Next Article in Journal
Association of Major Adverse Cardiac Events and Beta-Blockers in Patients with and without Atherosclerotic Cardiovascular Disease: Long-Term Follow-Up Results of the T-SPARCLE and T-PPARCLE Registry in Taiwan
Previous Article in Journal
After SARS-CoV-2 Pandemics: New Insights into ICU-Acquired Pneumonia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Secondary Cardiovascular Prevention after Acute Coronary Syndrome: Emerging Risk Factors and Novel Therapeutic Targets

by
Angelo Silverio
1,†,
Francesco Paolo Cancro
1,†,
Luca Esposito
1,
Michele Bellino
1,
Debora D’Elia
1,
Monica Verdoia
2,
Maria Giovanna Vassallo
1,
Michele Ciccarelli
1,
Carmine Vecchione
1,
Gennaro Galasso
1,* and
Giuseppe De Luca
3,4
1
Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Baronissi, Italy
2
Division of Cardiology Ospedale degli Infermi, ASL, 13875 Biella, Italy
3
Department of Clinical and Experimental Medicine, Division of Cardiology, AOU Policlinico G. Martino, University of Messina, 98122 Messina, Italy
4
IRCCS Hospital Galeazzi- Sant’Ambrogio, 20161 Milano, Italy
*
Author to whom correspondence should be addressed.
These authors equally contributed to this work.
J. Clin. Med. 2023, 12(6), 2161; https://doi.org/10.3390/jcm12062161
Submission received: 5 February 2023 / Revised: 2 March 2023 / Accepted: 7 March 2023 / Published: 10 March 2023
(This article belongs to the Section Cardiovascular Medicine)

Abstract

:
The control of cardiovascular risk factors, the promotion of a healthy lifestyle, and antithrombotic therapy are the cornerstones of secondary prevention after acute coronary syndrome (ACS). However, many patients have recurrent ischemic events despite the optimal control of traditional modifiable risk factors and the use of tailored pharmacological therapy, including new-generation antiplatelet and lipid-lowering agents. This evidence emphasizes the importance of identifying novel risk factors and targets to optimize secondary preventive strategies. Lipoprotein(a) (Lp(a)) has emerged as an independent predictor of adverse events after ACS. New molecules such as anti-PCSK9 monoclonal antibodies, small interfering RNAs, and antisense oligonucleotides can reduce plasma Lp(a) levels and are associated with a long-term outcome benefit after the index event. The inflammatory stimulus and the inflammasome, pivotal elements in the development and progression of atherosclerosis, have been widely investigated in patients with coronary artery disease. More recently, randomized clinical trials including post-ACS patients treated with colchicine and monoclonal antibodies targeting cytokines yielded promising results in the reduction in major cardiovascular events after an ACS. Gut dysbiosis has also raised great interest for its potential pathophysiological role in cardiovascular disease. This evidence, albeit preliminary and needing confirmation by larger population-based studies, suggests the possibility of targeting the gut microbiome in particularly high-risk populations. The risk of recurrent ischemic events after ACS is related to the complex interaction between intrinsic predisposing factors and environmental triggers. The identification of novel risk factors and targets is fundamental to customizing patient clinical management with a precision medicine perspective.

1. Introduction

Coronary artery disease (CAD) is the leading cause of death worldwide, accounting for about 20% of cases in Europe [1]. In the last years, advances in preventive measures, pharmacological treatments [2,3,4], and percutaneous coronary interventional strategies [5,6] significantly improved life quality and expectancy after index myocardial infarction (MI), thus increasing the population of stable post-MI patients [7]. This novel epidemiological scenario emphasized the importance of implementing secondary preventive strategies, as these patients have a very high risk of further atherothrombotic events and mortality [8,9,10,11,12].
The control of traditional risk factors, the promotion of a healthy lifestyle, and antithrombotic therapy have been the cornerstones of secondary prevention after MI for decades. However, many patients continue to experience recurrent ischemic events despite lower residual cardiovascular risk and guideline-directed antithrombotic therapy [13].
In this new era of precision medicine, controlling traditional risk factors may not be sufficient for further reducing individual patient risk. Substantial attention has been focused on the identification of new risk factors [14,15,16] and new targets to develop personalized preventive strategies after an acute coronary syndrome (ACS). The aim of this review is to summarize the potential pathophysiological mechanisms underlying residual cardiovascular risk in post-ACS patients, describe novel risk factors associated with recurrent atherothrombotic events, and describe novel potential therapeutic targets that might be considered in this very high-risk clinical setting.

1.1. Risk of Recurrent Ischemic Events in Post-ACS Patients

The main pathogenetic mechanism of ACS is the rupture of thin-capped atherosclerotic plaque with a large lipid or necrotic core, with consequent exposure to prothrombotic material, a release of pro-thrombotic mediators, and ultimately, formation of a flow-limiting thrombus [17]. In the last years, we have understood that this mechanism is only a part of a much more complex pathophysiological framework. Intravascular imaging studies showed that subclinical plaque ruptures often occur in patients with CAD [18,19] and that some “vulnerable plaques” may evolve into a condition of stability, probably through the cycles of rupture and healing [20,21]. Indeed, many stable plaques causing significant flow-limiting stenoses show morphological features of previous plaque rupture and healing [22,23]. For reasons that we do not fully understand, in some patients, prothrombotic activation prevails over the healing response, leading to the formation of a flow-limiting thrombus and the clinical manifestation of an ACS. This pathophysiological evidence suggests that patients with a history of ACS represent a vulnerable niche of patients with CAD characterized by a biological predisposition to the occurrence of acute atherothrombotic events.
Prior studies showed that post-ACS patients have a higher risk of ischemic events compared to stable CAD patients with no history of ACS [24,25]. The risk of recurrent events is higher during the first year after the index event but continues to increase over later years [9,26]. Data from large-scale real-world registries reported long-term annual rates of recurrent events between 4.4% and 6.7% in stable post-ACS patients [9,27]. All this evidence confirms that the “residual cardiovascular risk” remains clinically meaningful in post-ACS patients despite the adoption of optimal pharmacological therapy.

1.2. Residual Cardiovascular Risk after ACS: Limitations of Traditional Risk Factors

The estimate of individual residual risk after ACS is a complex multiparametric process that should theoretically account for many aspects including lifestyle habits, thrombotic cascade, lipoprotein particles, and inflammatory cells. Traditionally, the thrombotic and low-density lipoprotein cholesterol (LDL-C) pathophysiological pathways have been identified as the leading mechanisms involved in ACS recurrence, and many drugs have been developed for controlling them. The control of thrombotic phenomena after ACS patients refers to a holistic prothrombotic milieu that goes beyond the simple prevention of stent thrombosis, encompassing instead the risk of the destabilization of non-culprit lesions [28]. Dual antiplatelet therapy (DAPT) is the cornerstone of medical therapy after ACS. To overcome “residual thrombotic risk”, long-term DAPT strategies have also been developed for selected very-high-risk patient cohorts [29]. In the PEGASUS-TIMI 54 (Prevention of Cardiovascular Events in Patients with Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin–Thrombolysis in Myocardial Infarction 54) trial, long-term DAPT with ticagrelor in addition to aspirin provided a 16% reduction in adverse ischemic events in post-MI patients [29]. The benefits of a prolonged DAPT were particularly relevant in patients with high atherosclerotic burden, such as those with coexistent peripheral artery disease [30]. Furthermore, the benefit of prolonged DAPT in PEGASUS was observed regardless of prior coronary stenting [31]. The reduction in ischemic events was counterbalanced by an increase in major bleeding, albeit with no difference in terms of fatal and intracranial bleeding.
An alternative strategy for the long-term prevention of recurrent thrombotic events is the combination of antiplatelet and low-dose anticoagulant agents. In the COMPASS (Cardiovascular Outcomes for People Using Anticoagulation Strategies) trial, the use of low-dose rivaroxaban (2.5 mg twice daily) in addition to aspirin in patients with atherosclerotic disease (including 61.8% of post-MI patients) significantly reduced the incidence of ischemic events at follow-up at the cost of higher rates of major bleeding [32].
These results emphasize the importance of a personalized approach based on the assessment of net ischemic and bleeding risks instead of a “one-size fits all” strategy. This may be difficult due to the substantial overlap of the predictors of ischemic and bleeding events and the difficulties inherent in balancing their individual contributions in each patient [33,34,35].
LDL-C has been recognized as a causal factor for atherosclerotic disease [36]. Landmark trials showed that LDL-c-lowering agents, such as statins and ezetimibe, significantly reduced the incidence of adverse ischemic events in patients with CAD [37,38]. However, several studies on lipid-lowering therapies showed a persistent residual risk of ischemic events despite an aggressive LDL-C control [37,39,40]. The issue of “residual cholesterol risk” has been partially addressed with the introduction of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors that dramatically reduce LDL-c levels when added to statin therapy, showing a significant reduction in cardiovascular events in patients with CAD [41,42]. However, a subanalysis from the FOURIER (Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk) trial showed that in patients treated with PCSK9 inhibitors, although an aggressive reduction in LDL-c up to 20–30 mg/dL was achieved, the high-sensitivity C-reactive protein (hsCRP) remained a strong predictor of recurrent ischemic events, confirming the central role of inflammation in atherogenesis in spite of optimal LDL-c levels [13,43].
These data demonstrated that we are still missing some pieces of the complex puzzle of residual cardiovascular risks. We will try to discuss the role of emerging risk factors of recurrent atherothrombotic events in post-ACS patients below, with a focus on potential therapeutic strategies.

2. Lipoprotein(a)

Lipoprotein(a) (Lp(a)) is a plasma lipoprotein composed of an LDL-rich particle with an apolipoprotein B100 molecule bound via a single disulphide bond to apolipoprotein(a) (apo(a)), a plasminogen-like glycoprotein. This is a pathogenetic component of Lp(a) containing, similarly to plasminogen, several three-dimensional domains called kringles [44].

2.1. Genetics and Activity of Lp(a)

Lp(a) concentrations vary from <0.1 mg/dL to >300 mg/dL (<0.2 to 750 nmol/L) and are mostly (>90%) determined by genetic variability in the LPA locus with negligible influence attributable to dietary and environmental factors [44]. Kringle-IV (K-IV) polymorphisms are responsible for the great variability in the plasma concentration of Lp(a). A low number (<23) of K-IV repeat polymorphisms are associated with smaller apo(a) isoforms and higher plasma Lp(a) levels than a higher number of K-IV repeats and larger apo(a) isoforms [45].
In vitro and animal studies have emphasized the central role of Lp(a) in different pathophysiological pathways of atherosclerosis, including foam cell formation, smooth muscle cell proliferation, inflammation, and plaque instability [46]. Indeed, Lp(a) not only appears to contribute to atherosclerosis through the same mechanisms of LDL but also presents additional apo(a)-related effects. Apo(a), due to its close homology with plasminogen, competitively inhibits fibrinolysis by binding fibrin and thus promotes thrombotic phenomena [47]. Moreover, apo(a) contains lysine binding sites, permitting its interaction with the exposed portions of the damaged endothelium and its penetration and accumulation in subintimal spaces and conferring a particular propensity of Lp(a) for vascular walls [48]. Furthermore, Lp(a) is one of the main carriers of oxidized phospholipids (Ox-PL) within the vessel wall, enhancing atherogenesis and inflammation [49]. In an in vivo study, patients with elevated Lp(a) showed an increased accumulation of 18-fluorodeoxyglucose in the carotid arteries and aorta, indicating the inflammation of arterial walls. In addition, the monocytes of these patients showed increased responsiveness in the production of proinflammatory cytokines and a marked tendency to transmigrate, which could reflect their capacity to penetrate the vessel wall [50]. Moreover, Lp(a) seems to upregulate the expression of proinflammatory genes and promote the release of inflammatory cytokines and chemokines, such as interleukin-8 and monocyte chemoattractant protein-1 [51,52].

2.2. Lipoprotein(a) as a Cardiovascular Risk Factor

Over the past two decades, many studies conducted in populations without a history of atherosclerotic cardiovascular disease (ASCVD), including observational studies, meta-analyses, mendelian randomization studies, and genome-wide association studies, have shown that high levels of Lp(a) are linearly associated with an increased risk of developing ASCVD [53,54,55,56,57]. The Copenhagen City Heart Study, which included 9330 subjects followed for 10 years, showed a linear increase in the risk of cardiovascular events as the level of Lp(a) increased, reaching a 3.6-fold greater risk in subjects with extremely elevated Lp(a) levels (>120 mg/dL) [53]. A recent Mendelian randomization study matching 9015 patients with acute MI and 8629 controls from the Pakistan Risk of Myocardial Infarction Study (PROMIS) suggested that both smaller apo(a) isoform size and increased Lp(a) concentration are independent and causal risk factors for CAD [58]. An individual patient data meta-analysis, which included 29,609 patients from seven randomized clinical trials, showed that high Lp(a) levels are linearly associated with the risk of cardiovascular adverse events independently of LDL levels and statin treatment [59]. This evidence established Lp(a) as a risk factor and a potential therapeutic target for primary prevention in healthy subjects. Indeed, the European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) recommend measuring Lp(a) at least once in adults, preferably in the first lipid profile, and they suggest a desirable plasma level of <30 mg/dL (IIaC) [60].

2.3. Usefulness of Lipoprotein(a) in Secondary Prevention

In patients with established CAD, the prognostic role of Lp(a) levels has long been debated. Recently, several studies show the association between Lp(a) and adverse events in ASCVD settings [8,42,61,62,63,64,65,66,67,68]. A meta-analysis that included three randomized trials and eight secondary prevention studies, including a total of 18,978 patients, found that subjects with Lp(a) levels above the highest quintile were at increased risk of cardiovascular events but with a high heterogeneity between studies. However, this correlation was not significant for subjects whose plasma LDL levels were <130 mg/dL [65].
In the Copenhagen General Population Study, the 5-year risk for recurrent major adverse cardiovascular events (MACEs) in subjects with pre-existing ASCVD was higher for Lp(a) levels ≥50 mg/dL, independently of LDL plasma levels [69]. Recently, a pre-specified analysis from the ODISSEY Outcomes trial, which randomized 18,924 post-ACS patients on high-intensity statin treatment relative to alirocumab vs. placebo, found that baseline Lp(a) levels were linearly associated with MACE (composite of coronary death, MI, stroke, or unstable angina), fatal and non-fatal MI, and cardiovascular death, independently of baseline LDL values. They also found that the reduction in Lp(a) levels given by alirocumab independently reduced the risk of MACE [66]. Furthermore, a recent observational study including 12,064 consecutive unselected real-world patients who underwent PCI with drug-eluting stents showed that high plasma levels of Lp(a) were significantly and independently associated with a higher risk of recurrent ischemic events and repeated revascularization at a long-term follow-up [67]. More recently, Lp(a) emerged as an independent predictor of recurrent MI after ACS, particularly in patients without diabetes [8,64].
These pieces of evidence emphasize the importance of including baseline Lp(a) plasma levels in the prognostic stratification of patients with established ASCVD and the increasing need for pharmacological agents to target this molecule.

2.4. Current and Emerging Therapies for High Lipoprotein(a)

To date, the only available and effective therapy for reducing Lp(a) levels is lipoprotein apheresis, indicated for secondary prevention in patients with extremely high Lp(a) levels and recurrent cardiovascular events despite optimal medical therapy. Since lipoprotein apheresis is an invasive, expensive, and not risk-free procedure, it is used only in a few selected patients in third-level dedicated centers [70].
There are no approved pharmacological agents for Lp(a) nor preliminary data from randomized clinical trials on Lp(a) reduction. Although statins are a milestone treatment of dyslipidaemia, they have failed to show any effect in lowering Lp(a) plasma concentrations [71]. In the AIM-HIGH (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglyceride and Impact on Global Health Outcomes) trial, niacin reduced Lp(a) plasma levels by 21% but without a significant influence on the cardiovascular risk [62].
Mipomersen, an antisense oligonucleotide (ASO) targeting apolipoprotein B synthesis, has been shown to reduce Lp(a) levels by 26% in four phase 3 trials, but its clinical impact remains unclear [72].
PCSK9 inhibitors are currently the most promising molecules liable to reduce the plasma levels of Lp(a). A post hoc analysis from the FOURIER trial, in which 25,096 patients with stable CAD were randomized relative to evolocumab vs. placebo, showed a reduction in Lp(a) plasma levels of 27% with evolocumab associated with a significant decrease in adverse cardiovascular outcomes (composite of coronary heart disease, death, myocardial infarction, and urgent revascularization) with an absolute risk reduction of 2.49% and number needed to treat of 40 up to 3 years in patients with Lp(a) plasma values above the median 37 nmol/L serum level [68]. Similarly, a post hoc analysis of the ODISSEY Outcomes trial showed that alirocumab was associated with a reduction in Lp(a) plasma levels of 23%, leading to a significant reduction in long-term cardiovascular adverse events in patients with ACS; however, this outcome benefit was principally due to the reduction in non-fatal MI, ischemia-driven coronary revascularization, and major PAD events rather than mortality. Moreover, in this study, baseline Lp(a) levels were independently associated not only with the patients’ cardiovascular risk but also with the expected benefit from alirocumab treatment [73].
The ORION-10 and ORION-11 trials showed that Inclisiran, a small-interfering RNA (siRNA) PCSK9 inhibitor, was associated with a 20% reduction in Lp(a) levels, but the trial did not investigate the effect on cardiovascular outcomes [74]. To date, PCSK9 inhibitors are recommended by ESC/EAS guidelines in subjects with familial hypercholesterolemia and high Lp(a) (IIa) [75].
Eventually, newer emerging therapies targeting specifically apo(a) via ASOs (Pelacarsen) and siRNAs (Olpasiran) are being investigated in phase II and III trials, with encouraging preliminary results in terms of safety and efficacy in reducing plasma Lp(a) levels, and data on the clinical outcome are expected in the coming years [76,77,78].
Table 1 summarizes the emerging pharmacological therapies for lowering Lp(a) plasma levels.

3. Inflammation

Atherosclerosis is an unsolved progressive chronic inflammatory process related to cholesterol deposits within the arterial intima, which plays a central role in developing type I MI [79]. The outcome benefit associated with anti-inflammatory therapies in patients with established ASCVD emphasizes the importance of inflammation in the development and progression of atherosclerosis independently of the optimal control of other traditional cardiovascular risk factors, including LDL cholesterol [80,81,82,83].

3.1. Inflammation, Inflammasome, and Atherosclerosis

The damaged and activated endothelium in the initial phase of atherosclerosis favors the recruitment of circulating monocytes, T-cells, and mast cells that infiltrate the atheroma [84,85] and participates in the release of a high quantity of inflammatory cytokines [86,87]. In this environment, there is a vicious cycle related to apoptised intimal cells, which are not engulfed and efficiently cleared by macrophages and favor the build-up of the necrotic core. The persistence of these necrotic elements further promotes the activation and release of more inflammatory molecules by the cells of innate immunity [88,89]. Therefore, inflammation plays a crucial role in the pathogenesis and progression of atherosclerosis and actively participates in increasing the residual risk in patients with history of ACS.
A meta-analysis involving 160,309 subjects from 54 long-term prospective studies showed a continuous association between CRP levels and CAD (RR 1.32; 95% CI 1.18 to 1.49) [90]. Moreover, a secondary analysis from the CANTOS (The Canakinumab Anti-Inflammatory Thrombosis Outcomes Study) trial, which evaluated the efficacy of canakinumab vs. placebo in 1061 patients with a history of MI, showed that patients who reached high-sensitivity CRP levels <2 mg/dL had a significant reduction (25%) in MACE (HR: 0.75, 95% CI 0.66 to 0.85) than those who had high-sensitivity CRP plasma levels >2 mg/dL [91].
In recent years, the inflammasome, which indicates the mechanism for caspase-1 activation and interleukin-1b processing, emerged as one of the main actors and facilitators of the inflammatory process [92,93]. In particular, the nucleotide-binding oligomerization domain and leucine-rich repeat-containing receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome have emerged to be big players in the development of atherosclerosis [93].

3.2. NLRP3 in Atherosclerosis and Cardiovascular Risk

Different stimuli can activate NLRP3 [94]. In the peculiar context of atherosclerosis, shear stress on the endothelial wall, cholesterol crystals, and Ox-PL seem to be the primers of NLRP3 activation [95,96,97,98]. After activation, NLRP3 enhances via the caspase-1 activation of proinflammatory cytokine interleukin-1β (IL-1β), which is the circulating form of interleukin-1 (IL-1) cytokine [99]. IL-1 exhibits a wide variety of cardiovascular effects: 99 It induces the production of additional proinflammatory mediators, such as tumor necrosis factors, interleukin-6 (IL-6), and chemoattractant molecules, implicated in the tissue invasion of inflammatory cells into the atheroma. IL-6 drives the secretion of acute-phase reactants, such as fibrinogen and plasminogen activator inhibitor-1 (PAI-1), facilitating prothrombotic and antifibrinolytic impairment [100,101]. Notably, IL-1 facilitates its own gene expression, maintaining a self-sustained inflammatory response [102].
In experimental studies, IL-1 worsened ischemic-reperfusion injury after MI and enhanced negative cardiac remodeling; conversely, IL-1 inhibition mitigated negative cardiac remodeling and decreased acute phase protein release [103,104,105,106]. This evidence highlights the contribution of the NLRP3-mediated inflammatory cascade in the development and progression of atherosclerosis in patients with established ASCVD, thus increasing their residual risk of recurrent ischemic events.

3.3. Anti-Inflammatory Therapies in ASCVD Patients

Anti-inflammatory therapies have been proposed as potential game changers in the future management of patients with established ASCVD, and NLRP3 inflammasome and its products could be targets for future drugs.
Among the anti-inflammatory agents, colchicine has been widely studied in patients with established ASCVD [82,83,107]. By inhibiting polymerization and microtubule formation, colchicine could interfere with NLRP3 formation [108]. Data from two large randomized controlled trials, COLCOT and LoDoCo2, showed that the daily 0.5 mg of colchicine significantly reduced the risk of adverse cardiovascular events by 23% and 30%, respectively, in cohorts of CAD patients [82,83]. Recently, a meta-analysis including 12,869 patients from 11 randomized controlled trials reported that the use of colchicine in patients with CAD was effective in reducing major adverse cardiovascular and cerebrovascular events (with an acceptable risk profile in terms of drug-related adverse events), but it did not significantly reduce the risk of cardiovascular and non-cardiovascular death. Moreover, a prior meta-analysis showed a higher, but not significant, incidence of non-cardiovascular deaths in patients treated with low-dose colchicine [109]. The 2021 ESC guidelines on cardiovascular disease prevention suggest that low-dose colchicine (0.5 mg daily) may be considered in secondary prevention, especially if the other risk factors are insufficiently controlled or if recurrent cardiovascular events occur under optimal therapy (IIb) [110].
Canakinumab, a human monoclonal antibody that can hamper the interaction between IL-1β and its receptor, has been tested in the CANTOS trial, which randomized 10,061 patients with established CAD to different doses of subcutaneous canakinumab (50 mg, 150 mg, and 300 mg every 3 months) vs. the placebo. The 150 mg dose significantly reduced the risk of adverse cardiovascular events vs. the placebo. However, the effect on the primary endpoint (composite of non-fatal MI, non-fatal stroke, and cardiovascular death) was largely driven by a reduction in non-fatal MI, whereas a non-significant reduction in long-term mortality was observed [81]. Therefore, these results should be interpreted with caution since non-fatal MI is not validated as a valid surrogate of all-cause or cardiovascular mortality in clinical trials enrolling patients with CAD [111]. Moreover, a CANTOS pre-specified analysis showed that canakinumab led to a significant reduction in IL-6 and hs-CRP [91]. However, caution is necessary since canakinumab may alter immune homeostasis, as it acts systemically; in fact, patients treated with this drug reported a higher incidence of infections, even fatal ones, compared to the placebo [81].
Methotrexate (MTX), a competitive folic acid antagonist, is a widely used drug in rheumatic diseases. A meta-analysis including 10 cohort studies conducted on patients with rheumatoid arthritis receiving MTX showed a significant reduction in cardiovascular events [112]. However, the recent Cardiovascular Inflammation Reduction trial (CIRT), conducted in 4786 patients with stable atherosclerosis receiving low doses of MTX (15–20 mg), did not show a significant reduction in cardiovascular events, including hs-CRP, IL-1β, and IL-6 plasma levels [113]. MTX delivered in LDL nanoparticles is currently being studied in ASCVD patients in two ongoing trials, including stable CAD patients and STEMI patients (NCT04616872; NCT 03516903).
Anakinra, an IL-1 receptor antagonist, significantly reduced the acute inflammatory response, as demonstrated in a phase II trial including 99 patients with STEMI and acute decompensated heart failure; however, no significant differences were found in terms of clinical outcomes compared to the placebo [114].
Tocilizumab and Ziltivekimab, two human IgG1 monoclonal antibodies targeting IL-6 and the IL-6 ligand, have recently been studied in two different randomized controlled trials [115,116]. In the ASSAIL-MI (ASSessing the effect of Anti-IL-6 treatment in MI) trial, Tocilizumab at a 280 mg dose increased myocardial salvage in STEMI patients [115]. In the Trial to Evaluate Reduction in Inflammation in Patients with Advanced Chronic Renal Disease Utilizing Antibody Mediated IL-6 Inhibition (RESCUE), Ziltivekimab, a monoclonal antibody inhibiting the IL-6 ligand, markedly reduced the serum biomarkers of inflammation and thrombosis in patients with advanced chronic kidney disease [116].
Arglabin is a molecule isolated from different plant species that seems to interfere with the stimulation of NLRP3 in macrophages, and in animal models, it has been shown to significantly reduce NLRP3-related inflammatory molecules and has a beneficial impact on lipid profiles [117]. MCC950 is a selective NLRP3 inhibitor that, in several animal models, has been shown to decrease IL-1β production, reduce atherosclerotic plaque size, and reduce the infarct area in experimental models of myocardial infarction [80,118].
Although anti-inflammatory therapies could represent the way forward for the treatment of patients with established ASCVD, their long-term efficacy and safety is still unknown, and some of them are forbiddingly expensive. In the forthcoming years, it will be necessary to find drugs that are both safe and effective up until the long-term follow-up and with affordable costs.
Anti-inflammatory and immune therapies tested in patients with established CAD are summarized in Table 2.

4. Gut Microbiota

The gut microbiota consists of innumerable microbes that colonize the entire digestive tract from the stomach down to the colon. Its composition could be influenced by genetic, dietary, and environmental factors [119,120]. The bacteria and viruses that make it up cause a continuous inflammatory trigger, which is outweighed by the host’s innate immunity [121]. Gut dysbiosis, a condition in which the microbial composition of microbiota is altered, has been linked to numerous pathological conditions as well as intestinal diseases, type 2 diabetes, obesity, and ischemic stroke [122]. Recently, emerging interest has grown on the possibility that the permeation of specific bacterial products of altered microbiota, such as lipopolysaccharide (LPS) and trimethylamine-N-oxide (TMAO), within the systemic circulation may promote the onset of atherothrombotic and ischemic cardiovascular events [123]. Animal models with ApoE−/− mice showed that increased plasma levels of TMAO lead to an augmented atherosclerotic burden [124]; in addition, a reduction in TMAO plasma levels showed an abortion of the atherosclerotic effect with a decrease in plaque size [125]. However, clinical studies in humans showed conflicting results upon the correlation between TMAO, atherosclerotic burden, and cardiovascular events; thus, further studies are needed to clarify the role of TMAO in the development and progression of atherosclerosis [126,127,128,129,130]. A nested case–control study from the PEGASUS-TIMI 54 trial including 597 post-MI patients and 1206 controls showed that higher TMAO levels were associated with cardiovascular death and stroke but not with recurrent MI in patients with a history of prior MI [131].
Emerging evidence suggests that low-grade endotoxemia driven by circulating LPS could lead to atherothrombosis and cardiovascular disease [129]. LPS is detected by the receptors of the immune system, such as toll-like receptors, a group of membrane receptors involved in atherosclerotic and thrombotic processes [132]. Previous studies linked the LPS circulating levels with a long-term risk of developing ASCVD; however, these studies were conducted in healthy subjects, and the role of LPS in patients with established ASCVD has not yet been evaluated [133,134,135].
A case–control study including 50 patients diagnosed with STEMI, 50 patients with stable CAD, and 50 healthy controls demonstrated an augmented gut permeability during myocardial ischemia and, consequently, a significant role of circulating LPS in favoring thrombus growth during STEMI [136]. Recently, a study conducted in a population of post-STEMI patients showed that the permeation of LPS in the systemic circulatory system during the acute phase of MI significantly correlated with future adverse cardiovascular outcomes at 3 years follow-up [137]. Despite these results, further efforts are needed to better understand the potential role of gut dysbiosis on the individual cardiovascular risk of patients with established ASCVD.
The next challenge could be the possibility of directly targeting the gut microbiome in high-risk patients. Indeed, fibre-rich diets such as a Mediterranean diet, prebiotics, probiotics, and faecal microbiome transplantation have been shown to positively regulate gut permeability and can reduce circulating levels of bacterial products. However, this evidence is limited to animal models and small-sampled clinical studies [123,129].

5. Conclusions and Future Perspectives

Secondary prevention in post-ACS patients is a major health problem. Despite significant improvements in behavioral, pharmacological, and invasive treatments, these patients still show a high risk of recurrent atherothrombotic events and mortality. The residual risk of post-ACS patients is related to the interaction between intrinsic predisposing factors and environmental triggers, combined in a complex pathophysiological framework. The impossibility to simplify an articulated biological phenomenon calls for a tailored clinical approach for post-ACS patients, including the identification of novel risk factors and targets.
Lp(a) is a non-traditional cardiovascular risk factor that has progressively assumed relevance over the last years, and ongoing trials will clarify the best use of Lp(a) as a therapeutic target after ACS.
Chronic inflammation is recognized as a key pathogenetic element of atherosclerosis, and recent evidence from large-scale randomized trials supports the use of anti-inflammatory drugs for secondary prevention in patients with recurrent ACS. However, further studies are needed to definitely clarify the safety and cost-effectiveness profile of these therapeutic options.
The characterization of the systemic implications of gut dysbiosis is a new field of translational medicine. The link between dysfunctional microbiota and atherothrombosis could provide the rationale for novel therapies in the future.
The mission of the cardiology community is to pursue the search for novel risk factors and targets, develop new therapies, and ultimately improve patient outcomes after ACS.

Author Contributions

A.S., F.P.C., L.E. and M.B.: Drafting of the manuscript. D.D., M.V., M.G.V. and M.C.: Critically revising the manuscript. C.V., G.G. and G.D.L.: Final approval of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

No conflicts of interest or any financial support to declare.

References

  1. Nichols, M.; Townsend, N.; Scarborough, P.; Rayner, M. Cardiovascular disease in Europe: Epidemiological update. Eur. Heart J. 2013, 34, 3028–3034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. De Luca, G.; Suryapranata, H.; Stone, G.W.; Antoniucci, D.; Tcheng, J.E.; Neumann, F.J.; Bonizzoni, E.; Topol, E.J.; Chiariello, M. Relationship between patient’s risk profile and benefits in mortality from adjunctive abciximab to mechanical revascularization for ST-segment elevation myocardial infarction: A meta-regression analysis of randomized trials. J. Am. Coll. Cardiol. 2006, 47, 685–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Verdoia, M.; Schaffer, A.; Barbieri, L.; Cassetti, E.; Piccolo, R.; Galasso, G.; Marino, P.; Sinigaglia, F.; De Luca, G. Benefits from new ADP antagonists as compared with clopidogrel in patients with stable angina or acute coronary syndrome undergoing invasive management: A meta-analysis of randomized trials. J. Cardiovasc. Pharmacol. 2014, 63, 339–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Costa, F.; Montalto, C.; Branca, M.; Hong, S.J.; Watanabe, H.; Franzone, A.; Vranckx, P.; Hahn, J.Y.; Gwon, H.C.; Feres, F.; et al. Dual antiplatelet therapy duration after percutaneous coronary intervention in high bleeding risk: A meta-analysis of randomized trials. Eur. Heart J. 2022, ehac706. [Google Scholar] [CrossRef] [PubMed]
  5. De Luca, G.; Smits, P.; Hofma, S.H.; Di Lorenzo, E.; Vlachojannis, G.J.; Van’t Hof, A.W.J.; van Boven, A.J.; Kedhi, E.; Stone, G.W.; Suryapranata, H. Everolimus eluting stent vs first generation drug-eluting stent in primary angioplasty: A pooled patient-level meta-analysis of randomized trials. Int. J. Cardiol. 2017, 244, 121–127. [Google Scholar] [CrossRef]
  6. De Luca, G.; Schaffer, A.; Wirianta, J.; Suryapranata, H. Comprehensive meta-analysis of radial vs femoral approach in primary angioplasty for STEMI. Int. J. Cardiol. 2013, 168, 2070–2081. [Google Scholar] [CrossRef]
  7. Szummer, K.; Wallentin, L.; Lindhagen, L.; Alfredsson, J.; Erlinge, D.; Held, C.; James, S.; Kellerth, T.; Lindahl, B.; Ravn-Fischer, A.; et al. Improved outcomes in patients with ST-elevation myocardial infarction during the last 20 years are related to implementation of evidence-based treatments: Experiences from the SWEDEHEART registry 1995-2014. Eur. Heart J. 2017, 38, 3056–3065. [Google Scholar] [CrossRef] [Green Version]
  8. Galasso, G.; De Angelis, E.; Silverio, A.; Di Maio, M.; Cancro, F.P.; Esposito, L.; Bellino, M.; Scudiero, F.; Damato, A.; Parodi, G.; et al. Predictors of Recurrent Ischemic Events in Patients With ST-Segment Elevation Myocardial Infarction. Am. J. Cardiol. 2021, 159, 44–51. [Google Scholar] [CrossRef]
  9. De Luca, G.; Dirksen, M.T.; Spaulding, C.; Kelbæk, H.; Schalij, M.; Thuesen, L.; van der Hoeven, B.; Vink, M.A.; Kaiser, C.; Musto, C.; et al. Time course, predictors and clinical implications of stent thrombosis following primary angioplasty. Insights from the DESERT cooperation. Thromb. Haemost. 2013, 110, 826–833. [Google Scholar]
  10. Jernberg, T.; Hasvold, P.; Henriksson, M.; Hjelm, H.; Thuresson, M.; Janzon, M. Cardiovascular risk in post-myocardial infarction patients: Nationwide real world data demonstrate the importance of a long-term perspective. Eur. Heart J. 2015, 36, 1163–1170. [Google Scholar] [CrossRef] [Green Version]
  11. Kernis, S.J.; Harjai, K.J.; Stone, G.W.; Grines, L.L.; Boura, J.A.; Yerkey, M.W.; O’Neill, W.; Grines, C.L. The incidence, predictors, and outcomes of early reinfarction after primary angioplasty for acute myocardial infarction. J. Am. Coll. Cardiol. 2003, 42, 1173–1177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. De Rosa, R.; Silverio, A.; Varricchio, A.; De Luca, G.; Di Maio, M.; Radano, I.; Belmonte, M.; De Angelis, M.C.; Moscarella, E.; Citro, R.; et al. Meta-Analysis Comparing Outcomes After Everolimus-Eluting Bioresorbable Vascular Scaffolds Versus Everolimus-Eluting Metallic Stents in Patients with Acute Coronary Syndromes. Am. J. Cardiol. 2018, 122, 61–68. [Google Scholar] [CrossRef] [PubMed]
  13. Ridker, P.M. Residual inflammatory risk: Addressing the obverse side of the atherosclerosis prevention coin. Eur. Heart J. 2016, 37, 1720–1722. [Google Scholar] [CrossRef] [PubMed]
  14. De Luca, G.; Verdoia, M.; Cassetti, E.; Schaffer, A.; Cavallino, C.; Bolzani, V.; Marino, P. High fibrinogen level is an independent predictor of presence and extent of coronary artery disease among Italian population. J. Thromb. Thrombolysis 2011, 31, 458–463. [Google Scholar] [CrossRef] [PubMed]
  15. Verdoia, M.; Barbieri, L.; Di Giovine, G.; Marino, P.; Suryapranata, H.; De Luca, G. Neutrophil to Lymphocyte Ratio and the Extent of Coronary Artery Disease: Results From a Large Cohort Study. Angiology 2016, 67, 75–82. [Google Scholar] [CrossRef]
  16. Barbieri, L.; Verdoia, M.; Schaffer, A.; Marino, P.; Suryapranata, H.; De Luca, G. Impact of sex on uric acid levels and its relationship with the extent of coronary artery disease: A single-centre study. Atherosclerosis 2015, 241, 241–248. [Google Scholar] [CrossRef] [PubMed]
  17. Virmani, R.; Burke, A.P.; Farb, A.; Kolodgie, F.D. Pathology of the vulnerable plaque. J. Am. Coll. Cardiol. 2006, 47, C13–C18. [Google Scholar] [CrossRef] [Green Version]
  18. Roleder-Dylewska, M.; Gasior, P.; Hommels, T.M.; Roleder, T.; Berta, B.; Ang, H.Y.; Ng, J.C.K.; Hermanides, R.S.; Fabris, E.; IJsselmuiden, A.J.J.; et al. Morphological characteristics of lesions with thin cap fibroatheroma-a substudy from the COMBINE (OCT-FFR) trial. Eur. Heart J. Cardiovasc. Imaging 2022, jeac218. [Google Scholar] [CrossRef]
  19. Kedhi, E.; Berta, B.; Roleder, T.; Hermanides, R.S.; Fabris, E.; IJsselmuiden, A.J.J.; Kauer, F.; Alfonso, F.; von Birgelen, C.; Escaned, J.; et al. Thin-cap fibroatheroma predicts clinical events in diabetic patients with normal fractional flow reserve: The COMBINE OCT-FFR trial. Eur. Heart J. 2021, 42, 4671–4679. [Google Scholar] [CrossRef]
  20. Arbab-Zadeh, A.; Fuster, V. The myth of the “vulnerable plaque”: Transitioning from a focus on individual lesions to atherosclerotic disease burden for coronary artery disease risk assessment. J. Am. Coll. Cardiol. 2015, 65, 846–855. [Google Scholar] [CrossRef] [Green Version]
  21. Kubo, T.; Maehara, A.; Mintz, G.S.; Doi, H.; Tsujita, K.; Choi, S.Y.; Katoh, O.; Nasu, K.; Koenig, A.; Pieper, M.; et al. The dynamic nature of coronary artery lesion morphology assessed by serial virtual histology intravascular ultrasound tissue characterization. J. Am. Coll. Cardiol. 2010, 55, 1590–1597. [Google Scholar] [CrossRef] [Green Version]
  22. Burke, A.P.; Kolodgie, F.D.; Farb, A.; Weber, D.K.; Malcom, G.T.; Smialek, J.; Virmani, R. Healed plaque ruptures and sudden coronary death: Evidence that subclinical rupture has a role in plaque progression. Circulation 2001, 103, 934–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Rioufol, G.; Finet, G.; Ginon, I.; André-Fouët, X.; Rossi, R.; Vialle, E.; Desjoyaux, E.; Convert, G.; Huret, J.F.; Tabib, A. Multiple atherosclerotic plaque rupture in acute coronary syndrome: A three-vessel intravascular ultrasound study. Circulation 2002, 106, 804–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Bhatt, D.L.; Eagle, K.A.; Ohman, E.M.; Hirsch, A.T.; Goto, S.; Mahoney, E.M.; Wilson, P.W.; Alberts, M.J.; D’Agostino, R.; Liau, C.S.; et al. Comparative determinants of 4-year cardiovascular event rates in stable outpatients at risk of or with atherothrombosis. JAMA 2010, 304, 1350–1357. [Google Scholar] [CrossRef] [Green Version]
  25. Kereiakes, D.J.; Yeh, R.W.; Massaro, J.M.; Cutlip, D.E.; Steg, P.G.; Wiviott, S.D.; Mauri, L. DAPT Score Utility for Risk Prediction in Patients With or Without Previous Myocardial Infarction. J. Am. Coll. Cardiol. 2016, 67, 2492–2502. [Google Scholar] [CrossRef]
  26. Smolina, K.; Wright, F.L.; Rayner, M.; Goldacre, M.J. Long-term survival and recurrence after acute myocardial infarction in England, 2004 to 2010. Circ. Cardiovasc. Qual. Outcomes 2012, 5, 532–540. [Google Scholar] [CrossRef] [Green Version]
  27. Kern, D.M.; Mellström, C.; Hunt, P.R.; Tunceli, O.; Wu, B.; Westergaard, M.; Hammar, N. Long-term cardiovascular risk and costs for myocardial infarction survivors in a US commercially insured population. Curr. Med. Res. Opin. 2016, 32, 703–711. [Google Scholar] [CrossRef]
  28. Stone, G.W.; Maehara, A.; Lansky, A.J.; de Bruyne, B.; Cristea, E.; Mintz, G.S.; Mehran, R.; McPherson, J.; Farhat, N.; Marso, S.P.; et al. A prospective natural-history study of coronary atherosclerosis. N. Engl. J. Med. 2011, 364, 226–235. [Google Scholar] [CrossRef]
  29. Bonaca, M.P.; Bhatt, D.L.; Cohen, M.; Steg, P.G.; Storey, R.F.; Jensen, E.C.; Magnani, G.; Bansilal, S.; Fish, M.P.; Im, K.; et al. Long-term use of ticagrelor in patients with prior myocardial infarction. N. Engl. J. Med. 2015, 372, 1791–1800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Bonaca, M.P.; Bhatt, D.L.; Storey, R.F.; Steg, P.G.; Cohen, M.; Kuder, J.; Goodrich, E.; Nicolau, J.C.; Parkhomenko, A.; López-Sendón, J.; et al. Ticagrelor for Prevention of Ischemic Events After Myocardial Infarction in Patients With Peripheral Artery Disease. J. Am. Coll. Cardiol. 2016, 67, 2719–2728. [Google Scholar] [CrossRef]
  31. Furtado, R.H.M.; Nicolau, J.C.; Magnani, G.; Im, K.; Bhatt, D.L.; Storey, R.F.; Steg, P.G.; Spinar, J.; Budaj, A.; Kontny, F.; et al. Long-term ticagrelor for secondary prevention in patients with prior myocardial infarction and no history of coronary stenting: Insights from PEGASUS-TIMI 54. Eur. Heart J. 2020, 41, 1625–1632. [Google Scholar] [CrossRef] [PubMed]
  32. Eikelboom, J.W.; Connolly, S.J.; Bosch, J.; Dagenais, G.R.; Hart, R.G.; Shestakovska, O.; Diaz, R.; Alings, M.; Lonn, E.M.; Anand, S.S.; et al. Rivaroxaban with or without Aspirin in Stable Cardiovascular Disease. N. Engl. J. Med. 2017, 377, 1319–1330. [Google Scholar] [CrossRef] [PubMed]
  33. Urban, P.; Mehran, R.; Colleran, R.; Angiolillo, D.J.; Byrne, R.A.; Capodanno, D.; Cuisset, T.; Cutlip, D.; Eerdmans, P.; Eikelboom, J.; et al. Defining high bleeding risk in patients undergoing percutaneous coronary intervention: A consensus document from the Academic Research Consortium for High Bleeding Risk. Eur. Heart J. 2019, 40, 2632–2653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Silverio, A.; Di Maio, M.; Buccheri, S.; De Luca, G.; Esposito, L.; Sarno, G.; Vecchione, C.; Galasso, G. Validation of the academic research consortium high bleeding risk criteria in patients undergoing percutaneous coronary intervention: A systematic review and meta-analysis of 10 studies and 67,862 patients. Int. J. Cardiol. 2022, 347, 8–15. [Google Scholar] [CrossRef] [PubMed]
  35. Scudiero, F.; Canonico, M.E.; Sanna, G.D.; Dossi, F.; Silverio, A.; Galasso, G.; Esposito, G.; Porto, I.; Parodi, G. Dual Antiplatelet Therapy with 3(rd) Generation P2Y(12) Inhibitors in STEMI Patients: Impact of Body Mass Index on Loading Dose-Response. Cardiovasc. Drugs Ther. 2022. [Google Scholar] [CrossRef]
  36. Ference, B.A.; Ginsberg, H.N.; Graham, I.; Ray, K.K.; Packard, C.J.; Bruckert, E.; Hegele, R.A.; Krauss, R.M.; Raal, F.J.; Schunkert, H.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2017, 38, 2459–2472. [Google Scholar] [CrossRef] [Green Version]
  37. Baigent, C.; Blackwell, L.; Emberson, J.; Holland, L.E.; Reith, C.; Bhala, N.; Peto, R.; Barnes, E.H.; Keech, A.; Simes, J.; et al. Efficacy and safety of more intensive lowering of LDL cholesterol: A meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 2010, 376, 1670–1681. [Google Scholar]
  38. Silverman, M.G.; Ference, B.A.; Im, K.; Wiviott, S.D.; Giugliano, R.P.; Grundy, S.M.; Braunwald, E.; Sabatine, M.S. Association Between Lowering LDL-C and Cardiovascular Risk Reduction Among Different Therapeutic Interventions: A Systematic Review and Meta-analysis. JAMA 2016, 316, 1289–1297. [Google Scholar] [CrossRef] [Green Version]
  39. Cannon, C.P.; Blazing, M.A.; Giugliano, R.P.; McCagg, A.; White, J.A.; Theroux, P.; Darius, H.; Lewis, B.S.; Ophuis, T.O.; Jukema, J.W.; et al. Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N. Engl. J. Med. 2015, 372, 2387–2397. [Google Scholar] [CrossRef] [Green Version]
  40. Silverio, A.; Benvenga, R.M.; Piscione, F.; Gulizia, M.M.; Meessen, J.; Colivicchi, F.; Nardi, F.; Baldi, C.; Galasso, G.; Vecchione, C.; et al. Prevalence and Predictors of Out-of-Target LDL Cholesterol 1 to 3 Years After Myocardial Infarction. A Subanalysis From the EYESHOT Post-MI Registry. J. Cardiovasc. Pharmacol. Ther. 2021, 26, 149–157. [Google Scholar] [CrossRef]
  41. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M.; et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef] [PubMed]
  42. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A.; et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef] [PubMed]
  43. Bohula, E.A.; Giugliano, R.P.; Leiter, L.A.; Verma, S.; Park, J.G.; Sever, P.S.; Lira Pineda, A.; Honarpour, N.; Wang, H.; Murphy, S.A.; et al. Inflammatory and Cholesterol Risk in the FOURIER Trial. Circulation. 2018, 138, 131–140. [Google Scholar] [CrossRef] [PubMed]
  44. Schmidt, K.; Noureen, A.; Kronenberg, F.; Utermann, G. Structure, function, and genetics of lipoprotein (a). J. Lipid Res. 2016, 57, 1339–1359. [Google Scholar] [CrossRef] [Green Version]
  45. Coassin, S.; Kronenberg, F. Lipoprotein(a) beyond the kringle IV repeat polymorphism: The complexity of genetic variation in the LPA gene. Atherosclerosis 2022, 349, 17–35. [Google Scholar] [CrossRef]
  46. Boffa, M.B.; Marcovina, S.M.; Koschinsky, M.L. Lipoprotein(a) as a risk factor for atherosclerosis and thrombosis: Mechanistic insights from animal models. Clin. Biochem. 2004, 37, 333–343. [Google Scholar] [CrossRef]
  47. Deb, A.; Caplice, N.M. Lipoprotein(a): New insights into mechanisms of atherogenesis and thrombosis. Clinical cardiology 2004, 27, 258–264. [Google Scholar] [CrossRef]
  48. Nielsen, L.B. Atherogenecity of lipoprotein(a) and oxidized low density lipoprotein: Insight from in vivo studies of arterial wall influx, degradation and efflux. Atherosclerosis 1999, 143, 229–243. [Google Scholar] [CrossRef]
  49. Tsimikas, S.; Brilakis, E.S.; Miller, E.R.; McConnell, J.P.; Lennon, R.J.; Kornman, K.S.; Witztum, J.L.; Berger, P.B. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N. Engl. J. Med. 2005, 353, 46–57. [Google Scholar] [CrossRef]
  50. van der Valk, F.M.; Bekkering, S.; Kroon, J.; Yeang, C.; Van den Bossche, J.; van Buul, J.D.; Ravandi, A.; Nederveen, A.J.; Verberne, H.J.; Scipione, C.; et al. Oxidized Phospholipids on Lipoprotein(a) Elicit Arterial Wall Inflammation and an Inflammatory Monocyte Response in Humans. Circulation 2016, 134, 611–624. [Google Scholar] [CrossRef] [Green Version]
  51. Scipione, C.A.; Sayegh, S.E.; Romagnuolo, R.; Tsimikas, S.; Marcovina, S.M.; Boffa, M.B.; Koschinsky, M.L. Mechanistic insights into Lp(a)-induced IL-8 expression: A role for oxidized phospholipid modification of apo(a). J. Lipid Res. 2015, 56, 2273–2285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Wiesner, P.; Tafelmeier, M.; Chittka, D.; Choi, S.H.; Zhang, L.; Byun, Y.S.; Almazan, F.; Yang, X.; Iqbal, N.; Chowdhury, P.; et al. MCP-1 binds to oxidized LDL and is carried by lipoprotein(a) in human plasma. J. Lipid Res. 2013, 54, 1877–1883. [Google Scholar] [CrossRef] [Green Version]
  53. Kamstrup, P.R.; Benn, M.; Tybjaerg-Hansen, A.; Nordestgaard, B.G. Extreme lipoprotein(a) levels and risk of myocardial infarction in the general population: The Copenhagen City Heart Study. Circulation 2008, 117, 176–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kamstrup, P.R.; Tybjaerg-Hansen, A.; Steffensen, R.; Nordestgaard, B.G. Genetically elevated lipoprotein(a) and increased risk of myocardial infarction. JAMA 2009, 301, 2331–2339. [Google Scholar] [CrossRef] [PubMed]
  55. Tsimikas, S.; Mallat, Z.; Talmud, P.J.; Kastelein, J.J.; Wareham, N.J.; Sandhu, M.S.; Miller, E.R.; Benessiano, J.; Tedgui, A.; Witztum, J.L.; et al. Oxidation-specific biomarkers, lipoprotein(a), and risk of fatal and nonfatal coronary events. J. Am. Coll. Cardiol. 2010, 56, 946–955. [Google Scholar] [CrossRef] [Green Version]
  56. Erqou, S.; Kaptoge, S.; Perry, P.L.; Di Angelantonio, E.; Thompson, A.; White, I.R.; Marcovina, S.M.; Collins, R.; Thompson, S.G.; Danesh, J. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality. JAMA 2009, 302, 412–423. [Google Scholar] [PubMed] [Green Version]
  57. Clarke, R.; Peden, J.F.; Hopewell, J.C.; Kyriakou, T.; Goel, A.; Heath, S.C.; Parish, S.; Barlera, S.; Franzosi, M.G.; Rust, S.; et al. Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N. Engl. J. Med. 2009, 361, 2518–2528. [Google Scholar] [CrossRef] [Green Version]
  58. Saleheen, D.; Haycock, P.C.; Zhao, W.; Rasheed, A.; Taleb, A.; Imran, A.; Abbas, S.; Majeed, F.; Akhtar, S.; Qamar, N.; et al. Apolipoprotein(a) isoform size, lipoprotein(a) concentration, and coronary artery disease: A mendelian randomisation analysis. Lancet Diabetes Endocrinol. 2017, 5, 524–533. [Google Scholar] [CrossRef] [Green Version]
  59. Willeit, P.; Ridker, P.M.; Nestel, P.J.; Simes, J.; Tonkin, A.M.; Pedersen, T.R.; Schwartz, G.G.; Olsson, A.G.; Colhoun, H.M.; Kronenberg, F.; et al. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: Individual patient-data meta-analysis of statin outcome trials. Lancet 2018, 392, 1311–1320. [Google Scholar] [CrossRef] [Green Version]
  60. Kronenberg, F.; Mora, S.; Stroes, E.S.G.; Ference, B.A.; Arsenault, B.J.; Berglund, L.; Dweck, M.R.; Koschinsky, M.; Lambert, G.; Mach, F.; et al. Lipoprotein(a) in atherosclerotic cardiovascular disease and aortic stenosis: A European Atherosclerosis Society consensus statement. Eur. Heart J. 2022, 43, 3925–3946. [Google Scholar] [CrossRef]
  61. Gencer, B.; Rigamonti, F.; Nanchen, D.; Vuilleumier, N.; Kern, I.; Aghlmandi, S.; Klingenberg, R.; Räber, L.; Auer, R.; Carballo, D.; et al. Prognostic value of elevated lipoprotein(a) in patients with acute coronary syndromes. Eur. J. Clin. Investig. 2019, 49, e13117. [Google Scholar] [CrossRef]
  62. Albers, J.J.; Slee, A.; O’Brien, K.D.; Robinson, J.G.; Kashyap, M.L.; Kwiterovich, P.O., Jr.; Xu, P.; Marcovina, S.M. Relationship of apolipoproteins A-1 and B, and lipoprotein(a) to cardiovascular outcomes: The AIM-HIGH trial (Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglyceride and Impact on Global Health Outcomes). J. Am. Coll. Cardiol. 2013, 62, 1575–1579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zewinger, S.; Kleber, M.E.; Tragante, V.; McCubrey, R.O.; Schmidt, A.F.; Direk, K.; Laufs, U.; Werner, C.; Koenig, W.; Rothenbacher, D.; et al. Relations between lipoprotein(a) concentrations, LPA genetic variants, and the risk of mortality in patients with established coronary heart disease: A molecular and genetic association study. Lancet Diabetes Endocrinol. 2017, 5, 534–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Silverio, A.; Cancro, F.P.; Di Maio, M.; Bellino, M.; Esposito, L.; Centore, M.; Carrizzo, A.; Di Pietro, P.; Borrelli, A.; De Luca, G.; et al. Lipoprotein(a) levels and risk of adverse events after myocardial infarction in patients with and without diabetes. J. Thromb. Thrombolysis 2022, 54, 382–392. [Google Scholar] [CrossRef] [PubMed]
  65. O’Donoghue, M.L.; Morrow, D.A.; Tsimikas, S.; Sloan, S.; Ren, A.F.; Hoffman, E.B.; Desai, N.R.; Solomon, S.D.; Domanski, M.; Arai, K.; et al. Lipoprotein(a) for risk assessment in patients with established coronary artery disease. J. Am. Coll. Cardiol. 2014, 63, 520–527. [Google Scholar] [CrossRef] [Green Version]
  66. Bittner, V.A.; Szarek, M.; Aylward, P.E.; Bhatt, D.L.; Diaz, R.; Edelberg, J.M.; Fras, Z.; Goodman, S.G.; Halvorsen, S.; Hanotin, C.; et al. Effect of Alirocumab on Lipoprotein(a) and Cardiovascular Risk After Acute Coronary Syndrome. J. Am. Coll. Cardiol. 2020, 75, 133–144. [Google Scholar] [CrossRef]
  67. Yoon, Y.H.; Ahn, J.M.; Kang, D.Y.; Lee, P.H.; Kang, S.J.; Park, D.W.; Lee, S.W.; Kim, Y.H.; Han, K.H.; Lee, C.W.; et al. Association of Lipoprotein(a) With Recurrent Ischemic Events Following Percutaneous Coronary Intervention. JACC Cardiovasc. Interv. 2021, 14, 2059–2068. [Google Scholar] [CrossRef]
  68. O’Donoghue, M.L.; Fazio, S.; Giugliano, R.P.; Stroes, E.S.G.; Kanevsky, E.; Gouni-Berthold, I.; Im, K.; Lira Pineda, A.; Wasserman, S.M.; Češka, R.; et al. Lipoprotein(a), PCSK9 Inhibition, and Cardiovascular Risk. Circulation 2019, 139, 1483–1492. [Google Scholar] [CrossRef]
  69. Madsen, C.M.; Kamstrup, P.R.; Langsted, A.; Varbo, A.; Nordestgaard, B.G. Lipoprotein(a)-Lowering by 50 mg/dL (105 nmol/L) May Be Needed to Reduce Cardiovascular Disease 20% in Secondary Prevention: A Population-Based Study. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 255–266. [Google Scholar] [CrossRef]
  70. Thompson, G.R. Recommendations for the use of LDL apheresis. Atherosclerosis 2008, 198, 247–255. [Google Scholar] [CrossRef]
  71. Tsimikas, S.; Gordts, P.; Nora, C.; Yeang, C.; Witztum, J.L. Statin therapy increases lipoprotein(a) levels. Eur. Heart J. 2020, 41, 2275–2284. [Google Scholar] [CrossRef] [PubMed]
  72. Santos, R.D.; Raal, F.J.; Catapano, A.L.; Witztum, J.L.; Steinhagen-Thiessen, E.; Tsimikas, S. Mipomersen, an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various populations with hypercholesterolemia: Results of 4 phase III trials. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 689–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Szarek, M.; Bittner, V.A.; Aylward, P.; Baccara-Dinet, M.; Bhatt, D.L.; Diaz, R.; Fras, Z.; Goodman, S.G.; Halvorsen, S.; Harrington, R.A.; et al. Lipoprotein(a) lowering by alirocumab reduces the total burden of cardiovascular events independent of low-density lipoprotein cholesterol lowering: ODYSSEY OUTCOMES trial. Eur. Heart J. 2020, 41, 4245–4255. [Google Scholar] [CrossRef] [PubMed]
  74. Ray, K.K.; Wright, R.S.; Kallend, D.; Koenig, W.; Leiter, L.A.; Raal, F.J.; Bisch, J.A.; Richardson, T.; Jaros, M.; Wijngaard, P.L.J.; et al. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N. Engl. J. Med. 2020, 382, 1507–1519. [Google Scholar] [CrossRef]
  75. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur. Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Koren, M.J.; Moriarty, P.M.; Baum, S.J.; Neutel, J.; Hernandez-Illas, M.; Weintraub, H.S.; Florio, M.; Kassahun, H.; Melquist, S.; Varrieur, T.; et al. Preclinical development and phase 1 trial of a novel siRNA targeting lipoprotein(a). Nat. Med. 2022, 28, 96–103. [Google Scholar] [CrossRef]
  77. Tsimikas, S.; Karwatowska-Prokopczuk, E.; Gouni-Berthold, I.; Tardif, J.C.; Baum, S.J.; Steinhagen-Thiessen, E.; Shapiro, M.D.; Stroes, E.S.; Moriarty, P.M.; Nordestgaard, B.G.; et al. Lipoprotein(a) Reduction in Persons with Cardiovascular Disease. N. Engl. J. Med. 2020, 382, 244–255. [Google Scholar] [CrossRef]
  78. O’Donoghue, M.L.; Rosenson, R.S.; Gencer, B.; López, J.A.G.; Lepor, N.E.; Baum, S.J.; Stout, E.; Gaudet, D.; Knusel, B.; Kuder, J.F.; et al. Small Interfering RNA to Reduce Lipoprotein(a) in Cardiovascular Disease. N. Engl. J. Med. 2022, 387, 1855–1864. [Google Scholar] [CrossRef]
  79. Hansson, G.K. Inflammation, atherosclerosis, and coronary artery disease. N. Engl. J. Med. 2005, 352, 1685–1695. [Google Scholar] [CrossRef] [Green Version]
  80. van Hout, G.P.; Bosch, L.; Ellenbroek, G.H.; de Haan, J.J.; van Solinge, W.W.; Cooper, M.A.; Arslan, F.; de Jager, S.C.; Robertson, A.A.; Pasterkamp, G.; et al. The selective NLRP3-inflammasome inhibitor MCC950 reduces infarct size and preserves cardiac function in a pig model of myocardial infarction. Eur. Heart J. 2017, 38, 828–836. [Google Scholar] [CrossRef] [Green Version]
  81. 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.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
  82. Nidorf, S.M.; Fiolet, A.T.L.; Mosterd, A.; Eikelboom, J.W.; Schut, A.; Opstal, T.S.J.; The, S.H.K.; Xu, X.F.; Ireland, M.A.; Lenderink, T.; et al. Colchicine in Patients with Chronic Coronary Disease. N. Engl. J. Med. 2020, 383, 1838–1847. [Google Scholar] [CrossRef] [PubMed]
  83. Tardif, J.C.; Kouz, S.; Waters, D.D.; Bertrand, O.F.; Diaz, R.; Maggioni, A.P.; Pinto, F.J.; Ibrahim, R.; Gamra, H.; Kiwan, G.S.; et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N. Engl. J. Med. 2019, 381, 2497–2505. [Google Scholar] [CrossRef] [PubMed]
  84. Stary, H.C.; Chandler, A.B.; Dinsmore, R.E.; Fuster, V.; Glagov, S.; Insull, W.; Jr Rosenfeld, M.E.; Schwartz, C.J.; Wagner, W.D.; Wissler, R.W. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 1995, 92, 1355–1374. [Google Scholar] [CrossRef] [PubMed]
  85. Kovanen, P.T.; Kaartinen, M.; Paavonen, T. Infiltrates of activated mast cells at the site of coronary atheromatous erosion or rupture in myocardial infarction. Circulation 1995, 92, 1084–1088. [Google Scholar] [CrossRef]
  86. Frostegård, J.; Ulfgren, A.K.; Nyberg, P.; Hedin, U.; Swedenborg, J.; Andersson, U.; Hansson, G.K. Cytokine expression in advanced human atherosclerotic plaques: Dominance of pro-inflammatory (Th1) and macrophage-stimulating cytokines. Atherosclerosis 1999, 145, 33–43. [Google Scholar] [CrossRef]
  87. van der Wal, A.C.; Becker, A.E.; van der Loos, C.M.; Das, P.K. Site of intimal rupture or erosion of thrombosed coronary atherosclerotic plaques is characterized by an inflammatory process irrespective of the dominant plaque morphology. Circulation 1994, 89, 36–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Moore, K.J.; Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 2011, 145, 341–355. [Google Scholar] [CrossRef] [Green Version]
  89. Libby, P.; Tabas, I.; Fredman, G.; Fisher, E.A. Inflammation and its resolution as determinants of acute coronary syndromes. Circ. Res. 2014, 114, 1867–1879. [Google Scholar] [CrossRef] [Green Version]
  90. Kaptoge, S.; Di Angelantonio, E.; Lowe, G.; Pepys, M.B.; Thompson, S.G.; Collins, R.; Danesh, J. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: An individual participant meta-analysis. Lancet 2010, 375, 132–140. [Google Scholar]
  91. Ridker, P.M.; MacFadyen, J.G.; Everett, B.M.; Libby, P.; Thuren, T.; Glynn, R.J. 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, 319–328. [Google Scholar] [CrossRef] [PubMed]
  92. Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nuñez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Schroder, K.; Tschopp, J. The inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Baldrighi, M.; Mallat, Z.; Li, X. NLRP3 inflammasome pathways in atherosclerosis. Atherosclerosis 2017, 267, 127–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Xiao, H.; Lu, M.; Lin, T.Y.; Chen, Z.; Chen, G.; Wang, W.C.; Marin, T.; Shentu, T.P.; Wen, L.; Gongol, B.; et al. Sterol regulatory element binding protein 2 activation of NLRP3 inflammasome in endothelium mediates hemodynamic-induced atherosclerosis susceptibility. Circulation 2013, 128, 632–642. [Google Scholar] [CrossRef] [Green Version]
  96. Rajamäki, K.; Lappalainen, J.; Oörni, K.; Välimäki, E.; Matikainen, S.; Kovanen, P.T.; Eklund, K.K. Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: A novel link between cholesterol metabolism and inflammation. PLoS ONE 2010, 5, e11765. [Google Scholar] [CrossRef] [Green Version]
  97. Yeh, M.; Cole, A.L.; Choi, J.; Liu, Y.; Tulchinsky, D.; Qiao, J.H.; Fishbein, M.C.; Dooley, A.N.; Hovnanian, T.; Mouilleseaux, K.; et al. Role for sterol regulatory element-binding protein in activation of endothelial cells by phospholipid oxidation products. Circ. Res. 2004, 95, 780–788. [Google Scholar] [CrossRef] [Green Version]
  98. He, Y.; Hara, H.; Núñez, G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem. Sci. 2016, 41, 1012–1021. [Google Scholar] [CrossRef] [Green Version]
  99. Libby, P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J. Am. Coll. Cardiol. 2017, 70, 2278–2289. [Google Scholar] [CrossRef]
  100. Wang, J.M.; Sica, A.; Peri, G.; Walter, S.; Padura, I.M.; Libby, P.; Ceska, M.; Lindley, I.; Colotta, F.; Mantovani, A. Expression of monocyte chemotactic protein and interleukin-8 by cytokine-activated human vascular smooth muscle cells. Arterioscler. Thromb. 1991, 11, 1166–1174. [Google Scholar] [CrossRef] [Green Version]
  101. Loppnow, H.; Libby, P. Adult human vascular endothelial cells express the IL6 gene differentially in response to LPS or IL1. Cell. Immunol. 1989, 122, 493–503. [Google Scholar] [CrossRef]
  102. Warner, S.J.; Auger, K.R.; Libby, P. Human interleukin 1 induces interleukin 1 gene expression in human vascular smooth muscle cells. J. Exp. Med. 1987, 165, 1316–1331. [Google Scholar] [CrossRef] [PubMed]
  103. Suzuki, K.; Murtuza, B.; Smolenski, R.T.; Sammut, I.A.; Suzuki, N.; Kaneda, Y.; Yacoub, M.H. Overexpression of interleukin-1 receptor antagonist provides cardioprotection against ischemia-reperfusion injury associated with reduction in apoptosis. Circulation 2001, 104, I-308–I-313. [Google Scholar] [CrossRef] [PubMed]
  104. Abbate, A.; Salloum, F.N.; Van Tassell, B.W.; Vecile, E.; Toldo, S.; Seropian, I.; Mezzaroma, E.; Dobrina, A. Alterations in the interleukin-1/interleukin-1 receptor antagonist balance modulate cardiac remodeling following myocardial infarction in the mouse. PLoS ONE 2011, 6, e27923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Abbate, A.; Dinarello, C.A. Anti-inflammatory therapies in acute coronary syndromes: Is IL-1 blockade a solution? Eur. Heart J. 2015, 36, 337–339. [Google Scholar] [CrossRef] [Green Version]
  106. Morton, A.C.; Rothman, A.M.; Greenwood, J.P.; Gunn, J.; Chase, A.; Clarke, B.; Hall, A.S.; Fox, K.; Foley, C.; Banya, W.; et al. The effect of interleukin-1 receptor antagonist therapy on markers of inflammation in non-ST elevation acute coronary syndromes: The MRC-ILA Heart Study. Eur. Heart J. 2015, 36, 377–384. [Google Scholar] [CrossRef] [Green Version]
  107. Andreis, A.; Imazio, M.; Piroli, F.; Avondo, S.; Casula, M.; Paneva, E.; De Ferrari, G.M. Efficacy and safety of colchicine for the prevention of major cardiovascular and cerebrovascular events in patients with coronary artery disease: A systematic review and meta-analysis on 12 869 patients. Eur. J. Prev. Cardiol. 2022, 28, 1916–1925. [Google Scholar] [CrossRef]
  108. Martinon, F.; Burns, K.; Tschopp, J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
  109. Fiolet, A.T.L.; Opstal, T.S.J.; Mosterd, A.; Eikelboom, J.W.; Jolly, S.S.; Keech, A.C.; Kelly, P.; Tong, D.C.; Layland, J.; Nidorf, S.M.; et al. Efficacy and safety of low-dose colchicine in patients with coronary disease: A systematic review and meta-analysis of randomized trials. Eur. Heart J. 2021, 42, 2765–2775. [Google Scholar] [CrossRef]
  110. Visseren, F.L.J.; Mach, F.; Smulders, Y.M.; Carballo, D.; Koskinas, K.C.; Bäck, M.; Benetos, A.; Biffi, A.; Boavida, J.M.; Capodanno, D.; et al. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice. Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef]
  111. O’Fee, K.; Deych, E.; Ciani, O.; Brown, D.L. Assessment of Nonfatal Myocardial Infarction as a Surrogate for All-Cause and Cardiovascular Mortality in Treatment or Prevention of Coronary Artery Disease: A Meta-analysis of Randomized Clinical Trials. JAMA Intern. Med. 2021, 181, 1575–1587. [Google Scholar] [CrossRef] [PubMed]
  112. Micha, R.; Imamura, F.; Wyler von Ballmoos, M.; Solomon, D.H.; Hernán, 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, 1362–1370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ridker, P.M.; Everett, B.M.; Pradhan, A.; MacFadyen, J.G.; Solomon, D.H.; Zaharris, E.; Mam, V.; Hasan, A.; Rosenberg, Y.; Iturriaga, E.; et al. Low-Dose Methotrexate for the Prevention of Atherosclerotic Events. N. Engl. J. Med. 2019, 380, 752–762. [Google Scholar] [CrossRef] [PubMed]
  114. Abbate, A.; Trankle, C.R.; Buckley, L.F.; Lipinski, M.J.; Appleton, D.; Kadariya, D.; Canada, J.M.; Carbone, S.; Roberts, C.S.; Abouzaki, N.; et al. Interleukin-1 Blockade Inhibits the Acute Inflammatory Response in Patients With ST-Segment-Elevation Myocardial Infarction. J. Am. Heart Assoc. 2020, 9, e014941. [Google Scholar] [CrossRef]
  115. Broch, K.; Anstensrud, A.K.; Woxholt, S.; Sharma, K.; Tøllefsen, I.M.; Bendz, B.; Aakhus, S.; Ueland, T.; Amundsen, B.H.; Damås, J.K.; et al. Randomized Trial of Interleukin-6 Receptor Inhibition in Patients With Acute ST-Segment Elevation Myocardial Infarction. J. Am. Coll. Cardiol. 2021, 77, 1845–1855. [Google Scholar] [CrossRef]
  116. Ridker, P.M.; Devalaraja, M.; Baeres, F.M.M.; Engelmann, M.D.M.; Hovingh, G.K.; Ivkovic, M.; Lo, L.; Kling, D.; Pergola, P.; Raj, D.; et al. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2021, 397, 2060–2069. [Google Scholar] [CrossRef]
  117. Abderrazak, A.; Couchie, D.; Mahmood, D.F.; Elhage, R.; Vindis, C.; Laffargue, M.; Matéo, V.; Büchele, B.; Ayala, M.R.; El Gaafary, M.; et al. Anti-inflammatory and antiatherogenic effects of the NLRP3 inflammasome inhibitor arglabin in ApoE2.Ki mice fed a high-fat diet. Circulation 2015, 131, 1061–1070. [Google Scholar] [CrossRef] [Green Version]
  118. Jiang, M.; Li, R.; Lyu, J.; Li, X.; Wang, W.; Wang, Z.; Sheng, H.; Zhang, W.; Karhausen, J.; Yang, W. MCC950, a selective NLPR3 inflammasome inhibitor, improves neurologic function and survival after cardiac arrest and resuscitation. J. Neuroinflamm. 2020, 17, 256. [Google Scholar] [CrossRef]
  119. Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef] [Green Version]
  120. Tousoulis, D.; Guzik, T.; Padro, T.; Duncker, D.J.; De Luca, G.; Eringa, E.; Vavlukis, M.; Antonopoulos, A.S.; Katsimichas, T.; Cenko, E.; et al. Mechanisms, therapeutic implications, and methodological challenges of gut microbiota and cardiovascular diseases: A position paper by the ESC Working Group on Coronary Pathophysiology and Microcirculation. Cardiovasc. Res. 2022, 118, 3171–3182. [Google Scholar] [CrossRef]
  121. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Schroeder, B.O.; Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef] [PubMed]
  123. Ascher, S.; Reinhardt, C. The gut microbiota: An emerging risk factor for cardiovascular and cerebrovascular disease. Eur. J. Immunol. 2018, 48, 564–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef] [Green Version]
  125. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [Green Version]
  126. Senthong, V.; Li, X.S.; Hudec, T.; Coughlin, J.; Wu, Y.; Levison, B.; Wang, Z.; Hazen, S.L.; Tang, W.H. Plasma Trimethylamine N-Oxide, a Gut Microbe-Generated Phosphatidylcholine Metabolite, Is Associated With Atherosclerotic Burden. J. Am. Coll. Cardiol. 2016, 67, 2620–2628. [Google Scholar] [CrossRef]
  127. Senthong, V.; Wang, Z.; Fan, Y.; Wu, Y.; Hazen, S.L.; Tang, W.H. Trimethylamine N-Oxide and Mortality Risk in Patients With Peripheral Artery Disease. J. Am. Heart Assoc. 2016, 5, e004237. [Google Scholar] [CrossRef]
  128. Skagen, K.; Trøseid, M.; Ueland, T.; Holm, S.; Abbas, A.; Gregersen, I.; Kummen, M.; Bjerkeli, V.; Reier-Nilsen, F.; Russell, D.; et al. The Carnitine-butyrobetaine-trimethylamine-N-oxide pathway and its association with cardiovascular mortality in patients with carotid atherosclerosis. Atherosclerosis 2016, 247, 64–69. [Google Scholar] [CrossRef] [Green Version]
  129. Violi, F.; Cammisotto, V.; Bartimoccia, S.; Pignatelli, P.; Carnevale, R.; Nocella, C. Gut-derived low-grade endotoxaemia, atherothrombosis and cardiovascular disease. Nat. Rev. Cardiol. 2023, 20, 24–37. [Google Scholar] [CrossRef]
  130. Mueller, D.M.; Allenspach, M.; Othman, A.; Saely, C.H.; Muendlein, A.; Vonbank, A.; Drexel, H.; von Eckardstein, A. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis 2015, 243, 638–644. [Google Scholar] [CrossRef]
  131. Gencer, B.; Li, X.S.; Gurmu, Y.; Bonaca, M.P.; Morrow, D.A.; Cohen, M.; Bhatt, D.L.; Steg, P.G.; Storey, R.F.; Johanson, P.; et al. Gut Microbiota-Dependent Trimethylamine N-oxide and Cardiovascular Outcomes in Patients With Prior Myocardial Infarction: A Nested Case Control Study From the PEGASUS-TIMI 54 Trial. J. Am. Heart Assoc. 2020, 9, e015331. [Google Scholar] [CrossRef] [PubMed]
  132. den Dekker, W.K.; Cheng, C.; Pasterkamp, G.; Duckers, H.J. Toll like receptor 4 in atherosclerosis and plaque destabilization. Atherosclerosis 2010, 209, 314–320. [Google Scholar] [CrossRef] [PubMed]
  133. Wiedermann, C.J.; Kiechl, S.; Dunzendorfer, S.; Schratzberger, P.; Egger, G.; Oberhollenzer, F.; Willeit, J. Association of endotoxemia with carotid atherosclerosis and cardiovascular disease: Prospective results from the Bruneck Study. J. Am. Coll. Cardiol. 1999, 34, 1975–1981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Asada, M.; Oishi, E.; Sakata, S.; Hata, J.; Yoshida, D.; Honda, T.; Furuta, Y.; Shibata, M.; Suzuki, K.; Watanabe, H.; et al. Serum Lipopolysaccharide-Binding Protein Levels and the Incidence of Cardiovascular Disease in a General Japanese Population: The Hisayama Study. J. Am. Heart Assoc. 2019, 8, e013628. [Google Scholar] [CrossRef]
  135. Pussinen, P.J.; Tuomisto, K.; Jousilahti, P.; Havulinna, A.S.; Sundvall, J.; Salomaa, V. Endotoxemia, immune response to periodontal pathogens, and systemic inflammation associate with incident cardiovascular disease events. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 1433–1439. [Google Scholar] [CrossRef] [Green Version]
  136. Carnevale, R.; Sciarretta, S.; Valenti, V.; di Nonno, F.; Calvieri, C.; Nocella, C.; Frati, G.; Forte, M.; d’Amati, G.; Pignataro, M.G.; et al. Low-grade endotoxaemia enhances artery thrombus growth via Toll-like receptor 4: Implication for myocardial infarction. Eur. Heart J. 2020, 41, 3156–3165. [Google Scholar] [CrossRef]
  137. Zhou, X.; Li, J.; Guo, J.; Geng, B.; Ji, W.; Zhao, Q.; Li, J.; Liu, X.; Liu, J.; Guo, Z.; et al. Gut-dependent microbial translocation induces inflammation and cardiovascular events after ST-elevation myocardial infarction. Microbiome 2018, 6, 66. [Google Scholar] [CrossRef] [Green Version]
Table 1. Emerging therapies for lowering Lp(a) serum levels.
Table 1. Emerging therapies for lowering Lp(a) serum levels.
DrugPrincipal Mechanism of ActionMechanism of Lp(a) LoweringStudyDesignPopulationFindings
EvolocumabMonoclonal antibody inhibiting LDL-R degradation by targeting PCSK9Inhibition of apo(a) secretionFOURIER
(NCT01764633)
Sub-analysis of phase III RCT25,096 stable-CAD ptsEvolocumab significantly reduced Lp(a) by a median of 26.9% [54];
Evolocumab reduced the risk of death, MI, or PCI by 23% in patients with a baseline Lp(a) > median value [54]
AlirocumabMonoclonal antibody inhibiting LDL-R degradation by targeting PCSK9Inhibition of apo(a) secretionODISSEY Outcomes
(NCT01663402)
Sub-analysis of phase III RCT18,924 post-ACS ptsAlirocumab significantly reduced Lp(a) by 23% [61];
Alirocumab independently reduced the risk of CV adverse outcomes [61]
InclisiransiRNA inhibiting LDL-R degradation targeting PCSK9Inhibition of apo(a) secretionORION-10
ORION-11
(NCT03399370;
NCT03400800)
Phase III RCT2178 pts with ASCVD (1975) or ASCVD risk equivalent (203)Inclisiran reduces Lp(a) plasma levels by 19–22% [62];
the effect on CV outcomes is unknown
MipomersenASO inhibiting apo(B) synthesis-----Four phase III trials
(NCT00607373;
NCT00706849;
NCT00770146;
NCT00794664)
Four phase III RCTs382 pts diagnosed with:
- HoFH (51),
- HeFH with CAD (123),
- severe HC (57),
- HC at risk of CAD (151)
Mipomersen reduced Lp(a) plasma levels from 20% to 40% [60];
the effects on CV outcomes are unknown
OlpasiransiRNA targeting apo(a) mRNA and leads to
degradation
-----OCEAN[a]-DOSE
(NCT04270760)
Phase II RCT281 pts with high Lp(a) and ASCVDOlpasiran reduced Lp(a) plasma levels from 67% to 97% [66];
the effect on CV outcomes is unclear
PelacarsenASO targeting apo(a) mRNA and leads to
degradation
-----AKCEA-APO(a)-LRx
(NCT03070782)
Phase II RCT286 pts with high Lp(a) and CVDPelacarsen reduced Lp(a) plasma levels by 80% [65]
A phase III RCT evaluating the effect on CV outcomes is ongoing (NCT04023552)
ACS, acute coronary syndromes; Apo(a), apolipoprotein(a); Apo(B), apolipoprotein(B); ASCVD, atherosclerotic cardiovascular disease; ASO, antisense oligonucleotide; CAD, coronary artery disease; CV, cardiovascular; CVD, cardiovascular disease; HC, hypercholesterolemia; HeFH, heterozygous familial hypercholesterolemia; HoFH, homozygous familial hypercholesterolemia; LDL-R, low-density lipoprotein receptor; Lp(a), lipoprotein(a); MI, myocardial infarction; PCI, percutaneous coronary intervention; PCSK9, proprotein convertase subtilisin/kexin type 9; pts, patients; RCT, randomized controlled trial.
Table 2. Anti-inflammatory and immune therapies in patients with established CAD.
Table 2. Anti-inflammatory and immune therapies in patients with established CAD.
DrugPrincipal Mechanism of ActionStudyDesignPopulationFindings
ColchicineInhibition of microtubules polymerisationCOLCOT
(NCT02551094)
LoDoCo2
(ACTRN12614000093684)
Phase III RCT4745 pts with recent MI (COLCOT)
5522 pts with chronic CAD (LoDoCo2)
Colchicine 0.5 mg significantly reduced the risk of ischemic CV events [70,71]
Canakinumabanti-IL-1β monoclonal antibodyCANTOS
(NCT01327846)
Phase III RCT10,061 pts with previous MI and hs-CRP >2 mg/LCanakinumab 150 mg every 3 months significantly reduced the rate of recurrent CV events [69]
MethotrexateFolate pathway antagonistCIRT
(NCT01594333)
Phase III RCT4786 pts with previous MI or MVDMTX 15–20 mg weekly did not reduce levels of IL-1β, IL-6, or CRP and did not reduce CV events [99];
a phase III RCT evaluating MTX delivered in LDL-nanoparticles is ongoing (NCT04616872)
AnakinraInhibition of the interaction between IL-1 and IL-1RVCU-ART3
(NCT01950299)
Phase II RCT99 pts with STEMI and acute HFAnakinra significantly reduces the systemic inflammatory response after STEMI;
no difference in terms of CV outcomes [100]
TocilizumabInhibits IL-6RASSAIL-MI
(NCT03004703)
Phase II RCT199 pts with STEMITocilizumab increased myocardial salvage;
no difference in terms of CV outcomes [101]
ZiltivekimabInhibits IL-6 ligandRESCUE
(NCT03926117)
Phase II RCT264 pts with CKDZiltivekimab reduced biomarkers of inflammation and thrombosis [102];
phase III RCT evaluating the effect on CV outcomes is ongoing (NCT05021835)
CAD, coronary artery disease; CKD, chronic kidney disease; CRP, C-reactive protein; CV, cardiovascular events; HF, heart failure; hs-CRP, high-sensitivity C-reactive protein; IL-1, interleukin-1; IL-1R, interleukin-1 receptor; IL-1β, interleukin-1β; IL-6, interleukin-6; IL-6R, interleukin-6 receptor; LDL, low-density lipoprotein; MI, myocardial infarction; MTX, methotrexate; MVD, multivessel disease; STEMI, ST-elevation myocardial infarction; pts, patients; RCT, randomized controlled trials.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Silverio, A.; Cancro, F.P.; Esposito, L.; Bellino, M.; D’Elia, D.; Verdoia, M.; Vassallo, M.G.; Ciccarelli, M.; Vecchione, C.; Galasso, G.; et al. Secondary Cardiovascular Prevention after Acute Coronary Syndrome: Emerging Risk Factors and Novel Therapeutic Targets. J. Clin. Med. 2023, 12, 2161. https://doi.org/10.3390/jcm12062161

AMA Style

Silverio A, Cancro FP, Esposito L, Bellino M, D’Elia D, Verdoia M, Vassallo MG, Ciccarelli M, Vecchione C, Galasso G, et al. Secondary Cardiovascular Prevention after Acute Coronary Syndrome: Emerging Risk Factors and Novel Therapeutic Targets. Journal of Clinical Medicine. 2023; 12(6):2161. https://doi.org/10.3390/jcm12062161

Chicago/Turabian Style

Silverio, Angelo, Francesco Paolo Cancro, Luca Esposito, Michele Bellino, Debora D’Elia, Monica Verdoia, Maria Giovanna Vassallo, Michele Ciccarelli, Carmine Vecchione, Gennaro Galasso, and et al. 2023. "Secondary Cardiovascular Prevention after Acute Coronary Syndrome: Emerging Risk Factors and Novel Therapeutic Targets" Journal of Clinical Medicine 12, no. 6: 2161. https://doi.org/10.3390/jcm12062161

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