Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

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

  • Tomohiro Koga ,

    tkoga@nagasaki-u.ac.jp

    Affiliations 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

  • Akitomo Okada,

    Affiliation Japanese Red Cross Nagasaki Genbaku Hospital, Department of Rheumatology, Mori-machi 3–15, Nagasaki, Japan

  • Takaaki Fukuda,

    Affiliation Kurume University Medical Center, Department of Rheumatology, Kokubun 155–1, Kurume, Japan

  • Toshihiko Hidaka,

    Affiliation Zenjinkai Shimin-no-Mori Hospital, Shioji 2783–37, Miyazaki, Japan

  • Tomonori Ishii,

    Affiliation Tohoku University Hospital, Department of Hematology and Rheumatology, Aoba-ku, seiryo 1–1, Sendai, Japan

  • Yukitaka Ueki,

    Affiliation Sasebo Chuo Hospital, Rheumatic and Collagen Disease Center, Yamato 15, Sasebo, Japan

  • Takao Kodera,

    Affiliation Tohoku Medical and Pharmaceutical University Hospital, Aoba-ku, komatsujima 4-4-1, Sendai, Japan

  • Munetoshi Nakashima,

    Affiliations 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

  • Yuichi Takahashi,

    Affiliation Yu Family Clinic, Rifu-aza sinkan 2–5, Miyagi, Japan

  • Seiyo Honda,

    Affiliation Kurume University School of Medicine, asahi-machi 67, Kurume, Japan

  • Yoshiro Horai,

    Affiliation Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan

  • Ryu Watanabe,

    Affiliation Tohoku University Hospital, Department of Hematology and Rheumatology, Aoba-ku, seiryo 1–1, Sendai, Japan

  • Hiroshi Okuno,

    Affiliation Tohoku University Hospital, Department of Orthopaedic Surgery, Hirase 9–3, Sasebo, Japan

  • Toshiyuki Aramaki,

    Affiliations 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,

    Affiliation East Sendai Rheumatism and internal medicine Clinic, Miyagino, Nittahigashi 1-17-5, Sendai, Japan

  • Osamu Takai,

    Affiliation Osaki Citizen Hospital, Furukawa-honami 3-8-1, Osaki, Japan

  • Taiichiro Miyashita,

    Affiliation NHO Nagasaki Medical Center, Kubara 2-1001-1, Omura, Nagasaki Japan

  • Shuntaro Sato,

    Affiliation Clinical Research Center, Nagasaki University Hospital, Sakamoto 1-7-1, Nagasaki, Japan

  • Shin-ya Kawashiri,

    Affiliations 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,

    Affiliation Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan

  • Kunihiro Ichinose,

    Affiliation Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan

  • Mami Tamai,

    Affiliation Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan

  • Tomoki Origuchi,

    Affiliation Department of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-12-4, Nagasaki, Japan

  • Hideki Nakamura,

    Affiliation Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan

  • Kiyoshi Aoyagi,

    Affiliation Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-12-4, Nagasaki, Japan

  • Katsumi Eguchi,

    Affiliation Sasebo City General Hospital, Hirase 9–3, Sasebo, Japan

  • Atsushi Kawakami,

    Affiliation Unit of Translational Medicine, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan

  •  [ ... ],
  • the Japanese RA Patients with RRP Study Group

    The complete membership of the author group can be found in the Acknowledgments.

  • [ view all ]
  • [ view less ]

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.

Introduction

Rheumatoid arthritis (RA) is a systemic inflammatory disease characterized by autoimmune disorder and progressive joint destruction, leading to impaired quality of life [1, 2]. The therapeutic strategies for RA have developed remarkably, and the treat-to-target (T2T) strategy described in the European League Against Rheumatism (EULAR) recommendations is to aim for remission or low disease activity (LDA) [3]. However, some RA patients develop clinically relevant radiographic progression (CRRP) despite the achievement of remission or LDA by the T2T strategy with disease-modifying antirheumatic drug (DMARDs) [4].

We suspect that the development of CRRP is due to subclinical/residual synovitis. A number of previous studies attempted to identify promising prognostic markers of CRRP. Various clinical and biological markers including C-reactive protein (CRP) at baseline, erosion score at baseline, and the presence of autoantibodies have been identified as risk factors for CRRP in RA patients with high disease activity [58]. Importantly, the achievement of clinical remission defined by, for example, the Disease Activity Score in 28 joints (DAS28), the Simplified Disease Activity Index (SDAI) and the Clinical Disease Activity Index (CDAI) is not always associated with good structural and functional outcomes [9, 10]. In addition, subclinical synovitis or residual synovitis is well recognized in patients with RA and has been demonstrated by ultrasonography (US) [1113] and MRI [14].

A Japanese Institute of Rheumatoid Arthritis (IORRA) observational study showed that more than one-half of real-world RA patients achieved clinical remission or LDA [15]. In daily practice it would be very useful to have prognostic markers for CRRP in 'good responders' among RA patients. However, little is known about variables that could be used to predict CRRP among RA patients who achieve clinical remission or LDA with DMARDs, especially conventional synthetic DMARDs (csDMARDs).

In the present study, using data from a nationwide, multicenter, prospective study in Japan, we evaluated a large number of clinical variables for their ability to predict the development of joint damage as CRRP after 1 year in RA patients who achieved remission or LDA with csDMARDs.

Methods

Patients

We performed a secondary analysis using data from a prospective, observational cohort study registered with the University Hospital Medical Information Network Clinical Trials Registry (UMIN-CTR) [http://www.umin.ac.jp/ctr/] (#UMIN000014791), conducted in daily clinical practices for RA in Japan. Overall, 887 RA patients from 26 centers affiliated with Nagasaki University or Tohoku University in Japan were recruited as the study cohort between May 2009 and March 2012. All of the patients were examined and treated by Japan College of Rheumatology (JCR)-certified rheumatologists.

Using this observational cohort, we recently reported prognostic factors for CRRP in RA patients whose clinical disease activity was moderate to high at enrollment [16]. In this second investigation, we focused on the prognostic factors for CRRP in 198 RA patients who achieved remission or LDA at enrollment.

When a patient relapsed during the present study, one of the participating JCR-certified rheumatologists treated the patient by using a T2T strategy that included the use of bDMARDs. We observed all of the patients for 1 year after their respective enrollment and assessed their RA disease activity every 3 months, using the DAS28-ESR and the Health Assessment Questionnaire (HAQ) [17]. The cumulative inflammatory burden was estimated by the time-integrated values (area under the curve-AUC) of DAS28-ESR. We constructed plots of longitudinal data of DAS28-ESR (baseline, 3 month, 6 month, 9 month and 12 month after enrollment) and calculated the area under the curve as described previously [18, 19].

All patients gave their signed informed consent to be subjected to the protocol, which was approved by the Institutional Review Board of Nagasaki University (approval number: 10022570–2), Tohoku University and the related centers.

Structural damage assessment

To evaluated the patients' structural damage, radiographs of each patient's hands and feet were obtained at baseline and at 1 year, and the images were evaluated by two independent rheumatologists blinded to the clinical evaluation, using the van der Heijde-mTSS system as described [16, 20]. In our 2016 study [16], the interobserver reliability (as determined by the interclass correlation coefficient) was 0.97, and the smallest detectable change of mTSS in the present study was calculated as 2.96 as described [21]. Accordingly, we defined an annual increase of the mTSS >3.0 units as the development of CRRP, as was done in a previous study [22].

Statistical analysis

The baseline demographic, clinical, and radiographic characteristics of the RA patients with and without CRRP were compared with Fisher's exact test for discrete variables, and with Wilcoxon’s test for continuous variables. To determine the independent predictive factors of the development of CRRP at 1 year, we performed a multiple logistic regression analysis. We selected variables with p-values <0.05 by univariate analyses as model 1. We then determined the final model by selecting the variables with p-values <0.05 in model 1. We also performed subgroup analysis based on disease activity at enrollment (remission vs. LDA). Each subgroup was analyzed with a multiple logistic regression analysis by selecting variables we used the final model. To convert continuous variables to binary variables, we defined the cut-off values by constructing a receiver operator characteristic (ROC) curve. Since treatment is an important confounder for radiographic progression, we performed a sensitivity analysis by removing patients treated with bDMARDs. Separation occurs in subgroup analysis because a contingency table of CRRP and ACPA had a zero cell. Separation leads diverges to infinity of estimated parameters. To solve this problem, we performed a Firth’s bias-reduced penalized-likelihood logistic regression [23]. This method is a modified logistic regression which can obtain estimates which do not diverge even in separation. The statistical analyses were performed using SAS 9.4 software (SAS Institute, Cary, NC, USA). A p-value <0.05 was considered significant.

Results

Patients

We analyzed 198 patients with remission or LDA at enrollment in this study. The patients’ characteristics are shown in Table 1. Their mean age was 58.8 years, and the median disease duration was 5.1 years. The mean DAS28-ESR at baseline was 2.33. Among the 198 patients, the treatment of 67 patients (34%) was strengthened with one or more csDMARDs or bDMARDs according to the EULAR recommendation, as decided by the participating rheumatologists. For eight patients, the bDMARDs (adalimumab [ADA], n = 1; etanercept [ETA], n = 2; infliximab [IFX], n = 2; tocilizumab [TCZ], n = 1; and abatacept [ABT], n = 2) were initiated within 3 months after the enrollment. In contrast, there were five patients (3%) whose treatment was weakened during the 1 year.

thumbnail
Table 1. Association between baseline characteristics and CRRP (univariate analyses)*.

https://doi.org/10.1371/journal.pone.0175281.t001

The therapeutic course during the 1 year following the baseline is shown in Fig 1. CRRP was observed in 15 of the 198 patients (7.6%). Among the 198 patients, 142 (72%) achieved sustained clinical remission or LDA during the 1-year observation. Cumulative probability plots during the 1 year post-baseline as assessed by mTSS are shown in Fig 2.

thumbnail
Fig 1. Patient enrollment flow chart and the therapeutic course during the 1-year observation after the baseline in our RA cohort.

HDA: high disease activity; MDA: moderate disease activity; LDA: low disease activity; csDMARDs: conventional synthetic disease-modifying antirheumatic drugs; bDMARDs: biological disease-modifying antirheumatic drugs.

https://doi.org/10.1371/journal.pone.0175281.g001

thumbnail
Fig 2. Cumulative probability plots of actual radiographic progression assessed by mTSS (U/year) in the cohort (n = 198).

mTSS: modified total Sharp score.

https://doi.org/10.1371/journal.pone.0175281.g002

Prediction of CRRP at 1 year in the RA patients

To determine which variables are associated with the development of CRRP at 1 year, we evaluated the 21 variables shown in Table 1. We found that the following eight variables were significantly associated with CRRP in the univariate analyses: ACPA positivity, time-integrated DAS28-ESR during the 1 year post-baseline, relapse during the follow-up, the CRP level at baseline, the HAQ score at baseline, the total mTSS at baseline, the erosion score at baseline, and the joint space narrowing score at baseline.

We selected these variables for a logistic regression analysis and determined the final model by selecting the variables with p-values <0.05 in the first model and identified three independent prognostic factors of CRRP, as follows: ACPA positivity (odds ratio [OR] = 15.2, 95% confidence interval [CI] 2.64–299, p = 0.0007), time-integrated DAS28-ESR during the 1 year post-baseline (7.85-unit increase, OR = 1.83, 95%CI 1.03–3.45, p = 0.036), and mTSS at baseline (13-unit increase, OR = 1.22, 95%CI 1.06–1.42, p = 0.0073) (Table 2). We also show logistic analyses with all variables converted to continuous variables (Table 3) or binary variables (Table 4). These binary variables are determined by constructing ROC curves. Taken together, we determined that ACPA positivity, time-integrated DAS28-ESR during the 1 year post-baseline and mTSS at baseline are independent prognostic factors of CRRP. The cumulative probability plots of changes in mTSS for 1 year in the ACPA-positive patients (n = 113) versus the ACPA-negative patients (n = 85) are shown in Fig 3.

thumbnail
Fig 3. Cumulative probability plots of changes in mTSS for 1 year in the ACPA-positive patients versus the ACPA-negative patients.

ACPA: anti-citrullinated peptide antibodies; mTSS: modified total Sharp score.

https://doi.org/10.1371/journal.pone.0175281.g003

thumbnail
Table 2. Prediction of CRRP in multiple logistic regression analysis*.

https://doi.org/10.1371/journal.pone.0175281.t002

thumbnail
Table 3. Prediction of CRRP in multiple logistic regression analysis (continuous variables)*.

https://doi.org/10.1371/journal.pone.0175281.t003

thumbnail
Table 4. Prediction of CRRP in multiple logistic regression analysis (binary variables)*.

https://doi.org/10.1371/journal.pone.0175281.t004

Since several studies have demonstrated better radiographic response of bDMARDs above csDMARDs, we performed a sensitivity analysis in which patients treated with bDMARDs are omitted, showing that all of the results did not change significantly after the exclusion (data not shown).

Prediction of CRRP at 1 year post-baseline in subgroups defined by disease activity

To further examine predictors of radiographic progression in the absence of inflammatory activity for its potential clinical utility, we stratified patients into two groups according to their disease activity at entry: remission (n = 120) or LDA (n = 78). The patients’ characteristics according to disease activity are shown in Table 5. There was no significant difference in the percentage of CRRP between the two groups (remission, 6%; LDA, 10%; p = 0.28).

thumbnail
Table 5. Patients’ characteristics with disease activity (univariate analyses)*.

https://doi.org/10.1371/journal.pone.0175281.t005

We subsequently performed a Firth’s bias-reduced penalized-likelihood logistic regression analysis by using the final model created in this study. In the group with remission, ACPA positivity at baseline (OR = 13.3, 95%CI 1.46–176, p = 0.017), and mTSS at baseline (11-unit increase, OR = 1.27, 95%CI 1.02–1.64, p = 0.0316) are independent variables to predict the development of CRRP. Especially, all of the patients who developed CRRP had positive ACPA at baseline. Meanwhile, we found that ACPA positivity at baseline (OR = 8.10, 95%CI 1.14–181, p = 0.0345) was the only variable for predicting CRRP in the group with LDA at enrollment. Taken together, our results show that the prognostic factors of CRRP differ according to the disease activity.

Discussion

Although achieving clinical remission is a primary goal of RA, it should be noted that some RA patients in remission experience joint destruction. In the present study we sought to determine the predictive factors for CRRP in bDMARD-naïve RA patients who achieved remission or LDA by treatment with conventional DMARDs, and our analyses revealed that ACPA positivity was the strongest predictor of CRRP among these patients.

Studies that investigated early RA patients with high disease activity revealed that autoantibodies such as ACPA and rheumatoid factor (RF) were predictive factors for CRRP [7, 8, 24]. However, little is known the effect of autoantibodies on CRRP in patients with RA who achieve remission or LDA. Our present findings demonstrated a direct association between ACPA positivity and CRRP among Japanese RA patients in real-world daily practices, which suggests that physicians should consider the possibility of subclinical or residual synovitis in RA patients with ACPA positivity even if the patients have achieved remisson by csDMARD treatment.

Our subgroup analysis showed that ACPA positivity is a potent prognostic factor toward CRRP among patients with remission compared with those with LDA. Of note, a systematic review and meta-analysis reported that the presence of synovial hypertrophy on US, power Doppler activity or synovitis shown by MRI at baseline in RA patients in clinical remission were each significantly associated with structural progression at 1 year, even in asymptomatic joints [25]. Thus, it is important for clinicians to assess subclinical/residual synovitis by using MRI or US in ACPA-positive patients who achived remission.

Schett et al. recently reviewed the predictors of relapse in RA patients in clinical remission during the tapering of biologic and conventional DMARDs [26]. With respect to serum biomarkers, the best-studied predictor of relapse to date is ACPA positivity, and the second is RF positivity [26]. The clinical situations are different between this review and our cohort since we did not intend to taper or stop the DMARDs; however, ACPA is thought to be an indispensable biomarker of unfavorable outcome regardless of continuing, tapering or stopping DMARDs in RA patients with clinical remission or LDA. In fact, in the present study the rate of relapse during the 1-year observation was 35% in the ACPA-positive patients and 19% in the ACPA-negative patients (p = 0.011).

In the present study RF, unlike ACPA, did not predict the development of CRRP. It is well known that RF positivity, unlike that of ACPA, can be changed to negativity by DMARD treatment [27, 28]. The ACPA and RF serostatus in our patients was obtained at the latest data collection, and thus the serostatus of RF, but not that of ACPA, might have been influenced by the introduction of DMARDs and might not accurately reflect the immunological activities of the patients.

The multiple logistic regression analysis revealed that time-integrated DAS28-ESR during the 1 year post-baseline was also significantly associated with CRRP. This finding is consistent with the previous report that radiographic progression was prominent in the subgroup of patients with disease exacerbation in a cohort of European RA patients in clinical remission at enrollment [9]. Our data also confirm the importance of the maintenance of favorable clinical disease activity achieved by the T2T strategy in the EULAR recommendations [3, 29].

Our group [16] and others [24, 30] reported that the CRP level at baseline is a predictive factor for CRRP in RA patients with moderate to high disease activity. In contrast, the present investigation of RA patients in remission or with LDA showed that the CRP level at baseline was not associated with CRRP. Taken together, these findings indicated that the CRP level at baseline is a predictive factor for CRRP only in RA patients with obvious disease activity, since the patients' CRP levels at enrollment were considerably low (median 0.15 mg/dl). In addition, joint damage at baseline was reported to be a significant predictor of CRRP in RA patients with high disease activity [7, 8]. In line with these observations, the multiple logistic regression analysis in the present study revealed that the mTSS at baseline was an independent variable to predict CRRP; this finding reconfirms that joint damage sustains joint inflammation [31]. These processes might be more apparent in RA patients in remission or with LDA compared to patients with moderate to high disease activity, because our present findings demonstrated that mTSS but not CRP at baseline is predictive of CRRP.

There are some study limitations to acknowledge. First, the follow-up period was only 1 year. Since the structural damage in RA progresses over several years, long-term verification studies are needed to confirm our results. Second, although Takahashi et al. reported that Japanese patients with RA had higher concentrations of the active form of methotrexate (MTX)-polyglutamate in their red blood cells than RA patients in other counties [32], the dosage of MTX in our cohort was lower than that in the Takahashi cohort. Accordingly, we cannot exclude the possibility that the efficacy of MTX was underestimated in the present study. Finally, we were unable to investigate the period during which our patients achieved remission or LDA prior to their entry in the study. Recruited patients were in remission at the time of this entry, but we did not have detailed information on the recent treatment change and the clinical course before the start of observation. There is a possibility that strengthening treatment just before starting observation have influenced the outcome of this study.

In conclusion, our findings indicate that CRRP in patients with rheumatoid arthritis in remission or LDA was associated with ACPA positivity, mTSS at baseline and time-integrated DAS28-ESR, the strongest predictor being ACPA. Since structural damage is strongly associated with functional impairment in RA patients [33], our results offer important information that can be used to help avoid radiographic progression in RA patients who have achieved a favorable clinical response with DMARD treatment.

Acknowledgments

The authors thank all the Japanese RA Patients with RRP Study Group members (a lead author: Atsushi Kawakami, e-mail: atsushik@nagasaki-u.ac.jp) include, in alphabetical order of their affiliations: Tomomasa Izumiyama (East Sendai Rheumatism and internal medicine Clinic, Sendai); Yasuhiko Hirabayashi (Hikarigaoka Supellman Hospital, Sendai); Takemasa Matsuda (Japanese Red Cross Kagoshima Hospital); Munetoshi Nakashima (Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki); Toshiaki Tsukada (JCHO Isahaya General Hospital, Isahaya); Michishi Tsukano (Kumamoto Orthopaedic Hospital, Kumamoto); Akira Rikimaru (Kurokawa Hospital, Kurokawa); Hiroaki Ida (Kurume University Hospital); Takaaki Fukuda (Kurume Univercity Medical Center); Naoki Matsuoka (Nagasaki Medical Hospital of Rheumatology); Atsushi Kawakami (Nagasaki University Hospital, Nagasaki); Eiichi Suematsu (NHO Kyushu Medical Center, Fukuoka); Kiyoshi Migita (NHO Nagasaki Medical Center, Omura); Youjirou kawabe (NHO Ureshino Medical Center, Ureshino); Motohiro Oribe (Oribe Rheumachika Naika Clinic, Oita); Osamu Takai (Osaki Citizen Hospital, Osaki); Yukio Sekiguchi (NTT East Tohoku Hospital); Yukitaka Ueki (Sasebo Chuo Hospital, Sasebo); Katsumi Eguchi (Sasebo City General Hospital, Sasebo); Yasuhiko Munakata (Sendai Taihaku Hospital, Sendai); Masanobu Mine (Suga Orthopaedic Hospital, Isahaya); Takao Kodera (Tohoku Pharmaceutical University Hospital, Sendai); Akira Hatayama (Tohoku Rosai Hospital, Sendai); Ken Ogura (Tohoku University School of Medicine, Department of Orthopaedic Surgery, Sendai); Yuichi Takahashi (Yu Family Clinic, Miyagi); Toshihiko Hidaka (Zenjinkai Shimin-no-Mori Hospital, Miyazaki).

Author Contributions

  1. Conceptualization: AK KE.
  2. Formal analysis: T. Koga AO SS KA.
  3. Funding acquisition: AK KE.
  4. Investigation: TF TH T. Ishii YU T. Kodera MN YT SH TA T. Izumiyama OT YH RW HO TM SK NI KI MT TO HN.
  5. Project administration: T. Koga AK KE.
  6. Supervision: AK KE.
  7. Writing – original draft: T. Koga.
  8. Writing – review & editing: T. Koga AK.

References

  1. 1. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO 3rd, et al. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann Rheum Dis. 2010;69(9):1580–8. pmid:20699241
  2. 2. Arend WP, Firestein GS. Pre-rheumatoid arthritis: predisposition and transition to clinical synovitis. Nature reviews Rheumatology. 2012;8(10):573–86. pmid:22907289
  3. 3. Smolen JS, Landewe R, Breedveld FC, Buch M, Burmester G, Dougados M, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2013 update. Ann Rheum Dis. 2014;73(3):492–509. PubMed Central PMCID: PMC3933074. pmid:24161836
  4. 4. Cohen G, Gossec L, Dougados M, Cantagrel A, Goupille P, Daures JP, et al. Radiological damage in patients with rheumatoid arthritis on sustained remission. Ann Rheum Dis. 2007;66(3):358–63. PubMed Central PMCID: PMCPMC1856000. pmid:16935911
  5. 5. Smolen JS, Han C, Bala M, Maini RN, Kalden JR, van der Heijde D, et al. Evidence of radiographic benefit of treatment with infliximab plus methotrexate in rheumatoid arthritis patients who had no clinical improvement: a detailed subanalysis of data from the anti-tumor necrosis factor trial in rheumatoid arthritis with concomitant therapy study. Arthritis Rheum. 2005;52(4):1020–30. pmid:15818697
  6. 6. Meyer O, Labarre C, Dougados M, Goupille P, Cantagrel A, Dubois A, et al. Anticitrullinated protein/peptide antibody assays in early rheumatoid arthritis for predicting five year radiographic damage. Ann Rheum Dis. 2003;62(2):120–6. PubMed Central PMCID: PMC1754441. pmid:12525380
  7. 7. Visser K, Goekoop-Ruiterman YP, de Vries-Bouwstra JK, Ronday HK, Seys PE, Kerstens PJ, et al. A matrix risk model for the prediction of rapid radiographic progression in patients with rheumatoid arthritis receiving different dynamic treatment strategies: post hoc analyses from the BeSt study. Ann Rheum Dis. 2010;69(7):1333–7. pmid:20498212
  8. 8. Vastesaeger N, Xu S, Aletaha D, St Clair EW, Smolen JS. A pilot risk model for the prediction of rapid radiographic progression in rheumatoid arthritis. Rheumatology (Oxford). 2009;48(9):1114–21.
  9. 9. Molenaar ET, Voskuyl AE, Dinant HJ, Bezemer PD, Boers M, Dijkmans BA. Progression of radiologic damage in patients with rheumatoid arthritis in clinical remission. Arthritis Rheum. 2004;50(1):36–42. pmid:14730597
  10. 10. Brown AK, Conaghan PG, Karim Z, Quinn MA, Ikeda K, Peterfy CG, et al. An explanation for the apparent dissociation between clinical remission and continued structural deterioration in rheumatoid arthritis. Arthritis Rheum. 2008;58(10):2958–67. pmid:18821687
  11. 11. Karim Z, Wakefield RJ, Quinn M, Conaghan PG, Brown AK, Veale DJ, et al. Validation and reproducibility of ultrasonography in the detection of synovitis in the knee: a comparison with arthroscopy and clinical examination. Arthritis Rheum. 2004;50(2):387–94. pmid:14872480
  12. 12. Szkudlarek M, Klarlund M, Narvestad E, Court-Payen M, Strandberg C, Jensen KE, et al. Ultrasonography of the metacarpophalangeal and proximal interphalangeal joints in rheumatoid arthritis: a comparison with magnetic resonance imaging, conventional radiography and clinical examination. Arthritis Res Ther. 2006;8(2):R52. PubMed Central PMCID: PMCPMC1526591. pmid:16519793
  13. 13. Kane D, Balint PV, Sturrock RD. Ultrasonography is superior to clinical examination in the detection and localization of knee joint effusion in rheumatoid arthritis. J Rheumatol. 2003;30(5):966–71. pmid:12734890
  14. 14. Johnson K, Wittkop B, Haigh F, Ryder C, Gardner-Medwin JM. The early magnetic resonance imaging features of the knee in juvenile idiopathic arthritis. Clin Radiol. 2002;57(6):466–71. pmid:12069461
  15. 15. Yamanaka H, Seto Y, Tanaka E, Furuya T, Nakajima A, Ikari K, et al. Management of rheumatoid arthritis: the 2012 perspective. Mod Rheumatol. 2013;23(1):1–7. pmid:22772460
  16. 16. Koga T, Okada A, Fukuda T, Hidaka T, Ishii T, Ueki Y, et al. 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. 2016;95(17):e3476. pmid:27124044
  17. 17. Fries JF, Spitz P, Kraines RG, Holman HR. Measurement of patient outcome in arthritis. Arthritis Rheum. 1980;23(2):137–45. pmid:7362664
  18. 18. Salaffi F, Carotti M, Ciapetti A, Gasparini S, Filippucci E, Grassi W. Relationship between time-integrated disease activity estimated by DAS28-CRP and radiographic progression of anatomical damage in patients with early rheumatoid arthritis. BMC musculoskeletal disorders. 2011;12:120. PubMed Central PMCID: PMCPMC3123613. pmid:21624120
  19. 19. Matthews JN, Altman DG, Campbell MJ, Royston P. Analysis of serial measurements in medical research. BMJ. 1990;300(6719):230–5. PubMed Central PMCID: PMCPMC1662068. pmid:2106931
  20. 20. Takeuchi T, Yamanaka H, Ishiguro N, Miyasaka N, Mukai M, Matsubara T, et al. Adalimumab, a human anti-TNF monoclonal antibody, outcome study for the prevention of joint damage in Japanese patients with early rheumatoid arthritis: the HOPEFUL 1 study. Ann Rheum Dis. 2014;73(3):536–43. PubMed Central PMCID: PMC4151516. pmid:23316080
  21. 21. Bruynesteyn K, Boers M, Kostense P, van der Linden S, van der Heijde D. Deciding on progression of joint damage in paired films of individual patients: smallest detectable difference or change. Ann Rheum Dis. 2005;64(2):179–82. PubMed Central PMCID: PMC1755378. pmid:15286006
  22. 22. Bruynesteyn K, Van Der Heijde D, Boers M, Saudan A, Peloso P, Paulus H, et al. Detecting radiological changes in rheumatoid arthritis that are considered important by clinical experts: influence of reading with or without known sequence. J Rheumatol. 2002;29(11):2306–12. pmid:12415585
  23. 23. Heinze G, Schemper M. A solution to the problem of separation in logistic regression. Stat Med. 2002;21(16):2409–19. pmid:12210625
  24. 24. Fautrel B, Granger B, Combe B, Saraux A, Guillemin F, Le Loet X. Matrix to predict rapid radiographic progression of early rheumatoid arthritis patients from the community treated with methotrexate or leflunomide: results from the ESPOIR cohort. Arthritis Res Ther. 2012;14(6):R249. PubMed Central PMCID: PMC3674616. pmid:23164197
  25. 25. Colebatch AN, Edwards CJ, Ostergaard M, van der Heijde D, Balint PV, D'Agostino MA, et al. EULAR recommendations for the use of imaging of the joints in the clinical management of rheumatoid arthritis. Ann Rheum Dis. 2013;72(6):804–14. pmid:23520036
  26. 26. Schett G, Emery P, Tanaka Y, Burmester G, Pisetsky DS, Naredo E, et al. Tapering biologic and conventional DMARD therapy in rheumatoid arthritis: current evidence and future directions. Ann Rheum Dis. 2016;75(8):1428–37. pmid:27261493
  27. 27. Bohler C, Radner H, Smolen JS, Aletaha D. Serological changes in the course of traditional and biological disease modifying therapy of rheumatoid arthritis. Ann Rheum Dis. 2013;72(2):241–4. pmid:23087181
  28. 28. Aletaha D, Bluml S. Therapeutic implications of autoantibodies in rheumatoid arthritis. RMD Open. 2016;2(1):e000009. PubMed Central PMCID: PMCPMC4879342. pmid:27252890
  29. 29. Smolen JS, Aletaha D, Bijlsma JW, Breedveld FC, Boumpas D, Burmester G, et al. Treating rheumatoid arthritis to target: recommendations of an international task force. Ann Rheum Dis. 2010;69(4):631–7. PubMed Central PMCID: PMC3015099. pmid:20215140
  30. 30. Lillegraven S, Paynter N, Prince FH, Shadick NA, Haavardsholm EA, Frits ML, et al. Performance of matrix-based risk models for rapid radiographic progression in a cohort of patients with established rheumatoid arthritis. Arthritis Care Res (Hoboken). 2013;65(4):526–33. PubMed Central PMCID: PMC3594116.
  31. 31. Smolen JS, Aletaha D, Steiner G. Does damage cause inflammation? Revisiting the link between joint damage and inflammation. Ann Rheum Dis. 2009;68(2):159–62. pmid:19139203
  32. 32. Takahashi C, Kaneko Y, Okano Y, Ooshima H, Takeuchi T. THU0118 Methotrexate Polyglutamates in Erythrocytes Correlates with Clinical Response in Japanese Patients with Rheumatoid Arthritis. Annals of the Rheumatic Diseases. 2014;73(Suppl 2):218–9.
  33. 33. Aletaha D, Smolen J, Ward MM. Measuring function in rheumatoid arthritis: Identifying reversible and irreversible components. Arthritis Rheum. 2006;54(9):2784–92. pmid:16947781