Skip to main content
Sage Choice logoLink to Sage Choice
. 2024 Oct 1;62(1):3–21. doi: 10.1177/00045632241285843

Biomarkers in the diagnosis, prognosis and management of rheumatoid arthritis: A comprehensive review

Didem Sahin 1,2, Andrea Di Matteo 1,2, Paul Emery 1,2,
PMCID: PMC11707974  PMID: 39242085

Abstract

Rheumatoid arthritis (RA) is a chronic, systemic, autoimmune condition that primarily affects the joints and periarticular soft tissues. In the past two decades, the discovery of new biomarkers has contributed to advances in the understanding of the pathogenesis and natural history of RA. These biomarkers, including genetic, clinical, serological and imaging biomarkers, play a key role in the different stages and aspects of RA, from the so called ‘pre-clinical RA’, which is characterized by subclinical pathological events, such as autoimmunity and inflammation, to diagnosis (including differential diagnosis), treatment decision making and disease monitoring.

This review will provide an overview on the current role of traditional and newer biomarkers in the main aspects of RA management, from the identification of individuals ‘at-risk’ of RA who are likely to progress to clinically evident disease, to ‘early’ diagnosis of RA, prognosis, precision medicine, and prediction of response to treatment.

Keywords: Rheumatoid arthritis, biomarkers, autoantibodies, cytokines

Introduction

Rheumatoid arthritis (RA) is a chronic, systemic, autoimmune condition that primarily affects the joints and peri-articular soft tissues. 1 The prevalence of RA is approximately 1% in the adult population. 1 Females are more affected than males with a ratio of 2.45. 2 RA has significant negative impact on patient’s quality of life. Indeed, if left untreated, RA leads to irreversible joint damage, development of extra-articular manifestations, disability, and increased mortality.

An early diagnosis and prompt treatment initiation, especially when poor prognostic factors are present, are crucial for optimal management of RA patients. 3 In 2010, the American College of Rheumatology (ACR) and the European Alliance of Associations for Rheumatology (EULAR) updated their classification criteria for RA; four main domains were delineated, including joint involvement (i.e. number of joints and small vs large joints), serology (i.e. RA-related autoantibodies), acute phase reactants (APRs), such as increased C-reactive protein (CRP) and/or erythrocyte sedimentation rate (ESR), and duration of symptoms (i.e. more or less than 6 weeks).4,5

The development of a swollen joint is traditionally regarded as the beginning of RA. In recent years, the concept of RA as a ‘disease continuum’ has emerged. According to this concept, RA starts with a pre-clinical phase, in which individuals ‘at-risk’ of this disease (because of the presence of genetic/environmental risk factors) go through different phases of autoimmunity and sub-clinical inflammation, before evolving into a chronic (and clinically evident) disease state. 6 This RA ‘disease continuum’ encompasses complex disease mechanisms and diversity in immune cell profiles among individual patients. In the past decade, multiple studies have attempted to prevent the development of RA (or delay its onset) in individuals ‘at-risk’, with some encouraging results. 7 An important aspect to take into consideration is that not all ‘at-risk’ individuals will develop clinical RA. Therefore, the identification of reliable biomarkers is crucial for the risk-stratification of these individuals (i.e. differentiation between those at low-risk and those at high-risk of developing the disease) and their management, including consideration for participation in prevention trials.

The management of RA has undergone significant advances over the last two decades. Our understanding of the RA pathogenesis has improved considerably, so has the ability to diagnose RA patients early, using biomarkers including serum and imaging. Cutting-edge treatments have been developed to selectively inhibit specific immune cells or cytokines, making a treat-to-target approach feasible. Nevertheless, in some patients, achieving a status of disease remission can be challenging, highlighting the complexity and heterogeneity of RA. Addressing this heterogeneity is essential for optimizing the therapeutic management of RA patients (i.e. precision medicine).

Therefore, the field of biomarker research in RA holds great promise for improving patients long-term outcomes. From new genes being discovered to autoantibodies, serology and advanced imaging techniques, the range of available biomarkers continues to expand. This review will provide an overview on the status of traditional and new biomarkers in the diagnosis (including the identification of ‘at-risk’ individuals) and management of RA, including prognosis, precision medicine, and treatment monitoring. In addition, the role of each biomarker in different stages of RA continuum are summarized in Table 1.

Table 1.

The role of each biomarker in RA.

Pre-clinical RA Diagnosis Prognosis
Genetic biomarkers Increased risk of RA development
Autoantibody production
No routine use Possible association with radiographic progression
Conflicting data for treatment response
Association with extra-articular manifestations (e.g. Felty syndrome and rheumatoid vasculitis)
Autoantibodies Precede development of musculoskeletal symptoms and subclinical synovitis on US
Potential link with bone loss
Increase risk of RA development
2010 ACR/EULAR classification criteria (RF/ACPA) Poor prognostic factor (i.e. association with joint damage, high disease activity, less drug-free remission and extra-articular manifestations)
Possible association with response to individual bDMARDs
APRs Increased RA risk (especially ESR) 2010 ACR/EULAR classification criteria (CRP/ESR) Poor prognostic factor (i.e. active disease, association with joint damage, and less drug-free remission)
Part of disease activity scores (i.e. DAS 28-ESR)
Weak correlation with PROs
Cytokines Increased serum levels before clinical disease
No clear association with RA development
No routine use
Increased levels compared to other arthritides/healthy controls
Joint damage
High disease activity
Target molecules for bDMARDs (i.e. TNFi and IL-6 inhibitor)
14-3-3η No clear role No routine use
Positive correlation with autoantibodies
Possible association with radiographic progression, high disease activity, and response to tocilizumab
MicroRNAs Increased serum levels before clinical disease No routine use
Increased levels compared to other arthritides/healthy controls
High disease activity
Treatment response to DMARDs (i.e. TNFi/csDMARD combination and rituximab)
MMPs No clear role No routine use
Increased levels compared to other arthritides/healthy controls
Association with joint damage, high disease activity
T cells Increased RA risk (result of the dysregulation) No clinical use Association with treatment response (i.e. methotrexate), drug-free remission
Imaging Increased RA risk (subclinical synovitis-tenosynovitis-erosions) a 1987 ACR classification criteria (erosions-periarticular osteopenia on X-Ray)
Additional criteria 2010 ACR/EULAR classification criteria
Helpful in differential diagnosis
Joint damage evaluation b
Joint damage prediction
Treatment response to DMARDs
Consider intensive medication when erosions present
Increased risk of flares
Less drug-free remission (when subclinical inflammation on US)

Abbreviations: APRs: Acute phase reactants. ACPA: Anti-citrullinated protein antibodies. ACR: American College of Rheumatology. bDMARDs: biological disease-modifying antirheumatic drugs.csDMARDs: conventional synthetic DMARDs. CRP: C-reactive protein. DAS28: Disease activity score 28. ESR: Erythrocyte sedimentation rate. csDMARD: EULAR: European Alliance of Associations for Rheumatology. IL-6: Interleukin-6. miR: microRNAs. MMPs: Matrix metalloproteinases. PRO: Patient reported outcomes. RA: Rheumatoid arthritis. RF: Rheumatoid factor. TNFi: Tumour necrosis factor inhibitors. US: Ultrasound.

aUltrasound and/or magnetic resonance imaging detected.

bX-Ray, ultrasound or magnetic resonance imaging.

Biomarkers

Genetic biomarkers

The likelihood of developing RA is influenced by genetic factors. Multiethnic genome-wide association studies revealed more than 100 genetic loci associated with RA, highlighting the polygenic nature of the disease.811 The familial heritability of RA is around 60%, and is more prominent in younger and seropositive RA patients than in older and seronegative patients (i.e. 50% for anti-citrullinated protein antibodies (ACPA) positive RA and 20% for ACPA negative RA).8,10 The HLA-DRB1 allele, also called shared epitope (HLA-SE), has been traditionally regarded as the most important genetic risk factor for RA development.12,13 HLA-SE has a strong association with ACPA, and studies have shown an allele-dose effect on the levels of these antibodies.1418 Smoking exposure in combination with two copies of HLA-SE increases significantly (20-fold) the risk of developing RA.14,16,19,20 Nevertheless, HLA-SE explains only 18% of the genetic variance in ACPA positive RA, and only 2.4% of ACPA negative RA. 21 On the other hand, some HLA-DRB1 alleles have showed a protective effect on the disease development, such as HLA-DRB1*1301, HLA-DRB1*1302, and DERAA-encoding HLA-DRB1 alleles.2225

The presence of HLA-SE has been associated with radiographic progression.2628 A previous study highlighted a robust association between the presence of valine at position 11 of the HLA-DRB1 gene and increased susceptibility to radiological damage. 29 On the other hand, the impact of HLA-DRB1 presence on the response to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) and biological DMARDs (bDMARDs) remains uncertain.2934 Similarly, while one study found no correlation between HLA-SE and disease remission with csDMARD therapy, another demonstrated that the absence of HLA-SE predicted DMARD-free remission in RA patients.31,35 Finally, certain HLA-DRB1 alleles were found to be associated with RA extra-articular manifestations (i.e. Felty’s syndrome and rheumatoid vasculitis) and increased mortality.29,36,37

Other than the HLA-SE, the protein tyrosine phosphatase non-receptor type 22 (PTPN22) gene has been reported to be an important genetic risk factor for RA. PTPN22 encodes for lymphoid tyrosine phosphatase (LYP), which is a 110-kd protein and a critical regulator of T-cells, B-cells and other immune cells activation. 38 The single nucleotide polymorphism (SNP) rs2476601 in the PTPN22 gene has been consistently associated with an increased risk of RA.39,40 This variant results in the substitution of arginine with tryptophan at position 620 (R620W) in the LYP protein. 38 The altered function of LYP has been implicated in dysregulated immune responses, contributing to the pathogenesis (via augmented T cell signalling) of RA. In a retrospective study on blood donors, the polymorphism in PTPN22 along with ACPA positivity predicted future development of RA, with a specificity of 100%. 41 Variable predictive values were reported for the progression from undifferentiated arthritis to RA.4244 PTPN22 was found to be associated with an earlier disease onset and ACPA positivity in multiple studies,18,45 as well as with baseline radiographic erosions and radiographic progression. 46 However, no correlation was found between this gene and treatment response to methotrexate or tumour necrosis factor inhibitors (TNFi).47,48

Other genes, which could potentially play a role in RA disease susceptibility, include TNFAIP3, PADI4, STAT4, TRAF1C5, and CTLA4. In addition, epigenetic mechanisms, such as DNA methylation and histone acetylation, and microRNAs, have also been implicated in the pathogenesis pathways (i.e. regulator of immune cell development and function) that are involved in the RA disease onset and perpetuation.49,50

Autoantibodies

Anti-cyclic citrullinated peptide antibodies and rheumatoid factor

Autoantibodies play a key role in the pathogenesis of RA. Different autoantibodies target different and specific antigens. ACPAs and rheumatoid factor (RF) represent specific and early serological markers for the diagnosis of RA. ACPAs are a group of antibodies directed against proteins (such as filaggrin, vimentin, α-enolase, and fibrinogen) that have undergone citrullination, which is a post-translational modification where the amino acid arginine is converted to citrulline. 51 On the other hand, RF is an autoantibody primarily of the IgM class, which targets the Fc portion of IgG antibodies, forming immune complexes that can contribute to tissue damage and inflammation. For ACPA detection, second-generation anti-cyclic citrullinated peptide (CCP2) assay is widely used in Europe, while third generation anti-cyclic citrullinated peptide (CCP3) are commonly used in the United States. The technological differences between CCP2 and CCP3 assays are not completely known as they’ve not been disclosed by the companies (e.g. targeted antigens).

ACPA and RF are important diagnostic biomarkers. Indeed, the presence of ACPA and/or RF is one of the four cardinal features of the 2010 ACR/EULAR classification criteria for RA. 4 The frequency of ACPA and/or RF positivity in the general population has been reported to range from 1% to 2.8%.52,53 ACPA showed similar sensitivity but higher specificity than RF for the diagnosis of RA (67% vs 69% and 95% vs 85%, respectively). 54

ACPAs and RF can precede the development of RA by up to 18 years. 55 The detection of these autoantibodies, especially ACPAs, has been associated with an increased risk of developing RA in different ‘at-risk’ populations.5659 In a previous study of our research group involving different ‘at-risk’ individuals with musculoskeletal symptoms, a positive anti-CCP3 antibody test increased significantly the risk of developing RA.60,61 Conversely, a negative anti-CCP3 test in these anti-CCP2 positive individuals decreased remarkably the risk of developing RA, especially in those with high titer anti-CCP2 antibodies. Subsequently, in a similar population of anti-CCP2 positive ‘at-risk’ individuals with musculoskeletal symptoms (with no clinical or sub-clinical joint involvement), anti-CCP3 antibodies were associated with the development of joint inflammation on ultrasound (US). 62 The presence of ACPA has also been linked with bone loss in individuals ‘at-risk’ of RA, long before the onset of clinical synovitis.63,64 Interestingly, in a population-based study including ACPA IgG-positive healthy individuals, the presence of different ACPAs/RF isotypes (e.g. ACPA IgA) and other autoantibodies was associated with the development of clinically suspect arthralgia. 65 ACPAs have also showed a distinct association with different T-cells profiles in patients with early RA. In a study, lower naive and regulatory T cells (Tregs) frequencies were found in ACPA+ ‘at-risk’ individuals than in those without autoantibodies. 66

RA-related antibodies have also been associated with a more aggressive RA disease phenotype, which is characterized by more joint damage, higher disease activity, and extra-articular manifestations.6773 Previous studies have shown that seropositive RA patients could potentially respond better to certain bDMARDs, particularly rituximab and abatacept, and to some extent tocilizumab, than to TNFi.48,7476 Positive results in terms of treatment response have also emerged in seropositive RA patients who are treated with tofacitinib, which is a Jak-inhibitor. 77 Other studies have also revealed that seropositivity was associated with good treatment response, but less long-term drug free remission in early RA patients.78,79 An important aspect to consider is that the levels of antibodies may fluctuate (particularly RF), primarily due to changes in medication intensity, but this has less impact on disease activity indices and long-term outcomes. 80 Additionally, seroconversion (i.e. negativization of autoantibodies) appears to be a very rare occurrence in RA patients, also in those who have achieved persistent and drug-free remission. 81 These results emphasize the dynamic picture of RA pathogenesis and management.

Other novel autoantibodies

Anti-Carbamylated Protein Antibodies (anti-CarP) antibodies target proteins that have undergone non-enzymatic post-translational modification, where cyanate binds to lysine residues, forming homocitrulline. 82 In contrast to ACPAs, no associations have been found between HLA-SE and anti-CarP antibodies, emphasizing the diverse nature of the immune responses and antibody profiles involved in the pathogenesis of RA. 83

Studies in preclinical RA demonstrated that, like RF and ACPAs, anti-CarP antibodies can be identified in the serum of ‘at-risk’ individuals years before the diagnosis of RA. However, adding anti-CarP to conventional antibodies (i.e. RF and ACPAs) did not improve the prediction for progression to RA in a study. 84

Anti-CarP antibodies can be found in 30%–47% of established RA and can be detected in both seropositive and seronegative (i.e. for RF or ACPAs) patients.8589 Anti-CarP seems to play a relevant role especially in ACPA negative patients, where they might be useful for RA classification. 88 In a meta-analysis, triple positivity for RF, ACPAs, and anti-CarP showed a very high specificity for the diagnosis of RA (98%–100%), in spite of a sub-optimal sensitivity (11%–39%). 90

Positive anti-CarP antibodies were found to be associated with worse radiographic outcomes in both ACPA positive and negative RA patients, as well as in individuals ‘at-risk’. 91

Elevated levels of anti-CarP antibodies and anti-peptidylarginine deaminases-4 antibodies (anti-PAD4) were associated with poorer response to TNFi. 92 Another study showed a positive correlation between the presence of anti-CarP antibodies and response to abatacept. 87

Another novel class of autoantibodies identified in RA is anti-PAD4. Increased levels of anti-PAD4 were detected in the serum samples of the patients before RA development. 93 A recent meta-analysis showed a pooled sensitivity of 34% and specificity of 94% for RA diagnosis. 94 When combined with ACPA, a slight improvement in the sensitivity was observed compared to ACPA alone (3.8%). 94 Positive anti-PAD4 was also shown to be associated with radiographic joint damage in RA patients.95,96

Antibodies against other post-translational modifications have also been described, including anti-acetylated protein antibodies and anti-malondialdehyde-acetaldehyde antibodies, mainly in ACPA positive patients.97,98 However, their additional value in comparison to the more traditional anti-CCP and RF is unclear; therefore, they are not used in routine clinical practice.

Acute phase reactants

C-reactive protein and erythrocyte sedimentation rate

CRP is an acute phase protein, which is produced by the liver in response to inflammatory stimuli. In RA, the production of pro-inflammatory cytokines, particularly interleukin-6 (IL-6), leads to an elevation of CRP levels. 99

CRP is a useful biomarker for the diagnosis and assessment of disease activity of RA patients. Indeed, the presence of elevated APR, such as CRP and/or ESR, represents a main domain of the 2010 ACR/EULAR classification criteria for RA. 4 Previous studies have demonstrated that elevated CRP levels may occur in the pre-clinical stages of RA. 100 However, conflicting results were found regarding the predictive value of CRP levels for progression to clinical synovitis in different at-risk populations.57,59,101 This might stem from its short half-life, mainly reflecting the current presence of inflammation rather than more chronic or slowly developing processes.

CRP level is a component of various comprehensive measures of disease activity, such as the Disease Activity Index 28 (DAS28)-CRP score, Simplified Disease Activity Index (SDAI), and ACR/EULAR Boolean definition of remission.102104 Higher serum CRP levels showed a positive correlation with joint inflammation. 105 However, CRP levels are not invariably elevated with RA disease activity. For example, in a large cohort of RA patients with high disease activity according to Clinical Disease Activity Index (CDAI), more than half of these patients had normal CRP levels. 106 Additionally, joint inflammation on histology has been noted in nearly half of RA patients with normal CRP levels. 105 Moreover, CRP and ESR demonstrated a weak correlation with subjective measures of RA disease activity reported by patients, such as early morning stiffness and global pain, and fatigue. 107 In addition, CRP level is not a reliable indicator of inflammatory activity in patients using IL-6 blockers by blocking IL-6 to induce hepatic acute phase response. 108 These results underscore the complexity of RA, and the importance of a multifaceted approach in the assessment and management of RA patients.

Monitoring CRP levels can have a potential utility in the RA prognosis and guiding treatment. Higher baseline CRP levels were linked to radiographic joint damage and disability in different RA populations.71,72,109 According to EULAR, an elevated CRP should be considered in treatment decision-making of RA patients (i.e. consideration of bDMARDs in patients who failed a csDMARD instead of combination therapy with two csDMARDs). 110

ESR evaluates the rate (mm/h) at which red blood cells settle in plasma within a vertical tube. Like CRP, ESR is regarded as a non-specific inflammatory biomarker. However, unlike CRP, ESR levels can also be influenced by other factors, such as age, sex, immunoglobulin levels, and anaemia. 111 Furthermore, ESR response is slower than CRP to resolve due to longer half-life of components. 111

An increased ESR was shown to be independently associated with RA development in a large population of CCP2 + at-risk individuals. 112 Similar to CRP, ESR is used for disease monitoring, as part of the DAS28-ESR score. 103 Elevated ESR levels are regarded as poor prognostic factors in RA patients.73,110 Systematic reviews and meta-analyses showed an association between baseline ESR levels and poorer response to both csDMARD and bDMARD therapy.113,114

In conclusion, APRs such as CRP and ESR play a key role in the diagnosis and monitoring of RA patients. ESR is more complex with multiple causes for elevation but because of this more sensitive at presentation than CRP. However, CRP and ESR levels might differ and contribute separately to predicting outcomes in RA patients. 106 Therefore, in our opinion, it is important in routine practice to assess both these inflammatory markers in RA patients.

Serum calprotectin

Calprotectin, also known as S100A8/A9 or MRP8/14, is a heterodimeric complex composed of two subunits, S100A8 and S100A9, which contributes to chemotaxis, phagocyte migration, and macrophage activation. 115 It has been shown that serum calprotectin levels increase during the preclinical stages of RA. 116 A study by Baillet et al., showed higher calprotectin levels in synovial fluid of RA patients compared to osteoarthritis and other inflammatory arthritides. 117 In addition, higher calprotectin levels were detected in erosive RA compared to non-erosive RA. 118 Several studies have also demonstrated a good correlation between serum calprotectin and disease activity markers, such as DAS28-CRP and US detected synovitis.119122

Data regarding the impact of baseline calprotection levels to treatment response are inconclusive.120,123 In a large cohort study involving 470 RA patients treated with either adalimumab or etanercept, baseline calprotectin levels did not offer any additional value for the prediction of TNFi treatment response over CRP levels. 122 However, in a different study, reduced calprotectin levels after 1 month of bDMARD therapy were associated with better treatment response at 12 months. 119 Other studies explored the link between baseline calprotectin levels and RA flares in RA patients who were in sustained remission, producing controversial results regarding the effectiveness of calprotectin as a predictor for future flares in RA patients.124127

Serum Amyloid A

Serum Amyloid A (SAA) is an acute phase protein which is produced primarily by the liver in response to inflammation. Serum concentration of SAA can increase dramatically (by up to 1000-fold or more) in patients with inflammatory conditions. 128 Higher serum SAA levels were found in RA patients compared to healthy controls and patients with osteoarthritis. 128 SAA also contributes to the formation of the ‘rheumatoid pannus’ (i.e. hyperplasia of the normal synovial tissue, neovascularization, and a heterogeneous inflammatory cell infiltrate) and joint destruction by activating cytokines, inducing adhesion molecules expression and matrix degradation.128,129 In a recent systematic review and meta-analysis, which included a large number of RA patients and healthy controls, SAA levels were found to be higher in RA patients, showing a significant correlation with DAS28, APRs (i.e. ESR and CRP) and inflammatory cytokines in these patients. 130 Furthermore, in the same systematic literature review, it was shown that the SAA 1.3 allele could determine an elevated risk of developing RA in ‘at-risk’ individuals, and RA-associated amyloidosis in patients with established disease. 130

Cytokines

Cytokines, such as tumour necrosis factor α (TNF-α), IL-6, IL-1, IL-8, IL-17, IL-23, and GM-CSF, play a key role in the pathogenesis of RA. Interestingly, abnormal levels of cytokines and chemokines have been found in pre-clinical RA; their increase, however, has been observed later than the development of RA related autoantibodies. 55 Increasing evidence suggests that pro-inflammatory cytokines contribute to initiation and perpetuation of systemic inflammation in ‘at-risk’ individuals. TNF-α and IL-6 have been the most extensively studied and characterized cytokines in the pathogenesis of RA.

TNF-α is produced by macrophages, neutrophils, and activated T cells. TNF-α exerts multiple actions, including activating immune cells, stimulating production of other cytokines, and triggering tissue remodeling and bone resorption via stimulating osteoclasts, thus contributing to the development of bone erosions. 131 Serum levels of TNF-α increase in patients with active RA. 132 Consequently, molecules targeting TNF-α, so called TNFi, such as infliximab, adalimumab, etanercept, certolizumab, and golimumab, have been developed and have been used for more than two decades in RA patients. These agents have revolutionized the outcome of most RA patients, especially those who failed multiple csDMARDs. The safety of TNFi has been documented in several studies, with an increasing risk of infections being the most common side effect of these medications. Interestingly, therapy with TNFi has been associated with reduced cardiovascular risk and better survival rates in RA patients.133136 In addition, a previous study showed that TNFi might induce repair of bone erosions compared to methotrexate. 137

IL-6 is another abundant key cytokine in RA, which shows similar effector functions as TNF-α. 138 Additionally, IL-6 contributes to the production of APRs, such as CRP, thus leading to systemic inflammation. 99 Elevated serum IL-6 levels correlate with high disease activity in RA patients.138,139 Agents inhibiting IL-6 include monoclonal antibodies, such as tocilizumab and sarilumab, which target both soluble and membrane-bound IL-6 receptors. These treatments have been proven to be efficacious and safe options for RA patients, also in monotherapy (i.e. without methotrexate). 136 Multiple studies have also showed the ability of IL-6 inhibition to reduce bone resorption markers, thus potentially inhibiting joint damage and inducing bone erosions repair.140142 Increased IL-6 levels in serum and synovial fluid indicate better responses to IL-6 inhibition.139,143 Additionally, a study including RA patients in remission and those with low disease activity showed that low levels of IL-6 at the time of tocilizumab discontinuation were associated with a low rate of disease relapse at 52 weeks. 144

Nevertheless, neither TNF-α nor IL-6 is used for diagnostic or monitoring purposes in the context of RA on a daily basis, given the fact that its levels are also found to be high in other inflammatory conditions.145,146

14-3-3η proteins

A crucial family of intracellular regulators, 14-3-3 proteins modulate diverse cellular processes by binding to specific proteins, with 14-3-3η eta emerging as a distinctive isoform in this family. Notably, the 14-3-3η exhibited higher levels in both synovial fluid and serum of inflammatory arthritis patients including RA, compared to healthy controls. 147 A subsequent study showed higher 14-3-3η levels in early and established RA patients compared to disease controls and healthy subjects. In this study, elevated 14-3-3η levels correlated with ACPA and RF levels. 148 A recent meta-analysis showed that 14-3-3η proteins had a pooled sensitivity and specificity of 0.73 and 0.88 for the diagnosis of RA, respectively. 149 In addition, 14-3-3η levels were associated with radiographic progression, and this was more evident in treatment-naïve early RA patients than in those with established disease. 150 Another study showed a significant association between 14-3-3η levels and higher disease activity (including elevated APRs) in newly diagnosed RA patients and baseline levels of this protein predicted response to tocilizumab. 151 All these studies highlighted the potential diagnostic and prognostic implications of 14-3-3η in patients with RA.

MicroRNAs

MicroRNAs (miR) are short, non-coding RNA molecules, which are typically composed of about 21 to 23 nucleotides; these molecules regulate post-trascriptional gene expression mainly by binding the 3’ untranslated region of target messenger RNAs, thereby inhibiting their translation or promoting their degradation. MiRNAs have been implicated in various aspects of RA pathogenesis, including immune system dysregulation, inflammation, and joint tissue destruction. Many studies have revealed miRNAs dysregulation in synovial fibroblasts,152,153 macrophages,154,155 and peripheral blood mononuclear cells 156 of RA patients.

In a longitudinal study including ACPA+ ‘at-risk’ individuals, miR-22, miR-382, and miR-486-3p levels increased when patients progressed to clinical disease compared to baseline. In addition, baseline miR-22 levels were higher in progressors to clinical disease compared to non progressors, suggesting that miR-22 could be a potential biomarker for predicting RA development. 157 In a different study, a significant upregulation of miR-103a-3p and miR-346 was observed in ACPA + first-degree relatives of RA patients and patients themselves, as opposed to healthy controls where downregulation of these microRNAs was noted. 158 In another cross-sectional study, miR-126-3p, let-7d-5p, miR-431-3p, miR-221-3p, miR-24-3p, and miR-130a-3p were elevated in ACPA + at-risk individuals and treatment-naïve RA patients compared to healthy controls. 159 In a Japanese study, high plasma concentrations of miR-24 and miR-125a-5p emerged as potential diagnostic biomarkers in both seronegative and seropositive RA patients. 160 Furthermore, the combination of miR-125a-5p, miR-24-3p, and miR-26a-5p was found to have the strongest diagnostic accuracy in Caucasian RA patients. 161 Some miRs, such as miR-24, miR-16, miR-146a, and miR-223, also showed a positive correlation with DAS28 and APRs.160,162,163 Preliminary studies including a small number of patients also showed that miR-23 and miR-223 could potentially predict treatment response to TNFi/csDMARD combination, while other studies observed an association between high levels of miR-125 and response to rituximab.164,165 In a randomized, double-blinded trial, higher baseline miR-27a-3p levels were associated with adalimumab-induced remission at 12 months, but not methotrexate. 166 These studies have showed the promising role of miR in the diagnosis and prognosis of RA patients.

Matrix metalloproteinases

Matrix metalloproteinases (MMPs) constitute a group of zinc-dependent endopeptidases, which are engaged in the physiological processes of tissue remodeling and repair. Dysregulation of MMP activity leads to excessive degradation of collagen, proteoglycans, and other matrix components in the synovium, which is a key contributor to the progressive joint damage observed in RA patients. 167

Approximately 20 distinct MMPs have been identified in humans, with MMP-1 (collagenase-1) and MMP-3 (stromelysin-1) being those most extensively studied in RA. Higher serum levels of MMP-1 and/or MMP-3 have been detected in RA patients compared to healthy controls.168172 In patients with RA, increased serum levels of MMP-3 were associated with higher disease activity, APRs, and more disability.170,172,173 In early RA patients, serum MMP-3 levels at the time of diagnosis showed an association with radiographic joint damage. 169 Subsequent studies demonstrated that higher baseline levels of MMPs in newly diagnosed RA patients were predictive of radiographic damage development.170,173

T cells

Significant T cell abnormalities have been described in various stages of RA. T-cell subsets dysregulations, particularly those that involve CD4 + T cells, CD8 + T cells, and Tregs, play a pivotal role in the initiation and perpetuation of systemic inflammation, and the development of joint damage in RA patients.174176 In a study which included 103 ACPA + at-risk individuals, the inclusion of CD4 + T cells – naïve cells, Tregs, and inflammation-related cells (IRCs) – in a complex statistical model (which also considered clinical, serological, and imaging biomarkers) improved significantly the prediction of inflammatory arthritis development in these individuals.59,177

Changes in T cell subsets have been shown to occur before the development of clinical synovitis in ‘at-risk’ individuals. 177 Interestingly, normal levels of baseline naïve T-cells were associated with higher remission rates in treatment-naïve early RA patients who started methotrexate. 66 In another study, which included RA patients in sustained clinical remission, lower rates of disease reactivation after TNFi discontinuation were observed in RA patients who had normal baseline levels of naïve T-cells and Tregs, and low levels of IRCs levels. 178 In addition, increased levels of baseline IRCs predicted flares in RA patients in clinical remission who underwent csDMARDs tapering. 179 Moreover, some studies have also showed that csDMARDs and bDMARDs can determine a shift in T-cell subsets frequencies, leading to an increase in Tregs levels.180,181 A very recent study demonstrated the value of CD4 + T cell subsets in different stages of the ‘RA continuum’. 182 In this study, T-cells subsets improved prediction of inflammatory arthritis development in individuals ‘at-risk’, and treatment response in those with established disease, including prediction of flares after therapy discontinuation. 182

Imaging biomarkers

X-rays

Conventional radiography (i.e. X-rays) is regarded as the traditional gold standard for the evaluation of structural damage (i.e. bone erosions and cartilage damage) in patients with RA. The presence of radiographic erosions and/or periarticular osteopenia in hand/wrist joints on X-ray was part of the 1987 ACR classification criteria for RA. 5 However, X-rays have limited sensitivity in the detection of early changes (i.e. bone erosions) and soft tissue abnormalities (i.e. synovitis and tenosynovitis). Therefore, other imaging tools with a higher sensitivity for the detection of these findings have taken place in the diagnostic work-up of RA, such as US and magnetic resonance imaging (MRI).183,184 Nevertheless, X-rays are still widely used in current practice and have been designated as the first-line imaging technique for the detection of RA-related joint damage by EULAR. 185

In a study on ‘at-risk’ individuals with anti-CCP2 + antibodies and musculoskeletal symptoms, bone erosions on X-ray were uncommon (4.1%) and not associated with inflammatory arthritis/RA development, thus suggesting that prevention studies with DMARDs should have the potential at least to prevent X-ray damage in these ‘at-risk’ individuals. 186 In patients with undifferentiated arthritis, the presence of ≥2 erosions at baseline increased the risk of progression to RA by 53%. 187 In RA patients, the detection of erosions at the time of diagnosis has been associated with further radiographic progression and poor prognosis.73,110,188 Prospective cohort studies also showed the association of lower baseline Sharp-van der Heijde score (a method used to quantify radiographic joint damage) with higher remission rates at 3 years, including sustained DMARD-free remission.35,189,190

Ultrasound

US can detect synovitis, tenosynovitis, bone erosions, cartilage damage, and tendon tears. 191 Therefore, this imaging tool is widely used in the diagnostic work-up and monitoring of RA patients. US is a bedside tool, which makes it feasible to use in everyday clinical practice. Operator dependency is the main limitation of US.

Several studies have demonstrated the predictive value for future RA development of US-detected sub-clinical inflammation and/or structural damage in different populations of ‘at-risk’ individuals.192,193 In a very recent study from our research group, the detection of US synovitis and/or bone erosions in only three joints (i.e. wrists, knee and MTP5) improved prediction of inflammatory arthritis progression over and above clinical and serological markers in more than 400 CCP2 + at-risk individuals with musculoskeletal symptoms. 194 In addition, synovitis and tenosynovitis were found to be predictor of progression to inflammatory arthritis in different cohorts of ‘at-risk’ individuals with clinically suspect arthralgia.195,196

US has demonstrated to have a higher sensitivity for the detection of bone erosions than X-ray in RA patients.183,184 The presence of bone erosions on US in certain joints (MCP 2 and 5, MTP 5 and ulnar styloid) was sensitive and specific for RA compared to other rheumatic diseases, including psoriatic arthritis (PsA), osteoarthritis, gout, as well as healthy individuals. 197 The development of bone erosions early in the disease course is regarded as a poor prognostic factor for RA by EULAR, which recommends considering a bDMARD in patients with bone erosions who have failed a csDMARD. 110 US has a very important role in the differential diagnosis between RA and other arthritides, such as PsA, crystal arthritis, and connective tissue diseases.198200

The presence of subclinical inflammation on US has been documented in a substantial number of RA patients in clinical remission, showing also an association with an increased risk of flares and structural progression in these patients.201203 A recent systematic literature review showed a pivotal role of US in different aspects of RA patients management (i.e. diagnosis, treat to target and monitoring, including prediction of disease relapse/response to therapy). 204 Multiple studies have documented that US sub-clinical inflammation can predict disease reactivation after csDMARDs or bDMARDs discontinuation in RA patients in clinical remission.179,205208 Despite these promising results, the role of US in the treatment decision strategy of RA patients remains unclear. Indeed, in two recent randomized clinical trials (ARCTIC and TaSER), an US driven treat-to-target strategy was not superior to a conventional (‘clinical based’) approach in RA patients.209,210

Magnetic resonance imaging

MRI scan is able to detect synovitis, tenosynovitis, bone erosions, cartilage damage, and bone marrow oedema (BME), which is also called osteitis. Costs and time are the major drawbacks of MRI use in daily practice.

In ‘at-risk’ patients with clinically suspect arthralgia, the presence of baseline MRI inflammation predicted the development of clinical synovitis, with tenosynovitis being the strongest predictor, while MRI-detected erosions were not predictive for this outcome.57,211 Furthermore, interosseous tendon inflammation on MRI scan was an early feature of ACPA+ ‘at-risk’ individuals in a different study, and it was also detected in early and established RA patients, but not in healthy controls. 212 In a systematic literature review, the sensitivity and specificity of MRI synovitis in the prediction of progression from undifferentiated arthritis to RA was 93% and 25%, respectively. 213 The specificity of BME was high (76%), whereas the sensitivity of this finding was 51%. 213 Higher BME scores at baseline were associated with radiographic progression (i.e. bone erosions development) in multiple studies, which also included RA patients in sustained remission.214217 However, in a randomized clinical trial, a treatment strategy aiming at ‘MRI remission’ did not show superiority to the conventional (i.e. clinically driven) treat-to-target strategy. 218 In addition, while MRI pathological features at RA diagnosis were found not to be predictive of DMARD-free sustained remission in another study, results from AVERT study showed that MRI detected bone erosions were related to flares in patients who tapered therapy with abatacept.219,220

Integrating biomarkers: Multibiomarker disease activity score

The multibiomarker disease activity (MBDA) score system was developed to determine disease activity in RA as it is the cornerstone to reach sustained remission or at least low disease activity according to treat-to-target strategy. 110 MBDA scoring was developed with the combination of the serum level of 12 biomarkers (IL-6, TNF receptor type 1 (TNFR1), vascular cell adhesion molecule 1 (VCAM-1), epidermal growth factor (EGF), vascular EGF A (VEGF-A), bone glycoprotein 39 (YKL-40), MMP-1, MMP-3, CRP, SAA, leptin, and resistin). The equation used in the MBDA scoring algorithm is akin to that of the DAS28-CRP, incorporating the serum level of each biomarker for different components of the DAS28-CRP scoring system. 221 This results in a scale from 0 to 100, with higher scores indicating greater disease activity in RA patients. MBDA score was also found to be correlated with other disease activity indices, such as DAS28-ESR, SDAI, and CDAI. 222 A recent systematic review and meta-analysis demonstrated positive correlations with baseline MBDA scores and baseline DAS28-CRP and -ESR, CDAI, 28 joint-tender and swollen joint counts, CRP, and ESR. 223 High MBDA scores were shown to be strongly related to radiographic damage, independent of other predictors of this outcome, including seropositivity, CRP level and radiographic damage score at baseline.223,224 Overall, the MBDA test represents a promising tool for assessing disease activity in RA, offering a more comprehensive and objective approach compared to individual biomarkers or composite indices.

Conclusion

The current review provides an overview on the role of different biomarkers across the RA continuum, from prediction of inflammatory arthritis development in at-risk individuals, to diagnosis (including differential diagnosis), prognosis, and monitoring of RA.

The discovery and advent of new genetic, serological, and imaging biomarkers have advanced our understanding of the pathogenesis of RA and the natural history of the disease. From this biomarker pool, only autoantibodies such as RF and ACPA, and APRs such as CRP and ESR, and imaging modalities are routinely used in clinical practice for the diagnosis and monitoring of disease activity. However, normal results in these tests do not rule out an RA diagnosis and/or active disease, especially in the early phase. A combination of clinical assessment and multiple tests, especially in patients with initially negative results, is recommended for a more accurate diagnosis and effective monitoring of RA. Consequently, precision medicine remains an elusive concept in Rheumatology, as reliable biomarkers for diagnosis, prognosis, and especially for therapy, have yet to be identified. While challenges remain, ongoing research continues to refine the role of biomarkers in predicting disease progression and treatment response, ultimately improving the quality of care for RA patients.

Acknowledgements

This article was prepared at the invitation of the Clinical Sciences Reviews Committee of the Association for Clinical Biochemistry and Laboratory Medicine.

Footnotes

The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: DS declares no COI. ADM has received speaking fees from Janssen in the past 36 months. PE has received consulting fees from Abbvie, Activa, Astra-Zeneca, BMS, Boehringer Ingelheim, Galapagos, Gilead, Immunovant, Lilly, and Novartis in the past 36 months.

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

Ethical approval: Not needed.

Guarantor: PE.

Contributorship: DS, ADM, and PE discussed and agreed on the structure of the review. DS prepared the first draft of the manuscript, which was critically reviewed by ADM and PE. All the authors have seen and approved of the final version of the manuscript.

ORCID iD

Didem Sahin https://orcid.org/0000-0003-3558-54000

References

  • 1.Di Matteo A, Bathon JM, Emery P. Rheumatoid arthritis. Lancet 2023; 402: 2019–2033. DOI: 10.1016/s0140-6736(23)01525-8. [DOI] [PubMed] [Google Scholar]
  • 2.GBD 2021 Osteoarthritis Collaborators . Global, regional, and national burden of rheumatoid arthritis, 1990-2020, and projections to 2050: a systematic analysis of the Global Burden of Disease Study 2021. Lancet Rheumatol 2023; 5: e594-e610. DOI: 10.1016/s2665-9913(23)00211-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Smolen JS, Aletaha D, Bijlsma JW, et al. Treating rheumatoid arthritis to target: recommendations of an international task force. Ann Rheum Dis 2010; 69: 631–637. DOI: 10.1136/ard.2009.123919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League against rheumatism collaborative initiative. Ann Rheum Dis 2010; 69: 1580–1588. DOI: 10.1136/ard.2010.138461. [DOI] [PubMed] [Google Scholar]
  • 5.Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 1988; 31: 315–324. DOI: 10.1002/art.1780310302. [DOI] [PubMed] [Google Scholar]
  • 6.Smolen JS, Aletaha D, Barton A, et al. Rheumatoid arthritis. Nat Rev Dis Primers 2018; 4: 18001. DOI: 10.1038/nrdp.2018.1. [DOI] [PubMed] [Google Scholar]
  • 7.Frazzei G, Musters A, de Vries N, et al. Prevention of rheumatoid arthritis: a systematic literature review of preventive strategies in at-risk individuals. Autoimmun Rev 2023; 22: 103217. DOI: 10.1016/j.autrev.2022.103217. [DOI] [PubMed] [Google Scholar]
  • 8.MacGregor AJ, Snieder H, Rigby AS, et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum 2000; 43: 30–37. DOI: . [DOI] [PubMed] [Google Scholar]
  • 9.Svendsen AJ, Kyvik KO, Houen G, et al. On the origin of rheumatoid arthritis: the impact of environment and genes--a population based twin study. PLoS One 2013; 8: e57304. DOI: 10.1371/journal.pone.0057304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Frisell T, Holmqvist M, Källberg H, et al. Familial risks and heritability of rheumatoid arthritis: role of rheumatoid factor/anti-citrullinated protein antibody status, number and type of affected relatives, sex, and age. Arthritis Rheum 2013; 65: 2773–2782. DOI: 10.1002/art.38097. [DOI] [PubMed] [Google Scholar]
  • 11.Ishigaki K, Sakaue S, Terao C, et al. Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis. Nat Genet 2022; 54: 1640–1651. DOI: 10.1038/s41588-022-01213-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.MacGregor A, Ollier W, Thomson W, et al. HLA-DRB1*0401/0404 genotype and rheumatoid arthritis: increased association in men, young age at onset, and disease severity. J Rheumatol 1995; 22: 1032–1036. [PubMed] [Google Scholar]
  • 13.Morgan AW, Haroon-Rashid L, Martin SG, et al. The shared epitope hypothesis in rheumatoid arthritis: evaluation of alternative classification criteria in a large UK Caucasian cohort. Arthritis Rheum 2008; 58: 1275–1283. DOI: 10.1002/art.23432. [DOI] [PubMed] [Google Scholar]
  • 14.Klareskog L, Stolt P, Lundberg K, et al. A new model for an etiology of rheumatoid arthritis: smoking may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis Rheum 2006; 54: 38–46. DOI: 10.1002/art.21575. [DOI] [PubMed] [Google Scholar]
  • 15.van der Helm-van Mil AHM, Verpoort KN, Breedveld FC, et al. The HLA-DRB1 shared epitope alleles are primarily a risk factor for anti-cyclic citrullinated peptide antibodies and are not an independent risk factor for development of rheumatoid arthritis. Arthritis Rheum 2006; 54: 1117–1121. DOI: 10.1002/art.21739. [DOI] [PubMed] [Google Scholar]
  • 16.van der Helm-van Mil AHM, Verpoort KN, le Cessie S, et al. The HLA-DRB1 shared epitope alleles differ in the interaction with smoking and predisposition to antibodies to cyclic citrullinated peptide. Arthritis Rheum 2007; 56: 425–432. DOI: 10.1002/art.22373. [DOI] [PubMed] [Google Scholar]
  • 17.Karlson EW, Chibnik LB, Cui J, et al. Associations between human leukocyte antigen, PTPN22, CTLA4 genotypes and rheumatoid arthritis phenotypes of autoantibody status, age at diagnosis and erosions in a large cohort study. Ann Rheum Dis 2008; 67: 358–363. DOI: 10.1136/ard.2007.071662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Morgan AW, Thomson W, Martin SG, et al. Reevaluation of the interaction between HLA-DRB1 shared epitope alleles, PTPN22, and smoking in determining susceptibility to autoantibody-positive and autoantibody-negative rheumatoid arthritis in a large UK Caucasian population. Arthritis Rheum 2009; 60: 2565–2576. DOI: 10.1002/art.24752. [DOI] [PubMed] [Google Scholar]
  • 19.Kim K, Jiang X, Cui J, et al. Interactions between amino acid-defined major histocompatibility complex class II variants and smoking in seropositive rheumatoid arthritis. Arthritis Rheumatol 2015; 67: 2611–2623. DOI: 10.1002/art.39228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Michou L, Teixeira VH, Pierlot C, et al. Associations between genetic factors, tobacco smoking and autoantibodies in familial and sporadic rheumatoid arthritis. Ann Rheum Dis 2008; 67: 466–470. DOI: 10.1136/ard.2007.075622. [DOI] [PubMed] [Google Scholar]
  • 21.van der Woude D, Houwing-Duistermaat JJ, Toes RE, et al. Quantitative heritability of anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum 2009; 60: 916–923. DOI: 10.1002/art.24385. [DOI] [PubMed] [Google Scholar]
  • 22.van der Woude D, Lie BA, Lundström E, et al. Protection against anti-citrullinated protein antibody-positive rheumatoid arthritis is predominantly associated with HLA-DRB1*1301: a meta-analysis of HLA-DRB1 associations with anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis in four European populations. Arthritis Rheum 2010; 62: 1236–1245. DOI: 10.1002/art.27366. [DOI] [PubMed] [Google Scholar]
  • 23.Lundström E, Källberg H, Smolnikova M, et al. Opposing effects of HLA-DRB1*13 alleles on the risk of developing anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum 2009; 60: 924–930. DOI: 10.1002/art.24410. [DOI] [PubMed] [Google Scholar]
  • 24.van der Helm-van Mil AHM, Huizinga TW, Schreuder GM, et al. An independent role of protective HLA class II alleles in rheumatoid arthritis severity and susceptibility. Arthritis Rheum 2005; 52: 2637–2644. DOI: 10.1002/art.21272. [DOI] [PubMed] [Google Scholar]
  • 25.Oka S, Furukawa H, Kawasaki A, et al. Protective effect of the HLA-DRB1*13:02 allele in Japanese rheumatoid arthritis patients. PLoS One 2014; 9: e99453. DOI: 10.1371/journal.pone.0099453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gorman JD, Lum RF, Chen JJ, et al. Impact of shared epitope genotype and ethnicity on erosive disease: a meta-analysis of 3,240 rheumatoid arthritis patients. Arthritis Rheum 2004; 50: 400–412. DOI: 10.1002/art.20006. [DOI] [PubMed] [Google Scholar]
  • 27.Moreno I, Valenzuela A, García A, et al. Association of the shared epitope with radiological severity of rheumatoid arthritis. J Rheumatol 1996; 23: 6–9. [PubMed] [Google Scholar]
  • 28.Lard LR, Boers M, Verhoeven A, et al. Early and aggressive treatment of rheumatoid arthritis patients affects the association of HLA class II antigens with progression of joint damage. Arthritis Rheum 2002; 46: 899–905. DOI: 10.1002/art.10151. [DOI] [PubMed] [Google Scholar]
  • 29.Viatte S, Plant D, Han B, et al. Association of HLA-DRB1 haplotypes with rheumatoid arthritis severity, mortality, and treatment response. JAMA 2015; 313: 1645–1656. DOI: 10.1001/jama.2015.3435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.O'Dell JR, Nepom BS, Haire C, et al. HLA-DRB1 typing in rheumatoid arthritis: predicting response to specific treatments. Ann Rheum Dis 1998; 57: 209–213. DOI: 10.1136/ard.57.4.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Möttönen T, Hannonen P, Korpela M, et al. Delay to institution of therapy and induction of remission using single-drug or combination-disease-modifying antirheumatic drug therapy in early rheumatoid arthritis. Arthritis Rheum 2002; 46: 894–898. DOI: 10.1002/art.10135. [DOI] [PubMed] [Google Scholar]
  • 32.Miceli-Richard C, Comets E, Verstuyft C, et al. A single tumour necrosis factor haplotype influences the response to adalimumab in rheumatoid arthritis. Ann Rheum Dis 2008; 67: 478–484. DOI: 10.1136/ard.2007.074104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Criswell LA, Lum RF, Turner KN, et al. The influence of genetic variation in the HLA-DRB1 and LTA-TNF regions on the response to treatment of early rheumatoid arthritis with methotrexate or etanercept. Arthritis Rheum 2004; 50: 2750–2756. DOI: 10.1002/art.20469. [DOI] [PubMed] [Google Scholar]
  • 34.Skapenko A, Smolen JS, Kavanaugh A, et al. Genetic markers associated with clinical and radiographic response in adalimumab plus methotrexate- or methotrexate-treated rheumatoid arthritis patients in OPTIMA. Clin Exp Rheumatol 2019; 37: 783–790. [PubMed] [Google Scholar]
  • 35.van der Woude D, Young A, Jayakumar K, et al. 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. DOI: 10.1002/art.24661. [DOI] [PubMed] [Google Scholar]
  • 36.Turesson C, Schaid DJ, Weyand CM, et al. The impact of HLA-DRB1 genes on extra-articular disease manifestations in rheumatoid arthritis. Arthritis Res Ther 2005; 7: R1386–R1393. DOI: 10.1186/ar1837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gorman JD, David-Vaudey E, Pai M, et al. Particular HLA-DRB1 shared epitope genotypes are strongly associated with rheumatoid vasculitis. Arthritis Rheum 2004; 50: 3476–3484. DOI: 10.1002/art.20588. [DOI] [PubMed] [Google Scholar]
  • 38.Gianchecchi E, Palombi M, Fierabracci A. The putative role of the C1858T polymorphism of protein tyrosine phosphatase PTPN22 gene in autoimmunity. Autoimmun Rev 2013; 12: 717–725. DOI: 10.1016/j.autrev.2012.12.003. [DOI] [PubMed] [Google Scholar]
  • 39.Begovich AB, Carlton VE, Honigberg LA, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet 2004; 75: 330–337. DOI: 10.1086/422827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hinks A, Barton A, John S, et al. Association between the PTPN22 gene and rheumatoid arthritis and juvenile idiopathic arthritis in a UK population: further support that PTPN22 is an autoimmunity gene. Arthritis Rheum 2005; 52: 1694–1699. DOI: 10.1002/art.21049. [DOI] [PubMed] [Google Scholar]
  • 41.Johansson M, Arlestig L, Hallmans G, et al. PTPN22 polymorphism and anti-cyclic citrullinated peptide antibodies in combination strongly predicts future onset of rheumatoid arthritis and has a specificity of 100% for the disease. Arthritis Res Ther 2006; 8: R19. DOI: 10.1186/ar1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Orozco G, Pascual-Salcedo D, López-Nevot MA, et al. Auto-antibodies, HLA and PTPN22: susceptibility markers for rheumatoid arthritis. Rheumatology (Oxford) 2008; 47: 138–141. DOI: 10.1093/rheumatology/kem343. [DOI] [PubMed] [Google Scholar]
  • 43.Feitsma AL, Toes RE, Begovich AB, et al. Risk of progression from undifferentiated arthritis to rheumatoid arthritis: the effect of the PTPN22 1858T-allele in anti-citrullinated peptide antibody positive patients. Rheumatology (Oxford) 2007; 46: 1092–1095. DOI: 10.1093/rheumatology/kem006. [DOI] [PubMed] [Google Scholar]
  • 44.Goëb V, Dieudé P, Daveau R, et al. Contribution of PTPN22 1858T, TNFRII 196R and HLA-shared epitope alleles with rheumatoid factor and anti-citrullinated protein antibodies to very early rheumatoid arthritis diagnosis. Rheumatology (Oxford) 2008; 47: 1208–1212. DOI: 10.1093/rheumatology/ken192. [DOI] [PubMed] [Google Scholar]
  • 45.Kokkonen H, Johansson M, Innala L, et al. The PTPN22 1858C/T polymorphism is associated with anti-cyclic citrullinated peptide antibody-positive early rheumatoid arthritis in Northern Sweden. Arthritis Res Ther 2007; 9: R56. DOI: 10.1186/ar2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lie BA, Viken MK, Odegård S, et al. Associations between the PTPN22 1858C->T polymorphism and radiographic joint destruction in patients with rheumatoid arthritis: results from a 10-year longitudinal study. Ann Rheum Dis 2007; 66: 1604–1609. DOI: 10.1136/ard.2006.067892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Majorczyk E, Pawlik A, Kuśnierczyk P. PTPN22 1858C>T polymorphism is strongly associated with rheumatoid arthritis but not with a response to methotrexate therapy. Int Immunopharmacol 2010; 10: 1626–1629. DOI: 10.1016/j.intimp.2010.09.008. [DOI] [PubMed] [Google Scholar]
  • 48.Potter C, Hyrich KL, Tracey A, et al. Association of rheumatoid factor and anti-cyclic citrullinated peptide positivity, but not carriage of shared epitope or PTPN22 susceptibility variants, with anti-tumour necrosis factor response in rheumatoid arthritis. Ann Rheum Dis 2009; 68: 69–74. DOI: 10.1136/ard.2007.084715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Perricone C, Ceccarelli F, Valesini G. An overview on the genetic of rheumatoid arthritis: a never-ending story. Autoimmun Rev 2011; 10: 599–608. DOI: 10.1016/j.autrev.2011.04.021. [DOI] [PubMed] [Google Scholar]
  • 50.Nemtsova MV, Zaletaev DV, Bure IV, et al. Epigenetic changes in the pathogenesis of rheumatoid arthritis. Front Genet 2019; 10: 570. DOI: 10.3389/fgene.2019.00570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.van Delft MAM, Huizinga TWJ. An overview of autoantibodies in rheumatoid arthritis. J Autoimmun 2020; 110: 102392. DOI: 10.1016/j.jaut.2019.102392. [DOI] [PubMed] [Google Scholar]
  • 52.Tasliyurt T, Kisacik B, Kaya SU, et al. The frequency of antibodies against cyclic citrullinated peptides and rheumatoid factor in healthy population: a field study of rheumatoid arthritis from northern Turkey. Rheumatol Int 2013; 33: 939–942. DOI: 10.1007/s00296-012-2458-5. [DOI] [PubMed] [Google Scholar]
  • 53.van Zanten A, Arends S, Roozendaal C, et al. Presence of anticitrullinated protein antibodies in a large population-based cohort from The Netherlands. Ann Rheum Dis 2017; 76: 1184–1190. DOI: 10.1136/annrheumdis-2016-209991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nishimura K, Sugiyama D, Kogata Y, et al. Meta-analysis: diagnostic accuracy of anti-cyclic citrullinated peptide antibody and rheumatoid factor for rheumatoid arthritis. Ann Intern Med 2007; 146: 797–808. DOI: 10.7326/0003-4819-146-11-200706050-00008. [DOI] [PubMed] [Google Scholar]
  • 55.Jørgensen KT, Wiik A, Pedersen M, et al. Cytokines, autoantibodies and viral antibodies in premorbid and postdiagnostic sera from patients with rheumatoid arthritis: case-control study nested in a cohort of Norwegian blood donors. Ann Rheum Dis 2008; 67: 860–866. DOI: 10.1136/ard.2007.073825. [DOI] [PubMed] [Google Scholar]
  • 56.Bos WH, Wolbink GJ, Boers M, et al. Arthritis development in patients with arthralgia is strongly associated with anti-citrullinated protein antibody status: a prospective cohort study. Ann Rheum Dis 2010; 69: 490–494. DOI: 10.1136/ard.2008.105759. [DOI] [PubMed] [Google Scholar]
  • 57.van Steenbergen HW, Mangnus L, Reijnierse M, et al. Clinical factors, anticitrullinated peptide antibodies and MRI-detected subclinical inflammation in relation to progression from clinically suspect arthralgia to arthritis. Ann Rheum Dis 2016; 75: 1824–1830. DOI: 10.1136/annrheumdis-2015-208138. [DOI] [PubMed] [Google Scholar]
  • 58.Bemis EA, Demoruelle MK, Seifert JA, et al. Factors associated with progression to inflammatory arthritis in first-degree relatives of individuals with RA following autoantibody positive screening in a non-clinical setting. Ann Rheum Dis 2021; 80: 154–161. DOI: 10.1136/annrheumdis-2020-217066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rakieh C, Nam JL, Hunt L, et al. Predicting the development of clinical arthritis in anti-CCP positive individuals with non-specific musculoskeletal symptoms: a prospective observational cohort study. Ann Rheum Dis 2015; 74: 1659–1666. DOI: 10.1136/annrheumdis-2014-205227. [DOI] [PubMed] [Google Scholar]
  • 60.Di Matteo A, Mankia K, Duquenne L, et al. Third-generation anti-cyclic citrullinated peptide antibodies improve prediction of clinical arthritis in individuals at risk of rheumatoid arthritis. Arthritis Rheumatol 2020; 72: 1820–1828. DOI: 10.1002/art.41402. [DOI] [PubMed] [Google Scholar]
  • 61.Di Matteo A, Mankia K, Garcia-Montoya L, et al. Utility of testing for third-generation anticyclic citrullinated peptide (anti-CCP3) antibodies in individuals who present with new musculoskeletal symptoms but have a negative second-generation anticyclic citrullinated peptide (anti-CCP2) antibody test. RMD Open 2024; 10: e003927. DOI: 10.1136/rmdopen-2023-003927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Di Matteo A, Duquenne L, Cipolletta E, et al. Ultrasound subclinical synovitis in anti-CCP-positive at-risk individuals with musculoskeletal symptoms: an important and predictable stage in the rheumatoid arthritis continuum. Rheumatology (Oxford) 2022; 61: 3192–3200. DOI: 10.1093/rheumatology/keab862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kleyer A, Finzel S, Rech J, et al. Bone loss before the clinical onset of rheumatoid arthritis in subjects with anticitrullinated protein antibodies. Ann Rheum Dis 2014; 73: 854–860. DOI: 10.1136/annrheumdis-2012-202958. [DOI] [PubMed] [Google Scholar]
  • 64.Di Matteo A, Mankia K, Duquenne L, et al. Ultrasound erosions in the feet best predict progression to inflammatory arthritis in anti-CCP positive at-risk individuals without clinical synovitis. Ann Rheum Dis 2020; 79: 901–907. DOI: 10.1136/annrheumdis-2020-217215. [DOI] [PubMed] [Google Scholar]
  • 65.Westra J, Brouwer E, Raveling-Eelsing E, et al. Arthritis autoantibodies in individuals without rheumatoid arthritis: follow-up data from a Dutch population-based cohort (Lifelines). Rheumatology (Oxford) 2021; 60: 658–666. DOI: 10.1093/rheumatology/keaa219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ponchel F, Goëb V, Parmar R, et al. An immunological biomarker to predict MTX response in early RA. Ann Rheum Dis 2014; 73: 2047–2053. DOI: 10.1136/annrheumdis-2013-203566. [DOI] [PubMed] [Google Scholar]
  • 67.van den Broek M, Dirven L, Klarenbeek NB, et al. The association of treatment response and joint damage with ACPA-status in recent-onset RA: a subanalysis of the 8-year follow-up of the BeSt study. Ann Rheum Dis 2012; 71: 245–248. DOI: 10.1136/annrheumdis-2011-200379. [DOI] [PubMed] [Google Scholar]
  • 68.Turesson C, Jacobsson LT, Sturfelt G, et al. Rheumatoid factor and antibodies to cyclic citrullinated peptides are associated with severe extra-articular manifestations in rheumatoid arthritis. Ann Rheum Dis 2007; 66: 59–64. DOI: 10.1136/ard.2006.054445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Westerlind H, Rönnelid J, Hansson M, et al. Anti-citrullinated protein antibody specificities, rheumatoid factor isotypes, and incident cardiovascular events in patients with rheumatoid arthritis. Arthritis Rheumatol 2020; 72: 1658–1667. DOI: 10.1002/art.41381. [DOI] [PubMed] [Google Scholar]
  • 70.Westerlind H, Kastbom A, Rönnelid J, et al. The association between autoantibodies and risk for venous thromboembolic events among patients with rheumatoid arthritis. Rheumatology (Oxford) 2023; 62: 2106–2112. DOI: 10.1093/rheumatology/keac601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Jansen LM, van der Horst-Bruinsma IE, van Schaardenburg D, et al. Predictors of radiographic joint damage in patients with early rheumatoid arthritis. Ann Rheum Dis 2001; 60: 924–927. DOI: 10.1136/ard.60.10.924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Vanier A, Smolen JS, Allaart CF, et al. An updated matrix to predict rapid radiographic progression of early rheumatoid arthritis patients: pooled analyses from several databases. Rheumatology (Oxford) 2020; 59: 1842–1852. DOI: 10.1093/rheumatology/kez542. [DOI] [PubMed] [Google Scholar]
  • 73.Rydell E, Forslind K, Nilsson J, et al. Predictors of radiographic erosion and joint space narrowing progression in patients with early rheumatoid arthritis: a cohort study. Arthritis Res Ther 2021; 23: 27. DOI: 10.1186/s13075-020-02413-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chatzidionysiou K, Lie E, Nasonov E, et al. Highest clinical effectiveness of rituximab in autoantibody-positive patients with rheumatoid arthritis and in those for whom no more than one previous TNF antagonist has failed: pooled data from 10 European registries. Ann Rheum Dis 2011; 70: 1575–1580. DOI: 10.1136/ard.2010.148759. [DOI] [PubMed] [Google Scholar]
  • 75.Narvaez J, Díaz-Torné C, Ruiz JM, et al. Predictors of response to rituximab in patients with active rheumatoid arthritis and inadequate response to anti-TNF agents or traditional DMARDs. Clin Exp Rheumatol 2011; 29: 991–997. [PubMed] [Google Scholar]
  • 76.Courvoisier DS, Chatzidionysiou K, Mongin D, et al. The impact of seropositivity on the effectiveness of biologic anti-rheumatic agents: results from a collaboration of 16 registries. Rheumatology (Oxford) 2021; 60: 820–828. DOI: 10.1093/rheumatology/keaa393. [DOI] [PubMed] [Google Scholar]
  • 77.Bird P, Hall S, Nash P, et al. Treatment outcomes in patients with seropositive versus seronegative rheumatoid arthritis in Phase III randomised clinical trials of tofacitinib. RMD Open 2019; 5: e000742. DOI: 10.1136/rmdopen-2018-000742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.de Moel EC, Derksen V, Stoeken G, et al. Baseline autoantibody profile in rheumatoid arthritis is associated with early treatment response but not long-term outcomes. Arthritis Res Ther 2018; 20: 33. DOI: 10.1186/s13075-018-1520-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Heimans L, Akdemir G, Boer KV, et al. Two-year results of disease activity score (DAS)-remission-steered treatment strategies aiming at drug-free remission in early arthritis patients (the IMPROVED-study). Arthritis Res Ther 2016; 18: 23. DOI: 10.1186/s13075-015-0912-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.de Moel EC, Derksen V, Trouw LA, et al. In rheumatoid arthritis, changes in autoantibody levels reflect intensity of immunosuppression, not subsequent treatment response. Arthritis Res Ther 2019; 21: 28. DOI: 10.1186/s13075-019-1815-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Boeters DM, Burgers LE, Toes RE, et al. Does immunological remission, defined as disappearance of autoantibodies, occur with current treatment strategies? A long-term follow-up study in rheumatoid arthritis patients who achieved sustained DMARD-free status. Ann Rheum Dis 2019; 78: 1497–1504. DOI: 10.1136/annrheumdis-2018-214868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Shi J, van Veelen PA, Mahler M, et al. Carbamylation and antibodies against carbamylated proteins in autoimmunity and other pathologies. Autoimmun Rev 2014; 13: 225–230. DOI: 10.1016/j.autrev.2013.10.008. [DOI] [PubMed] [Google Scholar]
  • 83.Jiang X, Trouw LA, van Wesemael TJ, et al. Anti-CarP antibodies in two large cohorts of patients with rheumatoid arthritis and their relationship to genetic risk factors, cigarette smoking and other autoantibodies. Ann Rheum Dis 2014; 73: 1761–1768. DOI: 10.1136/annrheumdis-2013-205109. [DOI] [PubMed] [Google Scholar]
  • 84.Gan RW, Trouw LA, Shi J, et al. Anti-carbamylated protein antibodies are present prior to rheumatoid arthritis and are associated with its future diagnosis. J Rheumatol 2015; 42: 572–579. DOI: 10.3899/jrheum.140767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Shi J, van de Stadt LA, Levarht EW, et al. Anti-carbamylated protein (anti-CarP) antibodies precede the onset of rheumatoid arthritis. Ann Rheum Dis 2014; 73: 780–783. DOI: 10.1136/annrheumdis-2013-204154. [DOI] [PubMed] [Google Scholar]
  • 86.Brink M, Verheul MK, Rönnelid J, et al. Anti-carbamylated protein antibodies in the pre-symptomatic phase of rheumatoid arthritis, their relationship with multiple anti-citrulline peptide antibodies and association with radiological damage. Arthritis Res Ther 2015; 17: 25. DOI: 10.1186/s13075-015-0536-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kumar R, Piantoni S, Boldini M, et al. Anti-carbamylated protein antibodies as a clinical response predictor in rheumatoid arthritis patients treated with abatacept. Clin Exp Rheumatol 2021; 39: 91–97. [DOI] [PubMed] [Google Scholar]
  • 88.Ponchel F, van Delft MAM, Xie X, et al. Anti-carbamylated protein antibodies: are they useful for the diagnosis of rheumatoid arthritis? Clin Exp Rheumatol 2021; 39: 146–150. [DOI] [PubMed] [Google Scholar]
  • 89.Vega-Morales D, Garza-Elizondo MA, Trouw LA, et al. Anti-carbamylated protein antibodies positivity and disease activity in Hispanic patients with established rheumatoid arthritis: an observational study. Mod Rheumatol 2022; 32: 330–337. DOI: 10.1080/14397595.2021.1913277. [DOI] [PubMed] [Google Scholar]
  • 90.Verheul MK, Böhringer S, van Delft MAM, et al. Triple positivity for anti-citrullinated protein autoantibodies, rheumatoid factor, and anti-carbamylated protein antibodies conferring high specificity for rheumatoid arthritis: implications for very early identification of at-risk individuals. Arthritis Rheumatol 2018; 70: 1721–1731. DOI: 10.1002/art.40562. [DOI] [PubMed] [Google Scholar]
  • 91.Shi J, Knevel R, Suwannalai P, et al. Autoantibodies recognizing carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. Proc Natl Acad Sci U S A 2011; 108: 17372–17377. DOI: 10.1073/pnas.1114465108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Julià A, López-Lasanta M, Blanco F, et al. Interactions between rheumatoid arthritis antibodies are associated with the response to anti-tumor necrosis factor therapy. BMC Musculoskelet Disord 2021; 22: 372. DOI: 10.1186/s12891-021-04248-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kolfenbach JR, Deane KD, Derber LA, et al. Autoimmunity to peptidyl arginine deiminase type 4 precedes clinical onset of rheumatoid arthritis. Arthritis Rheum 2010; 62: 2633–2639. DOI: 10.1002/art.27570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhang G, Xu J, Du D, et al. Diagnostic values, association with disease activity and possible risk factors of anti-PAD4 in rheumatoid arthritis: a meta-analysis. Rheumatology (Oxford) 2023; 63: 914–924. DOI: 10.1093/rheumatology/kead545. [DOI] [PubMed] [Google Scholar]
  • 95.Halvorsen EH, Pollmann S, Gilboe IM, et al. Serum IgG antibodies to peptidylarginine deiminase 4 in rheumatoid arthritis and associations with disease severity. Ann Rheum Dis 2008; 67: 414–417. DOI: 10.1136/ard.2007.080267. [DOI] [PubMed] [Google Scholar]
  • 96.Halvorsen EH, Haavardsholm EA, Pollmann S, et al. Serum IgG antibodies to peptidylarginine deiminase 4 predict radiographic progression in patients with rheumatoid arthritis treated with tumour necrosis factor-alpha blocking agents. Ann Rheum Dis 2009; 68: 249–252. DOI: 10.1136/ard.2008.094490. [DOI] [PubMed] [Google Scholar]
  • 97.Juarez M, Bang H, Hammar F, et al. Identification of novel antiacetylated vimentin antibodies in patients with early inflammatory arthritis. Ann Rheum Dis 2016; 75: 1099–1107. DOI: 10.1136/annrheumdis-2014-206785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Thiele GM, Duryee MJ, Anderson DR, et al. Malondialdehyde-acetaldehyde adducts and anti-malondialdehyde-acetaldehyde antibodies in rheumatoid arthritis. Arthritis Rheumatol 2015; 67: 645–655. DOI: 10.1002/art.38969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Tyrrell DJ, Goldstein DR. Ageing and atherosclerosis: vascular intrinsic and extrinsic factors and potential role of IL-6. Nat Rev Cardiol 2021; 18: 58–68. DOI: 10.1038/s41569-020-0431-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nielen MM, van Schaardenburg D, Reesink HW, et al. Increased levels of C-reactive protein in serum from blood donors before the onset of rheumatoid arthritis. Arthritis Rheum 2004; 50: 2423–2427. DOI: 10.1002/art.20431. [DOI] [PubMed] [Google Scholar]
  • 101.van de Stadt LA, Witte BI, Bos WH, et al. A prediction rule for the development of arthritis in seropositive arthralgia patients. Ann Rheum Dis 2013; 72: 1920-1926. DOI: 10.1136/annrheumdis-2012-202127. [DOI] [PubMed] [Google Scholar]
  • 102.Smolen JS, Breedveld FC, Schiff MH, et al. A simplified disease activity index for rheumatoid arthritis for use in clinical practice. Rheumatology (Oxford) 2003; 42: 244–257. DOI: 10.1093/rheumatology/keg072. [DOI] [PubMed] [Google Scholar]
  • 103.England BR, Tiong BK, Bergman MJ, et al. 2019 update of the American College of Rheumatology recommended rheumatoid arthritis disease activity measures. Arthritis Care Res (Hoboken) 2019; 71: 1540–1555. DOI: 10.1002/acr.24042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Felson DT, Smolen JS, Wells G, et al. American College of Rheumatology/European League against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical trials. Ann Rheum Dis 2011; 70: 404–413. DOI: 10.1136/ard.2011.149765. [DOI] [PubMed] [Google Scholar]
  • 105.Orr CK, Najm A, Young F, et al. The utility and limitations of CRP, ESR and DAS28-CRP in appraising disease activity in rheumatoid arthritis. Front Med (Lausanne) 2018; 5: 185. DOI: 10.3389/fmed.2018.00185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kay J, Morgacheva O, Messing SP, et al. Clinical disease activity and acute phase reactant levels are discordant among patients with active rheumatoid arthritis: acute phase reactant levels contribute separately to predicting outcome at one year. Arthritis Res Ther 2014; 16: R40. DOI: 10.1186/ar4469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Keenan RT, Swearingen CJ, Yazici Y. Erythrocyte sedimentation rate and C-reactive protein levels are poorly correlated with clinical measures of disease activity in rheumatoid arthritis, systemic lupus erythematosus and osteoarthritis patients. Clin Exp Rheumatol 2008; 26: 814–819. [PubMed] [Google Scholar]
  • 108.Aletaha D, Kerschbaumer A, Kastrati K, et al. Consensus statement on blocking interleukin-6 receptor and interleukin-6 in inflammatory conditions: an update. Ann Rheum Dis 2023; 82: 773–787. DOI: 10.1136/ard-2022-222784. [DOI] [PubMed] [Google Scholar]
  • 109.Combe B, Cantagrel A, Goupille P, et al. Predictive factors of 5-year health assessment questionnaire disability in early rheumatoid arthritis. J Rheumatol 2003; 30: 2344–2349. [PubMed] [Google Scholar]
  • 110.Smolen JS, Landewé RBM, Bergstra SA, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2022 update. Ann Rheum Dis 2023; 82: 3–18. DOI: 10.1136/ard-2022-223356. [DOI] [PubMed] [Google Scholar]
  • 111.Emery P, Gabay C, Kraan M, et al. Evidence-based review of biologic markers as indicators of disease progression and remission in rheumatoid arthritis. Rheumatol Int 2007; 27: 793–806. DOI: 10.1007/s00296-007-0357-y. [DOI] [PubMed] [Google Scholar]
  • 112.Duquenne L, Hensor EM, Wilson M, et al. Predicting inflammatory arthritis in at-risk persons: development of scores for risk stratification. Ann Intern Med 2023; 176: 1027–1036. DOI: 10.7326/m23-0272. [DOI] [PubMed] [Google Scholar]
  • 113.Majorczyk E, Mazurek-Mochol M, Pawlik A, et al. Clinical factors and the outcome of treatment with methotrexate in rheumatoid arthritis: role of rheumatoid factor, erosive disease and high level of erythrocyte sedimentation rate. J Clin Med 2022; 11: 6078. DOI: 10.3390/jcm11206078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Khader Y, Beran A, Ghazaleh S, et al. Predictors of remission in rheumatoid arthritis patients treated with biologics: a systematic review and meta-analysis. Clin Rheumatol 2022; 41: 3615–3627. DOI: 10.1007/s10067-022-06307-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Inciarte-Mundo J, Frade-Sosa B, Sanmartí R. From bench to bedside: calprotectin (S100A8/S100A9) as a biomarker in rheumatoid arthritis. Front Immunol 2022; 13: 1001025. DOI: 10.3389/fimmu.2022.1001025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Bettner LF, Peterson RA, Bergstedt DT, et al. Combinations of anticyclic citrullinated protein antibody, rheumatoid factor, and serum calprotectin positivity are associated with the diagnosis of rheumatoid arthritis within 3 years. ACR Open Rheumatol 2021; 3: 684–689. DOI: 10.1002/acr2.11309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Baillet A, Trocmé C, Berthier S, et al. Synovial fluid proteomic fingerprint: S100A8, S100A9 and S100A12 proteins discriminate rheumatoid arthritis from other inflammatory joint diseases. Rheumatology (Oxford) 2010; 49: 671–682. DOI: 10.1093/rheumatology/kep452. [DOI] [PubMed] [Google Scholar]
  • 118.Liao H, Wu J, Kuhn E, et al. Use of mass spectrometry to identify protein biomarkers of disease severity in the synovial fluid and serum of patients with rheumatoid arthritis. Arthritis Rheum 2004; 50: 3792–3803. DOI: 10.1002/art.20720. [DOI] [PubMed] [Google Scholar]
  • 119.Nordal HH, Brokstad KA, Solheim M, et al. Calprotectin (S100A8/A9) has the strongest association with ultrasound-detected synovitis and predicts response to biologic treatment: results from a longitudinal study of patients with established rheumatoid arthritis. Arthritis Res Ther 2017; 19: 3. DOI: 10.1186/s13075-016-1201-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Nordal HH, Brun JG, Hordvik M, et al. Calprotectin (S100A8/A9) and S100A12 are associated with measures of disease activity in a longitudinal study of patients with rheumatoid arthritis treated with infliximab. Scand J Rheumatol 2016; 45: 274–281. DOI: 10.3109/03009742.2015.1107128. [DOI] [PubMed] [Google Scholar]
  • 121.Torgutalp M, Yayla ME, Eroglu DS, et al. Serum calprotectin is indicating clinical and ultrasonographic disease activity in rheumatoid arthritis, even with normal C-reactive protein levels. Mediterr J Rheumatol 2021; 32: 56–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Smith SL, Alexander S, Nair N, et al. Pre-treatment calprotectin (MRP8/14) provides no added value to testing CRP alone in terms of predicting response to TNF inhibitors in rheumatoid arthritis in a post hoc analysis. Ann Rheum Dis 2023; 82: 611–620. DOI: 10.1136/ard-2022-222519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Andrés Cerezo L, Mann H, Pecha O, et al. Decreases in serum levels of S100A8/9 (calprotectin) correlate with improvements in total swollen joint count in patients with recent-onset rheumatoid arthritis. Arthritis Res Ther 2011; 13: R122. DOI: 10.1186/ar3426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Bechman K, Tweehuysen L, Garrood T, et al. Flares in rheumatoid arthritis patients with low disease activity: predictability and association with worse clinical outcomes. J Rheumatol 2018; 45: 1515–1521. DOI: 10.3899/jrheum.171375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Romand X, Clapasson M, Chuong MV, et al. Serum calprotectin levels do not predict subsequent relapse in rheumatoid arthritis in remission: a post-hoc analysis of STRASS study. RMD Open 2023; 9: e003198. DOI: 10.1136/rmdopen-2023-003198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Inciarte-Mundo J, Ramirez J, Hernández MV, et al. Calprotectin strongly and independently predicts relapse in rheumatoid arthritis and polyarticular psoriatic arthritis patients treated with tumor necrosis factor inhibitors: a 1-year prospective cohort study. Arthritis Res Ther 2018; 20: 275. DOI: 10.1186/s13075-018-1764-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.de Moel EC, Rech J, Mahler M, et al. Circulating calprotectin (S100A8/A9) is higher in rheumatoid arthritis patients that relapse within 12 months of tapering anti-rheumatic drugs. Arthritis Res Ther 2019; 21: 268. DOI: 10.1186/s13075-019-2064-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sorić Hosman I, Kos I, Lamot L. Serum Amyloid A in inflammatory rheumatic diseases: a compendious review of a renowned biomarker. Front Immunol 2020; 11: 631299. DOI: 10.3389/fimmu.2020.631299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Mullan RH, Bresnihan B, Golden-Mason L, et al. Acute-phase serum amyloid A stimulation of angiogenesis, leukocyte recruitment, and matrix degradation in rheumatoid arthritis through an NF-kappaB-dependent signal transduction pathway. Arthritis Rheum 2006; 54: 105–114. DOI: 10.1002/art.21518. [DOI] [PubMed] [Google Scholar]
  • 130.Zhou J, Dai Y, Lin Y, et al. Association between serum amyloid A and rheumatoid arthritis: a systematic review and meta-analysis. Semin Arthritis Rheum 2022; 52: 151943. DOI: 10.1016/j.semarthrit.2021.12.011. [DOI] [PubMed] [Google Scholar]
  • 131.Noack M, Miossec P. Selected cytokine pathways in rheumatoid arthritis. Semin Immunopathol 2017; 39: 365–383. DOI: 10.1007/s00281-017-0619-z. [DOI] [PubMed] [Google Scholar]
  • 132.Tetta C, Camussi G, Modena V, et al. Tumour necrosis factor in serum and synovial fluid of patients with active and severe rheumatoid arthritis. Ann Rheum Dis 1990; 49: 665–667. DOI: 10.1136/ard.49.9.665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Burmester GR, Panaccione R, Gordon KB, et al. Adalimumab: long-term safety in 23 458 patients from global clinical trials in rheumatoid arthritis, juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis and Crohn’s disease. Ann Rheum Dis 2013; 72: 517–524. DOI: 10.1136/annrheumdis-2011-201244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Jacobsson LT, Turesson C, Nilsson JA, et al. Treatment with TNF blockers and mortality risk in patients with rheumatoid arthritis. Ann Rheum Dis 2007; 66: 670–675. DOI: 10.1136/ard.2006.062497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Solomon DH, Giles JT, Liao KP, et al. Reducing cardiovascular risk with immunomodulators: a randomised active comparator trial among patients with rheumatoid arthritis. Ann Rheum Dis 2023; 82: 324–330. DOI: 10.1136/ard-2022-223302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Sepriano A, Kerschbaumer A, Bergstra SA, et al. Safety of synthetic and biological DMARDs: a systematic literature review informing the 2022 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis 2023; 82: 107–118. DOI: 10.1136/ard-2022-223357. [DOI] [PubMed] [Google Scholar]
  • 137.Finzel S, Rech J, Schmidt S, et al. Repair of bone erosions in rheumatoid arthritis treated with tumour necrosis factor inhibitors is based on bone apposition at the base of the erosion. Ann Rheum Dis 2011; 70: 1587–1593. DOI: 10.1136/ard.2010.148395. [DOI] [PubMed] [Google Scholar]
  • 138.Madhok R, Crilly A, Watson J, et al. Serum interleukin 6 levels in rheumatoid arthritis: correlations with clinical and laboratory indices of disease activity. Ann Rheum Dis 1993; 52: 232–234. DOI: 10.1136/ard.52.3.232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Shimamoto K, Ito T, Ozaki Y, et al. Serum interleukin 6 before and after therapy with tocilizumab is a principal biomarker in patients with rheumatoid arthritis. J Rheumatol 2013; 40: 1074–1081. DOI: 10.3899/jrheum.121389. [DOI] [PubMed] [Google Scholar]
  • 140.Garnero P, Thompson E, Woodworth T, et al. Rapid and sustained improvement in bone and cartilage turnover markers with the anti-interleukin-6 receptor inhibitor tocilizumab plus methotrexate in rheumatoid arthritis patients with an inadequate response to methotrexate: results from a substudy of the multicenter double-blind, placebo-controlled trial of tocilizumab in inadequate responders to methotrexate alone. Arthritis Rheum 2010; 62: 33–43. DOI: 10.1002/art.25053. [DOI] [PubMed] [Google Scholar]
  • 141.Kremer JM, Blanco R, Brzosko M, et al. Tocilizumab inhibits structural joint damage in rheumatoid arthritis patients with inadequate responses to methotrexate: results from the double-blind treatment phase of a randomized placebo-controlled trial of tocilizumab safety and prevention of structural joint damage at one year. Arthritis Rheum 2011; 63: 609–621. DOI: 10.1002/art.30158. [DOI] [PubMed] [Google Scholar]
  • 142.Finzel S, Rech J, Schmidt S, et al. Interleukin-6 receptor blockade induces limited repair of bone erosions in rheumatoid arthritis: a micro CT study. Ann Rheum Dis 2013; 72: 396–400. DOI: 10.1136/annrheumdis-2011-201075. [DOI] [PubMed] [Google Scholar]
  • 143.Wright HL, Mewar D, Bucknall RC, et al. Synovial fluid IL-6 concentrations associated with positive response to tocilizumab in an RA patient with failed response to anti-TNF and rituximab. Rheumatology (Oxford) 2015; 54: 743–744. DOI: 10.1093/rheumatology/keu488. [DOI] [PubMed] [Google Scholar]
  • 144.Nishimoto N, Amano K, Hirabayashi Y, et al. Drug free REmission/low disease activity after cessation of tocilizumab (Actemra) monotherapy (DREAM) study. Mod Rheumatol 2014; 24: 17–25. DOI: 10.3109/14397595.2013.854079. [DOI] [PubMed] [Google Scholar]
  • 145.Usón J, Balsa A, Pascual-Salcedo D, et al. Soluble interleukin 6 (IL-6) receptor and IL-6 levels in serum and synovial fluid of patients with different arthropathies. J Rheumatol 1997; 24: 2069–2075. [PubMed] [Google Scholar]
  • 146.Bystrom J, Clanchy FI, Taher TE, et al. TNFα in the regulation of Treg and Th17 cells in rheumatoid arthritis and other autoimmune inflammatory diseases. Cytokine 2018; 101: 4–13. DOI: 10.1016/j.cyto.2016.09.001. [DOI] [PubMed] [Google Scholar]
  • 147.Kilani RT, Maksymowych WP, Aitken A, et al. Detection of high levels of 2 specific isoforms of 14-3-3 proteins in synovial fluid from patients with joint inflammation. J Rheumatol 2007; 34: 1650–1657. [PubMed] [Google Scholar]
  • 148.Maksymowych WP, Naides SJ, Bykerk V, et al. Serum 14-3-3η is a novel marker that complements current serological measurements to enhance detection of patients with rheumatoid arthritis. J Rheumatol 2014; 41: 2104–2113. DOI: 10.3899/jrheum.131446. [DOI] [PubMed] [Google Scholar]
  • 149.Wang D, Cui Y, Lei H, et al. Diagnostic accuracy of 14-3-3 η protein in rheumatoid arthritis: a meta-analysis. Int J Rheum Dis 2020; 23: 1443–1451. DOI: 10.1111/1756-185x.13921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Maksymowych WP, van der Heijde D, Allaart CF, et al. 14-3-3η is a novel mediator associated with the pathogenesis of rheumatoid arthritis and joint damage. Arthritis Res Ther 2014; 16: R99. DOI: 10.1186/ar4547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hirata S, Marotta A, Gui Y, et al. Serum 14-3-3η level is associated with severity and clinical outcomes of rheumatoid arthritis, and its pretreatment level is predictive of DAS28 remission with tocilizumab. Arthritis Res Ther 2015; 17: 280. DOI: 10.1186/s13075-015-0799-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Stanczyk J, Pedrioli DM, Brentano F, et al. Altered expression of MicroRNA in synovial fibroblasts and synovial tissue in rheumatoid arthritis. Arthritis Rheum 2008; 58: 1001–1009. DOI: 10.1002/art.23386. [DOI] [PubMed] [Google Scholar]
  • 153.Stanczyk J, Ospelt C, Karouzakis E, et al. Altered expression of microRNA-203 in rheumatoid arthritis synovial fibroblasts and its role in fibroblast activation. Arthritis Rheum 2011; 63: 373–381. DOI: 10.1002/art.30115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kurowska-Stolarska M, Alivernini S, Ballantine LE, et al. MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis. Proc Natl Acad Sci U S A 2011; 108: 11193–11198. DOI: 10.1073/pnas.1019536108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ogando J, Tardáguila M, Díaz-Alderete A, et al. Notch-regulated miR-223 targets the aryl hydrocarbon receptor pathway and increases cytokine production in macrophages from rheumatoid arthritis patients. Sci Rep 2016; 6: 20223. DOI: 10.1038/srep20223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Pauley KM, Satoh M, Chan AL, et al. Upregulated miR-146a expression in peripheral blood mononuclear cells from rheumatoid arthritis patients. Arthritis Res Ther 2008; 10: R101. DOI: 10.1186/ar2493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Ouboussad L, Hunt L, Hensor EMA, et al. Profiling microRNAs in individuals at risk of progression to rheumatoid arthritis. Arthritis Res Ther 2017; 19: 288. DOI: 10.1186/s13075-017-1492-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Anaparti V, Smolik I, Meng X, et al. Whole blood microRNA expression pattern differentiates patients with rheumatoid arthritis, their seropositive first-degree relatives, and healthy unrelated control subjects. Arthritis Res Ther 2017; 19: 249. DOI: 10.1186/s13075-017-1459-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Cunningham CC, Wade S, Floudas A, et al. Serum miRNA signature in rheumatoid arthritis and “At-Risk individuals”. Front Immunol 2021; 12: 633201. DOI: 10.3389/fimmu.2021.633201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Murata K, Furu M, Yoshitomi H, et al. Comprehensive microRNA analysis identifies miR-24 and miR-125a-5p as plasma biomarkers for rheumatoid arthritis. PLoS One 2013; 8: e69118. DOI: 10.1371/journal.pone.0069118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Ormseth MJ, Solus JF, Vickers KC, et al. Utility of select plasma MicroRNA for disease and cardiovascular risk assessment in patients with rheumatoid arthritis. J Rheumatol 2015; 42: 1746–1751. DOI: 10.3899/jrheum.150232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Murata K, Yoshitomi H, Tanida S, et al. Plasma and synovial fluid microRNAs as potential biomarkers of rheumatoid arthritis and osteoarthritis. Arthritis Res Ther 2010; 12: R86. DOI: 10.1186/ar3013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Filková M, Aradi B, Senolt L, et al. Association of circulating miR-223 and miR-16 with disease activity in patients with early rheumatoid arthritis. Ann Rheum Dis 2014; 73: 1898–1904. DOI: 10.1136/annrheumdis-2012-202815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Castro-Villegas C, Pérez-Sánchez C, Escudero A, et al. Circulating miRNAs as potential biomarkers of therapy effectiveness in rheumatoid arthritis patients treated with anti-TNFα. Arthritis Res Ther 2015; 17: 49. DOI: 10.1186/s13075-015-0555-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Duroux-Richard I, Pers YM, Fabre S, et al. Circulating miRNA-125b is a potential biomarker predicting response to rituximab in rheumatoid arthritis. Mediators Inflamm 2014; 2014: 342524. DOI: 10.1155/2014/342524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Sode J, Krintel SB, Carlsen AL, et al. Plasma MicroRNA profiles in patients with early rheumatoid arthritis responding to adalimumab plus methotrexate vs methotrexate alone: a placebo-controlled clinical trial. J Rheumatol 2018; 45: 53–61. DOI: 10.3899/jrheum.170266. [DOI] [PubMed] [Google Scholar]
  • 167.Grillet B, Pereira RVS, Van Damme J, et al. Matrix metalloproteinases in arthritis: towards precision medicine. Nat Rev Rheumatol 2023; 19: 363–377. DOI: 10.1038/s41584-023-00966-w. [DOI] [PubMed] [Google Scholar]
  • 168.Manicourt DH, Fujimoto N, Obata K, et al. Levels of circulating collagenase, stromelysin-1, and tissue inhibitor of matrix metalloproteinases 1 in patients with rheumatoid arthritis. Relationship to serum levels of antigenic keratan sulfate and systemic parameters of inflammation. Arthritis Rheum 1995; 38: 1031–1039. DOI: 10.1002/art.1780380803. [DOI] [PubMed] [Google Scholar]
  • 169.Posthumus MD, Limburg PC, Westra J, et al. Serum levels of matrix metalloproteinase-3 in relation to the development of radiological damage in patients with early rheumatoid arthritis. Rheumatology (Oxford) 1999; 38: 1081–1087. DOI: 10.1093/rheumatology/38.11.1081. [DOI] [PubMed] [Google Scholar]
  • 170.Green MJ, Gough AK, Devlin J, et al. Serum MMP-3 and MMP-1 and progression of joint damage in early rheumatoid arthritis. Rheumatology (Oxford) 2003; 42: 83–88. DOI: 10.1093/rheumatology/keg037. [DOI] [PubMed] [Google Scholar]
  • 171.Keyszer G, Lambiri I, Nagel R, et al. Circulating levels of matrix metalloproteinases MMP-3 and MMP-1, tissue inhibitor of metalloproteinases 1 (TIMP-1), and MMP-1/TIMP-1 complex in rheumatic disease. Correlation with clinical activity of rheumatoid arthritis versus other surrogate markers. J Rheumatol 1999; 26: 251–258. [PubMed] [Google Scholar]
  • 172.Ribbens C, Martin y Porras M, Franchimont N, et al. Increased matrix metalloproteinase-3 serum levels in rheumatic diseases: relationship with synovitis and steroid treatment. Ann Rheum Dis 2002; 61: 161–166. DOI: 10.1136/ard.61.2.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Yamanaka H, Matsuda Y, Tanaka M, et al. Serum matrix metalloproteinase 3 as a predictor of the degree of joint destruction during the six months after measurement, in patients with early rheumatoid arthritis. Arthritis Rheum 2000; 43: 852–858. DOI: . [DOI] [PubMed] [Google Scholar]
  • 174.Ponchel F, Morgan AW, Bingham SJ, et al. Dysregulated lymphocyte proliferation and differentiation in patients with rheumatoid arthritis. Blood 2002; 100: 4550–4556. DOI: 10.1182/blood-2002-03-0671. [DOI] [PubMed] [Google Scholar]
  • 175.Carvalheiro H, Duarte C, Silva-Cardoso S, et al. CD8+ T cell profiles in patients with rheumatoid arthritis and their relationship to disease activity. Arthritis Rheumatol 2015; 67: 363–371. DOI: 10.1002/art.38941. [DOI] [PubMed] [Google Scholar]
  • 176.Lawson CA, Brown AK, Bejarano V, et al. Early rheumatoid arthritis is associated with a deficit in the CD4+CD25high regulatory T cell population in peripheral blood. Rheumatology (Oxford) 2006; 45: 1210–1217. DOI: 10.1093/rheumatology/kel089. [DOI] [PubMed] [Google Scholar]
  • 177.Hunt L, Hensor EM, Nam J, et al. T cell subsets: an immunological biomarker to predict progression to clinical arthritis in ACPA-positive individuals. Ann Rheum Dis 2016; 75: 1884–1889. DOI: 10.1136/annrheumdis-2015-207991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Saleem B, Keen H, Goeb V, 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. DOI: 10.1136/ard.2009.117341. [DOI] [PubMed] [Google Scholar]
  • 179.Gul HL, Di Matteo A, Mankia K, et al. Can biomarkers predict successful tapering of conventional disease-modifying therapy in rheumatoid arthritis patients in stable remission? Clin Exp Rheumatol 2023; 41: 126–136. [DOI] [PubMed] [Google Scholar]
  • 180.Szalay B, Vásárhelyi B, Cseh A, et al. The impact of conventional DMARD and biological therapies on CD4+ cell subsets in rheumatoid arthritis: a follow-up study. Clin Rheumatol 2014; 33: 175–185. DOI: 10.1007/s10067-013-2352-x. [DOI] [PubMed] [Google Scholar]
  • 181.Avdeeva A, Rubtsov Y, Dyikanov D, et al. Regulatory T cells in patients with early untreated rheumatoid arthritis: phenotypic changes in the course of methotrexate treatment. Biochimie 2020; 174: 9–17. DOI: 10.1016/j.biochi.2020.03.014. [DOI] [PubMed] [Google Scholar]
  • 182.Ponchel F, Burska AN, Hunt L, et al. T-cell subset abnormalities predict progression along the Inflammatory Arthritis disease continuum: implications for management. Sci Rep 2020; 10: 3669. DOI: 10.1038/s41598-020-60314-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Wakefield RJ, Gibbon WW, Conaghan PG, et al. The value of sonography in the detection of bone erosions in patients with rheumatoid arthritis: a comparison with conventional radiography. Arthritis Rheum 2000; 43: 2762–2770. DOI: . [DOI] [PubMed] [Google Scholar]
  • 184.Baillet A, Gaujoux-Viala C, Mouterde G, et al. Comparison of the efficacy of sonography, magnetic resonance imaging and conventional radiography for the detection of bone erosions in rheumatoid arthritis patients: a systematic review and meta-analysis. Rheumatology (Oxford) 2011; 50: 1137–1147. DOI: 10.1093/rheumatology/keq437. [DOI] [PubMed] [Google Scholar]
  • 185.Colebatch AN, Edwards CJ, Østergaard M, et al. EULAR recommendations for the use of imaging of the joints in the clinical management of rheumatoid arthritis. Ann Rheum Dis 2013; 72: 804–814. DOI: 10.1136/annrheumdis-2012-203158. [DOI] [PubMed] [Google Scholar]
  • 186.Di Matteo A, Mankia K, Nam JL, et al. In anti-CCP+ at-risk individuals, radiographic bone erosions are uncommon and are not associated with the development of clinical arthritis. Rheumatology (Oxford) 2021; 60: 3156–3164. DOI: 10.1093/rheumatology/keaa761. [DOI] [PubMed] [Google Scholar]
  • 187.Thabet MM, Huizinga TW, van der Heijde DM, et al. The prognostic value of baseline erosions in undifferentiated arthritis. Arthritis Res Ther 2009; 11: R155. DOI: 10.1186/ar2832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.van Nies JA, van Steenbergen HW, Krabben A, et al. Evaluating processes underlying the predictive value of baseline erosions for future radiological damage in early rheumatoid arthritis. Ann Rheum Dis 2015; 74: 883–889. DOI: 10.1136/annrheumdis-2013-204659. [DOI] [PubMed] [Google Scholar]
  • 189.Gossec L, Dougados M, Goupille P, et al. Prognostic factors for remission in early rheumatoid arthritis: a multiparameter prospective study. Ann Rheum Dis 2004; 63: 675–680. DOI: 10.1136/ard.2003.010611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Quintana-Duque MA, Rondon-Herrera F, Mantilla RD, et al. Predictors of remission, erosive disease and radiographic progression in a Colombian cohort of early onset rheumatoid arthritis: a 3-year follow-up study. Clin Rheumatol 2016; 35: 1463–1473. DOI: 10.1007/s10067-016-3246-5. [DOI] [PubMed] [Google Scholar]
  • 191.Filippucci E, Cipolletta E, Mashadi Mirza R, et al. Ultrasound imaging in rheumatoid arthritis. Radiol Med 2019; 124: 1087–1100. DOI: 10.1007/s11547-019-01002-2. [DOI] [PubMed] [Google Scholar]
  • 192.Di Matteo A, Corradini D, Mankia K. What is the value of ultrasound in individuals ‘at-risk’ of rheumatoid arthritis who do not have clinical synovitis? Healthcare (Basel) 2021; 9: 752. DOI: 10.3390/healthcare9060752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Harnden K, Di Matteo A, Mankia K. When and how should we use imaging in individuals at risk of rheumatoid arthritis? Front Med (Lausanne) 2022; 9: 1058510. DOI: 10.3389/fmed.2022.1058510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Di Matteo A, De Lorenzis E, Duquenne L, et al. Ultrasound in anti-CCP+ at-risk individuals without clinical synovitis: development of a novel 6-joint protocol for feasible risk prediction. Rheumatology (Oxford) 2023; 63: 2213–2221. DOI: 10.1093/rheumatology/kead570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Molina Collada J, López Gloria K, Castrejón I, et al. Ultrasound in clinically suspect arthralgia: the role of power Doppler to predict rheumatoid arthritis development. Arthritis Res Ther 2021; 23: 299. DOI: 10.1186/s13075-021-02685-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Zufferey P, Rebell C, Benaim C, et al. Ultrasound can be useful to predict an evolution towards rheumatoid arthritis in patients with inflammatory polyarthralgia without anticitrullinated antibodies. Joint Bone Spine 2017; 84: 299–303. DOI: 10.1016/j.jbspin.2016.05.011. [DOI] [PubMed] [Google Scholar]
  • 197.Zayat AS, Ellegaard K, Conaghan PG, et al. The specificity of ultrasound-detected bone erosions for rheumatoid arthritis. Ann Rheum Dis 2015; 74: 897–903. DOI: 10.1136/annrheumdis-2013-204864. [DOI] [PubMed] [Google Scholar]
  • 198.Gutierrez M, Filippucci E, Salaffi F, et al. Differential diagnosis between rheumatoid arthritis and psoriatic arthritis: the value of ultrasound findings at metacarpophalangeal joints level. Ann Rheum Dis 2011; 70: 1111–1114. DOI: 10.1136/ard.2010.147272. [DOI] [PubMed] [Google Scholar]
  • 199.Cipolletta E, Filippucci E, Abhishek A, et al. In patients with acute mono/oligoarthritis, a targeted ultrasound scanning protocol shows great accuracy for the diagnosis of gout and CPPD. Rheumatology (Oxford) 2023; 62: 1493–1500. DOI: 10.1093/rheumatology/keac479. [DOI] [PubMed] [Google Scholar]
  • 200.Di Matteo A, De Angelis R, Cipolletta E, et al. Systemic lupus erythematosus arthropathy: the sonographic perspective. Lupus 2018; 27: 794–801. DOI: 10.1177/0961203317747716. [DOI] [PubMed] [Google Scholar]
  • 201.Nguyen H, Ruyssen-Witrand A, Gandjbakhch F, et al. 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 (Oxford) 2014; 53: 2110–2118. DOI: 10.1093/rheumatology/keu217. [DOI] [PubMed] [Google Scholar]
  • 202.Naredo E, Valor L, De la Torre I, et al. Ultrasound joint inflammation in rheumatoid arthritis in clinical remission: how many and which joints should be assessed? Arthritis Care Res (Hoboken) 2013; 65: 512–517. DOI: 10.1002/acr.21869. [DOI] [PubMed] [Google Scholar]
  • 203.Terslev L, Brahe CH, Østergaard M, et al. Using a DAS28-CRP-steered treat-to-target strategy does not eliminate subclinical inflammation as assessed by ultrasonography in rheumatoid arthritis patients in longstanding clinical remission. Arthritis Res Ther 2021; 23: 48. DOI: 10.1186/s13075-021-02426-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Silvagni E, Zandonella Callegher S, Mauric E, et al. Musculoskeletal ultrasound for treating rheumatoid arthritis to target-a systematic literature review. Rheumatology (Oxford) 2022; 61: 4590–4602. DOI: 10.1093/rheumatology/keac261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Naredo E, Valor L, De la Torre I, et al. Predictive value of Doppler ultrasound-detected synovitis in relation to failed tapering of biologic therapy in patients with rheumatoid arthritis. Rheumatology (Oxford) 2015; 54: 1408–1414. DOI: 10.1093/rheumatology/kev006. [DOI] [PubMed] [Google Scholar]
  • 206.Terslev L, Ostergaard M, Georgiadis S, et al. Flare during tapering of biological DMARDs in patients with rheumatoid arthritis in routine care: characteristics and predictors. RMD Open 2022; 8: e002796. DOI: 10.1136/rmdopen-2022-002796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Terslev L, Brahe CH, Hetland ML, et al. Doppler ultrasound predicts successful discontinuation of biological DMARDs in rheumatoid arthritis patients in clinical remission. Rheumatology (Oxford) 2021; 60: 5549–5559. DOI: 10.1093/rheumatology/keab276. [DOI] [PubMed] [Google Scholar]
  • 208.Kawashiri SY, Fujikawa K, Nishino A, et al. Ultrasound-detected bone erosion is a relapse risk factor after discontinuation of biologic disease-modifying antirheumatic drugs in patients with rheumatoid arthritis whose ultrasound power Doppler synovitis activity and clinical disease activity are well controlled. Arthritis Res Ther 2017; 19: 108. DOI: 10.1186/s13075-017-1320-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Haavardsholm EA, Aga AB, Olsen IC, et al. Ultrasound in management of rheumatoid arthritis: ARCTIC randomised controlled strategy trial. Bmj 2016; 354: i4205. DOI: 10.1136/bmj.i4205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Dale J, Stirling A, Zhang R, et al. Targeting ultrasound remission in early rheumatoid arthritis: the results of the TaSER study, a randomised clinical trial. Ann Rheum Dis 2016; 75: 1043–1050. DOI: 10.1136/annrheumdis-2015-208941. [DOI] [PubMed] [Google Scholar]
  • 211.Wouters F, Matthijssen X, Boeters DM, et al. Do magnetic resonance imaging-detected erosions predict progression to rheumatoid arthritis in patients presenting with clinically suspect arthralgia? A longitudinal study. Scand J Rheumatol 2020; 49: 461–467. DOI: 10.1080/03009742.2020.1737221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Mankia K, D’Agostino MA, Rowbotham E, et al. MRI inflammation of the hand interosseous tendons occurs in anti-CCP-positive at-risk individuals and may precede the development of clinical synovitis. Ann Rheum Dis 2019; 78: 781–786. DOI: 10.1136/annrheumdis-2018-214331. [DOI] [PubMed] [Google Scholar]
  • 213.de Pablo P, Dinnes J, Berhane S, et al. Systematic review of imaging tests to predict the development of rheumatoid arthritis in people with unclassified arthritis. Semin Arthritis Rheum 2022; 52: 151919. DOI: 10.1016/j.semarthrit.2021.10.003. [DOI] [PubMed] [Google Scholar]
  • 214.Haavardsholm EA, Bøyesen P, Østergaard M, et al. Magnetic resonance imaging findings in 84 patients with early rheumatoid arthritis: bone marrow oedema predicts erosive progression. Ann Rheum Dis 2008; 67: 794–800. DOI: 10.1136/ard.2007.071977. [DOI] [PubMed] [Google Scholar]
  • 215.Hetland ML, Ejbjerg B, Hørslev-Petersen K, et al. MRI bone oedema is the strongest predictor of subsequent radiographic progression in early rheumatoid arthritis. Results from a 2-year randomised controlled trial (CIMESTRA). Ann Rheum Dis 2009; 68: 384–390. DOI: 10.1136/ard.2008.088245. [DOI] [PubMed] [Google Scholar]
  • 216.Lisbona MP, Pàmies A, Ares J, et al. Association of bone edema with the progression of bone erosions quantified by hand magnetic resonance imaging in patients with rheumatoid arthritis in remission. J Rheumatol 2014; 41: 1623–1629. DOI: 10.3899/jrheum.130902. [DOI] [PubMed] [Google Scholar]
  • 217.Gandjbakhch F, Foltz V, Mallet A, et al. Bone marrow oedema predicts structural progression in a 1-year follow-up of 85 patients with RA in remission or with low disease activity with low-field MRI. Ann Rheum Dis 2011; 70: 2159–2162. DOI: 10.1136/ard.2010.149377. [DOI] [PubMed] [Google Scholar]
  • 218.Møller-Bisgaard S, Hørslev-Petersen K, Ejbjerg B, et al. Effect of magnetic resonance imaging vs conventional treat-to-target strategies on disease activity remission and radiographic progression in rheumatoid arthritis: the IMAGINE-RA randomized clinical trial. JAMA 2019; 321: 461–472. DOI: 10.1001/jama.2018.21362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Burgers LE, Boeters DM, Reijnierse M, et al. Does the presence of magnetic resonance imaging-detected osteitis at diagnosis with rheumatoid arthritis lower the risk for achieving disease-modifying antirheumatic drug-free sustained remission: results of a longitudinal study. Arthritis Res Ther 2018; 20: 68. DOI: 10.1186/s13075-018-1553-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Ahmad HA, Baker JF, Conaghan PG, et al. Prediction of flare following remission and treatment withdrawal in early rheumatoid arthritis: post hoc analysis of a phase IIIb trial with abatacept. Arthritis Res Ther 2022; 24: 47. DOI: 10.1186/s13075-022-02735-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Centola M, Cavet G, Shen Y, et al. Development of a multi-biomarker disease activity test for rheumatoid arthritis. PLoS One 2013; 8: e60635. DOI: 10.1371/journal.pone.0060635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Hirata S, Dirven L, Shen Y, et al. A multi-biomarker score measures rheumatoid arthritis disease activity in the BeSt study. Rheumatology (Oxford) 2013; 52: 1202–1207. DOI: 10.1093/rheumatology/kes362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Meznerics FA, Kemény LV, Gunther E, et al. Multibiomarker disease activity score: an objective tool for monitoring rheumatoid arthritis? A systematic review and meta-analysis. Rheumatology (Oxford) 2023; 62: 2048–2059. DOI: 10.1093/rheumatology/keac715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Curtis JR, Weinblatt ME, Shadick NA, et al. Validation of the adjusted multi-biomarker disease activity score as a prognostic test for radiographic progression in rheumatoid arthritis: a combined analysis of multiple studies. Arthritis Res Ther 2021; 23: 1. DOI: 10.1186/s13075-020-02389-4. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Annals of Clinical Biochemistry are provided here courtesy of SAGE Publications

RESOURCES