Abstract

HomeCirculationVol. 109, No. 25Clopidogrel Resistance Free AccessArticle CommentaryPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessArticle CommentaryPDF/EPUBClopidogrel ResistanceA New Chapter in a Fast-Moving Story Stephen D. Wiviott, MD and Elliott M. Antman, MD Stephen D. WiviottStephen D. Wiviott From the TIMI Study Group of the Cardiovascular Division, Brigham and Women’s Hospital, Boston, Mass. Search for more papers by this author and Elliott M. AntmanElliott M. Antman From the TIMI Study Group of the Cardiovascular Division, Brigham and Women’s Hospital, Boston, Mass. Search for more papers by this author Originally published29 Jun 2004https://doi.org/10.1161/01.CIR.0000134701.40946.30Circulation. 2004;109:3064–3067Although platelets lack nuclei and are the smallest circulating human cells, they play an integral and complex role in the process of thrombosis, both physiological and pathophysiological. Activation and aggregation of platelets play a central role in the propagation of intracoronary thrombi after (1) spontaneous atherosclerotic plaque disruption that results in myocardial ischemia or infarction in the acute coronary syndromes (ACS), or (2) the mechanical disruption that results from percutaneous coronary intervention (PCI). Platelets initially adhere to collagen and von Willebrand factor at the site of the disrupted plaque, resulting in an initial platelet monolayer. After activation, platelets release secondary agonists such as thromboxane A2 and adenosine diphosphate (ADP), which in combination with thrombin generated by the coagulation cascade result in stimulation and recruitment of additional platelets.1,2 With this pathophysiological background, it is not surprising that antiplatelet therapy is a cornerstone of the management of patients with ACS, especially those undergoing PCI.3–5See p 3171Antiplatelet AgentsAspirin inhibits cyclooxygenase (COX) by irreversible acetylation, which prevents the production of thromboxane A2. The antithrombotic effect of aspirin results from the decreased production of this prothrombotic, vasoconstrictive substance. Aspirin is effective in the short- and long-term prevention of adverse vascular events in high-risk patient groups, including those with ACS, stroke and peripheral arterial disease.6 Aspirin also has been shown to reduce the frequency of ischemic complications after PCI.7,8Despite the impressive and consistent effects of aspirin in reducing adverse events in a variety of ischemic heart disease states, a significant rate of such events persists, and more potent antiplatelet agents, glycoprotein IIb/IIIa inhibitors, and thienopyridines have been developed. The thienopyridines irreversibly inhibit ADP binding to the P2Y12 receptor on the platelet surface. By blocking this receptor, these agents interfere with platelet activation, degranulation, and—by inhibiting the modification of the glycoprotein IIb/IIIa receptor—aggregation. Currently available thienopyridine antiplatelet agents include ticlopidine and clopidogrel. Both agents are rapidly absorbed prodrugs that are modified hepatically to active metabolites.2 The agents have similar platelet effects and have been shown to be clinically efficacious. However, clopidogrel has largely replaced ticlopidine because of an improved safety profile, with a lower incidence of hematologic complications (neutropenia and pancytopenia) than ticlopidine.9 The effects of clopidogrel are time and dose dependent, with a ceiling effect at approximately 50% to 60% inhibition of platelet aggregation.2 Loading doses of clopidogrel of 300 to 600 mg reach near steady-state levels of platelet aggregation by 4 to 24 hours, whereas daily maintenance dosing with 75 mg daily without a preload results in steady-state levels within 4 to 7 days.2,10,11Clopidogrel has been compared with aspirin in the Clopidogrel versus Aspirin in Patients at Risk of Ischemic Events (CAPRIE) trial12 in patients with vascular disease (stroke, myocardial infarction, or peripheral arterial disease) and was found to produce a relative reduction in vascular events by 8.7%. Clopidogrel has been tested in combination with aspirin (versus aspirin alone) in the Clopidogrel in Unstable angina Recurrent Events (CURE) trial13 in patients with unstable angina/non–ST-segment–elevation myocardial infarction (UA/NSTEMI). Significant reductions in clinical outcomes were seen as early as 24 hours after randomization and persisted to study completion (average of 9-month follow-up) and were seen across multiple treatment and risk groups.13 Studies of thienopyridines as a component of acute treatment for STEMI are ongoing.The Clopidogrel for the Reduction of Events During Observation (CREDO) trial14 examined the effects of a loading dose (300 mg) of clopidogrel before PCI followed by maintenance dosing versus maintenance alone and found a significant reduction of early events only when pretreatment was given >6 hours before PCI. A subsequent analysis of the CREDO trial suggests the minimal efficacious time interval may be on the order of 12 to 15 hours.15 Data from a modest-sized trial examining high-dose (600-mg) clopidogrel pretreatment in the setting of low-risk PCI suggest there is no incremental advantage of adding an intravenous glycoprotein IIb/IIIa inhibitor in reducing peri-PCI ischemic events.16Resistance to Antiplatelet AgentsAlthough antiplatelet agents reduce ischemic events, “resistance” to their effects continues to occur. The term resistance in this setting is problematic in that it has been variably used to indicate failure of an agent to prevent the clinical condition for which it is intended or failure of the agent to achieve the biochemical (pharmacokinetic and/or pharmacodynamic) effect. Because of the complex pathophysiology of ischemic heart disease, involving thrombosis, inflammation, vascular biology, hemodynamics, etc, no single agent can be expected to abolish ischemic events. Furthermore, a patient may have the appropriate platelet response to a given therapy but have recurrent events mediated by nonplatelet factors. For these reasons, it would be reasonable to classify patients who have recurrent events on therapy as having failure of therapy, while limiting the term resistance to those patients for whom the agent does not achieve its pharmacological effect. The key clinical question is what role resistance to an agent plays in failure of therapy.To identify the failure to achieve a pharmacological effect, one must be able to measure it reliably. Several assays are available to measure platelet function and effects of antiplatelet agents.17 A commonly used test of platelet function measures platelet aggregation by light transmittance (optical aggregometry) in platelet-rich plasma in response to an agonist (arachidonic acid, ADP, collagen, epinephrine, or a thrombin receptor–activating peptide).17 This mechanism allows monitoring of different drug effects by allowing choice of agonist (eg, ADP for thienopyridines). Because of inter- and intra-patient variability, standardized responses are not meaningful, and results are often reported as a percentage of a baseline value. Other methods include the cone and plate(let) analyzer,18 a rapid test that measures whole blood platelet aggregation under conditions of high shear stress.The presence of aspirin treatment failures in several disease states led to the concern that selected patients may be resistant to the effects of aspirin. Prevalence of resistance varied by disease condition and platelet function methodology.19 There is evidence of a link between aspirin resistance and clinical events. In patients with a prior stroke, those with aspirin resistance were 89% more likely to have a recurrent cerebrovascular accident within 2 years than were responders.20,21 Similar results were seen in patients after peripheral intervention, with an increase in arterial reocclusion among aspirin responders.22 A case-control study from the Heart Outcomes Prevention Evaluation (HOPE) study measured urinary 11-dehydro thromboxane B2 (a stable arachidonic acid metabolite produced by activated platelets). They found that among patients treated with aspirin, those with the highest levels (suggesting incomplete thromboxane inhibition) had a 3.5-fold increase in cardiovascular mortality and a doubling of myocardial infarction.23 The definitive cause(s) of aspirin resistance are not known; however, several possible mechanisms have been proposed and include extrinsic factors (eg, cigarette smoking, drug–drug interactions, inadequate aspirin dosing) and intrinsic factors (eg, inducible COX-2 not inhibited by aspirin, variation in COX-1 structure preventing acetylation, thromboxane production by nonplatelet cells).19More recently, a similar story of variable platelet response and potential resistance to therapy has emerged with thienopyridines. Analogous to aspirin resistance, there is no clear and accepted definition for clopidogrel resistance. Studies have shown a dose- and time-dependent variability in response to clopidogrel as measured by optical platelet aggregometry in response to ADP.10,11,24,25 In a study by Gurbel et al,10 96 patients undergoing elective coronary stenting were monitored before and at multiple time points after standard clopidogrel therapy (300-mg loading dose followed by 75 mg daily). Clopidogrel resistance, empirically defined as <10% reduction in aggregation in response to 5 μmol/L ADP compared with pretreatment values, was seen in 63% of patients at 2 hours, 31% at 24 hours, 31% at 5 days, and 15% at 30 days.10 Patients with the highest pretreatment values had the least antithrombotic protection over the first 5 days.10 In another report, Muller et al11 defined nonresponders as those with <10% reduction in platelet aggregation to ADP and semiresponders as those with 10% to 29% reduction 4 hours after 600-mg clopidogrel load,11 as no additional effect was seen with this treatment regimen at 24 hours. This study found that to 5 μmol/L ADP, 5% were nonresponders and 9% were semiresponders, and to 20 μmol/L ADP, 11% were nonresponders and 26% were semiresponders.11 Although not designed to evaluate clinical outcomes, an intriguing finding in the Muller study was that 2 patients (of 105 tested) developed subacute stent thrombosis, and both met the definition of clopidogrel nonresponse. An additional report correlated anginal class to platelet inhibition and found that patients with higher anginal class on presentation had less inhibition of platelet aggregation after loading with 450 mg of ADP.26Several mechanisms of clopidogrel resistance are possible. Extrinsic mechanisms include inappropriate dosing or underdosing of clopidogrel and drug–drug interactions, including a possible interaction between clopidogrel and atorvastatin.27,28 There is a positive correlation of clopidogrel response with CYP3A4 activity (measured by erythromycin breath test),29 suggesting that an important mechanism may be variable conversion to the active metabolite. Other potential extrinsic mechanisms could include variable absorption of the prodrug or clearance of the active metabolite. Intrinsic mechanisms could include P2Y12 receptor variability, increase in number of receptors, increased release of ADP, or upregulation of other platelet activation pathways. In contrast to aspirin resistance, there has not previously been a link between clopidogrel resistance as measured by platelet assays and clinical adverse events.In the present issue of Circulation, Matetzky and colleagues30 add a new and important piece to the emerging clopidogrel resistance picture: correlation of a laboratory measure of clopidogrel nonresponse with clinical outcomes. Patients who underwent primary PCI (n=60) with stenting and 10 patients who underwent primary angioplasty for STEMI received 300 mg aspirin on admission and eptifibatide and heparin during PCI. Those who received stents were treated with clopidogrel: 300 mg immediately after PCI and 75 mg daily for 3 months. Platelet function tests were performed with turbidometric analysis after stimulation with ADP (5 μmol/L) and epinephrine (10 μmol/L), as well as separate assays of platelet function using a cone and plate(let) analyzer.17 Patients were divided into quartiles of inhibition of platelet aggregation (platelet aggregation compared with baseline platelet aggregation), with the first quartile being considered nonresponders (day 6 aggregation 103±8% compared with baseline). Patients in quartiles 2 through 4 had varying levels of response, with platelet aggregation of 69%, 58%, and 33% of baseline values. During 6-month follow-up, 7 patients (40%) in quartile 1 (nonresponders) had 8 clinical events, including stent thrombosis, myocardial infarction, recurrent ACS, and peripheral arterial occlusion. One patient in the second quartile (6.7%) and no patients in quartiles 3 or 4 had recurrent events (P trend=0.007). Although the study population was small, these data strongly suggest that there is individual variability in response to clopidogrel in the setting of PCI after STEMI and more broadly that clopidogrel resistance may be a marker for increased risk of recurrent cardiovascular events.Several questions arise from the growing literature on clopidogrel resistance. Should patients with ACS or those undergoing PCI routinely have platelet function measured? If so, how should it be measured? What should be considered the appropriate definition of clopidogrel resistance? What therapeutic maneuvers should clinicians undertake when they encounter a patient with clopidogrel resistance? Are there actions that can be taken prospectively to avoid the problem of resistance?Before the first question is answered in the affirmative, the observations by Matetzky et al30 need to be reproduced in larger datasets. Before these measurements become clinically useful for risk stratification, there should be easily performed and reproducible ways to measure platelet aggregation, with standardized definitions of response that correlate with clinical outcomes. Finally, to allow these observations to improve the care of patients, therapies must be found that can overcome the resistance to platelet aggregation inhibition. One approach to managing clopidogrel resistance may involve giving higher loading and maintenance doses. Another promising approach may be to utilize alternative thienopyridine agents such as CS-747 (LY640315),31 nonthienopyridine P2Y12 inhibitors such as AR-C69931MX,2,32 or antagonists of other platelet targets. As we learn more about the variable response to antiplatelet drugs, will the time soon come for us to think of antiplatelet agents like antibiotics, tailoring therapy when resistance is observed in the laboratory?The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.FootnotesCorrespondence to Stephen D. Wiviott, MD, The TIMI Study Group, Cardiovascular Division, Brigham and Women’s Hospital, 75 Francis St, Boston, MA 02115. E-mail [email protected] References 1 McNicol A, Israels SJ. Platelets and anti-platelet therapy. J Pharmacol Sci. 2003; 93: 381–396.CrossrefMedlineGoogle Scholar2 Patrono C, Bachmann F, Baigent C, et al. Expert consensus document on the use of antiplatelet agents. The Task Force on the Use of Antiplatelet Agents in Patients with Atherosclerotic Cardiovascular Disease of the European Society of Cardiology. Eur Heart J. 2004; 25: 166–181.CrossrefMedlineGoogle Scholar3 Antman EM, Anbe DT, Armstrong PW, et al. ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction: executive summary: a report of the ACC/AHA Task Force on Practice Guidelines (Committee to Revise the 1999 Guidelines on the Management of Patients With Acute Myocardial Infarction). Circulation. In press.Google Scholar4 Braunwald E, Antman EM, Beasley JW, et al. ACC/AHA guideline update for the management of patients with unstable angina and non–ST-segment elevation myocardial infarction—2002: summary article: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Committee on the Management of Patients With Unstable Angina). Circulation. 2002; 106: 1893–1900.LinkGoogle Scholar5 Smith SC Jr, Dove JT, Jacobs AK, et al. ACC/AHA guidelines for percutaneous coronary intervention (revision of the 1993 PTCA guidelines)—executive summary: a report of the American College of Cardiology/American Heart Association task force on practice guidelines (Committee to Revise the 1993 Guidelines for Percutaneous Transluminal Coronary Angioplasty) endorsed by the Society for Cardiac Angiography and Interventions. Circulation. 2001; 103: 3019–3041.CrossrefMedlineGoogle Scholar6 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.CrossrefMedlineGoogle Scholar7 Savage MP, Goldberg S, Bove AA, et al. Effect of thromboxane A2 blockade on clinical outcome and restenosis after successful coronary angioplasty. Multi-Hospital Eastern Atlantic Restenosis Trial (M-HEART II). Circulation. 1995; 92: 3194–3200.CrossrefMedlineGoogle Scholar8 Schwartz L, Bourassa MG, Lesperance J, et al. Aspirin and dipyridamole in the prevention of restenosis after percutaneous transluminal coronary angioplasty. N Engl J Med. 1988; 318: 1714–1719.CrossrefMedlineGoogle Scholar9 Elias M, Reichman N, Flatau E. Bone marrow aplasia associated with ticlopidine therapy. Am J Hematol. 1993; 44: 289–290.Google Scholar10 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.LinkGoogle Scholar11 Muller I, Besta F, Schulz C, et al. Prevalence of clopidogrel non-responders among patients with stable angina pectoris scheduled for elective coronary stent placement. Thromb Haemost. 2003; 89: 783–787.CrossrefMedlineGoogle Scholar12 CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet. 1996; 348: 1329–1339.CrossrefMedlineGoogle Scholar13 Yusuf S, Zhao F, Mehta SR, et al. 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.CrossrefMedlineGoogle Scholar14 Steinhubl SR, Berger PB, Mann JT 3rd, et al. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. JAMA. 2002; 288: 2411–2420.CrossrefMedlineGoogle Scholar15 Steinhubl SR, Darrah S, Brennan D, et al. Optimal duration of pretreatment with clopidogrel prior to PCI: data from the CREDO trial. Circulation. 2003; 108: IV-374.Google Scholar16 Kastrati A, Mehilli J, Schuhlen H, et al. A clinical trial of abciximab in elective percutaneous coronary intervention after pretreatment with clopidogrel. N Engl J Med. 2004; 350: 232–238.CrossrefMedlineGoogle Scholar17 Rand ML, Leung R, Packham MA. Platelet function assays. Transfus Apheresis Sci. 2003; 28: 307–317.CrossrefMedlineGoogle Scholar18 Varon D, Lashevski I, Brenner B, et al. Cone and plate(let) analyzer: monitoring glycoprotein IIb/IIIa antagonists and von Willebrand disease replacement therapy by testing platelet deposition under flow conditions. Am Heart J. 1998; 135: S187–S193.CrossrefMedlineGoogle Scholar19 McKee SA, Sane DC, Deliargyris EN. Aspirin resistance in cardiovascular disease: a review of prevalence, mechanisms, and clinical significance. Thromb Haemost. 2002; 88: 711–715.CrossrefMedlineGoogle Scholar20 Grotemeyer KH. Effects of acetylsalicylic acid in stroke patients: evidence of nonresponders in a subpopulation of treated patients. Thromb Res. 1991; 63: 587–593.CrossrefMedlineGoogle Scholar21 Grotemeyer KH, Scharafinski HW, Husstedt IW. Two-year follow-up of aspirin responder and aspirin non responder: a pilot-study including 180 post-stroke patients. Thromb Res. 1993; 71: 397–403.CrossrefMedlineGoogle Scholar22 Mueller MR, Salat A, Stangl P, et al. Variable platelet response to low-dose ASA and the risk of limb deterioration in patients submitted to peripheral arterial angioplasty. Thromb Haemost. 1997; 78: 1003–1007.CrossrefMedlineGoogle Scholar23 Eikelboom JW, Hirsh J, Weitz JI, et al. Aspirin-resistant thromboxane biosynthesis and the risk of myocardial infarction, stroke, or cardiovascular death in patients at high risk for cardiovascular events. Circulation. 2002; 105: 1650–1655.LinkGoogle Scholar24 Jaremo P, Lindahl TL, Fransson SG, et al. Individual variations of platelet inhibition after loading doses of clopidogrel. J Intern Med. 2002; 252: 233–238.CrossrefMedlineGoogle Scholar25 Gurbel PA, Bliden KP. Durability of platelet inhibition by clopidogrel. Am J Cardiol. 2003; 91: 1123–1125.CrossrefMedlineGoogle Scholar26 Soffer D, Moussa I, Harjai KJ, et al. Impact of angina class on inhibition of platelet aggregation following clopidogrel loading in patients undergoing coronary intervention: do we need more aggressive dosing regimens in unstable angina? Catheter Cardiovasc Interv. 2003; 59: 21–25.CrossrefMedlineGoogle Scholar27 Saw J, Steinhubl SR, Berger PB, et al. Lack of adverse clopidogrel-atorvastatin clinical interaction from secondary analysis of a randomized, placebo-controlled clopidogrel trial. Circulation. 2003; 108: 921–924.LinkGoogle Scholar28 Lau WC, Waskell LA, Watkins PB, et al. Atorvastatin reduces the ability of clopidogrel to inhibit platelet aggregation: a new drug–drug interaction. Circulation. 2003; 107: 32–37.LinkGoogle Scholar29 Lau WC, Gurbel PA, Watkins PB, et al. Contribution of hepatic cytochrome P450 3A4 metabolic activity to the phenomenon of clopidogrel resistance. Circulation. 2004; 109: 166–171.LinkGoogle Scholar30 Matetzky S, Shenkman B, Guetta V, et al. Clopidogrel resistance is associated with increased risk of recurrent atherothrombotic events in patients with acute myocardial infarction. Circulation. 2004; 109: 3171–3175.LinkGoogle Scholar31 Sugidachi A, Asai F, Ogawa T, et al. The in vivo pharmacological profile of CS-747, a novel antiplatelet agent with platelet ADP receptor antagonist properties. Br J Pharmacol. 2000; 129: 1439–1446.CrossrefMedlineGoogle Scholar32 Storey F. The P2Y12 receptor as a therapeutic target in cardiovascular disease. Platelets. 2001; 12: 197–209.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Tam D and Fremes S (2022) Commentary: Microvesicles, personalized surgery, and tailored medical therapy to improve coronary artery bypass grafting outcomes, The Journal of Thoracic and Cardiovascular Surgery, 10.1016/j.jtcvs.2020.11.142, 163:2, (701-702), Online publication date: 1-Feb-2022. van Stijn D, Schoenmaker N, Planken R, Koolbergen D, Gouw S, Kuijpers T, Blom N and Kuipers I (2022) Myocardial infarction due to thrombotic occlusion despite anticoagulation in Kawasaki disease – a case report, BMC Pediatrics, 10.1186/s12887-022-03151-2, 22:1, Online publication date: 1-Dec-2022. Farrokh S, Owusu K, Lara L, Nault K, Hui F and Spoelhof B (2019) Neuro-Interventional Use of Oral Antiplatelets: A Survey of Neuro-Endovascular Centers in the United States and Review of the Literature, Journal of Pharmacy Practice, 10.1177/0897190019854868, 34:2, (207-215), Online publication date: 1-Apr-2021. Zheng N, Yin F, Yu Q, Zhong J, Yang J, Xu Z, Su J and Chen X (2021) Associations of PER3 polymorphisms with clopidogrel resistance among Chinese Han people treated with clopidogrel , Journal of Clinical Laboratory Analysis, 10.1002/jcla.23713, 35:4, Online publication date: 1-Apr-2021. Jafari M, Nguyen T, Ortega-Gutierrez S, Hussain M, Hassan A, Ikram A, Eliyas J, Rodriguez G and Divani A (2021) Current Advances in Endovascular Treatment of Intracranial Atherosclerotic Disease and Future Prospective, Journal of Stroke and Cerebrovascular Diseases, 10.1016/j.jstrokecerebrovasdis.2020.105556, 30:3, (105556), Online publication date: 1-Mar-2021. Li C, Jia W, Li J, Li F, Ma J and Zhou L (2021) Association with CYP2C19 polymorphisms and Clopidogrel in treatment of elderly stroke patients, BMC Neurology, 10.1186/s12883-021-02127-6, 21:1, Online publication date: 1-Dec-2021. Zhang L, Li X, Wang D, Lv H, Si X, Li X, Sun Y, Wang D, Chen K, Kang X, Lou X, Zhang G and Ma N Risk Factors of Recurrent Ischemic Events after Acute Noncardiogenic Ischemic Stroke, Current Pharmaceutical Design, 10.2174/1381612825666191029103756, 25:45, (4827-4834) Zhu Y, Gao M, Zhou T, Xie M, Mao A, Feng L, Yao X, Wong W and Ma X (2019) The TRPC5 channel regulates angiogenesis and promotes recovery from ischemic injury in mice, Journal of Biological Chemistry, 10.1074/jbc.RA118.005392, 294:1, (28-37), Online publication date: 1-Jan-2019. Aguilar-Salinas P, Brasiliense L, Lima J, Aghaebrahim A, Sauvageau E and Hanel R (2019) Complex Stent Reconstruction for the Treatment of Intracranial Aneurysms Management of Cerebrovascular Disorders, 10.1007/978-3-319-99016-3_13, (207-232), . Wang D, Yang X, Zhang J, Li R, Jia M and Cui X (2018) Compared efficacy of clopidogrel and ticagrelor in treating acute coronary syndrome: a meta-analysis, BMC Cardiovascular Disorders, 10.1186/s12872-018-0948-4, 18:1, Online publication date: 1-Dec-2018. Divanji P and Shunk K Modern Antiplatelet Therapy: When Is Clopidogrel the Right Choice?, Cardiovascular Innovations and Applications, 10.15212/CVIA.2017.0049, 3:2, (175-202) Kitazono T, Ikeda Y, Nishikawa M, Yoshiba S, Abe K and Ogawa A (2018) Influence of cytochrome P450 polymorphisms on the antiplatelet effects of prasugrel in patients with non-cardioembolic stroke previously treated with clopidogrel, Journal of Thrombosis and Thrombolysis, 10.1007/s11239-018-1714-2, 46:4, (488-495), Online publication date: 1-Nov-2018. Wang B, Li X, Ma N, Mo D, Gao F, Sun X, Xu X, Liu L, Song L, Li X, Zhao Z, Zhao X and Miao Z (2015) Association of thrombelastographic parameters with post-stenting ischemic events, Journal of NeuroInterventional Surgery, 10.1136/neurintsurg-2015-011687, 9:2, (192-195), Online publication date: 1-Feb-2017. Tan S, Fong A, Mejin M, Gerunsin J, Kong K, Chin F, Tiong L, Lim M, Said A, Khiew N, Voon C, Mohd Amin N, Cham Y, Koh K, Oon Y and Ong T (2017) Association of CYP2C19*2 polymorphism with clopidogrel response and 1-year major adverse cardiovascular events in a multiethnic population with drug-eluting stents , Pharmacogenomics, 10.2217/pgs-2017-0078, 18:13, (1225-1239), Online publication date: 1-Aug-2017. Ramachandran R, Mihara K, Thibeault P, Vanderboor C, Petri B, Saifeddine M, Bouvier M and Hollenberg M (2017) Targeting a Proteinase-Activated Receptor 4 (PAR4) Carboxyl Terminal Motif to Regulate Platelet Function, Molecular Pharmacology, 10.1124/mol.116.106526, 91:4, (287-295), Online publication date: 1-Apr-2017. Lettino M, Leonardi S, De Maria E and Halvorsen S (2017) Antiplatelet and antithrombotic treatment for secondary prevention in ischaemic heart disease, European Journal of Preventive Cardiology, 10.1177/2047487317707854, 24:3_suppl, (61-70), Online publication date: 1-Jun-2017. Chhonker Y, Pandey C, Chandasana H, Laxman T, Prasad Y, Narain V, Dikshit M and Bhatta R (2015) Simultaneous quantitation of acetylsalicylic acid and clopidogrel along with their metabolites in human plasma using liquid chromatography tandem mass spectrometry, Biomedical Chromatography, 10.1002/bmc.3573, 30:3, (466-473), Online publication date: 1-Mar-2016. Dalal J, Digrajkar A and Gandhi A (2016) Oral antiplatelet therapy and platelet inhibition: An experience from a tertiary care center, Indian Heart Journal, 10.1016/j.ihj.2015.12.022, 68:5, (624-631), Online publication date: 1-Sep-2016. Marcone S, Dervin F and Fitzgerald D (2015) Proteomic signatures of antiplatelet drugs: new approaches to exploring drug effects, Journal of Thrombosis and Haemostasis, 10.1111/jth.12943, 13, (S323-S331), Online publication date: 1-Jun-2015. Holinstat M and Reheman A (2015) Dual antiplatelet therapy for PCI: Are we tailored to all?, Thrombosis Research, 10.1016/j.thromres.2015.04.008, 135:6, (1045-1046), Online publication date: 1-Jun-2015. Yetgin T, van der Linden M, de Vries A, Smits P, Boersma E, van Geuns R and Zijlstra F (2013) Adoption of prasugrel into routine practice: rationale and design of the Rijnmond Collective Cardiology Research (CCR) study in percutaneous coronary intervention for acute coronary syndromes, Netherlands Heart Journal, 10.1007/s12471-013-0472-1, 22:2, (55-61), Online publication date: 1-Feb-2014. Akturk I, Caglar F, Erturk M, Tuncer N, Yalcın A, Surgit O, Uzun F and Caglar I (2013) Hypertension as a Risk Factor for Aspirin and Clopidogrel Resistance in Patients With Stable Coronary Artery Disease, Clinical and Applied Thrombosis/Hemostasis, 10.1177/1076029613481102, 20:7, (749-754), Online publication date: 1-Oct-2014. Louca J, Mina G, Habib B and Sadek S (2014) The effect of doubling the dose of clopidogrel on platelet aggregation in patients with clopidogrel resistance, The Egyptian Heart Journal, 10.1016/j.ehj.2013.08.009, 66:3, (259-262), Online publication date: 1-Sep-2014. Ge C, Yuan F, Feng L, Lv S, Liu H, Song X, Chen X and Huo Y (2013) C

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call