1,191
Views
21
CrossRef citations to date
0
Altmetric
Review

The genetic basis of antiplatelet and anticoagulant therapy: A pharmacogenetic review of newer antiplatelets (clopidogrel, prasugrel and ticagrelor) and anticoagulants (dabigatran, rivaroxaban, apixaban and edoxaban)

ORCID Icon, &
Pages 725-739 | Received 22 Jun 2016, Accepted 31 May 2017, Published online: 13 Jun 2017

References

  • Scott SA. Personalizing medicine with clinical pharmacogenetics. Genet Med. 2011;13:987–995.
  • FDA Drug Safety Communication: Reduced effectiveness of Plavix (clopidogrel) in patients who are poor metabolizers of the drug. 2010. Available from: https://www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/ucm203888.htm.
  • Gage BF, Lesko LJ. Pharmacogenetics of warfarin: regulatory, scientific, and clinical issues. J Thromb Thrombolysis. 2008;25:45–51.
  • Weiss HJ, Aledort LM. Impaired platelet-connective-tissue reaction in man after aspirin ingestion. Lancet. 1967;2:495–497.
  • Jneid H, Anderson JL, Wright RS, et al. 2012 ACCF/AHA focused update of the guideline for the management of patients with unstable angina/non-ST-elevation myocardial infarction (updating the 2007 guideline and replacing the 2011 focused update): a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2012;60:645–681.
  • Levine GN, Bates ER, Blankenship JC, et al. 2011 ACCF/AHA/SCAI Guideline for percutaneous coronary intervention: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines and the society for cardiovascular angiography and interventions. Circulation. 2011;124:e574–651.
  • O’Gara PT, Kushner FG, Ascheim DD, et al.; American College of Cardiology F, American Heart Association Task Force on Practice G, American College of Emergency P, Society for Cardiovascular A, Interventions. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: executive summary: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines: developed in collaboration with the American College of Emergency Physicians and Society for Cardiovascular Angiography and Interventions. Catheter Cardiovasc Interv. 2013;82:E1–27.
  • Hamm CW, Bassand JP, Agewall S, et al., European Society of C, [ESC guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. The task force for the management of acute coronary syndromes (ACS) in patients presenting without persistent ST-segment elevation of the European Society of Cardiology (ESC)]. G Ital Cardiol (Rome). 2012;13:171–228.
  • Fitzpatrick FA. Cyclooxygenase enzymes: regulation and function. Curr Pharm Des. 2004;10:577–588.
  • Paikin JS, Eikelboom JW. Cardiology patient page: aspirin. Circulation. 2012;125:e439–42.
  • Halvorsen S, Andreotti F, Ten Berg JM, et al. Aspirin therapy in primary cardiovascular disease prevention: a position paper of the European Society of cardiology Working Group on Thrombosis. J Am Coll Cardiol. 2014;64:319–327.
  • Fitzgerald R, Pirmohamed M. Aspirin resistance: effect of clinical, biochemical and genetic factors. Pharmacol Ther. 2011;130:213–225.
  • Lordkipanidze M, Pharand C, Schampaert E, et al. A comparison of six major platelet function tests to determine the prevalence of aspirin resistance in patients with stable coronary artery disease. Eur Heart J. 2007;28:1702–1708.
  • Sofi F, Marcucci R, Gori AM, et al. Residual platelet reactivity on aspirin therapy and recurrent cardiovascular events–a meta-analysis. Int J Cardiol. 2008;128:166–171.
  • Gum PA, Kottke-Marchant K, Poggio ED, et al. Profile and prevalence of aspirin resistance in patients with cardiovascular disease. Am J Cardiol. 2001;88:230–235.
  • Hankey GJ, Eikelboom JW. Aspirin resistance. Lancet. 2006;367:606–617.
  • O’Donnell CJ, Larson MG, Feng D, et al. Genetic and environmental contributions to platelet aggregation: the Framingham heart study. Circulation. 2001;103:3051–3056.
  • Cooke GE, Liu-Stratton Y, Ferketich AK, et al. Effect of platelet antigen polymorphism on platelet inhibition by aspirin, clopidogrel, or their combination. J Am Coll Cardiol. 2006;47:541–546.
  • Maree AO, Curtin RJ, Chubb A, et al. Cyclooxygenase-1 haplotype modulates platelet response to aspirin. J Thromb Haemost. 2005;3:2340–2345.
  • Halushka MK, Walker LP, Halushka PV. Genetic variation in cyclooxygenase 1: effects on response to aspirin. Clin Pharmacol Ther. 2003;73:122–130.
  • Goodman T, Sharma P, Ferro A. The genetics of aspirin resistance. Int J Clin Pract. 2007;61:826–834.
  • Weng Z, Li X, Li Y, et al. The association of four common polymorphisms from four candidate genes (COX-1, COX-2, ITGA2B, ITGA2) with aspirin insensitivity: a meta-analysis. PLoS One. 2013;8:e78093.
  • Gonzalez-Conejero R, Rivera J, Corral J, et al. Biological assessment of aspirin efficacy on healthy individuals: heterogeneous response or aspirin failure? Stroke. 2005;36:276–280.
  • Kranzhofer R, Ruef J. Aspirin resistance in coronary artery disease is correlated to elevated markers for oxidative stress but not to the expression of cyclooxygenase (COX) 1/2, a novel COX-1 polymorphism or the PlA(1/2) polymorphism. Platelets. 2006;17:163–169.
  • Lepantalo A, Mikkelsson J, Resendiz JC, et al. Polymorphisms of COX-1 and GPVI associate with the antiplatelet effect of aspirin in coronary artery disease patients. Thromb Haemost. 2006;95:253–259.
  • Cipollone F, Rocca B, Patrono C. Cyclooxygenase-2 expression and inhibition in atherothrombosis. Arterioscler Thromb Vasc Biol. 2004;24:246–255.
  • Patrono C, Coller B, Dalen JE, et al. Platelet-active drugs: the relationships among dose, effectiveness, and side effects. Chest. 2001;119:39S–63S.
  • Sharma V, Kaul S, Al-Hazzani A, et al. Association of COX-2 rs20417 with aspirin resistance. J Thromb Thrombolysis. 2013;35:95–99.
  • Mukherjee D, Nissen SE, Topol EJ. Risk of cardiovascular events associated with selective COX-2 inhibitors. Jama. 2001;286:954–959.
  • Kearney PM, Baigent C, Godwin J, et al. Do selective cyclo-oxygenase-2 inhibitors and traditional non-steroidal anti-inflammatory drugs increase the risk of atherothrombosis? Meta-analysis of randomised trials. Bmj. 2006;332:1302–1308.
  • Investigators A, Connolly SJ, Pogue J, et al. Effect of clopidogrel added to aspirin in patients with atrial fibrillation. N Engl J Med. 2009;360:2066–2078.
  • Yusuf S, Zhao F, Mehta SR, et al. Clopidogrel in unstable angina to prevent recurrent events trial I. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med. 2001;345:494–502.
  • Investigators DT, Gerstein HC, Yusuf S, et al. Effect of rosiglitazone on the frequency of diabetes in patients with impaired glucose tolerance or impaired fasting glucose: a randomised controlled trial. Lancet. 2006;368:1096–1105.
  • Anand SS, Dagenais GR, Mohan V, et al. Glucose levels are associated with cardiovascular disease and death in an international cohort of normal glycaemic and dysglycaemic men and women: the EpiDREAM cohort study. Eur J Prev Cardiol. 2012;19:755–764.
  • Investigators O, Yusuf S, Teo KK, et al. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358:1547–1559.
  • Connolly SJ, Ezekowitz MD, Yusuf S, et al.; Committee R-LS, Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361:1139–1151.
  • Ridker PM, Chasman DI, Zee RY, et al. Women’s genome health study working G. Rationale, design, and methodology of the Women’s Genome Health Study: a genome-wide association study of more than 25,000 initially healthy American women. Clin Chem. 2008;54:249–255.
  • Ridker PM, Cook NR, Lee IM, et al. A randomized trial of low-dose aspirin in the primary prevention of cardiovascular disease in women. N Engl J Med. 2005;352:1293–1304.
  • Yusuf S, Hawken S, Ounpuu S, et al. Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study. Lancet. 2004;364:937–952.
  • Blankenhorn DH, Azen SP, Kramsch DM, et al., Group MR, Coronary angiographic changes with lovastatin therapy. The Monitored Atherosclerosis Regression Study (MARS). Ann Intern Med. 1993;119:969–976.
  • Ross S, Eikelboom J, Anand SS, et al. Association of cyclooxygenase-2 genetic variant with cardiovascular disease. Eur Heart J. 2014;35:2242–8a.
  • Watson SP, Gibbins J. Collagen receptor signalling in platelets: extending the role of the ITAM. Immunol Today. 1998;19:260–264.
  • Kunicki TJ, Kritzik M, Annis DS, et al. Hereditary variation in platelet integrin alpha 2 beta 1 density is associated with two silent polymorphisms in the alpha 2 gene coding sequence. Blood. 1997;89:1939–1943.
  • Nanda N, Bao M, Lin H, et al. Platelet endothelial aggregation receptor 1 (PEAR1), a novel epidermal growth factor repeat-containing transmembrane receptor, participates in platelet contact-induced activation. J Biol Chem. 2005;280:24680–24689.
  • Wurtz M, Nissen PH, Grove EL, et al. Genetic determinants of on-aspirin platelet reactivity: focus on the influence of PEAR1. PLoS One. 2014;9:e111816.
  • Sherry ST, Ward MH, Kholodov M, et al. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res. 2001;29:308–311.
  • 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 Mar;10(2):102–109.
  • 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.
  • Vijayan KV, Bray PF. Molecular mechanisms of prothrombotic risk due to genetic variations in platelet genes: enhanced outside-in signaling through the Pro33 variant of integrin beta3. Exp Biol Med (Maywood). 2006;231:505–513.
  • Sangkuhl K, Klein TE, Altman RB. Clopidogrel pathway. Pharmacogenet Genomics. 2010;20:463–465. Available from: https://www.pharmgkb.org/pathway/PA154424674
  • Gurbel PA, Bliden KP, Hiatt BL, et al. Clopidogrel for coronary stenting: response variability, drug resistance, and the effect of pretreatment platelet reactivity. Circulation. 2003;107:2908–2913.
  • Bal Dit Sollier C, Berge N, Boval B, et al. Functional variability of platelet response to clopidogrel correlates with P2Y(12) receptor occupancy. Thromb Haemost. 2009;101:116–122.
  • Angiolillo DJ, Bernardo E, Sabate M, et al. Impact of platelet reactivity on cardiovascular outcomes in patients with type 2 diabetes mellitus and coronary artery disease. J Am Coll Cardiol. 2007;50:1541–1547.
  • Collet JP, Cuisset T, Range G, et al. Bedside monitoring to adjust antiplatelet therapy for coronary stenting. N Engl J Med. 2012;367:2100–2109.
  • Price MJ, Berger PB, Teirstein PS, et al. Standard- vs high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention: the GRAVITAS randomized trial. Jama. 2011;305:1097–1105.
  • Simon T, Verstuyft C, Mary-Krause M, et al. French registry of acute STE, non STEMII. Genetic determinants of response to clopidogrel and cardiovascular events. N Engl J Med. 2009;360:363–375.
  • Taubert D, Von Beckerath N, Grimberg G, et al. Impact of P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther. 2006;80:486–501.
  • Spiewak M, Malek LA, Kostrzewa G, et al. Influence of C3435T multidrug resistance gene-1 (MDR-1) polymorphism on platelet reactivity and prognosis in patients with acute coronary syndromes. Kardiol Pol. 2009;67:827–834.
  • Kim IS, Jeong YH, Park Y, et al. Interaction analysis between genetic polymorphisms and pharmacodynamic effect in patients treated with adjunctive cilostazol to dual antiplatelet therapy: results of the ACCEL-TRIPLE (Accelerated platelet inhibition by triple antiplatelet therapy according to gene polymorphism) study. Br J Clin Pharmacol. 2012;73:629–640.
  • Jeong YH, Tantry US, Kim IS, et al. Effect of CYP2C19*2 and *3 loss-of-function alleles on platelet reactivity and adverse clinical events in East Asian acute myocardial infarction survivors treated with clopidogrel and aspirin. Circ Cardiovasc Interv. 2011;4:585–594.
  • Jeong YH, Kim IS, Park Y, et al. Carriage of cytochrome 2C19 polymorphism is associated with risk of high post-treatment platelet reactivity on high maintenance-dose clopidogrel of 150 mg/day: results of the ACCEL-DOUBLE (Accelerated platelet inhibition by a double dose of clopidogrel according to gene polymorphism) study. JACC Cardiovasc Interv. 2010;3:731–741.
  • Kazui M, Nishiya Y, Ishizuka T, et al. Identification of the human cytochrome P450 enzymes involved in the two oxidative steps in the bioactivation of clopidogrel to its pharmacologically active metabolite. Drug Metab Dispos. 2010;38:92–99.
  • Savi P, Pereillo JM, Uzabiaga MF, et al. Identification and biological activity of the active metabolite of clopidogrel. Thromb Haemost. 2000;84:891–896.
  • Tang M, Mukundan M, Yang J, et al. Antiplatelet agents aspirin and clopidogrel are hydrolyzed by distinct carboxylesterases, and clopidogrel is transesterificated in the presence of ethyl alcohol. J Pharmacol Exp Ther. 2006;319:1467–1476.
  • Zhu HJ, Wang X, Gawronski BE, et al. Carboxylesterase 1 as a determinant of clopidogrel metabolism and activation. J Pharmacol Exp Ther. 2013;344:665–672.
  • 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.
  • 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.
  • Schror K, Siller-Matula JM, Huber K. Pharmacokinetic basis of the antiplatelet action of prasugrel. Fundam Clin Pharmacol. 2012;26:39–46.
  • Bauer T, Bouman HJ, Van Werkum JW, et al. Impact of CYP2C19 variant genotypes on clinical efficacy of antiplatelet treatment with clopidogrel: systematic review and meta-analysis. Bmj. 2011;343:d4588.
  • Holmes MV, Perel P, Shah T, et al. CYP2C19 genotype, clopidogrel metabolism, platelet function, and cardiovascular events: a systematic review and meta-analysis. Jama. 2011;306:2704–2714.
  • Hulot JS, Bura A, Villard E, et al. Cytochrome P450 2C19 loss-of-function polymorphism is a major determinant of clopidogrel responsiveness in healthy subjects. Blood. 2006;108:2244–2247.
  • Hulot JS, Collet JP, Silvain J, et al. Cardiovascular risk in clopidogrel-treated patients according to cytochrome P450 2C19*2 loss-of-function allele or proton pump inhibitor coadministration: a systematic meta-analysis. J Am Coll Cardiol. 2010;56:134–143.
  • Jin B, Ni HC, Shen W, et al. Cytochrome P450 2C19 polymorphism is associated with poor clinical outcomes in coronary artery disease patients treated with clopidogrel. Mol Biol Rep. 2011;38:1697–1702.
  • Li Y, Tang HL, Hu YF, et al. The gain-of-function variant allele CYP2C19*17: a double-edged sword between thrombosis and bleeding in clopidogrel-treated patients. J Thromb Haemost. 2012;10:199–206.
  • Liu YP, Hao PP, Zhang MX, et al. Association of genetic variants in CYP2C19 and adverse clinical outcomes after treatment with clopidogrel: an updated meta-analysis. Thromb Res. 2011;128:593–594.
  • Mega JL, Simon T, Collet JP, et al. Reduced-function CYP2C19 genotype and risk of adverse clinical outcomes among patients treated with clopidogrel predominantly for PCI: a meta-analysis. Jama. 2010;304:1821–1830.
  • Osnabrugge RL, Head SJ, Zijlstra F, et al. A systematic review and critical assessment of 11 discordant meta-analyses on reduced-function CYP2C19 genotype and risk of adverse clinical outcomes in clopidogrel users. Genet Med. 2015;17:3–11.
  • Sofi F, Giusti B, Marcucci R, et al. Cytochrome P450 2C19*2 polymorphism and cardiovascular recurrences in patients taking clopidogrel: a meta-analysis. Pharmacogenomics J. 2011;11:199–206.
  • Zabalza M, Subirana I, Sala J, et al. Meta-analyses of the association between cytochrome CYP2C19 loss- and gain-of-function polymorphisms and cardiovascular outcomes in patients with coronary artery disease treated with clopidogrel. Heart. 2012;98:100–108.
  • Strom CM, Goos D, Crossley B, et al. Testing for variants in CYP2C19: population frequencies and testing experience in a clinical laboratory. Genet Med. 2012;14:95–100.
  • 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.
  • Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 polymorphisms and response to clopidogrel. N Engl J Med. 2009;360:354–362.
  • 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.
  • Desta Z, Zhao X, Shin JG, et al. Clinical significance of the cytochrome P450 2C19 genetic polymorphism. Clin Pharmacokinet. 2002;41:913–958.
  • Mackness B, Mackness MI, Arrol S, et al. Effect of the human serum paraoxonase 55 and 192 genetic polymorphisms on the protection by high density lipoprotein against low density lipoprotein oxidative modification. FEBS Lett. 1998;423:57–60.
  • Bouman HJ, Schomig E, Van Werkum JW, et al. Paraoxonase-1 is a major determinant of clopidogrel efficacy. Nat Med. 2011;17:110–116.
  • Humbert R, Adler DA, Disteche CM, et al. The molecular basis of the human serum paraoxonase activity polymorphism. Nat Genet. 1993;3:73–76.
  • Mackness B, Mackness MI, Arrol S, et al. Effect of the molecular polymorphisms of human paraoxonase (PON1) on the rate of hydrolysis of paraoxon. Br J Pharmacol. 1997;122:265–268.
  • 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.
  • Zhou SF. Drugs behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. Curr Drug Metab. 2008;9:310–322.
  • Elens L, Van Gelder T, Hesselink DA, et al. CYP3A4*22: promising newly identified CYP3A4 variant allele for personalizing pharmacotherapy. Pharmacogenomics. 2013;14:47–62.
  • Lee JS, Cheong HS, Kim LH, et al. Screening of Genetic Polymorphisms of CYP3A4 and CYP3A5 Genes. Korean J Physiol Pharmacol. 2013;17:479–484.
  • Klein K, Zanger UM. Pharmacogenomics of cytochrome P450 3A4: recent progress toward the “Missing heritability” problem. Front Genet. 2013;4:12.
  • Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, et al. Contribution of gene sequence variations of the hepatic cytochrome P450 3A4 enzyme to variability in individual responsiveness to clopidogrel. Arterioscler Thromb Vasc Biol. 2006;26:1895–1900.
  • Smith SM, Judge HM, Peters G, et al. Common sequence variations in the P2Y12 and CYP3A5 genes do not explain the variability in the inhibitory effects of clopidogrel therapy. Platelets. 2006;17:250–258.
  • Suh JW, Koo BK, Zhang SY, et al. Increased risk of atherothrombotic events associated with cytochrome P450 3A5 polymorphism in patients taking clopidogrel. Cmaj. 2006;174:1715–1722.
  • Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–2015.
  • Farid NA, Smith RL, Gillespie TA, et al. The disposition of prasugrel, a novel thienopyridine, in humans. Drug Metab Dispos. 2007;35:1096–1104.
  • 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.
  • Williams ET, Jones KO, Ponsler GD, et al. The biotransformation of prasugrel, a new thienopyridine prodrug, by the human carboxylesterases 1 and 2. Drug Metab Dispos. 2008;36:1227–1232.
  • Jones RD, Taylor AM, Tong EY, et al. Carboxylesterases are uniquely expressed among tissues and regulated by nuclear hormone receptors in the mouse. Drug Metab Dispos. 2013;41:40–49.
  • Wiviott SD, Antman EM, Braunwald E. Prasugrel. Circulation. 2010;122:394–403.
  • Rehmel JL, Eckstein JA, Farid NA, et al. Interactions of two major metabolites of prasugrel, a thienopyridine antiplatelet agent, with the cytochromes P450. Drug Metab Dispos. 2006;34:600–607.
  • Mega JL, Close SL, Wiviott SD, et al. Cytochrome P450 genetic polymorphisms and the response to prasugrel: relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Circulation. 2009;119:2553–2560.
  • Cuisset T, Loosveld M, Morange PE, et al. CYP2C19*2 and *17 alleles have a significant impact on platelet response and bleeding risk in patients treated with prasugrel after acute coronary syndrome. JACC Cardiovasc Interv. 2012;5:1280–1287.
  • Dansette PM, Rosi J, Debernardi J, et al. Metabolic activation of prasugrel: nature of the two competitive pathways resulting in the opening of its thiophene ring. Chem Res Toxicol. 2012;25:1058–1065.
  • Mega JL, Close SL, Wiviott SD, et al. PON1 Q192R genetic variant and response to clopidogrel and prasugrel: pharmacokinetics, pharmacodynamics, and a meta-analysis of clinical outcomes. J Thromb Thrombolysis. 2016;41:374–383.
  • Xiang Q, Cui Y, Zhao X, et al. Identification of PEAR1 SNPs and their influences on the variation in prasugrel pharmacodynamics. Pharmacogenomics. 2013;14:1179–1189.
  • Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045–1057.
  • Siller-Matula JM, Jilma B. Ticagrelor: from discovery to phase III clinical trial. Future Cardiol. 2010;6:753–764.
  • Teng R, Butler K. Pharmacokinetics, pharmacodynamics, tolerability and safety of single ascending doses of ticagrelor, a reversibly binding oral P2Y(12) receptor antagonist, in healthy subjects. Eur J Clin Pharmacol. 2010;66:487–496.
  • Teng R. Pharmacokinetic, pharmacodynamic and pharmacogenetic profile of the oral antiplatelet agent ticagrelor. Clin Pharmacokinet. 2012;51:305–318.
  • Gurbel PA, Bliden KP, Butler K, et al. Randomized double-blind assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coronary artery disease: the ONSET/OFFSET study. Circulation. 2009;120:2577–2585.
  • Gurbel PA, Bliden KP, Butler K, et al. Response to ticagrelor in clopidogrel nonresponders and responders and effect of switching therapies: the RESPOND study. Circulation. 2010;121:1188–1199.
  • Tantry US, Bliden KP, Wei C, et al. First analysis of the relation between CYP2C19 genotype and pharmacodynamics in patients treated with ticagrelor versus clopidogrel: the ONSET/OFFSET and RESPOND genotype studies. Circ Cardiovasc Genet. 2010;3:556–566.
  • Varenhorst CMH, Eriksson N, Johansson A, et al. Ticagrelor plasma levels but not clinical outcomes are associated with transporter and metabolism enzyme genetic polymorphisms. Journal of the American College of Cardiology. 63(12):A25, April 2014
  • Hirsh J, Fuster V, Ansell J, et al. American College of Cardiology F. American Heart Association/American College of Cardiology Foundation guide to warfarin therapy. Circulation. 2003;107:1692–1711.
  • Breckenridge A. Oral anticoagulant drugs: pharmacokinetic aspects. Semin Hematol. 1978;15:19–26.
  • Kelly JG, O’Malley K. Clinical pharmacokinetics of oral anticoagulants. Clin Pharmacokinet. 1979;4:1–15.
  • O’Reilly RA. Warfarin metabolism and drug-drug interactions. Adv Exp Med Biol. 1987;214:205–212.
  • Choonara IA, Haynes BP, Cholerton S, et al. Enantiomers of warfarin and vitamin K1 metabolism. Br J Clin Pharmacol. 1986;22:729–732.
  • Jansing RL, Chao ES, Kaminsky LS. Phase II metabolism of warfarin in primary culture of adult rat hepatocytes. Mol Pharmacol. 1992;41:209–215.
  • Aithal GP, Day CP, Kesteven PJ, et al. Association of polymorphisms in the cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding complications. Lancet. 1999;353:717–719.
  • Higashi MK, Veenstra DL, Kondo LM, et al. Association between CYP2C9 genetic variants and anticoagulation-related outcomes during warfarin therapy. Jama. 2002;287:1690–1698.
  • Limdi NA, Brown TM, Yan Q, et al. Race influences warfarin dose changes associated with genetic factors. Blood. 2015;126:539–545.
  • Wajih N, Hutson SM, Owen J, et al. Increased production of functional recombinant human clotting factor IX by baby hamster kidney cells engineered to overexpress VKORC1, the vitamin K 2,3-epoxide-reducing enzyme of the vitamin K cycle. J Biol Chem. 2005;280:31603–31607.
  • Nunnelee JD. Review of an article: the International Warfarin Pharmacogenetics Consortium (2009). Estimation of the warfarin dose with clinical and pharmacogenetic data. NEJM 360 (8): 753-64. J Vasc Nurs. 2009;27:109.
  • Yuan HY, Chen JJ, Lee MT, et al. A novel functional VKORC1 promoter polymorphism is associated with inter-individual and inter-ethnic differences in warfarin sensitivity. Hum Mol Genet. 2005;14:1745–1751.
  • Schwarz UI, Ritchie MD, Bradford Y, et al. Genetic determinants of response to warfarin during initial anticoagulation. N Engl J Med. 2008;358:999–1008.
  • Nakamura K, Obayashi K, Araki T, et al. CYP4F2 gene polymorphism as a contributor to warfarin maintenance dose in Japanese subjects. J Clin Pharm Ther. 2012;37:481–485.
  • Caldwell MD, Awad T, Johnson JA, et al. CYP4F2 genetic variant alters required warfarin dose. Blood. 2008;111:4106–4112.
  • Teichert M, Eijgelsheim M, Rivadeneira F, et al. A genome-wide association study of acenocoumarol maintenance dosage. Hum Mol Genet. 2009;18:3758–3768.
  • 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.
  • Epstein RS, Moyer TP, Aubert RE, et al. Warfarin genotyping reduces hospitalization rates results from the MM-WES (Medco-Mayo Warfarin Effectiveness study). J Am Coll Cardiol. 2010;55:2804–2812.
  • You JH, Tsui KK, Wong RS, et al. Cost-effectiveness of dabigatran versus genotype-guided management of warfarin therapy for stroke prevention in patients with atrial fibrillation. PLoS One. 2012;7:e39640.
  • Shi C, Yan W, Wang G, et al. Pharmacogenetics-based versus conventional dosing of warfarin: a meta-analysis of randomized controlled trials. PLoS One. 2015;10:e0144511.
  • Johnson JA, Gong L, Whirl-Carrillo M, et al. Clinical pharmacogenetics implementation C. Clinical pharmacogenetics implementation consortium guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin Pharmacol Ther. 2011;90:625–629.
  • Garcia DA, Witt DM, Hylek E, et al. Delivery of optimized anticoagulant therapy: consensus statement from the anticoagulation forum. Ann Pharmacother. 2008;42:979–988.
  • Hankey GJ, Eikelboom JW. Dabigatran etexilate: a new oral thrombin inhibitor. Circulation. 2011;123:1436–1450.
  • Stangier J, Rathgen K, Stahle H, et al. The pharmacokinetics, pharmacodynamics and tolerability of dabigatran etexilate, a new oral direct thrombin inhibitor, in healthy male subjects. Br J Clin Pharmacol. 2007;64:292–303.
  • Ebner T, Wagner K, Wienen W. Dabigatran acylglucuronide, the major human metabolite of dabigatran: in vitro formation, stability, and pharmacological activity. Drug Metab Dispos. 2010;38:1567–1575.
  • Pare G, Eriksson N, Lehr T, et al. Genetic determinants of dabigatran plasma levels and their relation to bleeding. Circulation. 2013;127:1404–1412.
  • Dimatteo C, D’Andrea G, Vecchione G, et al. Pharmacogenetics of dabigatran etexilate interindividual variability. Thromb Res. 2016;144:1–5.
  • Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883–891.
  • Gnoth MJ, Buetehorn U, Muenster U, et al. In vitro and in vivo P-glycoprotein transport characteristics of rivaroxaban. J Pharmacol Exp Ther. 2011;338:372–380.
  • Mueck W, Kubitza D, Becka M. Co-administration of rivaroxaban with drugs that share its elimination pathways: pharmacokinetic effects in healthy subjects. Br J Clin Pharmacol. 2013;76:455–466.
  • Cusatis G, Sparreboom A. Pharmacogenomic importance of ABCG2. Pharmacogenomics. 2008;9:1005–1009.
  • Cusatis G, Gregorc V, Li J, et al. Pharmacogenetics of ABCG2 and adverse reactions to gefitinib. J Natl Cancer Inst. 2006;98:1739–1742.
  • Woodward OM, Tukaye DN, Cui J, et al. Gout-causing Q141K mutation in ABCG2 leads to instability of the nucleotide-binding domain and can be corrected with small molecules. Proc Natl Acad Sci U S A. 2013;110:5223–5228.
  • Gong IY, Mansell SE, Kim RB. Absence of both MDR1 (ABCB1) and breast cancer resistance protein (ABCG2) transporters significantly alters rivaroxaban disposition and central nervous system entry. Basic Clin Pharmacol Toxicol. 2013;112:164–170.
  • Sim SC, Ingelman-Sundberg M. Update on allele nomenclature for human cytochromes P450 and the human cytochrome P450 allele (CYP-allele) nomenclature database. Methods Mol Biol. 2013;987:251–259.
  • Solus JF, Arietta BJ, Harris JR, et al. Genetic variation in eleven phase I drug metabolism genes in an ethnically diverse population. Pharmacogenomics. 2004;5:895–931.
  • King LM, Gainer JV, David GL, et al. Single nucleotide polymorphisms in the CYP2J2 and CYP2C8 genes and the risk of hypertension. Pharmacogenet Genomics. 2005;15:7–13.
  • Wu SN, Zhang Y, Gardner CO, et al. Evidence for association of polymorphisms in CYP2J2 and susceptibility to essential hypertension. Ann Hum Genet. 2007;71:519–525.
  • Granger CB, Alexander JH, McMurray JJ, et al.; Committees A, Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365:981–992.
  • Alberts MJ, Eikelboom JW, Hankey GJ. Antithrombotic therapy for stroke prevention in non-valvular atrial fibrillation. Lancet Neurol. 2012;11:1066–1081.
  • Wang L, Zhang D, Raghavan N, et al. In vitro assessment of metabolic drug-drug interaction potential of apixaban through cytochrome P450 phenotyping, inhibition, and induction studies. Drug Metab Dispos. 2010;38:448–458.
  • Raghavan N, Frost CE, Yu Z, et al. Apixaban metabolism and pharmacokinetics after oral administration to humans. Drug Metab Dispos. 2009;37:74–81.
  • Nutescu E, Chuatrisorn I, Hellenbart E. Drug and dietary interactions of warfarin and novel oral anticoagulants: an update. J Thromb Thrombolysis. 2011;31:326–343.
  • Wang L, Raghavan N, He K, et al. Sulfation of o-demethyl apixaban: enzyme identification and species comparison. Drug Metab Dispos. 2009;37:802–808.
  • Nagar S, Walther S, Blanchard RL. Sulfotransferase (SULT) 1A1 polymorphic variants *1, *2, and *3 are associated with altered enzymatic activity, cellular phenotype, and protein degradation. Mol Pharmacol. 2006;69:2084–2092.
  • Giugliano RP, Ruff CT, Braunwald E, et al. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093–2104.
  • Bathala MS, Masumoto H, Oguma T, et al. Pharmacokinetics, biotransformation, and mass balance of edoxaban, a selective, direct factor Xa inhibitor, in humans. Drug Metab Dispos. 2012;40:2250–2255.
  • Bounameaux H, Camm AJ. Edoxaban: an update on the new oral direct factor Xa inhibitor. Drugs. 2014;74:1209–1231.
  • Matsushima N, Lee F, Sato T, et al. Bioavailability and safety of the factor xa inhibitor edoxaban and the effects of quinidine in healthy subjects. Clin Pharmacol Drug Dev. 2013;2:358–366.
  • Shuldiner AR, Palmer K, Pakyz RE, et al. Implementation of pharmacogenetics: the University of Maryland personalized anti-platelet pharmacogenetics program. Am J Med Genet C Semin Med Genet. 2014;166C:76–84.
  • Levine GN, Bates ER, Bittl JA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2016;68:1082–1115.
  • Scott SA, Sangkuhl K, Stein CM, et al. Clinical pharmacogenetics implementation C. Clinical pharmacogenetics implementation consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update. Clin Pharmacol Ther. 2013;94:317–323.
  • You JH. Pharmacogenetic-guided selection of warfarin versus novel oral anticoagulants for stroke prevention in patients with atrial fibrillation: a cost-effectiveness analysis. Pharmacogenet Genomics. 2014;24:6–14.
  • Mega JL, Walker JR, Ruff CT, et al. Genetics and the clinical response to warfarin and edoxaban: findings from the randomised, double-blind ENGAGE AF-TIMI 48 trial. Lancet. 2015;385:2280–2287.
  • Su J, Xu J, Li X, et al. ABCB1 C3435T polymorphism and response to clopidogrel treatment in coronary artery disease (CAD) patients: a meta-analysis. PLoS One. 2012;7:e46366.
  • Tantisira KG, Lasky-Su J, Harada M, et al. Genomewide association between GLCCI1 and response to glucocorticoid therapy in asthma. N Engl J Med. 2011;365:1173–1183.
  • Parra EJ, Botton MR, Perini JA, et al. Genome-wide association study of warfarin maintenance dose in a Brazilian sample. Pharmacogenomics. 2015;16:1253–1263.
  • Meneveau NFSF, Obert L, Capellier G, et al. Influence of ABCB1 polymorphisms on plasma concentrations of new oral anticoagulants in case of serious adverse events (Pgp NACO). Clinicaltrialsgov Identifier. 2014;NCT02103101.

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.