Next Article in Journal
Analysis of Ferritin, Hepcidin, Zinc, C-Reactive Protein and IL-6 Levels in COVID-19 in Patients Living at Different Altitudes in Peru
Previous Article in Journal
Diabetes Mellitus in Non-Functioning Adrenal Incidentalomas: Analysis of the Mild Autonomous Cortisol Secretion (MACS) Impact on Glucose Profile
Previous Article in Special Issue
APOA1/C3/A4/A5 Gene Cluster at 11q23.3 and Lipid Metabolism Disorders: From Epigenetic Mechanisms to Clinical Practices
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Treating Cardiovascular Disease in the Inflammatory Setting of Rheumatoid Arthritis: An Ongoing Challenge

by
Saloni Godbole
,
Jenny Lue Solomon
,
Maryann Johnson
,
Ankita Srivastava
,
Steven E. Carsons
,
Elise Belilos
,
Joshua De Leon
and
Allison B. Reiss
*
Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(7), 1608; https://doi.org/10.3390/biomedicines12071608
Submission received: 29 April 2024 / Revised: 30 June 2024 / Accepted: 17 July 2024 / Published: 19 July 2024
(This article belongs to the Special Issue Emerging Trends in Lipoprotein and Cardiovascular Diseases)

Abstract

:
Despite progress in treating rheumatoid arthritis, this autoimmune disorder confers an increased risk of developing cardiovascular disease (CVD). Widely used screening protocols and current clinical guidelines are inadequate for the early detection of CVD in persons with rheumatoid arthritis. Traditional CVD risk factors alone cannot be applied because they underestimate CVD risk in rheumatoid arthritis, missing the window of opportunity for prompt intervention to decrease morbidity and mortality. The lipid profile is insufficient to assess CVD risk. This review delves into the connection between systemic inflammation in rheumatoid arthritis and the premature onset of CVD. The shared inflammatory and immunologic pathways between the two diseases that result in subclinical atherosclerosis and disrupted cholesterol homeostasis are examined. The treatment armamentarium for rheumatoid arthritis is summarized, with a particular focus on each medication’s cardiovascular effect, as well as the mechanism of action, risk–benefit profile, safety, and cost. A clinical approach to CVD screening and treatment for rheumatoid arthritis patients is proposed based on the available evidence. The mortality gap between rheumatoid arthritis and non-rheumatoid arthritis populations due to premature CVD represents an urgent research need in the fields of cardiology and rheumatology. Future research areas, including risk assessment tools and novel immunotherapeutic targets, are highlighted.

1. Introduction

Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by progressive inflammation in the synovial joints and extra-articular structures [1,2]. It is a systemic disease that can severely impair mobility and quality of life. Approximately 1% of the global population is affected [3,4,5,6]. The peak incidence of RA is between the ages of 30 and 50 years, and females are 2 to 3 times more commonly affected than males [7,8].
The incidence of coronary artery atherosclerosis is higher in persons with RA, making cardiovascular disease (CVD) a significant cause of morbidity and mortality for this patient group [9,10,11]. Individuals with RA have a 50% increased risk of developing CVD compared to the general population, and it has been estimated that persons with RA above age 45 have an almost three-fold higher risk of CVD death [12]. The prevalence of subclinical atherosclerosis is higher in RA patients than in healthy persons, and this difference remains after adjusting for traditional CVD risk factors [13]. In addition to sub-clinical atherosclerosis, CVD in RA may also manifest as silent ischemia and silent myocardial infarction [14,15]. Unfortunately, the accurate prediction of CVD in RA is difficult, and the measurement of Framingham risk factors or RA disease activity markers underestimate this risk [16,17,18,19]. The lack of biomarkers has impeded efforts to detect CVD before it manifests when preventive therapy or lifestyle modification could be instituted [20,21]. Further, clinical guidelines offer little direction in the selection of tailored therapeutics for the prevention and treatment of CVD in the setting of RA.
The purpose of this narrative review is to report the present state of understanding of how to best manage CVD and minimize CVD risk in persons diagnosed with RA. This review covers the effects of commonly used anti-rheumatic, atheroprotective, and lipid-lowering drugs on heart health in RA patients. A pragmatic approach to clinical care of this atherosclerosis-prone population is discussed.

2. Pathophysiology of Cardiovascular Diseases

CVD is an umbrella term for a group of diseases affecting the heart and blood vessels. There are many different types of CVD, which include coronary artery disease, heart failure, valvular heart disease, arrhythmia, peripheral artery disease, aortic disease, and deep vein thrombosis [22,23]. CVD is the leading cause of death globally. The incidence of CVD increases with increases in plasma cholesterol levels, arterial stiffness, and peripheral vascular resistance [24,25].
Atherosclerosis is the major cause of CVD-related death worldwide. Atherosclerosis is a chronic disease characterized by increased deposition of lipids, fibrous elements, and calcium within large arterial walls [26]. This process is triggered by damage to the endothelial cell monolayer of the arterial wall, resulting in endothelial activation. This further stimulates changes in transendothelial permeability and accumulation of lipids, particularly particles of low-density lipoprotein (LDL) cholesterol into the intima of the arterial wall [27]. Modified forms of LDL such as oxidized LDL also amass in the vascular wall and are strong chemoattractants for monocytes [28,29]. The activated endothelium expresses adhesion molecules, which also recruit monocytes that can then migrate into the subendothelial space, where they differentiate into resident macrophages and avidly take up modified LDLs, transforming into foam cells [30,31].
Oxidized LDL enters macrophages primarily via CD36 scavenger receptors. Internalized oxidized LDL can then induce pro-inflammatory cytokine and chemokine expression [32]. These processes lead to macrophage retention of a massive burden of intracellular cholesterol leading to macrophage foam cell transformation [33]. The foam cells produced will then become a crucial constituent of the fatty streak that forms in the subendothelial layer of the vessel wall and represents early atherosclerosis [34,35]. The activated macrophage secretion of various inflammatory cytokines and growth factors draws more circulating monocytes to the atherosclerotic lesion and stimulates smooth muscle cell proliferation and migration [36]. These processes constitute a summary of the major steps in atherosclerotic plaque pathogenesis.

3. Mechanisms and Role of Inflammation in Cardiovascular Disease (CVD)

The pathophysiology of RA involves inflammatory cascades, which lead to persistent synovial inflammation and eventually damage to surrounding cartilage and bone [37]. The same inflammatory cascades that lead to joint inflammation and destruction also contribute to increased atherosclerotic burden in patients with RA. Measures of disease activity in RA do not correlate directly with cardiovascular risk, and RA disease measures are not reliable for the prediction of CV events or mortality [38,39,40]. However, there are some markers with potential utility. For instance, the overexpression of tumor necrosis factor (TNF)-like cytokine 1A in the synovium of rheumatoid arthritis patients correlates with both disease activity and atheromatous plaque height and the formation of new plaques [41].
Cytokines are a key player in the inflammatory process and contribute to cardiovascular risk by recruiting immune cells, activating the endothelium, differentiating monocytes, forming foam cells, and causing plaque rupture and thrombosis [42,43]. In fact, the pro-inflammatory cytokines involved in the pathogenesis of atherosclerosis and RA have substantial overlap [44,45]. TNF-α, IL-6, and IL-1β are examples of cytokines involved in both RA and atherosclerotic CVD [46]. Interactions between T and B lymphocytes, fibroblasts, and macrophages lead to the overproduction of TNF-α, which in turn stimulates other cytokines known to promote inflammation, including IL-1β and IL-6 [47]. TNF-α drives endothelial dysfunction, one of the earliest stages of plaque formation in atherosclerosis [48]. It also lowers nitric oxide and thrombomodulin levels, creating a prothrombotic environment [49,50,51]. IL-1β provokes endothelial cells to upregulate surface adhesion molecule expression, thereby bringing about macrophage recruitment to the arterial intima [52]. Once the endothelial layer has been breached by inflammatory cells, cholesterol buildup occurs inside the artery. Monocytes are recruited into the subendothelial space, where they differentiate into macrophages, internalize oxidized lipoproteins, and transform into atherogenic foam cells [53]. As foam cells gather in particular locations in the arterial intima, they form a fatty streak, a precursor to the atherosclerotic plaque. Another key cytokine involved in all stages of atherogenesis is IFN-y. In addition to damaging vasculature via oxidative stress, IFN-y leads to plaque destabilization via smooth muscle cell apoptosis and the release of matrix metalloproteinases [54,55]. Not all cytokines involved in atherosclerosis are pro-inflammatory, however. TGF-β and IL-10 exert an atheroprotective role when released by regulatory T-cells [56,57]. IL-10 has been shown to have anti-inflammatory properties, which decelerate the progression of atherosclerosis by inhibiting inflammation and cell apoptosis [58]. Murine models of acute myocardial infarction treated with IL-10 demonstrated decreased inflammation and improved left ventricular function [59]. Conversely, atherosclerosis was accelerated in vivo with genetic inactivation of IL-10 [45]. Orecchioni et al. used the apolipoprotein (apo)E-deficient mouse model of atherosclerosis to show that resident and inflammatory macrophages in the aorta are major producers of IL-10 [60]. They then generated apoE-deficient mice with myeloid-specific inactivation of IL-10 and demonstrated that these mice exhibit a pro-inflammatory state, with significantly increased expression of TNF-ɑ, and CCL2, a chemokine linked to atherosclerosis as well as a trend toward increased IL-1β. This resulted in significantly increased total cholesterol levels and, when these mice were fed a Western diet, a five-fold increase in the size of atherosclerotic lesions.
The inflammatory pathways involved in RA not only cause endothelial activation as a route to cholesterol accumulation within vasculature but also disrupt cholesterol homeostasis, further increasing cardiovascular risk [61,62] (Figure 1). Cholesterol equilibrium in macrophages is maintained by three processes: cholesterol ingress through designated receptors, intracellular degradation by enzymes, and cholesterol movement out of the cell. Cell culture experiments in which human macrophages are incubated in human plasma have found that the plasma of persons diagnosed with RA causes a diminution in the level of macrophage cholesterol efflux proteins while also upregulating scavenger receptors that influx cholesterol. This pattern amplifies the macrophage cholesterol burden and foam cell formation [63]. The pervasive inflammatory milieu that exists in the RA patient influences the level and function of both low-density lipoprotein (LDL) and high-density lipoprotein (HDL). Studies have shown that when exposed to inflammation and oxidative stress, HDL can become pro-atherogenic rather than atheroprotective [64,65,66]. During acute or chronic inflammation, the HDL structure changes to include oxidized lipids and acute phase reactants such as serum amyloid A and ceruloplasmin, rather than cholesterol-transport proteins such as apoA-1 and antioxidant enzymes [67]. This pro-inflammatory HDL, or piHDL, results in impaired reverse cholesterol transport, decreased lipoprotein synthesis, and increased oxidation of LDL. RA patients were found to have a higher percentage of the most electronegative subfraction of HDL, which correlated with subclinical atherosclerosis [68]. Further confirming the relationship of RA to abnormal HDL, HDL from RA patients with high disease activity was shown to have a decreased ability to promote cholesterol efflux compared to HDL from patients with low disease activity or in clinical remission [69]. These researchers also found a significant inverse correlation of the erythrocyte sedimentation rate (ESR) with cholesterol efflux after controlling for confounding variables including diabetes, glucocorticoid use, and smoking. Paraoxonase (PON)-1, a glycoprotein located in the HDL particle, is responsible for the antioxidant properties of HDL. In RA patients, both the concentration of PON-1 and its enzymatic activity are reduced, and this reduction correlates with an increased carotid plaque burden found using ultrasound [70].
Inflammation-related disturbance in HDL functions, such as the inhibition of LDL oxidation and reverse cholesterol transport, may help explain why traditional lipid profile evaluations are insufficient for predicting cardiovascular risk for RA patients. Not only is the composition of HDL affected in RA, but LDL cholesterol is also different, with more citrullinated and homocitrullinated forms detected in RA patients than in healthy subjects [71]. Citrullinated and homocitrullinated LDL has greater atherogenic potential and has been shown to induce foam cell formation in cultured macrophages [72].

4. Current Treatment Options

The therapeutic armamentarium in RA has varying pro- and anti-atherogenic properties. Some agents, such as corticosteroids and non-steroidal anti-inflammatory drugs (NSAIDs), are well known to enhance atherosclerotic risk [73]. For others, such as methotrexate and biologics, the evidence is accumulating that certain agents may be beneficial for both RA management and risk attenuation for atherosclerosis. The following subsections describe recent scientific data on the most commonly used agents for RA treatment (summarized in Table 1).

4.1. Methotrexate

Methotrexate is the cornerstone of RA treatment [74]. The mechanism of action of methotrexate is the inhibition of the enzyme dihydrofolate reductase, which prevents the purine and pyrimidine formation needed for DNA and RNA synthesis [75]. Methotrexate offers protection against CVD both indirectly, through the reduction in overall inflammation, and directly via anti-atherogenic effects. Methotrexate has been shown to promote the outflow of cholesterol from macrophages, the enhancement of free radical scavenging, and the improvement in endothelial function [47]. Levels of adenosine, an endogenous nucleoside with many cardioprotective qualities, are increased by methotrexate [76,77]. Cardioprotective effects of adenosine include coronary artery vasodilation, the reduction in myocardial oxygen demand, and anti-platelet activity. Through the increased generation of adenosine with consequent adenosine A2A receptor activation, methotrexate promotes reverse cholesterol transport and limits foam cell formation in cultured THP-1 human macrophages, a widely used cellular model for macrophage behavior during atherosclerosis [78,79]. Furthermore, methotrexate treatment is associated with reduced risk of type 2 diabetes by improving insulin sensitivity via the increased extracellular adenosine concentration, promoting metabolism and transport of glucose [80]. As it is well-established that patients with diabetes are at higher mortality risk from CVD, treatment with methotrexate may have further value. The numerous cardioprotective mechanisms wielded by methotrexate add up to a real-world, tangible benefit for patients with RA. By some estimates, methotrexate use is associated with a 70% reduction in cardiovascular mortality and a 60% reduction in all-cause mortality [81]. Methotrexate treatment was shown to reduce cardiovascular risk in RA in a prospective cohort study using the Veterans Affairs Rheumatoid Arthritis (VARA) registry [82]. Data obtained on 2044 US veterans with RA showed that the lower CVD risk associated with methotrexate was independent of RA disease activity, age, body mass index, and traditional CVD risk factors such as hypertension, hyperlipidemia, and diabetes. These beneficial cardiovascular effects, along with their lower cost, established the effectiveness and safety profile that underscore methotrexate’s role as an initial treatment for RA patients [68].

4.2. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs)

NSAIDS are frequently employed as an initial or adjunctive strategy in a regimen designed to relieve the symptoms of pain and stiffness in RA and other rheumatologic conditions. The mechanism of action of NSAIDs is via the inhibition of vasodilatory cyclooxygenase (COX), which reduces pain and inflammation through the inhibition of prostaglandins [83,84]. COX-2 inhibitors preserve platelet function, thus favoring thrombosis. Even short-term use of NSAIDs has been demonstrated to raise the risk of cardiovascular events and stroke. A meta-analysis of 34 papers, with 28 focused on patients with RA and 6 directed at patients with psoriasis or psoriatic arthritis, found that CVD risk was higher for both groups with either non-selective or COX-2 NSAIDs [85]. Though the exact mechanism has not yet been determined, cell culture studies in THP-1 human macrophages show that the COX-2-selective inhibitor NS398 enhances lipid-overload and foam cell transformation via the downregulation of the reverse cholesterol transport proteins cholesterol 27-hydroxylase and ATP-binding cassette transporter (ABC)A1 [86]. In cultured human macrophages, celecoxib and rofecoxib increased foam cell formation, likely via the suppression of the cholesterol efflux proteins 27-hydroxylase and ABCA1 [87]. Celecoxib also upregulated CD36, a scavenger receptor responsible for cholesterol uptake, which would add to the macrophage lipid burden. Therefore, NSAIDs such as COX-2 inhibitors like celecoxib can both benefit RA patients while also negatively impacting lipid metabolism via interference with the transport of cholesterol. The Standard Care versus celecoxib Outcome Trial (SCOT) randomized 7297 persons over age 60 years with either RA or osteoarthritis to continue on their previously prescribed non-selective NSAID or switch to celecoxib, and after a median follow-up of 3 years, outcomes were comparable, with similar cardiovascular event rates in both COX-2 and non-selective NSAID treatment groups [88]. Though NSAIDs help manage RA symptoms, they are also known to cause gastrointestinal events, including iron deficiency anemia, and they carry the risk of nephrotoxicity, which is a consideration for certain patient populations. In the Prospective Randomized Evaluation of Celecoxib Integrated Safety vs. Ibuprofen or Naproxen (PRECISION) trial, the group of patients taking celecoxib had significantly fewer gastrointestinal events than the naproxen or ibuprofen groups, thus other comorbidities should be considered when choosing an NSAID for treating RA [83].

4.3. Leflunomide

The mechanism of action of leflunomide is the inhibition of dihydroorotate dehydrogenase, an enzyme essential for pyrimidine synthesis and, therefore, the proliferation of activated T-lymphocytes [89]. Direct associations between leflunomide and cardiovascular risk factors are not well-described. Leflunomide may indirectly affect cardiovascular risk via its effects on blood pressure, body weight, and plasma glucose concentrations. A post hoc analysis of 169 patients with RA showed that leflunomide use was associated with both decreased body weight and lower plasma glucose concentrations [90]. Ribosomal protein S6 kinase 1 (S6K1), which is a serine/threonine kinase downstream of mTOR, under high nutrition circumstances will remain activated, which induces insulin resistance and obesity; however, in mouse models, leflunomide inhibited S6K1 activity and promoted autophagy of adipocytes [91]. Leflunomide is associated with an increased incidence of hypertension. Drug-related hypertension occurs in approximately 2 to 4% of leflunomide-treated patients compared to 2% for comparator drugs including sulfasalazine and methotrexate [92]. Regarding safety profile, in a cohort study of RA patients taking leflunomide, severe infection occurred with an incidence of 3.3%, though these patients were also taking additional medications such as glucocorticoids or methotrexate, therefore making it difficult to assess leflunomide’s individual risk for infection [93]. While the most common side effect of leflunomide is gastrointestinal disturbance, hepatotoxicity is a concern, which is why it carries a black-boxed warning to monitor liver enzymes [89].

4.4. Sulfasalazine

The exact mechanism of action of sulfasalazine is not fully understood. Sulfasalazine is made up of sulfapyridine, an antibiotic, and 5-aminosalicyclic acid, an NSAID. It can inhibit platelet thromboxane synthetase and prevent arachidonic acid-mediated platelet aggregation, similar to the mechanism of aspirin [94]. Furthermore, it increases HDL, which is cardioprotective [95]. It also decreases inflammatory cytokines by inhibiting nuclear factor-κB (NF-κB), which promotes both inflammation and the adhesion that leads to monocyte recruitment in nascent atherogenic plaques. The inhibitory effect of sulfasalazine on NF-κB occurs due to the halting of the phosphorylation and breakdown of NF-κB’s inhibitory subunit IκB [47]. Of note, though the downregulation of TNF-α gene expression was observed in isolated mononuclear cells after exposure to sulfasalazine, this did not appear to correlate to changes in systemic inflammatory markers [96]. Sulfasalazine has also been associated with a small but significant reduction in cardiovascular events [97]. Sulfasalazine can have positive effects on the vasculature by inducing the enzyme HO-1, which, in turn, lowers reactive oxygen radicals and oxidative stress and may improve endothelial function in vessel walls [98,99,100]. Sulfasalazine is conditionally recommended by the American College of Rheumatology for patients with low disease activity over methotrexate because of its decreased immunosuppressive activity and for patients who wish to avoid the adverse effects of methotrexate [74].

4.5. Hydroxychloroquine

Hydroxychloroquine accumulates in and alkalinizes lysosomes, thereby inhibiting their functions [101]. Though the exact mechanism of action of hydroxychloroquine is unknown, it is thought to immunosuppress by blocking the proliferation of T-cells, halting macrophage cytokine production, and preventing toll-like receptor stimulation [102,103]. Hydroxychloroquine has been well-established to improve joint pain and prevent cartilage destruction, and it is safe to prescribe in pregnancy, though retinopathy is a concern for long-term use, for which patients should undergo regular ophthalmological screenings [104]. In a retrospective study of a cohort of 1266 RA patients, the risk of incident CVD was reduced by 72% with hydroxychloroquine treatment [105]. Part of its effect on preventing CVD may be in reducing cytokines like IL-1, IL-6, and TNF-α, which contribute to the formation of plaques [106]. Studies suggest that hydroxychloroquine is potentially anti-thrombotic via mechanisms involving platelet adhesion and interaction with coagulation factors [107]. In murine studies of RA by Shi et al., hydroxychloroquine both reduced atherosclerosis and increased microbiota flora in the gut [108]. The authors hypothesized that the atheroprotection was conferred due to decreased platelet aggregation and the cleaving of arachidonic acid from platelets as well as the suppression of antiphospholipid antibodies. The beneficial effects of hydroxychloroquine on the gut microbiota were attributed to an increase in Akkermansia muciniphila, which produces enzymes that decrease inflammation, produce an anti-atherosclerotic effect, and may prevent obesity. Additionally, hydroxychloroquine may improve outcomes in diabetic patients, mitigating a major risk factor for CVD. Hydroxychloroquine has been shown to increase the binding of insulin to its receptor, and it has also been shown to have a significant impact on increased insulin sensitivity [109].

4.6. Glucocorticoids

Glucocorticoids such as prednisone are a common treatment to suppress inflammation caused by RA. Glucocorticoids exert most of their effects by binding to intracellular glucocorticoid receptors, translocating to the nucleus, and changing gene expression [110]. They function as agonists of glucocorticoid and mineralocorticoid receptors, impacting ligand-activated transcription factors and directly affecting endothelial function [111]. However, their use may also increase the risk of cardiovascular events. They can promote oxidative stress and decrease nitric oxide (NO) in the body via downregulating endothelial NO gene transcription and expression as well as enhancing the production of reactive oxygen species, thereby diminishing NO bioavailability in the vasculature [111]. Yet, they are so frequently used due to their quick onset to relieve joint inflammation and pain as well as their affordable cost. Steroids have also been shown to increase cardiovascular events such as myocardial infarction, stroke, and heart failure in patients being treated for RA [85,112]. A study of steroid dose thresholds associated with all-cause and cardiovascular mortality revealed an incidence ratio of 1.89 for cardiovascular-related deaths associated with glucocorticoid use [113]. For these reasons, glucocorticoids are a clear example of the “double-edged sword” due to their swift therapeutic effect; therefore, from the available evidence, patients should be treated with the lowest dose for the shortest time possible to control the disease [114]. Moreover, glucocorticoids are well known to increase the risk of insulin resistance and hypertension [111,115,116]. For these reasons, glucocorticoids are an example of the “double-edged sword”, signifying that they have utility due to their swift therapeutic effect but also confer greater risk for CVD; therefore, patients should be treated with the lowest dose for the shortest time possible to control the disease [114]. Interestingly, in active RA patients, data are conflicting as to whether endothelial function is truly negatively impacted by glucocorticoids [111,115]. Confounding factors in prior studies, including the co-administration of other DMARDs, participants with a wide range of disease stages, and non-standardized dosages of glucocorticoids further affect the external validity of prior findings [111]. More research is necessary to clarify the effect of glucocorticoids on endothelial function, specifically in the setting of RA.

4.7. Janus Kinase Inhibitors

Janus kinase (JAK) inhibitors are non-receptor tyrosine kinases. Their mechanism of action is the transduction of cytokine signaling through the JAK signal transducer and activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) transcription pathway, which in turn controls the transcription of genes that modulate inflammatory conditions [117].
The ORAL Surveillance randomized controlled trial comparing tofacitinib 5 mg twice daily, tofacitinib 10 mg twice daily, and the TNF inhibitor adalimumab for RA demonstrated an increased risk of major adverse cardiovascular events (MACE) with a hazard ratio of 1.33 [118]. This led the FDA to issue black-box warnings for tofacitinib, baricitinib, and upadacitinib. Sakai et al. also found increased CVD event risk in RA patients given JAK inhibitor therapy in a retrospective longitudinal study from Japan [119]. Real-world registries have generated conflicting results. A nationwide population-based cohort study of the French national health system did not show a significantly increased risk of MACE in patients exposed to JAK inhibitors [120].
JAK inhibitor treatment can lead to increased total cholesterol, LDL cholesterol, HDL cholesterol, and triglycerides, though it has not been shown to result in a significantly higher risk of cardiovascular events [114,121]. Tofacitinib has been shown to increase the levels of HDL and LDL more than TNF inhibitors, possibly by reversing cholesterol ester catabolism [122,123]. Low doses of atorvastatin reverse the induced dyslipidemia [124]. Higher HDL levels with tofacitinib are associated with decreased adverse cardiovascular events in patients with RA via a proposed mechanism of alterations to HDL cholesterol fractions that convert them to become anti-inflammatory [125]. There is an inverse correlation between inflammation (as measured by CRP levels) and the levels of small-sized atheroprotective HDL particles [126]. Treatment of RA patients with JAK inhibitors was found to increase the levels of small-sized, atheroprotective HDL molecules. Interestingly, an open-label, prospective study of 46 patients with active RA treated with tofacitinib for 54 weeks resulted in decreased measures of atherosclerosis, including carotid intima-media thickness and pulse-wave velocity [127]. An in vitro study using THP-1 human macrophages showed that tofacitinib decreases intracellular cholesterol and the formation of foam cells by promoting cholesterol efflux to extracellular acceptors such as apoA-I, HDL, and whole normolipidemic human serum [128]. This effect may come about through several mechanisms, one of which is an increase in ABCA1. In addition, based on prior work showing that JAK-2 inhibition in human macrophages downregulates acyl-CoA cholesterol acyltransferase (ACAT), the authors postulate that another mechanism by which tofacitinib may improve cholesterol efflux is by enhancing ACAT expression and thus promoting cholesterol efflux [129]. As noted in this study, tofacitinib decreased intracellular cholesterol in the serum of patients with active inflammatory disease (specifically juvenile idiopathic arthritis), despite the inflammatory environment and defective lipoproteins. A small prospective observational study of microvascular and macrovascular function in 11 RA patients at baseline and 3 months after initiation of JAK inhibitor therapy showed no change in macrovascular structure or function but a significant change in capillaroscopic parameters consistent with vascular impairment [130]. The authors urge further investigation to confirm their findings and in order to assess the ramifications for the long-term cardiovascular impact. Another aspect to be considered in overall RA treatment is the steroid-sparing effect of using JAK inhibitors since reducing the use of glucocorticoids has cardiovascular benefits [131].

4.8. Abatacept

Abatacept is a selective co-stimulation modulator. Its mechanism of action is blocking T-cell activity via the CD80/CD86-CD28 costimulatory signal [132]. Treatment with abatacept has been associated with increased insulin sensitivity, which could mitigate the risk of CVD in RA patients with diabetes [133]. Compared to TNF inhibitors, Jin et al. found abatacept to be more protective against a new cardiovascular event and was not associated with higher venous thromboembolic events [134]. In an open-label, prospective, observational study from Japan, Yamada et al. also saw favorable effects of abatacept on the intima-media thickness of the common carotid artery in RA patients treated with abatacept, with smaller increases in the plaque score with abatacept [135]. However, other studies have shown no advantage when comparing biologic treatments and the risk of acute coronary syndrome [136]. Abatacept’s use appears to be generally safe and unlikely to increase the incidence of cardiovascular events [114,135]. Recently, synovial power Doppler ultrasound signals (PDUSs) have been studied in relation to abatacept treatment. Patients treated with abatacept for 3 months had greater decreases in PDUSs and improvement in HDL function compared to those with a lesser change in PDUSs [137]. For patients who were naïve to treatment with biologics, a PDUS signal was associated with significant antioxidant activity of HDL. Conversely, patients with the worst HDL function had greater synovitis. Furthermore, in a population of seropositive patients with moderate to severe RA, abatacept (as compared to adalimumab) had both better clinical efficacy and lower cost per responder due to its higher clinical efficacy [138].

4.9. TNF-α Inhibitors

Tumor necrosis factor alpha (TNF-α) inhibitors are a class of biologic medications that work by blocking TNF-α, a protein that causes inflammation and can lead to autoimmune diseases [139]. TNF-α also plays a pivotal role in inflammation and the development of atherosclerosis. Despite efficacy in treating joint disease, the effect of TNF inhibitors on cardiovascular risk remains a topic of debate [140]. Data from a Swedish registry demonstrated a mixed effect of TNF inhibitors on cardiovascular outcomes. Following the initiation of RA therapy, the efficacy of treatment can be assessed in patients via the European League Against Rheumatism (EULAR) response, in which a good response is defined as a significant change in the disease activity score (DAS)28 from baseline. RA patients with a good response to TNF inhibition at 5 ± 3 months exhibited a 40–50% decrease in risk for acute coronary syndrome at 1 year. RA patients who were non-responders and did not have a significant change in DAS28 had rates of acute coronary syndrome at 1 year, more than two times higher than the age- and sex-matched general population [141]. Hussain et al. hypothesized that the varying effects of TNF inhibition on the cardiovascular system are due to the differing roles of tumor necrosis factor receptor 1 (TNFR1) and tumor necrosis factor receptor 2 (TNFR2) in cardiac tissue [142]. TNFR1 is an apoptotic receptor, and its inhibition by TNF-α inhibitors is subsequently cardioprotective. However, TNFR2 is primarily cardioprotective, and its greater inhibition may explain the cardiovascular morbidity associated with TNF-α inhibitors. A systematic review undertaken by Nair et al. found that treatment of RA patients with a TNF-α inhibitor did reduce cardiovascular events, including myocardial infarction, stroke, transient ischemic attack, and coronary artery disease [143]. Other studies have not shown a lower incidence of MI with TNF inhibitor use compared with RA patients treated with traditional DMARDs [144]. This finding may be explained by the neutral effect of TNF inhibition on cholesterol transport at the level of the macrophage. In work from our lab using cultured human macrophages, adalimumab did not affect the levels of multiple reverse cholesterol transport protein mRNAs, including ABCA1, ABCG1, and 27-hydroxylase (unpublished data). This was similarly reflected in a lack of change in macrophage lipid efflux with no significant difference in effluxed cholesterol as a percent of the total (intracellular and extracellular) cholesterol either with or without exposure to adalimumab (U = 19, p = 0.5350, n = 6 per group). TNF inhibitors may counteract atherosclerosis development in patients with RA by reducing levels of cell surface heparan sulfate proteoglycans and improving endothelial dysfunction [145,146,147].

4.10. Tocilizumab

Tocilizumab is a humanized anti-IL-6 receptor antibody that binds to both soluble and membrane-bound IL-6 receptors. Its mechanism of action is the inhibition of IL-6-mediated signaling through these receptors [148]. IL-6 is associated with plaque destabilization, microvascular flow dysfunction, and adverse outcomes in the setting of acute ischemia [149]. Theoretically, IL-6 inhibition should decrease cardiovascular risk. The findings in the literature are mixed. One systematic review and meta-analysis of 14 cohort studies found that tocilizumab was associated with a lower risk of MACE as compared to TNF inhibitors [150]. The ENTRACTE trial, a randomized controlled trial investigating the cardiovascular safety of tocilizumab versus etanercept, found no significant difference in the number of MACE events between the two groups [151]. The tocilizumab group had a median 11.1% greater increase in LDL cholesterol levels, a median 5.4% greater increase in HDL cholesterol levels, and a median 13.6% greater increase in triglyceride levels compared to the etanercept group at 4 weeks (each p < 0.001), which persisted over time. Tocilizumab-induced hyperlipidemia has been shown in multiple other studies, without a corresponding increase in cardiovascular risk [152,153].

4.11. Rituximab

Rituximab is a chimeric anti-CD-20 monoclonal antibody that exerts its mechanism of action via the depletion of B cells [154]. It promotes both antibody-dependent and complement-dependent cytotoxicity along with upregulating B cell apoptosis, though the exact mechanism underlying its effectiveness in RA treatment is not wholly understood [155]. It is an approved therapeutic agent in RA for patients who do not respond to DMARD therapy. Its efficacy has been established by various studies. Rituximab has been found to significantly improve patient-reported symptoms, decrease joint damage confirmed by radiographic imaging, and initiate remission for some patients, in one case for almost a decade [154]. The side-effect profile of rituximab mainly concerns developing or reactivating infections as well as the decreased function of immunizations. Rituximab has not been shown to increase the risk of MACE events or cause cardiac dysfunction [155]. An open-label longitudinal study found that there were significant reductions in inflammatory markers, disease activity scores, and pulse-wave velocities over time in patients who received rituximab [156]. Improvement in endothelial function can be seen after a single dose of rituximab within 24 weeks [157]. Patients who responded clinically to rituximab were also noted to have improved lipid profiles, decreased arterial stiffness, and carotid intima-media thickness, whereas non-responders did not have significant changes in the aforementioned values [158]. Whether these changes culminate in a clinically significant decrease in cardiovascular risk remains to be seen.

4.12. Statins

Statins inhibit the enzyme HMG-CoA reductase and block the conversion of HMG-CoA to mevalonic acid. This reduces hepatic cholesterol synthesis and increases cell surface LDL receptor expression [159]. In the general population, statins are used to decrease the risk of cardiovascular events. The role of statins in mitigating the adverse effects of dyslipidemia in RA is not clear. Statins decrease vascular inflammation with postulated mechanisms involving downregulated IL-6 expression and lowering of homocysteine levels [160,161,162,163]. However, in the RA patient, this is complicated by the lipid paradox. The lipid paradox refers to the association between low lipid levels and active inflammation, leading to worse symptoms. For instance, patients with high disease activity and elevated inflammatory markers often have low LDL and total cholesterol, though with a higher risk of CVD [164]. Studies such as the Trial of Atorvastatin for the Primary Prevention of Cardiovascular Events in Patients with Rheumatoid Arthritis (TRACE RA), which was the largest academically led RA clinical trial, have not been able to establish whether statin therapy would benefit RA patients for primary prevention [165]. The findings from TRACE RA suggest that RA patients experience the same benefits for cardiovascular protection as the general population. However, RA patients had lower rates of cardiovascular events than expected, and this caused the study to be terminated early. Therefore, this study did not support using statins as primary prevention against cardiovascular events in RA patients. The decision to prescribe statins should be made on a case-by-case basis, taking into account the full clinical picture. Smaller studies have shown that statins reduce arterial stiffness and carotid plaque; moreover, other studies have found that stopping statins was associated with poorer outcomes [166]. High-intensity statins not only stopped the formation of new atherosclerotic plaques but they have also been shown to promote the regression of existing plaques as well as encourage the calcification and stabilization of plaques, thereby decreasing the long-term cardiovascular risk of RA patients [167]. Further trials with large sample sizes and longer follow-ups are needed to better define the role of statins in the management of CVD in RA.
Table 1. Medications used in rheumatoid arthritis and their effect on cardiovascular risk.
Table 1. Medications used in rheumatoid arthritis and their effect on cardiovascular risk.
Medication Name or CategoryNet Effect on
Cardiovascular Risk
Putative
Mechanism
References
MethotrexateBeneficialIncreased adenosine via inhibition of adenosine deamination[74,75,76,77,78,79,80,81]
NSAIDs HarmfulInhibition of cyclooxygenase
enzymes, interferes with
reverse cholesterol transport
[83,84,85,86,87,88]
LeflunomideNeutralInhibition of dihydroorotate
dehydrogenase
[89,90,91,92]
SulfasalazineLikely BeneficialDecreased platelet aggregation,
inhibition of NF-κB, improved
endothelial function
[74,94,95,96,97,98,99,100]
HydroxychloroquineBeneficialOverall reduction in inflammation,
possibly anti-thrombotic
[101,102,103,104,105,106,107,108,109]
GlucocorticoidsHarmfulIncreased oxidative stress
and decreased nitric oxide.
Increased adiposity and
insulin resistance.
[85,110,111,112,113,114,115,116]
JAK inhibitorsPotential for harm in
certain patients
Suppress the JAK-STAT
signaling pathway
[117,118,119,120,121,122,123,124,125,126,127,128,129,130]
AbataceptNeutralCD80/CD86-CD28
costimulatory modulator
[132,133,134,135,136,137,138]
TNF-α inhibitorsLikely beneficialInhibition of TNFR1,
amelioration of endothelial
dysfunction
[139,140,141,142,143,144,145,146,147]
TocilizumabNeutralAnti-IL-6 receptor antibody, may
increase cholesterol levels
[148,149,150,151,152,153]
RituximabNeutralAnti-CD20[154,155,156,157,158]
StatinsBeneficialLower LDL
cholesterol, improve lipid profile, anti-inflammatory
[159,160,161,162,163,164,165,166]
Abbreviations: IL—interleukin; JAK-STAT—Janus kinase-signal transducer and activator of transcription; LDL—low-density lipoprotein; NF-κB—nuclear factor-κB; NSAIDs—non-steroidal anti-inflammatory drugs; TNF—tumor necrosis factor; TNFR1—tumor necrosis factor receptor 1.

5. Clinical Context

In the absence of established treatment guidelines for CVD in RA, we propose the following screening and treatment paradigm based on the available evidence. One recommendation made by the American Heart Association (AHA) is to assess the risk for atherosclerotic CVD during times of low disease activity because low cholesterol levels during flares of disease can underestimate the atherosclerotic CVD risk [168]. Furthermore, in patients for whom CAD is suspected, coronary computed tomography angiography (CTA), comprised of a non-enhanced and a contrast-enhanced CT scan, may be used to evaluate the coronary artery calcification score to guide treatment. The non-contrast portion of the procedure provides a coronary artery calcium score and, in a large-scale cohort study from Tinggaard et al., a trend toward increased prevalence of calcification was found for seropositive RA patients and patients requiring treatment for relapse or flare [169]. Further compounding the issue of risk assessment in the context of RA is the fact that the atherosclerotic CVD risk calculator is not validated for RA patients and does not consider several factors, for example, the use of glucocorticoids.
Currently, there are no established total cholesterol or LDL goals for RA patients; however, applying goals for diabetic patients may be helpful, considering a similarly elevated risk. The AHA/ACC recommends initiating statin therapy for adults 40 to 75 without diabetes and an intermediate ten-year risk (7.5% to 19.9%) and “risk-enhancing factors”, which include chronic inflammatory disorders such as RA [168]. Inflammation can cause instability and rupture of atherosclerotic plaques, so the risk of myocardial infarction in a patient with RA has been estimated to be similar to patients without RA who have diabetes or are ten years older [170]. Patients with untreated RA or those with a strong disease burden tend to have lower total cholesterol, HDL-C, LDL-C, and triglycerides [171]. This is potentially due to a high level of inflammation limiting the efflux of HDL-C [172]. Treatment to decrease inflammation, in fact, can increase lipid levels but also decrease cardiovascular risk. Total cholesterol had a weak association with myocardial infarction in RA patients as compared to the general population, as found by the AMORIS study, implying that decreasing lipid levels of patients with RA may not be effective as a protective factor against myocardial infarction [173]. However, TRACE RA found that after the initiation of atorvastatin 40 mg daily, there was a decrease of 42% in cardiovascular event risk reduction per mmole/Liter decrease in LDL, yet this treatment did not achieve a significant reduction in cardiovascular risk (hazard ratio [HR] 0.66, 95% CI 0.39–1.11) [165].
Previous sections of this paper have highlighted how inflammatory cytokines such as TNF-α, IL-6, and IL-1β contribute to both the RA disease process and atherosclerosis [46,174] (Figure 2). Observational data suggest that controlling inflammation in RA may favorably affect some CVD risk factors and reduce the development and progression of CVD [175]. The data support the first-line use of methotrexate for RA and its cardiovascular manifestations: among eight large cohorts, methotrexate use was associated with a pooled 28% reduction in CVD event risk [85]. TNF inhibitors are likely a reasonable second choice if methotrexate therapy is contraindicated or insufficient, though evidence for the cardiovascular benefit is mixed. JAK inhibitors, particularly tofacitinib, should be avoided in patients over the age of 65 with other cardiovascular risk factors based on the results of the ORAL surveillance study [118]. Likewise, glucocorticoid and NSAID use in patients with pre-existing cardiovascular risk factors should be avoided given the higher associated risk for adverse cardiovascular events [113,176].
It is also vital to acknowledge how the biases of the studies referred to in this paper may impact the selection of treatment regimens for RA patients in clinical practice. Some limitations for generalizability include the exclusion of diabetic patients in TRACE RA, which may be an important confounding factor as diabetes is a major risk factor for CVD [165]. The predominantly older male population of veterans studied for methotrexate’s impact on CVD risk affects the generalizability to the generally younger female population that is most affected by RA [82]. The ORAL surveillance study could not fully assess the effect of pack years of smoking for RA patients and MACE risk when smoking is a major risk factor for the severity of RA as well as for CVD [118]. This review lays the foundation for clinicians to guide the selection of RA medications based on the existing data (see Table 1).

6. Conclusions and Future Perspectives

The mortality gap between RA and non-RA populations due to premature ASCVD represents an urgent research need in the fields of cardiology and rheumatology [177]. A recent study conducted in Norway has shown that patients with RA who are on stable anti-rheumatic therapy that included methotrexate in over 97% of subjects had no elevation in CVD risk above the general population, while the non-stable group had elevated CVD risk [178]. This kind of study may help to distinguish the higher-CVD-risk RA patients who need more intense monitoring [179]. Important areas of focus include a cardiovascular risk assessment tool validated for use in patients with RA. Such a tool could help identify at-risk patients earlier than the widely used Framingham risk assessment score [180]. Artificial intelligence may increase accuracy in ASCVD risk prediction in RA [181]. The lipid paradox in RA, discussed previously, is a second possible area for intervention. A method other than statin therapy is essential to alter pro-inflammatory HDL and inhibit atherosclerosis [67]. Current agents under investigation include those directed at raising the anti-atherogenic lipid apo-A1, others that inhibit cholesteryl ester transfer protein (CETP), which increases HDL cholesterol, and bempedoic acid, which inhibits the de novo synthesis of cholesterol and triglycerides [182,183,184]. These approaches have yielded mixed results [185]. Another potential immunotherapeutic target is IL-10, which, along with TGF-β, inhibits the development of atheromas [57,186]. The management of hypertension is also crucial [187]. Furthermore, the development of specialized centers focused on CVD screening and treatment within rheumatic diseases is warranted.
RA is an independent risk factor for coronary artery disease [188,189]. The chronic activation of inflammatory cytokine pathways may be the shared mechanism that leads to both joint symptoms and an increased atherosclerotic burden. Several medications for RA have cardiovascular benefits. However, CVD remains the leading cause of death in patients with RA, despite major advancements in the treatment of joint disease. As we await new developments in preventing and treating CVD in RA patients, rheumatologists, cardiologists, and primary care providers must recognize and convey the heightened risk to patients, aggressively screen for and treat traditional CVD risk factors, and treat RA disease activity to a target of low disease activity or remission.

Author Contributions

Conceptualization, A.B.R., S.E.C. and E.B.; methodology, E.B., S.G. and A.B.R.; validation, M.J., A.S. and J.L.S.; investigation, S.G., A.S. and A.B.R.; writing—original draft preparation, S.G., J.L.S., J.D.L. and A.B.R.; writing—review and editing, A.B.R., J.D.L. and E.B.; visualization, M.J.; supervision, A.B.R. and E.B.; project administration, A.B.R. and S.E.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This study was conducted in memory of Elizabeth Daniell and Lt. Col. E.L. Daniell. We thank Robert Buescher for his dedication to medical research. We offer a special thank you to Amy Glass for formatting references. With immense gratitude, we thank Maggie Lachcik and Rachel Schnabl from our development team. We offer a special thank you to Maryann Johnson for figure preparation.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cai, Y.; Zhang, J.; Liang, J.; Xiao, M.; Zhang, G.; Jing, Z.; Lv, L.; Nan, K.; Dang, X. The Burden of Rheumatoid Arthritis: Findings from the 2019 Global Burden of Diseases Study and Forecasts for 2030 by Bayesian Age-Period-Cohort Analysis. J. Clin. Med. 2023, 12, 1291. [Google Scholar] [CrossRef]
  2. Nie, Q.; Luo, Q.; Yan, W.; Zhang, T.; Wang, H.; Wu, J. Rheumatoid arthritis and coronary atherosclerosis: A two-sample Mendelian randomization study. Front. Cardiovasc. Med. 2023, 10, 1033644. [Google Scholar] [CrossRef]
  3. Turesson, C. Comorbidity in rheumatoid arthritis. Swiss Med. Wkly. 2016, 146, 14290. [Google Scholar] [CrossRef]
  4. van der Woude, D.; van der Helm-van Mil, A.H.M. Update on the epidemiology, risk factors, and disease outcomes of rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2018, 32, 174–187. [Google Scholar] [CrossRef]
  5. Lin, Y.J.; Anzaghe, M.; Schülke, S. Update on the Pathomechanism, Diagnosis, and Treatment Options for Rheumatoid Arthritis. Cells 2020, 9, 880. [Google Scholar] [CrossRef]
  6. Jahid, M.; Khan, K.U.; Rehan-Ul-Haq; Ahmed, R.S. Overview of Rheumatoid Arthritis and Scientific Understanding of the Disease. Mediterr. J. Rheumatol. 2023, 34, 284–291. [Google Scholar] [CrossRef]
  7. Finckh, A.; Gilbert, B.; Hodkinson, B.; Bae, S.C.; Thomas, R.; Deane, K.D.; Alpizar-Rodriguez, D.; Lauper, K. Global epidemiology of rheumatoid arthritis. Nat. Rev. Rheumatol. 2022, 18, 591–602. [Google Scholar] [CrossRef]
  8. Yu, F.; Chen, H.; Li, Q.; Tao, M.; Jin, Z.; Geng, L.; Sun, L. Secular trend of mortality and incidence of rheumatoid arthritis in global, 1990-2019: An age period cohort analysis and joinpoint analysis. BMC Pulm. Med. 2023, 23, 356. [Google Scholar] [CrossRef]
  9. Ali, A.; Ali, A.; Kumar, D.; Kumar, R.; Elahi, K.; Suman, F.; Anjum, Z.; Tharwani, P.; Jahangir, M.; Rizwan, A. Comparison of Incidence of Myocardial Infarction in Patients with Rheumatoid Arthritis and Diabetes Mellitus. Cureus 2021, 13, e15716. [Google Scholar] [CrossRef]
  10. Taylor, P.C.; 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, 509. [Google Scholar] [CrossRef]
  11. Guo, Y.; Chung, W.; Shan, Z.; Zhu, Z.; Costenbader, K.H.; Liang, L. Genome-Wide Assessment of Shared Genetic Architecture Between Rheumatoid Arthritis and Cardiovascular Diseases. J. Am. Heart Assoc. 2023, 12, e030211. [Google Scholar] [CrossRef]
  12. Widdifield, J.; Paterson, J.M.; Huang, A.; Bernatsky, S. Causes of death in rheumatoid arthritis: How do they compare to the general population? Arthritis Care Res. 2018, 70, 1748–1755. [Google Scholar] [CrossRef]
  13. Arora, A.; Ingle, V.; Joshi, R.; Malik, R.; Khandelwal, G. Exploring the Subclinical Atherosclerotic Load in Patients With Rheumatoid Arthritis: A Cross-Sectional Study. Cureus 2022, 14, e32644. [Google Scholar] [CrossRef]
  14. Bernardes, M.; Vieira, T.S.; Martins, M.J.; Lucas, R.; Costa, L.; Pereira, J.G.; Ventura, F.; Martins, E. Myocardial Perfusion in Rheumatoid Arthritis Patients: Associations with Traditional Risk Factors and Novel Biomarkers. BioMed Res Int. 2017, 2017, 6509754. [Google Scholar] [CrossRef]
  15. Rouached, L.; Tekaya, R.; Ben Ayed, H.; Bouden, S.; Ben Tekaya, A.; Ben Ahmed, H.; Mahmoud, I.; Saidane, O.; Abdelmoula, L. Screening of Silent Myocardial Ischaemia Using a Stress Test in Rheumatoid Arthritis Patients: Association with Traditional Risk Factors and Disease Activity. Musculoskelet. Care 2023, 21, 997–1004. [Google Scholar] [CrossRef]
  16. Arts, E.E.; Popa, C.; Den Broeder, A.A.; Semb, A.G.; Toms, T.; Kitas, G.D.; van Riel, P.L.; Fransen, J. Performance of four current risk algorithms in predicting cardiovascular events in patients with early rheumatoid arthritis. Ann. Rheum. Dis. 2015, 74, 668–674. [Google Scholar] [CrossRef]
  17. Chodara, A.M.; Wattiaux, A.; Bartels, C.M. Managing Cardiovascular Disease Risk in Rheumatoid Arthritis: Clinical Updates and Three Strategic Approaches. Curr. Rheumatol. Rep. 2017, 19, 16. [Google Scholar] [CrossRef]
  18. Nikiphorou, E.; de Lusignan, S.; Mallen, C.D.; Khavandi, K.; Bedarida, G.; Buckley, C.D.; Galloway, J.; Raza, K. Cardiovascular risk factors and outcomes in early rheumatoid arthritis: A population-based study. Heart 2020, 106, 1566–1572. [Google Scholar] [CrossRef]
  19. Dessein, P.H.; Gonzalez-Gay, M.A. Management of Cardiovascular Disease Risk in Rheumatoid Arthritis. J. Clin. Med. 2022, 11, 3487. [Google Scholar] [CrossRef]
  20. Crowson, C.S.; Matteson, E.L.; Roger, V.L.; Therneau, T.M.; Gabriel, S.E. Usefulness of risk scores to estimate the risk of cardiovascular disease in patients with rheumatoid arthritis. Am. J. Cardiol. 2012, 110, 420–424. [Google Scholar] [CrossRef]
  21. Södergren, A.; Karp, K.; Bengtsson, C.; Möller, B.; Rantapää-Dahlqvist, S.; Wållberg-Jonsson, S. Biomarkers associated with cardiovascular disease in patients with early rheumatoid arthritis. PLoS ONE 2019, 14, e0220531. [Google Scholar] [CrossRef]
  22. Frąk, W.; Wojtasińska, A.; Lisińska, W.; Młynarska, E.; Franczyk, B.; Rysz, J. Pathophysiology of Cardiovascular Diseases: New Insights into Molecular Mechanisms of Atherosclerosis, Arterial Hypertension, and Coronary Artery Disease. Biomedicines 2022, 10, 1938. [Google Scholar] [CrossRef]
  23. Scott, J. Pathophysiology and biochemistry of cardiovascular disease. Curr. Opin. Genet. Dev. 2004, 14, 271–279. [Google Scholar] [CrossRef]
  24. Ciumărnean, L.; Milaciu, M.V.; Runcan, O.; Vesa, S.C.; Răchișan, A.L.; Negrean, V.; Perné, M.; Donca, V.I.; Alexescu, T.; Para, I.; et al. The Effects of Flavonoids in Cardiovascular Diseases. Molecules 2020, 25, 4320. [Google Scholar] [CrossRef]
  25. Baba, M.; Maris, M.; Jianu, D.; Luca, C.T.; Stoian, D.; Mozos, I. The Impact of the Blood Lipids Levels on Arterial Stiffness. J. Cardiovasc. Dev. Dis. 2023, 10, 127. [Google Scholar] [CrossRef]
  26. Jebari-Benslaiman, S.; Galicia-García, U.; Larrea-Sebal, A.; Olaetxea, J.R.; Alloza, I.; Vandenbroeck, K.; Benito-Vicente, A.; Martin, C. Pathophysiology of Atherosclerosis. Int. J. Mol. Sci. 2022, 3, 3346. [Google Scholar] [CrossRef]
  27. Mundi, S.; Massaro, M.; Scoditti, E.; Carluccio, M.A.; van Hinsbergh, V.W.M.; Iruela-Arispe, M.L.; De Caterina, R. Endothelial permeability, LDL deposition, and cardiovascular risk factors—A review. Cardiovasc. Res. 2018, 114, 35–52. [Google Scholar] [CrossRef]
  28. Bergheanu, S.C.; Bodde, M.C.; Jukema, J.W. Pathophysiology and treatment of atherosclerosis: Current view and future perspective on lipoprotein modification treatment. Neth. Heart J. 2017, 25, 231–242. [Google Scholar] [CrossRef]
  29. Kowara, M.; Cudnoch-Jedrzejewska, A. Pathophysiology of Atherosclerotic Plaque Development–Contemporary Experience and New Directions in Research. Int. J. Mol. Sci. 2021, 22, 3513. [Google Scholar] [CrossRef]
  30. Mahdinia, E.; Shokri, N.; Taheri, A.T.; Asgharzadeh, S.; Elahimanesh, M.; Najafi, M. Cellular crosstalk in atherosclerotic plaque microenvironment. Cell Commun. Signal. 2023, 21, 125. [Google Scholar] [CrossRef]
  31. Allahverdian, S.; Chehroudi, A.C.; McManus, B.M.; Abraham, T.; Francis, G.A. Contribution of Intimal Smooth Muscle Cells to Cholesterol Accumulation and Macrophage-Like Cells in Human Atherosclerosis. Circulation 2014, 129, 1551–1559. [Google Scholar] [CrossRef]
  32. Sakakura, K.; Nakano, M.; Otsuka, F.; Ladich, E.; Kolodgie, F.D.; Virmani, R. Pathophysiology of atherosclerosis plaque progression. Heart Lung Circ. 2013, 22, 399–411. [Google Scholar] [CrossRef]
  33. Mushenkova, N.V.; Bezsonov, E.E.; Orekhova, V.A.; Popkova, T.V.; Starodubova, A.V.; Orekhov, A.N. Recognition of Oxidized Lipids by Macrophages and Its Role in Atherosclerosis Development. Biomedicines 2021, 9, 915. [Google Scholar] [CrossRef]
  34. Stary, H.C.; Chandler, A.B.; Glagov, S.; Guyton, J.R.; Insull, W.; Rosenfeld, M.E.; Schaffer, S.A.; Schwartz, C.J.; Wagner, W.D.; Wissler, R.W. A definition of initial, fatty streak, and intermediate lesions of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 1994, 89, 2462–2478. [Google Scholar] [CrossRef]
  35. Reiss, A.B.; Cronstein, B.N. Regulation of foam cells by adenosine. Arterioscler. Thromb. Vasc. Biol. 2012, 3, 879–886. [Google Scholar] [CrossRef]
  36. Cao, G.; Xuan, X.; Hu, J.; Zhang, R.; Jin, H.; Dong, H. How vascular smooth muscle cell phenotype switching contributes to vascular disease. Cell Commun. Signal. 2022, 20, 180. [Google Scholar] [CrossRef]
  37. Scott, D.L.; Wolfe, F.; Huizinga, T.W. Rheumatoid arthritis. Lancet 2010, 376, 1094–1108. [Google Scholar] [CrossRef]
  38. Book, C.; Saxne, T.; Jacobsson, L.T. Prediction of mortality in rheumatoid arthritis based on disease activity markers. J. Rheumatol. 2005, 32, 430–434. [Google Scholar]
  39. Kramer, H.R.; Giles, J.T. Cardiovascular disease risk in rheumatoid arthritis: Progress, debate, and opportunity. Arthritis Care Res. 2011, 63, 484–499. [Google Scholar] [CrossRef]
  40. Bathon, J.M.; Centola, M.; Liu, X.; Jin, Z.; Ji, W.; Knowlton, N.S.; Ferraz-Amaro, I.; Fu, Q.; Giles, J.T.; Wasko, M.C.; et al. Identification of novel biomarkers for the prediction of subclinical coronary artery atherosclerosis in patients with rheumatoid arthritis: An exploratory analysis. Arthritis Res. Ther. 2023, 25, 213. [Google Scholar] [CrossRef]
  41. Bamias, G.; Stamatelopoulos, K.; Zampeli, E.; Protogerou, A.; Sigala, F.; Papamichael, C.; Christopoulos, P.; Kitas, G.D.; Sfikakis, P.P. Circulating levels of TNF-like cytokine 1A correlate with the progression of atheromatous lesions in patients with rheumatoid arthritis. Clin. Immunol. 2013, 147, 144–150. [Google Scholar] [CrossRef]
  42. Arida, A.; Protogerou, A.D.; Kitas, G.D.; Sfikakis, P.P. Systemic Inflammatory Response and Atherosclerosis: The Paradigm of Chronic Inflammatory Rheumatic Diseases. Int. J. Mol. Sci. 2018, 19, 1890. [Google Scholar] [CrossRef]
  43. Markin, A.M.; Markina, Y.V.; Bogatyreva, A.I.; Tolstik, T.V.; Chakal, D.A.; Breshenkov, D.G.; Charchyan, E.R. The Role of Cytokines in Cholesterol Accumulation in Cells and Atherosclerosis Progression. Int. J. Mol. Sci. 2023, 24, 6426. [Google Scholar] [CrossRef]
  44. Cavagna, L.; Boffini, N.; Cagnotto, G.; Inverardi, F.; Grosso, V.; Caporali, R. Atherosclerosis and rheumatoid arthritis: More than a simple association. Mediat. Inflamm. 2012, 2012, 147354. [Google Scholar] [CrossRef]
  45. Popescu, D.; Rezus, E.; Badescu, M.C.; Dima, N.; Seritean Isac, P.N.; Dragoi, I.-T.; Rezus, C. Cardiovascular Risk Assessment in Rheumatoid Arthritis: Accelerated Atherosclerosis, New Biomarkers, and the of Biological Therapy. Life 2023, 13, 319. [Google Scholar] [CrossRef]
  46. Ku, I.A.; Imboden, J.B.; Hsue, P.Y.; Ganz, P. Rheumatoid arthritis: Model of systemic inflammation driving atherosclerosis. Circ. J. 2009, 73, 977–985. [Google Scholar] [CrossRef]
  47. Ahmed, S.; Jacob, B.; Carsons, S.E.; De Leon, J.; Reiss, A.B. Treatment of Cardiovascular Disease in Rheumatoid Arthritis: A Complex Challenge with Increased Atherosclerotic Risk. Pharmaceuticals 2021, 15, 11. [Google Scholar] [CrossRef]
  48. Gimbrone, M.A., Jr.; García-Cardeña, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620. [Google Scholar] [CrossRef]
  49. Searles, C.D. Transcriptional and posttranscriptional regulation of endothelial nitric oxide synthase expression. Am. J. Physiol. Cell Physiol. 2006, 291, C803–C816. [Google Scholar] [CrossRef]
  50. Forstermann, U.; Xia, N.; Li, H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef]
  51. Page, M.J.; Bester, J.; Pretorius, E. The inflammatory effects of TNF and complement component 3 on coagulation. Sci. Rep. 2018, 8, 1812. [Google Scholar] [CrossRef] [PubMed]
  52. Schunk, S.J.; Triem, S.; Schmit, D.; Zewinger, S.; Sarakpi, T.; Becker, E.; Hütter, G.; Wrublewsky, S.; Küting, F.; Hohl, M.; et al. Interleukin-1α Is a Central Regulator of Leukocyte-Endothelial Adhesion in Myocardial Infarction and in Chronic Kidney Disease. Circulation 2021, 144, 893–908. [Google Scholar] [CrossRef] [PubMed]
  53. Poznyak, A.V.; Nikiforov, N.G.; Starodubova, A.V.; Popkova, T.V.; Orekhov, A.N. Macrophages and Foam Cells: Brief Overview of Their Role, Linkage, and Targeting Potential in Atherosclerosis. Biomedicines 2021, 9, 1221. [Google Scholar] [CrossRef]
  54. Voloshyna, I.; Littlefield, M.J.; Reiss, A.B. Atherosclerosis and interferon-γ: New insights and therapeutic targets. Trends Cardiovasc. Med. 2014, 24, 45–51. [Google Scholar] [CrossRef] [PubMed]
  55. Elyasi, A.; Voloshyna, I.; Ahmed, S.; Kasselman, L.J.; Behbodikhah, J.; De Leon, J.; Reiss, A.B. The role of interferon-γ in cardiovascular disease: An update. Inflamm. Res. 2020, 69, 975–988. [Google Scholar] [CrossRef] [PubMed]
  56. Kim, M.; Sahu, A.; Hwang, Y.; Kim, G.B.; Nam, G.H.; Kim, I.S.; Chan Kwon, I.; Tae, G. Targeted delivery of anti-inflammatory cytokine by nanocarrier reduces atherosclerosis in Apo E-/- mice. Biomaterials 2020, 226, 119550. [Google Scholar] [CrossRef] [PubMed]
  57. Tabares-Guevara, J.H.; Villa-Pulgarin, J.A.; Hernandez, J.C. Atherosclerosis: Immunopathogenesis and strategies for immunotherapy. Immunotherapy 2021, 13, 1231–1244. [Google Scholar] [CrossRef]
  58. Filatova, A.Y.; Pylaeva, E.A.; Potekhina, A.V.; Ruleva, N.Y.; Klesareva, E.A.; Radyukhina, N.V.; Masenko, V.P.; Shchinova, A.M.; Noeva, E.A.; Provatorov, S.I.; et al. Low Blood content of IL-10-producing C4+ T cells as a risk factor for progression of coronary atherosclerosis. Bull. Exp. Biol. Med. 2019, 166, 330–333. [Google Scholar] [CrossRef] [PubMed]
  59. Krishnamurthy, P.; Rajasingh, J.; Lambers, E.; Qin, G.; Losordo, D.W.; Kishore, R. IL-10 inhibits inflammation and attenuates left ventricular remodeling after myocardial infarction via activation of STAT3 and suppression of HuR. Circ. Res. 2009, 104, e9–e18. [Google Scholar] [CrossRef]
  60. Orecchioni, M.; Wolf, D.; Suryawanshi, V.; Winkels, H.; Kobiyama, K.; Makings, J.; Kiosses, W.B.; Ley, K. Deleting interleukin-10 from myeloid cells exacerbates atherosclerosis in Apoe-/- mice. Cell. Mol. Life Sci. 2022, 80, 10. [Google Scholar] [CrossRef]
  61. Coomes, E.; Chan, E.S.; Reiss, A.B. Methotrexate in atherogenesis and cholesterol metabolism. Cholesterol 2011, 2011, 503028. [Google Scholar] [CrossRef] [PubMed]
  62. Daidone, M.; Del Cuore, A.; Casuccio, A.; Di Chiara, T.; Guggino, G.; Di Raimondo, D.; Puleo, M.G.; Ferrante, A.; Scaglione, R.; Pinto, A.; et al. Vascular health in subjects with rheumatoid arthritis: Assessment of endothelial function indices and serum biomarkers of vascular damage. Intern. Emerg. Med. 2023, 18, 467–475. [Google Scholar] [CrossRef] [PubMed]
  63. Voloshyna, I.; Modayil, S.; Littlefield, M.J.; Belilos, E.; Belostocki, K.; Bonetti, L.; Rosenblum, G.; Carsons, S.E.; Reiss, A.B. Plasma from rheumatoid arthritis patients promotes pro-atherogenic cholesterol transport gene expression in THP-1 human macrophages. Exp. Biol. Med. 2013, 238, 1192–1197. [Google Scholar] [CrossRef]
  64. Alisik, T.; Alisik, M.; Nacir, B.; Ayhan, F.F.; Genc, H.; Erel, O. Evaluation of dysfunctional high-density lipoprotein levels with myeloperoxidase/paraoxonase-1 ratio in rheumatoid arthritis. Int. J. Clin. Pract. 2021, 75, e14172. [Google Scholar] [CrossRef] [PubMed]
  65. Vyletelová, V.; Nováková, M.; Pašková, Ľ. Alterations of HDL’s to piHDL’s Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals 2022, 15, 1278. [Google Scholar] [CrossRef] [PubMed]
  66. García-Gómez, M.C.; Padró, T.; Muñoz-García, N.; Bianchi, M.; Álvarez, L.; Badimon, L.; Corbella, E.; Pintó, X. Dysfunctional antioxidant capacity of high-density lipoprotein in rheumatoid arthritis. Eur. J. Clin. Investig. 2023, 53, e13999. [Google Scholar] [CrossRef] [PubMed]
  67. Hahn, B.H.; Grossman, J.; Ansell, B.J.; Skaggs, B.J.; McMahon, M. Altered lipoprotein metabolism in chronic inflammatory states: Proinflammatory high-density lipoprotein and accelerated atherosclerosis in systemic lupus erythematosus and rheumatoid arthritis. Arthritis Res. Ther. 2008, 10, 213. [Google Scholar] [CrossRef] [PubMed]
  68. Chang, C.K.; Cheng, W.C.; Ma, W.L.; Chen, P.K.; Chen, C.H.; Shen, P.C.; Chen, C.C.; Chang, S.H.; Lai, Y.H.; Chen, D.Y. The Potential Role of Electronegative High-Density Lipoprotein H5 Subfraction in RA-Related Atherosclerosis. Int. J. Mol. Sci. 2021, 22, 11419. [Google Scholar] [CrossRef] [PubMed]
  69. Charles-Schoeman, C.; Lee, Y.Y.; Grijalva, V.; Amjadi, S.; FitzGerald, J.; Ranganath, V.K.; Taylor, M.; McMahon, M.; Paulus, H.E.; Reddy, S.T. Cholesterol efflux by high density lipoproteins is impaired in patients with active rheumatoid arthritis. Ann. Rheum. Dis. 2012, 71, 1157–1162. [Google Scholar] [CrossRef]
  70. Parada-Turska, J.; Wójcicka, G.; Beltowski, J. Paraoxonase 1 Phenotype and Protein N-Homocysteinylation in Patients with Rheumatoid Arthritis: Implications for Cardiovascular Disease. Antioxidants 2020, 9, 899. [Google Scholar] [CrossRef]
  71. de Groot, L.; Hinkema, H.; Westra, J.; Smit, A.J.; Kallenberg, C.G.; Bijl, M.; Posthumus, M.D. Advanced glycation endproducts are increased in rheumatoid arthritis patients with controlled disease. Arthritis Res. Ther. 2011, 13, R205. [Google Scholar] [CrossRef]
  72. Rajamohan, A.; Heit, B.; Cairns, E.; Barra, L. Citrullinated and homocitrullinated low-density lipoprotein in rheumatoid arthritis. Scand. J. Rheumatol. 2021, 50, 343–350. [Google Scholar] [CrossRef]
  73. Giles, J.T. Cardiovascular disease in rheumatoid arthritis: Current perspectives on assessing and mitigating risk in clinical practice. Best Pract. Res. Clin. Rheumatol. 2015, 29, 597–613. [Google Scholar] [CrossRef]
  74. Fraenkel, L.; Bathon, J.M.; England, B.R.; St Clair, E.W.; Arayssi, T.; Carandang, K.; Deane, K.D.; Genovese, M.; Huston, K.K.; Kerr, G.; et al. 2021 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Care Res. 2021, 73, 924–939. [Google Scholar] [CrossRef]
  75. Sramek, M.; Neradil, J.; Veselska, R. Much more than you expected: The non-DHFR-mediated effects of methotrexate. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 499–503. [Google Scholar] [CrossRef]
  76. Friedman, B.; Cronstein, B. Methotrexate mechanism in treatment of rheumatoid arthritis. Jt. Bone Spine 2019, 86, 301–307. [Google Scholar] [CrossRef] [PubMed]
  77. Reiss, A.B.; Grossfeld, D.; Kasselman, L.J.; Renna, H.A.; Vernice, N.A.; Drewes, W.; Konig, J.; Carsons, S.E.; DeLeon, J. Adenosine and the Cardiovascular System. Am. J. Cardiovasc. Drugs 2019, 19, 449–464. [Google Scholar] [CrossRef] [PubMed]
  78. Reiss, A.B.; Carsons, S.E.; Anwar, K.; Rao, S.; Edelman, S.D.; Zhang, H.; Fernandez, P.; Cronstein, B.N.; Chan, E.S. Atheroprotective effects of methotrexate on reverse cholesterol transport proteins and foam cell transformation in human THP-1 monocyte/macrophages. Arthritis Rheum. 2008, 58, 3675–3683. [Google Scholar] [CrossRef] [PubMed]
  79. Qin, Z. The use of THP-1 cells as a model for mimicking the function and regulation of monocytes and macrophages in the vasculature. Atherosclerosis 2012, 221, 2–11. [Google Scholar] [CrossRef]
  80. Yang, D.; Haemmig, S.; Zhou, H.; Perez-Cremades, D.; Sun, X.; Chen, L.; Li, J.; Haneo-Mejia, J.; Yang, T.; Hollan, I.; et al. Methotrexate attenuates vascular inflammation through an adenosine-microRNA-dependent pathway. eLife 2021, 10, e58064. [Google Scholar] [CrossRef]
  81. Choi, H.K.; Hernán, M.A.; Seeger, J.D.; Robins, J.M.; Wolfe, F. Methotrexate and mortality in patients with rheumatoid arthritis: A prospective study. Lancet 2002, 359, 1173–1177. [Google Scholar] [CrossRef] [PubMed]
  82. Johnson, T.M.; Sayles, H.R.; Baker, J.F.; George, M.D.; Roul, P.; Zheng, C.; Sauer, B.; Liao, K.P.; Anderson, D.R.; Mikuls, T.R.; et al. Investigating changes in disease activity as a mediator of cardiovascular risk reduction with methotrexate use in rheumatoid arthritis. Ann. Rheum. Dis. 2021, 80, 1385–1392. [Google Scholar] [CrossRef]
  83. Nissen, S.E.; Yeomans, N.D.; Solomon, D.H.; Lüscher, T.F.; Libby, P.; Husni, M.E.; Graham, D.Y.; Borer, J.S.; Wisniewski, L.M.; Wolski, K.E.; et al. Cardiovascular Safety of Celecoxib, Naproxen, or Ibuprofen for Arthritis. N. Engl. J. Med. 2016, 375, 2519–2529. [Google Scholar] [CrossRef] [PubMed]
  84. Ghosh, R.; Alajbegovic, A.; Gomes, A.V. NSAIDs and cardiovascular diseases: Role of reactive oxygen species. Oxid. Med. Cell. Longev. 2015, 2015, 536962. [Google Scholar] [CrossRef] [PubMed]
  85. Roubille, C.; Richer, V.; Starnino, T.; McCourt, C.; McFarlane, A.; Fleming, P.; Siu, S.; Kraft, J.; Lynde, C.; Pope, J.; et al. The effects of tumour necrosis factor inhibitors, methotrexate, non-steroidal anti-inflammatory drugs and corticosteroids on cardiovascular events in rheumatoid arthritis, psoriasis and psoriatic arthritis: A systematic review and meta-analysis. Ann. Rheum. Dis. 2015, 74, 480–489. [Google Scholar] [CrossRef]
  86. Chan, E.S.; Zhang, H.; Fernandez, P.; Edelman, S.D.; Pillinger, M.H.; Ragolia, L.; Palaia, T.; Carsons, S.; Reiss, A.B. Effect of COX inhibition on cholesterol efflux proteins and atheromatous foam cell transformation in THP-1 human macrophages: A possible mechanism for increased cardiovascular risk. Arthritis Res. Ther. 2007, 9, R4. [Google Scholar] [CrossRef]
  87. Voloshyna, I.; Kasselman, L.J.; Carsons, S.E.; Littlefield, M.J.; Gomolin, I.H.; De Leon, J.; Reiss, A.B. COX-2-dependent and independent effects of COX-2 inhibitors and NSAIDs on proatherogenic changes in human monocytes/macrophages. J. Investig. Med. 2017, 65, 694–704. [Google Scholar] [CrossRef]
  88. MacDonald, T.M.; Hawkey, C.J.; Ford, I.; McMurray, J.J.V.; Scheiman, J.M.; Hallas, J.; Findlay, E.; Grobbee, D.E.; Hobbs, F.D.R.; Ralston, S.H.; et al. Randomized trial of switching from prescribed non-selective non-steroidal anti-inflammatory drugs to prescribed celecoxib: The Standard care vs. Celecoxib Outcome Trial (SCOT). Eur. Heart J. 2016, 38, 1843–1850. [Google Scholar] [CrossRef]
  89. Alamri, R.D.; Elmeligy, M.A.; Albalawi, G.A.; Alquayr, S.M.; Alsubhi, S.S.; El-Ghaiesh, S.H. Leflunomide an immunomodulator with antineoplastic and antiviral potentials but drug-induced liver injury: A comprehensive review. Int. Immunopharmacol. 2021, 93, 107398. [Google Scholar] [CrossRef]
  90. Rho, Y.H.; Oeser, A.; Chung, C.P.; Milne, G.L.; Stein, C.M. Drugs Used in the Treatment of Rheumatoid Arthritis: Relationship between Current Use and Cardiovascular Risk Factors. Arch. Drug Inf. 2009, 2, 34–40. [Google Scholar] [CrossRef]
  91. Ji, X.; Chen, J.; You, C.; Sun, J.; Xu, X. Leflunomide alleviates obesity via activation of the TAK1-AMPK pathway and induction of lipophagy. FASEB J. 2023, 37, e23227. [Google Scholar] [CrossRef] [PubMed]
  92. Nurmohamed, M.T.; van Halm, V.P.; Dijkmans, B.A.C. Cardiovascular Risk Profile of Antirheumatic Agents in Patients with Osteoarthritis and Rheumatoid Arthritis. Drugs 2002, 62, 1599–1609. [Google Scholar] [CrossRef]
  93. Jenks, K.A.; Stamp, L.K.; O’Donnell, J.L.; Savage, R.L.; Chapman, P.T. Leflunomide-associated infections in rheumatoid arthritis. J. Rheumatol. 2007, 34, 2201–2203. [Google Scholar]
  94. MacMullan, P.A.; Madigan, A.M.; Paul, N.; Peace, A.J.; Alagha, A.; Nolan, K.B.; McCarthy, G.M.; Kenny, D. Sulfasalazine and its metabolites inhibit platelet function in patients with inflammatory arthritis. Clin. Rheumatol. 2016, 35, 447–455. [Google Scholar] [CrossRef]
  95. Park, Y.B.; Choi, H.K.; Kim, M.Y.; Lee, W.K.; Song, J.; Kim, D.K.; Lee, W.K.; Song, J.; Kim, D.K.; Lee, S.K. Effects of antirheumatic therapy on serum lipid levels in patients with rheumatoid arthritis: A prospective study. Am. J. Med. 2002, 113, 188–193. [Google Scholar] [CrossRef]
  96. Tabit, C.E.; Holbrook, M.; Shenouda, S.M.; Dohadwala, M.M.; Widlansky, M.E.; Frame, A.A.; Kim, B.H.; Duess, M.A.; Kluge, M.A.; Levit, A.; et al. Effect of sulfasalazine on inflammation and endothelial function in patients with established coronary artery disease. Vasc. Med. 2012, 17, 101–107. [Google Scholar] [CrossRef] [PubMed]
  97. Naranjo, A.; Sokka, T.; Descalzo, M.A.; Calvo-Alén, J.; Hørslev-Petersen, K.; Luukkainen, R.K.; Combe, B.; Burmester, G.R.; Devlin, J.; Ferraccioli, G.; et al. Cardiovascular disease in patients with rheumatoid arthritis: Results from the QUEST-RA study. Arthritis Res. Ther. 2008, 10, R30. [Google Scholar] [CrossRef] [PubMed]
  98. Kim, J.Y.; Cho, H.J.; Sir, J.J.; Kim, B.K.; Hur, J.; Youn, S.W.; Yang, H.M.; Jun, S.I.; Park, K.W.; Hwang, S.J.; et al. Sulfasalazine induces haem oxygenase-1 via ROS-dependent Nrf2 signalling, leading to control of neointimal hyperplasia. Cardiovasc. Res. 2009, 82, 550–560. [Google Scholar] [CrossRef]
  99. Brownfoot, F.C.; Hannan, N.J.; Cannon, P.; Nguyen, V.; Hastie, R.; Parry, L.J.; Senadheera, S.; Tuohey, L.; Tong, S.; Kaitu’u-Lino, T. Sulfasalazine reduces placental secretion of antiangiogenic factors, up-regulates the secretion of placental growth factor and rescues endothelial dysfunction. EBioMedicine 2019, 41, 636–648. [Google Scholar] [CrossRef]
  100. Sonmez, M.I.; Shahzadi, A.; Kose, C.; Sonmez, H.; Ozyazgan, S.; Akkan, A.G. Effect of sulfasalazine on endothelium-dependent vascular response by the activation of Nrf2 signalling pathway. Front. Pharmacol. 2022, 13, 979300. [Google Scholar] [CrossRef]
  101. Ponticelli, C.; Moroni, G. Hydroxychloroquine in systemic lupus erythematosus (SLE). Expert Opin. Drug Saf. 2017, 16, 411–419. [Google Scholar] [CrossRef] [PubMed]
  102. Kuznik, A.; Bencina, M.; Svajger, U.; Jeras, M.; Rozman, B.; Jerala, R. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J. Immunol. 2011, 186, 4794–4804. [Google Scholar] [CrossRef] [PubMed]
  103. Wallace, D.J.; Gudsoorkar, V.S.; Weisman, M.H.; Venuturupalli, S.R. New insights into mechanisms of therapeutic effects of antimalarial agents in SLE. Nat. Rev. Rheumatol. 2012, 8, 522–533. [Google Scholar] [CrossRef] [PubMed]
  104. Nirk, E.L.; Reggiori, F.; Mauthe, M. Hydroxychloroquine in rheumatic autoimmune disorders and beyond. EMBO Mol. Med. 2020, 12, e12476. [Google Scholar] [CrossRef] [PubMed]
  105. Sharma, T.S.; Wasko, M.C.; Tang, X.; Vedamurthy, D.; Yan, X.; Cote, J.; Bili, A. Hydroxychloroquine Use Is Associated with Decreased Incident Cardiovascular Events in Rheumatoid Arthritis Patients. J. Am. Heart Assoc. 2016, 5, e002867. [Google Scholar] [CrossRef] [PubMed]
  106. Silva, J.C.; Mariz, H.A.; Rocha, L.F.; Oliveira, P.S.; Dantas, A.T.; Duarte, A.L.; da Rocha Pitta, I.; Galdino, S.L.; Pitta, M.G. Hydroxychloroquine decreases Th17-related cytokines in systemic lupus erythematosus and rheumatoid arthritis patients. Clinics 2013, 68, 766–771. [Google Scholar] [CrossRef] [PubMed]
  107. Ruiz-Irastorza, G.; Egurbide, M.V.; Pijoan, J.I.; e Garmendia, M.; Villar, I.; Martinez-Berriotxoa, A.; Erdozain, J.G.; Aguirre, C. Effect of antimalarials on thrombosis and survival in patients with systemic lupus erythematosus. Lupus 2006, 15, 77–583. [Google Scholar] [CrossRef] [PubMed]
  108. Shi, N.; Zhang, S.; Silverman, G.; Mengtao, L.; Cai, J.; Niu, H. Protective effect of hydroxychloroquine on rheumatoid arthritis-associated atherosclerosis. Anim. Model. Exp. Med. 2019, 2, 98–106. [Google Scholar] [CrossRef] [PubMed]
  109. Hage, M.P.; Al-Badri, M.R.; Azar, S.T. A favorable effect of hydroxychloroquine on glucose and lipid metabolism beyond its anti-inflammatory role. Ther. Adv. Endocrinol. Metab. 2014, 5, 77–85. [Google Scholar] [CrossRef] [PubMed]
  110. Verhoeven, F.; Prati, C.; Maguin-Gaté, K.; Wendling, D.; Demougeot, C. Glucocorticoids and endothelial function in inflammatory diseases: Focus on rheumatoid arthritis. Arthritis Res. Ther. 2016, 18, 258. [Google Scholar] [CrossRef]
  111. Ramamoorthy, S.; Cidlowski, J.A. Corticosteroids: Mechanisms of Action in Health and Disease. Rheum. Dis. Clin N. Am. 2016, 42, 15–31. [Google Scholar] [CrossRef] [PubMed]
  112. Ocon, A.J.; Reed, G.; Pappas, D.A.; Curtis, J.R.; Kremer, J.M. Short-term dose and duration-dependent glucocorticoid risk for cardiovascular events in glucocorticoid-naive patients with rheumatoid arthritis. Ann. Rheum. Dis. 2021, 80, 1522–1529. [Google Scholar] [CrossRef] [PubMed]
  113. del Rincón, I.; Battafarano, D.F.; Restrepo, J.F.; Erikson, J.M.; Escalante, A. Glucocorticoid dose thresholds assoiated with all-cause and cardiovascular mortality in rheumatoid arthritis. Arthritis Rheumatol. 2014, 66, 264–272. [Google Scholar] [CrossRef] [PubMed]
  114. Atzeni, F.; Rodríguez-Carrio, J.; Popa, C.D.; Nurmohamed, M.T.; Szűcs, G.; Szekanecz, Z. Cardiovascular effects of approved drugs for rheumatoid arthritis. Nat. Rev. Rheumatol. 2021, 17, 270–290. [Google Scholar] [CrossRef] [PubMed]
  115. Petersons, C.J.; Mangelsdorf, B.L.; Poljak, A.; Smith, M.D.; Greenfield, J.R.; Thompson, C.H.; Burt, M.G. Low dose prednisolone and insulin sensitivity differentially affect arterial stiffness and endothelial function: An open interventional and cross-sectional study. Atherosclerosis 2017, 258, 34–39. [Google Scholar] [CrossRef] [PubMed]
  116. Pofi, R.; Caratti, G.; Ray, D.W.; Tomlinson, J.W. Treating the Side Effects of Exogenous Glucocorticoids; Can We Separate the Good From the Bad? Endocr. Rev. 2023, 44, 975–1011. [Google Scholar] [CrossRef] [PubMed]
  117. Xu, P.; Shen, P.; Yu, B.; Xu, X.; Ge, R.; Cheng, X.; Chen, Q.; Bian, J.; Li, Z.; Wang, J. Janus kinases (JAKs): The efficient therapeutic targets for autoimmune diseases and myeloproliferative disorders. Eur. J. Med. Chem. 2020, 192, 112155. [Google Scholar] [CrossRef] [PubMed]
  118. Ytterberg, S.R.; Bhatt, D.L.; Mikuls, T.R.; Koch, G.G.; Fleischmann, R.; Rivas, J.L.; Germino, R.; Menon, S.; Sun, Y.; Wang, C.; et al. Cardiovascular and Cancer Risk with Tofacitinib in Rheumatoid Arthritis. N. Engl. J. Med. 2022, 386, 316–326. [Google Scholar] [CrossRef] [PubMed]
  119. Sakai, R.; Tanaka, E.; Inoue, E.; Harigai, M. Increased risk of cardiovascular events under the treatments with Janus kinase inhibitors versus biological disease-modifying antirheumatic drugs in patients with rheumatoid arthritis: A retrospective longitudinal population-based study using the Japanese health insurance database. RMD Open 2024, 10, e003885. [Google Scholar] [CrossRef] [PubMed]
  120. Hoisnard, L.; Pina Vegas, L.; Dray-Spira, R.; Weill, A.; Zureik, M.; Sbidian, E. Risk of major adverse cardiovascular and venous thromboembolism events in patients with rheumatoid arthritis exposed to JAK inhibitors versus adalimumab: A nationwide cohort study. Ann. Rheum. Dis. 2023, 82, 182–188. [Google Scholar] [CrossRef]
  121. Corrao, S. Crucial safety issues on Janus kinase inhibitors in rheumatoid arthritis might be associated with the lack of LDL-cholesterol management: A reasoned literature analysis. Intern. Emerg. Med. 2023, 18, 2157–2161. [Google Scholar] [CrossRef] [PubMed]
  122. Alzahrani, Z.; Alhazmi, A.; Almalki, H.; Aljehani, N.; Dumyati, M.; Alabdali, H. Efficacy and Safety of Tofacitinib in Rheumatoid Arthritis (RA): A Retrospective Study from Two Centers in Jeddah, Saudi Arabia. Cureus 2022, 14, e32240. [Google Scholar] [CrossRef]
  123. Czókolyová, M.; Hamar, A.; Pusztai, A.; Tajti, G.; Végh, E.; Pethő, Z.; Bodnár, N.; Horváth, Á.; Soós, B.; Szamosi, S.; et al. Effects of One-Year Tofacitinib Therapy on Lipids and Adipokines in Association with Vascular Pathophysiology in Rheumatoid Arthritis. Biomolecules 2022, 12, 1483. [Google Scholar] [CrossRef]
  124. McInnes, I.B.; Kim, H.Y.; Lee, S.H.; Mandel, D.; Song, Y.W.; Connell, C.A.; Luo, Z.; Brosnan, M.J.; Zuckerman, A.; Zwillich, S.H.; et al. Open-label and double-blind atorvastatin in rheumatoid arthritis patients: A randomised study. Ann. Rheum. Dis. 2014, 73, 124–131. [Google Scholar] [CrossRef] [PubMed]
  125. Charles-Schoeman, C.; DeMasi, R.; Valdez, H.; Soma, K.; Hwang, L.J.; Boy, M.G.; Biswas, P.; McInnes, I.B. Risk factors for major adverse cardiovascular events in phase III and long-term extension studies of tofacitinib in patients with rheumatoid arthritis. Arthritis Rheumatol. 2019, 71, 1450–1459. [Google Scholar] [CrossRef]
  126. Chang, C.K.; Chiang, E.I.; Chang, K.H.; Tang, K.T.; Chen, P.K.; Yip, H.T.; Chen, C.H.; Chen, D.Y. The Sizes and Composition of HDL-Cholesterol Are Significantly Associated with Inflammation in Rheumatoid Arthritis Patients. Int. J. Mol. Sci. 2023, 24, 10645. [Google Scholar] [CrossRef]
  127. Kume, K.; Amano, K.; Yamada, S.; Kanazawa, T.; Ohta, H.; Hatta, K.; Amano, K.; Kuwaba, N. Tofacitinib improves atherosclerosis despite up-regulating serum cholesterol in patients with active rheumatoid arthritis: A cohort study. Rheumatol. Int. 2017, 37, 2079–2085. [Google Scholar] [CrossRef] [PubMed]
  128. Adorni, M.P.; Papotti, B.; Borghi, M.O.; Raschi, E.; Zimetti, F.; Bernini, F.; Meroni, P.L.; Ronda, N. Effect of the JAK/STAT Inhibitor Tofacitinib on Macrophage Cholesterol Metabolism. Int. J. Mol. Sci. 2023, 24, 12571. [Google Scholar] [CrossRef]
  129. Hongo, S.; Watanabe, T.; Arita, S.; Kanome, T.; Kageyama, H.; Shioda, S.; Miyazaki, A. Leptin modulates ACAT1 expression and cholesterol efflux from human macrophages. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E474–E482. [Google Scholar] [CrossRef]
  130. Anyfanti, P.; Angeloudi, E.; Dara, A.; Pagkopoulou, E.; Moysidou, G.S.; Deuteraiou, K.; Boutel, M.; Bekiari, E.; Doumas, M.; Kitas, G.D.; et al. Non-Invasive Assessment of Micro- and Macrovascular Function after Initiation of JAK Inhibitors in Patients with Rheumatoid Arthritis. Diagnostics 2024, 14, 834. [Google Scholar] [CrossRef]
  131. Conigliaro, P.; Minerba, C.; Vendola, A.; Fiannacca, L.; Triggianese, P.; Kroegler, B.; Greco, E.; Bergamini, A.; Chimenti, M.S. The steroid-sparing effect of JAK inhibitors across multiple patient populations. Front. Immunol. 2024, 15, 1376476. [Google Scholar] [CrossRef] [PubMed]
  132. Bonelli, M.; Scheinecker, C. How does abatacept really work in rheumatoid arthritis? Curr. Opin. Rheumatol. 2018, 30, 295–300. [Google Scholar] [CrossRef] [PubMed]
  133. Ursini, F.; Russo, E.; Letizia Hribal, M.; Mauro, D.; Savarino, F.; Bruno, C.; Tripolino, C.; Rubino, M.; Naty, S.; Grembiale, R.D. Abatacept improves whole-body insulin sensitivity in rheumatoid arthritis: An observational study. Medicine 2015, 94, e888. [Google Scholar] [CrossRef] [PubMed]
  134. Jin, Y.; Kang, E.H.; Brill, G.; Desai, R.J.; Kim, S.C. Cardiovascular (CV) risk after initiation of abatacept versus TNF inhibitors in rheumatoid arthritis patients with and without baseline CV disease. J. Rheumatol. 2018, 45, 1240–1248. [Google Scholar] [CrossRef] [PubMed]
  135. Yamada, Z.; Muraoka, S.; Kawazoe, M.; Hirose, W.; Kono, H.; Yasuda, S.; Sugihara, T.; Nanki, T. Long-term effects of abatacept on atherosclerosis and arthritis in older vs. younger patients with rheumatoid arthritis: 3-year results of a prospective, multicenter, observational study. Arthritis Res. Ther. 2024, 26, 87. [Google Scholar] [CrossRef] [PubMed]
  136. Delcoigne, B.; Ljung, L.; Provan, S.A.; Glintborg, B.; Hetland, M.L.; Grøn, K.L.; Peltomaa, R.; Relas, H.; Turesson, C.; Gudbjornsson, B.; et al. Short-term, intermediate-term and long-term risks of acute coronary syndrome in cohorts of patients with RA starting biologic DMARDs: Results from four Nordic countries. Ann. Rheum. Dis. 2022, 81, 789–797. [Google Scholar] [CrossRef] [PubMed]
  137. Charles-Schoeman, C.; Wang, J.; Shahbazian, A.; Wilhalme, H.; Brook, J.; Kaeley, G.S.; Oganesian, B.; Ben-Artzi, A.; Elashoff, D.A.; Ranganath, V.K. Power doppler ultrasound signal predicts abnormal HDL function in patients with rheumatoid arthritis. Rheumatol. Int. 2023, 43, 1041–1053. [Google Scholar] [CrossRef] [PubMed]
  138. Park, S.H.; Han, X.; Lobo, F.; Nanji, S.; Patel, D. A Cost per Responder Model for Abatacept versus Adalimumab Among Rheumatoid Arthritis Patients with Seropositivity. Clinicoecon. Outcomes Res. 2020, 12, 589–594. [Google Scholar] [CrossRef] [PubMed]
  139. Jang, D.I.; Lee, A.H.; Shin, H.Y.; Song, H.R.; Park, J.H.; Kang, T.B.; Lee, S.R.; Yang, S.H. The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int. J. Mol. Sci. 2021, 22, 2719. [Google Scholar] [CrossRef]
  140. Delbaere, Q.; Chapet, N.; Huet, F.; Delmas, C.; Mewton, N.; Prunier, F.; Angoulvant, D.; Roubille, F. Anti-Inflammatory Drug Candidates for Prevention and Treatment of Cardiovascular Diseases. Pharmaceuticals 2023, 16, 78. [Google Scholar] [CrossRef]
  141. Ljung, L.; Rantapää-Dahlqvist, S.; Jacobsson, L.T.; Askling, J. Response to biological treatment and subsequent risk of coronary events in rheumatoid arthritis. Ann. Rheum. Dis. 2016, 75, 2087–2094. [Google Scholar] [CrossRef] [PubMed]
  142. Hussain, A.; Tarahomi, T.; Singh, L.; Bollampally, M.; Heydari-Kamjani, M.; Kesselman, M.M. Cardiovascular Risk Associated With TNF Alpha Inhibitor Use in Patients With Rheumatoid Arthritis. Cureus 2021, 13, e17938. [Google Scholar] [CrossRef] [PubMed]
  143. Nair, S.; Singh Kahlon, S.; Sikandar, R.; Peddemul, A.; Tejovath, S.; Hassan, D.; Patel, K.K.; Mostafa, J.A. Tumor Necrosis Factor-Alpha Inhibitors and Cardiovascular Risk in Rheumatoid Arthritis: A Systematic Review. Cureus 2022, 14, e26430. [Google Scholar] [CrossRef] [PubMed]
  144. Avouac, J.; Allanore, Y. Cardiovascular risk in rheumatoid arthritis: Effects of anti-TNF drugs. Expert Opin. Pharmacother. 2008, 9, 1121–1128. [Google Scholar] [CrossRef] [PubMed]
  145. Calvo Alén, J.; Lavin-Gomez, B.A.; Aurrecoechea, E.; Guerra Ruiz, A.R.; Martínez Taboada, V.; Gómez Gerique, J. TNF Inhibitors Exert a “Hidden” Beneficial Effect in the Cardiovascular Lipoprotein Profile of RA Patients. Biologics 2022, 16, 187–197. [Google Scholar] [CrossRef] [PubMed]
  146. Szeremeta, A.; Jura-Półtorak, A.; Zoń-Giebel, A.; Olczyk, K.; Komosińska-Vassev, K. TNF-α Inhibitors in Combination with MTX Reduce Circulating Levels of Heparan Sulfate/Heparin and Endothelial Dysfunction Biomarkers (sVCAM-1, MCP-1, MMP-9 and ADMA) in Women with Rheumatoid Arthritis. J. Clin. Med. 2022, 11, 4213. [Google Scholar] [CrossRef] [PubMed]
  147. Thota, L.N.R.; Chignalia, A.Z. The role of the glypican and syndecan families of heparan sulfate proteoglycans in cardiovascular function and disease. Am. J. Physiol. Cell Physiol. 2022, 323, C1052–C1060. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, M.; Chen, L.; He, J.; Xia, W.; Ye, Z.; She, J. Structural insights into IL-6 signaling inhibition by therapeutic antibodies. Cell Rep. 2024, 43, 113819. [Google Scholar] [CrossRef] [PubMed]
  149. Ridker, P.M. From C-Reactive Protein to Interleukin-6 to Interleukin-1: Moving Upstream to Identify Novel Targets for Atheroprotection. Circ. Res. 2016, 118, 145–156. [Google Scholar] [CrossRef]
  150. Singh, S.; Fumery, M.; Singh, A.G.; Singh, N.; Prokop, L.J.; Dulai, P.S.; Sandborn, W.J.; Curtis, J.R. 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. 2020, 72, 561–576. [Google Scholar] [CrossRef]
  151. Giles, J.T.; Sattar, N.; Gabriel, S.; Ridker, P.M.; Gay, S.; Warne, C.; Musselman, D.; Brockwell, L.; Shittu, E.; Klearman, M.; et al. Cardiovascular Safety of Tocilizumab Versus Etanercept in Rheumatoid Arthritis: A Randomized Controlled Trial. Arthritis Rheumatol. 2020, 72, 31–40. [Google Scholar] [CrossRef]
  152. Castagné, B.; Viprey, M.; Martin, J.; Schott, A.M.; Cucherat, M.; Soubrier, M. Cardiovascular safety of tocilizumab: A systematic review and network meta-analysis. PLoS ONE 2019, 14, e02s20178. [Google Scholar] [CrossRef] [PubMed]
  153. Alsulaim, T.; Alhassan, N.; Khalil, H.; Almutlaq, A. Tocilizumab Effect on Lipid Profile in Correlation to Cardiovascular Events: A Retrospective Cohort Study. Int. J. Rheumatol. 2021, 2021, 5535486. [Google Scholar] [CrossRef] [PubMed]
  154. Tavakolpour, S.; Alesaeidi, S.; Darvishi, M.; GhasemiAdl, M.; Darabi-Monadi, S.; Akhlaghdoust, M.; likaei Behjati, S.; Jafarieh, A. A comprehensive review of rituximab therapy in rheumatoid arthritis patients. Clin. Rheumatol. 2019, 38, 2977–2994. [Google Scholar] [CrossRef] [PubMed]
  155. Cohen, M.D.; Keystone, E. Rituximab for Rheumatoid Arthritis. Rheumatol. Ther. 2015, 2, 99–111. [Google Scholar] [CrossRef]
  156. Provan, S.A.; Berg, I.J.; Hammer, H.B.; Mathiessen, A.; Kvien, T.K.; Semb, A.G. 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 Tociliziumab. PLoS ONE 2015, 10, e0130709. [Google Scholar] [CrossRef] [PubMed]
  157. Hsue, P.Y.; Scherzer, R.; Grunfeld, C.; Imboden, J.; Wu, Y.; Del Puerto, G.; Nitta, E.; Shigenaga, J.; Schnell Heringer, A.; Ganz, P.; et al. Depletion of B-cells with rituximab improves endothelial function and reduces inflammation among individuals with rheumatoid arthritis. J. Am. Heart Assoc. 2014, 3, e001267. [Google Scholar] [CrossRef] [PubMed]
  158. Novikova, D.S.; Popkova, T.V.; Lukina, G.V.; Luchikhina, E.L.; Karateev, D.E.; Volkov, A.V.; Novikov, A.A.; Aleksandrova, E.N.; Nasonov, E.L. The Effects of Rituximab on Lipids, Arterial Stiffness and Carotid Intima-Media Thickness in Rheumatoid Arthritis. J. Korean Med. Sci. 2016, 31, 202–207. [Google Scholar] [CrossRef] [PubMed]
  159. Ward, N.C.; Watts, G.F.; Eckel, R.H. Statin Toxicity. Circ. Res. 2019, 124, 328–350. [Google Scholar] [CrossRef]
  160. Lüftjohann, D.; Sigit, J.I.; Locatelli, S.; von Bergmann, K.; Schmidt, H.H. High-dose simvastin (80 mg/day) decreases plasma concentrations of total homocysteine in patients with hypercholesteromia. Atherosclerosis 2001, 155, 265–266. [Google Scholar] [CrossRef]
  161. Jougasaki, M.; Ichiki, T.; Takenoshita, Y.; Setoguchi, M. Statins suppress interleukin-6-induced monocyte chemo-attractant protein-1 by inhibiting Janus kinase/signal transducers and activators of transcription pathways in human vascular endothelial cells. Br. J. Pharmacol. 2010, 159, 1294–1303. [Google Scholar] [CrossRef]
  162. Tremoulet, A.H. The role of statins in inflammatory vasculitides. Autoimmunity 2015, 48, 177–180. [Google Scholar] [CrossRef] [PubMed]
  163. Koushki, K.; Shahbaz, S.K.; Mashayekhi, K.; Sadeghi, M.; Zayeri, Z.D.; Taba, M.Y.; Banach, M.; Al-Rasadi, K.; Johnston, T.P.; Sahebkar, A. Anti-inflammatory Action of Statins in Cardiovascular Disease: The Role of Inflammasome and Toll-Like Receptor Pathways. Clinic. Rev. Allerg. Immunol. 2021, 60, 175–199. [Google Scholar] [CrossRef] [PubMed]
  164. Venetsanopoulou, A.I.; Pelechas, E.; Voulgari, P.V.; Drosos, A.A. The lipid paradox in rheumatoid arthritis: The dark horse of the augmented cardiovascular risk. Rheumatol. Int. 2020, 40, 1181–1191. [Google Scholar] [CrossRef] [PubMed]
  165. Kitas, G.D.; Nightingale, P.; Armitage, J.; Sattar, N.; Belch, J.J.F.; Symmons, D.P.M.; TRACE RA Consortium. A Multicenter, Randomized, Placebo-Controlled Trial of Atorvastatin for the Primary Prevention of Cardiovascular Events in Patients With Rheumatoid Arthritis. Arthritis Rheumatol. 2019, 71, 1437–1449. [Google Scholar] [CrossRef]
  166. De Vera, M.A.; Choi, H.; Abrahamowicz, M.; Kopec, J.; Lacaille, D. Impact of statin discontinuation on mortality in patients with rheumatoid arthritis: A population-based study. Arthritis Care Res. 2012, 64, 809–816. [Google Scholar] [CrossRef] [PubMed]
  167. Karpouzas, G.A.; Ormseth, S.R.; Hernandez, E.; Budoff, M.J. The impact of statins on coronary atherosclerosis progression and long-term cardiovascular disease risk in rheumatoid arthritis. Rheumatology 2022, 61, 1857–1866. [Google Scholar] [CrossRef] [PubMed]
  168. Grundy, S.M.; Stone, N.J.; Bailey, A.L.; Beam, C.; Birtcher, K.K.; Blumenthal, R.S.; Braun, L.T.; de Ferranti, S.; Faiella-Tommasino, J.; Forman, D.E.; et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: A report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 139, e1082–e1143. [Google Scholar] [CrossRef]
  169. Tinggaard, A.B.; de Thurah, A.; Andersen, I.T.; Riis, A.H.; Therkildsen, J.; Winther, S.; Hauge, E.M.; Bøttcher, M. Rheumatoid Arthritis as a Risk Factor for Coronary Artery Calcification and Obstructive Coronary Artery Disease in Patients with Chest Pain: A Registry Based Cross-Sectional Study. Clin. Epidemiol. 2020, 12, 679–689. [Google Scholar] [CrossRef]
  170. Lindhardsen, J.; Ahlehoff, O.; Gislason, G.H.; Madsen, O.R.; Olesen, J.B.; Torp-Pedersen, C.; Hansen, P.R. The risk of myocardial infarction in rheumatoid arthritis and diabetes mellitus: A Danish nationwide cohort study. Ann. Rheum. Dis. 2011, 70, 929. [Google Scholar] [CrossRef]
  171. Myasoedova, E.; Crowson, C.S.; Kremers, H.M.; Roger, V.L.; Fitz-Gibbon, P.D.; Therneau, T.M.; Gabriel, S.E. Lipid paradox in rheumatoid arthritis: The impact of serum lipid measures and systemic inflammation on the risk of cardiovascular disease. Ann. Rheum. Dis. 2011, 70, 482–487. [Google Scholar] [CrossRef] [PubMed]
  172. Ronda, N.; Favari, E.; Borghi, M.O.; Ingegnoli, F.; Gerosa, M.; Chighizola, C.; Zimetti, F.; Adorni, M.P.; Bernini, F.; Meroni, P.L. Impaired serum cholesterol efflux capacity in rheumatoid arthritis and systemic lupus erythematosus. Ann. Rheum. Dis. 2014, 73, 609–615. [Google Scholar] [CrossRef] [PubMed]
  173. Semb, A.G.; Kvien, T.K.; Aastveit, A.H.; Jungner, I.; Pedersen, T.R.; Walldius, G.; Holme, I. Lipids, myocardial infarction and ischaemic stroke in patients with rheumatoid arthritis in the apolipoprotein-related mortality RISk (AMORIS) study. Ann. Rheum. Dis. 2010, 69, 1996–2001. [Google Scholar] [CrossRef] [PubMed]
  174. Bedeković, D.; Bošnjak, I.; Šarić, S.; Kirner, D.; Novak, S. Role of Inflammatory Cytokines in Rheumatoid Arthritis and Development of Atherosclerosis: A Review. Medicina 2023, 59, 1550. [Google Scholar] [CrossRef]
  175. Hall, F.C.; Dalbeth, N. Disease modification and cardiovascular risk reduction: Two sides of the same coin? Rheumatology 2005, 44, 1473–1482. [Google Scholar] [CrossRef] [PubMed]
  176. Barber, C.E.; Smith, A.; Esdaile, J.M.; Barnabe, C.; Martin, L.O.; Faris, P.; Hazlewood, G.; Noormohamed, R.; Alvarez, N.; Mancini, G.B.; et al. Best Practices for Cardiovascular Disease Prevention in Rheumatoid Arthritis: A Systematic Review of Guideline Recommendations and Quality Indicators. Arthritis Care Res. 2015, 67, 169–179. [Google Scholar] [CrossRef] [PubMed]
  177. Farhat, H.; Irfan, H.; Muthiah, K.; Pallipamu, N.; Taheri, S.; Thiagaraj, S.S.; Shukla, T.S.; Gutlapalli, S.D.; Giva, S.; Penumetcha, S.S. Increased Risk of Cardiovascular Diseases in Rheumatoid Arthritis: A Systematic Review. Cureus 2022, 14, e32308. [Google Scholar] [CrossRef]
  178. Ikdahl, E.; Semb, A.G.; Kerola, A.M. No sign of elevated cardiovascular risk in patients with seropositive rheumatoid arthritis who remain on stable antirheumatic therapy: Results from the nationwide Norwegian Cardio-Rheuma Register. Ann. Rheum. Dis. 2024. advance online publication. [Google Scholar] [CrossRef] [PubMed]
  179. Giles, J.T.; Solomon, D.H.; Liao, K.P.; Rist, P.M.; Fayad, Z.A.; Tawakol, A.; Bathon, J.M. Association of the multi-biomarker disease activity score with arterial 18-fluorodeoxyglucose uptake in rheumatoid arthritis. Rheumatology 2024. advance online publication. [Google Scholar] [CrossRef] [PubMed]
  180. Mandel, A.; Schwarting, A.; Cavagna, L.; Triantafyllias, K. Novel Surrogate Markers of Cardiovascular Risk in the Setting of Autoimmune Rheumatic Diseases: Current Data and Implications for the Future. Front. Med. 2022, 9, 820263. [Google Scholar] [CrossRef]
  181. Jamthikar, A.D.; Gupta, D.; Puvvula, A.; Johri, A.M.; Khanna, N.N.; Saba, L.; Mavrogeni, S.; Laird, J.R.; Pareek, G.; Miner, M.; et al. Cardiovascular risk assessment in patients with rheumatoid arthritis using carotid ultrasound B-mode imaging. Rheumatol. Int. 2020, 40, 1921–1939. [Google Scholar] [CrossRef] [PubMed]
  182. Schmidt, A.F.; Hunt, N.B.; Gordillo-Marañón, M.; Charoen, P.; Drenos, F.; Kivimaki, M.; Lawlor, D.A.; Giambartolomei, C.; Papacosta, O.; Chaturvedi, N.; et al. Cholesteryl ester transfer protein (CETP) as a drug target for cardiovascular disease. Nat. Commun. 2021, 12, 5640. [Google Scholar] [CrossRef] [PubMed]
  183. Biolo, G.; Vinci, P.; Mangogna, A.; Landolfo, M.; Schincariol, P.; Fiotti, N.; Mearelli, F.; Di Girolamo, F.G. Mechanism of action and therapeutic use of bempedoic acid in atherosclerosis and metabolic syndrome. Front. Cardiovasc. Med. 2022, 9, 1028355. [Google Scholar] [CrossRef]
  184. Yuan, W.; Ernst, K.; Kuai, R.; Morin, E.E.; Yu, M.; Sviridov, D.O.; Tang, J.; Mei, L.; Li, D.; Ackermann, R.; et al. Systematic evaluation of the effect of different apolipoprotein A-I mimetic peptides on the performance of synthetic high-density lipoproteins in vitro and in vivo. Nanomedicine 2023, 48, 102646. [Google Scholar] [CrossRef] [PubMed]
  185. Ridker, P.M.; Lei, L.; Ray, K.K.; Ballantyne, C.M.; Bradwin, G.; Rifai, N. Effects of bempedoic acid on CRP, IL-6, fibrinogen and lipoprotein(a) in patients with residual inflammatory risk: A secondary analysis of the CLEAR harmony trial. J. Clin. Lipidol. 2023, 17, 297–302. [Google Scholar] [CrossRef] [PubMed]
  186. Bu, T.; Li, Z.; Hou, Y.; Sun, W.; Zhang, R.; Zhao, L.; Wei, M.; Yang, G.; Yuan, L. Exosome-mediated delivery of inflammation-responsive Il-10 mRNA for controlled atherosclerosis treatment. Theranostics 2021, 11, 9988–10000. [Google Scholar] [CrossRef] [PubMed]
  187. Barozet, M.; Le Tilly, O.; Bejan-Angoulvant, T.; Fesler, P.; Roubille, C. Hypertension and Cardiovascular Outcomes in Inflammatory and Autoimmune Diseases: A Systematic Review and Meta-analysis. Curr. Hypertens. Rep. 2024. advance online publication. [Google Scholar] [CrossRef] [PubMed]
  188. Chung, C.P.; Giles, J.T.; Petri, M.; Szklo, M.; Post, W.; Blumenthal, R.S.; Gelber, A.C.; Ouyang, P.; Jenny, N.S.; Bathon, J.M. Prevalence of traditional modifiable cardiovascular risk factors in patients with rheumatoid arthritis: Comparison with control subjects from the multi-ethnic study of atherosclerosis. Semin. Arthritis Rheum. 2012, 41, 535–544. [Google Scholar] [CrossRef]
  189. Løgstrup, B.B.; Olesen, K.K.W.; Masic, D.; Gyldenkerne, C.; Thrane, P.G.; Ellingsen, T.; Bøtker, H.E.; Maeng, M. Impact of rheumatoid arthritis on major cardiovascular events in patients with and without coronary artery disease. Ann. Rheum. Dis. 2020, 79, 1182–1188. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of common mechanisms in the pathogenesis of cardiovascular disease and rheumatoid arthritis. Increased levels of pro-inflammatory cytokines are found in the joint space and the systemic circulation. The combination of pro-inflammatory mediators and reduced levels of atheroprotective nitric oxide and IL-10 trigger endothelial dysfunction. Dysfunctional endothelium, poorly functioning pro-inflammatory HDL, highly atherogenic citrullinated LDL, and an altered blood lipid profile contribute to atherosclerosis and thrombosis, which favor accelerated development of cardiovascular disease in patients with rheumatoid arthritis. ↑ = increases; ↓ = decreases; → = progresses to.
Figure 1. Schematic representation of common mechanisms in the pathogenesis of cardiovascular disease and rheumatoid arthritis. Increased levels of pro-inflammatory cytokines are found in the joint space and the systemic circulation. The combination of pro-inflammatory mediators and reduced levels of atheroprotective nitric oxide and IL-10 trigger endothelial dysfunction. Dysfunctional endothelium, poorly functioning pro-inflammatory HDL, highly atherogenic citrullinated LDL, and an altered blood lipid profile contribute to atherosclerosis and thrombosis, which favor accelerated development of cardiovascular disease in patients with rheumatoid arthritis. ↑ = increases; ↓ = decreases; → = progresses to.
Biomedicines 12 01608 g001
Figure 2. Illustration of the effects of three cytokines, TNF-α, IL-1β, and IFN-γ on the musculoskeletal system (left side) and cardiovascular system (right side).
Figure 2. Illustration of the effects of three cytokines, TNF-α, IL-1β, and IFN-γ on the musculoskeletal system (left side) and cardiovascular system (right side).
Biomedicines 12 01608 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Godbole, S.; Solomon, J.L.; Johnson, M.; Srivastava, A.; Carsons, S.E.; Belilos, E.; De Leon, J.; Reiss, A.B. Treating Cardiovascular Disease in the Inflammatory Setting of Rheumatoid Arthritis: An Ongoing Challenge. Biomedicines 2024, 12, 1608. https://doi.org/10.3390/biomedicines12071608

AMA Style

Godbole S, Solomon JL, Johnson M, Srivastava A, Carsons SE, Belilos E, De Leon J, Reiss AB. Treating Cardiovascular Disease in the Inflammatory Setting of Rheumatoid Arthritis: An Ongoing Challenge. Biomedicines. 2024; 12(7):1608. https://doi.org/10.3390/biomedicines12071608

Chicago/Turabian Style

Godbole, Saloni, Jenny Lue Solomon, Maryann Johnson, Ankita Srivastava, Steven E. Carsons, Elise Belilos, Joshua De Leon, and Allison B. Reiss. 2024. "Treating Cardiovascular Disease in the Inflammatory Setting of Rheumatoid Arthritis: An Ongoing Challenge" Biomedicines 12, no. 7: 1608. https://doi.org/10.3390/biomedicines12071608

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop