2,752
Views
14
CrossRef citations to date
0
Altmetric
Review Article

Molecular aspects of hypercholesterolemia treatment: current perspectives and hopes

ORCID Icon, , & ORCID Icon
Pages 303-311 | Received 01 Feb 2018, Accepted 21 Mar 2018, Published online: 10 Apr 2018

Abstract

Hypercholesterolemia is a pathological condition which has been reported in 39% of the worlds’ adult population. We aimed to review molecular aspects of current and novel therapeutic approaches based on low-density lipoprotein cholesterol lowering strategies. Pathogenic mutations in the LDLR, ApoB, PCSK9 and LDLRAP genes cause deficient clearance of circulating low-density lipoprotein cholesterol particles via hepatic LDL receptor. This leads to increased plasma LDL cholesterol levels from birth, which can cause LDL depositions in the arterial walls. Ultimately, it progresses to atherosclerosis and an increased risk of premature cardiovascular diseases. Currently, statins, Ezetimibe, Bile acid sequestrants and PCSK9 inhibitors are the main therapeutic agents for the treatment of hypercholesterolemia. Moreover, novel RNA-based therapy had a strong impact on therapeutic strategies in recent decades. Additional development in understanding of the molecular basis of hypercholesterolemia will provide opportunities for the development of targeted therapy in the near future.

    Key Messages

  • The most common genes involved in hypercholesterolemia are LDLR, PCSK9 and ApoB.

  • Pharmacogenetic effects are typically constrained to pathways closely related to the pharmacodynamics and pharmacokinetics.

  • Change in lifestyle and diet along with treatment of the underlying disease and drug therapy are the current therapeutic strategies.

Introduction

Hypercholesterolemia is a pathological condition which has been reported in 38% of the worlds’ adult population. Raised cholesterol is defined as total cholesterol value ≥ 5.0 mmol/L (190 mg/dL) [Citation1]. Hypercholesterolemia is characterized by elevated low-density lipoprotein-cholesterol (LDL-C) levels and is closely associated with the development of coronary artery disease (CAD) [Citation2]. Hypercholesterolemia is usually caused by a combination of environmental and genetic factors [Citation3]. In developed countries, hypercholesterolemia is often due to both poor dietary habits and lifestyle, and is linked to the so called “metabolic syndrome”, a worldwide epidemic disease mainly associated with the increased onset of health problems and mortality () [Citation4]. Familial hypercholesterolemia (FH) is mostly caused by mutations in genes coding for the LDLR, ApoB, PCSK9 and LDLRAP. Recently, it has been predicted that heterozygous familial hypercholesterolemia (HeFH) is quite a common disease with a prevalence between 1:200 and 1:250 [Citation5,Citation6]. Identification of novel genes through Next Generation Sequencing (NGS) techniques such as whole exome sequencing (WES) might explain the FH phenotypes. This in turn, may expand the basic knowledge about the molecular basis of the disease, that is essential for diagnosis and paves the path to better therapeutic strategies. In a recent study in the UK, among patients with FH in whom a monogenic cause was found, almost 93% have a mutation in LDLR, 5% in ApoB and 2% in PCSK9 (). Although mutations in other genes have been found, none have been independently confirmed [Citation7]. These pathogenic mutations cause deficient clearance of circulating LDL particles via hepatic LDLR leading to increased plasma LDL-C levels from birth and deposition in the arterial wall, thus accelerating atherosclerosis and the risk of premature coronary artery disease or PCAD [Citation8]. While hypercholesterolemia may be caused by specific genetic alterations, most cases of hypercholesterolemia are influenced by other factors, such as endocrine or metabolic diseases, lifestyle (exercise and diet) and drugs [Citation9]. As shown in , statins, ezetimibe, bile acid sequestrants and recent PCSK9 inhibitors are the main therapeutic drugs for the treatment of hypercholesterolemia which act through increased LDLR activity and LDL-C clearance [Citation10]. Furthermore, with the remarkable development of molecular techniques, novel RNA-based therapeutic approaches for therapeutic intervention had a strong impact on therapeutic strategies in recent decades [Citation11]. We aimed to review molecular aspects of current and novel therapeutic approaches based on LDL-C lowering strategies ().

Figure 1. Genetic aspects of different hypercholesterolemia forms and outcomes.

Figure 1. Genetic aspects of different hypercholesterolemia forms and outcomes.

Figure 2. Therapeutic strategies using ASO approach.

Figure 2. Therapeutic strategies using ASO approach.

Table 1. Summary of the most common genes involved in hypercholesterolemia.

Table 2. Summary of current LDL-C lowering therapies (based on FDA and EMA reports).

Bile acid sequestrants (BAS)

Bile acids are the amphipathic agents which are produced as the final products of cholesterol metabolism in the liver. These products have significant roles in the formation of micelles and intestinal lipid absorption. In addition to their classical functions, bile acids act as modulators of a number of intracellular signalling cascades involved in immune response, apoptosis and carcinogenesis [Citation12]. Data suggest that bile acids in the small and large intestine regulate gut microbiota, incretin secretion and production of fibroblast growth factor 15/19 (FGF15/19), which modulate total body lipid, glucose and energy homeostasis [Citation13]. Bile acids are regulators of metabolism acting in an integrated inter-organ manner via farnesoid X receptor (FXR) and Takeda G-protein-coupled receptor 5 (TGR5). It has been demonstrated that FXR has bile salts as ligands that represses bile acid synthesis [Citation14]. Furthermore, besides their role as physiological detergents for intestinal fat, sterol and vitamin absorption, bile acids also activate FXR, inducing multiple signalling pathways to ensure optimal liver metabolism [Citation15]. Modulation of the bile acid pool size and composition, and selective interference with their receptors could therefore be a therapeutic approach to correct hypercholesterolemia. Even though clinical cardiovascular outcome studies using BAS are still lacking, the existing data point to BAS as an efficacious pharmacological approach to reduce cardiovascular risk factors [Citation16]. Resins, including Colestipol, cholestyramine and Colesevelam, were the primary cholesterol-lowering agents before statins were introduced [Citation17]. Molecular evidence has revealed the importance of the enterohepatic bile salt cycle. Undeniably, bile salts activate transcription factors in the nucleus, promoting feed-forward cholesterol catabolic pathways that produce bile salts, as well as the feedback regulation of bile acid synthesis [Citation18].

HMG-CoA inhibitors

HMG-CoA reductase inhibitors or statins are lipid-lowering agents which were established to decrease CAD morbidity and mortality [Citation19]. Statins are analogues of 3-hydroxy-3-methylglutarylCoA (HMG-CoA) reductase substrate. They act not only as a competent with the normal substrate in the enzymes active site but also modify the conformation of the enzyme when they bind to its active site. This process prevents HMG-CoA reductase from reaching a functional structure. Conformation change at the active site makes these drugs very effective and specific. The resultant reduction in hepatocyte cholesterol concentration triggers increased expression of hepatic LDL receptors, which clear LDL and LDL precursors from the circulation [Citation20,Citation21]. Currently available statins include Atorvastatin, Simvastatin, Fluvastatin, Lovastatin, Pitvastatin, Rosuvastatin and Pravastatin. Competitive inhibition of HMG-CoA conversion to mevalonate, as a rate limiting step in de novo cholesterol biosynthesis, is done via binding of statins to HMG-CoA reductase. This causes a temporary and modest reduction in cellular cholesterol concentration [Citation22]. Statins reduce LDL-C by up to 55% and are used as the first-line drugs for the prevention of primary and secondary atherosclerotic cardiovascular disease (ACVD) [Citation23]. Additional to the cholesterol-lowering effect, statins: reduce inflammation via attenuating the expression of several proteins that are involved in the formation of atherosclerotic plaque, induce apoptosis leading to reduced hyperplasia and restenosis, inhibit immune cell proliferation and activation, and improve endothelial function [Citation24].

Despite statins’ benefits, some side-effects such as myopathy have been reported [Citation25]. According previous studies the risk for myopathies is different in various ethnicities and between genders. There is a threefold increased risk of myopathy in women, whereas the risk for men is around six folds [Citation26]. It has been demonstrated that the individual’s genetic architecture has an important role in drug response to statin therapy [Citation27–29]. Variation in several genes, including those encoding cholesterol-7-alpha-hydroxylase (CYP7A1) and ATP-binding cassette transporters G5 (ABCG5) involved in lipid metabolism (pharmacodynamics factors), and ATP-binding cassette protein B1/multidrug resistance protein 1 (ABCB1/MDR1), Cytochrome P450 3A4 (CYP3A4) and solute carrier organic anion transporter family member 1B1 (SLCO1B1) involved in statin metabolism (pharmacokinetic factors), have revealed an association with variability in statin efficacy [Citation29–31].

Intestinal sterol absorption inhibitors

Ezetimibe is an intestinal cholesterol absorption inhibitor which was approved for clinical application (year 2002). It has been available as a single agent or in combination with a statin such as simvastatin and, more recently approved, a fixed-dose of atorvastatin [Citation32]. Ezetimibe prevents intestinal absorption of both biliary and dietary cholesterol via interacting with the Niemann-Pick C1-like 1 (NPC1L1) sterol transporter located on the brush border membrane of proximal jejunum enterocytes [Citation33]. Ezetimibe can reduce plasma LDL-C by 20% and is approved for both primary and secondary prevention of ACVD as a monotherapy in patients intolerant of statins or in combination with statins in high-risk or refractory patients with unsatisfactory statin therapy consequences [Citation34]. However, when the patients were stratified by response, it became clear that some familial hypercholesteraemic patients were at the extremes of response. Exceptional responders, those in the top 10%, had large ≥ 35% reductions and low-responders, those in the bottom 10%, had very small 10% reductions in their LDL-cholesterol [Citation34]. Cholesterol uptake in the intestine is an active process involving the action of transporters [Citation35]. It has been recognized that trans-intestinal cholesterol excretion (TICE) is an important alternative route to the hepatobiliary pathway [Citation36]. A recent study showed that TICE is essential to macrophage reverse cholesterol transport in mice suggesting a potential antiatherogenic role for TICE [Citation37]. Niemann-Pick C 1-like 1 (NPC1L1) gene was identified as related to the Niemann-Pick C1 (NPC1) gene [Citation38]. Both proteins are involved in cholesterol transport and show homology to the patched family of proteins. NPC1L1 was later shown to be responsible for intestinal absorption of dietary cholesterol [Citation39]. Additional research identified NPC1L1 as the protein target for Ezetimibe [Citation40–42] and the extracellular loop C of NPC1L1 as the site of Ezetimibe binding [Citation43]. Sequencing and description of genes in exceptional responders have confirmed valuable to detect genetic variations that change protein function [Citation44]. Whereas understanding and refining the effectiveness of these potential pharmacological agents has yet to be considered for cardiovascular disease, it is evidently likely to become an essential weapon in cardiovascular disease treatment. It has been demonstrated that there is a large inconsistency in individual response to several different statins and doses. This rather unpredictable response is supposedly due to a complex interaction between genetic and environmental factors [Citation45]. Combination of Ezetimibe–statins provides statistically significant improvements in triglycerides (TG), HDL-C, non-HDL-C, apolipoprotein B (ApoB) and high-sensitivity C-reactive protein (hs-CRP) [Citation46]. Data on cardiovascular risk reduction with statin–Ezetimibe combination therapy have been demonstrated in randomized clinical trials (RCTs) in several clinical settings. Though they were all performed using simvastatin, their results serve as a “‘proof-of-concept” for the clinical benefits of other statin–Ezetimibe combinations [Citation32,Citation47,Citation48].

Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors

Proprotein convertase subtilisin/kexin-type 9 (PCSK9) is a serine protease that reduces both hepatic and extrahepatic LDLR levels and increases plasma LDL-C [Citation49]. More than 30 PCSK9 gain of function (GOF) mutations have been reported to have a role in FH [Citation50]. Several studies have shown that mutations in PCSK9 gene decreased its binding to LDLR by greater than 90% [Citation51]. PCSK9 overexpression in the liver results in hypercholesterolemia by reducing LDLR number [Citation52,Citation53]. Gain-of-function mutations of PCSK9 gene leads to increase in LDL receptor degradation causing a reduction in the LDL receptor expression on the cell surface which leads to accumulation of LDL-C in plasma [Citation2]. In contrast, it has been shown that mutations that inactivate PCSK9 (i.e. loss-of-function mutations) lead to lower LDL-C levels and reduced risk of CVD [Citation54,Citation55]. Since the discovery of PCSK9 in 2003, its role in LDLR regulation and plasma LDL-C has produced the most exciting and promising therapeutic strategy in the last three decades [Citation56]. A number of methods which target PCSK9 have been developed. The most successful approach has been the anti-PCSK9 monoclonal antibodies. Three human monoclonal antibodies (Alirocumab, Evolocumab and Bococizumab) have been approved for clinical use before cardiovascular outcomes trials were completed [Citation57] based on their powerful LDL-C lowering properties (60–80%) when used alone or on top of statins [Citation58–60]. Despite the powerful effects of these inhibitors, there was a concern that low levels of LDL-C that result from their use were associated with cognitive deficits. However, in a randomized trial including patients who received either evolocumab or placebo in addition to statin therapy, no significant difference between study groups in cognitive function was observed over a median of 19 months [Citation61].

Inhibition of ANGPTL3 in homozygous familial hypercholesterolemia

Responses to statins and PCSK9 antibodies have limited function due to the dependency on LDL receptor interaction for their activity. Recent drugs such as lomitapide and mipomersen with independent action from LDL receptors for treating HoFH have also recently been approved [Citation62]. Angiopoietin-like 3 (ANGPTL3) is a secreted protein expressed in the liver. It increases plasma levels of TG, LDL-C and HDL-C. ANGPTL3 is an endogenous inhibitor of lipoprotein lipase (LPL) which is related to ANGPTL4. It has been shown that rare loss of function variants in ANGPTL3 gene was associated with decreased triglyceride levels as well as decreased LDL-C and HDL-C levels in family and general population studies [Citation63]. Previous preclinical studies showed that suppression of hepatic Angptl3 protein production in mice resulted in significant reductions in serum levels of TG, LDL-C and non-HDL-C and an increase in insulin sensitivity, which can in turn lead to a reduction in atherosclerosis progression [Citation64]. Genetic and therapeutic antagonists of ANGPTL3 in human and of Angptl3 in mice were associated with decreased levels of all three major lipid fractions and a decreased risk of ACVD [Citation65]. Importantly, recent data demonstrate that ANGPTL3 targeting seems to reduce LDL-C levels independently of the LDL receptor expression and thereby in a recent study where homozygous FH patients were receiving ANGPTL3 inhibitor (Evinacumab) a significant reduction of LDL-C was obtained on top of reductions already achieved with other used lipid-lowering therapy [Citation66]. This data strongly suggests that inhibition of ANGPTL3 function may be a valid approach to reduce residual risk in homozygous FH patients.

Therapeutic strategies using RNA

Small interfering RNAs (siRNAs) and miRNAs have been recently known as important regulators of genes involved in cholesterol homeostasis and potential novel therapeutic targets for hypercholesterolemia [Citation67,Citation68]. MicroRNAs are about 22 nucleotide RNAs that mediate post-transcriptional gene processing. They bind with an argonaute protein to form a silencing complex and act as specific sequence guides which are directing the silencing complex to transcripts within the 3′ untranslated regions (3′ UTRs) of target RNAs [Citation69]. Several recent studies and clinical trials focused on using these small RNAs to target the most causative genes including LDLR [Citation70], ApoB [Citation71,Citation72], PCSK9 [Citation73] and LDLRAP [Citation74,Citation75] as culprit genes in FH. In a recent study, evidence has been presented which supports the potential of miR-27a as a novel therapeutic target for the prevention of atherosclerosis. The inhibition of miR-27a using locked nucleic acids increased LDLR levels by up to 70% [Citation74].

MiR-27 inhibits the expression of two master regulators of adipogenesis PPAR-γ and C/EBP-α [Citation76,Citation77]. A recent study showed the relationship between GLP-1 and miR-27a in lipid metabolism which suggested that treatment with GLP-1 may reduce the expression of miR-27a, with or without cholesterol. Thus, GLP-1 may have a significant role through inhibiting miR-27a [Citation78].

It has been shown that downregulation of sterol regulatory element-binding proteins (SREBPs), as transcription factors of PCSK9, could decrease the amount of PCSK9 transcripts indirectly [Citation79–81]. Interestingly, RNA interference (RNAi) provides alternative treatment options when current drug technology fails. Downregulation of PCSK9 via antisense oligonucleotides (ASO) or small interfering RNA (siRNA) has been developed in the recent years with variable outcomes [Citation82–84]. Advances such as improved lipid nanoparticles (LNPs) and N-acetylgalactosamine (GalNAc) chemistry have targeted liver for clinically approved RNAi therapeutics. Inclisiran (formerly known as ALN-PCS) is a siRNA designed to inhibit the intracellular synthesis of the PCSK9 transcript and consequently decrease plasma PCSK9, and has been reported to be well tolerated and significantly lower PCSK9 and LDL-C after a single intravenous dose in patients at high risk of cardiovascular risk [Citation85–87].

Conclusion

The main reason for dyslipidemia treatment is to decrease the related CVD risk through the therapeutic options such as change in lifestyle and diet, treatment of the underlying disease and drug therapy [Citation88]. Since it has been shown that atherosclerotic process initiates in childhood in which it is mediated by several known risk factors. Children, adolescents and young adults (<20 years) with LDL cholesterol ≥ 160 mg/dL or non-HDL cholesterol ≥ 190 mg/dL should be considered as the first step in hypercholesterolemia screening [Citation89,Citation90]. Treatment should be given at the age of 8 years or older. However, in special cases, such as those with homozygous FH, treatment might need to be started at earlier ages [Citation91,Citation92]. Considering the fact that cholesterol is an essential substance for many biological pathways (e.g. steroid hormones, cell membrane, etc.), treatment at earlier ages should be monitored carefully. Several studies focused on the safety of lipid lowering therapy in FH children ≤ 10 years old and reported no significant concerns about recent drugs [Citation93–95]. Regardless of the reduction in cardiovascular outcomes and mortality through statin therapy, remarkable residual risk remains, especially in severe familial hypercholesterolemia. Recent therapeutic strategies to achieve even lower LDL levels are available at this time (), such as the addition of ezetimibe to inhibit cholesterol absorption, and the recently PCSK9 monoclonal antibodies (Alirolumab and Evolocumab and Bococizumab). Moreover, new LDL-C lowering agents may be effectively administrated in those patients who cannot tolerate statins [Citation96]. More than 20 RNAi-based therapeutics () are now in clinical trials, and a number of these are Phase III trials. Sustained positive results from these trials may support further attempts to develop clinically relevant RNAi therapies. To improve treatment efficacy and safety, pharmacogenetic effects are typically constrained to pathways closely related to the primary effects on the drug target (pharmacodynamics) and aspects of drug absorption, distribution, metabolism and elimination (pharmacokinetics) [Citation97]. Additional development in understanding the molecular basis of hypercholesterolemia will provide opportunities for the development of targeted therapy in the near future.

Figure 3. Summary of current therapeutic targets aimed to reduce serum LDL cholesterol. (1) Bile acid sequestrants, (2) HMG-CoA inhibition, (3) intestinal sterol absorption inhibition, (4) PCSK9 inhibition, (5) miRNA therapy: inhibition of translation of related mRNA.

Figure 3. Summary of current therapeutic targets aimed to reduce serum LDL cholesterol. (1) Bile acid sequestrants, (2) HMG-CoA inhibition, (3) intestinal sterol absorption inhibition, (4) PCSK9 inhibition, (5) miRNA therapy: inhibition of translation of related mRNA.

Disclosure statement

The authors declare that they have no competing interests.

References

  • WHO. Noncommunicable diseases country profiles 2011. WHO Library Cataloguing-in-Publication Data; 2011.
  • Paththinige CS, Sirisena ND, Dissanayake V. Genetic determinants of inherited susceptibility to hypercholesterolemia - a comprehensive literature review. Lipids Health Dis. 2017;16:103.
  • Bhatnagar D, Soran H, Durrington PN. Hypercholesterolaemia and its management. BMJ. 2008;337:a993.
  • Santini A, Novellino E. Nutraceuticals in hypercholesterolaemia: an overview. Br J Pharmacol. 2017;174:1450–1463.
  • Nanchen D, Gencer B, Auer R, et al. Prevalence and management of familial hypercholesterolaemia in patients with acute coronary syndromes. Eur Heart J. 2015;36:2438–2445.
  • Benn M, Watts GF, Tybjaerg-Hansen A, et al. Mutations causative of familial hypercholesterolaemia: screening of 98 098 individuals from the Copenhagen general population study estimated a prevalence of 1 in 217. Eur Heart J. 2016;37:1384–1394.
  • Swerdlow DI, Humphries SE. Genetics of CHD in 2016: common and rare genetic variants and risk of CHD. Nature Rev Cardiol. 2017;14:73–74.
  • Cuchel M, Bruckert E, Ginsberg HN, et al. Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society. Eur Heart J. 2014;35:2146–2157.
  • Ornish D, Scherwitz LW, Billings JH, et al. Intensive lifestyle changes for reversal of coronary heart disease. JAMA. 1998;280:2001–2007.
  • Momtazi AA, Banach M, Pirro M, et al. MicroRNAs: new therapeutic targets for familial hypercholesterolemia? Clin Rev Allergy Immunol. 2018;54:224–233.
  • Gholamin S, Pasdar A, Sadegh Khorrami M, et al. The potential for circulating microRNAs in the diagnosis of myocardial infarction: a novel approach to disease diagnosis and treatment. Curr Pharm Design. 2016;22:397–403.
  • Yamanashi Y, Takada T, Suzuki H. Bile acid as therapeutic agents. In: Tazuma S, Takikawa H, editors. Bile acids in gastroenterology. Tokyo: Springer; 2017. p. 61–90.
  • Li T, Chiang JY. Bile acids as metabolic regulators. Curr Opin Gastroenterol. 2015;31:159.
  • Fiorucci S, Mencarelli A, Palladino G, et al. Bile-acid-activated receptors: targeting TGR5 and farnesoid-X-receptor in lipid and glucose disorders. Trends Pharmacol Sci. 2009;30:570–580.
  • Chiang JY. Bile acids: regulation of synthesis. J Lipid Res. 2009;50:1955–1966.
  • Spinelli V, Chávez-Talavera O, Tailleux A, et al. Metabolic effects of bile acid sequestration: impact on cardiovascular risk factors. Curr Opin Endocrinol Diabetes Obes. 2016;23:138–144.
  • Chang Y, Robidoux J. Dyslipidemia management update. Curr Opin Pharmacol. 2017;33:47–55.
  • Hofmann AF. The enterohepatic circulation of bile acids in mammals: form and functions. Front Biosci. 2009;14:2584–2598.
  • Maron DJ, Fazio S, Linton MF. Current perspectives on statins. Circulation. 2000;101:207–213.
  • Brown MS, Goldstein JL. A receptor-mediated pathway for cholesterol homeostasis. Science. 1986;232:34–47.
  • Sehayek E, Butbul E, Avner R, et al. Enhanced cellular metabolism of very low density lipoprotein by simvastatin. A novel mechanism of action of HMG-CoA reductase inhibitors. Eur J Clin Invest. 1994;24:173–178.
  • Buhaescu I, Izzedine H. Mevalonate pathway: a review of clinical and therapeutical implications. Clin Biochem. 2007;40:575–584.
  • Baigent C, Blackwell L, Emberson J, et al. Efficacy and safety of more intensive lowering of LDL cholesterol: a meta-analysis of data from 170,000 participants in 26 randomised trials. Lancet. 2010;376:1670–1681.
  • Sadowitz B, Maier KG, Gahtan V. Basic science review: statin therapy – Part I: the pleiotropic effects of statins in cardiovascular disease. Vasc Endovascular Surg. 2010;44:241–251.
  • Fadini GP, Manzato E, Crepaldi C, et al. Two cases of statin-induced rhabdomyolysis associated with mononeuropathy. Clin Drug Invest. 2010;30:347–350.
  • Hippisley-Cox J, Coupland C. Individualising the risks of statins in men and women in England and Wales: population-based cohort study. Heart. 2010;96:939–947.
  • Postmus I, Verschuren JJ, De Craen AJ, et al. Pharmacogenetics of statins: achievements, whole-genome analyses and future perspectives. Pharmacogenomics. 2012;13:831–840.
  • Mangravite L, Thorn C, Krauss R. Clinical implications of pharmacogenomics of statin treatment. Pharmacogenomics J. 2006;6:360.
  • Kajinami K, Takekoshi N, Brousseau ME, et al. Pharmacogenetics of HMG-CoA reductase inhibitors: exploring the potential for genotype-based individualization of coronary heart disease management. Atherosclerosis. 2004;177:219–234.
  • Thompson J, Man M, Johnson K, et al. An association study of 43 SNPs in 16 candidate genes with atorvastatin response. Pharmacogenomics J. 2005;5:352.
  • Ruano G, Thompson PD, Windemuth A, et al. Physiogenomic association of statin-related myalgia to serotonin receptors. Muscle Nerve. 2007;36:329–335.
  • Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med. 2015;372:2387–2397.
  • Kotseva K, Wood D, De Bacquer D, et al. EUROASPIRE IV: a European society of cardiology survey on the lifestyle, risk factor and therapeutic management of coronary patients from 24 European countries. Eur J Prev Cardiolog. 2016;23:636–648.
  • Stone NJ, Robinson J, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults. J Am Coll Cardiol. 2014;63:2889–2934.
  • Kruit JK, Groen AK, van BTJ, et al. Emerging roles of the intestine in control of cholesterol metabolism. World J Gastroenterol. 2006;12:6429.
  • Van der Velde AE, Brufau G, Groen AK. Transintestinal cholesterol efflux. Curr Opin Lipidol. 2010;21:167–171.
  • Temel RE, Sawyer JK, Yu L, et al. Biliary sterol secretion is not required for macrophage reverse cholesterol transport. Cell Metab. 2010;12:96–102.
  • Davies JP, Levy B, Ioannou YA. Evidence for a Niemann-pick C (NPC) gene family: identification and characterization of NPC1L1. Genomics. 2000;65:137–145.
  • Altmann SW, Davis HR, Zhu L-J, et al. Niemann-Pick C1 Like 1 protein is critical for intestinal cholesterol absorption. Science. 2004;303:1201–1204.
  • García-García AB, González C, Real JT, et al. Influence of microsomal triglyceride transfer protein promoter polymorphism −493 GT on fasting plasma triglyceride values interaction with treatment response to atorvastatin in subjects with heterozygous familial hypercholesterolaemia. Pharmacogenet Genom. 2005;15:211–218.
  • Hawes BE, O’Neill KA, Yao X, et al. In vivo responsiveness to ezetimibe correlates with niemann-pick C1 like-1 (NPC1L1) binding affinity: comparison of multiple species NPC1L1 orthologs. Mol Pharmacol. 2007;71:19–29.
  • Xie C, Zhou Z-S, Li N, et al. Ezetimibe blocks the internalization of NPC1L1 and cholesterol in mouse small intestine. J Lipid Res. 2012;53:2092.
  • Weinglass AB, Kohler M, Schulte U, et al. Extracellular loop C of NPC1L1 is important for binding to ezetimibe. Proc Nat Acad Sci. 2008;105:11140–11145.
  • Chang DK, Grimmond SM, Evans TJ, et al. Mining the genomes of exceptional responders. Nat Rev Cancer. 2014;14:291.
  • Ferreira AM, da Silva PM. Defining the place of ezetimibe/atorvastatin in the management of hyperlipidemia. Am J Cardiovasc Drugs. 2017;17:169–181.
  • Catapano A, Toth PP, Tomassini JE, et al. The efficacy and safety of ezetimibe coadministered with statin therapy in various patient groups. Clin Lipidol. 2013;8:13–41.
  • Rossebø AB, Pedersen TR, Boman K, et al. Intensive lipid lowering with simvastatin and ezetimibe in aortic stenosis. N Engl J Med. 2008;359:1343–1356.
  • Baigent C, Landray MJ, Reith C, 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.
  • Schmidt RJ, Beyer TP, Bensch WR, et al. Secreted proprotein convertase subtilisin/kexin type 9 reduces both hepatic and extrahepatic low-density lipoprotein receptors in vivo. Biochem Biophys Res Commun. 2008;370:634–640.
  • Varret M, Abifadel M, Rabès JP, et al. Genetic heterogeneity of autosomal dominant hypercholesterolemia. Clin Genet. 2008;73:1–13.
  • Kwon HJ, Lagace TA, McNutt MC, et al. Molecular basis for LDL receptor recognition by PCSK9. Proc Nat Acad Sci. 2008;105:1820–1825.
  • Maxwell KN, Fisher EA, Breslow JL. Overexpression of PCSK9 accelerates the degradation of the LDLR in a post-endoplasmic reticulum compartment. Proc Nat Acad Sci. 2005;102:2069–2074.
  • Park SW, Moon Y-A, Horton JD. Post-transcriptional regulation of low density lipoprotein receptor protein by proprotein convertase subtilisin/kexin type 9a in mouse liver. J Biol Chem. 2004;279:50630–50638.
  • Cohen JC, Boerwinkle E, Mosley TH Jr, et al. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354:1264–1272.
  • Cohen J, Pertsemlidis A, Kotowski IK, et al. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat Genet. 2005;37:161.
  • Stein EA. PCSK9: the critical role of familial hypercholesterolemia from discovery to benefit for all: editorial to: “Efficacy and safety of alirocumab in patients with heterozygous familial hypercholesterolemia and LDL-C of 160 mg/Dl or higher” by Henry N. Ginsberg et al. Cardiovasc Drugs Ther. 2016;30:427–431.
  • Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;2017:1713–1722.
  • White CM. Therapeutic potential and critical analysis of the PCSK9 monoclonal antibodies evolocumab and alirocumab. Ann Pharmacother. 2015;49:1327–1335.
  • Farnier M. An evaluation of alirocumab for the treatment of hypercholesterolemia. Expert Rev Cardiovasc Ther. 2015;13:1307–1323.
  • Langslet G, Emery M, Wasserman SM. Evolocumab (AMG 145) for primary hypercholesterolemia. Expert Rev Cardiovasc Ther. 2015;13:477–488.
  • Giugliano RP, Mach F, Zavitz K, et al. Cognitive function in a randomized trial of evolocumab. N Engl J Med. 2017;377:633–643.
  • Rader DJ, Kastelein JJ. Lomitapide and mipomersen: two first-in-class drugs for reducing low-density lipoprotein cholesterol in patients with homozygous familial hypercholesterolemia. Circulation. 2014;129:1022–1032.
  • Romeo S, Yin W, Kozlitina J, et al. Rare loss-of-function mutations in ANGPTL family members contribute to plasma triglyceride levels in humans. J Clin Invest. 2009;119:70–79.
  • Graham MJ, Lee RG, Brandt TA, et al. Cardiovascular and metabolic effects of ANGPTL3 antisense oligonucleotides. N Engl J Med. 2017;377:222–232.
  • Dewey FE, Gusarova V, Dunbar RL, et al. Genetic and pharmacologic inactivation of ANGPTL3 and cardiovascular disease. N Engl J Med. 2017;377:211–221.
  • Gaudet D, Gipe DA, Pordy R, et al. ANGPTL3 inhibition in homozygous familial hypercholesterolemia. N Engl J Med. 2017;377:296–297.
  • Ray KK, Landmesser U, Leiter LA, et al. siRNA to PCSK9 in patients with high cardiovascular risk and elevated LDL-C: the ORION 1 trial. Atherosclerosis. 2017;263:e9–e10.
  • Dávalos A, Fernández-Hernando C. From evolution to revolution: miRNAs as pharmacological targets for modulating cholesterol efflux and reverse cholesterol transport. Pharmacol Res. 2013;75:60–72.
  • Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233.
  • Agarwal V, Bell GW, Nam J-W, et al. Predicting effective microRNA target sites in mammalian mRNAs. eLife. 2015;4:e05005.
  • Soh J, Hussain MM. Supplementary site interactions are critical for the regulation of microsomal triglyceride transfer protein by microRNA-30c. Nutr Metab. 2013;10:56.
  • Hsu S-H, Wang B, Kota J, et al. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest. 2012;122:2871.
  • Zhang X-L, Zhu Q-Q, Zhu L, et al. Safety and efficacy of anti-PCSK9 antibodies: a meta-analysis of 25 randomized, controlled trials. BMC Med. 2015;13:123.
  • Alvarez ML, Khosroheidari M, Eddy E, et al. MicroRNA-27a decreases the level and efficiency of the LDL receptor and contributes to the dysregulation of cholesterol homeostasis. Atherosclerosis. 2015;242:595–604.
  • Goedeke L, Rotllan N, Ramírez CM, et al. miR-27b inhibits LDLR and ABCA1 expression but does not influence plasma and hepatic lipid levels in mice. Atherosclerosis. 2015;243:499–509.
  • Sun L, Trajkovski M. MiR-27 orchestrates the transcriptional regulation of brown adipogenesis. Metab Clin Exp. 2014;63:272–282.
  • Lin Q, Gao Z, Alarcon RM, et al. A role of miR-27 in the regulation of adipogenesis. FEBS J. 2009;276:2348–2358.
  • Noyan-Ashraf MH, Shikatani EA, Schuiki I, et al. A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation. 2013;127:74–85.
  • Li Y, Zhang J, He J, et al. MicroRNA-132 cause apoptosis of glioma cells through blockade of the SREBP-1c metabolic pathway related to SIRT1. Biomed Pharmacother. 2016;78:177–184.
  • Zhang H, Feng Z, Huang R, et al. MicroRNA-449 suppresses proliferation of hepatoma cell lines through blockade lipid metabolic pathway related to SIRT1. Int J Oncol. 2014;45:2143–2152.
  • Li X, Chen Y-T, Josson S, et al. MicroRNA-185 and 342 inhibit tumorigenicity and induce apoptosis through blockade of the SREBP metabolic pathway in prostate cancer cells. PloS One. 2013;8:e70987.
  • Gupta N, Fisker N, Asselin MC, et al. A locked nucleic acid antisense oligonucleotide (LNA) silences PCSK9 and enhances LDLR expression in vitro and in vivo. PloS One. 2010;5:e10682.
  • Yamamoto T, Harada-Shiba M, Nakatani M, et al. Cholesterol-lowering action of BNA-based antisense oligonucleotides targeting PCSK9 in atherogenic diet-induced hypercholesterolemic mice. Mol Ther Nucleic Acids. 2012;1:e22.
  • Norata GD, Tibolla G, Catapano AL. Targeting PCSK9 for hypercholesterolemia. Annu Rev Pharmacol Toxicol. 2014;54:273–293.
  • Fitzgerald K, Frank-Kamenetsky M, Shulga-Morskaya S, et al. Effect of an RNA interference drug on the synthesis of proprotein convertase subtilisin/kexin type 9 (PCSK9) and the concentration of serum LDL cholesterol in healthy volunteers: a randomised, single-blind, placebo-controlled, phase 1 trial. Lancet. 2014;383:60–68.
  • Bobbin ML, Rossi JJ. RNA interference (RNAi)-based therapeutics: delivering on the promise? Annu Rev Pharmacol Toxicol. 2016;56:103–122.
  • Ray KK, Landmesser U, Leiter LA, et al. Inclisiran in patients at high cardiovascular risk with elevated LDL cholesterol. N Engl J Med. 2017;376:1430–1440.
  • Iughetti L, Bruzzi P, Predieri B. Evaluation and management of hyperlipidemia in children and adolescents. Curr Opin Pediatr. 2010;22:485–493.
  • McMahan CA, Gidding SS, Fayad ZA, et al. Risk scores predict atherosclerotic lesions in young people. Arch Intern Med. 2005;165:883–890.
  • Hopkins PN, Toth PP, Ballantyne CM, et al. Familial hypercholesterolemias: prevalence, genetics, diagnosis and screening recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol. 2011;5:S9–S17.
  • Goldberg AC, Hopkins PN, Toth PP, et al. Familial hypercholesterolemia: screening, diagnosis and management of pediatric and adult patients: clinical guidance from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol. 2011;5:133–140.
  • Lughetti L, Predieri B, Balli F, et al. Rational approach to the treatment for heterozygous familial hypercholesterolemia in childhood and adolescence: a review. J Endocrinol Invest. 2007;30:700–719.
  • Bellunghi MS, Miname M, Jannes CE, et al. Familial hypercholesterolemia in children and safety of early lipid-lowering treatment. Circulation. 2017;136:A14877.
  • Saltijeral A, de Isla LP, Alonso R, et al. Attainment of LDL cholesterol treatment goals in children and adolescents with familial hypercholesterolemia. The SAFEHEART Follow-up Registry. Rev Esp Cardiol (English Edition). 2017;70:444–450.
  • Civeira F, Plana N. Treatment of heterozygous familial hypercholesterolemia in children and adolescents: an unsolved problem. Rev Esp Cardiol. 2017;70:423–424.
  • Paragh G, Karádi I. Up to date lipid lowering treatment. Orv Hetil. 2016;157:1219–1223.
  • Paynter NP, Ridker PM, Chasman DI. Are genetic tests for atherosclerosis ready for routine clinical use? Circ Res. 2016;118:607–619.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.