9-Updates in Antiplatelet Agents 2013 RCT

Embed Size (px)

Citation preview

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    1/12

    Cardiovascular Pharmacology Core Review

    Updates in Antiplatelet AgentsUsed in Cardiovascular Diseases

    Judy W. M. Cheng, BS, PharmD, MPH, FCCP, BCPS1

    Abstract

    Background: Antiplatelet therapy is a cornerstone in coronary artery disease management. However, patients with acutecoronary syndrome still remain at risk of recurrent cardiovascular events despite the advance of medical therapy.Objective:This article provides a review of antiplatelet agents used in cardiovascular diseases and focus on updates in the past 5 years.Method: Peer-reviewed clinical trials and relevant treatment guidelines were identified from MEDLINE and Current Contentdatabase (from 1966 to April 15, 2013) using search terms aspirin, clopidogrel, prasugrel, ticagrelor, glycoprotein IIb/IIIa inhibitors,antiplatelet agents, coronary artery disease, acute coronary syndrome, pharmacology, pharmacokinetics, and pharmacodynamics.Citations from the available articles were also reviewed for additional references. Results: In unstable angina and non-ST-

    segment elevation myocardial infarction (MI), dual antiplatelet therapy (aspirin and clopidogrel) demonstrated a reduction in deathfrom cardiovascular causes, nonfatal MI, or stroke (relative risk 0.80; 95% confidence interval [CI], 0.72-0.90). In ST-segment ele-vation MI, dual antiplatelet therapy reduced the rate of occluded infarct-related artery/death or recurrent MI (95% CI, 24%-47%).Newer agents such as prasugrel, when compared to clopidogrel, reduced death from vascular causes, MI, or stroke in patientsundergoing percutaneous coronary intervention (PCI; hazard ratio [HR], 0.81; 95% CI 0.73-0.90) but not in those receiving med-ical management only. When compared to clopidogrel, ticagrelor reduces death from vascular causes, MI, or stroke (HR: 0.84;

    95% CI, 0.77-0.92) in patients undergoing PCI or receiving medical management only. Both the agents, however, increase the riskof bleeding in certain patient population. Conclusions: In the last 5 years, newer antiplatelet agents, including prasugrel andticagrelor, have been demonstrated to reduce recurrent cardiovascular events compared to standard therapy and, however, alsocaused increase bleeding in selected patient populations. Newer agents including shorter acting P2Y12 inhibitor or antiplateletsthat target other receptors are being evaluated to improve/maintain therapeutic efficacy yet minimize the risk of bleeding.

    Keywords

    antiplatelet agents, prasugrel, ticagrelor, acute coronary syndrome

    Introduction

    Platelets play an important role in normal homeostasis and

    atherothrombosis by adhering to injured vascular wall, releasing

    vasoactive and prothrombotic mediators that trigger vasocon-

    striction and promote coagulation.1 However, uncontrolled

    progression of this process can lead to intraluminal thrombus

    formation, vascular occlusion, and subsequent ischemia or

    infarction. Coronary artery diseases including stable angina

    and acute coronary syndromes (ACS) are the leading causes

    of morbidity and mortality in the United States.2 In 2010, app-

    roximately 8 million cases of myocardial infarction (MI) were

    diagnosed.2 Thrombotic occlusion of the coronary arteries

    underlies the pathophysiology of each of these conditions. This

    occlusion occurs when atherosclerotic plaque ruptures, and

    platelet aggregation takes place at the site of rupture.1

    Antiplatelet therapy is a cornerstone in coronary artery dis-

    ease management. They interfere with one or more steps of the

    process of platelet release and aggregation and reduce the risk

    of thrombosis. However, the beneficial effect cannot be

    dissociated from an increased risk of bleeding. Aspirin, thieno-

    pyridine P2Y12 receptor antagonists (eg, clopidogrel), and gly-

    coprotein IIb/IIIa inhibitors (eg, abciximab and eptifibatide)

    are standard therapy, well established to prevent and manage

    arterial thrombotic events.3,4 The clinical benefits of dual anti-

    platelet agent (aspirin second generation thienopyridine, and

    clopidogrel) in the management of ACS, especially in patients

    receiving percutaneous coronary intervention (PCI), are alsowell established.57 In unstable angina and non-ST-segment

    elevation MI (NSTEMI), dual antiplatelet therapy with aspirin

    and clopidogrel has been demonstrated to reduce death from

    1 Department of Pharmacy Practice

    Manuscript submitted:April 27, 2013; accepted: July 03, 2013.

    Corresponding Author:

    Judy W. M. Cheng, MCPHS University, 179 Longwood Avenue, Boston, MA

    02115, USA.

    Email: [email protected]

    Journal of Cardiovascular

    Pharmacology and Therapeutics

    18(6) 514-524

    The Author(s) 2012

    Reprints and permission:

    sagepub.com/journalsPermissions.nav

    DOI: 10.1177/1074248413499971

    cpt.sagepub.com

    http://www.sagepub.com/journalsPermissions.navhttp://cpt.sagepub.com/http://cpt.sagepub.com/http://www.sagepub.com/journalsPermissions.nav
  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    2/12

    cardiovascular causes, nonfatal MI, or stroke by a relative risk

    of 20% (clopidogrel: 9.3%, placebo: 11.4%: relative risk [RR]

    0.80; 95% confidence interval [CI], 0.72-0.90; P< .001).5,6 In

    STEMI, dual antiplatelet therapy reduces the rate of occluded

    infarct-related artery on angiography or death or recurrent MI

    before angiography by 36% (placebo: 21.7%, clopidogrel:

    15.0%, (95% CI, 24%-47%; P< .001).7 The ClOpidogrel andMetoprolol in Myocardial Infarction Trial study also demon-

    strated that clopidogrel added to aspirin therapy reduces death,

    reinfarction, and stroke (9.2% clopidogrel vs 10.1% placebo,

    P .002).8 In the last 5 years, newer antiplatelet agents have

    also become available, and even more agents are in the develop-

    mental pipeline. This continues to refine the role of antiplatelet

    agents used in cardiovascular diseases (CVDs). The latest Amer-

    ican College of Cardiology Foundation and American Heart

    Association (AHA) Guidelines on unstable angina and NSTEMI

    management recommend that dual antiplatelet agents be used

    for either medical management or patients undergoing PCI.3

    For medical management patients, in addition to aspirin, bothclopidogrel and ticagrelor are therapeutic options as the second

    antiplatelet agent. For patients undergoing PCI, in addition to

    aspirin, clopidogrel, prasugrel, or ticagrelor are all acceptable

    therapeutic option as the second antiplatelet agent.3 This article

    provides a review of the pharmacology and clinical efficacy/

    adverse effects of these agents with focus on update of the newer

    antiplatelet agents used in this area in the past 5 years.

    Method

    Peer-reviewed clinical trials, review articles, and relevant treat-

    ment guidelines were identified from MEDLINE and CurrentContent database (both from 1966 to April 15, 2013) using

    search terms aspirin, clopidogrel, prasugrel, ticagrelor, glyco-

    protein IIb/IIIa inhibitors, antiplatelet agents, coronary artery

    disease, acute coronary syndrome, pharmacology, pharmacoki-

    netics, and pharmacodynamics. Citations from available arti-

    cles were also reviewed for additional references.

    Clinical Pharmacology of Antiplatelet Agents

    Table 1 summarizes the mechanism of action, pharmacoki-

    netics, and optimal dosing of different antiplatelet agents on the

    US market.

    Aspirin

    Aspirin is the first antiplatelet agent established for its cardio-

    vascular beneficial effect and is the most widely studied and

    used antiplatelet drug. The best-characterized mechanism of

    action of aspirin is related to its capacity to permanently inhibit

    the cyclooxygenase (COX) activity of prostaglandin H-synthase

    1 and prostaglandin H-synthase 2 (COX-1 and COX-2, respec-

    tively).9 The COX isozymes catalyze the conversion of arachi-

    donic acid to prostaglandin H2 (PGH2). The PGH2 is the

    immediate precursor of thromboxane A2 (TXA2) and prostacy-

    clin (PGI2). The TXA2 induces platelet aggregation and

    vasoconstriction, whereas PGI2 inhibits platelet aggregation

    and induces vasodilation. Because TXA2 is largely derived

    from COX-1 (mostly from platelets), it is highly responsive

    to aspirin inhibition.9

    Aspirin is rapidly absorbed in the stomach and upper intes-

    tine. Plasma levels peak 30 to 40 minutes after ingestion, and

    inhibition of platelet function is evident within an hour. In con-trast, it can take 3 to 4 hours to reach peak plasma levels after

    administration of enteric-coated aspirin. Therefore, if a rapid

    effect is required and only enteric-coated tablets are available,

    the tablets should be chewed instead of swallowed intact. The

    oral bioavailability of regular aspirin tablets is 40% to 50%.

    Aspirin has a half-life of 15 to 20 minutes.10 Despite rapid

    clearance of aspirin from the circulation, the platelet inhibitory

    effects last the life span of the platelet, because aspirin irrever-

    sibly inactivates platelet COX-1. Aspirin also acetylates mega-

    karyocyte COX-1, thereby inhibiting thromboxane production

    in newly released platelets as well as those already in the circu-

    lation. The mean life span of human platelets is approximately10 days, which means that approximately 10% to 12% of the

    circulating platelets are replaced every day.9 Major side effect

    of aspirin is dose-related bleeding and gastrointestinal distress.

    Low-dose aspirin (75-325 mg per day) use has been long

    established to be associated with a significant reduction in the

    risk of cardiovascular events.11,12 The role of low-dose aspirin

    for the secondary prevention (in individuals with coronary

    artery disease, peripheral vascular disease, or cerebrovascular

    disease) of cardiovascular events is well established, while its

    use in primary prevention is more controversial.13,14 The deci-

    sion of use of aspirin as primary prevention therapy is depen-

    dent on a balance of an individuals risk of cardiovascularevents and adverse treatment effects, such as bleeding.15 Odd

    ratios for bleeding, in casecontrol studies of low-dose aspirin,

    range between 1.3 and 3.2.16 The US Food and Drug Admin-

    istration has not been adequately persuaded that there is suf-

    ficient evidence of a net benefit for aspirin use in primary

    prevention in all patients.17 The AHA, however, recommends

    low-dose aspirin in individuals with an estimated10% risk

    of a cardiovascular event over a 10-year period.18 Similarly, the

    US Preventive Services Taskforce recommends aspirin in men

    aged 45 to 79 years in whom the benefit of a reduction in MI

    outweighs the harm of an increased risk of gastrointestinal

    bleeding, and in women aged 55 to 79 years in whom the ben-

    efit of a reduction in the risk of ischemic stroke outweighs the

    same risk of harm.19 For older adults, they recommend a 12%

    risk of a cardiovascular event over 10 years as the cutoff when

    the benefit exceeds the risk in those aged 70 to 79 years. For

    people with diabetes, the American Diabetes Association

    acknowledges the lack of a clear role for aspirin in primary

    prevention and currently recommends its use in patients with

    diabetes who have a 10-year CVD risk of over 10%.20,21

    Glycoprotein IIb/IIIa Inhibitors

    Glycoprotein (Gp) IIb/IIIa inhibitors prevent platelet aggrega-

    tion by blocking fibrinogen binding to the GpIIb/IIIa

    Cheng 515

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    3/12

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    4/12

    receptors on the platelet, thus preventing linking of platelets,

    the final step of platelet aggregation. Three parenteral glyco-

    protein IIb/IIIa inhibitors are available on the US market.

    Abciximab is a humanized version of a Fab fragment of a

    murine antibody directed against GpIIb-IIIa. Platelet aggre-

    gation is nearly completely abolished with >80% receptor

    blockade.22 After intravenous bolus administration, pharma-cokinetic data indicate that free plasma abciximab concentra-

    tions decrease rapidly (initial half-life of about 30 minutes),

    reflecting the rapid binding of the antibody to GpIIb-IIIa.22

    Peak effects on receptor blockade, platelet aggregation, and

    bleeding time were observed at 2 hours. This was followed

    by gradual recovery of platelet function, with bleeding times

    returning to baseline by 12 hours.22,23 Thrombocytopenia

    occurs in 1% to 2% of the patients treated with abciximab.

    The risk of thrombocytopenia appears to be increased with

    abciximab readministration. Typically, the decrease in plate-

    let count occurs within 24 hours of initiation of treatment but

    may begin to fall as early as 2 hours after the treatment starts.9

    Abciximab has been demonstrated to decrease the risk of

    events compared with placebo in high-risk patients with

    NSTEMI scheduled for PCI after treatment with clopidogrel.

    In a meta-analysis of studies assessing patients presenting

    for primary PCI and stenting of STEMI (n 1101), death

    or reinfarction was also reduced in patients receiving abcixi-

    mab versus placebo.24

    Tirofiban is a nonpeptide tyrosine derivative that selectively

    binds to GpIIb-IIIa. The plasma half-life of tirofiban is 1.5 to 2

    hours, and both renal and biliary excretion contribute to tirofi-

    ban clearance with unchanged tirofiban found in urine and

    feces.

    25

    Dose adjustment is required in patients with renalinsufficiency but not in patients with hepatic disease. Severe,

    but reversible, thrombocytopenia has been reported in a small

    percentage of patients treated with tirofiban.26 In a meta-

    analysis (n 20 006), tirofiban used in patients with unstable

    angina and NSTEMI scheduled for PCI was significantly more

    effective than placebo at reducing the risk of mortality or the

    composite of death and MI at 30 days.27

    Eptifibatide is a synthetic disulfide-linked cyclic heptapep-

    tide. It is formulated after the KGD sequence found in the snake

    venom disintegrin obtained from Sistrurus miliarius barbouri

    (barbourin) and has high specificity for GpIIb-IIIa.9 Because

    the drug is cleared by the kidneys, patients with renal impair-

    ment exhibit prolonged inhibition of platelet function after

    receiving eptifibatide and required dosage adjustment.28 Eptifi-

    batide treatment has also been associated with thrombocytope-

    nia, and an immunologic mechanism has been identified in

    some patients.26 In one study in patients with unstable angina

    and NSTEMI undergoing PCI, eptifibatide did not show signif-

    icant benefit when compared with placebo.29 However, in a

    subsequent trial investigating higher doses, a significant reduc-

    tion in the risk of death, MI, urgent coronary revascularization,

    and bail-out use of GPIIb/IIIa inhibitors was demonstrated ver-

    sus placebo.30 More recently, it has been shown that early

    administration of eptifibatide presented no advantage over

    postangiographic administration.31

    Dipyridamole

    Dipyridamole is a pyrimidopyrimidine derivative with vasodi-

    lator and antiplatelet properties. The mechanism of action of

    dipyridamole as an antiplatelet agent is controversial. Both

    inhibition of cyclic nucleotide phosphodiesterase (the enzyme

    that degrades cyclic adenosine monophosphate [cAMP]) and

    blockade of the uptake of adenosine (which binds to A2 recep-

    tors, stimulates platelet adenylyl cyclase, and increases cAMP)

    have been suggested.9 The cAMP is an inhibitor for platelet

    aggregation. The absorption of dipyridamole is variable and

    results in low-systemic bioavailability of the drug. A modified

    release formulation of dipyridamole with improved bioavailabil-

    ity has been developed in a combination pill with low-dose

    aspirin.32 Dipyridamole is highly protein bound to albumin,

    eliminated primarily by biliary excretion as a glucuronide con-

    jugate, and is subject to enterohepatic recirculation. Half-life of

    dipyridamole is 10 hours.9 The use of dipyridamole for primary

    or secondary prevention of coronary artery disease is not well

    established.

    Cilostazil

    Cilostazol is a 2-oxoquinolone derivative that is reported to

    have vasodilatory and antiplatelet properties, via phosphodies-

    terase 3 inhibitory effect, as well as antiproliferative properties

    reducing smooth muscle cell proliferation and neointimal

    hyperplasia after endothelial injury. Cilostazol is contraindi-

    cated in patients with heart failure because of the potential to

    trigger ventricular tachycardia. There is substantial variability

    in the absorption of orally administered cilostazol. Cilostazol

    is highly albumin bound and is extensively metabolized by

    cytochrome P450 (CYP450) enzymes with excretion of meta-

    bolites in the urine. It has a half-life of 11 hours.9 Cilostazil

    is indicated for symptomatic peripheral arterial disease, but its

    role in primary or secondary prevention of coronary artery

    disease is not well established.

    P2Y12 receptor antagonists

    Ticlopidine, clopidogrel, and prasugrel represent 3 generations

    of oral thienopyridines that inhibit adenosine diphosphate

    (ADP)-induced platelet aggregation. The use of first-genera-

    tion agent ticlopidine was limited by its bone marrow toxicity

    (neutropenia) and has largely been replaced by clopidogrel that

    has become established as standard therapy across the spectrum

    of patients with coronary artery disease and in those under-

    going PCI. However, clopidogrel also has limitations, includ-

    ing variable absorption; variable antiplatelet effects related,

    at least in part, to common polymorphisms in the genes that

    regulate the metabolic activation of clopidogrel; and a delayed

    onset and offset of action. Prasugrel, a more recently available,

    third-generation thienopyridine, has a more rapid onset of

    action, is more potent than clopidogrel, and produces more

    consistent platelet inhibition. All 3 thienopyridines are pro-

    drugs that must undergo metabolic activation through the

    Cheng 517

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    5/12

    hepatic CYP450 system to generate the active metabolites that

    exert their pharmacologic action.

    Ticlopidine.Up to 90% of a single oral dose of ticlopidine is rap-

    idly absorbed.18 Plasma concentrations peak 1 to 3 hours after

    a single-oral dose of 250 mg. More than 98% of the absorbed

    ticlopidine is reversibly bound to plasma proteins, primarilyalbumin. Ticlopidine is metabolized rapidly and extensively.

    A total of 13 metabolites have been identified in humans.18 The

    apparent half-life of ticlopidine is 24 to 36 hours after a single

    oral dose and up to 96 hours after 14 days of repeated dosing.

    The standard dosing regimen of ticlopidine is 250 mg twice

    a day.33

    Clopidogrel. Clopidogrel is also rapidly absorbed and metabo-

    lized through a 2-step process to generate an active metabolite

    that irreversibly binds to the platelet P2Y12 receptor.34 On

    repeated daily dosing of 50 to 100 mg of clopidogrel in healthy

    volunteers, ADP-induced platelet aggregation was inhibitedfrom the second day of treatment (25%-30% inhibition) and

    reached a steady state (50%-60% inhibition) after 4 to 7 days.

    Such a level of inhibition was comparable to that achieved with

    ticlopidine (500 mg/d), although the antiplatelet effects of the

    latter were more delayed than that of clopidogrel.34 Loading

    dose (eg, 300 mg) of clopidogrel results in more rapid platelet

    inhibition (6-12 hours) than that achieved with the 75 mg

    dose.35 After loading with 600 mg of clopidogrel, the full anti-

    platelet effect of the drug was achieved after 2 to 4 hours.36

    Moreover, a loading dose of 600 mg resulted in higher plasma

    concentrations of the active metabolite and the inactive car-

    boxyl metabolite compared with a loading dose of 300 mg.

    36

    Inhibition of ADP-induced platelet aggregation was also signif-

    icantly greater with a 600 mg loading dose of clopidogrel

    compared with a 300 mg loading dose.37 The incremental anti-

    platelet effect of 900 mg over 600 mg of clopidogrel appears

    marginal.37,38 Platelet function returns to baseline 7 to 10 days

    after the last dose of clopidogrel. This also justifies a once-

    daily regimen for aspirin and clopidogrel in patients with

    normal rates of platelet turnover despite short half-life of both

    the drugs in the circulation.

    Prasugrel.Prasugrel is rapidly absorbed after oral administration

    and converted into its active metabolite, which reaches peak

    concentrations within 30 minutes of dosing. The active meta-

    bolite has a half-life of approximately 4 hours, and renal excre-

    tion is the major route for elimination.39 Initial pharmaco-

    logical studies with prasugrel in healthy individuals39 and in

    patients with stable coronary artery disease32 showed that pra-

    sugrel has a more rapid onset of action than clopidogrel and

    achieves more consistent and complete inhibition of ADP-

    induced platelet aggregation.40,41 The more rapid onset of

    action of prasugrel may in part reflect the hepatic conversion

    to its active metabolite by CYP450 enzymes in a single step,

    which contrasts with that of clopidogrel that undergoes a

    2-step hepatic conversion process.42 Unlike clopidogrel, evi-

    dence showed that polymorphisms in CYP2C19 or the

    concomitant use of proton-pump inhibitors did not interfere

    with the metabolism of prasugrel.43,44 Clinical studies on the

    use of prasugrel in patients with coronary artery disease will

    be discussed in greater depth in the clinical trial section.

    Ticagrelor

    Ticagrelor is the first member of a new class of antiplatelet

    agent, the cyclopentyl-triazolopyrimidines.45 Ticagrelor is an

    orally active, selective antagonist of the P2Y12 receptor, inhi-

    biting ADP-mediated platelet response. Ticagrelor demon-

    strates noncompetitive antagonistic activity for ADP-induced

    platelet aggregation.46 Unlike clopidogrel and prasugrel, tica-

    grelor demonstrates platelet inhibitory activity without the

    need for metabolic activation. In addition, its active metabolite

    (AR-C124910XX) is also active and demonstrates P2Y12

    receptor antagonism activity equipotent to ticagrelor.47,48

    Ticagrelor was rapidly absorbed with a maximum plasma

    concentration occurring at 1.5 hours. The major active metabo-

    lite, AR-C124910XX, is formed by O-deethylation (via

    CYP3A4) and represents approximately 40% of the parent

    concentration. The plasma elimination half-life of ticagrelor

    was found to be 7.2 hours.49 Clinical trials evaluating the use

    of ticagrelor in patients with coronary artery disease will be

    discussed subsequently.

    Clinical Trials Updates: Updates Fromthe Last 5 years

    Prasugrel

    The majority of the clinical outcomes data for prasugrel comes

    from the phase III Trial to Access Improvement in Therapeutic

    Outcomes by Optimizing Platelet Inhibition with Prasugrel-

    Thrombolysis in Myocardial Infarction (TRITON-TIMI 38)

    trial. In this study, 13 608 patients with ACS with planned

    PCI (10 074 patients with moderate to high-risk unstable

    angina and NSTEMI and 3534 patients with STEMI) were ran-

    domized to receive either clopidogrel 300 mg loading dose

    followed by 75 mg daily or prasugrel 60 mg loading dose fol-

    lowed by 10 mg daily.50 Patients were treated for a median of

    14.5 months. The primary end point was the composite of death

    from cardiovascular causes, nonfatal MI, or nonfatal stroke.

    Patients randomized to prasugrel had fewer primary end point

    events compared with clopidogrel (9.9% vs 12.1%; hazard ratio

    [HR], 0.81; 95% CI, 0.73 to 0.90; P < .001). The reduction in

    clinical ischemic events was also notable for a reduction in MI

    (7.4% vs 9.7%,P < .001) and urgent target vessel revasculari-

    zation (2.5% vs 3.7%;P < .001). The major safety end point of

    noncoronary artery bypass graft (CABG)-related TIMI major

    bleeding was significantly higher with prasugrel (2.4% vs

    1.8%; HR, 1.32; 95% CI, 1.03-1.68; P < .03). There was also

    significant increase in non-CABG-related TIMI major or minor

    bleeding (5.0% vs 3.8%; HR, 1.31; 95% CI, 1.11-1.56; P 75 years of age, 9 spontaneous fatal hemorrhages

    were observed with prasugrel and 0 with clopidogrel. In patients

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    7/12

    in intensity and often resolved during continued treatment.57 In

    a substudy,199 patients from PLATO (101 ticagrelor and 98

    clopidogrel) underwent pulmonary function testing irrespective

    to whether they reported dyspnea.58 There was no significant

    difference between treatment groups for forced expiratory

    volume in the first second of expiration (ticagrelor 2.81 L and

    clopidogrel 2.70 L). There was no indication of an adverseeffect on pulmonary function assessed after 1 month or after

    at least 6 months of chronic treatment.

    Ticagrelor has also been shown to increase the occurrence of

    Holter-detected bradyarrhythmias (including ventricular

    pauses).59 In a Holter substudy, a total of 2908 patients were

    included in the continuous electrographic assessment, of whom

    2866 (98.5%) had week 1 recordings, 1991 (68.4%) had

    1-month recordings, and 1949 (67.0%) had both. During the

    first week after randomization, ventricular pauses 3 seconds

    occurred more frequently in patients receiving ticagrelor than

    clopidogrel (5.8% vs 3.6%; RR: 1.61; P .006). At 1 month,

    pauses 3 seconds occurred overall less frequently and weresimilar between treatments (2.1% vs 1.7%). There were no dif-

    ferences between ticagrelor and clopidogrel in the incidence of

    clinically reported bradycardic adverse events. Ticagrelor is

    structurally similar to adenosine57 and therefore may partially

    explain some of its side effects similar to that of adenosine such

    as dyspnea and bradyarrhythmia.

    Several subanalyses were performed with PLATO. One sub-

    analysis was performed in patients who underwent planned

    invasive strategy (PCI or CABG) for management of their

    ACS.60 In all, 6732 patients undergoing invasive strategy were

    assigned to ticagrelor and 6676 patients were assigned to clopi-

    dogrel. The primary composite end point occurred in fewerpatients in the ticagrelor group than in the clopidogrel group

    (9.0% vs 10.7%, HR 0.84, 95% CI 0.75-0.94; P .0025).

    There was no difference between clopidogrel and ticagrelor

    groups in the rates of total major bleeding (11.6% vs 11.5%,

    HR 0.99, 95% CI 0.89-1.10; P .8803) or severe bleeding

    (3.2% vs 2.9%, HR 0.91, 95% CI 0.74-1.12; P .3785).

    Another PLATO substudy was also performed looking at

    patients with ACS who were managed by medical treatment

    only.61 In all, 3143 patients were managed noninvasively. The

    incidence of the primary end point was lower with ticagrelor

    than with clopidogrel (12.0% vs 14.3%; HR 0.85, 95% CI

    0.73-1.00; P .04). Overall mortality was also lower (6.1%

    vs 8.2%; HR 0.75, 95% CI 0.61-0.93;P .01). The incidence

    of total major bleeding (11.9% vs 10.3%; HR 1.17, 95% CI

    0.98-1.39; P .08) and non-CABG-related major bleeding

    (4.0% vs 3.1%; HR 1.30, 0.95-1.77; P .10) was not signifi-

    cantly different between ticagrelor and clopidogrel.

    The subpopulation in the PLATO trial that underwent

    CABG was also examined in a separate subanalysis.62 The pro-

    tocol recommended ticagrelor to be withheld for 24 to 72 hours

    and clopidogrel for 5 days preoperatively. In all, 1261 patients

    underwent CABG and were receiving study drug treatment

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    8/12

    majority of patients (*70%) recovered more than 60% of their

    baseline aggregation response by 1 hour after infusion.67 The

    potential advantage of a short-acting P2Y12 inhibitor is that

    there will be less delay should patients require bypass surgery.

    Cangrelor was studied in 2 large phase III trials with PCI

    (CHAMPION-PLATFORM and CHAPMION-PHEONIX) and

    demonstrated significant reduction in rate of ischemic event,including stent thrombosis.68,69 Compared with placebo, major

    bleeding appeared to be more frequent due to increased groin

    hematomas.68 Similar to ticagrelor, cangrelor is associated with

    higher frequencies of dyspnea.68,69 The fast onset and offset of

    action of cangrelor may make it a desirable therapeutic option

    for patients with ACS undergoing PCI before committing

    patients to one of the longer acting oral antiplatelet agents for

    chronic therapy.

    Protease Activated Receptor 1 Inhibitors. Thrombin is one of the

    most potent platelet activators, and protease activated recep-

    tor 1 (PAR-1) is the principal thrombin receptor on human

    platelets. However, unlike ADP and TXA2 pathways, which

    are crucial in both physiological hemostasis and pathological

    thrombosis, preclinical evidence suggests PAR-1-mediated

    platelet activation does not appear to be involved in protective

    hemostasis. It has been hypothesized that combining a PAR-1

    inhibitor with existing treatment may increase platelet inhibi-

    tion without further increasing the risk of bleeding.70 Cur-

    rently, 2 PAR-1 inhibitors are under clinical investigation,

    vorapaxar and atopaxar, and vorapaxar has entered Phase III

    clinical trials. Vorapaxar is a high affinity, oral antagonist

    of the PAR-1 receptor.71 In a recent large clinical trial,

    Thrombin Receptor Antagonist for Clinical Event Reduction

    in Acute Coronary Syndrome patients (n 13 000) receiving

    vorapaxar did not experience improvement in the primary

    efficacy end point (a composite of death from cardiovascular

    causes, MI, stroke, recurrent ischemia with rehospitalization,

    or urgent coronary revascularization) versus placebo. Further-

    more, vorapaxar significantly increased the incidence of

    moderate and severe bleeding as well as intracranial haemor-

    rhage.72 This is consistent with a large-scale secondary pre-

    vention trial (Thrombin Receptor Antagonist in Secondary

    Prevention of Atherothrombotic Ischemic Events [TRA-2P/

    TIMI 50]), where an increase in intracranial hemorrhage was

    detected in patients with a history of stroke. This trend contin-ued in patients in the TRA-2P/TIMI 50 trial, where the data

    and safety monitoring board recommended the discontinua-

    tion of vorapaxar in patients with a history of stroke on the

    basis of an excess of intracranial hemorrhage. The overall

    trial, however, showed that in 26 449 patients with a history

    of MI, ischemic stroke, or peripheral arterial disease (PAD),

    vorapaxar 2.5 mg daily significantly reduced the primary

    efficacy end point, a composite of death from cardiovascular

    causes, MI, or stroke compared with placebo (9.3% vs 10.5%,

    respectively; HR 0.87 [95% CI 0.80, 0.94]; P < .001).73 The

    role of PAR-1 inhibitor in management of coronary artery dis-

    ease needs to be further defined.

    Conclusions

    Antiplatelet therapy, including aspirin, clopidogrel, and glyco-

    protein IIb/IIIa inhibitors, is a cornerstone in coronary artery

    disease management. However, with the advances made in the

    area of antiplatelet therapy in the last few years, patients with

    ACS still remain at risk of recurrent cardiovascular events. In

    the last 5 years, newer antiplatelet agents including prasugrel

    and ticagrelor have become available. Each has demonstrated

    reduced recurrent cardiovascular events compared to standard

    therapy and, however, also caused increased bleeding in

    selected patient populations. Newer agents including shorter

    acting P2Y12 inhibitors, such as cangrelor or antiplatelets that

    target other receptors, are being evaluated to attempt to

    improve/maintain therapeutic efficacy yet minimize the risk

    of bleeding.

    Declaration of Conflicting Interests

    The author(s) declared no potential conflicts of interest with respect tothe research, authorship, and/or publication of this article.

    Funding

    The author(s) received no financial support for the research, author-

    ship, and/or publication of this article.

    References

    1. Dav G, Patrono C. Platelet activation and atherothrombosis.

    N Engl J Med. 2007;357(24):2482-2494.

    2. Go AS, Mozaffarian D, Roger V, et al. Heart disease and stroke

    statistics2013 update: a report from the American heart associ-ation.Circulation. 2013;127(1):143-152.

    3. 2012 ACCF/AHA Focused Update Incorporated Into the ACC/

    AHA 2007 Guidelines for the Management of Patients With

    Unstable Angina/Non-ST Elevation Myocardial Infarction: A

    Report of the American College of Cardiology Foundation/

    American Heart association Task Force on Practice Guidelines;

    2013. http://circ.ahajournals.org/content/127/23/e663.full.pdf

    html. Accessed July 2, 2013.

    4. OGara PT, Kushner FG, Ascheim DD, et al. 2013 ACCF/AHA

    guideline for the management of ST elevation myocardial infarc-

    tion.Circulation. 2013;127(4):e362-e425.

    5. Yusuf S, Zhao F, Mehta SR, et al. Effects of clopidogrel in addi-

    tion to aspirin in patients with acute coronary syndromes without

    ST-segment elevation.N Engl J Med. 2001;345(7):494-502.

    6. Mehta SR, Yusuf S, Peters RJ, et al. Effects of pretreatment with

    clopidogrel and aspirin followed by long-term therapy in patients

    undergoing percutaneous coronary intervention: the PCI-CURE

    study.Lancet. 2001;358(9281):527-533.

    7. Sabatine MS, Cannon CP, Gibson CM, et al. Addition of clopido-

    grel to aspirin and fibrinolytic therapy for myocardial infarction

    with ST-segment elevation. N Engl J Med . 2005;352(12):

    1179-1189.

    8. Chen ZM, Jiang LX, Chen YP, et al. Addition of clopidogrel to

    aspirin in 45852 patients with acute myocardial infarction: rando-

    mized placebo controlled trial.Lancet. 2005;366(9497):1607-1621.

    Cheng 521

    http://circ.ahajournals.org/content/127/23/e663.full.pdf+htmlhttp://circ.ahajournals.org/content/127/23/e663.full.pdf+htmlhttp://circ.ahajournals.org/content/127/23/e663.full.pdf+htmlhttp://circ.ahajournals.org/content/127/23/e663.full.pdf+html
  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    9/12

    9. Eikelboom JW, Hirsh J, Spencer FA, Baglin TP, Weitz JI. Anti-

    platelet drugs: antithrombotic therapy and prevention of thrombo-

    sis, 9th ed: American college of chest physicians evidence based

    clinical practice guidelines. Chest. 2012;141(2 suppl):e89S-

    e119S.

    10. Pedersen AK, FitzGerald GA. Dose-related kinetics of aspirin.

    presystemic acetylation of platelet cyclooxygenase. N EnglJ Med. 1984;311(19):1206-1211.

    11. Hennekens CH. Aspirin in the treatment and prevention of cardi-

    ovascular disease: current perspective and future directions. Curr

    Atheroscler Rep. 2007;9(5):409-416.

    12. Baigent C, Blackwell L, Collins R, et al. Aspirin in the primary

    and secondary prevention of vascular disease: collaborative

    meta-analysis of individual participant data from randomized

    trials.Lancet. 2009;373(9678):1849-1860.

    13. Berger JA, Lala A, Jrantz MJ, Baker GS, Hiatt WR. Aspirin for

    the prevention of cardiovascular events in patients without clini-

    cal cardiovascular disease: a meta-analysis of randomized trial.

    Am Heart J. 2011;162(1):115-124.14. Seshaspirini SR, Wijesuriya S, Sivakumaran R, et al. Effect of

    aspirin on vascular and non-vascular outcomes. meta-analysis of

    randomized controlled trials. Arch Intern Med. 2012;172(3):

    209-216.

    15. Sussman JB, Vijan S, Choi H, et al. Individual and population

    benefits of daily aspirin therapy: a proposal for personalizing

    national guidelines. Cir Cardiovasc Qual Outcomes. 2011;4(3):

    268-275.

    16. Shiotani A, Manabe N, Kamada T, Fujimura Y, Sakakibara, Har-

    uma K. Risk and prevention factors of low-dose aspirin-induced

    gastroduodenal injuries: a comprehensive review.J Gastroenterol

    Hepatol. 2012;27(suppl 3):8-12.

    17. Nicolucci A, De Berardis G, Sacco M, Tognoni G. AHA/ADA

    vs. ESC/EASD recommendations on aspirin as a primary

    prevention strategy in people with diabetes: how the same data

    generate divergent conclusions. Eur Heart J. 2007;28(16):

    1925-1927.

    18. Pearson TA, Blair SN, Daniels SR, et al. AHA Guidelines for pri-

    mary prevention of cardiovascular disease and stroke: 2002

    update: consensus panel guide to comprehensive risk reduction

    for adult patients without coronary or other atherosclerotic vascu-

    lar diseases. American heart association science advisory and

    coordinating committee. Circulation. 2002;106(3):388-391.

    19. US Preventive Services Task Force. Aspirin for the prevention of

    cardiovascular disease: U.S. preventive services task force recom-

    mendation statement. Ann Intern Med. 2009;150(6):396-404.

    20. Pignone M, Alberts MJ, Colwell JA, et al. Aspirin for primary

    prevention of cardiovascular events in people with diabetes:

    a position statement of the American diabetes association, a scien-

    tific statement of the American heart association, and an expert

    consensus document of the American college of cardiology foun-

    dation.Diabetes Care. 2010;33(6):1395-1402.

    21. Pignone M, Williams CD. Aspirin for primary prevention of car-

    diovascular disease in diabetes mellitus. Nat Rev Endocrinol.

    2010;6(11):619-628.

    22. Tcheng JE, Ellis SG, George BS, et al. Pharmacodynamics of chi-

    meric glycoprotein IIb/IIIa integrin antiplatelet antibody Fab 7E3

    in high-risk coronary angioplasty. Circulation. 1994;90(4):

    1757-1764.

    23. Mascelli MA, Lance ET, Damaraju L, Wagner CL, Weisman HF,

    Jordan RE. Pharmacodynamic profile of short-term abciximab

    treatment demonstrates prolonged platelet inhibition with gradual

    recovery from GP IIb/IIIa receptor blockade. Circulation. 1998;

    97(17):1680-1688.24. Montalescot G, Antoniucci D, Kastrati A. Abciximab in primary

    coronary stenting of ST-elevation myocardial infarction: a Eur-

    opean meta-analysis on individual patients data with long-term

    follow-up.Eur Heart J. 2007;28(4):443-449.

    25. Vickers S, Theoharides AD, Arison B, et al. In vitro and in vivo

    studies on the metabolism of tirofiban. Drug Metab Dispos.

    1999;27(11):1360-1366.

    26. Madan M, Berkowitz SD. Understanding thrombocytopenia and

    antigenicity with glycoprotein IIb-IIIa inhibitors. Am Heart J.

    1999;138(4 pt 2):317-326.

    27. Valgimigli M, Biondi-Zoccai G, Tebaldi M, et al. Tirofiban as

    adjunctive therapy for acute coronary syndromes and percuta-neous coronary intervention: a meta-analysis of randomized trials.

    Eur Heart J. 2010;31(1):35-49.

    28. Smith BS, Gandhi PJ. Pharmacokinetics and pharmacodynamics

    of low-molecular-weight heparins and glycoprotein IIb/IIIa

    receptor antagonists in renal failure. J Thromb Thrombolysis.

    2001;11(1):39-48.

    29. IMPACT-II Investigators. Randomized placebo-controlled trial of

    effect of eptifibatide on complications of percutaneous coronary

    intervention: IMPACT-II. Integrilin to minimize platelet aggrega-

    tion and coronary thrombosis-II. Lancet. 1997;349(9063):

    1422-1428.

    30. ESPRIT-II Investigators. Novel dosing regimen of eptifibatide in

    planned coronary stent implantation (ESPRIT): a randomized,

    placebo-controlled trial. Lancet. 2000;356(9247):2037-2044.

    31. Giugliano RP, White JA, Bode C, et al. Early versus delayed, pro-

    visional eptifibatide in acute coronary syndromes. N Engl J Med.

    2009;360(21):2176-2190.

    32. Muller TH, Su CA, Weisenberger H, Brickl R, Nehmiz G, Eisert

    WG. Dipyridamole alone or combined with low-dose acetylsa-

    licylic acid inhibits platelet aggregation in human whole blood

    ex vivo.Br J Clin Pharmacol. 1990;30(2):179-186.

    33. Savi P, Herbert JM. Clopidogrel and ticlopidine: P2Y12 adeno-

    sine diphosphate-receptor antagonists for the prevention of ather-

    othrombosis.Semin Thromb Hemost. 2005;31(2):174-183.

    34. Savi P, Pereillo JM, Uzabiaga MF, et al. Identification and biolo-

    gical activity of the active metabolite of clopidogrel. Thromb

    Haemost. 2000;84(5):891-896.

    35. Savcic M, Hauert J, Bachmann F, Wyld PJ, Geudelin B, Cariou R.

    Clopidogrel loading dose regimens: kinetic profile of pharmaco-

    dynamic response in healthy subjects. Semin Thromb Hemost.

    1999;25(suppl 2):15-19.

    36. Hochholzer W, Trenk D, Frundi D, et al. Time dependence of pla-

    telet inhibition after a 600-mg loading dose of clopidogrel in a

    large, unselected cohort of candidates for percutaneous coronary

    intervention.Circulation. 2005;111(20):2560-2564.

    37. von Beckerath N, Taubert D, Pogatsa-Murray G, Schomig E,

    Kastrati A, Schomig A. Absorption, metabolization, and antiplatelet

    522 Journal of Cardiovascular Pharmacology and Therapeutics 18(6)

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    10/12

    effects of 300-, 600-, and 900-mg loading doses of clopidogrel:

    results of the ISAR-CHOICE(intracoronarystenting and antithrom-

    botic regimen: choose between 3 high oral doses for immediate clo-

    pidogrel effect) trial.Circulation. 2005;112(19):2946-2950.

    38. Cuisset T, Frere C, QuiliciJ, et al. Benefit of a 600-mg loading dose

    of clopidogrel on plateletreactivity and clinicaloutcomesin patients

    with non-ST-segment elevation acute coronary syndromeunder-going coronary stenting.J Am Coll Cardiol. 2006;48(7):1339-1345.

    39. Montalescot G, Sideris G, Meuleman C, et al. ALBION Trial

    Investigators. A randomized comparison of high clopidogrel

    Cheng 9 loading doses in patients with non-ST-segment elevation

    acutecoronary syndromes: the ALBION (assessment of the best

    loading dose of clopidogrel to blunt platelet activation, inflamma-

    tion and ongoing necrosis) trial. J Am Coll Cardiol. 2006;48(5):

    931-938.

    40. Farid NA, Smith RL, Gillespie TA, et al. The disposition of pra-

    sugrel, a novel thienopyridine, in humans. Drug Metab Dispos.

    2007;35(7):1096-1104.

    41. Armstrong PC, Dhanji AR, Tucker AT, Mitchell JA, Warner TD.Reduction of platelet thromboxane A2 production ex vivo and in

    vivo by clopidogrel therapy. J Thromb Haemost. 2010;8(3):

    613-615.

    42. Jernberg T, Payne CD, Winters KJ, et al. Prasugrel achieves

    greater inhibition of platelet aggregation and a lower rate of

    non-responders compared with clopidogrel in aspirin treated

    patients with stable coronary artery disease. Eur Heart J. 2006;

    27(10):1166-1173.

    43. Farid NA, Payne CD, Ernest CS II, et al. Prasugrel, a new thieno-

    pyridine antiplatelet drug, weakly inhibits cytochrome P450 2B6

    in humans.J Clin Pharmacol. 2008;48(1):53-59.

    44. Brandt JT, Payne CD, Wiviott SD, et al. A comparison of prasu-

    grel and clopidogrel loading doses on platelet function: magnitude

    of platelet inhibition is related to active metabolite formation. Am

    Heart J. 2007;153(1):66.e9-e16.

    45. Wiviott SD, Braunwald E, McCabe CH, et al; TRITON TIMI 38

    Investigators. Prasugrel versus clopidogrel in patients with acute

    coronary syndromes.N Engl J Med. 2007;357(20):2001-2015.

    46. Capodanno D, Dharmashankar K, Angiolillo DJ. Mechanism of

    action and clinical development of ticagrelor, a novel platelet

    ADP P2Y12 receptor antagonist. Expert Rev Cardiovasc Ther.

    2010;8(2):151-158.

    47. Herbert JM, Frehel D, Vallee E, et al. Clopidogrel, a novel anti-

    platelet and antithrombotic agent. Cardiovasc Drug Rev. 1993;

    11(2):180-198.

    48. Storey RF. Pharmacology and clinical trials of reversibly binding

    P2Y12 inhibitors.Thromb Haemost. 2011;105(suppl 1):S75-S81.

    49. Brilinta prescribing information; 2012. http://www1.astraze-

    neca-us.com/pi/brilinta.pdf. Accessed January 5, 2012.

    50. Teng R, Oliver S, Hayes MA, Butler K. Absorption, distribution,

    metabolism and excretion of ticagrelor in healthy subjects. Drug

    Metab Dispos. 2010;38(9):1514-1521.

    51. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus

    clopidogrel in patients with acute coronary syndromes. N Engl

    J Med. 2007;357(20):2001-2015.

    52. Wiviott SD, Braunwald E, McCabe CH, et al. Intensive oral anti-

    platelet therapy for reduction of ischemic events including stent

    thrombosis in patients with acute coronary syndromes treated

    with percutaneous coronary intervention and stenting in the

    TRITON-TIMI 38 trial: a subanalysis of a randomized trial. Lan-

    cet. 2008;371(9621):1353-1363.

    53. Wiviott SD, Braunwald E, Angiolillo DJ, et al. Greater clinical

    benefit of more intensive oral antiplatelet therapy with prasugrel

    in patients with diabetes mellitus in the trial to assess improve-ment in therapeutic outcomes by optimizing platelet inhibition

    with prasugrel-thrombolysis in myocardial infarction 38. Circula-

    tion. 2008;118(16):1626-1636.

    54. Montalescot G, Wiviott SD, Braunwald E, et al. Prasugrel

    compared with clopidogrel in patients undergoing percutaneous

    coronary intervention for ST-elevation myocardial infarction

    (TRITON-TIMI 38): double-blind, randomized controlled trial.

    Lancet. 2009;373(9665):723-731.

    55. Mega JL, Close SL, Wiviott SD, et al. Cytochrome p-450 poly-

    morphisms and response to prasugrel: relationship to pharmacoki-

    netic, pharmacodynamic, and clinical outcomes. Circulation.

    2009;119(19):2553-2560.56. Roe MT, Armstrong PW, Fox KA, et al. Prasugrel versus clopido-

    grel for acute coronary syndromes without revascularization.

    N Engl J Med. 2012;367(14):1297-1309.

    57. Wallentin L, Richard BC, Budaj A, et al. Ticagrelor versus clopi-

    dogrel in patients with acute coronary syndromes. N Engl J Med.

    2009;361(11):1045-1057.

    58. Storey RF, Becker RC, Harrington RA, et al. Pulmonary function

    in patients with acute coronary syndrome treated with ticagrelor

    or clopidogrel (from the platelet inhibition and platelet outcome

    (PLATO) pulmonary function substudy). Am J Cardiol. 2011;

    108(11):1542-1546.

    59. Scirica BM, Cannon CP, Emanuelsson H, et al. The incidence of

    bradyarrhythmias and clinical bradyarrhythmic events in patients

    with acute coronary syndromes treated with ticagrelor or clopido-

    grel in PLATO (platelet inhibition and patient outcomes) trial:

    results of the continuous electrocardiographic assessment sub-

    study.J Am Coll Cardiol. 2011;57(19):1908-1916.

    60. Cannon CP, Harrington RA, James S, et al. Comparison of tica-

    grelor with clopidogrel in patients with planned invasive strategy

    for acute coronary syndromes (PLATO): a randomized double-

    blind study.Lancet. 2010;375(9711):283-293.

    61. James SK, Row MT, Cannon CP, et al. Ticagrelor versus clopido-

    grel in patients with acute coronary syndromes intended for non-

    invasive management: substudy from prospective randomized

    platelet inhibition and patient outcomes (PLATO) trial. BMJ.

    2011;342:d3527.

    62. Held C, AsenbladN, Bassand JP,et al. Ticagrelor versus clopidogrel

    in patients with acute coronary syndromes undergoing coronary

    artery bypass surgery: results from the PLATO (platelet inhibition

    and patient outcomes) trial.J Am Coll Cardiol. 2011;57(6):672-684.

    63. Steg PG, James S, Harrington RA, et al. Ticagrelor versus clopido-

    grel in patients with ST elevation acute coronary syndromes

    intended for reperfusion with primary percutaneous coronary inter-

    vention: a platelet inhibition and patient outcomes (PLATO) trial

    subgroup analysis.Circulation. 2010;122(21):2131-2141.

    64. James S, Budaj A, Aylward P, et al. Ticagrelor versus clopidogrel

    in acute coronary syndromes in relation to renal function: results

    Cheng 523

    http://www1.astrazeneca-us.com/pi/brilinta.pdfhttp://www1.astrazeneca-us.com/pi/brilinta.pdfhttp://www1.astrazeneca-us.com/pi/brilinta.pdfhttp://www1.astrazeneca-us.com/pi/brilinta.pdf
  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    11/12

    from the platelet inhibition and patient outcomes (PLATO) trial.

    Circulation. 2010;122(11):1056-1067.

    65. James S, Angiolillo DJ, Cornel JH, et al. Ticagrelor vs. clopido-

    grel in patients with acute coronary syndromes and diabetes:

    a substudy from the platelet inhibition and patient outcomes

    (PLATO) trial.Eur Heart J. 2010;31(24):3006-3016.

    66. Ferreiro JL, Ueno M, Angiolillo DJ. Cangrelor: a review on itsmechanism of action and clinical development. Expert Rev Car-

    diovasc Ther. 2009;7(10):1195-1201.

    67. van GiezenJJ, HumphriesRG. Preclinical andclinicalstudies with

    selective reversible direct P2Y12 antagonists. Semin Thromb

    Hemost. 2005;31(2):195-204.

    68. Bhatt DL, Lincoff AM, Gibson CM, et al. Intravenous platelet

    blockade with cangrelor during PCI. N Engl J Med. 2009;

    361(24):2330-2341.

    69. Bhatt DL, Stone GW, Mahaffey KW, et al. Effect of platelet inhi-

    bition with cangrelor during PCI on ischemic events. N Engl

    J Med. 2013;368(14):1303-1313.

    70. Angiolillo DJ, Capodanno D, Goto S. Platelet thrombin receptor

    antagonism and atherothrombosis.Eur Hear J. 2010;31(1):17-28.

    71. Chackalamannil S, Xia Y, Greenlee WJ, et al. Discovery of potent

    orally active thrombin receptor (protease activated receptor 1)antagonists as novel antithrombotic agents. J Med Chem. 2005;

    48(19):5884-5887.

    72. Tricoci P, Huang Z, Held C, et al. Thrombin-receptor antagonist

    vorapaxar in acute coronary syndromes. N Engl J Med. 2012;

    366(1):20-33.

    73. Morrow DA, Braunwald E, Bonaca MP, et al. Vorapaxar in the

    secondary prevention of atherothrombotic events. N Engl J Med.

    2012;366(15):1404-1413.

    524 Journal of Cardiovascular Pharmacology and Therapeutics 18(6)

  • 8/12/2019 9-Updates in Antiplatelet Agents 2013 RCT

    12/12

    C o p y r i g h t o f J o u r n a l o f C a r d i o v a s c u l a r P h a r m a c o l o g y & T h e r a p e u t i c s i s t h e p r o p e r t y o f S a g e

    P u b l i c a t i o n s I n c . a n d i t s c o n t e n t m a y n o t b e c o p i e d o r e m a i l e d t o m u l t i p l e s i t e s o r p o s t e d t o a

    l i s t s e r v w i t h o u t t h e c o p y r i g h t h o l d e r ' s e x p r e s s w r i t t e n p e r m i s s i o n . H o w e v e r , u s e r s m a y p r i n t ,

    d o w n l o a d , o r e m a i l a r t i c l e s f o r i n d i v i d u a l u s e .