Abstracting and Indexing

  • PubMed NLM
  • Google Scholar
  • Semantic Scholar
  • Scilit
  • CrossRef
  • WorldCat
  • ResearchGate
  • Academic Keys
  • DRJI
  • Microsoft Academic
  • Academia.edu
  • OpenAIRE
  • Scribd
  • Baidu Scholar

Factors Underlying Failure of Methotrexate Treatment in Rheumatoid Arthritis: Implications in Personalized Care

Ananta Srivastava1, Stefanie Au1, Sumanjali Reddy Kanmantha Reddy1, Emmanuel Katsaros2, Devendra K. Agrawal1*

1Departments of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA

2Internal Medicine, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences,

Pomona, California 91766 USA

*Corresponding author: Devendra K. Agrawal, MSc, PhD (Biochem), PhD (Med Sci), MBA, MS (ITM), FAAAAI, FAHA, FAPS, FIACS, Professor and Director, Department of Translational Research, Western University of Health Sciences 309 E. Second Street, Pomona, California 91766, USA

Received: 14 April 2025; Accepted: 21 April 2025; Published: 22 April 2025

Article Information

Citation: Ananta Srivastava, Stefanie Au, Sumanjali Reddy Kanmantha Reddy, Emmanuel Katsaros, Devendra K. Agrawal. Factors Underlying Failure of Methotrexate Treatment in Rheumatoid Arthritis: Implications in Personalized Care. Archives of Internal Medicine Research. 8 (2025): 121-131.

DOI: 10.26502/aimr.0203

View / Download Pdf Share at Facebook

Abstract

Rheumatoid arthritis (RA) is a chronic inflammatory disease that can be managed with a range of therapeutic treatments. Methotrexate (MTX) is a first-line treatment for RA; however, its metabolism in RA patients can be complicated by multiple factors. Therefore, understanding these specific factors is crucial for optimizing the efficacy of MTX to provide improved therapeutic outcomes for patients. This article explores existing literature to examine how MTX metabolism in RA patients is impacted by other commonly used medications for RA. Additionally, the review explores the role of genetics by investigating the impact that single nucleotide polymorphisms (SNPs) have on MTX metabolism. Key findings from this review highlight how MTX metabolism can be enhanced or impaired based on specific combination therapies and how alternative treatments are considered with MTX treatment failure. MTX metabolism can also vary across different racial, ethnic, and population-based groups due to the presence of distinct SNPs in their genetic profiles. These results underscore the importance of personalized treatment approaches when treating RA patients with MTX, as its metabolism is influenced by factors such as drug interactions and SNPs. Future research is needed to expand our understanding of these factors to further improve therapeutic outcomes in RA patients.

Keywords

Autoantibodies; Autoimmune disease; B-cell depletion therapy; Corticosteroid; Disease-modifying antirheumatic drugs; Genetic polymorphism; IL-6 antagonist; Inflammation; JAK inhibitor; Methotrexate; Methotrexate metabolism; NSAIDs; Rheumatoid arthritis; SNPs; TNF inhibitor.

Autoantibodies articles; Autoimmune disease articles; B-cell depletion therapy articles; Corticosteroid articles; Disease-modifying antirheumatic drugs articles; Genetic polymorphism articles; IL-6 antagonist articles; Inflammation articles; JAK inhibitor articles; Methotrexate articles; Methotrexate metabolism articles; NSAIDs articles; Rheumatoid arthritis articles; SNPs articles; TNF inhibitor articles.

Article Details

Introduction

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by the immune system attacking the lining of the joint [1,2]. The hallmark symptoms of RA include symmetrical joint pain, particularly in the hands and feet, along with joint stiffness in the morning that typically lasts more than 30 minutes [1,3]. RA may manifest in other organs, such as the lungs, heart, and eyes while presenting with pericarditis, pleural effusion, or interstitial lung disease [1,3]. As there is no single diagnostic method that is considered the gold standard for RA, its diagnosis relies on clinical evaluation and is supported by the presentation of symptoms, serologic tests, and imaging findings [1,3,4]. Additionally, key laboratory tests for RA include rheumatoid factor and anti-cyclic citrullinated antibodies [1,3,5,6]. However, despite the presence of disease, one in three individuals may still test negative for these markers (seronegative) [3].

Certain risk factors can increase the likelihood of developing RA. Some examples include smoking, obesity, a family history of the disease, and genetics [3,4]. RA is also debilitating, with 80% of patients who are not properly treated having misaligned joints and 40% being unable to continue working within a decade from the manifestation of the disease [7]. Therefore, early and effective treatment is crucial. While there have been advances in the diagnosis and management of RA, challenges still exist with achieving an early diagnosis and timely referral of patients to the appropriate specialists [4].

One of the primary treatments used for RA is methotrexate (MTX), a disease-modifying antirheumatic drug (DMARD). MTX alters the activity of dihydrofolate reductase, a part of the folate metabolism pathway, resulting in the inhibition of DNA synthesis and purine metabolism [7,8]. It is usually administered as a weekly dose and is often given as an initial monotherapy for patients with moderate to severe RA [1,3]. MTX is not fully effective as a treatment, as only an estimated 40%-50% of RA patients achieve disease remission or have reduction in disease activity [1,9]. While the medication possesses potent anti-inflammatory and anti-proliferative properties, the individual treatment responses can differ due to a myriad of underlying factors [10].

One retrospective study in a tertiary care hospital in Pakistan identified that RA patients who had an increased body mass index (BMI), diabetes, being female, and smoking correlated with increased risk of MTX treatment failure [11]. Another crucial factor that can influence responsiveness to MTX in individuals is genetic polymorphisms [10]. Identifying variations in gene polymorphisms can aid as a tool for understanding the effectiveness and progression of MTX treatment in RA patients [10,12]. Previous studies have identified 120 single nucleotide polymorphisms (SNPs), a type of genetic polymorphism, that may potentially impact MTX treatment response in RA patients [13]. The review critically examined the current literature on the existing therapeutic options and their connections to treatment with MTX for RA patients, SNPs that influence MTX metabolism in these patients, and explored potential variations in gene polymorphisms across different racial, ethnic, and population-based groups.

Method

To select research findings for this article on MTX metabolism in RA patients, with a focus on current therapeutics, SNPs, and potential polymorphisms prevalent in different groups, a comprehensive search was conducted using databases such as PubMed and Google Scholar from 2005 - 2025. Key search terms included: "MTX metabolism," "rheumatoid arthritis," "SNP," "DMARD," "corticosteroid," "conventional synthetic DMARD," "TNF inhibitor," "IL-6 antagonist," "B-cell depletion therapy," "JAK inhibitors," and "NSAID." Articles excluded from consideration were those not written in English or those available only as abstracts.

Prevalence and Incidence

The global prevalence of RA varies, with higher rates reported in industrialized countries, likely influenced by environmental exposures, genetic predispositions, and demographic differences [14,71] (Figure 1). In the United States (U.S.), RA affects approximately 1.3 million adults, representing 0.6% to 1% of the population [15,16]. As previously mentioned, common risk factors include both non-modifiable factors, such as genetics and sex, and modifiable factors, such as smoking and obesity [3,4,15]. The disease imposes significant societal and individual burdens, with approximately 35% of patients experiencing work disability. Indirect costs, driven by lost work productivity, are estimated to be nearly three times greater than direct treatment costs. In the U.S. alone, the total annual healthcare expenditure for RA is approximately $19.3 billion [15,17].

fortune-biomass-feedstock

Figure 1: Incidence Rates of RA. This table demonstrates the global incidence of rheumatoid arthritis and a breakdown of the rates per 10,000 in different continents.

The prevalence of RA may have increased in recent years, with emerging evidence suggesting that infections, particularly COVID-19, may influence the incidence and exacerbation of autoimmune diseases like RA. A large population-based study conducted from 2018 to 2023 found that individuals who developed COVID-19 had twice the incidence rate of RA-related diagnoses compared to those without COVID-19. This aligns with earlier research highlighting infections as environmental triggers for autoimmune processes. While these findings emphasize a potential link, further longitudinal studies are needed to explore contributing factors, such as the effects of vaccination, other environmental exposures, and long-term outcomes in RA development after COVID-19 [18].

Pathophysiology of Rheumatoid Arthritis

The autoimmune response in RA is driven by several inflammatory pathways, with Nuclear Factor Kappa-Β (NF-κB) playing a central role in regulating transcription factors that amplify inflammation. Activated by pro-inflammatory cytokines such as IL-1, TNFα, and IL-6, NF-κB promotes joint inflammation, bone erosion, and disease progression. Another critical pathway involves the Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL), which drives osteoclast differentiation and bone destruction, contributing to the osteoclastogenesis and bone degradation by activated NF-κB [19]. The persistent production of these cytokines disrupts the balance of bone remodeling, favoring resorption and leading to progressive joint damage and systemic bone loss.

Joint swelling in RA results from synovial inflammation driven by immune activation and leukocyte infiltration into the synovial compartment. This complex interplay involves both innate and adaptive immune responses, with dendritic cells (DCs), monocytes, mast cells, innate lymphoid cells, T-helper cells, B cells, and plasma cells playing significant roles. Environmental and genetic factors can provoke DCs, which activate T cells and stimulate B cells, macrophages, synoviocytes, and osteoclasts. These cells release inflammatory mediators such as IL-1β, IL-6, TNF-α, and matrix metalloproteinases (MMPs), perpetuating chronic inflammation and driving tissue remodeling. The integration of these immune pathways in the synovium and adjacent bone marrow perpetuates inflammation, tissue damage, and leukocyte migration into affected joints [20].

The synovial tissue undergoes profound structural changes that contribute to joint destruction in RA. The hyperplastic synovial lining layer, composed of synovial fibroblasts (SFs) and macrophages, forms an invasive pannus that erodes cartilage and subchondral bone. The sub-lining layer, infiltrated with immune cells and enriched with memory CD45RO+ T cells, further supports inflammation. These T cells, drawn by chemokines and sustained by survival signals like IL-7 and IL-15, create a persistent inflammatory environment. SFs in RA adopt an aggressive, tumor-like phenotype, acting similarly to T and B cells in producing matrix-degrading enzymes, such as MMPs and cathepsins, that degrade cartilage and secrete RANKL [21].

Each of these mechanisms illustrates the complex interplay of immune cells, cytokines, and autoantibodies in driving inflammation and bone erosion in RA (Figure 2). This aggressive remodeling and immune cell interaction underpin the chronic and self-sustaining nature disease progression as well as explains the many different therapies required to tackle different inflammatory processes. Important markers and inflammatory mediators such as IL-1β, IL-6, IL-7, IL-15, RANKL, and TNF-α are important when it comes to therapeutic measures, personalized medicine, and are key in understanding the complexities of RA.

fortune-biomass-feedstock

Figure 2: Inflammatory Pathways Contributing to the Pathophysiology of Rheumatoid Arthritis. This figure demonstrates the various mechanisms and inflammatory pathways contributing to the development of rheumatoid arthritis.

Methotrexate and Current Therapies in Rheumatoid Arthritis

(i) Further Insights into Methotrexate (MTX)

Treating RA patients with MTX can be challenging because there are varying degrees of response depending on the individual. To determine how patients may respond to MTX, one study evaluated 312 patients to assess clinical factors, such as age and levels of markers for inflammation. Their findings indicated that poorer response to MTX was associated with patients who had a younger age of disease onset, were rheumatoid factor positive, and had elevated levels of erythrocyte sedimentation rate [22]. Additionally, to improve the bioavailability of the drug, one study explored the use of microneedle patches for treatment, which is beneficial because it provides a localized response and reduces invasiveness. After treating RA guinea pigs with the microneedle patches, they found that there were reduced levels of key RA-associated diagnostic markers, less swelling and bone erosion, and minimal long-term side effects [23]. In all, MTX is a first-line therapy and cornerstone treatment for RA, but its effectiveness and bioavailability can differ based on the administration route. Similarly, treatment outcomes can vary due to influencing clinical factors. Given these limitations, MTX may not be sufficient as a monotherapy for all patients. Therefore, it is essential to explore additional therapies that may modify or enhance the effectiveness of MTX and assess how these treatments can impact the overall outcomes for RA patients.

(ii) Conventional Synthetic Disease Modifying Antirheumatic Drugs (DMARDs)

Conventional synthetic DMARDs include sulfasalazine, hydroxychloroquine, and leflunomide. Sulfasalazine is a pro-drug, metabolized into sulphapyridine and 5-aminosalicylic. It has an unclear mechanism but demonstrates anti-inflammatory and immunoregulatory properties [7,24]. Hydroxychloroquine is an antimalarial that possesses anti-inflammatory properties by suppression of autoreactive T cells [7,25]. Leflunomide works by arresting nucleotide synthesis, leading to inhibition of T lymphocyte proliferation and ultimately reducing immune reactions [26].

For the treatment of RA, there are many possible combinations when using conventional synthetic DMARDs. One common therapy is a triple combination of MTX, sulfasalazine, and hydroxychloroquine, while another possibility is a double therapy of MTX and leflunomide. In a study comparing the rates at which patients adhered to therapy, they concluded that while there were longer retention rates of therapy use with the triple combination compared to the double, the findings were not statistically significant [27].

When evaluating how MTX may interact with other DMARDs, one study looked at the incidence rate of serious adverse events with the combination of MTX + leflunomide versus MTX + sulfasalazine. They found that there was a better safety profile with MTX + leflunomide compared to MTX + sulfasalazine [28]. Yet, when directly comparing the toxicity and efficacy differences in monotherapy of leflunomide to MTX + leflunomide, there was no statistically significant difference [26]. Another study also revealed that there was no difference between the safety profiles of MTX + leflunomide and MTX+ hydroxychloroquine in RA patients who had inadequate response to MTX alone; however, the median time to remission was quicker with MTX + hydroxychloroquine, taking 11 months, compared to 16 months with MTX + leflunomide [29].

To further evaluate the adverse effects of these therapies, especially in RA patients with increased cardiovascular (CV) risk, one study compared hydroxychloroquine and MTX. In patients without pre-existing CV risk, both medications had similar rates of sudden cardiac arrest, ventricular arrhythmia, and major adverse cardiovascular events [30]. However, in patients with a history of heart failure who started hydroxychloroquine, there was an increased risk of CV-related adverse events. Additionally, regardless of prior heart failure history, initiating hydroxychloroquine was linked to a higher risk of hospitalization for heart failure [30].

Conventional synthetic DMARDs can be used in combination with MTX to reduce inflammation. However, when determining suitable treatment for a patient, these studies showed it is crucial to account for factors such as medication adherence, safety profile, duration of use, and the patient’s past medical history.

(iii) Biologic and Targeted DMARDs

Biologic DMARDs (bDMARDs) are another available therapy for RA patients. BDMARDs, infliximab and adalimumab, have been shown to have enhanced efficacy when used with MTX [31]. In addition, the combination of bDMARD + MTX led to a 24% decreased risk of RA patients having cardiovascular disease events [31].

Another study compared the monotherapy of bDMARD etanercept, a TNF inhibitor, to the monotherapy of MTX to assess whether it was more effective in maintaining remission rates. 49.5% of patients in the etanercept group-maintained remission compared to 28.7% of patients in the MTX group (p-value = 0.006) [32].

Likewise, tocilizumab, an IL-6 antagonist, is a bDMARD that can be used to treat RA as monotherapy or with MTX. IL-6 is a cytokine that can inhibit T-reg cells and impair the body’s ability to differentiate between self and non-self while also promoting inflammation [33,34]. Antagonizing IL-6 can be beneficial for treating RA patients who have not responded to previous standard treatments [34]. One study found that the combination of tocilizumab + MTX was more impactful in preventing the worsening of radiographic progression of joint damage compared to tocilizumab monotherapy [35]. However, there was no difference noted in the effectiveness of treating patients with early RA who had more joint damage and those with prolonged durations of RA [24].

Rituximab is also used for treating RA when standard therapeutics, such as MTX, are not adequate for controlling the disease [36]. This drug is an anti-CD20 monoclonal antibody that promotes B cell depletion by inducing cell death, and thus, modulating the immune system [37,38]. When comparing RA patients who maintained the standard dose versus switching to a low-dose regimen of rituximab once they demonstrated improvement in their condition, the low-dose regimen led to less chance of relapse and serious adverse events [36].

Another class of drugs that can treat RA are targeted DMARDs, such as JAK inhibitors tofacitinib and baricitinib. Their mechanism of action is to block the signaling of cytokines that promote inflammation [39]. These drugs can be used as monotherapy or with MTX. A study showed that there was remarked improvement in RA patients who were treated with JAK inhibitors after failure with MTX and other bDMARDs [39]. When directly comparing the safety profile and effectiveness of baricitinib and tofacitinib used in combination with MTX, they were similar. However, when it came to pain relief, baricitinib was more effective [40]. Additionally, another study showed that monotherapy baricitinib, when compared to MTX, showed better rates of remission of RA—62% and 48%, respectively (p-value = 0.03) [41]. Patients using baricitinib were also able to achieve remission quicker and had less radiographic progression in seropositive RA patients [41].

Finally, when comparing the first-line therapies of etanercept, adalimumab, and JAK inhibitors in RA patients, one study analyzed patient data from the CorEvitas RA Registry and demonstrated that there were no significant differences in the effectiveness of monotherapy or combination therapy with any of these treatments [42].

Although MTX is the most used first-line therapy for RA, bDMARDs and targeted DMARDs can be used concomitantly and can be effective in those who had an inadequate response to MTX. In some cases, bDMARDs and targeted DMARDs also may have a better efficacy and safety profile than MTX. These studies highlight the complexity of RA as a disease and importance of providing individualized treatment to achieve disease remission.

(iv) Non-steroidal anti-inflammatory drugs

Non-steroidal anti-inflammatory drugs (NSAIDs), such as ibuprofen and naproxen, can be used to reduce acute inflammation in RA by the inhibition of cyclooxygenase (COX) [43]. Non-selective NSAIDs can cause adverse side effects, such as gastrointestinal ulceration and renal injury [43,44]. Therefore, the administration of COX-2 selective inhibitors, like celecoxib, can mitigate some of these effects [43]. As NSAIDs are used intermittently to help with RA flare-ups, it is important to understand their interaction with standard medications, such as MTX. One study noted that low-dose MTX (less than 30 mg) was primarily safe to use alongside NSAIDs as it did not pose a significant risk of interacting or diminishing effectiveness [45]. Yet, when MTX is used alongside aspirin, the combination can increase toxicity levels in the liver and kidney dysfunction [45,46].

Altogether, these studies reinforce the value of having a multi-pronged approach for treating RA patients. While NSAIDS may not be the primary treatment for RA, they can be used to alleviate acute episodes of inflammation and provide therapeutic support alongside MTX.

(v) Glucocorticoids

Glucocorticoids (GCs) are used intermittently for acute flare-ups of RA, as they possess anti-inflammatory properties but are limited in use because of causing systemic toxicity, like increasing risk of osteoporosis and hyperglycemia [47,48]. Although GCs are used for short-term treatment in RA, the CareRA trial demonstrated their long-term benefits when used in the initial treatment with MTX of early RA patients, who had a favorable prognosis. This subpopulation had lower long-term utilization of NSAIDs and other analgesics compared to patients treated with MTX alone [49].

Short-term GC use can be beneficial for controlling sudden exacerbations of inflammation and reduce the future use of additional therapeutic support when used with MTX. Again, these findings emphasize the need for providing tailored treatments to maximize the therapeutic efficacy while minimizing risks.

Table 1: Mechanism of Actions of Drugs in the Treatment of Rheumatoid Arthritis. This table highlights many of the therapeutics discussed for RA treatment, focusing specifically on an overview of the relevant mechanism of action.

Drug

Mechanism of Action

methotrexate

Affects the folate metabolism pathway via inhibition of dihydrofolate reductase to disrupt DNA synthesis and purine metabolism

sulfasalazine

Unclear mechanism of action; pro-drug metabolized into sulfapyridine and 5- aminosalicylic

hydroxychloroquine

Suppression of autoreactive T cells

leflunomide

Inhibition of T lymphocyte proliferation via arresting nucleotide synthesis

etanercept

TNF inhibition

tocilizumab

IL-6 antagonist

rituximab

Anti-CD20 monoclonal antibody that induces cell death to to promote B cell depletion

tofacitinib/baricitinib

Inhibition of janus kinases (JAK) to block signaling of cytokines that induce inflammation

non-steroid anti-inflammatory drugs (NSAIDs)

Reduction of acute inflammation via inhibition of cyclooxygenase (COX)

Single Nucleotide Polymorphisms (SNPs) Impacting MTX Metabolism in RA Patients

When evaluating clinical studies published within the past five years exploring the impact of SNPs on MTX metabolism in RA patients, certain SNPs were associated with treatment failure and increased toxicity of the drug or its metabolite, MTX-7-OH, while others were correlated with increased tolerance or showed no significant correlation (Table 2).

When assessing the plasma of RA patients, one study concluded that the SNP rs4149081 (SLCO1B1 gene) was associated with higher concentrations of MTX and MTX-7-OH (p < 0.05) [50]. In the same study, SNP rs1476413 (MTHFR gene) TT or CT alleles were correlated with increased levels of MTX metabolites and severity of disease symptoms (p < 0.001) [50]. Similarly, another study identified the correlation of SNP rs1476413 (MTHFR gene) with MTX monotherapy failure after one to two years of treatment, alongside SNPs rs246240 (ABCC1 gene), rs2259571 (AIF-1 gene), and rs2274808 (SLC19A1 gene) [51]. Other SNPs that were associated with inadequate response to MTX include rs3821353 (ATIC gene) and rs7279445 (SLC19A1 gene) [52]. By contrast, the SNP rs10072026 (DHFR) was correlated with adequate response to MTX [52]. Likewise, a separate study identified SNPs with contrasting effects on MTX tolerance, with GGH-T401C (rs11545078) associated with increased intolerance and ABCC2-C24T (rs717620) with increased tolerance [53]. In another study, they found that out of the 20 patients who were unresponsive to MTX, 19 patients had SNP C3435T (ABCB1 gene) (p <0.001) [54].

SNPs may also play a role in influencing adverse drug effects. A study identified SNPs specifically on the SLCO1A2 gene of RA patients treated with MTX monotherapy for a minimum

Table 2: Impact of Single Nucleotide Polymorphisms (SNPs) on MTX Metabolism. This table summarizes the SNPs and their relationship with MTX metabolism.

SNPs

Impact of MTX Metabolism

rs4149081 (SLCO1B1 gene)

Associated with higher concentrations of MTX and MTX-7-OH

rs1476413 (MTHFR gene)

Associated with increased MTX metabolite levels and severity of disease symptoms and with MTX monotherapy failure post-treatment for one to two years

rs246240 (ABCC1 gene)

Associated with MTX monotherapy failure post-treatment for one to two years

rs2259571 (AIF-1 gene)

Associated with MTX monotherapy failure post-treatment for one to two years

rs2274808 (SLC19A1 gene)

Associated with MTX monotherapy failure post-treatment for one to two years

rs3821353 (ATIC gene)

Inadequate treatment response to MTX

rs7279445 (SLC19A1 gene)

Inadequate treatment response to MTX

s10072026 (DHFR gene)

Adequate treatment response to MTX

GGH-T401C/ rs11545078

Increased intolerance to MTX treatment

ABCC2-C24T/ rs717620

Increased tolerance to MTX treatment

C3435T (ABCB1 gene)

Unresponsive to MTX treatment

G550A (SLCO1A2 gene)

Increased risk of MTX adverse drug effects

1994A > G (FPGS gene)

Increased risk of MTX adverse drug effects

C677T/ rs1801133 (MTHFR gene)

Not associated with impacting MTX metabolism

A1298C/rs180113 (MTHFR gene)

Not associated with impacting MTX metabolism

of six months. The G550A polymorphism was correlated with increased risk of adverse drug effects with MTX [55]. Another study identified that increased adverse effects were associated with the SNP 1994A > G (FPGS gene) in RA patients who had been treated with MTX for 24 weeks [56].

Finally, genes such as MTHFR, which have SNPs known to influence MTX metabolism, may also contain variants that do not impact metabolism. For instance, one study evaluating SNPs in early RA patients on MTX monotherapy found that C677T (rs1801133) and A1298C (rs180113) on the MTHFR gene were not associated with changes in MTX metabolism [57].

Overall, these findings highlight the role that SNPs have in influencing MTX rates of metabolism, treatment efficacy, and adverse effects. It also underscores the complexity and the potential that the identification of genetic markers, like SNPs, can play when determining the best individualized treatment for RA patients.

SNPs Impacting MTX Metabolism in Different Racial, Ethnic, and Population-Based RA Patient Groups

Identifying SNPs that influence MTX metabolism in various racial, ethnic, and population-based groups is crucial, as genetic differences can provide valuable insights into how patients respond to therapy.

Regarding race-specific research related to SNP and MTX metabolism, one study found that ABCC2 IVS 23+56 T → C SNP was associated with MTX toxicity-related dose adjustments or drug discontinuation in Caucasians with RA but not in African Americans [58]. This intronic SNP may serve as a genetic marker of MTX toxicity or occur in linkage disequilibrium with a variant that determines toxicity in Caucasian populations. Likewise, another study that examined genetic and population-based differences in MTX efficacy, toxicity, and related allelic variations among RA patients of African American and Caucasian populations that the 1298A allele in Caucasians was associated with a significant increase in MTX-related adverse events, whereas in African Americans, the rs4846051C allele and its haplotypes were associated with MTX toxicity [59]. Yet, other factors that may play a role in MTX efficacy and metabolism are those of lifestyle, such as differences in folate status [60,61], socioeconomic factors, diet, lifestyle, and education [62]. These factors likely contribute to population-specific variability in MTX adverse events, independent of SNPs or genetics [62]. For example, a survey on RA patients found that African American patients exhibited higher risk aversion related to MTX toxicity (adjusted OR 8.4, 95% CI 3.1–23.1), emphasizing risks over benefits compared to white patients [63]. Additionally, studies often lack large and representative African American sample populations, limiting their ability to draw definitive conclusions [62]. Enhanced risk communication, aversion, and education as well as emphasizing the expected benefits could reduce disparities in treatment acceptance, and eventually, patient outcomes.

Other studies have focused on findings related to the impact of SNPs on MTX metabolism within the context of ethnicity and population-based groups. In contrast to the association of ABCC2 IVS 23+56 T → C SNP in Caucasians previously mentioned, this SNP had higher allele frequencies in Japanese populations [64]. In another study conducted in Malaysia on 647 RA patients, none of the SNPs were significantly associated with MTX efficacy or toxicity based on the Chi-square test and logistic regression [65]. However, stratification by ancestry revealed that among patients of Malay descent, two SNPs—ATIC C347G (rs2372536) (OR 0.5478, 95% CI 0.3396–0.8835, p = 0.01321) and ATIC T675C (rs4673993) (OR 0.5247, 95% CI 0.3248–0.8478, p = 0.008111)—were significantly associated with an adequate MTX response (p <0.05), but no SNPs were linked to MTX toxicity [65]. A study conducted on Iraqi patients in 2024 found that gene polymorphisms of the TYMS gene: TCrs2853741 and ACrs260641, predict non-responsiveness while the CTAT genotype predicts a successful MTX response [66]. The same authors later concluded in a separate study of Iraqi patients that TTrs1801131 genotypes were associated with unresponsiveness, and the G-carried allele for rs1801131 has an association with response to MTX [67]. Looking at Egyptian adult RA patients treated with MTX, a study identified MTHFR A1298 CC as the primary SNP likely to predict both MTX toxicity and efficacy [68]. Finally, a 2017 meta-analysis on European RA patients concluded the significant association between the toxicity of MTX and the RFC-1 80G > A (rs1051266) polymorphism [69]. These various studies demonstrate that there can be significant associations between SNP and MTX metabolism in different ethnic and population-based groups.

Despite some insights, there remains no clear consensus on how MTX metabolism varies across many racial, ethnic, and population-based groups and how SNPs may contribute, as most past research focuses primarily on comparisons specifically between African American and Caucasian RA patients. Knowing these genetic differences allows for tailored therapy, enabling patients to respond more effectively to treatment and minimizing adverse outcomes [70]. To bridge existing gaps, future studies must prioritize larger, diverse cohorts and focus on integrating genetic, environmental, and socioeconomic variables into treatment strategies, ensuring equitable care for all patients with RA.

Conclusion

RA is a complex, systemic autoimmune disease that utilizes MTX, a DMARD, as the first-line treatment. However, its efficacy can vary significantly between patients due to multiple influences. Understanding how MTX interacts with other medications commonly used in treatment with RA can provide insight into personalizing therapies to optimize clinical outcomes. Similarly, SNPs can impact the response to MTX treatment by affecting its drug metabolism and may be associated with adverse drug effects. Due to the presence of SNPs, tailoring treatment to individuals based on their genetic profile, including racial, ethnic, and population-based background, is valuable in providing personalized care for the management of RA. Future research into pharmacogenomics and identification of genetic markers associated with MTX metabolism can further our understanding of RA pathogenicity. Advancements in our knowledge of genetic polymorphisms on drug efficacy can enhance the effectiveness of RA treatment and, therefore, improve therapeutic outcomes.

Key points, outstanding questions, and challenges

  • Methotrexate (MTX) metabolism in rheumatoid arthritis (RA) patients is influenced by combination therapies and genetic polymorphisms (SNPs), impacting treatment efficacy and toxicity.
  • Further research is essential to better understand the complex interplay of drug interactions, genetic factors (SNPs), and demographic variables (race, ethnicity, socioeconomic factors) in MTX metabolism to improve therapeutic outcomes for all RA patients.
  • Are there specific biomarkers (beyond genetic markers) that can predict how a patient's MTX metabolism will be affected by combination therapies, allowing for more proactive and personalized treatment decisions?
  • How do gene-environment interactions (e.g., the interplay between specific SNPs and factors like diet, smoking, or folate levels) influence MTX metabolism and treatment outcomes in RA patients?
  • Can population-specific pharmacokinetic and pharmacodynamic studies of MTX be conducted to establish more tailored dosing guidelines for different ethnic groups?
  • Currently, there is not adequate literature to cover neither the epigenetics of many ethnic groups nor the socioeconomic factors involved in RA. Additional studies are warranted in these areas.

Funding:

The research work of DKA is supported by the R25AI179582 and R01 HL147662 grants from the National Institutes of Health, USA. The contents of this article are solely the responsibility of the authors and do not necessarily represent the official views of the National Institutes of Health.

Competing interests:

All authors have read the manuscript and declare no conflict of interest. No writing assistance was utilized in the production of this manuscript.

Consent for publication:

All authors have read the manuscript and consented for publication.

References

  1. Smith MH, Berman JR. What Is Rheumatoid Arthritis? JAMA 327 (2022):1194-1194.
  2. Gao Y, Zhang Y, Liu X. Rheumatoid arthritis: pathogenesis and therapeutic advances. MedComm 5 (2024): e509.
  3. Sparks JA. Rheumatoid Arthritis. Annals of Internal Medicine 170 (2019): ITC1.
  4. Cush JJ. Rheumatoid Arthritis: Early Diagnosis and Treatment. Rheum Dis Clin North Am 48 (2022): 537-547.
  5. Van Delft MAM, Huizinga TWJ. An overview of autoantibodies in rheumatoid arthritis. Journal of Autoimmunity 110 (2020): 102392.
  6. Sokolova MV, Schett G, Steffen U. Autoantibodies in Rheumatoid Arthritis: Historical Background and Novel Findings. Clin Rev Allergy Immunol 63 (2022): 138-151.
  7. Lin YJ, Anzaghe M, Schülke S. Update on the Pathomechanism, Diagnosis, and Treatment Options for Rheumatoid Arthritis. Cells 9 (2020): 880.
  8. Nayak RR, Alexander M, Deshpande I, et al. Methotrexate impacts conserved pathways in diverse human gut bacteria leading to decreased host immune activation. Cell Host & Microbe 29 (2021): 362-377.e11.
  9. Chatzidionysiou K, Sfikakis PP. Low rates of remission with methotrexate monotherapy in rheumatoid arthritis: review of randomised controlled trials could point towards a paradigm shift. RMD Open 5 (2019): e000993.
  10. Zhao X, Wu P, Yang Z, et al. Relationship between the efficacy and adverse effects of methotrexate and gene polymorphism. Egyptian Journal of Medical Human Genetics 25 (2024).
  11. Siddiqui A, Totonchian A, Jabar Ali JB, et al. Risk Factors Associated With Non-Respondence to Methotrexate in Rheumatoid Arthritis Patients. Cureus. Published online September 20 (2021).
  12. Lim AJW, Lim LJ, Ooi BNS, et al. Functional coding haplotypes and machine-learning feature elimination identifies predictors of Methotrexate Response in Rheumatoid Arthritis patients. eBioMedicine 75 (2022): 103800.
  13. Ceballos FC, Chamizo-Carmona E, Mata-Martín C, et al. Pharmacogenetic Sex-Specific Effects of Methotrexate Response in Patients with Rheumatoid Arthritis. Pharmaceutics 15 (2023): 1661.
  14. Finckh A, Gilbert B, Hodkinson B, et al. Global epidemiology of rheumatoid arthritis. Nature Reviews Rheumatology 18 (2022).
  15. Xu Y, Wu Q. Prevalence Trend and Disparities in Rheumatoid Arthritis among US Adults, 2005–2018. Journal of Clinical Medicine 10 (2021): 3289.
  16. Jang S, Kwon EJ, Lee JJ. Rheumatoid Arthritis: Pathogenic Roles of Diverse Immune Cells. IJMS 23 (2022): 905.
  17. Chen CI, Wang L, Wei W, et al. Burden of rheumatoid arthritis among US Medicare population: co-morbidities, health-care resource utilization and costs. Rheumatol Adv Pract 2 (2018): rky005.
  18. Marín JS, Mazenett-Granados EA, Salazar-Uribe JC, et al. Increased incidence of rheumatoid arthritis after COVID-19. Autoimmunity Reviews 22 (2023):103409.
  19. Ilchovska D (Daisy), Barrow DM. An Overview of the NF-kB mechanism of pathophysiology in rheumatoid arthritis, investigation of the NF-kB ligand RANKL and related nutritional interventions. Autoimmunity Reviews 20 (2021): 102741.
  20. Ding Q, Hu W, Wang R, et al. Signaling pathways in rheumatoid arthritis: implications for targeted therapy. Signal Transduction and Targeted Therapy 8 (2023): 1-24.
  21. Araki Y, Mimura T. The Mechanisms Underlying Chronic Inflammation in Rheumatoid Arthritis from the Perspective of the Epigenetic Landscape. Journal of Immunology Research (2016): 1-10.
  22. Edyta Majorczyk, Małgorzata Mazurek-Mochol, 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 11 (2022): 6078-6078.
  23. Rajendran K, Pahal S, Badnikar K, et al. Methotrexate delivering microneedle patches for improved therapeutic efficacy in treatment of rheumatoid arthritis. International Journal of Pharmaceutics 642 (2023): 123184.
  24. Zhao C, Yu Y, Yin G, et al. Sulfasalazine promotes ferroptosis through AKT-ERK1/2 and P53-SLC7A11 in rheumatoid arthritis. Inflammopharmacol 32 (2024): 1277-1294.
  25. Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat Rev Rheumatol 16 (2020): 155-166.
  26. Guadagnin DA, Mazzali LV, Skare TL, et al. Treating rheumatoid arthritis with leflunomide monotherapy versus combination therapy with methotrexate. European Journal of Rheumatology 8 (2021): 12-15.
  27. Bhavsar SV, Movahedi M, Cesta A, et al. Retention of triple therapy with methotrexate, sulfasalazine, and hydroxychloroquine compared to combination methotrexate and leflunomide in rheumatoid arthritis. Joint Bone Spine 91 (2024): 105732.
  28. Bredemeier M, Ranza R, Kakehasi AM, et al. Safety of the Methotrexate–leflunomide Combination in Rheumatoid Arthritis: Results of a Multicentric, Registry-based, Cohort Study (BiobadaBrasil). The Journal of Rheumatology 48 (2021): 1519-1527.
  29. Zhang L, Chen F, Geng S, et al. Methotrexate (MTX) Plus Hydroxychloroquine versus MTX Plus Leflunomide in Patients with MTX-Resistant Active Rheumatoid Arthritis: A 2-Year Cohort Study in Real World. JIR 13 (2020): 1141-1150.
  30. D’Andrea E, Desai R, He M, et al. Cardiovascular Risks of Hydroxychloroquine vs Methotrexate in Patients With Rheumatoid Arthritis. Journal of the American College of Cardiology 80 (2022): 36-46.
  31. Xie F, Chen L, Yun H, et al. Benefits of Methotrexate Use on Cardiovascular Disease Risk Among Rheumatoid Arthritis Patients Initiating Biologic Disease-modifying Antirheumatic Drugs. The Journal of Rheumatology 48 (2020): 804-812.
  32. Curtis JR, Emery P, Karis E, et al. Etanercept or Methotrexate Withdrawal in Rheumatoid Arthritis Patients in Sustained Remission on Combination Therapy: A Randomized, Double-Blind Trial. Arthritis & Rheumatology. Published online November 18 (2020).
  33. Chatzidionysiou K, Circiumaru A, Rethi B, et al. Tocilizumab decreases T cells but not macrophages in the synovium of patients with rheumatoid arthritis while it increases the levels of serum interleukin-6 and RANKL. RMD Open 7 (2021): e001662.
  34. Pandolfi F, Franza L, Carusi V, et al. Interleukin-6 in Rheumatoid Arthritis. International Journal of Molecular Sciences 21 (2020): 5238.
  35. Verhoeven MMA, Janneke Tekstra, Johannes, et al. Efficacy of Tocilizumab Monotherapy Versus Tocilizumab and Methotrexate Combination Therapy in the Prevention of Radiographic Progression in Rheumatoid Arthritis: An Analysis Using Individual Patient Data From Multiple Clinical Trials. Arthritis Care and Research 74 (2022): 889-895.
  36. Bertsias A, Avgoustidis N, Papalopoulos I, et al. Rheumatoid arthritis patients initiating rituximab with low number of previous bDMARDs failures may effectively reduce rituximab dose and experience fewer serious adverse events than patients on full dose: a 5-year cohort study. Arthritis Res Ther 24 (2022): 132.
  37. Kim JW, Jung JY, Shin K, et al. Factors Determining Retreatment Time Interval of Rituximab in Korean Patients With Rheumatoid Arthritis. Front Med 8 (2021): 765535.
  38. Hanif N, Anwer F. Rituximab. PubMed. Published 2021. https://www.ncbi.nlm.nih.gov/books/NBK564374/
  39. Fitton J, Melville AR, Emery P, et al. Real-world single centre use of JAK inhibitors across the rheumatoid arthritis pathway. Rheumatology (Oxford, England) 60 (2021): 4048-4054.
  40. Liao X, Huo W, Zeng W, et al. Efficacy and safety of different Janus kinase inhibitors combined with methotrexate for the treatment of rheumatoid arthritis: a single-center randomized trial. Advances in Rheumatology 63 (2023).
  41. Baset MA, Rumi MA, Karim M. Comparative Efficacy of Methotrexate and Baricitinib as First-Line Therapy in Seropositive Rheumatoid Arthritis; A Clinical Study. Asia Pacific Journal of Medical Innovations 1 (2024): 14-23.
  42. Pappas DA, O’Brien J, Guo L, et al. Treatment patterns and clinical outcomes in patients with rheumatoid arthritis initiating etanercept, adalimumab, or Janus kinase inhibitor as first-line therapy: results from the real-world CorEvitas RA Registry. Arthritis Res Ther 25 (2023): 166.
  43. Radu AF, Bungau SG. Management of Rheumatoid Arthritis: an Overview. Cells 10 (2021): 2857.
  44. Akram M, Daniyal M, Sultana S, et al. Traditional and modern management strategies for rheumatoid arthritis. Clinica Chimica Acta 512 (2021): 142-155.
  45. Honma T, Onda K, Koichi Masuyama. Drug-drug interaction assessment based on a large-scale spontaneous reporting system for hepato- and renal-toxicity, and thrombocytopenia with concomitant low-dose methotrexate and analgesics use. BMC Pharmacology and Toxicology 25 (2024).
  46. Colebatch AN, Marks JL, Van Der Heijde DM, et al. Safety of Nonsteroidal Antiinflammatory Drugs and/or Paracetamol in People Receiving Methotrexate for Inflammatory Arthritis: A Cochrane Systematic Review. The Journal of Rheumatology Supplement 90 (2012): 62-73.
  47. Metselaar JM, Middelink LM, Wortel CH, et al. Intravenous pegylated liposomal prednisolone outperforms intramuscular methylprednisolone in treating rheumatoid arthritis flares: A randomized controlled clinical trial. Journal of Controlled Release 341 (2022): 548-554.
  48. Crowson LP, Davis JM, Hanson AC, et al. Time Trends in Glucocorticoid Use in Rheumatoid Arthritis During the Biologics Era: 1999-2018. Semin Arthritis Rheum 61 (2023): 152219.
  49. Pazmino S, Boonen A, Cock DD, et al. Short-term glucocorticoids reduce risk of chronic NSAID and analgesic use in early methotrexate-treated rheumatoid arthritis patients with favourable prognosis: subanalysis of the CareRA randomised controlled trial. RMD Open 7 (2021): e001615.
  50. D’Cruz LG, McEleney KG, Tan KBC, et al. Clinical and Laboratory Associations with Methotrexate Metabolism Gene Polymorphisms in Rheumatoid Arthritis. Journal of Personalized Medicine 10 (2020): 149.
  51. Kolan SS, Li G, Grimolizzi F, et al. Identification of SNPs associated with methotrexate treatment outcomes in patients with early rheumatoid arthritis. Frontiers in Pharmacology 13 (2022).
  52. Abdallah HY, Ibrahim ME, Noha, et al. Pharmacogenomics of Methotrexate Pathway in Rheumatoid Arthritis Patients: Approach toward Personalized Medicine. Diagnostics 12 (2022): 1560-1560.
  53. Escudero-Contreras A, López-Medina C, Collantes-Estévez E, et al. Genetic Polymorphisms of GGH and ABCC2 Are Associated with Methotrexate Intolerance in Patients with Rheumatoid Arthritis. Journal of Clinical Medicine 10 (2021): 4070.
  54. Melikoglu MA, Balkan E. Can we predict unresponsiveness to methotrexate in rheumatoid arthritis? A pharmacogenetic study. Inflammopharmacol 30 (2022): 193-197.
  55. Wang J, Yin J, Li W, et al. Association between SLCO1A2 genetic variation and methotrexate toxicity in human rheumatoid arthritis treatment. J Biochem & Molecular Tox. 34 (2020): e22513.
  56. Sandhu A, Dhir V, Ahmad S, et al. Clinico-genetic model to predict methotrexate intolerance in rheumatoid arthritis. Clin Rheumatol 39 (2020): 201-206.
  57. Zhang Q, Fu P, Cao Z, et al. MTHFR and MTRR Genetic Polymorphism of Methotrexate Therapy Outcomes in Early Rheumatoid Arthritis. PGPM 16 (2023): 407-423.
  58. Ranganathan, Prabha, Robert Culverhouse, et al. “Methotrexate (MTX) Pathway Gene Polymorphisms and Their Effects on MTX Toxicity in Caucasian and African American Patients with Rheumatoid Arthritis.” The Journal of Rheumatology 35 (2008): 572–579.
  59. Hughes LB, Beasley TM, Patel H, et al. Racial or ethnic differences in allele frequencies of single-nucleotide polymorphisms in the methylenetetrahydrofolate reductase gene and their influence on response to methotrexate in rheumatoid arthritis. Annals of the Rheumatic Diseases 65 (2006): 1213-1218.
  60. Perry CA, Renna SA, Khitun E, et al. Ethnicity and race influence the folate status response to controlled folate intakes in young women. J Nutrition 134 (2004): 1786-1792.
  61. Barry Shane, Faith Pangilinan, James L Mills, et al, The 677C→T variant of MTHFR is the major genetic modifier of biomarkers of folate status in a young, healthy Irish population, The American Journal of Clinical Nutrition, Volume 108, Issue 6, 2018, Pages 1334-1341.
  62. Pharmacogenomics of Methotrexate in Rheumatoid Arthritis: Does Race Make a Difference? Accessed January 5, 2025. https://www.jrheum.org/content/jrheum/35/4/550.full.pdf
  63. Constantinescu F, Goucher S, Weinstein A, et al. Understanding why rheumatoid arthritis patient treatment preferences differ by race. Arthritis & Rheumatism 61 (2009): 413-418.
  64. Hashiguchi M, Shimizu M, Jun Hakamata, et al. Genetic polymorphisms of enzyme proteins and transporters related to methotrexate response and pharmacokinetics in a Japanese population. Journal of Pharmaceutical Health Care and Sciences 2 (2016).
  65. Sha HX, Veerapen K, Chow SK, et al. Genetic variations in methotrexate metabolic pathway genes influence methotrexate responses in rheumatoid arthritis patients in Malaysia. Scientific Reports 12 (2022).
  66. Mutlak, Qassim Mahdi PhDa,*; Kasim, Ali Abdulhussain PhDb; et al. Impact of TYMS gene polymorphism on the outcome of methotrexate treatment in a sample of Iraqi rheumatoid arthritis patients – identification of novel single nucleotide polymorphism: Cross-sectional study. Medicine 103 (2024): p e38448.
  67. Mutlak, Q.M., Kasim, A.A. Impact of MTHFR gene polymorphism on the outcome of methotrexate treatment in a sample of Iraqi rheumatoid arthritis patients. Sci Rep 14 (2024): 15119.
  68. Sharaki OA, Elgerby AH, Nassar ES, et al. Impact of methylenetetrahydrofolate reductase (MTHFR) A1298C gene polymorphism on the outcome of methotrexate treatment in a sample of Egyptian rheumatoid arthritis patients. Alexandria Journal of Medicine 54 (2018): 633-638.
  69. Qiu, Qi PhDa; Huang, Jing BSb,c; et al. Polymorphisms and pharmacogenomics for the toxicity of methotrexate monotherapy in patients with rheumatoid arthritis: A systematic review and meta-analysis. Medicine 96 (2017): p e6337.
  70. Lima A, Bernardes M, Azevedo R, et al. Moving toward personalized medicine in rheumatoid arthritis: SNPs in methotrexate intracellular pathways are associated with methotrexate therapeutic outcome. Pharmacogenomics 17 (2016): 1649-1674.
  71. Yu F, Chen H, Li Q. et al. Secular trend of mortality and incidence of rheumatoid arthritis in global ,1990–2019: an age period cohort analysis and joinpoint analysis. BMC Pulm Med 23 (2023): 356.

Journal Statistics

Impact Factor: * 3.6

Acceptance Rate: 78.21%

Time to first decision: 10.4 days

Time from article received to acceptance: 2-3 weeks

Discover More: Recent Articles

Grant Support Articles

© 2016-2025, Copyrights Fortune Journals. All Rights Reserved!