713
Views
18
CrossRef citations to date
0
Altmetric
Review

Recent developments and future directions for the use of pharmacogenomics in cardiovascular disease treatments

&
Pages 973-983 | Received 17 May 2017, Accepted 01 Aug 2017, Published online: 20 Aug 2017

References

  • Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366:1267–1278.
  • Jacobson TA, Ito MK, Maki KC, et al. National lipid association recommendations for patient-centered management of dyslipidemia: part 1–full report. J Clin Lipidol. 2015;9:129–169.
  • Jacobson TA, Maki KC, Orringer CE, et al. National lipid association recommendations for patient-centered management of dyslipidemia: part 2. J Clin Lipidol. 2015;9:S1–122 e1.
  • Stone NJ, Robinson JG, Lichtenstein AH, et al. American College of Cardiology/American Heart Association Task Force on Practice G. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation. 2014;129:S1–45.
  • Stroes ES, Thompson PD, Corsini A, et al. Statin-associated muscle symptoms: impact on statin therapy-European Atherosclerosis Society Consensus Panel Statement on Assessment, Aetiology and Management. Eur Heart J. 2015;36:1012–1022.
  • Bruckert E, Hayem G, Dejager S, et al. Mild to moderate muscular symptoms with high-dosage statin therapy in hyperlipidemic patients–the PRIMO study. Cardiovasc Drugs Ther. 2005;19:403–414.
  • Law M, Rudnicka AR. Statin safety: a systematic review. Am J Cardiol. 2006;97:52C–60C.
  • Simpson RJ Jr., Mendys P. The effects of adherence and persistence on clinical outcomes in patients treated with statins: a systematic review. J Clin Lipidol. 2010;4:462–471.
  • de Denus S, Spinler SA, Miller K, et al. Statins and liver toxicity: a meta-analysis. Pharmacotherapy. 2004;24:584–591.
  • Suraweera C, De Silva V, Hanwella R. Simvastatin-induced cognitive dysfunction: two case reports. J Med Case Rep. 2016;10:83.
  • Link E, Parish S, Armitage J, et al. SLCO1B1 variants and statin-induced myopathy–a genomewide study. N Engl J Med. 2008;359:789–799.
  • Kadam P, Ashavaid TF, Ponde CK, et al. Genetic determinants of lipid-lowering response to atorvastatin therapy in an Indian population. J Clin Pharm Ther. 2016;41:329–333.
  • Chasman DI, Giulianini F, MacFadyen J, et al. Genetic determinants of statin-induced low-density lipoprotein cholesterol reduction: the Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) trial. Circ Cardiovasc Genet. 2012;5:257–264.
  • Donnelly LA, Doney AS, Tavendale R, et al. Common nonsynonymous substitutions in SLCO1B1 predispose to statin intolerance in routinely treated individuals with type 2 diabetes: a go-DARTS study. Clin Pharmacol Ther. 2011;89:210–216.
  • Jiang J, Tang Q, Feng J, et al. Association between SLCO1B1-521T>C and −388A>G polymorphisms and risk of statin-induced adverse drug reactions: a meta-analysis. Springerplus. 2016;5:1368.
  • Prado Y, Saavedra N, Zambrano T, et al. SLCO1B1 c.388A>G polymorphism is associated with HDL-C levels in response to atorvastatin in Chilean individuals. Int J Mol Sci. 2015;16:20609–20619.
  • Hoenig MR, Walker PJ, Gurnsey C, et al. The C3435T polymorphism in ABCB1 influences atorvastatin efficacy and muscle symptoms in a high-risk vascular cohort. J Clin Lipidol. 2011;5:91–96.
  • Ferrari M, Guasti L, Maresca A, et al. Association between statin-induced creatine kinase elevation and genetic polymorphisms in SLCO1B1, ABCB1 and ABCG2. Eur J Clin Pharmacol. 2014;70:539–547.
  • Wang D, Guo Y, Wrighton SA, et al. Intronic polymorphism in CYP3A4 affects hepatic expression and response to statin drugs. Pharmacogenomics J. 2011;11:274–286.
  • Elens L, Becker ML, Haufroid V, et al. Novel CYP3A4 intron 6 single nucleotide polymorphism is associated with simvastatin-mediated cholesterol reduction in the Rotterdam Study. Pharmacogenet Genomics. 2011;21:861–866.
  • Westlind-Johnsson A, Malmebo S, Johansson A, et al. Comparative analysis of CYP3A expression in human liver suggests only a minor role for CYP3A5 in drug metabolism. Drug Metab Dispos. 2003;31:755–761.
  • Kim KA, Park PW, Lee OJ, et al. Effect of polymorphic CYP3A5 genotype on the single-dose simvastatin pharmacokinetics in healthy subjects. J Clin Pharmacol. 2007;47:87–93.
  • Kivistö KT, Niemi M, Schaeffeler E, et al. Lipid-lowering response to statins is affected by CYP3A5 polymorphism. Pharmacogenetics. 2004;14:523–525.
  • Hirano M, Maeda K, Shitara Y, et al. Contribution of OATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans. J Pharmacol Exp Ther. 2004;311:139–146.
  • Pasanen MK, Neuvonen M, Neuvonen PJ, et al. SLCO1B1 polymorphism markedly affects the pharmacokinetics of simvastatin acid. Pharmacogenet Genomics. 2006;16:873–879.
  • Kitzmiller JP, Mikulik EB, Dauki AM, et al. Pharmacogenomics of statins: understanding susceptibility to adverse effects. Pharmgenomics Pers Med. 2016;9:97–106.
  • Keskitalo JE, Kurkinen KJ, Neuvoneni PJ, et al. ABCB1 haplotypes differentially affect the pharmacokinetics of the acid and lactone forms of simvastatin and atorvastatin. Clin Pharmacol Ther. 2008;84:457–461.
  • Hermann M, Bogsrud MP, Molden E, et al. Exposure of atorvastatin is unchanged but lactone and acid metabolites are increased several-fold in patients with atorvastatin-induced myopathy. Clin Pharmacol Ther. 2006;79:532–539.
  • Danielson PB. The cytochrome P450 superfamily: biochemistry, evolution and drug metabolism in humans. Curr Drug Metab. 2002;3:561–597.
  • Williams D, Feely J. Pharmacokinetic-pharmacodynamic drug interactions with HMG-CoA reductase inhibitors. Clin Pharmacokinet. 2002;41:343–370.
  • Wang D, Sadee W. CYP3A4 intronic SNP rs35599367 (CYP3A4*22) alters RNA splicing. Pharmacogenet Genomics. 2016;26:40–43.
  • Tsamandouras N, Dickinson G, Guo Y, et al. Identification of the effect of multiple polymorphisms on the pharmacokinetics of simvastatin and simvastatin acid using a population-modeling approach. Clin Pharmacol Ther. 2014;96:90–100.
  • Ragia G, Kolovou V, Tavridou A, et al. No effect of CYP3A4 intron 6 C>T polymorphism (CYP3A4*22) on lipid-lowering response to statins in Greek patients with primary hypercholesterolemia. Drug Metab Pers Ther. 2015;30:43–48.
  • Hu M, Mak VW, Xiao Y, et al. Associations between the genotypes and phenotype of CYP3A and the lipid response to simvastatin in Chinese patients with hypercholesterolemia. Pharmacogenomics. 2013;14:25–34.
  • Park JE, Kim KB, Bae SK, et al. Contribution of cytochrome P450 3A4 and 3A5 to the metabolism of atorvastatin. Xenobiotica. 2008;38:1240–1251.
  • Angiolillo DJ. Variability in responsiveness to oral antiplatelet therapy. Am J Cardiol. 2009;103:27A–34A.
  • Mega JL, Hochholzer W, Frelinger AL 3rd, et al. Dosing clopidogrel based on CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovascular disease. JAMA. 2011;306:2221–2228.
  • Geisler T, Schaeffeler E, Dippon J, et al. CYP2C19 and nongenetic factors predict poor responsiveness to clopidogrel loading dose after coronary stent implantation. Pharmacogenomics. 2008;9:1251–1259.
  • Collet JP, Hulot JS, Pena A, et al. Cytochrome P450 2C19 polymorphism in young patients treated with clopidogrel after myocardial infarction: a cohort study. Lancet. 2009;373:309–317.
  • Sim SC, Risinger C, Dahl ML, et al. A common novel CYP2C19 gene variant causes ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and antidepressants. Clin Pharmacol Ther. 2006;79:103–113.
  • Sibbing D, Koch W, Gebhard D, et al. Cytochrome 2C19*17 allelic variant, platelet aggregation, bleeding events, and stent thrombosis in clopidogrel-treated patients with coronary stent placement. Circulation. 2010;121:512–518.
  • Maree AO, Curtin RJ, Chubb A, et al. Cyclooxygenase-1 haplotype modulates platelet response to aspirin. J Thromb Haemost. 2005;3:2340–2345.
  • Goodman T, Ferro A, Sharma P. Pharmacogenetics of aspirin resistance: a comprehensive systematic review. Br J Clin Pharmacol. 2008;66:222–232.
  • Li XL, Cao J, Fan L, et al. Genetic polymorphisms of HO-1 and COX-1 are associated with aspirin resistance defined by light transmittance aggregation in Chinese Han patients. Clin Appl Thromb Hemost. 2013;19:513–521.
  • Cao L, Zhang Z, Sun W, et al. Impacts of COX-1 gene polymorphisms on vascular outcomes in patients with ischemic stroke and treated with aspirin. Gene. 2014;546:172–176.
  • Colaizzo D, Fofi L, Tiscia G, et al. The COX-2 G/C −765 polymorphism may modulate the occurrence of cerebrovascular ischemia. Blood Coagul Fibrinolysis. 2006;17:93–96.
  • Orbe J, Beloqui O, Rodriguez JA, et al. Protective effect of the G-765C COX-2 polymorphism on subclinical atherosclerosis and inflammatory markers in asymptomatic subjects with cardiovascular risk factors. Clin Chim Acta. 2006;368:138–143.
  • Cipollone F, Toniato E, Martinotti S, et al. A polymorphism in the cyclooxygenase 2 gene as an inherited protective factor against myocardial infarction and stroke. JAMA. 2004;291:2221–2228.
  • Rudock ME, Liu Y, Ziegler JT, et al. Association of polymorphisms in cyclooxygenase (COX)-2 with coronary and carotid calcium in the Diabetes Heart Study. Atherosclerosis. 2009;203:459–465.
  • Lee CR, North KE, Bray MS, et al. Cyclooxygenase polymorphisms and risk of cardiovascular events: the Atherosclerosis Risk in Communities (ARIC) study. Clin Pharmacol Ther. 2008;83:52–60.
  • Packer M, Bristow MR, Cohn JN, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. U.S. Carvedilol Heart Failure Study Group. N Engl J Med. 1996;334:1349–1355.
  • Zaiou M, El Amri H. Cardiovascular pharmacogenetics: a promise for genomically-guided therapy and personalized medicine. Clin Genet. 2017;91:355–370.
  • Johnson JA, Zineh I, Puckett BJ, et al. Beta 1-adrenergic receptor polymorphisms and antihypertensive response to metoprolol. Clin Pharmacol Ther. 2003;74:44–52.
  • Liggett SB, Mialet-Perez J, Thaneemit-Chen S, et al. A polymorphism within a conserved beta(1)-adrenergic receptor motif alters cardiac function and beta-blocker response in human heart failure. Proc Natl Acad Sci U S A. 2006;103:11288–11293.
  • Magvanjav O, McDonough CW, Gong Y, et al. Pharmacogenetic associations of beta1-adrenergic receptor polymorphisms with cardiovascular outcomes in the SPS3 trial (Secondary Prevention of Small Subcortical Strokes). Stroke. 2017;48:1337–1343.
  • Johnson JA, Cavallari LH. Warfarin pharmacogenetics. Trends Cardiovasc Med. 2015;25:33–41.
  • Rau T, Wuttke H, Michels LM, et al. Impact of the CYP2D6 genotype on the clinical effects of metoprolol: a prospective longitudinal study. Clin Pharmacol Ther. 2009;85:269–272.
  • Dean L. Metoprolol therapy and CYP2D6 genotype. In: Pratt V, McLeod H, Dean L, et al., editors. Medical genetics summaries. NCBI: 2012.
  • Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med. 2009;360:363–375.
  • Shuldiner AR, O’Connell JR, Bliden KP, et al. Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA. 2009;302:849–857.
  • Taubert D, Von Beckerath N, Grimberg G, et al. Impact of P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther. 2006;80:486–501.
  • Nakamura T, Sakaeda T, Horinouchi M, et al. Effect of the mutation (C3435T) at exon 26 of the MDR1 gene on expression level of MDR1 messenger ribonucleic acid in duodenal enterocytes of healthy Japanese subjects. Clin Pharmacol Ther. 2002;71:297–303.
  • Woodahl EL, Ho RJ. The role of MDR1 genetic polymorphisms in interindividual variability in P-glycoprotein expression and function. Curr Drug Metab. 2004;5:11–19.
  • Mega JL, Close SL, Wiviott SD, et al. Genetic variants in ABCB1 and CYP2C19 and cardiovascular outcomes after treatment with clopidogrel and prasugrel in the TRITON-TIMI 38 trial: a pharmacogenetic analysis. Lancet. 2010;376:1312–1319.
  • Zhai Y, He H, Ma X, et al. Meta-analysis of effects of ABCB1 polymorphisms on clopidogrel response among patients with coronary artery disease. Eur J Clin Pharmacol. 2017;73:843–854.
  • Tarkiainen EK, Tornio A, Holmberg MT, et al. Effect of carboxylesterase 1 c.428G > A single nucleotide variation on the pharmacokinetics of quinapril and enalapril. Br J Clin Pharmacol. 2015;80:1131–1138.
  • Lewis JP, Horenstein RB, Ryan K, et al. The functional G143E variant of carboxylesterase 1 is associated with increased clopidogrel active metabolite levels and greater clopidogrel response. Pharmacogenet Genomics. 2013;23:1–8.
  • Zhao Z, Li X, Sun S, et al. Impact of genetic polymorphisms related to clopidogrel or acetylsalicylic acid pharmacology on clinical outcome in Chinese patients with symptomatic extracranial or intracranial stenosis. Eur J Clin Pharmacol. 2016;72:1195–1204.
  • Zhu HJ, Patrick KS, Yuan HJ, et al. Two CES1 gene mutations lead to dysfunctional carboxylesterase 1 activity in man: clinical significance and molecular basis. Am J Hum Genet. 2008;82:1241–1248.
  • Farid NA, Kurihara A, Wrighton SA. Metabolism and disposition of the thienopyridine antiplatelet drugs ticlopidine, clopidogrel, and prasugrel in humans. J Clin Pharmacol. 2010;50:126–142.
  • Bouman HJ, Schömig E, Van Werkum JW, et al. Paraoxonase-1 is a major determinant of clopidogrel efficacy. Nat Med. 2011;17:110–116.
  • Trenk D, Hochholzer W, Fromm MF, et al. Paraoxonase-1 Q192R polymorphism and antiplatelet effects of clopidogrel in patients undergoing elective coronary stent placement. Circ Cardiovasc Genet. 2011;4:429–436.
  • Sibbing D, Koch W, Massberg S, et al. No association of paraoxonase-1 Q192R genotypes with platelet response to clopidogrel and risk of stent thrombosis after coronary stenting. Eur Heart J. 2011;32:1605–1613.
  • Antithrombotic Trialists C. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324:71–86.
  • Hankey GJ, Eikelboom JW. Aspirin resistance. Lancet. 2006;367:606–617.
  • Kauskot A, Di Michele M, Loyen S, et al. A novel mechanism of sustained platelet alphaIIbbeta3 activation via PEAR1. Blood. 2012;119:4056–4065.
  • Herrera-Galeano JE, Becker DM, Wilson AF, et al. A novel variant in the platelet endothelial aggregation receptor-1 gene is associated with increased platelet aggregability. Arterioscler Thromb Vasc Biol. 2008;28:1484–1490.
  • Lewis JP, Ryan K, O’Connell JR, et al. Genetic variation in PEAR1 is associated with platelet aggregation and cardiovascular outcomes. Circ Cardiovasc Genet. 2013;6:184–192.
  • Backman JD, Yerges-Armstrong LM, Horenstein RB, et al. Prospective evaluation of genetic variation in platelet endothelial aggregation receptor 1 reveals aspirin-dependent effects on platelet aggregation pathways. Clin Transl Sci. 2017;10:102–109.
  • Faraday N, Yanek LR, Yang XP, et al. Identification of a specific intronic PEAR1 gene variant associated with greater platelet aggregability and protein expression. Blood. 2011;118:3367–3375.
  • Floyd CN, Ferro A. The platelet fibrinogen receptor: from megakaryocyte to the mortuary. JRSM Cardiovasc Dis. 2012;1:1-13.
  • Floyd CN, Ferro A. The PlA1/A2 polymorphism of glycoprotein IIIa in relation to efficacy of antiplatelet drugs: a systematic review and meta-analysis. Br J Clin Pharmacol. 2014;77:446–457.
  • Li Q, Chen BL, Ozdemir V, et al. Frequency of genetic polymorphisms of COX1, GPIIIa and P2Y1 in a Chinese population and association with attenuated response to aspirin. Pharmacogenomics. 2007;8:577–586.
  • Lev EI, Patel RT, Guthikonda S, et al. Genetic polymorphisms of the platelet receptors P2Y(12), P2Y(1) and GP IIIa and response to aspirin and clopidogrel. Thromb Res. 2007;119:355–360.
  • Yi X, Zhou Q, Wang C, et al. Platelet receptor Gene (P2Y12, P2Y1) and platelet glycoprotein Gene (GPIIIa) polymorphisms are associated with antiplatelet drug responsiveness and clinical outcomes after acute minor ischemic stroke. Eur J Clin Pharmacol. 2017;73:437–443.
  • Wang H, Liu J, Liu K, et al. beta1-adrenoceptor gene Arg389Gly polymorphism and essential hypertension risk in general population: a meta-analysis. Mol Biol Rep. 2013;40:4055–4063.
  • Wain LV, Verwoert GC, O’Reilly PF, et al. Genome-wide association study identifies six new loci influencing pulse pressure and mean arterial pressure. Nat Genet. 2011;43:1005–1011.
  • Ganesh SK, Tragante V, Guo W, et al. Loci influencing blood pressure identified using a cardiovascular gene-centric array. Hum Mol Genet. 2013;22:1663–1678.
  • Liu J, Liu ZQ, Yu BN, et al. Beta1-adrenergic receptor polymorphisms influence the response to metoprolol monotherapy in patients with essential hypertension. Clin Pharmacol Ther. 2006;80:23–32.
  • Wu D, Li G, Deng M, et al. Associations between ADRB1 and CYP2D6 gene polymorphisms and the response to beta-blocker therapy in hypertension. J Int Med Res. 2015;43:424–434.
  • Aleong RG, Sauer WH, Sauer WH, et al. Prevention of atrial fibrillation by bucindolol is dependent on the beta(1)389 Arg/Gly adrenergic receptor polymorphism. JACC Heart Fail. 2013;1:338–344.
  • Aleong RG, Sauer WH, Robertson AD, et al. Adrenergic receptor polymorphisms and prevention of ventricular arrhythmias with bucindolol in patients with chronic heart failure. Circ Arrhythm Electrophysiol. 2013;6:137–143.
  • O’Shaughnessy KM, Fu B, Dickerson C, et al. The gain-of-function G389R variant of the beta1-adrenoceptor does not influence blood pressure or heart rate response to beta-blockade in hypertensive subjects. Clin Sci (Lond). 2000;99:233–238.
  • Karlsson J, Lind L, Hallberg P, et al. Beta1-adrenergic receptor gene polymorphisms and response to beta1-adrenergic receptor blockade in patients with essential hypertension. Clin Cardiol. 2004;27:347–350.
  • Nagele P, Liggett SB. Genetic variation, beta-blockers, and perioperative myocardial infarction. Anesthesiology. 2011;115:1316–1327.
  • Gardemann A, Humme J, Stricker J, et al. Association of the platelet glycoprotein IIIa PlA1/A2 gene polymorphism to coronary artery disease but not to nonfatal myocardial infarction in low risk patients. Thromb Haemost. 1998;80:214–217.
  • Ham AC, Van Dijk SC, Swart KMA, et al. Beta-blocker use and fall risk in older individuals: original results from two studies with meta-analysis. Br J Clin Pharmacol. 2017.
  • Blake CM, Kharasch ED, Schwab M, et al. A meta-analysis of CYP2D6 metabolizer phenotype and metoprolol pharmacokinetics. Clin Pharmacol Ther. 2013;94:394–399.
  • Owen RP, Altman RB, Klein TE. PharmGKB and the International Warfarin Pharmacogenetics Consortium: the changing role for pharmacogenomic databases and single-drug pharmacogenetics. Hum Mutat. 2008;29:456–460.
  • Dean L. Warfarin therapy and the genotypes CYP2C9 and VKORC1. In: Pratt V, McLeod H, Dean L, et al., editors. Medical genetics summaries. NCBI: 2012.
  • Leemann T, Transon C, Dayer P. Cytochrome P450TB (CYP2C): a major monooxygenase catalyzing diclofenac 4ʹ-hydroxylation in human liver. Life Sci. 1993;52:29–34.
  • Takeuchi F, McGinnis R, Bourgeois S, et al. A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. PLoS Genet. 2009;5:e1000433.
  • Cavallari LH, Langaee TY, Momary KM, et al. Genetic and clinical predictors of warfarin dose requirements in African Americans. Clin Pharmacol Ther. 2010;87:459–464.
  • Pirmohamed M, Burnside G, Eriksson N, et al. A randomized trial of genotype-guided dosing of warfarin. N Engl J Med. 2013;369:2294–2303.
  • Kimmel SE, French B, Kasner SE, et al. A pharmacogenetic versus a clinical algorithm for warfarin dosing. N Engl J Med. 2013;369:2283–2293.
  • Jarrar YB, Lee SJ. Molecular functionality of CYP2C9 polymorphisms and their influence on drug therapy. Drug Metabol Drug Interact. 2014;29:211–220.
  • Rosenson RS, Brewer HB Jr., Davidson WS, et al. Cholesterol efflux and atheroprotection: advancing the concept of reverse cholesterol transport. Circulation. 2012;125:1905–1919.
  • Norata GD, Catapano AL. Molecular mechanisms responsible for the antiinflammatory and protective effect of HDL on the endothelium. Vasc Health Risk Manag. 2005;1:119–129.
  • Ansell BJ, Navab M, Watson KE, et al. Anti-inflammatory properties of HDL. Rev Endocr Metab Disord. 2004;5:351–358.
  • Rosenson RS, Brewer HB Jr., Chapman MJ, et al. HDL measures, particle heterogeneity, proposed nomenclature, and relation to atherosclerotic cardiovascular events. Clin Chem. 2011;57:392–410.
  • Schwartz GG, Olsson AG, Abt M, et al. Effects of dalcetrapib in patients with a recent acute coronary syndrome. N Engl J Med. 2012;367:2089–2099.
  • Pitts R, Gunzburger E, Ballantyne CM, et al. Treatment with dalcetrapib modifies the relationship between high-density lipoprotein cholesterol and C-reactive protein. J Am Coll Cardiol. 2016;68:2488–2490.
  • Remaley AT. HDL cholesterol/HDL particle ratio: a new measure of HDL function? J Am Coll Cardiol. 2015;65:364–366.
  • Tardif JC, Rheaume E, Lemieux Perreault LP, et al. Pharmacogenomic determinants of the cardiovascular effects of dalcetrapib. Circ Cardiovasc Genet. 2015;8:372–382.
  • Tardif JC, Rhainds D, Brodeur M, et al. Genotype-dependent effects of dalcetrapib on cholesterol efflux and inflammation: concordance with clinical outcomes. Circ Cardiovasc Genet. 2016;9:340–348.
  • Westra HJ, Peters MJ, Esko T, et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet. 2013;45:1238–1243.
  • Steinberg D, Parthasarathy S, Carew TE, et al. Beyond cholesterol. Modifications of low-density lipoprotein that increase its atherogenicity. N Engl J Med. 1989;320:915–924.
  • Kagan VE, Serbinova EA, Forte T, et al. Recycling of vitamin E in human low density lipoproteins. J Lipid Res. 1992;33:385–397.
  • Rimm EB, Stampfer MJ, Ascherio A, et al. Vitamin E consumption and the risk of coronary heart disease in men. N Engl J Med. 1993;328:1450–1456.
  • Stampfer MJ, Hennekens CH, Manson JE, et al. Vitamin E consumption and the risk of coronary disease in women. N Engl J Med. 1993;328:1444–1449.
  • Carr AC, McCall MR, Frei B. Oxidation of LDL by myeloperoxidase and reactive nitrogen species: reaction pathways and antioxidant protection. Arterioscler Thromb Vasc Biol. 2000;20:1716–1723.
  • Vardi M, Levy NS, Levy AP. Vitamin E in the prevention of cardiovascular disease: the importance of proper patient selection. J Lipid Res. 2013;54:2307–2314.
  • Levy AP, Gerstein HC, Miller-Lotan R, et al. The effect of vitamin E supplementation on cardiovascular risk in diabetic individuals with different haptoglobin phenotypes. Diabetes Care. 2004;27:2767.
  • Levy AP, Hochberg I, Jablonski K, et al. Haptoglobin phenotype is an independent risk factor for cardiovascular disease in individuals with diabetes: the Strong Heart Study. J Am Coll Cardiol. 2002;40:1984–1990.
  • Milman U, Blum S, Shapira C, et al. Vitamin E supplementation reduces cardiovascular events in a subgroup of middle-aged individuals with both type 2 diabetes mellitus and the haptoglobin 2-2 genotype: a prospective double-blinded clinical trial. Arterioscler Thromb Vasc Biol. 2008;28:341–347.
  • Murray RK, Connell GE, Pert JH. The role of haptoglobin in the clearance and distribution of extracorpuscular hemoglobin. Blood. 1961;17:45–53.
  • Navab M, Anantharamaiah GM, Reddy ST, et al. Mechanisms of disease: proatherogenic HDL–an evolving field. Nat Clin Pract Endocrinol Metab. 2006;2:504–511.
  • Asleh R, Blum S, Kalet-Litman S, et al. Correction of HDL dysfunction in individuals with diabetes and the haptoglobin 2-2 genotype. Diabetes. 2008;57:2794–2800.
  • Levy AP, Purushothaman KR, Levy NS, et al. Downregulation of the hemoglobin scavenger receptor in individuals with diabetes and the Hp 2-2 genotype: implications for the response to intraplaque hemorrhage and plaque vulnerability. Circ Res. 2007;101:106–110.
  • Shin DG, Bayarsaihan D. A novel epi-drug therapy based on the suppression of BET family epigenetic readers. Yale J Biol Med. 2017;90:63–71.
  • Voelter-Mahlknecht S. Epigenetic associations in relation to cardiovascular prevention and therapeutics. Clin Epigenetics. 2016;8:4.
  • Wang B, Canestaro WJ, Choudhry NK. Clinical evidence supporting pharmacogenomic biomarker testing provided in US Food and Drug Administration drug labels. JAMA Intern Med. 2014;174:1938–1944.
  • Pulley JM, Denny JC, Peterson JF, et al. Operational implementation of prospective genotyping for personalized medicine: the design of the Vanderbilt PREDICT project. Clin Pharmacol Ther. 2012;92:87–95.

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.