Department of Rheumatology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
Background: Both articular and extra-articular structures are affected by rheumatoid arthritis (RA), a chronic inflammatory rheumatic illness that results in severe joint destruction, disability, and death. To increase the response rate and provide RA patients more options, there is an unmet need for the development of innovative treatment drugs. Evaluation of cellular, cytokine, genomic, and transcriptome profiles that would predict therapeutic response to biologic or targeted disease-modifying anti-rheumatic drugs (DMARDs) with various action modes is necessary for a customized therapy plan in RA. Owing to the development of new biologic medicines that target distinct mechanisms of action, the treatment algorithm for RA has undergone significant modification during the last one to two decades. More patients are now able to undergo biologic therapy early in the progression of their illness, thanks to the availability of less expensive biosimilars.
Objective: To summarize the efficacy and safety of biologic or targeted syntheticb/ts DMARDs in RA treatment based on the publications in the past year.
Material and Methods: We compiled the most recent findings from original research publications of 2024 about the effects of b/tsDMARDs on the treatment of RA. In addition, this review article also concluded the recent findings from original research publications of the year 2024 about the effects of b/tsDMARDs biosimilars for treating RA.
Conclusion: This article summarizes the evidence and safety of biologic DMARDs (bDMARDs) or targeted synthetic DMARDs (tsDMARDs) in the management of RA, including those that target tumor necrosis factor alpha, interleukin (IL)-6, B cells, T-cell co-stimulation, and Janus kinase (JAK) from original research articles published in 2024.
Key words: b/ts DMARDs, JAK, IL-6, rheumatoid arthritis, TNF-α
*Corresponding author: Daqing Nie, Department of Rheumatology, the Affiliated Hospital to Changchun University of Chinese Medicine, No. 1478, Gongnong Avenue, Changchun, Jilin 130021, China. Email address: [email protected]
Received 28 November 2024; Accepted 25 January 2025; Available online 1 May 2025
Copyright: Liu D, et al.
License: This open access article is licensed under Creative Commons Attribution 4.0 International (CC BY 4.0). http://creativecommons.org/licenses/by/4.0/
Inflammation is a hallmark of rheumatoid arthritis (RA), a chronic autoimmune illness. Patients with RA frequently experience bilateral joint stiffness, swelling, and discomfort.1 The etiology and pathophysiology of RA are unknown and incredibly complex. They include epigenetic changes, environmental stimuli, and inheritance.2 RA is a global health issue that affects around 240 out of every 100,000 individuals. Furthermore, since the world’s population is aging quickly, the prevalence of RA is increasing globally.3 Moreover, when RA progresses, harm to extra-articular organs and systems may happen. According to epidemiological data, people with RA have a 3–10 years lower life expectancy than people in the general population because they are more likely to experience cardiovascular (CV) effects, infections, pulmonary diseases, and various types of cancers.4,5 Thankfully, early detection of the condition, and the significant progress in managing RA over the past 10 years, can result in better health outcomes, quicker attainment of treatment objectives, and preservation of joint functions.6
The first-line management options for RA are disease-modifying anti-rheumatic drugs (DMARDs), which help to attenuate clinical manifestations as well as slow the course of joint deterioration. A significant percentage of patients are either nonresponders or insufficient responders to conventional synthetic DMARDs (csDMARDs), despite the fact that these medications are linked with better results.7 In the last one to two decades, the development of biologic DMARDs (bDMARDs) or targeted synthetic DMARDs (tsDMARDs) has completely changed the way RA is treated. According to the updated 2022 European Alliance of Associations for Rheumatology (EULAR) guidelines, short-term bridging glucocorticoids (GCs) and a single csDMARD, such as methotrexate (MTX), should be used to treat RA.8 In cases where methotrexate is contraindicated or intolerant, other csDMARDs include leuflunomide or sulphasalazine. bDMARDs or tsDMARDs may be appended as the second phase of therapy in patients with unfavorable prognostic factors if clinical improvement is not observed at 3 months or if the treatment goal (remission or state of low disease activity [LDA]) is not reached at 6 months.
The first class of bDMARDs to be licensed for the management of RA was anti-tumor necrosis factor (TNF) biologics. Several biologic agents that target interleukin (IL)-6, B cells, and co-stimulation of T cells were studied in RA. In the arsenal of RA treatment, there are now a number of specific tsDMARDs that mark the Janus kinase–signal transducer and activator of transcription (JAK-STAT) pathways. In this review article, we compiled the most recent findings from original research publications of 2024 about the effects of b/tsDMARDs on the treatment of RA (Figure 1). Besides, biosimilar drugs are potentially low-cost versions that have been used in RA treatment and have the potential to improve access to therapy.9,10 Moreover, several systematic reviews have compiled evidence on similar efficacy and safety between biosimilars and reference products for RA at both qualitative and quantitative levels.11,12 International guidelines and consensus recommend biosimilars for RA management.8,13–16 Thus, this review article also concluded the recent findings from original research publications of the year 2024 about the effects of b/tsDMARD biosimilars for treating RA.
Figure 1 b/tsDMARDs for RA treatment. TNFis (i.e., adalimumab, etanercept, golimumab, infliximab, certolizumab, and ozoralizumab), IL-6 inhibitors (i.e., tocilizumab, sarilumab, and olokizumab), anti-CD20 (rituximab), CTLA-4 immunoglobulin (abatacept), and JAKis (i.e., tofacitinib, baricitinib, and upadacitinib) were summarized in this mauscript for the treatment of RA.
TNF-α is essential to the pathophysiology of RA. Both synovium and synovial fluid are discovered to contain it.17 The pathophysiology of inflammatory osteolysis, osteoclast recruitment, and bone destruction are dependent on TNF-α; thus, it is essential for bone deterioration.18 The addition of TNF-α inhibitors to the RA therapy regimen has considerable improved the quality of life, joint injury prevention, and recovery of physical functions. Additionally, it is now easier to maintain control over RA manifestations.19 TNFi may weaken the immune system and raise the risk of severe infections, although it prevents TNF-α from stimulating proinflammatory pathway.20 For instance, JAK inhibitors (JAKi) and TNFi were used to treat a 59-year-old male with RA who developed iatrogenic Kaposi sarcoma.21 According to a case series, TNFi caused systemic lupus erythematosus in RA patients, suggesting that systemic lupus erythematosus should be monitored in RA patients receiving TNFi treatment.22 Although each TNFi had a unique positive adverse drug event (ADE), a real-world analysis based on the US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) database revealed that there were four common ADEs among TNFi: infection, pulmonary TB, hypersensitivity, and herpes zoster.23 TNFi administered to RA patients were linked to the development of Merkel cell cancer.24 As a result, TNFi are now the first option for RA patients who are intolerant of or do not respond to bDMARD treatment with csDMARDs. So far, six TNF-α targeted biologic agents—adalimumab, golimumab, certolizumab, etanercept, infliximab, and ozoralizumab—have received global approval for managing moderate-to-severe RA.
A critical component of the biologic treatment of RA is adalimumab, a TNFi that binds to TNF-α and prevents TNF-mediated cellular inflammation. Globally, it is among the most commonly prescribed biologic drugs.25 Adalimumab, either by itself or in conjunction with csDMARDs, such as methotrexate, has been shown in clinical trials and real-world investigations to considerably lower disease activity and improve patient outcomes.26,27
Fifty-six RA patients who did not respond to methotrexate were treated with adalimumab according to established therapeutic guidelines in a prospective, observational, single-center research. Twenty-four (42.9%) patients failed the American College of Rheumatology (ACR) 20 response after receiving continuous adalimumab treatment for 12 weeks.28 According to a retrospective analysis of 105 RA patients that received either adalimumab or etanercept, adalimumab outperformed etanercept in terms of a numerically higher response rate and comparable adverse effects.29 Tripterygium wilfordii Hook F could be an alternative management for the RA patients intolerant to methotrexate, according to the results of a multicenter, open-label, randomized controlled clinical trial that treated 64 RA patients with inadequate response to methotrexate with either adalimumab in combination with tripterygium wilfordii Hook F or methotrexate with tripterygium wilfordii. No significant differences in efficacy and safety was observed between adalimumab combined with tripterygium wilfordii Hook F and adalimumab combined with methotrexate.30 According to a population-based cohort research, adalimumab and infliximab were linked to the longest drug survival periods among late-onset RA patients, compared to early-onset patients when it came to drug survival on first biologic therapy.31 Adalimumab-treated RA patients had a decreased incidence of dyslipidemia than tofacitinib-treated RA patients, according to a real-world cohort research involving 7580 patients; however, there were no differences in major adverse CV events (MACEs) or all-cause mortality.32 A 47-year-old female with RA who was diagnosed with multiple sclerosis was treated with adalimumab for 2 years.33 According to Shirah et al., a patient with RA experienced demyelination of the central nervous system because of adalimumab.34
Additionally, a few biosimilars of adalimumab have also shown promising results in treating RA because of their lower cost, bioequivalence efficacy, and safety. Phase I and III clinical trials, pre-clinical and analytical data, and proof of similar efficacy, pharmacokinetics, safety, and immunogenicity profiles of the reference adalimumab were the basis for the 2017 European approval of adalimumab biosimilar SB5.35 Patients with RA who started routine SB5 medication as their first adalimumab (naïve) or when they switched from another adalimumab (switched) were the subjects of a 12-month observational, multi-center cohort trial. The results showed that naïve patients’ 12-month remission/low-activity proportions for RA were 58%, significantly higher than baseline, while switched patients’ remission proportions were constant between baseline and month 12 (M12) for all reasons.36 With a safety profile in keeping with the one reported for other biosimilars, a retrospective and multicenter investigation demonstrated that adalimumab biosimilar MSB11022 preserved the efficacy advantages offered by prior adalimumab therapies.37 Adalimumab and biosimilar (GP2017) patient groups did not differ in disease activity levels, according to Colina et al.38 During the first 18 months of treatment, all scores dropped dramatically across all treatments. They hit a low point at 24 months, which persisted for as long as 48 months, suggesting that adalimumab biosimilars were just as effective as their original medications over an extended period.
The recombinant human TNF receptor p75Fc fusion protein etanercept, which binds to either soluble or transmembrane TNF-α, is the first TNFi approved by the US FDA for rheumatic diseases.39 According to a population-based cohort research comparing the drug survival on first biologic therapy among late-onset RA patients to those with early-onset RA, etanercept was linked to the longest drug survival period of the first biologic in early-onset patients.31 According to the propensity score-matched analysis from the Czech ATTRA registry, patients with RA treated with etanercept had a longer median survival period than those treated with monoclonal antibody tumor necrosis factor-α inhibitors.40 A two-year pragmatic, randomized CareRA2020 trial assessing the efficacy of methotrexate and bridging glucocorticoids with or without early introduction of a 6-month course of etanercept in early RA discovered that adding etanercept for 6 months did not improve disease control over 104 weeks, compared to adding leflunomide first in early insufficient responders. However, etanercept’s brief introduction resulted in better disease management immediately following randomization and fewer patients on b/tsDMARDs at 104 weeks.41
Regarding safety, a 68-year-old RA patient experienced aseptic meningitis during etanercept and methotrexate treatment, according to Kassabian et al.42 A 35-year-old female with RA since the age of 18 years developed clinical manifestations similar to multiple sclerosis as a result of using etanercept.43 Ramsay Hunt syndrome was identified in a 38-year-old RA patient receiving etanercept.44 A 67-year-old male with RA on etanercept therapy developed pulmonary embolism (PE).45 One patient using methotrexate and etanercept for RA developed multifocal leukoencephalopathy.46 According to a population-based cohort study on the incidence and risk factors of stopping tofacitinib and bDMARDs (etanercept, adalimumab, golimumab, tocilizumab, or abatacept) in RA patients, etanercept had the highest proportion (43.27%), while tofacitinib had the lowest proportion (21.8%) of drug discontinuation. Higher steroid dosage and concurrent connective tissue diseases were identified as risk factors for stopping the medication.47
Numerous etanercept biosimilars have been used in clinical settings. According to an open-label, randomized, comparative, multicenter report evaluating biosimilar etanercept’s efficacy, safety, and immunogenicity in RA patients, compared to the reference formulation of original etanercept, the American College of Rheumatology 20 (ACR20) response proportions in the biosimilar etanercept and etanercept groups were 82.3% and 90.9%, respectively, with no significant difference. Furthermore, there were no appreciable variations in the frequency of adverse responses or incidents, regardless of their intensity, between the two groups. According to the findings, the reference formulation and biosimilar etanercept were identical.48
According to a retrospective 12-month follow-up research, RA patients who used the original etanercept to achieve remission were able to keep it going even after switching to biosimilar YLB113.49 Following the release of etanercept biosimilar, a 49-year-old female with RA acquired antimelanoma differentiation-associated gene 5 antibody-positive dermatomyositis.50 Etanercept biosimilar LBEC0101 and reference were compared for safety and efficacy in RA patients using the Kyoto University Rheumatoid Arthritis Management Alliance (KURAMA) cohort. According to the results, LBEC0101 and the reference product were comparable in terms of continuation rate, introduction efficacy, and impact permanence both before and after switching in clinical practice.51
According to Colina et al.,38 no variations in disease activity levels were observed between patient groups receiving etanercept and biosimilars (GP2015/SB4). During the first 18 months of treatment, all scores dropped dramatically across all treatments. The scores hit a low point at 24 months, which persisted for as long as 48 months, suggesting that biosimilars are just as effective as their original medications over an extended period. Owing to the relative efficacy of etanercept originator and biosimilar in treating RA, treatment persistence was comparable when taking either one of the two.52 The European Commission has approved SDZ ETN as an etanercept biosimilar for the same indications as the reference etanercept, including RA, axial-spondyloarthritis (axSpA), psoriatic arthritis (PsA), and juvenile idiopathic arthritis (JIA). SDZ ETN has been approved by the US FDA to treat ankylosing spondylitis (AS), RA, PsA, and JIA.53 According to a multi-country COMPACT research, At 12 months, patients with PsA, axSpA, or RA receiving SDZ ETN demonstrated good treatment persistence.54 According a matched-analysis of the BSRBR-RA, among terms of disease activity and treatment persistence, individuals who transitioned from etanercept originator to etanercept biosimilar among 1024 RA patients appeared to perform similarly, compared to those who remained on originator.55 In a JET observational study carried out in Japanese clinical practice, the treatment continuity, utility, and nocebo effect of transferring from the original etanercept to its biosimilar in patients with RA were investigated. Even in ordinary clinical practice at rheumatology clinics in Japan, the results showed that moving from etanercept to biosimilar was successful, and no nocebo effects were observed.56
Approved to treat rheumatic illnesses, golimumab is a fully humanized monoclonal IgG1κ antibody with a high affinity for TNF-α.57,58 The GO-BEYOND study, which was conducted from 2017 to 2019, involved 39 RA patients who were started on golimumab following the failure of a first-line TNFi. Of these patients, 80% achieved at least LDA, with 60% achieving complete remission, and 88.2% showed a good/moderate EULAR response after a 12-month follow-up.59 A pooled analysis of European prospective observational studies showed improvements in disease activity, quality of life, and treatment persistence rate without any new safety signals when golimumab was used to treat patients with active RA, PsA, or axSpA who had not responded to treatment with an initial TNFi.60 A post hoc analysis of the GO-FORTH research employing cluster analysis for efficacy of golimumab in RA patients at high risk of a bad prognosis discovered that the drug was efficacious in a subset of RA patients at high risk of poor prognostic characteristics. For this cohort, a dose of 100 mg would be more beneficial in halting radiographic progression.61 A cohort study of RA patients’ self-reported experiences and satisfaction with golimumab and etanercept therapies discovered no differences between the two groups’ satisfaction levels with global disease improvement, symptom alleviation, and speed of action. In contrast to the etanercept group, the golimumab group experienced less discomfort, swelling, pain, and burning, and was more satisfied with the global injection experience, injection device, frequency, and convenience.62 Golimumab has six distinct positive ADEs, such as pneumonia cryptococcal, device failure, pneumonia bacterial, device issue, pneumocystis pneumonia, polyneuropathy, etc., according to a real-world study from the US FAERS database for the safety of TNFi.23 An 87-year-old female RA patient who was on golimumab and methotrexate experienced severe pneumocystis pneumonia.63
A chimeric monoclonal antibody called infliximab exhibits high affinity for both membrane-bound and soluble forms of TNF-α. According to a population-based cohort research, both adalimumab and infliximab were linked to the longest drug survival periods among late-onset RA patients, compared with early-onset patients when it came to drug survival on first biologic therapy.31 In a study on the features and 6-month results of RA patients, beginning infliximab biosimilar IFX-dyyb in a real-world setting, the Clinical Disease Activity Index (CDAI) remained constant in patients switching from reference infliximab/IFX biosimilar, but improved in patients switching from a non-IFX b/tsDMARD and in b/tsDMARD-naïve patients.64 When compared to originators, such as infliximab, etanercept, and adalimumab, an observational analysis on the French National Health Data System revealed that the safety and persistence of biosimilar TNFi were reassuring.65
Based on the US FAERS database for safety of TNFi, a real-world study discovered that infliximab has 60 different positive ADEs, such as Hodgkin’s disease, metastatic neoplasm, non-Hodgkin’s lymphoma, and others.23 After receiving long-term treatment with methotrexate, leflunomide, and infliximab, a 60-year-old female with more than 10 years of RA developed an actinomycosis infection.66 Infliximab-induced plaque psoriasis eruption on the palms and soles occurred in a 70-year-old female with a RA history.67 A 25-year-old female with RA using infliximab was diagnosed with new-onset uveitis after presenting with bilateral impaired vision.68 A female with RA was suspected of having colitis brought on by rituximab.69 After receiving treatment with methotrexate, bucillamine, prednisolone, and infliximab for RA, a 73-year-old female was diagnosed with the disease at the age of 60.70
A PEGylated, Fc-free TNFi is certolizumab. Numerous clinical trials that demonstrated a significant and long-lasting reduction in disease manifestations, suppression of illness progression, and improvements in physical performance and quality of life have encouraged the global adoption of certolizumab for the management of RA. Certolizumab pegol’s safety and efficacy in Japanese RA patients were confirmed by a post-marketing surveillance trial that lasted for up to 3 years.71 Certolizumab has 24 distinct positive ADEs, according to a real-world study from the US FAERS database for the safety of TNFi. These include exposure by the mother before becoming pregnant, early rupture of the membranes, exposure through breast milk, erysipelas, low birth weight, herpes virus infection, premature delivery, and staphylococcal sepsis.23
The sixth TNF inhibitor approved in Japan, ozoralizumab, was approved in September 2022. It is a ground-breaking next-generation TNFi known as Nanobody®, which has changeable heavy-chain domains of heavy-chain-only antibodies. No new safety issues were discovered in an integrated analysis of the OHZORA, NATSUZORA, and HOSHIZORA studies, and long-term ozoralizumab treatment preserved efficacy in RA patients.72 An 81-year-old female patient with RA, who had not reacted favorably to previous TNFi, experienced heart failure with reduced ejection fraction following her first dose of ozoralizumab.73
Interleukin-6 is a key target for therapies that aim to reduce the systemic and local inflammatory features of RA.74 It plays a significant role in the pathophysiology of RA by regulating the chronic inflammation that underlies both local and systemic clinical manifestations of RA through cell signaling influenced by membrane-bound and soluble IL-6 receptor (IL-6R).75,76 Significantly elevated IL-6 levels in RA, in conjunction with many growth factors mostly generated by macrophages and synovial-like fibroblasts, are essential for the clinical course of the illness. Because IL-6 is harmful and mediates inflammation and context-driven signaling, blocking it could be a powerful target for RA treatment.
By blocking both soluble and membrane-bound IL-6 receptors, tocilizumab, the first IL-6 inhibitor to be approved, lowers the inflammatory cascade. With a low risk of immunogenicity and a flexible administration method (subcutaneous or intravenous), tocilizumab can be self-administered once a week.77 Tocilizumab-treated 189 RA patients displayed notably lower levels of all disease activity indices than the control group in a prospective observational analytical analysis.78
In a Saudi Arabian single-center research, RA patients treated with tocilizumab experienced greater mean decrease in their levels of erythrocyte sedimentation proportion and C-reactive protein (CRP) than those treated with adalimumab and etanercept, although at a higher cost.79 In a multicenter, post-marketing, non-interventional, observational study of the persistence of subcutaneous tocilizumab as monotherapy or in combination with csDMARDs in RA patients, it was determined that the drug was a very safe and effective treatment option for patients with moderate-to-severe RA in Greece.80 Real-world experience from the TReasure Registry (a multicentre, web-based registry of RA and spondyloarthritis patients across Turkey), which demonstrated notable decrease in the disease activity score-28 as well as in the simplified disease activity index (SDAI), CDAI, and health assessment questionnaire (HAQ) scores, corroborated the impact of tocilizumab in managing RA with a good safety profile.81 When tocilizumab, sarilumab, and olokizumab were compared for efficacy and safety in individuals with active RA, they were discovered to be more effective than adalimumab and to have comparable efficacy and safety in RA patients who did not respond well to methotrexate.82 According to a Korean multi-center registry research involving 893 patients, tocilizumab was discovered to have a considerably lower treatment discontinuation rate than TNFi in biologic-naïve patients with RA, especially those with anemia.83 Infliximab-induced plaque psoriatic eruption of the palms and soles occurred in a 70-year-old female with a RA history. It recurred after tocilizumab relay and disappeared following tocilizumab interruption.67
After using tocilizumab for 2 years to treat RA, a 75-year-old female was diagnosed with liver dysfunction a year later. Additional investigation revealed that tocilizumab caused iron buildup.84 Following the administration of methotrexate and tocilizumab, a 59-year-old female RA patient developed medication-related osteonecrosis of the jaw.85 The safety outcomes of tocilizumab and tofacitinib in RA patients were compared using data from the Taiwan National Health Insurance Research Database. The findings indicated that the incidence proportions of other safety concerns and mortality proportions were similar in both groups, with a lower incident rate of herpes zoster in the tocilizumab group.86 Prosthetic infections were reported by RA patients treated with tocilizumab.87 In contrast to TNFi-treated and bDMARD-naïve RA patients, a Danish cohort study discovered that in a real-world setting, treating RA patients with tocilizumab/sarilumab did not increase their chance of acquiring cancer.88
In a phase 3 randomized, double-blind, active-controlled clinical trial, individuals with moderate-to-severe RA who did not respond well to methotrexate were compared with the tocilizumab reference product using the tocilizumab biosimilar BAT1806/BIIB800 for analysis of two treatment periods (weeks 24 to 48), the tocilizumab, tocilizumab to BAT1806/BIIB800, and BAT1806/BIIB800 groups were shown to have similar effectiveness, safety, immunogenicity, and pharmacokinetic profiles.89 A double-blind, randomized phase III study discovered that tocilizumab biosimilar CT-P47 or tocilizumab (8 mg/kg) given every 4 weeks to RA patients demonstrated efficacy equivalence as well as comparable pharmacokinetic, safety, and immunogenicity profiles between CT-P47 and tocilizumab, including after switching from tocilizumab to CT-P47.90 According to a randomized double-blind study, the suggested biosimilar MSB11456 and the tocilizumab reference product had the same level of safety and efficacy in patients with moderate-to-severe RA.91 According to a phase 3 randomized, multicenter, double-blind, active-controlled clinical trial, BAT1806/BIIB800 demonstrated similar safety, immunogenicity, and pharmacokinetic characteristics to cilizumab, along with equivalent effectiveness.92
Human monoclonal antibody sarilumab stops IL-6 from binding to membrane-bound and soluble IL-6R-α. It can be used to treat RA in adults either alone or in combination with csDMARDs.93,94 A 1-year PROspective sarilumab (preFILled syringe/pen) international observational (PROFILE) trial revealed that sarilumab treatment enhanced physical performance and quality of life while lowering the CDAI score. With 66.2% of patients reporting a treatment-emergent adverse event (TEAE) or serious adverse event (SAE), neutropenia was the most frequently reported TEAE of interest.95 Tocilizumab, sarilumab, and olokizumab were discovered to be more successful than adalimumab when assessed for efficacy and safety in people with active RA. They also showed comparable efficacy and safety in RA patients who did not respond well to methotrexate.82 In an interim analysis of a post-marketing surveillance for the safety and effectiveness of the medication in Japanese RA patients who had not responded to previous treatments, sarilumab was well tolerated and no new safety signals were discovered.96 A multicenter, retrospective, inverse probability of treatment-weighted analysis based on the Fukuoka RA Biologics (FRAB) Registry discovered that sarilumab’s effectiveness was unaffected by body mass index (BMI), comorbidities, methotrexate use, or the number of prior b/tsDMARDs.97 Furthermore, there were no apparent additional safety concerns in RA patients. In a Japanese population with RA, data from an interim analysis of a post-marketing monitoring study showed that sarilumab was well tolerated by RA patients, with no new safety signals, regardless of the age group.98 In contrast to TNFi-treated and bDMARD-naïve RA patients, a Danish cohort study discovered that in a real-world setting, treating RA patients with tocilizumab/sarilumab did not increase their chance of acquiring cancer.88 According to Tada et al., sarilumab was effective in treating RA without causing a recurrence of lymphoproliferative diseases, and two individuals with RA flared after stopping methotrexate because of these conditions.99
A new IL-6 inhibitor called olokizumab has shown promising results in RA patients.100 The results of a Clopidogrel for the Reduction of Events During Observation (CREDO) I, a randomized controlled trial (RCT), comparing the effectiveness of olokizumab subcutaneous injections at a dose of 64 mg every 2 and 4 weeks, versus placebo, in 428 RA patients were recently published by Nasonov et al.101 Compared to a placebo, the treatment with olokizumab (of regimens) produced clinically and statistically significant improvements in all patient-reported outcomes (PRO), including pain, exhaustion, and functional impairment, in a timely and meaningful manner.101 A total of 183 patients with moderate-to-severe RA activity were included in an open-label observational non-interventional trial. Every 4 weeks, all patients got injections of olokizumab, 64 mg, along with methotrexate. The Disease Activity Score in 28 joints (DAS28)-CRP dropped to 3.3 ± 0.9 (P < 0.001) after 6 months, and a statistically significant decrease was observed in fatigue, patient global assessment (PGA), pain intensity, and functional impairment. In 24 weeks, the percentage of patients with Central Sensitization Inventory (CSI) score > 40 dropped from 71.0% to 21.0% (P < 0.001), and the percentage of patients with PainDETECT score > 18 dropped from 21.5% to 13.2%. Steroid use dropped from 54.2% to 32.6%, while use of nonsteroidal anti-inflammatory drugs (NSAID) dropped from 70.8% to 33.8% (P < 0.001). Three patients experienced serious events, while 14.2% of patients reported adverse effects. Olokizumab is useful in managing chronic pain associated with nociceptive system dysfunction and lowering RA activity.102 According to Lee et al., olokizumab at 64 mg/kg every 2 or 4 weeks produced a prominent ACR20 response relative to placebo.103
During the COVID-19 epidemic, it was demonstrated that RA patients could safely and effectively switch from rituximab to olokizumab. TJC28 significantly decreased after 8 and 12 weeks of treatment, and TSC28 decreased after 4, 8, and 12 weeks of treatment with olokizumab, according to the findings of a 12-week open-label, non-interventional study on the safety and effectiveness of olokizumab therapy in patients with RA who had switched from rituximab during the SARS-CoV-2 pandemic.104 The safety and effectiveness of olokizumab plus methotrexate were examined over 106 weeks of treatment using pooled data from three randomized clinical trials followed by an open-label extension study. The findings demonstrated that the efficacy of olokizumab was sustained through week 106 and that the long-term safety and tolerability of olokizumabin combined with methotrexate remained stable.105 Tocilizumab, sarilumab, and olokizumab were discovered to be more effective than adalimumab and to have comparable efficacy and safety in RA patients who did not respond well to methotrexate, according to a network meta-analysis of RCTs comparing the efficacy and safety of these medications in RA patients.106
It’s still unclear how exactly B cells enhance RA pathophysiology.107 The identification of autoantibodies directed against citrullinated peptides (anti-CCPs) was linked to the resurgence of interest in B cells in RA. Because B cells also produce pro-inflammatory cytokines108 and act as antigen-presenting cells,109 reduction of B cells has been explored as a potential therapy approach for RA.
The CD20 protein on the surface of B lymphocytes carrying this antigen is the target of rituximab, a mouse/human chimeric monoclonal antibody. B cells in bone marrow and synovial tissue are somewhat depleted with rituximab treatment, while B cells in peripheral blood are temporarily but almost completely reduced.110 In patients who are not responding to traditional and other bDMARDs, rituximab, an approved and successful medication for the treatment of RA, has been demonstrated to reduce disease manifestations.111,112 Early RA patients who get repeated management with rituximab + methotrexate report better physical functioning and better clinical results at the 2-year follow-up.113,114
A single dose of either 1000-mg rituximab + 100-mg methylprednisolone and antihistamines or a placebo was administered to 78 RA-risk patients in a phase II randomized, double-blind, and placebo-controlled clinical trial. In seropositive RA-risk patients, 1000-mg rituximab demonstrated a considerable delay in the onset of arthritis by up to 12 months;115 however, there was no appreciable and noteworthy improvement in patient-reported outcomes over a 2-year follow-up period.116 A 104-week open-label multicenter randomized controlled superiority trial comparing treat-to-target fixed-dose rituximab retreatment and fixed interval retreatment with disease activity-guided rituximab dose optimization for RA patients was described by De Meyst et al.117 Compared to JAKi, IL6Ri, and TNFi, the patients with difficult-to-treat RA (D2TRA) showed a higher proportion of survival following rituximab treatment.118 According to 15 years of data from the Quebec registry RHUMADATA, rituximab seems to be the most effective course of treatment for RA patients who did not respond to a first-line TNFi.119 In clinical practice, the reduced dosage of rituximab is successful for many RA patients and results in relevant reduction in dose, according to a retrospective cohort analysis of RA patients with mild disease activity on rituximab.120
Adalimumab medication led to the diagnosis of multiple sclerosis in a 47-year-old RA patient, who subsequently experienced free manifestations after switching to rituximab.33 In an 83-year-old male, RA and anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis were successfully treated with rituximab.121 A 31-year-old female patient with RA and TNFi-induced immunoglobulin A (IgA) vasculitis was effectively treated with rituximab and methotrexate.122 The treatment (rituximab and methotrexate, and hydroxychloroquine) showed good response and resolved anasarca and albuminuria in a 45-year-old female with verified membranous nephropathy and RA.123
In RA patients receiving rituximab, a retrospective cohort study was performed to examine the incidence, timing, and dose-dependency of infections. The findings revealed that, among 490 patients, 819 infections occurred over 1254 patient years, with respiratory tract infections being the most common type of infection. Additionally, compared to 500- and 1000-mg rituximab, ultra-low dosages (200 mg) are linked to a decreased incidence of infections in RA.124 RA patients who responded to conventional doses of rituximab were discovered to benefit from long-term usage of ultralow dosages of the drug, according to the observational extension of the REDO (REtreatment with Rituximab in RhEmatoid arthritis: Disease Outcome after Dose Optimization) study.125 After 34 patients received 76 infusions, data from a Tertiary Veterans Affairs Center for the analysis of safety in RA patients with the implementation of an accelerated infusion protocol (90-min infusion) of rituximab revealed that only two infusion-related reactions were noted: one was chest pain and dyspnea, and the other was itching and sore throat.126 According to a Danish cohort research, rituximab treatment for RA patients did not raise their risk of developing cancer in a real-world context, compared to the TNFi-treated and bDMARD-naïve RA patients.88 According to a retrospective cohort research for the examination of cancer risk following the use of bDMARD or tsDMARD, rituximab was linked to a higher incidence of incident cancer than TNFis.127
T cells penetrate the synovial membrane in RA patients, where they activate synovial fibroblasts and macrophages and turn them into effector cells that damage tissues.128 For complete T-cell activation, first a specific antigen must be recognized by a T-cell receptor, followed by a co-stimulatory signal, such as the binding of cluster of differentiation (CD)80 and/or CD86 on the surface of antigen-presenting cells to the CD28 receptor on T cells. Activated T cells express cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), which binds to CD80 and CD86 more firmly and strongly than CD28, thereby acting as a negative regulator of T cell-mediated immune responses.
By preventing co-stimulation, the CTL4-Fc fusion protein abatacept prevents T-cell activation.129 Abatacept binds specifically to CD80 and CD86, thereby inhibiting T-cell activation.130 DAS28-CRP significantly decreased in 53 RA patients who received continuous abatacept medication for 3 months or a longer period, and the decline in DAS28 was age-related.131 After 274 patients received abatacept, their CDAI scores after 4, 12, 24, and 52 weeks were considerably lower than their baseline scores. At 4, 12, and 24 weeks, however, the impact of abatacept on CDAI scores was noticeably greater than that of the baricitinib group.132 According to a retrospective cohort trial, abatacept, when used as a first-line biologic for patients with RA that is anti-citrullinated protein antibody (ACPA)- and rheumatoid factor-positive, improved clinical outcomes and remission rates at 3, 6, and 12 months.133 According to a population-based cohort research, abatacept was linked to a longer medication survival period for patients with early onset as opposed to those with late onset.31 Comparing the performance of abatacept and TNFi in RA patients who are anti-citrullinated protein antibodies and shared epitope positive, analyses revealed that abatacept outperformed TNFi in terms of efficacy results.134 In order to assess the effectiveness and safety of abatacept in preclinical RA, Asif et al. implemented a systematic review and meta-analysis of RCTs.135 Their findings indicated that abatacept is a promising treatment option for delaying the onset of RA in high-risk individuals with a positive safety profile.
A 3-year efficacy and safety was validated by the interim findings of ORIGAMI, a long-term observational multicenter research conducted in Japan. Additionally, the study identified potential determinants linked to J-HAQ remission in biologic-naïve RA patients receiving abatacept in actual clinical settings.136 According to a study on the drug’s clinical effectiveness and retention in RA patients, the 24-month retention rate of abatacept was 59.9%. Abatacept was also linked to better clinical results and was well-tolerated in Taiwan’s real-world environment.137 When methotrexate is not used at the beginning of the treatment, abatacept is the most commonly used medication, followed by IL-6 receptor inhibitor, according to a retrospective hospital-based administrative claims database study for the disease-modifying antirheumatic drug selection in Japanese patients with RA treated with biologics or JAK inhibitors without methotrexate.138 A 6-month, multinational, multicenter, double-blind, placebo-controlled trial discovered that abatacept medication lowers MRI inflammation, clinical manifestations, and the risk of RA in high-risk individuals.139 The intervention’s effects last for a year-long drug-free observation period. A randomized, double-blind, multicentre, parallel, placebo-controlled, phase 2b clinical trial discovered that blocking T-cell co-stimulation with abatacept for a year slows the development of RA. Following the course of treatment, there is evidence of continued efficacy and no new safety concerns.140
Hypervirulent Klebsiella pneumoniae was detected in a 79-year-old male with RA using abatacept.141 Abatacept was used for 7 months to treat RA in a female in her seventies who was diagnosed with eruptions similar to Mycosis fungoides.142 According to a retrospective cohort study, in the first 2 years following the start of biologic or tsDMARDs, RA patients starting abatacept, rituximab, and JAKis showed higher incidence rates and statistically significant higher risks of incident cancers than those starting TNFis.127 According to a Danish cohort research, in a real-world scenario, RA patients treated with abatacept did not have higher cancer risks than those treated with TNFi or bDMARD-naïve RA patients.88 Comparing patients treated with abatacept to those treated with csDMARDs or other b/tsDMARDs, the results of a 10-year international post-marketing research did not reveal a statistically significant increase in the risk of total malignancies in pooled data.143 However, relative to csDMARDs, a thorough assessment of observational data and RCTs revealed a possible increase in the incidence of nonmelanoma skin cancer with abatacept treatment.144
tsDMARDs known as JAK inhibitors prevent certain cytokines from signaling intracellularly.145,146 TYK2, JAK1, JAK2, and JAK3 are the four JAK isoforms. While JAK3 is mostly expressed in vascular smooth muscles, endothelial, and hematopoietic cells, JAK1, JAK2, and TYK2 are present in nearly all cells. Different JAK proteins are connected to the corresponding signal transducers and transcription activators (STAT1–STAT6). Different JAK-STAT combinations determine which immunological activities are mediated and which cytokine signaling pathways these molecules are involved in.145–147 Common γ-chain cytokines, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, for instance, mediate lymphocyte growth, proliferation, and homeostasis via signaling through JAK1 and JAK3. IL-6, IL-11, IL-12, IL-27, leukemia inhibitory factor, and oncostatin M use JAK1, JAK2, and TYK2 in the development of T cells and inflammation. Common β-chain cytokines, such granulocyte, granulocyte–monocyte colony-stimulating factors, erythropoietin, thrombopoietin, IL-3, and IL-5, only entail JAK2 signaling. These cytokines are involved in myelopoiesis, erythropoiesis, thrombopoiesis, and allergy reactions. Type I interferons, which signal through JAK1 and TYK2, include IFNα and IFNβ, IL-10, IL-20, IL-22, and IL-28. They mainly mediate autoimmunity, B cell development, and innate immune responses. Finally, through IFNγ, IL-12, and IL-23 signaling, JAK1 and JAK2 contribute to inflammation and antiviral immunity.145,147 These details are important because the safety and efficacy characteristics of different JAK inhibitors may be determined by their unique cytokine inhibition patterns.145–147
A total of 18 RCTs totaling 21,432 patients and 57,040 patient-years were included in a traditional and Bayesian network meta-analysis of randomized clinical trials to report on the CV safety of JAKi or tocilizumab class in comparison to TNFi in RA patients. Findings demonstrated that, in comparison to TNFi, JAKi was associated with a non-statistically significant increase in the risk of severe adverse CV events and all-cause deaths. According to Bayesian analysis, the JAKi group had a higher clinical probability of experiencing significant adverse CV events and all-cause deaths more frequently than the TNFi group.148 JAK drugs (upadacitinib, baricitinib, tofacitinib, and filgotinib) helped 48.89% of patients reach remission and 26.67% achieve low activity in a single-center observational, descriptive, retrospective study of RA patients. There have been reports of adverse effects or consequences, including herpes zoster infection, increased transaminase levels, and minor upper respiratory tract infections (pharyngitis/pharyngotonsillitis).149 Colonization of Pneumocystis jirovecii is about six times more likely to occur in RA patients taking bDMARDs (such as TNFi [etanercept, adalimumab, and golimumab], IL-6 inhibitors [tocilizumab], CTLA-4 inhibitors [abatacept], and anti-CD20 Ab [rituximab]) or JAKi (tofacitinib, baricitinib, and upadacitinib).150
In patients with D2TRA and those without D2TRA, JAKi significantly decreased CDAI-LDA.151 JAKi effectively decreased disease activity in D2TRA patients up to the same degree as active non-D2TRA patients, and tolerability profiles were comparable, according to a real-world evaluation of the efficacy and tolerability profile of JAKi in D2TRA patients. The results were primarily influenced by the presence of age and/or CV risk factors.152
According to a propensity score-matched study comparing creatine kinase elevation brought on by JAKi and IL-6 inhibitors in RA patients, mild creatine kinase elevations with JAK inhibitors were not a specific clinical issue, and creatine kinase elevation may be unique to JAK inhibitors.153 Remission rates with JAKi were significantly greater in RA patients who did not respond well to methotrexate, according to a network meta-analysis comparing the relative remission proportions of JAK inhibitors and adalimumab in patients with active RA.154
Tofacitinib, baricitinib, upadacitinib, filgotinib, and peficitinib are the five JAK inhibitors approved for the treatment of RA. Upadacitinib and filgotinib mostly inhibit JAK1, while tofacitinib preferentially inhibits JAK1 and JAK3, and baricitinib inhibits JAK1 and JAK2, and so on.146,147
The first JAK inhibitor approved for the treatment of autoimmune illnesses was tofacitinib (JAK1/JAK3, with limited efficacy against JAK2). For this reason, maximum information is available for the medication. According to study findings, tofacitinib is safe and effective if used in conjunction with other csDMARDs to treat PsA, SpA, and RA.155,156
In comparison to the reference strategy, which included the four classes of biologics commonly used in France (TNFi, tocilizumab, abatacept, and rituximab), a multi-state Markov model analysis revealed that the introduction of tofacitinib was a dominant strategy. This strategy also produced maximum cost savings and increased quality-adjusted life period by 0.29 years.157 According to a study on the safety and effectiveness of tofacitinib in RA, 9 years of real-world data showed that the drug was consistently successful, with notable decrease in disease activity indices at 3 and 6 months after beginning tofacitinib.158
According to the Oral Surveillance clinical trial, tofacitinib was shown to have a higher risk of major adverse cardiovascular event (MACE) and malignancies (apart from nonmelanoma skin cancer [NMSC]) than TNFi. A multicenter, observational, prospective cohort trial (CANTORAL) included 504 adult Canadian patients with moderate-to-severe RA starting tofacitinib. These patients were separated into cardiovascular (CV), risk-enriched (CV+), and CV– cohorts. The findings demonstrated that CDAI-LDA and remission rates in CV+ and CV- cohorts were 51.5%/54.6% and 12.0%/19.6%, respectively, at month 6; DAS28-4(CRP) < 3.2/ < 2.6 rates were 44.0%/39.3% and 31.5%/28.8%, respectively; and the efficacy was mostly sustained through month 18. In both cohorts, the side effects included safety, TEAEs, SAEs, fatalities, serious infections, herpes zoster, MACE, malignancies (not including NMSC), NMSC, and venous thromboembolism (VTE).159
According to a retrospective research, tofacitinib in combination with bDMARDs was discovered to be both safe and effective for RA patients not responding well to bDMARDs.160 According to clinical trials and real-world data, tofacitinib monotherapy showed clinically significant responses/persistence in RCT/LTE analyses, with effectiveness and persistence comparable to combo therapy.161
According to the Tofacitinib in Rheumatoid Arthritis a Real-Life experience in Italy (ToRaRI), there were advantages in adopting tofacitinib as first-line therapy, demonstrating that the drug was safe, efficacious, and could increase remission rates in patients who were unfamiliar with bDMARDs.162 The interim analysis of post-marketing surveillance in Japan showed that disease activity improved over a 6-month period, and safety was consistent with the characteristics of RA patients receiving tofacitinib.163 Regardless of the regimen, line of therapy, period of beginning, or dose, tofacitinib efficacy (CDAI-LDA) was demonstrated in a real-world US patient scenario with RA, according to results from the CorEvitas Rheumatoid Arthritis Registry.164
According to a prospective observational analysis, the remission rates of tofacitinib and TNFi groups at 12 months did not differ significantly. Regarding safety, tofacitinib had more adverse events (AEs) than TNFi; however, both groups experienced SAEs at similar proportions.165 Tofacitinib was associated with greater proportions of PE and VTE than TNFi, and these proportions were mostly stable across time.166 Patients with RA receiving tofacitinib and baricitinib were more likely to become infected than those receiving subcutaneous bDMARDs.167 A post hoc analysis of a phase 3b/4 randomized safety study discovered no difference between tofacitinib and TNFi in terms of the risk of composite CV endpoints, which comprise all ischemic CV events and heart failure.168 Patients on tofacitinib experienced higher proportion of AEs, such as major adverse CV events and malignancies other than nonmelanoma skin cancer, than those on TNFi, according to a post hoc analysis of the ORAL surveillance trial.169 Comparing generic tofacitinib to its original manufacturer, a prospective longitudinal cohort trial in RA patients revealed that it was more cost-effective and had comparable clinical efficacy and safety.170
The safety outcomes of tofacitinib and tocilizumab in RA patients were compared using data from the Taiwan National Health Insurance Research Database. The results showed that death proportions and other safety concerns were similar in both groups, even though the tocilizumab group had fewer herpes zoster incidents.86 According to a real-world cohort research with 7580 individuals, RA patients treated with tofacitinib had a greater incidence of dyslipidemia than RA patients treated with adalimumab; nevertheless, MACE and all-cause mortality did not vary.32
One RA patient using tofacitinib was reported to have a significant lung nodule.171 A RA patient on tofacitinib had disseminated cryptococcosis that manifested as a widespread cutaneous manifestation.172 After receiving tofacitinib therapy, a 70-year-old female with RA who had bilateral hip replacements later experienced a prosthetic joint infection caused by Listeria monocytogenes.173 On the sixth day after starting tofacitinib, a 58-year-old Tunisian female with a 4-year history of RA developed severe pancreatitis.174 Tofacitinib-treated RA patients frequently develop herpes zoster, and recent usage of bDMARDs (TNFi, tocilizumab, or abatacept) increased the likelihood of developing herpes zoster.175
JAK1/JAK2 is selectively inhibited by baricitinib. Its effectiveness in RA has been revealed in a number of phase III studies.176,177 In the Mahmoud et al. trial, 334 RA patients were divided into three groups178: the first group received baricitinib (4 mg daily), the second group received TNFi (golimumab at 50 mg/month, etanercept at 50 mg/week, and adalimumab at 40 mg/2 weeks), and the third group received cDMARDs. According to the findings, at weeks 12 and 24, patients in the baricitinib group showed a substantial improvement in all end measures, including TJC, SJC, VAS, DAS28, CDAI, HAQ-DI, ACR20, ACR50, and ACR70. Baricitinib performed noticeably better than cDMARDs and on par with TNF inhibitors. Infection, GIT, and CVS problems were the most frequent adverse outcomes in the baricitinib group. However, this study was a single-center experiment; thus, multicenter studies are recommended to support the results. According to the composite measures of disease activity 28-joint count and CDAI, after roughly 6–12 months of treatment, 40.7–93.8% and 55.6–88.0% of patients achieved remission and LDA, respectively, according to real-world data analyzed by Edwards et al.179 This suggests that baricitinib monotherapy can be a suitable treatment option in routine clinical practice for patients with RA. However, several limitations were there in this study. First, the conclusions were limited by the inconsistent categories of data provided by each registry and the different trends observed across registries. Second, the number of patients with post-baseline data available for composite measures was low in each registry and the number of patients with missing data was not available. Third, the patients included in the registries reviewed may not be representative of patients currently eligible for baricitinib monotherapy, limiting the generalizability of findings. Fourth, it was unable to consider the safety of baricitinib monotherapy, as only the Erlangen baricitinib cohort data included safety information.
Results of a long-term study to assess the effectiveness of baricitinib in patients with moderate-to-severe RA up to 6.5 years of treatment showed that treatment with 4 mg or 2 mg of baricitinib was effective for up to 6.5 years, with maintained LDA/remission results across SDAI, CDAI, and DAS28-hsCRP consistent with previously published data.180 However, the included patients were fully LTE-compliant, which was one of the limitations of this descriptive study. Besides, the fact that a large proportion of patients were discontinued before the final study visit when the sponsor ended LTE because of the fulfillment of objectives, also limited this study. A retrospective cohort research comprising 78 patients, 33 of whom had D2TRA and 45 without D2TRA, discovered that baricitinib shared similar safety and efficacy profiles between these two groups despite the non-D2TRA group exhibiting higher proportions at 24 months.181 However, a limited number of patients with the retrospective analysis was the flaw of this study.
After 241 patients received baricitinib treatment, their CDAI scores at 4, 12, 24, and 52 weeks were considerably lower than their baseline scores. Furthermore, at 4, 12, and 24 weeks, baricitinib’s impact on CDAI scores was noticeably less than that of the abatacept group.132 However, this was a non-randomized observational study, and there may occur the selection bias related to different treatment periods. In addition, the registry did not include radiographic data and the relatively short follow-up period may limit the generalizability of results.
In patients with RA who were not responding to csDMARDs, the PERFECTRA (a pragmatic, multicentre, real-life) study indicated that starting with baricitinib was better than starting with TNFi in terms of response at 12 weeks and improved outcomes across all clinical measures and Patient-Reported Outcome Measures (PROMs) during the study period.182 The limited number of patients and study duration, however, were the limitations of this study.
In a Swiss cohort study of RA patients, a comparison of the efficacy of baricitinib and other bDMARDs revealed differences in clinical outcomes, with baricitinib exhibiting a considerably higher medication maintenance proportion than TNFi.183 However, this was a non-randomized study, and the average length of follow-up was short, which were the limitations of this study.
A 3-year post-marketing monitoring analysis of baricitinib was conducted on Japanese RA patients. The results showed that the 3-year persistence proportion of patients in the safety population, who received baricitinib, was 45.4% based on Kaplan–Meier analysis; 10.42% patients had SAEs, and there were 0.43% deaths. AEs included herpes zoster, serious infection, malignancy, major adverse CV events, and VTE.184 In a long-term study for the drug’s real-world effectiveness, persistence, adherence, and safety in RA conducted by Calvo-Garcia et al.,185 baricitinib exhibited efficacy, significant persistence, high adherence to treatment, and a tolerable safety profile. Therein, 15.2% patients had AEs, and 3.5% had SAEs. AEs included anemia, infection, hypercholesterolemia, abnormal liver enzymes, nausea and vomiting, alopecia, skin disorders, asthenia, weight gain, VTE, hypertriglyceridemia, rhabdomyolysis and increase in platelets. SAEs included bacterial pneumonia with intravenous treatment, cancer, abnormal liver enzymes, VTE, hypertriglyceridemia, skin disorders (urticaria), and increase in platelets. Also, baricitinib was linked to a slightly increased risk of infection and a higher risk of herpes zoster, specifically in RA patients, relative to bDMARDs.167 In a rare instance, a patient with RA receiving baricitinib was shown to have elevated creatine kinase.186
A new-generation JAKi called upadacitinib is thought to be more specific for JAK1 because it is 74 times more selective for JAK1 than for JAK2. Patients with moderate-to-severe RA who are not improving with methotrexate or anti-TNF medications have been the subjects of two multicenter randomized, double-blind, placebo-controlled phase II trials. Both studies demonstrated that upadacitinib quickly improved the DAS28 with CRP and ACR 20/50/70 response criteria, compared to a placebo. RA, SpA, and PsA patients are currently treated with upadacitinib.187,188 Overall length and size were the limitations of these two studies. Upadacitinib-treated RA patients experienced a noteworthy proportion of disease remission or LDA, according to a multicenter observational research on the medication’s effectiveness in RA.189
Barešić et al.,190 after a thorough work-up, reported a Caucasian patient from South-Eastern Europe who had developed extended eosinophilia while receiving upadacitinib for RA. Despite living in a nonendemic part of the world, the patient was diagnosed with strongyloidiasis. According to a prospective, non-randomized pilot trial that included 20 adult patients with active RA, both upadacitinib and csDMARDs lessened RA disease activity. However, those who took upadacitinib demonstrated a differential regression of erosion on high-resolution peripheral quantitative computed tomography (HR-pQCT).191 Lack of randomization and blinding, small sample size, and lack of the longer-term effect were the limitations of this study.
The 5-year benefit-risk profile for upadacitinib in RA remained good, according to a research on the medication’s efficacy and safety as well as insufficient response to csDMARDs.192 Lack of a placebo control group beyond week 12 and the possible effect of background medication during LTE are the main limitations of this study. According to the prospective observational CLOSE-UP study’s interim results, upadacitinib treatment lessened disease activity and improved patient-reported outcomes in the real world. These results were consistent with clinical trial data of Canadian patients who had previously been exposed to therapy and who were receiving upadacitinib monotherapy. Overall, the benefit-risk profile was favorable.193 However, this is an interim analysis of an ongoing trial, and long-term benefit–risk profile is still needed in the future.
Regardless of previous TNFi experience, line of therapy, or concurrent use of conventional medications, upadacitinib commencement was linked with improvements in clinical and patient-reported outcomes, according to a study from the CorEvitas registry.194 However, the generalizability of the findings, sample size, follow-up time and descriptive data were the limitations of this study. A prospective longitudinal multicentric study’s real-world experience validated upadacitinib’s effectiveness in improving clinical and ultrasonographic outcomes while exhibiting a favorable safety profile.195 Study limitations included relatively small sample size and lack of a control group for comparative analysis. It was discovered that upadacitinib effectively treats seronegative RA that is anti-PTX3 positive.196
Upadacitinib may increase CV risk, especially when taken at a dose of 30 mg, according to a systematic review and meta-analysis that included six studies with a total of 4202 respondents.197 In a study conducted by Charles-Schoeman et al.,198 discontinuation of study treatment due to AEs were rare, occurring in ≤ 2.5% patients across all treatment groups. AEs included increase in alanine aminotransferase (ALT) and aspartate aminotransferase (AST), anemia, lymphopenia, neutropenia, and elevation in creatinine and total serum creatinine phosphokinase (CPK) levels. Overall, 15-mg upadacitinib once a day for moderate-to-severe RA was shown to have a satisfactory safety profile based on integrated data spanning 6.5 years about the drug’s effects on laboratory parameters and associated AEs in RA patients. The SELECT-BEYOND study’s results over a 5-year period demonstrated that 15/30-mg upadacitinib remained effective in treating clinical and functional outcomes in RA patients, and the safety profile over that period was in line with previous study-specific and integrated evaluations of upadacitinib treatment. In all, 93% of all AEs were mild-to-moderate in severity and included infections, herpes zoster, malignancy, hepatic disorder, anemia, neutropenia, lymphopenia, CPK elevation, VTE, and COVID-19.199
The phase 3 randomized SELECT-COMPARE study’s 5-year data demonstrated that there were no new safety hazards and upadacitinib’s safety profile remained consistent with its established safety profile. The most common TEAEs were serious infection, opportunistic infection, herpes zoster, active tuberculosis, malignancy, adjudicated MACE, adjudicated VTE, adjudicated gastrointestinal perforation, renal dysfunction, anemia, lymphopenia, neutropenia, CPK elevation and hepatic disorder. At 5 years, upadacitinib produced quantitatively greater clinical responses than adalimumab. According to these findings, upadacitinib had a favorable benefit–risk profile for the long-term management of RA.200 According to the SELECT-COMPARE 5-year data, RA patients who did not respond or responded partially to the first dose of upadacitinib or adalimumab were moved to the alternative treatment by week 26. Through 228 weeks after the transition, clinically significant improvements were observed in all effectiveness measures. These benefits were largely comparable across groups, with minor numerical variations primarily supporting the switch to upadacitinib. Additionally, while the proportions of TEAEs were comparable between groups, switching to upadacitinib increased the incidence of herpes zoster, lymphopenia, and creatine phosphokinase elevation.201
Compared to patients who cycled TNFi or transitioned to an advanced medication with a different mechanism of action, RA patients who switched from TNFi to upadacitinib saw noticeably improved clinical outcomes of remission, no discomfort, and full adherence.202 According to the SELECT-EARLY randomized controlled study, upadacitinib had higher incidences of AEs, particularly in the 30-mg group, than methotrexate, but it also showed stronger clinical responses in patients with RA over the course of the 5-year trial. TEAEs in patients receiving upadacitinib included hepatic disorder, neutropenia, CPK elevation, serious infections, herpes zoster, and nonmelanoma skin cancer.203 A SELECT-BEYOND phase 3 trial evaluated the long-term sustainability of response to upadacitinib among patients with active RA refractory to biological treatments. The results showed that more than three-quarters of patients with bDMARD intolerance or inadequate response were able to attain CDAI-LDA with upadacitinib, and over half of those patients were able to maintain LDA for 240 weeks of follow-up, according to SELECT-BEYOND results of up to 5 years.204 Compared to the primary analyses at week 24, the results of the SELECT-CHOICE study for the safety and effectiveness of upadacitinib in patients with RA, who were refractory to bDMARDs, showed that efficacy responses were maintained or improved further with 15-mg upadacitinib through week 216. AEs included infection, herpes zoster, COVID-19, adjudicated gastrointestinal perforation, hepatic disorder, anemia, neutropenia, lymphopenia, CPK elevation, malignancy, lymphoma, adjudicated MACE, and adjudicated VTE.205
After 12 and 24 weeks of upadacitinib treatment, 40% and 63.6% of patients, respectively, achieved ultrasonography plus clinical remission, according to the UPAdacitinib Rheumatoid Arthritis REmission UltraSonography (UPARAREMUS) real-life study on the clinical and ultrasonographic remission in RA patients who were bio-naïve and bio-failure.206
Filgotinib inhibits JAK1 and JAK2 simultaneously; however, for JAK1, it is 30 times more selective. Filgotinib was discovered to be more effective than a placebo if used alone in patients with active RA who did not respond well to methotrexate. Two phase IIb trials, DARWIN1 and DARWIN2, supported this conclusion.207,208
In FINCH 1–3 studies, which were phase 3 RCTs, filgotinib was evaluated in patients who had not reacted well to either bDMARDs (FINCH 2) or methotrexate (FINCH 3).209,210 Each study’s primary goal was met by demonstrating that a substantially larger proportion of filgotinib-treated patients achieved the ACR20 response at week 12 (FINCH 1 and FINCH 2) or methotrexate at week 24 (FINCH 3) than those treated with either placebo.209,210 Furthermore, filgotinib treatment improved several endpoints linked to RA manifestations and indicators.209,210 Filgotinib was typically well tolerated in the clinical trial program for RA.211 Filgotinib 200 mg or 100 mg is administered de novo to patients enrolled in the ongoing long-term extension of FINCH 4 study, or they continue to receive filgotinib 200 mg or 100 mg from FINCH 1, 2, or 3 studies. At week 156, 60.2% and 54.6% of patients who received de novo filgotinib 200 mg and 100 mg, and 67.3% and 59.5% of those who continued to receive filgotinib 200 mg and 100 mg, respectively, had an ACR20. Boolean remission 1.0 was observed in 18.8% and 15.4% of patients treated with de novo filgotinib 200 mg and 100 mg at week 156, respectively, and in 21.1% and 18.5% of patients treated with Boolean 2.0 criteria. Data on effectiveness were similar for patients in FINCH 2 and FINCH 3 studies. The safety information matched the known safety profile of filgotinib. These results indicated that filgotinib 200 mg and 100 mg (continuous or de novo) in FINCH 4 demonstrated maintained efficacy up to week 156 for participants enrolled from FINCH 1, 2, or 3, with no unanticipated negative effects.212
Filgotinib 200 mg or 100 mg once daily, adalimumab 40 mg every 2 weeks, or a placebo were given to patients with active moderate-to-severe RA in FINCH 1 who did not respond well to methotrexate (n = 1755). Comparing the two filgotinib doses to a placebo at weeks 12 and 24, a greater proportion of patients achieved DAS28-CRP < 2.6, CDAI remission (≤2.8), LDA (DAS28-CRP ≤3.2 or CDAI ≤10), and ACR20/50/70 responses. Through week 52, filgotinib’s effectiveness remained on par with that of adalimumab’s effectivity. The frequency of adalimumab and filgotinib AEs was comparable. With an incidence rate of 40–53% in groups receiving active treatment, infections observed were the most frequent AEs.213
According to FINCH 4’s week 156 interim findings, Japanese patients generally tolerated well filgotinib 200 mg or 100 mg, with no new or unexpected AEs reported.214 With the exception of herpes zoster in the general population, TEAEs of interest were stable over time and comparable between filgotinib 100-mg and 200-mg dose groups, according to an integrated safety analysis of the drug in patients with moderate-to-severe RA over a treatment duration of up to 8.3 years. Additionally, a dose-dependent relationship between malignancies and all-cause mortality was suggested in patients aged ≥65 years.215
In German clinical practice, filgotinib was primarily used as monotherapy and was discovered to be both efficacious and generally well tolerated.216 Filgotinib was discovered to be safe and to have a decent efficacy profile if used alone or in combination with glucocorticoids, according to data from the Italian Group for the Study of Early Arthritis (GISEA) registry.217 The efficacy and safety of filgotinib in the treatment of RA, particularly in patients who have never used a bDMARD, were demonstrated by a real-world multicenter experience.218
A pan-JAK inhibitor called peficitinib (ASP015K) is presently in late-stage development in China219 and is approved for clinical usage in RA patients in Taiwan, South Korea, and Japan.220 A multicenter, randomized, double-blind, placebo-controlled phase 3 study discovered that peficitinib 100 mg and 150 mg were better than placebo for lessening RA manifestations, and were well tolerated in Asian patients with RA who had an inadequate response or intolerance to methotrexate.221 Long-term peficitinib treatment at a dose of 100 mg/day was usually well tolerated and, after induction therapy, maintained effectiveness through week 48, according to a long-term open-label extension study conducted in Japan, South Korea, and Taiwan (RAJ2).222 Peficitinib showed persistent efficacy in clinical remission up to week 52, according to a post hoc analysis of patients with RA in clinical remission in two Japanese phase 3 trials of the medication (RAJ3 and RAJ4). Baseline characteristics linked to CDAI remission were largely in line with earlier research using other disease-modifying antirheumatic medications.223
Juvenile idiopathic arthritis is one of the most prevalent chronic conditions affecting children. JIA is defined as chronic joint inflammation that appears in children aged <16 years and lasts for more than 6 weeks without any other discernible cause (e.g., infections, metabolic illnesses, malignancies, etc.).224 The International League of Associations for Rheumatology (ILAR) has classified JIA into a number of clinical types, such as: systemic (sJIA), polyarticular (pJIA, rheumatoid factor negative [RF-] or rheumatoid factor positive [RF+]), oligoarticular (oJIA, persistent or extended), psoriatic (psJIA), enthesitis-related arthritis (ERA), and undifferentiated types.225
Biologic drugs are used as therapeutic options in the treatment of JIA, including TNFi, IL-6 inhibitors, IL-1β inhibitors, and biologics acting directly on cellular function and/or interaction.226,227 There are five TNFis used in the treatment of JIA: etanercept, infliximab, adalimumab, golimumab, and certolizumab. Etanercept is indicated for the treatment of pJIA patients aged <2 years, PsJIA patients aged >12 years, and ERA patients aged >12 years. Infliximab is off-label for the treatment of JIA-related uveitis and pJIA based on the recommendations of the European Medicines Agency (EMA). Adalimumab is indicated for the treatment of pJIA patients aged >2 years and ERA patients aged >6 years but is off-label for the treatment of uveitis. Golimumab is recommended for the treatment of pJIA only in the experimental stage.
Tocilizumab, an IL-6 inhibitor, is indicated for the treatment of sJIA and pJIA patients aged >2 years. Three IL-1β inhibitors are used in the treatment of JIA. Among them, anakinra is off-label for the treatment of sJIA. Canakinumab is indicated for the treatment of sJIA patients aged >2 years. Two biologics acting directly on cellular function and/or interaction include abatacept and rituximab. Abatacept is indicated for the treatment of pJIA patients aged >6 years, while rituximab is off-label for the treatment of pJIA.
We have compiled the original research papers on b/tsDMARDs for the treatment of RA that were published within the past year, along with their recently introduced biosimilar compounds. The indications and mechanism action of biologics in the treatment of RA were listed in Table 1. The development of more b/tsDMARDs and adherence to the treat-to-target principle are essential for improving RA prognosis. Early treatment during the window of opportunity is also a key. More RA patients being able to employ biologic agents earlier in the course of their disease is made possible by the availability of b/tsDMARDs, which is equally significant for improving therapeutic response and the final prognosis. However, the fact that many patients do not respond well or experience AEs that outweigh the benefits should be addressed when intending these therapies for RA treatment. In addition, it should be noted that some questions, such as small sample size, lack of long-term follow-up, and potential biases, exist in these clinical studies. Future research with a larger cohort and refined methodological approaches is necessary to validate and expand upon these preliminary findings. Thus, it is important to highlight a balanced perspective by focusing the risks and limitations of these treatments. Besides, owing to lower cost, bioequivalence efficacy, and safety, exploration that biosimilars fit into the treatment paradigm for RA has prominent value, which may significantly alter treatment strategies.
Table 1 The indications and mechanism action of biologics in the treatment of RA.
Biologic | Indications | Mechanism action |
---|---|---|
TNFi | ||
Adalimumab | Rheumatoid arthritis Ankylosing spondylitis (AS) Psoriasis Psoriatic arthritis JIA Crohn’s disease (including childhood Crohn’s disease) Ulcerative colitis Hidradenitis suppurativa uveitis |
Binds to TNF-α and prevents TNF-mediated cellular inflammation |
Etanercept | Adult patients with moderately to severely active rheumatoid arthritis, when refractory to DMARDs, including methotrexate Adult patients with severe active AS, when unresponsive to conventional treatment |
Binds to either soluble or transmembrane TNF-α |
Golimumab | Adult patients with moderate-to-severely active rheumatoid arthritis, when refractory to DMARDs, including methotrexate Adult patients with active AS |
Binds to TNF-α with high affinity |
Infliximab | Rheumatoid arthritis AS Psoriatic arthritis Crohn’s disease |
Binds to both membrane-bound and soluble forms ofTNF-α with high affinity |
Certolizumab | Moderate-to-severe rheumatoid arthritis | A PEGylated, Fc-free TNFi |
Ozoralizumab | Rheumatoid arthritis | Humanized antibody against TNFα |
IL-6 inhibitors | ||
Tocilizumab | Adult patients with moderate-to-severe active rheumatoid arthritis who do not respond well to treatment with one or more TNF antagonists. | Blocking both soluble and membrane-bound IL-6 receptors |
Sarilumab | Rheumatoid arthritis | Stops IL-6 from binding to membrane-bound and soluble IL-6R-α |
Olokizumab | Rheumatoid arthritis | A humanized monoclonal antibody targeting IL-6 |
Anti-CD20 | ||
Rituximab | Rheumatoid arthritis Chronic lymphocytic leukemia Relapsed or refractory follicular central lymphoma |
Targets the CD20 protein on the surface of B lymphocytes |
CTLA-4 immunoglobulin | ||
Abatacept | Adult patients with moderate-to-severe active rheumatoid arthritis who had inadequate response to one or more DMARDs, such as methotrexate (MTX) and TNFi | Prevents T-cell activation by preventing co-stimulation |
JAK inhibitors | ||
Tofacitinib | Rheumatoid arthritis Ulcerative colitis Psoriasis |
Inhibiting JAK1/JAK3, with limited efficacy against JAK2 |
Baricitinib | Adult patients with moderate-to-severe active rheumatoid arthritis | Selectively inhibits JAK1/JAK2 |
Upadacitinib | Patients with refractory, moderate-to-severe atopic dermatitis in adults and adolescents, aged ≥12 years who have had a poor response to or are not suitable for other systemic therapies Adults with moderate-to-severe active rheumatoid arthritis who have had a poor response to or intolerance to one or more TNFi Adult patients with active psoriatic arthritis who have a poor response to or intolerance to one or more DMARDs Adult patients with moderate-to-severe active ulcerative colitis who have a poor response to one or more TNF inhibitors or who are intolerant or contraindicated |
Blocking the activity of Janus kinases in the JAK-STAT signaling pathway |
All data generated or analyzed during this study are included in this published article. The datasets used and/or analyzed in the present study are available from the corresponding author on reasonable request.
Di Liu and Guimei Yu designed the study, completed the experiment, and supervised data collection. Na Yuan analyzed and interpreted the data. Daqing Nie prepared the manuscript for publication and reviewed the draft of the manuscript. All authors had read and approved the final manuscript.
The authors stated that there was no conflict of interest to disclose.
The fund was provided by Natural Science Foundation of Jilin Province (Norman Bethune Special Program) (Grant No. 20200201433JC).
1 Figus FA, Piga M, Azzolin I, McConnell R, Iagnocco A. Rheumatoid arthritis: Extra-articular manifestations and comorbidities. Autoimmun Rev. 2021;20(4):102776. 10.1016/j.autrev.2021.102776
2 Fu J, Nogueira SV, Drongelen VV, Coit P, Ling S, Rosloniec EF, et al. Shared epitope-aryl hydrocarbon receptor crosstalk underlies the mechanism of gene-environment interaction in autoimmune arthritis. Proc Natl Acad Sci USA. 2018;115(18):4755–60. 10.1073/pnas.1722124115
3 Safiri S, Kolahi AA, Hoy D, Smith E, Bettampadi D, Mansournia MA, et al. Global, regional and national burden of rheumatoid arthritis 1990–2017: A systematic analysis of the Global Burden of Disease study 2017. Ann Rheum Dis. 2019;78(11):1463–71. 10.1136/annrheumdis-2019-215920
4 Carmona L, Cross M, Williams B, Lassere M, March L. Rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2010;24(6):733–45. 10.1016/j.berh.2010.10.001
5 Mok CC, Kwok CL, Ho LY, Chan PT, Yip SF. Life expectancy, standardized mortality ratios, and causes of death in six rheumatic diseases in Hong Kong, China. Arthritis Rheum. 2011;63(5):1182–9. 10.1002/art.30277
6 Cush JJ. Rheumatoid arthritis: Early diagnosis and treatment. Med Clin North Am. 2021;105(2):355–65. 10.1016/j.mcna.2020.10.006
7 Fraenkel L, Bathon JM, England BR, St Clair EW, Arayssi T, Carandang K, et al. 2021 American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken). 2021;73(7):924–39. 10.1002/acr.24596
8 Smolen JS, Landewé RBM, Bergstra SA, Kerschbaumer A, Sepriano A, Aletaha D, 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. 10.1136/ard-2022-223356
9 Kim H, Alten R, Avedano L, Dignass A, Gomollón F, Greveson K, et al. The future of biosimilars: Maximizing benefits across immune-mediated inflammatory diseases. Drugs. 2020;80(2):99–113. 10.1007/s40265-020-01256-5
10 Kvien TK, Patel K, Strand V. The cost savings of biosimilars can help increase patient access and lift the financial burden of health care systems. Semin Arthritis Rheum. 2022;52:151939. 10.1016/j.semarthrit.2021.11.009
11 Chingcuanco F, Segal JB, Kim SC, Alexander GC. Bioequivalence of biosimilar tumor necrosis factor-α inhibitors compared with their reference biologics: A systematic review. Ann Intern Med. 2016;165(8):565–74. 10.7326/M16-0428
12 Ascef BdO, Almeida MO, Medeiros-Ribeiro ACd, Oliveira de Andrade DC, Oliveira Junior HAd, de Soárez PC. Therapeutic equivalence of biosimilar and reference biologic drugs in rheumatoid arthritis: A systematic review and meta-analysis. JAMA Network Open. 2023;6(5):e2315872-e. 10.1001/jamanetworkopen.2023.15872
13 Fraenkel L, Bathon JM, England BR, St Clair EW, Arayssi T, Carandang K, et al. American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 2021;73(7):1108–23. 10.1002/art.41752
14 Ho CTK, Mok CC, Cheung TT, Kwok KY, Yip RML. Management of rheumatoid arthritis: 2019 updated consensus recommendations from the Hong Kong Society of Rheumatology. Clin Rheumatol. 2019;38(12):3331–50. 10.1007/s10067-019-04761-5
15 Parisi S, Bortoluzzi A, Sebastiani GD, Conti F, Caporali R, Ughi N, et al. The Italian Society for Rheumatology clinical practice guidelines for rheumatoid arthritis. Reumatismo. 2019;71(S1):22–49. 10.4081/reumatismo.2019.1202
16 Kawahito Y, Morinobu A, Kaneko Y, Kohno M, Hirata S, Kishimoto M, et al. Drug treatment algorithm and recommendations from the 2020 update of the Japan College of Rheumatology clinical practice guidelines for the management of rheumatoid arthritis-secondary publication. Mod Rheumatol. 2023;33(1):21–35. 10.1093/mr/roac017
17 Petrović-Rackov L. Evaluation of the degree of clinical rheumatoid arthritis activity based on the concentrations of cytokines TNF-alpha, IL-12, IL-15, and IL-18 in serum and synovial fluid. Vojnosanit Pregl. 2006;63(1):21–6. 10.2298/VSP0601021P
18 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(6):R211. 10.1186/ar3188
19 Emery P, Breedveld F, van der Heijde D, Ferraccioli G, Dougados M, Robertson D, et al. Two-year clinical and radiographic results with combination etanercept-methotrexate therapy versus monotherapy in early rheumatoid arthritis: A two-year, double-blind, randomized study. Arthritis Rheum. 2010;62(3):674–82. 10.1002/art.27268
20 Ruderman EM. Overview of safety of non-biologic and biologic DMARDs. Rheumatology (Oxford). 2012;51(Suppl 6):vi37–43. 10.1093/rheumatology/kes283
21 Garza-Elizondo AK, Oscherwitz M, Cervantes-Ramirez V, Salazar-Marentes E, Galarza-Delgado DA, Cardenas-de la Garza JA. Case report: Latrogenic Kaposi sarcoma secondary to Janus kinase and tumor necrosis factor-alpha inhibitors in rheumatoid arthritis. Int J Rheum Dis. 2024;27(10):e15365. 10.1111/1756-185X.15365
22 Oliveira D, Martins A, Martins F, Gomes C, Pimenta S, Vaz C, et al. Systemic lupus erythematosus induced by anti-tumor necrosis factor α therapy in inflammatory rheumatic diseases: A case series. Reumatismo. 2024;76(4):286-90. 10.4081/reumatismo.2024.1658
23 Zheng B, Liu M, Dai D, Shang Y, Dou X, Liu B, et al. Safety of TNF-α inhibitors: A real-world study based on the US FDA Adverse Event Reporting System database. Medicine (Baltimore). 2024;103(29):e39012. 10.1097/MD.0000000000039012
24 Almashali M, Alotaibi MA, Shadid A. Merkel cell carcinoma associated with TNF inhibitor therapy: A systematic review of case reports. Dermatol Online J. 2024;30(2). 10.5070/D330263575
25 Pelechas E, Voulgari PV, Drosos AA. Preclinical discovery and development of adalimumab for the treatment of rheumatoid arthritis. Expert Opin Drug Discov. 2021;16(3):227–34. 10.1080/17460441.2021.1846516
26 Tamai H, Ikeda K, Miyamoto T, Taguchi H, Kuo CF, Shin K, et al. Reduced versus maximum tolerated methotrexate dose concomitant with adalimumab in patients with rheumatoid arthritis (MIRACLE): A randomised, open-label, non-inferiority trial. Lancet Rheumatol. 2023;5(4):e215–24. 10.1016/S2665-9913(23)00070-X
27 Machado MA, Maciel AA, de Lemos LL, Costa JO, Kakehasi AM, Andrade EI, et al. Adalimumab in rheumatoid arthritis treatment: A systematic review and meta-analysis of randomized clinical trials. Rev Bras Reumatol. 2013;53(5):419–30. 10.1016/S2255-5021(13)70113-5
28 Li JB, Liu PC, Chen L, Wu R. Infiltrations of plasma cells in synovium predict inadequate response to adalimumab in rheumatoid arthritis patients. Arthritis Res Ther. 2024;26(1):186. 10.1186/s13075-024-03426-2
29 Yu Z, Gao L, Zang Y, Cheng L, Gao W, Xu Y. Adalimumab exhibits superiority over etanercept in terms of a numerically higher response rate and equivalent adverse events: A real-world finding. Immun Inflamm Dis. 2024;12(2):e1166. 10.1002/iid3.1166
30 Luo F, Gau SY, Wu YX, Liao HL, Tang F, Zhong Q, et al. Efficacy of adalimumab combined with tripterygium wilfordii Hook F in the treatment of patient with rheumatoid arthritis: A multicenter, open-label, randomized-controlled trial. Int J Rheum Dis. 2024;27(1):e15031. 10.1111/1756-185X.15031
31 Jeries H, Daood R, Hijazi B, Golan-Cohen A, Green I, Merzon E, et al. Drug survival on first biologic therapy among late-onset rheumatoid arthritis patients compared to early-onset patients: A population-based cohort study. Musculoskeletal Care. 2024;22(3):e1928. 10.1002/msc.1928
32 Ma XN, Shi MF, Wang SI, Feng W, Chen SL, Zhong XQ, et al. Risk of dyslipidemia and major adverse cardiac events with tofacitinib versus adalimumab in rheumatoid arthritis: A real-world cohort study from 7580 patients. Front Pharmacol. 2024;15:1370661. 10.3389/fphar.2024.1370661
33 Khosravi S, Zamani B, Motamed MR, Akhoundi FH. Adalimumab-induced CNS demyelination in a patient with multiple pre-existing autoimmune diseases; treatment contemplation. Mod Rheumatol Case Rep. 2024;9(1):75-8. 10.1093/mrcr/rxae069
34 Shirah B, Altwirgi S, Faridoon I, Alghamdi S. Adalimumab-induced central nervous system demyelination in a patient with rheumatoid arthritis. Neuroradiol J. 2024;37(2):234–6. 10.1177/19714009231173102
35 Weinblatt ME, Baranauskaite A, Niebrzydowski J, Dokoupilova E, Zielinska A, Jaworski J, et al. Phase III randomized study of SB5, an adalimumab biosimilar, versus reference adalimumab in patients with moderate-to-severe rheumatoid arthritis. Arthritis Rheumatol. 2018;70(1):40–8. 10.1002/art.40336
36 Fautrel B, Bouhnik Y, Salliot C, Carbonnel F, Fumery M, Bernardeau C, et al. Real-world evidence of clinical outcomes of the use of the adalimumab biosimilar SB5 in rheumatic and gastrointestinal immune-mediated inflammatory diseases: 12-Month data from the PERFUSE study. Drugs Real World Outcomes. 2024;11(4):573-91. 10.1007/s40801-024-00459-6
37 García Miguel J, Yankova Komsalova L, Mata Arnaiz C, Alegre-Sancho JJ, González Polo J, Torrente-Segarra V, et al. Clinical outcomes of switching to adalimumab biosimilar (MSB11022) in patients with rheumatoid arthritis: RESTART Spanish Registry. Curr Med Res Opin. 2024:40(8):1431–1438. 10.1080/03007995.2024.2372295
38 Colina M, Khodeir M, Rimondini R, Valentini M, Campomori F, Corvaglia S, et al. Forty-Eight-month monitoring of disease activity in patients with long-standing rheumatoid arthritis treated with TNF-α inhibitors: Time for clinical outcome prediction and biosimilar vs biologic originator performance. Clin Drug Investig. 2024;44(3):141-8. 10.1007/s40261-024-01341-7
39 Hassett B, Singh E, Mahgoub E, O’Brien J, Vicik SM, Fitzpatrick B. Manufacturing history of etanercept (Enbrel®): Consistency of product quality through major process revisions. MAbs. 2018;10(1):159–65. 10.1080/19420862.2017.1388483
40 Tichý Š, Nekvindová L, Baranová J, Vencovský J, Pavelka K, Horák P, et al. Drug survival analysis of etanercept compared with monoclonal antibody tumour necrosis factor-α inhibitors in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: A propensity score-matched analysis from the Czech ATTRA registry. Scand J Rheumatol. 2024:1–8. 10.1080/03009742.2024.2381746. Epub ahead of print.
41 Bertrand D, Joly J, Neerinckx B, Durez P, Lenaerts J, Joos R, et al. Effectiveness of methotrexate and bridging glucocorticoids with or without early introduction of a 6-month course of etanercept in early RA: Results of the 2-year, pragmatic, randomised CareRA2020 trial. RMD Open. 2024;10(3):e004535. 10.1136/rmdopen-2024-004535
42 Kassabian B, Facco M, Miscioscia A, Carraro S, Rinaldi F, Gallo P, et al. Case report: Breaking CNS immuno-privilege: TNFα-inhibitor triggers aseptic meningitis in a patient with rheumatoid arthritis. Front Immunol. 2024;15:1432360. 10.3389/fimmu.2024.1432360
43 Hosseinpour A, Haj Mohamad Ebrahim Ketabforoush A, Daneshzad E, Babanezhad Gajouti A, Hajimollarabi S. Unexpected multiple sclerosis-like symptoms in a rheumatoid arthritis patient treated with etanercept: A case report. Clin Case Rep. 2024;12(11):e9486. 10.1002/ccr3.9486
44 Robinson SE, Varman R. Ramsay Hunt syndrome associated with etanercept treatment for rheumatoid arthritis: A case report and a review of literature. Cureus. 2024;16(8):e66531. 10.7759/cureus.66531
45 Roy M, Rathore H, Roy AK. Pulmonary embolism in a patient with rheumatoid arthritis on etanercept therapy. J Community Hosp Intern Med Perspect. 2024;14(1):39–42. 10.55729/2000-9666.1294
46 Yu T, Yang C. Multifocal leukoencephalopathy in a patient medicated with etanercept and methotrexate for rheumatoid arthritis. Z Rheumatol. 2024;83(4):297–302. 10.1007/s00393-023-01430-6
47 Shih PC, Hung PC, Leong PY, Hsu JN, Yang CC, Wei JCC, et al. Incidence and risk factors of discontinuation of tofacitinib and biologic disease-modifying anti-rheumatic drugs among patients with rheumatoid arthritis: A population-based cohort study. Clin Rheumatol. 2024;43(12):3625-37. 10.1007/s10067-024-07161-6
48 Pekhenko V, Udovitskiy V, Barbukho O. Efficacy, safety and immunogenicity of the biosimilar etanercept compared to the reference formulation original etanercept in patients with rheumatoid arthritis: An open-label, randomized, comparative, multicenter study. Medicine (Baltimore). 2024;103(30):e39060. 10.1097/MD.0000000000039060
49 Sakane H, Yonemoto Y, Okamura K, Suto T, Inoue M, Mitomi H, et al. Efficacy of switching from originator etanercept to biosimilar YLB113 in real-world patients with rheumatoid arthritis: A retrospective 12 months follow-up study. J Orthop Surg (Hong Kong). 2024;32(2):10225536241265818. 10.1177/10225536241265818
50 Okazaki S, Shirai T, Sato H, Ishii T, Fujii H. Development of anti-MDA5 autoantibody-positive dermatomyositis following the use of etanercept biosimilar in rheumatoid arthritis. Mod Rheumatol Case Rep. 2024;8(2):296-301. 10.1093/mrcr/rxae013
51 Kawakami T, Masui S, Onishi A, Onizawa H, Fujii T, Murakami K, et al. Comparison of safety and effectiveness between etanercept biosimilar LBEC0101 and reference in patients with rheumatoid arthritis in real-world data using the KURAMA cohort. Mod Rheumatol. 2024;34(6):1135-41. 10.1093/mr/roae018
52 Deakin CT, Littlejohn GO, Griffiths H, Ciciriello S, O’Sullivan C, Smith T, et al. Comparative effectiveness of etanercept originator and biosimilar for treating rheumatoid arthritis: implications for cost-savings. Intern Med J. 2024;54(5):795-801. 10.1111/imj.16296
53 Deeks ED. GP2015: An etanercept biosimilar. BioDrugs. 2017;31(6):555–8. 10.1007/s40259-017-0246-1
54 Schmalzing M, Kellner H, Askari A, De Toro Santos J, Vazquez Perez-Coleman JC, Foti R, et al. Real-world effectiveness and safety of SDZ ETN, an etanercept biosimilar, in patients with rheumatic diseases: Final results from multi-country COMPACT study. Adv Ther. 2024;41(1):315-30. 10.1007/s12325-023-02706-8
55 Kearsley-Fleet L, Rokad A, Tsoi MF, Zhao SS, Lunt M, Watson KD, et al. Outcomes following switching from etanercept originator to etanercept biosimilar in 1024 patients with RA: A matched-analysis of the BSRBR-RA. Rheumatology (Oxford). 2024;63(8):2082–92. 10.1093/rheumatology/kead470
56 Matsubara T, Katayama K, Sagawa A, Yoshida M, Mitsuka T, Hashimoto K, et al. Investigation of treatment continuity, usefulness, and nocebo effect in switching from the original etanercept to its biosimilar in patients with rheumatoid arthritis: A JET observational study in Japanese clinical practice. Mod Rheumatol. 2024;34(2):307–12. 10.1093/mr/road027
57 Melo AT, Campanilho-Marques R, Fonseca JE. Golimumab (anti-TNF monoclonal antibody): Where we stand today. Hum Vaccin Immunother. 2021;17(6):1586–98. 10.1080/21645515.2020.1836919
58 Husni ME, Deodhar A, Schwartzman S, Chakravarty SD, Hsia EC, Leu JH, et al. Pooled safety results across phase 3 randomized trials of intravenous golimumab in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis. Arthritis Res Ther. 2022;24(1):73. 10.1186/s13075-022-02753-6
59 D’Angelo S, Tirri E, Giardino AM, De Rosa T, Matucci-Cerinic M, Dagna L, et al. Real-world effectiveness and persistence of golimumab as second-line anti-TNFα drug in rheumatoid arthritis, psoriatic arthritis, and axial spondyloarthritis patients in Italy: GO-BEYOND, a 12-month prospective observational study. Int J Rheum Dis. 2024;27(3):e15091. 10.1111/1756-185X.15091
60 Govoni M, Batalov A, Boumpas DT, D’Angelo S, De Keyser F, Flipo RM, et al. Real-world effectiveness of golimumab in the treatment of patients with active rheumatoid arthritis, psoriatic arthritis, or axial spondyloarthritis who failed initial TNF-α inhibitor therapy: A pooled analysis of European prospective observational study. Clin Exp Rheumatol. 2024;42(3):642–50. 10.55563/clinexprheumatol/7sr1ni
61 Asano T, Ishii Y, Tsuchiya H, Masuda J. Efficacy of golimumab in rheumatoid arthritis patients at high risk of a poor prognosis: Post hoc analysis of GO-FORTH study using cluster analysis. Mod Rheumatol. 2024;34(6):1125–34. 10.1093/mr/roae037
62 Xia W, Zhou L, Gao W, Zhang Y, Si F, Bai F, et al. Patient self-reported experience and satisfaction with golimumab and etanercept treatments for rheumatic diseases: A cohort study. Medicine (Baltimore). 2024;103(8):e36982. 10.1097/MD.0000000000036982
63 Yanagihara T, Oka Y, Moriwaki A, Moriuchi Y, Ogata H, Ishimatsu A, et al. A case of pneumocystis pneumonia developed during rheumatoid arthritis treatment with methotrexate and golimumab. Cureus. 2024;16(1):e52944. 10.7759/cureus.52944
64 Baker JF, Bakewell C, Dikranian A, Lam G, O’Brien J, Moore PC, et al. Characteristics and 6-month outcomes in patients with rheumatoid arthritis initiating infliximab biosimilar IFX-dyyb in a real-world setting. Rheumatol Ther. 2024;11(3):841-53. 10.1007/s40744-024-00653-6
65 Jourdain H, Hoisnard L, Sbidian E, Zureik M. Persistence and safety of anti-TNF biosimilars versus originators in immune-mediated inflammatory diseases: An observational study on the French National Health Data System. RMD Open. 2024;10(1):e003531. 10.1136/rmdopen-2023-003531
66 Gan H, Ren X, Zou Y, Li L, Ding J, Peng L, et al. Rheumatoid arthritis complicated with cervical actinomycosis and ureteral obstruction: A case report and literature review. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2024;49(5):818–24.
67 Nielly H, Bialé L, Gilardin L, Carmoi T, Éon A, Vanquaethem H, et al. Tocilizumab-induced psoriatic eruption : A case report and a case-based review. Rheumatol Int. 2024;44(10):2205–12. 10.1007/s00296-024-05663-9
68 Bouladi M, Ben Othmen A, Sawssen M, Kalou M, Laater A, El Fekih L. New onset of uveitis during infliximab treatment: A case report. Tunis Med. 2024;102(1):58–60. 10.62438/tunismed.v102i1.4564
69 Martinez Perez P, Hanna L, Jaynes E, Gwiggner M. Infliximab rescue therapy in a case of severe granulomatous colitis associated with rituximab use. BMJ Case Rep. 2024;17(2):e257729. 10.1136/bcr-2023-257729.
70 Fukumoto K, Watanabe R, Tsutsumi M, Takakuwa T, Miyamoto M, Hayashi N, et al. Lymphoproliferative disorder progressing after partial remission following immunosuppressive drugs withdrawal in a patient with rheumatoid arthritis. Mod Rheumatol Case Rep. 2024;8(2):243-8. 10.1093/mrcr/rxae007
71 Kameda H, Nishida K, Nanki T, Watanabe A, Oshima Y, Momohara S. Safety and effectiveness of certolizumab pegol in Japanese patients with rheumatoid arthritis: Up to 3-year results from a postmarketing surveillance study. Mod Rheumatol. 2024;34(6):1103-14. 10.1093/mr/roae019
72 Tanaka Y, Miyazaki Y, Kawanishi M, Yamasaki H, Takeuchi T. Long-term safety and efficacy of anti-TNF multivalent VHH antibodies ozoralizumab in patients with rheumatoid arthritis. RMD Open. 2024;10(3):e004480. 10.1136/rmdopen-2024-004480
73 Ito T, Miyoshi Y. Acute heart failure with reduced ejection fraction following ozoralizumab in a patient with rheumatoid arthritis: A case report. Mod Rheumatol Case Rep. 2024;9(1):1-5. 10.1093/mrcr/rxae053
74 Srivastava S, Rasool M. Underpinning IL-6 biology and emphasizing selective JAK blockade as the potential alternate therapeutic intervention for rheumatoid arthritis. Life Sci. 2022;298:120516. 10.1016/j.lfs.2022.120516
75 Favalli EG. Understanding the role of interleukin-6 (IL-6) in the joint and beyond: A comprehensive review of IL-6 inhibition for the management of rheumatoid arthritis. Rheumatol Ther. 2020;7(3):473–516. 10.1007/s40744-020-00219-2
76 Hennigan S, Kavanaugh A. Interleukin-6 inhibitors in the treatment of rheumatoid arthritis. Ther Clin Risk Manag. 2008;4(4):767–75. 10.2147/TCRM.S3470
77 Hsieh PH, Wu O, Geue C, McIntosh E, McInnes IB, Siebert S. Economic burden of rheumatoid arthritis: A systematic review of literature in biologic era. Ann Rheum Dis. 2020;79(6):771–7. 10.1136/annrheumdis-2019-216243
78 Jocić J, Lučić AT, Jovanović I, Jurišević M, Stanisavljević I, Stamenković B, et al. Interleukin 41 as a potential predictor of bio-therapy efficacy in patients with rheumatoid arthritis: A prospective observational study. Int J Med Sci. 2024;21(13):2518–24. 10.7150/ijms.98752
79 Albahdal AS, Alotaibi AM, Alanazi MA, Abanmy N, Alwhaibi M, AlRuthia Y. Cost-consequence analysis of tocilizumab versus adalimumab and etanercept among rheumatoid arthritis patients in Saudi Arabia: A single-center study. Cost Eff Resour Alloc. 2024;22(1):14. 10.1186/s12962-024-00522-7
80 Athanassiou P, Katsimbri P, Bounas A, Gazi S, Sarikoudis T, Syrigou V, et al. Persistence on subcutaneous tocilizumab as monotherapy or in combination with synthetic disease-modifying anti-rheumatic drugs in rheumatoid arthritis patients in Greece (EMBRACE study): A multicenter, post-marketing, non-interventional, observational trial. Clin Rheumatol. 2024;43(5):1469-78. 10.1007/s10067-024-06916-5
81 Karadag O, Farisogullari B, Yagiz B, Erden A, Ademoglu Z, Kimyon G, et al. Tocilizumab as a first line biologic agent in rheumatoid arthritis patients with inadequate response to disease-modifying anti-rheumatic drugs: Real life experience from the TReasure Registry. Clin Exp Rheumatol. 2024;42(1):130-7. 10.55563/clinexprheumatol/2h6ma1
82 Ho Lee Y, Gyu Song G. Comparison of the efficacy and safety of tocilizumab, sarilumab, and olokizumab in patients with active rheumatoid arthritis: A network meta-analysis of randomized controlled trials. Z Rheumatol. 2024;83(Suppl 1):97-106. 10.1007/s00393-022-01315-0
83 Kim JG, Koo BS, Lee JH, Yoon BY. Anemia as an indicator of a higher retention rate for tocilizumab versus tumor necrosis factor inhibitors in patients with rheumatoid arthritis from a Korean multi-center registry. J Rheum Dis. 2024;31(4):212–22. 10.4078/jrd.2024.0026
84 Harada M, Honma Y, Shiba E, Tomosugi N, Harada R. Tocilizumab, a humanized anti-interleukin-6 receptor antibody, induces hepatic iron overload in a susceptible patient. Intern Med. 2024. 10.2169/internalmedicine.4329-24
85 Seki K, Kobayashi A, Takahashi N, Kamimoto A. Medication-related osteonecrosis of the jaw in a patient with rheumatoid arthritis with suspected involvement of methotrexate and tocilizumab. J Dent Sci. 2024;19(4):2428–9. 10.1016/j.jds.2024.07.024
86 Fang YF, Chang SH, Kuo CF, See LC. Safety outcomes of tocilizumab and tofacitinib treatment for rheumatoid arthritis: Target trial emulation. Int J Rheum Dis. 2024;27(11):e15406. 10.1111/1756-185X.15406
87 Louters A, Van Oosterhout M, Burgemeister L, Jessurun NT, Van Lint JA. Prosthetic infections following tocilizumab treatment in patients with rheumatoid arthritis. Rheumatology (Oxford). 2024;63(10):e290-1. 10.1093/rheumatology/keae228
88 Westermann R, Cordtz RL, Duch K, Mellemkjaer L, Hetland ML, Magnussen B, et al. Cancer risk with tocilizumab/sarilumab, abatacept, and rituximab treatment in patients with rheumatoid arthritis: A Danish cohort study. Rheumatology (Oxford). 2024. 10.1093/rheumatology/keae140
89 Leng X, Leszczyński P, Jeka S, Liu S, Liu H, Miakisz M, et al. A phase 3, randomized, double-blind, active-controlled clinical trial to compare BAT1806/BIIB800, a tocilizumab biosimilar, with tocilizumab reference product in participants with moderate-to-severe rheumatoid arthritis with inadequate response to methotrexate: Treatment period 2 analysis (week 24 to week 48). Arthritis Res Ther. 2024;26(1):157. 10.1186/s13075-024-03375-w
90 Smolen JS, Trefler J, Racewicz A, Jaworski J, Zielińska A, Krogulec M, et al. Efficacy and safety of CT-P47 versus reference tocilizumab: 32-Week results of a randomised, active-controlled, double-blind, phase III study in patients with rheumatoid arthritis, including 8 weeks of switching data from reference tocilizumab to CT-P47. RMD Open. 2024;10(4):e004514. 10.1136/rmdopen-2024-004514
91 Zubrzycka-Sienkiewicz A, Klama K, Ullmann M, Petit-Frere C, Baker P, Monnet J, et al. Comparison of the efficacy and safety of a proposed biosimilar MSB11456 with tocilizumab reference product in subjects with moderate-to-severe rheumatoid arthritis: Results of a randomised double-blind study. RMD Open. 2024;10(1):e003596. 10.1136/rmdopen-2023-003596
92 Leng X, Leszczyński P, Jeka S, Liu SY, Liu H, Miakisz M, et al. Comparing tocilizumab biosimilar BAT1806/BIIB800 with reference tocilizumab in patients with moderate-to-severe rheumatoid arthritis with an inadequate response to methotrexate: A phase 3, randomised, multicentre, double-blind, active-controlled clinical trial. Lancet Rheumatol. 2024;6(1):e40–50. 10.1016/S2665-9913(23)00237-0
93 Salvarani C, Cantini F, Niccoli L, Macchioni P, Consonni D, Bajocchi G, et al. Acute-phase reactants and the risk of relapse/recurrence in polymyalgia rheumatica: A prospective followup study. Arthritis Rheum. 2005;53(1):33–8. 10.1002/art.20901
94 van der Geest KS, Abdulahad WH, Rutgers A, Horst G, Bijzet J, Arends S, et al. Serum markers associated with disease activity in giant cell arteritis and polymyalgia rheumatica. Rheumatology (Oxford). 2015;54(8):1397–402. 10.1093/rheumatology/keu526
95 Kivitz A, Gottenberg JE, Bergman M, Qiu C, van Hoogstraten H, de Nijs R, et al. Real-world effectiveness of sarilumab in RA: Results from the open-label, prospective, single-arm observational PROFILE study. Rheumatol Ther. 2024;11(6):1533-50. 10.1007/s40744-024-00715-9
96 Kameda H, Tasaka S, Takahashi T, Suzuki K, Soeda N, Tanaka Y. Safety and effectiveness of sarilumab in Japanese patients with rheumatoid arthritis refractory to previous treatments: An interim analysis of a post-marketing surveillance. Mod Rheumatol. 2024;34(3):444-52. 10.1093/mr/road055
97 Harada H, Kondo M, Maeyama A, Fukuda T, Ikemura S, Shono E, et al. Effectiveness and safety of sarilumab in patients with rheumatoid arthritis: A multicenter, retrospective, inverse probability of treatment-weighted analysis based on the FRAB-registry. Clin Rheumatol. 2024;43(5):1447-59. 10.1007/s10067-024-06907-6
98 Kameda H, Tasaka S, Takahashi T, Suzuki K, Soeda N, van Hoogstraten H, et al. Safety of sarilumab in a Japanese population with rheumatoid arthritis by age group: Data from an interim analysis of a post-marketing surveillance study. Mod Rheumatol. 2024;35(1):42-9. 10.1093/mr/roae051
99 Tada Y, Maeyama A, Hagio T, Sakai M, Maruyama A, Yamamoto T. Usefulness of sarilumab in patients with rheumatoid arthritis after regression of lymphoproliferative disorders. Case Rep Rheumatol. 2023;2023:5780733. 10.1155/2023/5780733
100 Smolen JS, Feist E, Fatenejad S, Grishin SA, Korneva EV, Nasonov EL, et al. Olokizumab versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med. 2022;387(8):715–26. 10.1056/NEJMoa2201302
101 Nasonov EL, Lisitsyna TA, Zonova EV, Kuzkina S. The effect of olokizumab on rheumatoid arthritis patient’s reported outcomes: Results of a double-blind randomized placebo-controlled multicenter phase III trial (CREDO 1). Rheumatol Sci Pract. 2021. 10.47360/1995-4484-2021-62-69
102 Karateev A, Polishchuk EY, Filatova ES, Amirdzhanova VN, Khlaboshina VN, Lila A, et al. Olokizumab effect on chronic pain in rheumatoid arthritis: Results of the PROLOGUE observational study. Int J Rheum Dis. 2024;27(10):e15320. 10.1111/1756-185X.15320
103 Lee YH, Song GG. Comparison of the efficacy and safety of olokizumab at different dosages in patients with active rheumatoid arthritis: A network meta-analysis of randomized controlled trials. Z Rheumatol. 2024;83(Suppl 1):107–14. 10.1007/s00393-023-01367-w
104 Akimova AA, Banshchikova NE, Sizikov AE, Mullagaliev AA, Letyagina EA, Ilina NA, et al. Results of a 12-week open-label, non-interventional study of the efficacy and safety of olokizumab therapy in patients with rheumatoid arthritis after switching from anti-B-cell therapy during the SARS-CoV-2 pandemic. Dokl Biochem Biophys. 2024;518(1):291-9. 10.1134/S1607672924701060
105 Feist E, Fleischmann RM, Fatenejad S, Bukhanova D, Grishin S, Kuzkina S, et al. Olokizumab plus methotrexate: safety and efficacy over 106 weeks of treatment. Ann Rheum Dis. 2024;83(11):1454-64. 10.14412/1996-7012-2024-6-21-35
106 Ho Lee Y, Gyu Song G. Comparison of the efficacy and safety of tocilizumab, sarilumab, and olokizumab in patients with active rheumatoid arthritis: A network meta-analysis of randomized controlled trials. Z Rheumatol. 2024;83(Suppl 1):97–106. 10.1007/s00393-022-01315-0
107 Panayi GS. B cells: A fundamental role in the pathogenesis of rheumatoid arthritis? Rheumatology (Oxford). 2005; 44(Suppl 2):ii3–ii7. 10.1093/rheumatology/keh616
108 Marston B, Palanichamy A, Anolik JH. B cells in the pathogenesis and treatment of rheumatoid arthritis. Curr Opin Rheumatol. 2010;22(3):307–15. 10.1097/BOR.0b013e3283369cb8
109 Takemura S, Klimiuk PA, Braun A, Goronzy JJ, Weyand CM. T cell activation in rheumatoid synovium is B cell dependent. J Immunol. 2001;167(8):4710–8. 10.4049/jimmunol.167.8.4710
110 Mok CC. Rituximab for the treatment of rheumatoid arthritis: An update. Drug Des Devel Ther. 2013;8:87–100. 10.2147/DDDT.S41645
111 Cohen SB, Emery P, Greenwald MW, Dougados M, Furie RA, Genovese MC, et al. Rituximab for rheumatoid arthritis refractory to anti-tumor necrosis factor therapy: Results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial evaluating primary efficacy and safety at twenty-four weeks. Arthritis Rheum. 2006;54(9):2793–806. 10.1002/art.22025
112 Emery P, Fleischmann R, Filipowicz-Sosnowska A, Schechtman J, Szczepanski L, Kavanaugh A, et al. The efficacy and safety of rituximab in patients with active rheumatoid arthritis despite methotrexate treatment: Results of a phase IIB randomized, double-blind, placebo-controlled, dose-ranging trial. Arthritis Rheum. 2006;54(5):1390–400. 10.1002/art.21778
113 Rigby W, Ferraccioli G, Greenwald M, Zazueta-Montiel B, Fleischmann R, Wassenberg S, et al. Effect of rituximab on physical function and quality of life in patients with rheumatoid arthritis previously untreated with methotrexate. Arthritis Care Res (Hoboken). 2011;63(5):711–20. 10.1002/acr.20419
114 Tak PP, Rigby W, Rubbert-Roth A, Peterfy C, van Vollenhoven RF, Stohl W, et al. Sustained inhibition of progressive joint damage with rituximab plus methotrexate in early active rheumatoid arthritis: 2-Year results from the randomised controlled trial IMAGE. Ann Rheum Dis. 2012;71(3):351–7. 10.1136/annrheumdis-2011-200170
115 Gerlag DM, Safy M, Maijer KI, Tang MW, Tas SW, Starmans-Kool MJF, et al. Effects of B-cell directed therapy on the preclinical stage of rheumatoid arthritis: The PRAIRI study. Ann Rheum Dis. 2019;78(2):179–85. 10.1136/annrheumdis-2017-212763
116 Frazzei G, Cramer SHM, Landewé RBM, Maijer KI, Gerlag DM, Tak PP, et al. The effect of rituximab on patient reported outcomes in the preclinical phase of rheumatoid arthritis: 2 Year data from the PRAIRI study. RMD Open. 2024;10(4):e004622. 10.1136/rmdopen-2024-004622
117 De Meyst E, Bertrand D, Joly J, Doumen M, Marchal A, Thelissen M, et al. Treat-to-target fixed dose rituximab retreatment versus fixed interval retreatment with disease activity-guided rituximab dose optimisation for patients with rheumatoid arthritis: Study protocol for a multicentre randomised controlled superiority trial focusing on long-term disease impact (RITUXERA). Trials. 2024;25(1):681. 10.1186/s13063-024-08542-7
118 Novella-Navarro M, Ruiz-Esquide V, López-Juanes N, Chacur CA, Monjo-Henry I, Nuño L, et al. Subsequent biologic and targeted synthetic disease modifying anti rheumatic drugs after fulfilling difficult-to-treat rheumatoid arthritis criteria: A survival analysis. Clin Rheumatol. 2024;43(9):2817-23. 10.1007/s10067-024-07070-8
119 Choquette D, Haraoui B, Movahedi M, Bessette L, Sauvageau LC, Ferdinand I, et al. Which advanced treatment should be used following the failure of a first-line anti-TNF in patients with rheumatoid arthritis? 15 Years of evidence from the Quebec registry RHUMADATA. Rheumatology (Oxford). 2024. 10.1093/rheumatology/keae283
120 Wientjes MHM, van Huissteden J, van Herwaarden N, Verhoef LM, den Broeder AA. Dose reduction of rituximab in clinical practice: A retrospective cohort study of rheumatoid arthritis patients in low disease activity on rituximab. Clin Exp Rheumatol. 2024;42(11):2215-20. 10.55563/clinexprheumatol/lrctjp
121 Santos Faria M, Godinho I, Nogueira E, Tavares J, Carvalho D, Ponte C. Rheumatoid arthritis and ANCA-associated vasculitis–An unusual overlap successfully treated with rituximab. ARP Rheumatol. 2024;3(3):248–9. 10.63032/ZTDH8563
122 Janda P, Sączek A, Milan-Ciesielska K, Dolipska J, Kraśniak A, Krzanowski M. IgA vasculitis with Koebner’s phenomenon and renal involvement induced by anti-TNF-α therapy in the treatment of rheumatoid arthritis. Pol Arch Intern Med. 2025;135:16854. 10.20452/pamw.16854
123 Kumar S, Singh SK, Arora R, Kashif AW. Membranous nephropathy in a treatment naïve patient of rheumatoid arthritis. Indian J Nephrol. 2024;34(5):543–4. 10.25259/IJN_40_2024
124 Veeken LD, Opdam MAA, Verhoef LM, Popa C, van Crevel R, den Broeder AA. Infection incidence, timing and dose dependency in rheumatoid arthritis patients treated with rituximab: a retrospective cohort study. Rheumatology (Oxford). 2024;63(5):1246–50. 10.1093/rheumatology/kead328
125 den Broeder N, Verhoef L, de Man YA, Kok MR, Thurlings R, van der Weele W, et al. Long-term clinical and radiological effectiveness and safety of ultralow doses of rituximab in rheumatoid arthritis: observational extension of the REDO trial. RMD Open. 2024;10(2):e003659. 10.1136/rmdopen-2023-003659
126 Nahra V, Panning D, Anthony D, Mattar M. Implementation of an accelerated infusion protocol (90-minute infusion) of rituximab and its safety in patients with autoimmune rheumatic diseases at a tertiary veterans affairs center. Cureus. 2024;16(5):e60558. 10.7759/cureus.60558
127 Sendaydiego X, Gold LS, Dubreuil M, Andrews JS, Reid P, Liew DFL, et al. Use of biologic or targeted synthetic disease-modifying antirheumatic drugs and cancer risk. JAMA Netw Open. 2024;7(11):e2446336. 10.1001/jamanetworkopen.2024.46336
128 Weyand CM, Bryl E, Goronzy JJ. The role of T cells in rheumatoid arthritis. Arch Immunol Ther Exp (Warsz). 2000;48(5):429–35. PMid: 11140470
129 Bonelli M, Scheinecker C. How does abatacept really work in rheumatoid arthritis? Curr Opin Rheumatol. 2018;30(3):295–300. 10.1097/BOR.0000000000000491
130 Walker LS, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol. 2011;11(12):852–63. 10.1038/nri3108
131 Jiang H, Tang S, Li Y, Wang J, Ying Z. Clinical efficacy study of abatacept in the treatment of rheumatoid arthritis patients in China. Int J Rheum Dis. 2024;27(10):e15300. 10.1111/1756-185X.15300
132 Asai S, Takahashi N, Terabe K, Yoshioka Y, Kojima T, Kobayakawa T, et al. Clinical effectiveness of baricitinib and abatacept in patients with rheumatoid arthritis. Int J Rheum Dis. 2024;27(11):e15414. 10.1111/1756-185X.15414
133 Balanean A, Brown-Bickerstaff C, Klink A, Patel V, Zheng H, N’Dri L, et al. Real-world clinical outcomes and rationale for initiating abatacept as a first-line biologic for patients with anticitrullinated protein antibody-and rheumatoid factor-positive rheumatoid arthritis. J Comp Eff Res. 2024:13(12):e230144. 10.57264/cer-2023-0144.
134 Harrold LR, Wittstock K, Kelly S, Han X, Zhuo J, Schrader A, et al. Comparative effectiveness of abatacept versus TNF inhibitors in rheumatoid arthritis patients who are ACPA and shared epitope positive. Adv Rheumatol. 2024;64(1):10. 10.1186/s42358-024-00352-4
135 Asif M, Asif A, Rahman UA, Haseeb A, Jafar U, Farooq H. Efficacy and safety of abatacept in preclinical rheumatoid arthritis: A systematic review and meta-analysis of randomized controlled trials. Semin Arthritis Rheum. 2024;69:152562. 10.1016/j.semarthrit.2024.152562
136 Misaki K, Tanaka E, Inoue E, Tamura N, Hirano F, Taniguchi Y, et al. Factors contributing to the improvement in J-HAQ after 3 years of treatment with abatacept in biologic-naïve rheumatoid arthritis patients: Interim results of a long-term, observational, multicentre study in Japan (ORIGAMI). Mod Rheumatol. 2024;35(1):17-26. 10.1093/mr/roae043
137 Chen KH, Li KJ, Fang YF, Hsieh SC, Chen YC, Lee CS, et al. Abatacept retention and clinical effectiveness in patients with rheumatoid arthritis in a real-world setting in Taiwan. Int J Rheum Dis. 2024;27(7):e15199. 10.1111/1756-185X.15199
138 Miyakawa N, Tsuritani K, Teixeira BC, Fujio K. Disease-modifying anti-rheumatic drug selection in Japanese patients with rheumatoid arthritis treated with biologics or JAK inhibitors without methotrexate: A retrospective hospital-based administrative claims database study. Mod Rheumatol. 2024;34(5):900-9. 10.1093/mr/roae021
139 Rech J, Tascilar K, Hagen M, Kleyer A, Manger B, Schoenau V, et al. Abatacept inhibits inflammation and onset of rheumatoid arthritis in individuals at high risk (ARIAA): A randomised, international, multicentre, double-blind, placebo-controlled trial. Lancet. 2024;403(10429):850-9. 10.1016/S0140-6736(23)02650-8
140 Cope AP, Jasenecova M, Vasconcelos JC, Filer A, Raza K, Qureshi S, et al. Abatacept in individuals at high risk of rheumatoid arthritis (APIPPRA): A randomised, double-blind, multicentre, parallel, placebo-controlled, phase 2B clinical trial. Lancet. 2024;403(10429):838-49. 10.1016/S0140-6736(23)02649-1
141 Sunaga A, Inoue T. Hypervirulent Klebsiella pneumoniae in rheumatoid arthritis on abatacept. J Clin Rheumatol. 2025;31(1):e4. 10.1097/RHU.0000000000002167
142 Hoshina D, Nagata Y. Mycosis fungoides-like eruptions associated with abatacept. Australas J Dermatol. 2024;65(3):e77-8. 10.1111/ajd.14249
143 Simon TA, Suissa S, Boers M, Hochberg MC, Skovron ML, Askling J, et al. Malignancy outcomes in patients with rheumatoid arthritis treated with abatacept and other disease-modifying antirheumatic drugs: Results from a 10-year international post-marketing study. Semin Arthritis Rheum. 2024:64:152240. 10.1016/j.semarthrit.2023.152240
144 Simon TA, Dong L, Suissa S, Michaud K, Pedro S, Hochberg M, et al. Abatacept and nonmelanoma skin cancer in patients with rheumatoid arthritis: A comprehensive evaluation of randomised controlled trials and observational studies. Ann Rheum Dis. 2024;83(2):177-83. 10.1136/ard-2023-224356
145 O’Shea J J. Targeting the Jak/STAT pathway for immunosuppression. Ann Rheum Dis. 2004;63(Suppl 2):ii67–ii71. 10.1136/ard.2004.028290
146 Fleischmann R. Novel small-molecular therapeutics for rheumatoid arthritis. Curr Opin Rheumatol. 2012;24(3):335–41. 10.1097/BOR.0b013e32835190ef
147 Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–43. 10.1038/nrrheum.2017.23
148 Pugliesi A, Oliveira DGC, Filho VAS, Machado JO, Pereira AG, Bichuette JCS, et al. Cardiovascular safety of the class of JAK inhibitors or tocilizumab compared with TNF inhibitors in patients with rheumatoid arthritis: Systematic review and a traditional and Bayesian network meta-analysis of randomized clinical trials. Semin Arthritis Rheum. 2024;69:152563. 10.1016/j.semarthrit.2024.152563
149 Ramirez Huaranga MA, Calvo Pascual LA, Velasco Sanchez D, Martin de la Sierra Lopez L, Jimenez Rodriguez L, Lopez Menchero Mora A, et al. Real-world experience with janus kinase inhibitors in immune-mediated diseases: Clinical experience of a university hospital. Cureus. 2024;16(8):e67729. 10.7759/cureus.67729
150 Huang YC, Lee NY, Weng MY. Increased risk of Pneumocystis jirovecii colonization in rheumatoid arthritis patients on biologics and Janus kinase inhibitor. J Microbiol Immunol Infect. 2025;58(1):112-9. 10.1016/j.jmii.2024.08.013
151 Hayashi S, Nakano N, Tsubosaka M, Kamenaga T, Kuroda Y, Matsumoto T, et al. Real-world study comparing the efficacy of Janus kinase inhibitors in patients with difficult-to-treat rheumatoid arthritis. Clin Rheumatol. 2024;43(11):3285-92. 10.1007/s10067-024-07117-w
152 Al Tabaa O, Hecquet S, Thomas M, Carvès S, Combier A, Miceli-Richard C, et al. Real-world assessment of the efficacy and tolerability profile of JAK inhibitors in difficult-to-treat rheumatoid arthritis. Semin Arthritis Rheum. 2024;69:152572. 10.1016/j.semarthrit.2024.152572
153 Tada M, Okano T, Mamaoto K, Yamada Y, Orita K, Mandai K, et al. Comparison of creatine kinase elevation caused by Janus kinase inhibitors and interleukin-6 inhibitors in patients with rheumatoid arthritis: A propensity score-matched study. Arch Rheumatol. 2024;39(3):350–7. 10.46497/ArchRheumatol.2024.10645
154 Lee YH, Song GG. Relative remission rates of Janus kinase inhibitors in comparison with adalimumab in patients with active rheumatoid arthritis: A network meta-analysis. Z Rheumatol. 2024;83(Suppl 1):88–96. 10.1007/s00393-022-01165-w
155 Genovese MC, van Vollenhoven RF, Wilkinson B, Wang L, Zwillich SH, Gruben D, et al. Switching from adalimumab to tofacitinib in the treatment of patients with rheumatoid arthritis. Arthritis Res Ther. 2016;18:145. 10.1186/s13075-016-1049-3
156 Papp KA, Krueger JG, Feldman SR, Langley RG, Thaci D, Torii H, et al. Tofacitinib, an oral Janus kinase inhibitor, for the treatment of chronic plaque psoriasis: Long-term efficacy and safety results from 2 randomized phase-III studies and 1 open-label long-term extension study. J Am Acad Dermatol. 2016;74(5):841–50. 10.1016/j.jaad.2016.01.013
157 Fournier J, Boussat B, Dervaux B, Gaudin P, Romand X. Cost-utility of tofacitinib in the treatment of moderate-to-severe rheumatoid arthritis in France: A multi-state Markov model analysis. Clin Exp Rheumatol. 2025;43(1):62-9. 10.55563/clinexprheumatol/3f60yv
158 Ekin A, Misirci S, İldemir S, Coskun BN, Yagiz B, Dalkilic E, et al. Efficacy and safety of tofacitinib in rheumatoid arthritis: Nine years of real-world data. Clin Transl Sci. 2024;17(11):e70084. 10.1111/cts.70084
159 Haraoui B, Khraishi M, Choquette D, Fortin I, Kinch CD, Galos C, et al. Tofacitinib safety and effectiveness in Canadian patients with rheumatoid arthritis by cardiovascular risk enrichment: Subanalysis of the CANTORAL study. Rheumatol Ther. 2024;11(6):1629-48. 10.1007/s40744-024-00719-5
160 Chang J, Wang G. A retrospective study of efficacy of tofacitinib combined with bDMARDs in the treatment of rheumatoid arthritis patients with inadequate response to bDMARDs. Int J Rheum Dis. 2024;27(9):e15311. 10.1111/1756-185X.15311
161 Pope J, Finckh A, Silva-Fernández L, Mandl P, Fan H, Rivas JL, et al. Tofacitinib monotherapy in rheumatoid arthritis: Clinical trials and real-world data contextualization of patients, efficacy, and treatment retention. Open Access Rheumatol. 2024;16:115–26. 10.2147/OARRR.S446431
162 D’Alessandro F, Cazzato M, Laurino E, Morganti R, Bardelli M, Frediani B, et al. ToRaRI (Tofacitinib in Rheumatoid Arthritis a Real-Life experience in Italy): Effectiveness, safety profile of tofacitinib and concordance between patient-reported outcomes and physician’s global assessment of disease activity in a retrospective study in central-Italy. Clin Rheumatol. 2024;43(2):657-65. 10.1007/s10067-023-06836-w
163 Kuwana M, Sugiyama N, Momohara S, Atsumi T, Takei S, Tamura N, et al. Six-month safety and effectiveness of tofacitinib in patients with rheumatoid arthritis in Japan: Interim analysis of post-marketing surveillance. Mod Rheumatol. 2024;34(2):272-86. 10.1093/mr/road063
164 Pappas DA, O’Brien J, Moore PC, Dodge R, Germino R, Masri KR, et al. Treatment patterns and effectiveness of tofacitinib in patients initiating therapy for rheumatoid arthritis: Results from the CorEvitas rheumatoid arthritis registry. J Rheumatol. 2024;51(5):452-61. 10.3899/jrheum.2023-0752
165 Cho SK, Song YJ, Kim HW, Nam E, Jeon JY, Yoo HJ, et al. Comparative effectiveness of tofacitinib and tumour necrosis factor inhibitors in patients with rheumatoid arthritis in real-world practice: A prospective observational study. Rheumatology (Oxford). 2025;64(2):541-7. 10.1093/rheumatology/keae109
166 Charles-Schoeman C, Fleischmann R, Mysler E, Greenwald M, Ytterberg SR, Koch GG, et al. Risk of venous thromboembolism with tofacitinib versus tumor necrosis factor inhibitors in cardiovascular risk-enriched rheumatoid arthritis patients. Arthritis Rheumatol. 2024;76(8):1218-29. 10.1002/art.42846
167 Opdam MAA, Broeder ND, van den Bemt BJF, Mulder K, van de Wiel KM, van Ballegooijen H, et al. Higher infection risk for JAK inhibitors tofacitinib and baricitinib compared to subcutaneous biological DMARDs. Clin Rheumatol. 2024;43(6):2133-8. 10.1007/s10067-024-06980-x
168 Buch MH, Bhatt DL, Charles-Schoeman C, Giles JT, Mikuls T, Koch GG, et al. Risk of extended major adverse cardiovascular event endpoints with tofacitinib versus TNF inhibitors in patients with rheumatoid arthritis: A post hoc analysis of a phase 3b/4 randomised safety study. RMD Open. 2024;10(2):e003912. 10.1136/rmdopen-2023-003912
169 Citera G, Mysler E, Kakehasi AM, Pascual-Ramos V, Masson W, Cadatal MJ, et al. Cardiovascular events, malignancies, and efficacy outcomes in Latin American patients with rheumatoid arthritis receiving tofacitinib or tumor necrosis factor inhibitors: A post hoc analysis of the ORAL Surveillance Study. J Clin Rheumatol. 2024;30(5):208-16. 10.1097/RHU.0000000000002106
170 Zhao J, Huang H, Wang Y, Deng X, Geng Y, Zhang X, et al. Real-world clinical equivalence of generic and branded tofacitinib: A prospective longitudinal cohort study in patients with rheumatoid arthritis. Mayo Clin Proc. 2024;99(1):26–38. 10.1016/j.mayocp.2023.08.029
171 Her M, Park J, Lee SG. A large pulmonary nodule in a rheumatoid arthritis patient treated with tofacitinib. Int J Rheum Dis. 2024;27(1):e15013. 10.1111/1756-185X.15013
172 Li Z, Shan Y, Dong J, Mei H, Kong Q, Li Y. Disseminated cryptococcosis presenting with generalized cutaneous involvement in a rheumatoid arthritis patient receiving tofacitinib: A case report. J Dermatol. 2024;51(2):e39-41. 10.1111/1346-8138.16960
173 Deng C, Chiu KH, Lou N, Xing F. Case report: Occult Listeria monocytogenes invasion leading to prosthetic hip joint infection in a patient with rheumatoid arthritis taking tofacitinib. Front Med (Lausanne). 2023;10:1322993. 10.3389/fmed.2023.1322993
174 Boussaa H, Hamdi O, Miladi S, Makhlouf Y, Abdelghani KB, Fazaa A, et al. Tofacitinib-induced acute pancreatitis in a patient with rheumatoid arthritis: A case report and review of the literature. Curr Drug Saf. 2024. 10.2174/0115748863300565240819114551
175 Sun YS, Huang DF, Chen WS, Liao HT, Chen MH, Tsai HC, et al. Real-world risk factors for herpes zoster in patients with rheumatoid arthritis undergoing tofacitinib treatment. J Chin Med Assoc. 2024;87(11):988-92. 10.1097/JCMA.0000000000001159
176 Keystone EC, Taylor PC, Tanaka Y, Gaich C, DeLozier AM, Dudek A, et al. Patient-reported outcomes from a phase 3 study of baricitinib versus placebo or adalimumab in rheumatoid arthritis: Secondary analyses from the RA-BEAM study. Ann Rheum Dis. 2017;76(11):1853–61. 10.1136/annrheumdis-2017-211259
177 Dougados M, van der Heijde D, Chen YC, Greenwald M, Drescher E, Liu J, et al. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: Results from the RA-BUILD study. Ann Rheum Dis. 2017;76(1):88–95. 10.1136/annrheumdis-2016-210094
178 Mahmoud EM, Radwan A, Elsayed SA. A prospective randomized-controlled non-blinded comparative study of the JAK inhibitor (baricitinib) with TNF-α inhibitors and conventional DMARDs in a sample of Egyptian rheumatoid arthritis patients. Clin Rheumatol. 2024;43(12):3657-68. 10.1007/s10067-024-07194-x
179 Edwards CJ, Takeuchi T, Finckh A, Kleyer A, Haladyj E, Gerwien JG, et al. Baricitinib as monotherapy for treatment of rheumatoid arthritis: Analysis of real-world data. Curr Med Res Opin. 2024:40(11):1993–2002. 10.1080/03007995.2024.2416979
180 Caporali R, Taylor PC, Aletaha D, Sanmartí R, Takeuchi T, Mo D, et al. Efficacy of baricitinib in patients with moderate-to-severe rheumatoid arthritis up to 6.5 years of treatment: Results of a long-term study. Rheumatology (Oxford). 2024;63(10):2799-809. 10.1093/rheumatology/keae012
181 Ekin A, Misirci S, Görünen A, Coskun BN, Yagiz B, Dalkilic E, et al. Is baricitinib effective and safe for patients with difficult-to-treat rheumatoid arthritis? Comparative data with the RA group of rheumatoid arthritis not difficult to treat. Med Princ Pract. 2025;34(1):75–86. 10.1159/000541488
182 van de Laar CJ, Oude Voshaar MAH, Ten Klooster P, Tedjo DI, Bos R, Jansen T, et al. PERFECTRA: A pragmatic, multicentre, real-life study comparing treat-to-target strategies with baricitinib versus TNF inhibitors in patients with active rheumatoid arthritis after failure on csDMARDs. RMD Open. 2024;10(2):e004291. 10.1136/rmdopen-2024-004291
183 Gilbert BTP, Mongin D, Aymon R, Lauper K, Laedermann C, Perrier C, et al. Comparative effectiveness of baricitinib and alternative biological DMARDs in a Swiss cohort study of patients with RA. BMJ Open. 2024;14(3):e072300. 10.1136/bmjopen-2023-072300
184 Okamoto N, Atsumi T, Takagi M, Takahashi N, Takeuchi T, Tamura N, et al. Safety of baricitinib in Japanese patients with rheumatoid arthritis in clinical use: 3-Year data of all-case post-marketing surveillance study. Mod Rheumatol. 2024;35(2):215-24. 10.1093/mr/roae064
185 Calvo-Garcia A, Ramírez Herráiz E, Llorente Cubas IM, Varas De Dios B, Benedí González J, Morell Baladrón A, et al. The real-world effectiveness, persistence, adherence, and safety of Janus kinase inhibitor baricitinib in rheumatoid arthritis: A long-term study. J Clin Med. 2024;13(9):2517. 10.3390/jcm13092517
186 Mariani FM, Alunno A, Carubbi F, Ferri C. A rare case of symptomatic creatine kinase elevation in a patient with rheumatoid arthritis treated with baricitinib. Reumatismo. 2024;76(2). 10.4081/reumatismo.2024.1620
187 Genovese MC, Smolen JS, Weinblatt ME, Burmester GR, Meerwein S, Camp HS, et al. Efficacy and safety of ABT-494, a selective JAK-1 inhibitor, in a phase IIB study in patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Rheumatol. 2016;68(12):2857–66. 10.1002/art.39808
188 Kremer JM, Emery P, Camp HS, Friedman A, Wang L, Othman AA, et al. A phase IIB study of ABT-494, a selective JAK-1 inhibitor, in patients with rheumatoid arthritis and an inadequate response to anti-tumor necrosis factor therapy. Arthritis Rheumatol. 2016;68(12):2867–77. 10.1002/art.39801
189 Lo Gullo A, Parisi S, Becciolini A, Paroli M, Bravi E, Andracco R, et al. Multicenter observational study on the efficacy of selective Janus kinase-1 inhibitor upatacitinib in rheumatoid arthritis. Minerva Med. 2024;115(4):430–8. 10.23736/S0026-4806.24.09409-6
190 Barešić M, Kilić P, Balen Topić M, Anić B. Prolonged eosinophilia caused by strongyloidiasis in a patient with rheumatoid arthritis treated with upadacitinib. Mod Rheumatol Case Rep. 2025;9(1):6-9. 10.1093/mrcr/rxae061
191 So H, Cheng I, Chow E, Wu Q, Li M, Hung V, et al. Erosion regression in patients with rheumatoid arthritis after upadacitinib-a pilot study using high resolution peripheral quantitative computed tomography. Rheumatology (Oxford). 2024. 10.1142/S2661341724740286
192 Burmester GR, Van den Bosch F, Tesser J, Shmagel A, Liu Y, Khan N, et al. Upadacitinib in rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying antirheumatic drugs: Efficacy and safety through 5 years (SELECT-NEXT). J Rheumatol. 2024;51(7):663-72. 10.3899/jrheum.2023-1062
193 Bessette L, Chan J, Chow A, Lisnevskaia L, Richard N, Fournier PA, et al. Real-world effectiveness of upadacitinib for treatment of rheumatoid arthritis in Canadian patients: Interim results from the prospective observational Close-Up study. Rheumatol Ther. 2024;11(3):563-82. 10.1007/s40744-024-00651-8
194 Baker JF, Zueger P, Ali M, Bennett D, Yu M, Munoz Maldonado Y, et al. Real-world use and effectiveness outcomes in patients with rheumatoid arthritis treated with upadacitinib: An analysis from the CorEvitas registry. Rheumatol Ther. 2024;11(2):363-80. 10.1007/s40744-024-00639-4
195 Baldi C, Parisi S, Falsetti P, Sota J, Ditto MC, Capassoni M, et al. Efficacy and safety of upadacitinib in rheumatoid arthritis: Real-life experience from a prospective longitudinal multicentric study. J Clin Med. 2024;13(2):401. 10.3390/jcm13020401
196 Xu H, Su B, Li R, Wang L, Zhou J, Zhao L. Successful treatment of anti-PTX3 positive seronegative rheumatoid arthritis with upadacitinib. Int J Rheum Dis. 2024;27(1):e14899. 10.1111/1756-185X.14899
197 Omidi F, Delkash P, Nasiri MJ, Mirsaeidi M. Upadacitinib and cardiovascular adverse events in rheumatoid arthritis: A systematic review and meta-analysis. Curr Rheumatol Rev. 2024. 10.1101/2023.10.24.23297509
198 Charles-Schoeman C, Giles JT, Lane NE, Choy E, Furst DE, Vencovský J, et al. Impact of upadacitinib on laboratory parameters and related adverse events in patients with RA: Integrated data up to 6.5 years. Rheumatol Ther. 2024;11(1):157-75. 10.1007/s40744-023-00624-3
199 Fleischmann R, Meerwein S, Charles-Schoeman C, Combe B, Hall S, Khan N, et al. Efficacy and safety of upadacitinib in patients with rheumatoid arthritis and inadequate response or intolerance to biological treatments: Results through 5 years from the SELECT-BEYOND study. RMD Open. 2024;10(3):e003918. 10.1136/rmdopen-2023-003918
200 Fleischmann R, Swierkot J, Penn SK, Durez P, Bessette L, Bu X, et al. Long-term safety and efficacy of upadacitinib versus adalimumab in patients with rheumatoid arthritis: 5-Year data from the phase 3, randomised SELECT-COMPARE study. RMD Open. 2024;10(2):e004007. 10.1136/rmdopen-2023-004007
201 Fleischmann R, Blanco R, Van den Bosch F, Bessette L, Song Y, Penn SK, et al. Long-term efficacy and safety following switch between upadacitinib and adalimumab in patients with rheumatoid arthritis: 5-Year data from SELECT-COMPARE. Rheumatol Ther. 2024;11(3):599-615. 10.1007/s40744-024-00658-1
202 Caporali R, Kadakia A, Howell O, Patel J, Milligan J, Strengholt S, et al. A real-world comparison of clinical effectiveness in patients with rheumatoid arthritis treated with upadacitinib, tumor necrosis factor inhibitors, and other advanced therapies after switching from an initial tumor necrosis factor inhibitor. Adv Ther. 2024;41(9):3706-21. 10.1007/s12325-024-02948-0
203 van Vollenhoven R, Strand V, Takeuchi T, Chávez N, Walter PM, Singhal A, et al. Upadacitinib monotherapy versus methotrexate monotherapy in patients with rheumatoid arthritis: Efficacy and safety through 5 years in the SELECT-EARLY randomized controlled trial. Arthritis Res Ther. 2024;26(1):143. 10.1186/s13075-024-03358-x
204 van Vollenhoven RF, Hall S, Wells AF, Meerwein S, Song Y, Tanjinatus O, et al. Long-term sustainability of response to upadacitinib among patients with active rheumatoid arthritis refractory to biological treatments: Results up to 5 years from SELECT-BEYOND. RMD Open. 2024;10(3):e004037. 10.1136/rmdopen-2023-004037.
205 Rubbert-Roth A, Kato K, Haraoui B, Rischmueller M, Liu Y, Khan N, et al. Safety and efficacy of upadacitinib in patients with rheumatoid arthritis refractory to biologic DMARDs: Results through week 216 from the SELECT-CHOICE Study. Rheumatol Ther. 2024;11(5):1197-215. 10.1007/s40744-024-00694-x
206 Picchianti Diamanti A, Cattaruzza MS, Salemi S, Di Rosa R, Sesti G, De Lorenzo C, et al. Clinical and ultrasonographic remission in bio-naïve and bio-failure patients with rheumatoid arthritis at 24 weeks of upadacitinib treatment: The UPARAREMUS real-life study. Rheumatol Ther. 2024;11(5):1347-61. 10.1007/s40744-024-00712-y
207 Namour F, Desrivot J, Van der Aa A, Harrison P, Tasset C, van’t Klooster G. Clinical confirmation that the selective JAK1 inhibitor filgotinib (GLPG0634) has a low liability for drug-drug interactions. Drug Metab Lett. 2016;10(1):38–48. 10.2174/1872312810666151223103353
208 Kavanaugh A, Kremer J, Ponce L, Cseuz R, Reshetko OV, Stanislavchuk M, et al. Filgotinib (GLPG0634/GS-6034), an oral selective JAK1 inhibitor, is effective as monotherapy in patients with active rheumatoid arthritis: Results from a randomised, dose-finding study (DARWIN 2). Ann Rheum Dis. 2017;76(6):1009–19. 10.1136/annrheumdis-2016-210105
209 Combe B, Kivitz A, Tanaka Y, van der Heijde D, Simon JA, Baraf HSB, et al. Filgotinib versus placebo or adalimumab in patients with rheumatoid arthritis and inadequate response to methotrexate: A phase III randomised clinical trial. Ann Rheum Dis. 2021;80(7):848–58. 10.1136/annrheumdis-2020-219214
210 Westhovens R, Rigby WFC, van der Heijde D, Ching DWT, Stohl W, Kay J, et al. Filgotinib in combination with methotrexate or as monotherapy versus methotrexate monotherapy in patients with active rheumatoid arthritis and limited or no prior exposure to methotrexate: The phase 3, randomised controlled FINCH 3 trial. Ann Rheum Dis. 2021;80(6):727–38. 10.1136/annrheumdis-2020-219213
211 Winthrop KL, Tanaka Y, Takeuchi T, Kivitz A, Matzkies F, Genovese MC, et al. Integrated safety analysis of filgotinib in patients with moderately to severely active rheumatoid arthritis receiving treatment over a median of 1.6 years. Ann Rheum Dis. 2022;81(2):184–92. 10.1136/annrheumdis-2021-221051
212 Buch MH, Aletaha D, Combe BG, Tanaka Y, Caporali R, Schulze-Koops H, et al. Efficacy and safety of filgotinib in patients with rheumatoid arthritis: Week 156 interim results from a long-term extension study. RMD Open. 2024;10(4): e004476. 10.1136/rmdopen-2024-004476.
213 Buch MH, Walker D, Edwards CJ, Barry J, Akroyd L, Omoruyi EVE, et al. Efficacy and safety of filgotinib in patients with moderately active rheumatoid arthritis and an inadequate response to methotrexate. Rheumatology (Oxford). 2024. 10.1093/rheumatology/keae486
214 Tanaka Y, Matsubara T, Atsumi T, Amano K, Ishiguro N, Hirata S, et al. Safety and efficacy of filgotinib in Japanese patients with rheumatoid arthritis: Week 156 interim results in FINCH 4. Mod Rheumatol. 2024. 10.1093/mr/roae099
215 Burmester GR, Gottenberg JE, Caporali R, Winthrop KL, Tanaka Y, Ekoka Omoruyi EV, et al. Integrated safety analysis of filgotinib in patients with moderate-to-severe rheumatoid arthritis over a treatment duration of up to 8.3 years. Ann Rheum Dis. 2024;83(9):1110-7. 10.1136/ard-2024-225759
216 Schultz O, Fiehn C, Kneitz C, Picker N, Kromer D, Zignani M, et al. Real-world experience with filgotinib for rheumatoid arthritis in Germany: A retrospective chart review. Z Rheumatol. 2024;84:39-47. 10.1007/s00393-024-01506-x
217 Fornaro M, Caporali R, Biggioggero M, Bugatti S, De Stefano L, Cauli A, et al. Effectiveness and safety of filgotinib in rheumatoid arthritis patients: Data from the GISEA registry. Clin Exp Rheumatol. 2024;42(5):1043-50. 10.55563/clinexprheumatol/b40rv4
218 La Barbera L, Rizzo C, Camarda F, Atzeni F, Miceli G, Molica Colella AB, et al. Effectiveness and safety of filgotinib in rheumatoid arthritis: A real-life multicentre experience. Clin Exp Rheumatol. 2024;42(5):991-8. 10.55563/clinexprheumatol/k78ug3
219 Gao X, He X, Oshima H, Miyatake D, Otsuka Y, Kato K, et al. Pharmacokinetics and safety of single and multiple doses of peficitinib (ASP015K) in healthy Chinese subjects. Drug Des Devel Ther. 2022;16:1365–81. 10.2147/DDDT.S359501
220 Tanaka Y, Izutsu H. Peficitinib for the treatment of rheumatoid arthritis: An overview from clinical trials. Expert Opin Pharmacother. 2020;21(9):1015–25. 10.1080/14656566.2020.1739649
221 Yang Y, Li J, Liu J, Liu L, Wang Y, Hu J, et al. Safety and efficacy of peficitinib in Asian patients with rheumatoid arthritis who had an inadequate response or intolerance to methotrexate: Results of a multicenter, randomized, double-blind, placebo-controlled phase 3 study. Lancet Reg Health West Pac. 2024;42:100925. 10.1016/j.lanwpc.2023.100925
222 Tanaka Y, Takeuchi T, Morita Y, Kaneko Y, Terada W. Safety and effectiveness of peficitinib 100 mg/day in patients achieving clinical remission from a long-term open-label extension study in Japan, Korea, and Taiwan (RAJ2). Mod Rheumatol. 2024;34(5):918-26. 10.1093/mr/road110
223 Tanaka Y, Takeuchi T, Morita Y, Kato D, Kaneko Y, Terada W. Post hoc analysis of patients with rheumatoid arthritis under clinical remission in two Japanese phase 3 trials of peficitinib treatment (RAJ3 and RAJ4). Mod Rheumatol. 2024;34(3):453–65. 10.1093/mr/road059
224 Prakken B, Albani S, Martini A. Juvenile idiopathic arthritis. Lancet. 2011;377(9783):2138–49. 10.1016/S0140-6736(11)60244-4
225 Petty RE, Southwood TR, Manners P, Baum J, Glass DN, Goldenberg J, et al. International League of Associations for Rheumatology classification of juvenile idiopathic arthritis: Second revision, Edmonton, 2001. J Rheumatol. 2004;31(2):390–2.
226 Srinivasalu H, Simpson J, Stoll ML. Drug therapy in juvenile spondyloarthritis. Curr Opin Rheumatol. 2024;36(4):295–301. 10.1097/BOR.0000000000001016
227 Poddighe D, Romano M, Gattinara M, Gerloni V. Biologics for the treatment of juvenile idiopathic arthritis. Curr Med Chem. 2018;25(42):5860–93. 10.2174/0929867325666180522085716