IMR Press / FBL / Volume 25 / Issue 2 / DOI: 10.2741/4805
Review
Anti-inflammatory therapy in atherosclerosis
Show Less
1 Cardiology Clinics, Afyonkarahisar Dinar State Hospital, 03400, Afyonkarahisar, Turkey
2 Department of Basic Medical Sciences, Adnan Menderes University Faculty of Medicine, 09010, Aydin, Turkey
3 Department of Cardiology, Hacettepe University Faculty of Medicine, 06100, Ankara, Turkey
Send correspondence to: Duygu Kocyigit, Cardiology Clinics, Afyonkarahisar Dinar State Hospital, 03400, Afyonkarahisar, Turkey, Tel: 0090-272-3536099, Fax: 0090-272-3534017, E-mail: kocyigitduygu@yahoo.com
Front. Biosci. (Landmark Ed) 2020, 25(2), 242–269; https://doi.org/10.2741/4805
Published: 1 January 2020
(This article belongs to the Special Issue New trends in atherosclerosis research)
Abstract

Cardiovascular disease (CVD) is the leading cause of death. Although once considered merely as a lipid storage disease, studies indicate the role of inflammation in initiation and progression of atherosclerotic CVD, as well as the development of thrombotic complications. Despite significant advances in treatment of atherosclerosis, there still exists a residual risk for CVD-related morbidity and mortality. Even with optimal treatment, the rate of a new event after an index acute coronary syndrome event, such as myocardial ischemia or infarction, in the first three years has been reported to be as high as 20%. In the last decades, inflammation due to apoB-lipoproteins and other traditional risk factors, such as hypertension, diabetes and smoking, is accepted as a new target for CVD prevention. Up to now, several anti-inflammatory drugs have been tested for use in atherosclerosis. This review focuses on the current status of anti-inflammatory drug therapy for atherosclerotic CVD in humans.

Keywords
Inflammation
Atherosclerosis
Human
Clinical Trials
Review
2. INTRODUCTION

Cardiovascular disease (CVD) is the leading cause of death (1). According to 2015 World Health Organization data, CVD mortality accounts for 31% of all deaths worldwide (1). The most common form of CVD is atherosclerosis. Although once considered merely as a lipid storage disease, studies indicate the role of inflammation in the initiation and progression of the atherosclerotic CVD, as well as the development of thrombotic complications.

It has been believed that the main initiators of inflammation in atherosclerotic CVD are apolipoprotein B-containing lipoproteins (apoB-LPs). Besides causing inflammation in plaques, they also cause progression of the disease via interacting with other traditional CVD risk factors, such as hypertension, diabetes and smoking (2). Despite significant advances in treatment of atherosclerosis, there still exists a residual risk for CVD-related morbidity and mortality. Even with optimal treatment, the rate of a new event after an index acute coronary syndrome (ACS) event, such as myocardial ischemia or infarction, in the first three years has been reported to be as high as 20% (3). In the last decades, inflammation due to apoB-LPs and other traditional risk factors, such as hypertension, diabetes and smoking, is accepted as a new target for CVD prevention. Up to now, several anti-inflammatory drugs have been tested for use in atherosclerotic CVD.

Randomized clinical trials (RCTs) have demonstrated that in addition to their lipid lowering effects, statins are useful in the primary (4) and secondary (5) prevention from CVD due to their anti-inflammatory potential. The landmark Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) trial has suggested that in normolipidemic subjects (low-density lipoprotein [LDL]-cholesterol< 130 mg/dL) without evident coronary heart disease but with elevated C-reactive protein (CRP) levels (> 2.0 mg/L), suppression of low-grade inflammation by rosuvastatin 20 mg/day resulted in a risk reduction of 0.56 (95% CI: 0.46-0.69, p<0.00001) for the combined primary endpoint when compared to placebo at a median follow-up of 1.9 years(6). A recent plaque imaging study in 218 patients using intravascular ultrasound has shown that after 12-month rosuvastatin treatment a greater high sensitivity-CRP (hs-CRP) reduction, but not LDL-cholesterol, was associated with a greater decrease in percent necrotic core volume and absence of intravascular ultrasound defined thin-cap fibroatheroma, suggesting a link between the anti-inflammatory action of statins and plaque stabilization (7). This review focuses on the current status of anti-inflammatory drug therapy other than statins in atherosclerotic CVD in humans.

3. ANTI-INFLAMMATORY DRUGS
3.1. Anti-inflammatory drugs with low probability to improve cardiovascular outcomes
3.1.1. Corticosteroids

Corticosteroids are among the most prescribed anti-inflammatory drugs. They have a wide spectrum of anti-inflammatory actions, including increasing transcription of genes coding anti-inflammatory proteins (such as lipocortin-1, interleukin-10, interleukin-l receptor antagonist), inhibiting expression of various inflammatory genes (such as cytokines, enzymes, receptors and adhesion molecules)- possibly via activated transcription of nuclear factor-κB, activator protein-1- and directly inhibiting inflammatory cells (such as macrophages, dendritic cells, mast cells) (8). Glucocorticoids (GCs) were already established therapeutic agents in various inflammatory diseases prior to introduction of RCTs. Therefore, their effects on cardiovascular (CV) outcomes have been evaluated in epidemiological studies only.

Increased risk of death from myocardial infarction (MI) or stroke at early ages before surgical treatment of Cushing’s syndrome has supported a link between accelerated atherosclerosis and long-term excessive GC exposure (9). Observational studies suggested that rheumatoid arthritis (RA) and systemic lupus erythematosus patients under corticosteroids have significantly more atherosclerosis than those not treated with steroids and the risk of atherosclerosis is related to the cumulative exposure to corticosteroids (10-12). However, extent of inflammation is an important confounder in atherosclerosis associated with rheumatologic disease. This limitation could also apply to population-based observational studies, which were designed to assess the relationship between GC therapy and CV events (13, 14), owing to the fact that increased odds ratio (OR) of any CV or cerebrovascular outcome may be attributable to the underlying disease being treated rather than to the use of GCs. However, in both studies (13, 14), effects wereconsistent across different disease indications.

Nevertheless, in other studies involving cohorts of RA patients, there was evidence of interactions between effects of GCs and the underlying inflammatory disease on CV risk. In an US cohort (n= 603), a dose-dependent risk of CVD with GC exposure was confirmed, but that risk was only detectable among rheumatoid factor-positive patients (15). Other studies, which had relatively smaller study populations, revealed contradictory findings (12, 13). Adjustment for the presence of components of the metabolic syndrome did not eliminate the effect of GCs (13, 14, 16), raising the possibility that the influence of GCs on CV outcomes is not mediated exclusively by traditional CV risk factors.

Various clinical trials have been undertaken to test the effects of GCs administered in the first days after MI. A meta-analysis of 11 controlled trials (n= 2.646) (18) has revealed a 26% decrease in mortality with corticosteroids (OR: 0.74, 95% CI: 0.59-0.94). Sensitivity analyses limited to large studies and RCTs revealed ORs of 0.76 (95% CI: 0.53-1.09) and 0.95 (95% CI: 0.72-1.26), respectively, suggesting that corticosteroids at least did not cause any harm in acute MI. However, the increased incidence of heart failure amongst GC users in the epidemiological studies described above (13, 14) raises the possibility that longer-term GC therapy has an adverse effect on cardiac remodelling.

Corticosteroids have also been used to prevent restenosis after angioplasty. Systemic GCs have been beneficial in some (19-22), but not all (23-27) clinical studies. Discrepancy in the results may be explained by the fact that the benefits of local anti-inflammatory effects may be offset by adverse systemic effects. Second, the anti-inflammatory effects of GR activation may be offset by pro-inflammatory MR activation (28). To overcome these limitations, GC-eluting stents have been proposed to deliver local anti-inflammatory therapy (29-31) using GCs devoid of affinity for MR, such as dexamethasone and prednisolone. However, there still exists the risk that GR activation may induce changes within the vessel which may offset any benefit of conventional anti-inflammatory effects, such as increasing local angiotensin-II (32, 33) or endothelin-1 (34) generation or decreasing endothelial nitric oxide generation (35).

3.1.2. Pexelizumab

The classical complement pathway, which is one of three pathways that activate the complement system, may directly damage the myocardium (36). Complement 5 inhibitor, pexelizumab, is a humanized monoclonal antibody inhibiting C5 complement, thereby blocking its cleavage into active forms: C5a and C5b-9. C5a is a powerful peptide mediator of inflammation, whereas C5b triggers the late events in which the terminal components of complement assemble into a membrane-attack complex that can damage the membrane of certain pathogens (37).

In a systemic review and meta-analysis of seven trials (n= 15.196, 7.019 patients with ST segment elevation MI (STEMI) and 8.177 undergoing coronary artery bypass grafting (CABG), the impact of pexelizumab versus placebo on CV outcomes was investigated. Endpoints were the composite risk of all-cause death, MI and thromboembolic stroke; the risk of single end points and heart failure. There was not any benefit of pexelizumab on the endpoints in STEMI, however it was associated with a 26% reduction of death risk in the setting of CABG (OR: 0.74, 95% CI: 0.58-0.94, p= 0.01) (38).

3.1.3. Tranilast

Mast cells stabilizers block mast cell degranulation, thereby preventing the release of histamine and related mediators. Tranilast is an anti-fibrotic drug which prevents collagen synthesis by inhibiting the release of mast cell factors, including histamine, prostaglandins, platelet derived growth factor, transforming growth factor-beta and interleukin (IL)-1beta (39). Three RCTs evaluating the impact of tranilast on CV outcomes are summarized in Table 1. The largest trial, Prevention of REStenosis with Tranilast and its Outcomes (PRESTO), failed to demonstrate beneficial effects of tranilast on CV outcomes (40).

Table 1 Summary of three randomized controlled trials that evaluate the impact of tranilast on cardiovascular outcomes
Trial Study protocol Outcomes Results
The Tranilast REstenosis following Angioplasty Trial (TREAT) (137) - Double-blind, placebo-controlled trial - n= 255 patients -Randomization: 600 mg/day tranilast po, 300 mg/day tranilast po or placebo po for 3 months after successful PTCA Restenosis rates after successful PTCA - loss of ≥50% of the initial gain - % stenosis of ≥50% at follow-up - 14.7% in the 600 mg/day tranilast group, 35.2% in the 300 mg/day tranilast group and 46.5% in the placebo group (p< 0.0001 for 600 mg/day tranilast vs placebo) - 17.6% in the 600 mg/day tranilast group, 38.6% in the 300 mg/day tranilast group, and 39.4% in the placebo group (p= 0.005 for 600 mg/day tranilast vs placebo)
TREAT-2 (138) - Double-blind, placebo-controlled trial - n= 297 patients - Randomization: 600 mg/day tranilast po or placebo po for 3 months after successful PTCA Restenosis rates after successful PTCA - loss of ≥50% of the initial gain - % stenosis of ≥50% at follow-up - 18.8% in the tranilast group vs. 44.1% in the placebo group, p<0.001 -- in de novo lesions: 18.9% in the tranilast group vs. 41.2% in the placebo group, p= 0.003 -- in restenotic lesions: 18.4% in the tranilast group vs. 53.3% in the placebo group, p= 0.004 - 25.9% in the tranilast group vs. 41.9% in the placebo group, p= 0.012
Prevention of REStenosis with Tranilast and its Outcomes (PRESTO) trial (40) - Double-blind, placebo-controlled trial - n= 11.484 patients - Randomization: 300 and 450 mg twice daily tranilast or placebo for 1 or 3 months within 4 hours after successful percutaneous coronary intervention of at least 1 vessel - Primary endpoint: the first occurrence of death, MI or ischemia-driven target vessel revascularization within 9 months - In the angiographic substudy (n= 2.018 patients): MLD measured by quantitative coronary angiography -- a subset of patients (n= 1.107) underwent intravascular ultrasound at follow-up - 15.8% in the placebo group, 15.5% to 16.1% in the tranilast groups (p= 0.77 and p= 0.81) - MLD was 1.76±0.77 mm in the placebo group; 1.72±0.76, p= 0.49 and 1.78±0.80 mm, p= 0.89 in the tranilast groups -- plaque volume was 39.3 mm3 in the placebo and 37.5 and 46.1 mm3 in the tranilast groups (p= 0.16 and p= 0.72, respectively)
CI confidence interval, MI myocardial infarction, MLD minimal lumen diameter, PTCA percutaneous transluminal coronary angioplasty.
3.1.4. Anti-tumor necrosis factor (anti-TNF) therapy

Anti-tumor necrosis factor (anti-TNF) therapy is a widely used biological therapy of several immune-mediated inflammatory diseases such as RA, Crohn’s disease, psoriatic arthritis, ankylosing spondylitis, ulcerative colitis and severe chronic plaque psoriasis. Infliximab, etanercept and adalimumab are the most commonly used anti-TNF treatments. Infliximab is a recombinant humanized monoclonal anti-TNF-alpha antibody, whereas etanercept binds and inactivates TNF. Adalimumab is also a recombinant human monoclonal anti-TNF-alpha antibody; in addition, it lyses surface TNF-expressing cells in vitro and modulates biologic responses responsible for leukocyte migration.

Data from the South Swedish Arthritis Treatment Group (SSATG) has shown that RA patients treated with etanercept or infliximab had a reduced incidence of first CV event compared to patients not exposed to anti-TNF (14.0/1000 person-years at risk, 95% CI: 5.7-22.4 vs. 35.4/1000 person-years, 95% CI: 16.5-54.4), but not overall events (41). The analysis of patients from a different register, the British Society for Rheumatology Biologics Register (BSRBR) has shown no reduction in the incidence of MI in 8.670 patients treated with anti-TNF when compared to 2.170 patients treated with standard disease modifying anti-rheumatic drugs (DMARDs) (incidence rate ratio: 1.44, 95% CI: 0.56-3.67) (42). However, responders to anti-TNF therapy in the first 6 months had a decrease in MI compared to non-responders (adjusted incidence rate ratio: 0.36, 95% CI: 0.19-0.69), which suggests that inflammation control may be associated with lowering the CV burden in these patients. Data from 10.156 RA patients enrolled in the Consortium of Rheumatology Researchers of North America RA registry has shown that patients under anti-TNF therapy had a reduced risk of the primary composite CV endpoint compared with users of non-biological DMARDs (hazards ratio [HR]: 0.39, 95% CI 0.19-0.82). The risk reduction associated with TNF antagonists was also observed for non-fatal CV events (HR: 0.35, 95% CI 0.16-0.74) (43). Another study has compared the composite CV endpoint of MI, stroke or coronary revascularization at 6 months between 11.587 new users of anti-TNF therapy and 8.656 new users of a non-biological DMARDs. Incidence rates per 100 person-years for the composite CV end point were 2.52 (95% CI: 2.12-2.98) for TNF-α blocking agents and 3.05 (95% CI: 2.54-3.65) for DMARDs (44).

The Pilot Study on the Effect of Adalimumab on Vascular Inflammation in Patients with Moderate to Severe Plaque Psoriasis has sought to determine the effect of adalimumab on inflammation of blood vessels. This RCT included 30 patients with moderate-severe psoriasis and a history or multiple risk factors of coronary atherosclerosis. Patients were randomized to receive either adalimumab subcutaneously for 4 months or control non-systemic treatment (topical therapies or phototherapy). Vascular inflammation was measured in the carotid artery and ascending aorta at baseline and week 15 by 18F-fluorodeoxyglucose uptake on positron emission tomography. However, the change in the FDG measurement (target:background ratio) in patients randomized to adalimumab was similar to that in controls (45).

3.1.5. Phospholipase A2 (PLA2) inhibitors

Phospholipase A2 (PLA2) inhibitors are inhibitors of phospholipases, the enzymes that promote inflammation by producing precursors of arachidonic acid from membrane glycerophospholipids (46). There is a soluble (sPLA2) and a lipoprotein-associated form (Lp-PLA2). Circulating levels and enzymatic activity of these two families of PLA2 enzymes have been evaluated as biomarkers of CV risk in population-based studies including subjects with and without established coronary heart disease (47, 48). Darapladib inhibits Lp-PLA2 activity, whereas varespladib is an inhibitor of sPLA2. Three RCTs evaluating the impact of PLA2 inhibitors on CV outcomes are summarized in Table 2. However, these agents failed to demonstrate beneficial effects on the primary composite endpoint of CV death, MI and stroke. Indeed, sPLA2 inhibition with varespladib has been shown to be harmful after ACS (49).

Table 2 Summary of three randomized controlled trials that evaluate the impact of phospholipase A2 inhibitors on cardiovascular outcomes
Trial Study protocol Median follow-up Outcomes Results
The Stabilization Of pLaques usIng Darapladib-Thrombolysis In Myocardial Infarction 52 (SOLID-TIMI-52) trial (139) - Double-blind, placebo-controlled trial - n= 13.026 patients - Randomization: 160 mg/day darapladib po or placebo po within 30 days of hospitalization with MI 2.5 years - Primary endpoint: the composite of CV death, MI or urgent coronary revascularization for myocardial ischemia - The composite of CV death, MI or stroke - in 903 patients in the darapladib group (16.3%) and 910 in the placebo group (15.6%), HR: 1.00 (95% CI, 0.91-1.09), p= 0.93 - in 824 patients in the darapladib group (15.0%) and 838 in the placebo group (15.0%), HR: 0.99 (95% CI, 0.90-1.09), p= 0.78
The Stabilization of Atherosclerotic Plaque by Initiation of Darapladib Therapy (STABILITY) trial (140) - Double-blind, placebo-controlled trial - n= 15.828 patients (with at least one of the following: previous MI, previous PCI or CABG, or multivessel coronary artery disease - Randomization: 160 mg/day darapladib po (median drug exposure: 3.5 years) or placebo po (median drug exposure: 3.6 years) 3.7 years - Primary endpoint: the composite of CV death, MI or stroke - Secondary endpoints: the components of the primary endpoint as well as major coronary events (death from coronary heart disease, MI or urgent coronary revascularization for myocardial ischemia) and total coronary events (death from coronary heart disease, MI, hospitalization for unstable angina, or any coronary revascularization) - in 769 of 7.924 patients (9.7%) in the darapladib group and 819 of 7.904 patients (10.4%) in the placebo group, HR: 0.94, (95% CI, 0.85-1.03), p= 0.20 - no significant between-group differences in the rates of the individual components of the primary endpoint or in all-cause mortality - reduction in the rate of major coronary events: 9.3% in the darapladib group vs. 10.3% in the placebo group, HR: 0.90, (95% CI, 0.82-1.00), p= 0.045 - reduction in the rate of total coronary events: 14.6% in the darapladib group vs. 16.1% in the placebo group, HR: 0.91, (95% CI: 0.84-0.98), p= 0.02
The Evaluation of Safety and Efficacy of Short-term A-002 Treatment in Subjects with Acute Coronary Syndrome (VISTA-16) trial (47) - Double-blind, placebo-controlled trial - Halted prematurely. Aimed to randomize 5.145 patients within 96 hours of presentation of an acute coronary syndrome. - Randomization: 500 mg/day varespladib po or placebo po for 16 weeks - Halted prematurely at a pre-specified interim analysis, including 212 primary endpoint events. - Primary endpoint: the composite of CV mortality, non-fatal MI, nonfatal stroke or unstable angina with evidence of ischemia requiring hospitalization at 16 weeks - Secondary endpoint: the composite of CV mortality, MI and stroke - in 136 patients (6.1%) in the varespladib group and in 109 patients (5.1%) in the placebo group, HR: 1.25, (95% CI, 0.97-1.61), p= 0.08 - in 107 patients (4.6%) in the varespladib group and in 79 patients (3.8%) in the placebo group, HR: 1.36, (95% CI, 1.02-1.82), p= 0.04
CABG coronary-artery bypass grafting, CI confidence interval, CV cardiovascular, MI myocardial infarction, PCI percutaneous coronary intervention.
3.1.6. Losmapimod

p38 mitogen-activated protein kinase (MAPK) inhibitors inhibit the p38 MAPK, which is activated in the CV system by a variety of stressors, including oxidized LDL-cholesterol, hypertension, ischemia and volume overload (50). Selective inhibitors of p38 MAPK have been shown to inhibit lipopolysaccharide-stimulated IL-1 and TNF-alpha production in human monocytes and the production of several other cytokines, including IL-6, IL-8, and granulocyte-macrophage colony-stimulating factor (51, 52). Losmapimod is a selective, reversible, competitive inhibitor of p38 MAPK with onset as early as 30 minutes after oral dosing. RCTs evaluating the impact of p38 MAPK inhibitors on CV outcomes are summarized in Table 3.

Table 3 Summary of randomized controlled trials that evaluate the impact of p38 mitogen-activated protein kinase inhibitors on cardiovascular outcomes
Trial Study protocol Outcomes Results
A Study to Evaluate the Safety of 12 Weeks of Dosing with GW856553 and Its Effects on Inflammatory Markers, Infarct Size, and Cardiac Function in Subjects with Myocardial Infarction without ST-segment Elevation (SOLSTICE) trial (141) - Double-blind, placebo-controlled trial - n= 526 NSTEMI patients - Randomization: losmapimod po (7.5 mg or 15 mg loading dose followed by 7.5 mg twice daily) or matching placebo - Primary endpoints: hsCRP and BNP concentrations at 72 h and 12 weeks and troponin I AUC over 72 h - Secondary inflammation measures: hsCRP concentrations at 14 weeks and IL-6 concentrations at 24 h and 12 weeks. - Secondary infarct size measures: CK-MB AUC and peak troponin I concentration over 72 h or discharge - The primary substudy endpoints: infarct size (% of the left ventricle) on delayed-enhancement MRI and LV ejection fraction at days 3-5 and week 12 - Secondary MRI endpoints: LV end-diastolic and end-systolic volumes at days 3-5 and week 12 - geometric mean hsCRP concentrations at 72 h: lower in the losmapimod group than in the placebo group (64.1 nmol/L, 95% CI 53.0–77.6 vs 110.8 nmol/L, 95% CI: 83.1–147.7, p=0.0009) - hsCRP concentrations at 12 weeks: similar - geometric mean BNP concentrations at 72 h: similar - geometric mean BNP concentrations at 12 weeks: lower in the losmapimod group (37.2 ng/L, 95% CI 32.3–42.9 vs 49.4 ng/L, 38.7–63.0, p=0.04) - reduced geometric mean IL-6 at 72 hours in the losmapimod group (6.6 ng/L, 95% CI: 5.8-7.4 vs. 10.6 ng/L, 95% CI: 8.6-13.1, p< 0.001), but similar in the losmapimod and placebo groups at 12 weeks - CK-MB AUC over 72 h and peak troponin I at 72 h or discharge similar in the losmapimod and placebo groups - similar infarct size in the losmapimod and placebo groups - higher mean LVEF both at days 3-5 (56.86% vs. 52.13%, p= 0.05) and week 12 (60.28% vs. 55.14%, p= 0.039) in the losmapimod group - lower mean LV end-diastolic and end- systolic volumes both at days 3-5 (127.18 mL vs. 147.06 mL, p= 0.039 and 56.13 mL vs. 71.65 mL, p= 0.023) and week 12 (127.74 mL vs. 147.83 mL, p= 0.020 and 51.47 mL vs. 67.99 mL, p= 0.010) in the losmapimod group
LosmApimod To Inhibit p38 MAP Kinase as a TherapeUtic Target and moDify Outcomes After an Acute Coronary syndromE (LATITUDE)-TIMI 60 trial (142) - Double-blind, placebo-controlled, parallel-group trial - Part A: a leading cohort (n= 3.503) to provide an initial assessment of safety and exploratory efficacy before considering progression to part B Part B: approximately 22.000 patients - Subjects hospitalized with an acute MI and had at least 1 additional predictor of CV risk - Randomization: twice-daily losmapimod (7.5 mg) or matching placebo for 12 weeks Primary endpoint: the composite of CV death, MI or severe recurrent ischemia requiring urgent coronary revascularization at week 12. - In part A, the primary endpoint occurred by 12 weeks in 123 patients treated with placebo (7.0%) and 139 patients treated with losmapimod (8.1%) (HR: 1.16; (95% CI, 0.91-1.47), p=<0.24). The on-treatment rates of serious adverse events were 16.0% with losmapimod and 14.2% with placebo. - The results of part A did not justify proceeding to part B
AUC area under the curve, BNP B-type natriuretic peptide, CI confidence interval, CK-MB creatine kinase-myocardial band, CV cardiovascular, hsCRP high-sensitivity C-reactive protein, IL interleukin, LV left ventricular, MI myocardial infarction, MRI magnetic resonance imaging, NSTEMI non-ST segment elevation myocardial infarction.
3.1.7. Salsalate

Salsalate is a prodrug dimer of salicylates marketed for relief of arthritic pain. Anti-inflammatory actions of salicylates mainly focus on their ability to inhibit the proinflammatory kinase I kappa B kinase (IKK) (53), which activates the pro-inflammatory and pro-atherosclerotic transcription mediator NF-κB (54-55). However, its anti-inflammatory effects independent of NF-κB have also been shown (56-59).

The Targeting Inflammation Using Salsalate in Cardiovascular Disease (TINSAL-CVD) trial is a double-blind, placebo-controlled trial that randomized 257 overweight and obese statin-using patients with established, stable coronary heart disease to salsalate (3.5 g/day) (n= 129) or placebo (n= 128) orally over 30 months. The primary endpoint was progression of noncalcified coronary artery plaque assessed by multidetector computed tomographic angiography. 190 participants (n= 89 in the salsalate group, n=101 in the placebo group) completed the study. Compared with baseline, there was no increase in non-calcified plaque volume in the placebo-treated patients and no difference in change between the salsalate and placebo groups (mean difference: −1 mm3, 95% CI,: −11 to 9 mm3, p=<0.87). The absence of progression of noncalcified plaque volume in the placebo group has been suggested to limit interpretation of the trial results (60).

3.1.8. Interleukin-1 receptor antagonists (IL-1Ra)

Interleukin-1 receptor antagonists (IL-1Ra) block the signaling receptor that binds either IL-1 alpha (IL-1ɑ) or beta (IL-1β). There is a wide spectrum for the role of IL-1 in CVD, such as activation of endothelial cells (61) and smooth muscle cell proliferation (62), and IL-6 secretion that in turn promotes thrombosis and limits fibrinolysis (63). Taking the human genetic studies revealing a causal link between IL-6 and coronary heart disease into account (64, 65), IL-1, as an inducer of IL-6, has considerable importance in atherothrombosis pathogenesis.

Anakinra is a short-acting recombinant IL-1Ra that blocks both IL-1ɑ and IL-1β signalling via binding to the IL-1 type I receptor. It is currently approved for use in RA, cryopyrin-associated periodic syndromes and Still disease. MRC-ILA Heart Study has been undertaken to examine the effect of anakinra on hsCRP levels in patients with NSTE-ACS (66). In this phase II, double-blinded, placebo-controlled study, 182 patients were randomized within <48 h from onset of chest pain to either daily subcutaneous anakinra or placebo for 14 days. Treatment compliance was 85% at 7 days. The primary endpoint (area under curve [AUC] for CRP over the first 7 days) was lower in the anakinra group (geometric mean ratio= 0.51 mg/L, 95% CI 0.32–0.79, p= 0.003). In the anakinra group, 14-day achieved hsCRP (p< 0.001) and IL-6 levels (p= 0.02) were lower than day 1, however on day 30 (16 days after discontinuation of the treatment) hsCRP levels rose again (IL-1ra group:; 3.50 mg/L [2.65–4.62], placebo group: 2.21 mg/L [1.67–2.92], p= 0.022). Major adverse CV events at day 30 and 3 months were similar but at 1 year there was a significant excess of events in the anakinra group (66). This finding may be supported by a genome-wide association study on genetic variants upstream of IL1RN, which is the gene encoding the IL-1Ra, showing that dual IL-1α/β inhibition provided by IL-1Ra could increase CV risk, probably due to an increase in pro-atherogenic lipid concentrations (67).

Following two pilot studies undertaken in patients with STEMI (68, 69), the extended follow-up data of these 40 patients with STEMI randomized to anakinra 100 mg/day for 14 days or placebo has been published. Endpoints included death, cardiac death, recurrent acute MI, stroke, unstable angina and symptomatic heart failure. At a median follow-up of 28 months, treatment with anakinra was associated with a HR of 1.08 (95% CI 0.31-3.74, p= 0.90) for the combined endpoint of death, recurrent acute MI, unstable angina pectoris or stroke and a HR of 0.16 (95% CI 0.03-0.76, p= 0.008) for death or heart failure. These data have suggested that IL-1 blockade with anakinra for 2 weeks in STEMI patients may prevent new-onset heart failure in long term after STEMI despite neutral effects on recurrent ischemic events (70).

3.1.9. Methotrexate

Methotrexate (MTX) is a dihydrofolate reductase inhibitor, and when given at high doses, it prevents the synthesis of both purine and pyrimidine nucleotides. Low-dose MTX (LDM) has been suggested to exert atheroprotective effects via various mechanisms. These include inhibition of cytokine production (71), induction of inflammatory cell apoptosis (72), increased adenosine release and subsequent agonism of the adenosine A2A receptor, which in turn induces the expression of key proteins in reverse cholesterol transport (73) and reduces expression of adhesion molecules commonly associated with the development of atheroma, such as vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 (74). Direct anti-inflammatory effects of LDM at the level of the vascular endothelium have also been suggested (75).

Low dose methotrexate (which partially acts on the IL-6 pathway as demonstrated by reduction of IL-6 plasma levels) has been shown to reduce CV events in patients with RA in several studies, where HRs ranged between 0.30 and 0.85 (76-78). The Cardiovascular Inflammation Reduction Trial (CIRT), which is a double-blind, placebo-controlled study, aimed to randomize a total of 7.000 stable coronary artery disease patients with either type 2 diabetes or metabolic syndrome to LDM (15-20 mg/week) or placebo (79). The primary trial endpoint was determined initially as the rate of non-fatal MI, non-fatal stroke or CV death. Near the conclusion of the trial, but before unblinding, hospitalization for unstable angina that led to urgent revascularization was added to the primary end point. The trial was ended prematurely in May 2018 after 4.786 patients of the 7.000 patients were enrolled, not due to any apparent safety concerns but to acquisition of enough data to answer the main question of the study. Results of the trial came out recently in the American Heart Association Scientific Sessions 2018. At a median follow-up of 2.3 years, LDM did not lower IL-1β, IL-6 or CRP levels compared to placebo. The final primary end point occurred in 201 patients in the LDM group and in 207 in the placebo group (incidence rate: 4.13 vs. 4.31 per 100 person-years; HR: 0.96; 95% CI: 0.79-1.16). The original primary end point occurred in 170 patients in the methotrexate group and in 167 in the placebo group (incidence rate: 3.46 vs. 3.43 per 100 person-years; HR: 1.01; 95% CI: 0.82-1.25). Despite the low dose, MTX was associated with a higher incidence of non–basal-cell skin cancers than placebo (31 vs. 10, rate ratio: 3.08, p=0.002) (81), as well as elevations in liver-enzyme levels, reductions in leukocyte counts and hematocrit levels.

3.2. Anti-inflammatory drugs with probability to improve cardiovascular outcomes
3.2.1. Colchicine

Colchicine has been used for gout for at least several hundred years. It downregulates inflammation through blocking microtubule spindle formation, disrupting inflammasome function (particularly NLRP3 inflammasome, a cytosolic complex responsible for the production of IL-1β and IL-18) (82), inhibiting cytokine production (82, 83) and impeding neutrophil chemotaxis (84).

Colchicine has been demonstrated to have no effect on angiographic restenosis in patients undergoing balloon angioplasty (85) but reduce the risk of neo-intimal hyperplasia in diabetic patients undergoing coronary stenting (86). Regarding its effects on ischemia/ reperfusion injury, perioperative oral colchicine has been reported to reduce the levels of troponin and creatine kinase- myocardial band in patients undergoing CABG (87).

The potential benefit of long-term colchicine in patients with stable coronary disease was suggested in retrospective studies in patients with familial Mediterranean fever (88) and gout (89), and was demonstrated prospectively in the LoDoCo trial, which tested the same low dose of colchicine used for secondary prevention in gout (90). In LoDoCo trial, which had a prospective, randomized, observer-blinded endpoint design, 532 patients with stable coronary disease were randomized to either colchicine 0.5 mg/day or no colchicine and followed for a median of 3 years. The primary outcome was the composite incidence of ACS, out-of-hospital cardiac arrest, or non-cardioembolic ischemic stroke. The primary analysis was by intention-to-treat. The primary outcome occurred in 15 of 282 patients (5.3%) who received colchicine and 40 of 250 patients (16.0%) assigned to no colchicine (HR: 0.33; 95% CI 0.18-0.59, p < 0.001). In a pre-specified secondary on-treatment analysis that excluded 32 patients (11%) assigned to colchicine, who withdrew within 30 days due to intestinal intolerance and a further 7 patients (2%) who did not start treatment, the primary outcome occurred in 4.5% vs. 16.0% (HR: 0.29; 95% CI: 0.15- 0.56, p< 0.001) (90).

A meta-analysis of 15 colchicine RCTs (n= 3.431 patients, median treatment 3 months, median follow-up 15 months, 1 mg/day colchicine except for 2 patients) has pointed towards an overall benefit on CV risk reduction, in addition to its ability to reduce pericarditis (85). In 5 trials that included patients with CVD (n= 1.301), colchicine reduced composite CV outcomes (risk ratio: 0.44, 95% CI 0.28-0.69, p< 0.001) and showed a trend towards lower all-cause mortality though not reaching statistical significance (risk ratio: 0.50, 95% CI 0.23-1.08, p=0.08) (91). The potential association between colchicine and CV risk and all-cause mortality has also been evaluated in a population-based case-control study that included subjects diagnosed with gout using data from an electronic medical record database linked with Medicare claims (2006–2011) (92). 501 colchicine users were matched with an equal number of non-users and followed-up at a median of 16.5 months for the primary outcome, a composite of MI, stroke or transient ischemic attack. Incidence rates per 1.000 person-years were 35.6 for users and 81.8 for non-users. After full adjustment, colchicine use was associated with a lower risk (HR 0.51, 95% CI: 0.30–0.88) in the primary CV outcome, as well as a reduction in all-cause mortality (HR 0.27, 95% CI: 0.17–0.43) (92).

Recently, in a prospective, non-randomized study of 80 patients with recent ACS (<1 month), colchicine 0.5 mg/day has been reported to result in a significant reduction of low attenuation plaque volume (LAPV) as assessed by coronary computed tomography angiography (mean 15.9 mm3 (-40.9%) vs. 6.6 mm3 (-17.0%); p= 0.008) at a mean follow-up of 12.6 months. This change in LAPV was positively associated to a reduction in hsCRP levels in those patients receiving colchicine (mean 1.10 mg/L (-37.3%) vs. 0.38 mg/L (-14.6%); p < 0.001). There was also a significant linear association (p< 0.001) and strong positive correlation (r= 0.578) between change in LAPV and hsCRP (93).

The promise of low dose colchicine in patients with coronary artery disease is being explored in two trials. The LoDoCo2 study is designed to determine its effects in ~5.000 patients with stable coronary disease (94) and the COLCOT study of similar size, is examining its effects in patients with recent ACS (95).

3.2.2. Tocilizumab

IL-6 signalling serves as a potential target in coronary atherosclerotic disease. A clear association between IL-6 and atherogenesis has been suggested by a meta-analysis of 82 studies published in 2012 (64). Indeed, it has also been proposed to have a causal role in coronary artery disease (65). Furthermore, IL-6 is involved in ischaemia–reperfusion injury (96), and is associated with increased myocardial injury and mortality in ACS patients (97). Tocilizumab (TCZ) is a monoclonal antibody that competitively inhibits the binding of IL-6 to its receptor. It is currently used in patients with RA, giant cell arteritis and cytokine release syndrome. RCTs evaluating the impact of IL-6 signalling inhibitors on CV outcomes are summarized in Table 4.

Table 4 Summary of randomized controlled trials that evaluate the impact of p38 mitogen-activated protein kinase inhibitors on cardiovascular outcomes
Trial Study protocol Outcomes Results
Effect of a single dose of the interleukin-6 receptor antagonist tocilizumab on inflammation and troponin T release in patients with non-ST-elevation myocardial infarction (143) - Double-blind, placebo-controlled trial - n= 117 patients with NSTEMI (at a median of 2 days after symptom onset) - Randomization: Single dose intravenous 280 mg TCZ (n = 58) or placebo (n = 59), administered prior to coronary angiography - Primary endpoint: AUC for hsCRP at days 1-3 - Secondary endpoint: AUC for hsTnT at days 1-3 - median AUC for hsCRP was lower in the TCZ group (2.0 mg/L/h vs. 4.2 mg/L/h, p< 0.001) - TCZ-induced reduction in hsCRP was more pronounced in patients treated with percutaneous coronary intervention - median AUC for hsTnT was lower in the TCZ group (159 ng/L/h vs. 234 ng/L/h, p= 0.007)
A Clinical Outcomes Study to Evaluate the Effects of IL-6 Receptor Blockade with Tocilizumab in Comparison with Etanercept on the Rate of Cardiovascular Events in Patients with Moderate to Severe Rheumatoid Arthritis (ENTRACTE) (144) - An open-label study - RA patients who newly started TCZ or TNF inhibitor; all patients were required to have previously used a different TNF inhibitor: abatacept or tofacitinib. - Randomization: intravenous 8 mg/kg TCZ every 4 weeks (n= 9.218) or subcutaneous 50 mg TNF inhibitor etanercept weekly (n= 18.810), with or without non-biologic DMARD Primary endpoint: a composite CV endpoint of hospitalization for MI or stroke at a mean study period of 0.9 years 0.52 (95% CI: 0.37-0.71) per 100 person years in the TCZ group and 0.59 (95% CI: 0.47–0.72) per 100 person years for etanercept
The Short-Term Application of Tocilizumab during Myocardial Infarction (STAT-MI) study (145) - Double-blind, placebo-controlled trial - n= 28 patients admitted with MI - Randomization: a single TCZ dose of 162 mg subcutaneously (n= 12) or placebo (n= 16). - Primary endpoint: 30-day MACE defined as recurrent MI, development of a new arrhythmia, new septal/valve rupture, evidence of dissection, pericarditis, or tamponade. - Secondary efficacy outcomes: readmission rates within 30 days of receiving the study medication, differences in CRP levels at 30 days as compared to levels before receiving the study medication and changes in QT/corrected QT - was similar in both groups (9/12 in TCZ group vs. 3/16 in placebo group) - was similar in both groups
AUC area under curve, CRP C-reactive protein, CV cardiovascular, DMARD disease modifying anti-rheumatic drug, hsCRP high-sensitivity C- reactive protein, hsTnT high-sensitivity TrT, MACE major adverse cardiovascular event, MI myocardial infarction, TCZ tocilizumab, TNF tumor necrosis factor.

Evidence suggest that IL-6 has a positive impact on the lipid handling system through upregulation of ATP binding cassette transporter (ABC)A1, a protein involved in macrophage lipid efflux (98) and that IL-6 may inhibit several pro-inflammatory cytokines (99-102). Changes in lipid profile have been reported with TCZ use. TCZ has been shown to increase body weight and elevate triglycerides and cholesterol (LDL-cholesterol, high density lipoprotein [HDL]-cholesterol, total cholesterol, triglycerides, total cholesterol to HDL ratio) (103). A recent proteomic study in 20 female subjects with moderate-severe RA has shown that TCZ changed HDL composition, reducing atherogenic proteins such as serum amyloid A (104). TCZ has also been shown to reduce circulating levels of lipoprotein (Lp) (a), a predictor of vascular events and an independent risk factor for atherosclerotic CVD containing a highly glycosylated apolipoprotein (a), covalently attached to apolipoprotein B-100 (105, 106). After 12 weeks of TCZ treatment, patients with RA had reduced apolipoprotein (a) within LDL particles (104).

Currently, an ongoing phase IV trial named “Effect of Subcutaneous Actemra on Inflamed Atherosclerotic Plaques in Patients With Rheumatoid Arthritis” aims to test the hypothesis that subcutaneous TCZ 62 mg/week (in addition to MTX or monotherapy) is effective for reducing plaque inflammation measured by FDG-PET in patients with RA who are inadequate responders to DMARDs and are naive to biologic therapy. Effect of TCZ on arterial plaque inflammation will be examined by measuring the change in FDG uptake in the carotid arteries, coronary arteries and aorta between the baseline and 12-week FDG-PET/MRI (107).

3.3. Anti-inflammatory drugs currently being tested in cardiovascular outcome studies
3.3.1. Canakinumab

Anti-IL-1β inhibitor canakinumab, is approved for use in systemic juvenile idiopathic arthritis and cryopyrin-associated periodic syndromes. It is a fully human monoclonal antibody that directly and specifically inhibits soluble IL-1β, sparing IL-1α signalling. IL-1β is believed to be a key pro-atherogenic cytokine that stimulates cytokine secretion, activation of endothelial cells (108) and leukocytes (109) and fibroblast proliferation (110, 111). IL-1β levels have been shown to correlate with coronary artery disease severity (112).

Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) was a double-blind, placebo-controlled, event-driven trial of quarterly subcutaneous canakinumab in the prevention of recurrent CV events among stable post-MI patients with elevated hsCRP. It has tested the hypothesis that canakinumab treatment of patients with MI at least one month prior to study entry and elevated hsCRP (≥ 2 mg/L) would prevent recurrent CV events. A total of 10.061 subjects were randomized to either 50 mg, 150 mg, 300 mg canakinumab or placebo. The primary endpoint of the study was time to first occurrence of major adverse CV event, a composite of CV death, non-fatal MI and non-fatal stroke. At a median follow-up of 3.7 years, the primary outcome occurred in 4.11/100 person-years of the 50 mg group vs. 3.86/100 person-years of the 150 mg group vs. 3.90/100 person-years of the 300 mg group vs. 4.50/100 person-years of the placebo group (p = 0.02 for 150 mg group vs. placebo; other comparisons non-significant). Secondary endpoints included time to first occurrence of the composite CV endpoint consisting of CV death, non-fatal MI, non-fatal stroke and hospitalization for unstable angina requiring unplanned revascularization; time to new onset type 2 diabetes among people with pre-diabetes at randomization; time to occurrence of non-fatal MI, non-fatal stroke or all-cause mortality; and time to all-cause mortality. hsCRP reduction from baseline vs. placebo was 26% greater in the 50 mg group, 37% greater in the 150 mg group and 41% greater in the 300 mg group (p< 0.001 for all comparisons with placebo). Fatal infection or sepsis rate was higher in the canakinumab group (0.31/100 person-year in the combined canakinumab vs. 0.18/100 person-year in the placebo group, p = 0.02). The overall rates of adverse events (AEs), serious AEs and discontinuations due to AEs were similar to placebo across all doses (113). Results of secondary analysis addressing relationship of hsCRP reduction to event reduction showed that among patients who were randomized to canakinumab and achieved on-treatment hsCRP< 2 mg/L, there was a reduction in major adverse CV events (HR 0.75, 95% CI 0.66-0.85, p < 0.0001), compared with no benefit among those with on-treatment hsCRP≥ 2 mg/L (HR 0.90, 95% CI 0.79-1.02, p = 0.11) (114). In a similar way, secondary analysis to investigate CV outcomes in correspondence to the amount of reduction in IL-6 demonstrated that among those who experienced an on-treatment reduction in IL-6 below the median value, there was a 32% reduction in major adverse CV events for canakinumab vs. placebo (p< 0.0001) (115). CANTOS trial is of particular importance to suggest that in patients with a history of MI and elevated hsCRP, 150 mg dose of canakinumab is effective at preventing major adverse CV events over a median of 3.7 years, supporting the inflammation hypothesis of atherosclerosis. This trial validates for the first time that anti-inflammatory therapies may address the residual risk in CVD that exists despite intense lipid-lowering therapy. FDA has recently declared that the modest absolute clinical benefit of canakinumab cannot currently justify its routine use in patients with previous MI unless more data about its efficacy and safety is obtained and cost-effectiveness evaluation supports it (116).

In an ongoing substudy of CANTOS, it is aimed to evaluate the effect of quarterly subcutaneous canakinumab treatment for 24 months compared with placebo on the carotid plaque burden measured by vascular MRI in patients enrolled in CANTOS (117).

3.3.2. Rituximab

Rituximab is an anti-CD20 antibody that is used clinically to treat inflammatory autoimmune diseases and B-lymphocyte cancers. Rituximab in Patients with Acute ST-elevation Myocardial Infarction (RITA-MI) study is currently recruiting participants to test its clinical efficacy and safety in acute MI (118).

3.4. Possible future anti-inflammatory drug targets for treatment of atheroclerosis

Possible anti-inflammatory drugs may target NLRP3 inflammasome (119, 120); IL-1; IL-2; IL-18; IL-33; MCP-1 (CCL-2)/ CCR-2 (121-123); CCL5/CXCR4 (124, 125); CD40/ CD40L, tumor necrosis factor receptor- associated factor-6 (TRAF-6) (126-130); MPO (131) and glucagon-like peptide-1 (GLP-1) (132) for treatment of atherosclerosis.

Currently only IL-2 is under assessment in a clinical trial for investigating its safety and efficacy in the setting of coronary artery disease. Recent in vivo studies have suggested that low-dose IL-2 can increase the number of regulatory T cells, which are known to induce immune tolerance and preserve immune homeostasis (graft-versus-host disease (133, 134) and in hepatitis C virus-induced vasculitis (135)). Aldesleukin is a human recombinant form of IL-2 that has been used therapeutically in several autoimmune diseases. Low Dose Interleukin-2 in Patients with Stable Ischemic Heart Disease and Acute Coronary Syndromes (LILACS) is a double-blind, placebo-controlled trial will allow assessment of the safety and efficacy of low-dose IL-2 in patients with stable ischamic heart disease and in ACS patients, both conditions where it is currently contraindicated (136).

References
[2]
Bentzon J. F. Targeting Inflammation in Atherosclerosis J Am Coll Cardiol 2016 68 25 2794 2796 DOI: 10.1016/j.jacc.2016.11.005
[3]
Stone G. W. Maehara A. Lansky A. J. Bruyne B. de Cristea E. Mintz G. S. Mehran R. McPherson J. Farhat N. Marso S. P. Parise H. Templin B. White R. Zhang Z. Serruys P. W. Investigators P. A prospective natural-history study of coronary atherosclerosis N Engl J Med 2011 364 3 226 35 DOI: 10.1056/NEJMoa1002358
[4]
Downs J. R. Clearfield M. Weis S. Whitney E. Shapiro D. R. Beere P. A. Langendorfer A. Stein E. A. Kruyer W. Gotto Jr.: A. M. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas Coronary Atherosclerosis Prevention Study JAMA 1998 279 20 1615 22 DOI: 10.1001/jama.279.20.1615
[5]
G. Long-Term Intervention with Pravastatin in Ischaemic Disease Study: Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels N Engl J Med 1998 339 19 1349 57 DOI: 10.1056/NEJM199811053391902
[6]
Ridker P. M. Danielson E. Fonseca F. A. Genest J. Gotto Jr. A. M. Kastelein J. J. Koenig W. Libby P. Lorenzatti A. J. MacFadyen J. G. Nordestgaard B. G. Shepherd J. Willerson J. T. Glynn R. J. Group J. S. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein N Engl J Med 2008 359 21 2195 207 DOI: 10.1056/NEJMoa0807646
[7]
Kwon O. Kang S. J. Kang S. H. Lee P. H. Yun S. C. Ahn J. M. Park D. W. Lee S. W. Kim Y. H. Lee C. W. Han K. H. Park S. W. Park S. J. Relationship Between Serum Inflammatory Marker Levels and the Dynamic Changes in Coronary Plaque Characteristics After Statin Therapy Circ Cardiovasc Imaging 2017 10 7 DOI: 10.1161/CIRCIMAGING.116.005934
[8]
Barnes P. J. Anti-inflammatory actions of glucocorticoids: molecular mechanisms Clin Sci (Lond) 1998 94 6 557 72 DOI: 10.1042/cs0940557
[9]
Colao A. Pivonello R. Spiezia S. Faggiano A. Ferone D. Filippella M. Marzullo P. Cerbone G. Siciliani M. Lombardi G. Persistence of increased cardiovascular risk in patients with Cushing's disease after five years of successful cure J Clin Endocrinol Metab 1999 84 8 2664 72 DOI: 10.1210/jcem.84.8.5.896
[10]
Vlachoyiannopoulos P. G. Kanellopoulos P. G. Ioannidis J. P. Tektonidou M. G. Mastorakou I. Moutsopoulos H. M. Atherosclerosis in premenopausal women with antiphospholipid syndrome and systemic lupus erythematosus: a controlled study Rheumatology (Oxford) 2003 42 5 645 51 DOI: 10.1093/rheumatology/keg182
[11]
Manger K. Kusus M. Forster C. Ropers D. Daniel W. G. Kalden J. R. Achenbach S. Manger B. Factors associated with coronary artery calcification in young female patients with SLE Ann Rheum Dis 2003 62 9 846 50 DOI: 10.1136/ard.62.9.846
[12]
Rincon I. del O'Leary D. H. Haas R. W. Escalante A. Effect of glucocorticoids on the arteries in rheumatoid arthritis Arthritis Rheum 2004 50 12 3813 22 DOI: 10.1002/art.20661
[13]
Souverein P. C. Berard A. Staa T. P. Van Cooper C. Egberts A. C. Leufkens H. G. Walker B. R. Use of oral glucocorticoids and risk of cardiovascular and cerebrovascular disease in a population based case-control study Heart 2004 90 8 859 65 DOI: 10.1136/hrt.2003.020180
[14]
Wei L. MacDonald T. M. Walker B. R. Taking glucocorticoids by prescription is associated with subsequent cardiovascular disease Ann Intern Med 2004 141 10 764 70 DOI: 10.7326/0003-4819-141-10-200411160-00007
[15]
Davis 3rd J. M. Kremers H. Maradit Crowson C. S. Nicola P. J. Ballman K. V. Therneau T. M. Roger V. L. Gabriel S. E. Glucocorticoids and cardiovascular events in rheumatoid arthritis: a population-based cohort study Arthritis Rheum 2007 56 3 820 30 DOI: 10.1002/art.22418
[16]
Uddhammar A. Eriksson A. L. Nystrom L. Stenling R. Rantapaa-Dahlqvist S. Increased mortality due to cardiovascular disease in patients with giant cell arteritis in northern Sweden J Rheumatol 2002 29 4 737 42
[17]
Kremers H. Maradit Reinalda M. S. Crowson C. S. Davis 3rd J. M. Hunder G. G. Gabriel S. E. Glucocorticoids and cardiovascular and cerebrovascular events in polymyalgia rheumatica Arthritis Rheum 2007 57 2 279 86 DOI: 10.1002/art.22548
[18]
Giugliano G. R. Giugliano R. P. Gibson C. M. Kuntz R. E. Meta-analysis of corticosteroid treatment in acute myocardial infarction Am J Cardiol 2003 91 9 1055 9 DOI: 10.1016/S0002-9149(03)00148-6
[19]
Versaci F. Gaspardone A. Tomai F. Ribichini F. Russo P. Proietti I. Ghini A. S. Ferrero V. Chiariello L. Gioffre P. A. Romeo F. Crea F. Implantation S. Immunosuppressive Therapy for the Prevention of Restenosis after Coronary Artery Stent Immunosuppressive Therapy for the Prevention of Restenosis after Coronary Artery Stent Implantation (IMPRESS Study) J Am Coll Cardiol 2002 40 11 1935 42 DOI: 10.1016/S0735-1097(02)02562-7
[20]
Kakio T. Matsumori A. Ohashi N. Yamada T. Saito T. Kawamoto A. Taguchi A. Morita Y. Takahashi M. Sasayama S. Hydrocortisone reduces restenosis after stenting of small coronary arteries J Interv Cardiol 2004 17 5 295 300 DOI: 10.1111/j.1540-8183.2004.02999.x
[21]
Kakio T. Matsumori A. Ohashi N. Yamada T. Nobuhara M. Saito T. Kawamoto A. Taguchi A. Morita Y. Takahashi M. Sasayama S. The effect of hydrocortisone on reducing rates of restenosis and target lesion revascularization after coronary stenting less than 3 mm in stent diameter Intern Med 2003 42 11 1084 9
[22]
Ferrero V. Ribichini F. Rognoni A. Marino P. Brunelleschi S. Vassanelli C. Comparison of efficacy and safety of lower-dose to higher-dose oral prednisone after percutaneous coronary interventions (the IMPRESS-LD study) Am J Cardiol 2007 99 8 1082 6 DOI: 10.1016/j.amjcard.2006.11.064
[23]
Pepine C. J. Hirshfeld J. W. Macdonald R. G. Henderson M. A. Bass T. A. Goldberg S. Savage M. P. Vetrovec G. Cowley M. Taussig A. S. A controlled trial of corticosteroids to prevent restenosis after coronary angioplasty. M-HEART Group Circulation 1990 81 6 1753 61 https://10.1161/01. CIR.81.6.1753
[24]
Lee C. W. Chae J. K. Lim H. Y. Hong M. K. Kim J. J. Park S. W. Park S. J. Prospective randomized trial of corticosteroids for the prevention of restenosis after intracoronary stent implantation Am Heart J 1999 138 1 Pt 1 60 3 DOI: 10.1016/S0002-8703(99)70247-4
[25]
Stone G. W. Rutherford B. D. McConahay D. R. Johnson W. L. Giorgi L. V. Ligon R. W. Hartzler G. O. A randomized trial of corticosteroids for the prevention of restenosis in 102 patients undergoing repeat coronary angioplasty Cathet Cardiovasc Diagn 1989 18 4 227 31 DOI: 10.1002/ccd.1810180407
[26]
Reimers B. Moussa I. I. Akiyama T. Kobayashi Y. Albiero R. Francesco L. Di Mario C. Di Colombo A. Persistent High Restenosis After Local Intrawall Delivery of Long-Acting Steroids Before Coronary Stent Implantation J Invasive Cardiol 1998 10 6 323 331
[27]
Rab S. T. King 3rd S. B. Roubin G. S. Carlin S. Hearn J. A. Douglas Jr.: J. S. Coronary aneurysms after stent placement: a suggestion of altered vessel wall healing in the presence of anti-inflammatory agents J Am Coll Cardiol 1991 18 6 1524 8 DOI: 10.1016/j.jcin.2007.10.004
[28]
Ward M. R. Kanellakis P. Ramsey D. Funder J. Bobik A. Eplerenone suppresses constrictive remodeling and collagen accumulation after angioplasty in porcine coronary arteries Circulation 2001 104 4 467 72 DOI: 10.1161/hc3001.091458
[29]
Patti G. Chello M. Pasceri V. Colonna D. Carminati P. Covino E. Germano S. Di Dexamethasone-eluting stents and plasma concentrations of adhesion molecules in patients with unstable coronary syndromes: results of the historically controlled SESAME study Clin Ther 2005 27 9 1411 9 DOI: 10.1016/j.clinthera.2005.09.009
[30]
Patti G. Pasceri V. Carminati P. D'Ambrosio A. Carcagni A. Sciascio G. Di Effect of dexamethasone-eluting stents on systemic inflammatory response in patients with unstable angina pectoris or recent myocardial infarction undergoing percutaneous coronary intervention Am J Cardiol 2005 95 4 502 5 DOI: 10.1016/j.amjcard.2004.10.021
[31]
Liu X. Huang Y. Hanet C. Vandormael M. Legrand V. Dens J. Vandenbossche J. L. Missault L. Vrints C. Scheerder I. De Study of antirestenosis with the BiodivYsio dexamethasone-eluting stent (STRIDE): a first-in-human multicenter pilot trial Catheter Cardiovasc Interv 2003 60 2 discussion 179 DOI: 10.1002/ccd.10636
[32]
Fishel R. S. Eisenberg S. Shai S. Y. Redden R. A. Bernstein K. E. Berk B. C. Glucocorticoids induce angiotensin-converting enzyme expression in vascular smooth muscle Hypertension 1995 25 3 343 9 DOI: 10.1161/01.HYP.25.3.343
[33]
Mendelsohn F. A. Lloyd C. J. Kachel C. Funder J. W. Induction by glucocorticoids of angiotensin converting enzyme production from bovine endothelial cells in culture and rat lung in vivo J Clin Invest 1982 70 3 684 92 DOI: 10.1172/JCI110663
[34]
Morin C. Asselin C. Boudreau F. Provencher P. H. Transcriptional regulation of pre-pro-endothelin-1 gene by glucocorticoids in vascular smooth muscle cells Biochem Biophys Res Commun 1998 244 2 583 7 DOI: 10.1006/bbrc.1998.8300
[35]
Mangos G. J. Walker B. R. Kelly J. J. Lawson J. A. Webb D. J. Whitworth J. A. Cortisol inhibits cholinergic vasodilation in the human forearm Am J Hypertens 2000 13 11 1155 60 DOI: 10.1016/S0895-7061(00)01201-2
[36]
Yasojima K. Schwab C. McGeer E. G. McGeer P. L. Human heart generates complement proteins that are upregulated and activated after myocardial infarction Circ Res 1998 83 8 860 9 DOI: 10.1161/01. RES.83.8.860
[37]
Markiewski M. M. Nilsson B. Ekdahl K. N. Mollnes T. E. Lambris J. D. Complement and coagulation: strangers or partners in crime? Trends Immunol, 2007 28 4 184 92 DOI: 10.1016/j.it.2007.02.006
[38]
Testa L. Van Gaal W. J. Bhindi R. Biondi-Zoccai G. G. Abbate A. Agostoni P. Porto I. Andreotti F. Crea F. Banning A. P. Pexelizumab in ischemic heart disease: a systematic review and meta-analysis on 15,196 patients J Thorac Cardiovasc Surg 2008 136 4 884 93 DOI: 10.1016/j.jtcvs.2007.12.062
[39]
Suzawa H. Kikuchi S. Arai N. Koda A. The mechanism involved in the inhibitory action of tranilast on collagen biosynthesis of keloid fibroblasts Jpn J Pharmacol 1992 60 2 91 6 DOI: 10.1254/jjp.60.91
[40]
Holmes Jr. D. R. Savage M. LaBlanche J. M. Grip L. Serruys P. W. Fitzgerald P. Fischman D. Goldberg S. Brinker J. A. Zeiher A. M. Shapiro L. M. Willerson J. Davis B. R. Ferguson J. J. Popma J. King 3rd S. B. Lincoff A. M. Tcheng J. E. Chan R. Granett J. R. Poland M. Results of Prevention of REStenosis with Tranilast and its Outcomes (PRESTO) trial Circulation 2002 106 10 1243 50 DOI: 10.1161/01. CIR.0000028335.31300. DA
[41]
Jacobsson L. T. Turesson C. Gulfe A. Kapetanovic M. C. Petersson I. F. Saxne T. Geborek P. Treatment with tumor necrosis factor blockers is associated with a lower incidence of first cardiovascular events in patients with rheumatoid arthritis J Rheumatol 2005 32 7 1213 8
[42]
Dixon W. G. Watson K. D. Lunt M. Hyrich K. L. Centre C. British Society for Rheumatology Biologics Register Control Silman A. J. Symmons D. P. Biologics R. British Society for Rheumatology Reduction in the incidence of myocardial infarction in patients with rheumatoid arthritis who respond to anti-tumor necrosis factor alpha therapy: results from the British Society for Rheumatology Biologics Register Arthritis Rheum 2007 56 9 2905 12 DOI: 10.1002/art.22809
[43]
Greenberg J. D. Kremer J. M. Curtis J. R. Hochberg M. C. Reed G. Tsao P. Farkouh M. E. Nasir A. Setoguchi S. Solomon D. H. Investigators C. Tumour necrosis factor antagonist use and associated risk reduction of cardiovascular events among patients with rheumatoid arthritis Ann Rheum Dis 2011 70 4 576 82 DOI: 10.1136/ard.2010.129916
[44]
Solomon D. H. Curtis J. R. Saag K. G. Lii J. Chen L. Harrold L. R. Herrinton L. J. Graham D. J. Kowal M. K. Kuriya B. Liu L. Griffin M. R. Lewis J. D. Rassen J. A. Cardiovascular risk in rheumatoid arthritis: comparing TNF-alpha blockade with nonbiologic DMARDs Am J Med 2013 126 8 e9 730 e17 DOI: 10.1016/j.amjmed.2013.02.016
[45]
Bissonnette R. Tardif J. C. Harel F. Pressacco J. Bolduc C. Guertin M. C. Effects of the tumor necrosis factor-alpha antagonist adalimumab on arterial inflammation assessed by positron emission tomography in patients with psoriasis: results of a randomized controlled trial Circ Cardiovasc Imaging 2013 6 1 83 90 DOI: 10.1161/CIRCIMAGING.112.975730
[46]
Corson M. A. Phospholipase A2 inhibitors in atherosclerosis: the race is on Lancet 2009 373 9664 608 10 DOI: 10.1016/S0140-6736(09)60378-0
[47]
Lp P. L. A. S. C. Thompson A. Gao P. Orfei L. Watson S. Angelantonio E. Di Kaptoge S. Ballantyne C. Cannon C. P. Criqui M. Cushman M. Hofman A. Packard C. Thompson S. G. Collins R. Danesh J. Lipoprotein-associated phospholipase A (2) and risk of coronary disease, stroke, and mortality: collaborative analysis of 32 prospective studies Lancet 2010 375 9725 1536 44 DOI: 10.1016/S0140-6736(10)60319-4
[48]
Hatoum I. J. Cook N. R. Nelson J. J. Rexrode K. M. Rimm E. B. Lipoprotein-associated phospholipase A2 activity improves risk discrimination of incident coronary heart disease among women Am Heart J 2011 161 3 516 22 DOI: 10.1016/j.ahj.2010.11.007
[49]
Nicholls S. J. Kastelein J. J. Schwartz G. G. Bash D. Rosenson R. S. Cavender M. A. Brennan D. M. Koenig W. Jukema J. W. Nambi V. Wright R. S. Menon V. Lincoff A. M. Nissen S. E. Investigators V.- Varespladib and cardiovascular events in patients with an acute coronary syndrome: the VISTA-16 randomized clinical trial JAMA 2014 311 3 252 62 DOI: 10.1001/jama.2013.282836
[50]
Marber M. S. Rose B. Wang Y. The p38 mitogen-activated protein kinase pathway--a potential target for intervention in infarction, hypertrophy, and heart failure J Mol Cell Cardiol 2011 51 4 485 90 DOI: 10.1016/j.yjmcc.2010.10.021
[51]
Lee J. C. Kassis S. Kumar S. Badger A. Adams J. L. p38 mitogen-activated protein kinase inhibitors--mechanisms and therapeutic potentials Pharmacol Ther 1999 82 2-3 389 97 DOI: 10.1016/S0163-7258(99)00008-X
[52]
Kumar S. Boehm J. Lee J. C. p38 MAP kinases: key signalling molecules as therapeutic targets for inflammatory diseases Nat Rev Drug Discov 2003 2 9 717 26 DOI: 10.1038/nrd1177
[53]
Yin M. J. Yamamoto Y. Gaynor R. B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of I (kappa)B kinase-beta Nature 1998 396 6706 77 80 DOI: 10.1038/23948
[54]
Chiba T. Kondo Y. Shinozaki S. Kaneko E. Ishigami A. Maruyama N. Umezawa K. Shimokado K. A selective NFkappaB inhibitor, DHMEQ, reduced atherosclerosis in ApoE-deficient mice J Atheroscler Thromb 2006 13 6 308 13 DOI: 10.5551/jat.13.308
[55]
Gareus R. Kotsaki E. Xanthoulea S. Made I. van der Gijbels M. J. Kardakaris R. Polykratis A. Kollias G. Winther M. P. de Pasparakis M. Endothelial cell-specific NF-kappaB inhibition protects mice from atherosclerosis Cell Metab 2008 8 5 372 83 DOI: 10.1016/j.cmet.2008.08.016
[56]
Mitchell J. A. Saunders M. Barnes P. J. Newton R. Belvisi M. G. Sodium salicylate inhibits cyclo-oxygenase-2 activity independently of transcription factor (nuclear factor kappaB) activation: role of arachidonic acid Mol Pharmacol 1997 51 6 907 12 DOI: 10.1124/mol.51.6.907
[57]
Schwenger P. Alpert D. Skolnik E. Y. Vilcek J. Activation of p38 mitogen-activated protein kinase by sodium salicylate leads to inhibition of tumor necrosis factor-induced IkappaB alpha phosphorylation and degradation Mol Cell Biol 1998 18 1 78 84 https:/doi:10.1128/MCB.18.1.78
[58]
Cronstein B. N. Montesinos M. C. Weissmann G. Salicylates and sulfasalazine, but not glucocorticoids, inhibit leukocyte accumulation by an adenosine-dependent mechanism that is independent of inhibition of prostaglandin synthesis and p105 of NFkappaB Proc Natl Acad Sci U S A 1999 96 11 6377 81 DOI: 10.1073/pnas.96.11.6377
[59]
Xu X. M. Sansores-Garcia L. Chen X. M. Matijevic-Aleksic N. Du M. Wu K. K. Suppression of inducible cyclooxygenase 2 gene transcription by aspirin and sodium salicylate Proc Natl Acad Sci U S A 1999 96 9 5292 7 DOI: 10.1073/pnas.96.9.5292
[60]
Hauser T. H. Salastekar N. Schaefer E. J. Desai T. Goldfine H. L. Fowler K. M. Weber G. M. Welty F. Clouse M. Shoelson S. E. Goldfine A. B. Study T. Targeting Inflammation Using Salsalate in Cardiovascular Disease Effect of Targeting Inflammation With Salsalate: The TINSAL-CVD Randomized Clinical Trial on Progression of Coronary Plaque in Overweight and Obese Patients Using Statins JAMA Cardiol 2016 1 4 413 23 DOI: 10.1001/jamacardio.2016.0605
[61]
Bevilacqua M. P. Pober J. S. Wheeler M. E. Cotran R. S. Gimbrone Jr.: M. A. Interleukin-1 activation of vascular endothelium. Effects on procoagulant activity and leukocyte adhesion Am J Pathol 1985 121 3 394 403
[62]
Libby P. Warner S. J. 1 G. B. Friedman: Interleukin a mitogen for human vascular smooth muscle cells that induces the release of growth-inhibitory prostanoids J Clin Invest 1988 81 2 487 98 DOI: 10.1172/JCI113346
[63]
Loppnow H. Libby P. Proliferating or interleukin 1-activated human vascular smooth muscle cells secrete copious interleukin 6 J Clin Invest 1990 85 3 731 8 DOI: 10.1172/JCI114498
[64]
Collaboration I. R. G. C. E. R. F. Sarwar N. Butterworth A. S. Freitag D. F. Gregson J. Willeit P. Gorman D. N. Gao P. Saleheen D. Rendon A. Nelson C. P. Braund P. S. Hall A. S. Chasman D. I. Tybjaerg-Hansen A. Chambers J. C. Benjamin E. J. Franks P. W. Clarke R. Wilde A. A. Trip M. D. Steri M. Witteman J. C. Qi L. Schoot C. E. van der Faire U. de Erdmann J. Stringham H. M. Koenig W. Rader D. J. Melzer D. Reich D. Psaty B. M. Kleber M. E. Panagiotakos D. B. Willeit J. Wennberg P. Woodward M. Adamovic S. Rimm E. B. Meade T. W. Gillum R. F. Shaffer J. A. Hofman A. Onat A. Sundstrom J. Wassertheil-Smoller S. Mellstrom D. Gallacher J. Cushman M. Tracy R. P. Kauhanen J. Karlsson M. Salonen J. T. Wilhelmsen L. Amouyel P. Cantin B. Best L. G. Ben-Shlomo Y. Manson J. E. Davey-Smith G. Bakker P. I. de O'Donnell C. J. Wilson J. F. Wilson A. G. Assimes T. L. Jansson J. O. Ohlsson C. Tivesten A. Ljunggren O. Reilly M. P. Hamsten A. Ingelsson E. Cambien F. Hung J. Thomas G. N. Boehnke M. Schunkert H. Asselbergs F. W. Kastelein J. J. Gudnason V. Salomaa V. Harris T. B. Kooner J. S. Allin K. H. Nordestgaard B. G. Hopewell J. C. Goodall A. H. Ridker P. M. Holm H. Watkins H. Ouwehand W. H. Samani N. J. Kaptoge S. Angelantonio E. Di Harari O. Danesh J. Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies Lancet 2012 379 9822 1205 13 DOI: 10.1016/S0140-6736(11)61931-4
[65]
Analysis C. Interleukin-6 Receptor Mendelian Randomisation Swerdlow D. I. Holmes M. V. Kuchenbaecker K. B. Engmann J. E. Shah T. Sofat R. Guo Y. Chung C. Peasey A. Pfister R. Mooijaart S. P. Ireland H. A. Leusink M. Langenberg C. Li K. W. Palmen J. Howard P. Cooper J. A. Drenos F. Hardy J. Nalls M. A. Li Y. R. Lowe G. Stewart M. Bielinski S. J. Peto J. Timpson N. J. Gallacher J. Dunlop M. Houlston R. Tomlinson I. Tzoulaki I. Luan J. Boer J. M. Forouhi N. G. Onland-Moret N. C. Schouw Y. T. van der Schnabel R. B. Hubacek J. A. Kubinova R. Baceviciene M. Tamosiunas A. Pajak A. Topor-Madry R. Malyutina S. Baldassarre D. Sennblad B. Tremoli E. Faire U. de Ferrucci L. Bandenelli S. Tanaka T. Meschia J. F. Singleton A. Navis G. Leach I. Mateo Bakker S. J. Gansevoort R. T. Ford I. Epstein S. E. Burnett M. S. Devaney J. M. Jukema J. W. Westendorp R. G. Borst G. Jan de Graaf Y. van der Jong P. A. de Zee A. H. Mailand-van der Klungel O. H. Boer A. de Doevendans P. A. Stephens J. W. Eaton C. B. Robinson J. G. Manson J. E. Fowkes F. G. Frayling T. M. Price J. F. Whincup P. H. Morris R. W. Lawlor D. A. Smith G. D. Ben-Shlomo Y. Redline S. Lange L. A. Kumari M. Wareham N. J. Verschuren W. M. Benjamin E. J. Whittaker J. C. Hamsten A. Dudbridge F. Delaney J. A. Wong A. Kuh D. Hardy R. Castillo B. A. Connolly J. J. Harst P. van der Brunner E. J. Marmot M. G. Wassel C. L. Humphries S. E. Talmud P. J. Kivimaki M. Asselbergs F. W. Voevoda M. Bobak M. Pikhart H. Wilson J. G. Hakonarson H. Reiner A. P. Keating B. J. Sattar N. Hingorani A. D. Casas J. P. The interleukin-6 receptor as a target for prevention of coronary heart disease: a mendelian randomisation analysis Lancet 2012 379 9822 1214 24 DOI: 10.1016/S0140-6736(12)60110-X
[66]
Morton A. C. Rothman A. M. Greenwood J. P. Gunn J. Chase A. Clarke B. Hall A. S. Fox K. Foley C. Banya W. Wang D. Flather M. D. Crossman D. C. The effect of interleukin-1 receptor antagonist therapy on markers of inflammation in non-ST elevation acute coronary syndromes: the MRC-ILA Heart Study Eur Heart J 2015 36 6 377 84 DOI: 10.1093/eurheartj/ehu272
[67]
Genetics C. Interleukin 1 Cardiometabolic effects of genetic upregulation of the interleukin 1 receptor antagonist: a Mendelian randomisation analysis Lancet Diabetes Endocrinol 2015 3 4 243 53 DOI: 10.1016/S2213-8587(15)00034-0
[68]
Abbate A. Kontos M. C. Grizzard J. D. Biondi-Zoccai G. G. Tassell B. W. Van Robati R. Roach L. M. Arena R. A. Roberts C. S. Varma A. Gelwix C. C. Salloum F. N. Hastillo A. Dinarello C. A. Vetrovec G. W. Investigators V.-A. Interleukin-1 blockade with anakinra to prevent adverse cardiac remodeling after acute myocardial infarction (Virginia Commonwealth University Anakinra Remodeling Trial (VCU-ART) Pilot study) Am J Cardiol 2010 105 10 1371 1377 e1 DOI: 10.1016/j.amjcard.2009.12.059
[69]
Abbate A. Tassell B. W. Van Biondi-Zoccai G. Kontos M. C. Grizzard J. D. Spillman D. W. Oddi C. Roberts C. S. Melchior R. D. Mueller G. H. Abouzaki N. A. Rengel L. R. Varma A. Gambill M. L. Falcao R. A. Voelkel N. F. Dinarello C. A. Vetrovec G. W. Effects of interleukin-1 blockade with anakinra on adverse cardiac remodeling and heart failure after acute myocardial infarction (from the Virginia Commonwealth University-Anakinra Remodeling Trial (2) (VCU-ART2) pilot study) Am J Cardiol 2013 111 10 1394 400 DOI: 10.1016/j.amjcard.2013.01.287
[70]
Abbate A. Kontos M. C. Abouzaki N. A. Melchior R. D. Thomas C. Tassell B. W. Van Oddi C. Carbone S. Trankle C. R. Roberts C. S. Mueller G. H. Gambill M. L. Christopher S. Markley R. Vetrovec G. W. Dinarello C. A. Biondi-Zoccai G. Comparative safety of interleukin-1 blockade with anakinra in patients with ST-segment elevation acute myocardial infarction (from the VCU-ART and VCU-ART2 pilot studies) Am J Cardiol 2015 115 3 288 92 DOI: 10.1016/j.amjcard.2014.11.003
[71]
Gerards A. H. Lathouder S. de Groot E. R. de Dijkmans B. A. Aarden L. A. Inhibition of cytokine production by methotrexate. Studies in healthy volunteers and patients with rheumatoid arthritis Rheumatology (Oxford) 2003 42 10 1189 96 DOI: 10.1093/rheumatology/keg323
[72]
Herman S. Zurgil N. Langevitz P. Ehrenfeld M. Deutsch M. Methotrexate selectively modulates TH1/TH2 balance in active rheumatoid arthritis patients Clin Exp Rheumatol 2008 26 2 317 23
[73]
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 12 3675 83 DOI: 10.1002/art.24040
[74]
McPherson J. A. Barringhaus K. G. Bishop G. G. Sanders J. M. Rieger J. M. Hesselbacher S. E. Gimple L. W. Powers E. R. Macdonald T. Sullivan G. Linden J. Sarembock I. J. Adenosine A (2A) receptor stimulation reduces inflammation and neointimal growth in a murine carotid ligation model Arterioscler Thromb Vasc Biol 2001 21 5 791 6 DOI: 10.1161/01. ATV.21.5.791
[75]
Bulgarelli A. Dias A. A. Martins Caramelli B. Maranhao R. C. Treatment with methotrexate inhibits atherogenesis in cholesterol-fed rabbits J Cardiovasc Pharmacol 2012 59 4 308 14 DOI: 10.1097/FJC.0b013e318241c385
[76]
Choi H. K. Hernan M. A. Seeger J. D. Robins J. M. Wolfe F. Methotrexate and mortality in patients with rheumatoid arthritis: a prospective study Lancet 2002 359 9313 1173 7 DOI: 10.1016/S0140-6736(02)08213-2
[77]
Ridker P. M. Testing the inflammatory hypothesis of atherothrombosis: scientific rationale for the cardiovascular inflammation reduction trial (CIRT) J Thromb Haemost 2009 7 Suppl 1 332 9 DOI: 10.1111/j.1538-7836.2009.03404.x
[78]
Micha R. Imamura F. Ballmoos M. Wyler von Solomon D. H. Hernan M. A. Ridker P. M. Mozaffarian D. Systematic review and meta-analysis of methotrexate use and risk of cardiovascular disease Am J Cardiol 2011 108 9 1362 70 DOI: 10.1016/j.amjcard.2011.06.054
[81]
P. M. E. Ridker B.M. Pradhan A. MacFadyen J.G. Solomon D.H. Zaharris E. Mam V. Hasan A. Rosenberg Y. Iturriaga E. Gupta M. Tsigoulis M. Verma S. Clearfield M. Libby P. Goldhaber S.Z. Seagle R. Ofori C. Saklayen M. Butman S. Singh N. Le May M. Bertrand O. Johnston J. Paynter N.P. Glynn R.J. for the CIRT Investigators: Low-Dose Methotrexate for the Prevention of Atherosclerotic Events New Engl J Med 2018 DOI: 10.1056/NEJMoa1809798
[82]
Robertson S. Martinez G. J. Payet C. A. Barraclough J. Y. Celermajer D. S. Bursill C. PaTel S. Colchicine therapy in acute coronary syndrome patients acts on caspase-1 to suppress NLRP3 inflammasome monocyte activation Clin Sci (Lond) 2016 130 14 1237 46 DOI: 10.1042/CS20160090
[83]
Martinez G. J. Robertson S. Barraclough J. Xia Q. Mallat Z. Bursill C. Celermajer D. S. PaTel S. Colchicine Acutely Suppresses Local Cardiac Production of Inflammatory Cytokines in Patients With an Acute Coronary Syndrome J Am Heart Assoc 2015 4 8 e002128 DOI: 10.1161/JAHA.115.002128
[84]
Colchicine G. Nuki its mechanism of action and efficacy in crystal-induced inflammation Curr Rheumatol Rep 2008 10 3 218 27 DOI: 10.1007/s11926-008-0036-3
[85]
O'Keefe Jr. J. H. McCallister B. D. Bateman T. M. Kuhnlein D. L. Ligon R. W. Hartzler G. O. Ineffectiveness of colchicine for the prevention of restenosis after coronary angioplasty J Am Coll Cardiol 1992 19 7 1597 600 DOI: 10.1016/0735-1097(92)90624-V
[86]
Deftereos S. Giannopoulos G. Raisakis K. Kossyvakis C. Kaoukis A. Panagopoulou V. Driva M. Hahalis G. Pyrgakis V. Alexopoulos D. Manolis A. S. Stefanadis C. Cleman M. W. Colchicine treatment for the prevention of bare-metal stent restenosis in diabetic patients J Am Coll Cardiol 2013 61 16 1679 85 DOI: 10.1016/j.jacc.2013.01.055
[87]
Giannopoulos G. Angelidis C. Kouritas V. K. Dedeilias P. Filippatos G. Cleman M. W. Panagopoulou V. Siasos G. Tousoulis D. Lekakis J. Deftereos S. Usefulness of colchicine to reduce perioperative myocardial damage in patients who underwent on-pump coronary artery bypass grafting Am J Cardiol 2015 115 10 1376 81 DOI: 10.1016/j.amjcard.2015.02.036
[88]
Langevitz P. Livneh A. Neumann L. Buskila D. Shemer J. Amolsky D. Pras M. Prevalence of ischemic heart disease in patients with familial Mediterranean fever Isr Med Assoc J 2001 3 1 9 12
[89]
Crittenden D. B. Lehmann R. A. Schneck L. Keenan R. T. Shah B. Greenberg J. D. Cronstein B. N. Sedlis S. P. Pillinger M. H. Colchicine use is associated with decreased prevalence of myocardial infarction in patients with gout J Rheumatol 2012 39 7 1458 64 DOI: 10.3899/jrheum.111533
[90]
Nidorf S. M. Eikelboom J. W. Budgeon C. A. Thompson P. L. Low-dose colchicine for secondary prevention of cardiovascular disease J Am Coll Cardiol 2013 61 4 404 10 DOI: 10.1016/j.jacc.2012.10.027
[91]
Verma S. Eikelboom J. W. Nidorf S. M. Al-Omran M. Gupta N. Teoh H. Friedrich J. O. Colchicine in cardiac disease: a systematic review and meta-analysis of randomized controlled trials BMC Cardiovasc Disord 2015 15 96 DOI: 10.1186/s12872-015-0068-3
[92]
Solomon D. H. Liu C. C. Kuo I. H. Zak A. Kim S. C. Effects of colchicine on risk of cardiovascular events and mortality among patients with gout: a cohort study using electronic medical records linked with Medicare claims Ann Rheum Dis 2016 75 9 1674 9 DOI: 10.1136/annrheumdis-2015-207984
[93]
Vaidya K. Arnott C. Martinez G. J. Ng B. McCormack S. Sullivan D. R. Celermajer D. S. PaTel S. Colchicine Therapy and Plaque Stabilization in Patients With Acute Coronary Syndrome: A CT Coronary Angiography Study JACC Cardiovasc Imaging 2018 11 2 Pt 2 305 316 DOI: 10.1016/j.jcmg.2017.08.013
[96]
Sawa Y. Ichikawa H. Kagisaki K. Ohata T. Matsuda H. Interleukin-6 derived from hypoxic myocytes promotes neutrophil-mediated reperfusion injury in myocardium J Thorac Cardiovasc Surg 1998 116 3 511 7 DOI: 10.1016/S0022-5223(98)70018-2
[97]
Zamani P. Schwartz G. G. Olsson A. G. Rifai N. Bao W. Libby P. Ganz P. Kinlay S. Study I. Myocardial Ischemia Reduction with Aggressive Cholesterol Lowering Inflammatory biomarkers, death, and recurrent nonfatal coronary events after an acute coronary syndrome in the MIRACL study J Am Heart Assoc 2013 2 1 e003103 DOI: 10.1161/JAHA.112.003103
[98]
Frisdal E. Lesnik P. Olivier M. Robillard P. Chapman M. J. Huby T. Guerin M. Goff W. Le Interleukin-6 protects human macrophages from cellular cholesterol accumulation and attenuates the proinflammatory response J Biol Chem 2011 286 35 30926 36 DOI: 10.1074/jbc.M111.264325
[99]
Starkie R. Ostrowski S. R. Jauffred S. Febbraio M. Pedersen B. K. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production in humans FASEB J 2003 17 8 884 6 DOI: 10.1096/fj.02-0670fje
[100]
Tilg H. Dinarello C. A. Mier J. W. IL-6 and APPs: anti-inflammatory and immunosuppressive mediators Immunol Today 1997 18 9 428 32 DOI: 10.1016/S0167-5699(97)01103-1
[101]
Tilg H. Trehu E. Atkins M. B. Dinarello C. A. Mier J. W. Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55 Blood 1994 83 1 113 8
[102]
Schindler R. Mancilla J. Endres S. Ghorbani R. Clark S. C. Dinarello C. A. Correlations and interactions in the production of interleukin-6 (IL-6), IL-1, and tumor necrosis factor (TNF) in human blood mononuclear cells: IL-6 suppresses IL-1 and TNF Blood 1990 75 1 40 7
[103]
Gabay C. McInnes I. B. Kavanaugh A. Tuckwell K. Klearman M. Pulley J. Sattar N. Comparison of lipid and lipid-associated cardiovascular risk marker changes after treatment with tocilizumab or adalimumab in patients with rheumatoid arthritis Ann Rheum Dis 2016 75 10 1806 12 DOI: 10.1136/annrheumdis-2015-207872
[104]
Lee J. S. Chapman M. J. Piraino P. Lamerz J. Schindler T. Cutler P. Dernick G. Remodeling of plasma lipoproteins in patients with rheumatoid arthritis: Interleukin-6 receptor-alpha inhibition with tocilizumab Proteomics Clin Appl 2016 10 2 183 94 DOI: 10.1002/prca.201500036
[105]
Muller N. Schulte D. M. Turk K. Freitag-Wolf S. Hampe J. Zeuner R. Schroder J. O. Gouni-Berthold I. Berthold H. K. Krone W. Rose-John S. Schreiber S. Laudes M. IL-6 blockade by monoclonal antibodies inhibits apolipoprotein (a) expression and lipoprotein (a) synthesis in humans J Lipid Res 2015 56 5 1034 42 DOI: 10.1194/jlr.P052209
[106]
Schultz O. Oberhauser F. Saech J. Rubbert-Roth A. Hahn M. Krone W. Laudes M. Effects of inhibition of interleukin-6 signalling on insulin sensitivity and lipoprotein (a) levels in human subjects with rheumatoid diseases PLoS One 2010 5 12 e14328 DOI: 10.1371/journal.pone.0014328
[108]
Wang X. Feuerstein G. Z. Gu J. L. Lysko P. G. Yue T. L. Interleukin-1 beta induces expression of adhesion molecules in human vascular smooth muscle cells and enhances adhesion of leukocytes to smooth muscle cells Atherosclerosis 1995 115 1 89 98 DOI: 10.1016/0021-9150(94)05503-B
[109]
Mantovani A. Cassatella M. A. Costantini C. Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity Nat Rev Immunol 2011 11 8 519 31 DOI: 10.1038/nri3024
[110]
Lambert J. M. Lopez E. F. Lindsey M. L. Macrophage roles following myocardial infarction Int J Cardiol 2008 130 2 1477 58 DOI: 10.1016/j.ijcard.2008.04.059
[111]
Tedgui A. Mallat Z. Cytokines in atherosclerosis: pathogenic and regulatory pathways Physiol Rev 2006 86 2 515 81 DOI: 10.1152/physrev.00024.2005
[112]
Galea J. Armstrong J. Gadsdon P. Holden H. Francis S. E. Holt C. M. Interleukin-1 beta in coronary arteries of patients with ischemic heart disease Arterioscler Thromb Vasc Biol 1996 16 8 1000 6 DOI: 10.1161/01.ATV.16.8.1000
[113]
Ridker P. M. Everett B. M. Thuren T. MacFadyen J. G. Chang W. H. Ballantyne C. Fonseca F. Nicolau J. Koenig W. Anker S. D. Kastelein J. J. P. Cornel J. H. Pais P. Pella D. Genest J. Cifkova R. Lorenzatti A. Forster T. Kobalava Z. Vida-Simiti L. Flather M. Shimokawa H. Ogawa H. Dellborg M. Rossi P. R. F. Troquay R. P. T. Libby P. Glynn R. J. Group C. T. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease New Engl J Med 2017 377 12 1119 1131 DOI: 10.1056/NEJMoa1707914
[114]
Ridker P. M. MacFadyen J. G. Everett B. M. Libby P. Thuren T. Glynn R. J. Group C. T. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial Lancet 2018 391 10118 319 328 DOI: 10.1016/S0140-6736(17)32814-3
[115]
Ridker P. M. Libby P. MacFadyen J. G. Thuren T. Ballantyne C. Fonseca F. Koenig W. Shimokawa H. Everett B. M. Glynn R. J. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS) Eur Heart J 2018 39 38 3499 3507 DOI: 10.1093/eurheartj/ehy310
[119]
Heijden T. van der Kritikou E. Venema W. Duijn J. van Santbrink P. J. van Slutter B. Foks A. C. Bot I. Kuiper J. NLRP3 Inflammasome Inhibition by MCC950 Reduces Atherosclerotic Lesion Development in Apolipoprotein E-Deficient Mice-Brief Report Arterioscler Thromb Vasc Biol 2017 37 8 1457 1461 DOI: 10.1161/ATVBAHA.117.309575
[120]
Abderrazak A. Couchie D. Mahmood D. F. Elhage R. Vindis C. Laffargue M. Mateo V. Buchele B. Ayala M. R. Gaafary M. El Syrovets T. Slimane M. N. Friguet B. Fulop T. Simmet T. Hadri K. El Rouis M. Anti-inflammatory and antiatherogenic effects of the NLRP3 inflammasome inhibitor arglabin in ApoE2. Ki mice fed a high-fat diet Circulation 2015 131 12 1061 70 DOI: 10.1161/CIRCULATIONAHA.114.013730
[121]
Gu L. Okada Y. Clinton S. K. Gerard C. Sukhova G. K. Libby P. Rollins B. J. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice Mol Cell 1998 2 2 275 81 DOI: 10.1016/S1097-2765(00)80139-2
[122]
Gosling J. Slaymaker S. Gu L. Tseng S. Zlot C. H. Young S. G. Rollins B. J. Charo I. F. MCP-1 deficiency reduces susceptibility to atherosclerosis in mice that overexpress human apolipoprotein B J Clin Invest 1999 103 6 773 8 DOI: 10.1172/JCI5624
[123]
Boring L. Gosling J. Cleary M. Charo I. F. Decreased lesion formation in CCR2-/- mice reveals a role for chemokines in the initiation of atherosclerosis Nature 1998 394 6696 894 7 DOI: 10.1038/29788
[124]
Piconi S. Pocaterra D. Rainone V. Cossu M. Masetti M. Rizzardini G. Clerici M. Trabattoni D. Maraviroc Reduces Arterial Stiffness in PI-Treated HIV-infected Patients Sci Rep 2016 6 28853 DOI: 10.1038/srep28853
[125]
Koenen R. R. Hundelshausen P. von Nesmelova I. V. Zernecke A. Liehn E. A. Sarabi A. Kramp B. K. Piccinini A. M. Paludan S. R. Kowalska M. A. Kungl A. J. Hackeng T. M. Mayo K. H. Weber C. Disrupting functional interactions between platelet chemokines inhibits atherosclerosis in hyperlipidemic mice Nat Med 2009 15 1 97 103 DOI: 10.1038/nm.1898
[126]
Lutgens E. Lievens D. Beckers L. Wijnands E. Soehnlein O. Zernecke A. Seijkens T. Engel D. Cleutjens J. Keller A. M. Naik S. H. Boon L. Oufella H. A. Mallat Z. Ahonen C. L. Noelle R. J. Winther M. P. de Daemen M. J. Biessen E. A. Weber C. Deficient CD40-TRAF6 signaling in leukocytes prevents atherosclerosis by skewing the immune response toward an antiinflammatory profile J Exp Med 2010 207 2 391 404 DOI: 10.1084/jem.20091293
[127]
Donners M. M. Beckers L. Lievens D. Munnix I. Heemskerk J. Janssen B. J. Wijnands E. Cleutjens J. Zernecke A. Weber C. Ahonen C. L. Benbow U. Newby A. C. Noelle R. J. Daemen M. J. Lutgens E. The CD40-TRAF6 axis is the key regulator of the CD40/CD40L system in neointima formation and arterial remodeling Blood 2008 111 9 4596 604 DOI: 10.1182/blood-2007-05-088906
[128]
Seijkens T. T. P. Tiel C. M. van Kusters P. J. H. Atzler D. Soehnlein O. Zarzycka B. Aarts S. Lameijer M. Gijbels M. J. Beckers L. Toom M. den Slutter B. Kuiper J. Duchene J. Aslani M. Megens R. T. A. Veer C. van 't Kooij G. Schrijver R. Hoeksema M. A. Boon L. Fay F. Tang J. Baxter S. Jongejan A. Moerland P. D. Vriend G. Bleijlevens B. Fisher E. A. Duivenvoorden R. Gerdes N. Winther M. P. J. de Nicolaes G. A. Mulder W. J. M. Weber C. Lutgens E. Targeting CD40-Induced TRAF6 Signaling in Macrophages Reduces Atherosclerosis J Am Coll Cardiol 2018 71 5 527 542 DOI: 10.1016/j.jacc.2017.11.055
[129]
Mach F. Schonbeck U. Sukhova G. K. Atkinson E. Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling Nature 1998 394 6689 200 3 DOI: 10.1038/28204
[130]
Schonbeck U. Sukhova G. K. Shimizu K. Mach F. Libby P. Inhibition of CD40 signaling limits evolution of established atherosclerosis in mice Proc Natl Acad Sci U S A 2000 97 13 7458 63 DOI: 10.1073/pnas.97.13.7458
[131]
Rashid I. Maghzal G. J. Chen Y. C. Cheng D. Talib J. Newington D. Ren M. Vajandar S. K. Searle A. Maluenda A. Lindstedt E. L. Jabbour A. Kettle A. J. Bongers A. Power C. Michaelsson E. Peter K. Stocker R. Myeloperoxidase is a potential molecular imaging and therapeutic target for the identification and stabilization of high-risk atherosclerotic plaque Eur Heart J 2018 39 35 3301 3310 DOI: 10.1093/eurheartj/ehy419
[132]
Tashiro Y. Sato K. Watanabe T. Nohtomi K. Terasaki M. Nagashima M. Hirano T. A glucagon-like peptide-1 analog liraglutide suppresses macrophage foam cell formation and atherosclerosis Peptides 2014 54 19 26 DOI: 10.1016/j.peptides.2013.12.015
[133]
Koreth J. Matsuoka K. Kim H. T. McDonough S. M. Bindra B. Alyea 3rd E. P. Armand P. Cutler C. Ho V. T. Treister N. S. Bienfang D. C. Prasad S. Tzachanis D. Joyce R. M. Avigan D. E. Antin J. H. Ritz J. Soiffer R. J. Interleukin-2 and regulatory T cells in graft-versus-host disease N Engl J Med 2011 365 22 2055 66 DOI: 10.1056/NEJMoa1108188
[134]
Matsuoka K. Koreth J. Kim H. T. Bascug G. McDonough S. Kawano Y. Murase K. Cutler C. Ho V. T. Alyea E. P. Armand P. Blazar B. R. Antin J. H. Soiffer R. J. Ritz J. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease Sci Transl Med 2013 5 179 ra43 DOI: 10.1126/scitranslmed.3005265
[135]
Saadoun D. Rosenzwajg M. Joly F. Six A. Carrat F. Thibault V. Sene D. Cacoub P. Klatzmann D. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis N Engl J Med 2011 365 22 2067 77 DOI: 10.1056/NEJMoa1105143
[137]
Tamai H. Katoh O. Suzuki S. Fujii K. Aizawa T. Takase S. Kurogane H. Nishikawa H. Sone T. Sakai K. Suzuki T. Impact of tranilast on restenosis after coronary angioplasty: tranilast restenosis following angioplasty trial (TREAT) Am Heart J 1999 138 5 Pt 1 968 75 DOI: 10.1016/S0002-8703(99)70025-6
[138]
Tamai H. Katoh K. Yamaguchi T. Hayakawa H. Kanmatsuse K. Haze K. Aizawa T. Nakanishi S. Suzuki S. Suzuki T. Takase S. Nishikawa H. Katoh O. The impact of tranilast on restenosis after coronary angioplasty: the Second Tranilast Restenosis Following Angioplasty Trial (TREAT-2) Am Heart J 2002 143 3 506 13 DOI: 10.1067/mhj.2002.120770
[139]
O'Donoghue M. L. Braunwald E. White H. D. Lukas M. A. Tarka E. Steg P. G. Hochman J. S. Bode C. Maggioni A. P. Im K. Shannon J. B. Davies R. Y. Murphy S. A. Crugnale S. E. Wiviott S. D. Bonaca M. P. Watson D. F. Weaver W. D. Serruys P. W. Cannon C. P. Investigators S.-T. Steen D. L. Effect of darapladib on major coronary events after an acute coronary syndrome: the SOLID-TIMI 52 randomized clinical trial JAMA 2014 312 10 1006 15 DOI: 10.1001/jama.2014.11061
[140]
Investigators S. White H. D. Held C. Stewart R. Tarka E. Brown R. Davies R. Y. Budaj A. Harrington R. A. Steg P. G. Ardissino D. Armstrong P. W. Avezum A. Aylward P. E. Bryce A. Chen H. Chen M. F. Corbalan R. Dalby A. J. Danchin N. Winter R. J. De Denchev S. Diaz R. Elisaf M. Flather M. D. Goudev A. R. Granger C. B. Grinfeld L. Hochman J. S. Husted S. Kim H. S. Koenig W. Linhart A. Lonn E. Lopez-Sendon J. Manolis A. J. Mohler 3rd E. R. Nicolau J. C. Pais P. Parkhomenko A. Pedersen T. R. Pella D. Ramos-Corrales M. A. Ruda M. Sereg M. Siddique S. Sinnaeve P. Smith P. Sritara P. Swart H. P. Sy R. G. Teramoto T. Tse H. F. Watson D. Weaver W. D. Weiss R. Viigimaa M. Vinereanu D. Zhu J. Cannon C. P. Wallentin L. Darapladib for preventing ischemic events in stable coronary heart disease N Engl J Med 2014 370 18 1702 11 DOI: 10.1056/NEJMoa1315878
[141]
Newby L. K. Marber M. S. Melloni C. Sarov-Blat L. Aberle L. H. Aylward P. E. Cai G. Winter R. J. de Hamm C. W. Heitner J. F. Kim R. Lerman A. Patel M. R. Tanguay J. F. Lepore J. J. Al-Khalidi H. R. Sprecher D. L. Granger C. B. Investigators S. Losmapimod, a novel p38 mitogen-activated protein kinase inhibitor, in non-ST-segment elevation myocardial infarction: a randomised phase 2 trial Lancet 2014 384 9949 1187 95 DOI: 10.1016/S0140-6736(14)60417-7
[142]
O'Donoghue M. L. Glaser R. Cavender M. A. Aylward P. E. Bonaca M. P. Budaj A. Davies R. Y. Dellborg M. Fox K. A. Gutierrez J. A. Hamm C. Kiss R. G. Kovar F. Kuder J. F. Im K. A. Lepore J. J. Lopez-Sendon J. L. Ophuis T. O. Parkhomenko A. Shannon J. B. Spinar J. Tanguay J. F. Ruda M. Steg P. G. Theroux P. Wiviott S. D. Laws I. Sabatine M. S. Morrow D. A. Investigators L.-T. Effect of Losmapimod on Cardiovascular Outcomes in Patients Hospitalized With Acute Myocardial Infarction: A Randomized Clinical Trial JAMA 2016 315 15 1591 9 DOI: 10.1001/jama.2016.3609
[143]
Kleveland O. Kunszt G. Bratlie M. Ueland T. Broch K. Holte E. Michelsen A. E. Bendz B. Amundsen B. H. Espevik T. Aakhus S. Damas J. K. Aukrust P. Wiseth R. Gullestad L. Effect of a single dose of the interleukin-6 receptor antagonist tocilizumab on inflammation and troponin T release in patients with non-ST-elevation myocardial infarction: a double-blind, randomized, placebo-controlled phase 2 trial Eur Heart J 2016 37 30 2406 13 DOI: 10.1093/eurheartj/ehw171
[144]
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 6 1154 1164 DOI: 10.1002/art.40084
[145]
Carroll M. B. Haller C. Smith C. Short-term application of tocilizumab during myocardial infarction (STAT-MI) Rheumatol Int 2018 38 1 59 66 DOI: 10.1007/s00296-017-3842-y
Share
Back to top