Next Article in Journal
Nuclear Imaging in Infective Endocarditis
Next Article in Special Issue
Empagliflozin—A New Chance for Patients with Chronic Heart Failure
Previous Article in Journal
Radioimmunotherapy Targeting IGF2R on Canine-Patient-Derived Osteosarcoma Tumors in Mice and Radiation Dosimetry in Canine and Pediatric Models
Previous Article in Special Issue
A Novel Statin Compound from Monacolin J Produced Using CYP102A1-Catalyzed Regioselective C-Hydroxylation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Treatment of Cardiovascular Disease in Rheumatoid Arthritis: A Complex Challenge with Increased Atherosclerotic Risk

Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine and NYU Langone Hospital, Long Island, Mineola, NY 11501, USA
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(1), 11; https://0-doi-org.brum.beds.ac.uk/10.3390/ph15010011
Submission received: 12 October 2021 / Revised: 13 December 2021 / Accepted: 17 December 2021 / Published: 22 December 2021

Abstract

:
Rheumatoid arthritis (RA) carries significant risk for atherosclerotic cardiovascular disease (ASCVD). Traditional ASCVD risk factors fail to account for this accelerated atherosclerosis. Shared inflammatory pathways are fundamental in the pathogenesis of both diseases. Considering the impact of RA in increasing cardiovascular morbidity and mortality, the characterization of therapies encompassing both RA and ASCVD management merit high priority. Despite little progress, several drugs discussed here promote remission and or lower rheumatoid disease activity while simultaneously conferring some level of atheroprotection. Methotrexate, a widely used disease-modifying drug used in RA, is associated with significant reduction in cardiovascular adverse events. MTX promotes cholesterol efflux from macrophages, upregulates free radical scavenging and improves endothelial function. Likewise, the sulfonamide drug sulfasalazine positively impacts the lipid profile by increasing HDL-C, and its use in RA has been correlated with reduced risk of myocardial infraction. In the biologic class, inhibitors of TNF-α and IL-6 contribute to improvements in endothelial function and promote anti-atherogenic properties of HDL-C, respectively. The immunosuppressant hydroxychloroquine positively affects insulin sensitization and the lipid profile. While no individual therapy has elicited optimal atheroprotection, further investigation of combination therapies are ongoing.

Graphical Abstract

1. Introduction

Rheumatoid arthritis (RA) is a chronic immune-mediated inflammatory disorder that leads to the breakdown of immune tolerance [1]. It affects women disproportionately and usually presents between the ages of 40–60 [2,3]. Worldwide prevalence of RA is approximately 0.5–1% [4]. This disorder primarily manifests as joint pain, swelling and stiffness. Over time, uncontrolled inflammation leads to disease progression, irreversible destruction of cartilage and bone, and ultimately to reduced range of motion and severe physical disability [5]. Since RA is a systemic disease, extra-articular manifestations can affect multiple organ systems. Lung involvement is the most common, but vasculitis and rheumatoid nodules on the skin are also observed [6,7,8].
Dyslipidemia and atherosclerosis are major comorbidities associated with RA [9]. It is well established that RA confers increased risk for atherosclerotic cardiovascular disease (ASCVD), a chronic inflammatory and lipid-depository disease [10,11]. Traditional ASCVD risk factors fail to fully account for this increase, making it difficult to identify and treat these patients at the preclinical stage [12,13,14]. In comparison to the general population, RA patients have a CVD risk approximately 50% higher and a 1.6 times higher rate of acute myocardial infarction and ischemic stroke [15,16]. The risk of myocardial infarction is on a par with that of persons with diabetes mellitus [17].
ASCVD adds to the substantial burden on RA patients, contributing to overall morbidity, decline in physical function, and diminished quality of life [18,19].

2. Methods

An extensive literature review was performed using PubMed and Google with regard to the drugs used in rheumatoid arthritis and their cardiovascular effects. The search terms included “rheumatoid arthritis“, “atherosclerosis”, “rheumatoid arthritis AND atherosclerosis AND mechanism”, “cardiovascular disease”, “treatment”, “medications”, “DMARDs”, “methotrexate AND cardiovascular” and “TNF-α AND cardiovascular.” Studies available in English from 1989 onward were included. This yielded approximately 600 manuscripts from which we narrowed the scope by looking at human studies (observational, randomized, prospective and retrospective) as well as meta-analysis papers and reviews of mechanism of action of individual drugs. We excluded cancer, diabetes and autoimmune rheumatic disorders other than rheumatoid arthritis. The final number of papers reviewed after applying these selection criteria was 212.

3. Background

3.1. Causes of Increased ASCVD Risk in RA

Lipid disturbances are considered one of the primary factors underlying the increased risk for ASCVD in the RA population (Figure 1). Similar to the general population, circulating levels of high density lipoprotein (HDL) are inversely proportional to incidence of cardiovascular events, but beyond quantity, HDL functionality also affects ASCVD risk in RA [20,21,22,23]. Under normal circumstances, HDL is highly atheroprotective, acting as an antioxidant while facilitating cholesterol efflux [24]. However, these functions are impaired in the RA setting [25]. Paraoxonase (PON)-1 activity, the component responsible for HDL antioxidant properties, is decreased in RA correlating with an increased carotid plaque burden [26,27,28]. HDL in patients with high disease activity has also been found to have reduced cholesterol efflux capacity [29,30,31]. This reduced efflux capacity may result from impaired binding of ATP to the ATP binding cassette transporter (ABC)G1 [32].
In addition, RA plasma can foster a combination of attenuated cholesterol outflow and increased scavenger receptor-mediated oxidized low density lipoprotein (LDL) uptake by monocytes and macrophages via the downregulation of a set of cholesterol efflux proteins (ABCA1, ABCG1, 27-hydroxylase) coupled with the upregulation of scavenger receptors (CD36, LOX1 and CXCL16). This is a highly favorable setting for macrophage cholesterol overload and foam cell formation [33,34,35]. Changes in LDL composition resulting in greater levels of glycated end-products, oxidation, enhanced phagocytosis, and fatty acid accumulation have also been observed [36,37,38]. Citrullinated and homocitrullinated forms of LDL have atherogenic properties and may also be present in RA plasma [39]. Further, Lipoprotein a (Lp(a)), an atherogenic lipoprotein complex containing apolipoprotein (apo)A and apo B, is found at high levels in RA and is predictive of increased ASCVD risk [40]. A deeper understanding of these aberrations in lipoprotein function in rheumatoid arthritis patients may provide insight into methods for alleviating the cardiovascular burden.
Damage to endothelium occurs early in the atherosclerotic process and leads to endothelial dysfunction and activation, upregulation of adhesion molecules and interruption of the monolayer. The pro-inflammatory cytokine tumor necrosis factor (TNF)-α, produced in excess in RA, causes the release of adhesion molecules and is one factor that drives endothelial dysfunction [41]. Damaged endothelium allows oxidized LDL (oxLDL) and inflammatory cells to accumulate in the arterial intima where monocyte-to-macrophages differentiation, oxLDL internalization, and foam cell formation occur [42,43]. A cross-sectional study of 50 newly diagnosed early RA patients by Salem et al. [42] found that serum levels of vascular cell adhesion molecule (VCAM)-1 were elevated and correlated with disease activity and oxidative stress. VCAM-1 levels, considered an indicator of endothelial dysfunction, were significantly lower in treated patients compared to newly diagnosed patients. Higher serum levels of a soluble form of VCAM-1 are associated with accelerated progression of subclinical atherosclerosis [44].
In RA, damage to endothelium can also occur due impaired functioning of endothelial nitric oxide synthase which reduces availability of nitric oxide, a potent anti-inflammatory and vasodilator [45,46]. RA patients have high circulating concentrations of the methylated arginine asymmetric dimethylarginine (ADMA), a potent endogenous inhibitor of endothelial nitric oxide synthase [47].

3.2. RA and CVD: Shared Pathways

Development of CVD is a life-threatening systemic consequence of RA [48,49,50]. While chronic inflammation, a hallmark of RA, is likely responsible for much of the elevated CVD risk, traditional CVD risk factors such as obesity, diabetes, smoking and hypertension are over-represented in RA as well [51,52,53,54,55,56]. As the strong link between the two disorders worsens prognosis, it is essential to explore their connection in order to minimize health burden in RA. Doing so reveals shared inflammatory pathways fundamental to both disease processes with many pro-inflammatory cytokines implicated in RA also playing a central role in ASCVD [20,57,58,59]. Most prominent of the cytokines involved in both RA and ASCVD are TNF-α, interleukin (IL)-6 and IL-1β [60,61]. Elevated concentrations of each of these three cytokines have been correlated to the presence of coronary artery calcification, a severe complication of the atherosclerotic process [62,63,64,65]. A critical inflammatory pathway involved in the formation of atherosclerotic plaque is the oxLDL-stimulated production of a broad spectrum of cytokines by macrophages, with TNF-α and IL-1β playing major roles [66].
TNF-α is a key pro-inflammatory mediator in the pathogenesis of RA that promotes the development of atherosclerosis [67]. Elevated serum TNF-α is associated with dyslipidemia in RA patients. The cytokine not only impairs the anti-oxidant effects of HDL, but also promotes the formation of oxLDL within the arterial intima [68,69,70].
TNF-α has been linked to vascular injury in both acute and chronic inflammatory states. It promotes endothelial expression of chemokines and adhesion molecules that bring about leukocyte adherence and migration into the intimal layer of the arterial wall [71,72]. TNF-α also causes endothelial apoptosis leading to breakdown of endothelial barrier integrity and a pro-thrombotic environment [73,74]. Nitric oxide and thrombomodulin production are decreased in the setting of elevated TNF-α, further encouraging thrombotic events [71,75]. The inflammatory milieu of RA promotes formation of atherosclerotic plaques that are unstable and vulnerable to rupture [76,77].
IL-1β is a secretory protein produced by macrophages and endothelial cells. It induces endothelial cells to express surface adhesion molecules that foster recruitment of macrophages into the arterial intima [78].
Numerous additional inflammatory cytokines foster a pro-atherogenic, pro-thrombotic state, including IL-17, IL-18, and IL-33 [20,79,80,81]. Many of these cytokines act in synergy with each other and are involved in multiple signaling events that occur simultaneously [82]. Of the myriad mediators, nuclear factor (NF)-κB, stands out as a “master switch” that turns on multiple pro-atherosclerotic genes, including VCAM-1, intercellular adhesion molecule-1, and macrophage-colony-stimulating factor and induces vascular inflammation [83,84]. NF-κB stimulates production of C-reactive protein, IL-6, and TNF-α [85,86].
Serum amyloid A (SAA), an acute-phase protein found at elevated levels in RA [87]. SAA is linked to atherosclerosis by its ability to bind to HDL and attenuate its anti-oxidative properties [88,89].
In RA, matrix metalloproteinases (MMPs), which degrade collagen as well as non-collagen components of the joint extracellular matrix, are produced by invading synovial pannus. Serum concentrations of MMP-1 and MMP-3 correlate with RA disease activity and predict progression of joint destruction [90,91,92]. MMPs are also associated with plaque instability and rupture [77,93,94] (Figure 2). Atherosclerotic plaque formation in RA may be accelerated due not only to MMPs, but also due to inflammatory cytokines which stimulate vascular smooth muscle proliferation and migration, leading to arterial intimal thickening, while also promoting plaque destabilizing microcalcification [95,96].
The CD40-CD40L axis, which mediates pro-inflammatory and pro-fibrotic pathways, is another shared process in both RA and ASCVD [97,98,99]. CD40, a member of the TNF receptor superfamily, is linked to cardiovascular events including myocardial infarction, plaque rupture and thrombosis [100,101,102].
Oxidative stress, the imbalance of reactive oxygen species generation and antioxidant defense, contributes to the pathogenesis of both RA and ASCVD [103,104,105]. Plasma antioxidant capacity is reduced in RA and oxidative stress increased [106,107]. Reactive oxygen species are chemically reactive free radicals that mediate tissue damage within joints and other body systems by inciting lipid peroxidation, protein oxidation and DNA damage [108,109]. Oxidative stress drives the atherosclerotic process and RA creates a source of excessive reactive oxygen species, thus contributing to a pro-atherosclerotic milieu [110]. Reactive oxygen species impair functionality of vascular endothelium and participate in oxidation of LDL, two critical processes involved in ASCVD [111]. In a cell culture model using ECV304, a spontaneously transformed cell line from human umbilical vein endothelial cells, plasma from RA patients was shown to reduce nitric oxide synthesis and increase reactive oxygen species production [112].

4. Treatment of RA—Does It Address CVD Risk?

Considering the significant impact on RA morbidity and mortality, the development of treatments that incorporate ASCVD management would be expected to merit high priority, but surprisingly little progress has been achieved. The current goal of RA therapy is remission or low disease activity, characterized as a state with minimal joint pain and swelling, prevention of joint deformities and radiographic damage, maintenance of quality of life and control of extra-articular manifestations [113,114,115] obtainable in approximately 10–50% of patients
Given the substantial strain this chronic disease carries for both the individual and society, extensive research into its treatment has been conducted. DMARDs, which are slow-acting agents beneficial in improving the symptoms and radiographic progression, remain at the forefront of therapy [116,117,118]. Methotrexate has dominated the DMARDs as the medication of choice; however, alternatives include leflunomide, sulfasalazine and hydroxychloroquine. Numerous studies have led to the understanding that combination therapies are more effective than mono-therapies. Considering the significant role cytokines and other molecules play in the pathology of the disease, relatively newer treatments utilizing these biological targets have also emerged in recent times [116,117]. These drugs accomplish their goal via four main mechanism: blockade of TNF-α, inhibiting IL-6, blocking T-cell co-stimulation as well as depletion of B-cell. While non-steroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids can relieve pain and inflammation, they do not prevent progressive joint damage and their long-term use may be harmful [118,119]. Ancillary therapies including physical activity, exercise, muscle strengthening and diets supporting a healthy microbiome are also implemented. In severe cases, when the damage is too advanced total joint replacement, most commonly of the hip or knee, may be required.
The NIH funded clinical trial, Treatments Against RA and Effect on FDG-PET/CT (fluorodeoxyglucose-positron emission tomography with computed tomography) (TARGET, NCT02374021) [120], aims to determine whether anti-inflammatory DMARDs used to treat RA also reduced the risk of CVD. This will be accomplished by comparing two separate therapies, administration of methotrexate (MTX) combined with a TNF-α inhibitor or triple therapy with MTX, sulfasalazine and hydroxychloroquine. This paper explores the effects of cholesterol-lowering therapy and of the individual drugs used in TARGET on development of ASCVD as we move toward discovering which, if any, combinations confer optimal atheroprotection.

5. ASCVD Management in RA

5.1. Cardiovascular-Specific Use of Statins

Currently, the front-line pre-emptive method to address atherosclerosis is the use of statins regardless of LDL levels [121,122,123]. The beneficial effects of statins include not only lipid-lowering, but also immunomodulation and quelling of inflammation [124]. Statins subdue vascular inflammation by reducing expression of IL-6, high-sensitivity C-reactive protein, and homocysteine [125,126]. Their cholesterol-independent pleiotropic effects extend to improving endothelial function and lowering oxidative stress [127]. They inhibit vascular smooth muscle proliferation and platelet aggregation [128]. They modulate the functions of adhesion molecules and reduce monocytes adhesion to endothelial cells. Statins have not been found to change the course of RA itself and thus are given specifically for cardiovascular risk reduction [129].

5.2. Drugs That Treat RA and Their Impact on ASCVD

Therapies directed against RA that also mitigate some of the associated CVD risk would be particularly valuable and there are several to be discussed here and summarized in Table 1. Disease-modifying anti-rheumatic drugs (DMARDs), which decrease the inflammation common to RA and ASCVD have a variable track record in atheroprotection. Studies such as TARGET are intended to delve into whether a reduction in inflammation via DMARDs does, in fact, reduce the risk of CVD [120].

5.2.1. Methotrexate

One such DMARD investigated by the TARGET study is MTX, a folic acid analog that inhibits dihydrofolate reductase and reduces the levels of tetrahydrofolate in cells. MTX also inhibits purines and pyrimidines necessary for DNA and RNA synthesis [130]. Another mechanism by which it reduces inflammation is by lowering levels of NF-κB activity [131]. MTX is widely used in the treatment of various inflammatory and autoimmune disorders and it is the initial therapy of choice for RA [132,133]. MTX possesses cardioprotective properties that may reduce CVD morbidity and mortality in patients with RA [134,135,136,137].
Low dose MTX, the gold standard RA treatment, broadly inhibits several immune pathways involved in purine and pyrimidine synthesis, transmethylation reactions, translocation of NF-κB to the nucleus, signaling via the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway and nitric oxide production [138]. The benefits of MTX in reducing joint symptoms in RA can be attributed to these anti-inflammatory mechanisms described above. The protective effects of MTX against CVD can occur indirectly via anti-inflammatory actions and also directly via antiatherogenic effects, such as enhancing cholesterol transport out of monocytes and macrophages and preventing the differentiation and activation of foams cells [139,140,141]. Low-dose MTX also promotes release of the anti-inflammatory endogenous nucleoside adenosine, which acts on its receptors to increase production of ABCA1 and ABCG1, proteins involved in cholesterol efflux from macrophages [142]. MTX also upregulates free radical scavenging and improves endothelial function by reducing the expression of adhesion molecules, consequently inhibiting the recruitment of proinflammatory T cells and monocytes [143]. Of note, by depleting tetrahydrofolate, which is needed for conversion of homocysteine to methionine, MTX may have negative effects on cardiovascular health by raising plasma homocysteine levels, thus contributing to oxidative stress [144]. However, folate supplementation can mitigate this rise in homocysteine, promoting an overall anti-atherogenic effect [143].
In a multicenter prospective cohort study of 2044 US veterans with RA, Johnson et al. [145] found that the use of MTX was associated with a significant reduction in CVD related adverse events. In this study, CVD risk reduction by MTX was independent of age, BMI, traditional CVD risk factors, and RA disease activity. The cardioprotective properties of MTX were not solely due to amelioration of RA disease activity, suggesting that MTX may have direct cardiovascular advantages for patients with RA.

5.2.2. Sulfasalazine

Sulfasalazine, an older anti-rheumatic drug also being investigated in the TARGET study, is a prodrug synthesized from the fusion of the antibiotic sulfapyridine and an NSAID, 5-aminosalicylic acid via an azo bond. This oral sulfonamide drug possesses antithrombotic properties. It inhibits platelet thromboxane synthetase and therefore thromboxane production [146]. Sulfasalazine can prevent arachidonic acid-mediated platelet aggregation, an inhibition equivalent to that produced by aspirin given for protection against CVD [147]. Sulfasalazine decreases proliferation of lymphocytes, and adhesion of monocytes and leukocytes [148]. Sulfasalazine reduces production of inflammatory cytokines, likely through inhibition of NF-κB. As noted previously, NF-κB is a major regulator of genes that promote inflammation and adhesion in cell types involved in atherogenesis [83,84]. Sulfasalazine acts on the NF-κB pathway by preventing the phosphorylation and successive degradation of IκB, the inhibitory subunit of NF-κB [149,150]. Inhibition of NF-κB is considered a viable approach to protection from atherosclerosis development [151].
Sulfasalazine positively impacts the lipid profile, increasing HDL-C and reducing the atherogenic ratio [152,153,154]. Observational data from a QUEST-RA longitudinal cohort study also indicated that longer exposure to sulfasalazine correlated with a reduced risk of CVD events and myocardial infarction [155].

5.2.3. TNF-α Inhibitors

Based upon the pro-atherogenic actions of TNF-α, the reduction in systemic inflammation with the use of biologics such as TNF inhibitors (TNFi) could be expected to contribute to improvements in endothelial function and confer atheroprotection [156,157,158,159]. Commonly used TNF-α inhibitors include etanercept, infliximab and adalimumab [160]. Etanercept is a dimeric fusion protein consisting of the extracellular portion of the human TNF receptor linked to the Fc portion of human immunoglobulin 1 (IgG1). It targets free TNF-α and TNF-β. Infliximab is a chimeric monoclonal antibody with a murine variable region that specifically binds to human TNF-α. Adalimumab is a fully human monoclonal anti-TNF-α antibody. All exert their effect by neutralizing circulating TNF-α.
A 5-year study that compared RA patients who were biologic-naïve receiving only synthetic DMARDs and those taking a TNFi (either etanercept, infliximab or adalimumab) found that the risk of myocardial infarction decreased by 39% in the latter group [161]. Vegh et al. showed that one year of TNFi treatment significantly improved pulse–wave velocity, a marker of arterial stiffness and resulted in no significant increase in common carotid intima-media thickness in a mixed cohort of RA and ankylosing spondylitis patients [162]. Serum levels of biomarkers for atherosclerosis may be reduced by TNFi treatment in RA patients. Pusztai et al. followed 53 RA patients on TNFi for one year and found that this treatment significantly decreased the level of circulating complexes of oxLDL bound to β2-glycoprotein [163]. These complexes can be taken up into macrophages to form foam cells and are associated with the development of acute coronary syndromes [164].
Additionally, higher levels of TNF-α may amplify standard cardiac risk factors such as diabetes and dyslipidemia in patients with RA [165,166]. Insulin resistance has been associated with elevated levels of TNF-α in RA patients [167]. TNF-α promotes insulin resistance by directly suppressing activities of insulin-induced insulin receptor substrate-1 and peroxisome proliferator-activated receptor gamma (PPAR-γ) [165,166]. TNFi increases insulin sensitivity in RA patients compared to patients not receiving biologics [168].
The effects of TNFi on lipid profile are unclear, with some studies showing an increase in LDL with treatment [169]. However, other studies show no effect, and a recent study from Korea found no detrimental impact over a four year period [170,171,172].

5.2.4. IL-6 Inhibitors

The introduction of biologic drugs has considerably improved the outcome for patients with RA, but at the same time certain drugs, specifically the IL-6 inhibitors (i.e., tocilizumab), have been a source of controversy in terms of their effects on cardiovascular health, especially with regard to their effects on the lipid profile [173]. A representative prospective study revealed that in patients undergoing tocilizumab therapy, serum concentrations of total cholesterol, LDL, HDL and triglycerides increased during the first 24 weeks of treatment [174]. The results of several randomized clinical trials also showed increased LDL levels in tocilizumab-treated RA patients, of a magnitude greater than the increase seen in RA patients treated with conventional DMARDs [175,176]. Nevertheless, recent studies have shown encouraging data regarding major adverse cardiovascular events with tocilizumab compared to other biologic DMARDs. Two studies conducted underscored a significant decrease in cardiovascular events with this drug [177,178]. Zhang et al., revealed that tocilizumab reduced the risk of myocardial infarction significantly more than abatacept, another biologic DMARD [177]. Kim et al. indicated better cardiovascular outcomes with tocilizumab when compared to TNFi [178]. Furthermore, tocilizumab decreases inflammatory proteins such as serum amyloid A and may restore the anti-atherogenic function of HDL [179]. Effects on HDL are observed directly in recent studies that found tocilizumab to increase HDL cholesterol efflux capacity in RA [180,181]. Cholesterol efflux capacity is considered a strong negative ASCVD risk determinant [182]. The atherogenic lipoprotein Lp(a) was also reduced by tocilizumab [183,184].

5.2.5. JAK Kinase Inhibitors

JAK inhibitors, small molecules that target the JAK-STAT signaling pathway, are targeted synthetic DMARDs for the treatment of RA and other immune-mediated inflammatory diseases [185,186]. Unlike the protein-based biologic DMARDs, the JAK inhibitors do not cause formation of anti-drug antibodies and can be taken orally [187]. Unfortunately, the JAK inhibitors are associated with risk of venous thromboembolism and pulmonary embolism [188]. The effect of these drugs on cardiovascular risk has not yet been determined as data is inconclusive. Charles-Schoeman et al. pooled data from multiple studies on the first generation JAK inhibitor tofacitinib showed low incidence of cardiac adverse events, comparable to that found with placebo [189]. A further study by Charles-Shoeman with post hoc analysis, after 24 weeks of tofacitinib showed increased HDL levels which was associated with lower cardiovascular event risk [190]. A long-term study over 9.5 years found incidence rates for major adverse cardiovascular event with tofacitinib of 0.4 with events per 100 patient-years, which is comparable to those seen with biologic agents [191]. Upadacitinib, a newer and more selective JAK1 inhibitor, has shown comparable effects on cardiovascular risk to placebo and other JAK inhibitors. It raised both HDL and LDL while leaving the HDL-to-LDL ratio constant [192]. The cardiac event risk from these drugs appears low, but more studies are in progress [193,194].

5.2.6. HCQ: A Potential Double Agent

Despite its original intended purpose as an anti-parasitic agent, the therapeutic impact of HCQ extends beyond malaria to multiple autoimmune diseases in which it acts as an immunomodulator and its potential benefits continue to emerge [118,195,196,197]. HCQ can improve the survival rates of patients with various autoimmune diseases including RA and systemic lupus erythematosus (SLE) via its immunosuppressive and anti-inflammatory activity [198,199,200].
HCQ is a weak basic 4-aminoquinoline compound that differs from chloroquine by the placement of a single hydroxyl group [197,198]. This subtle alteration decreases toxicity while conserving efficacy, rendering it safer and therefore preferable to chloroquine [196]. The drug is administered orally, as a sulfate tablet, and appears to be well-tolerated, with efficient absorption, a large volume of distribution and bioavailability, as well as a prolonged half-life of about 40–50 days [201]. Approximately 50% of HCQ remains plasma bound and is metabolized into three metabolites, desethyl-chloroquine, desethyl-hydroxychloroquine, and bis-desethyl-hydroxychloroquine, in the liver.
Although the exact mechanisms of action of this compound remain uncertain, it is postulated that immunosuppression results from its ability to block the stimulation of toll-like receptors, suppress T-cell proliferation, inhibit autophagy and reduce macrophage-mediated cytokine production and calcium signaling in B and T cells [202,203]. Its atheroprotective attributes may be due to the reduction in circulating cytokines such as Il-1, IL-6 and TNF-α [204].
In addition to its role as an anti-rheumatic drug, HCQ has anti-diabetic and cardioprotective capabilities. It improves glycemic control, positively impacts insulin sensitivity and metabolism, favorably influences the lipid profile and has anti-thrombotic and anticoagulant properties [196,198,205,206,207,208]. In RA patients, HCQ improves microvascular endothelial function in RA [209].
Additionally, HCQ is a favorable candidate for not only RA treatment but also the management of CVD in these patients as there are few contraindications and adverse effects. The most common side effects predominantly relate to the gastrointestinal issues, such as nausea, vomiting and diarrhea [210]. Although rare, the possibility of developing retinal, neuromuscular, cardiac, and hematological impairments, including retinopathy, does exist [211]. Despite its limited efficacy in the treatment of RA when used alone, HCQ’s utilization in combination therapies may have a significant impact on the cardiovascular risks associated with this rheumatic disease, thereby reducing the burden for both the individual and society [212].

6. Conclusions

One of the most life-threatening complications associated with RA is the development of CVD. RA patients have a 30–50% increased risk of cardiac events compared to the general population [15,16]. CVD may go undiagnosed and untreated for prolonged periods in RA patients as it is often subclinical and asymptomatic. Factors responsible for elevated CVD rates in RA include traditional CVD risk factors and an environment induced by RA of persistent inflammation and oxidative stress with constant exposure to high levels of inflammatory cytokines. The excess mortality resulting from ASCVD makes it imperative that consideration be given to the cardiovascular system when developing an RA treatment plan. Despite the clear need to include heart health in clinical management of RA, there has been little progress in this respect and the ongoing primary goal of RA therapies is to achieve disease remission (focused on joints) or at least low disease activity. Cardiovascular-specific management of ASCVD in RA is often limited to use of statins to improve lipid profile and anti-hypertensive medications in patients with elevated blood pressure. Several anti-rheumatic drugs possess established or likely cardiovascular benefits. These include methotrexate, sulfasalazine, TNF-α inhibitors, Il-6 inhibitors and hydroxychloroquine. The ongoing TARGET study holds promise because it is designed to explore which combination of DMARDs will best reduce the risk of CVD in RA. More such prospective trials are needed, and it is hoped that new therapies will bring life-prolonging outcomes for the RA population.

Author Contributions

A.B.R. and S.A. conceptualized the topic of the review. The manuscript was drafted by B.J., A.B.R., S.A., and J.D.L. and critically revised by J.D.L. and S.E.C. Figures were designed by B.J. and S.A. All authors have read and agreed to the published version of the manuscript.

Funding

Not applicable—no funding was needed for preparation of this manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

In memory of Elizabeth Daniell and Lt. Col. E.L. Daniell. We thank Robert Buescher for his dedication to medical research. We thank Lynn Drucker.

Conflicts of Interest

All authors declare that they have no conflict of interest or competing interests.

References

  1. Gibofsky, A. Epidemiology, pathophysiology, and diagnosis of rheumatoid arthritis: A synopsis. Am. J. Manag. Care 2014, 20 (Suppl. 7), S128–S135. [Google Scholar]
  2. Safiri, S.; Kolahi, A.A.; Hoy, D.; Smith, E.; Bettampadi, D.; Mansournia, M.A.; Almasi-Hashiani, A.; Ashrafi-Asgarabad, A.; Moradi-Lakeh, M.; Qorbani, M.; et al. Global, regional and national burden of rheumatoid arthritis 1990–2017: A systematic analysis of the global burden of disease study 2017. Ann. Rheum. Dis. 2019, 78, 1463–1471. [Google Scholar] [CrossRef]
  3. Tedeschi, S.K.; Bermas, B.; Costenbader, K.H. Sexual disparities in the incidence and course of SLE and RA. Clin. Immunol. 2013, 149, 211–218. [Google Scholar] [CrossRef]
  4. Hunter, T.M.; Boytsov, N.N.; Zhang, X.; Schroeder, K.; Michaud, K.; Araujo, A.B. Prevalence of rheumatoid arthritis in the United States adult population in healthcare claims databases, 2004–2014. Rheumatol. Int. 2017, 37, 1551–1557. [Google Scholar] [CrossRef]
  5. McInnes, I.B.; Schett, G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 2017, 389, 2328–2337. [Google Scholar] [CrossRef] [Green Version]
  6. García-Patos, V. Rheumatoid nodule. Semin. Cutan. Med. Surg. 2007, 26, 100–107. [Google Scholar] [CrossRef]
  7. Yunt, Z.X.; Solomon, J.J. Lung Disease in Rheumatoid Arthritis. Rheum. Dis. Clin. N. Am. 2015, 41, 225–236. [Google Scholar] [CrossRef] [Green Version]
  8. Prete, M.; Racanelli, V.; Digiglio, L.; Vacca, A.; Dammacco, F.; Perosa, F. Extra-articular manifestations of rheumatoid arthritis: An update. Autoimmun. Rev. 2011, 11, 123–131. [Google Scholar] [CrossRef]
  9. Toms, T.E.; Symmons, D.P.; Kitas, G.D. Dyslipidaemia in rheumatoid arthritis: The role of inflammation, drugs, lifestyle and genetic factors. Curr. Vasc. Pharmacol. 2010, 8, 301–326. [Google Scholar] [CrossRef]
  10. Holmqvist, M.E.; Wedrén, S.; Jacobsson, L.T.; Klareskog, L.; Nyberg, F.; Rantapää-Dahlqvist, S.; Alfredsson, L.; Askling, J. Rapid increase in myocardial infarction risk following diagnosis of rheumatoid arthritis amongst patients diagnosed between 1995 and 2006. J. Intern. Med. 2010, 268, 578–585. [Google Scholar] [CrossRef] [Green Version]
  11. Koren Krajnc, M.; Hojs, R.; Holc, I.; Knez, Ž.; Pahor, A. Accelerated atherosclerosis in premenopausal women with rheumatoid arthritis—15-year follow-up. Bosn. J. Basic Med. Sci. 2021, 21, 477–483. [Google Scholar] [CrossRef]
  12. Skeoch, S.; Bruce, I.N. Atherosclerosis in rheumatoid arthritis: Is it all about inflammation? Nat. Rev. Rheumatol. 2015, 11, 390–400. [Google Scholar] [CrossRef]
  13. del Rincón, I.D.; Williams, K.; Stern, M.P.; Freeman, G.L.; Escalante, A. High incidence of cardiovascular events in a rheumatoid arthritis cohort not explained by traditional cardiac risk factors. Arthritis Rheum. 2001, 44, 2737–2745. [Google Scholar] [CrossRef]
  14. Björsenius, I.; Rantapää-Dahlqvist, S.; Berglin, E.; Södergren, A. Extent of atherosclerosis after 11-year prospective follow-up in patients with early rheumatoid arthritis was affected by disease severity at diagnosis. Scand. J. Rheumatol. 2020, 49, 443–451. [Google Scholar] [CrossRef]
  15. Aviña-Zubieta, J.A.; Choi, H.K.; Sadatsafavi, M.; Etminan, M.; Esdaile, J.M.; Lacaille, D. Risk of cardiovascular mortality in patients with rheumatoid arthritis: A meta-analysis of observational studies. Arthritis Rheum. 2008, 59, 1690–1697. [Google Scholar] [CrossRef]
  16. 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]
  17. 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, 929–934. [Google Scholar] [CrossRef] [Green Version]
  18. Cross, M.; Smith, E.; Hoy, D.; Carmona, L.; Wolfe, F.; Vos, T.; Williams, B.; Gabriel, S.; Lassere, M.; Johns, N.; et al. The global burden of rheumatoid arthritis: Estimates from the global burden of disease 2010 study. Ann. Rheum. Dis. 2014, 73, 1316–1322. [Google Scholar] [CrossRef]
  19. Ghosh-Swaby, O.R.; Kuriya, B. Awareness and perceived risk of cardiovascular disease among individuals living with rheumatoid arthritis is low: Results of a systematic literature review. Arthritis Res. Ther. 2019, 21, 33. [Google Scholar] [CrossRef] [Green Version]
  20. England, B.R.; Thiele, G.M.; Anderson, D.R.; Mikuls, T.R. Increased cardiovascular risk in rheumatoid arthritis: Mechanisms and implications. BMJ 2018, 361, k1036. [Google Scholar] [CrossRef]
  21. Navarro-Millán, I.; Yang, S.; DuVall, S.L.; Chen, L.; Baddley, J.; Cannon, G.W.; Delzell, E.S.; Zhang, J.; Safford, M.M.; Patkar, N.M.; et al. Association of hyperlipidaemia, inflammation and serological status and coronary heart disease among patients with rheumatoid arthritis: Data from the National Veterans Health Administration. Ann. Rheum. Dis. 2016, 75, 341–347. [Google Scholar] [CrossRef]
  22. Zhang, J.; Chen, L.; Delzell, E.; Muntner, P.; Hillegass, W.B.; Safford, M.M.; Millan, I.Y.; Crowson, C.S.; Curtis, J.R. The association between inflammatory markers, serum lipids and the risk of cardiovascular events in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2014, 73, 1301–1308. [Google Scholar] [CrossRef]
  23. Liao, K.P.; Liu, J.; Lu, B.; Solomon, D.H.; Kim, S.C. Association between lipid levels and major adverse cardiovascular events in rheumatoid arthritis compared to non-rheumatoid arthritis patients. Arthritis Rheumatol. 2015, 67, 2004–2010. [Google Scholar] [CrossRef] [Green Version]
  24. Brites, F.; Martin, M.; Guillas, I.; Kontush, A. Antioxidative activity of high-density lipoprotein (HDL): Mechanistic insights into potential clinical benefit. BBA Clin. 2017, 8, 66–77. [Google Scholar] [CrossRef]
  25. 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]
  26. 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]
  27. Charles-Schoeman, C.; Watanabe, J.; Lee, Y.Y.; Furst, D.E.; Amjadi, S.; Elashoff, D.; Park, G.; McMahon, M.; Paulus, H.E.; Fogelman, A.M.; et al. Abnormal function of high-density lipoprotein is associated with poor disease control and an altered protein cargo in rheumatoid arthritis. Arthritis Rheum. 2009, 60, 2870–2879. [Google Scholar] [CrossRef] [Green Version]
  28. Charles-Schoeman, C.; Lee, Y.Y.; Shahbazian, A.; Gorn, A.H.; Fitzgerald, J.; Ranganath, V.K.; Taylor, M.; Ragavendra, N.; McMahon, M.; Elashoff, D.; et al. Association of paraoxonase 1 gene polymorphism and enzyme activity with carotid plaque in rheumatoid arthritis. Arthritis Rheum. 2013, 65, 2765–2772. [Google Scholar] [CrossRef] [Green Version]
  29. Xie, B.; He, J.; Liu, Y.; Liu, T.; Liu, C. A meta-analysis of HDL cholesterol efflux capacity and concentration in patients with rheumatoid arthritis. Lipids Health Dis. 2021, 20, 18. [Google Scholar] [CrossRef]
  30. 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]
  31. Liao, K.P.; Playford, M.P.; Frits, M.; Coblyn, J.S.; Iannaccone, C.; Weinblatt, M.E.; Shadick, N.S.; Mehta, N.N. The association between reduction in inflammation and changes in lipoprotein levels and HDL cholesterol efflux capacity in rheumatoid arthritis. J. Am. Heart. Assoc. 2015, 4, e001588. [Google Scholar] [CrossRef] [Green Version]
  32. 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]
  33. 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] [Green Version]
  34. Turgunova, L.G.; Shalygina, A.A.; Zalkalns, J.P.; Klyuyev, D.A.; Akhmaltdinova, L.L.; Dosmagambetova, R.S. Assessment of Adipokines, CXCL16 Chemokine Levels in Patients with Rheumatoid Arthritis Combined with Metabolic Syndrome. Clin. Med. Insights Arthritis Musculoskelet. Disord. 2021, 14, 1179544120985860. [Google Scholar] [CrossRef]
  35. Dragoljevic, D.; Kraakman, M.J.; Nagareddy, P.R.; Ngo, D.; Shihata, W.; Kammoun, H.L.; Whillas, A.; Lee, M.K.S.; Al-Sharea, A.; Pernes, G.; et al. Defective cholesterol metabolism in haematopoietic stem cells promotes monocyte-driven atherosclerosis in rheumatoid arthritis. Eur. Heart J. 2018, 39, 2158–2167. [Google Scholar] [CrossRef] [Green Version]
  36. Li, Y.; Khan, M.S.; Akhter, F.; Husain, F.M.; Ahmad, S.; Chen, L. The non-enzymatic glycation of LDL proteins results in biochemical alterations—A correlation study of Apo B100-AGE with obesity and rheumatoid arthritis. Int. J. Biol. Macromol. 2019, 122, 195–200. [Google Scholar] [CrossRef]
  37. Kim, J.Y.; Lee, E.Y.; Park, J.K.; Song, Y.W.; Kim, J.R.; Cho, K.H. Patients with Rheumatoid Arthritis Show Altered Lipoprotein Profiles with Dysfunctional High-Density Lipoproteins that Can Exacerbate Inflammatory and Atherogenic Process. PLoS ONE 2016, 11, e0164564. [Google Scholar] [CrossRef]
  38. 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] [Green Version]
  39. 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]
  40. Govindan, K.P.; Basha, S.; Ramesh, V.; Kumar, C.N.; Swathi, S. A comparative study on serum lipoprotein (a) and lipid profile between rheumatoid arthritis patients and normal subjects. J. Pharm. Bioallied Sci. 2015, 7, S22–S25. [Google Scholar] [CrossRef]
  41. Yang, X.; Chang, Y.; Wei, W. Endothelial dysfunction and inflammation: Immunity in rheumatoid arthritis. Mediators Inflamm. 2016, 2016, 6813016. [Google Scholar] [CrossRef] [Green Version]
  42. Salem, H.R.; Zahran, E.S. Vascular cell adhesion molecule-1 in rheumatoid arthritis patients: Relation to disease activity, oxidative stress, and systemic inflammation. Saudi Med. J. 2021, 42, 620–628. [Google Scholar] [CrossRef]
  43. Gimbrone, M.A.; Garcia-Cardena, G., Jr. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Davies, R.; Williams, J.; Sime, K.; Jin, H.S.; Thompson, C.; Jordan, L.; Lang, D.; Halcox, J.P.; Ellins, E.; Jones, G.W.; et al. The role of interleukin-6 trans-signalling on cardiovascular dysfunction in inflammatory arthritis. Rheumatology 2021, 60, 2852–2861. [Google Scholar] [CrossRef]
  45. Totoson, P.; Maguin-Gaté, K.; Prati, C.; Wendling, D.; Demougeot, C. Mechanisms of endothelial dysfunction in rheumatoid arthritis: Lessons from animal studies. Arthritis Res. Ther. 2014, 16, 202. [Google Scholar] [CrossRef] [Green Version]
  46. Su, J.B. Vascular endothelial dysfunction and pharmacological treatment. World J. Cardiol. 2015, 7, 719–741. [Google Scholar] [CrossRef]
  47. Akhmedov, A.; Crucet, M.; Simic, B.; Kraler, S.; Bonetti, N.R.; Ospelt, C.; Distler, O.; Ciurea, A.; Liberale, L.; Jauhiainen, M.; et al. TNFα induces endothelial dysfunction in rheumatoid arthritis via LOX-1 and arginase 2: Reversal by monoclonal TNFα antibodies. Cardiovasc. Res. 2021, cvab005. [Google Scholar] [CrossRef]
  48. Lassere, M.N.; Rappo, J.; Portek, I.J.; Sturgess, A.; Edmonds, J.P. How many life years are lost in patients with rheumatoid arthritis? Secular cause-specific and all-cause mortality in rheumatoid arthritis, and their predictors in a long-term Australian cohort study. Intern. Med. J. 2013, 43, 66–72. [Google Scholar] [CrossRef]
  49. van den Hoek, J.; Boshuizen, H.C.; Roorda, L.D.; Tijhuis, G.J.; Nurmohamed, M.T.; van den Bos, G.A.M.; Dekker, J. Mortality in patients with rheumatoid arthritis: A 15-year prospective cohort study. Rheumatol. Int. 2017, 37, 487–493. [Google Scholar] [CrossRef] [Green Version]
  50. Sokka, T.; Abelson, B.; Pincus, T. Mortality in rheumatoid arthritis: 2008 update. Clin. Exp. Rheumatol. 2008, 26 (Suppl. 51), S35–S61. [Google Scholar] [PubMed]
  51. Solomon, D.H.; Reed, G.W.; Kremer, J.M.; Curtis, J.R.; Farkouh, M.E.; Harrold, L.R.; Hochberg, M.C.; Tsao, P.; Greenberg, J.D. Disease activity in rheumatoid arthritis and the risk of cardiovascular events. Arthritis Rheumatol. 2015, 67, 1449–1455. [Google Scholar] [CrossRef]
  52. Chang, K.; Yang, S.M.; Kim, S.H.; Han, K.H.; Park, S.J.; Shin, J.I. Smoking and rheumatoid arthritis. Int. J. Mol. Sci. 2014, 15, 22279–22295. [Google Scholar] [CrossRef] [Green Version]
  53. Sandoo, A.; Chanchlani, N.; Hodson, J.; Smith, J.P.; Douglas, K.M.; Kitas, G.D. Classical cardiovascular disease risk factors associate with vascular function and morphology in rheumatoid arthritis: A six-year prospective study. Arthritis Res. Ther. 2013, 15, R203. [Google Scholar] [CrossRef] [Green Version]
  54. Chung, C.P.; Giles, J.T.; Kronmal, R.A.; Post, W.S.; Gelber, A.C.; Petri, M.; Szklo, M.; Detrano, R.; Budoff, M.J.; Blumenthal, R.S.; et al. Progression of coronary artery atherosclerosis in rheumatoid arthritis: Comparison with participants from the Multi-Ethnic Study of Atherosclerosis. Arthritis Res. Ther. 2013, 15, R134. [Google Scholar] [CrossRef] [Green Version]
  55. Jafri, K.; Bartels, C.M.; Shin, D.; Gelfand, J.M.; Ogdie, A. Incidence and Management of Cardiovascular Risk Factors in Psoriatic Arthritis and Rheumatoid Arthritis: A Population-Based Study. Arthritis Care Res. 2017, 69, 51–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. An, J.; Alemao, E.; Reynolds, K.; Kawabata, H.; Solomon, D.H.; Liao, K.P.; Niu, F.; Cheetham, T.C. Cardiovascular Outcomes Associated with Lowering Low-density Lipoprotein Cholesterol in Rheumatoid Arthritis and Matched Nonrheumatoid Arthritis. J. Rheumatol. 2016, 43, 1989–1996. [Google Scholar] [CrossRef]
  57. Willerson, J.T.; Ridker, P.M. Inflammation as a cardiovascular risk factor. Circulation 2004, 109 (Suppl. 1), II2–II10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Hansson, G.K. Inflammation, atherosclerosis, and coronary artery disease. N. Engl. J. Med. 2005, 352, 1685–1695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Geovanini, G.R.; Libby, P. Atherosclerosis and inflammation: Overview and updates. Clin. Sci. 2018, 132, 1243–1252. [Google Scholar] [CrossRef]
  60. Interleukin-6 Receptor Mendelian Randomisation Analysis (IL6R MR) Consortium; Swerdlow, D.I.; Holmes, M.V.; Kuchenbaecker, K.B.; Engmann, J.E.; Shah, T.; Sofat, R.; Guo, Y.; Chung, C.; Peasey, A.; et al. The interleukin-6 receptor as a target for prevention of coronary heart disease: A mendelian randomisation analysis. Lancet 2012, 379, 1214–1224. [Google Scholar] [CrossRef] [Green Version]
  61. 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] [Green Version]
  62. Larsen, B.A.; Laughlin, G.A.; Cummins, K.; Barrett-Connor, E.; Wassel, C.L. Adipokines and severity and progression of coronary artery calcium: Findings from the Rancho Bernardo Study. Atherosclerosis 2017, 265, 1–6. [Google Scholar] [CrossRef]
  63. López-Mejías, R.; González-Gay, M.A. IL-6: Linking chronic inflammation and vascular calcification. Nat. Rev. Rheumatol. 2019, 15, 457–459. [Google Scholar] [CrossRef]
  64. Rho, Y.H.; Chung, C.P.; Oeser, A.; Solus, J.; Asanuma, Y.; Sokka, T.; Pincus, T.; Raggi, P.; Gebretsadik, T.; Shintani, A.; et al. Inflammatory mediators and premature coronary atherosclerosis in rheumatoid arthritis. Arthritis Rheum. 2009, 61, 1580–1585. [Google Scholar] [CrossRef] [Green Version]
  65. Shobeiri, N.; Bendeck, M.P. Interleukin-1β Is a Key Biomarker and Mediator of Inflammatory Vascular Calcification. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 179–180. [Google Scholar] [CrossRef] [Green Version]
  66. Libby, P.; Ridker, P.M.; Hansson, G.K. Progress and challenges in translating the biology of atherosclerosis. Nature 2011, 473, 317–325. [Google Scholar] [CrossRef] [PubMed]
  67. Wei, S.T.; Sun, Y.H.; Zong, S.H.; Xiang, Y.B. Serum levels of IL-6 and TNF-α may correlate with activity and severity of rheumatoid arthritis. Med. Sci. Monit. 2015, 21, 4030–4038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Popa, C.; van Tits, L.J.; Barrera, P.; Lemmers, H.L.; van den Hoogen, F.H.; van Riel, P.L.; Radstake, T.R.; Netea, M.G.; Roest, M.; Stalenhoef, A.F. Anti-inflammatory therapy with tumour necrosis factor alpha inhibitors improves high-density lipoprotein cholesterol antioxidative capacity in rheumatoid arthritis patients. Ann. Rheum. Dis. 2009, 68, 868–872. [Google Scholar] [CrossRef]
  69. 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]
  70. Bartoloni, E.; Alunno, A.; Bistoni, O.; Gerli, R. How early is the atherosclerotic risk in rheumatoid arthritis? Autoimmun. Rev. 2010, 9, 701–707. [Google Scholar] [CrossRef]
  71. Page, M.J.; Bester, J.; Pretorius, E. The inflammatory effects of TNF-α and complement component 3 on coagulation. Sci. Rep. 2018, 8, 1812. [Google Scholar] [CrossRef] [Green Version]
  72. Chen, Z.; Tang, M.; Huang, D.; Jiang, W.; Li, M.; Ji, H.; Park, J.; Xu, B.; Atchison, L.J.; Truskey, G.A.; et al. Real-time observation of leukocyte-endothelium interactions in tissue-engineered blood vessel. Lab Chip 2018, 18, 2047–2054. [Google Scholar] [CrossRef]
  73. Barbati, C.; Colasanti, T.; Vomero, M.; Ceccarelli, F.; Celia, A.I.; Perricone, C.; Spinelli, F.R.; Conti, F.; Valesini, G.; Alessandri, C. Up-regulation of autophagy by etanercept treatment results in TNF-induced apoptosis reduction in EA.hy926 endothelial cell line. Clin. Exp. Rheumatol. 2021, 39, 606–611. [Google Scholar] [PubMed]
  74. Mussbacher, M.; Salzmann, M.; Brostjan, C.; Hoesel, B.; Schoergenhofer, C.; Datler, H.; Hohensinner, P.; Basílio, J.; Petzelbauer, P.; Assinger, A.; et al. Cell Type-Specific Roles of NF-κB Linking Inflammation and Thrombosis. Front. Immunol. 2019, 10, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Jia, Z.; Nallasamy, P.; Liu, D.; Shah, H.; Li, J.Z.; Chitrakar, R.; Si, H.; McCormick, J.; Zhu, H.; Zhen, W.; et al. Luteolin protects against vascular inflammation in mice and TNF-alpha-induced monocyte adhesion to endothelial cells via suppressing IKBα/NF-κB signaling pathway. J. Nutr. Biochem. 2015, 26, 293–302. [Google Scholar] [CrossRef] [Green Version]
  76. Gunter, S.; Solomon, A.; Tsang, L.; Woodiwiss, A.J.; Robinson, C.; Millen, A.M.; Norton, G.R.; Dessein, P.H. Apelin concentrations are associated with altered atherosclerotic plaque stability mediator levels and atherosclerosis in rheumatoid arthritis. Atherosclerosis 2017, 256, 75–81. [Google Scholar] [CrossRef] [PubMed]
  77. Karpouzas, G.A.; Malpeso, J.; Choi, T.Y.; Li, D.; Munoz, S.; Budoff, M.J. Prevalence, extent and composition of coronary plaque in patients with rheumatoid arthritis without symptoms or prior diagnosis of coronary artery disease. Ann. Rheum. Dis. 2014, 73, 1797–1804. [Google Scholar] [CrossRef] [PubMed]
  78. Mai, W.; Liao, Y. Targeting IL-1β in the Treatment of Atherosclerosis. Front. Immunol. 2020, 11, 589654. [Google Scholar] [CrossRef] [PubMed]
  79. Ahmed, A.; Hollan, I.; Curran, S.A.; Riggio, M.P.; Mikkelsen, K.; Almdahl, S.M.; Aukrust, P.; McInnes, I.B.; Goodyear, C.S. Brief Report: Proatherogenic Cytokine Microenvironment in the Aortic Adventitia of Patients with Rheumatoid Arthritis. Arthritis Rheumatol. 2016, 68, 1361–1366. [Google Scholar] [CrossRef]
  80. Winchester, R.; Giles, J.T.; Nativ, S.; Downer, K.; Zhang, H.Z.; Bag-Ozbek, A.; Zartoshti, A.; Bokhari, S.; Bathon, J.M. Association of Elevations of Specific T Cell and Monocyte Subpopulations in Rheumatoid Arthritis with Subclinical Coronary Artery Atherosclerosis. Arthritis Rheumatol. 2016, 68, 92–102. [Google Scholar] [CrossRef] [PubMed]
  81. Hot, A.; Lenief, V.; Miossec, P. Combination of IL-17 and TNFα induces a pro-inflammatory, pro-coagulant and pro-thrombotic phenotype in human endothelial cells. Ann. Rheum. Dis. 2012, 71, 768–776. [Google Scholar] [CrossRef]
  82. Ramji, D.P.; Davies, T.S. Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev. 2015, 26, 673–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kim, I.; Moon, S.O.; Kim, S.H.; Kim, H.J.; Koh, Y.S.; Koh, G.Y. Vascular endothelial growth factor expression of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin through nuclear factor-kappa B activation in endothelial cells. J. Biol. Chem. 2001, 276, 7614–7620. [Google Scholar] [CrossRef] [Green Version]
  84. Pamukcu, B.; Lip, G.Y.; Shantsila, E. The nuclear factor—kappa B pathway in atherosclerosis: A potential therapeutic target for atherothrombotic vascular disease. Thromb. Res. 2011, 128, 117–123. [Google Scholar] [CrossRef]
  85. Li, Q.; Verma, I.M. NF-kappaB regulation in the immune system. Nat. Rev. Immunol. 2002, 2, 725–734. [Google Scholar] [CrossRef] [PubMed]
  86. Chung, H.Y.; Kim, D.H.; Lee, E.K.; Chung, K.W.; Chung, S.; Lee, B.; Seo, A.Y.; Chung, J.H.; Jung, Y.S.; Im, E.; et al. Redefining Chronic Inflammation in Aging and Age-Related Diseases: Proposal of the Senoinflammation Concept. Aging Dis. 2019, 10, 367–382. [Google Scholar] [CrossRef] [Green Version]
  87. Shen, C.; Sun, X.G.; Liu, N.; Mu, Y.; Hong, C.C.; Wei, W.; Zheng, F. Increased serum amyloid A and its association with autoantibodies, acute phase reactants and disease activity in patients with rheumatoid arthritis. Mol. Med. Rep. 2015, 11, 1528–1534. [Google Scholar] [CrossRef] [Green Version]
  88. Sack, G.H., Jr. Serum amyloid A (SAA) proteins. Subcell. Biochem. 2020, 94, 421–436. [Google Scholar] [CrossRef] [PubMed]
  89. Sato, M.; Ohkawa, R.; Yoshimoto, A.; Yano, K.; Ichimura, N.; Nishimori, M.; Okubo, S.; Yatomi, Y.; Tozuka, M. Effects of serum amyloid A on the structure and antioxidant ability of high-density lipoprotein. Biosci. Rep. 2016, 36, e00369. [Google Scholar] [CrossRef] [Green Version]
  90. Green, M.M.; Gough, A.A.; Devlin, J.; Smith, J.; Astin, P.; Taylor, D.; Emery, P. Serum MMP-3 and MMP-1 and progression of joint damage in early rheumatoid arthritis. Rheumatology 2003, 42, 83–88. [Google Scholar] [CrossRef] [Green Version]
  91. Fiedorczyk, M.; Klimiuk, P.A.; Sierakowski, S.; Gindzienska-Sieskiewicz, E.; Chwiecko, J. Serum matrix metalloproteinases and tissue inhibitors of metalloproteinases in patients with early rheumatoid arthritis. J. Rheumatol. 2006, 33, 1523–1529. [Google Scholar]
  92. Tchetverikov, I.; Lard, L.R.; DeGroot, J.; Verzijl, N.; TeKoppele, J.M.; Breedveld, F.C.; Huizinga, T.W.; Hanemaaijer, R. Matrix metalloproteinases-3, -8, -9 as markers of disease activity and joint damage progression in early rheumatoid arthritis. Ann. Rheum. Dis. 2003, 62, 1094–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Newby, A.C. Metalloproteinases and vulnerable atherosclerotic plaques. Trends Cardiovasc. Med. 2007, 17, 253–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Olejarz, W.; Łacheta, D.; Kubiak-Tomaszewska, G. Matrix Metalloproteinases as Biomarkers of Atherosclerotic Plaque Instability. Int. J. Mol. Sci. 2020, 21, 3946. [Google Scholar] [CrossRef]
  95. Tousoulis, D.; Oikonomou, E.; Economou, E.K.; Crea, F.; Kaski, J.C. Inflammatory cytokines in atherosclerosis: Current therapeutic approaches. Eur. Heart J. 2016, 37, 1723–1732. [Google Scholar] [CrossRef] [Green Version]
  96. Bennett, M.R.; Sinha, S.; Owens, G.K. Vascular smooth muscle cells in atherosclerosis. Circ. Res. 2016, 118, 692–702. [Google Scholar] [CrossRef] [PubMed]
  97. Espié, P.; He, Y.; Koo, P.; Sickert, D.; Dupuy, C.; Chokoté, E.; Schuler, R.; Mergentaler, H.; Ristov, J.; Milojevic, J.; et al. First-in-human clinical trial to assess pharmacokinetics, pharmacodynamics, safety, and tolerability of iscalimab, an anti-CD40 monoclonal antibody. Am. J. Transplant. 2020, 20, 463–473. [Google Scholar] [CrossRef]
  98. Peters, A.L.; Stunz, L.L.; Bishop, G.A. CD40 and autoimmunity: The dark side of a great activator. Semin. Immunol. 2009, 21, 293–300. [Google Scholar] [CrossRef] [Green Version]
  99. Antoniades, C.; Bakogiannis, C.; Tousoulis, D.; Antonopoulos, A.S.; Stefanadis, C. The CD40/CD40 ligand system: Linking inflammation with atherothrombosis. J. Am. Coll. Cardiol. 2009, 54, 669–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Mälarstig, A.; Lindahl, B.; Wallentin, L.; Siegbahn, A. Soluble CD40L levels are regulated by the -3459 A>G polymorphism and predict myocardial infarction and the efficacy of antithrombotic treatment in non-ST elevation acute coronary syndrome. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 1667–1673. [Google Scholar] [CrossRef] [Green Version]
  101. Wang, C.; Yan, J.; Yang, P.; Du, R.; Chen, G. The relationship between CD40 gene polymorphism and unstable coronary atherosclerotic plaques. Clin. Cardiol. 2010, 33, E55–E60. [Google Scholar] [CrossRef]
  102. Michel, N.A.; Zirlik, A.; Wolf, D. CD40L and Its Receptors in Atherothrombosis-An Update. Front. Cardiovasc. Med. 2017, 4, 40. [Google Scholar] [CrossRef] [Green Version]
  103. Mateen, S.; Moin, S.; Khan, A.Q.; Zafar, A.; Fatima, N. Increased reactive oxygen species formation and oxidative stress in rheumatoid arthritis. PLoS ONE 2016, 11, e0152925. [Google Scholar] [CrossRef] [Green Version]
  104. Quiñonez-Flores, C.M.; González-Chávez, S.A.; Del Río Nájera, D.; Pacheco-Tena, C. Oxidative Stress Relevance in the Pathogenesis of the Rheumatoid Arthritis: A Systematic Review. Biomed. Res. Int. 2016, 2016, 6097417. [Google Scholar] [CrossRef] [Green Version]
  105. Harrison, D.; Griendling, K.K.; Landmesser, U.; Hornig, B.; Drexler, H. Role of oxidative stress in atherosclerosis. Am. J. Cardiol. 2003, 91, 7A–11A. [Google Scholar] [CrossRef]
  106. Sarban, S.; Kocyigit, A.; Yazar, M.; Isikan, U.E. Plasma total antioxidant capacity, lipid peroxidation, and erythrocyte antioxidant enzyme activities in patients with rheumatoid arthritis and osteoarthritis. Clin. Biochem. 2005, 38, 981–986. [Google Scholar] [CrossRef]
  107. Oğul, Y.; Gür, F.; Cengiz, M.; Gür, B.; Sarı, R.A.; Kızıltunç, A. Evaluation of oxidant and intracellular anti-oxidant activity in rheumatoid arthritis patients: In vivo and in silico studies. Int. Immunopharmacol. 2021, 97, 107654. [Google Scholar] [CrossRef] [PubMed]
  108. Wang, Y.; Tabas, I. Emerging roles of mitochondria ROS in atherosclerotic lesions: Causation or association? J. Atheroscler. Thromb. 2014, 21, 381–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Kattoor, A.J.; Pothineni, N.V.K.; Palagiri, D.; Mehta, J.L. Oxidative stress in atherosclerosis. Curr. Atheroscler. Rep. 2017, 19, 42. [Google Scholar] [CrossRef]
  110. Datta, S.; Kundu, S.; Ghosh, P.; De, S.; Ghosh, A.; Chatterjee, M. Correlation of oxidant status with oxidative tissue damage in patients with rheumatoid arthritis. Clin. Rheumatol. 2014, 33, 1557–1564. [Google Scholar] [CrossRef]
  111. Marchio, P.; Guerra-Ojeda, S.; Vila, J.M.; Aldasoro, M.; Victor, V.M.; Mauricio, M.D. Targeting early atherosclerosis: A focus on oxidative stress and inflammation. Oxid. Med. Cell Longev. 2019, 2019, 8563845. [Google Scholar] [CrossRef] [PubMed]
  112. Herlitz-Cifuentes, H.; Vejar, C.; Flores, A.; Jara, P.; Bustos, P.; Castro, I.; Poblete, E.; Saez, K.; Opazo, M.; Gajardo, J.; et al. Plasma from Patients with Rheumatoid Arthritis Reduces Nitric Oxide Synthesis and Induces Reactive Oxygen Species in A Cell-Based Biosensor. Biosensors 2019, 9, 32. [Google Scholar] [CrossRef] [Green Version]
  113. Solomon, D.H.; Bitton, A.; Katz, J.N.; Radner, H.; Brown, E.M.; Fraenkel, L. Review: Treat to target in rheumatoid arthritis: Fact, fiction, or hypothesis? Arthritis Rheumatol. 2014, 66, 775–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Bugatti, S.; De Stefano, L.; Manzo, A.; Sakellariou, G.; Xoxi, B.; Montecucco, C. Limiting factors to Boolean remission differ between autoantibody-positive and -negative patients in early rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis. 2021, 13, 1759720X211011826. [Google Scholar] [CrossRef]
  115. Smolen, J.S.; Landewé, R.; Bijlsma, J.; Burmester, G.; Chatzidionysiou, K.; Dougados, M.; Nam, J.; Ramiro, S.; Voshaar, M.; van Vollenhoven, R.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann. Rheum. Dis. 2017, 76, 960–977. [Google Scholar] [CrossRef]
  116. Wasserman, A.M. Diagnosis and management of rheumatoid arthritis. Am. Fam. Physician 2011, 84, 1245–1252. [Google Scholar] [PubMed]
  117. Burmester, R.; Pope, J.E. Novel treatment strategies in rheumatoid arthritis. Lancet 2017, 389, 2338–2348. [Google Scholar] [CrossRef]
  118. Abbasi, M.; Mousavi, M.J.; Jamalzehi, S.; Alimohammadi, R.; Bezvan, M.H.; Mohammadi, H.; Aslani, S. Strategies toward rheumatoid arthritis therapy; the old and the new. J. Cell Physiol. 2019, 234, 10018–10031. [Google Scholar] [CrossRef]
  119. Kavanaugh, A.; Wells, A.F. Benefits and risks of low-dose glucocorticoid treatment in the patient with rheumatoid arthritis. Rheumatology 2014, 53, 1742–1751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Giles, J.T.; Rist, P.M.; Liao, K.P.; Tawakol, A.; Fayad, Z.A.; Mani, V.; Paynter, N.P.; Ridker, P.M.; Glynn, R.J.; Lu, F.; et al. Testing the Effects of Disease-Modifying Antirheumatic Drugs on Vascular Inflammation in Rheumatoid Arthritis: Rationale and Design of the TARGET Trial. ACR Open Rheumatol. 2021, 3, 371–380. [Google Scholar] [CrossRef] [PubMed]
  121. Liao, K.P. Cardiovascular disease in patients with rheumatoid arthritis. Trends Cardiovasc. Med. 2017, 27, 136–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Masson, W.; Rossi, E.; Alvarado, R.N.; Cornejo-Peña, G.; Damonte, J.I.; Fiorini, N.; Mora-Crespo, L.M.; Tobar-Jaramillo, M.A.; Scolnik, M. Rheumatoid Arthritis, Statin Indication and Lipid Goals: Analysis According to Different Recommendations. Rheumatol. Clin. 2021. [Google Scholar] [CrossRef]
  123. Chhibber, A.; Hansen, S.; Biskupiak, J. Statin use and mortality in rheumatoid arthritis: An incident user cohort study. J. Manag. Care Spec. Pharm. 2021, 27, 296–305. [Google Scholar] [CrossRef]
  124. Abeles, A.M.; Pillinger, M.H. Statins as anti-inflammatory and immunomodulatory agents: A future in rheumatologic therapy? Arthritis Rheum. 2006, 54, 393–407. [Google Scholar] [CrossRef]
  125. Asher, J.; Houston, M. Statins and C-reactive protein levels. J. Clin. Hypertens. 2007, 9, 622–628. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Oesterle, A.; Laufs, U.; Liao, J.K. Pleiotropic effects of statins on the cardiovascular system. Circ. Res. 2017, 120, 229–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Oesterle, A.; Liao, J.K. The Pleiotropic effects of statins—From coronary artery disease and stroke to atrial fibrillation and ventricular tachyarrhythmia. Curr. Vasc. Pharmacol. 2019, 17, 222–232. [Google Scholar] [CrossRef] [PubMed]
  129. van Boheemen, L.; Turk, S.; Beers-Tas, M.V.; Bos, W.; Marsman, D.; Griep, E.N.; Starmans-Kool, M.; Popa, C.D.; van Sijl, A.; Boers, M.; et al. Atorvastatin is unlikely to prevent rheumatoid arthritis in high risk individuals: Results from the prematurely stopped STAtins to Prevent Rheumatoid Arthritis (STAPRA) trial. RMD Open 2021, 7, e001591. [Google Scholar] [CrossRef] [PubMed]
  130. 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] [PubMed]
  131. Pang, Z.; Wang, G.; Ran, N.; Lin, H.; Wang, Z.; Guan, X.; Yuan, Y.; Fang, K.; Liu, J.; Wang, F. Inhibitory Effect of Methotrexate on Rheumatoid Arthritis Inflammation and Comprehensive Metabolomics Analysis Using Ultra-Performance Liquid Chromatography-Quadrupole Time of Flight-Mass Spectrometry (UPLC-Q/TOF-MS). Int. J. Mol. Sci. 2018, 19, 2894. [Google Scholar] [CrossRef] [Green Version]
  132. Singh, J.A.; Saag, K.G.; Bridges, S.L., Jr.; Akl, E.A.; Bannuru, R.R.; Sullivan, M.C.; Vaysbrot, E.; McNaughton, C.; Osani, M.; Shmerling, R.H.; et al. 2015 American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Care Res. 2016, 68, 1–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Smolen, J.S.; Landewé, R.B.M.; Bijlsma, J.W.J.; Burmester, G.R.; Dougados, M.; Kerschbaumer, A.; McInnes, I.B.; Sepriano, A.; van Vollenhoven, R.F.; de Wit, M.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann. Rheum. Dis. 2020, 79, 685–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. 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]
  135. Verhoeven, F.; Prati, C.; Chouk, M.; Demougeot, C.; Wendling, D. Methotrexate and cardiovascular risk in rheumatic diseases: A comprehensive review. Expert Rev. Clin. Pharmacol. 2021, 1–8. [Google Scholar] [CrossRef]
  136. Sun, K.J.; Liu, L.L.; Hu, J.H.; Chen, Y.Y.; Xu, D.Y. Methotrexate can prevent cardiovascular events in patients with rheumatoid arthritis: An updated meta-analysis. Medicine 2021, 100, e24579. [Google Scholar] [CrossRef]
  137. Coomes, E.; Chan, E.S.; Reiss, A.B. Methotrexate in atherogenesis and cholesterol metabolism. Cholesterol 2011, 2011, 503028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Cronstein, B.N.; Aune, T.M. Methotrexate and its mechanisms of action in inflammatory arthritis. Nat. Rev. Rheumatol. 2020, 16, 145–154. [Google Scholar] [CrossRef]
  139. Reiss, A.B.; Cronstein, B.N. Regulation of foam cells by adenosine. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 879–886. [Google Scholar] [CrossRef] [Green Version]
  140. 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] [Green Version]
  141. Chen, D.Y.; Chih, H.M.; Lan, J.L.; Chang, H.Y.; Chen, W.W.; Chiang, E.P. Blood lipid profiles and peripheral blood mononuclear cell cholesterol metabolism gene expression in patients with and without methotrexate treatment. BMC Med. 2011, 9, 4. [Google Scholar] [CrossRef] [Green Version]
  142. 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]
  143. 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. Rheumat. 2021, 17, 270–290. [Google Scholar] [CrossRef] [PubMed]
  144. Chaabane, S.; Messedi, M.; Akrout, R.; Ben Hamad, M.; Turki, M.; Marzouk, S.; Keskes, L.; Bahloul, Z.; Rebai, A.; Ayedi, F.; et al. Association of hyperhomocysteinemia with genetic variants in key enzymes of homocysteine metabolism and methotrexate toxicity in rheumatoid arthritis patients. Inflamm. Res. 2018, 67, 703–710. [Google Scholar] [CrossRef]
  145. 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, annrheumdis-2021-220125. [Google Scholar] [CrossRef] [PubMed]
  146. Stenson, W.F.; Lobos, E.A. Inhibition of platelet thromboxane synthetase by sulphasalazine. Biochem. Pharmacol. 1983, 32, 2205–2209. [Google Scholar] [CrossRef]
  147. 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]
  148. Day, A.L.; Singh, J.A. Cardiovascular Disease Risk in Older Adults and Elderly Patients with Rheumatoid Arthritis: What Role Can Disease-Modifying Antirheumatic Drugs Play in Cardiovascular Risk Reduction? Drugs Aging 2019, 36, 493–510. [Google Scholar] [CrossRef]
  149. 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] [Green Version]
  150. Wahl, C.; Liptay, S.; Adler, G.; Schmid, R.M. Sulfasalazine: A potent and specific inhibitor of nuclear factor kappa B. J. Clin. Investig. 1998, 101, 1163–1174. [Google Scholar] [CrossRef] [Green Version]
  151. Pateras, I.; Giaginis, C.; Tsigris, C.; Patsouris, E.; Theocharis, S. NF-κB signaling at the crossroads of inflammation and atherogenesis: Searching for new therapeutic links. Expert Opin. Ther. Targets 2014, 18, 1089–1101. [Google Scholar] [CrossRef]
  152. Park, Y.B.; Choi, H.K.; Kim, M.Y.; 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]
  153. Atzeni, F.; Turiel, M.; Caporali, R.; Cavagna, L.; Tomasoni, L.; Sitia, S.; Sarzi-Puttini, P. The effect of pharmacological therapy on the cardiovascular system of patients with systemic rheumatic diseases. Autoimmun. Rev. 2010, 9, 835–839. [Google Scholar] [CrossRef] [PubMed]
  154. Karimifar, M.; Sepehrifar, M.S.; Moussavi, H.; Sepehrifar, M.B.; Mottaghi, P.; Siavash, M.; Karimifar, M. The effects of conventional drugs in the treatment of rheumatoid arthritis on the serum lipids. J. Res. Med. Sci. 2018, 23, 105. [Google Scholar] [CrossRef] [PubMed]
  155. 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] [Green Version]
  156. Zanten, J.; Sandoo, A.; Metsios, G.S.; Kalinoglou, A.; Ntoumanis, N.; Kitas, G.D. Comparison of the effects of exercise and anti-TNF treatment on cardiovascular health in rheumatoid arthritis: Results from two controlled trials. Rheumatol. Int. 2019, 39, 219–225. [Google Scholar] [CrossRef] [Green Version]
  157. Barnabe, C.; Martin, B.J.; Ghali, W.A. Systematic review and meta-analysis: Anti-tumor necrosis factor α therapy and cardiovascular events in rheumatoid arthritis. Arthritis Care Res. 2011, 63, 522–529. [Google Scholar] [CrossRef]
  158. Ntusi, N.A.B.; Francis, J.M.; Sever, E.; Liu, A.; Piechnik, S.K.; Ferreira, V.M.; Matthews, P.M.; Robson, M.D.; Wordsworth, P.B.; Neubauer, S.; et al. Anti-TNF modulation reduces myocardial inflammation and improves cardiovascular function in systemic rheumatic diseases. Int. J. Cardiol. 2018, 270, 253–259. [Google Scholar] [CrossRef] [PubMed]
  159. Ait-Oufella, H.; Libby, P.; Tedgui, A. Anticytokine Immune Therapy and Atherothrombotic Cardiovascular Risk. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1510–1519. [Google Scholar] [CrossRef]
  160. Thalayasingam, N.; Isaacs, J.D. Anti-TNF therapy. Best Pract. Res. Clin. Rheumatol. 2011, 25, 549–567. [Google Scholar] [CrossRef]
  161. Low, A.S.; Symmons, D.P.; Lunt, M.; Mercer, L.K.; Gale, C.P.; Watson, K.D.; Dixon, W.G.; Hyrich, K.L.; British Society for Rheumatology Biologics Register for Rheumatoid Arthritis, BSRBR-RA; BSRBR Control Centre Consortium. Relationship between exposure to tumour necrosis factor inhibitor therapy and incidence and severity of myocardial infarction in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2017, 76, 654–660. [Google Scholar] [CrossRef] [Green Version]
  162. Végh, E.; Kerekes, G.; Pusztai, A.; Hamar, A.; Szamosi, S.; Váncsa, A.; Bodoki, L.; Pogácsás, L.; Balázs, F.; Hodosi, K.; et al. Effects of 1-year anti-TNF-α therapy on vascular function in rheumatoid arthritis and ankylosing spondylitis. Rheumatol. Int. 2020, 40, 427–436. [Google Scholar] [CrossRef] [Green Version]
  163. Pusztai, A.; Hamar, A.; Horváth, Á.; Gulyás, K.; Végh, E.; Bodnár, N.; Kerekes, G.; Czókolyová, M.; Szamosi, S.; Bodoki, L.; et al. Soluble Vascular Biomarkers in Rheumatoid Arthritis and Ankylosing Spondylitis: Effects of 1-year Antitumor Necrosis Factor-α Therapy. J. Rheumatol. 2021, 48, 821–828. [Google Scholar] [CrossRef] [PubMed]
  164. Greco, T.P.; Conti-Kelly, A.M.; Anthony, J.R.; Greco, T., Jr.; Doyle, R.; Boisen, M.; Kojima, K.; Matsuura, E.; Lopez, L.R. Oxidized-LDL/beta2-glycoprotein I complexes are associated with disease severity and increased risk for adverse outcomes in patients with acute coronary syndromes. Am. J. Clin. Pathol. 2010, 133, 737–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Karpouzas, G.A.; Bui, V.L.; Ronda, N.; Hollan, I.; Ormseth, S.R. Biologics and atherosclerotic cardiovascular risk in rheumatoid arthritis: A review of evidence and mechanistic insights. Expert Rev. Clin. Immunol. 2021, 17, 355–374. [Google Scholar] [CrossRef] [PubMed]
  166. Grimble, R.F. Inflammatory status and insulin resistance. Curr. Opin. Clin. Nutr. Metab. Care 2002, 5, 551–559. [Google Scholar] [CrossRef] [PubMed]
  167. Chung, C.P.; Oeser, A.; Solus, J.F.; Gebretsadik, T.; Shintani, A.; Avalos, I.; Sokka, T.; Raggi, P.; Pincus, T.; Stein, C.M. Inflammation-associated insulin resistance: Differential effects in rheumatoid arthritis and systemic lupus erythematosus define potential mechanisms. Arthritis Rheum. 2008, 58, 2105–2112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Avouac, J.; Elhai, M.; Forien, M.; Sellam, J.; Eymard, F.; Molto, A.; Banal, F.; Damiano, J.; Dieudé, P.; Larger, E.; et al. Influence of inflammatory and non-inflammatory rheumatic disorders on the clinical and biological profile of type-2 diabetes. Rheumatology 2021, 60, 3598–3606. [Google Scholar] [CrossRef] [PubMed]
  169. Wijbrandts, C.A.; Leuven, S.I.; Boom, H.D.; Gerlag, D.M.; Stroes, E.G.; Kastelein, J.J.; Tak, P.P. Sustained changes in lipid profile and macrophage migration inhibitory factor levels after anti-tumor necrosis factor therapy in rheumatoid arthritis. Ann. Rheum. Dis. 2009, 68, 1316–1321. [Google Scholar] [CrossRef] [PubMed]
  170. Min, H.K.; Kim, H.R.; Lee, S.H.; Shin, K.; Kim, H.A.; Park, S.H.; Kwok, S.K. Four-year follow-up of atherogenicity in rheumatoid arthritis patients: From the nationwide Korean College of Rheumatology Biologics Registry. Clin. Rheumatol. 2021, 40, 3105–3113. [Google Scholar] [CrossRef] [PubMed]
  171. Cacciapaglia, F.; Anelli, M.G.; Rinaldi, A.; Serafino, L.; Covelli, M.; Scioscia, C.; Iannone, F.; Lapadula, G. Lipid profile of rheumatoid arthritis patients treated with anti-tumor necrosis factor-alpha drugs changes according to disease activity and predicts clinical response. Drug Dev. Res. 2014, 75 (Suppl. 1), S77–S80. [Google Scholar] [CrossRef]
  172. Daïen, C.I.; Duny, Y.; Barnetche, T.; Daurès, J.P.; Combe, B.; Morel, J. Effect of TNF inhibitors on lipid profile in rheumatoid arthritis: A systematic review with meta-analysis. Ann. Rheum. Dis. 2012, 71, 862–868. [Google Scholar] [CrossRef] [PubMed]
  173. Cacciapaglia, F.; Anelli, M.G.; Rinaldi, A.; Fornaro, M.; Lopalco, G.; Scioscia, C.; Lapadula, G.; Iannone, F. Lipids and Atherogenic Indices Fluctuation in Rheumatoid Arthritis Patients on Long-Term Tocilizumab Treatment. Mediat. Inflamm. 2018, 2018, 2453265. [Google Scholar] [CrossRef]
  174. Jones, G.; Sebba, A.; Gu, J.; Lowenstein, M.B.; Calvo, A.; Gomez-Reino, J.J.; Siri, D.A.; Tomsic, M.; Alecock, E.; Woodworth, T.; et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: The AMBITION study. Ann. Rheum. Dis. 2010, 69, 88–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Kawashiri, S.Y.; Kawakami, A.; Yamasaki, S.; Imazato, T.; Iwamoto, N.; Fujikawa, K.; Aramaki, T.; Tamai, M.; Nakamura, H.; Ida, H.; et al. Effects of the anti-interleukin-6 receptor antibody, tocilizumab, on serum lipid levels in patients with rheumatoid arthritis. Rheumatol. Int. 2011, 31, 451–456. [Google Scholar] [CrossRef]
  176. Smolen, J.S.; Beaulieu, A.; Rubbert-Roth, A.; Ramos-Remus, C.; Rovensky, J.; Alecock, E.; Woodworth, T.; Alten, R.; OPTION Investigators. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): A double-blind, placebo-controlled, randomised trial. Lancet 2008, 371, 987–997. [Google Scholar] [CrossRef]
  177. Zhang, J.; Xie, F.; Yun, H.; Chen, L.; Muntner, P.; Levitan, E.B.; Safford, M.M.; Kent, S.T.; Osterman, M.T.; Lewis, J.D.; et al. Comparative effects of biologics on cardiovascular risk among older patients with rheumatoid arthritis. Ann. Rheum. Dis. 2016, 75, 1813–1818. [Google Scholar] [CrossRef]
  178. Kim, S.C.; Solomon, D.H.; Rogers, J.R.; Gale, S.; Klearman, M.; Sarsour, K.; Schneeweiss, S. Cardiovascular Safety of Tocilizumab Versus Tumor Necrosis Factor Inhibitors in Patients With Rheumatoid Arthritis: A Multi-Database Cohort Study. Arthritis Rheumatol. 2017, 69, 1154–1164. [Google Scholar] [CrossRef] [Green Version]
  179. 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, e0220178. [Google Scholar] [CrossRef]
  180. Cacciapaglia, F.; Perniola, S.; Venerito, V.; Anelli, M.G.; Härdfeldt, J.; Fornaro, M.; Moschetta, A.; Iannone, F. The Impact of Biologic Drugs on High-Density Lipoprotein Cholesterol Efflux Capacity in Rheumatoid Arthritis Patients. J. Clin. Rheumatol. 2020. [Google Scholar] [CrossRef] [PubMed]
  181. Ormseth, M.J.; Yancey, P.G.; Solus, J.F.; Bridges, S.L., Jr.; Curtis, J.R.; Linton, M.F.; Fazio, S.; Davies, S.S.; Roberts, L.J., 2nd; Vickers, K.C.; et al. Effect of Drug Therapy on Net Cholesterol Efflux Capacity of High-Density Lipoprotein-Enriched Serum in Rheumatoid Arthritis. Arthritis Rheumatol. 2016, 68, 2099–2105. [Google Scholar] [CrossRef] [PubMed]
  182. Anastasius, M.; Luquain-Costaz, C.; Kockx, M.; Jessup, W.; Kritharides, L. A critical appraisal of the measurement of serum “cholesterol efflux capacity” and its use as surrogate marker of risk of cardiovascular disease. Biochim. Biophys. Acta. Mol. Cell. Biol Lipids 2018, 1863, 1257–1273. [Google Scholar] [CrossRef]
  183. Pierini, F.S.; Botta, E.; Soriano, E.R.; Martin, M.; Boero, L.; Meroño, T.; Saez, M.S.; Lozano Chiappe, E.; Cerda, O.; Brites, F. Effect of Tocilizumab on LDL and HDL Characteristics in Patients with Rheumatoid Arthritis. An Observational Study. Rheumatol. Ther. 2021, 8, 803–815. [Google Scholar] [CrossRef]
  184. Ferraz-Amaro, I.; Hernández-Hernández, M.V.; Tejera-Segura, B.; Delgado-Frías, E.; Macía-Díaz, M.; Machado, J.D.; Diaz-González, F. Effect of IL-6 Receptor Blockade on Proprotein Convertase Subtilisin/Kexin Type-9 and Cholesterol Efflux Capacity in Rheumatoid Arthritis Patients. Horm. Metab. Res. 2019, 51, 200–209. [Google Scholar] [CrossRef] [PubMed]
  185. Bechman, K.; Yates, M.; Galloway, J.B. The new entries in the therapeutic armamentarium: The small molecule JAK inhibitors. Pharmacol. Res. 2019, 147, 104392. [Google Scholar] [CrossRef]
  186. Moura, R.A.; Fonseca, J.E. JAK inhibitors and modulation of B cell immune responses in rheumatoid arthritis. Front. Med. 2020, 7, 607725. [Google Scholar] [CrossRef] [PubMed]
  187. Strand, V.; Goncalves, J.; Isaacs, J.D. Immunogenicity of biologic agents in rheumatology. Nat. Rev. Rheumatol. 2021, 17, 81–97. [Google Scholar] [CrossRef] [PubMed]
  188. Mori, S.; Ogata, F.; Tsunoda, R. Risk of venous thromboembolism associated with Janus kinase inhibitors for rheumatoid arthritis: Case presentation and literature review. Clin. Rheumatol. 2021, 40, 4457–4471. [Google Scholar] [CrossRef] [PubMed]
  189. Charles-Schoeman, C.; Wicker, P.; Gonzalez-Gay, M.A.; Boy, M.; Zuckerman, A.; Soma, K.; Geier, J.; Kwok, K.; Riese, R. Cardiovascular safety findings in patients with rheumatoid arthritis treated with tofacitinib, an oral Janus kinase inhibitor. Semin. Arthritis Rheum. 2016, 46, 261–271. [Google Scholar] [CrossRef]
  190. 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] [Green Version]
  191. Wollenhaupt, J.; Lee, E.B.; Curtis, J.R.; Silverfield, J.; Terry, K.; Soma, K.; Mojcik, C.; DeMasi, R.; Strengholt, S.; Kwok, K.; et al. Safety and efficacy of tofacitinib for up to 9.5 years in the treatment of rheumatoid arthritis: Final results of a global, open-label, long-term extension study. Arthritis Res. Ther. 2019, 21, 89. [Google Scholar] [CrossRef] [Green Version]
  192. Conaghan, P.G.; Mysler, E.; Tanaka, Y.; Da Silva-Tillmann, B.; Shaw, T.; Liu, J.; Ferguson, R.; Enejosa, J.V.; Cohen, S.; Nash, P.; et al. Upadacitinib in Rheumatoid Arthritis: A Benefit-Risk Assessment Across a Phase III Program. Drug Saf. 2021, 44, 515–530. [Google Scholar] [CrossRef] [PubMed]
  193. Xie, W.; Huang, Y.; Xiao, S.; Sun, X.; Fan, Y.; Zhang, Z. Impact of janus kinase inhibitors on risk of cardiovascular events in patients with rheumatoid arthritis: Systematic review and meta-analysis of randomised controlled trials. Ann. Rheum. Dis. 2019, 78, 1048–1054. [Google Scholar] [CrossRef]
  194. Xie, W.; Zhang, Z. Tofacitinib in cardiovascular outcomes: Friend or foe? Rheumatology 2020, 59, 1797–1798. [Google Scholar] [CrossRef] [Green Version]
  195. Rainsford, K.D.; Parke, A.L.; Clifford-Rashotte, M.; Kean, W.F. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology 2015, 23, 231–269. [Google Scholar] [CrossRef] [PubMed]
  196. Wu, C.L.; Chang, C.C.; Kor, C.T.; Yang, T.H.; Chiu, P.F.; Tarng, D.C.; Hsu, C.C. Hydroxychloroquine Use and Risk of CKD in Patients with Rheumatoid Arthritis. Clin. J. Am. Soc. Nephrol. 2018, 13, 702–709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Ben-Zvi, I.; Kivity, S.; Langevitz, P.; Shoenfeld, Y. Hydroxychloroquine: From malaria to autoimmunity. Clin. Rev. Allergy Immunol. 2012, 42, 145–153. [Google Scholar] [CrossRef]
  198. Schrezenmeier, E.; Dörner, T. Mechanisms of action of hydroxychloroquine and chloroquine: Implications for rheumatology. Nat. Rev. Rheumatol. 2020, 16, 155–166. [Google Scholar] [CrossRef] [PubMed]
  199. Shi, N.; Zhang, S.; Silverman, G.; Li, M.; Cai, J.; Niu, H. Protective effect of hydroxychloroquine on rheumatoid arthritis-associated atherosclerosis. Anim. Model Exp. Med. 2019, 2, 98–106. [Google Scholar] [CrossRef]
  200. Hu, C.; Lu, L.; Wan, J.P.; Wen, C. The Pharmacological Mechanisms and Therapeutic Activities of Hydroxychloroquine in Rheumatic and Related Diseases. Curr. Med. Chem. 2017, 24, 2241–2249. [Google Scholar] [CrossRef]
  201. Fan, H.W.; Ma, Z.X.; Chen, J.; Yang, X.Y.; Cheng, J.L.; Li, Y.B. Pharmacokinetics and Bioequivalence Study of Hydroxychloroquine Sulfate Tablets in Chinese Healthy Volunteers by LC-MS/MS. Rheumatol. Ther. 2015, 2, 183–195. [Google Scholar] [CrossRef] [Green Version]
  202. 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]
  203. Eugenia Schroeder, M.; Russo, S.; Costa, C.; Hori, J.; Tiscornia, I.; Bollati-Fogolín, M.; Zamboni, D.S.; Ferreira, G.; Cairoli, E.; Hill, M. Pro-inflammatory Ca++-activated K+ channels are inhibited by hydroxychloroquine. Sci. Rep. 2017, 15, 1892. [Google Scholar] [CrossRef]
  204. Silva, J.C.; Mariz, H.A.; Rocha, L.F., Jr.; Oliveira, P.S.; Dantas, A.T.; Duarte, A.L.; 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]
  205. Mercer, E.; Rekedal, L.; Garg, R.; Lu, B.; Massarotti, E.M.; Solomon, D.H. Hydroxychloroquine improves insulin sensitivity in obese non-diabetic individuals. Arthritis Res. Ther. 2012, 14, R135. [Google Scholar] [CrossRef] [Green Version]
  206. 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] [Green Version]
  207. Wasko, M.C.; Hubert, H.B.; Lingala, V.B.; Elliott, J.R.; Luggen, M.E.; Fries, J.F.; Ward, M.M. Hydroxychloroquine and risk of diabetes in patients with rheumatoid arthritis. JAMA 2007, 298, 187–193. [Google Scholar] [CrossRef] [Green Version]
  208. Guevara, M.; Ng, B. Positive effect of hydroxychloroquine on lipid profiles of patients with rheumatoid arthritis: A Veterans Affair cohort. Eur. J. Rheumatol. 2020, 8, 62–66. [Google Scholar] [CrossRef] [PubMed]
  209. Mangoni, A.A.; Woodman, R.J.; Piga, M.; Cauli, A.; Fedele, A.L.; Gremese, E.; Erre, G.L.; EDRA Study Group. Patterns of Anti-Inflammatory and Immunomodulating Drug Usage and Microvascular Endothelial Function in Rheumatoid Arthritis. Front. Cardiovasc. Med. 2021, 8, 681327. [Google Scholar] [CrossRef]
  210. Kedia, A.K.; Mohansundaram, K.; Goyal, M.; Ravindran, V. Safety of long-term use of four common conventional disease modifying anti-rheumatic drugs in rheumatoid arthritis. J. R. Coll. Physicians Edinb. 2021, 51, 237–245. [Google Scholar] [CrossRef] [PubMed]
  211. Doyno, C.; Sobieraj, D.M.; Baker, W.L. Toxicity of chloroquine and hydroxychloroquine following therapeutic use or overdose. Clin. Toxicol. 2021, 59, 12–23. [Google Scholar] [CrossRef] [PubMed]
  212. Rempenault, C.; Combe, B.; Barnetche, T.; Gaujoux-Viala, C.; Lukas, C.; Morel, J.; Hua, C. Metabolic and cardiovascular benefits of hydroxychloroquine in patients with rheumatoid arthritis: A systematic review and meta-analysis. Ann. Rheum. Dis. 2018, 77, 98–103. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pro-atherogenic disordered lipid transport and processing in RA. Both low density lipoprotein (LDL) and high density lipoprotein (HDL) are impacted by the inflammatory milieu in RA. Disruptions in lipid composition and transport observed in RA include (1) Citrullination. Citrullinated and homocitrullinated forms of LDL have atherogenic properties and may be abundant in RA plasma. (2) HDL abnormalities. Circulating levels of HDL may be low and paraoxonase 1 (PON1) activity of HDL impaired. Poorly functioning PON1 reduces antioxidant capacity of HDL. (3) Enhanced foam cell formation. Downregulation of cholesterol efflux proteins coupled with upregulation of scavenger receptors attenuates cholesterol outflow and increases oxidized LDL uptake within macrophages leading to lipid overload and formation of foam cells. TNF-α increases levels of oxidatively modified LDL via augmentation of reactive oxygen species generation.
Figure 1. Pro-atherogenic disordered lipid transport and processing in RA. Both low density lipoprotein (LDL) and high density lipoprotein (HDL) are impacted by the inflammatory milieu in RA. Disruptions in lipid composition and transport observed in RA include (1) Citrullination. Citrullinated and homocitrullinated forms of LDL have atherogenic properties and may be abundant in RA plasma. (2) HDL abnormalities. Circulating levels of HDL may be low and paraoxonase 1 (PON1) activity of HDL impaired. Poorly functioning PON1 reduces antioxidant capacity of HDL. (3) Enhanced foam cell formation. Downregulation of cholesterol efflux proteins coupled with upregulation of scavenger receptors attenuates cholesterol outflow and increases oxidized LDL uptake within macrophages leading to lipid overload and formation of foam cells. TNF-α increases levels of oxidatively modified LDL via augmentation of reactive oxygen species generation.
Pharmaceuticals 15 00011 g001
Figure 2. Shared inflammatory pathways are fundamental in the development of rheumatoid synovitis and ASCVD. The initial phase involves endothelial dysfunction which promotes inflammatory cell infiltration within the joint capsule and the inception of plaque formation within the sub-intima of arteries. The reduction in anti-oxidative capacity in RA patients not only accelerates joint tissue damage, but also promotes LDL oxidation and foam cell formation. Increased matrix metalloproteases (MMPs) evident in RA plasma then stimulates cartilage degradation in the joint and initiate deterioration of the fibrous cap surrounding the atherosclerotic plaque. This encourages plaque instability and eventual rupture which can lead to acute myocardial infarctions and strokes, depending on vessel location.
Figure 2. Shared inflammatory pathways are fundamental in the development of rheumatoid synovitis and ASCVD. The initial phase involves endothelial dysfunction which promotes inflammatory cell infiltration within the joint capsule and the inception of plaque formation within the sub-intima of arteries. The reduction in anti-oxidative capacity in RA patients not only accelerates joint tissue damage, but also promotes LDL oxidation and foam cell formation. Increased matrix metalloproteases (MMPs) evident in RA plasma then stimulates cartilage degradation in the joint and initiate deterioration of the fibrous cap surrounding the atherosclerotic plaque. This encourages plaque instability and eventual rupture which can lead to acute myocardial infarctions and strokes, depending on vessel location.
Pharmaceuticals 15 00011 g002
Table 1. Therapies Effective Against RA and ASCVD.
Table 1. Therapies Effective Against RA and ASCVD.
DMARDs
Name of DrugMechanism of Action
Anti-rheumatic Properties Atheroprotective Properties
MethotrexateInhibits dihydrofolate reductase
and several immune pathways involved in purine and pyrimidine synthesis.
Enhances macrophage cholesterol efflux and prevents foams cell differentiation and activation. Upregulates free radical scavenging; improves
endothelial function.
SulfasalazineReduces production of inflammatory cytokines, likely through
inhibition of NF-κB activation.
Prevents arachidonic acid-mediated platelet aggregation, decreases adhesion of monocytes and leukocytes, and increases HDL-C.
HydroxychloroquineInterferes with toll-like receptor signaling, reduces calcium signaling in B and T cells and matrix metalloprotease activityPositively impacts insulin sensitization, promotes anti-atherogenic lipid profile. Anti-thrombotic and anticoagulant properties.
Tumor Necrosis Factor (TNF)-α Inhibitors
Etanercept
Infliximab
Adalimumab
Biologics that inactivate TNF-α.
Etanercept is a fusion protein of human immunoglobulin 1 Fc domain and TNF-α receptor.
Infliximab is a mouse-human chimeric anti-human TNF-α antibody
Adalimumab is a human anti-human TNF-α antibody
TNF-α promotes numerous inflammatory responses associated with atherosclerosis, including induction of vascular adhesion and monocyte/macrophage proliferation. TNF-α impacts lipid metabolism by stimulating liver triglyceride production.
IL-6 Inhibitors
TocilizumabInhibits IL-6 which contributes to
inflammation and antibody production through its action on T cells, B cells, monocytes and neutrophils
Decreases inflammatory proteins such as serum amyloid A, and restores the anti-atherogenic function of HDL by increasing HDL cholesterol efflux capacity.
JAK Kinase Inhibitors
TofacitinibSmall molecules that target the JAK-STAT signaling pathway. Reduce expression of cytokine related genes.Risk of adverse cardiovascular events still being evaluated. Many studies show no difference compared to placebo or biologic
UpadacitinibMore JAK1 selective
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

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 2022, 15, 11. https://0-doi-org.brum.beds.ac.uk/10.3390/ph15010011

AMA Style

Ahmed S, Jacob B, Carsons SE, De Leon J, Reiss AB. Treatment of Cardiovascular Disease in Rheumatoid Arthritis: A Complex Challenge with Increased Atherosclerotic Risk. Pharmaceuticals. 2022; 15(1):11. https://0-doi-org.brum.beds.ac.uk/10.3390/ph15010011

Chicago/Turabian Style

Ahmed, Saba, Benna Jacob, Steven E. Carsons, Joshua De Leon, and Allison B. Reiss. 2022. "Treatment of Cardiovascular Disease in Rheumatoid Arthritis: A Complex Challenge with Increased Atherosclerotic Risk" Pharmaceuticals 15, no. 1: 11. https://0-doi-org.brum.beds.ac.uk/10.3390/ph15010011

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