Switching First-Line Targeted Therapy After Not Reaching Low Disease Activity Within Six Months is Superior to Conservative Approach: A Propensity Score Matched Analysis From The ATTRA Registry

DOI: https://doi.org/10.21203/rs.3.rs-76194/v1

Abstract

Background: Treat-to-target (T2T) is a widely accepted strategy for patients with rheumatoid arthritis (RA). It recommends attaining a goal of at least low disease activity (LDA) within six months; otherwise, the current therapy should be modified. We aimed to investigate whether switching a first-line targeted therapy (TT) in patients not reaching LDA within six months leads to a higher probability of meeting LDA at 12‑month visit in daily clinical practice using data from Czech registry ATTRA.

Methods: We included patients with RA starting the first-line TT from 1 January 2012 to 31 January 2017 with at least one-year follow-up. We created four mutually exclusive cohorts based on 1) switching to another TT within the first year and 2) reaching a treatment target (DAS28-ESR ≤ 3.2) at the 6‑month visit. The primary outcome was the comparison of odds for reaching remission (REM) or LDA at the 12-month visit between patients switching and not switching TT after not reaching treatment target at six months. Before using logistic regression to estimate the odds ratio, we employed the propensity score to match patients at the 6-month visit.

Results: A total of 1275 patients were eligible for the analysis. Sixty-two patients switched within the first five months of the treatment before evaluating treatment response at the 6-month visit (C1); 598 patients reached the treatment target within six months of therapy (C2); 124 patients did not reach treatment response at 6-month visit and switched to another therapy (C3), and 491 patients continued with the same treatment despite not reaching LDA at the 6‑month visit (C4). We matched 75 patients from cohort C3 and 75 patients from C4 using the propensity score. Patients following T2T principle (C3) showed 2.8 (95% CI 1.4–5.8; p=0.005) times increased likelihood of achieving REM/LDA at 12‑month visit compared to patients not following T2T strategy (C4).

Conclusions: In daily clinical practice, the application of T2T strategy is underused. Switching TT after not reaching REM/LDA within the first six months leads to a higher probability of achieving REM/LDA in RA patients at the 12-month visit.

Background

Currently, patients with rheumatoid arthritis (RA) have multiple drug options with different mechanisms of actions to address the heterogeneous nature of the disease. Patients may require multiple successive therapies throughout their lives. In 2010, the European League Against Rheumatism (EULAR) developed its first recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs (DMARDs) (1). There were several updates throughout the years with the last update so far at the end of 2019 (2). Treating toward a target of remission (REM) or at least a low disease activity (LDA) has become the standard of care for patients. Achievement of the treatment target often requires switching between drugs. According to EULAR recommendations, therapy with conventional synthetic (cs) DMARDs should be started as soon as the diagnosis of RA is made, and methotrexate (MTX) should be the first choice. If the treatment target is not reached with the first csDMARDs, and poor prognostic factors are present (i.e. presence of rheumatoid factor/anti-citrullinated protein antibodies, high disease activity early, joint damage, failure of two or more csDMARDs), a biological (b)DMARD or targeted synthetic (ts)DMARD should be added. If there is no improvement within three months after the start of treatment or if patients have not reached the treatment target with bDMARD/tsDMARD by six months, therapy should be adjusted, and treatment with another bDMARD or tsDMARD should be considered (2).

The approach currently recommended for RA treatment involves titrating medication dosages until pre-specified disease activity targets (either REM or LDA) have been met and maintaining these targets over time. Such so-called treat-to-target strategies (T2T) have proven to be more effective and to generate better outcomes than usual care (3, 4). Efficacy of T2T approach has been evaluated in many randomised controlled clinical trials (511). Even though the T2T strategy has been widely applied in daily clinical practice nowadays, studies from daily clinical practice concerning the advantage of following T2T over usual care are still required. Several studies evaluating the efficacy of T2T in real clinical practice have been already done (4, 1217), but more evidence through real-life data is needed to support the implementation of T2T.

The primary aim of this study was to assess whether following a T2T strategy after not reaching treatment target (REM/LDA) within first six months leads to a higher probability of meeting the treatment target at the 12-month visit in daily clinical practice. We also described four groups of patients based on different courses of their treatment with the fist bDMARD/tsDMARD.

Methods

Study setting and data source

The ATTRA registry, established in 2002, is a prospective, national, observational cohort study. Its main purpose is to evaluate the safety and effectiveness of bDMARDs (and lately also tsDMARDs) in patients with chronic inflammatory rheumatic diseases. Patients with RA (and ankylosing spondylitis, psoriatic arthritis, and juvenile idiopathic arthritis) starting bDMARDs or tsDMARDs are recruited from fifty practices sites (private or academic), and captures more than 95% of patients with RA treated with bDMARDs/tsDMARDs in the Czech Republic (CZ). Targeted therapy (TT) has been reimbursed for patients with RA if DAS28 > 5.1 despite therapy with csDMARDs until 2019. Since 2019 the cut-off for DAS28 was lowered to 3.2 in CZ. Initial TT should include either TNF inhibitors (TNFis) or tsDMARDs. At the time of this analysis, the ATTRA database included information on 5050 patients with RA.

At the start of therapy, baseline data are collected including demographics (gender, age at diagnosis, age at the start of 1st line treatment, height, weight, smoking status, presence comorbidities), disease characteristics (disease duration, presence of rheumatoid factor (RF) and anti-citrullinated protein antibodies (ACPA), presence of joint erosions on X-ray), disease activity (swollen and tender joint count (0–28), patient global assessment (PTGA) of disease activity and physician global assessment of disease activity (MDGA) on a 100-mm visual analogue scale (VAS; 0 – best, 100 – worst), erythrocyte sedimentation rate (ESR, mg/h) and C-reactive protein (CRP, mg/L)) and 28-joint disease activity score index (DAS28; 0–10) (18), simplified disease activity index (SDAI, 0–86) (19), Health Assessment Questionnaire (HAQ) for patient function with values from 0 to 3 (the higher, the worse disability) (20), EuroQol EQ-5D questionnaire for quality of life with values from –0.59 to 1 (the higher, the better quality of life) (21), and current or previous anti-rheumatic therapies (csDMARDs, bDMARDs, tsDMARDs) and therapy with glucocorticoids (GCs). Follow-up data on disease activity, disease function and anti-rheumatic therapies are collected after three and six months, and then every six months for three years, with disease activity and anti-rheumatic therapy data collected annually thereafter.

Ethics approval for ATTRA was granted by the Czech Multicentre Research Ethics Committee, no. 201611 S300 and Institutional Ethics Committee of Institute of Rheumatology, Prague, Czech Republic, no. 10113/2016. No additional ethical approval was required for the current analysis. All subjects provided their written consent for the collection and storage of data before participation. All procedures were performed following the Declaration of Helsinki.

Study population

In this study, we included all bio-naive adult patients diagnosed with RA starting bDMARDs/tsDMARDs within a period from 1 January 2012 to 31 January 2017 with at least one-year follow-up. Patients without available DAS28-ESR at baseline, 6-month and 12-month visit or without HAQ and EQ-5D at baseline and 12-month visit were excluded (see Supplementary Figure 1).

Study design

We divided patients into four cohorts based on a treatment result at the 6-month visit and based on switches to another therapy during the first year of the treatment (Figure 1). First, we evaluated whether patients switched to another therapy within the first five months of the treatment. Next, we assessed if patients reached remission or low disease activity at the 6-month visit (defined as DAS28‑ESR ≤ 3.2). Finally, we checked whether patients changed the therapy within months 6–11 provided they did not achieve the treatment target. Cohort C1 includes patients that changed bDMARD/tsDMARD therapy during the first months (usually at 3-month visit) before evaluating treatment response at the 6‑month visit. These patients were either not responding to the treatment at all, or were not tolerating the treatment (e.g., side effects) within the first months of the first-line therapy. Cohort C2 consists of patients ideally responding to the treatment because they achieved the treatment target after six months of therapy without a need to switch. Cohort C3 comprises the group of patients not responding to the treatment, because they did not achieve the treatment target after the first six months of therapy. Following T2T principles, they switched to a different treatment. The last cohort C4 is represented by patients not responding to the treatment since they did not achieve the treatment target (similarly to C3 cohort). Regardless of T2T principles, they continued with the same treatment.

Objectives and Outcome measures

The primary objective of this study was to compare odds for the achievement of REM or at least LDA after one year of the treatment between patients following and not following T2T strategy (C3 vs C4). We assessed disease activity DAS28-ESR index; specifically, LDA was defined as DAS28-ESR ≤ 3.2 and REM was defined as DAS28-ESR < 2.6. In terms of the secondary outcomes, we compared treatment results based on DAS28-ESR after 12 months between all studied cohorts. The proportion of patients with remission (REM; DAS28‑ESR < 2.6), low disease activity (LDA; 2.6 ≤ DAS28-ESR ≤ 3.2), medium disease activity (MDA; 3.2 < DAS28-ESR ≤ 5.1) and high disease activity (HDA; DAS28-ESR > 5.1) at baseline and 12-month visit were compared across the studied cohorts C1–C4. Next, we compared changes in parameters related to disease activity (DAS28‑ESR, SDAI, tender and swollen joint count, CRP, ESR, PTGA, MDGA) and quality of life (HAQ-DI, EQ-5D) after 6 and 12 months of the bDMARDs/tsDMARDs treatment between cohorts C3 and C4.

Statistical methods

Descriptive summary of patients’ demographic and treatment characteristics and disease activity measurements was performed for all four studied cohorts C1–C4. For continuous variables, we calculated the median with interquartile range (IQR, 25th–75th percentiles). For a description of categorical variables, we used absolute and relative frequencies (i.e., percentages). To test differences between two patients’ groups, we performed the non-parametric Mann-Whitney U test for continuous variables and Pearson’s chi-squared test for categorical variables. The magnitude of changes in parameters over two visits was tested through the paired Wilcoxon test. For all tests, P‑values < 0.05 were considered to be statistically significant. We did not impute missing data in this analysis and performed a complete-case analysis instead. The percentage of missing data in outcome variables (i.e. DAS28-ESR, HAQ and EQ-5D at baseline, 6 and 12 months) was relatively small; we excluded 1.8% of patients in total.

We used propensity score matching to match patients not switching to another therapy after not reaching treatment target at 6-month visit (C4) to patients switching to a different treatment after not reaching treatment target (C3). For matching, we performed logistic regression with outcome variable C3 (=1) vs C4 (=0) and covariates: gender, age at the start of 1st line therapy, disease duration, number of previous csDMARDs, glucocorticoids in previous therapy, swollen joint count, tender joint count, PTGA, ESR, CRP, HAQ, RF positivity, presence of comorbidities, smoking, csDMARDs in concomitant therapy and glucocorticoids in previous therapy. We chose the matching ratio 1:1 and set the caliper to a value 0.2. We used matching to make both groups comparable in characteristics at 6-month visit and to minimise confounding by other factors in the evaluation of achieving REM/LDA at the 12-month visit. After we carried out propensity score matching, we employed binary logistic regression to determine the odds for reaching of REM/LDA at the 12-month visit in cohort C3 and C4. We did all descriptive statistics and testing using IBM SPSS Statistics 25.0. Propensity score model was performed in R (version 3.5.3).

Results

Patients’ characteristics at baseline

In total, we included 1275 patients fulfilling the inclusion criteria into the analysis (see Supplementary Figure 1). Cohort C1 was represented by 62 (4.9%) patients, C2 consisted of 598 (46.9%) patients, C3 included 124 (9.7%) patients and 491 (38.5%) patients belonged to C4 subgroup (see Supplementary Figure 1). The most frequently administered drug was bio-original adalimumab (ranging from 27.4% to 40.3% in studied cohorts), bio-original etanercept (from 10.5% to 32.3%) and golimumab (from 6.5% to 15.5%). We present baseline characteristics of all four studied cohorts in Table 1. Median age at the start of the first bDMARD/tsDMARD was between 51 years (C1) and 55 years (C4 cohort). Females represented from 72.1% (C2 cohort) to 83.9% (C1 cohort) patients. Patients from all studied cohorts had high baseline disease activity according to DAS28-ESR score with median 6.4 (5.7–7.0) in C1, 5.9 (5.3–6.5) in C2, 6.2 (5.6–6.8) in C3 and 6.3 (5.8–6.8) in C4. Patient from cohorts C3 and C4 significantly differed only in age at the start of the first therapy (p=0.016) and the number of previous csDMARDs (p=0.025). The median age was 52.0 (44.5–61.0) years in C3 and 55.0 (48.0–63.0) years in C4. Cohorts C1 and C2 significantly differed in gender (84% vs 72% females; p=0.046) and in almost all parameters related to baseline disease activity and quality of life. Patients from cohort C1 had higher disease activity than C2 cohort according to DAS28-ESR (median 6.4 vs 5.9; p<0.001), TJC (median 14 vs 12; p=0.005), CRP (median 22 vs 12; p=0.002), PTGA (median 78 vs 70; p=0.008), MDGA (median 74 vs 65; p=0.015) and worse physical function and quality of life according to HAQ-DI (median 1.8 vs 1.4; p=0.001) and EQ-5D (median 0.1 vs 0.2; p=0.048) resp.

Disease activity after 12 months in C1–C4

Comparison of disease activity according to the DAS28-ESR score after one year of treatment in cohorts C1–C4 is illustrated in Figure 2. We could see the best treatment results after 12 months in the group C2 with almost 79% patients with REM/LDA compared to 48% patients in group C1 (p<0.001), 40% patients in group C3 (p<0.001) and 32% in group C4 (p<0.001). Although there was no statistically significant difference in the proportion of patients with REM/LDA between groups C3 (following T2T strategy) and C4 (not following T2T strategy) after 12 months (p=0.095), we could observe slightly better results in the group C3 (40% vs 32% with REM/LDA). We observed the lowest median value of DAS28-ESR in the group C2, and it falls within the level of remission. The median value of DAS28-ESR in group C1 corresponded to the upper bound of low disease activity, median DAS28-ESR values in groups C3 and C4 were within the range of medium disease activity.

Comparison of cohorts C3 and C4 at 6-month and 12-month visit

At the 6-month visit, patients from group C3 and C4 differed in all tested parameters related to disease activity and quality of life (Table 2). We observed lower disease activity and better quality of life in C4. Patients from C3 and C4 did not significantly differ in concomitant therapy, but numerically more changes in dosage of glucocorticoids and methotrexate have been observed in the C3 cohort compared to C4 cohort between M6-M12 (see Supplementary Table 1). At the 12-month visit, patients from both groups did not significantly differ in most of the parameters related to disease activity; they only differed in PTGA (p=0.044) and EQ-5D (p=0.017). In terms of the magnitude of changes across the two visits, patients from C3 significantly improved in all parameters related to disease activity and quality of life (see Supplementary Table 2). Patients from C4 did not significantly improve in CRP and HAQ-DI. In the comparison of the size of changes between the two groups, patients from C3 showed better results (i.e., more significant improvements) in all tested parameters (see Supplementary Table 2).

Odds for treatment target in C3 vs C4 at the 12-month visit

We employed propensity score matching to reduce selection bias by adjusting for potential confounding factors at the 6-month visit. We show a description of patients’ characteristics at 6-month visit after using propensity score matching in Table 3. Both groups included 75 patients after the matching. Density plots of propensity score before and after matching are displayed in Supplementary Figure 2. Patients did not differ anymore in parameters related to disease activity and quality of life (see Table 3). The most frequently administered drugs at the 12-mont visit were tocilizumab (27%), certolizumab (17%), abatacept (15%) and etanercept (12%) in C3. Patients from C4 were most frequently treated with adalimumab (35%), etanercept (21%), golimumab (16%) and certolizumab (13%). To compare odds for reaching treatment target at the 12-month visit in patients following T2T principle at 6-month visit (C3) vs patients staying on the first treatment (C4), we employed logistic regression model with outcome DAS28-ESR ≤ 3.2. Patients following T2T principle (C3) showed 2.8 (CI 1.4–5.8) times higher odds for reaching at least LDA at the 12-month visit (p=0.005) compared to patients not following T2T principle (C4). In the group C3, 41% of patients achieved at least REM/LDA at the 12-month visit, while in the group C4, it was 20%.

Discussion

In this prospective observational cohort study from real clinical practice in the Czech Republic, we have shown that following the T2T strategy and switching the targeted drug to another therapy after not reaching REM/LDA at 6-month visit increases the chance of achieving REM/LDA at the 12-month visit as opposed to patients not following the treatment target. This finding support results from previous studies showing that T2T is efficient in daily clinical practice. Our study also provided a summary of four different courses of treatment management during the first year of bDMARD/tsDMARD therapy. We created four patients’ cohorts based on switching the treatment and based on reaching a treatment target at six months. We described all four patients’ groups at baseline and compared their treatment results after one year of treatment. Furthermore, we evaluated disease activity and quality of life at six months in groups C3 and C4 and compared the sizes of changes from the 6-month to the 12-month visit. We observed that patients not following the T2T at the 6-month visit (C4) had lower disease activity and better quality of life at six months than patients following T2T and switching to another therapy after not reaching the treatment target (C3). However, patients following the T2T strategy showed a more significant improvement both in disease activity and quality of life within the period from the 6-month visit to 12month visit. Patients from cohort C3 also had a higher rate of REM/LDA at 12 months in comparison with C4 (though not statistically significant; p = 0.095).

A similar study investigated whether a tight control treatment strategy (i.e. optimising treatment by measurement of disease activity in order to make treatment adjustments to reach a predefined target LDA/REM) in early RA is more effective than treatment according to usual care in reaching REM (DAS28 < 2.6) after one year (4). They compared two distinct early RA cohorts from two different regions in the Netherlands: the usual care cohort and the ‘tight control’ cohort. The OR adjusted for baseline DAS28 was 3.1 (95% CI 1.8–5.2). Therefore, patients treated according to tight control had approximately three times higher odds to reach REM at one year after the baseline. This result is very similar to the OR obtained in our study, though we evaluated achievement of LDA/REM instead. In another similar study, Norwegian authors compared patients following a T2T strategy (2010–2015) with patients from the pre-T2T cohort (2006–2009) following routine care (13). They assessed the twoyear effect on disease activity and health-related quality of life and showed significantly higher odds (multivariable OR 1.89, 95% CI 1.33–2.68) for SDAI remission (≤ 3.3) in patients following a T2T strategy. Within secondary outcomes, they also evaluated REM, according to DAS28 (OR 2.15, 95% CI 1.51–3.06).

A Dutch study investigated the 3-year results of a protocolised T2T strategy in daily clinical practice (16). Authors found out that T2T leads to hight remission rates, improved physical function and quality of life, and limited radiographic damage after three years in daily clinical practice. In another study from the Netherlands, authors described a five-year continuous application of a T2T strategy in patients with early RA in daily clinical practice and confirmed the favourable disease- and patientrelated outcomes (12). A longitudinal study of RA patients from 10 countries (RA BIODAM) investigated whether following a T2T strategy in daily clinical practice leads to more patients meeting REM (14). Application of T2T every three months did not yield a higher likelihood of REM according to DAS44 and DAS28 three months later, but sustained T2T (i.e. T2T followed in at least two consecutive visits) resulted in an increased likelihood of achieving DAS44 REM (OR 1.19, 95% CI 1.03–1.39).

Although the present study has a limitation of the absence of randomisation, we have partially overcome this problem by employing the propensity score matching at the 6-month visit. Thus, we have minimised confounding by other factors, and we obtained the effect of following/notfollowing T2T principle in the evaluation of REM/LDA at the 12-month visit. A possible limitation of this study could be an absence of monitoring treatment intensification through increased dosages. Further, our study only concerned the firstline bDMARD/tsDMARD therapy. Thus, evaluating of implementation of T2T strategy within subsequent lines of therapy could be a possible subject for future studies.

Conclusion

In conclusion, application of T2T principles and switching to another bDMARD/tsDMARD after not reaching REM/LDA within the first six months of bDMARD/tsDMARD treatment leads to a higher probability of achieving REM/LDA in RA patients at the 12-month visit. In this study, the T2T strategy showed superiority over traditional routine care in daily clinical practice.

List Of Abbreviations

ACPA: anti-citrullinated protein antibodies

bDMARDs: biological disease-modifying anti-rheumatic drugs

CI: confidence interval

CRP: C-reactive protein

csDMARDs: conventional synthetic disease-modifying anti-rheumatic drugs

CZ: Czech Republic

DAS28-ESR: 28-joint disease activity score using the erythrocyte sedimentation rate

DMARD: disease-modifying anti-rheumatic drugs

EQ-5D: EuroQol-5 Dimensions

ESR: erythrocyte sedimentation rate

EULAR: European League Against Rheumatism

GCs: glucocorticoids

HAQ-DI: health assessment questionnaire disability index

HDA: high disease activity

IQR: interquartile range

LDA: low disease activity

MDA: medium disease activity

MDGA: physician global assessment of disease activity

MTX: methotrexate

OR: odds ratio

PTGA: patient global assessment of disease activity

RA: rheumatoid arthritis

REM: remission

RF: rheumatoid factor

SDAI: simplified disease activity index

T2T: treat-to-target

TNFi: tumour necrosis factor inhibitor

tsDMARDs: targeted disease-modifying anti-rheumatic drugs

TT: targeted therapy

VAS: visual analogue scale

Declarations

Ethics approval and consent to participate

All procedures in this study were in accordance with the ethical standards of the institutional and national research committee (Czech Multicentre Research Ethics Committee, no. 201611 S300 and Institutional Ethics Committee of Institute of Rheumatology, Prague, Czech Republic, no. 10113/2016) and with the 1964 Helsinki declaration and its later amendments. All subjects provided their written consent for the collection and storage of data before participation.

Consent for publication

Not applicable

Availability of data and materials

The datasets analysed during the current study are available from the corresponding author on reasonable request. Requests will be considered by the Czeach Rheamatological Society.

Competing interests

The authors declare that they have no competing interests

Funding

This work was supported by the project (Ministry of Health, Czech Republic) for conceptual development of research organisation 00023728 (Institute of Rheumatology).

Authors’ contributions

All authors were involved in drafting the manuscript or revising it critically for content. LN planned and performed the analysis, interpreted patients’ data and wrote the manuscript. JV, KP and PH revised the manuscript. ZK managed the project. JZ designed the project, supervised its conduct, and helped to write the manuscript. All authors read and approved the final manuscript.

Acknowledgements

Not applicable

References

  1. Smolen JS, Landewé R, Breedveld FC, Dougados M, Emery P, Gaujoux-Viala C, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs. Ann Rheum Dis. 2010 Jun;69(6):964–75.
  2. Smolen JS, Landewé RBM, Bijlsma JWJ, Burmester GR, Dougados M, Kerschbaumer A, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann Rheum Dis. 2020 Jan 22;79:685-699.
  3. Schoels M, Wong J, Scott DL, Zink A, Richards P, Landewé R, et al. Economic aspects of treatment options in rheumatoid arthritis: a systematic literature review informing the EULAR recommendations for the management of rheumatoid arthritis. Annals of the Rheumatic Diseases. 2010 Jun 1;69(6):995–1003.
  4. Schipper LG, Vermeer M, Kuper HH, Hoekstra MO, Haagsma CJ, Broeder AAD, et al. A tight control treatment strategy aiming for remission in early rheumatoid arthritis is more effective than usual care treatment in daily clinical practice: a study of two cohorts in the Dutch Rheumatoid Arthritis Monitoring registry. Annals of the Rheumatic Diseases. 2012 Jun 1;71(6):845–50.
  5. Grigor C, Capell H, Stirling A, McMahon AD, Lock P, Vallance R, et al. Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet. 2004 Jul 17;364(9430):263–9.
  6. Verstappen SMM, Jacobs JWG, van der Veen MJ, Heurkens AHM, Schenk Y, ter Borg EJ, et al. Intensive treatment with methotrexate in early rheumatoid arthritis: aiming for remission. Computer Assisted Management in Early Rheumatoid Arthritis (CAMERA, an open-label strategy trial). Ann Rheum Dis. 2007 Nov;66(11):1443–9.
  7. Goekoop-Ruiterman YPM, de Vries-Bouwstra JK, Allaart CF, van Zeben D, Kerstens PJSM, Hazes JMW, et al. Clinical and radiographic outcomes of four different treatment strategies in patients with early rheumatoid arthritis (the BeSt study): a randomized, controlled trial. Arthritis Rheum. 2005 Nov;52(11):3381–90.
  8. Goekoop-Ruiterman YPM, de Vries-Bouwstra JK, Kerstens PJSM, Nielen MMJ, Vos K, van Schaardenburg D, et al. DAS-driven therapy versus routine care in patients with recent-onset active rheumatoid arthritis. Ann Rheum Dis. 2010 Jan;69(1):65–9.
  9. Schipper LG, van Hulst LTC, Grol R, van Riel PLCM, Hulscher MEJL, Fransen J. Meta-analysis of tight control strategies in rheumatoid arthritis: protocolized treatment has additional value with respect to the clinical outcome. Rheumatology (Oxford). 2010 Nov;49(11):2154–64.
  10. Stoffer MA, Schoels MM, Smolen JS, Aletaha D, Breedveld FC, Burmester G, et al. Evidence for treating rheumatoid arthritis to target: results of a systematic literature search update. Ann Rheum Dis. 2016 Jan;75(1):16–22.
  11. Schoels M, Knevel R, Aletaha D, Bijlsma JWJ, Breedveld FC, Boumpas DT, et al. Evidence for treating rheumatoid arthritis to target: results of a systematic literature search. Ann Rheum Dis. 2010 Apr;69(4):638–43.
  12. Versteeg GA, Steunebrink LMM, Vonkeman HE, ten Klooster PM, van der Bijl AE, van de Laar MAFJ. Long-term disease and patient-reported outcomes of a continuous treat-to-target approach in patients with early rheumatoid arthritis in daily clinical practice. Clin Rheumatol. 2018 May 1;37(5):1189–97.
  13. Brinkmann GH, Norvang V, Norli ES, Grøvle L, Haugen AJ, Lexberg ÅS, et al. Treat to target strategy in early rheumatoid arthritis versus routine care – A comparative clinical practice study. Seminars in Arthritis and Rheumatism. 2019 Apr 1;48(5):808–14.
  14. Ramiro S, Landewé RB, Heijde D van der, Sepriano A, FitzGerald O, Ostergaard M, et al. Is treat-to-target really working in rheumatoid arthritis? a longitudinal analysis of a cohort of patients treated in daily practice (RA BIODAM). Annals of the Rheumatic Diseases. 2020 Apr 1;79(4):453–9.
  15. Vermeer M, Kuper HH, Hoekstra M, Haagsma CJ, Posthumus MD, Brus HLM, et al. Implementation of a treat-to-target strategy in very early rheumatoid arthritis: results of the Dutch Rheumatoid Arthritis Monitoring remission induction cohort study. Arthritis Rheum. 2011 Oct;63(10):2865–72.
  16. Vermeer M, Kuper HH, Moens HJB, Drossaers‐Bakker KW, Bijl AE van der, Riel PLCM van, et al. Sustained Beneficial Effects of a Protocolized Treat-to-Target Strategy in Very Early Rheumatoid Arthritis: Three-Year Results of the Dutch Rheumatoid Arthritis Monitoring Remission Induction Cohort. Arthritis Care & Research. 2013;65(8):1219–26.
  17. Steunebrink LMM, Versteeg GA, Vonkeman HE, Ten Klooster PM, Kuper HH, Zijlstra TR, et al. Initial combination therapy versus step-up therapy in treatment to the target of remission in daily clinical practice in early rheumatoid arthritis patients: results from the DREAM registry. Arthritis Res Ther. 2016 Mar 8;18:60.
  18. Prevoo ML, van ’t Hof MA, Kuper HH, van Leeuwen MA, van de Putte LB, van Riel PL. Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum. 1995 Jan;38(1):44–8.
  19. Smolen JS, Breedveld FC, Schiff MH, Kalden JR, Emery P, Eberl G, et al. A simplified disease activity index for rheumatoid arthritis for use in clinical practice. Rheumatology (Oxford). 2003 Feb;42(2):244–57.
  20. Bruce B, Fries JF. The Health Assessment Questionnaire (HAQ). Clin Exp Rheumatol. 2005 Oct;23(5 Suppl 39):S14-18.
  21. EuroQol Group. EuroQol--a new facility for the measurement of health-related quality of life. Health Policy. 1990 Dec;16(3):199–208.

Tables

Table 1

Baseline characteristics of patients in cohorts C1–C4 (N = 1275)

 

C1 (n = 62)

C2 (n = 598)

C3 (n = 124)

C4 (n = 491)

Female, n (%)

52 (83.9%)

431 (72.1%)

102 (82.3%)

390 (79.4%)

Age at diagnosis, years, median (IQR)

44.0 (34.0–52.0)

43.5 (33.0–52.0)

45.0 (34.0–51.5)

47.0 (38.0–54.0)

Age at start of 1st line, years, median (IQR)

51.0 (42.0–58.0)

53.0 (41.0–60.0)

52.0 (44.5–61.0)

55.0 (48.0–63.0)

Disease duration, years, median (IQR)

5.6 (3.0–7.8)

6.0 (2.5–11.8)

5.0 (2.2–12.1)

6.2 (3.0–12.9)

RF positive, n (%)

47 (75.8%)

428 (71.6%)

92 (74.2%)

389 (79.6%)

ACPA positive, n/total (%)

44/61 (72.1%)

399/587 (68.0%)

91/120 (75.8%)

348/480 (72.5%)

Presence of erosions, n/total (%)

25/38 (65.8%)

210/295 (71.2%)

46/67 (68.7%)

22/290 (76.6%)

Presence of comorbidities, n (%)

44 (71.0%)

375 (62.7%)

79 (63.7%)

337 (68.6%)

Currently smoking, n/total (%)

10/53 (18.9%)

102/504 (20.2%)

26/103 (25.2%)

93/413 (22.5%)

Number of previous csDMARDs, n (%)

       

0

0 (0.0%)

8/592 (1.4%)

2 (1.6%)

6/484 (1.2%)

1

13 (21.0%)

226/592 (38.2%)

27 (21.8%)

131/484 (27.1%)

2

18 (29.0%)

147/592 (24.8%)

30 (24.2%)

165/484 (34.1%)

3

17 (27.4%)

112/592 (18.9%)

35 (28.2%)

113/484 (23.3%)

4+

14 (22.6%)

99/592 (16.7%)

30 (24.2%)

69/484 (14.3%)

Glucocorticoids in previous history, n (%)

56 (90.3%)

519/597 (86.9%)

112 (90.3%)

442 (90.0%)

Concomitant csDMARDs, n (%)

54 (87.1%)

549 (91.8%)

107 (86.3%)

440 (89.6%)

Concomitant MTX, n (%)

44 (71.0%)

454 (75.9%)

77 (62.1%)

349 (71.1%)

Concomitant GCs, n (%)

49 (79.0%)

446 (74.6%)

96 (77.4%)

402 (81.9%)

DAS28-ESR (0–10), median (IQR)

6.4 (5.7–7.0)

5.9 (5.3–6.5)

6.2 (5.6–6.8)

6.3 (5.8–6.8)

TJC (28 joints), median (IQR)

14.0 (11.0–19.0)

12.0 (9.0–16.0)

14.5 (9.0–19.0)

13.0 (10.0–18.0)

SJC (28 joints), median (IQR)

10.0 (7.0–13.0)

9.0 (6.0–12.0)

9.5 (6.0–12.5)

10.0 (7.0–13.0)

ESR (mm/h), median (IQR)a

33.5 (16.0–53.0)

28.0 (16.0–40.0)

32.0 (18.0–50.0)

34.0 (23.0–50.0)

CRP (mg/l), median (IQR)b

22.0 (9.4–34.0)

12.0 (5.3–23.5)

15.0 (7.9–31.0)

16.8 (8.0–33.1)

SDAI (0–86), median (IQR)c

40.5 (32.7–47.8)

35.6 (29.5–42.4)

39.3 (33.0–48.2)

39.0 (32.6–45.9)

PTGA (0–100), median (IQR)

78.0 (69.0–84.0)

70.0 (59.0–80.0)

75.0 (62.5–81.5)

75.0 (60.0–85.0)

MDGA (0–100), median (IQR)d

74.0 (60.0–80.0)

65.0 (52.0–75.0)

70.0 (58.5–80.0)

70.0 (60.0–80.0)

HAQ-DI (0–3), median (IQR)

1.8 (1.3–2.0)

1.4 (1.0–1.8)

1.8 (1.4–2.0)

1.8 (1.4–2.0)

EQ-5D (-0.59–1), median (IQR)

0.1 (0.0–0.6)

0.2 (0.1–0.7)

0.1 (0.0–0.5)

0.1 (0.0–0.5)

IQR interquartile range; RF rheumatoid factor; ACPA anti-citrullinated protein; csDMARDs conventional synthetic disease-modifying anti-rheumatic drugs; MTX methotrexate; DAS28-ESR 28-joint disease activity score with ESR; TJC tender joint count; SJC swollen joint count; ESR erythrocyte sedimentation rate; CRP C-reactive protein; SDAI Simplified Disease Activity Index; PTGA patient general assessment of disease activity; MDGA physician general assessment of disease activity; HAQDI Health Assessment Questionnaire; EQ-5D EuroQol 5 Dimension for measuring the quality of life
a n = 62 (C1), n = 573 (C2), n = 118 (C3), n = 486 (C4)
b n = 60 (C1), n = 576 (C2), n = 123 (C3), n = 480 (C4)
c n = 55 (C1), n = 560 (C2), n = 119 (C3), n = 465 (C4)
d n = 57 (C1), n = 582 (C2), n = 120 (C3), n = 476 (C4)

Table 2

Comparison of parameters related to disease activity, quality of life and concomitant therapy between C3 and C4 cohort at the 6-month and 12-month visit

 

6 months

 

12 months

 
 

C3 (n = 124)

C4 (n = 491)

P-value

C3 (n = 124)

C4 (n = 491)

P-value

DAS28-ESR (0–10)

5.4 (4.6–6.3)

4.0 (3.5–4.5)

< 0.001

3.7 (2.6–4.7)

3.7 (3.0–4.4)

0.710

TJC (28 joints)

9.0 (4.0–14.0)

3.0 (2.0–5.0)

< 0.001

3.0 (1.0–7.0)

2.0 (1.0–5.0)

0.490

SJC (28 joints)

6.0 (2.0–9.5)

2.0 (1.0–4.0)

< 0.001

2.0 (0.0–4.0)

2.0 (0.0–3.0)

0.498

ESR (mm/h)

28.0 (16.5–46.5)

22.0 (13.0–33.0)

< 0.001

16.5 (6.5–32.0)

19.0 (11.0–30.5)

0.052

CRP (mg/l)

15.0 (7.9–28.9)

5.7 (2.5–13.7)

< 0.001

4.7 (1.6–17.0)

5.0 (2.3–11.3)

0.766

SDAI (0–86)

30.2 (19.7–39.5)

13.9 (10.7–18.3)

< 0.001

13.8 (8.0–20.9)

11.3 (7.7–17.4)

0.093

PTGA (0–100)

61.0 (50.0–75.0)

40.0 (26.0–50.0)

< 0.001

36.0 (25.0–60.0)

33.0 (20.0–50.0)

0.044

MDGA (0–100)

58.0 (40.0–70.0)

30.0 (20.0–40.0)

< 0.001

25.0 (15.0–45.0)

25.0 (15.0–40.0)

0.812

HAQ-DI (0–3)

1.5 (1.1–1.9)

1.3 (0.9–1.6)

< 0.001

1.3 (0.9–1.9)

1.3 (0.9–1.6)

0.140

EQ-5D (-0.59–1)

0.2 (0.1–0.7)

0.7 (0.5–0.7)

< 0.001

0.6 (0.1–0.7)

0.7 (0.5–0.8)

0.017

Concomitant csDMARDs

98 (79.0%)

414 (84.3%)

0.159

94 (75.8%)

407 (82.9%)

0.070

Concomitant MTX

79 (63.7%)

341 (69.5%)

0.220

77 (62.1%)

332 (67.6%)

0.245

Concomitant GCs

95 (76.6%)

374 (76.2%)

0.918

92 (74.2%)

370 (75.4%)

0.789

DAS28-ESR 28-joint disease activity score with ESR; TJC tender joint count; SJC swollen joint count; ESR erythrocyte sedimentation rate; CRP C-reactive protein; SDAI Simplified Disease Activity Index; PTGA patient general assessment of disease activity; MDGA physician general assessment of disease activity; HAQDI Health Assessment Questionnaire; EQ-5D EuroQol 5 Dimension for measuring the quality of life; csDMARDs conventional synthetic disease-modifying anti-rheumatic drugs; MTX methotrexate; GCs glucocorticoids
Continuous variables are described through the median (interquartile range); categorical variables are characterised by n (%).


Table 3 Description of patients from C3 and C4 cohort at 6-month visit after applying propensity score matching

 

C3 (n=75)

C4 (n=75)

P-value

Female, n (%)*

60 (80.0%)

61 (81.3%)

0.836

Age at diagnosis, years, median (IQR)

45.0 (36.0–53.0)

45.0 (37.0–53.0)

0.678

Age at start of 1st line, years, median (IQR)*

52.0 (45.0–61.0)

55.0 (44.0–61.0)

0.811

Disease duration, years, median (IQR)*

5.0 (2.4–12.7)

5.8 (3.0–13.1)

0.937

RF positive, n (%)*

60 (80.0%)

54 (72.0%)

0.251

Presence of comorbidities, n (%)*

54 (72.0%)

55 (73.3%)

0.855

Currently smoking, n (%)*

21 (28.0%)

21 (28.0%)

1.000

Number of previous csDMARDs, n (%)*

 

 

 

0

2 (2.7%)

0 (0.0%)

0.230

1

16 (21.3%)

15 (20.0%)

2

20 (26.7%)

28 (37.3%)

3

17 (22.7%)

20 (26.7%)

4+

20 (26.7%)

12 (16.0%)

Glucocorticoids in previous history, n (%)*

67 (89.3%)

66 (88.0%)

0.797

Concomitant csDMARDs, n (%)*

61 (81.3%)

63 (84.0%)

0.666

Concomitant GCs, n (%)*

56 (74.7%)

55 (73.3%)

0.852

DAS28-ESR (0–10), median (IQR)

5.0 (4.2–5.9)

5.0 (4.1–5.7)

0.717

TJC (28 joints), median (IQR)*

8.0 (4.0–12.0)

6.0 (3.0–11.0)

0.677

SJC (28 joints), median (IQR)*

4.0 (2.0–8.0)

4.0 (2.0–7.0)

0.973

ESR (mm/h), median (IQR)*

27.0 (15.0–37.0)

25.0 (12.0–41.0)

0.844

CRP (mg/l), median (IQR)*

15.0 (8.0–22.2)

8.4 (3.5–25.7)

0.090

SDAI (0–86), median (IQR)

25.5 (15.6–34.9)

22.7 (16.1–30.9)

0.531

PTGA (0–100), median (IQR)*

60.0 (40.0–71.0)

50.0 (40.0–71.0)

0.519

MDGA (0–100), median (IQR)

55.0 (35.0–70.0)

45.0 (30.0–60.0)

0.059

HAQ-DI (0–3), median (IQR)*

1.5 (1.1–1.9)

1.5 (1.1–1.9)

0.877

EQ-5D (-0.59–1), median (IQR)a

0.2 (0.1–0.7)

0.6 (0.1–0.7)

0.290

IQR interquartile range; RF rheumatoid factor; csDMARDs conventional synthetic disease-modifying anti-rheumatic drugs; GCs glucocorticoids; DAS28-ESR 28-joint disease activity score with ESR; TJC tender joint count; SJC swollen joint count; ESR erythrocyte sedimentation rate; CRP C-reactive protein; SDAI Simplified Disease Activity Index; PTGA patient general assessment of disease activity; MDGA physician general assessment of disease activity; HAQ‑DI Health Assessment Questionnaire; EQ-5D EuroQol 5 Dimension for measuring the quality of life
a n=74 (C3), n=75 (C4)

* These parameters were included in the propensity score model.