Preprint
Review

This version is not peer-reviewed.

Modern Pharmacologic Management of Rheumatoid Arthritis: From Methotrexate to Targeted Therapies

Submitted:

15 December 2025

Posted:

16 December 2025

You are already at the latest version

Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune pathology characterized by symmetric synovitis, pannus formation, and the potential for severe extra-articular manifestations. Despite historical associations with high morbidity, the 'treat-to-target' strategy has revolutionized patient outcomes. This review analyzes the pharmacological evolution of RA management over the last decade. We examine the foundational role of Methotrexate (MTX)—specifically its adenosine-mediated mechanism—and the stratification of biologic disease-modifying antirheumatic drugs (b-DMARDs), including TNF inhibitors, IL-6 receptor antagonists, and B-cell depleting agents. Crucially, we discuss the recent paradigm shift in the use of Janus Kinase (JAK) inhibitors following the 2021 ACR Guidelines and emerging safety data regarding cardiovascular and malignancy risks (the ORAL Surveillance trial). Finally, we explore the horizon of RA treatment, including GM-CSF inhibition, the complex management of difficult-to-treat (D2T) phenotypes, and the increasing integration of biosimilars to improve global treatment access.
Keywords: 
;  ;  ;  ;  ;  ;  

1. Introduction

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder that affects approximately 0.5 to 1.0 percent of individuals globally. The incidence is two to three times higher in women and increases with age, reaching a peak in the fifth decade [1,2,3]. RA is characterized clinically by destructive, inflammatory synovitis that results in joint damage and substantial physical impairment; however, the underlying pathology begins long before clinical onset.
Pathophysiology: The Cytokine Cascade Although the etiology of RA is multifactorial, involving a complex interplay of genetic susceptibility (e.g., HLA-DRB1 alleles) and environmental triggers (smoking, mucosal dysbiosis), the final common pathway involves the loss of tolerance to self-antigens (such as citrullinated proteins). Auto-reactive T lymphocytes (Th1 and Th17 subsets) infiltrate the synovium and interact with macrophages and B-cells [4].
This interaction triggers a massive release of pro-inflammatory cytokines, specifically Tumor Necrosis Factor (TNF), Interleukin-6 (IL-6), and Interleukin-1 (IL-1). These cytokines drive the proliferation of fibroblast-like synoviocytes (FLS), leading to the formation of invasive pannus tissue. Crucially, the expression of Receptor Activator of Nuclear Factor Kappa-B Ligand (RANKL) is upregulated, promoting the differentiation of osteoclasts which actively resorb bone, leading to the hallmark erosions seen on radiography [5]. Extra-articular manifestations—such as pleuritis, pericarditis, interstitial lung disease (ILD), and rheumatoid nodules—occur in up to 40% of patients and are driven by this same systemic cytokine burden [6].
The current RA treatment approach is "treat-to-target," utilizing early aggressive therapy to disrupt this cascade before permanent joint damage occurs.

2. Review

2.1. Methotrexate’s Crucial Role (MTX)

Methotrexate (MTX), a folate antagonist, remains the "anchor drug" in the treatment of RA for the majority of patients [7,8,9]. While originally designed as an anti-proliferative chemotherapy agent, its mechanism in RA is distinct. At low doses, MTX inhibits the enzyme AICAR transformylase, leading to the intracellular accumulation of AICAR (5-aminoimidazole-4-carboxamide ribonucleotide). This accumulation inhibits adenosine deaminase and increases the release of adenosine into the extracellular space. Adenosine acts as a potent anti-inflammatory mediator by binding to A2A receptors on immune cells, downregulating their function [10].
MTX typically achieves an ACR-20 response in approximately 70% of DMARD-naive patients. However, its use requires careful monitoring for hepatotoxicity, myelosuppression, and pulmonary toxicity. Folic acid supplementation (typically 1–5 mg daily) is mandatory to reduce common adverse effects such as stomatitis and GI distress without compromising efficacy.
Guideline Update on Glucocorticoids: Historically, glucocorticoids were used liberally as "bridge therapy" while waiting for MTX to take effect. The 2021 ACR Guidelines and 2022 EULAR Recommendations now strongly emphasize minimizing glucocorticoid exposure. They recommend that if glucocorticoids are used, they should be tapered and discontinued as rapidly as possible (ideally within 3 months) due to long-term toxicity (osteoporosis, metabolic syndrome), favoring rapid escalation of DMARDs instead [11,12].

2.2. Biologic Disease-Modifying Anti-Rheumatic Drugs (b-DMARDs)

For patients who fail to respond to MTX, b-DMARDs are the standard of care. These large protein molecules target specific components of the immune cascade:
  • TNF Inhibitors (TNFi): Agents like adalimumab, etanercept, and infliximab bind to soluble and transmembrane TNF, preventing it from binding to its receptors. They remain the most common first-line biologic.
  • IL-6 Receptor Antagonists: Tocilizumab and sarilumab block the IL-6 receptor. Since IL-6 is the primary driver of hepatic acute-phase reactant production, these drugs are particularly effective at normalizing C-reactive protein (CRP) levels and improving the "systemic" symptoms of RA such as anemia and fatigue.
  • B-Cell Depletion: Rituximab targets CD20-positive B cells. By depleting these cells, it reduces autoantibody production (Rheumatoid Factor and ACPA) and inhibits B-cell antigen presentation to T cells.
  • T-Cell Co-stimulation Modulation: Abatacept (CTLA-4 Ig) binds to CD80/86 on antigen-presenting cells, preventing the "second signal" required for T-cell activation.
Comorbidity-Guided Selection:
  • Interstitial Lung Disease (ILD): For patients with RA-ILD, retrospective studies suggest that Abatacept or Rituximab may be preferred over TNFi. Some cohorts have suggested TNFi might be associated with ILD exacerbation, whereas Abatacept has shown a stabilizing effect on lung function in observational studies [13,14].
  • Monotherapy: In patients unable to tolerate MTX, Tocilizumab and Sarilumab have demonstrated superiority over adalimumab when used as monotherapy, likely due to their profound effect on the IL-6 signaling axis [15].

3. Molecular-Targeted Agents: The JAK Inhibitor Safety Shift

Targeted synthetic DMARDs (ts-DMARDs) represent a major advancement in oral therapy. Unlike biologics, which target extracellular cytokines, JAK inhibitors (tofacitinib, baricitinib, upadacitinib) enter the cell and inhibit the Janus Kinase (JAK) enzymes (JAK1, JAK2, JAK3, and TYK2) attached to the cytoplasmic tail of cytokine receptors. This blockade prevents the phosphorylation of STAT proteins and subsequent gene transcription [16,17].
Efficacy vs. Safety (The ORAL Surveillance Trial): While JAK inhibitors demonstrated rapid efficacy, often superior to MTX, their safety profile has come under intense scrutiny. The ORAL Surveillance trial (NCT02092467) [18], published in 2022, was a mandatory post-marketing safety study comparing tofacitinib (5mg and 10mg BID) against TNFi in high-risk patients (age ≥50 with ≥1 cardiovascular risk factor).

Key Findings:

  • MACE (Major Adverse Cardiovascular Events): The hazard ratio (HR) for MACE with tofacitinib was 1.33 (95% CI, 0.91 to 1.94) compared to TNFi, failing the non-inferiority criteria.
  • Malignancy: The HR for malignancies (excluding non-melanoma skin cancer) was 1.48 (95% CI, 1.04 to 2.09).
  • Thrombosis: An increased, dose-dependent risk of venous thromboembolism (VTE) and pulmonary embolism was also observed.
Clinical Implications: Due to these findings, the FDA issued "Black Box" warnings for the entire class of JAK inhibitors. The 2021 ACR Guidelines were updated to conditionally recommend attempting a TNFi or non-TNF biologic before initiating a JAK inhibitor [12]. JAK inhibitors are now generally reserved for patients who have had an inadequate response or intolerance to at least one TNF inhibitor, particularly in patients with existing cardiovascular risk factors.

4. Emerging Therapies and Future Directions

Difficult-to-Treat (D2T) RA: A growing area of research is "Difficult-to-Treat" (D2T) RA, formally defined by EULAR in 2021. This classification applies to patients who have failed treatment with ≥2 b/tsDMARDs with different mechanisms of action and who display signs of active or progressive disease [19]. The definition acknowledges that "resistance" is not always inflammatory; it encompasses non-inflammatory pain mechanisms (such as central sensitization or secondary fibromyalgia) that do not respond to immunosuppression. Management of D2T RA requires a holistic approach, distinguishing true synovial inflammation (requiring drug escalation) from non-inflammatory pain (requiring adjunctive analgesia or physical therapy).

Novel Targets:

  • GM-CSF Inhibitors: Therapies targeting Granulocyte-macrophage colony-stimulating factor (GM-CSF), such as otilimab and mavrilimumab, target the innate immune activation of macrophages. Otilimab has shown promise in pain reduction endpoints even where inflammation suppression was comparable to current standards, suggesting a potential role in decoupling pain from inflammation [20].
  • Bioelectronic Medicine: The "inflammatory reflex" describes the vagus nerve's ability to inhibit splenic cytokine production. Small trials utilizing implantable Vagus Nerve Stimulation (VNS) devices have shown reductions in TNF production and disease activity scores in drug-refractory RA, offering a potential non-pharmacologic future avenue [21].
  • Biosimilars: The widespread approval of biosimilars for adalimumab, etanercept, infliximab, and rituximab continues to lower costs. "Switch studies" (like NOR-SWITCH) have largely confirmed that switching from a bio-originator to a biosimilar is safe and effective, increasing global access to these life-changing therapies [22,23].

5. Conclusions

The management of Rheumatoid Arthritis has evolved from simple immunosuppression to precision medicine. While Methotrexate remains the foundational anchor, the arsenal has expanded to include diverse biologics and targeted synthetic drugs. The choice of therapy is now highly personalized: it must balance the superior oral efficacy of JAK inhibitors against their established cardiovascular safety signals and consider extra-articular comorbidities like ILD when selecting biologics. The updated 2021 ACR and 2022 EULAR guidelines reflect this complexity, prioritizing safety and strictly limiting glucocorticoid toxicity. Future advances in GM-CSF inhibition and bioelectronic medicine may further refine the management of difficult-to-treat disease, moving us closer to the goal of drug-free remission.

Authors' Contributions

Mohammad Asim Amjad: Conceptualization, literature search, data synthesis, drafting of the manuscript, and critical revision. Zamara Hamid: Literature search, data verification, reviewing, and editing of the final manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Ethical Approval

Not applicable. This article is a review of existing literature and does not contain any studies with human participants or animals performed by any of the authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Guo, Q; Wang, Y; Xu, D; Nossent, J; Pavlos, NJ; Xu, J. Rheumatoid arthritis: pathological mechanisms and modern pharmacologic therapies. Bone Res. 2018, 6, 15. [Google Scholar] [CrossRef]
  2. McInnes, IB; Schett, G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 2017, 389(10086), 2328–2337. [Google Scholar] [CrossRef]
  3. Donahue, KE; Gartlehner, G; Schulman, ER; et al. Drug Therapy for Early Rheumatoid Arthritis: A Systematic Review Update. In AHRQ Comparative Effectiveness Reviews; 2018. [Google Scholar]
  4. Furst, DE; Emery, P. Rheumatoid arthritis pathophysiology: update on emerging cytokine and cytokine-associated cell targets. Rheumatology (Oxford) 2014, 53(9), 1560–1569. [Google Scholar] [CrossRef]
  5. Tanaka, Y. Current concepts in the management of rheumatoid arthritis. Korean J Intern Med. 2016, 31(2), 210–218. [Google Scholar] [CrossRef]
  6. Wijbrandts, CA; Tak, PP. Prediction of Response to Targeted Treatment in Rheumatoid Arthritis. Mayo Clin Proc. 2017, 92(7), 1129–1143. [Google Scholar] [CrossRef] [PubMed]
  7. Burmester, GR; Pope, JE. Novel treatment strategies in rheumatoid arthritis. Lancet 2017, 389(10086), 2338–2348. [Google Scholar] [CrossRef]
  8. Wilsdon, TD; Hill, CL. Managing the drug treatment of rheumatoid arthritis. Aust Prescr. 2017, 40(2), 51–58. [Google Scholar] [CrossRef]
  9. Aletaha, D; Smolen, JS. Diagnosis and Management of Rheumatoid Arthritis: A Review. JAMA 2018, 320(13), 1360–1372. [Google Scholar] [CrossRef]
  10. Alfaro-Lara, R; Espinosa-Ortega, HF; Arce-Salinas, CA. Systematic review and meta-analysis of the efficacy and safety of leflunomide and methotrexate in the treatment of rheumatoid arthritis. Reumatol Clin. 2019, 15(3), 133–139. [Google Scholar] [CrossRef] [PubMed]
  11. Smolen, JS; Landewé, RBM; Bergstra, SA; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2022 update. Ann Rheum Dis. 2023, 82(1), 3–18. [Google Scholar] [CrossRef] [PubMed]
  12. Fraenkel, L; Bathon, JM; England, BR; et al. 2021 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol. 2021, 73(7), 1108–1123. [Google Scholar] [CrossRef] [PubMed]
  13. Cassone, G; Manfredi, A; Vacchi, C; et al. Treatment of Rheumatoid Arthritis-Associated Interstitial Lung Disease: Lights and Shadows. J Clin Med. 2020, 9(4), 1082. [Google Scholar] [CrossRef] [PubMed]
  14. Fernandez-Diaz, C; Loricera, J; Castañeda, S; et al. Abatacept in patients with rheumatoid arthritis and interstitial lung disease: A national multicenter study of 63 patients. Semin Arthritis Rheum. 2018, 48(1), 22–27. [Google Scholar] [CrossRef]
  15. Mahajan, TD; Mikuls, TR. Recent advances in the treatment of rheumatoid arthritis. Curr Opin Rheumatol. 2018, 30(3), 231–237. [Google Scholar] [CrossRef]
  16. Rivellese, F; Lobasso, A; Barbieri, L; Liccardo, B; De Paulis, A; Rossi, FW. Novel therapeutic approaches in Rheumatoid Arthritis: Role of Janus Kinases Inhibitors. Curr Med Chem. 2019, 26(9), 1497–1510. [Google Scholar] [CrossRef]
  17. Yamaoka, K. Janus kinase inhibitors for rheumatoid arthritis. Curr Opin Chem Biol. 2016, 32, 29–33. [Google Scholar] [CrossRef]
  18. Ytterberg, SR; Bhatt, DL; Mikuls, TR; et al. Cardiovascular and Cancer Risk with Tofacitinib in Rheumatoid Arthritis. N Engl J Med. 2022, 386(4), 316–326. [Google Scholar] [CrossRef]
  19. Nagy, G; Roodenrijs, NMT; Welsing, PM; et al. EULAR definition of difficult-to-treat rheumatoid arthritis. Ann Rheum Dis. 2021, 80(1), 31–35. [Google Scholar] [CrossRef]
  20. Buckley, CD; Isa, F; Nash, P; et al. Otilimab in moderate to severe rheumatoid arthritis (contRAst 1 and 2): two randomised, double-blind, placebo-controlled, phase 3 trials. Lancet Rheumatol. 2023, 5(11), e636–e648. [Google Scholar]
  21. Genovese, MC; Gaylis, NB; Sikes, D; et al. Safety and Efficacy of Neurostimulation With a Miniaturized Vagus Nerve Stimulator in Patients With Multidrug-Refractory Rheumatoid Arthritis. Lancet Rheumatol. 2020, 2(9), e527–e538. [Google Scholar] [CrossRef]
  22. Pisetsky, DS. Advances in the Treatment of Rheumatoid Arthritis: Costs and Challenges. N C Med J 2017, 78(5), 337–340. [Google Scholar] [CrossRef] [PubMed]
  23. Bortoluzzi, A; Furini, F; Generali, E; Silvagni, E; Luciano, N; Scirè, CA. One year in review 2018: novelties in the treatment of rheumatoid arthritis. Clin Exp Rheumatol. 2018, 36(3), 347–361. [Google Scholar] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated