A systematic literature review until July 2013 was performed by two independent reviewers using the Medline and Embase databases. Longitudinal studies assessing the relationship between DAIs and single instruments and radiographic progression were included. The results were grouped based on the means of measurement (baseline vs time integrated) and analysis (univariable or multivariable). RESULTS Fifty-seven studies from 1232 hits were included. All published studies that assessed the relationship between any time-integrated DAI including joint count and radiographic progression reached a statistically significant association. Among the single instruments, only swollen joint count and ESR were associated with radiographic progression, while no significant association was found for tender joint count. Data with respect to CRP are conflicting. Data on patient's global health, pain assessment and evaluator's global assessment are limited and do not support a positive association with progression of joint damage. CONCLUSION Published data indicate that all DAIs that include swollen joints are related to radiographic progression while, of the individual components, only swollen joints and acute phase reactants are associated. Therefore composite DAIs are the optimal tool to monitor disease activity in patients with RA.
Collapse
Affiliation(s)
- Victoria Navarro-Compán
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands
| | - Ana Maria Gherghe
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands
| | - Josef S Smolen
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands
| | - Daniel Aletaha
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands
| | - Robert Landewé
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands
| | - Désirée van der Heijde
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands, Department of Rheumatology, University Hospital La Paz, Madrid, Spain, Department of Rheumatology and Internal Medicine, Cantacuzino Clinical Hospital, Bucharest, Romania, Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Second Department of Medicine, Hietzing Hospital, Vienna, Austria and Amsterdam Rheumatology Center, Amsterdam, and Atrium Medical Center, Heerlen, The Netherlands
| |
Collapse
|