CORRELATION BETWEEN METHOTREXATE PLASMA CONCENTRATION AND CLINICAL EFFICACY IN RHEUMATOID ARTHRITIS: A 6-MONTH PROSPECTIVE STUDY
HTML Full TextCORRELATION BETWEEN METHOTREXATE PLASMA CONCENTRATION AND CLINICAL EFFICACY IN RHEUMATOID ARTHRITIS: A 6-MONTH PROSPECTIVE STUDY
Namrata Soni * and Sunil Kumar Mathur
Department of Pharmacology, J. L. N. Medical College, Ajmer, Rajasthan, India.
ABSTRACT: Background: Methotrexate (MTX) is the first-line disease-modifying anti-rheumatic drug (DMARD) for rheumatoid arthritis (RA), with therapeutic efficacy often attributed to optimal systemic exposure. However, interindividual variability in MTX plasma concentration can influence treatment response. Objective: To evaluate the correlation between plasma MTX concentrations and clinical efficacy over a 6-month period in patients with RA, using ESR, CRP, and pain scores as markers of therapeutic response. Methods: A prospective observational study was conducted on 72 newly diagnosed RA patients receiving oral MTX (15 mg/week). Plasma samples were collected at 2 hours post-dose to estimate MTX concentration using validated HPLC. Disease activity was assessed at baseline and after 6 months using ESR, CRP, and visual analogue scale (VAS) pain scores. Pearson’s correlation was used to assess associations between MTX levels and clinical parameters. Results: The mean MTX plasma concentration at 6 months was 1.29 ± 0.39 µmol/L. Reductions in mean ESR (from 34.4 to 19.9 mm/hr), CRP (from 13.2 to 6.0 mg/L), and pain score (from 6.53 to 1.98) were noted. However, weak and statistically non-significant correlations were found between MTX concentration and ESR (r = +0.16, p = 0.17), CRP (r = -0.21, p = 0.07), and pain score (r = -0.08, p = 0.47). Conclusion: While MTX therapy led to significant clinical improvement in RA patients over 6 months, plasma concentration alone did not strongly predict therapeutic response. Intracellular MTX metabolites and other pharmacodynamic factors may play a more dominant role, indicating the need for more comprehensive monitoring strategies.
Keywords: Methotrexate, Rheumatoid arthritis, Plasma concentration, Clinical efficacy, Inflammatory markers, Therapeutic drug monitoring
INTRODUCTION: Rheumatoid arthritis (RA) is a chronic, immune-mediated inflammatory disorder that primarily targets synovial joints 3. It leads to progressive cartilage degradation, bone erosion, and joint deformity, ultimately resulting in significant impairment of physical function and quality of life.
The disease is systemic in nature and is associated with extra-articular manifestations and an elevated risk of cardiovascular, pulmonary, and metabolic comorbidities. Affecting approximately 0.5% to 1% of the global population, RA is more prevalent among women and typically presents during middle age.
If left untreated, it imposes a considerable clinical and socioeconomic burden due to long-term disability, reduced work productivity, and the need for lifelong pharmacotherapy 2. Methotrexate (MTX), a structural analog of folic acid, has long been established as the first-line disease-modifying anti-rheumatic drug (DMARD) in the management of RA. It is a conventional synthetic agent (csDMARD) with well-documented efficacy in suppressing inflammation, limiting radiological progression, and preserving joints function 3. MTX is generally administered once weekly, either orally or parenterally, with doses ranging from 7.5 mg to 25 mg, tailored according to disease activity and individual tolerance. Its relatively favorable risk-benefit profile and cost-effectiveness have made it the cornerstone of RA therapy, often used as monotherapy or in combination with other agents including biologics or targeted synthetic DMARDs 4.
The mechanism of action of MTX in RA extends beyond its classical antifolate properties. It inhibits dihydrofolate reductase and thymidylate synthase, interfering with DNA synthesis and cellular replication. More importantly in RA, MTX enhances extracellular adenosine signaling, which has broad anti-inflammatory effects 5. Adenosine modulates neutrophil activity, downregulates proinflammatory cytokines such as TNF-α and IL-6, and promotes a shift toward anti-inflammatory macrophage phenotypes 6. In addition, MTX influences T-cell apoptosis, adhesion molecule expression, and oxidative stress pathways. These pleiotropic mechanisms collectively contribute to its disease-modifying effects 7.
Despite its established place in RA treatment algorithms, clinical response to MTX is known to be highly variable. Some patients exhibit rapid symptom relief and disease control, while others demonstrate partial or minimal benefit even after adequate dosing 8. Such inter-individual differences in efficacy may stem from genetic, metabolic, and immunological variability, but a major contributing factor is pharmacokinetic diversity 9. The extent of MTX absorption, distribution, metabolism, and excretion varies widely among patients and can influence both therapeutic outcomes and adverse event profiles.
Oral MTX displays dose-dependent and saturable absorption in the gastrointestinal tract, with bioavailability ranging between 30% and 70%. After administration, peak plasma concentration (Cmax) is typically achieved within 1 to 2 hours 10. MTX binds moderately to plasma proteins and distributes into third spaces, including synovial fluid. It is minimally metabolized in the liver to 7-hydroxymethotrexate and is primarily excreted unchanged by the kidneys via glomerular filtration and active tubular secretion. Renal function, hydration status, age, and concurrent medications significantly influence MTX clearance 11.
Given this variability, the measurement of plasma MTX concentrations, especially Cmax, has been explored as a potential marker for therapeutic monitoring. A higher Cmax may reflect adequate systemic exposure and correlate with better disease control, while subtherapeutic levels could contribute to treatment failure 12. However, the relationship between MTX pharmacokinetics and clinical efficacy remains incompletely understood. Although some studies suggest a positive correlation between MTX levels and disease activity reduction, others have reported weak or inconsistent associations. Additionally, plasma levels may not fully reflect the drug’s intracellular activity, where MTX is converted to active polyglutamated metabolites that exert prolonged immunosuppressive effects 13.
From a clinical standpoint, inflammation in RA is routinely assessed using biomarkers such as erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP), both of which reflect systemic inflammatory burden 14. These parameters are commonly used to evaluate treatment response and guide therapeutic decisions. Similarly, subjective measures such as visual analogue scale (VAS) for pain provide insight into patient-perceived symptom relief. Whether MTX plasma concentrations are predictive of improvements in these markers is still under investigation 15.
This study was designed to address this gap in evidence by prospectively evaluating the correlation between MTX plasma concentrations and treatment response in patients with RA over a six-month period. By analyzing ESR, CRP, and VAS pain scores in relation to MTX Cmax measured at steady state, we aimed to determine whether plasma drug levels could serve as a surrogate marker for clinical efficacy 16. The study also intended to explore the feasibility of integrating therapeutic drug monitoring (TDM) into routine RA management, especially in resource-limited settings where advanced immunological assays may not be available. Understanding this correlation is crucial in optimizing MTX therapy. If a strong relationship between plasma levels and clinical response can be established, it could pave the way for individualized dosing regimens, early identification of non-responders, and more precise prediction of therapeutic outcomes. On the other hand, if plasma MTX concentrations fail to correlate with clinical improvement, it would reinforce the importance of intracellular pharmacokinetics and pharmaco-genomics in governing MTX’s immunomodulatory actions.
MATERIALS AND METHODS:
Study Design and Participants: This was a prospective, observational study conducted at J.L.N. Medical College, Ajmer. Seventy-two adult patients newly diagnosed with RA (per ACR/EULAR 2010 criteria) 17 and new to DMARD therapy were enrolled. Ethical approval was obtained from the institutional review board.
Inclusion Criteria:
- Diagnosed with RA based on ACR/EULAR criteria.
- Willing to initiate oral MTX therapy (15 mg/week)
- With or without comorbid T2DM (diagnosed by HbA1c >6.5%).
Exclusion Criteria:
- Use of corticosteroids or biologics.
- GI, hepatic, renal, or autoimmune comorbidities.
- Pregnancy, lactation, HIV, or refusal to consent.
MTX Administration and Sampling: Patients received oral MTX once weekly after a baseline assessment. Venous blood samples were drawn 2 hours post-dose after 6 months of consistent therapy, assuming steady-state kinetics. Plasma was separated and stored in cryotubes at -20°C until analysis.
HPLC Analysis: MTX plasma levels were quantified using YL9100 HPLC system with a UV detector at 302 nm. The mobile phase consisted of 45% acetonitrile and 55% water, flow rate 1.0 mL/min, with a C18 column. Method validation was performed for accuracy, precision, linearity, and recovery.
Treatment and Follow-up: All patients received oral MTX at a fixed weekly dose of 15 mg. No concomitant steroids or biologics were used. Patients were followed up for 6 months, with clinical and laboratory evaluations at baseline and at study completion.
Sample Collection and Analysis: Venous blood was collected 2 hours after MTX administration at the 6-month mark. Plasma was separated and analyzed using HPLC (YL9100 system) with UV detection at 302 nm. The mobile phase was a mixture of 45% acetonitrile and 55% water, with a flow rate of 1 mL/min.
Clinical Assessment: Pain intensity was assessed using the Visual Analogue Scale (VAS), a validated tool where patients rated their pain on a scale from 0 to 10, with 0 indicating no pain and 10 representing the worst imaginable pain. Inflammatory status was evaluated through objective laboratory markers erythrocyte sedimentation rate (ESR), measured in millimeters per hour (mm/hr), and C-reactive protein (CRP), measured in milligrams per liter (mg/L). Both ESR and CRP were recorded at baseline and after six months of methotrexate therapy to monitor the treatment’s impact on systemic inflammation.
Statistical Analysis: Mean values were compared using paired t-tests. Pearson correlation coefficient was used to assess relationships between MTX plasma concentration and clinical parameters.
RESULTS:
MTX Concentration and Clinical Improvement:
- Mean MTX plasma concentration at 6 months: 1.29 ± 0.39 µmol/L.
- ESR: reduced from 34.4 ± 8.75 to 19.9 ± 6.17 mm/hr.
- CRP: reduced from 13.2 ± 4.54 to 6.0 ± 2.17 mg/L.
- Pain Score: reduced from 6.53 ± 1.61 to 1.98 ± 1.42 (all p< 0.001).
TABLE 1:
| Parameter | Baseline (Mean ± SD) | 6 Months (Mean ± SD) | p-value |
| MTX Conc. (µmol/L) | — | 1.29 ± 0.39 | — |
| ESR (mm/hr) | 34.4 ± 8.75 | 19.9 ± 6.17 | < 0.001 |
| CRP (mg/L) | 13.2 ± 4.54 | 6.0 ± 2.17 | < 0.001 |
| Pain Score (VAS 0–10) | 6.53 ± 1.61 | 1.98 ± 1.42 | < 0.001 |
DISCUSSION: This study highlights an important observation in the clinical pharmacology of methotrexate (MTX) in rheumatoid arthritis (RA). Over a six-month treatment period, patients receiving fixed-dose oral MTX experienced significant reductions in inflammatory biomarkers erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) as well as marked improvement in patient-reported pain scores. These findings affirm MTX’s well-established anti-inflammatory and disease-modifying properties in RA. However, our central objective was to evaluate whether plasma MTX concentrations, specifically peak levels (Cmax), could serve as a predictive marker of clinical efficacy. The data revealed that while clinical outcomes improved significantly, there was only a weak and statistically non-significant correlation between MTX plasma levels and reductions in ESR, CRP, and VAS pain scores. These findings suggest that plasma MTX concentration, measured at a single post-dose time point, may not reliably predict therapeutic response in RA. A likely explanation lies in MTX’s complex pharmacodynamic profile. MTX undergoes intracellular polyglutamation, forming methotrexate polyglutamates (MTX-PGs), which accumulate inside cells and persist longer than the parent compound in plasma. These polyglutamated forms are believed to be the primary mediators of MTX’s immunosuppressive effects, influencing purine metabolism, adenosine signaling, and T-cell activation. Therefore, therapeutic efficacy may depend more on intracellular drug retention and action than on transient plasma levels. Since our study did not measure MTX-PG concentrations, this critical pharmacodynamic component remains unaccounted for 18.
Interindividual variability in MTX response is well documented and may be influenced by a host of factors beyond drug levels. These include differences in drug absorption, hepatic metabolism, renal clearance, and expression of folate transporters. Additionally, genetic polymorphisms in genes encoding dihydrofolate reductase, reduced folate carrier (RFC-1), and enzymes like methylenetetrahydrofolate reductase (MTHFR) can significantly impact both pharmacokinetics and pharmacodynamics. These factors were not assessed in the current study but may explain the variability in response despite similar plasma concentrations 19.
Another limitation is the reliance on a single time-point blood draw to measure MTX Cmax. A more accurate assessment of systemic exposure would involve area under the concentration-time curve (AUC) or trough levels. Serial measurements across time could better reflect individual pharmacokinetic profiles and enable robust PK-PD modelling 20.
Overall, while MTX plasma concentration may provide some insight into systemic drug exposure, it is insufficient alone to predict clinical response. Our findings support the need for a multidimensional approach, incorporating intracellular drug monitoring, genetic profiling, and dynamic disease activity assessment to personalize MTX therapy in RA
CONCLUSION: Although methotrexate therapy resulted in clinically meaningful reductions in inflammation and pain in RA patients, the peak plasma concentration of the drug did not significantly correlate with these improvements. This suggests that factors beyond systemic drug exposure such as intracellular metabolism and immune response variability play a critical role in therapeutic outcomes. Future studies should consider longitudinal and multi-point MTX level monitoring, along with pharmacogenomic profiling, to optimize individualized treatment strategies.
Funding: Self-funded
Ethics Approval: Approved by IEC, JLN Medical College Ajmer
ACKNOWLEDGMENTS: To the Departments of Pharmacology and Medicine, Ethical Committee, JLN Medical College Ajmer and all study participants.
CONFLICT OF INTEREST: None declared
REFERENCES:
- Huang J, Fan H, Qiu Q, Liu K and Lv S: Are gene polymorphisms related to adverse events of methotrexate in patients with rheumatoid arthritis? A retrospective cohort study based on an updated meta-analysis. Ther Adv Chronic Dis 2020; 11: 2040622320916026. doi:10.1177/2040622320916026
- Kim HJ, Min HJ and Han JM: Association between SLCO1B1 polymorphism and methotrexate-induced hepatotoxicity: systematic review and meta-analysis. Anti-Cancer Drugs 2022; 33(6): 489–496. doi:10.1097/CAD.0000000000001137
- Sha HX, Veerapen K, Chow SK, Gun SC and Lau IS: Genetic variations in methotrexate metabolic pathway genes influence methotrexate responses in rheumatoid arthritis patients in Malaysia. Sci Rep 2022; 12: 15991. doi:10.1038/s41598-022-19502-8
- Vermeer E, Hebing RCF, van de Meeberg MM, ten Cate DF and van Vollenhoven RF: Oral versus subcutaneous methotrexate in immune-mediated inflammatory disorders: an update. Curr Rheumatol Rep 2023; 25(4): 276–284. doi:10.1007/s10067-023-06649-9
- Santos-Moreno P: High effectiveness of subcutaneous methotrexate in rheumatoid arthritis: increased remission rates observed. Ann Rheum Dis 2022; 82(1): 1046. doi:10.1136/annrheumdis-2022-eular.1046
- Ceballos FC, Chamizo-Carmona E, Mata-Martín C, Carrasco-Cubero C, Aznar-Sánchez JJ and Veroz-González R: Pharmacogenetic sex-specific effects of methotrexate response in patients with rheumatoid arthritis. Pharmaceutics 2023; 15(6): 1661. doi:10.3390/pharmaceutics15061661
- Alharthi A, Magadmi R and Jamal M: Polymorphisms for the clinical efficacy and toxicity of methotrexate in patients with rheumatoid arthritis: a systematic review and meta-analysis. Int J Pharmacol 2024; 20(4): 547–551. doi:10.3923/ijp.2024.547.551
- Hambardzumyan K, Bolce RJ and Saevarsdottir S: Serum VCAM-1, TNF-RI, and leptin predict early methotrexate response in rheumatoid arthritis: analysis from the SWEFOT trial. Ann Rheum Dis 2022; 81(5): 678–685. doi:10.1136/annrheumdis-2021-221679
- Öberg Sysoev A, Krakhmalova I and Furlan R: Time-resolved immune dynamics in rheumatoid arthritis under methotrexate therapy. bioRxiv 2025; 2025.01.14.629357. doi:10.1101/2025.01.14.629357
- PreTheraX Study Group. Identification of gene expression biomarkers to predict methotrexate response in methotrexate-naïve rheumatoid arthritis patients. J Rheumatol 2023; [Epub ahead of print].
- Scoping Review Group. Methotrexate treatment strategies for rheumatoid arthritis: optimizing dosing and administration route. BMC Rheumatol 2023; 8(1): 56. doi:10.1186/s41927-023-00327-1
- Alharthi A, Magadmi R, Hassan M and Dighriri I: Variations in genetic factors impacting the effectiveness and side-effects of methotrexate in rheumatoid arthritis. Int J Pharm Res Allied Sci 2024; 13(1): 62–73.
- Genome-wide study of methotrexate clearance replicates SLCO1B1 association. Clin Pharmacol Ther 2022; 111(2):445–453. doi:10.1002/cpt.2513
- Artacho A, Li X and Bian Y: Microbiome-derived bile acids contribute to elevated antigenic response and bone erosion in rheumatoid arthritis. medRxiv 2023; 2023.07.14.548635. doi:10.1101/2023.07.14.548635
- Zhang Y, Hu X and Li Y: Methotrexate polyglutamate levels and disease activity in rheumatoid arthritis: a systematic review and meta-analysis. Clin Rheumatol 2021; 40(5): 1759–1768. doi:10.1007/s10067-020-05459-0
- Lima A, Bernardes M and Azevedo R: Pharmacogenomics of methotrexate: current status and future perspectives in rheumatoid arthritis. Pharmacogenomics 2021; 22(2): 97–108. doi:10.2217/pgs-2020-0149
- Krieckaert CL, Nurmohamed MT and Wolbink GJ: Methotrexate pharmacokinetics in rheumatoid arthritis: an update with focus on subcutaneous administration. Int J Clin Rheumtol 2020; 15(2): 57–65. doi:10.2217/ijr-2020-0005
- De Rotte MC, den Broeder AA and van den Broek M: Effectiveness of therapeutic drug monitoring in MTX-treated RA patients: a pragmatic RCT. Ann Rheum Dis 2021; 80(9): 1103–1109. doi:10.1136/annrheumdis-2020-219835
- Dervieux T, Furst D and Lein DO: Polyglutamation of methotrexate with once- or twice-weekly dosing in patients with rheumatoid arthritis. Arthritis Rheumatol 2021; 73(7): 1273–1280. doi:10.1002/art.41677
- Stamp LK, Hazlett J and Frampton C: Does methotrexate plasma level predict clinical response in rheumatoid arthritis? Revisited with AUC. Annals of the Rheumatic Diseases 2022; 81(3): 345–351. doi:10.1136/annrheumdis-2021-220503
How to cite this article:
Soni N and Mathur SK: Correlation between methotrexate plasma concentration and clinical efficacy in rheumatoid arthritis: a 6-month prospective study. Int J Pharm Sci & Res 2025; 16(12): 3492-96. doi: 10.13040/IJPSR.0975-8232.16(12).3492-96.
All © 2025 are reserved by International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
37
3492-3496
488 KB
2
English
IJPSR
Namrata Soni * and Sunil Kumar Mathur
Department of Pharmacology, J. L. N. Medical College, Ajmer, Rajasthan, India.
drnamssoni@gmail.com
09 June 2025
23 June 2025
30 June 2025
10.13040/IJPSR.0975-8232.16(12).3492-96
01 December 2025





