Skip to main content

Tailored therapeutic decision of rheumatoid arthritis using proteomic strategies: how to start and when to stop?

Abstract

Unpredictable treatment responses have been an obstacle for the successful management of rheumatoid arthritis. Although numerous serum proteins have been proposed, there is a lack of integrative survey to compare their relevance in predicting treatment outcomes in rheumatoid arthritis. Also, little is known about their applications in various treatment stages, such as dose modification, drug switching or withdrawal. Here we present an in-depth exploration of the potential usefulness of serum proteins in clinical decision-making and unveil the spectrum of immunopathology underlying responders to different drugs. Patients with robust autoimmunity and inflammation are more responsive to biological treatments and prone to relapse during treatment de-escalation. Moreover, the concentration changes of serum proteins at the beginning of the treatments possibly assist early recognition of treatment responders. With a better understanding of the relationship between the serum proteome and treatment responses, personalized medicine in rheumatoid arthritis will be more achievable in the near future.

Introduction

Rheumatoid arthritis (RA) is a highly heterogeneous autoimmune disease characterized by chronic inflammation and joint destruction. Recent advances in biological disease-modifying anti-rheumatic drugs (bDMARDs) have revolutionized the management of RA [1], with four main groups available: anti-CD20 antibody rituximab, cytotoxic T lymphocyte-associated antigen 4‐immunoglobulin (CTLA4-Ig) abatacept, interleukin (IL)-6 receptor inhibitors (such as tocilizumab and sarilumab), and tumor necrosis factor inhibitors (TNFi) which include infliximab, adalimumab, golimumab, certolizumab, and etanercept. Furthermore, targeted small molecule inhibitors (e.g., tofacitinib and baricitinib) have emerged as new therapeutic options. Nevertheless, timely selections of appropriate treatments for individual RA patients remains challenging. Biomarkers for the characterization of different RA phenotypes are urgently needed to develop personalized treatment plans.

Proteomics is a valuable research for identifying functional molecules directly involved in the pathophysiology of RA. Serum protein analysis has been widely adopted in clinical practice for its less invasive nature and easy reproducibility compared with synovial sampling [2]. Mass spectrometry (MS) and immunoassays are two conventional methodologies for serum protein analysis. The former is a high-throughput technique to identify and quantify proteins by measuring their mass-to-charge ratios (m/z) and signal intensities. Proteins are ionized by electron ionization, electrospray ionization, or matrix-assisted laser desorption/ionization, and then separated based on mass-to-charge ratios before detection. MS is a powerful tool for identifying unknown compounds, determining the purity of a sample, and analyzing complex mixtures. On the other hand, immunoassays are used to detect and quantify specific proteins in a sample based on the specific binding between an antibody and its corresponding target molecules. In a typical immunoassay, a specific antibody is coated on a plate, and the sample is added to the plate. If the target molecule is present in the sample, it will bind to the antibody, forming an antigen–antibody complex that can be detected using a secondary antibody conjugated with an enzyme or fluorophore. The intensity of the signal generated by the enzyme or fluorophore is proportional to the amount of the target molecule in the sample, allowing for quantification. Both MS and immunoassays have their own unique strengths and limitations, and the choice of techniques mostly depends on specific experiment goals and sample characteristics [3].

In RA patients, serum proteins derived from the inflamed joints can exert systemic effects through interaction with liver, adipose tissue, connective tissues and circulating blood cells (Fig. 1a). Understanding the serum proteome has the potential to uncover fundamental factors that drive the immunopathogenesis of RA, thereby improving its management. Despite growing interests in serum proteomics, there is currently a lack of collective evidence to clarify their relevance in predicting treatment responses. To bridge this gap, our study aims to explore the clinical applications of serum proteins in four treatment stages of RA (Fig. 1b), including (1) predicting clinical responses before treatments. Since there is currently no effective cure, a trial-and-error process remains a common strategy for RA patients, which is very time-consuming and expensive. It is estimated that about one third of RA patients do not respond to the initial medications [4]. Bergman et al. have shown that predicting responses before treatment can offer several advantages for RA patients, such as improving outcomes, reducing costs, and minimizing exposure to ineffective medications [5]. Therefore, in the first stage, we will investigate the potential of serum proteins as indicators before the treatments. (2) early evaluation of therapeutic effectiveness for treatment adjustments. Most physicians rely on clinical experiences and patient-reported outcomes to evaluate treatment effectiveness, which could be subjective and may not truly reflect the underlying disease activity. In the second stage, we will discuss whether biomarker dynamics can be used as an objective measure of early response or non-response to facilitate treatment adjustments. (3) therapeutic drug monitoring during treatments. RA is a chronic disease that requires ongoing management, and the drug concentrations in serum can vary between individuals. Additionally, some RA medications can have potential side effects, so monitoring is important to ensure that the treatment is working well and not causing any harm. In the third stage, we will explore the utility of drug levels and anti-drug antibodies in guiding dose modification strategies. (4) predicting successful treatment withdrawal after achieving clinical remission. In some cases, it may be possible to withdraw or reduce the dose of RA medication if the disease is well-controlled and the patient has been in remission for a certain period of time. However, the timing and process of treatment withdrawal should be carefully managed to avoid disease flares or recurrence. Molecular remission, referring to a state in which the disease activity is undetectable at the molecular level, has increasingly become an important goal in the treatments of RA [6, 7]. In the fourth stage, we will investigate the relationship between the RA patients’ serum proteins and molecular remission to offer insights on successful treatment discontinuation. The possible bottlenecks occurred in each stage are summarized in Box 1.

Fig. 1
figure 1

Serum proteome in the therapeutic decision of rheumatoid arthritis. a The serum proteome results from the interaction between inflamed joints and various tissues, such as liver, adipose tissue, connective tissues, and circulating blood cells. macrophages, DCs dendritic cells, DAMPs danger-associated molecular pattern molecules, ECM extracellular matrix. b Serum proteins can aid in clinical decision-making in different stages of RA treatments. The trapezoid shadow represents the period of treatment administration followed by gradual tapering. The dashed line depicts the changes in disease activity in patients who achieved remission with treatments but relapsed after treatment withdrawal. The solid line illustrates the changes in disease activity in non-responders

To explore the clinical applications of serum proteins in RA treatments, a literature search was performed using PubMed, Embase, and Cochrane Library databases from four aspects: "the patient populations (rheumatoid arthritis)", " the purpose of the study (predict)", "serum protein biomarkers", and "outcome parameters". All keywords are listed in (Additional file 2: Table S1). As a result, 1553 articles were found, among which 476 were duplicates, and only the articles focused on serum protein biomarkers in the observational cohorts or clinical trials of RA patients were kept through manual inspection. Finally, a total of 276 articles were selected and discussed in the following paragraphs.

Predicting clinical responses before treatments

Predicting treatment responses remains challenging for RA patients. Serum proteins claimed to be useful for predicting therapeutic effectiveness in some studies may be considered irrelevant in other studies. Therefore, we will thoroughly discuss the relationship between serum proteins and the prediction of clinical responses in this section.

Autoantibodies


A number of autoantibodies have shown promise in predicting treatment responses, as illustrated in Fig. 2a. Rheumatoid factors (RF) and antibodies against cyclic citrullinated peptide (anti‐CCP) are two autoantibodies that play pivotal roles in the diagnosis and classification of RA. RF is an antibody against the fragment crystallizable (Fc) region of immunoglobulin G (IgG) and anti‐CCP is a subset of anti-citrullinated protein antibodies (ACPAs) targeting citrullinated antigens. Based on our survey, both RF and anti-CCP are positively associated with treatment responses to rituximab, and are insignificantly associated with TNFi (Fig. 2a), indicating that RF and anti-CCP are potentially useful for the prediction of treatment responses to rituximab. This investigation is also consistent with a pooled analysis of 16 RA registries [8]. Interestingly, the relationships between autoantibodies and treatment responses are not always consistent for the drugs belonging to the same group. For example, both sarilumab and tocilizumab are IL-6 receptor inhibitors, and the presence of RF or anti-CCP is positively associated with the treatment responses to sarilumab but not to tocilizumab [9]. A similar phenomenon is observed in the small molecule inhibitors, where tofacitinib is positively associated with RF or anti-CCP while baricitinib is not [10, 11] (Fig. 2a).

Fig. 2
figure 2

Autoantibodies in the prediction of treatment responses. a The relationship between the single positivity of antibodies and the treatment outcomes in RA patients, with red lines indicating a positive association, double dashed green lines indicating an inconsistent association, and dashed grey lines indicating no significant association. b The coexistence of antibodies associated with favorable treatment responses is illustrated by connected areas, while the combination of antibodies associated with unfavourable responses is depicted by dashed lines. The references are provided in Additional file 2: Table S2. Anti-PAD4 anti-peptidylarginine deiminase 4, anti-Pg anti-Porphyromonas gingivalis, anti-CarP anti–carbamylated protein, anti-MAA anti-malondialdehyde-acetaldehyde adducts, anti-hnRNP-DL anti-heterogeneous nuclear ribonucleoproteins-D-like protein, anti-CENPF anti-centromere protein F, anti-MCV anti-mutated citrullinated vimentin, AAPA anti-acetylated peptide antibodies

Proteins participating in the process of citrullination, such as the enzymes peptidylarginine deiminase 4 (PAD4) and the periodontitis-causing bacteria Porphyromonas gingivalis (Pg), can also trigger the production of autoantibodies (anti-PAD4 and anti-Pg) in RA patients. Nevertheless, based on our investigation, it is still inconclusive to justify the use of anti-PAD4 and anti-Pg as the predictors of treatment responses in RA [12,13,14,15,16,17]. In addition to citrullination, there are several autoantibodies produced against other post-translational modifications (PTMs) of proteins, including carbamylation (non-enzymatic conversion of lysine to homocitrulline in the presence of cyanate), acetylation (enzymatic addition of an acetyl group) and lipid peroxidation-generated adducts, which may also be helpful for the prediction of treatment responses [18,19,20]. For example, the presence of anti–carbamylated protein (anti-CarP) antibodies are reported to be associated with better clinical improvement using abatacept [21], but the similar observation was not found in treatments with TNFi [12]. Another example is antibodies against malondialdehyde-acetaldehyde adducts (anti-MAA). MAA adducts generated through lipid peroxidation are overexpressed in a variety of conditions with oxidative stress. Although the circulating levels of anti-MAA correlate with the extent of tissue damage [22, 23], no significant association was found between anti-MAA status and treatment responses to TNFi, tocilizumab, abatacept, and rituximab [24, 25] (Fig. 2a).

Noteworthy, autoantibodies against nuclear antigen recently emerged as potential predictors for TNFi, including antibodies against heterogeneous nuclear ribonucleoproteins-D-like protein (anti-hnRNP-DL) and centromere protein F (anti-CENPF) [26, 27]. These antibodies may be elicited during externalization of intracellular neoepitopes via neutrophil extracellular traps. A prospective investigation is desirable to warrant its use in clinical practice.

Combination of autoantibodies


Growing evidences suggested that combining different autoantibodies are helpful for predicting treatment responses (Fig. 2b). For example, Julià et al. have demonstrated that the coexistence of RF and anti-CCP predicts a better response to TNFi, while having both anti-CarP and anti-PAD4 implies unresponsiveness [12]. On the other hand, combining anti-CCP and anti-MCV (anti-mutated citrullinated vimentin) antibodies can better identify RA patients who are more likely to have unfavorable TNFi responses [28]. Additionally, while anti-MMA alone is not associated with TNFi response, combining it with RF and anti-CCP leads to an increasing odds ratio for responders in a dose-dependent manner [24, 25]. Moel et al. reported that patients with a variety of antibodies against citrullinated, carbamylated, and acetylated peptides (ACPA, anti-CarP, AAPA) were found to have better responses to RA treatments [29]. These results corroborate earlier studies that a higher number of positive autoantibodies is correlated with a greater likelihood of positive treatment responses [25, 29,30,31]. This could be due to the increased inflammatory burden from the loss of self-tolerance to multiple autoantigens, rendering patients more susceptible to anti-inflammatory treatments.

Myeloid and lymphoid markers


Myeloid markers are serum proteins that originate from myeloid cells, and some of them have been found to be correlated with treatment responses, such as 14–3-3η and calprotectin. 14–3-3η are primarily intracellular chaperones overexpressed in synovial macrophages [32]. Since 14–3-3η are released into extracellular space upon TNF-α stimulation [33, 34], a lower serum level of 14–3-3η possibly indicates less involvement of TNF-α in the nature of disease. In such cases, therapeutic approaches alternative to TNFi may be more effective. As reported by Hirata et al., patients with lower 14–3-3η before treatment are more likely to achieve remission in treatment with tocilizumab [35].

Calprotectin is a heterodimer consisting of two small calcium-binding proteins S100A8 and S100A9. When calprotectin is released by neutrophils and macrophages in response to cell stress, it acts as danger-associated molecular patterns (DAMPs) to promote inflammation and joint destruction [36]. Upon our integrative survey, patients with elevated levels of calprotectin before treatments are more likely to have better treatment outcomes (Additional file 1: Fig. S1). Intriguingly, the interpretation of the relationship between calprotectin levels and treatment outcomes may be affected by the analytical methods. Studies using MS have reported a positive association between elevated levels of calprotectin and treatment responses to etanercept, whereas studies using immunoassays do not.

Lymphoid markers are serum proteins associated with lymphoid cells. C-X-C motif chemokine 13 (CXCL13), an important chemokine involved in the migration and development of B cell follicles within the synovium [37,38,39,40], is initially proposed to be a negative indicator of TNFi response [41]. Nevertheless, the pre-treatment levels of CXCL13 demonstrate conflicting associations with the clinical responsiveness to TNFi upon collective investigation [42] (Additional file 1: Fig. S1).

The variation in immune cell populations has contributed to differences in the serum proteome (Fig. 3a) and is associated with the choice of therapeutic approaches (Fig. 3b). For example, RA patients with robust humoral immunity usually exhibit higher levels of autoantibodies, and rituximab and abatacept are recommended for these seropositive patients. In comparison, TNFi appears to be more effective for innate cell-mediated RA, especially for those enriched in myeloid markers, as proposed by Dennis et al. in which responders to TNFi were positively associated with synovial myeloid pathotypes [41]. Noteworthy, patients with multiple antibodies positive also benefit from TNFi, suggesting that TNFi may be effective across a wide range of both cell-mediated and humoral immune responses. The four groups of bDMARDs could be complementary with each other, and alternative treatments targeting different immunopathologies are suggested for inadequate responders to TNFi.

Fig. 3
figure 3

The immunopathology contributes to differences in serum proteome and the choice of treatments. a The spectrum of RA immunopathology ranges from innate immunity, cell-mediated to humoral immune response. The predominantly involved immune cells contribute to the distinctive compositions of serum proteins. DCs, dendritic cells; NETome, proteome associated with neutrophil extracellular traps. b Responders to four groups of bDMARDs are characterized by different biomarker spectrum, as illustrated by the color gradients. The darker areas of gradient lines represent enrichment of corresponding biomarkers, indicating the effective range of each bDMARD. The references are listed in Additional file 2: Table S3

Early evaluation of therapeutic effectiveness for treatment adjustments

Early changes in the concentrations of serum proteins can also serve as predictors of treatment outcomes. For example, a prominent decrease in C-reactive protein (CRP) within the first two weeks of treatment is associated with a favorable outcome at week 12 [43, 44]. A reduction in haptoglobin and other acute phase reactants by week 4 is also linked to better responses at week 14 [45]. In addition, similar observations have been noticed in several serum proteins, including autoantibodies, inflammatory mediators and extracellular matrix (ECM) markers. The relationship between the dynamics of these serum proteins and the prediction of subsequent treatment responses has been reported in treatments with TNFi (infliximab, adalimumab, golimumab), tocilizumab, abatacept and rituximab, as summarized in Fig. 4.

Fig. 4
figure 4

The concentration changes of serum proteins in the prediction of treatment responses. Biomarker concentrations were measured at treatment initiation (circles) and the early stage (squares), while clinical responses were evaluated at the end of treatment (triangles). The decrease in the levels of the listed biomarkers was associated with positive responses at the end of treatment. The references are provided in Additional file 2: Table S4, and the full names of biomarkers are listed in Additional file 2: Table S5. ECM extracellular matrix

Autoantibodies

The extent to which RF levels decrease serves as an early indicator of therapeutic effectiveness in several TNFi (infliximab and adalimumab) and tocilizumab [46,47,48,49]. Patients with decreasing RF levels during treatments ultimately have better treatment outcomes (Fig. 4). However, the degree of autoantibodies changes is unable to predict clinical improvement in rituximab [88,89,90,91].

The glycosylation profile of autoantibodies offers an additional insight. The effector functions of antibodies are modulated by the glycan structures on the Fc portion of IgG. The IgG glycosylation profile varies in different pathological conditions. It has been proposed that effective RA treatments would lead to a significantly increase of glycosylation [50,51,52,53,54], and patients with more increases in IgG galactosylation at the initial stage are more likely to have clinical improvement at the final assessment [53, 54]. Meanwhile, as reported by Ciregia et al., patients attaining clinical responses exhibit higher degrees of glycosylation in corticosteroid-binding globulin and lipopolysaccharide-binding protein after 12 months of treatments with corticoids, methotrexate, and bDMARDs [55]. Further research on the glycosylation profiles of serum proteins may be important in discovering predictors of RA treatment responses.

Inflammatory mediators

The dysregulation of immunological pathways in RA leads to the aberrant activation of inflammatory mediators, followed by erosion of cartilage and bone in joints [36]. Calprotectin has been regarded as a potential biomarker as the serum and synovial fluid levels of calprotectin are significantly increased in RA patients. Studies have demonstrated that patients with remarkable decreases in calprotectin levels after treatments with bDMARDs are more likely to experience favorable responses [56,57,58]. Similar observation is also found in 14–3-3η upon treatments with TNFi, tocilizumab and tofacitinib [32, 35, 59,60,61].

In contrast to calprotectin and 14–3-3η, other therapeutic targets, such as TNF-α and IL-6, are barely modulated by their corresponding inhibitors [54, 62,63,64]. The fast-acting and short-lived nature of cytokines make it difficult for accurate quantification. To address the challenge, Schotte et al. [65] and Dissanayake et al. [66] have utilized the enzyme-linked immunosorbent spot assay (ELISpot) to assess cytokines secreted by peripheral blood mononuclear cells. Still, cytokines do not serve as an ideal indicator for the treatment responses.

Extracellular matrix (ECM) remodelling

While it remains challenging to characterize cytokines, the downstream products such as the components of ECM are relatively long-lasting and directly reflect structural changes in joints. There are three categories of ECM markers, including bone remodelling, cartilage modulation, and synovial inflammation. Most bDMARDs demonstrated a positive effect on bone remodelling, with varying degrees of reductions in bone resorption markers and increments in bone formation markers [67,68,69]. Patients with early reduction in bone resorption markers, such as the decrease of receptor activator of nuclear factor kappa-B ligand, are more likely to experience favorable outcomes after six months of treatments with abatacept [70, 71]. As opposed to bone remodelling, most bDMARDs exert modest effects on cartilage modulation [72,73,74].

Matrix metalloproteinases (MMPs) are crucial mediators in proteolytic degradation and synovial destruction. A rapid downregulation of matrix metalloproteinases 3 (MMP-3) is found to be an early indicator of a favorable outcome to TNFi, tocilizumab, sarilumab and abatacept [17, 44, 68, 75], suggesting that successful suppression of MMP-3 activity at the beginning of treatment is able to impede progressive joint damage. On the other hand, there are several MMP-mediated products resulting from the degradation of type I collagen (C1M), type II collagen (C2M), type III collagen (C3M), type IV collagen (C4M) and CRP (CRPM). Decreases in the levels of these products indicate the reversal of ongoing synovial damage and better clinical responses [44, 75,76,77,78]. The extent of reduction in these markers within the first four weeks of tocilizumab treatments can aid in early distinguishing between responders and non-responders [77, 79, 80], and a similar phenomenon is also found in baricitinib [81]. While ECM markers were less useful for predicting therapeutic responses before treatment, the robustness of these findings suggested that the reduction in ECM markers may precede clinical outcome and enable early prediction of treatment results.

Combination of biomarkers

Considering the heterogeneous nature of RA, utilizing multiple biomarkers for prediction is an alternative solution. The multi-biomarker disease activity (MBDA) score, which is based on the measurement of 12 serum proteins (cytokines, growth factors, ECM and stress-related proteins), has been recognized as an objective assessment of disease activity and a predictor of radiographic progression [82,83,84,85]. Moreover, early changes in MBDA scores have been reported to be associated with clinical remission in treatments with TNFi (infliximab, adalimumab, and etanercept), rituximab, and tofacitinib [73,74,75,76,77].

In an era of high-throughput technology and advanced computational analysis, combining various molecular signatures to characterize clinical response has become a popular strategy. For instance, early changes in immunophenotyping and synovial cellular population enable the prediction of treatment responses [86, 87]. Tasaki et al. have developed statistical models to calculate the probability of patients classified as RA from the aspects of transcripts, proteins, and immunophenotypes [6]. They noticed that the difference in reduction of RA probability between responders and inadequate responders is more prominent based on a proteomic model than a transcriptional model, implying that serum proteome provides a stronger evidence towards healthy states. Furthermore, a greater reduction in the calculated probability after the first month of treatment is associated with a higher likelihood of clinical responsiveness at the 24th week. The early alteration in molecular signatures facilitates the prediction of subsequent responses, enabling a timely decision for clinical treatments.

Therapeutic drug monitoring during treatment

The variability of drug concentrations in serum is a critical issue for precision medicine. Therapeutic drug monitoring (TDM, monitoring the drug concentrations during treatments) provides a clue for the optimization of personalized treatments. Additionally, the production of anti-drug antibodies (ADAs) upon exposure to exogenous therapeutic proteins emerges as another important consideration. ADAs partly diminish the therapeutic effects of patients’ medications and the measurement of ADAs is potentially helpful in determining the cause of suboptimal drug level and aids in treatment modifications. The application of TDM in the management of RA has received growing attention (Table 1).

Table 1 Therapeutic drug monitoring in the prediction of rheumatoid arthritis treatment

Measurement of drug levels

The drug concentrations directly reflect the effectors of clinical responsiveness. Patients with higher drug levels are expected to experience clinical improvement. Studies have shown that the drug levels of infliximab, adalimumab, etanercept, and tocilizumab are positively associated with their clinical responses [88,89,90,91,92,93,94,95]. Nevertheless, it is not encouraged to reduce the dose of infliximab, adalimumab, and etanercept based on serum drug concentrations given current evidences [96,97,98,99].

Measurement of anti-drug antibodies

The formation of anti-drug antibodies (ADAs) is a major reason for inter-individual variations in serum drug levels [100, 101]. The prevalence of ADAs ranges from nearly undetectable in etanercept to 67% in infliximab and adalimumab [102, 103]. Factors influencing the emergence of ADAs include genetic predisposition, smoking habits, manufacturing process of bDMARDs, and the prolonged exposure of bDMARDs, in which the last factor is the riskiest [104]. Serum drug levels are affected by ADAs through various biological mechanisms, such as competitive inhibition or enhanced drug clearance. Most studies agree that ADAs against infliximab and adalimumab negatively impact the drug level [102, 103, 105,106,107,108,109,110,111,112,113], thereby hampering the accomplishment of therapeutic responses [89, 90, 102, 103, 105, 108, 109, 114,115,116,117]. Conversely, ADAs against tocilizumab, sarilumab, and rituximab are less relevant to clinical responses [102, 107, 113, 118,119,120,121]. Ongoing monitoring is still necessary to establish the clinical relevance, especially for patients with a higher risk of immunogenicity [122].

For patients with inadequate improvement under a given treatment, switching to another class of bDMARDs may be a more effective strategy than dose escalation [123]. Jamnitski et al. proposed that patients who developed ADAs to infliximab or adalimumab are more likely to have favorable responses after switching to etanercept [124]. Nevertheless, the use of ADAs for drug switching remains controversial [125].

Predicting successful treatment withdrawal

When the clinical symptoms of RA are well-controlled, physicians may start to explore the possibility of treatment de-escalation. Although treatment tapering has been recommended in recent guidelines [1], scarcely had it been implemented in clinical practice under the concern of disease flares [126, 127]. It would be of great help if one could identify patients most likely to benefit from treatment withdrawal.

Common clinical variables (e.g. age and swollen joint count) have been extensively discussed, but they are not recommended as ideal predictors [128, 129]. Molecular remission, on the other hand, has drawn an increasing attention because it requires a more stringent criterion that patients’ molecular profiles need to be close to those of healthy individuals. Identifying the serum proteome associated with the molecular remission has become a fascinating field. Table 2 summarizes the patient cohorts, interventions, primary outcomes, and clinical implications of recent efforts, in which nine studies agreed that seropositive patients have higher rates of disease relapses during tapering of tocilizumab and TNFi [29, 130,131,132,133,134,135,136,137]. In particular, the risk of relapses increases as more antibody reactivities are involved [29, 134]. Patients positive for a broad spectrum of autoantibodies are not only more responsive to bDMARDs, but also more likely to experience disease relapses during treatment discontinuation, highlighting the need of continuous treatments for these seropositive patients.

Table 2 Potential biomarkers associated with successful treatment withdrawal in rheumatoid arthritis

In our literature survey, two studies have demonstrated that patients with higher calprotectin levels at the moment of dose reduction were predisposed to relapse [138, 139], and four studies have investigated the application of MBDA in making treatment adjustments [133, 140,141,142]. Higher MBDA scores at the moment of intervention were positively associated with the risk of flares after treatment tapering [133] or cessation [140], but similar observations were not reported in other cohorts [141, 142]. More importantly, patients with a low MBDA score (< 30 units) and negative anti-CCP are at a lower risk of relapses, suggesting that the combination of MBDA score and anti-CCP status enables risk stratification for treatment withdrawal [143].

Generally speaking, lower levels of autoantibodies, calprotectin, and MBDA are associated with reduced risks of relapses. Persistent elevations of these serum proteins are implicated in subclinical inflammation and may hinder successful treatment de-escalation. The state in which the levels of molecules are akin to healthy controls is considered a more stabilized condition for tapering of bDMARDs. Patients attaining molecular remission are more appropriate for treatment adjustment as compared with those only having clinical remission, and the probability of sustained disease inactivation increases when more molecular classes achieve remission [6, 144]. A recent study reported by Inamo et al. has elucidated a distinct subset of CD4 + and CD8 + as the key components associated with molecular remission [7]. With multi-omics approaches, Tasaki et al. concluded that downregulation of neutrophils (and upregulation of natural killer cells) are correlated to remission in transcript-based models, whereas inactivation of the complement pathway is associated with remission in protein-based models [6, 145].

Conclusions and future directions

Personalized medicine remains an unmet need for RA patients. The considerable heterogeneity at the proteomic level has contributed to variations in therapeutic responses. In this review, we comprehensively summarize the clinical applications of serum proteins in different treatment stages. Upon collective investigation, some serum proteins that were initially suggested to be promising predictors in individual studies were found to have inconsistent associations. Our survey suggests that autoantibodies, calprotectin and 14–3-3η have a more consistent potential in predicting therapeutic outcomes, giving directions for further validation. We also shed light on the spectrum of RA immunopathology underlying between responders to different treatments. RA patients who exhibit dominant humoral immunity are more likely to respond to rituximab and abatacept, but they are also prone to relapse upon treatment cessation. On the other hand, RA patients with predominant cell-mediated immunity and myeloid cells tend to respond to TNFi. Serum protein profiling reveals a novel insight for personalized treatment strategies.

MS and immunoassays are currently the major analytical platforms for proteomic analyses. In our survey, complement components and apolipoproteins are commonly reported in the articles using MS, while cytokines, chemokines, and 14–3-3η proteins are frequently discussed in the studies using immunoassays (the analytical strategies of these articles are listed in Additional file 2: Table S3). Our observation is consistent with the article reported by Skalnikova et al. that shotgun proteomics might have difficulties in the detection of low abundant proteins (such as cytokines) without enrichment [3]. Another possible reason is the low molecular weights of cytokines, (e.g., the protein sequence lengths of IL-6 and TNF-α are 212 and 233, respectively) further pose challenges for their detection by shotgun proteomics. It is noteworthy that calprotectin can be identified using immunoassay-based and MS-based proteomics, yet the statistical significance varies between the two platforms, which highlights the need for a cross-platform normalization tool [146].

While current studies have identified several serum protein candidates, their validation in large clinical cohorts is essential for clinical practice. The challenges of biomarker discovery include the heterogeneous size and clinical features of patient cohorts, variability in analytical platforms, and the lack of reproducibility. As incorrect prediction may pose additional side effects and costs to the patients, ongoing researches are dedicated to combining different biomarkers using omics approaches, and refining proteomic techniques to address these issues. More advanced proteomic techniques, such as single-cell proteomics and imaging MS, are expected to improve the identification of protein biomarkers with higher sensitivity and specificity [147]. Recently, the incorporation of serum proteins as prediction models into the clinical care of RA has drawn increasing attention because it not only facilitates clinical decision-making, maximizes the possibility of successful treatments, but also reduces costs [5, 148]. In summary, our study provides a comprehensive discussion of the relationship between serum proteins and RA treatments, hoping to pave the way for precision medicine and overcome the limitation of the traditional "one-size-fits-all" approach for RA patients.

Availability of data and materials

Data are available from the corresponding author upon reasonable request.

Abbreviations

ACPAs:

Anti-citrullinated protein antibodies

ADAs:

Anti-drug antibodies

Anti-CarP:

Anti–carbamylated protein

Anti‐CCP:

Antibodies against cyclic citrullinated peptide

bDMARDs:

Biological disease-modifying anti-rheumatic drugs

DFR:

DMARD-free remission

ECM:

Extracellular matrix

IL:

Interleukin

MAA:

Malondialdehyde-acetaldehyde adducts

MBDA:

Multi-biomarker disease activity

MMPs:

Matrix metalloproteinases

RA:

Rheumatoid arthritis

RF:

Rheumatoid factors

TDM:

Therapeutic drug monitoring

TNFi:

Tumor necrosis factor inhibitors

References

  1. Smolen JS, Landewé RB, Bijlsma JW, Burmester GR, Dougados M, Kerschbaumer A, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann Rheum Dis. 2020;79(6):685–99. https://doi.org/10.1136/annrheumdis-2019-216655.

    Article  CAS  PubMed  Google Scholar 

  2. Tsuchiya H, Fujio K. Title current status of the search for biomarkers for optimal therapeutic drug selection for patients with rheumatoid arthritis. IJMS. 2021;22(17):9534. https://doi.org/10.3390/ijms22179534.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kupcova Skalnikova H, Cizkova J, Cervenka J, Vodicka P. Advances in proteomic techniques for cytokine analysis: focus on melanoma research. IJMS. 2017;18(12):2697. https://doi.org/10.3390/ijms18122697.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Law-Wan J, Sparfel M-A, Derolez S, Azzopardi N, Goupille P, Detert J, et al. Predictors of response to TNF inhibitors in rheumatoid arthritis: an individual patient data pooled analysis of randomised controlled trials. RMD Open. 2021;7(3):e001882. https://doi.org/10.1136/rmdopen-2021-001882.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Bergman MJ, Kivitz AJ, Pappas DA, Kremer JM, Zhang L, Jeter A, et al. Clinical utility and cost savings in predicting inadequate response to anti-TNF therapies in rheumatoid arthritis. Rheumatol Ther. 2020;7:775–92. https://doi.org/10.1007/s40744-020-00226-3.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Tasaki S, Suzuki K, Kassai Y, Takeshita M, Murota A, Kondo Y, et al. Multi-omics monitoring of drug response in rheumatoid arthritis in pursuit of molecular remission. Nat Commun. 2018;9(1):1–12. https://doi.org/10.1038/s41467-018-05044-4.

    Article  CAS  Google Scholar 

  7. Inamo J, Suzuki K, Takeshita M, Kondo Y, Okuzono Y, Koga K, et al. Molecular remission at T cell level in patients with rheumatoid arthritis. Sci Rep. 2021;11(1):1–9. https://doi.org/10.1038/s41598-021-96300-z.

    Article  CAS  Google Scholar 

  8. Courvoisier DS, Chatzidionysiou K, Mongin D, Lauper K, Mariette X, Morel J, et al. The impact of seropositivity on the effectiveness of biologic anti-rheumatic agents: results from a collaboration of 16 registries. Rheumatology. 2021;60(2):820–8. https://doi.org/10.1093/rheumatology/keaa393.

    Article  CAS  PubMed  Google Scholar 

  9. Genovese MC, Fleischmann R, Kivitz A, Lee E-B, Van Hoogstraten H, Kimura T, et al. Efficacy and safety of sarilumab in combination with csDMARDs or as monotherapy in subpopulations of patients with moderately to severely active rheumatoid arthritis in three phase III randomized, controlled studies. Arthritis Res Ther. 2020;22(1):1–17. https://doi.org/10.1186/s13075-020-02194-z.

    Article  CAS  Google Scholar 

  10. Bird P, Hall S, Nash P, Connell CA, Kwok K, Witcombe D, et al. Treatment outcomes in patients with seropositive versus seronegative rheumatoid arthritis in Phase III randomised clinical trials of tofacitinib. RMD Open. 2019;5(1):e000742. https://doi.org/10.1136/rmdopen-2018-000742.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Takahashi N, Asai S, Kobayakawa T, Kaneko A, Watanabe T, Kato T, et al. Predictors for clinical effectiveness of baricitinib in rheumatoid arthritis patients in routine clinical practice: data from a Japanese multicenter registry. Sci Rep. 2020;10(1):21907. https://doi.org/10.1038/s41598-020-78925-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Julià A, López-Lasanta M, Blanco F, Gómez A, Haro I, Mas AJ, et al. Interactions between rheumatoid arthritis antibodies are associated with the response to anti-tumor necrosis factor therapy. BMC Musculoskelet Disord. 2021;22(1):1–7. https://doi.org/10.1186/s12891-021-04248-y.

    Article  CAS  Google Scholar 

  13. Darrah E, Yu F, Cappelli LC, Rosen A, O’Dell JR, Mikuls TR. Association of baseline peptidylarginine deiminase 4 autoantibodies with favorable response to treatment escalation in rheumatoid arthritis. Arthritis Rheumatol. 2019;71(5):696–702. https://doi.org/10.1002/art.40791.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Halvorsen E, Haavardsholm E, Pollmann S, Boonen A, Van Der Heijde D, Kvien T, et al. Serum IgG antibodies to peptidylarginine deiminase 4 predict radiographic progression in patients with rheumatoid arthritis treated with tumour necrosis factor-α blocking agents. Ann Rheum Dis. 2009;68(2):249–52. https://doi.org/10.1136/ard.2008.094490.

    Article  CAS  PubMed  Google Scholar 

  15. Ling SF, Nair N, Verstappen SM, Barton A, Zucht H-D, Budde P, et al. Proteomic analysis to define predictors of treatment response to adalimumab or methotrexate in rheumatoid arthritis patients. Pharmacogenomics J. 2020;20(3):516–23. https://doi.org/10.1038/s41397-019-0139-4.

    Article  CAS  PubMed  Google Scholar 

  16. Arvikar SL, Collier DS, Fisher MC, Unizony S, Cohen GL, McHugh G, et al. Clinical correlations with Porphyromonas gingivalis antibody responses in patients with early rheumatoid arthritis. Arthritis Res Ther. 2013;15(5):1–12. https://doi.org/10.1186/ar4289.

    Article  CAS  Google Scholar 

  17. Rinaudo-Gaujous M, Blasco-Baque V, Miossec P, Gaudin P, Farge P, Roblin X, et al. Infliximab induced a dissociated response of severe periodontal biomarkers in rheumatoid arthritis patients. J Clin Med. 2019;8(5):751. https://doi.org/10.3390/jcm8050751.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Trouw LA, Rispens T, Toes RE. Beyond citrullination: other post-translational protein modifications in rheumatoid arthritis. Nat Rev Rheumatol. 2017;13(6):331–9. https://doi.org/10.1038/nrrheum.2017.15.

    Article  CAS  PubMed  Google Scholar 

  19. Carubbi F, Alunno A, Gerli R, Giacomelli R. Post-translational modifications of proteins: novel insights in the autoimmune response in rheumatoid arthritis. Cells. 2019;8(7):657. https://doi.org/10.3390/cells8070657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wu C-Y, Yang H-Y, Luo S-F, Lai J-H. From rheumatoid factor to anti-citrullinated protein antibodies and anti-carbamylated protein antibodies for diagnosis and prognosis prediction in patients with rheumatoid arthritis. Int J Mol Sci. 2021;22(2):686. https://doi.org/10.3390/ijms22020686.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kumar R, Piantoni S, Boldini M, Garrafa E, Bazzani C, Fredi M, et al. Anti-carbamylated protein antibodies as a clinical response predictor in rheumatoid arthritis patients treated with abatacept. Clin Exp Rheumatol. 2021;39(1):91–7. https://doi.org/10.55563/clinexprheumatol/g8xqxr.

    Article  PubMed  Google Scholar 

  22. Rolla R, Vay D, Mottaran E, Parodi M, Traverso N, Aricó S, et al. Detection of circulating antibodies against malondialdehyde-acetaldehyde adducts in patients with alcohol-induced liver disease. Hepatology. 2000;31(4):878–84. https://doi.org/10.1053/he.2000.5373.

    Article  CAS  PubMed  Google Scholar 

  23. Anderson DR, Duryee MJ, Hunter CD, Garvin RP, Shurmur SW, Klassen LW, et al. Differentiating normal coronary arteries, stable atheromatous lesions and unstable atheromatous lesions: MAA-protein adducts and anti-MAA antibodies isotypes in patients with atherosclerotic disease and acute myocardial infarction. ATVB. 2012. https://doi.org/10.1161/atvb.32.suppl_1.A303.

    Article  Google Scholar 

  24. Mikuls TR, Coburn B, Sayles H, Yu F, Brophy M, O’Dell JR, et al. Antibody to malondialdehyde-acetaldehyde (MAA) adducts serve as biomarkers of treatment response in rheumatoid arthritis. Arthritis Rheumatol. 2015;67(3):645–55.

    PubMed  PubMed Central  Google Scholar 

  25. Petro AD, Dougherty J, England BR, Sayles H, Duryee MJ, Hunter CD, et al. Associations between an expanded autoantibody profile and treatment responses to biologic therapies in patients with rheumatoid arthritis. Int Immunopharmacol. 2021;91:107260. https://doi.org/10.1016/j.intimp.2020.107260.

    Article  CAS  PubMed  Google Scholar 

  26. Marklein B, Jenning M, Konthur Z, Häupl T, Welzel F, Nonhoff U, et al. The citrullinated/native index of autoantibodies against hnRNP-DL predicts an individual “window of treatment success” in RA patients. Arthritis Res Ther. 2021;23(1):1–16. https://doi.org/10.1186/s13075-021-02603-x.

    Article  CAS  Google Scholar 

  27. Lourido L, Ruiz-Romero C, Picchi F, Diz-Rosales N, Vilaboa-Galán S, Fernández-López C, et al. Association of serum anti-centromere protein F antibodies with clinical response to infliximab in patients with rheumatoid arthritis: A prospective study. Semin Arthritis Rheum. 2020. https://doi.org/10.1016/j.semarthrit.2020.06.010.

    Article  PubMed  Google Scholar 

  28. Fisher BA, Plant D, Lundberg K, Charles P, Barton A, Venables PJ, et al. Heterogeneity of anticitrullinated peptide antibodies and response to anti-tumor necrosis factor agents in rheumatoid arthritis. J Rheumatol. 2012;39(5):929–32. https://doi.org/10.3899/jrheum.111315.

    Article  CAS  PubMed  Google Scholar 

  29. de Moel EC, Derksen VF, Stoeken G, Trouw LA, Bang H, Goekoop RJ, et al. Baseline autoantibody profile in rheumatoid arthritis is associated with early treatment response but not long-term outcomes. Arthritis Res Ther. 2018;20(1):1–12. https://doi.org/10.1186/s13075-018-1520-4.

    Article  CAS  Google Scholar 

  30. de Moel EC, Derksen VF, Trouw LA, Bang H, Collée G, Lard LR, et al. In rheumatoid arthritis, changes in autoantibody levels reflect intensity of immunosuppression, not subsequent treatment response. Arthritis Res Ther. 2019;21(1):1–8. https://doi.org/10.1186/s13075-019-1815-0.

    Article  Google Scholar 

  31. Ferraccioli G, Tolusso B, Bobbio-Pallavicini F, Gremese E, Ravagnani V, Benucci M, et al. Biomarkers of good EULAR response to the B cell depletion therapy in all seropositive rheumatoid arthritis patients: clues for the pathogenesis. PLoS ONE. 2012. https://doi.org/10.1371/journal.pone.0040362.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Zeng T, Tan L. 14–3-3η protein: a promising biomarker for rheumatoid arthritis. Biomarkers Med. 2018;12(8):917–25. https://doi.org/10.1371/journal.pone.0040362.

    Article  CAS  Google Scholar 

  33. Trimova G, Yamagata K, Iwata S, Hirata S, Zhang T, Uemura F, et al. Tumour necrosis factor alpha promotes secretion of 14–3–3η by inducing necroptosis in macrophages. Arthritis Res Ther. 2020;22(1):1–11. https://doi.org/10.1186/s13075-020-2110-9.

    Article  CAS  Google Scholar 

  34. Munier CC, Ottmann C, Perry MW. 14–3-3 modulation of the inflammatory response. Pharmacol Res. 2021;163:105236. https://doi.org/10.1016/j.phrs.2020.105236.

    Article  CAS  PubMed  Google Scholar 

  35. Hirata S, Marotta A, Gui Y, Hanami K, Tanaka Y. 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(1):1–10. https://doi.org/10.1186/s13075-015-0799-7.

    Article  CAS  Google Scholar 

  36. Wang Q, Chen W, Lin J. The role of calprotectin in rheumatoid arthritis. J Transl Int Med. 2019;7(4):126–31. https://doi.org/10.2478/jtim-2019-0026.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Bechman K, Dalrymple A, Southey-Bassols C, Cope AP, Galloway JB. A systematic review of CXCL13 as a biomarker of disease and treatment response in rheumatoid arthritis. BMC Rheumatol. 2020;4(1):1–9. https://doi.org/10.1186/s41927-020-00154-3.

    Article  Google Scholar 

  38. Armas-González E, Domínguez-Luis MJ, Díaz-Martín A, Arce-Franco M, Castro-Hernández J, Danelon G, et al. Role of CXCL13 and CCL20 in the recruitment of B cells to inflammatory foci in chronic arthritis. Arthritis Res Ther. 2018;20(1):1–12. https://doi.org/10.1186/s13075-018-1611-2.

    Article  CAS  Google Scholar 

  39. Mandik-Nayak L, Huang G, Sheehan KC, Erikson J, Chaplin DD. Signaling through TNF receptor p55 in TNF-α-deficient mice alters the CXCL13/CCL19/CCL21 ratio in the spleen and induces maturation and migration of anergic B cells into the B cell follicle. J Immunol. 2001;167(4):1920–8. https://doi.org/10.4049/jimmunol.167.4.1920.

    Article  CAS  PubMed  Google Scholar 

  40. Corsiero E, Bombardieri M, Manzo A, Bugatti S, Uguccioni M, Pitzalis C. Role of lymphoid chemokines in the development of functional ectopic lymphoid structures in rheumatic autoimmune diseases. Immunol Lett. 2012;145(1–2):62–7. https://doi.org/10.1016/j.imlet.2012.04.013.

    Article  CAS  PubMed  Google Scholar 

  41. Dennis G, Holweg CT, Kummerfeld SK, Choy DF, Setiadi AF, Hackney JA, et al. Synovial phenotypes in rheumatoid arthritis correlate with response to biologic therapeutics. Arthritis Res Ther. 2014;16(2):1–18. https://doi.org/10.1186/ar4555.

    Article  CAS  Google Scholar 

  42. Zhao J, Ye X, Zhang Z. The predictive value of serum soluble ICAM-1 and CXCL13 in the therapeutic response to TNF inhibitor in rheumatoid arthritis patients who are refractory to csDMARDs. Clin Rheumatol. 2020;39:2573–81. https://doi.org/10.1007/s10067-020-05043-1.

    Article  PubMed  Google Scholar 

  43. Buch MH, Seto Y, Bingham SJ, Bejarano V, Bryer D, White J, et al. C-reactive protein as a predictor of infliximab treatment outcome in patients with rheumatoid arthritis: defining subtypes of nonresponse and subsequent response to etanercept. Arthritis Rheum. 2005;52(1):42–8. https://doi.org/10.1002/art.20711.

    Article  CAS  PubMed  Google Scholar 

  44. Gabay C, Msihid J, Zilberstein M, Paccard C, Lin Y, Graham NM, et al. Identification of sarilumab pharmacodynamic and predictive markers in patients with inadequate response to TNF inhibition: a biomarker substudy of the phase 3 TARGET study. RMD Open. 2018;4(1):e000607. https://doi.org/10.1136/rmdopen-2017-000607.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Visvanathan S, Rahman MU, Keystone E, Genovese M, Klareskog L, Hsia E, et al. Association of serum markers with improvement in clinical response measures after treatment with golimumab in patients with active rheumatoid arthritis despite receiving methotrexate: results from the GO-FORWARD study. Arthritis Res Ther. 2010;12:1–11. https://doi.org/10.1186/ar3188.

    Article  CAS  Google Scholar 

  46. Bobbio-Pallavicini F, Caporali R, Bugatti S, Montecucco C. What can we learn from treatment-induced changes in rheumatoid factor and anti-citrullinated peptide antibodies? J Rheumatol. 2008;35(10):1903–5.

    CAS  PubMed  Google Scholar 

  47. Modi S, Soejima M, Levesque M. The effect of targeted rheumatoid arthritis therapies on anti-citrullinated protein autoantibody levels and B cell responses. Clin Exp Immunol. 2013;173(1):8–17. https://doi.org/10.1111/cei.12114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Smolen JS, Beaulieu A, Rubbert-Roth A, Ramos-Remus C, Rovensky J, Alecock E, et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet. 2008;371(9617):987–97. https://doi.org/10.1016/S0140-6736(08)60453-5.

    Article  CAS  PubMed  Google Scholar 

  49. Yamada H, Tsuru T, Otsuka T, Maekawa M, Harada H, Fukuda T, et al. Abatacept reduces disease activity of rheumatoid arthritis independently of modulating anti-citrullinated peptide antibody production. Immunol Med. 2020;43(2):87–91. https://doi.org/10.1080/25785826.2020.1718833.

    Article  PubMed  Google Scholar 

  50. Gińdzieńska-Sieśkiewicz E, Radziejewska I, Domysławska I, Klimiuk PA, Sulik A, Rojewska J, et al. Changes of glycosylation of IgG in rheumatoid arthritis patients treated with methotrexate. Adv Med Sci. 2016;61(2):193–7. https://doi.org/10.1016/j.advms.2015.12.009.

    Article  PubMed  Google Scholar 

  51. Pasek M, Duk M, Podbielska M, Sokolik R, Szechiński J, Lisowska E, et al. Galactosylation of IgG from rheumatoid arthritis (RA) patients–changes during therapy. Glycoconj J. 2006;23(7):463–71. https://doi.org/10.1007/s10719-006-5409-0.

    Article  CAS  PubMed  Google Scholar 

  52. Van Beneden K, Coppieters K, Laroy W, De Keyser F, Hoffman IE, Van den Bosch F, et al. Reversible changes in serum immunoglobulin galactosylation during the immune response and treatment of inflammatory autoimmune arthritis. Ann Rheum Dis. 2009;68(8):1360–5. https://doi.org/10.1136/ard.2008.089292.

    Article  CAS  PubMed  Google Scholar 

  53. Croce A, Firuzi O, Altieri F, Eufemi M, Agostino R, Priori R, et al. Effect of infliximab on the glycosylation of IgG of patients with rheumatoid arthritis. J Clin Lab Anal. 2007;21(5):303–14. https://doi.org/10.1002/jcla.20191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mesko B, Poliska S, Szamosi S, Szekanecz Z, Podani J, Varadi C, et al. Peripheral blood gene expression and IgG glycosylation profiles as markers of tocilizumab treatment in rheumatoid arthritis. J Rheumatol. 2012;39(5):916–28. https://doi.org/10.3899/jrheum.110961.

    Article  CAS  PubMed  Google Scholar 

  55. Ciregia F, Baiwir D, Cobraiville G, Dewael T, Mazzucchelli G, Badot V, et al. Glycosylation deficiency of lipopolysaccharide-binding protein and corticosteroid-binding globulin associated with activity and response to treatment for rheumatoid arthritis. J Transl Med. 2020;18(1):1–17. https://doi.org/10.1186/s12967-019-02188-9.

    Article  CAS  Google Scholar 

  56. Nordal HH, Brokstad KA, Solheim M, Halse A-K, Kvien TK, Hammer HB. 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(1):1–10. https://doi.org/10.1186/s13075-016-1201-0.

    Article  CAS  Google Scholar 

  57. Choi IY, Gerlag DM, Herenius MJ, Thurlings RM, Wijbrandts CA, Foell D, et al. MRP8/14 serum levels as a strong predictor of response to biological treatments in patients with rheumatoid arthritis. Ann Rheum Dis. 2015;74(3):499–505. https://doi.org/10.1136/annrheumdis-2013-203923.

    Article  CAS  PubMed  Google Scholar 

  58. Drynda S, Ringel B, Kekow M, Kühne C, Drynda A, Glocker MO, et al. Proteome analysis reveals disease-associated marker proteins to differentiate RA patients from other inflammatory joint diseases with the potential to monitor anti-TNFα therapy. Pathol Res Pract. 2004;200(2):165–71. https://doi.org/10.1016/j.prp.2004.02.011.

    Article  CAS  PubMed  Google Scholar 

  59. van Schaardenburg D, Murphy M, Gui Y, Turk S, Maksymowych WP, Marotta A, editors. Change in 14-3-3 eta expression in early RA patients treated with Dmards corresponds with change in DAS28 and good EULAR responses. Arthritis Rheumatol 2014.

  60. Alashkar DS, Elkhouly RM, Abd Elnaby AY, Nada DW. Will 14–3–3η be a new diagnostic and prognostic biomarker in rheumatoid arthritis? A prospective study of its utility in early diagnosis and response to treatment. Autoimmune Dis. 2022. https://doi.org/10.1155/2022/1497748.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Shovman O, Gilburd B, Watad A, Amital H, Langevitz P, Bragazzi N, et al. Decrease in 14–3-3η protein levels is correlated with improvement in disease activity in patients with rheumatoid arthritis treated with Tofacitinib. Pharmacol Res. 2019;141:623–6. https://doi.org/10.1016/j.phrs.2018.11.009.

    Article  CAS  PubMed  Google Scholar 

  62. Diaz-Torne C, Moya P, Hernandez MV, Reina D, Castellvi I, De Agustin JJ, et al. The combination of IL-6 and its soluble receptor is associated with the response of rheumatoid arthritis patients to tocilizumab. Semin Arthritis Rheum. 2018. https://doi.org/10.1016/j.semarthrit.2017.10.022.

    Article  PubMed  Google Scholar 

  63. Chen J, Tang M-S, Xu L-C, Li S, Ge Y, Du J-F, et al. Proteomic analysis of biomarkers predicting the response to triple therapy in patients with rheumatoid arthritis. Biomed Pharmacother. 2019;116:109026. https://doi.org/10.1016/j.biopha.2019.109026.

    Article  CAS  PubMed  Google Scholar 

  64. Fabre S, Dupuy A, Dossat N, Guisset C, Cohen J, Cristol J, et al. Protein biochip array technology for cytokine profiling predicts etanercept responsiveness in rheumatoid arthritis. Clin Exp Immunol. 2008;153(2):188–95. https://doi.org/10.1111/j.1365-2249.2008.03691.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Schotte H, Schluter B, Willeke P, Mickholz E, Schorat M, Domschke W, et al. Long-term treatment with etanercept significantly reduces the number of proinflammatory cytokine-secreting peripheral blood mononuclear cells in patients with rheumatoid arthritis. Rheumatology. 2004;43(8):960–4. https://doi.org/10.1093/rheumatology/keh178.

    Article  CAS  PubMed  Google Scholar 

  66. Dissanayake K, Jayasinghe C, Wanigasekara P, Sominanda A. Potential applicability of cytokines as biomarkers of disease activity in rheumatoid arthritis: enzyme-linked immunosorbent spot assay-based evaluation of TNF-α, IL-1β, IL-10 and IL-17A. PLoS ONE. 2021;16(1):e0246111. https://doi.org/10.1371/journal.pone.0246111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Chopin F, Garnero P, le Henanff A, Debiais F, Daragon A, Roux C, et al. Long-term effects of infliximab on bone and cartilage turnover markers in patients with rheumatoid arthritis. Ann Rheum Dis. 2008;67(3):353–7. https://doi.org/10.1136/ard.2007.076604.

    Article  CAS  PubMed  Google Scholar 

  68. Garnero P, Thompson E, Woodworth T, Smolen JS. 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(1):33–43. https://doi.org/10.1002/art.25053.

    Article  CAS  PubMed  Google Scholar 

  69. Karsdal MA, Schett G, Emery P, Harari O, Byrjalsen I, Kenwright A, et al., editors. IL-6 receptor inhibition positively modulates bone balance in rheumatoid arthritis patients with an inadequate response to anti-tumor necrosis factor therapy: biochemical marker analysis of bone metabolism in the tocilizumab RADIATE study (NCT00106522). Semin Arthritis Rheum 2012. 10.1016/j.semarthrit.2012.01.004

  70. Kawashiri S-Y, Endo Y, Nishino A, Okamoto M, Tsuji S, Takatani A, et al. Effect of abatacept treatment on serum osteoclast-related biomarkers in patients with rheumatoid arthritis (RA): a multicenter RA ultrasound prospective cohort in Japan. Medicine. 2021. https://doi.org/10.1097/MD.0000000000026592.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Kawashiri S-Y, Endo Y, Nishino A, Okamoto M, Tsuji S, Takatani A, et al. Association between serum bone biomarker levels and therapeutic response to abatacept in patients with rheumatoid arthritis (RA): a multicenter, prospective, and observational RA ultrasound cohort study in Japan. BMC Musculoskelet Disord. 2021;22(1):1–10. https://doi.org/10.1186/s12891-021-04392-5.

    Article  CAS  Google Scholar 

  72. Szeremeta A, Jura-Półtorak A, Koźma EM, Głowacki A, Kucharz EJ, Kopeć-Mędrek M, et al. Effects of a 15-month anti-TNF-α treatment on plasma levels of glycosaminoglycans in women with rheumatoid arthritis. Arthritis Res Ther. 2018;20(1):1–10. https://doi.org/10.1186/s13075-018-1711-z.

    Article  CAS  Google Scholar 

  73. Marotte H, Gineyts E, Miossec P, Delmas PD. Effects of infliximab therapy on biological markers of synovium activity and cartilage breakdown in patients with rheumatoid arthritis. Ann Rheum Dis. 2009;68(7):1197–200. https://doi.org/10.1136/ard.2008.096057.

    Article  CAS  PubMed  Google Scholar 

  74. Niki Y, Takeuchi T, Nakayama M, Nagasawa H, Kurasawa T, Yamada H, et al. Clinical significance of cartilage biomarkers for monitoring structural joint damage in rheumatoid arthritis patients treated with anti-TNF therapy. PLoS ONE. 2012;7(5):e37447. https://doi.org/10.1371/journal.pone.0037447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Boyapati A, Msihid J, Fiore S, van Adelsberg J, Graham NM, Hamilton JD. Sarilumab plus methotrexate suppresses circulating biomarkers of bone resorption and synovial damage in patients with rheumatoid arthritis and inadequate response to methotrexate: a biomarker study of MOBILITY. Arthritis Res Ther. 2016;18(1):1–12. https://doi.org/10.1186/s13075-016-1132-9.

    Article  CAS  Google Scholar 

  76. Bay-Jensen AC, Platt A, Byrjalsen I, Vergnoud P, Christiansen C, Karsdal MA. Effect of tocilizumab combined with methotrexate on circulating biomarkers of synovium, cartilage, and bone in the LITHE study. Semin Arthritis Rheum. 2014;43(4):470–8. https://doi.org/10.1016/j.semarthrit.2013.07.008.

    Article  CAS  PubMed  Google Scholar 

  77. Bay-Jensen AC, Platt A, Siebuhr AS, Christiansen C, Byrjalsen I, Karsdal MA. Early changes in blood-based joint tissue destruction biomarkers are predictive of response to tocilizumab in the LITHE study. Arthritis Res Ther. 2016;18(1):1–9. https://doi.org/10.1186/s13075-015-0913-x.

    Article  CAS  Google Scholar 

  78. Juhl P, Thudium CS, Gudmann NS, Karsdal MA, Bay-Jensen A-C, Siebuhr AS. IL-6 receptor inhibition modulates type III collagen and C-reactive protein degradation in rheumatoid arthritis patients with an inadequate response to anti-tumour necrosis factor therapy: analysis of connective tissue turnover in the tocilizumab RADIATE study. Clin Exp Rheumatol. 2018;36(4):568–74.

    PubMed  Google Scholar 

  79. Gudmann NS, Junker P, Juhl P, Thudium CS, Siebuhr AS, Byrjalsen I, et al. Type IV collagen metabolism is associated with disease activity, radiographic progression and response to tocilizumab in rheumatoid arthritis. Clin Exp Rheumatol. 2018;36(5):829–35.

    PubMed  Google Scholar 

  80. Gudmann NS, Hirata S, Karsdal MA, Kubo S, Bay-Jensen A-C, Tanaka Y. Increased remodelling of interstitial collagens and basement membrane is suppressed by treatment in patients with rheumatoid arthritis: serological evaluation of a one-year prospective study of 149 Japanese patients. Clin Exp Rheumatol. 2018;36(3):462–70.

    PubMed  Google Scholar 

  81. Thudium CS, Bay-Jensen AC, Cahya S, Dow ER, Karsdal MA, Koch AE, et al. The Janus kinase 1/2 inhibitor baricitinib reduces biomarkers of joint destruction in moderate to severe rheumatoid arthritis. Arthritis Res Ther. 2020;22(1):1–11. https://doi.org/10.1186/s13075-020-02340-7.

    Article  CAS  Google Scholar 

  82. Curtis JR, van der Helm-van Mil AH, Knevel R, Huizinga TW, Haney DJ, Shen Y, et al. Validation of a novel multibiomarker test to assess rheumatoid arthritis disease activity. Arthritis Care Res. 2012;64(12):1794–803. https://doi.org/10.1002/acr.21767.

    Article  Google Scholar 

  83. Centola M, Cavet G, Shen Y, Ramanujan S, Knowlton N, Swan KA, et al. Development of a multi-biomarker disease activity test for rheumatoid arthritis. PLoS ONE. 2013;8(4):e60635. https://doi.org/10.1371/journal.pone.0060635.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Curtis JR, Brahe CH, Østergaard M, Lund Hetland M, Hambardzumyan K, Saevarsdottir S, et al. Predicting risk for radiographic damage in rheumatoid arthritis: comparative analysis of the multi-biomarker disease activity score and conventional measures of disease activity in multiple studies. Curr Med Res Opin. 2019;35(9):1483–93. https://doi.org/10.1080/03007995.2019.1585064.

    Article  CAS  PubMed  Google Scholar 

  85. Curtis JR, Weinblatt ME, Shadick NA, Brahe CH, Østergaard M, Hetland ML, 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):1–13. https://doi.org/10.1186/s13075-020-02389-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Chara L, Sánchez-Atrio A, Pérez A, Cuende E, Albarrán F, Turrión A, et al. Monocyte populations as markers of response to adalimumab plus MTX in rheumatoid arthritis. Arthritis Res Ther. 2012;14(4):1–11. https://doi.org/10.1186/ar3928.

    Article  CAS  Google Scholar 

  87. Thurlings RM, Vos K, Wijbrandts CA, Zwinderman A, Gerlag DM, Tak P. Synovial tissue response to rituximab: mechanism of action and identification of biomarkers of response. Ann Rheum Dis. 2008;67(7):917–25. https://doi.org/10.1136/ard.2007.080960.

    Article  CAS  PubMed  Google Scholar 

  88. van den Bemt BJ, den Broeder AA, Wolbink GJ, van den Maas A, Hekster YA, van Riel PL, et al. The combined use of disease activity and infliximab serum trough concentrations for early prediction of (non-) response to infliximab in rheumatoid arthritis. Br J Clin Pharmacol. 2013;76(6):939–45. https://doi.org/10.1111/bcp.12142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Rosas J, Llinares-Tello F, de la Torre I, Santos-Ramírez C, Senabre-Gallego JM, Valor L, et al. Clinical relevance of monitoring serum levels of adalimumab in patients with rheumatoid arthritis in daily practice. Clin Exp Rheumatol. 2014;32(6):942–8.

    PubMed  Google Scholar 

  90. Jani M, Chinoy H, Warren RB, Griffiths CE, Plant D, Fu B, et al. Clinical utility of random anti-tumor necrosis factor drug-level testing and measurement of antidrug antibodies on the long-term treatment response in rheumatoid arthritis. Arthritis Rheumatol. 2015;67(8):2011–9. https://doi.org/10.1002/art.39169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Arad U, Elkayam O. Association of serum tocilizumab trough concentrations with clinical disease activity index scores in adult patients with rheumatoid arthritis. J Rheumatol. 2019;46(12):1577–81. https://doi.org/10.3899/jrheum.181431.

    Article  CAS  PubMed  Google Scholar 

  92. Jurado T, Plasencia-Rodríguez C, Martínez-Feito A, Navarro-Compán V, Rispens T, de Vries A, et al. Predictive value of serum infliximab levels at induction phase in rheumatoid arthritis patients. Open Rheumatol J. 2017;11:75. https://doi.org/10.2174/1874312901711010075.

    Article  CAS  PubMed Central  Google Scholar 

  93. Takeuchi T, Miyasaka N, Inoue K, Abe T, Koike T. Impact of trough serum level on radiographic and clinical response to infliximab plus methotrexate in patients with rheumatoid arthritis: results from the RISING study. Modern Rheumatol. 2009;19(5):478–87. https://doi.org/10.3109/s10165-009-0195-8.

    Article  CAS  Google Scholar 

  94. Jamnitski A, Krieckaert C, Nurmohamed M, Hart M, Dijkmans B, Aarden L, et al. Patients non-responding to etanercept obtain lower etanercept concentrations compared with responding patients. Ann Rheum Dis. 2012;71(1):88–91. https://doi.org/10.1136/annrheumdis-2011-200184.

    Article  CAS  PubMed  Google Scholar 

  95. Kneepkens E, van den Oever I, Plasencia C, Pascual-Salcedo D, De Vries A, Hart M, et al. Serum tocilizumab trough concentration can be used to monitor systemic IL-6 receptor blockade in patients with rheumatoid arthritis: a prospective observational cohort study. Scand J Rheumatol. 2017;46(2):87–94. https://doi.org/10.1080/03009742.2016.1183039.

    Article  CAS  PubMed  Google Scholar 

  96. Paramarta JE, Baeten DL. Adalimumab serum levels and antidrug antibodies towards adalimumab in peripheral spondyloarthritis: no association with clinical response to treatment or with disease relapse upon treatment discontinuation. Arthritis Res Ther. 2014;16(4):1–9. https://doi.org/10.1186/ar4675.

    Article  Google Scholar 

  97. van Herwaarden N, Bouman CA, van der Maas A, van Vollenhoven RF, Bijlsma JW, van den Hoogen FH, et al. Adalimumab and etanercept serum (anti) drug levels are not predictive for successful dose reduction or discontinuation in rheumatoid arthritis. Ann Rheum Dis. 2015;74(12):2260–1. https://doi.org/10.1136/annrheumdis-2015-207814.

    Article  CAS  PubMed  Google Scholar 

  98. Marotte H, Rinaudo M, Paul S, Fautrel B. No prediction of relapse by TNF blocker concentrations or detection of antibodies against anti-TNF: data from STRASS study. Ann Rheum Dis. 2016. https://doi.org/10.1136/annrheumdis-2016-eular.5823.

    Article  Google Scholar 

  99. Chen D-Y, Chen Y-M, Hsieh T-Y, Hung W-T, Hsieh C-W, Chen H-H, et al. Drug trough levels predict therapeutic responses to dose reduction of adalimumab for rheumatoid arthritis patients during 24 weeks of follow-up. Rheumatology. 2016;55(1):143–8. https://doi.org/10.1093/rheumatology/kev298.

    Article  CAS  PubMed  Google Scholar 

  100. Ruscitti P, Sinigaglia L, Cazzato M, Grembiale RD, Triolo G, Lubrano E, et al. Dose adjustments and discontinuation in TNF inhibitors treated patients: when and how. A systematic review of literature. Rheumatology. 2018;57(Supplement 7):vii23-vii31. https://doi.org/10.1093/rheumatology/key132

  101. Gunn GR, Sealey DC, Jamali F, Meibohm B, Ghosh S, Shankar G. From the bench to clinical practice: understanding the challenges and uncertainties in immunogenicity testing for biopharmaceuticals. Clin Exp Immunol. 2016;184(2):137–46. https://doi.org/10.1111/cei.12742.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Strand V, Goncalves J, Isaacs JD. Immunogenicity of biologic agents in rheumatology. Nat Rev Rheumatol. 2021;17(2):81–97.

    Article  PubMed  Google Scholar 

  103. Strand V, Balsa A, Al-Saleh J, Barile-Fabris L, Horiuchi T, Takeuchi T, et al. Immunogenicity of biologics in chronic inflammatory diseases: a systematic review. BioDrugs. 2017;31(4):299–316. https://doi.org/10.1007/s40259-017-0231-8

  104. Parikh CR, Ponnampalam JK, Seligmann G, Coelewij L, Pineda-Torra I, Jury EC, et al. Impact of immunogenicity on clinical efficacy and toxicity profile of biologic agents used for treatment of inflammatory arthritis in children compared to adults. Ther Adv Musculoskelet Dis. 2021. https://doi.org/10.1177/1759720X211002685.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Bartelds GM, Krieckaert CL, Nurmohamed MT, van Schouwenburg PA, Lems WF, Twisk JW, et al. Development of antidrug antibodies against adalimumab and association with disease activity and treatment failure during long-term follow-up. JAMA. 2011;305(14):1460–8. https://doi.org/10.1001/jama.2011.406.

    Article  CAS  PubMed  Google Scholar 

  106. Balsa A, Sanmarti R, Rosas J, Martin V, Cabez A, Gómez S, et al. Drug immunogenicity in patients with inflammatory arthritis and secondary failure to tumour necrosis factor inhibitor therapies: the REASON study. Rheumatology. 2018;57(4):688–93. https://doi.org/10.1093/rheumatology/kex474.

    Article  CAS  PubMed  Google Scholar 

  107. Wells AF, Parrino J, Mangan EK, Paccaly A, Lin Y, Xu C, et al. Immunogenicity of sarilumab monotherapy in patients with rheumatoid arthritis who were inadequate responders or intolerant to disease-modifying antirheumatic drugs. Rheumatol Ther. 2019;6(3):339–52. https://doi.org/10.1007/s40744-019-0157-3.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Moots RJ, Xavier RM, Mok CC, Rahman MU, Tsai W-C, Al-Maini MH, et al. The impact of anti-drug antibodies on drug concentrations and clinical outcomes in rheumatoid arthritis patients treated with adalimumab, etanercept, or infliximab: Results from a multinational, real-world clinical practice, non-interventional study. PLoS ONE. 2017;12(4):e0175207. https://doi.org/10.1371/journal.pone.0175207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Maid PJ, Xavier R, Real RM, Pedersen R, Shen Q, Marshall L, et al. Incidence of antidrug antibodies in rheumatoid arthritis patients from Argentina treated with adalimumab, etanercept, or infliximab in a real-world setting. JCR. 2018;24(4):177–82. https://doi.org/10.1097/RHU.0000000000000612.

    Article  PubMed  Google Scholar 

  110. Ducourau E, Mulleman D, Paintaud G, Miow Lin DC, Lauféron F, Ternant D, et al. Antibodies toward infliximab are associated with low infliximab concentration at treatment initiation and poor infliximab maintenance in rheumatic diseases. Arthritis Res Ther. 2011;13(3):1–7. https://doi.org/10.1186/ar3386.

    Article  CAS  Google Scholar 

  111. Siljehult F, Ärlestig L, Eriksson C, Rantapää-Dahlqvist S. Concentrations of infliximab and anti-drug antibodies in relation to clinical response in patients with rheumatoid arthritis. Scand J Rheumatol. 2018;47(5):345–50. https://doi.org/10.1080/03009742.2018.1433232.

    Article  CAS  PubMed  Google Scholar 

  112. Hambardzumyan K, Hermanrud C, Marits P, Vivar N, Ernestam S, Wallman J, et al. Association of female sex and positive rheumatoid factor with low serum infliximab and anti-drug antibodies, related to treatment failure in early rheumatoid arthritis: results from the SWEFOT trial population. Scand J Rheumatol. 2019;48(5):362–6. https://doi.org/10.1080/03009742.2019.1602670.

    Article  CAS  PubMed  Google Scholar 

  113. Xu C, Su Y, Paccaly A, Kanamaluru V. Population pharmacokinetics of sarilumab in patients with rheumatoid arthritis. Clin Pharmacokinet. 2019;58(11):1455–67. https://doi.org/10.1007/s40262-019-00765-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Hoxha A, Calligaro A, Tonello M, Ramonda R, Carletto A, Paolazzi G, et al. The clinical relevance of early anti-adalimumab antibodies detection in rheumatoid arthritis, ankylosing spondylitis and psoriatic arthritis: a prospective multicentre study. Joint Bone Spine. 2016;83(2):167–71. https://doi.org/10.1016/j.jbspin.2015.04.020.

    Article  CAS  PubMed  Google Scholar 

  115. Quistrebert J, Hässler S, Bachelet D, Mbogning C, Musters A, Tak PP, et al. Incidence and risk factors for adalimumab and infliximab anti-drug antibodies in rheumatoid arthritis: a European retrospective multicohort analysis. Semin Arthritis Rheum. 2019. https://doi.org/10.1016/j.semarthrit.2018.10.006.

    Article  PubMed  Google Scholar 

  116. Wolbink GJ, Vis M, Lems W, Voskuyl AE, De Groot E, Nurmohamed MT, et al. Development of antiinfliximab antibodies and relationship to clinical response in patients with rheumatoid arthritis. Arthritis Rheum. 2006;54(3):711–5. https://doi.org/10.1002/art.21671.

    Article  CAS  PubMed  Google Scholar 

  117. Doeleman MJ, Van Maarseveen EM, Swart JF. Immunogenicity of biologic agents in juvenile idiopathic arthritis: a systematic review and meta-analysis. Rheumatology. 2019;58(10):1839–49. https://doi.org/10.1093/rheumatology/kez030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Burmester GR, Choy E, Kivitz A, Ogata A, Bao M, Nomura A, et al. Low immunogenicity of tocilizumab in patients with rheumatoid arthritis. Ann Rheum Dis. 2017;76(6):1078–85. https://doi.org/10.1136/annrheumdis-2016-210297.

    Article  CAS  PubMed  Google Scholar 

  119. Sigaux J, Hamze M, Daien C, Morel J, Krzysiek R, Pallardy M, et al. Immunogenicity of tocilizumab in patients with rheumatoid arthritis. Joint Bone Spine. 2017;84(1):39–45. https://doi.org/10.1016/j.jbspin.2016.04.013.

    Article  CAS  PubMed  Google Scholar 

  120. Genovese MC, Fleischmann R, Kivitz AJ, Rell-Bakalarska M, Martincova R, Fiore S, et al. Sarilumab plus methotrexate in patients with active rheumatoid arthritis and inadequate response to methotrexate: results of a phase III study. Arthritis Rheumatol. 2015;67(6):1424–37. https://doi.org/10.1002/art.39093.

    Article  CAS  PubMed  Google Scholar 

  121. Thurlings RM, Teng O, Vos K, Gerlag DM, Aarden L, Stapel SO, et al. Clinical response, pharmacokinetics, development of human anti-chimaeric antibodies, and synovial tissue response to rituximab treatment in patients with rheumatoid arthritis. Ann Rheum Dis. 2010;69(2):409–12. https://doi.org/10.1136/ard.2009.109041.

    Article  CAS  PubMed  Google Scholar 

  122. den Broeder AA, van Herwaarden N, van den Bemt BJ. Therapeutic drug monitoring of biologicals in rheumatoid arthritis: a disconnect between beliefs and facts. Curr Opin Rheumatol. 2018;30(3):266–75. https://doi.org/10.1097/BOR.0000000000000487.

    Article  CAS  Google Scholar 

  123. Harrold LR, Litman HJ, Connolly SE, Alemao E, Kelly S, Rebello S, et al. Comparative effectiveness of abatacept versus tumor necrosis factor inhibitors in patients with rheumatoid arthritis who are anti-CCP positive in the United States Corrona Registry. Rheumatol Ther. 2019;6(2):217–30. https://doi.org/10.1007/s40744-019-0149-3.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Jamnitski A, Bartelds GM, Nurmohamed MT, van Schouwenburg PA, van Schaardenburg D, Stapel SO, et al. The presence or absence of antibodies to infliximab or adalimumab determines the outcome of switching to etanercept. Ann Rheum Dis. 2011;70(2):284–8. https://doi.org/10.1136/ard.2010.135111.

    Article  CAS  PubMed  Google Scholar 

  125. Ulijn E, den Broeder N, Wientjes M, van Herwaarden N, Meek I, Tweehuysen L, et al. Therapeutic drug monitoring of adalimumab in RA: no predictive value of adalimumab serum levels and anti-adalimumab antibodies for prediction of response to the next bDMARD. Ann Rheum Dis. 2020;79(7):867–73. https://doi.org/10.1136/annrheumdis-2020-216996.

    Article  CAS  PubMed  Google Scholar 

  126. Kerschbaumer A, Sepriano A, Smolen JS, van der Heijde D, Dougados M, van Vollenhoven R, et al. Efficacy of pharmacological treatment in rheumatoid arthritis: a systematic literature research informing the 2019 update of the EULAR recommendations for management of rheumatoid arthritis. Ann Rheum Dis. 2020;79(6):744–59. https://doi.org/10.1136/annrheumdis-2019-216656.

    Article  CAS  PubMed  Google Scholar 

  127. O’Mahony R, Richards A, Deighton C, Scott D. Withdrawal of disease-modifying antirheumatic drugs in patients with rheumatoid arthritis: a systematic review and meta-analysis. Ann Rheum Dis. 2010;69(10):1823–6. https://doi.org/10.1136/ard.2008.105577.

    Article  CAS  PubMed  Google Scholar 

  128. Verstappen M, van Mulligen E, de Jong P, van der Helm-Van MA. DMARD-free remission as novel treatment target in rheumatoid arthritis: a systematic literature review of achievability and sustainability. RMD Open. 2020;6(1):e001220. https://doi.org/10.1136/rmdopen-2020-001220.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Tweehuysen L, van den Ende CH, Beeren FM, Been EM, van den Hoogen FH, den Broeder AA. Little evidence for usefulness of biomarkers for predicting successful dose reduction or discontinuation of a biologic agent in rheumatoid arthritis: a systematic review. Arthritis Rheum. 2017;69(2):301–8. https://doi.org/10.1002/art.39946.

    Article  Google Scholar 

  130. van der Woude D, Young A, Jayakumar K, Mertens BJ, Toes RE, van der Heijde D, 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(8):2262–71. https://doi.org/10.1002/art.24661.

    Article  PubMed  Google Scholar 

  131. Valor L, Garrido J, Martínez-Estupiñán L, Hernández-Flórez D, Janta I, López-Longo FJ, et al. Identifying markers of sustained remission in rheumatoid arthritis patients on long-term tapered biological disease-modifying antirheumatic drugs. Rheumatol Int. 2018;38(8):1465–70. https://doi.org/10.1007/s00296-018-4087-0.

    Article  CAS  PubMed  Google Scholar 

  132. Haschka J, Englbrecht M, Hueber AJ, Manger B, Kleyer A, Reiser M, et al. Relapse rates in patients with rheumatoid arthritis in stable remission tapering or stopping antirheumatic therapy: interim results from the prospective randomised controlled RETRO study. Arthritis Res Ther. 2016;75(1):45–51. https://doi.org/10.1136/annrheumdis-2014-206439.

    Article  Google Scholar 

  133. Rech J, Hueber AJ, Finzel S, Englbrecht M, Haschka J, Manger B, et al. Prediction of disease relapses by multibiomarker disease activity and autoantibody status in patients with rheumatoid arthritis on tapering DMARD treatment. Arthritis Res Ther. 2016;75(9):1637–44. https://doi.org/10.1136/annrheumdis-2015-207900.

    Article  CAS  Google Scholar 

  134. Figueiredo CP, Bang H, Cobra JF, Englbrecht M, Hueber AJ, Haschka J, et al. Antimodified protein antibody response pattern influences the risk for disease relapse in patients with rheumatoid arthritis tapering disease modifying antirheumatic drugs. Arthritis Res Ther. 2017;76(2):399–407. https://doi.org/10.1136/annrheumdis-2016-209297.

    Article  CAS  Google Scholar 

  135. Heimans L, Akdemir G, Boer KV, Goekoop-Ruiterman YP, Molenaar ET, van Groenendael JH, 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(1):1–11. https://doi.org/10.1186/s13075-015-0912-y.

    Article  CAS  Google Scholar 

  136. Tanaka Y, Oba K, Koike T, Miyasaka N, Mimori T, Takeuchi T, et al. Sustained discontinuation of infliximab with a raising-dose strategy after obtaining remission in patients with rheumatoid arthritis: the RRRR study, a randomised controlled trial. Ann Rheum Dis. 2020;79(1):94–102. https://doi.org/10.1136/annrheumdis-2019-216169.

    Article  CAS  PubMed  Google Scholar 

  137. Baker KF, Skelton AJ, Lendrem DW, Scadeng A, Thompson B, Pratt AG, et al. Predicting drug-free remission in rheumatoid arthritis: a prospective interventional cohort study. J Autoimmun. 2019;105:102298. https://doi.org/10.1016/j.jaut.2019.06.009.

    Article  PubMed  PubMed Central  Google Scholar 

  138. de Moel EC, Rech J, Mahler M, Roth J, Vogl T, Schouffoer A, 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(1):1–8. https://doi.org/10.1186/s13075-019-2064-y.

    Article  CAS  Google Scholar 

  139. Bechman K, Tweehuysen L, Garrood T, Scott DL, Cope AP, Galloway JB, et al. Flares in rheumatoid arthritis patients with low disease activity: predictability and association with worse clinical outcomes. J Rheumatol. 2018;45(11):1515–21. https://doi.org/10.3899/jrheum.171375.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. GhitiMoghadam M, Lamers-Karnebeek FB, Vonkeman HE, Ten Klooster PM, Tekstra J, Schilder AM, et al. Multi-biomarker disease activity score as a predictor of disease relapse in patients with rheumatoid arthritis stopping TNF inhibitor treatment. PLoS ONE. 2018;13(5):e0192425. https://doi.org/10.1371/journal.pone.0192425.

    Article  CAS  Google Scholar 

  141. Kossi S, Sasso EH, Marotte H, Liu X, Tubach F, Hajage D, et al. Ability of the multi-biomarker disease activity score to identify rheumatoid arthritis patients in remission at risk of relapse after TNF-blocker tapering. An ancillary study of the STRASS trial. BMJ Publishing Group Ltd; 2019. p. 1430. https://doi.org/10.1136/annrheumdis-2019-eular.5988.

  142. Bouman CA, van der Maas A, van Herwaarden N, Sasso EH, van den Hoogen FH, den Broeder AA. A multi-biomarker score measuring disease activity in rheumatoid arthritis patients tapering adalimumab or etanercept: predictive value for clinical and radiographic outcomes. Rheumatology. 2017;56(6):973–80. https://doi.org/10.1093/rheumatology/kex003

  143. Hagen M, Englbrecht M, Haschka J, Reiser M, Kleyer A, Hueber A, et al. Cost-effective tapering algorithm in patients with rheumatoid arthritis: combination of multibiomarker disease activity score and autoantibody status. J Rheumatol. 2019;46(5):460–6. https://doi.org/10.3899/jrheum.180028.

    Article  CAS  PubMed  Google Scholar 

  144. Tanaka Y, Smolen JS, Jones H, Szumski A, Marshall L, Emery P. The effect of deep or sustained remission on maintenance of remission after dose reduction or withdrawal of etanercept in patients with rheumatoid arthritis. Arthritis Res Ther. 2019;21(1):1–16. https://doi.org/10.1186/s13075-019-1937-4.

    Article  CAS  Google Scholar 

  145. Farutin V, Prod’homme T, McConnell K, Washburn N, Halvey P, Etzel CJ, et al. Molecular profiling of rheumatoid arthritis patients reveals an association between innate and adaptive cell populations and response to anti-tumor necrosis factor. Arthritis Res Ther. 2019;21(1):1–14. https://doi.org/10.1186/s13075-019-1999-3.

    Article  CAS  Google Scholar 

  146. Smith SL, Plant D, Eyre S, Hyrich K, Morgan AW, Wilson AG, et al. The predictive value of serum S100A9 and response to etanercept is not confirmed in a large UK rheumatoid arthritis cohort. Rheumatology. 2017;56(6):1019–24. https://doi.org/10.1093/rheumatology/kew387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Petrosius V, Schoof EM. Recent advances in the field of single-cell proteomics. Transl Oncol. 2023;27:101556. https://doi.org/10.1016/j.tranon.2022.101556.

    Article  CAS  PubMed  Google Scholar 

  148. Freites-Núñez D, Baillet A, Rodriguez-Rodriguez L, Nguyen MVC, Gonzalez I, Pablos JL, et al. Efficacy, safety and cost-effectiveness of a web-based platform delivering the results of a biomarker-based predictive model of biotherapy response for rheumatoid arthritis patients: a protocol for a randomized multicenter single-blind active controlled clinical trial (PREDIRA). Trials. 2020;21(1):1–15. https://doi.org/10.1186/s13063-020-04683-7.

    Article  Google Scholar 

  149. Bartelds GM, Wijbrandts CA, Nurmohamed MT, Stapel S, Lems WF, Aarden L, et al. Anti-infliximab and anti-adalimumab antibodies in relation to response to adalimumab in infliximab switchers and anti-tumour necrosis factor naive patients: a cohort study. Ann Rheum Dis. 2010;69(5):817–21. https://doi.org/10.1136/ard.2009.112847.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Supported by the Ministry of Science and Technology, Taiwan [MOST-110-2320-B-008-001-MY2].

Author information

Authors and Affiliations

Authors

Contributions

S-FC collected and performed the literature review, and was the major contributor in writing the manuscript. F-CY and H-YC selected the clinically relevant topic and provided real-world experience from the clinical and proteomic aspect. F-CY, C-YC, and H-YC helped revise and finalize the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Hui-Yin Chang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1

. A pdf file including Fig. S1. Association between the pretreatment levels of serum proteins and clinical responses of RA treatments.

Additional file 2.

An excel file including Table S1. Search strategy. Table S2. Studies associated with autoantibodies (References of Fig. 2). Table S3. Studies associated with serum proteins other than autoantibodies (References of Fig. 3b and Fig. S1). Table S4. References of Fig. 4. Table S5. Serum proteins and their full names.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, SF., Yeh, FC., Chen, CY. et al. Tailored therapeutic decision of rheumatoid arthritis using proteomic strategies: how to start and when to stop?. Clin Proteom 20, 22 (2023). https://doi.org/10.1186/s12014-023-09411-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12014-023-09411-2

Keywords