Next Article in Journal
Photon-Counting Detector Computed Tomography (PCD-CT): A New Era for Cardiovascular Imaging? Current Status and Future Outlooks
Previous Article in Journal
Novel Medical Treatments and Devices for the Management of Heart Failure with Reduced Ejection Fraction
Previous Article in Special Issue
Neutrophil Extracellular Traps (NETs) and Atherosclerosis: Does Hypolipidemic Treatment Have an Effect?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Atherosclerosis Residual Lipid Risk-Overview of Existing and Future Pharmacotherapies

by
Muntaser Omari
1 and
Mohammad Alkhalil
1,2,*
1
Cardiothoracic Centre, Freeman Hospital, Newcastle-upon-Tyne NE7 7DN, UK
2
Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK
*
Author to whom correspondence should be addressed.
J. Cardiovasc. Dev. Dis. 2024, 11(4), 126; https://doi.org/10.3390/jcdd11040126
Submission received: 29 March 2024 / Revised: 15 April 2024 / Accepted: 18 April 2024 / Published: 21 April 2024

Abstract

:
Patients with atherosclerotic disease remain at increased risk of future events despite receiving optimal medical treatment. This residual risk is widely heterogeneous, but lipoprotein particles and their content play a major role in determining future cardiovascular events. Beyond low-density lipoprotein cholesterol (LDL-c), other lipoprotein particles have not demonstrated similar contribution to the progression of atherosclerosis. Statins, ezetimibe, and more recently, proprotein convertase subtilisin kexin 9 (PCSK9) inhibitors and bempedoic acid have confirmed the causal role of LDL-c in the development of atherosclerosis. Data on high-density lipoprotein cholesterol (HDL-c) suggested a possible causal role for atherosclerosis; nonetheless, HDL-c-raising treatments, including cholesteryl-ester transfer protein (CETP) inhibitors and niacin, failed to confirm this relationship. On the other hand, mendelian randomisation revealed that triglycerides are more implicated in the development of atherosclerosis. Although the use of highly purified eicosapentaenoic acid (EPA) was associated with a reduction in the risk of adverse cardiovascular events, this beneficial effect did not correlate with the reduction in triglycerides level and has not been consistent across large phase 3 trials. Moreover, other triglyceride-lowering treatments, such as fibrates, were not associated with a reduction in future cardiovascular risk. Studies assessing agents targeting angiopoietin-like 3 (lipoprotein lipase inhibitor) and apolipoprotein C3 antisense will add further insights into the role of triglycerides in atherosclerosis. Emerging lipid markers such as lipoprotein (a) and cholesterol efflux capacity may have a direct role in the progression of atherosclerosis. Targeting these biomarkers may provide incremental benefits in reducing cardiovascular risk when added to optimal medical treatment. This Review aims to assess available therapies for current lipid biomarkers and provide mechanistic insight into their potential role in reducing future cardiovascular risk.

1. Introduction

Despite advances in revascularisation strategies as well as pharmacotherapies, acute coronary syndrome is one of the leading causes of morbidity and mortality in the world [1]. One in five patients who survive acute coronary syndrome presentation returns with a second event within the following five years despite being on the guidelines-directed optimal medical therapy [2,3]. This residual risk is rather heterogeneous and not identical among patients re-presenting with a secondary ACS. Therefore, characterising the risk at an individual level may allow tailored therapy and precise intervention to reduce future cardiovascular events. The use of blood, imaging, or genomic biomarkers may enable us to understand future risks and offer patients intensive therapies accordingly [4,5,6,7]. This is important given the recent advances in pharmacotherapy, particularly in the field of lipidology.
The intricate relationship between lipoprotein particles and their content plays a significant role in determining future cardiovascular events. The role of low-density lipoprotein cholesterol (LDL-c) was recognised early, and recent studies have confirmed its causal relationship with the development of atherosclerotic disease [8,9,10,11]. However, data on other lipoprotein particles, such as high-density lipoprotein (HDL) or triglycerides, were less consistent, and their direct role in reducing future cardiovascular risk could not be ascertained [12,13,14,15]. Nonetheless, new triglyceride-lowering treatments targeting angiopoietin-like 3 (lipoprotein lipase inhibitor) and apolipoprotein C3 antisense will add further insights into the role of triglycerides in the progression of atherosclerosis [16].
Lipoprotein (a) and cholesterol efflux capacity are emerging as a therapeutic target for patients at high risk of future cardiovascular events. Targeting these biomarkers may provide incremental benefits in reducing cardiovascular risk when added to optimal medical treatment. This Review aims to assess available therapies for current lipid biomarkers and provide mechanistic insight into their potential role in reducing future cardiovascular risk.

2. Low-Density Lipoprotein Cholesterol Lowering Treatment

Numerous primary and secondary prevention trials have consistently shown a strong and direct correlation between LDL-c levels and atherosclerotic cardiovascular disease risk [17]. LDL and other apoB-containing particles are required to initiate and propagate atherosclerotic plaques [18]. Retention and accumulation of apoB lipoproteins (>90% are LDL particles) in the arterial wall initiate atherosclerotic plaque formation [19]. Correspondingly, the benefit of lipid-lowering therapies is proportional to the degree of reduction in apoB, and this benefit appears to increase over time [20]. Therefore, guidelines have recommended targets for reducing LDL-c with a lower threshold for patients who are considered at very high risk or recurrent events [21].
Statins are the cornerstone of preventing and treating atherosclerotic disease. Statins inhibit the rate-limiting step of cholesterol biosynthesis, hydroxymethylglutaryl (HMG)-CoA reductase and block the endogenic cholesterol synthase pathway, up-regulating hepatic LDL receptors (LDLR) expression, resulting in lower plasma LDL-c. The reduction in cardiovascular risk is proportionate to the decrease in LDL-c. For every 1 mmol/L reduction in LDL-c, there is a 22% relative risk reduction in future cardiovascular events [17]. Moreover, the benefits were more apparent in patients with high baseline clinical risk, such as post-coronary artery bypass grafts or polyvascular disease [22,23]. Statins are known to have pleiotropic features, including anti-inflammatory and plaque stabilisation effects [24,25]. There are some variations in the pharmacodynamic properties among different statins (Table 1). Mechanistically, it is presumed that lipid evacuation and replacement with fibrosis, in response to lowering LDL-c, is the plausible statin mechanism [4]. This would render the plaque more stable and less likely to rupture and cause future events. Statins are usually well-tolerated. Myalgia remains the most common side effect of statin use, with documented rates from 1 to 10%. Rhabdomyolysis, on the other hand, is the most serious adverse effect, but its occurrence is rare (less than 0.1%). There are a few conditions that may increase the risk of rhabdomyolysis, which include hypothyroidism, alcohol abuse and poly-pharmacology. Liver function derangement is not uncommon and occurs in about 1%. There is an increased risk of diabetes associated with statins, but this risk is small, and the benefits of statins significantly outweigh this risk [26]. Additionally, statins are associated with a paradoxical increase of reductase protein that may attenuate their efficacy and reduce their safety. Degradation of reductase protein (HMG-CoA reductase degrader) would eliminate this risk. The recent development of compounds that share structure-activity with sterol analogues may help with this phenomenon, lower cholesterol, and reduce atherosclerotic plaques in experimental studies [27].
The main driver of clinical benefits in response to statin treatment has always been debated [28]. The statin versus LDL-c hypothesis seems to have shifted with the results of ezetimibe in the Improved Reduction of Outcome: Vytorin Efficacy International Trial (IMPROVE-IT) [11]. Ezetimibe reduces intestinal cholesterol absorption by targeting the Niemann-Pick C1-like 1 (NPC1L1) protein. It is worth noting that the absorption of ezetimibe differs according to age, and it is higher in older adults [29]. It is associated with an 18–22% reduction in LDL-c, which resulted in a 2% absolute risk reduction in patients presenting with acute coronary syndrome when combined with simvastatin after a median follow-up of 6 years [11]. The Plaque Regression With Cholesterol Absorption Inhibitor or Synthesis Inhibitor Evaluated by Intravascular Ultrasound (PRECISE-IVUS) study showed greater plaque regression with the combination of ezetimibe and atorvastatin compared to atorvastatin only [30]. Ezetimibe is well-tolerated and has a good safety profile, with few to no adverse effects reported in the literature.
More recently, inhibiting proprotein convertase subtilisin–kexin type 9 with monoclonal antibodies using evolocumab and alirocumab has been added to the LDL-c reduction armamentarium. PCSK9 is responsible for extracellular binding and subsequent degradation of LDLRs, which stops LDL clearance from circulation. Monoclonal antibodies targeting PCSK9 maintain the number of LDLRs and subsequently lead to a reduction in LDL-c. Large phase 3 outcome studies showed a significant decrease in LDL-c, which was translated into a significant reduction in adverse clinical outcomes [31,32]. The Further Cardiovascular Outcomes Research with PCSK9 Inhibition in Subjects with Elevated Risk (FOURIER) reported a 59% reduction in LDL-c using evolocumab on top of statin treatment. This led to a 1.5% absolute risk reduction in major adverse cardiovascular events over 2.2 years of follow-up in patients with stable atherosclerotic disease [31]. Similarly, the use of alirocumab in the ODYSSEY OUTCOMES trial resulted in a 1.6% absolute risk reduction after 2.8 years of follow-up [32]. Interestingly, there was a mortality benefit associated with the use of alirocumab, which may reflect the high-risk nature of the targeted cohort of acute coronary syndrome as opposed to stable patients in the FOURIER study. Both monoclonal antibodies are safe, with the most common adverse effect being a mild injection site reaction (itching, rash and swelling). There is no evidence to suggest worsening cognitive function with the use of PCSK9i despite achieving very low LDL-c [33]. One limitation to the routine use of PCSK9i is related to its costs, and various strategies were implemented to improve patient’s access to this promising treatment [34].
Inclisiran is a long-acting synthetic small interfering RNA inhibitor that selectively and catalytically silences the translation of PCSK9 mRNA in the hepatocytes. It is administered subcutaneously once every 6 months, but initially, early administration of three months following the first injection is required to boost the LDL-c reduction effect. This provides an advantage over monoclonal antibodies when inhibiting PCSK9 and may help overcome issues related to adherence to lipid-lowering therapies [35]. Several trials have effectively demonstrated that adding inclisiran to the maximum tolerated statin has resulted in a significant reduction in LDL-c, up to 50% [36]. Notably, the magnitude of LDL-c reduction with inclisiran is slightly lower than that of inhibiting PCSK9 using monoclonal antibodies [36]. Whether reducing LDL-c using inclisiran will translate into better clinical outcomes is yet to be determined. Like evolocumab and alirocumab, inclisiran is associated with infrequent side effects and predominately are related to local reactions at the administration site alongside muscular pain. Other side effects, such as upper respiratory tract symptoms, have also been reported with its use [37].
Bempedoic acid is the latest LDL-c-specific lowering treatment. It is an oral prodrug, and its metabolites target ATP-citrate lyase (ACL), an enzyme that is upstream from HMG coenzyme A reductase, the enzyme inhibited by statins [38]. Bempedoic acid provides a potential alternative for patients who are statin intolerant, but it could also be used in combination with statins [38,39]. Several studies highlighted its effect on lowering LDL-c either in combination with maximally tolerated statins or when used alone in statin-intolerable patients [40,41,42,43,44]. The CLEAR (Cholesterol Lowering via Bempedoic Acid [ECT1002], an ACL-Inhibiting Regimen) Outcomes trial demonstrated that bempedoic acid was associated with a 21% decrease in LDL-c after 6 months of treatment compared with placebo in patients who were intolerant to statins who had or were at high risk for, cardiovascular disease. Such difference in LDL-c has led to a 13% relative risk reduction in the incidence of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularisation when compared to placebo [44]. Importantly, there was an increase in the incidence of gout, cholelithiasis, and small derangements in serum creatinine and liver enzymes associated with the use of bempedoic acid compared to the placebo group [44]. The effect of bempedoic acid on creatinine has been small, stable and consistent regardless of baseline renal function. This is presumed to be related to the inhibition of tubular reabsorption of creatinine, although bempedoic acid has also been associated with small changes in urea, which appears early and is stable over time irrespective of baseline renal function [45]. Using another biomarker, such as cystatin C which is a muscle mass-independent marker, may provide more insights into the role of bempedoic acid on renal function [46].
Overall, the reduction in LDL-c, irrespective of the mechanism of the used drug, was consistently associated with improved clinical outcomes. Nonetheless, a significant proportion of patients remained at increased risk despite achieving an ‘optimal’ LDL-c level. Therefore, there was a rationale to target other lipoprotein particles aiming to reduce lipid-related atherosclerotic risk.

3. High-Density Lipoprotein Cholesterol Raising Treatment

Low plasma level of high-density lipoprotein cholesterol (HDL-c) is a recognised risk marker in patients with previous and recurrent cardiovascular event [47]. Data from the Framingham Heart Study highlighted a unique association between low HDL-c and cardiovascular death with less correlation with other causes of deaths [48]. However, subsequent Mendelian randomisation challenged the concept that raising HDL-c would translate into lowering cardiovascular risk [49]. This finding matched the results of pharmacotherapies aiming to increase HDL-c.
Cholesteryl ester transfer protein (CETP) inhibitors were used early to reduce future cardiovascular risk, with the aim of increasing HDL-c levels. CETP facilitates the exchange of triglycerides and cholesteryl ester between high-density lipoprotein (HDL) and apoB100-containing lipoproteins [50]. Evidence from genetic studies highlighted that variants in the CETP gene were associated with higher levels of HDL-c, lower LDL-c, and lower risk of coronary artery disease [50]. Therefore, inhibiting CETP may provide a similar lipid profile, which may translate into better clinical outcomes. The first CETP inhibitor, torcetrapib, was associated with a 25% increased risk of cardiovascular events despite achieving high HLD-c and low LDL-c levels [13]. The failure of this agent was thought to be related to off-target effects, including a rise in blood pressure and aldosterone levels [13]. Importantly, torcetrapib did not reduce the progression of coronary atheroma in the Investigation of Lipid Level Management Using Coronary Ultrasound to Assess Reduction of Atherosclerosis by CETP Inhibition and HDL Elevation (ILLUSTRATE) trial [51]. Nonetheless, all subsequent CETP inhibitors failed to show any incremental benefits in reducing clinical outcomes [3,4,52]. Anacetrapib was associated with a modest 9% relative risk reduction in major coronary events in patients with atherosclerotic vascular disease who were receiving intensive statin therapy. However, this effect was likely driven by the reduction in LDL-c of 18% associated with its use [12]. Obicetrapib is the newest CETP inhibitor and appears to be the most potent one by increasing HDL-c by 165% and reducing LDL-c by up to 51% in patients on high-intensity statin treatment [53,54]. The ongoing phase 3 Randomized Study to Evaluate the Effect of Obicetrapib on Top of Maximum Tolerated Lipid-Modifying Therapies (BROADWAY) trial will help ascertain the effect of raising HDL-c in the management of atherosclerotic disease.
Niacin is known to alter the lipid profile of patients with atherosclerotic disease by raising apo A1-containing HDL, reducing apoB lipoprotein particles, and inhibiting the synthesis of triglycerides in the hepatic cells, thus decreasing the secretion of LDL and VLDL particles [55]. Despite these favourable effects on lipoprotein particles, two large randomised trials did not demonstrate any reduction in cardiovascular events when patients were subjected to niacin compared to placebo [56,57]. Niacin was reported to increase the incidence of diabetes by 1.3%, as well as disturbing diabetic control by 3.7%. There was an unexpected increase in infection rate by 1.4% with the use of niacin [57]. Overall, this has led to limited use of niacin in the management of atherosclerotic disease.

4. Triglycerides Lowering Treatment

The relationship between hypertriglyceridemia and future adverse cardiovascular events is well established. Patients with mildly elevated triglycerides of more than 170 mg/dL had almost 50% increase in mortality [58]. A causal link between triglyceride-rich lipoprotein particles and the risk of ischaemic heart disease was suggested from previous Mendelian randomisation studies [59,60]. This was supported by statin trials suggesting a 10% reduction in the risk of death or recurrent myocardial infarction for each 1 mmol/L decrease in triglycerides level [61]. Treatments targeting hypertriglyceridemia have not been consistent in lowering future cardiovascular events.
Fibrates are one of the early therapies that lowered triglycerides via the nuclear receptor peroxisome proliferator-activated receptor α (PPARα). In statin-treated patients, fibrate did not reduce adverse cardiovascular events when compared to placebo [62,63,64]. Nonetheless, subgroup analyses have strongly linked a reduction in cardiovascular risk in response to fibrate treatment in patients with high triglycerides levels and low HDL-c, particularly if they have concomitant type 2 diabetes [65,66]. The Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients with Diabetes (PROMINENT) study assessed the use of pemafibrate, potent and selective PPARα modulator, in patients with type 2 diabetes, mild-to-moderate hypertriglyceridemia (200 to 499 mg/dL), and HDL-c ≤ 40 mg/dL [63]. The primary endpoint, defined as the composite of cardiovascular death, nonfatal myocardial infarction, ischemic stroke, and coronary revascularisation, was comparable to placebo [63].
Omega-3 fatty acids have historically been used to lower triglycerides. However, their data on reducing adverse clinical outcomes are conflicting [67,68]. The active metabolites of omega-3 fatty acids, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), have been assessed as adjunctive treatment to reduce future cardiovascular risk [69]. Whilst both fibrate and omega-3 fatty acids share a similar mechanism in reducing triglycerides by targeting PPARα, the former lacks the ability to increase the clearance of cholesterol remnants particles, resulting in no change and, sometimes, a slight increase in apo-B lipoprotein particles [69]. Early studies showed potential benefits in using omega-3 fatty acids. The JAPAN EPA Lipid Intervention Study (JELIS) assessed the role of high-dose purified EPA in 18,645 statin-treated patients with a total cholesterol ≥6.5 mmol/L [70]. There was almost 20% relative risk reduction in the composite endpoint of sudden cardiac death, fatal and non-fatal myocardial infarction, and other non-fatal events, including unstable angina pectoris, angioplasty, stenting, or coronary artery bypass grafting in patients receiving EPA [70]. The Reduction of Cardiovascular Events with Icosapent Ethyl–Intervention Trial (REDUCE-IT) highlighted almost 5% absolute risk reduction when subjecting statin-treated patients to an even higher dose of purified EPA (4 g/day) after a median follow-up of 4.9 years [67]. Notably, the clinical benefits were not related to the level of triglycerides but to the treatment of EPA, which may explain the inconsistency in the results of the studies assessing the role of omega-3 fatty acids [68,69]. There is a slight increase in the risk of bleeding and the incidence of atrial fibrillation associated with the use of high-dose EPA [67].
Emerging therapies targeting angiopoietin-like 3 protein (ANGPTL3) may add benefits to patients at increased cardiovascular risk. Blocking ANGPTL3 will disinhibit lipoprotein and endothelial lipase, leading to a reduction in triglycerides and LDL-c [16]. Evinacumab is a fully human monoclonal antibody inhibitor of ANGPTL3 and is currently approved for treating familial hypercholesterolemia [71]. It is administered intravenously every four weeks and has few reported adverse effects, including upper respiratory tract infections and flu-like syndromes. Like other intravenously administered drugs, evinacumab has the potential to cause severe allergic reactions or even anaphylaxis [72]. Vupanorsen is an antisense oligonucleotide that inhibits the liver synthesis of ANGPTL3. However, a recent clinical trial showed severe hepatic toxicity, questioning its safety clinical profile [71].

5. Lipoprotein-Specific Therapies

The fundamental steps in developing atherosclerosis are related to the accumulation and progression of cholesterol within the intima of vascular walls, triggering an inflammatory cellular cascade. This process is largely controlled by the influx of apo-B lipoproteins and the cholesterol efflux that is mediated by apo-A1. Cholesterol efflux capacity from macrophages is considered a metric of HDL functionality and is significantly determined by apo-A1 level [73]. Moreover, it has an inverse strong association with the development of atherosclerosis and the incidence of cardiovascular events [73,74]. Intuitively, increasing Apo-A1 resulted in an increase in ex vivo cholesterol efflux capacity in patients with acute myocardial infarction [75]. However, this did not translate into a reduction in cardiovascular events in the Rationale and Design of ApoA-I Event Reducing in Ischemic Syndromes II (AEGIS-II) study [76]. A novel approach has recently been proposed to decrease cholesterol by promoting its excretion. Loss-of-function variants of Asialoglycoprotein receptor 1 (ASGR1) were associated with low cholesterol and a reduction in cardiovascular disease [77]. ASGR1 deficiency was associated with increased cholesterol reverse transport via ABCA1 and ABCG5/G8 on HDL particles. The prevailing mechanism is related to stabilising LXRα, which subsequently upregulates ABCA1 and ABCG5/G8, enhancing cholesterol excretion to bile and faeces [77].
Since LDL-c is directly related to the development of atherosclerosis, reducing LDL may provide a therapeutic option to reduce future cardiovascular risks. Apolipoprotein B is one of the main components of LDL and VLDL particles and is central in packaging and redistributing cholesterol within the body. Antisense oligonucleotide treatments were developed to target hepatic synthesis of apoB100 and showed promising results by reducing apo-B up to 50% [78,79]. Mipomersen is a second-generation antisense that targets the coding region of the human apoB mRNA [79]. It is FDA-approved but has not yet been approved for use in Europe by the European Medicines Agency (EMA) [80]. A recent meta-analysis of conducted randomised clinical trials highlighted a mean LDL-c reduction of 26% in response to mipomersen [81]. However, mipomersen was associated with a few adverse effects, including elevation of liver enzymes, injection site reactions, and flu-like symptoms, which subsequently led to discontinuation in up to 20% of the patients investigated [81].
Numerous studies have demonstrated the importance of apoC-III in hypertriglyceridemia and its intrinsic proatherogenic effects [82,83]. ApoC-III is the current target for a new class of drugs aimed at lowering triglyceride levels and indirectly preventing cardiovascular incidents and pancreatitis episodes in patients with hypertriglyceridemia. The first of these drugs is volanesorsen, an antisense oligonucleotide that binds to the apoC-III mRNA and disrupts apoC-III translation, resulting in lower levels of chylomicrons and triglycerides [84]. Volanesorsen was associated with a 72% reduction in triglyceride levels, alongside pancreatic events, compared to placebo after three months [85]. The most common adverse effect of volanesorsen is injection site reaction; however, severe thrombocytopenia has also been observed [85]. Similar to volanesorsen, olezarsen is an antisense oligonucleotide targeting apoC-III, and through apoC-III translation disruption, chylomicrons and triglyceride levels are reduced. It is currently being studied, and initial results did not suggest thrombocytopenic risk [86].
Lipoprotein(a), Lp(a), is considered a variant of LDL as both contain apo-B lipoprotein, but Lp(a) carries lipoprotein (a), which is due to its structure is a promotor of inflammation and thrombosis. Lp(a) is genetically determined and not modified by exercise or diet. It is known to be an independent risk factor for atherosclerosis and cardiovascular events [87]. Therefore, it is currently being pursued as a therapeutic target to reduce future cardiovascular risk. Existing therapies such as statins are not associated with a clinically significant reduction in Lp(a) level. Likewise, PCSK9 inhibitors have a relatively marginal lowering effect, causing a 20–25% reduction in Lp(a) [31,32]. The HORIZON (Assessing the Impact of Lipoprotein(a) Lowering with pelacarsen (TQJ230) on Major Cardiovascular Events in Patients With CVD) outcomes trial is a phase 3 study that is currently assessing the role of pelacarsen, an antisense oligonucleotide, on reducing future cardiovascular risk [88]. Pelacarsen is a subcutaneously administered medication that binds to hepatocyte apo(a) mRNA, preventing the translation of apolipoprotein(a). It was associated with a significant reduction, up to 67%, in the level of circulating Lp(a) [89]. Olpasiran is another small-interfering RNA that reduces hepatic Lp(a) synthesis. In the phase 2 study, olpasiran was associated with a significant reduction in placebo-adjusted mean percent changes of up to 100% [90]. The outcomes of the phase 3 trial will be eagerly awaited.

6. Conclusions

Statins remain the cornerstone in the management of patients with atherosclerotic disease. However, a significant proportion of patients remain at increased risk, despite receiving maximal statin dose. This risk is related to sub-optimal LDL-c level but may also be derived from other lipoprotein-related risk. It is important to recognise the availability of other therapies that may reduce this residual lipid risk (Summary of landmark trials and mechanisms of available treatments is presented in Table 2 and Figure 1).

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Collet, J.P.; Thiele, H.; Barbato, E.; Barthelemy, O.; Bauersachs, J.; Bhatt, D.L.; Dendale, P.; Dorobantu, M.; Edvardsen, T.; Folliguet, T.; et al. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Eur. Heart J. 2021, 42, 1289–1367. [Google Scholar] [CrossRef]
  2. Abu-Assi, E.; Lopez-Lopez, A.; Gonzalez-Salvado, V.; Redondo-Dieguez, A.; Pena-Gil, C.; Bouzas-Cruz, N.; Raposeiras-Roubin, S.; Riziq-Yousef Abumuaileq, R.; Garcia-Acuna, J.M.; Gonzalez-Juanatey, J.R. The Risk of Cardiovascular Events After an Acute Coronary Event Remains High, Especially During the First Year, Despite Revascularization. Rev. Esp. Cardiol. 2016, 69, 11–18. [Google Scholar] [CrossRef]
  3. Alkhalil, M. Mechanistic Insights to Target Atherosclerosis Residual Risk. Curr. Probl. Cardiol. 2021, 46, 100432. [Google Scholar] [CrossRef]
  4. Alkhalil, M.; Chai, J.T.; Choudhury, R.P. Plaque imaging to refine indications for emerging lipid-lowering drugs. Eur. Heart J. Cardiovasc. Pharmacother. 2017, 3, 58–67. [Google Scholar] [CrossRef]
  5. Li, C.; Pan, Y.; Zhang, R.; Huang, Z.; Li, D.; Han, Y.; Larkin, C.; Rao, V.; Sun, X.; Kelly, T.N. Genomic Innovation in Early Life Cardiovascular Disease Prevention and Treatment. Circ. Res. 2023, 132, 1628–1647. [Google Scholar] [CrossRef]
  6. Alkhalil, M.; Edmond, E.; Edgar, L.; Digby, J.E.; Omar, O.; Robson, M.D.; Choudhury, R.P. The relationship of perivascular adipose tissue and atherosclerosis in the aorta and carotid arteries, determined by magnetic resonance imaging. Diabetes Vasc. Dis. Res. 2018, 15, 286–293. [Google Scholar] [CrossRef]
  7. Oikonomou, E.K.; Marwan, M.; Desai, M.Y.; Mancio, J.; Alashi, A.; Hutt Centeno, E.; Thomas, S.; Herdman, L.; Kotanidis, C.P.; Thomas, K.E.; et al. Non-invasive detection of coronary inflammation using computed tomography and prediction of residual cardiovascular risk (the CRISP CT study): A post-hoc analysis of prospective outcome data. Lancet 2018, 392, 929–939. [Google Scholar] [CrossRef]
  8. Serruys, P.W.; de Feyter, P.; Macaya, C.; Kokott, N.; Puel, J.; Vrolix, M.; Branzi, A.; Bertolami, M.C.; Jackson, G.; Strauss, B.; et al. Fluvastatin for prevention of cardiac events following successful first percutaneous coronary intervention: A randomized controlled trial. JAMA 2002, 287, 3215–3222. [Google Scholar] [CrossRef]
  9. Castelli, W.P.; Garrison, R.J.; Wilson, P.W.; Abbott, R.D.; Kalousdian, S.; Kannel, W.B. Incidence of coronary heart disease and lipoprotein cholesterol levels. The Framingham Study. JAMA 1986, 256, 2835–2838. [Google Scholar] [CrossRef]
  10. Cannon, C.P.; Steinberg, B.A.; Murphy, S.A.; Mega, J.L.; Braunwald, E. Meta-analysis of cardiovascular outcomes trials comparing intensive versus moderate statin therapy. J. Am. Coll. Cardiol. 2006, 48, 438–445. [Google Scholar] [CrossRef]
  11. 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]
  12. Group, H.T.R.C.; Bowman, L.; Hopewell, J.C.; Chen, F.; Wallendszus, K.; Stevens, W.; Collins, R.; Wiviott, S.D.; Cannon, C.P.; Braunwald, E.; et al. Effects of Anacetrapib in Patients with Atherosclerotic Vascular Disease. N. Engl. J. Med. 2017, 377, 1217–1227. [Google Scholar] [CrossRef]
  13. Barter, P.J.; Caulfield, M.; Eriksson, M.; Grundy, S.M.; Kastelein, J.J.; Komajda, M.; Lopez-Sendon, J.; Mosca, L.; Tardif, J.C.; Waters, D.D.; et al. Effects of torcetrapib in patients at high risk for coronary events. N. Engl. J. Med. 2007, 357, 2109–2122. [Google Scholar] [CrossRef]
  14. Fayad, Z.A.; Mani, V.; Woodward, M.; Kallend, D.; Abt, M.; Burgess, T.; Fuster, V.; Ballantyne, C.M.; Stein, E.A.; Tardif, J.C.; et al. Safety and efficacy of dalcetrapib on atherosclerotic disease using novel non-invasive multimodality imaging (dal-PLAQUE): A randomised clinical trial. Lancet 2011, 378, 1547–1559. [Google Scholar] [CrossRef]
  15. Virani, S.S. The Fibrates Story—A Tepid End to a PROMINENT Drug. N. Engl. J. Med. 2022, 387, 1991–1992. [Google Scholar] [CrossRef]
  16. Lang, W.; Frishman, W.H. Angiopoietin-Like 3 Protein Inhibition: A New Frontier in Lipid-Lowering Treatment. Cardiol. Rev. 2019, 27, 211–217. [Google Scholar] [CrossRef]
  17. Cholesterol Treatment Trialists, C.; Baigent, C.; Blackwell, L.; Emberson, J.; Holland, L.E.; Reith, C.; Bhala, N.; Peto, R.; Barnes, E.H.; Keech, A.; 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] [CrossRef]
  18. Williams, K.J.; Tabas, I. The response-to-retention hypothesis of early atherogenesis. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 551–561. [Google Scholar] [CrossRef]
  19. Boren, J.; Chapman, M.J.; Krauss, R.M.; Packard, C.J.; Bentzon, J.F.; Binder, C.J.; Daemen, M.J.; Demer, L.L.; Hegele, R.A.; Nicholls, S.J.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease: Pathophysiological, genetic, and therapeutic insights: A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2020, 41, 2313–2330. [Google Scholar] [CrossRef]
  20. Ference, B.A.; Kastelein, J.J.P.; Catapano, A.L. Lipids and Lipoproteins in 2020. JAMA 2020, 324, 595–596. [Google Scholar] [CrossRef]
  21. 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]
  22. Alkhalil, M. Effects of intensive lipid-lowering therapy on mortality after coronary bypass surgery: A meta-analysis of 7 randomised trials. Atherosclerosis 2020, 293, 75–78. [Google Scholar] [CrossRef]
  23. Alkhalil, M.; Kuzemczak, M.; Whitehead, N.; Kavvouras, C.; Dzavik, V. Meta-Analysis of Intensive Lipid-Lowering Therapy in Patients with Polyvascular Disease. J. Am. Heart Assoc. 2021, 10, e017948. [Google Scholar] [CrossRef]
  24. Ridker, P.M.; Danielson, E.; Fonseca, F.A.; Genest, J.; Gotto, A.M., Jr.; Kastelein, J.J.; Koenig, W.; Libby, P.; Lorenzatti, A.J.; Macfadyen, J.G.; et al. Reduction in C-reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: A prospective study of the JUPITER trial. Lancet 2009, 373, 1175–1182. [Google Scholar] [CrossRef]
  25. Alkhalil, M.; Biasiolli, L.; Akbar, N.; Galassi, F.; Chai, J.T.; Robson, M.D.; Choudhury, R.P. T2 mapping MRI technique quantifies carotid plaque lipid, and its depletion after statin initiation, following acute myocardial infarction. Atherosclerosis 2018, 279, 100–106. [Google Scholar] [CrossRef]
  26. Ramkumar, S.; Raghunath, A.; Raghunath, S. Statin Therapy: Review of Safety and Potential Side Effects. Acta Cardiol. Sin. 2016, 32, 631–639. [Google Scholar] [CrossRef]
  27. Jiang, S.Y.; Li, H.; Tang, J.J.; Wang, J.; Luo, J.; Liu, B.; Wang, J.K.; Shi, X.J.; Cui, H.W.; Tang, J.; et al. Discovery of a potent HMG-CoA reductase degrader that eliminates statin-induced reductase accumulation and lowers cholesterol. Nat. Commun. 2018, 9, 5138. [Google Scholar] [CrossRef]
  28. Jarcho, J.A.; Keaney, J.F., Jr. Proof That Lower Is Better--LDL Cholesterol and IMPROVE-IT. N. Engl. J. Med. 2015, 372, 2448–2450. [Google Scholar] [CrossRef]
  29. Kosoglou, T.; Statkevich, P.; Johnson-Levonas, A.O.; Paolini, J.F.; Bergman, A.J.; Alton, K.B. Ezetimibe: A review of its metabolism, pharmacokinetics and drug interactions. Clin. Pharmacokinet. 2005, 44, 467–494. [Google Scholar] [CrossRef]
  30. Tsujita, K.; Sugiyama, S.; Sumida, H.; Shimomura, H.; Yamashita, T.; Yamanaga, K.; Komura, N.; Sakamoto, K.; Oka, H.; Nakao, K.; et al. Impact of Dual Lipid-Lowering Strategy with Ezetimibe and Atorvastatin on Coronary Plaque Regression in Patients With Percutaneous Coronary Intervention: The Multicenter Randomized Controlled PRECISE-IVUS Trial. J. Am. Coll. Cardiol. 2015, 66, 495–507. [Google Scholar] [CrossRef]
  31. 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]
  32. 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]
  33. Giugliano, R.P.; Mach, F.; Zavitz, K.; Kurtz, C.; Im, K.; Kanevsky, E.; Schneider, J.; Wang, H.; Keech, A.; Pedersen, T.R.; et al. Cognitive Function in a Randomized Trial of Evolocumab. N. Engl. J. Med. 2017, 377, 633–643. [Google Scholar] [CrossRef]
  34. Alkhalil, M. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Inhibitors, Reality or Dream in Managing Patients with Cardiovascular Disease. Curr. Drug Metab. 2019, 20, 72–82. [Google Scholar] [CrossRef]
  35. Fitzgerald, K.; White, S.; Borodovsky, A.; Bettencourt, B.R.; Strahs, A.; Clausen, V.; Wijngaard, P.; Horton, J.D.; Taubel, J.; Brooks, A.; et al. A Highly Durable RNAi Therapeutic Inhibitor of PCSK9. N. Engl. J. Med. 2017, 376, 41–51. [Google Scholar] [CrossRef]
  36. 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]
  37. Mercep, I.; Friscic, N.; Strikic, D.; Reiner, Z. Advantages and Disadvantages of Inclisiran: A Small Interfering Ribonucleic Acid Molecule Targeting PCSK9-A Narrative Review. Cardiovasc. Ther. 2022, 2022, 8129513. [Google Scholar] [CrossRef]
  38. Pinkosky, S.L.; Newton, R.S.; Day, E.A.; Ford, R.J.; Lhotak, S.; Austin, R.C.; Birch, C.M.; Smith, B.K.; Filippov, S.; Groot, P.H.E.; et al. Liver-specific ATP-citrate lyase inhibition by bempedoic acid decreases LDL-C and attenuates atherosclerosis. Nat. Commun. 2016, 7, 13457. [Google Scholar] [CrossRef]
  39. Bardolia, C.; Amin, N.S.; Turgeon, J. Emerging Non-statin Treatment Options for Lowering Low-Density Lipoprotein Cholesterol. Front. Cardiovasc. Med. 2021, 8, 789931. [Google Scholar] [CrossRef]
  40. Laufs, U.; Banach, M.; Mancini, G.B.J.; Gaudet, D.; Bloedon, L.T.; Sterling, L.R.; Kelly, S.; Stroes, E.S.G. Efficacy and Safety of Bempedoic Acid in Patients with Hypercholesterolemia and Statin Intolerance. J. Am. Heart Assoc. 2019, 8, e011662. [Google Scholar] [CrossRef]
  41. Goldberg, A.C.; Leiter, L.A.; Stroes, E.S.G.; Baum, S.J.; Hanselman, J.C.; Bloedon, L.T.; Lalwani, N.D.; Patel, P.M.; Zhao, X.; Duell, P.B. Effect of Bempedoic Acid vs Placebo Added to Maximally Tolerated Statins on Low-Density Lipoprotein Cholesterol in Patients at High Risk for Cardiovascular Disease: The CLEAR Wisdom Randomized Clinical Trial. JAMA 2019, 322, 1780–1788. [Google Scholar] [CrossRef]
  42. Ray, K.K.; Bays, H.E.; Catapano, A.L.; Lalwani, N.D.; Bloedon, L.T.; Sterling, L.R.; Robinson, P.L.; Ballantyne, C.M.; Trial, C.H. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N. Engl. J. Med. 2019, 380, 1022–1032. [Google Scholar] [CrossRef]
  43. Ballantyne, C.M.; Banach, M.; Mancini, G.B.J.; Lepor, N.E.; Hanselman, J.C.; Zhao, X.; Leiter, L.A. Efficacy and safety of bempedoic acid added to ezetimibe in statin-intolerant patients with hypercholesterolemia: A randomized, placebo-controlled study. Atherosclerosis 2018, 277, 195–203. [Google Scholar] [CrossRef]
  44. Nissen, S.E.; Lincoff, A.M.; Brennan, D.; Ray, K.K.; Mason, D.; Kastelein, J.J.P.; Thompson, P.D.; Libby, P.; Cho, L.; Plutzky, J.; et al. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N. Engl. J. Med. 2023, 388, 1353–1364. [Google Scholar] [CrossRef]
  45. Bays, H.E.; Bloedon, L.T.; Lin, G.; Powell, H.A.; Louie, M.J.; Nicholls, S.J.; Lincoff, A.M.; Nissen, S.E. Safety of bempedoic acid in patients at high cardiovascular risk and with statin intolerance. J. Clin. Lipidol. 2024, 18, e59–e69. [Google Scholar] [CrossRef]
  46. Groothof, D.; Post, A.; Polinder-Bos, H.A.; Erler, N.S.; Flores-Guerrero, J.L.; Kootstra-Ros, J.E.; Pol, R.A.; de Borst, M.H.; Gansevoort, R.T.; Gans, R.O.B.; et al. Muscle mass and estimates of renal function: A longitudinal cohort study. J. Cachexia Sarcopenia Muscle 2022, 13, 2031–2043. [Google Scholar] [CrossRef]
  47. Di Vito, L.; Di Giusto, F.; Bruscoli, F.; Scalone, G.; Silenzi, S.; Mariani, L.; Selimi, A.; Delfino, D.; Grossi, P. Recurrent events after acute ST-segment elevation myocardial infarction: Predictors and features of plaque progression and stent failure. Coron. Artery Dis. 2024. [Google Scholar] [CrossRef]
  48. Wilson, P.W.; Abbott, R.D.; Castelli, W.P. High density lipoprotein cholesterol and mortality. The Framingham Heart Study. Arteriosclerosis 1988, 8, 737–741. [Google Scholar] [CrossRef]
  49. Voight, B.F.; Peloso, G.M.; Orho-Melander, M.; Frikke-Schmidt, R.; Barbalic, M.; Jensen, M.K.; Hindy, G.; Holm, H.; Ding, E.L.; Johnson, T.; et al. Plasma HDL cholesterol and risk of myocardial infarction: A mendelian randomisation study. Lancet 2012, 380, 572–580. [Google Scholar] [CrossRef]
  50. Armitage, J.; Holmes, M.V.; Preiss, D. Cholesteryl Ester Transfer Protein Inhibition for Preventing Cardiovascular Events: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2019, 73, 477–487. [Google Scholar] [CrossRef]
  51. Nissen, S.E.; Tardif, J.C.; Nicholls, S.J.; Revkin, J.H.; Shear, C.L.; Duggan, W.T.; Ruzyllo, W.; Bachinsky, W.B.; Lasala, G.P.; Tuzcu, E.M.; et al. Effect of torcetrapib on the progression of coronary atherosclerosis. N. Engl. J. Med. 2007, 356, 1304–1316. [Google Scholar] [CrossRef]
  52. Alkhalil, M.; Choudhury, R.P. Current concepts in atherosclerosis. Indian J. Thorac. Cardiovasc. Surg. 2018, 34, 198–205. [Google Scholar] [CrossRef]
  53. Nurmohamed, N.S.; Navar, A.M.; Kastelein, J.J.P. New and Emerging Therapies for Reduction of LDL-Cholesterol and Apolipoprotein B: JACC Focus Seminar 1/4. J. Am. Coll. Cardiol. 2021, 77, 1564–1575. [Google Scholar] [CrossRef]
  54. Nicholls, S.J.; Ditmarsch, M.; Kastelein, J.J.; Rigby, S.P.; Kling, D.; Curcio, D.L.; Alp, N.J.; Davidson, M.H. Lipid lowering effects of the CETP inhibitor obicetrapib in combination with high-intensity statins: A randomized phase 2 trial. Nat. Med. 2022, 28, 1672–1678. [Google Scholar] [CrossRef]
  55. Kamanna, V.S.; Kashyap, M.L. Mechanism of action of niacin. Am. J. Cardiol. 2008, 101, 20B–26B. [Google Scholar] [CrossRef]
  56. Investigators, A.-H.; Boden, W.E.; Probstfield, J.L.; Anderson, T.; Chaitman, B.R.; Desvignes-Nickens, P.; Koprowicz, K.; McBride, R.; Teo, K.; Weintraub, W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N. Engl. J. Med. 2011, 365, 2255–2267. [Google Scholar] [CrossRef]
  57. Group, H.T.C.; Landray, M.J.; Haynes, R.; Hopewell, J.C.; Parish, S.; Aung, T.; Tomson, J.; Wallendszus, K.; Craig, M.; Jiang, L.; et al. Effects of extended-release niacin with laropiprant in high-risk patients. N. Engl. J. Med. 2014, 371, 203–212. [Google Scholar] [CrossRef]
  58. Nordestgaard, B.G.; Benn, M.; Schnohr, P.; Tybjaerg-Hansen, A. Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. JAMA 2007, 298, 299–308. [Google Scholar] [CrossRef]
  59. Varbo, A.; Benn, M.; Tybjaerg-Hansen, A.; Nordestgaard, B.G. Elevated remnant cholesterol causes both low-grade inflammation and ischemic heart disease, whereas elevated low-density lipoprotein cholesterol causes ischemic heart disease without inflammation. Circulation 2013, 128, 1298–1309. [Google Scholar] [CrossRef]
  60. Varbo, A.; Benn, M.; Tybjaerg-Hansen, A.; Jorgensen, A.B.; Frikke-Schmidt, R.; Nordestgaard, B.G. Remnant cholesterol as a causal risk factor for ischemic heart disease. J. Am. Coll. Cardiol. 2013, 61, 427–436. [Google Scholar] [CrossRef]
  61. Miller, M.; Cannon, C.P.; Murphy, S.A.; Qin, J.; Ray, K.K.; Braunwald, E.; Investigators, P.I.-T. Impact of triglyceride levels beyond low-density lipoprotein cholesterol after acute coronary syndrome in the PROVE IT-TIMI 22 trial. J. Am. Coll. Cardiol. 2008, 51, 724–730. [Google Scholar] [CrossRef]
  62. Arai, H.; Yamashita, S.; Yokote, K.; Araki, E.; Suganami, H.; Ishibashi, S.; Group, K.S. Efficacy and safety of K-877, a novel selective peroxisome proliferator-activated receptor alpha modulator (SPPARMalpha), in combination with statin treatment: Two randomised, double-blind, placebo-controlled clinical trials in patients with dyslipidaemia. Atherosclerosis 2017, 261, 144–152. [Google Scholar] [CrossRef]
  63. Das Pradhan, A.; Glynn, R.J.; Fruchart, J.C.; MacFadyen, J.G.; Zaharris, E.S.; Everett, B.M.; Campbell, S.E.; Oshima, R.; Amarenco, P.; Blom, D.J.; et al. Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N. Engl. J. Med. 2022, 387, 1923–1934. [Google Scholar] [CrossRef]
  64. Group, A.S.; Ginsberg, H.N.; Elam, M.B.; Lovato, L.C.; Crouse, J.R., 3rd; Leiter, L.A.; Linz, P.; Friedewald, W.T.; Buse, J.B.; Gerstein, H.C.; et al. Effects of combination lipid therapy in type 2 diabetes mellitus. N. Engl. J. Med. 2010, 362, 1563–1574. [Google Scholar] [CrossRef]
  65. Jun, M.; Foote, C.; Lv, J.; Neal, B.; Patel, A.; Nicholls, S.J.; Grobbee, D.E.; Cass, A.; Chalmers, J.; Perkovic, V. Effects of fibrates on cardiovascular outcomes: A systematic review and meta-analysis. Lancet 2010, 375, 1875–1884. [Google Scholar] [CrossRef]
  66. Sacks, F.M.; Carey, V.J.; Fruchart, J.C. Combination lipid therapy in type 2 diabetes. N. Engl. J. Med. 2010, 363, 692–694; author reply 694–695. [Google Scholar] [CrossRef]
  67. Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T., Jr.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N. Engl. J. Med. 2019, 380, 11–22. [Google Scholar] [CrossRef]
  68. Nicholls, S.J.; Lincoff, A.M.; Garcia, M.; Bash, D.; Ballantyne, C.M.; Barter, P.J.; Davidson, M.H.; Kastelein, J.J.P.; Koenig, W.; McGuire, D.K.; et al. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020, 324, 2268–2280. [Google Scholar] [CrossRef]
  69. Gupta, A.; Alkhalil, M. The Emerging Role of Icosapent Ethyl in Patients with Cardiovascular Disease: Mechanistic Insights and Future Applications. J. Clin. Med. 2023, 12, 3758. [Google Scholar] [CrossRef]
  70. Yokoyama, M.; Origasa, H.; Matsuzaki, M.; Matsuzawa, Y.; Saito, Y.; Ishikawa, Y.; Oikawa, S.; Sasaki, J.; Hishida, H.; Itakura, H.; et al. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): A randomised open-label, blinded endpoint analysis. Lancet 2007, 369, 1090–1098. [Google Scholar] [CrossRef]
  71. Mercep, I.; Strikic, D.; Sliskovic, A.M.; Reiner, Z. New Therapeutic Approaches in Treatment of Dyslipidaemia-A Narrative Review. Pharmaceuticals 2022, 15, 839. [Google Scholar] [CrossRef] [PubMed]
  72. Raal, F.J.; Rosenson, R.S.; Reeskamp, L.F.; Hovingh, G.K.; Kastelein, J.J.P.; Rubba, P.; Ali, S.; Banerjee, P.; Chan, K.C.; Gipe, D.A.; et al. Evinacumab for Homozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 383, 711–720. [Google Scholar] [CrossRef] [PubMed]
  73. Khera, A.V.; Cuchel, M.; de la Llera-Moya, M.; Rodrigues, A.; Burke, M.F.; Jafri, K.; French, B.C.; Phillips, J.A.; Mucksavage, M.L.; Wilensky, R.L.; et al. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N. Engl. J. Med. 2011, 364, 127–135. [Google Scholar] [CrossRef] [PubMed]
  74. Rohatgi, A.; Khera, A.; Berry, J.D.; Givens, E.G.; Ayers, C.R.; Wedin, K.E.; Neeland, I.J.; Yuhanna, I.S.; Rader, D.R.; de Lemos, J.A.; et al. HDL cholesterol efflux capacity and incident cardiovascular events. N. Engl. J. Med. 2014, 371, 2383–2393. [Google Scholar] [CrossRef] [PubMed]
  75. Michael Gibson, C.; Korjian, S.; Tricoci, P.; Daaboul, Y.; Yee, M.; Jain, P.; Alexander, J.H.; Steg, P.G.; Lincoff, A.M.; Kastelein, J.J.; et al. Safety and Tolerability of CSL112, a Reconstituted, Infusible, Plasma-Derived Apolipoprotein A-I, After Acute Myocardial Infarction: The AEGIS-I Trial (ApoA-I Event Reducing in Ischemic Syndromes I). Circulation 2016, 134, 1918–1930. [Google Scholar] [CrossRef] [PubMed]
  76. Gibson, C.M.; Kastelein, J.J.P.; Phillips, A.T.; Aylward, P.E.; Yee, M.K.; Tendera, M.; Nicholls, S.J.; Pocock, S.; Goodman, S.G.; Alexander, J.H.; et al. Rationale and design of ApoA-I Event Reducing in Ischemic Syndromes II (AEGIS-II): A phase 3, multicenter, double-blind, randomized, placebo-controlled, parallel-group study to investigate the efficacy and safety of CSL112 in subjects after acute myocardial infarction. Am. Heart J. 2021, 231, 121–127. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, J.Q.; Li, L.L.; Hu, A.; Deng, G.; Wei, J.; Li, Y.F.; Liu, Y.B.; Lu, X.Y.; Qiu, Z.P.; Shi, X.J.; et al. Inhibition of ASGR1 decreases lipid levels by promoting cholesterol excretion. Nature 2022, 608, 413–420. [Google Scholar] [CrossRef] [PubMed]
  78. Kastelein, J.J.; Wedel, M.K.; Baker, B.F.; Su, J.; Bradley, J.D.; Yu, R.Z.; Chuang, E.; Graham, M.J.; Crooke, R.M. Potent reduction of apolipoprotein B and low-density lipoprotein cholesterol by short-term administration of an antisense inhibitor of apolipoprotein B. Circulation 2006, 114, 1729–1735. [Google Scholar] [CrossRef]
  79. Visser, M.E.; Witztum, J.L.; Stroes, E.S.; Kastelein, J.J. Antisense oligonucleotides for the treatment of dyslipidaemia. Eur. Heart J. 2012, 33, 1451–1458. [Google Scholar] [CrossRef]
  80. Geary, R.S.; Baker, B.F.; Crooke, S.T. Clinical and preclinical pharmacokinetics and pharmacodynamics of mipomersen (kynamro®): A second-generation antisense oligonucleotide inhibitor of apolipoprotein B. Clin. Pharmacokinet. 2015, 54, 133–146. [Google Scholar] [CrossRef]
  81. Fogacci, F.; Ferri, N.; Toth, P.P.; Ruscica, M.; Corsini, A.; Cicero, A.F.G. Efficacy and Safety of Mipomersen: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Drugs 2019, 79, 751–766. [Google Scholar] [CrossRef]
  82. Reiner, Z. Hypertriglyceridaemia and risk of coronary artery disease. Nat. Rev. Cardiol. 2017, 14, 401–411. [Google Scholar] [CrossRef] [PubMed]
  83. Hegele, R.A.; Tsimikas, S. Lipid-Lowering Agents. Circ. Res. 2019, 124, 386–404. [Google Scholar] [CrossRef]
  84. Witztum, J.L.; Gaudet, D.; Freedman, S.D.; Alexander, V.J.; Digenio, A.; Williams, K.R.; Yang, Q.; Hughes, S.G.; Geary, R.S.; Arca, M.; et al. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N. Engl. J. Med. 2019, 381, 531–542. [Google Scholar] [CrossRef]
  85. Gouni-Berthold, I.; Alexander, V.J.; Yang, Q.; Hurh, E.; Steinhagen-Thiessen, E.; Moriarty, P.M.; Hughes, S.G.; Gaudet, D.; Hegele, R.A.; O’Dea, L.S.L.; et al. Efficacy and safety of volanesorsen in patients with multifactorial chylomicronaemia (COMPASS): A multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol. 2021, 9, 264–275. [Google Scholar] [CrossRef]
  86. Tardif, J.C.; Karwatowska-Prokopczuk, E.; Amour, E.S.; Ballantyne, C.M.; Shapiro, M.D.; Moriarty, P.M.; Baum, S.J.; Hurh, E.; Bartlett, V.J.; Kingsbury, J.; et al. Apolipoprotein C-III reduction in subjects with moderate hypertriglyceridaemia and at high cardiovascular risk. Eur. Heart J. 2022, 43, 1401–1412. [Google Scholar] [CrossRef]
  87. 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]
  88. Malick, W.A.; Goonewardena, S.N.; Koenig, W.; Rosenson, R.S. Clinical Trial Design for Lipoprotein(a)-Lowering Therapies: JACC Focus Seminar 2/3. J. Am. Coll. Cardiol. 2023, 81, 1633–1645. [Google Scholar] [CrossRef]
  89. Yeang, C.; Karwatowska-Prokopczuk, E.; Su, F.; Dinh, B.; Xia, S.; Witztum, J.L.; Tsimikas, S. Effect of Pelacarsen on Lipoprotein(a) Cholesterol and Corrected Low-Density Lipoprotein Cholesterol. J. Am. Coll. Cardiol. 2022, 79, 1035–1046. [Google Scholar] [CrossRef]
  90. O’Donoghue, M.L.; Rosenson, R.S.; Gencer, B.; Lopez, 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]
  91. Baigent, C.; Landray, M.J.; Reith, C.; Emberson, J.; Wheeler, D.C.; Tomson, C.; Wanner, C.; Krane, V.; Cass, A.; Craig, J.; et al. The effects of lowering LDL cholesterol with simvastatin plus ezetimibe in patients with chronic kidney disease (Study of Heart and Renal Protection): A randomised placebo-controlled trial. Lancet 2011, 377, 2181–2192. [Google Scholar] [CrossRef]
  92. Ouchi, Y.; Sasaki, J.; Arai, H.; Yokote, K.; Harada, K.; Katayama, Y.; Urabe, T.; Uchida, Y.; Hayashi, M.; Yokota, N.; et al. Ezetimibe Lipid-Lowering Trial on Prevention of Atherosclerotic Cardiovascular Disease in 75 or Older (EWTOPIA 75): A Randomized, Controlled Trial. Circulation 2019, 140, 992–1003. [Google Scholar] [CrossRef]
  93. Ray, K.K.; Troquay, R.P.T.; Visseren, F.L.J.; Leiter, L.A.; Scott Wright, R.; Vikarunnessa, S.; Talloczy, Z.; Zang, X.; Maheux, P.; Lesogor, A.; et al. Long-term efficacy and safety of inclisiran in patients with high cardiovascular risk and elevated LDL cholesterol (ORION-3): Results from the 4-year open-label extension of the ORION-1 trial. Lancet Diabetes Endocrinol. 2023, 11, 109–119. [Google Scholar] [CrossRef]
  94. Koren, M.J.; Lundqvist, P.; Bolognese, M.; Neutel, J.M.; Monsalvo, M.L.; Yang, J.; Kim, J.B.; Scott, R.; Wasserman, S.M.; Bays, H.; et al. Anti-PCSK9 monotherapy for hypercholesterolemia: The MENDEL-2 randomized, controlled phase III clinical trial of evolocumab. J. Am. Coll. Cardiol. 2014, 63, 2531–2540. [Google Scholar] [CrossRef]
  95. Nicholls, S.J.; Puri, R.; Anderson, T.; Ballantyne, C.M.; Cho, L.; Kastelein, J.J.; Koenig, W.; Somaratne, R.; Kassahun, H.; Yang, J.; et al. Effect of Evolocumab on Progression of Coronary Disease in Statin-Treated Patients: The GLAGOV Randomized Clinical Trial. JAMA 2016, 316, 2373–2384. [Google Scholar] [CrossRef]
  96. Toth, P.P. Emerging LDL therapies: Mipomersen-antisense oligonucleotide therapy in the management of hypercholesterolemia. J. Clin. Lipidol. 2013, 7, S6–S10. [Google Scholar] [CrossRef]
  97. Group, R.C.; Bowman, L.; Chen, F.; Sammons, E.; Hopewell, J.C.; Wallendszus, K.; Stevens, W.; Valdes- Marquez, E.; Wiviott, S.; Cannon, C.P.; et al. Randomized Evaluation of the Effects of Anacetrapib through Lipid-modification (REVEAL)-A large-scale, randomized, placebo-controlled trial of the clinical effects of anacetrapib among people with established vascular disease: Trial design, recruitment, and baseline characteristics. Am. Heart J. 2017, 187, 182–190. [Google Scholar] [CrossRef]
  98. Sosnowska, B.; Adach, W.; Surma, S.; Rosenson, R.S.; Banach, M. Evinacumab, an ANGPTL3 Inhibitor, in the Treatment of Dyslipidemia. J. Clin. Med. 2022, 12, 168. [Google Scholar] [CrossRef]
  99. Yokoyama, M. [Japan EPA Lipid Intervention Study (JELIS). Randomized clinical trial involving primary and secondary prevention of cardiovascular events with EPA in hypercholesterolemia]. Nihon Ronen Igakkai Zasshi 2009, 46, 22–25. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of non-statin drugs (ezetimibe, PCSK9 inhibitors, bempedoic acid, ApoB100 mRNA inhibitor, ApoCIII inhibitors, ANGPLT3 inhibitor, CETPi, MTP inhibitors, PCSK9 MAB, and PCSK9 mRNA inhibitor inclisiran). CoA, coenzyme A; ACLY, ATP citrate lyase; HMG-CR, HMG-CoA reductase; NPC1L1, Niemann-Pick C1-Like 1; mRNA, messenger RNA; LPL, lipoprotein lipase; MTP, Microsomal Triglyceride transfer protein; ANGPLT3i, Angiopoietin-Like Protein 3 (ANGPTL3) Inhibitors; PCSK9, proprotein convertase subtilisin/kexin type 9; PCSK9 MAB, proprotein convertase subtilisin/kexin type 9 monoclonal antibody; PCSK9 mRNA, proprotein convertase subtilisin/kexin type 9 messenger RNA; CETPi, cholesteryl ester transfer protein inhibitor; CM, Chylomicrons; VLDL, very low-density lipoprotein; IDL, intermediate-density lipoprotein; LDL, low-density lipoprotein; HDL, high-density lipoprotein; TG, Triglyceride; ApoCIIIi, Apolipoprotein C-III inhibitors; ApoB100 mRNA inhibitor, Apolipoprotein B100 messenger RNA inhibitors; ApoB100, Apolipoprotein B100; LDLR, low-density lipoprotein receptor.
Figure 1. Mechanisms of non-statin drugs (ezetimibe, PCSK9 inhibitors, bempedoic acid, ApoB100 mRNA inhibitor, ApoCIII inhibitors, ANGPLT3 inhibitor, CETPi, MTP inhibitors, PCSK9 MAB, and PCSK9 mRNA inhibitor inclisiran). CoA, coenzyme A; ACLY, ATP citrate lyase; HMG-CR, HMG-CoA reductase; NPC1L1, Niemann-Pick C1-Like 1; mRNA, messenger RNA; LPL, lipoprotein lipase; MTP, Microsomal Triglyceride transfer protein; ANGPLT3i, Angiopoietin-Like Protein 3 (ANGPTL3) Inhibitors; PCSK9, proprotein convertase subtilisin/kexin type 9; PCSK9 MAB, proprotein convertase subtilisin/kexin type 9 monoclonal antibody; PCSK9 mRNA, proprotein convertase subtilisin/kexin type 9 messenger RNA; CETPi, cholesteryl ester transfer protein inhibitor; CM, Chylomicrons; VLDL, very low-density lipoprotein; IDL, intermediate-density lipoprotein; LDL, low-density lipoprotein; HDL, high-density lipoprotein; TG, Triglyceride; ApoCIIIi, Apolipoprotein C-III inhibitors; ApoB100 mRNA inhibitor, Apolipoprotein B100 messenger RNA inhibitors; ApoB100, Apolipoprotein B100; LDLR, low-density lipoprotein receptor.
Jcdd 11 00126 g001
Table 1. Pharmacological properties of most commonly used statins.
Table 1. Pharmacological properties of most commonly used statins.
PotencyAbsorptionBioavailabilityElimination Half-Life (h)Metabolism
AtorvastatinHigh intensity at a dose ≥40 mgLipophilic12%14CYP 3A4
RosuvastatinHigh intensity at a dose ≥20 mgHydrophilic20%20CYP2C9 and CYP2C19
PravastatinLow intensityHydrophilic17%1–2OARPB3
FluvastatinLow intensityLipophilic24%1–2CYP2C9
SimvastatinLow intensity at dose ≤20 mgLipophilic<5%1–2CYP 3A4
Table 2. Summary of major lipid-lowering treatment studies.
Table 2. Summary of major lipid-lowering treatment studies.
Study NameTested Treatment Mechanism of Action of the Study DrugNumber of PatientsLDL-C Reduction Primary Outcome
Improve ITsimvastatin–ezetimibe group vs. Simvastatin monotherapy Reduces absorption of cholesterol from the small intestine18,14423–24%absolute risk reduction, 2.0 percentage points; hazard ratio, 0.936; 95% confidence interval, 0.89 to 0.99; p = 0.016
SHARP TRIAL [91]Simvastatin plus ezetimibe vs. Simvastatin and placeboReduces absorption of cholesterol from the small intestine9270 with chronic kidney disease -17% reduction in major atherosclerotic events in the ezetimibe arm
EWTOPIA 75 [92]Ezetimibe vs. usual care for patients above 75 years old Reduces absorption of cholesterol from the small intestine 3796 25.9% vs. 18.5%Ezetimibe reduced the incidence of the primary outcome by 34% (HR 0.66; p = 0.002). Additionally, composite cardiac events were reduced by 60% (HR 0.60; p = 0.039) and coronary revascularisation by 62% (HR 0.38; p = 0.007) in the ezetimibe group vs. the control group.
CLEAR Outcomes study [44]Bempedoic acid group vs. placeboInhibition of adenosine triphosphate citrate lyase 13,970 26% vs. 10%A primary endpoint event (death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization) occurred in (11.7%) of the bempedoic acid group and in (13.3%) of the placebo group (hazard ratio, 0.87; 95% CI, 0.79 to 0.96; p = 0.004)
FOURIER [31]Evolucumab and statin vs. statin and placebo Human monoclonal antibody (PCSK9),
Inhibition of PCSK9 protein results in more LDL receptors available and increased uptake of LDL-C into cells
27,564 59% at the end of 48 weeks The primary composite end point of cardiovascular death, myocardial infarction, stroke, hospitalization for unstable angina, or coronary revascularization. The primary endpoint occurred in (9.8%) in the evolocumab group and (11.3%) in the placebo group (hazard ratio, 0.85; 95% CI, 0.79 to 0.92; p < 0.001)
ODYSSEY OUTCOMES [32]Alirocumab with high-intensity statin vs. high-intensity statin and placeboHuman monoclonal antibody (PCSK9), Inhibition of PCSK9 protein resulting in more LDL receptors available, and increased uptake of LDL-C into cells18,92454.7–62.7% more LDL reduction in the alirocumab group A composite primary endpoint event occurred in (9.5%) in the alirocumab group and in (11.1%) in the placebo group (hazard ratio, 0.85; 95% confidence interval [CI], 0.78 to 0.93; p < 0.001).
ORION-3 [93]InclisiransiRNA therapeutic inclisiran, which reduces hepatic production of (PCSK9), results in sustained reductions in LDL cholesterol 49747.5%LDL cholesterol was reduced by 47.5% (95% CI 50.7–44.3) at day 210 and sustained over 1440 days. The 4-year averaged mean reduction of LDL-C cholesterol was 44.2% (95% CI: 47.1–41.4),
Mendel-2 [94]Both alirocumab and evolocumab have been studied as monotherapy vs. Ezetimibe.Human monoclonal antibody (PCSK9),
Inhibition of PCSK9 protein results in more LDL receptors available and increased uptake of LDL-C into cells
614evolocumab group 57%, ezetimibe 18% compared to placebo. Evolocumab yielded significant LDL-C reductions compared with placebo or Ezetimibe and was well tolerated in patients with hypercholesterolemia.
GLAGOV TRIAL [95]Evolucumab and statin vs. statin and placebo to study the
effect of Evolocumab on Progression of Coronary Disease in Statin-Treated Patients
Human monoclonal antibody (PCSK9),
Inhibition of PCSK9 protein results in more LDL receptors available and increased uptake of LDL-C into cells
968 There was a marked decrease in LDL-C levels in the evolocumab group (Placebo 93 mg/dL vs. evolocumab 37 mg/dL; p < 0.001)Percent atheroma volume (PAV) increased 0.05% with placebo and decreased 0.95% with evolocumab (p  <  0.001), total atheroma volume (TAV) decreased 0.9 mm3 with placebo and 5.8 mm3 with evolocumab (p  <  0.001).
The phase 3 Lp(a) HORIZON [88] Pelacarsen antisense oligonucleotides, small-interfering RNA-based therapies, gene editing, lowering of Lp(a) 8323-The estimated date of study end is May 2025
Pivotal trial [96]Mipomersen Antisense oligonucleotide targeting hepatic apoB100 mRNA 51 patients with Homozygote Familial HypercholesterolemiaMipomersen lowered LDL-C levels by 21% and apolipoprotein B levels by 24% compared to placeboIn addition, non-HDL-C was decreased by 21.6%, triglycerides by 17%, and Lp(a) by 23%, while HDL and apolipoprotein A-I were increased by 11.2% and 3.9% respectively.
REVEAL (Randomised Evaluation of the Effects of Anacetrapib Through Lipid-Modification) [97]AnacetrapibCholesteryl ester transfer protein (CETP) inhibitor30,449HDL-C was increased by 104%, whereas LDL-C and apoB were reduced by 17% and 18%, respectively.-
ANGPTL3 inhibitor phase 3 trial [98]Evinacumabfully human monoclonal antibody inhibits ANGPTL3-49%-
REDUCE-IT [67]Icosapent ethyl highly purified and stable EPA ethyl ester, lower triglyceride levels and is used as an adjunct to diet8179 -The primary endpoint was a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or unstable angina, which occurred in 17.2% of the patients in the icosapent ethyl group, as compared with 22.0% of the patients in the placebo group (hazard ratio, 0.75; 95% confidence interval [CI], 0.68 to 0.83; p < 0.001)
Japan EPA Lipid Intervention Study (JELIS) [99]eicosapentaenoic acid (EPA) of 1.8 gm daily plus statin vs. statin alone Lower the triglyceride level 18,645-The risk of major coronary events was significantly lower, by 19%, in the group that received EPA plus statin therapy than in the group that received statin therapy alone.
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

Omari, M.; Alkhalil, M. Atherosclerosis Residual Lipid Risk-Overview of Existing and Future Pharmacotherapies. J. Cardiovasc. Dev. Dis. 2024, 11, 126. https://doi.org/10.3390/jcdd11040126

AMA Style

Omari M, Alkhalil M. Atherosclerosis Residual Lipid Risk-Overview of Existing and Future Pharmacotherapies. Journal of Cardiovascular Development and Disease. 2024; 11(4):126. https://doi.org/10.3390/jcdd11040126

Chicago/Turabian Style

Omari, Muntaser, and Mohammad Alkhalil. 2024. "Atherosclerosis Residual Lipid Risk-Overview of Existing and Future Pharmacotherapies" Journal of Cardiovascular Development and Disease 11, no. 4: 126. https://doi.org/10.3390/jcdd11040126

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