High-Density Lipoproteins and Cardiovascular Disease - European Medical Journal

High-Density Lipoproteins and Cardiovascular Disease

8 Mins
Cardiology
Download PDF
Cardiology CP
Authors:
*Stephen J. Nicholls, Hannah Brown, Kristen Bubb
Disclosure:

Dr Nicholls has received research support from AstraZeneca, Amgen, Anthera Pharmaceuticals, CSL Behring, Cerenis Therapeutics, Eli Lilly and Company, Esperion Therapeutics, Resverlogix, Novartis, Infraredx, and Sanofi-Regeneron; and is a consultant for Amgen, Akcea Therapeutics, AstraZeneca, Boehringer Ingelheim, CSL Behring, Eli Lilly and Company, Esperion Therapeutics, Kowa, Merck, Takeda, Pfizer, Sanofi-Regeneron, and Novo Nordisk. All other authors have declared no conflicts of interest.

Received:
19.02.20
Accepted:
19.03.20
Citation:
EMJ Cardiol. ; 10.33590/emjcardiol/20-00038. https://doi.org/10.33590/emjcardiol/20-00038.
Keywords:
Atherosclerosis, cardiovascular disease, clinical trials, high-density lipoproteins (HDL), risk factors.

Each article is made available under the terms of the Creative Commons Attribution-Non Commercial 4.0 License.

Abstract

In the search to develop new cardioprotective therapies, considerable interest has focussed on approaches for targeting the biological functions of high-density lipoproteins (HDL). This is based on data from population and animal studies demonstrating a potentially protective impact of HDL on cardiovascular risk. The findings of recent clinical trials of a range of therapeutic interventions aimed at promoting HDL have been disappointing and raise considerable uncertainty regarding the potential utility of this target. More recent evidence has highlighted the importance of HDL functionality, which may ultimately be important in terms of its association with cardiovascular risk. This has led to ongoing efforts to develop new risk markers and therapeutics focussing on HDL quality as opposed to quantity. The evidence supporting a protective role for HDL and findings of clinical trials of HDL-targeted therapies are reviewed here.

INTRODUCTION

For >20 years, clinical trials have consistently demonstrated that the reduction of levels of low-density lipoprotein (LDL) cholesterol results in lower cardiovascular event rates, in both the primary and secondary prevention setting.1 While guidelines for cardiovascular prevention have promoted widespread use of LDL cholesterol-lowering agents for patients at high vascular risk, many continue to experience clinical events.2This residual risk highlights the need to develop additional strategies to achieve a more effective reduction of cardiovascular risk in patients with atherosclerotic cardiovascular disease.

EVIDENCE SUPPORTING A PROTECTIVE ROLE OF HIGH-DENSITY LIPOPROTEINS

Following the early evidence that patients admitted to the coronary care unit with myocardial infarction had lower levels of high-density lipoprotein (HDL) cholesterol, large population studies demonstrated an inverse association between HDL cholesterol levels and prospective cardiovascular risk.3-6 Similar findings were observed in patients achieving very low LDL cholesterol levels, in which low HDL cholesterol continued to be associated with higher rates of cardiovascular events.4 Animal studies demonstrated that promoting HDL functionality, via direct infusion or transgenic expression of its major proteins, had a favourable impact on both the burden and composition of atherosclerotic plaque.7-12 Functional studies revealing that HDL exert favourable effects on inflammatory, oxidative, thrombotic, and apoptotic pathways, in addition to its well-characterised role in reverse cholesterol transport, are likely to underscore its impact on plaque.9,13-16

ESTABLISHED LIPID-MODIFYING STRATEGIES AND HIGH-DENSITY LIPOPROTEINS

Changes in lifestyle factors, including weight loss, accompanied by a reduction in abdominal adiposity and moderate alcohol consumption, have been reported to result in modest increases in HDL cholesterol.17 While statin therapy can raise HDL cholesterol by 3–15%, in clinical trials this has been reported to be independently associated with favourable effects of statins on both plaque progression and cardiovascular events.18,19Fibrates raise HDL cholesterol by 5–20%, showing variable effects on cardiovascular events in multiple clinical trials.20-22 Where a clinical benefit was observed, this was associated with an increase in small HDL particle concentration but not HDL cholesterol concentration overall.23 Niacin is the most effective HDL cholesterol-raising agent currently used in clinical practice. While early studies using immediate-release formulations of niacin reported favourable effects on both angiographic disease and cardiovascular events,24 more recent clinical trials of sustained formulations failed to demonstrate reductions in risk in statin-treated patients.25,26Niacin can prove challenging for patients because many experience the side effect of flushing. Efforts to administer niacin in combination with a prostanoid receptor antagonist reduced the rate of flushing, but did not produce clinical benefit.25,26 Clinical development programmes, accordingly, have sought to develop more effective approaches for targeting HDL (Table 1).

Table 1: High-density lipoprotein-targeted therapies and cardiovascular effects in trials.
Approx.: approximately; CETP: cholesteryl ester transfer protein; HDL: high-density lipoprotein.

HIGH-DENSITY LIPOPROTEIN INFUSIONS

On the basis of favourable effects on atherosclerosis and in-stent restenosis with infusions of reconstituted HDL,7-9,12 interest has focussed on the potential benefits of this approach in humans. Early mechanistic studies in humans demonstrated that HDL infusions increased faecal sterol excretion, a surrogate measure of reverse cholesterol transport, and improved endothelial function.27,28A number of small clinical trials employed serial vascular imaging to evaluate the impact of various formulations of delipidated HDL on atherosclerotic plaque. The first study demonstrated that administration of five weekly intravenous infusions of a HDL mimetic containing recombinant ApoA-I Milano (previously known as ETC-216, now known as MDCO-216) promoted rapid regression of coronary atherosclerosis in patients following an acute coronary syndrome.29 This provided a biological rationale to support observations that humans carrying ApoA-I Milano demonstrated a greater likelihood of longevity and protection from cardiovascular disease.30 It also reaffirmed findings from preclinical studies, demonstrating atheroprotective properties of ApoA-I Milano.31-33 The finding of regression at both 15 and 45 mg/kg doses in the human imaging study suggested a potential saturation effect of this mimetic on lipid transport out of the vessel wall.29

A second clinical development programme of a HDL mimetic, a combination of wild-type ApoA-I and a phospholipid (CSL-111), demonstrated a reduction in plaque lipid and macrophage content when administered 5–7 days prior to femoral endarterectomy.34 A subsequent coronary imaging study in patients following an acute coronary syndrome reported a non-significant trend towards plaque regression and an improvement in plaque echogenicity, suggesting potentially favourable effects on plaque stability.35 The potential for both mimetics, with different forms of ApoA-I, to exert beneficial effects supported the potential of delipidated HDL, as opposed to specific properties related to ApoA-I Milano. This was reaffirmed by the report that selective delipidation of a patient’s HDL, followed by autologous infusion, as similarly associated with plaque regression.36

Synthesis of HDL mimetics, in quantities sufficient for human use, has proven to be a challenge and has required refinement of the manufacturing process to produce purified mimetics with a low potential for toxicity. This has led to a second generation of clinical studies which evaluate the impact of HDL mimetics on a background of more intensive lipid-lowering therapy. The findings to date have been variable. A repeat coronary imaging study of the mimetic containing ApoA-I Milano failed to demonstrate plaque regression.37-39 Another mimetic, which contains recombinant ApoA-I and sphingomyelin (CER-001), has been evaluated in numerous imaging-based trials. While the first study failed to demonstrate a favourable benefit for plaque burden,40 a post hoc analysis revealed regression in those treated with the lowest dose (3 mg/kg) and in patients with the greatest burden of plaque at baseline.41 This observation was further tested; however, 10 weekly infusions failed to be beneficial.42 Similarly, early observations of the potential benefit of CER-001 on carotid plaque volume and inflammatory activity failed to be replicated in prospective, randomised clinical trials.43

Despite the disappointing results of these trials, leading to cessation of the MDCO-216 and CER-001 development programmes, hope remains that other HDL mimetics may produce cardiovascular benefit. Refined manufacturing to produce CSL-112 has progressed to clinical evaluation. A large safety study of this formulation revealed no adverse clinical effects of any concern and a substantial increase in ex vivocholesterol efflux capacity.44 The potential improvement in lipid-transporting activity is much greater than that observed with other mimetics. Given the reported association between cholesterol efflux activity and protection from adverse cardiovascular outcomes, there remains considerable interest in the development of this mimetic. Accordingly, a large cardiovascular outcomes trial is currently in progress to determine whether administration of four intravenous infusions of CSL-112 will reduce cardiovascular event rates in patients following an acute coronary syndrome. This represents the first definitive attempt to determine whether the infusion of some form of HDL will favourably reduce clinical events.

CHOLESTERYL ESTER TRANSFER PROTEIN INHIBITORS

Cholesteryl ester transfer protein (CETP) facilitates the movement of esterified cholesterol from HDL to very-low-density lipoproteins and LDL in exchange for triglycerides.45 CETP inhibition has received considerable attention, given its potential to substantially raise HDL cholesterol levels. Observational studies have reported an association between CETP activity and cardiovascular risk, supported by findings of genomic investigations.45-48 In animal models, therapeutic lowering of CETP activity, using antisense oligonucleotides, vaccines, and small-molecule inhibitors, has been reported to have a favourable impact on plaque burden.49-52 As a result, multiple development programmes have aimed to develop CETP inhibitors, with the potential to target the residual cardiovascular risk observed in many statin-treated patients.

However, experience to date with this class of agents has proved a challenge. The first CETP inhibitor to reach an advanced stage of development, torcetrapib, increased the risk of cardiovascular events and all-cause mortality in a large clinical outcomes trial.53 In parallel, the ability of torcetrapib to raise HDL cholesterol by >60% and lower LDL cholesterol by 20%, in addition to statin therapy, failed to favourably impact the progression of carotid intima-media thickness and coronary atherosclerosis.54-56 Subsequent studies revealed that torcetrapib possessed off-target effects, including adrenal excretion of aldosterone and cortisol, aortic wall expression of endothelin, and small elevations in blood pressure.53,57,58 Because these changes were observed in murine models, which do not express CETP, they are likely to reflect a torcetrapib-specific effect that is not as a result of CETP inhibition. Accordingly, there remains interest in the development of other CETP inhibitors that lack such toxic effects.

A number of CETP inhibitors continued to progress in development on the basis that they lack torcetrapib-like toxicity. Dalcetrapib is a modest CETP inhibitor and raises HDL cholesterol by approximately 25% with no effect on LDL cholesterol levels.59,60 A large clinical outcomes trial was terminated prematurely because dalcetrapib did not reduce cardiovascular event rates.61 Evacetrapib is a potent CETP inhibitor and raises HDL cholesterol by >120% and lowers LDL cholesterol by 25–30% when administered as either monotherapy or in combination with a statin.62 However, these more potent lipid effects did not translate to clinical benefit, with another outcome trial stopped due to futility.63 Anacetrapib is also a potent CETP inhibitor and raises HDL cholesterol by >130% and lowers LDL cholesterol by 30%.64 A clinical trial with more extended treatment demonstrated a significant, albeit modest, reduction in cardiovascular events when anacetrapib was administered in combination with a statin.65 This clinical benefit correlated with the degree of lowering of atherogenic lipoproteins, as opposed to the raising of HDL cholesterol. While this result provided some clinical validation that CETP inhibition may be cardioprotective, anacetrapib accumulates within adipose tissue66 and has not progressed to clinical use.

A number of observations from these CETP inhibitor studies provide insights on potential clinical utility. All of the trials have demonstrated that CETP inhibitor use was associated with a lower rate of diagnosis of Type 2 diabetes mellitus.67,68 In those patients with diabetes, CETP inhibitor use was associated with an improvement in glycaemic control.67,68 Whether this reflects favourable effects of HDL on pancreatic β-cell function,69,70 or an additional effect of CETP inhibition, remains uncertain. It may also have implications for broader use in patients at higher risk, who have evidence of prediabetes or insulin resistance.

Pharmacogenomic analysis of the dalcetrapib trial demonstrated a potential clinical benefit in patients harbouring a polymorphism of the ADCY9 gene on chromosome 16. Patients with the AA genotype demonstrated a 39% reduction in cardiovascular events with dalcetrapib compared with placebo, while patients with the GG phenotype demonstrated an increase in events.71 This finding was supported by the demonstration of a greater increase in cholesterol efflux activity and lesser rise in C-reactive protein levels with dalcetrapib treatment of patients with the AA polymorphism.71 This has led to a new clinical trial of dalcetrapib, performed exclusively in patients identified to have the AA polymorphism.72 A similar relationship was not observed with either evacetrapib or anacetrapib.73,74 If demonstrated to be effective in a prospective trial, it would appear to reflect a dalcetrapib-specific phenomenon.

Additional genomic analyses and Mendelian randomisation have demonstrated the cardioprotective effect of having low CETP activity and levels.46,47,75 This benefit was associated with reductions in levels of atherogenic lipoproteins, rather than raising HDL cholesterol. This benefit also appears greater in the presence, rather than absence, of HMGCR, the target of statins. If correct, this would suggest CETP inhibitors are more likely to be protective in the absence of concomitant statin therapy.75 Accordingly, any further development of CETP inhibitors might focus more on LDL-lowering capability, as opposed to raising of HDL cholesterol. Obicetrapib is the most potent CETP inhibitor developed to date, with more profound effects on LDL and HDL cholesterol, using much lower doses than other agents. How development of this agent will proceed remains to be determined.

ADDITIONAL HIGH-DENSITY LIPOPROTEIN-TARGETED STRATEGIES

A number of other approaches have been investigated, with regard to their potential impact on HDL and cardiovascular risk. Pharmacological agonists of the PPAR family have modest effects on HDL, with variable impact on clinical outcomes. Fibrates are modest PPAR-α agonists, with modest HDL cholesterol raising capabilities and evidence of cardiovascular benefit in some,76,77 but not all,22,78 clinical trials. Meta-analyses of these trials demonstrated a borderline clinical benefit of widespread fibrate use, but more definitive benefit when used in patients with evidence of hypertriglyceridaemia at baseline.20 Subsequent analysis of the gemfibrozil studies suggested that its cardiovascular benefit may be associated with the observed 21% increase in small HDL particles.23 In a similar fashion, thiazolidinediones are PPAR-γ agonists which mildly raise HDL cholesterol in addition to their primary role in improving insulin sensitivity. The effect of these agents on HDL is likely to contribute to their potential clinical benefit, with evidence that reducing the triglyceride/HDL cholesterol ratio is most strongly associated with the ability of these agents to slow progression of coronary atherosclerosis on serial imaging.79 Attempts to develop more potent PPAR agonists,80 or agents targeting multiple PPAR,81 have had difficulty with either toxicity or a lack of clinical benefit. The recent development of selective PPAR modulators has the potential to derive specific metabolic benefits without the toxicity observed with standard PPAR agents,82 although the clinical benefit of this approach has yet to be established.

Apabetalone is a selective inhibitor of bromodomain and extraterminal proteins: epigenetic regulators of lipoprotein metabolism, inflammation, and vascular calcification.83,84 Early studies of apabetalone focussed primarily on its potential impact on HDL functionality, on the basis of reports of stimulated ApoA-I synthesis and increase in cholesterol efflux capacity in non-human primates.85 Early clinical trials of apabetalone in statin-treated patients demonstrated modest effects on HDL cholesterol levels86 and progression of coronary atherosclerosis87 with short-term treatment. Pooled analyses of these trials demonstrated fewer cardiovascular events in apabetalone-treated patients compared with those who received placebo.87 This led to a moderate-sized clinical outcomes trial, which failed to demonstrate a significant reduction in clinical events with apabetalone when administered in patients with diabetes and low HDL cholesterol levels, following an acute coronary syndrome.88

Additional approaches to targeting HDL have focussed on specific factors implicated in reverse cholesterol transport. Upregulation of ABCA1, a pivotal factor facilitating cholesterol efflux to receptors such as HDL particles, may mobilise lipids without necessarily modulating systemic HDL concentrations.89 LCAT promotes esterification of cholesterol on the surface of HDL particles, which is subsequently stored within the particle core, maintaining a low concentration of free cholesterol and an environment favouring ongoing transfer of lipids to HDL. Pharmacological LCAT agonists are currently undergoing early evaluation in human studies.90,91 Advocates of genetic replacement therapy have proposed that the ability to upregulate hepatic ApoA-I expression may have the potential to increase generation of nascent HDL particles.92 While early proponents of such an approach suggested a potential role in patients with genetically low ApoA-I levels, evolving approaches in gene editing may provide a more widespread therapeutic option in the future.

DYSFUNCTIONAL HIGH-DENSITY LIPOPROTEINS

While the early work in HDL therapeutics focussed on raising HDL cholesterol levels, the poor results of clinical trials and lack of association between genetic polymorphisms regulating HDL cholesterol levels and cardiovascular risk93 have suggested this may not prove useful. In parallel, the potential benefit of HDL infusions, in the absence of raising HDL cholesterol levels and in addition to reports of the association of cholesterol efflux activity with cardiovascular risk,94 highlights the likelihood that HDL functionality may be more important. This is supported by reports of cardiovascular events in patients with very high HDL cholesterol levels95 and by reports that the functional properties of HDL may be impaired in the setting of hyperglycaemia, inflammation, and carbamylation.96-101 HDL circulates as a heterogenous population of particles, varying in size and composition of both protein and lipid. It may be that different sub-particles may possess varying functional activity. The small, lipid-deplete particles, as evidenced in infusion mimetics, are potent promoters of cholesterol efflux activity. Therapeutic increases in small HDL particles were reported to be associated with the clinical benefit of gemfibrozil.23 In contrast, generation of large, cholesterol-rich HDL particles with CETP inhibition, though proposed to potentially impair efflux activity, was never demonstrated. Nevertheless, future approaches to HDL therapeutics might consider targeting specific HDL subgroups. Measures of dysfunctional HDL may also play a role in risk prediction.102Furthermore, it is possible that modulating factors that render HDL dysfunctional may provide a novel approach to therapeutic targeting of HDL in patients at a high risk. This remains an active area of research interest for investigators still working to develop HDL-focussed approaches to reducing cardiovascular risk.

CONCLUSION

Considerable work has tried to enhance potentially protective properties of HDL to reduce the residual cardiovascular risk in statin-treated patients. While raising HDL cholesterol has proven to be a disappointing strategy, ongoing efforts have focussed on targeting the functional activities of HDL. Whether any such strategies are ultimately likely to be of clinical benefit will require more clinical trials. To facilitate this, the field will need to embrace the need to pivot the HDL story as a narrative worth pursuing. Only time will tell whether this will alter the course of cardiovascular risk.

References
Baigent C 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;376:1670-81. Libby P. The forgotten majority: unfinished business in cardiovascular risk reduction. J Am Col Cardiol. 2005;46(7):1225-8. Barter P et al.; Treating to New Targets Investigators. HDL cholesterol, very low levels of LDL cholesterol, and cardiovascular events. N Engl J Med. 2007;357(13):1301-10. Gordon DJ et al. High-density lipoprotein cholesterol and cardiovascular disease. Four prospective American studies. Circulation. 1989;79(1):8-15. Gordon DJ, Rifkind BM. High-density lipoprotein-the clinical implications of recent studies. N Engl J Med. 1989;321(19):1311-6. Gordon T et al. High density lipoprotein as a protective factor against coronary heart disease. The Framingham Study. Am J Med. 1977;62(5):707-14. Badimon JJ et al. Regression of atherosclerotic lesions by high density lipoprotein plasma fraction in the cholesterol-fed rabbit. J Clin Invest. 1990;85(4):1234-41. Badimon JJ et al. High density lipoprotein plasma fractions inhibit aortic fatty streaks in cholesterol-fed rabbits. Lab Invest. 1989;60(3):455-61. Nicholls SJ et al. Impact of short-term administration of high-density lipoproteins and atorvastatin on atherosclerosis in rabbits. Arterioscler Thromb Vasc Biol. 2005;25(11):2416-21. Plump AS et al. Human apolipoprotein A-I gene expression increases high density lipoprotein and suppresses atherosclerosis in the apolipoprotein E-deficient mouse. Proc Natl Acad Sci U S A. 1994;91(20):9607-11. Rubin EM et al. Inhibition of early atherogenesis in transgenic mice by human apolipoprotein AI. Nature. 1991;353(6341):265-7. Shah PK et al. High-dose recombinant apolipoprotein A-I(milano) mobilizes tissue cholesterol and rapidly reduces plaque lipid and macrophage content in apolipoprotein e-deficient mice. Potential implications for acute plaque stabilization. Circulation. 2001;103(25):3047-50. Barter PJ et al. Anti-inflammatory properties of HDL. Circ Res. 2004;95(8):764-72. Brewer HB Jr. HDL metabolism and the role of HDL in the treatment of high-risk patients with cardiovascular disease. Curr Cardiol Rep. 2007;9(6):486-92. Nicholls SJ et al. Reconstituted high-density lipoproteins inhibit the acute pro-oxidant and proinflammatory vascular changes induced by a periarterial collar in normocholesterolemic rabbits. Circulation. 2005;111(12):1543-50. Seetharam D et al. High-density lipoprotein promotes endothelial cell migration and reendothelialization via scavenger receptor-B type I. Circ Res. 2006;98(1):63-72. Kraus WE et al. Effects of the amount and intensity of exercise on plasma lipoproteins. N Engl J Med. 2002;347(19):1483-92. Nicholls SJ et al. Statins, high-density lipoprotein cholesterol, and regression of coronary atherosclerosis. JAMA. 2007;297(5):499-508. Cui Y et al. Effects of increasing high-density lipoprotein cholesterol and decreasing low-density lipoprotein cholesterol on the incidence of first acute coronary events (from the Air Force/Texas Coronary Atherosclerosis Prevention Study). Am J Cardiol. 2009;104(6):829-34. Jun M et al. Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet. 2010;375(9729):1875-84. Effect of fenofibrate on progression of coronary-artery disease in Type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet. 2001;357(9260):905-10. Keech A et al.; FIELD study investigators. Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with Type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet. 2005;366(9500):1849-61. Asztalos BF et al. Relation of gemfibrozil treatment and high-density lipoprotein subpopulation profile with cardiovascular events in the Veterans Affairs High-Density Lipoprotein Intervention Trial. Metabolism. 2008;57(1):77-83. AIM-HIGH Investigators; Boden WE et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365(24):2255-67. Bloomfield HE. ACP Journal Club: adding niacin plus laropiprant to statins did not reduce vascular events and increased serious adverse events. Ann Intern Med. 2014;161(10):JC8. HPS2-THRIVE Collaborative Group; Landray MJ et al. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med. 2014;371(3):203-12. Spieker LE et al. High-density lipoprotein restores endothelial function in hypercholesterolemic men. Circulation. 2002;105:1399-402. Tardy C et al. CER-001, a HDL-mimetic, stimulates the reverse lipid transport and atherosclerosis regression in high cholesterol diet-fed LDL-receptor deficient mice. Atherosclerosis. 2014;232(1):110-8. Nissen SE et al. Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. JAMA. 2003;290(17):2292-300. Calabresi L et al. Recombinant apolipoprotein A-IMilano for the treatment of cardiovascular diseases. Curr Atheroscler Rep. 2006;8(2):163-7. Marchesi M et al. Apolipoprotein A-IMilano and 1-palmitoyl-2-oleoyl phosphatidylcholine complex (ETC-216) protects the in vivo rabbit heart from regional ischemia-reperfusion injury. J Pharmacol Exp Ther. 2004;311(3):1023-31. Speidl WS et al. Recombinant apolipoprotein A-I Milano rapidly reverses aortic valve stenosis and decreases leaflet inflammation in an experimental rabbit model. Eur Heart J. 2010;31(16):2049-57. Marchesi M et al. Apolipoprotein A-IMilano/POPC complex attenuates post-ischemic ventricular dysfunction in the isolated rabbit heart. Atherosclerosis. 2008;197(2):572-8. Shaw JA et al. Infusion of reconstituted high-density lipoprotein leads to acute changes in human atherosclerotic plaque. Circ Res. 2008;103(10):1084-91. Tardif JC et al.; Effect of rHDL on Atherosclerosis-Safety and Efficacy (ERASE) Investigators. Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial. JAMA. 2007;297(15):1675-82. Waksman R et al. A first-in-man, randomized, placebo-controlled study to evaluate the safety and feasibility of autologous delipidated high-density lipoprotein plasma infusions in patients with acute coronary syndrome. J Am Coll Cardiol. 2010;55(24):2727-35. Kallend DG et al. A single infusion of MDCO-216 (ApoA-1 Milano/POPC) increases ABCA1-mediated cholesterol efflux and pre-beta 1 HDL in healthy volunteers and patients with stable coronary artery disease. Eur Heart J Cardiovasc Pharmacother. 2016;2(1):23-9. Kempen HJ et al. High-density lipoprotein subfractions and cholesterol efflux capacities after infusion of MDCO-216 (apolipoprotein A-Imilano/palmitoyl-oleoyl-phosphatidylcholine) in healthy volunteers and stable coronary artery disease patients. Arterioscler Thromb Vasc Biol. 2016;36:736-42. Reijers JAA et al. MDCO-216 does not induce adverse immunostimulation, in contrast to its predecessor ETC-216. Cardiovasc Drugs Ther. 2017;31(4):381-9. Tardif JC et al.; Can HDL Infusions Significantly QUicken Atherosclerosis REgression (CHI-SQUARE) Investigators. Effects of the high-density lipoprotein mimetic agent CER-001 on coronary atherosclerosis in patients with acute coronary syndromes: a randomized trial. Eur Heart J. 2014;35(46):3277-86. Kataoka Y et al. Regression of coronary atherosclerosis with infusions of the high-density lipoprotein mimetic CER-001 in patients with more extensive plaque burden. Cardiovasc Diagn Ther. 2017;7(3):252-63. Andrews J et al. Effect of serial infusions of reconstituted high-density lipoprotein (CER-001) on coronary atherosclerosis: rationale and design of the CARAT study. Cardiovasc Diagn Ther. 2017;7(1):45-51. Zheng KH et al. HDL mimetic CER-001 targets atherosclerotic plaques in patients. Atherosclerosis. 2016;251:381-8. Gibson CM 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(24):1918-30. Barter PJ et al. Cholesteryl ester transfer protein. A novel target for raising HDL and inhibiting atherosclerosis. Arterioscler Thromb Vasc Biol. 2003;23(2):160-7. Thompson A et al. Association of cholesteryl ester transfer protein genotypes with CETP mass and activity, lipid levels, and coronary risk. Jama. 2008;299(23):2777-88. Johannsen TH et al. Genetic inhibition of CETP, ischemic vascular disease and mortality, and possible adverse effects. J Am Coll Cardiol. 2012;60(20):2041-8. Brousseau ME et al. Effects of an inhibitor of cholesteryl ester transfer protein on HDL cholesterol. N Engl J Med. 2004;350:1505-15. Rittershaus CW et al. Vaccine-induced antibodies inhibit CETP activity in vivo and reduce aortic lesions in a rabbit model of atherosclerosis. Arterioscler Thromb Vasc Biol. 2000;20(9):2106-12. Sugano M et al. Effect of antisense oligonucleotides against cholesteryl ester transfer protein on the development of atherosclerosis in cholesterol-fed rabbits. J Biol Chem. 1998;273(9):5033-6. Okamoto H et al. A cholesteryl ester transfer protein inhibitor attenuates atherosclerosis in rabbits. Nature. 2000;406(6792):203-7. Morehouse LA et al. Inhibition of CETP activity by torcetrapib reduces susceptibility to diet-induced atherosclerosis in New Zealand White rabbits. J Lipid Res. 2007;48(6):1263-72. Barter PJ et al. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007;357(21):2109-22. Nissen SE et al. Effect of torcetrapib on the progression of coronary atherosclerosis. N Engl J Med. 2007;356(13):1304-16. Bots ML et al. Torcetrapib and carotid intima-media thickness in mixed dyslipidaemia (RADIANCE 2 study): a randomised, double-blind trial. Lancet. 2007;370(9582):153-60. Kastelein JJ et al. Effect of torcetrapib on carotid atherosclerosis in familial hypercholesterolemia. N Engl J Med. 2007;356(16):1620-30. Barter P. Lessons learned from the Investigation of Lipid Level Management to Understand its Impact in Atherosclerotic Events (ILLUMINATE) trial. Am J Cardiol. 2009;104(10 Suppl):10E-5E. Vergeer M, Stroes ES. The pharmacology and off-target effects of some cholesterol ester transfer protein inhibitors. Am J Cardiol. 2009;104(10 Suppl):32E-8E. Luscher TF et al. Vascular effects and safety of dalcetrapib in patients with or at risk of coronary heart disease: the dal-VESSEL randomized clinical trial. Eur Heart J. 2012;33(7):857-65. Fayad ZA 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(9802):1547-59. Schwartz GG et al. Effects of dalcetrapib in patients with a recent acute coronary syndrome. New Engl J Med. 2012;367:2089-99. Nicholls SJ et al. Effects of the CETP inhibitor evacetrapib administered as monotherapy or in combination with statins on HDL and LDL cholesterol: a randomized controlled trial. JAMA. 2011;306(19):2099-109. Lincoff AM et al.; ACCELERATE Investigators. Evacetrapib and cardiovascular outcomes in high-risk vascular disease. N Engl J Med. 2017;376:1933-42. Cannon CP et al. Safety of anacetrapib in patients with or at high risk for coronary heart disease. N Engl J Med. 2010;363:2406-15. The HPS3/TIMI55–REVEAL Collaborative Group. Effects of anacetrapib in patients with atherosclerotic vascular disease. N Engl J Med. 2017;377:1217-27. Gotto AM Jr. et al.; DEFINE Investigators. Lipids, safety parameters, and drug concentrations after an additional 2 years of treatment with anacetrapib in the DEFINE study. J Cardiovasc Pharmacol Ther. 2014;19(6):543-9. Masson W et al. Therapy with cholesteryl ester transfer protein (CETP) inhibitors and diabetes risk. Diabetes Metab. 2018;44(6):508-13. Barter PJ et al. Effect of torcetrapib on glucose, insulin, and hemoglobin A1c in subjects in the Investigation of Lipid Level Management to Understand its Impact in Atherosclerotic Events (ILLUMINATE) trial. Circulation. 2011;124(5):555-62. Barter PJ et al. CETP inhibition, statins and diabetes. Atherosclerosis. 2018;278:143-6. von Eckardstein A, Widmann C. High-density lipoprotein, beta cells, and diabetes. Cardiovasc Res. 2014;103(3):384-94. Tardif JC et al. Pharmacogenomic determinants of the cardiovascular effects of dalcetrapib. Circ Cardiovasc Genet. 2015;8(2):372-82. Tardif JC et al. Study design of Dal-GenE, a pharmacogenetic trial targeting reduction of cardiovascular events with dalcetrapib. Am Heart J. 2020;222:157-65. Hopewell JC et al.; HPS3/TIMI55 - REVEAL Collaborative Group. Impact of ADCY9 genotype on response to anacetrapib. Circulation. 2019;140(11):891-8. Nissen SE et al. ADCY9 genetic variants and cardiovascular outcomes with evacetrapib in patients with high-risk vascular disease: a nested case-control study. JAMA Cardiol. 2018;3(5):401-8. Ference BA et al. Association of genetic variants related to CETP inhibitors and statins with lipoprotein levels and cardiovascular risk. JAMA. 2017;318(10):947-56. Frick MH et al. Helsinki Heart Study: primary-prevention trial with gemfibrozil in middle aged men with dyslipidemia. Safety of treatment, changes in risk factors and incidence of coronary heart disease. N Engl J Med. 1987;317(20):1237-45. Rubins HB et al. Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med. 1999;341(6):410-8. Buse JB et al. Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial: design and methods. Am J Cardiol. 2007;99(12A):21i-33i. Nicholls SJ et al. Lowering of the triglyceride/HDL cholesterol ratio predicts the benefit of pioglitazone on progression of coronary atherosclerosis in diabetic patients. Circulation. 2008;118:S1135. Nissen SE et al. Effects of a potent and selective PPAR-alpha agonist in patients with atherogenic dyslipidemia or hypercholesterolemia: two randomized controlled trials. JAMA. 2007;297(12):1362-73. Lincoff AM et al. Effect of aleglitazar on cardiovascular outcomes after acute coronary syndrome in patients with Type 2 diabetes mellitus: the AleCardio randomized clinical trial. JAMA. 2014;311(15):1515-25. Fruchart JC et al. The selective peroxisome proliferator-activated receptor alpha modulator (SPPARMalpha) paradigm: conceptual framework and therapeutic potential: a consensus statement from the International Atherosclerosis Society (IAS) and the Residual Risk Reduction Initiative (R3i) Foundation. Cardiovasc Diabetol. 2019;18(1):71. Gilham D et al. Apabetalone downregulates factors and pathways associated with vascular calcification. Atherosclerosis. 2019;280:75-84. Gilham D et al. RVX-208, a BET-inhibitor for treating atherosclerotic cardiovascular disease, raises ApoA-I/HDL and represses pathways that contribute to cardiovascular disease. Atherosclerosis. 2016;247:48-57. Bailey D et al. RVX-208: a small molecule that increases apolipoprotein A-I and high-density lipoprotein cholesterol in vitro and in vivo. J Am Coll Cardiol. 2010;55(23):2580-9. Nicholls SJ et al. Efficacy and safety of a novel oral inducer of apolipoprotein A-I synthesis in statin-treated patients with stable coronary artery disease a randomized controlled trial. J Am Coll Cardiol. 2011;57(9):1111-9. Nicholls SJ et al. Selective BET protein inhibition with apabetalone and cardiovascular events: a pooled analysis of trials in patients with coronary artery disease. Am J Cardiovasc Drugs. 2018;18(2):109-15. Ray KK et al. Effect of selective BET protein inhibitor apabetalone on cardiovascular outcomes in patients with acute coronary syndrome and diabetes: rationale, design, and baseline characteristics of the BETonMACE trial. Am Heart J. 2019;217:72-83. Hafiane A et al. Novel Apo E-derived ABCA1 agonist peptide (CS-6253) promotes reverse cholesterol transport and induces formation of prebeta-1 HDL in vitro. PloS one. 2015;10(7):e0131997. Manthei KA et al. Molecular basis for activation of lecithin:cholesterol acyltransferase by a compound that increases HDL cholesterol.Elife. 2018;7. Gunawardane RN et al. Agonistic human antibodies binding to lecithin-cholesterol acyltransferase modulate high density lipoprotein metabolism. J Biol Chem. 2016;291(6):2799-811. Kassim SH et al. Gene therapy for dyslipidemia: a review of gene replacement and gene inhibition strategies. Clin Lipidol.2010;5(6):793-809. Voight BF et al. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet. 2012;380(9841):572-80. Khera AV et al. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N Engl J Med. 2011;364(2):127-35. Ansell BJ et al. Inflammatory/antiinflammatory properties of high-density lipoprotein distinguish patients from control subjects better than high-density lipoprotein cholesterol levels and are favorably affected by simvastatin treatment. Circulation. 2003;108(22):2751-6. Ohgami N et al. Advanced glycation end products (AGE) inhibit scavenger receptor class B Type I-mediated reverse cholesterol transport: a new crossroad of AGE to cholesterol metabolism. J Atheroscler Thromb. 2003;10(1):1-6. Quintao EC et al. Reverse cholesterol transport in diabetes mellitus. Diabetes Metab Res Rev. 2000;16(4):237-50. Alwaili K et al. The HDL proteome in acute coronary syndromes shifts to an inflammatory profile. Biochim Biophys Acta. 2012;1821(3):405-15. Mani P et al. HDL function and subclinical atherosclerosis in juvenile idiopathic arthritis. Cardiovasc Diagn Ther. 2016;6(1):34-43. Ueyama K et al. Cholesterol efflux effect of high density lipoprotein is impaired by whole cigarette smoke extracts through lipid peroxidation. Free Radic Biol Med. 1998;24(1):182-90. Yamamoto S et al. Dysfunctional high-density lipoprotein in patients on chronic hemodialysis. J Am Coll Cardiol. 2012;60(23):2372-9. Bhattacharyya T et al. Relationship of paraoxonase 1 (PON1) gene polymorphisms and functional activity with systemic oxidative stress and cardiovascular risk. JAMA. 2008;299(11):1265-76.

Please rate the quality of this content

As you found this content interesting...

Follow us on social media!

We are sorry that this content was not interesting for you!

Let us improve this content!

Tell us how we can improve this content?

Keep your finger on the pulse

Join Now

Elevating the Quality of Healthcare Globally

>