Back to Journals » Open Access Rheumatology: Research and Reviews » Volume 17

Impact of Biological Therapies on Quality of Life in Rheumatoid Arthritis: A Narrative Review

Authors Mamasaidov A , Sakibaev K, Zhumabaeva S , Isakov U , Eshbaeva CA, Abdyllaev J, Abdikhalilov B, Salieva RS 

Received 20 February 2025

Accepted for publication 10 April 2025

Published 23 April 2025 Volume 2025:17 Pages 73—86

DOI https://doi.org/10.2147/OARRR.S523778

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 4

Editor who approved publication: Professor Chuan-Ju Liu



Abdimutalib Mamasaidov,1,* Kyazbek Sakibaev,1 Symbat Zhumabaeva,1 Ulanbek Isakov,1 Chynara Abdasbekovna Eshbaeva,1 Joldubai Abdyllaev,1 Bektur Abdikhalilov,1 Rana Sherbaevna Salieva2,*

1Medical Faculty, Osh State University, Osh, Kyrgyzstan; 2International Medical Faculty, Osh State University, Osh, Kyrgyzstan

*These authors contributed equally to this work

Correspondence: Rana Sherbaevna Salieva, International Medical Faculty, Osh State University, Osh, 723500, Kyrgyzstan, Email [email protected]

Abstract: Rheumatoid arthritis (RA) is a chronic inflammatory disease that causes joint damage, pain, and disability, leading to significant impairments in patients’ physical, mental, and social well-being. While biological disease-modifying antirheumatic drugs (bDMARDs) such as tumor necrosis factor (TNF) inhibitors, interleukin-6 (IL-6) inhibitors, and Janus kinase (JAK) inhibitors have revolutionized the treatment of RA by effectively controlling disease activity, their influence on patients’ quality of life (QoL) is crucial but not fully understood. The aim of this review is to evaluate the impact of bDMARDs on QoL in RA patients, particularly focusing on domains such as physical functioning, pain, fatigue, mental health, and social participation. A comprehensive literature search was conducted in databases such as PubMed and the Cochrane Library, including randomized controlled trials, cohort studies, and surveys assessing QoL outcomes in RA patients receiving bDMARD therapy. The review includes studies that utilized the Health Assessment Questionnaire (HAQ), EuroQol-5 Dimension (EQ-5D), and Short Form-36 (SF-36), Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F), Fatigue Severity Scale (FSS), and Patient Global Assessment (PtGA) QoL questionnaires, among others, to assess patient-reported outcomes. The findings of the current review suggest that bDMARDs significantly improve QoL in RA patients by reducing pain, fatigue, and disability while enhancing physical function and mental well-being. However, variability in patient responses, side effects, and the long-term impact of these therapies remain key concerns. Future studies with standardized QoL assessments and longer follow-up periods are needed to provide a more comprehensive understanding of the sustained effects of bDMARD therapy on RA patients’ overall well-being.

Keywords: rheumatoid arthritis, biological therapies, QoL, HAQ-DI, patient-reported outcomes, physical function, mental health, fatigue

Introduction

Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease characterized by persistent synovial inflammation, which leads to progressive joint damage, deformity, and functional disability.1 The pathogenesis of RA involves a complex interplay between genetic susceptibility and environmental factors, leading to immune dysregulation. Key mechanisms include the activation of antigen-presenting cells, autoreactive T and B cells, and the overproduction of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 (IL-1).2,3

The impact of RA extends beyond joint pathology, significantly impairing patients’ physical function and overall QoL. The progressive nature of the disease leads to limitations in daily activities, such as walking and climbing stairs, contributing to reduced independence.4 Persistent joint pain, fatigue, and stiffness not only limit mobility but also affect mental well-being, increasing the risk of anxiety and depression.5 Emerging evidence suggests that inflammation itself may contribute to neuropsychiatric symptoms through cytokine-mediated pathways, emphasizing the need for a holistic approach to RA management.6,7

QoL in RA is a multidimensional concept encompassing physical, emotional, and social well-being. It is commonly assessed using validated instruments such as the HAQ, the SF-36, and the EQ-5D.8 Effective disease management aims to alleviate pain, improve function, and enhance psychosocial health, as these factors collectively influence treatment adherence and patient satisfaction.9,10 Given the chronic nature of RA, it is essential to consider QoL as an outcome measure in the management of the disease, as it directly correlates with treatment satisfaction, adherence to therapy, and overall health perception.

The advent of biologic disease-modifying antirheumatic drugs (bDMARDs) has transformed RA treatment by targeting specific immune pathways involved in disease progression. TNF inhibitors, IL-6 receptor antagonists, and JAK inhibitors have demonstrated significant efficacy in reducing inflammation, preventing structural damage, and improving functional outcomes.11,12 However, while these therapies effectively control disease activity, their impact on QoL varies among patients due to factors such as treatment response, comorbidities, and socioeconomic status.13 Several studies indicate that bDMARDs not only improve physical function but also contribute to better mental health outcomes by reducing pain and fatigue. According to the European League Against Rheumatism (EULAR) 2022 update, biological disease-modifying antirheumatic drugs (bDMARDs) are recommended for patients with rheumatoid arthritis (RA) who have an inadequate response to conventional synthetic DMARDs (csDMARDs) and present poor prognostic factors. In such cases, adding any bDMARD to the csDMARD regimen is advised.14 Regarding utilization rates, a study analyzing US outpatient data from 2018 to 2022 found that approximately 31.7% of RA patients were treated with bDMARDs.15 These sources provide evidence for the recommendation of bDMARDs following inadequate response to csDMARDs and indicate that their utilization among RA patients is around 30%, highlighting the adherence to treatment guidelines in clinical practice.

Despite their proven efficacy in disease control, the impact on QoL remains heterogeneous across different patient populations, necessitating further investigation.

Quality of Life as a Key Outcome in Rheumatoid Arthritis

QoL in the context of RA refers to a multifaceted concept encompassing patients’ physical, emotional, and social well-being. The physical aspect of QoL in RA patients is closely tied to disease activity, joint damage, and functional disability. Chronic pain, fatigue, joint stiffness, and loss of mobility are some of the most debilitating symptoms that directly influence physical functioning.16,17 As RA progresses, joint deformities and decreased range of motion may further limit the ability to perform daily activities such as walking, dressing, or cooking. The severity of pain and the degree of physical disability are often quantified using scales like the Disease Activity Score (DAS28) and Visual Analog Scale (VAS), which assess physical functioning and disease burden.18–20 Although tools assess tools that assess aspects of QoL related to physical function and mental health, including Health Assessment Questionnaire – Disability Index (HAQ-DI) that evaluates functional disability and is frequently used in RA clinical trials; Patient-Reported Outcomes Measurement Information System (PROMIS) which covers multiple domains such as pain, fatigue, depression, and physical function; Functional Assessment of Chronic Illness Therapy – Fatigue Scale (FACIT-F) which measures fatigue impact in RA patients.21,22 Biological therapies, particularly in the treatment of autoimmune and chronic inflammatory diseases, have become a cornerstone of clinical management, significantly improving patients’ QoL. Numerous studies have demonstrated that biological therapies can positively affect various QoL domains, including physical functioning, emotional well-being, social participation, and productivity.23–25 In a cohort study by Ines et al reported an improvement of FACIT-F score by 15.06% after sixth month of treatment with biologics.26 A Similar result was reported in a multi-ethnic cohort of rheumatoid arthritis patients.27 Biologics, particularly TNF inhibitors (Etanercept, Infliximab, and Adalimumab) and Rituximab, have been shown to effectively reduce fatigue in RA patients. A systematic review by Almeida et al highlights that improvements in fatigue often occur before significant changes in clinical disease activity, suggesting that biologics may target fatigue through mechanisms that go beyond inflammation reduction. This finding aligns with the hypothesis that fatigue in RA is not solely an inflammatory symptom but may involve additional pathways, such as neuroimmune interactions, psychological factors, and central sensitization.28 One of the key takeaways from this review is the suggestion that fatigue reduction may not be entirely linked to improvements in disease activity. This is particularly important because fatigue is a chronic, multidimensional symptom that significantly impacts patients’ QoL, irrespective of visible joint inflammation. The potential mechanisms by which biologics reduce fatigue include immune modulation, central nervous system effects, and the restoration of circadian rhythm—areas that warrant further exploration. Bessette et al demonstrated statistically significant improvements of HAQ-DI from baseline in patients with moderate to severely active RA treated with abatacept.29

A cross-sectional, non-interventional study by Inotai and colleagues reported that RA patients treated with biological agents experience lower disease activity and improved health-related quality of life compared to those receiving non-biological treatments. These findings underscore the importance of considering patient-reported outcomes and utility measures when evaluating treatment effectiveness in RA.30 These results support the integration of biological therapies into RA treatment regimens to enhance patient well-being. However, the study also highlights the need for individualized treatment plans, as the benefits of biological agents must be weighed against factors such as cost and potential adverse effects.

One of the most direct impacts of biological therapies on QoL is through significant improvements in physical functioning. In RA, joint pain and stiffness are prevalent, causing major limitations in daily activities and physical mobility.31,32 Numerous studies have demonstrated that patients treated with biological therapies, particularly TNF inhibitors, experienced substantial alleviation of joint pain and stiffness, resulting in improved physical functioning.33–35 HAQ-DI scores, which is a widely used measure of functional disability. Similarly, pain reduction was commonly reported by patients, leading to a higher degree of physical independence and improved performance of daily tasks. This reduction in physical limitations allows individuals to engage in more physical activity, contributing to better overall health and well-being.4,24,25

Chronic inflammatory diseases often lead to psychological distress, including anxiety, depression, and stress, particularly due to persistent symptoms and uncertain disease progression. The improvement in disease control facilitated by biological therapies can have a profound impact on emotional well-being.36,37 Several observed that, alongside improvements in physical symptoms, patients on biological treatments reported a reduction in depressive symptoms and anxiety. This aligns with findings from other studies, such as those by38–40 which show that controlling disease activity in RA not only alleviates physical symptoms but also improves mood and overall mental health. Biological treatments seem to reduce the emotional burden of living with a chronic disease, leading to greater psychological resilience and a more optimistic outlook on life. Chronic inflammatory diseases can significantly impair patients’ ability to engage in social activities and maintain personal relationships, crucial aspects of quality of life. Biological therapies, particularly anti-TNF agents, have been shown to alleviate these challenges. A study on patients with chronic inflammatory arthritis undergoing biological therapy found that social support from family, friends, and colleagues was essential in influencing a good treatment response. Regular conversations and social interactions were highlighted as important components of this support.41 Furthermore, the improved QoL associated with biologic therapy can increase productivity and reduce the societal impact of rheumatoid arthritis. Earlier access to biologics for RA patients has been shown to have a positive effect on employment status, indicating enhanced social participation and engagement.42 These findings suggest that by reducing disease activity and improving physical function, biological therapies enable patients to re-engage in social activities and relationships that were previously hindered by their condition. This restoration of social participation contributes to a better sense of belonging, social satisfaction, and overall mental health, as it fosters a sense of community and reduces isolation.

Chronic diseases significantly impact work-related outcomes, including absenteeism and productivity.43,44 Biological therapies have been shown to facilitate a return to work and enhance productivity. A study on patients with RA treated with etanercept reported significant reductions in work-related absenteeism and improvements in productivity.45 Shim et al reported significant improvements of work productivity patients with axial spondyloarthritis undergoing biological therapy.46 These improvements in work capacity contribute not only to financial stability but also to a sense of personal achievement and identity, which are important aspects of QoL. However, we could not find studies on the relationship between work productivity and biological therapies in the case of rheumatoid arthritis. Further studies are needed for standardized, RA-specific work productivity assessment tools would be valuable.

Biological therapies have been shown to significantly enhance QoL in patients with chronic inflammatory diseases, particularly RA. These improvements encompass various domains, including physical functioning, pain intensity, fatigue, and sleep quality. Fatigue is a prevalent and debilitating symptom in autoimmune diseases. Studies have demonstrated that biological therapies can lead to notable reductions in fatigue levels. A Cochrane review reported that treatment with biologics resulted in small to moderate reductions in patient-reported fatigue compared to placebo.28 Improved sleep quality is closely associated with reduced disease activity and pain relief. Research indicates that patients with RA often experience poor sleep quality, which correlates with higher disease activity and pain levels. Effective management of RA through biological therapies has been linked to improvements in sleep quality.47

The reduction in disease activity, as measured by indices like the DAS28 in RA, has been directly correlated with improved QoL outcomes. Lower DAS28 scores, indicating reduced disease activity, are associated with better physical function, less pain, and overall enhanced QoL.11,42 The Table 1 captures the diverse range of study designs, biologic therapies, and outcome measures used to assess the impact of these therapies on quality of life in patients with RA. The QoL improvements outline the various QoL assessment tools used in the studies. Commonly used tools include HAQ-DI, SF-36, FACIT-F, PtGA, EQ-5D, FSS, and others like WPAI-RA.23–26,28,30,31,38,40,45–69 These tools measure different aspects of patients’ health, including physical function, fatigue, and overall well-being.

Table 1 Summary of Studies Assessing QoL in RA Patients Receiving Biologic Therapy

Factors Influencing Quality of Life Outcomes in Rheumatoid Arthritis

Disease Activity and Treatment Response

The level of RA disease control directly correlates with QoL outcomes. Studies consistently demonstrate that achieving low disease activity or remission improves physical function, reduces pain, and enhances mental health. The ACR and EULAR criteria for treatment response underscore the importance of early and sustained control of disease activity for optimal QoL benefits.70 Biologic therapies such as adalimumab, infliximab, and etanercept are associated with significant reductions in disease activity and improved patient-reported outcomes.11,45 Patients who achieve remission often report enhanced physical functioning and reduced fatigue, contributing to better overall QoL. Conversely, inadequate treatment response, marked by persistent joint inflammation and pain, correlates with poorer QoL scores.36 These findings highlight the necessity of personalized treatment strategies to optimize disease control and QoL outcomes.

A treat-to-target (T2T) strategy, which involves regular monitoring and therapy adjustments to achieve predefined disease activity goals, has been widely endorsed to improve clinical outcomes. A systematic review and meta-regression analysis reported that patients managed with a T2T approach experience higher rates of remission, better physical function, and lower healthcare costs compared to conventional treatment strategies.71

Patient Characteristics

Demographic and clinical characteristics significantly influence QoL in RA patients. Age, disease duration, baseline disease severity, and comorbidities play pivotal roles. Older patients often report lower QoL scores due to age-related functional limitations and increased comorbidities, such as cardiovascular diseases and depression.72 Additionally, longer disease duration correlates with accumulated joint damage and reduced functional capacity, further impacting QoL.73 Patients with high baseline disease activity tend to exhibit greater improvements in QoL when achieving disease control through biologics, but they may also have a higher burden of initial symptoms. Addressing comorbidities through integrated care is crucial, as these conditions exacerbate pain, disability, and psychological distress, all of which negatively influence QoL.

Adverse Drug Events of Biological Therapies

Although biologic therapies improve disease activity and QoL, their adverse effects may offset these benefits for some patients.74 Common adverse drug reactions include injection site reactions, increased infection risk, and gastrointestinal disturbances, which may contribute to treatment discontinuation.13,75 Serious infections associated with TNF inhibitors are a critical concern, particularly in patients with pre-existing risk factors.76 Patient-reported outcome measures (PROMs) reveal that side effects like fatigue, nausea, and anxiety due to medication concerns can significantly reduce QoL despite clinical improvements in RA symptoms.24,25,52 Strategies to minimize adverse effects, such as careful patient selection, regular monitoring, and patient education, are essential to maximizing QoL benefits.

Patient Expectations and Perceptions

Patient expectations and perceptions of biologic therapies significantly influence their reported QoL outcomes. Unrealistically high expectations may lead to dissatisfaction, even when clinical targets are met. Conversely, patients with realistic expectations are more likely to report improved QoL when experiencing symptom relief and functional gains.77 Studies suggest that shared decision-making, which incorporates patient preferences and educates them on realistic outcomes, enhances treatment satisfaction and perceived QoL improvements.78 Patient-centered communication that aligns therapeutic goals with individual expectations is thus integral to optimizing QoL outcomes.79 QoL outcomes in RA are multifactorial, influenced by disease activity, patient characteristics, treatment side effects, and patient expectations. Achieving low disease activity or remission remains the cornerstone of improving QoL, underscoring the importance of individualized treatment strategies.

Challenges and Limitations in Measuring Quality of Life in Rheumatoid Arthritis

Measuring QoL in patients with RA is a critical aspect of understanding the disease’s impact and evaluating treatment effectiveness. However, capturing the complexity of QoL is challenging due to the multidimensionality of the concept, the subjective nature of assessments, and methodological inconsistencies. QoL encompasses several domains, including physical, emotional, social, and psychological well-being, all influenced by disease activity, functional limitations, and treatment responses. Instruments like the HAQ and the SF-36 often focus on physical health, underrepresenting psychological and social aspects.24,80,81 This imbalance can lead to an incomplete assessment of QoL.

Inadequacy of Standardized Tools

Assessing health-related QoL in RA patients presents unique challenges due to the disease’s multifaceted nature. While widely used generic tools like the SF-36 and EQ-5D are valuable for evaluating overall health status, they may not fully capture RA-specific symptoms such as joint stiffness, fatigue, and morning stiffness. Studies have shown that patients with RA score significantly lower on physical functioning and bodily pain dimensions of the SF-36 compared to individuals without musculoskeletal diseases, highlighting the substantial impact of RA on physical aspects of health-related QoL.72 To address these limitations, disease-specific instruments like the Rheumatoid Arthritis Quality of Life (RAQoL) questionnaire have been developed.30,82 The RAQoL was created directly from patient interviews to ensure relevance to the RA experience and has demonstrated high internal consistency and test-retest reliability. Additionally, the RAQoL has been adapted and validated in multiple languages, facilitating its use in diverse populations.

Despite its robust psychometric properties, the RAQoL’s adoption in clinical practice remains limited, possibly due to a lack of awareness or training among healthcare providers. Integrating RA-specific QoL assessments like the RAQoL into routine care could enhance the understanding of patient experiences and improve treatment outcomes.

Dynamic and Fluctuating Nature of Rheumatoid Arthritis

RA is characterized by periods of flares and remission, leading to fluctuations in QoL over time.1 Cross-sectional assessments often fail to capture these dynamic changes, providing only a static view of the patient’s experience.41 RA flares involve a sudden worsening of symptoms such as joint pain, swelling, and stiffness, which can persist for days to weeks. These episodes disrupt daily functioning and reduce QoL, even when overall disease activity appears controlled. In contrast, periods of remission or low disease activity allow for better functioning and improved well-being.25 The cumulative effect of repeated flares contributes to joint damage, physical disability, and comorbid conditions such as cardiovascular disease.5 This progressive nature of RA, influenced by the frequency and severity of flares, underscores the need for effective management strategies. The cumulative effect of repeated flares contributes to joint damage, physical disability, and comorbid conditions such as cardiovascular disease. This progressive nature of RA, influenced by the frequency and severity of flares, underscores the need for effective management strategies.3 Longitudinal studies and repeated QoL assessments are more effective in reflecting the dynamic course of RA. Tools that track changes over time, such as the DAS28 combined with patient-reported outcomes, offer better insights into the interplay between disease activity and QoL.83 Including flare frequency, duration, and severity as part of routine QoL assessments provides a more comprehensive understanding of the disease’s impact. Such data are essential for tailoring treatment plans to minimize flares and improve patient outcomes.81

Clinical Implication

Impact of Comorbidities

Comorbidities are additional medical conditions that coexist with RA and significantly influence disease outcomes, treatment approaches, and patients’ QoL. Patients with RA are at increased risk of developing cardiovascular diseases, including atherosclerosis, myocardial infarction, and stroke. Chronic inflammation is a major contributing factor, as it accelerates vascular damage and lipid metabolism abnormalities.84–86 This increased cardiovascular risk often necessitates aggressive inflammation control and lifestyle modifications. Chronic inflammation, long-term glucocorticoid use, and reduced physical activity contribute to an elevated risk of osteoporosis and fractures in RA patients.87 Osteoporotic fractures can severely impair mobility and QoL, necessitating early screening and preventive strategies.88 Depression and anxiety are prevalent in RA patients, often linked to chronic pain, functional limitations, and social isolation. These mental health conditions further reduce QoL and can hinder adherence to treatment regimens.72 RA treatments, especially biologics and glucocorticoids, increase susceptibility to infections. This vulnerability complicates disease management, as infections can trigger RA flares and necessitate treatment interruptions.89 RA patients are more likely to develop metabolic syndrome, characterized by obesity, insulin resistance, and hypertension.90 This is partly due to systemic inflammation and corticosteroid use, which exacerbate metabolic dysregulation. The presence of comorbidities often limits therapeutic options. Cardiovascular risk factors may preclude the use of certain nonsteroidal anti-inflammatory drugs (NSAIDs), and infections may restrict the use of biologics. The systematic review and meta-analysis titled “Effect of TNF inhibitors on arterial stiffness and intima media thickness in rheumatoid arthritis” by Abdulmajid et al reported that TNF inhibitor therapy in RA patients may favorably influence certain surrogate markers of cardiovascular risk.91

RA is not limited to joint pathology; it also involves systemic inflammation that affects organs such as the lungs, heart, and skin.84–93 Common comorbidities include cardiovascular disease, interstitial lung disease, and osteoporosis. These conditions complicate disease management and contribute to increased morbidity and mortality.88,94

RA imposes substantial direct and indirect costs on patients and healthcare systems. Direct costs include medication, hospitalizations, and laboratory monitoring, while indirect costs arise from work absenteeism and loss of productivity. These economic implications underscore the need for cost-effective treatment approaches.95 Early diagnosis and treatment initiation are critical for preventing joint damage and systemic complications. However, the nonspecific early symptoms of RA often lead to delays in diagnosis, resulting in poorer outcomes.49 The presence of comorbid conditions complicates the choice of therapies. For example, cardiovascular risk may limit the use of NSAIDs, and infections may preclude biologic treatments.86,96 Adherence to long-term RA treatment is challenging due to medication side effects, the complexity of treatment regimens, and patient perceptions of efficacy.23 Nonadherence can lead to disease flares and worse outcomes. Treat-to-target strategies using DMARDs or biologics aim to achieve remission or low disease activity, thereby preventing joint damage and systemic complications.48 Collaboration between rheumatologists, primary care physicians, psychologists, and other specialists ensures comprehensive management of RA and its associated comorbidities. Educating patients about RA, its progression, and the importance of adherence to the treatment plan.

Conclusion

The findings from this review highlight the significant impact of bDMARDs on the QoL of patients with RA. While these therapies have revolutionized RA treatment by effectively reducing inflammation and preventing joint damage, their influence extends beyond physical health, encompassing mental, emotional, and social well-being. One of the most notable benefits of biologic therapies is their ability to improve physical function by alleviating pain, stiffness, and fatigue. Studies have consistently demonstrated improvements in HAQ-DI scores, supporting the role of biologics in maintaining functional independence. Furthermore, evidence suggests that fatigue reduction associated with biologic therapy occurs independently of inflammation control, potentially indicating additional mechanisms such as modulation of neuroimmune pathways.

The majority of reviewed studies overemphasized biological therapies. While biologics have revolutionized RA treatment, traditional DMARDs like methotrexate still play a crucial role, especially in resource-limited settings. Integrating biosimilars, personalized treatment approaches, and early intervention strategies has demonstrated potential for optimizing both clinical outcomes and economic efficiency. Holistic management, encompassing regular monitoring, patient education, and multidisciplinary care, remains essential for addressing the diverse challenges of RA.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Sharma S, Bluett J. Towards personalized medicine in rheumatoid arthritis. Open Access Rheumatol Res Rev. 2024;16:89–114. doi:10.2147/OARRR.S372610

2. Mueller AL, Payandeh Z, Mohammadkhani N, et al. Recent advances in understanding the pathogenesis of rheumatoid arthritis: new treatment strategies. Cells. 2021;10(11):3017. doi:10.3390/cells10113017

3. Gao Y, Zhang Y, Liu X. Rheumatoid arthritis: pathogenesis and therapeutic advances. MedComm. 2024;5(3):e509. doi:10.1002/mco2.509

4. Barton JL. Patient preferences and satisfaction in the treatment of rheumatoid arthritis with biologic therapy. Patient Prefer Adherence. 2009;3:335–344. doi:10.2147/ppa.s5835

5. Romão VC, Fonseca JE. Disease mechanisms in preclinical rheumatoid arthritis: a narrative review. Front Med. 2022;9:689711. doi:10.3389/fmed.2022.689711

6. Lopez-Olivo MA, Foreman JT, Leung C, et al. A randomized controlled trial evaluating the effects of social networking on chronic disease management in rheumatoid arthritis. Semin Arthritis Rheum. 2022;56:152072. doi:10.1016/j.semarthrit.2022.152072

7. Bertoldi I, Caporali R. Tofacitinib: real-world data and treatment persistence in rheumatoid arthritis. Open Access Rheumatol Res Rev. 2021;13:221–237. doi:10.2147/OARRR.S322086

8. Nagafusa T, Mizushima T, Suzuki M, Yamauchi K. Comprehensive relationship between disease activity indices, mTSS, and mHAQ and physical function evaluation and QOL in females with rheumatoid arthritis. Sci Rep. 2023;13(1):21905. doi:10.1038/s41598-023-49380-y

9. Aktekin LA, Eser F, Başkan BM, et al. Disability of Arm Shoulder and Hand Questionnaire in rheumatoid arthritis patients: relationship with disease activity, HAQ, SF-36. Rheumatol Int. 2011;31(6):823–826. doi:10.1007/s00296-010-1568-1

10. Wright G, Zueger P, Copley-Merriman C, et al. Health disparities in rheumatology in the United States. Open Access Rheumatol Res Rev. 2025;17:1–12. doi:10.2147/OARRR.S493457

11. Stevenson M, Archer R, Tosh J, et al. Adalimumab, etanercept, infliximab, certolizumab pegol, golimumab, tocilizumab and Abatacept for the treatment of rheumatoid arthritis not previously treated with disease-modifying antirheumatic drugs and after the failure of conventional disease-modifying antirheumatic drugs only: systematic review and economic evaluation. Health Technol Assess Winch Engl. 2016;20(35):1–610. doi:10.3310/hta20350

12. Pope J, Finckh A, Silva-Fernández L, et al. Tofacitinib monotherapy in rheumatoid arthritis: clinical trials and real-world data contextualization of patients, efficacy, and treatment retention. Open Access Rheumatol Res Rev. 2024;16:115–126. doi:10.2147/OARRR.S446431

13. Costa NT, Iriyoda TMV, Alfieri DF, Simão ANC, Dichi I. Influence of disease-modifying antirheumatic drugs on oxidative and nitrosative stress in patients with rheumatoid arthritis. Inflammopharmacology. 2018;26(5):1151–1164. doi:10.1007/s10787-018-0514-9

14. Smolen JS, Landewé RBM, Bergstra SA et al, 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(3):e76. doi:10.1136/ard-2022-223356corr1

15. Gaitonde P, Bozzi LM, Shaya FT. Factors associated with use of disease modifying agents for rheumatoid arthritis in the national hospital and ambulatory medical care survey. Semin Arthritis Rheum. 2018;47(5):649–653. doi:10.1016/j.semarthrit.2017.10.011

16. Beckers E, Hermans K, Van Tubergen A, Boonen A. Fatigue in patients with rheumatic and musculoskeletal diseases: a scoping review on definitions, measurement instruments, determinants, consequences and interventions. RMD Open. 2023;9(3):e003056. doi:10.1136/rmdopen-2023-003056

17. Wolfe F. Determinants of WOMAC function, pain and stiffness scores: evidence for the role of low back pain, symptom counts, fatigue and depression in osteoarthritis, rheumatoid arthritis and fibromyalgia. Rheumatol Oxf Engl. 1999;38(4):355–361. doi:10.1093/rheumatology/38.4.355

18. Revicki D, Ganguli A, Kimel M, et al. Reliability and validity of the work instability scale for rheumatoid arthritis. Value Health J Int Soc PharmacoEcon Outcomes Res. 2015;18(8):1008–1015. doi:10.1016/j.jval.2015.09.2941

19. Turesson Wadell A, Bärebring L, Hulander E, et al. Effects on health-related quality of life in the randomized, controlled crossover trial ADIRA (anti-inflammatory diet in rheumatoid arthritis). PLoS One. 2021;16(10):e0258716. doi:10.1371/journal.pone.0258716

20. Linde L, Sørensen J, Ostergaard M, Hørslev-Petersen K, Hetland ML. Health-related quality of life: validity, reliability, and responsiveness of SF-36, 15D, EQ-5D [corrected] RAQoL, and HAQ in patients with rheumatoid arthritis. J Rheumatol. 2008;35(8):1528–1537.

21. Hewlett S, Dures E, Almeida C. Measures of fatigue: bristol rheumatoid arthritis fatigue multi-dimensional questionnaire (BRAF MDQ), bristol rheumatoid arthritis fatigue numerical rating scales (BRAF NRS) for severity, effect, and coping, chalder fatigue questionnaire (CFQ), checklist individual strength (CIS20R and CIS8R), fatigue severity scale (FSS), functional assessment chronic illness therapy (fatigue) (FACIT-F), multi-dimensional assessment of fatigue (MAF), multi-dimensional fatigue inventory (MFI), pediatric quality of life (PedsQL) multi-dimensional fatigue scale, profile of fatigue (ProF), short form 36 vitality subscale (SF-36 VT), and visual analog scales (VAS). Arthritis Care Res. 2011;63(Suppl 11):S263–286. doi:10.1002/acr.20579

22. Beaumont JL, Davis ES, Curtis JR, Cella D, Yun H. Meaningful change thresholds for patient-reported outcomes measurement information system (PROMIS) fatigue and pain interference scores in patients with rheumatoid arthritis. J Rheumatol. 2021;48(8):1239–1242. doi:10.3899/jrheum.200990

23. Bingham CO, Walker D, Nash P, et al. The impact of filgotinib on patient-reported outcomes and health-related quality of life for patients with active rheumatoid arthritis: a post hoc analysis of Phase 3 studies. Arthritis Res Ther. 2022;24(1):11. doi:10.1186/s13075-021-02677-7

24. Bergman M, Tundia N, Martin N, et al. Patient-reported outcomes of upadacitinib versus Abatacept in patients with rheumatoid arthritis and an inadequate response to biologic disease-modifying antirheumatic drugs: 12- and 24-week results of a phase 3 trial. Arthritis Res Ther. 2022;24(1):155. doi:10.1186/s13075-022-02813-x

25. Smolen JS, Kremer JM, Gaich CL, et al. Patient-reported outcomes from a randomised Phase III study of baricitinib in patients with rheumatoid arthritis and an inadequate response to biological agents (RA-BEACON). Ann Rheum Dis. 2017;76(4):694–700. doi:10.1136/annrheumdis-2016-209821

26. Ines M, Aicha BT, Leila R, et al. Is improvement of fatigue in rheumatoid arthritis a proper effect of biologics? Rom J Intern Med Rev Roum Med Int. 2021;59(1):58–65. doi:10.2478/rjim-2020-0028

27. Lee HJ, Pok LSL, Ng CM, et al. Fatigue and associated factors in a multi-ethnic cohort of rheumatoid arthritis patients. Int J Rheum Dis. 2020;23(8):1088–1093. doi:10.1111/1756-185X.13897

28. Almeida C, Choy EHS, Hewlett S, et al. Biologic interventions for fatigue in rheumatoid arthritis. Cochrane Database Syst Rev. 2016;2016(6):CD008334. doi:10.1002/14651858.CD008334.pub2

29. Bessette L, Haraoui B, Rampakakis E, Dembowy J, Trépanier MO, Pope J. Effectiveness of a treat-to-target strategy in patients with moderate to severely active rheumatoid arthritis treated with Abatacept. Arthritis Res Ther. 2023;25(1):183. doi:10.1186/s13075-023-03151-2

30. Inotai A, Rojkovich B, Fülöp A, Jászay E, Agh T, Mészáros A. Health-related quality of life and utility in patients receiving biological and non-biological treatments in rheumatoid arthritis. Rheumatol Int. 2012;32(4):963–969. doi:10.1007/s00296-010-1721-x

31. Fleischmann R, Mysler E, Bessette L, et al. Long-term safety and efficacy of upadacitinib or Adalimumab in patients with rheumatoid arthritis: results through 3 years from the SELECT-COMPARE study. RMD Open. 2022;8(1):e002012. doi:10.1136/rmdopen-2021-002012

32. Zhang X, Miao M, Zhang R, et al. Efficacy and safety of low-dose interleukin-2 in combination with methotrexate in patients with active rheumatoid arthritis: a randomized, double-blind, placebo-controlled Phase 2 trial. Signal Transduct Target Ther. 2022;7(1):67. doi:10.1038/s41392-022-00887-2

33. Ward MM, Madanchi N, Yazdanyar A, Shah NR, Constantinescu F. Prevalence and predictors of sustained remission/low disease activity after discontinuation of induction or maintenance treatment with tumor necrosis factor inhibitors in rheumatoid arthritis: a systematic and scoping review. Arthritis Res Ther. 2023;25(1):222. doi:10.1186/s13075-023-03199-0

34. Kang JH, Park DJ, Lee JW, et al. Drug survival rates of tumor necrosis factor inhibitors in patients with rheumatoid arthritis and ankylosing spondylitis. J Korean Med Sci. 2014;29(9):1205–1211. doi:10.3346/jkms.2014.29.9.1205

35. Gonzalez-Juanatey C, Vazquez-Rodriguez TR, Miranda-Filloy JA, et al. Anti-TNF-alpha-Adalimumab therapy is associated with persistent improvement of endothelial function without progression of carotid intima-media wall thickness in patients with rheumatoid arthritis refractory to conventional therapy. Mediators Inflamm. 2012;2012:674265. doi:10.1155/2012/674265

36. Raslan MA, Raslan SA, Shehata EM, et al. Different modalities to manage rheumatoid arthritis: an A to Z story. Future Sci OA. 2024;10(1):FSO968. doi:10.2144/fsoa-2023-0134

37. St Clair EW, van der Heijde DMFM, Smolen JS, et al. Combination of infliximab and methotrexate therapy for early rheumatoid arthritis: a randomized, controlled trial. Arthritis Rheum. 2004;50(11):3432–3443. doi:10.1002/art.20568

38. Lipsky PE, van der Heijde DM, St Clair EW, et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-tumor necrosis factor trial in rheumatoid arthritis with concomitant therapy study group. N Engl J Med. 2000;343(22):1594–1602. doi:10.1056/NEJM200011303432202

39. Genovese MC, Kalunian K, Gottenberg JE, et al. Effect of filgotinib vs placebo on clinical response in patients with moderate to severe rheumatoid arthritis refractory to disease-modifying antirheumatic drug therapy: the FINCH 2 randomized clinical trial. JAMA. 2019;322(4):315–325. doi:10.1001/jama.2019.9055

40. Glatt S, Taylor PC, McInnes IB, et al. Efficacy and safety of bimekizumab as add-on therapy for rheumatoid arthritis in patients with inadequate response to certolizumab pegol: a proof-of-concept study. Ann Rheum Dis. 2019;78(8):1033–1040. doi:10.1136/annrheumdis-2018-214943

41. Larsson I. Patients’ conceptions of their own influence on good treatment response to biological therapy in chronic inflammatory arthritis. Patient Prefer Adherence. 2017;11:1057–1067. doi:10.2147/PPA.S131239

42. Baumgart DC, Misery L, Naeyaert S, Taylor PC. Biological therapies in immune-mediated inflammatory diseases: can biosimilars reduce access inequities? Front Pharmacol. 2019;10:279. doi:10.3389/fphar.2019.00279

43. Salazar-Mejía CE, Galarza-Delgado DÁ, Colunga-Pedraza IJ, et al. Relationship between work productivity and clinical characteristics in rheumatoid arthritis. Reumatol Clínica Engl Ed. 2019;15(6):327–332. doi:10.1016/j.reumae.2017.12.002

44. Blomjous B, Boers M, Den Uyl D, et al. Predictors of sick leave and improved worker productivity after 52 weeks of intensive treatment in patients with early rheumatoid arthritis. Scand J Rheumatol. 2019;48(4):271–278. doi:10.1080/03009742.2019.1570549

45. Globe D, Mazonson P, Santas C, et al. Impact of etanercept treatment on absenteeism and productivity: the work loss and productivity survey. Journal of American Health and Drug Benefits. 2010;3(3):191–200.

46. Shim J, Jones GT, Pathan EMI, Macfarlane GJ. Impact of biological therapy on work outcomes in patients with axial spondyloarthritis: results from the British Society for Rheumatology Biologics Register (BSRBR-AS) and meta-analysis. Ann Rheum Dis. 2018;77(11):1578–1584. doi:10.1136/annrheumdis-2018-213590

47. Westhovens R, Van Der Elst K, Matthys A, Tran M, Gilloteau I. Sleep problems in patients with rheumatoid arthritis. J Rheumatol. 2014;41(1):31–40. doi:10.3899/jrheum.130430

48. Burmester GR, Cohen SB, Winthrop KL, et al. Safety profile of upadacitinib over 15 000 patient-years across rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis and atopic dermatitis. RMD Open. 2023;9(1):e002735. doi:10.1136/rmdopen-2022-002735

49. Burmester GR, Rigby WF, van Vollenhoven RF, et al. Tocilizumab in early progressive rheumatoid arthritis: FUNCTION, a randomised controlled trial. Ann Rheum Dis. 2016;75(6):1081–1091. doi:10.1136/annrheumdis-2015-207628

50. Cohen SB, van Vollenhoven RF, Winthrop KL, et al. Safety profile of upadacitinib in rheumatoid arthritis: integrated analysis from the SELECT phase III clinical programme. Ann Rheum Dis. 2021;80(3):304–311. doi:10.1136/annrheumdis-2020-218510

51. Combe B, Allanore Y, Alten R, et al. Comparative efficacy of subcutaneous (CT-P13) and intravenous infliximab in adult patients with rheumatoid arthritis: a network meta-regression of individual patient data from two randomised trials. Arthritis Res Ther. 2021;23(1):119. doi:10.1186/s13075-021-02487-x

52. Fleischmann RM, Genovese MC, Enejosa JV, et al. Safety and effectiveness of upadacitinib or Adalimumab plus methotrexate in patients with rheumatoid arthritis over 48 weeks with switch to alternate therapy in patients with insufficient response. Ann Rheum Dis. 2019;78(11):1454–1462. doi:10.1136/annrheumdis-2019-215764

53. Genovese MC, Kremer J, Zamani O, et al. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med. 2016;374(13):1243–1252. doi:10.1056/NEJMoa1507247

54. Genovese MC, van Adelsberg J, Fan C, et al. Two years of sarilumab in patients with rheumatoid arthritis and an inadequate response to MTX: safety, efficacy and radiographic outcomes. Rheumatol Oxf Engl. 2018;57(8):1423–1431. doi:10.1093/rheumatology/key121

55. Genovese MC, van Vollenhoven RF, Wilkinson B, et al. Switching from Adalimumab to tofacitinib in the treatment of patients with rheumatoid arthritis. Arthritis Res Ther. 2016;18:145. doi:10.1186/s13075-016-1049-3

56. Gerlag DM, Safy M, Maijer KI, et al. Effects of B-cell directed therapy on the preclinical stage of rheumatoid arthritis: the PRAIRI study. Ann Rheum Dis. 2019;78(2):179–185. doi:10.1136/annrheumdis-2017-212763

57. Hetland ML, Haavardsholm EA, Rudin A, et al. Active conventional treatment and three different biological treatments in early rheumatoid arthritis: Phase IV investigator initiated, randomised, observer blinded clinical trial. BMJ. 2020;371:m4328. doi:10.1136/bmj.m4328

58. Humby F, Durez P, Buch MH, et al. Rituximab versus tocilizumab in anti-TNF inadequate responder patients with rheumatoid arthritis (R4RA): 16-week outcomes of a stratified, biopsy-driven, multicentre, open-label, phase 4 randomised controlled trial. Lancet Lond Engl. 2021;397(10271):305–317. doi:10.1016/S0140-6736(20)32341-2

59. Ibrahim F, Lorente-Cánovas B, Doré CJ, et al. Optimizing treatment with tumour necrosis factor inhibitors in rheumatoid arthritis-a proof of principle and exploratory trial: is dose tapering practical in good responders? Rheumatol Oxf Engl. 2017;56(11):2004–2014. doi:10.1093/rheumatology/kex315

60. Kremer JM, Westhovens R, Leon M, et al. Treatment of rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein CTLA4Ig. N Engl J Med. 2003;349(20):1907–1915. doi:10.1056/NEJMoa035075

61. Leng X, Tang X, Hu P, et al. Safety, tolerability, pharmacokinetics, pharmacodynamics, and efficacy of WBP216, a novel IL-6 monoclonal antibody, in patients with rheumatoid arthritis: a phase Ia randomized placebo-controlled study. Front Immunol. 2022;13:1110992. doi:10.3389/fimmu.2022.1110992

62. Martin DA, Churchill M, Flores-Suarez L, et al. A phase Ib multiple ascending dose study evaluating safety, pharmacokinetics, and early clinical response of brodalumab, a human anti-IL-17R antibody, in methotrexate-resistant rheumatoid arthritis. Arthritis Res Ther. 2013;15(5):R164. doi:10.1186/ar4347

63. Smolen JS, Agarwal SK, Ilivanova E, et al. A randomised phase II study evaluating the efficacy and safety of subcutaneously administered ustekinumab and guselkumab in patients with active rheumatoid arthritis despite treatment with methotrexate. Ann Rheum Dis. 2017;76(5):831–839. doi:10.1136/annrheumdis-2016-209831

64. Smolen JS, Pangan AL, Emery P, et al. Upadacitinib as monotherapy in patients with active rheumatoid arthritis and inadequate response to methotrexate (SELECT-MONOTHERAPY): a randomised, placebo-controlled, double-blind phase 3 study. Lancet Lond Engl. 2019;393(10188):2303–2311. doi:10.1016/S0140-6736(19)30419-2

65. Strand V, Kaine J, Alten R, et al. Associations between patient global assessment scores and pain, physical function, and fatigue in rheumatoid arthritis: a post hoc analysis of data from phase 3 trials of tofacitinib. Arthritis Res Ther. 2020;22(1):243. doi:10.1186/s13075-020-02324-7

66. Strand V, Mysler E, Moots RJ, et al. Patient-reported outcomes for tofacitinib with and without methotrexate, or Adalimumab with methotrexate, in rheumatoid arthritis: a phase IIIB/IV trial. RMD Open. 2019;5(2):e001040. doi:10.1136/rmdopen-2019-001040

67. Strand V, Rentz AM, Cifaldi MA, Chen N, Roy S, Revicki D. Health-related quality of life outcomes of adalimumab for patients with early rheumatoid arthritis: results from a randomized multicenter study. J Rheumatol. 2012;39(1):63–72. doi:10.3899/jrheum.101161

68. Feist E, Fleischmann RM, Fatenejad S, et al. Olokizumab plus methotrexate: safety and efficacy over 106 weeks of treatment. Ann Rheum Dis. 2024;83(11):1454–1464. doi:10.1136/ard-2023-225473

69. Tan BE, Lim AL, Kan SL, et al. Real-world clinical experience of biological disease modifying anti-rheumatic drugs in Malaysia rheumatoid arthritis patients. Rheumatol Int. 2017;37(10):1719–1725. doi:10.1007/s00296-017-3772-8

70. Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75(1):3–15. doi:10.1136/annrheumdis-2015-207524

71. Messelink MA, Den Broeder AA, Marinelli FE, et al. What is the best target in a treat-to-target strategy in rheumatoid arthritis? Results from a systematic review and meta-regression analysis. RMD Open. 2023;9(2):e003196. doi:10.1136/rmdopen-2023-003196

72. Matcham F, Scott IC, Rayner L, et al. The impact of rheumatoid arthritis on quality-of-life assessed using the SF-36: a systematic review and meta-analysis. Semin Arthritis Rheum. 2014;44(2):123–130. doi:10.1016/j.semarthrit.2014.05.001

73. Favalli EG, Maioli G, Caporali R. Biologics or Janus Kinase inhibitors in rheumatoid arthritis patients who are insufficient responders to conventional anti-rheumatic drugs. Drugs. 2024;84(8):877–894. doi:10.1007/s40265-024-02059-8

74. Wlassits R, Müller M, Fenzl K, Lamprecht T, Erlacher L. JAK-Inhibitors – a story of success and adverse events. Open Access Rheumatol Res Rev. 2024;16:43–53. doi:10.2147/OARRR.S436637

75. Yamanaka H, Tanaka Y, Hibino T, et al. Lower injection-site reactions and long-term safety, immunogenicity, and efficacy of etanercept biosimilar YLB113: results from a post-hoc analysis of a double-blind, randomized, phase III comparative study and its open-label extension in patients with rheumatoid arthritis. Int J Rheum Dis. 2023;26(1):108–115. doi:10.1111/1756-185X.14462

76. Mansilla-Polo M, Morgado-Carrasco D. Biologics versus JAK inhibitors. Part II: risk of infections. a narrative review. Dermatol Ther. 2024;14(8):1983–2038. doi:10.1007/s13555-024-01203-2

77. Muehlensiepen F, May S, Hadaschik K, et al. Digitally supported shared decision-making and treat-to-target in rheumatology: a qualitative study embedded in a multicenter randomized controlled trial. Rheumatol Int. 2023;43(4):695–703. doi:10.1007/s00296-022-05224-y

78. Jonge MJ S-D, Weijers JM, Teerenstra S, et al. Patient involvement in rheumatoid arthritis care to improve disease activity-based management in daily practice: a randomized controlled trial. Patient Educ Couns. 2022;105(5):1244–1253. doi:10.1016/j.pec.2021.08.013

79. Zak A, Corrigan C, Yu Z, et al. Barriers to treatment adjustment within a treat to target strategy in rheumatoid arthritis: a secondary analysis of the TRACTION trial. Rheumatol Oxf Engl. 2018;57(11):1933–1937. doi:10.1093/rheumatology/key179

80. Schalet BD, Revicki DA, Cook KF, Krishnan E, Fries JF, Cella D. Establishing a common metric for physical function: linking the HAQ-DI and SF-36 PF subscale to PROMIS(®) physical function. J Gen Intern Med. 2015;30(10):1517–1523. doi:10.1007/s11606-015-3360-0

81. Gwinnutt JM, Sharp CA, Symmons DPM, Lunt M, Verstappen SMM. Baseline patient reported outcomes are more consistent predictors of long-term functional disability than laboratory, imaging or joint count data in patients with early inflammatory arthritis: a systematic review. Semin Arthritis Rheum. 2018;48(3):384–398. doi:10.1016/j.semarthrit.2018.03.004

82. Greenwood MC, Hakim AJ, Doyle DV. A simple extension to the rheumatoid arthritis quality of life questionnaire (RAQol) to explore individual patient concerns and monitor group outcome in clinical practice. Rheumatol Oxf Engl. 2006;45(1):61–65. doi:10.1093/rheumatology/kei099

83. Lubeck DP. Patient-reported outcomes and their role in the assessment of rheumatoid arthritis. PharmacoEconomics. 2004;22(2 Suppl 1):27–38. doi:10.2165/00019053-200422001-00004

84. England BR, Thiele GM, Anderson DR, Mikuls TR. Increased cardiovascular risk in rheumatoid arthritis: mechanisms and implications. BMJ. 2018;361(k1036). doi:10.1136/bmj.k1036

85. Nagy G, Roodenrijs NMT, Welsing PMJ, et al. EULAR points to consider for the management of difficult-to-treat rheumatoid arthritis. Ann Rheum Dis. 2022;81(1):20–33. doi:10.1136/annrheumdis-2021-220973

86. Chen J, Norling LV, Cooper D. Cardiac dysfunction in rheumatoid arthritis: the role of inflammation. Cells. 2021;10(4):881. doi:10.3390/cells10040881

87. Taylor PC, Atzeni F, Balsa A, Gossec L, Müller-Ladner U, Pope J. The key comorbidities in patients with rheumatoid arthritis: a narrative review. J Clin Med. 2021;10(3):509. doi:10.3390/jcm10030509

88. Rodríguez-Vargas GS, Santos-Moreno P, Rubio-Rubio JA, et al. Vascular age, metabolic panel, cardiovascular risk and inflammaging in patients with rheumatoid arthritis compared with patients with osteoarthritis. Front Cardiovasc Med. 2022;9:894577. doi:10.3389/fcvm.2022.894577

89. Simon TA, Dong L, Winthrop KL. Risk of opportunistic infections in patients with rheumatoid arthritis initiating Abatacept: cumulative clinical trial data. Arthritis Res Ther. 2021;23(1):17. doi:10.1186/s13075-020-02399-2

90. Lee RH, Sloane R, Pieper C, et al. Glycemic control and insulin treatment alter fracture risk in older men with type 2 diabetes mellitus. J Bone Miner Res Off J Am Soc Bone Miner Res. 2019;34(11):2045–2051. doi:10.1002/jbmr.3826

91. Abdulmajid B, Blanken AB, van Geel EH, Daams JG, Nurmohamed MT. Effect of TNF inhibitors on arterial stiffness and intima media thickness in rheumatoid arthritis: a systematic review and meta-analysis. Clin Rheumatol. 2023;42(4):999–1011. doi:10.1007/s10067-023-06505-y

92. Mitrović J, Hrkač S, Tečer J, et al. Pathogenesis of extraarticular manifestations in rheumatoid arthritis-a comprehensive review. Biomedicines. 2023;11(5):1262. doi:10.3390/biomedicines11051262

93. Sherbaevna SR, Tashalievich MA, Momunovna AR, et al. The spectrum of airway disease associated with rheumatoid arthritis. Curr Respir Med Rev. 2022;18(3):179–189. doi:10.2174/1573398X18666220509153713

94. Rezuș E, Macovei LA, Burlui AM, Cardoneanu A, Rezuș C. Ischemic heart disease and rheumatoid arthritis-two conditions, the same background. Life Basel Switz. 2021;11(10):1042. doi:10.3390/life11101042

95. Manders SHM, Kievit W, Adang E, et al. Cost-effectiveness of Abatacept, rituximab, and TNFi treatment after previous failure with TNFi treatment in rheumatoid arthritis: a pragmatic multi-centre randomised trial. Arthritis Res Ther. 2015;17(1):134. doi:10.1186/s13075-015-0630-5

96. Khosrow-Khavar F, Kim SC, Lee H, Lee SB, Desai RJ. Tofacitinib and risk of cardiovascular outcomes: results from the safety of TofAcitinib in routine care patients with rheumatoid arthritis (STAR-RA) study. Ann Rheum Dis. 2022;81(6):798–804. doi:10.1136/annrheumdis-2021-221915

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.