Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3341 2022-06-15 13:30:30 |
2 format change -19 word(s) 3322 2022-06-16 03:59:16 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Gul, H.; Harnden, K.; Saleem, B. Strategies for Achieving Drug-Free Remission in Rheumatoid Arthritis. Encyclopedia. Available online: https://encyclopedia.pub/entry/24066 (accessed on 19 May 2024).
Gul H, Harnden K, Saleem B. Strategies for Achieving Drug-Free Remission in Rheumatoid Arthritis. Encyclopedia. Available at: https://encyclopedia.pub/entry/24066. Accessed May 19, 2024.
Gul, Hanna, Kate Harnden, Benazir Saleem. "Strategies for Achieving Drug-Free Remission in Rheumatoid Arthritis" Encyclopedia, https://encyclopedia.pub/entry/24066 (accessed May 19, 2024).
Gul, H., Harnden, K., & Saleem, B. (2022, June 15). Strategies for Achieving Drug-Free Remission in Rheumatoid Arthritis. In Encyclopedia. https://encyclopedia.pub/entry/24066
Gul, Hanna, et al. "Strategies for Achieving Drug-Free Remission in Rheumatoid Arthritis." Encyclopedia. Web. 15 June, 2022.
Strategies for Achieving Drug-Free Remission in Rheumatoid Arthritis
Edit

Rheumatoid arthritis (RA) is a chronic immune-mediated systemic disease, which affects approximately 1% of the population and is characterized by a symmetrical inflammatory polyarthropathy.  It has been demonstrated that drug-free remission (DFR) is possible in a proportion of RA patients achieving clinically defined remission (both on cs and b-DMARDS). Immunological, imaging and clinical associations with/predictors of DFR have all been identified, including the presence of autoantibodies, absence of Power Doppler (PD) signal on ultrasound (US), lower disease activity according to composite scores of disease activity and lower patient-reported outcome scores (PROs) at treatment cessation. 

rheumatoid arthritis remission drug-free remission b-DMARDs cs-DMARDs tapering

1. Introduction

Rheumatoid arthritis (RA) is a chronic immune-mediated systemic disease, which affects approximately 1% of the population and is characterized by a symmetrical inflammatory polyarthropathy [1].
Over recent years, there has been a paradigm shift in the treatment approach in RA from cautious escalation of therapies for symptomatic relief to the early and rapid control of inflammation soon after diagnosis, aimed to prevent structural damage and preserve function. This is in accordance with the ‘window of opportunity’ hypothesis, which suggests that in early RA, aggressive treatment can reverse underlying autoimmunity and induce immune tolerance (thus potentially modifying the disease course) [2]. In clinical practice, this is achieved using a treat-to-target (T2T) strategy. This strategy involves strict monitoring of disease activity using composite measures, e.g., disease activity score (DAS28) resulting in successive escalation of immunosuppressive agents (conventional synthetic and biologic disease-modifying drugs (cs-DMARDs and b-DMARDs, respectively). These drugs are used alone or in combination and with or without corticosteroids to control inflammation [3].
Experience with b-DMARD tapering is largely with tumor necrosis factor inhibitors (TNFis). Tapering of cs-DMARDs, notably methotrexate (MTX), is also desirable for patients concerned about long-term side-effects and the burden of taking tablets/self-injecting if they are well [4][5]. These frequently lead to poor treatment compliance, with approximately 15% of patients self-discontinuing treatment, which itself can lead to increased disease morbidity [5][6].

2. Defining Remission in RA

To be able to identify individuals who are more likely to achieve DFR, we first need to be able to define remission accurately. Remission in RA is currently defined clinically using a cut-off of the DAS28 (disease activity score). It incorporates a mathematical formula comprising the number of tender and swollen joints out of 28 (TJC28, SJC28), a serum marker of inflammation (e.g., C-reactive protein, CRP) and an optional measure of patients’ assessment of global health status (PGA) [7].
DAS28-remission has been defined as a score of <2.6 [8][9]. It is the standard measure used in clinical practice; however, it is not a precise assessment of remission. This score and tender joint count assessment may be influenced by physical comorbidities, e.g., osteoarthritis or psychosocial factors. Swollen joint counts may also be inaccurate in remission [10], while objective serological inflammatory markers (ESR and CRP) are non-specific to RA. Furthermore, the DAS28 joint count excludes the feet and ankles, therefore missing active disease in these areas [6]. It has been shown that some patients in remission do still have evidence of subclinical synovitis on musculoskeletal ultrasound (US) [11][12][13][14].
There have been multiple attempts to define clinical remission more stringently, including the ACR/EULAR 2011 Boolean remission criteria (TJC28, SCJ28, CRP and PGA all ≤1) [15][16], CDAI (TJC + SJC + PGA + Physician GA: remission = 0.0–2.8) [17] and SDAI (TJC + SJC + PGA + Physician GA + CRP: remission is ≤5) [18] scores (comprehensive and simplified disease activity scores, respectively); however, these still include subjective measures and potentially inaccurate joint counts [15][18]. The concept of ‘deep’ clinical remission has been considered (DAS28 < 1.98), which is suggested to reflect the absence of biological inflammation; however, longitudinal outcome data relating to this target have not yet been studied prospectively [19].
Physical examination is known to have a low sensitivity for the detection of mild synovitis, such as that found in clinical remission; however, musculoskeletal US has proven to be an excellent tool to identify subclinical inflammation that is associated with risk of relapse and structural damage [20][21][22]. Despite this, the definition of what constitutes imaging remission remains challenging [13][22][23]. More recently, immunological status has been shown to predict the likelihood of sustained remission in RA [24][25]. This adds another potential dimension to consider when defining the remission state in RA.
Schett et al. [26] have recently introduced the concept of ‘multi-level’ remission aimed to characterize remission more precisely (Figure 1). It involves the achievement of different levels/depths of remission. It suggests that a state of deep remission may be attained if all three categories are achieved; however, this has not yet been used prospectively.
Figure 1. Shell model of remission states [26].

3. DFR Remission in Patients with RA Treated with cs-DMARDs

DMARDs are indicated for the treatment of inflammatory arthritis, e.g., RA; however, they are also used to treat other disorders [27]. cs-DMARDs are typically used as first-line agents, alone or in combination. Commonly used cs-DMARDs include methotrexate (MTX), hydroxychloroquine (HCQ), leflunomide (LEF) and sulfasalazine (SSZ). They are mostly oral preparations (except for MTX, which can also be injected subcutaneously) [28].
Some of the earliest data on withdrawing cs-DMARDS come from historical observational studies. These studies often focus on older conventional cs-DMARDs, which are no longer used in first-line RA treatment, e.g., gold and d-penicillamine [29][30]. It has been demonstrated that DFR is possible in a minority of cases. Most of the evidence for discontinuing cs-DMARDs to achieve sustained DFR comes from randomized controlled trials (RCTs) for patients with stable RA on a range of monotherapies [29][31][32][33][34]. Many of the DMARDs studied, however, are now rarely used in practice. Additional evidence comes from RCTs and observational studies in which a step-down approach in treatment was followed (combination DMARDs reduced to monotherapy). These demonstrated sustained clinical response to treatment after tapering in early RA patients [35][36][37][38].
Table 1 summarizes the studies discussed.
Table 1. cs-DMARD DFR remission studies.

Study

Design

Authors

n

Treatment/Intervention

RA Disease Duration

Remission Criteria

%DFR Remission

DFR-Predicting Factors

Follow Up Period

Can disease-modifying anti-rheumatic drugs be discontinued in long standing rheumatoid arthritis? A 15-year follow-up

Observational

Tiippana et al., 2010

70

Single or combination Cs-DMARDS tapered

Early RA

5/6 ARA criteria fulfilled.

16%

N/A

15 years

Prevalence and predictive factors for sustained disease-modifying antirheumatic drug-free remission in rheumatoid arthritis: results from two large early arthritis cohorts

Observational

van der Woude et al., 2009

Leiden EAC cohort: 454

British EAC Cohort: 895

Single or combination Cs-DMARDS tapered

(MTX/SSZ/HcQ)

Early RA

Had to fulfil 3 criteria:

(1) No current use of DMARDs/corticosteroids, (2) No swollen joints, and (3) Classification as DMARD-free remission by the patient’s rheumatologist.

Leiden EAC cohort: 15%

British EAC Cohort: 9.4%

Absence of autoantibodies ((ACPA and IgM-RF) and short symptom duration at presentation

Minimum of 1 year after discontinuation of DMARD therapy

KIMERA

Observational

Jung et al., 2020

234

Single or combination therapy with cs DMARDs; methotrexate (MTX)/sulfasalazine combined with high-dose glucocorticoid; MTX combined with TNF-inhibitors tapered

Early RA

(1) Non-use of cs or bDMARDs and glucocorticoids, (2) DAS28 <2.6, and (3) no swollen joints.

46.1%

Early RA and lower disease activity (DAS28 <2.26) at csDMARD withdrawal

48 months

Randomized placebo-controlled study of stopping second-line drugs in RA

RCT

Ten Wolde et al., 1996

285

Placebo or withdrawal of at least one 2nd line cs-DMARD (chloroquine, HCQ, gold, d-penicillamine, SSZ, AZA or MTX)

Established RA. Median duration 8–9 years.

5/6 ARA criteria fulfilled

62%

Lower maintenance dose of second line drug and absence of RF

52 weeks

D-penicillamine withdrawal in rheumatoid arthritis

Double blind RCT

Ahern et al., 1984

38

Tapering of d-penicillamine

Established RA (6–11 years)

5/6 ARA criteria fulfilled

21%

None

12 months

BeST

Multi center randomized single blind trial

Markusse et al., 2015

508

MTX/combination cs DMARD/ combination cs-DMARD +prednisolone/combination cs DMARD with MTX and Infliximab

Early disease (symptom duration < 2 years)

DAS44 <1.6

14%

Absence of ACPA and using MTX rather than SSZ as the last csDMARD before withdrawal

10 years

tREACH

RCT

Kuijper et al., 2016

281

Triple cs-DMARD (MTX, SSZ and HCQ) with glucocorticoid bridging or MTX monotherapy with glucocorticoid bridging

TNFi and MTX if the DAS28 was >2.4.

Early RA

DAS28 <1.6

2.4%

N/A

2 year

IMPROVED

RCT

Heimans et al., 2016

610

MTX and prednisolone, then tapered

Early RA or Undifferentiated arthritis

DAS44 <1.6

21%

Absence of ACPA

2 year

BioRRA

Interventional cohort study

Baker et al., 2019

44

Cessation of cs-DMARDs

Established RA

DAS28-CRP < 2.4

48%

Absence of RF, shorter time from diagnosis to starting first DMARD, shorter symptom duration at time of diagnosis, longer disease duration fulfilment of ACR/EULAR Boolean remission criteria and longer time since last DMARD change

Absence of genes within peripheral CD4+ T cells; FAM102B and ENSG00000227070

Presence of gene within peripheral CD4+ T cells: ENSG00000228010

6 months

4. Predicting DFR for Patients Receiving Treatment with cs-DMARDs

Several factors have been demonstrated to predict the successful maintenance of remission after cs-DMARD withdrawal to achieve DFR. cs-DMARD-free remission is more likely to be achieved when T2T strategies have been employed with the goal of establishing remission earlier in the RA disease course. This supports the window of opportunity hypothesis for RA treatment [2]. In addition, other factors associated with cs-DMARD-free remission include a longer duration of sustained remission prior to drug withdrawal [13], the absence of autoantibodies (ACPA and RF) [31][39][40] and lower disease activity (DAS28 < 2.6) at the time of treatment cessation [39][40][41][42]. Using methotrexate as the last cs-DMARD before withdrawal has also been associated with a higher chance of achieving DFR [40][43].
The BioRRA study [41] is the most comprehensive study of biomarkers for predicting cs-DMARD remission to date. Baker et al. developed a composite score for the prediction of DFR including circulating inflammatory biomarkers, and peripheral CD4+ T-cell gene expression. This score was able to differentiate future flare from DFR with an AUROC (receiver–operator characteristic) of 0.96 (95% CI 0.91–1.00), sensitivity 0.91 (0.78–1.00) and specificity 0.95 (0.84–1.00). Limitations of the study include small patient numbers and the heterogeneity of cs-DMARDs included. Ultrasound biomarkers were not identified.
Overall, it has been shown that cs-DMARD-free remission can occur in 14–48% of patients achieving remission, although discontinuing cs-DMARDS carries a 38–80% risk of disease flare. This is probably unacceptably high for both patients and clinicians alike due to the negative impact of flare on QoL [44] and the risk of disease progression [45]; however, it has been reassuringly demonstrated that most patients can re-capture remission following treatment for flare. Identifying patients who can achieve successful withdrawal of cs-DMARDs remains an area of unmet clinical need, despite reports of potential predictive factors in the literature. However, progress in this field is promising.

5. DFR Remission in Patients with RA Treated with Biological Therapies (b-DMARDs)

b-DMARDs can target and inhibit specific pathways of the immune system and inflammatory cascade, each with a unique mechanism of action. TNFis include: etanercept (ETN), adalimumab (ADA), infliximab (IFX), golimumab (GOL) and certolizumab-pegol (CZP). Other agents include rituximab (RTX, anti-CD19 agent), abatacept (ABA, humanized fusion antibody), tocilizumab (TCZ, anti-IL6) and small molecule Janus Kinase (JAK) inhibitors, e.g., baricitinib/tofacitinib, amongst others [27].
Several studies have analyzed the effects of b-DMARD (mainly TNFi) withdrawal in RA patients after a successful remission induction regime and will be discussed. The remission induction regime varies between studies involving different drugs. Furthermore, there is inconsistency in the definitions of remission used; duration of remission; and the duration RA: from DMARD naïve to established RA. Often, drug tapering or successful dose reduction may be the primary outcome. To date, there is no evidence in the literature for discontinuation of RTX or JAK inhibitors.
Table 2 summarizes the studies discussed.
Table 2. Biologic DFR remission studies.

Study

Design

Authors

n

Treatment/Intervention

Drug Withdrawn in Italics

RA Disease Duration

Remission Criteria

%DFR Remission in Biologic Treatment Arm

DFR Predicting Factors

Follow Up Period

IVEA

Double blind RCT

Quinn MQ et al., 2006

20

1. Infliximab + MTX

2. MTX

6 months

DAS28

70

-

12 months

BeSt

RCT

van den Broek M et al., 2011

128

4th study arm: Combination with infliximab

23 months

DAS44

56

Lower baseline HAQ

ACPA negative

Lower baseline disease activity

Younger age

Non-smoker

24 months

IDEA

Double blind RCT

Nam JL et al., 2014

112

1. Infliximab +MTX

2. MTX + single dose IV methylprednisolone

78 weeks

DAS44

76%

-

78 weeks

HONOR

Open label non randomized

Yamaguchi A et al., 2020

52

Adalimumab

7 years

DAS28

21

A baseline DAS28 of <2.22 or <1.98

Shorter disease duration

60 months

RRR *

Observational

Tanaka Y et al., 2010

114

Infliximab

6 years

LDA

55

A baseline DAS28 of <2.22 or <1.98

12 months

OPTIMA

RCT

Smolen J et al., 2013

1032

Adalimumab + MTX

≤12 months

DAS28

66%

Good baseline functional status

52 weeks

PRIZE

Double blind RCT

Emery P et al., 2014

306

1. ½ dose Etanercept + MTX

2. Placebo + MTX

3. Placebo alone

≤12 months

DAS2

23–40%

-

39 weeks

CERTAIN

Double blind RCT

Smolen J et al., 2015

194

1. Certolizumab + MTX

2. Placebo

6 months–10 years

CDAI

18.8%

-

52 weeks

Patients with RA in remission on TNF blockers: when and in whom can TNF blocker therapy be stopped?

Observational

Saleem et al., 2011

47

TNFi (Various) + MTX

1. Initial therapy

2. Delayed therapy

12 months

DAS28

59%15%

Male gender

First line TNFi

Shorter disease duration

Higher and naïve T-cells and fewer IRCs at baseline

24 months

EMPIRE

Double blind RCT

Nam et al., 2013

110

1. Etanercept + MTX

2. MTX + placebo

≤3 months

DAS28

28.1%

Starting TNFi earlier in disease course

52 weeks

TARA

Single blind RCT

Van Mulligen et al., 2020

189

94 DMARD

95 TNFi

TNFi or csDMARD (Various)

1. csDMARD taper first

2. TNFi taper first

Not stated

DAS44

15%

-

24 months

AVERT

Double blind RCT

Emery P et al., 2015

351

Abatacept + MTX

<1 year

DAS28

15%

Lower baseline PRO scores

18 months

DREAM

Observational

Nishimoto N et al., 2014

187

Tocilizumab

7.8 years

LDA

9%

Lower multi-biomarker assay scores (serological)

RF negative

12 months

ACT RAY

RCT

Huizinga TW et al., 2015

556

Tocilizumab

8 years

DAS28

6%

Shorter disease duration, few/absent erosions

12 months

RETRO

RCT

Haschka J et al., 2016

101

Various

NK

DAS28

48.1%

ACPA negative

Lower baseline disease activity

Male gender

Lower multi-biomarker assay scores (serological)

RF negative

12 months

PredictRA

Double blind RCT

Emery et al., 2020

122

Adalimumab taper vs. withdrawal

Mean 12.9 years

DAS28

55% (withdrawal arm)

-

36 weeks

ANSWER

Cohort

Hashimoto et al., 2018

181

Various

NK

DAS28

21.5%

Boolean remission at baseline

Sustained remission period

No glucocorticoid use at time of discontinuation

TNFi discontinuation (vs. other b-DMARD)

12 months

* NK = not known.

6. Predictors of DFR for Patients Receiving Treatment with b-DMARDs

6.1. Clinical and Demographic Variables

Although several potential biomarkers of b-DMARD-free remission have been reported, validated measures are yet to be identified. Shorter disease duration [46][47][48][49][50], fewer or absence of erosions [51][52] and low disease activity at baseline [46][53][54][55][56][57] have been consistently associated with successful discontinuation in several studies.
There is potential reversibility of autoimmunity in early disease. Subsequently, remission induction during this phase can increase the chance of successful b-DMARD discontinuation. This reversibility decreases with time, following which chronic synovitis ensues, in addition to persistent cytokine abnormalities, which can lead to structural progression. Thus, the efficacy of treatment may be reduced for patients with longer disease duration, resulting in only moderate clinical benefit and a reduced chance for DFR [58]. This concept is supported by the observation that treatment responses in the first 3 months following diagnosis can predict the later achievement of remission [59].

6.2. Patient Reported Outcomes (PRO) Measures

Lower PRO scores at baseline have been shown to be associated with a better chance of successful maintenance of remission after b-DMARD withdrawal to achieve DFR. Good baseline functional status at ADA discontinuation (assessed by standardized patient questionnaires) has been shown to be predictive of low disease activity in the OPTIMA trial [60] and worsening functional disability has been shown to be associated with disease flare [19]. These findings are supported by the AVERT study [61], where lower baseline HAQ (health assessment questionnaire) was associated with successful DFR. The BeST study also found that lower HAQ score was associated with sustained DFR remission in their DAS-guided tapering cohort [39].

6.3. Imaging Variables

Musculoskeletal US has been shown to be a reliable method of predicting relapse in patients in clinical remission [62]; therefore, there is interest in using this tool to identify patients who may be able to taper or discontinue biologic therapy. Studies have revealed that the presence of synovitis (measured using power Doppler (PD) assessment) could predict failure of b-DMARD tapering for RA patients in clinical remission and that PD was a good predictor of disease flare within six months of tapering [63][64][65]. Additionally, grey scale synovial hypertrophy (a measure of damage secondary to prior inflammation) is predictive of flare. Furthermore, El Miedany et al. [19] concluded that US was superior to DAS28 in predicting relapse for RA patients in remission, and both PD synovitis and synovial hypertrophy were independent predictors of relapse. Interestingly, Alivernini et al. [66] found that PD synovitis correlated with the histological characteristics of synovial tissue in established RA patients, thus suggesting that US, when combined with clinical remission criteria, could be a useful tool to identify patients likely to achieve DFR.
In light of these findings, US assessment, either alone or in combination with clinical measures, could evaluate remission more objectively and could help identify the best candidates for b-DMARD tapering, towards DFR [48]. MRI findings, e.g., bone marrow oedema, can also identify subclinical synovitis in RA remission and has been shown to be predictive of structural progression [13][67][68][69].

6.4. Immunological Variables

To date, the best studied predictor of relapse on tapering/discontinuation of b-DMARDs is ACPA positivity. This indicated higher risk of relapse following dose reduction and lower chances of maintaining remission status [40][55][70][71].
IGM-RF was also associated with a reduced chance of TNFi-free remission [19][55][72][73][74].
Immune dysregulation is key to the pathogenesis of RA. Inflammation has a direct effect on T-cell differentiation and promotes the differentiation and proliferation of naïve CD4+ T-cells towards an abnormal phenotype. Characteristically, there is dysregulation of pro-inflammatory CD4+ T-helper cell subsets (naïve, regulatory (Treg) and inflammation-related cells (IRC)) [75][76]. Abnormalities in T-cell subsets have been found across the spectrum of RA and can predict progression, from ‘at-risk’ individuals to evolving RA and those in clinical remission [25]. In a study comparing the characteristics of 47 patients undergoing TNFi tapering, Saleem et al. [48] found that sustained remission was associated with T-cell subset immunological abnormalities. Patients who sustained remission for 24 months presented a higher frequency (%) of naïve T-cells and lower frequency of IRCs. Furthermore, the frequency of Treg cells was higher in the sustained remission group. These proportions were different for the patients receiving early, aggressive treatment compared to delayed treatment, for whom Treg frequency was higher.

6.5. Serum Biomarkers and Multi-Biomarker Assays

Multi-biomarker disease activity (MDBA) assays, developed to identify subclinical inflammation at the molecular level, have been investigated in several studies of RA patients in clinical remission. In general, studies have found that MDBA scores may be elevated in patients deemed to be in remission according to conventional clinical definitions [77][78][79][80]. These patients were also found to have a higher risk of structural joint damage [4][80].
One such score involves a total of 12 inflammation parameters, including markers linked to the acute phase [77]. It was initially developed and validated to correlate with the DAS28CRP score. Two studies have demonstrated that the score is better at predicting radiological progression than the DAS28CRP score [78][81]. In patients with high baseline MBDA scores at discontinuation of TNFi in the POET study, discontinuation may have allowed a recurrence of residual subclinical inflammation and the need to recommence TNFi treatment [50].
Collectively, these findings indicate that evaluating subclinical inflammation using serum biomarkers may be a useful tool to determine risk of flare/high risk candidates in whom tapering or discontinuation of therapy should not be initiated. Validation of this work is required.

6.6. Deep/Multi-Level Remission

As previously described, it is thought that achieving deep clinical remission is required to facilitate DFR [26].
Building on this, Gul et al. [82] aimed to define remission more precisely using a multi-dimensional model of remission using clinical, US and T-cell subset measures (for patients treated with either cs or b-DMARDs). In this cross-sectional study, considerable heterogeneity of DAS28 remission was observed with respect to these characteristics, with some patients showing evidence of high inflammatory markers and joint counts, evidence of synovitis on PD US and persistent T-cell subset abnormalities (which should not be present in remission). Definitions for clinical, US and T-cell subset remission were created and the achievement of all three was thought to represent a state of complete remission (multi-dimensional remission (MDR)). Out of approximately 200 patients, only 30% satisfied the criteria for MDR. These patients were found to have lower PRO scores. Further work has resulted in the development of a predictive model for successful tapering (towards DFR) of cs-DMARDs and work is underway to replicate this in a cohort of patients undergoing tapering and discontinuation of b-DMARDs [83]. This could help inform tapering strategies in clinical practice.

References

  1. Lee, D.M.; Weinblatt, M.E. Rheumatoid arthritis. Lancet 2001, 358, 903–911.
  2. Quinn, M.A.; Emery, P. Window of opportunity in early rheumatoid arthritis: Possibility of altering the disease process with early intervention. Clin. Exp. Rheumatol. 2003, 21, S154–S157.
  3. Smolen, J.S.; Aletaha, D.; Bijlsma, J.W.; Breedveld, F.C.; Boumpas, D.; Burmester, G.; Combe, B.; Cutolo, M.; de Wit, M.; Dougados, M.; et al. Treating rheumatoid arthritis to target: Recommendations of an international task force. Ann. Rheum. Dis. 2010, 69, 631–637.
  4. Hughes, L.D.; Done, J.; Young, A. A 5 item version of the Compliance Questionnaire for Rheumatology (CQR5) successfully identifies low adherence to DMARDs. BMC Musculoskelet. Disord. 2013, 14, 286.
  5. Grijalva, C.G.; Chung, C.P.; Arbogast, P.G.; Stein, C.M.; Mitchel, E.F., Jr.; Griffin, M.R. Assessment of adherence to and persistence on disease-modifying antirheumatic drugs (DMARDs) in patients with rheumatoid arthritis. Med. Care 2007, 45, S66–S76.
  6. Betegnie, A.L.; Gauchet, A.; Lehmann, A.; Grange, L.; Roustit, M.; Baudrant, M.; Bedouch, P.; Allenet, B. Why Do Patients with Chronic Inflammatory Rheumatic Diseases Discontinue Their Biologics? An Assessment of Patients’ Adherence Using a Self-report Questionnaire. J. Rheumatol. 2016, 43, 724–730.
  7. Cruyssen, B.; Looy, S.; Wyns, B.; Westhovens, R.; Durez, P.; Van den Bosch, F.; Veys, E.; Mielants, H.; Clerck, L.; Peretz, A.; et al. DAS28 best reflects the physician’s clinical judgment of response to infliximab therapy in rheumatoid arthritis patients: Validation of the DAS28 score in patients under infliximab treatment. Arthritis Res. Ther. 2005, 7, R1063–R1071.
  8. Van der Heijde, D.M.; van’t Hof, M.A.; van Riel, P.L.; Theunisse, L.A.; Lubberts, E.W.; van Leeuwen, M.A.; van Rijswijk, M.H.; van de Putte, L.B. Judging disease activity in clinical practice in rheumatoid arthritis: First step in the development of a disease activity score. Ann. Rheum. Dis. 1990, 49, 916–920.
  9. Van der Maas, A.; Lie, E.; Christensen, R.; Choy, E.; de Man, Y.A.; van Riel, P.; Woodworth, T.; den Broeder, A.A. Construct and criterion validity of several proposed DAS28-based rheumatoid arthritis flare criteria: An OMERACT cohort validation study. Ann. Rheum. Dis. 2013, 72, 1800–1805.
  10. Saleem, B.; Brown, A.K.; Keen, H.; Nizam, S.; Freeston, J.; Wakefield, R.; Karim, Z.; Quinn, M.; Hensor, E.; Conaghan, P.G.; et al. Should imaging be a component of rheumatoid arthritis remission criteria? A comparison between traditional and modified composite remission scores and imaging assessments. Ann. Rheum. Dis. 2011, 70, 792–798.
  11. Saleem, B.; Brown, A.K.; Keen, H.; Nizam, S.; Freeston, J.; Karim, Z.; Quinn, M.; Wakefield, R.; Hensor, E.; Conaghan, P.G.; et al. Disease remission state in patients treated with the combination of tumor necrosis factor blockade and methotrexate or with disease-modifying antirheumatic drugs: A clinical and imaging comparative study. Arthritis Rheum. 2009, 60, 1915–1922.
  12. Saleem, B.; Nizam, S.; Emery, P. Can remission be maintained with or without further drug therapy in rheumatoid arthritis? Clin. Exp. Rheumatol. 2006, 24, S33–S36.
  13. Brown, A.K.; Conaghan, P.G.; Karim, Z.; Quinn, M.A.; Ikeda, K.; Peterfy, C.G.; Hensor, E.; Wakefield, R.J.; O’Connor, P.J.; Emery, P. An explanation for the apparent dissociation between clinical remission and continued structural deterioration in rheumatoid arthritis. Arthritis Rheum. 2008, 58, 2958–2967.
  14. Brown, A.K.; Quinn, M.A.; Karim, Z.; Conaghan, P.G.; Peterfy, C.G.; Hensor, E.; Wakefield, R.J.; O’Connor, P.J.; Emery, P. Presence of significant synovitis in rheumatoid arthritis patients with disease-modifying antirheumatic drug-induced clinical remission: Evidence from an imaging study may explain structural progression. Arthritis Rheum. 2006, 54, 3761–3773.
  15. Felson, D.T.; Smolen, J.S.; Wells, G.; Zhang, B.; van Tuyl, L.H.; Funovits, J.; Aletaha, D.; Allaart, C.F.; Bathon, J.; Bombardieri, S.; et al. American College of Rheumatology/European League Against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical trials. Arthritis Rheum. 2011, 63, 573–586.
  16. Singh, J.A.; Saag, K.G.; Bridges, S.L., Jr.; Akl, E.A.; Bannuru, R.R.; Sullivan, M.C.; Vaysbrot, E.; McNaughton, C.; Osani, M.; Shmerling, R.H.; et al. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol. 2016, 68, 1–26.
  17. Aletaha, D.; Nell, V.P.; Stamm, T.; Uffmann, M.; Pflugbeil, S.; Machold, K.; Smolen, J.S. Acute phase reactants add little to composite disease activity indices for rheumatoid arthritis: Validation of a clinical activity score. Arthritis Res. Ther. 2005, 7, R796–R806.
  18. Bykerk, V.P.; Massarotti, E.M. The new ACR/EULAR remission criteria: Rationale for developing new criteria for remission. Rheumatol. 2012, 51 (Suppl. 6), 16–20.
  19. El Miedany, Y.; El Gaafary, M.; Youssef, S.; Ahmed, I.; Bahlas, S.; Hegazi, M.; Nasr, A. Optimizing therapy in inflammatory arthritis: Prediction of relapse after tapering or stopping treatment for rheumatoid arthritis patients achieving clinical and radiological remission. Clin. Rheumatol. 2016, 35, 2915–2923.
  20. Ben Abdelghani, K.; Miladi, S.; Souabni, L.; Kassab, S.; Chekili, S.; Laatar, A.; Zakraoui, L. Role of ultrasound in assessing remission in rheumatoid arthritis. Diagn. Interv. Imaging 2015, 96, 3–10.
  21. Nguyen, H.; Ruyssen-Witrand, A.; Gandjbakhch, F.; Constantin, A.; Foltz, V.; Cantagrel, A. Prevalence of ultrasound-detected residual synovitis and risk of relapse and structural progression in rheumatoid arthritis patients in clinical remission: A systematic review and meta-analysis. Rheumatology 2014, 53, 2110–2118.
  22. Raffeiner, B.; Grisan, E.; Botsios, C.; Stramare, R.; Rizzo, G.; Bernardi, L.; Punzi, L.; Ometto, F.; Doria, A. Grade and location of power Doppler are predictive of damage progression in rheumatoid arthritis patients in clinical remission by anti-tumour necrosis factor alpha. Rheumatology 2017, 56, 1320–1325.
  23. Peluso, G.; Michelutti, A.; Bosello, S.; Gremese, E.; Tolusso, B.; Ferraccioli, G. Clinical and ultrasonographic remission determines different chances of relapse in early and long standing rheumatoid arthritis. Ann. Rheum. Dis. 2011, 70, 172–175.
  24. Saleem, B.; Brown, A.K.; Quinn, M. Prediction offlare and long-term outcome in DMARD treated RA patients in remission: The value of imaging and new remission criteria. Ann. Rheum. Dis. 2011, 70, 88.
  25. Ponchel, F.; Burska, A.N.; Hunt, L.; Gul, H.; Rabin, T.; Parmar, R.; Buch, M.H.; Conaghan, P.G.; Emery, P. T-cell subset abnormalities predict progression along the Inflammatory Arthritis disease continuum: Implications for management. Sci. Rep. 2020, 10, 3669.
  26. Schett, G.; Emery, P.; Tanaka, Y.; Burmester, G.; Pisetsky, D.S.; Naredo, E.; Fautrel, B.; van Vollenhoven, R. Tapering biologic and conventional DMARD therapy in rheumatoid arthritis: Current evidence and future directions. Ann. Rheum. Dis. 2016, 75, 1428–1437.
  27. Benjamin, O.; Bansal, P.; Goyal, A.; Lappin, S.L. Disease Modifying Anti-Rheumatic Drugs (DMARD); StatPearls Publishing LLC.: Treasure Island, FL, USA, 2021.
  28. Abbasi, M.; Mousavi, M.J.; Jamalzehi, S.; Alimohammadi, R.; Bezvan, M.H.; Mohammadi, H.; Aslani, S. Strategies toward rheumatoid arthritis therapy; the old and the new. J. Cell. Physiol. 2019, 234, 10018–10031.
  29. Ahern, M.J.; Hall, N.D.; Case, K.; Maddison, P.J. D-penicillamine withdrawal in rheumatoid arthritis. Ann. Rheum. Dis. 1984, 43, 213–217.
  30. Van der Leeden, H.; Dijkmans, B.A.; Hermans, J.; Cats, A. A double-blind study on the effect of discontinuation of gold therapy in patients with rheumatoid arthritis. Clin. Rheumatol. 1986, 5, 56–61.
  31. ten Wolde, S.; Breedveld, F.C.; Hermans, J.; Vandenbroucke, J.P.; van de Laar, M.A.; Markusse, H.M.; Janssen, M.; van den Brink, H.R.; Dijkmans, B.A. Randomised placebo-controlled study of stopping second-line drugs in rheumatoid arthritis. Lancet 1996, 347, 347–352.
  32. Kremer, J.M.; Rynes, R.I.; Bartholomew, L.E. Severe flare of rheumatoid arthritis after discontinuation of long-term methotrexate therapy. Double-blind study. Am. J. Med. 1987, 82, 781–786.
  33. Gøtzsche, P.C.; Hansen, M.; Stoltenberg, M.; Svendsen, A.; Beier, J.; Faarvang, K.L.; Wangel, M.; Rydgren, L.; Halberg, P.; Juncker, P.; et al. Randomized, placebo controlled trial of withdrawal of slow-acting antirheumatic drugs and of observer bias in rheumatoid arthritis. Scand. J. Rheumatol. 1996, 25, 194–199.
  34. De Silva, M.; Hazleman, B.L. Long-term azathioprine in rheumatoid arthritis: A double-blind study. Ann. Rheum. Dis. 1981, 40, 560–563.
  35. Landewé, R.B.; Boers, M.; Verhoeven, A.C.; Westhovens, R.; van de Laar, M.A.; Markusse, H.M.; van Denderen, J.C.; Westedt, M.L.; Peeters, A.J.; Dijkmans, B.A.; et al. COBRA combination therapy in patients with early rheumatoid arthritis: Long-term structural benefits of a brief intervention. Arthritis Rheum. 2002, 46, 347–356.
  36. Mottonen, T.; Hannonen, P.; Leirisalo-Repo, M.; Nissila, M.; Kautiainen, H.; Korpela, M.; Laasonen, L.; Julkunen, H.; Luukkainen, R.; Vuori, K.; et al. Comparison of combination therapy with single-drug therapy in early rheumatoid arthritis: A randomised trial. FIN-RACo trial group. Lancet 1999, 353, 1568–1573.
  37. Marchesoni, A.; Battafarano, N.; Arreghini, M.; Panni, B.; Gallazzi, M.; Tosi, S. Radiographic progression in early rheumatoid arthritis: A 12-month randomized controlled study comparing the combination of cyclosporin and methotrexate with methotrexate alone. Rheumatology 2003, 42, 1545–1549.
  38. Klarenbeek, N.B.; Koevoets, R.; van der Heijde, D.M.; Gerards, A.H.; Ten Wolde, S.; Kerstens, P.J.; Huizinga, T.W.; Dijkmans, B.A.; Allaart, C.F. Association with joint damage and physical functioning of nine composite indices and the 2011 ACR/EULAR remission criteria in rheumatoid arthritis. Ann. Rheum. Dis. 2011, 70, 1815–1821.
  39. Van der Woude, D.; Young, A.; Jayakumar, K.; Mertens, B.J.; Toes, R.E.; van der Heijde, D.; Huizinga, T.W.; van der Helm-van Mil, A.H. Prevalence of and predictive factors for sustained disease-modifying antirheumatic drug-free remission in rheumatoid arthritis: Results from two large early arthritis cohorts. Arthritis Rheum. 2009, 60, 2262–2271.
  40. Klarenbeek, N.B.; van der Kooij, S.M.; Guler-Yuksel, M.; van Groenendael, J.H.; Han, K.H.; Kerstens, P.J.; Huizinga, T.W.; Dijkmans, B.A.; Allaart, C.F. Discontinuing treatment in patients with rheumatoid arthritis in sustained clinical remission: Exploratory analyses from the BeSt study. Ann. Rheum. Dis. 2011, 70, 315–319.
  41. Baker, K.F.; Skelton, A.J.; Lendrem, D.W.; Scadeng, A.; Thompson, B.; Pratt, A.G.; Isaacs, J.D. Predicting drug-free remission in rheumatoid arthritis: A prospective interventional cohort study. J. Autoimmun. 2019, 105, 102298.
  42. Kuijper, T.M.; Luime, J.J.; de Jong, P.H.; Gerards, A.H.; van Zeben, D.; Tchetverikov, I.; de Sonnaville, P.B.; van Krugten, M.V.; Grillet, B.A.; Hazes, J.M.; et al. Tapering conventional synthetic DMARDs in patients with early arthritis in sustained remission: 2-year follow-up of the tREACH trial. Ann. Rheum. Dis. 2016, 75, 2119–2123.
  43. Van Mulligen, E.; Weel, A.E.A.M.; Kuijper, T.M.; Hazes, J.M.W.; van der Helm- van Mil, A.H.M.; de Jong, P.H.P. The impact of a disease flare during tapering of DMARDs on the lives of rheumatoid arthritis patients. Semin. Arthritis Rheum. 2020, 50, 423–431.
  44. Flurey, C.A.; Morris, M.; Richards, P.; Hughes, R.; Hewlett, S. It’s like a juggling act: Rheumatoid arthritis patient perspectives on daily life and flare while on current treatment regimes. Rheumatology 2014, 53, 696–703.
  45. Markusse, I.M.; Dirven, L.; Gerards, A.H.; van Groenendael, J.H.; Ronday, H.K.; Kerstens, P.J.; Lems, W.F.; Huizinga, T.W.; Allaart, C.F. Disease flares in rheumatoid arthritis are associated with joint damage progression and disability: 10-year results from the BeSt study. Arthritis Res. Ther. 2015, 17, 232.
  46. Van den Broek, M.; Huizinga, T.W.; Dijkmans, B.A.; Allaart, C.F. Drug-free remission: Is it already possible? Curr. Opin. Rheumatol. 2011, 23, 266–272.
  47. Yamaguchi, A.; Hirata, S.; Kubo, S.; Fukuyo, S.; Hanami, K.; Nakano, K.; Nakayamada, S.; Saito, K.; Tanaka, Y. 5-year remission rate after the discontinuation of adalimumab in patients with rheumatoid arthritis: Long-term follow-up results of the HONOR study. Mod. Rheumatol. 2020, 30, 799–806.
  48. Saleem, B.; Keen, H.; Goeb, V.; Parmar, R.; Nizam, S.; Hensor, E.M.; Churchman, S.M.; Quinn, M.; Wakefield, R.; Conaghan, P.G.; et al. Patients with RA in remission on TNF blockers: When and in whom can TNF blocker therapy be stopped? Ann. Rheum. Dis. 2010, 69, 1636–1642.
  49. Tanaka, Y.; Hirata, S.; Kubo, S.; Fukuyo, S.; Hanami, K.; Sawamukai, N.; Nakano, K.; Nakayamada, S.; Yamaoka, K.; Sawamura, F.; et al. Discontinuation of adalimumab after achieving remission in patients with established rheumatoid arthritis: 1-year outcome of the HONOR study. Ann. Rheum. Dis. 2015, 74, 389–395.
  50. Ghiti Moghadam, M.; Vonkeman, H.E.; Ten Klooster, P.M.; Tekstra, J.; van Schaardenburg, D.; Starmans-Kool, M.; Brouwer, E.; Bos, R.; Lems, W.F.; Colin, E.M.; et al. Stopping Tumor Necrosis Factor Inhibitor Treatment in Patients with Established Rheumatoid Arthritis in Remission or With Stable Low Disease Activity: A Pragmatic Multicenter, Open-Label Randomized Controlled Trial. Arthritis Rheumatol. 2016, 68, 1810–1817.
  51. Westhovens, R.; Robles, M.; Ximenes, A.C.; Wollenhaupt, J.; Durez, P.; Gomez-Reino, J.; Grassi, W.; Haraoui, B.; Shergy, W.; Park, S.H.; et al. Maintenance of remission following 2 years of standard treatment then dose reduction with abatacept in patients with early rheumatoid arthritis and poor prognosis. Ann. Rheum. Dis. 2015, 74, 564–568.
  52. Huizinga, T.W.; Conaghan, P.G.; Martin-Mola, E.; Schett, G.; Amital, H.; Xavier, R.M.; Troum, O.; Aassi, M.; Bernasconi, C.; Dougados, M. Clinical and radiographic outcomes at 2 years and the effect of tocilizumab discontinuation following sustained remission in the second and third year of the ACT-RAY study. Ann. Rheum. Dis. 2015, 74, 35–43.
  53. Van den Broek, M.; Klarenbeek, N.B.; Dirven, L.; van Schaardenburg, D.; Hulsmans, H.M.; Kerstens, P.J.; Huizinga, T.W.; Dijkmans, B.A.; Allaart, C.F. Discontinuation of infliximab and potential predictors of persistent low disease activity in patients with early rheumatoid arthritis and disease activity score-steered therapy: Subanalysis of the BeSt study. Ann. Rheum. Dis. 2011, 70, 1389–1394.
  54. Tanaka, Y.; Takeuchi, T.; Mimori, T.; Saito, K.; Nawata, M.; Kameda, H.; Nojima, T.; Miyasaka, N.; Koike, T. Discontinuation of infliximab after attaining low disease activity in patients with rheumatoid arthritis: RRR (remission induction by Remicade in RA) study. Ann. Rheum. Dis. 2010, 69, 1286–1291.
  55. Haschka, J.; Englbrecht, M.; Hueber, A.J.; Manger, B.; Kleyer, A.; Reiser, M.; Finzel, S.; Tony, H.P.; Kleinert, S.; Feuchtenberger, M.; et al. Relapse rates in patients with rheumatoid arthritis in stable remission tapering or stopping antirheumatic therapy: Interim results from the prospective randomised controlled RETRO study. Ann. Rheum. Dis. 2016, 75, 45–51.
  56. Kavanaugh, A.; Smolen, J.S. The when and how of biologic agent withdrawal in rheumatoid arthritis: Learning from large randomised controlled trials. Clin. Exp. Rheumatol. 2013, 31, S19–S21.
  57. Kavanaugh, A.; Lee, S.J.; Curtis, J.R.; Greenberg, J.D.; Kremer, J.M.; Soto, L.; Etzel, C.J.; Cox, V.; Yoshida, K.; Reed, G.W.; et al. Discontinuation of tumour necrosis factor inhibitors in patients with rheumatoid arthritis in low-disease activity: Persistent benefits. Data from the Corrona registry. Ann. Rheum. Dis. 2015, 74, 1150–1155.
  58. Boers, M. Understanding the window of opportunity concept in early rheumatoid arthritis. Arthritis Rheum. 2003, 48, 1771–1774.
  59. Aletaha, D.; Funovits, J.; Keystone, E.C.; Smolen, J.S. Disease activity early in the course of treatment predicts response to therapy after one year in rheumatoid arthritis patients. Arthritis Rheum. 2007, 56, 3226–3235.
  60. Smolen, J.S.; Emery, P.; Fleischmann, R.; van Vollenhoven, R.F.; Pavelka, K.; Durez, P.; Guérette, B.; Kupper, H.; Redden, L.; Arora, V.; et al. Adjustment of therapy in rheumatoid arthritis on the basis of achievement of stable low disease activity with adalimumab plus methotrexate or methotrexate alone: The randomised controlled OPTIMA trial. Lancet 2014, 383, 321–332.
  61. Emery, P.; Burmester, G.R.; Bykerk, V.P.; Combe, B.G.; Furst, D.E.; Barré, E.; Karyekar, C.S.; Wong, D.A.; Huizinga, T.W. Evaluating drug-free remission with abatacept in early rheumatoid arthritis: Results from the phase 3b, multicentre, randomised, active-controlled AVERT study of 24 months, with a 12-month, double-blind treatment period. Ann. Rheum. Dis. 2015, 74, 19–26.
  62. Saleem, B.; Brown, A.K.; Quinn, M.; Karim, Z.; Hensor, E.M.; Conaghan, P.; Peterfy, C.; Wakefield, R.J.; Emery, P. Can flare be predicted in DMARD treated RA patients in remission, and is it important? A cohort study. Ann. Rheum. Dis. 2012, 71, 1316–1321.
  63. Scire, C.A.; Montecucco, C.; Codullo, V.; Epis, O.; Todoerti, M.; Caporali, R. Ultrasonographic evaluation of joint involvement in early rheumatoid arthritis in clinical remission: Power Doppler signal predicts short-term relapse. Rheumatology 2009, 48, 1092–1097.
  64. Naredo, E.; Valor, L.; De la Torre, I.; Montoro, M.; Bello, N.; Martinez-Barrio, J.; Martinez-Estupinan, L.; Nieto, J.C.; Ovalles-Bonilla, J.G.; Hernandez-Florez, D.; et al. Predictive value of Doppler ultrasound-detected synovitis in relation to failed tapering of biologic therapy in patients with rheumatoid arthritis. Rheumatology 2015, 54, 1408–1414.
  65. Iwamoto, T.; Ikeda, K.; Hosokawa, J.; Yamagata, M.; Tanaka, S.; Norimoto, A.; Sanayama, Y.; Nakagomi, D.; Takahashi, K.; Hirose, K.; et al. Prediction of relapse after discontinuation of biologic agents by ultrasonographic assessment in patients with rheumatoid arthritis in clinical remission: High predictive values of total gray-scale and power Doppler scores that represent residual synovial inflammation before discontinuation. Arthritis Care Res. 2014, 66, 1576–1581.
  66. Alivernini, S.; Peluso, G.; Fedele, A.L.; Tolusso, B.; Gremese, E.; Ferraccioli, G. Tapering and discontinuation of TNF-α blockers without disease relapse using ultrasonography as a tool to identify patients with rheumatoid arthritis in clinical and histological remission. Arthritis Res. Ther. 2016, 18, 39.
  67. McQueen, F.M.; Benton, N.; Perry, D.; Crabbe, J.; Robinson, E.; Yeoman, S.; McLean, L.; Stewart, N. Bone edema scored on magnetic resonance imaging scans of the dominant carpus at presentation predicts radiographic joint damage of the hands and feet six years later in patients with rheumatoid arthritis. Arthritis Rheum. 2003, 48, 1814–1827.
  68. Benton, N.; Stewart, N.; Crabbe, J.; Robinson, E.; Yeoman, S.; McQueen, F.M. MRI of the wrist in early rheumatoid arthritis can be used to predict functional outcome at 6 years. Ann. Rheum. Dis. 2004, 63, 555–561.
  69. Foltz, V.; Gandjbakhch, F.; Etchepare, F.; Rosenberg, C.; Tanguy, M.L.; Rozenberg, S.; Bourgeois, P.; Fautrel, B. Power Doppler ultrasound, but not low-field magnetic resonance imaging, predicts relapse and radiographic disease progression in rheumatoid arthritis patients with low levels of disease activity. Arthritis Rheum. 2012, 64, 67–76.
  70. Detert, J.; Bastian, H.; Listing, J.; Weiß, A.; Wassenberg, S.; Liebhaber, A.; Rockwitz, K.; Alten, R.; Krüger, K.; Rau, R.; et al. Induction therapy with adalimumab plus methotrexate for 24 weeks followed by methotrexate monotherapy up to week 48 versus methotrexate therapy alone for DMARD-naive patients with early rheumatoid arthritis: HIT HARD, an investigator-initiated study. Ann. Rheum. Dis. 2013, 72, 844–850.
  71. Lamers-Karnebeek, F.B.; Luime, J.J.; Ten Cate, D.F.; Teerenstra, S.; Swen, N.; Gerards, A.H.; Hendrikx, J.; van Rooyen, E.M.; Voorneman, R.; Haagsma, C.; et al. Limited value for ultrasonography in predicting flare in rheumatoid arthritis patients with low disease activity stopping TNF inhibitors. Rheumatology 2017, 56, 1560–1565.
  72. Fautrel, B.; Pham, T.; Alfaiate, T.; Gandjbakhch, F.; Foltz, V.; Morel, J.; Dernis, E.; Gaudin, P.; Brocq, O.; Solau-Gervais, E.; et al. Step-down strategy of spacing TNF-blocker injections for established rheumatoid arthritis in remission: Results of the multicentre non-inferiority randomised open-label controlled trial (STRASS: Spacing of TNF-blocker injections in Rheumatoid ArthritiS Study). Ann. Rheum. Dis. 2016, 75, 59–67.
  73. Nishimoto, N.; Amano, K.; Hirabayashi, Y.; Horiuchi, T.; Ishii, T.; Iwahashi, M.; Iwamoto, M.; Kohsaka, H.; Kondo, M.; Matsubara, T.; et al. Drug free REmission/low disease activity after cessation of tocilizumab (Actemra) Monotherapy (DREAM) study. Mod. Rheumatol. 2014, 24, 17–25.
  74. Tanaka, Y.; Hirata, S. Intensive intervention can lead to a treatment holiday from biological DMARDs in patients with rheumatoid arthritis. Drugs 2014, 74, 2129–2139.
  75. Cope, A.P. T cells in rheumatoid arthritis. Arthritis Res. Ther. 2008, 10 (Suppl. 1), S1.
  76. Ponchel, F.; Morgan, A.W.; Bingham, S.J.; Quinn, M.; Buch, M.; Verburg, R.J.; Henwood, J.; Douglas, S.H.; Masurel, A.; Conaghan, P.; et al. Dysregulated lymphocyte proliferation and differentiation in patients with rheumatoid arthritis. Blood 2002, 100, 4550–4556.
  77. Rech, J.; Hueber, A.J.; Finzel, S.; Englbrecht, M.; Haschka, J.; Manger, B.; Kleyer, A.; Reiser, M.; Cobra, J.F.; Figueiredo, C.; et al. Prediction of disease relapses by multibiomarker disease activity and autoantibody status in patients with rheumatoid arthritis on tapering DMARD treatment. Ann. Rheum. Dis. 2016, 75, 1637–1644.
  78. Van der Helm-van Mil, A.H.; Knevel, R.; Cavet, G.; Huizinga, T.W.; Haney, D.J. An evaluation of molecular and clinical remission in rheumatoid arthritis by assessing radiographic progression. Rheumatology 2013, 52, 839–846.
  79. Hambardzumyan, K.; Bolce, R.; Saevarsdottir, S.; Cruickshank, S.E.; Sasso, E.H.; Chernoff, D.; Forslind, K.; Petersson, I.F.; Geborek, P.; van Vollenhoven, R.F. Pretreatment multi-biomarker disease activity score and radiographic progression in early RA: Results from the SWEFOT trial. Ann. Rheum. Dis. 2015, 74, 1102–1109.
  80. Li, W.; Sasso, E.H.; Emerling, D.; Cavet, G.; Ford, K. Impact of a multi-biomarker disease activity test on rheumatoid arthritis treatment decisions and therapy use. Curr. Med. Res. Opin. 2013, 29, 85–92.
  81. Curtis, J.R.; van der Helm-van Mil, A.H.; Knevel, R.; Huizinga, T.W.; Haney, D.J.; Shen, Y.; Ramanujan, S.; Cavet, G.; Centola, M.; Hesterberg, L.K.; et al. Validation of a novel multibiomarker test to assess rheumatoid arthritis disease activity. Arthritis Care Res. 2012, 64, 1794–1803.
  82. Gul, H.L.; Eugenio, G.; Rabin, T.; Burska, A.; Parmar, R.; Wu, J.; Ponchel, F.; Emery, P. Defining remission in rheumatoid arthritis: Does it matter to the patient? A comparison of multi-dimensional remission criteria and patient reported outcomes. Rheumatology 2020, 59, 613–621.
  83. Gul, H.; Ponchel, F.; Emery, P. OP0182 IN RA PATIENTS IN REMISSION, WHICH BIOMARKERS PREDICT SUCCESSFUL TAPERING OF CSDMARDS? Ann. Rheum. Dis. 2021, 80, 110.
More
Information
Subjects: Rheumatology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 470
Revisions: 2 times (View History)
Update Date: 16 Jun 2022
1000/1000