220
Views
42
CrossRef citations to date
0
Altmetric
Review

Pharmacodynamic properties of antiplatelet agents: current knowledge and future perspectives

, , &
Pages 319-336 | Published online: 10 Jan 2014

References

  • Davi G, Patrono C. Platelet activation and atherothrombosis. N. Engl. J. Med.357, 2482–2494 (2007).
  • Varga-Szabo D, Pleines I, Nieswandt B. Cell adhesion mechanisms in platelets. Arterioscler. Thromb. Vasc. Biol.28, 403–412 (2008).
  • Jennings LK. Mechanisms of platelet activation: need for new strategies to protect against platelet-mediated atherothrombosis. Thromb. Haemost.102, 248–257 (2009).
  • Patrono C, Andreotti F, Arnesen H et al. Antiplatelet agents for the treatment and prevention of atherothrombosis. Eur. Heart J.32, 2922–2932 (2011).
  • Michelson AD. Antiplatelet therapies for the treatment of cardiovascular disease. Nat. Rev. Drug. Discov.9, 154–169 (2011).
  • Papathanasiou A, Goudevenos J, Tselepis AD. Resistance to aspirin and clopidogrel: possible mechanisms, laboratory investigation, and clinical significance. Hellenic J. Cardiol.48, 352–363 (2007).
  • Antithrombotic Trialists’ Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. BMJ324, 71–86 (2002).
  • Krasopoulos G, Brister S, Beattie WS, Buchanan MR. Aspirin ‘resistance’ and risk of cardiovascular morbidity: systematic review and meta-analysis. BMJ336, 195–198 (2008).
  • Snoep JD, Hovens MM, Eikenboom JC, van der Bom JG, Huisman MV. Association of laboratory-defined aspirin resistance with a higher risk of recurrent cardiovascular events: a systematic review and meta-analysis. Arch. Intern. Med.167, 1593–1159 (2007).
  • Loureiro-Silva MR, Kouyoumdjian M, Borges DR. Plasma kallikrein and thrombin are cleared through unrelated hepatic pathways. Blood Coagul. Fibrinolysis4, 551–554 (1993).
  • Undas A, Siudak R, Topór-Mądry R et al. Simvastatin administration reduces thromboxane production in subjects taking aspirin: links between aspirin resistance and thrombin generation. Int. J. Cardiol.154, 59–64 (2012).
  • Tantry US, Bliden KP, Gurbel PA. Overestimation of platelet aspirin resistance detection by thrombelastograph platelet mapping and validation by conventional aggregometry using arachidonic acid stimulation. J. Am. Coll. Cardiol.46, 1705–1709 (2005).
  • Di Minno G. Aspirin resistance and platelet turnover: a 25-year old issue. Nutr. Metab. Cardiovasc. Dis.21, 542–545 (2011).
  • Feher G, Feher A, Pusch G et al. The genetics of antiplatelet drug resistance. Clin. Genet.75, 1–18 (2009).
  • Anninos H, Andrikopoulos G, Pastromas S, Sakellariou D, Theodorakis G, Vardas P. Triflusal: an old drug in modern antiplatelet therapy. Review of its action, use, safety and effectiveness. Hellenic J. Cardiol.50, 199–207 (2009).
  • Valle M, Barbanoj M, Donner A et al. Access of HTB, main metabolite of triflusal, to celebrospinal fluid in healthy volunteers. Eur. J. Clin. Pharmacol.61, 103–111 (2005).
  • Antonijoan RM, Gish I, Azaro A et al. Gastrointestinal safety of triflusal solution in healthy volunteers: a proof of concept endoscopic study. Eur. J. Clin. Pharmacol.67, 663–669 (2011).
  • Matias-Guiu J, Ferro J, Álvarez-Sabin J et al. Comparison of triflusal and aspirin for prevention of vascular events in patients after cerebral infarction: the TACIP study: a randomized, double-blind, multicenter trial. Stroke34, 840–848 (2003).
  • Mega JL, Close S, Wiviott SD et al. Cytochrome P450 genetic polymorphisms and the response to prasugrel relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Circulation119, 2553–2560 (2009).
  • Bonello L, Camoin-Jau L, Armero S et al. Tailored clopidogrel loading dose according to platelet reactivity monitoring to prevent acute and subacute stent thrombosis. Am. J. Cardiol.103, 5–10 (2009).
  • Wallentin L. P2Y12 inhibitors: differences in properties and mechanisms of action and potential consequences for clinical use. Eur. Heart J.30, 1964–1977 (2009).
  • Aleil B, Jacquemin L, De Poli F et al. Clopidogrel 150 mg/day to overcome low responsiveness in patients undergoing elective percutaneous coronary intervention: results from the VASP-02 (Vasodilator-Stimulated Phosphoprotein-02) randomized study. JACC Cardiovasc. Interv.1, 631–638 (2008).
  • Bhatt DL, Bertrand ME, Berger PB et al. Meta-analysis of randomized and registry comparisons of ticlopidine with clopidogrel after stenting. J. Am. Coll. Cardiol.39, 9–14 (2002).
  • Tselepis AD, Tsoumani M, Kalantzi K et al. Influence of HDL and paraoxonase-1 on platelet reactivity in patients with acute coronary syndromes receiving clopidogrel therapy. J. Thromb. Haemost.9(12), 2371–2378 (2011).
  • Bouman HJ, Schömig E, van Werkum JW et al. Paraoxonase-1 is a major determinant of clopidogrel efficacy. Nat. Med.1, 110–116 (2011).
  • 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.32, 1605–1613 (2011).
  • Kalantzi KI, Dimitriou A, Goudevenos JA et al. The platelet hyporesponsiveness to clopidogrel in acute coronary syndrome patients treated with 75 mg/day clopidogrel may be overcome within one month of treatment. Platelets23(2), 121–131 (2011).
  • Tsoumani ME, Kalantzi K, Goudevenos I et al. Platelet-mediated inflammation in cardiovascular disease. Potential role of platelet-endothelium interactions. Curr. Vascul. Pharmacol. (2012) (In Press).
  • von Beckerath N, Kastrati A, Wieczorek A et al. A double-blind, randomized study on platelet aggregation in patients treated with a daily dose of 150 or 75 mg of clopidogrel for 30 days. Eur. Heart. J.28, 1814–1819 (2007).
  • Palmerini T, Barozzi C, Tomasi L et al. A randomised study comparing the antiplatelet and antinflammatory effect of clopidogrel 150 mg/day versus 75 mg/day in patients with ST-segment elevation acute myocardial infarction and poor responsiveness to clopidogrel: results from the DOUBLE study. Thromb. Res.125, 309–314 (2007).
  • Mega JL, Close S, Wiviott SD et al. Cytochrome p-450 polymorphisms and response to clopidogrel. N. Engl. J. Med.360, 354–362 (2009).
  • 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. JAMA304, 1821–1830 (2010).
  • Serebruany V, Cherala G, Williams C et al. Association of platelet responsiveness with clopidogrel metabolism: role of compliance in the assessment of ‘resistance’. Am. Heart. J.158, 925–932 (2009).
  • Qureshi Z, Hobson A. Clopidogrel ‘resistance’: where are we now? Cardiovasc. Ther. doi:10.1111/j.1755-5922.2011.00296.x (2011) (Epub ahead of print).
  • Sabatine MS, Cannon CP, Gibson CM et al. Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with ST-segment elevation. N. Engl. J. Med.352, 1179–1189 (2005).
  • Steinhubl SR, Berger PB, Mann JT 3rd et al. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. JAMA288, 2411–2420 (2002).
  • Kalantzi KI, Dimitriou A, Milionis HJ, Goudevenos IA, Tselepis AD. Clopidogrel differentially affects platelet-mediated thrombosis and inflammatory response in patients with acute coronary syndromes. J. Thromb. Haemost.4, 875–878 (2011).
  • Mega JL, Hochholzer W, Frelinger AL et al. Dosing clopidogrel based on CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovascular disease. JAMA306, 2221–2228 (2011).
  • Bonello L, Camoin-Jau L, Arques S et al. Adjusted clopidogrel loading doses according to vasodilator-stimulated phosphoprotein phosphorylation index decrease rate of major adverse cardiovascular events in patients with clopidogrel resistance: a multicenter randomized prospective study. J. Am. Coll. Cardiol.51, 1404–1411 (2008).
  • Price MJ, Berger P, Teirstein PS et al. Standard- vs high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention: the GRAVITAS randomized trial. JAMA305, 1097–1105 (2011).
  • Valgimigli M, Campo G, de Cesare N et al.; Tailoring Treatment with Tirofiban in Patients Showing Resistance to Aspirin and/or Resistance to Clopidogrel (3T/2R) Investigators. Intensifying platelet inhibition with tirofiban in poor responders to aspirin, clopidogrel, or both agents undergoing elective coronary intervention: results from the double-blind, prospective, randomized tailoring treatment with tirofiban in patients showing resistance to aspirin and/or resistance to clopidogrel study. Circulation119, 3215–3222 (2009).
  • Angiolillo DJ, Gibson CM, Cheng S et al. Differential effects of omeprazole and pantoprazole on the pharmacodynamics and pharmacokinetics of clopidogrel in healthy subjects: randomized, placebo-controlled, crossover comparison studies. Clin. Pharmacol. Ther.89, 65–74 (2011).
  • Fernando H, Dart A, Peter K, Shaw JA. Proton pump inhibitors, genetic polymorphisms and response to clopidogrel therapy. Thromb. Haemost.105, 933–944 (2011).
  • Bhatt DL, Cryer BL, Contant CF et al. Clopidogrel with or without omeprazole in coronary artery disease. N. Engl. J. Med.363, 1909–1917 (2010).
  • Abraham NS, Hlatky MA, Antman EM et al. ACCF/ACG/AHA 2010 expert consensus document on the concomitant use of proton pump inhibitors and thienopyridines: a focused update of the ACCF/ACG/AHA 2008 expert consensus document on reducing the gastrointestinal risks of antiplatelet therapy and NSAID use: a report of the American College of Cardiology Foundation Task Force on expert consensus documents. Circulation122, 2619–2633 (2010).
  • Neubauer H, Gunesdogan B, Hanefeld C, Spiecker M, Mugge A. Lipophilic statins interfere with the inhibitory effects of clopidogrel on platelet function – a flow cytometry study. Eur. Heart J.24, 1744–1749 (2003).
  • Lau WC, Waskell LA, Watkins PB et al. Atorvastatin reduces the ability of clopidogrel to inhibit platelet aggregation: a new drug–drug interaction. Circulation107, 32–37 (2003).
  • Mitsios JV, Papathanasiou AI, Rodis FI, Elisaf M, Goudevenos JA, Tselepis AD. Atorvastatin does not affect the antiplatelet potency of clopidogrel when it is administered concomitantly for 5 weeks in patients with acute coronary syndromes. Circulation109, 1335–1338 (2004).
  • Bates ER, Lau WC, Angiolillo DJ. Clopidogrel–drug interactions. J. Am. Coll. Cardiol.57, 1251–1263 (2011).
  • Smith SM, Judge HM, Peters G, Storey RF. Multiple antiplatelet effects of clopidogrel are not modulated by statin type in patients undergoing percutaneous coronary intervention. Platelets15, 465–474 (2004).
  • Saw J, Brennan DM, Steinhubl SR et al. Lack of evidence of a clopidogrel-statin interaction in the CHARISMA trial. J. Am. Coll. Cardiol.50, 291–295 (2007).
  • Kim SD, Kang W, Lee HW et al. Bioequivalence and tolerability of two clopidogrel salt preparations, besylate and bisulfate: a randomized, open-label, crossover study in healthy Korean male subjects. Clin. Ther.31, 793–803 (2009).
  • Park JY, Kim KA, Ryu JH, Lee GH, Jeon SH, Kim JS. Pharmacokinetics and the antiplatelet effect of a new clopidogrel formulation, clopidogrel besylate, in healthy subjects. Int. J. Clin. Pharmacol. Ther.48, 259–269 (2010).
  • Tsoumani ME, Kalantzi K, Dimitriou AA, Ntalas IV, Goudevenos IA, Tselepis AD. Antiplatelet efficacy of long-term treatment with clopidogrel besylate in patients with a history of acute coronary syndrome: comparison with clopidogrel hydrogen sulfate. Angiology doi:10.1177/0003319711427697 (2011) (Epub ahead of print).
  • Tsoumani ME, Kalantzi K, Dimitriou AA, Ntalas IV, Goudevenos IA, Tselepis AD. Effect of clopidogrel besylate on platelet reactivity in patients with acute coronary syndromes. Comparison with clopidogrel hydrogen sulfate. Exp. Opin. Pharmacother.13, 149–158 (2012).
  • Wiviott S, Trenk D, Frelinger AL et al. Prasugrel compared with high loading- and maintenance-dose clopidogrel in patients with planned percutaneous coronary intervention: the prasugrel in comparison to clopidogrel for inhibition of platelet activation and aggregation-thrombolysis in myocardial infarction 44 trial. Circulation116, 2923–2932 (2007).
  • Mega JL, Close S, 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. Lancet376, 1312–1319 (2010).
  • Alexopoulos D, Dimitropoulos G, Davlouros P et al. Prasugrel overcomes high on-clopidogrel platelet reactivity post-stenting more effectively than high-dose (150 mg) clopidogrel. The importance of CYP2C19*2 genotyping. J. Am. Coll. Cardiol.4, 403–410 (2011).
  • Bonello L, Tantry U, Marcucci R et al. Consensus and future directions on the definition of high on-treatment platelet reactivity to adenosine diphosphate. J. Am. Coll. Cardiol.56, 919–933 (2010).
  • Springthorpe B, Bailey A, Barton P et al. From ATP to AZD6140: the discovery of an orally active reversible P2Y12 receptor antagonist for the prevention of thrombosis. Bioorgan. Med. Chem. Lett.17, 6013–6018 (2007).
  • Storey RF. Pharmacology and clinical trials of reversibly-binding P2Y12 inhibitors. Thromb. Haemost.105, S75–S81 (2011).
  • Gurbel PA, Bliden K, 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. Circulation120, 2577–2585 (2009).
  • James S, Budaj A, Aylward P et al. Ticagrelor versus clopidogrel in acute coronary syndromes in relation to renal function: results from the platelet inhibition and patient outcomes (PLATO) trial. Circulation122, 1056–1067 (2010).
  • Wallentin L, James S, Storey RF et al. PLATO investigators. Effect of CYP2C19 and ABCB1 single nucleotide polymorphisms on outcomes of treatment with ticagrelor versus clopidogrel for acute coronary syndromes: a genetic substudy of the PLATO trial. Lancet376, 1320e8 (2010).
  • Angiolillo DJ, Bhatt D, Gurbel PA, Jennings LK. Advances in antiplatelet therapy: agents in clinical development. Am. J. Cardiol.103(Suppl.), 40A–51A (2009).
  • Steinhubl SR, Oh J, Oestreich JH et al. Transitioning patients from cangrelor to clopidogrel: pharmacodynamic evidence of a competitive effect. Thromb. Res.121, 527–534 (2008).
  • Bhatt DL, Lincoff AM, Gibson CM et al. Intravenous platelet blockade with cangrelor during PCI. N. Engl. J. Med.361, 2330–2341 (2009).
  • Harrington RA, Stone GW, McNulty S et al. Platelet inhibition with cangrelor in patients undergoing PCI. N. Engl. J. Med.361, 2318–2329 (2009).
  • Berger JS, Roe MT, Gibson CM et al. Safety and feasibility of adjunctive antiplatelet therapy with intravenous elinogrel, a direct-acting and reversible P2Y12 ADP-receptor antagonist, before primary percutaneous intervention in patients with ST-elevation myocardial infarction: the early rapid reversal of platelet thrombosis with intravenous elinogrel before PCI to optimize reperfusion in acute myocardial infarction (ERASE MI) pilot trial. Am. Heart J.158, 998–1004.E1 (2009).
  • Leonardi S, Rao S, Harrington RA et al. Rationale and design of the randomized, double-blind trial testing intravenous and oral administration of elinogrel, a selective and reversible P2Y(12)- receptor inhibitor, versus clopidogrel to valuate tolerability and efficacy in nonurgent percutaneous coronary interventions patients (INNOVATE-PCI). Am. Heart J.160, 65–72 (2010).
  • Gurbel PA, Bliden KP, Antonino MJ et al. The effect of elinogrel on high platelet reactivity during dual antiplatelet therapy and the relation to CYP 2C19*2 genotype: first experience in patients. J. Thromb. Haemost.8, 43–53 (2010).
  • Phillips DR, Charro I, Parise LV, Fitzgerald LA. The platelet membrane glycoprotein IIb–IIIa complex. Blood71, 831–843 (1988).
  • Wagner CL, Maselli M, Neblock DS, Weisman HF, Coller BS, Jordan RE. Analysis of GPIIb/IIIa receptor number by quantification of 7E3 binding to human platelets. Blood88, 907–914 (1996).
  • Mitsios JV, Tambaki A, Abatzis M et al. Effect of synthetic peptides corresponding to residues 313–332 of the aIIb subunit on platelet activation and fibrinogen binding to aIIbb3. Eur. J. Biochem.271, 855–862 (2004).
  • Kloczewiak M, Timmons S, Hawiger J. Localization of a site interacting with human platelet receptor on carboxy-terminal segment of human fibrinogen γ chain. Biochem. Biophys. Res. Commun.107, 181–187 (1982).
  • Vergara-Jimenez J, Tricoci P. Safety and efficacy of abciximab as an adjunct to percutaneous coronary intervention. Vasc. Health. Risk. Manag.6, 39–45 (2010).
  • Lippi G, Montagnana M, Danese E, Favaloro EJ, Franchini M. Glycoprotein IIb/IIIa inhibitors: an update on the mechanism of action and use of functional testing methods to assess anti-platelet efficacy. Biomark. Med.5, 63–70 (2011).
  • Goudevenos JA, Tselepis A. Basic and clinical aspects of platelet integrin αIIbβ3 receptor antagonists in acute coronary syndromes. Curr. Opin. Investig. Drugs2, 244–249 (2001).
  • Beavo JA. Cyclic nucleotide phosphodiesterases: functional implications of multiple isoforms. Physiol. Rev.75, 725–748 (1995).
  • Bender AT, Beavo JA. Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol. Rev.58, 488–520 (2006).
  • Ji X, Hou M. Novel agents for anti-platelet therapy. J. Hematol. Oncol.4, 44 (2011).
  • Born G, Patrono C. Antiplatelet drugs. Br. J. Pharmacol.147(Suppl. 1), S241–S251 (2006).
  • Weyrich AS, Denis MM, Kuhlmann-Eyre JR et al. Dipyridamole selectively inhibits inflammatory gene expression in platelet–monocyte aggregates. Circulation111, 633–642 (2005).
  • Muller TH, Su CA, Weisenberger H, Brickl R, Nehmiz G, Eisert WG. Dipyridamole alone or combined with low-dose acetylsalicylic acid inhibits platelet aggregation in human whole blood ex vivo. Br. J. Clin. Pharmacol.30, 179–186 (1990).
  • Diener HC, Cunha L, Forbes C, Sivenius J, Smets P, Lowenthal A. European Stroke Prevention study. 2. Dipyridamole and acetylsalicylic acid in the secondary prevention of stroke. J. Neurol. Sci.143, 1–13 (1996).
  • Halkes PH, van Gijn J, Kappelle LJ, Koudstaal PJ, Algra A. Aspirin plus dipyridamole versus aspirin alone after cerebral ischaemia of arterial origin (ESPRIT): randomised controlled trial. Lancet367, 1665–1673 (2006).
  • Verro P, Gorelick PB, Nguyen D. Aspirin plus dipyridamole versus aspirin for prevention of vascular events after stroke or TIA: a meta-analysis. Stroke39, 1358–1363 (2008).
  • Sacco RL, Diener HC, Yusuf S et al. Aspirin and extended-release dipyridamole versus clopidogrel for recurrent stroke. N. Engl. J. Med.359, 1238–1251 (2008).
  • Uchiyama S, Demaerschalk BM, Goto S et al. Stroke prevention by cilostazol in patients with atherothrombosis: meta-analysis of placebo-controlled randomized trials. J. Stroke Cerebrovasc. Dis.18, 482–490 (2009).
  • Schror K. The pharmacology of cilostazol. Diabetes Obes. Metab.4(Suppl. 2), S1–S19 (2002).
  • Jeong YH, Hwang JY, Kim IS et al. Adding cilostazol to dual antiplatelet therapy achieves greater platelet inhibition than high maintenance dose clopidogrel in patients with acute myocardial infarction: results of the Adjunctive Cilostazol Versus High Maintenance Dose Clopidogrel in Patients With AMI (ACCEL-AMI) study. Circ. Cardiovasc. Interv.3, 17–26 (2010).
  • Jeong YH, Lee SW, Choi BR et al. Randomized comparison of adjunctive cilostazol versus high maintenance dose clopidogrel in patients with high post-treatment platelet reactivity: results of the ACCEL-RESISTANCE (Adjunctive Cilostazol Versus High Maintenance Dose Clopidogrel in Patients With Clopidogrel Resistance) randomized study. J. Am. Coll. Cardiol.53, 1101–1109 (2009).
  • Kumbhani DJ, Bhatt DL. Secondary prevention of stroke: can we do better than aspirin? Lancet Neurol.9, 942–943 (2010).
  • Tamhane U, Meier P, Chetcuti S et al. Efficacy of cilostazol in reducing restenosis in patients undergoing contemporary stent based PCI: a meta-analysis of randomised controlled trials. EuroIntervention5, 384–393 (2009).
  • Suh JW, Lee SP, Park KW et al. Multicenter randomized trial evaluating the efficacy of cilostazol on ischemic vascular complications after drug-eluting stent implantation for coronary heart disease: results of the CILON-T (Influence of Cilostazol-Based Triple Antiplatelet Therapy on Ischemic Complication After Drug-Eluting Stent Implantation) trial. J. Am. Coll. Cardiol.57, 280–289 (2011).
  • Brummel KE, Paradis SG, Butenas S, Mann KG. Thrombin functions during tissue factor-induced blood coagulation. Blood100, 148–152 (2002).
  • Chackalamannil S. Thrombin receptor antagonists as novel therapeutic targets. Curr. Opin. Drug. Discov. Devel.4(4), 417–427 (2001).
  • Ossovskaya VS, Bunnett NW. Protease-activated receptors: contribution to physiology and disease. Physiol. Rev.84, 579–621 (2004).
  • Coughlin SR. Protease-activated receptors in hemostasis, thrombosis and vascular biology. J. Thromb. Haemost.3, 1800–1814 (2005).
  • Doller D, Chackalamannil S, Czarniecki M, McQuade R, Ruperto V. Design, synthesis, and structure–activity relationship studies of himbacine derived muscarinic receptor antagonists. Bioorg. Med. Chem. Lett.9, 901–906 (1999).
  • Oestreich J. SCH-530348, a thrombin receptor (PAR-1) antagonist for the prevention and treatment of atherothrombosis. Curr. Opin. Investig. Drugs10, 988–996 (2009).
  • Becker RC, Moliterno DJ, Jennings LK et al. Safety and tolerability of SCH 530348 in patients undergoing non-urgent percutaneous coronary intervention: a randomised, double-blind, placebo-controlled Phase II study. Lancet373, 919–928 (2009).
  • Morrow DA, Scirica BM, Fox KA et al. Evaluation of a novel antiplatelet agent for secondary prevention in patients with a history of atherosclerotic disease: design and rationale for the thrombin-receptor antagonist in secondary prevention of atherothrombotic ischemic events (TRA 2 degrees P)-TIMI 50 trial. Am. Heart J.158, 335–341.E3 (2009).
  • Ueno M, Ferreiro JL, Angiolillo DJ. Mechanism of action and clinical development of platelet thrombin receptor antagonists. Expert Rev. Cardiovasc. Ther.8(8), 1191–1200 (2010).
  • Chintala M, Shimizu K, Ogawa M, Yamaguchi H, Doi M, Jensen P. Basic and translational research on proteinase-activated receptors: antagonism of the proteinase-activated receptor 1 for thrombin, a novel approach to antiplatelet therapy for atherothrombotic disease. J. Pharmacol. Sci.108, 433–438 (2008).
  • White HD. Oral antiplatelet therapy for atherothrombotic disease: current evidence and new directions. Am. Heart J.161, 450–461 (2011).
  • Goto S, Yamaguchi T, Ikeda Y, Kato K, Yamaguchi H, Jensen P. Safety and exploratory efficacy of the novel thrombin receptor (PAR-1) antagonist SCH530348 for non-ST-segment elevation acute coronary syndrome. J. Atheroscler. Thromb.17, 156–164 (2010).
  • TRA*CER Executive and Steering Committees. The thrombin receptor antagonist for clinical event reduction in acute coronary syndrome (TRA*CER) trial: study design and rationale. Am. Heart J.158, 327–334.e4 (2009).
  • Tricoci P, Huang Z, Held C et al.; TRACER investigators. Thrombin-receptor antagonist vorapaxar in acute coronary syndromes. N. Engl. J. Med.366, 20–33 (2012).
  • Martorell L, Martinez-Gonzalez J, Rodriguez C, Gentile M, Calvayrac O, Badimon L. Thrombin and protease-activated receptors (PARs) in atherothrombosis. Thromb. Haemost.99, 305–315 (2008).
  • Kai Y, Hirano K, Maeda Y, Nishimura J, Sasaki T, Kanaide H. Prevention of the hypercontractile response to thrombin by proteinase-activated receptor-1 antagonist in subarachnoid hemorrhage. Stroke38, 3259–3265 (2007).
  • Tello-Montoliu A, Tomasello SD, Ueno M, Angiolillo DJ. Antiplatelet therapy: thrombin receptor antagonists. Br. J. Clin. Pharmacol.72, 658–671 (2011).
  • Wiviott SD, Flather MD, O’Donoghue ML et al. Randomized trial of atopaxar in the treatment of patients with coronary artery disease: the lessons from antagonizing the cellular effect of thrombin-coronary artery disease trial. Circulation123, 1854–1863 (2011).
  • Goto S, Ogawa H, Takeuchi M, Flather MD, Bhatt DL. Double-blind, placebo-controlled Phase II studies of the protease-activated receptor 1 antagonist E5555 (atopaxar) in Japanese patients with acute coronary syndrome or high-risk coronary artery disease. Eur. Heart J.31, 2601–2613 (2010).
  • O’Donoghue ML, Bhatt DL, Wiviott SD et al. Safety and tolerability of atopaxar in the treatment of patients with acute coronary syndromes: the lessons from antagonizing the cellular effects of thrombin-acute coronary syndromes trial. Circulation123, 1843–1853 (2011).
  • Giannarelli C, Zafar M, Badimon JJ. Prostanoid and TP-receptors in atherothrombosis: is there a role for their antagonism? Thromb. Haemost.104, 949–954 (2010).
  • Chamorro A. TP receptor antagonism: a new concept in atherothrombosis and stroke prevention. Cerebrovasc. Dis.27(Suppl. 3), 20–27 (2009).
  • Smith EM. Thromboxane and the thromboxane receptor in cardiovascular disease. Clin. Lipidol.5, 209–219 (2010).
  • Hong TT, Huang J, Driscoll E, Lucchesi BR. Preclinical evaluation of S18886 in an experimental model of coronary arterial thrombosis. J. Cardiovasc. Pharmacol.48, 239–248 (2006).
  • Vilahur G, Casani L, Badimon L. A thromboxane A2/prostaglandin H2 receptor antagonist (S18886) shows high antithrombotic efficacy in an experimental model of stent-induced thrombosis. Thromb. Haemost.98, 662–669 (2007).
  • Gaussem P, Reny JL, Thalamas C et al. The specific thromboxane receptor antagonist S18886: pharmacokinetic and pharmacodynamic studies. J. Thromb. Haemost.3, 1437–1345 (2005).
  • Bousser MG, Amarenco P, Chamorro A et al. Rationale and design of a randomized, double-blind, parallel-group study of terutroban 30 mg/day versus aspirin 100 mg/day in stroke patients: the prevention of cerebrovascular and cardiovascular events of ischemic origin with terutroban in patients with a history of ischemic stroke or transient ischemic attack (PERFORM) study. Cerebrovasc. Dis.27, 509–518 (2009).
  • Spiel AO, Gilbert JC, Jilma B. Von Willebrandt factor in cardiovascular disease: focus on acute coronary syndromes. Circulation117(11), 1449–1459 (2008).
  • Sarratt KL, Chen H, Zutter MM, Santoro SA, Hammer DA, Kahn ML. GPVI and α2β1 play independent critical roles during platelet adhesion and aggregate formation to collagen under flow. Blood106, 1268–1277 (2005).
  • Kageyama S, Yamamoto H, Nakazawa H et al. Pharmacokinetics and pharmacodynamics of AJW200, a humanized monoclonal antibody to von Willebrand factor, in monkeys. Arterioscler. Thromb.22, 187–192 (2002).
  • Vanhoorelbeke K, Depraetere H, Romijn RA, Huizinga EG, De Maeyer M, Deckmyn H. A consensus tetrapeptide selected by phage display adopts the conformation of a dominant discontinuous epitope of a monoclonal anti-vWF antibody that inhibits the von Willebrand factor-collagen interaction. J. Biol. Chem.278, 37815–37821 (2002).
  • Ulrichts H, Schoolmeester A, Hoefman S et al. Anti-thrombotic drug candidate ALX-0081 shows superior preclinical efficacy and safety compared with current marketed antiplatelet drugs. J. Thromb. Haemost. Suppl.118(3), 757–765 (2009).
  • Abd-Elaziz K, Kamphuisen P, Lyssesns C et al. Safety, tolerability, pharmacokinetics and pharmacodynamics of anti-vWF nanobody ALX-0681 after single and multiple subcutaneous administrations to healthy volunteers. Presented at: 51st ASH Annual Meeting and Exposition. New Orleans, LA, USA, 5–8 December 2009.
  • Gilbert JC, DeFeo-Fraulini T, Hutabarat RM et al. First-in-human evaluation of anti von Willebrand factor therapeutic aptamer ARC1779 in healthy volunteers. Circulation116, 2678–2686 (2007).
  • Bouchard PR, Hutabarat R, Thompson KM. Discovery and development of therapeutic aptamers. Annu. Rev. Pharmacol. Toxicol.50, 237–257 (2010).
  • Diener JL, Daniel Lagasse H, Duerschmied D et al. Inhibition of von Willebrand factor-mediated platelet activation and thrombosis by the anti-von Willebrand factor A1-domain aptamer ARC1779. J. Thromb. Haemost.7, 1155–1162 (2009).
  • Cauwenberghs N, Vanhoorelbeke K, Vauterin S et al. Epitope mapping of inhibitory antibodies against platelet glycoprotein Iba reveals interaction between the leucine-rich repeat N-terminal and C-terminal flanking domains of glycoprotein Iba. Blood98, 652–660 (2001).
  • Hennan JK, Swillo R, Morgan GA et al. Pharmacologic inhibition of platelet vWF–GPIb a interaction prevents coronary artery thrombosis. Thromb. Haemost.95, 469–475 (2006).
  • Yang J, Ji S, Dong N, Zhao Y, Ruan C. Engineering and characterization of a chimeric anti-platelet glycoprotein Iba monoclonal antibody and preparation of its Fab fragment. Hybridoma (Larchmt)29, 125–132 (2010).
  • Best D, Senis YA, Jarvis GE et al. GPVI levels in platelets: relationship to platelet function at high shear. Blood102, 2811–2818 (2003).
  • Li H, Lockyer S, Concepcion A et al. The Fab fragment of a novel anti-GPVI monoclonal antibody, OM4, reduces in vivo thrombosis without bleeding risk in rats. Arterioscler. Thromb. Vasc. Biol.27, 1199–1205 (2007).
  • Qian MD, Villeval JL, Xiong X et al. Anti GPVI human antibodies neutralizing collagen-induced platelet aggregation isolated from a combinatorial phage display library. Hum. Antibodies11, 97–105 (2002).
  • Smethurst PA, Joutsi-Korhonen L, O’Connor MN et al. Identification of the primary collagen-binding surface on human glycoprotein VI by site-directed mutagenesis and by a blocking phage antibody. Blood103, 903–911 (2004).
  • Gruner S, Prostredna M, Aktas B et al. Anti-glycoprotein VI treatment severely compromises hemostasis in mice with reduced α2β1 levels or concomitant aspirin therapy. Circulation110, 2946–2951 (2004).
  • Morrow DA, Braunwald E, Bonaca MP et al. Vorapaxar in the secondary prevention of atherothrombotic events. N. Engl. J. Med.366(15), 1404–1413 (2012).

Website

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.