Abstract
The association between chronic inflammation and increased risk of cardiovascular disease in rheumatoid arthritis (RA) is well established. In the general population, inflammation is an established independent risk factor for cardiovascular disease, and much interest is placed on controlling inflammation to reduce cardiovascular events. As inflammation encompasses numerous pathways, the development of targeted therapies in RA provides an opportunity to understand the downstream effect of inhibiting specific pathways on cardiovascular risk. Data from these studies can inform cardiovascular risk management in patients with RA, and in the general population. This Review focuses on pro-inflammatory pathways targeted by existing therapies in RA and with mechanistic data from the general population on cardiovascular risk. Specifically, the discussions include the IL-1, IL-6 and TNF pathways, as well as the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) signalling pathway, and the role of these pathways in RA pathogenesis in the joint alongside the development of atherosclerotic cardiovascular disease. Overall, some robust data support inhibition of IL-1 and IL-6 in decreasing the risk of cardiovascular disease, with growing data supporting IL-6 inhibition in both patients with RA and the general population to reduce the risk of cardiovascular disease.
Introduction
Targeting inflammation is an established and effective strategy for the treatment of rheumatoid arthritis (RA) that has been in place since the development of the first biologic therapy in the late 1990s1-3. Historically, the treatment of atherosclerotic cardiovascular disease (ASCVD) has focused on the management of traditional risk factors such as arterial hypertension and hypercholesterolaemia4,5. However, large-scale randomized controlled trials in the past few years have now established inflammation as a modifiable risk factor to be considered in the prevention of the development and progression of atherosclerosis in the general population4,6,7. With the increased adoption of targeted biologic therapies to reduce the risk of ASCVD, summarizing and examining these pathways and their roles in the pathogenesis of RA and ASCVD have become increasingly important for the management of both RA and ASCVD.
RA can be considered a human model of inflammation and can inform the impact of chronically increased levels of inflammation in the general population. Indeed, studies on RA have linked inflammation to an 1.5× increased risk of cardiovascular morbidity and mortality8-11. Evidence is also available showing direct effects of inflammation on the vascular endothelium as well as interactions between systemic inflammation with classic cardiovascular risk factors, further contributing to excess cardiovascular risk12,13. The inflammatory pathophysiology of RA shares important links with that of atherogenesis and atherothrombosis, with inflammation promoting the progression of atherosclerosis14-16, leading to a range of cardiovascular outcomes such as venous thromboembolism, heart failure and ASCVD; the latter, ASCVD, is the focus of this Review. From studies of targeted therapies in RA, evidence suggests that targeting certain pathways might have a more beneficial effect on cardiovascular risk than others17-19. Thus, recent studies in RA in the past few years have informed the cardiovascular literature. In turn, large randomized controlled trials (RCTs) in the general population can help to inform the management of ASCVD in patients with RA, which is otherwise a major challenge in RA owing to the difficulty in achieving the sample size needed for a sufficient number of hard cardiovascular endpoints in RA-only RCTs.
As the convergence of drug targets for RA and cardiovascular disease is a relatively new development, limited reviews are available that summarize the shared pathways and downstream effects in one resource. Thus, in this Review, we seek to address this gap and will focus on shared pathways for existing targeted therapies in RA where mechanistic data are available from both the RA and ASCVD literature, specifically, the IL-1, IL-6, and TNF and JAK–STAT pathways, where emerging data are available20. A more detailed understanding of shared pathways between RA and cardiovascular disease can better inform the management of RA for individuals with or at higher risk of ASCVD, as well as the management of inflammation for individuals in the general population with an increased cardiovascular risk owing to inflammation. In this Review, we provide a comparative overview of the pathways shared by RA and ASCVD and the known mechanistic pathways for the pathogenesis of both conditions using data from selected mechanistic, clinical trials and observational cohort studies. Last, we highlight areas for future research that might inform treatment strategies that can control disease activity in RA and mitigate the risk of ASCVD.
The IL-1 pathway
IL-1 was first characterized in the 1970s as an endogenous pyrogen that induced fevers and a mononuclear cell factor involved in tissue destruction in RA21. Eventually, examinations of synovial explants from patients with RA led to the characterization of IL-1 (refs. 21,22). Although the IL-1 family consists of 11 members, IL-1 typically refers to IL-1α and IL-1β (which are both pro-inflammatory cytokines involved in inflammasome signalling23). The IL-1 receptor antagonist (IL-1Rα) is an endogenous inhibitor that blocks the actions of IL-1α and IL-1β.
IL-1 in RA
Mice deficient in IL-1Rα develop spontaneous erosive arthritis24. Anakinra, an analogue of IL-1Rα, is approved for use in RA; however, this drug is less frequently used than other biologic therapies owing to its limited efficacy25,26. The limited efficacy has been attributed in part to a bypass of IL-1-dependent signalling through the adaptor protein MyD88, providing a mechanism for ongoing synovial Toll-like receptor (TLR) signalling27. IL-1 can stimulate receptor activator of nuclear factor kappa-B ligand (RANKL), linked to osteoclast activation, and certain matrix metalloproteinases (MMPs, including MMP1, MMP3 and MMP13), which are both involved in bone and cartilage destruction28-31 (Fig. 1a). In patients with RA, elevated plasma concentrations of IL-1 correlate with higher disease activity32,33.
IL-1 in ASCVD
The inflammasome, and specifically the IL-1β pathway, has multiple effects in many stages of atherosclerosis and ASCVD34. IL-1β induces the expression of adhesion factors, such as intercellular adhesion molecule (ICAM-1) and vascular cell adhesion molecule (VCAM-1), and chemokines, including monocyte chemoattractant protein (MCP1), on endothelial cells and promotes inflammatory cell recruitment into the vasculature via MCP1 (refs. 35,36). Enhanced expression of IL-1 itself can form a positive feedback loop and generate the production of IL-6 and MMPs, upon which IL-6 activates the acute phase response37. Certain matrix metalloproteins (MMPs, also known as collagenases), such as MMP1, MMP8 and MMP13, can disrupt the atherosclerotic fibrous cap, increasing susceptibility to rupture and thrombus formation38,39 (Fig. 1a). In an established atheroma, neutralization of IL-1β promotes monocytes to switch to an anti-inflammatory state and secrete IL-10 (a cytokine associated with beneficial plaque remodelling40).
IL-1 has an important role in ischaemia–reperfusion injury and, in mouse models, overexpression of IL-1Rα by gene transfection protects the myocardium by reducing levels of apoptosis and attenuating inflammation41. These findings and others provided the rationale for further studies evaluating the effect of IL-1 inhibition after acute myocardial infarction in humans42-47 (Table 1). Promising results have now led to a larger ongoing randomized clinical trial that will evaluate IL-1 blockade versus placebo in the treatment of patients with acute ST-segment elevation myocardial infarction (NCT04463251)48,49.
Table 1 ∣.
Target (drug) | Trial name | Dosing | Follow-up | Population | Primary end point | Outcome |
---|---|---|---|---|---|---|
IL-1 (anakinra) | MRC-ILA Heart44 | 100 mg daily | 7 days | NSTE-ACS | Change in hsCRP | Decrease in hsCRP |
VCU-ART45 and VCU-ART 246 | 100 mg daily | 14 days | STEMI | Change in LVESVi | No change in LVESVi; decrease in hsCRP | |
VCU-ART347 | 100 mg daily or 100 mg twice daily | 14 days | STEMI | Change in hsCRP | Decrease in hsCRP | |
IL-1β (canakinumab) | CANTOS6 | 50 mg, 150 mg, 300 mg every 3 months | 3 years | Prior myocardial infarction and hsCRP levels over 2 mg/l | MACE | 150-mg dose: decrease in recurrent cardiovascular events; no change in all-cause mortality |
IL-6R (tocilizumab) | STAT-MI184 | Single dose of 162 mg | 30 days | Acute coronary syndrome | MACE | No changes; trial stopped because of lack of effect |
NCT01491074 78 | Single dose of 280 mg | 1–3 days | NSTEMI | AUC for hsCRP and hsTnT | Reduction in median AUC for hsCRP and hsTNT compared with placebo | |
NCT01491074 185 | Single dose of 280 mg | 1–3 days, 3 and 6 months | NSTEMI | Inflammatory markers (27 cytokines) | Increase in serum levels of MIP1B and IP-10 | |
ASSAIL-MI79 | Single dose of 280 mg | 3–7 days | Acute coronary syndrome | Myocardial salvage index from cardiac MRI (3–7 days post-myocardial infarction) | Increase in myocardial salvage; no change in final infarct size | |
NCT05350592 186 | Single dose of 280 mg | 48 h | Acute myocardial infarction and cardiogenic shock | NT-proBNP | Trial underway | |
IL-6 (ziltivekimab) | ZEUS92 | 15 mg every month | 4 years | ASCVD and chronic kidney disease | MACE | Trial underway |
TNF (etanercept) | RENEWAL128 | 25 mg once, twice or three times a week | 24 weeks | NYHA III–IV heart failure | Composite death or heart failure hospitalization | No effect |
TNF (infliximab) | ATTACH127 | 5 mg/kg or 10 mg/kg at 0, 2 and 6 weeks | 14 weeks | NYHA III–IV heart failure | Clinical composite score | No effect on primary end point; high dose associated with worse outcome at 28 weeks |
Trials for heart failure included only for TNF inhibitors. aPWV, aortic pulse wave velocity; ASCVD, atherosclerotic cardiovascular disease; AUC, area under the curve; FMDBA, brachial artery flow-mediated dilation; hsCRP, high-sensitivity C-reactive protein; hsTNT, high-sensitivity cardiac troponin T; IP-10, interferon gamma-inducible protein; LVESVi, left ventricle end systolic volume index; MACE, major adverse cardiovascular events; MIP1b, macrophage inflammatory protein-1b; MMP8, matrix metalloproteinase 8; NSTE-ACS, non-ST-segment elevation acute coronary syndrome; NSTEMI, non-ST-elevation myocardial infarction; NT-proBNP, N-terminal pro B-type natriuretic peptide; STEMI, ST-elevation myocardial infarction.
Several IL-1-targeting biologic agents are available that have shown beneficial effects in cardiovascular conditions: these include anakinra, a recombinant IL-1 receptor antagonist that blocks IL-1α and IL-1β; rilonacept, a fusion protein that functions as a trap receptor for IL-1α, IL-1β and IL-1Rα; and canakinumab, an IgG human monoclonal that targets IL-1β (Fig. 1b, Table 1). However, only anakinra is approved for the treatment of RA50. In early-phase trials, rilonacept had some limited efficacy in RA51, and canakinumab has some beneficial effects in reducing RA disease activity52, but neither was ultimately pursued as a treatment for RA. Canakinumab was the focus of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS), the first large-scale proof-of-principle trial to validate the inflammation hypothesis in human atherosclerosis6 (Table 2). The CANTOS trial enrolled over 10,000 individuals with a history of myocardial infarction and a high sensitivity C-reactive protein (hsCRP) test result >2 mg/1; the hsCRP test cut-off was chosen as the median cut-off for the general population, pre-selecting for individuals who had a residual inflammatory burden despite guidelines-directed medical therapy. Individuals were randomly assigned to treatment with either canakinumab or a placebo. All participants received post-myocardial infarction standard-of-care treatment, including statin therapy. The results demonstrated an overall 15% reduction in major cardiovascular events, defined as myocardial infarction, stroke, or cardiovascular death, with canakinumab therapy. Additionally, a post hoc analysis of the CANTOS trial found that the magnitude of cytokine reduction was associated with extent of cardiovascular risk reduction; individuals with a greater than average reduction in levels of IL-6 or hsCRP (known as ‘cytokine responders’) had fewer cardiovascular events than the other study participants, with a 30% reduction in cardiovascular and all-cause mortality in this subset53,54.
Table 2 ∣.
Pathway | Target | Drug name | ASCVD in RA | ASCVD general population |
Comments |
---|---|---|---|---|---|
IL-1 | IL-1 | Anakinra | – | Decreased | Surrogate endpoints only |
IL-1β | Canakinumab | – | Decreased | CANTOS trial6 | |
IL-6 | IL-6R | Tocilizumab and sarilumab | Decreased | Decreased | Surrogate endpoints only |
IL-6 | Ziltivekimab | – | Unclear (studies ongoing) | ZEUS trial underway92 | |
TNF | TNF | Adalimumab, certolizumab, etanercept, golimumab and infliximab | Decreased | Uncleara | Reduced risk of ASCVD risk in patients with RA (majority observational data only); TARGET trial186 |
JAK–STAT | All JAK members | Tofacitinib | Increased (safety signal requires further investigation) | – | ORAL Surveillance20 |
JAK1 and JAK2 | Baricitinib | – | – | – | |
JAK1 only | Upadacitinib | – | – | – | |
Filgotinib | – | – | Approved in the EU and Japan for the treatment of RA, but not approved in the USA |
ASCVD, atherosclerotic cardiovascular disease; RA, rheumatoid arthritis; –, insufficient data to comment.
Most studies were in patients with heart failure and not ASCVD.
The IL-6 pathway
IL-6 is a pleiotropic cytokine with broad effects in immunity, inflammation and haematopoiesis. Multiple names were given for IL-6 until sequencing of cDNA clones revealed that these molecules were the same protein55-57. Interest in targeting IL-6 for the treatment of RA grew with the discovery that IL-6 knockout mice were resistant to antigen-induced experimental arthritis58,59. In vascular biology, IL-6 is secreted by multiple cell-types implicated in atherothrombosis, including macrophages, monocytes, fibroblasts and endothelial cells37. IL-6 is typically secreted by cells following IL-1 stimulation, and has three predominant signalling mechanisms: ‘classical’ IL-6 signalling, known as cis-signalling, where secreted IL-6 binds to the membrane bound IL-6 receptor (IL-6R), which subsequently binds to a second membrane-bound ubiquitous protein known as gp130 to activate intracellular signalling; trans-signalling occurs when IL-6 binds to soluble IL-6R and is associated with a signal-transducing glycoprotein 130 (gp130) to activate signalling; and trans-presentation is the last mechanism of IL-6 signalling that requires interaction between a dendritic cell and a T cell60-62.
IL-6 in RA
In the RA synovium, IL-6 is one of the most abundantly expressed cytokines. Levels of both IL-6 and soluble IL-6R correlate with histological evidence of synovial inflammation in patients with RA63,64. IL-6, through induction of vascular endothelial growth factor (VEGF), can further promote joint inflammation and damage by increasing vascular permeability and the migration of endothelial cells37. IL-6 also induces osteoclast recruitment, contributing to the destruction of bone and cartilage65 (Fig. 1a). IL-6 and sIL-6R trans-signalling can influence osteoclastogenesis through different pathways, inducing RANKL and osteoprotegerin (OPG)66. Human synovial fluid from patients with RA containing high levels of IL-6 and sIL-6R can stimulate the formation of osteoclast-like cells from murine osteoblastic cells, and this process is inhibited by adding an anti-IL-6R antibody67. Two approved therapies for RA, tocilizumab and sarilumab, are monoclonal antibodies that inhibit both the membrane and the soluble form of IL-6R68,69.
IL-6 might be associated with a higher risk of cardiovascular disease, independent of traditional cardiovascular risk factors, in patients with RA70,71. Despite observed improvements in articular and systemic inflammation with IL-6 inhibition, treatment was also associated with lipid profile changes across various clinical trials, including increases in LDL cholesterol (LDL-C) of 12–20%, as well as increases in total cholesterol and triglycerides (all typically associated with a pro-atherogenic profile), but also increases in HDL cholesterol (HDL-C; associated with an anti-atherogenic profile)72. In the MEASURE trial, the effects of tocilizumab on biomarkers of cardiovascular disease in patients with RA were evaluated73. Although the levels of LDL-C increased with tocilizumab therapy, the levels of more atherogenic lipoproteins, such as oxidized LDL, did not increase and pro-atherogenic proteins such as serum amyloid A, secretory phospholipase A2 and lipoprotein(a), decreased73. Smaller observational studies suggest that IL-6R blockade might also promote the levels and function of anti-atherogenic HDL particles74,75. The ENTRACTE trial was a non-inferiority phase IV randomized clinical trial that compared the risk of major cardiovascular events in patents with RA receiving tocilizumab treatment versus those patients receiving treatment with the TNF inhibitor etanercept76. A total of 3,080 participants with inadequate response to conventional DMARDs and at least one cardiovascular risk factor were enrolled and randomized in a 1:1 fashion. Although circulating concentrations of total cholesterol, LDL-C and triglycerides did increase in the tocilizumab-treatment group compared with the etanercept-treatment group, no concomitant increase in cardiovascular events was observed over the 5-year study period76. Together, these data suggest that tocilizumab treatment might have an overall neutral effect on ASCVD risk when compared with etanercept treatment, as indicated by the overall changes in both atherogenic and atheroprotective lipids along with a reduction in systemic inflammation.
IL-6 in ASCVD
A role for IL-6 reduction in decreasing the risk of cardiovascular disease was observed in 2017 in the aforementioned CANTOS trial, which targeted inflammation through IL-1α inhibition6. Further analysis of the CANTOS study data revealed that the magnitude of IL-6 reduction correlated with the overall reduction in major cardiovascular events77. In fact, in those participants who did not experience reductions in IL-6 levels with canakinumab treatment, the number of cardiovascular events did not decrease and remained similar to the levels observed in the placebo group. IL-6 is also upregulated during myocardial infarction and can affect myocardial remodelling and plaque destabilization77 (Fig. 1a). In two separate studies, a single dose of tocilizumab resulted in higher levels of CRP and cardiac troponin T in patients experiencing a non-ST-segment elevation myocardial infarction and in patients following an ST-segment elevation myocardial infarction78,79. Furthermore, in one of these studies (the ASSAIL-MI trial), a single dose of tocilizumab administered prior to a percutaneous coronary intervention showed the potential to reduce myocardial damage from ischaemia, as indicated by an increase in the myocardial salvage index (the difference between the actual and potential infarct size defined by the initial area at risk by coronary occlusion, as measured by cardiac MRI) and a reduction in circulating CRP concentrations78,79 (Table 1).
Genomic studies also support a key role for IL-6 in ASCVD. A known variant in IL6R (that leads to a aspartic acid to alanine substitution at position 358 (Asp358Ala) of the encoded protein) hinders the cell surface expression of membrane-bound IL6R, thus reducing IL-6 signalling in individuals with this variant80. Mendelian randomization studies reported an association of this variant with reduced risk of ASCVD81,82, an association that was later confirmed in follow-up genetic association studies that considered a variety of cardiovascular phenotypes, including myocardial infarction and peripheral arterial disease83-85.
Although mounting data are available suggesting that direct inhibition of IL-6 itself could maximize the anti-inflammatory benefit of inhibiting the IL-6 pathway in atherosclerosis, not all IL-6 antagonists likely share the same profile. Sirukumab, an IL-6 inhibitor that targets the cytokine rather than the cytokine receptor, did not receive US Food and Drug Administration (FDA) approval for the treatment of RA owing to concerns regarding the mortality attributed to cardiovascular events and malignancy in the treatment group of an ongoing clinical trial86; the mechanism for this safety signal is not known. By contrast, ziltivekimab, a humanized monoclonal antibody that blocks IL-6, had promising effects on reducing the cardiovascular risk of individuals in a 2021 randomized double-blind placebo-controlled phase II trial, known as RESCUE87-90 (Fig. 1b, Table 1). RESCUE evaluated the effects of ziltivekimab on biomarkers of inflammation and thrombosis in patients at a high cardiovascular risk, enrolling 264 patients with chronic kidney disease and increased levels of hsCRP (>2 mg/dl)91. Over a period of 24 weeks, ziltivekimab treatment resulted in a reduction in the levels of various biomarkers of inflammation and thrombosis, including hsCRP, lipoprotein(a), fibrinogen and soluble phospholipase A2 (sPLA2). Moreover, no notable effect was observed for ziltivekimab on the total cholesterol to high-density lipoprotein cholesterol ratio, unlike previous reports for other IL-6 antagonists87. Notably, however, prior studies on IL-6 blockade were performed largely in patients with RA, where factors such as higher starting levels of inflammation might influence changes in lipid levels. The ziltivekimab phase II findings laid the foundation for the current phase III study, the ZEUS study, which will compare ziltivekimab with placebo in approximately 6,000 patients with chronic kidney disease and elevated hsCRP levels to determine whether IL-6 inhibition reduces the rate of cardiovascular events92 (Table 2).
The TNF pathway
TNF is a pleiotropic cytokine that signals through two different receptors: TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). TNFR1 is ubiquitously expressed and present on most cells, whereas TNFR2 has more limited expression and is mainly expressed on certain immune cells (such as some T cell and B cell subsets) as well as fibroblasts, endothelial cells and neuronal cell subsets93. In general, signalling through soluble TNFR1 triggers pro-inflammatory pathways whereas TNFR2 signalling has an important role in immune modulation94.
TNF in RA
TNF is the most well-known and targeted cytokine in RA25,95. Overexpression of a TNF transgene in mouse models can induce spontaneous erosive inflammatory arthritis and inhibition of TNF via various methods, and can suppress arthritis in this model96,97. In fact, inhibition of TNF can ameliorate disease in several animal models of arthritis, including antigen-induced arthritis, adjuvant arthritis and the aforementioned human TNF transgene-induced arthritis98. In the human synovium, TNF can induce the proliferation of fibroblast-like synoviocytes and trigger cartilage destruction by inducing the production of collagenase by synovial cells, osteoclastogenesis and bone resorption99-101 (Fig. 1a). TNF inhibition can rapidly decrease levels of IL-6 and acute-phase reactants, suppress leukocyte migration and endothelial cell activation and improve regulatory T cell function102,103.
Five TNF inhibitors are available for the treatment of RA. In order of development, infliximab is a chimeric anti-TNF antibody comprising both a mouse Fc fragment and a human IgG1 antibody104,105. Etanercept is a recombinant IgG1 Fc fragment fused to two p75 TNF receptors106,107. Adalimumab is a fully humanized recombinant IgG1 antibody against TNF108. Golimumab and certolizumab are the two most recently approved TNF inhibitors and are both humanized anti-TNF monoclonal antibodies109-111. Multiple observational studies have demonstrated an association between TNF inhibitor use and reduction in cardiovascular events in patients with RA112-115 and most recently one trial (the TARGET trial) demonstrated a reduction in vascular inflammation with TNF inhibitor therapy116 (Table 2).
TNF in ASCVD and other cardiac conditions
In the general population, TNF inhibition has a more complicated history. Whether TNF inhibition can reduce the risk of cardiovascular disease in the general population has not been studied and limited studies are available regarding the relationship between the TNF pathway and ASCVD risk in the general population117-119. TNF has multiple effects on the endothelium, including promoting the production of reactive oxygen species (ROS), reducing the bioavailability of nitric oxide and increasing endothelial permeability117,120,121 (Fig. 1a); furthermore, TNF is present in human atherosclerotic plaques122. In some mouse models of atherosclerosis, deletion of Tnf can attenuate the development of disease123,124. Yet, other mouse models have yielded conflicting results125,126. In large-scale observational studies, higher plasma concentrations of TNF were associated with the recurrence of cardiovascular events even after adjusting for classic cardiovascular risk factors118. The most-studied effect of TNF inhibition in cardiovascular disease relates to the effects on heart failure127,128.
In experimental settings, TNF can induce cardiomyopathy and left ventricular dysfunction119,129. However, in trials of TNF inhibition (with etanercept and infliximab) in patients with established systolic heart failure, treatment had no beneficial effect on recurrent hospitalization or death (Table 1). Further, a higher hospitalization rate was observed in a subgroup of patients that received infliximab 10 mg/kg compared with placebo127,128. Thus, the data suggested that global TNF inhibition could cause dysregulation in both adverse and protective signalling in a failing heart and this treatment is generally avoided in patients with systolic heart failure130. However, it is important to note that these studies were performed in the general population and not in RA specifically, where TNF has a strong pathogenic role.
The JAK–STAT pathway
The JAK–STAT pathway is activated downstream of multiple pro-inflammatory cytokines and is one of the newest targets for RA. Studies are ongoing to address concerns regarding the association between JAK inhibition and risk of cardiovascular events20. The JAK-STAT pathway consists of four JAK proteins (JAK1, JAK2, JAK3 and TYK2) and seven STAT proteins. Activation of JAK–STAT requires a cytokine that binds to its receptor, which results in JAK phosphorylation, STAT binding and STAT activation. The activated STAT protein translocates to the nucleus and binds to enhancers to induce gene transcription131. Downstream effects of the JAK–STAT pathway are broad, ranging from immune cell development to the induction or prevention of apoptosis132,133.
JAK inhibition in RA
In RA, the JAK–STAT pathway is constitutively elevated and various proteins that function to inhibit the JAK–STAT pathway are thought to be deficient134. Blockade of the JAK–STAT pathway can improve clinical symptoms and slow disease progression in patients with RA135. To date, three oral JAK inhibitors are approved for the treatment of RA in the USA: tofacitinib, baricitinib and upadacitinib135. Filgotinib is also approved for the treatment of RA in Europe and Japan, but approval was rejected by the FDA in 2020 (ref. 136). Each agent differs in terms of its JAK receptor selectivity, although studies in RA have suggested that JAK1 inhibition is largely responsible for the efficacy of these drugs in RA137. Tofacitinib is a non-specific JAK inhibitor that has greater selectivity for JAK1 and JAK3 than for JAK2 and TYK2 (refs. 138-140); baricitinib targets JAK1 and JAK2, with some activity against TYK2 but minimal activity against JAK3; whereas upadacitinib and filgotinib are both JAK1 selective agents137,141,142. Inhibition of the JAK pathway can affect the signalling of multiple cytokine pathways, including IL-6 and TNF, which depends on the JAK selectivity of the agent135 (Fig. 1b).
In 2022, the cardiovascular safety of JAK inhibitors in RA was called into question with results from a post-marketing safety surveillance study of tofacitinib20. The ORAL Surveillance study was a prospective, phase IIIb/IV randomized, open-label, non-inferiority trial of major adverse cardiovascular events and malignancy in patients with RA receiving tofacitinib versus TNF inhibitors (etanercept or adalimumab). The trial, mandated by the FDA, enrolled patients with RA with an inadequate response to methotrexate, who were over 50 years old and had at least one cardiovascular risk factor (such as cigarette smoking, hypertension, hyperlipidaemia, diabetes mellitus, family history of premature coronary artery disease, prior history of coronary artery disease or extra-articular disease associated with RA). Patients were randomly assigned to receive tofacitinib (5 mg or 10 mg twice daily) or a TNF inhibitor. During the interim data analysis, tofacitinib 10 mg twice daily was associated with a higher risk of venous thromboembolism than treatment with a TNF inhibitor. As a result, the tofacitinib dose for all study participants was reduced to 5 mg twice daily. The results of ORAL Surveillance indicated a safety signal for increased major adverse cardiovascular events in tofacitinib versus TNF inhibition, on the basis of a pre-specified non-inferiority criterion (Table 2). Malignancy rates were also higher in the tofacitinib group for both doses than in the TNF inhibitor group. The signal for major adverse cardiovascular events was higher among patients who were older than 65 years, patients who had ever smoked and aspirin users143. Whether these results are specific to tofacitinib and not more selective JAK inhibitors such as upadacitinib, or whether the effect is specific to RA and not other indications for JAK inhibitors, remains to be seen. Furthermore, although tofacitinib treatment is associated with an increase in lipid levels138,140,144, the relevance of this effect on cardiovascular risk is less clear; one small mechanistic prospective study demonstrated that tofacitinib might have beneficial effects on atherosclerosis as measured by regression of carotid intima media thickness in patients who had elevated levels of carotid intima media thickness at baseline145. Another small study demonstrated that treatment with tofacitinib for 6 weeks was associated with an increase in HDL-C and LDL-C concentrations. Further functional studies of HDL suggested that the treatment led to a more anti-atherogenic profile of HDL146.
Notably, prior to the ORAL Surveillance study, in six phase III studies and two open-label long-term extension studies of tofacitinib in patients with RA, the overall incidence of cardiovascular events was low147. In a post hoc analysis that combined the phase III and long-term extension studies, higher levels of HDL-C were associated with lower rates of subsequent cardiovascular events. A meta-analysis of 26 RCTs found no difference in the rate of cardiovascular events among patients with RA being treated with a JAK inhibitor compared with placebo148. The STAR-RA population-based study evaluated patients initiating treatment with tofacitinib or a TNF inhibitor using real-world observational data from commercial and Medicare data149. The analysis found no difference in rates of myocardial infarction or stroke incidence in the overall population. However, when the analysis was restricted to patients with cardiovascular risk factors using the same criteria as the ORAL Surveillance study, a similar numerical increase in cardiovascular events was observed149,150. Thus far, JAK inhibition has been consistently associated with a signal for an elevated risk of cardiovascular events among older individuals with cardiovascular risk factors when compared with TNF inhibition. Further studies are needed to understand the absolute risk for a given patient and the underlying mechanisms151.
JAK inhibition in ASCVD
The JAK–STAT pathway has an important role in atherosclerosis, as is evident from the cytokines impacted by JAK inhibition involved in this disease (such as IL-6 and TNF, as discussed in the previous sections). For instance, IL-6 activates the JAK–STAT pathway after binding to its receptor152. IFNγ can also augment atherosclerosis and signals through both JAK1 and JAK2 (ref. 153). However, not every JAK–STAT process is pro-inflammatory. For instance, although STAT3 can promote the expression of angiotensinogen II (a known mediator of cardiac hypertrophy) by binding to its gene promoter in myocytes154,155, STAT3 can also mediate cardioprotective signalling in myocytes when activated by IL-10 (ref. 156). Unlike IL-1, IL-6 and TNF, no large-scale trials have yet assessed the effects of JAK inhibition in the prevention of cardiovascular events in the general population, although a multi-STAT inhibitory strategy has been proposed as a potential therapeutic avenue157. Future mechanistic investigations to understand the role of JAK inhibition in promoting or mitigating cardiovascular disease will be crucial to understanding how this class of drugs could be used in the future.
Broad inhibition of multiple cytokines
The majority of this Review focuses on the key cytokine pathways involved in both RA and ASCVD that are the subject of approved targeted therapies, whereas in this section we discuss methotrexate, a nonspecific drug that has been studied in both patients with RA and the general population for reducing the risk of ASCVD and that is believed to target the pathways downstream of IL-6, TNF and IL-1 (Fig. 1a). First developed as an anticancer therapy for childhood leukaemia, this drug was also found to inhibit the proliferation of connective tissue, leading to the first study of this drug in RA in 1951 (ref. 158). Methotrexate is a dihydrofolate reductase inhibitor that, when given at high doses, blocks the synthesis of DNA nucleotides159. For example, when used as part of a chemotherapy regimen for lymphoma, an individual could receive 5,600 mg of intravenous methotrexate in one dose160. In RA, this drug is used at much lower doses, for example, 25 mg once a week, together with folic acid supplementation, to reduce the toxicity profile of this drug without reducing its anti-inflammatory efficacy161. Various mechanisms have been proposed for the anti-inflammatory effects of methotrexate; in addition to the anti-metabolite effect of this drug, methotrexate can lower the production of key pathogenic cytokines including IL-1, IL-6 and TNF; other proposed mechanisms include inducing apoptosis of inflammatory cells and increasing adenosine signalling162.
Methotrexate in RA
Methotrexate is currently the first-line therapy for RA25. Although open-label studies of methotrexate date back to 1968 (ref. 163), it was not until 1983 that the first RCT of methotrexate was performed in patients with RA164. The drug showed promising efficacy in these patients and similar results were obtained in a second larger placebo-controlled trial the following year165, leading the FDA to approve methotrexate as a therapy for RA in 1988 (ref. 166).
Studies in RA have shown an association between methotrexate use and reduction in cardiovascular risk by surrogate endpoints. In the TEAR trial, which compared methotrexate monotherapy, methotrexate therapy in combination with etanercept and methotrexate therapy in combination with two other DMARDs (triple therapy) in patients with RA, all three approaches led to an improvement in measures related to the anti-atherogenic functions of HDL (as measured by paraoxonase 1 activity and apolipoprotein AI levels)167. In other studies, methotrexate therapy has led to improvements in endothelial function biomarkers in patients with RA, including carotid intima-media thickness168. In a systematic review and meta-analysis that included 10 observational studies of patients with immune-mediated inflammatory diseases taking methotrexate, methotrexate use was associated with a 21% overall lower risk of cardiovascular disease and a 18% lower risk of myocardial infarction169.
Methotrexate in ASCVD
In the general population, the CIRT (Cardiovascular Inflammation Reduction Trial) was the second large-scale trial to test the inflammation hypothesis of atherosclerosis following the CANTOS trial170. The rationale was to not only choose an agent that has anti-inflammatory properties without effects on lipids (such as statins) but also a drug that is well-tolerated with a reasonable safety profile. Given the widespread use of methotrexate and long-term data on the efficacy and toxicity of this drug in RA, methotrexate was an ideal agent. The primary end point of CIRT was reduction in the rate of a composite primary end point of non-fatal myocardial infarction, non-fatal stroke or cardiovascular death among patients with either a prior myocardial infarction or multivessel coronary artery disease who additionally had either type II diabetes or metabolic syndrome. Although aimed at enrolling up to 7,000 patients, the trial was stopped early owing to a lack of benefit of methotrexate and included a total of 4,786 patients. No difference was observed for the primary end point when comparing low-dose methotrexate (15–20 mg weekly) with placebo. Furthermore, no differences were observed for CRP, IL-1α or IL-6 levels in the two treatment arms. However, unlike the CANTOS trial, the entry criteria did not include a minimum CRP requirement and the average baseline levels for the population were relatively low at 1.6 mg/l (ref. 171), introducing a potential basement effect. At this level of CRP, inflammation could only be further reduced by a small or insignificant amount, which in turn would preclude the ability to detect associations with reductions in cardiovascular risk.
Other relevant inflammatory pathways
In addition to the IL-6, TNF and IL-1 pathways, other pro-inflammatory pathways might also have a role in in both RA and ASCVD. In this section we discuss pathways for which some data are available on the effects of targeting these pathways on cardiovascular risk in RA or ASCVD but not for both. In patients with RA, targeting the CD80–CD86 costimulatory pathway with abatacept might result in a lower cardiovascular risk compared with targeting the TNF pathway, particularly for patients with diabetes mellitus172,173. However, limited data are available so far for this drug.
Likewise, the inflammasome, an important component of innate immunity, is implicated in the pathogenesis of ASCVD but is not known to have a major role in RA pathogenesis. The inflammasome is an important component of innate immunity and is implicated in the pathogenesis of atherosclerosis174,175. Of the five pattern recognition receptors known to function as inflammasome receptors, NLRP3 is the major receptor involved in atherogenesis174,176. Colchicine is an NLRP3 inflammasome inhibitor and inhibits tubulin polymerization and microtubule formation177. Colchicine is not used in the treatment of RA but is a mainstay of treatment for gout and familial Mediterranean fever177. Studies in the general population have demonstrated that colchicine has some efficacy in reducing the risk of ASCVD7,178,179. In the COLCOT (Colchicine Cardiovascular Outcomes Trial) study, which tested low-dose colchicine as secondary prevention and reduce cardiovascular disease in patients with a recent acute myocardial infarction, colchicine lowered the risk and incidence of urgent hospitalization for angina leading to coronary revascularization. Evidence also exists for the use of colchicine in the treatment of stable coronary artery disease; in the LoDoCo (Low-Dose Colchicine) pilot trial, colchicine (0.5 mg once daily) reduced the rate of cardiovascular events in patients with chronic coronary artery disease178. As noted in the section on IL-1, therapies that block the IL-1 pathway also target the consequences of inflammasome activation, and some evidence suggest that this approach provides some benefit in the prevention of ASCVD; however, these drugs either have a low efficacy (such as anakinra) or are not currently used in the treatment of RA (such as canakinumab)180.
Synthesizing the data
IL-1, IL-6 and TNF have important roles in the pathogenesis of RA and ASCVD, and emerging data support the involvement of the JAK–STAT pathway (which functions downstream of these cytokines). Focusing on the pathways of targeted therapies enables us to more precisely understand the downstream effect of blocking these pathways. Although deciphering the exact hierarchy of IL-1, IL-6, TNF and the JAK–STAT pathway within the pathogenesis of RA and atherosclerosis is challenging, most compelling data point to the involvement of IL-6 in both RA and ASCVD (Table 2). Indeed, data from both mechanistic investigations and genomic studies support the important role of IL-6 as a central cardiometabolic signalling cytokine181.
Despite the importance of TNF in RA pathogenesis, and evidence from observational studies of TNF inhibitors reducing the cardiovascular risk of patients with RA, the complex role of TNF in vascular biology and prior clinical trials of TNF inhibitors in patients with heart failure have made this therapy a less attractive option for reducing the risk of cardiovascular disease in the general population. Data from studies of IL-1 inhibitors, as well as of inflammasome inhibition, suggest that the IL-1 pathway probably has a more general upstream role in the inflammatory cascade. This more general role might explain the lower efficacy of IL-1 inhibitors in treating articular signs and symptoms of patients with RA compared with inhibitors of other pathways (such as IL-6 and TNF inhibitors), which have more direct effects on controlling RA inflammation. In terms of cardiovascular effects, in a study comparing the effects of anakinra and tocilizumab in patients with RA, anakinra had a more salutary effect on cardiac function, whereas tocilizumab mainly had beneficial vascular effects182. Furthermore, specifically targeting IL-1α was associated with a reduction in ASCVD risk in the general population. Based on these data, in the future, IL-1 might play a larger role in reducing cardiovascular risk than in controlling RA disease activity alone170.
In terms of drugs that more broadly target these cytokine pathways, observational data suggest that methotrexate can reduce the cardiovascular risk of patients with RA, a population exposed to chronic moderate-to-high levels of inflammation. However, methotrexate did not reduce the rate of recurrent cardiovascular events among high-risk individuals in the general population who had a low inflammatory burden in the CIRT study171. Although it is unlikely that another trial will assess the effects of methotrexate on cardiovascular disease in the general population, the lack of benefit observed in the CIRT population (that is, in individuals with a low baseline hsCRP level) in a sense supported the notion of inflammation as an important risk factor for cardiovascular disease. Inhibition of the JAK–STAT pathway impacts both the IL-6, TNF and other pro-inflammatory pathways. However, this broader effect might also lead to the mixed signals regarding its effect on ASCVD risk.
The complex signalling of these pathways creates a challenge for both rheumatologists and cardiologists in that these results cannot be viewed in a vacuum. Each cytokine and pathway has pleotropic functions and can further induce the expression of chemokines, which directly modulate the migration of cells and regulate context-specific aspects of the acute and chronic inflammatory response in the synovium and in the vasculature183. This complexity has created a double-edged sword in that blockade of proximal pro-inflammatory mediators might yield differing off-target effects, as was observed in the ORAL Surveillance study20. Furthermore, the effect of targeting these pathways to reduce cardiovascular risk might differ in patients with RA compared with the general population.
Conclusion
The convergence of pathways implicated in RA and ASCVD have led to a paradigm shift with regard to how treatments are considered in both rheumatology and cardiology. With multiple targeted therapies available for the treatment of RA, and limited data regarding which treatment will best control a particular patient’s disease, treatment decisions are currently guided by a patient’s comorbidities and the safety profile of the drugs. Incorporating ASCVD risk in therapy selection for each individual patient could help to inform the optimal therapy beyond simply the control of joint inflammation as we refine our knowledge of the mechanistic interplay between synovial inflammation and atherosclerotic development. An improved understanding of RA and cardiovascular risk will allow consideration of mitigating not just RA disease activity but also cardiovascular risk. IL-6 targeting currently stands as the most promising candidate; for the majority of remaining targets, data regarding the use of these targeted therapies and overall cardiovascular events in the general population remain limited. Returning to the notion of RA as a human model of inflammation, more studies on the pathobiology of atherosclerosis are needed in RA to better understand the downstream cardiovascular effects of targeted therapies and the mechanism behind these effects. Although this Review provides a simplified version of the complex inflammatory pathways shared between the two conditions, it provides a starting point for summarizing to date the inflammatory pathways that converge in RA and ASCVD, informing future direction for studies to improve the management of ASCVD risk. Moreover, this Review highlights a movement towards a growing collaboration between rheumatology and cardiology in both research and clinical care in the years to come.
Key points.
Rheumatoid arthritis (RA) and atherosclerotic cardiovascular disease share pro-inflammatory pathways that promote the pathogenesis of both conditions, including the IL-1 and IL-6 pathways.
Inflammation encompasses numerous pathways, and data from studies of targeted therapies developed for RA can provide insight into the potential effects of these therapies on cardiovascular risk.
Targeting IL-6 is considered more effective in controlling RA disease activity than targeting IL-1 and robust data also suggest that this therapy can reduce atherosclerotic cardiovascular disease risk in the general population.
Targeting some pro-inflammatory pathway(s), such as the TNF pathway, might be more beneficial than targeting others, such as JAK–STAT signalling, in reducing cardiovascular risk.
Acknowledgements
The authors would like to acknowledge the following grant funding support: BNW, National Institutes of Health (NIH) K23 HL159276-01 and the American Heart Association 21CDA851511; KPL NIH R01 HL127118, the Harold and DuVal Bowen Fund and the Be Brave Fund.
Footnotes
Competing interests
J.T.G. has consulted for AbbVie, Bristol Myers Squibb, Eli Lilly, Gilead, Novartis, Pfizer and UCB. J.T.G. served as a co-author on secondary analyses for studies from the ORAL Surveillance study for which he received no compensation. B.N.W. has consulted for Horizon Therapeutics, Kinisika and NovoNordisk. K.P.L. declares no competing interests.
References
- 1.McInnes IB & Schett G The pathogenesis of rheumatoid arthritis. N. Engl. J. Med 365, 2205–2219 (2011). [DOI] [PubMed] [Google Scholar]
- 2.Smolen JS, Aletaha D & McInnes IB Rheumatoid arthritis. Lancet 388, 2023–2038 (2016). [DOI] [PubMed] [Google Scholar]
- 3.England BR et al. Update of the American College of Rheumatology Recommended Rheumatoid Arthritis Disease Activity Measures. Arthritis Care Res. 71, 1540–1555 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Arnett DK et al. ACC/AHA Guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol 74, e177–e232 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Dawber TR, Moore FE & Mann GV II Coronary heart disease in the Framingham Study. Am. J. Public Health Nations Health 47, 4–24 (1957). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ridker PM et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med 377, 1119–1131 (2017). [DOI] [PubMed] [Google Scholar]
- 7.Tardif J-C et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N. Engl. J. Med 381, 2497–2505 (2019). [DOI] [PubMed] [Google Scholar]
- 8.Solomon DH et al. Cardiovascular morbidity and mortality in women diagnosed with rheumatoid arthritis. Circulation 107, 1303–1307 (2003). [DOI] [PubMed] [Google Scholar]
- 9.Aviña-Zubieta JA et al. Risk of cardiovascular mortality in patients with rheumatoid arthritis: a meta-analysis of observational studies. Arthritis Rheum. 59, 1690–1697 (2008). [DOI] [PubMed] [Google Scholar]
- 10.del Rincón I, Freeman GL, Haas RW, O’Leary DH & Escalante A Relative contribution of cardiovascular risk factors and rheumatoid arthritis clinical manifestations to atherosclerosis. Arthritis Rheum. 52, 3413–3423 (2005). [DOI] [PubMed] [Google Scholar]
- 11.Myasoedova E et al. The role of rheumatoid arthritis (RA) flare and cumulative burden of RA severity in the risk of cardiovascular disease. Ann. Rheum. Dis 75, 560–565 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.del Rincón I et al. Systemic inflammation and cardiovascular risk factors predict rapid progression of atherosclerosis in rheumatoid arthritis. Ann. Rheum. Dis 74, 1118–1123 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yu Z et al. Association between inflammation and systolic blood pressure in RA compared to patients without RA. Arthritis Res. Ther 20, 107 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hansson GK, Robertson A-KL & Söderberg-Nauclér C Inflammation and atherosclerosis. Annu. Rev. Pathol 1, 297–329 (2006). [DOI] [PubMed] [Google Scholar]
- 15.Libby P Inflammation in atherosclerosis. Arterioscler. Thromb. Vasc. Biol 32, 2045–2051 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Libby P et al. Atherosclerosis. Nat. Rev. Dis. Prim 5, 56 (2019). [DOI] [PubMed] [Google Scholar]
- 17.Provan SA et al. The impact of newer biological disease modifying anti-rheumatic drugs on cardiovascular risk factors: a 12-month longitudinal study in rheumatoid arthritis patients treated with rituximab, abatacept and tocilizumab. PLoS One 10, e0130709 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Singh S et al. Comparative risk of cardiovascular events with biologic and synthetic disease-modifying antirheumatic drugs in patients with rheumatoid arthritis: a systematic review and meta-analysis. Arthritis Care Res. 72, 561–576 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Solomon DH et al. Explaining the cardiovascular risk associated with rheumatoid arthritis: traditional risk factors versus markers of rheumatoid arthritis severity. Ann. Rheum. Dis 69, 1920–1925 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ytterberg SR et al. Cardiovascular and cancer risk with tofacitinib in rheumatoid arthritis. N. Engl. J. Med 386, 316–326 (2022). [DOI] [PubMed] [Google Scholar]
- 21.Dayer J-M, Oliviero F & Punzi L A brief history of IL-1 and IL-1 Ra in rheumatology. Front. Pharmacol 8, 00293 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dayer JM, Graham R, Russell G & Krane SM Collagenase production by rheumatoid synovial cells: stimulation by a human lymphocyte factor. Science 195, 181–183 (1977). [DOI] [PubMed] [Google Scholar]
- 23.Chan AH & Schroder K Inflammasome signaling and regulation of interleukin-1 family cytokines. J. Exp. Med 217, e20190314 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Akitsu A et al. IL-1 receptor antagonist-deficient mice develop autoimmune arthritis due to intrinsic activation of IL-17-producing CCR2+Vγ6+γδ T cells. Nat. Commun 6, 7464 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Fraenkel L et al. American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 73, 1108–1123 (2021). [DOI] [PubMed] [Google Scholar]
- 26.Singh JA et al. A network meta-analysis of randomized controlled trials of biologics for rheumatoid arthritis: a Cochrane overview. CMAJ 181, 787–796 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Choe J-Y, Crain B, Wu SR & Corr M Interleukin 1 receptor dependence of serum transferred arthritis can be circumvented by toll-like receptor 4 signaling. J. Exp. Med 197, 537–542 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wei S, Kitaura H, Zhou P, Ross FP & Teitelbaum SL IL-1 mediates TNF-induced osteoclastogenesis. J. Clin. Invest 115, 282–290 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jh K et al. The mechanism of osteoclast differentiation induced by IL-1. J. Immunol 183, 1862–1870 (2009). [DOI] [PubMed] [Google Scholar]
- 30.Schönbeck U, Mach F & Libby P Generation of biologically active IL-1 β by matrix metalloproteinases: a novel caspase-1-independent pathway of IL-1 β processing. J. Immunol 161, 3340–3346 (1998). [PubMed] [Google Scholar]
- 31.Ito A et al. Degradation of interleukin 1β by matrix metalloproteinases*. J. Biol. Chem 271, 14657–14660 (1996). [DOI] [PubMed] [Google Scholar]
- 32.Schiff MH Role of interleukin 1 and interleukin 1 receptor antagonist in the mediation of rheumatoid arthritis. Ann. Rheum. Dis 59, i103–i108 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Eastgate J et al. Correlation of plasma interleukin 1 levels with disease activity in rheumatoid arthritis. Lancet 332, 706–709 (1988). [DOI] [PubMed] [Google Scholar]
- 34.Abbate A et al. Interleukin-1 and the inflammasome as therapeutic targets in cardiovascular disease. Circ. Res 126, 1260–1280 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Libby P, Warner SJ & Friedman GB Interleukin 1: a mitogen for human vascular smooth muscle cells that induces the release of growth-inhibitory prostanoids. J. Clin. Invest 81, 487–498 (1988). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bevilacqua MP, Pober JS, Wheeler ME, Cotran RS & Gimbrone MA Interleukin-1 activation of vascular endothelium. Effects on procoagulant activity and leukocyte adhesion. Am. J. Pathol 121, 394–403 (1985). [PMC free article] [PubMed] [Google Scholar]
- 37.Tanaka T, Narazaki M & Kishimoto T IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol 6, a016295 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Libby P Collagenases and cracks in the plaque. J. Clin. Invest 123, 3201–3203 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Loppnow H & Libby P Proliferating or interleukin 1-activated human vascular smooth muscle cells secrete copious interleukin 6. J. Clin. Invest 85, 731–738 (1990). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Vromman A et al. Stage-dependent differential effects of interleukin-1 isoforms on experimental atherosclerosis. Eur. Heart J 40, 2482–2491 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Suzuki K et al. Overexpression of interleukin-1 receptor antagonist provides cardioprotection against ischemia-reperfusion injury associated with reduction in apoptosis. Circulation 10.1161/circ.104.suppl_1.I-308 (2001). [DOI] [PubMed] [Google Scholar]
- 42.Del Buono MG et al. Effect of interleukin-1 blockade with anakinra on leukocyte count in patients with ST-segment elevation acute myocardial infarction. Sci. Rep 12, 1254 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Harouki N et al. The IL-1β antibody gevokizumab limits cardiac remodeling and coronary dysfunction in rats with heart failure. JACC Basic. Transl. Sci 2, 418–430 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Morton AC et al. The effect of interleukin-1 receptor antagonist therapy on markers of inflammation in non-ST elevation acute coronary syndromes: the MRC-ILA Heart Study. Eur. Heart J 36, 377–384 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Abbate A et al. Interleukin-1 blockade with anakinra to prevent adverse cardiac remodeling after acute myocardial infarction (Virginia Commonwealth University Anakinra Remodeling Trial [VCU-ART] Pilot study). Am. J. Cardiol 105, 1371–1377.e1 (2010). [DOI] [PubMed] [Google Scholar]
- 46.Abbate A et al. Effects of interleukin-1 blockade with anakinra on adverse cardiac remodeling and heart failure after acute myocardial infarction [from the Virginia Commonwealth University-Anakinra Remodeling Trial (2) (VCU-ART2) pilot study]. Am. J. Cardiol 111, 1394–1400 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Abbate A et al. Interleukin-1 blockade inhibits the acute inflammatory response in patients with ST-segment-elevation myocardial infarction. J. Am. Heart Assoc 9, e014941 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Samsonov M, Bogin V, Van Tassell BW & Abbate A Interleukin-1 blockade with RPH-104 in patients with acute ST-elevation myocardial infarction: study design and rationale. J. Transl. Med 19, 169 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Abbate A et al. Comparative safety of interleukin-1 blockade with anakinra in patients with ST-segment elevation acute myocardial infarction (from the VCU-ART and VCU-ART2 pilot studies). Am. J. Cardiol 115, 288–292 (2015). [DOI] [PubMed] [Google Scholar]
- 50.Food and Drug Administration (FDA). KINERET® (anakinra) injection, for subcutaneous use https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/103950s5189lbl.pdf (2020). [Google Scholar]
- 51.McDermott MF Rilonacept in the treatment of chronic inflammatory disorders. Drugs Today 45, 423–430 (2009). [DOI] [PubMed] [Google Scholar]
- 52.Alten R et al. Efficacy and safety of the human anti-IL-1β monoclonal antibody canakinumab in rheumatoid arthritis: results of a 12-week, phase II, dose-finding study. BMC Musculoskelet. Disord 12, 153 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ridker PM et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med 377, 1119–1131 (2017). [DOI] [PubMed] [Google Scholar]
- 54.Ridker PM et al. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial. Lancet 391, 319–328 (2018). [DOI] [PubMed] [Google Scholar]
- 55.Kishimoto T Factors affecting B-cell growth and differentiation. Annu. Rev. Immunol 3, 133–157 (1985). [DOI] [PubMed] [Google Scholar]
- 56.Kishimoto T The biology of interleukin-6. Blood 74, 1–10 (1989). [PubMed] [Google Scholar]
- 57.Hirano T et al. Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin. Nature 324, 73–76 (1986). [DOI] [PubMed] [Google Scholar]
- 58.Boe A, Baiocchi M, Carbonatto M, Papoian R & Serlupi-Crescenzi O Interleukin 6 knock-out mice are resistant to antigen-induced experimental arthritis. Cytokine 11, 1057–1064 (1999). [DOI] [PubMed] [Google Scholar]
- 59.Kopf M et al. Impaired immune and acute-phase responses in interleukin-6-deficient mice. Nature 368, 339–342 (1994). [DOI] [PubMed] [Google Scholar]
- 60.Tamura T et al. Soluble interleukin-6 receptor triggers osteoclast formation by interleukin 6. Proc. Natl Acad. Sci. USA 90, 11924–11928 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Heink S et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat. Immunol 18, 74–85 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jones SA & Rose-John S The role of soluble receptors in cytokine biology: the agonistic properties of the sIL-6R/IL-6 complex. Biochim. Biophys. Acta Mol. Cell Res 1592, 251–263 (2002). [DOI] [PubMed] [Google Scholar]
- 63.Madhok R, Crilly A, Watson J & Capell HA Serum interleukin 6 levels in rheumatoid arthritis: correlations with clinical and laboratory indices of disease activity. Ann. Rheum. Dis 52, 232–234 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sack U et al. Interleukin-6 in synovial fluid is closely associated with chronic synovitis in rheumatoid arthritis. Rheumatol. Int 13, 45–51 (1993). [DOI] [PubMed] [Google Scholar]
- 65.Srirangan S & Choy EH The role of interleukin 6 in the pathophysiology of rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis 2, 247–256 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Feng W et al. Combination of IL-6 and sIL-6R differentially regulate varying levels of RANKL-induced osteoclastogenesis through NF-κB, ERK and JNK signaling pathways. Sci. Rep 7, 41411 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kotake S et al. Interleukin-6 and soluble interleukin-6 receptors in the synovial fluids from rheumatoid arthritis patients are responsible for osteoclast-like cell formation. J. Bone Min. Res 11, 88–95 (2009). [DOI] [PubMed] [Google Scholar]
- 68.Food and Drug Administration (FDA). Sarilumab FDA https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/761037orig1s000medr.pdf (2017). [Google Scholar]
- 69.Food and Drug Administration (FDA). Toclizumab FDA https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125276s114lbl.pdf (2017). [Google Scholar]
- 70.Weber B et al. Relationship between risk of atherosclerotic cardiovascular disease, inflammation, and coronary microvascular dysfunction in rheumatoid arthritis. J. Am. Heart Assoc 11, e025467 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Davies R et al. The role of interleukin-6 trans-signalling on cardiovascular dysfunction in inflammatory arthritis. Rheumatology 60, 2852–2861 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Souto A et al. Lipid profile changes in patients with chronic inflammatory arthritis treated with biologic agents and tofacitinib in randomized clinical trials: a systematic review and meta-analysis. Arthritis Rheumatol. 67, 117–127 (2015). [DOI] [PubMed] [Google Scholar]
- 73.McInnes IB et al. Effect of interleukin-6 receptor blockade on surrogates of vascular risk in rheumatoid arthritis: MEASURE, a randomised, placebo-controlled study. Ann. Rheum. Dis 74, 694–702 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Albers JJ, Slee A, Fleg JL, O’Brien KD & Marcovina SM Relationship of baseline HDL subclasses, small dense LDL and LDL triglyceride to cardiovascular events in the AIM-HIGH clinical trial. Atherosclerosis 251, 454–459 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Pierini FS et al. Effect of tocilizumab on LDL and HDL characteristics in patients with rheumatoid arthritis. An observational study. Rheumatol. Ther 8, 803–815 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Giles JT et al. Cardiovascular safety of tocilizumab versus etanercept in rheumatoid arthritis: a randomized controlled trial. Arthritis Rheumatol. 72, 31–40 (2020). [DOI] [PubMed] [Google Scholar]
- 77.Ridker PM et al. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS). Eur. Heart J 39, 3499–3507 (2018). [DOI] [PubMed] [Google Scholar]
- 78.Kleveland O et al. Effect of a single dose of the interleukin-6 receptor antagonist tocilizumab on inflammation and troponin T release in patients with non-ST-elevation myocardial infarction: a double-blind, randomized, placebo-controlled phase 2 trial. Eur. Heart J 37, 2406–2413 (2016). [DOI] [PubMed] [Google Scholar]
- 79.Broch K et al. Randomized trial of interleukin-6 receptor inhibition in patients with acute ST-segment elevation myocardial infarction. J. Am. Coll. Cardiol 77, 1845–1855 (2021). [DOI] [PubMed] [Google Scholar]
- 80.Ferreira RC et al. Functional IL6R 358Ala allele impairs classical IL-6 receptor signaling and influences risk of diverse inflammatory diseases. PLoS Genet. 9, e1003444 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.IL6R Genetics Consortium Emerging Risk Factors Collaboration. et al. Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies. Lancet 379, 1205–1213 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ridker PM & Rane M Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease. Circ. Res 128, 1728–1746 (2021). [DOI] [PubMed] [Google Scholar]
- 83.Levin MG et al. A missense variant in the IL-6 receptor and protection from peripheral artery disease. Circ. Res 129, 968–970 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Interleukin-6 Receptor Mendelian Randomisation Analysis (IL6R MR) Consortium. et al. The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis. Lancet 379, 1214–1224 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cai T et al. Association of interleukin 6 receptor variant with cardiovascular disease effects of interleukin 6 receptor blocking therapy: a phenome-wide association study. JAMA Cardiol. 3, 849–857 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Taylor NP FDA panel snubs Johnson & Johnson’s rheumatoid arthritis drug sirukumab, citing worries about trial deaths https://www.fiercebiotech.com/biotech/fda-panel-snubs-johnson-johnson-s-rheumatoid-arthritis-drug-sirukumab-citing-worries-about (2017). [Google Scholar]
- 87.Ridker PM et al. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 397, 2060–2069 (2021). [DOI] [PubMed] [Google Scholar]
- 88.Aletaha D et al. Efficacy and safety of sirukumab in patients with active rheumatoid arthritis refractory to anti-TNF therapy (SIRROUND-T): a randomised, double-blind, placebo-controlled, parallel-group, multinational, phase 3 study. Lancet 389, 1206–1217 (2017). [DOI] [PubMed] [Google Scholar]
- 89.Takeuchi T et al. Sirukumab for rheumatoid arthritis: the phase III SIRROUND-D study. Ann. Rheum. Dis 76, 2001–2008 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Bartoli F, Bae S, Cometi L, Matucci Cerinic M & Furst DE Sirukumab for the treatment of rheumatoid arthritis: update on sirukumab, 2018. Expert. Rev. Clin. Immunol 14, 539–547 (2018). [DOI] [PubMed] [Google Scholar]
- 91.Ridker PM et al. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 397, 2060–2069 (2021). [DOI] [PubMed] [Google Scholar]
- 92.Ridker PM From RESCUE to ZEUS: will interleukin-6 inhibition with ziltivekimab prove effective for cardiovascular event reduction? Cardiovasc. Res 117, e138–e140 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Gough P & Myles IA Tumor necrosis factor receptors: pleiotropic signaling complexes and their differential effects. Front. Immunol 11, 585880 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yang S, Wang J, Brand DD & Zheng SG Role of TNF–TNF receptor 2 signal in regulatory T cells and its therapeutic implications. Front. Immunol 9, 784 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Smolen JS et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann. Rheum. Dis 79, 685–699 (2020). [DOI] [PubMed] [Google Scholar]
- 96.Li P & Schwarz EM The TNF-α transgenic mouse model of inflammatory arthritis. Springe. Semin. Immunopathol 25, 19–33 (2003). [DOI] [PubMed] [Google Scholar]
- 97.Keffer J et al. Transgenic mice expressing human tumour necrosis factor: a predictive genetic model of arthritis. EMBO J. 10, 4025–4031 (1991). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Feldmann M, Brennan FM & Maini RN Rheumatoid arthritis. Cell 85, 307–310 (1996). [DOI] [PubMed] [Google Scholar]
- 99.Brennan FM & McInnes IB Evidence that cytokines play a role in rheumatoid arthritis. J. Clin. Invest 118, 3537–3545 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Dayer JM, Beutler B & Cerami A Cachectin/tumor necrosis factor stimulates collagenase and prostaglandin E2 production by human synovial cells and dermal fibroblasts. J. Exp. Med 162, 2163–2168 (1985). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Bertolini DR, Nedwin GE, Bringman TS, Smith DD & Mundy GR Stimulation of bone resorption and inhibition of bone formation in vitro by human tumour necrosis factors. Nature 319, 516–518 (1986). [DOI] [PubMed] [Google Scholar]
- 102.Nadkarni S, Mauri C & Ehrenstein MR Anti-TNF-α therapy induces a distinct regulatory T cell population in patients with rheumatoid arthritis via TGF-β. J. Exp. Med 204, 33–39 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ehrenstein MR et al. Compromised function of regulatory T cells in rheumatoid arthritis and reversal by anti-TNFα therapy. J. Exp. Med 200, 277–285 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Elliott MJ et al. Treatment of rheumatoid arthritis with chimeric monoclonal antibodies to tumor necrosis factor α. Arthritis Rheum. 36, 1681–1690 (1993). [DOI] [PubMed] [Google Scholar]
- 105.Elliott MJ et al. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor α (cA2) versus placebo in rheumatoid arthritis. Lancet 344, 1105–1110 (1994). [DOI] [PubMed] [Google Scholar]
- 106.Bathon JM et al. A comparison of etanercept and methotrexate in patients with early rheumatoid arthritis. N. Engl. J. Med 343, 1586–1593 (2000). [DOI] [PubMed] [Google Scholar]
- 107.Weinblatt ME et al. A trial of etanercept, a recombinant tumor necrosis factor receptor:Fc fusion protein, in patients with rheumatoid arthritis receiving methotrexate. N. Engl. J. Med 340, 253–259 (1999). [DOI] [PubMed] [Google Scholar]
- 108.van de Putte LBA et al. Efficacy and safety of adalimumab as monotherapy in patients with rheumatoid arthritis for whom previous disease modifying antirheumatic drug treatment has failed. Ann. Rheum. Dis 63, 508–516 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Keystone EC et al. Golimumab, a human antibody to tumour necrosis factor α given by monthly subcutaneous injections, in active rheumatoid arthritis despite methotrexate therapy: the GO-FORWARD Study. Ann. Rheum. Dis 68, 789–796 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kirkham BW et al. Effects of golimumab, an anti-tumour necrosis factor-α human monoclonal antibody, on Lipids and markers of inflammation. Ann. Rheum. Dis 73, 161–169 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Keystone E et al. Certolizumab pegol plus methotrexate is significantly more effective than placebo plus methotrexate in active rheumatoid arthritis: findings of a fifty-two-week, phase III, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Arthritis Rheum. 58, 3319–3329 (2008). [DOI] [PubMed] [Google Scholar]
- 112.Barnabe C, Martin B-J & Ghali WA Systematic review and meta-analysis: anti-tumor necrosis factor α therapy and cardiovascular events in rheumatoid arthritis. Arthritis Care Res. 63, 522–529 (2011). [DOI] [PubMed] [Google Scholar]
- 113.Dixon WG et al. Reduction in the incidence of myocardial infarction in patients with rheumatoid arthritis who respond to anti–tumor necrosis factor α therapy: results from the British Society for Rheumatology Biologics Register. Arthritis Rheum. 56, 2905–2912 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Solomon DH et al. The relationship of cardiovascular risk in rheumatoid arthritis comparing TNFα blockade with non-biologic DMARDs. Am. J. Med 126, 730.e9–730.e17 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Low ASL et al. Relationship between exposure to tumour necrosis factor inhibitor therapy and incidence and severity of myocardial infarction in patients with rheumatoid arthritis. Ann. Rheum. Dis 76, 654–660 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Solomon DH et al. Reducing cardiovascular risk with immunomodulators: a randomised active comparator trial among patients with rheumatoid arthritis. Ann. Rheum. Dis 82, 324–330 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Zhang H et al. Role of TNF-α in vascular dysfunction. Clin. Sci 116, 219–230 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ridker PM et al. Elevation of tumor necrosis factor-α and increased risk of recurrent coronary events after myocardial infarction. Circulation 10.1161/01.CIR.101.18.2149 (2000). [DOI] [PubMed] [Google Scholar]
- 119.Hegewisch S, Weh H-J & Hossfeld DK TNF-induced cardiomyopathy. Lancet 335, 294–295 (1990). [DOI] [PubMed] [Google Scholar]
- 120.Yoshizumi M, Perrella MA, Burnett JC & Lee ME Tumor necrosis factor downregulates an endothelial nitric oxide synthase mRNA by shortening its half-life. Circ. Res 73, 205–209 (1993). [DOI] [PubMed] [Google Scholar]
- 121.Goodwin BL, Pendleton LC, Levy MM, Solomonson LP & Eichler DC Tumor necrosis factor-alpha reduces argininosuccinate synthase expression and nitric oxide production in aortic endothelial cells. Am. J. Physiol. Heart Circ. Physiol 293, H1115–H1121 (2007). [DOI] [PubMed] [Google Scholar]
- 122.Hansson GK & Libby P The immune response in atherosclerosis: a double-edged sword. Nat. Rev. Immunol 6, 508–519 (2006). [DOI] [PubMed] [Google Scholar]
- 123.Canault M et al. Exclusive expression of transmembrane TNF-alpha in mice reduces the inflammatory response in early lipid lesions of aortic sinus. Atherosclerosis 172, 211–218 (2004). [DOI] [PubMed] [Google Scholar]
- 124.Ohta H et al. Disruption of tumor necrosis factor-α gene diminishes the development of atherosclerosis in ApoE-deficient mice. Atherosclerosis 180, 11–17 (2005). [DOI] [PubMed] [Google Scholar]
- 125.Schreyer SA, Peschon JJ & LeBoeuf RC Accelerated atherosclerosis in mice Lacking tumor necrosis factor receptor p55. J. Biol. Chem 271, 26174–26178 (1996). [DOI] [PubMed] [Google Scholar]
- 126.Oberoi R et al. Anti-tumor necrosis factor-α therapy increases plaque burden in a mouse model of experimental atherosclerosis. Atherosclerosis 277, 80–89 (2018). [DOI] [PubMed] [Google Scholar]
- 127.Chung ES et al. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-α, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation 107, 3133–3140 (2003). [DOI] [PubMed] [Google Scholar]
- 128.Mann DL et al. Targeted anticytokine therapy in patients with chronic heart failure: results of the randomized etanercept worldwide evaluation (RENEWAL). Circulation 109, 1594–1602 (2004). [DOI] [PubMed] [Google Scholar]
- 129.Yokoyama T et al. Cellular basis for the negative inotropic effects of tumor necrosis factor-α in the adult mammalian heart. J. Clin. Invest 92, 2303–2312 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Mann DL, Bozkurt B, Torre-Amione G, Soran OZ & Sivasubramanian N Effect of the soluble TNF-antagonist etanercept on tumor necrosis factor bioactivity and stability. Clin. Transl. Sci 1, 142–145 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.O’Shea JL et al. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu. Rev. Med 66, 311–328 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Seif F et al. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun. Signal 15, 23 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hu X, Li J, Fu M, Zhao X & Wang W The JAK/STAT signaling pathway: from bench to clinic. Sig. Transduct. Target. Ther 6, 1–33 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Malemud CJ Negative regulators of JAK/STAT signaling in rheumatoid arthritis and osteoarthritis. Int. J. Mol. Sci 18, E484 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Tanaka Y, Luo Y, O’Shea JJ & Nakayamada S Janus kinase-targeting therapies in rheumatology: a mechanisms-based approach. Nat. Rev. Rheumatol 18, 133–145 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Taylor NP FDA rejects Gilead’s would-be blockbuster filgotinib over toxicity concerns https://www.fiercebiotech.com/biotech/fda-rejects-gilead-s-would-be-blockbuster-filgotinib-over-toxicity-concerns (2020). [Google Scholar]
- 137.Traves PG et al. JAK selectivity and the implications for clinical inhibition of pharmacodynamic cytokine signalling by filgotinib, upadacitinib, tofacitinib and baricitinib. Ann. Rheum. Dis 80, 865–875 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Burmester GR et al. Tofacitinib (CP-690,550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial. Lancet 381, 451–460 (2013). [DOI] [PubMed] [Google Scholar]
- 139.Lee EB et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N. Engl. J. Med 370, 2377–2386 (2014). [DOI] [PubMed] [Google Scholar]
- 140.van Vollenhoven RF et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N. Engl. J. Med 367, 508–519 (2012). [DOI] [PubMed] [Google Scholar]
- 141.Burmester GR et al. Safety and efficacy of upadacitinib in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying anti-rheumatic drugs (SELECT-NEXT): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 391, 2503–2512 (2018). [DOI] [PubMed] [Google Scholar]
- 142.Genovese MC et al. Safety and efficacy of upadacitinib in patients with active rheumatoid arthritis refractory to biologic disease-modifying anti-rheumatic drugs (SELECT-BEYOND): a double-blind, randomised controlled phase 3 trial. Lancet 391, 2513–2524 (2018). [DOI] [PubMed] [Google Scholar]
- 143.Charles-Schoeman C et al. Risk of major adverse cardiovascular events with tofacitinib versus tumour necrosis factor inhibitors in patients with rheumatoid arthritis with or without a history of atherosclerotic cardiovascular disease: a post hoc analysis from ORAL Surveillance. Ann. Rheum. Dis 82, 119–129 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kremer J et al. Tofacitinib in combination with nonbiologic disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis. Ann. Intern. Med 159, 253–261 (2013). [DOI] [PubMed] [Google Scholar]
- 145.Kume K et al. Tofacitinib improves atherosclerosis despite up-regulating serum cholesterol in patients with active rheumatoid arthritis: a cohort study. Rheumatol. Int 37, 2079–2085 (2017). [DOI] [PubMed] [Google Scholar]
- 146.Charles-Schoeman C et al. Potential mechanisms Leading to the abnormal lipid profile in patients with rheumatoid arthritis versus healthy volunteers and reversal by tofacitinib. Arthritis Rheumatol. 67, 616–625 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Charles-Schoeman C et al. Cardiovascular safety findings in patients with rheumatoid arthritis treated with tofacitinib, an oral Janus kinase inhibitor. Semin. Arthritis Rheum 46, 261–271 (2016). [DOI] [PubMed] [Google Scholar]
- 148.Xie W et al. Impact of Janus kinase inhibitors on risk of cardiovascular events in patients with rheumatoid arthritis: systematic review and meta-analysis of randomised controlled trials. Ann. Rheum. Dis 78, 1048–1054 (2019). [DOI] [PubMed] [Google Scholar]
- 149.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. 81, 798–804 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Winthrop KL & Cohen SB Oral surveillance and JAK inhibitor safety: the theory of relativity. Nat. Rev. Rheumatol 18, 301–304 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Misra DP, Pande G & Agarwal V Cardiovascular risks associated with Janus kinase inhibitors: peering outside the black box. Clin. Rheumatol 42, 621–632 (2023). [DOI] [PubMed] [Google Scholar]
- 152.Garbers C, Aparicio-Siegmund S & Rose-John S The IL-6/gp130/STAT3 signaling axis: recent advances towards specific inhibition. Curr. Opin. Immunol 34, 75–82 (2015). [DOI] [PubMed] [Google Scholar]
- 153.Ivashkiv LB IFNγ: signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat. Rev. Immunol 18, 545–558 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Hilfiker-Kleiner D et al. Signal transducer and activator of transcription 3 is required for myocardial capillary growth, control of interstitial matrix deposition, and heart protection from ischemic injury. Circ. Res 95, 187–195 (2004). [DOI] [PubMed] [Google Scholar]
- 155.Yue H, Li W, Desnoyer R & Karnik SS Role of nuclear unphosphorylated STAT3 in angiotensin II type 1 receptor-induced cardiac hypertrophy. Cardiovasc. Res 85, 90–99 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Krishnamurthy P et al. IL-10 inhibits inflammation and attenuates Left ventricular remodeling after myocardial infarction via activation of STAT-3 and suppression of HuR. Circ. Res 104, e9–e18 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Plens-Galaska M et al. Genome-wide inhibition of pro-atherogenic gene expression by multi-STAT targeting compounds as a novel treatment strategy of CVDs. Front. Immunol 9, 2141 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Weinblatt ME Methotrexate in rheumatoid arthritis: a quarter century of development. Trans. Am. Clin. Climatol. Assoc 124, 16–25 (2013). [PMC free article] [PubMed] [Google Scholar]
- 159.Cronstein BN & Aune TM Methotrexate and its mechanisms of action in inflammatory arthritis. Nat. Rev. Rheumatol 16, 145–154 (2020). [DOI] [PubMed] [Google Scholar]
- 160.Bobillo S et al. Prophylaxis with intrathecal or high-dose methotrexate in diffuse large B-cell lymphoma and high risk of CNS relapse. Blood Cancer J. 11, 1–6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Morgan SL et al. Supplementation with folic acid during methotrexate therapy for rheumatoid arthritis. A double-blind, placebo-controlled trial. Ann. Intern. Med 121, 833–841 (1994). [DOI] [PubMed] [Google Scholar]
- 162.Everett BM et al. Rationale and design of the cardiovascular inflammation reduction trial (CIRT): a test of the inflammatory hypothesis of atherothrombosis. Am. Heart J 166, 199–207.e15 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Black RL et al. Methotrexate therapy in psoriatic arthritis; double-blind study on 21 patients. J. Am. Med. Assoc 189, 743–747 (1964). [PubMed] [Google Scholar]
- 164.Weinblatt ME et al. Efficacy of low-dose methotrexate in rheumatoid arthritis. N. Engl. J. Med 312, 818–822 (1985). [DOI] [PubMed] [Google Scholar]
- 165.Thompson RN, Watts C, Edelman J, Esdaile J & Russell AS A controlled two-centre trial of parenteral methotrexate therapy for refractory rheumatoid arthritis. J. Rheumatol 11, 760–763 (1984). [PubMed] [Google Scholar]
- 166.Weinblatt ME Methotrexate: who would have predicted its importance in rheumatoid arthritis? Arthritis Res. Ther 20, 103 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Charles-Schoeman C et al. Improvement of high-density lipoprotein function in patients with early rheumatoid arthritis treated with methotrexate monotherapy or combination therapies in a randomized controlled trial. Arthritis Rheumatol. 69, 46–57 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Ristić GG et al. Rheumatoid arthritis is an independent risk factor for increased carotid intima-media thickness: impact of anti-inflammatory treatment. Rheumatology 49, 1076–1081 (2010). [DOI] [PubMed] [Google Scholar]
- 169.Micha R et al. Systematic review and meta-analysis of methotrexate use and risk of cardiovascular disease. Am. J. Cardiol 108, 1362–1370 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Ridker PM et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med 377, 1119–1131 (2017). [DOI] [PubMed] [Google Scholar]
- 171.Ridker PM et al. Low-dose methotrexate for the prevention of atherosclerotic events. N. Engl. J. Med 380, 752–762 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Desai RJ, Dejene S, Jin Y, Liu J & Kim SC Comparative risk of diabetes mellitus in patients with rheumatoid arthritis treated with biologic or targeted synthetic disease-modifying drugs: a cohort study. ACR Open Rheumatol. 2, 222–231 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Kang EH et al. Comparative cardiovascular risk of abatacept and tumor necrosis factor inhibitors in patients with rheumatoid arthritis with and without diabetes mellitus: a multidatabase cohort study. J. Am. Heart Assoc 7, e007393 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Grebe A, Hoss F & Latz E NLRP3 inflammasome and the IL-1 pathway in atherosclerosis. Circ. Res 122, 1722–1740 (2018). [DOI] [PubMed] [Google Scholar]
- 175.Duewell P et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464, 1357–1361 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Lamkanfi M & Dixit VM Mechanisms and functions of inflammasomes. Cell 157, 1013–1022 (2014). [DOI] [PubMed] [Google Scholar]
- 177.Leung YY, Yao Hui LL & Kraus VB Colchicine — update on mechanisms of action and therapeutic uses. Semin. Arthritis Rheum 45, 341–350 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Nidorf SM et al. Colchicine in patients with chronic coronary disease. N. Engl. J. Med 383, 1838–1847 (2020). [DOI] [PubMed] [Google Scholar]
- 179.Weber BN & Blankstein R Something old, something new: a paradigm for considering immune therapies for cardiovascular disease. Cardiovasc. Res 116, e51–e53 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Clark W, Jobanputra P, Barton P & Burls A The clinical and cost-effectiveness of anakinra for the treatment of rheumatoid arthritis in adults: a systematic review and economic analysis. Health Technol. Assess 8, 1–105 (2004). [DOI] [PubMed] [Google Scholar]
- 181.Libby P & Rocha VZ All roads lead to IL-6: a central hub of cardiometabolic signaling. Int. J. Cardiol 259, 213–215 (2018). [DOI] [PubMed] [Google Scholar]
- 182.Ikonomidis I et al. Differential effects of inhibition of interleukin 1 and 6 on myocardial, coronary and vascular function. Clin. Res. Cardiol 108, 1093–1101 (2019). [DOI] [PubMed] [Google Scholar]
- 183.Gencer S, Evans BR, van der Vorst EPC, Döring Y & Weber C Inflammatory chemokines in atherosclerosis. Cells 10, 226 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Carroll MB, Haller C & Smith C Short-term application of tocilizumab during myocardial infarction (STAT-MI). Rheumatol. Int 38, 59–66 (2018). [DOI] [PubMed] [Google Scholar]
- 185.Kleveland O et al. Interleukin-6 receptor inhibition with tocilizumab induces a selective and substantial increase in plasma IP-10 and MIP-1β in non-ST-elevation myocardial infarction. Int. J. Cardiol 271, 1–7 (2018). [DOI] [PubMed] [Google Scholar]
- 186.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT05350592 (2023). [DOI] [PubMed] [Google Scholar]