2,947
views
0
recommends
+1 Recommend
1 collections
    2
    shares

      CVIA now indexed by SCOPUS from February 2024. CVIA received its first Journal Impact Factor (0.5) in the 2023 Journal Citation Reports Release. 

      Interested in becoming a CVIA published author?

      • Platinum Open Access with no APCs. 
      • Fast peer review/Fast publication online after article acceptance.

      Submissions should be made electronically at: https://mc04.manuscriptcentral.com/cvia-journal.

      Please refer to the Author Guidelines at https://cvia-journal.org/instructions-to-authors/ before submission.

       

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Modern Antiplatelet Therapy: When Is Clopidogrel the Right Choice?

      review-article
      Bookmark

            Abstract

            Platelet inhibition with aspirin is the cornerstone of medical therapy for coronary artery disease. In the era of percutaneous coronary intervention with drug-eluting stents, dual anti-platelet therapy with the addition of clopidogrel (Plavix©, Bristol-Myers Squibb, New York, NY) became the mainstay of therapy. However, with the advent of newer oral antiplatelet medications, including prasugrel (Effient©, Eli Lilly, Indianapolis, Indiana) and ticagrelor (Brilinta©, Astra-Zeneca, Cambridge, United Kingdom), choosing the appropriate platelet inhibitor has become more nuanced. The optimal regimen differs based on patient characteristics, clinical situation, and the condition being treated, with differing risk and safety profiles for these medications. This review explores the appropriate use of antiplatelet therapy for stable ischemic heart disease, acute coronary syndrome, stroke, and peripheral vascular disease. Furthermore, we evaluate the data behind the use of antiplatelet therapy in patients on oral anticoagulation. By understanding the biochemistry of platelet aggregation, the pharmacology of platelet inhibitors, and the extensive clinical trial data that informs current guideline recommendations we aim to better understand the role of clopidogrel in patients with cardiovascular disease.

            Main article text

            Introduction

            Antiplatelet agents are protective in a variety of atherosclerotic cardiac and vascular diseases, including acute myocardial infarction (MI), ischemic stroke, non-ST elevation acute coronary syndrome (NSTE-ACS), stable angina, and peripheral artery disease (PAD). Although aspirin is the mainstay of antiplatelet therapy, an adjunctive second antiplatelet agent is often used to produce additional benefits in some clinical circumstances [1]. The oral antiplatelet drugs, including ticlopidine, clopidogrel, prasugrel, and ticagrelor, act on the platelet P2Y12 receptor to inhibit platelet activation and aggregation. More than 600,000 patients received coronary stents in the United States in 2017, necessitating the use of antiplatelet agents [2]. Thus a thorough understanding of the various platelet inhibitors, their mechanism of action, and their indications is paramount.

            The P2Y12 Receptor

            Thienopyridines (ticlopidine, clopidogrel, and prasugrel) bind to the extracellular nucleotide P2Y12 receptor, found on the cell surface of platelets, glial cells, and vascular smooth muscle cells, to inhibit platelet aggregation. When activated by its ligand, adenosine diphosphate (ADP), the Gi protein–coupled P2Y12 receptor initiates an intracellular cascade that culminates in amplified platelet aggregation (Figure 1). In response to ADP binding, the Gi protein releases alpha and beta subunits that serve specific roles in the intracellular process [3]. The alpha subunit inhibits adenylyl cyclase, a protein responsible for the production of cyclic adenosine monophosphate (cAMP). Decreased cAMP production leads to decreased phosphorylation of vasodilator-stimulated phosphoprotein (VASP). The dephosphorylated form of VASP serves to activate the glycoprotein IIb/IIIa receptor, a key component of platelet aggregation. Simultaneously, the beta subunit activates phosphatidylinositol 3-kinase. This enzyme prompts conformational activation of integrin αIIbβ3, a cell adhesion molecule important in platelet aggregation [4]. Phosphatidylinositol 3-kinase also activates the serine/threonine protein kinase B and Rap1b guanosine triphosphate binding proteins, which, in turn, amplify the glycoprotein IIb/IIIa receptor to potentiate platelet aggregation. Furthermore, the beta subunit prompts release of platelet granules, which contain platelet agonists such as ADP, fibrinogen, factor V, and P-selectin [5]. Through this proinflammatory cascade, activation of the P2Y12 receptor enhances platelet aggregation, prompts granule release, and increases the procoagulant/prothrombotic milieu.

            Figure 1

            ADP-Receptor Antagonists Bind Irreversibly (Clopidogrel and Prasugrel) or Reversibly (Ticagrelor and Cangrelor) to the P2Y12 Receptor, Thereby Inhibiting Ca2+ Mobilization and Activation of the GPIIb/IIIa Receptor.

            Inhibition of Ca2+ mobilization reduces secretion of pro-aggregetory substances and activation of the GPIIb/IIIa receptor, preventing conformational changes that result in platelet aggregation.

            Thienopyridines

            Ticlopidine

            Ticlopidine, the first compound of this class, came to the market in 1979 for stroke prevention. However, the medication is fraught with severe side effects, including gastrointestinal disturbances (vomiting, diarrhea, dyspepsia), rashes (purpura), hyponatremia, nephrotic syndrome, hepatotoxicity, neutropenia, and thrombocytopenia [6]. Since the introduction of newer agents, it has been largely supplanted by clopidogrel. As the drug is rarely used in modern clinical practice, it will not be discussed further in this review.

            Clopidogrel
            Pharmacology

            Introduced in the United States in 1998, the oral P2Y12 inhibitor clopidogrel bisulfate (Figure 1), a thienopyridine, is ingested as an inactive prodrug. Approximately 50% of this compound is absorbed in the gastrointestinal tract by the drug efflux transporter P-glycoprotein (encoded by the ABCB1 gene). The inactive metabolite is then delivered to the liver for further processing. In the liver, up to 85% of the delivered clopidogrel is hydrolyzed during first-pass metabolism by carboxylesterase 1 [7]. This converts the prodrug into an inactive carboxylic acid derivative that is excreted mainly via the renal (50%) and gastrointestinal (46%) systems. The remaining clopidogrel is metabolized via a group of cytochrome 450 enzymes (CYP) into its active form in a two-step process. In the first step, CYP1A2, CYP2B6, and CYP2C19 convert clopidogrel into 2-oxoclopidogrel, a still inactive intermediate compound. This compound is then converted to the clopidogrel active metabolite (R-130964) by CYP2B6, CYP2C9, CYP2C19, and CYP3A4. CYP2C19 is the most active enzyme of this group, accounting for approximately 50% of the clopidogrel active metabolite that is produced [8]. Importantly, both 2-oxoclopidogrel and the active metabolite are hydrolyzed by carboxylesterase 1, such that approximately 2% of the ingested prodrug form of clopidogrel enters the systemic circulation in the active form. The prodrug form of clopidogrel has a half-life of approximately 7–8 h, while the active metabolite has a half-life of only 30 min; however, because of its irreversible binding to the P2Y12 receptor, the drug persist for the lifespan of the affected platelet, approximately 7 days [9]. Most (>95%) of this circulating clopidogrel, including both the active metabolite and the inactive metabolite, is protein bound. Thus any factors affecting protein binding, including the relative concentration of active/inactive metabolites, pH, and albumin concentration, can affect the bioavailability of active clopidogrel. Furthermore, the extensive hepatic metabolism of clopidogrel by the inducible CYP system leads to many potential drug-drug interactions. The active metabolite binds irreversibly [10] to the P2Y12 receptor by forming disulfide bonds [11] with cysteine residues at the ADP-binding site, leaving the receptor nonfunctional for the remainder of the platelet lifespan (up to 9 days) [12]. In addition to direct platelet inhibition, clopidogrel also decreases the levels of markers of inflammation, with decreased production of C-reactive protein, CD40 ligand, and P-selectin (CD62) [13].

            Of the important attributes of antiplatelet agents, the time from administration to effective platelet inhibition is paramount. In patients with acute coronary syndrome (ACS), particularly those with ST elevation MI (STEMI), the time required to achieve adequate platelet inhibition is crucial to prevent complications such as early drug-eluting stent thrombosis. Platelet inhibition by clopidogrel is dose and time dependent; when clopidogrel is administered as a standard 75 mg dose, the time to initial activity of the active metabolite is 2–4 h, achieving a peak effect as late as 3–7 days [14]. In practice, clopidogrel is typically administered with a loading dose of either 300 or 600 mg to achieve platelet inhibition as rapidly as possible. Assessment of platelet reactivity by means of the maximal intensity of ADP-induced platelet aggregation suggests a peak effect at 3–5 h with a dose of 300 mg [14]. After the administration of 600 mg, the peak effect occurs at 2–3 h [15]. The dose-effect relationship with clopidogrel is nonlinear, with no incremental benefit to loading doses greater than 600 mg [16]. Studies in patients undergoing percutaneous coronary intervention (PCI) for stable coronary artery disease (CAD) or ACS (both non–ST elevation MI [NSTEMI] and STEMI) had reduced major adverse cardiovascular events and infarct size with a loading dose of 600 mg, driven primarily by a decrease in stent thrombosis. However, there was a signal for increased major bleeding with high-dose clopidogrel [1723].

            Pharmacogenetics

            The maximum effect of clopidogrel appears to be 50–60% platelet inhibition, although the individual’s response differs on the basis of a number of patient-specific factors. Up to 30% of white patients and 50% of Asian patients have an attenuated response to clopidogrel administration (with higher rates in people with diabetes), resulting in high on-treatment platelet reactivity, a phenomenon dubbed clopidogrel resistance [3, 2426]. Platelet activity can be assessed by a number of different methods, although the gold standard is light transmittance aggregometry in response to ADP [27, 28]. By aggregometry, an appropriate response to clopidogrel administration is defined as a more than 20% decrease in maximal platelet aggregation after administration of the P2Y12 agonist ADP. Clopidogrel resistance, then, is defined in the laboratory as a decrease in maximal aggregation of 10% or more from the baseline [29]. However, because of the cumbersome nature of this test, the more commonly used methods involve bedside evaluation with commercially available tests: flow-cytometric analysis of VASP; the PFA-100® analyzer (Siemens Healthcare Diagnostics, Deerfield, IL, United States) looks at platelet aggregation in response to collagen/epinephrine or collagen/ADP cartridges under high-shear conditions; the VerifyNow® P2Y12 test (Accumetrics, San Diego, CA, USA) looks at platelet aggregation of fibrinogen-coated beads in response to ADP [3032]. With these tests, clopidogrel resistance is defined as platelet reactivity index greater than 50% by analysis of VASP phosphorylation and greater than 240 P2Y12 reaction units by the VerifyNow assay [33]. Importantly, laboratory evidence of decreased clopidogrel response does not necessarily translate to clinical treatment failure; in practice, clopidogrel resistance is defined as recurrent thrombotic events despite appropriate clopidogrel therapy, suggesting high on-treatment platelet activity.

            There are several potential patient-specific mechanisms for decreased platelet inhibition in response to clopidogrel, including drug-drug interactions, variable absorption/metabolism of the prodrug, alternative pathways for platelet activation, and genetic polymorphisms [34].

            Clopidogrel and prasugrel (a third-generation thienopyridine) are subject to intestinal efflux by the ATP-dependent P-glycoprotein pump (also known as multidrug resistance protein 1), encoded by the ABCB1 gene [35]. P-glycoprotein [1] limits the oral bioavailability of these drugs via control of intestinal absorption. Accordingly, individuals with the C3435 TT genotype, which results in impaired function of the intestinal drug-efflux transporter [36, 37], have lower circulating level of the active metabolite, with the potential for increased risk of major adverse cardiovascular events [38], although this remains controversial [39, 40].

            Furthermore, the CYP system is susceptible to genetic polymorphisms that affect thienopyridine metabolism, with more than 48 CYP2C19 allelic variants described, ranging from null to increased enzymatic activity [41]. The two most common genetic variants responsible for decreased platelet inhibition are loss of function (LoF) in the CYP2C19 and CYP3A4/CYP3A5 genes, resulting in so-called poor metabolizers [3, 42]. As noted earlier, CYP2C19 is active in both of the sequential steps required to convert clopidogrel into its active metabolite. Reduced CYP2C19 activity results in shunting of clopidogrel toward esterification, increasing production of the inactive metabolite. Thus LoF results in a 32% reduction in the active metabolite and 9% reduction in maximal platelet aggregation [36, 43]. Together, the LoF CYP2C19 polymorphisms cause 12% of the variation of platelet inhibition seen in patients receiving clopidogrel [44]. In this setting, the specific CYP2C19*2 mutation accounts for most (>90%) of those with the reduced function allele [45]. This allele is present in 15% of white and black patients, and up to 35% of Asians, suggesting a large potential clinical impact [46]. CYP2C19 LoF mutations are associated with increased death from cardiovascular causes, nonfatal MI, and nonfatal stroke [26, 45, 4749]. The risk associated with LoF CYP2C19 variants increases significantly in patients undergoing PCI, who have an almost three-fold increase in stent thrombosis [43, 50, 51].

            The most common polymorphism responsible for a gain of function is the CYP2C19*17 variant, which is associated with enhanced enzyme activity [52, 53]. These so-called fast metabolizers have greater reduction in platelet aggregation and a resultant decrease in potential thrombotic events, but at the cost of increased bleeding [36, 54].

            Other mutations in the CYP 450 system have been postulated to contribute to clopidogrel resistance, with CYP3A4 inhibitors leading to decreased production of the clopidogrel active metabolite [55]. However, studies do not support a clinically significant role for these mutations in causing adverse cardiovascular events [56], although inducers of CYP3A4 may potentiate active metabolite formation, resulting in increased efficacy, but, again, at the cost of increased bleeding [55].

            More than 50% of the population is affected by either ABCB1 or CYP2C19 polymorphisms, raising concern for the potential risk of major adverse cardiovascular events due to high platelet reactivity while patients are receiving thienopyridines [4, 38]. Because of the risk for variable platelet inhibition and potential adverse clinical outcomes, platelet function testing to guide the choice of P2Y12 inhibitor has a class IIb recommendation for selected, high-risk patients undergoing PCI [5759]. Otherwise, current guidelines note that the routine use of platelet function and genetic testing is not recommended (class III: no benefit) [57]. For these reasons, the black box warning on the package insert for clopidogrel warns that “patients homozygous for nonfunctional alleles of CYP2C19 gene (CYP2C19 poor metabolizers) formed less active metabolite and had reduced antiplatelet activity at recommended doses; CYP2C19 genotype tests are available to identify CYP2C19 poor metabolizers; consider using other platelet P2Y12 inhibitor treatment in CYP2C19 poor metabolizers” [60]. However, it is important to remember that not all cases of laboratory resistance translate to clinical resistance.

            Drug-Drug Interactions

            Ingested as a prodrug, clopidogrel must undergo enzymatic conversion to its active form, requiring the complex pathway described earlier. Each of these steps is susceptible to interaction with commonly used medications, both cardiac and noncardiac. The CYP system is particularly susceptible to inducers and inhibitors of its function, with an extensive list of agents that can affect the clinical response to thienopyridines (Table 1). The most commonly used drugs with the potential to inhibit the effects of clopidogrel include proton pump inhibitors (PPIs), 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase) inhibitors (statins), and calcium channel blockers (CCBs).

            Table 1

            Antiplatelet Drug Comparison Chart.

            DrugASAClopidogrel (Plavix®)Prasugrel (Effient®)Ticagrelor (Brilinta®)
            Indications1° and 2° prevention of stroke and MI
            – ACS
            – PCI with stent
            – PVD
            – ASA intolerance or failure
            – 1° and 2° preventionof stroke and MI (±ASA)
            – ACS (+ ASA)
            – PCI (+ ASA)
            – PVD
            – With ASA, for treatment of ACS in patients treated with PCI
            Contraindicated if:
            age >75 years; OR
            wt <60 kg; OR
            history of stroke
            NON-FORMULARY
            – With ASA, for treatmentof ACS
            See BCHA restrictions below1
            Dose and durationLoad: 160–325 mg
            Maintenance: 80 or 81 mg daily
            Duration: Indefinite
            Load: 300–600 mg
            Maintenance: 75 mg daily
            Duration:ACS: up to 1 year
            BMS: minimum 30 days
            DES: minimum 1 year
            Load: 60 mg
            Maintenance: 10 mg daily
            Duration: up to 1 year
            Load: 180 mg
            Maintenance: 90 mg BID
            Duration: up to 1 year
            ClassNon-steroidal
            Anti-inflammatory agent
            Second generation thienopyridine (Prodrug)Third-generation thienopyridine (Prodrug)Cyto-pentyl-triazolopyrimidine
            Mechanism of platelet inhibitionIrreversible inhibitor of COX-1 causing decrease in thromboxane A2Irreversible inhibitor of P2Y12 component of ADP receptor (preventing ADP binding and activation of platelets)Irreversible inhibitor of P2Y12 component of ADP receptor (preventing ADP binding and activation of platelets)Reversibly modifies P2Y12 component of ADP receptor (preventing ADP binding and activation of platelets)
            Oral bioavailability50–75%>50% (active metabolite)>78% (active metabolite)30–42%
            Peak effect1–3 h6 h (after load)4 h (after load)2 h (after load)
            Half-life (active metabolite)3 h (salicylate)0.5 h7 h (range 2–15 h)9 h (range 6.7–9.1 h)
            EliminationHydrolyzed by esterases; Hepatic conjugationEsterases;Metabolism by CYP-450 enzymesEsterases;Metabolism by CYP-450 enzymesMetabolism by CYP-450 enzymes
            CYP metabolismNoCYP2C19CYP3A4, CYP2B6CYP3A4/5
            When to hold dose prior to surgery7 days (optional)5–7 days7 days5 days

            1Ticagrelor restricted to patients on prior to admission or those on ASA with ACS i.e. STEMI, non-STEMI, unstable angina (UA) AND one of the following: Failure on optimal doses of clopidogrel and ASA therapy or recurrent ACS after revascularization with PCI; OR STEMI and undergoing revascularization via PCI; OR Non-STEMI or UA and high risk angiographic anatomy and undergoing revascularization via PCI.

            ACS, acute coronary syndrome; PCI, percutaneous coronary intervention; PVD, peripheral vascular disease; BMS, bare metal stent; DES, drug-eluting stent; ADP, adenosine diphosphate.

            Table reproduced courtesy of Vancouver Acute Pharmaceutical Sciences, Vancouver, BC 2016, Canada. See ref [61].

            Proton Pump Inhibitors

            To reduce the risk of gastrointestinal bleeding in patients receiving antiplatelet therapy, PPIs are often prescribed. Omeprazole, the most widely prescribed PPI, is ingested as a prodrug, requiring activation by CYP2C19 and CYP3A4/CYP3A5, similarly to clopidogrel [62]. Of the PPIs, omeprazole has a greater affinity for CYP2C19 than for CYP3A4/CYP3A 5 than other PPIs, suggesting a greater potential for interaction with clopidogrel and the potential safety of medications such as pantoprazole or esomeprazole [63]. Accordingly, pharmacodynamic studies demonstrated decreased effectiveness of clopidogrel, but this finding was not consistent with the findings of observational studies [64]. However, a recent meta-analysis evaluating studies since 2012 suggested increased rates of major adverse cardiac events following PCI in patients taking omeprazole (but not pantoprazole) [65]. In the Clopidogrel and the Optimization of Gastrointestinal Events Trial (COGENT), the only randomized, double-blind study to evaluate the cardiovascular implications of combining clopidogrel and omeprazole, patients treated with this combination had a lower incidence of gastrointestinal bleeding, with an incidence of cardiovascular events similar to that with clopidogrel and placebo [66]. However, there were several limitations to this study, including premature termination, smaller than expected number of events, wide confidence intervals, and small percentage of patients with the CYP2C19 homozygous mutation, and definitive conclusions about this interaction could not be made [66]. In light of these limitations, the Food and Drug Administration affirmed the warning on the clopidogrel label that concomitant use of clopidogrel and omeprazole should be avoided [67]. Further randomized controlled trials are needed to elucidate this issue.

            HMG-CoA Reductase Inhibitors

            To mitigate the risk of atherosclerotic cardiovascular events, statin medications, which inhibit HMG-CoA reductase so as to reduce endogenous cholesterol production, are commonly prescribed to patients with CAD or PAD. Several statins, including atorvastatin, simvastatin, lovastatin, and fluvastatin, are metabolized by CYP3A4/CYP3A5, and can theoretically interact with the metabolism of clopidogrel [68]. In 2003, Lau et al. [69] studied a potential interaction between atorvastatin and clopidogrel, and they concluded that competitive inhibition of CYP3A4/CYP3A5 by atorvastatin decreased the activation of clopidogrel, resulting in decreased platelet inhibition. However, several subsequent studies evaluated the interaction of the statins and clopidogrel, revealing no significant change in platelet inhibition or cardiovascular outcomes, especially when the higher (600 mg) loading dose is used [7076]. For this reason, there are currently no contraindications to statin use in patients taking clopidogrel (Table 2).

            Table 2

            Advantages and Disadvantages of the P2Y12 Receptor Inhibitors.

            Pros of choosing ticagrelorCons of choosing ticagrelor
            Rapid and extensive platelet inhibitionExpensive
            Reversible inhibition and rapid offset of effectIncreased risk of non-CABG surgery bleeding vs clopidogrel
            Less susceptible to genetic variation and drug-drug interactionsTwice daily dosing
            Reduction in ischemic event rates vs clopidogrelDyspnea and ventricular pauses
            Similar overall bleeding risk vs clopidogrel
            Proven efficacy regardless of treatment strategy

            Pros of choosing prasugrelCons of choosing prasugrel

            Rapid and extensive platelet inhibitionExpensive
            Less susceptible to genetic variation and drug-drug interactionsHigh risk of major bleeding, especially in patients undergoing CABG surgery
            Reduction in ischemic event rates following PCI vs clopidogrelCoronary anatomy should be defined by angiography before initiation
            Greatest efficacy in patients with diabetes and STEMIQuestionable utility in patients undergoing procedures other than PCI
            Once-daily dosing

            Pros of choosing clopidogrelCons of choosing clopidogrel

            AffordableResponse variability, with a poor response associated with increased risk of thrombosis
            Long history of useSusceptible to genetic variation and drug-drug interactions
            Only agent with proven efficacy in patients undergoing thrombolysisQuestions regarding appropriate dosing
            Once-daily dosing

            CABG, coronary artery bypass graft; PCI, percutaneous coronary intervention; STEMI, ST elevation myocardial infarction. From Ref. [77].

            Calcium Channel Blockers

            Frequently used in the management of hypertension, atrial fibrillation, and angina, CCBs can inhibit CYP3A4/CYP3A5, resulting in decreased concentration of the clopidogrel active metabolite and subsequent high on-treatment platelet activity [78]. Many CCBs also inhibit P-glycoprotein, thereby decreasing clopidogrel efflux and resulting in higher circulating levels of the drug, attenuating the effect of CYP3A4/CYP3A5 inhibition. However, amlodipine, which does not inhibit P-glycoprotein, results in both decreased clopidogrel activation and persistent intestinal efflux, decreasing bioavailability of the active form [79]. The dihydropyridine class of CCBs have been implicated in this interaction, with several studies suggesting decreased platelet inhibition in patients receiving clopidogrel and concomitant CCBs [8082]. However, there are conflicting recent studies regarding the clinical impact of CCBs (including amlodipine) on clopidogrel metabolism, with no consensus reached thus far [8385]. Thus there are no significant restrictions on the use of CCBs with clopidogrel.

            Azoles

            The azole antifungal drugs, used for both treatment and prophylaxis of fungal infections in immunosuppressed patients, such as organ transplant recipients or patients with human immunodeficiency virus. These medications inhibit the CYP3A4/CYP3A5 isozyme, particularly ketoconazole, although the more commonly used medications are fluconazole and itraconazole [86]. Administration of ketoconazole leads to impaired platelet inhibition in patients taking clopidogrel, although there are no descriptions of clinically significant clopidogrel resistance in patients taking azole antifungals [87].

            Clinical Outcomes
            Coronary Artery Disease

            The clinical efficacy of clopidogrel was first established by the Clopidogrel versus Aspirin in Patients at Risk of Ischemic Events (CAPRIE) trial in 1996, which evaluated 19,195 patients with recent stroke, or established PAD [88]. In this study, clopidogrel monotherapy versus aspirin monotherapy was associated with a significant 8.7% relative risk reduction compared with aspirin for the primary end point of ischemic stroke, MI, or vascular death (5.32% per year compared with 5.83% per year; P = 0.043). However, this benefit was most pronounced in the PAD subgroup, while the benefit in the stroke subgroup was not statistically significant; risks of hemorrhage were similar between the groups. This was followed by a comparison of clopidogrel versus ticlopidine in patients with ACS or patients undergoing PCI, demonstrating comparable efficacy and a better safety profile, including less bleeding, neutropenia, and thrombotic thrombocytopenic purpura [8991]. The Clopidogrel in Unstable Angina to Prevent Recurrent Events (CURE) study firmly established the role of aspirin plus clopidogrel in the medical management of ACS, while the PCI substudy supported clopidogrel pretreatment followed by long-term therapy, each showing a reduction in major cardiovascular events at the cost of increased bleeding [92, 93]. Several subsequent trials furthered the role of long-term aspirin plus clopidogrel therapy after ACS, demonstrating a reduction in the relative risk of death, MI, and stroke after PCI [94, 95] or fibrinolysis [96]. In the CHARISMA trial, the use of dual antiplatelet therapy (DAPT) including aspirin and clopidogrel did not, however, show significant benefit in a broad population of patients at high-risk of cardiovascular events, including those with stable CAD [97]. However, post hoc analysis revealed a signal for benefit in patients with symptomatic atherothrombotic disease, at the cost of increased bleeding.

            With the efficacy of this clopidogrel established in ACS and stroke, trials began to focus on the optimal timing of clopidogrel administration and dose of clopidogrel. In patients undergoing PCI, a double dose regimen, including a 600 mg loading dose of clopidogrel followed by 150 mg for 1 week, reduced cardiovascular events and stent thrombosis when compared with the standard dose (300 mg load, 75 mg daily) [23]. However, extension of this double dose out to 6 months did not improve outcomes further [98]. The Atorvastatin for Reduction of Myocardial Damage during Angioplasty (ARMYDA)-2 trial investigated the dose-time relationship for clopidogrel in patients with stable CAD or NSTE-ACS about to undergo PCI. A significant decrease in major adverse cardiovascular events was discovered with the higher loading dose (600 mg) without an increase in major bleeding. Subsequent ARMYDA trials concluded that patients receiving long-term clopidogrel therapy with stable angina could safely undergo PCI without a reload, that patients receiving long-term clopidogrel therapy with ACS undergoing PCI had better outcomes with a 600 mg reload, and that giving the clopidogrel loading dose “in the lab” just before PCI was as safe and effective as a preload (6 h before PCI) [99101]. In STEMI patients, pretreatment with the 600 mg clopidogrel loading dose before primary PCI was associated with a reduction in infarct size and 30 day major adverse cardiovascular events, while the 150 mg maintenance dose caused more potent platelet inhibition and decreased inflammation [21, 102]. In this setting, in patients with ACS, the current American College of Cardiology (ACC)/American Hear Association (AHA) guidelines recommend a clopidogrel 300 mg bolus and 75 mg maintenance dose for 1 year if ACS is managed medically or with fibrinolytics, versus a 600 mg bolus and 75 mg maintenance dose for 1 year if it is managed with PCI [103].

            Ischemic Stroke

            Building on the results of the CAPRIE trial, multiple trials evaluated the efficacy and safety of clopidogrel in noncardioembolic stroke or transient ischemic attack (TIA), demonstrating that clopidogrel and aspirin were superior to aspirin alone if used early (<24 h to 21 days) in ischemic stroke, but with an increased risk of bleeding [104]. This confirms the findings of previous studies looking at ischemic stroke in patients with symptomatic carotid disease [105107]. However, in the CHARISMA trial, aspirin plus clopidogrel did not reduce the risk of the composite primary end point (MI, stroke of any cause, or death from cardiovascular causes) compared with aspirin alone, and the risk of bleeding was higher [97]. Subsequent trials confirmed the benefit from upfront DAPT using aspirin and clopidogrel, with no benefit and potential increased harm with long-term use [108, 109]. The role of DAPT in patients with ischemic stroke will be further evaluated in the upcoming Platelet-Oriented Inhibition in New TIA and Minor Ischemic Stroke (POINT) trial, a randomized, double-blind, multicenter clinical trial to determine whether clopidogrel at 75 mg/day (after a loading dose of 600 mg) is effective in improving survival free from major ischemic vascular events when treatment is initiated within 12 h in patients receiving aspirin [110]. Thus current guidelines offer a class IIb recommendation for the use of clopidogrel at 75 mg/day with aspirin for up to 90 days in patients with recent stroke or TIA (within 30 days) attributable to severe stenosis (70–99%) of a major intracranial artery, with the caveat that long-term use of DAPT with clopidogrel plus aspirin (i.e., >90 days) is not recommended because of the increased risk of bleeding and all-cause mortality, unless there is another indication (e.g., coronary stent). Furthermore, the guidelines offer a class IIa recommendation for clopidogrel monotherapy in place of aspirin monotherapy [111].

            Peripheral Artery Disease

            PAD is an independent risk factor for CAD, and both entities have atherothrombotic complications [112]. Management strategies include medical therapy, balloon angioplasty, and stent placement, with various strategies regarding the management of antiplatelet therapy. In the MATCH trial (aspirin and clopidogrel compared with clopidogrel alone after recent ischemic stroke or TIA in high-risk patients), PAD patients in the high-risk category derived significant cardiovascular risk benefit with DAPT with clopidogrel [108]. However, when clopidogrel therapy was added to aspirin therapy in PAD patients in the CHARISMA trial, they did not have significant improvement in the primary efficacy end point of MI, stroke, or vascular death, although those at highest risk (i.e., established PAD) did see a decrease in the rate of hospitalization for ischemic events, including MI [97, 113]. In the aforementioned CAPRIE trial, the benefit of clopidogrel over aspirin in terms of cardiovascular risk reduction was driven predominantly by the PAD subgroup [88]. On the basis of these results, the ACC/AHA guidelines offer a class I recommendation for the use of clopidogrel as a safe and effective alternative antiplatelet therapy to aspirin to reduce the risk of MI, ischemic stroke, or vascular death in individuals with symptomatic atherosclerotic lower extremity PAD. Even in asymptomatic individuals, antiplatelet therapy has a class IIa indication in this group [114].

            Atrial Fibrillation

            In the population of patients with an indication for DAPT and anticoagulation (i.e., CAD and atrial fibrillation), triple therapy with aspirin, clopidogrel, and warfarin incurs an up to threefold increase in bleeding risk, which portends worse prognosis and predicts increased mortality in patients after PCI [115117]. There is potentially a need for triple therapy in at least 5% of patients undergoing PCI, and the incidence of atrial fibrillation is expected to increase in the coming years [103]. The WOEST trial (use of clopidogrel with or without aspirin in patients taking oral anticoagulant therapy and undergoing PCI) evaluated triple therapy with DAPT and warfarin versus clopidogrel monotherapy and warfarin in patients undergoing PCI [118]. This small trial of 563 patients found that the warfarin plus clopidogrel alone (double therapy) was associated with a significant reduction in bleeding complications compared with warfarin plus DAPT (triple therapy), without an increase in thrombotic complications. The findings of this study were further bolstered by registry data [119, 120]. However, the most recent ACC/AHA NSTE-ACS guidelines continue to recommend triple therapy in patients with NSTE-ACS [121], although the 2014 atrial fibrillation guidelines offer a class IIb recommendation for double therapy in this setting [122]. With the advent of direct oral anticoagulants (dabigatran, rivaroxaban, apixaban, edoxaban) and their burgeoning use in clinical practice, the question of anticoagulation in patients with atrial fibrillation undergoing PCI must be considered in this context. Akin to the WOEST trial, investigators looked at rivaroxaban plus P2Y12 inhibitor therapy versus traditional triple therapy in atrial fibrillation patients undergoing coronary revascularization, discovering that a rivaroxaban-based strategy was associated with a lower frequency of clinically significant bleeding, all-cause death, and rehospitalization, with similar rates of major adverse cardiac events, including stent thrombosis [123]. Similarly, investigators evaluated double therapy with dabigatran and clopidogrel versus traditional triple therapy, finding that dual therapy with dabigatran was superior to triple therapy with warfarin in preventing bleeding, without an increase in the risk of death, MI, or stroke [124]. On the basis of the findings of these trials, the 2017 European Society of Cardiology guidelines recommend dual therapy with an oral anticoagulant after 1 month of triple therapy in patients with high bleeding risk, adding that direct oral anticoagulants may be considered in place of warfarin [125]. Importantly, all patients in the WOEST trail, more than 90% of patients in the PIONEER AF-PCI trail, and more than 80% of patients in the RE-DUAL PCI trial were taking clopidogrel as the P2Y12 inhibitor of choice, suggesting a significant role for clopidogrel in patients with atrial fibrillation undergoing PCI [118, 123, 124].

            After Transcatheter Aortic Valve Replacement

            Antiplatelet therapy has been a cornerstone of postprocedure management for patients undergoing transcatheter aortic valve replacement (TAVR) since the early clinical trials. In the United States, current practice involves upfront DAPT with clopidogrel at a dose of 75 mg for 6 months with balloon-expandable valves and for 3 months with self-expanding valves, along with aspirin at a dose of 75–100 mg lifelong [126]. This recommendation arises from the original TAVR approval data, likely due to the standard clinical practice for other endovascular stents [127132]. To better evaluate the role of antiplatelet therapy after TAVR, the Aspirin Versus Aspirin + Clopidogrel following Transcatheter Aortic Valve Implantation (ARTE) trial randomized 222 patients presenting for TAVR with the Edwards balloon-expandable valve to receive either aspirin alone or aspirin plus clopidogrel following the TAVR procedure. At 3 months, those receiving DAPT were more likely to experience a primary end point event (death, MI, stroke/TIA, or major/life-threatening bleeding) than those receiving aspirin alone (15.3 vs. 7.2%; P = 0.065), driven primarily by increased major and minor bleeding [133]. This is particularly important in the setting of recent data suggesting rates of subclinical leaflet thrombosis approaching 15%, with no significant difference between DAPT and single antiplatelet therapy [134, 135]. Furthermore, severer, clinically manifest leaflet thrombosis may be associated with stroke, cardiogenic shock, and death [136], emphasizing the need for further research. To better understand the role of antiplatelet therapy after TAVR, two larger-scale trials, POPular-TAVI and CLOE, will evaluate aspirin therapy alone versus aspirin and clopidogrel therapy in the first 3 months after TAVR [137]. Notably, anticoagulation therapy appears to more effectively prevent leaflet thrombosis in comparison with antithrombotic therapy, an issue that will be addressed by the upcoming AUREA (vitamin K antagonist), GALILEO (rivaroxaban), and ATLANTIS (apixaban) trials.

            Triple Antiplatelet Therapy

            Cilostazol is a selective inhibitor of phosphodiesterase type 3 that results in reversible inhibition of platelet aggregation, in addition to vasodilatory and antiproliferative effects [138]. The concept of triple antiplatelet therapy with aspirin, clopidogrel, and cilostazol was intended to bolster antiplatelet activity. This has been studied in the context of both CAD and PAD, with evidence to suggest that the addition of cilostazol to standard therapy with aspirin and clopidogrel significantly decreases platelet activity when compared to standard therapy alone [139141]. Initial work suggested that triple antiplatelet therapy with these agents may decrease the rates of stent thrombosis and target vessel failure without an increased risk of bleeding, both in CAD [141, 142] and in PAD [143145]. However, recent trial data suggest no clinical benefit with regard to the incidence of cardiac death, nonfatal MI, or stroke, even in complex PCI [140, 146, 147]. More research is needed in this area to better adjudicate the role of cilostazol in addition to aspirin and clopidogrel.

            Thus clopidogrel is used as an effective antiplatelet agent in a variety of situations, including ACS, stable CAD, PAD, stroke, after TAVR, and as double therapy for atrial fibrillation patients undergoing PCI. However, the extensive metabolism required for activation potentially limits its efficacy in patients with common genetic polymorphisms (CYP2C19, ABCB1), and drug-drug interactions may play a role, although this requires further investigation. Nonetheless, for the majority of patients requiring antiplatelet therapy beyond aspirin, clopidogrel can be considered a safe first option.

            Prasugrel
            Pharmacology

            Like clopidogrel, the third-generation thienopyridine prasugrel irreversibly binds the P2Y12 receptor and begins as a prodrug that requires transformation to its active form [148]. Prasugrel is administered at an oral loading dose of 60 mg, followed by daily maintenance doses of 10 mg, with the consideration of a 5 mg dose in those weighing less than 60 lb (27 kg) [149]. More than 80% of the prodrug is absorbed in the small intestine, where carboxylesterase 2 hydrolyzes the prodrug to 2-oxoprasugrel [150, 151]. The intermediate compound is then converted to the prasugrel active metabolite in the liver by the CYP system [152]. As opposed to the two-step CYP metabolism of clopidogrel involving CYP2C19, prasugrel is metabolized primarily by CYP3A4/CYP3A5 in one step, making it less susceptible to inactivation and interference from CYP inhibitors [153]. CYP3A4/CYP3A5 accounts of up to 70% of the conversion of 2-oxoprasugrel to the prasugrel active metabolite, with CYP2B6 accounting for up to 26% [154]. Prasugrel metabolism is highly efficient during both absorption and conversion to the active metabolite; the active metabolite is detected within 15 min, the mean time to peak plasma concentration is approximately 30 min, and the median plasma half-life is approximately 7.4 h [155, 156]. The drug is excreted mostly via the renal (70%) and gastrointestinal (30%) systems [87]. Prasugrel reaches higher concentrations of its active metabolite than clopidogrel, with the highest concentration typically found in Asian patients [157]. Given this rapid and efficient activation, prasugrel is tenfold more potent than clopidogrel [158], yielding prompt, irreversible platelet inhibition by forming a disulfide bridge with a free cysteine residue on the P2Y12 receptor [53]. Thus prasugrel administration results in faster, more consistent, and more potent inhibition of platelet aggregation when compared with clopidogrel [159, 160].

            Pharmacogenetics

            The extensive metabolism of prasugrel via CYP3A4 obviates the issue with incomplete response due to CYP2C19 mutations, as this isozyme accounts for only a minority of active metabolite production [151, 161]. Additionally, prasugrel does not have a significant inactivation pathway, resulting in consistent plasma levels of the active metabolite [162]. Accordingly, on-treatment platelet reactivity is much lower for prasugrel when compared with clopidogrel, with a significantly lower incidence of laboratory hyporesponsiveness to the third-generation drug [163]. The aforementioned common functional CYP genetic variants (such as CYP2C19*2) do not affect the prasugrel active metabolite levels, platelet inhibition, or clinical cardiovascular events [164, 165]. As previously mentioned, prasugrel is also subject to intestinal efflux by the ATP-dependent P-glycoprotein pump, encoded by the ABCB1 gene. Unlike clopidogrel, however, the ABCB1 3435 TT genotype was not associated with impaired platelet inhibition of adverse clinical outcomes in patients taking prasugrel [38]. Furthermore, in cases of clinical clopidogrel resistance, changing to prasugrel therapy appears to confer significant benefit in terms of platelet reactivity and major adverse cardiovascular events [38, 160, 164, 165]. Thus although sporadic reports of prasugrel resistance exist, there does not seem to be a clinical benefit to genetic testing in patients taking this drug [33].

            Drug-Drug Interactions

            As prasugrel increases bleeding risk, its use with other antiplatelet/anticoagulant medications, such as warfarin or long-term nonsteroidal anti-inflammatory drugs, may further exacerbate this risk. As CYP3A4/CYP3A5 accounts for most of prasugrel activation, inhibitors or inducers of this enzyme would be expected to impact bioavailability of the antiplatelet agent. However, for most drugs with a potential effect on this enzyme, there does not appear to be a clinically significant interaction. Prasugrel can be safely administered with common inhibitors of CYP3A4/CYP3A5, including statins, CCBs, or PPIs, without concern for impaired platelet inhibition [166]. Ketoconazole, a potent CYP3A4/CYP3A5 inhibitor that causes pharmacologic, although not clinical, clopidogrel resistance, does not appear to have a significant effect on prasugrel [87]. However, the protease inhibitor ritonavir, used commonly in patients with human immunodeficiency virus or hepatitis C virus infection, strongly inhibits both CYP3A and CYP2B6, leading to a significant reduction in prasugrel activation and bioavailability [167]. Although the magnitude of the effect with ritonavir is similar to that with ketoconazole, clinical outcome data are lacking [168]. Thus caution is advised when ritonavir and prasugrel are used in patients with ACS [169].

            Clinical Outcomes

            The phase 2 Joint Utilization of Medications to Block Platelets Optimally (JUMBO)–Thrombolysis in Myocardial Infarction (TIMI) 26 trial evaluated 904 patients undergoing PCI (excluding STEMI and shock). The patients were treated with aspirin at a dose of 325 mg and randomized to receive either clopidogrel (300 mg loading dose followed by 75 mg daily) or prasugrel (40 mg loading dose plus 7.5 mg daily, 60 mg loading dose plus 10 mg daily, or 60 mg loading dose plus 15 mg daily). The primary end point of this study, meant to evaluate safety, found no significant difference in non–coronary artery bypass graft (CABG) bleeding between those receiving prasugrel and those receiving clopidogrel at 30 days. Any bleeding, including TIMI major and minor bleeding, tended to be higher in the high-dose prasugrel group, but the difference between the pooled prasugrel group and the clopidogrel group was not significant [170].

            The Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition with Prasugrel (TRITON)–TIMI-38 and its substudies evaluated 13,608 patients with moderate- to high-risk ACS, including NSTE-ACS [159, 171] and STEMI [172, 173]. In this population, they compared compare clopidogrel (300 mg loading dose and 75 mg daily) versus prasugrel (60 mg loading dose and 10 mg daily) in patients undergoing PCI. Notably, before randomization, the coronary anatomy had to be defined and deemed suitable for PCI. Exclusion criteria included increased risk of bleeding, anemia, thrombocytopenia, known intracranial abnormalities, or the use of thienopyridines in the previous 5 days. Prasugrel proved to be more efficacious, with the primary end point (composite of cardiovascular death, nonfatal MI, or nonfatal stroke) occurring in 12.1% of those receiving clopidogrel, compared with 9.9% of those receiving prasugrel. The prasugrel group also showed a significant reduction in death from cardiovascular causes, urgent target vessel revascularization, or rehospitalization for ischemia. The benefit was seen as early as day 3, and continued through the remainder of study period across all types of ACS [174]. These findings were consistent, regardless of stent type (drug-eluting stent vs. bare metal stent), and the rates of stent thrombosis were also lower with prasugrel (1.13 vs. 2.35%) across all groups [175]. The use of glycoprotein IIb/IIIa inhibitors did not diminish this benefit, as prasugrel significantly reduced the risk of MI, urgent revascularization, and stent thrombosis irrespective of glycoprotein IIb/IIIa inhibitor use [176]. Furthermore, patients with diabetes seemed to derive a greater benefit, with a greater reduction in ischemic events without an observed increase in TIMI major bleeding [177].

            These impressive benefits, however, came with a significant increase in the risk of bleeding for patients taking prasugrel. The initial signal for increased bleeding came from the JUMBO-TIMI 26 trial [170], where there was a nonsignificant trend toward increased TIMI major and minor bleeding. In TRITON-TIMI-38, 2.4% of those taking prasugrel had non-CABG TIMI major bleeding, compared with 1.8% of those treated with clopidogrel, including fatal bleeding (0.4 vs. 0.1%). Additionally, in patients undergoing CABG, there was a significantly higher rate of bleeding in the prasugrel group (13.4 vs. 3.2%) [178]. Risk factors for bleeding included history of TIA/stroke, age more than 75 years, and body weight less than 60 kg, in addition to renal dysfunction and concomitant use of medications that increase the risk of bleeding [179]. Because of the increased bleeding risk, those with a previous TIA/stroke, age more than 75 years, and body weight less than 60 kg had no clinical benefit from prasugrel, especially because of the high morbidity and mortality of bleeding events in older patients [180].

            In patients with ACS who were treated without intervention, there was no clear benefit from the use of prasugrel over clopidogrel. In the Targeted Platelet Inhibition to Clarify the Optimal Strategy to Medically Manage Acute Coronary Syndromes (TRILOGY-ACS) trial, prasugrel did not significantly reduce the frequency of a composite of death from cardiovascular causes, nonfatal MI, or nonfatal stroke in patients younger than 75 years, despite more potent platelet inhibition than clopidogrel [181, 182]. Building on this data, the Testing Platelet Reactivity in Patients Undergoing Elective Stent Placement on Clopidogrel to Guide Alternative Therapy with Prasugrel (TRIGGER-PCI) trial discovered that switching from clopidogrel to prasugrel in patients with high on-treatment platelet reactivity was not a clinically useful strategy in the era of modern drug-eluting stents, despite more potent platelet inhibition [183]. Furthermore, the A Comparison of Prasugrel at the Time of Percutaneous Coronary Intervention or as Pretreatment at the Time of Diagnosis in Patients with Non-ST Elevation Myocardial Infarction (ACCOAST) trial suggested that administration of prasugrel before PCI was not effective in reducing ischemic events and could be harmful because of the increased risk of bleeding compared with the administration of a loading dose at the time of PCI [184].

            Thus the role of prasugrel in PAD or stable CAD is not well established, because of the lack of clinical efficacy and safety data in these conditions [185, 186]. On the basis of these findings, the current ACC/AHA guidelines offer only a class I recommendation for the use of prasugrel in patients presenting with ACS undergoing PCI (after the anatomy has been defined), adding a class IIb recommendation to choose prasugrel over clopidogrel for maintenance P2Y12 inhibitor therapy [103]. Prasugrel is contraindicated in patients with stroke, age more than 75 years, and body weight less than 60 kg. The role of prasugrel in stable CAD, PAD, and patients with atrial fibrillation undergoing PCI remains unclear.

            Cyclopentyltriazolopyrimidines

            Ticagrelor
            Pharmacology

            Ticagrelor is the first of a new class of drugs, known as cyclopentyltriazolopyrimidines, that also inhibit platelet aggregation by binding to the P2Y12 inhibitor [187, 188]. However, there are key differences in terms of its metabolism and mechanism of effect, leading to a drug profile distinct from that of the thienopyridines. Ticagrelor, given with a 180 mg loading dose followed by a 90 mg maintenance dose twice daily, is not a prodrug, and therefore does not require metabolic activation to have a clinical effect [42, 189]. In bypassing the need for hepatic transformation, ticagrelor is able to quickly and consistently inhibit platelet aggregation [190]. The ingested compound is absorbed in the small intestine, and, like clopidogrel, is subject to efflux via P-glycoprotein (encoded by ABCB1) [191]. After absorption, more than 99% of the parent compound binds to plasma proteins, with an estimated bioavailability of 36% [162, 192]. The initial antiplatelet effect occurs within 30 min, reaching the peak effect at approximately 2 h [193]. The mean plasma half-life of ticagrelor is 8 h, with elimination primarily via the gastrointestinal system [194]. Although activation is not required for the platelet inhibitory effect of ticagrelor, it does undergo significant hepatic metabolism via CYP3A4/CYP3A5, leading to potential drug interactions [195]. This produces the active metabolite AR-C124910XX, which is at least as potent an antagonist of the P2Y12 receptor as ticagrelor and is present in the circulation at approximately one-third of the concentration of the parent drug [192, 196]. The active metabolite reaches peak plasma concentration at 3 h, with a mean plasma half-life of 12 h after a loading dose [193]. Together, these compounds exhibit predictable linear pharmacokinetics, resulting in a rapid and potent clinical effect in a dose-dependent manner, achieving 80–90% platelet inhibition at peak concentration [188, 197]. Ticagrelor binds to the P2Y12 receptor at a site distinct from the ADP-binding active site [198]. Rather than competitively inhibiting the ADP-P2Y12 interaction, ticagrelor inhibits conformational change and G protein activation, rendering the receptor locked in the inactive state [4]. This inactive receptor is thus unable to promote the Gi protein–coupled cascade that culminates in platelet aggregation. No dose adjustments are needed on the basis of age or renal function [199, 200]. Given the reversible nature of this drug, discontinuation of use of the drug results in a rapid offset, with platelet function returning to normal within 2–3 days [193].

            Pleiotropic Effects of Ticagrelor

            In addition to the antiplatelet effect of P2Y12 inhibition, ticagrelor also inhibits adenosine reuptake, leading to coronary vasodilation and other potential unintended effects [201]. Ticagrelor is structurally similar to adenosine [202], and inhibits adenosine reuptake by erythrocytes via equilibrative nucleoside transporter 1 [203]. It is postulated that the increase in endogenous adenosine levels also contributes to ticagrelor’s antiplatelet activity [204]. Furthermore, in animal models it was noted that ticagrelor, but not clopidogrel, limited infarct size by augmenting coronary blood flow through vasodilation [205, 206]. This finding was supported by evaluation of adenosine-mediated increases in coronary blood flow in healthy males receiving ticagrelor [204], thought to be due to upregulation of endothelial nitric oxide synthase and increased cAMP levels [206, 207]. Attempts have been made to implicate the increased endogenous adenosine with several of the potential adverse effects associated with ticagrelor, including dyspnea and bradycardia, although the role of this interaction is unclear. Specifically, dyspnea typically abates with continuation of therapy, and most of the bradyarrhythmias appear to be of sinoatrial origin, nocturnal, and typically do not require intervention [202]. There are also reports to suggest other off-target effects, including increased serum creatinine levels, improved endothelial function, and protection from the consequences of pulmonary infection, although debate remains regarding this issue [208].

            Pharmacogenetics

            Unlike clopidogrel, ticagrelor is not subject to significant modulation by the aforementioned common genetic variants that lead to clopidogrel resistance [191, 209]. In several large analyses of CYP2C19 and ABCB1 genetic polymorphisms did not have a significant impact on the pharmacologic or clinical profile of ticagrelor, with no significant effect on platelet inhibition or patient outcomes [210]. Several single nucleotide polymorphisms affecting ticagrelor plasma levels were identified in genome-wide association studies in the Platelet Inhibition and Patient Outcomes (PLATO) trial, although they were not associated with clinical outcomes [211, 212].

            Drug-Drug Interactions

            CYP3A4/CYP3A5 metabolism yields the ticagrelor active metabolite, which accounts for approximately 30% of the circulating drug, raising the concern that CYP3A4/CYP3A5 inhibitors/inducers may affect ticagrelor metabolism and its clinical effect. However, as ticagrelor is an active drug that does not require metabolic transformation to exert its antiplatelet effect, it is less susceptible to drug-drug interactions [213]. For example, coadministration of ticagrelor with atorvastatin or simvastatin does not appear to have any clinically relevant impact on the effect of any of these drugs, although an increase circulating simvastatin level was seen, warranting caution with doses greater than 40 mg [194, 214]. Ticagrelor and its active metabolite are also substrates and inhibitors of P-glycoprotein, responsible for intestinal efflux of multiple drugs, including digoxin. Coadministration of ticagrelor and digoxin leads to increases (up to twofold) in digoxin concentration, warranting close monitoring of digoxin levels when ticagrelor is used [196]. Current recommendations discourage the use of strong CYP3A4 inhibitors (ketoconazole, voriconazole, clarithromycin, protease inhibitors) or inducers (rifampin, dexamethasone, phenytoin, carbamazepine) with ticagrelor, although moderate CYP3A4 inhibitors, such as diltiazem, can be safely coadministered [215, 216].

            Clinical Outcomes
            Coronary Artery Disease

            The phase 2 Dose Confirmation Study Assessing Anti-platelet Effects of AZD6140 vs. Clopidogrel in Non-ST-segment Elevation Myocardial Infarction 2 (DISPERSE-2) trial evaluated the safety and initial efficacy of ticagrelor (90 mg twice daily versus 180 mg twice daily) versus clopidogrel in 990 NSTE-ACS patients already receiving aspirin therapy. The trial showed no difference in major bleeding, but an increase in minor bleeding at the higher dose with a nonsignificant trend toward benefit with regard to the secondary end point, MI. Asymptomatic ventricular pauses longer than 2.5 s were more common with ticagrelor, particularly at 180 mg twice daily. Importantly, the trial demonstrated a decrease in major bleeding for patients undergoing CABG 1–5 days after stopping use of the drug, when compared to clopidogrel [217]. More recent work demonstrates that the 180 mg loading dose led to more prompt and potent platelet inhibition [218]. The ONSET/OFFSET study evaluated the onset and offset of platelet inhibition with ticagrelor (180 mg loading dose) versus clopidogrel (600 mg loading dose) in 123 patients with stable CAD. A more rapid onset of platelet inhibition was seen with ticagrelor compared with clopidogrel as early as 30 min, with higher platelet inhibition in the maintenance therapy phase as well. Platelet inhibition was more consistent for patients receiving ticagrelor, with 90% achieving greater than 70% inhibition of platelet activity. Finally, a faster offset rate was observed after the last dose of ticagrelor than for clopidogrel [193].

            The phase 3 PLATO trial and its substudies evaluated 18,624 patients with ACS (NSTE-ACS or STEMI) randomized to receive ticagrelor (180 mg loading dose followed by 90 mg twice daily) versus clopidogrel (300 mg loading dose followed by 75 mg daily), regardless of an invasive or conservative management strategy, representing an all-comers ACS population. In patients undergoing CABG, it was recommended that use of the study drug be withheld for 5 days in the clopidogrel group and for 24–72 h in the ticagrelor group. All patients received aspirin with a 325 mg loading dose (if thy were not already receiving the medication) followed by 75–100 mg daily. The primary efficacy end point was the composite of cardiovascular death, MI, and stroke; the primary safety end point included major bleeding defined according to study criteria. Ticagrelor was more effective than clopidogrel at reducing the primary outcome, driven by significant reductions in cardiovascular death and MI, with a significant 27% reduction in stent thrombosis. However, there was a statistically significant increase in non-CABG major bleeding and fatal intracranial bleeding, although net major (TIMI and by study criteria), life-threatening, and fatal bleeding was not statistically significant. Importantly, no specific subgroups had a higher rate of bleeding, including those with stroke [219]. PLATO substudies confirmed that this ischemic benefit was consistent in patients with STEMI, NSTE-ACS, planned invasive strategy, or noninvasive strategy, and in patients who underwent CABG, without significant increased bleeding [220224]. Furthermore, the results held in patients with chronic kidney disease, in patients with diabetes, and in smokers, irrespective of age or angiographic outcome [225229]. Further analysis revealed a reduction in first and subsequent recurrent cardiovascular events, when compared with clopidogrel [230]. However, patients from North America experienced a reduced benefit with ticagrelor over clopidogrel, thought to be due to higher doses of aspirin in that group, although the cause of this interaction remains uncertain [231].

            The subsequent Response to Ticagrelor in Clopidogrel Nonresponders and Responders and Effect of Switching Therapies (RESPOND) trial found that the antiplatelet effect of ticagrelor was consistent regardless of responsiveness to clopidogrel [232]. In 2015, the Prevention of Cardiovascular Events in Patients with Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin (PEGASUS)-TIMI-54 trial demonstrated that patients with prior MI receiving aspirin therapy benefitted from the addition of ticagrelor, which led to a reduction in the risk of cardiovascular death, MI, or stroke when compared with placebo. In this trial, ticagrelor was associated with an increase in TIMI major bleeding, but not intracranial hemorrhage [233]. The A 30 Day Study to Evaluate Efficacy and Safety of Pre-hospital vs. In-Hospital Initiation of Ticagrelor Therapy in STEMI Patients Planned for PCI (ATLANTIC) assessed the benefit of pretreatment with ticagrelor compared with placebo in STEMI patients, with co-primary end points of (1) ST-segment resolution on admission and (2) TIMI III flow before PCI. The trial demonstrated lack of superiority with ticagrelor, potentially due to the small difference in the time of administration (only 30 min) between the two groups. Importantly, more deaths occurred in the ticagrelor group (1.1 vs. 0.2%; P = 0.048) [234, 235].

            As noted in the aforementioned studies, ticagrelor is associated with adverse events, including bleeding, dyspnea, and bradycardia. Dyspnea occurs in approximately 14% of patients taking ticagrelor, compared with 8% of patients taking clopidogrel. Although adenosine effects have been implicated as a cause, there remains debate on this issue. Recent analysis of the tolerability of this ticagrelor found that nearly one-third patients receiving 90 mg twice daily discontinued use of the medication, with approximately one-fifth citing adverse events, including bleeding (mostly nonmajor) and dyspnea [236].

            Ischemic Stroke

            In the initial PLATO trial, patients with prior stroke/TIA had higher rates of MI, death, recurrent stroke, and intracranial bleeding than patients without prior stroke/TIA, although efficacy and bleeding results of ticagrelor in these high-risk patients were consistent with the overall trial population [237]. The Acute Stroke or Transient Ischaemic Attack Treated with Aspirin or Ticagrelor and Patient Outcomes (SOCRATES) trial, looking at 13,199 patients with stroke or TIA, found that ticagrelor was not superior to aspirin in reducing the rate of stroke, MI, or death at 90 days [238]. However, a subgroup analysis revealed superiority of ticagrelor in patients with ipsilateral atherosclerotic stenosis, suggesting that the cause of stroke matters when one is choosing a drug for secondary prevention [239]. In this setting, the role of ticagrelor in stroke prevention remains uncertain.

            Peripheral Artery Disease

            Building on the potential role for clopidogrel in PAD, the Examining Use of Ticagrelor in PAD (EUCLID) trial studied 13,885 patients with symptomatic PAD randomized to receive monotherapy with ticagrelor (90 mg twice daily) or clopidogrel (75 mg once daily). The primary outcome (incidence of cardiovascular death, MI, or ischemic stroke) was no different in the two groups [240], with additional lack of efficacy in the cohort with a history of lower extremity revascularization [241]. Thus there is currently no clear role for ticagrelor in PAD.

            With potential pleiotropic effects apart from platelet inhibition, ticagrelor confers a benefit over clopidogrel in terms of cardiovascular events and mortality in patients with ACS, regardless of invasive versus conservative management. On the basis of these findings, the current ACC/AHA guidelines recommend ticagrelor in patients with NSTE-ACS, independent of the management strategy, and in STEMI patients who have received a stent (class I), adding a class IIb recommendation to choose ticagrelor over clopidogrel for maintenance P2Y12 inhibitor therapy [103]. On the basis of the PEGASUS-TIMI 54 trial, the guidelines also support continuation of P2Y12 therapy beyond 12 months in prior MI patients who are not at high bleeding risk (class IIb). The role of ticagrelor in stroke, PAD, and patients with atrial fibrillation undergoing PCI remains unclear.

            Cangrelor
            Pharmacology

            Although not an oral agent, the intravenous, reversible, rapid-acting P2Y12 inhibitor cangrelor is an important component of modern antiplatelet therapy. Administered as an active compound that does not require activation, this cyclopentyltriazolopyrimidine medication exhibits a linear dose-dependent pharmacokinetic profile, resulting in platelet inhibition exceeding 90% [242]. Platelet inhibition typically begins within 2 min after administration of a bolus dose, with maximum inhibition achieved within 30 min [243]. With a half-life of 3–6 min, the drug undergoes deactivation to an inactive metabolite via dephosphorylation by a vascular endothelial endonucleotidase; this allows near complete platelet recovery within 60 min after discontinuation of the infusion [244].

            Clinical Outcomes

            Intended for use during PCI, cangrelor was investigated in a series of three related trials. The first of these, Cangrelor versus Standard Therapy to Achieve Optimal Management of Platelet Inhibition (CHAMPION) PLATFORM trial was a randomized, double-blind, placebo controlled trial that evaluated 5301 patients with unstable angina or NSTEMI undergoing PCI with or without stent implantation. Patients were randomized to receive either cangrelor (30 μg/kg bolus and 4 μg/kg per minute infusion) or placebo, followed by clopidogrel at a dose of 600 mg at the end of the infusion or procedure. From examination of the composite end point of death, MI, or ischemia-driven revascularization 48 h after PCI, cangrelor was not superior to placebo. However, the prespecified secondary end points of stent thrombosis and death were lower in the cangrelor group, with a signal for more major bleeding [245]. At the same time, the CHAMPION PCI trial compared cangrelor with 600 mg of orally administered clopidogrel given before PCI in 8722 patients with stable angina, unstable angina, NSTEMI, or STEMI. Again, cangrelor was not superior to 600 mg of clopidogrel when the composite primary end point of death from any cause, MI, or ischemia-driven revascularization at 48 h was examined [246].

            Subsequently, the 2013 CHAMPION PHOENIX trial randomized 11,145 patients with stable angina, unstable angina, NSTEMI, or STEMI undergoing urgent or elective PCI to receive cangrelor or clopidogrel. In this trial, the primary outcome was a composite of all-cause death, MI, ischemia-driven revascularization, or, unlike the prior trials, stent thrombosis. In this setting, cangrelor significantly reduced the risk of the primary outcome without increasing in the risk of severe bleeding [247]. A prespecified pooled analysis of these three CHAMPION studies suggested that cangrelor was better than the control clopidogrel in decreasing post-PCI thrombotic complications [248].

            On the basis of the findings of the CHAMPION PHOENIX trial, cangrelor gained Food and Drug Admiration approval for use during PCI. Notably, cangrelor treatment was associated with a higher risk of dyspnea, as with ticagrelor. At this point it remains unclear how cangrelor compares with ticagrelor and prasugrel.

            New and Upcoming Data

            The recent single-center observational CHANGE-DAPT study (clopidogrel or ticagrelor in ACS patients treated with newer-generation drug-eluting stents) noted a higher rate of major bleeding in ticagrelor-treated ACS patients compared with clopidogrel-treated ACS patients (2.7 vs. 1.2%; adjusted hazard ratio 2.75; 95% confidence interval 1.34–5.61), without significant ischemic benefit for the newer-generation drug [249]. This furthers the findings of the TOPIC randomized study (benefit of switching DAPT after acute coronary syndrome), which found that a strategy of switching from prasugrel or ticagrelor to clopidogrel 1 month after PCI for ACS was associated with fewer bleeding events, without a significant change in ischemic outcomes [250]. Furthermore, the optimal duration of DAPT is undergoing more nuanced exploration. In ACS, the 6 versus 12 Months of Dual Antiplatelet Therapy after Drug-Eluting Stent Implantation in ST-Elevation Myocardial Infarction (DAPT STEMI) trial showed that among patients who remain event-free at 6 months following PCI with a zotarolimus-eluting stent for STEMI, cessation of DAPT at 6 months was noninferior to continuation to 12 months [251]. Many of these new data are focused on the role of newer-generation drug-eluting stents, with smaller stent struts and a lower risk of stent thrombosis. In non-ACS PCI, a series of trials are evaluating 1–3 months of therapy with a P2Y12 inhibitor with newer-generation stents [252, 253]. The findings of these studies may have significant impact on the use of DAPT in patients who do not present with ACS. Although not practice-changing, these studies suggest that the role of these newer antiplatelet agents in ACS will need to be further explored in randomized controlled trials, particularly with the advent of new stent technology.

            Conclusion

            Of the P2Y12 inhibitors, clopidogrel is commonly prescribed in the United States, with an established track record of safety and efficacy in various conditions, including stable CAD, ACS, stroke, PAD, and atrial fibrillation as combined therapy. As a generic medication, its relative affordability makes it an appealing choice for many patients and providers. However, it is limited by common polymorphisms that promote high on-treatment platelet reactivity and thus a higher risk of recurrent ischemic events. Both ticagrelor and prasugrel are superior to clopidogrel in terms of ischemic outcomes in patients with ACS who undergo PCI. Preliminary evidence from a head-to-head comparison of these newer agents suggests that one is neither more effective nor safer than the other in this population [254, 255]. Ticagrelor, however, is limited by cost and adverse effects, such as dyspnea, leading to medication use discontinuation. Prasugrel has a limited scope of indications and several contraindications to its use due to a higher risk of bleeding. In the United States, economic analysis of patients from the PLATO trial suggests that for PLATO-eligible ACS patients, an aspirin plus ticagrelor regimen increases life expectancy at an incremental cost well within accepted benchmarks of good value for money [256], although further analysis is still needed [257, 258]. Although ticagrelor and prasugrel are the preferred options in patients with ACS undergoing PCI, clopidogrel remains a cost-effective antiplatelet agent with the widest range of potential applications, suggesting that for many of our patients it remains an excellent first choice.

            References

            1. Antithrombotic Trialists’Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. Br Med J 2002;324:71–86.

            2. ChildersCP, Maggard-GibbonsM, ShekellePG. Antiplatelet therapy in patients with coronary stents undergoing elective noncardiac surgery. J Am Med Assoc 2017;318:120–1.

            3. NguyenTA, DiodatiJG, PharandC. Resistance to clopidogrel: a review of the evidence. J Am Coll Cardiol 2005;45:1157–64.

            4. FerriN, CorsiniA, BellostaS. Pharmacology of the new P2Y12 receptor inhibitors: insights on pharmacokinetic and pharmacodynamic properties. Drugs 2013;73:1681–709.

            5. DorsamRT, KunapuliSP. Central role of the P2Y12 receptor in platelet activation. J Clin Invest 2004;113:340–5.

            6. FaridNA, KuriharaA, WrightonSA. Metabolism and disposition of the thienopyridine antiplatelet drugs ticlopidine, clopidogrel, and prasugrel in humans. J Clin Pharmacol 2010;50:126–42.

            7. XiaoFY, LuoJQ, LiuM, ChenBL, CaoS, LiuZQ, et al. Effect of carboxylesterase 1 S75N on clopidogrel therapy among acute coronary syndrome patients. Sci Rep 2017;7:7244.

            8. JiangX-L, SamantS, LeskoLJ, SchmidtS. Clinical pharmacokinetics and pharmacodynamics of clopidogrel. Clin Pharmacokinet 2015;54(2):147–66.

            9. SchrörK. Clinical pharmacology of the adenosine diphosphate (ADP) receptor antagonist, clopidogrel. Vasc Med 1998;3:247–51.

            10. VorchheimerDA, BeckerR. Platelets in atherothrombosis. Mayo Clin Proc 2006;81:59–68.

            11. ZhangH, LauWC, HollenbergPF. Formation of the thiol conjugates and active metabolite of clopidogrel by human liver microsomes. Mol Pharmacol 2012;82:302–9.

            12. MillsDC, PuriR, HuCJ, MinnitiC, GranaG, FreedmanMD, et al. Clopidogrel inhibits the binding of ADP analogues to the receptor mediating inhibition of platelet adenylate cyclase. Arterioscler Thromb 1992;12:430–6.

            13. QuinnMJ, BhattDL, ZidarF, VivekananthanD, ChewDP, EllisSG, et al. Effect of clopidogrel pretreatment on inflammatory marker expression in patients undergoing percutaneous coronary intervention. Am J Cardiol 2004;93:679–84.

            14. von BeckerathN, TaubertD, Pogatsa-MurrayG, SchömigE, KastratiA, SchömigA. Absorption, metabolization, and antiplatelet effects of 300-, 600-, and 900-mg loading doses of clopidogrel. Circulation 2005;112:2946–50.

            15. NeumannFJ, KastratiA, Pogatsa-MurrayG, MehilliJ, BollweinH, BestehornHP, et al. Evaluation of prolonged antithrombotic pretreatment (“cooling-off” strategy) before intervention in patients with unstable coronary syndromes. J Am Med Assoc 2003;290:189–98.

            16. MontalescotG, SiderisG, MeulemanC, Bal-dit-SollierC, LelloucheN, StegPG, et al. A randomized comparison of high clopidogrel loading doses in patients with non-ST-segment elevation acute coronary syndromes. J Am Coll Cardiol 2006;48:931–8.

            17. CuissetT, FrereC, QuiliciJ, MorangePE, Nait-SaidiL, CarvajalJ, et al. Benefit of a 600-mg loading dose of clopidogrel on platelet reactivity and clinical outcomes in patients with non-ST-segment elevation acute coronary syndrome undergoing coronary stenting. J Am Coll Cardiol 2006;48:1339–45.

            18. GurbelPA, BlidenKP, ZamanKA, YohoJA, HayesKM, TantryUS. Clopidogrel loading with eptifibatide to arrest the reactivity of platelets. Circulation 2005;111:1153–9.

            19. JungJ-H, MinP-K, LeeS-H, SungCW, ChoiS, ChoJR, et al. Clopidogrel pretreatment before primary percutaneous coronary stenting in patients with acute ST-segment elevation myocardial infarction: comparison of high loading dose (600 mg) versus low loading dose (300 mg). Coron Artery Dis 2009;20:150–4.

            20. SaltzmanAJ, StoneGW, ClaessenBE, NarulaA, Leon-ReyesS, WeiszG, et al. Long-term impact of chronic kidney disease in patients with ST-segment elevation myocardial infarction treated with primary percutaneous coronary intervention. JACC Cardiovasc Interv 2011;4:1011–9.

            21. PattiG, BárcziG, OrlicD, MangiacapraF, ColonnaG, PasceriV, et al. Outcome comparison of 600- and 300-mg loading doses of clopidogrel in patients undergoing primary percutaneous coronary intervention for ST-segment elevation myocardial infarction. J Am Coll Cardiol 2011;58:1592–9.

            22. CURRENT-OASIS 7Investigators. Dose comparisons of clopidogrel and aspirin in acute coronary syndromes. N Engl J Med 2010;363:930–42.

            23. MehtaSR, TanguayJF, EikelboomJW, JollySS, JoynerCD, GrangerCB, et al. Double-dose versus standard-dose clopidogrel and high-dose versus low-dose aspirin in individuals undergoing percutaneous coronary intervention for acute coronary syndromes (CURRENT-OASIS 7): a randomised factorial trial. Lancet 2010;376:1233–43.

            24. KimH, LeeHK, HanK, JeonH-K. Prevalence and risk factors for aspirin and clopidogrel resistance in patients with coronary artery disease or ischemic cerebrovascular disease. Ann Clin Lab Sci 2009;39:289–94.

            25. AngiolilloDJ, SuryadevaraS. Aspirin and clopidogrel: efficacy and resistance in diabetes mellitus. Best Pract Res Clin Endocrinol Metab 2009;23:375–88.

            26. PanY, ChenW, XuY, YiX, HanY, YangQ, et al. Genetic polymorphisms and clopidogrel efficacy for acute ischemic stroke or transient ischemic attack. A systematic review and meta-analysis. Circulation 2016;135:21–35.

            27. LingLQ, LiaoJ, NiuQ, WangX, JiaJ, ZuoCH, et al. Evaluation of an automated light transmission aggregometry. Platelets 2017;28:712–9.

            28. GremmelT, KoppensteinerR, PanzerS. Comparison of aggregometry with flow cytometry for the assessment of agonists-induced platelet reactivity in patients on dual antiplatelet therapy. PLoS ONE 2015;10(6):e0129666.

            29. TantryUS, BlidenKP, GurbelPA. Resistance to antiplatelet drugs: current status and future research. Expert Opin Pharmacother 2005;6:2027–45.

            30. WangTH, BhattDL, TopolEJ. Aspirin and clopidogrel resistance: an emerging clinical entity. Eur Heart J 2006;27:647–54.

            31. CuissetT, CaylaG, SilvainJ. Clopidogrel resistance: what’s new? Arch Cardiovasc Dis 2010;103:349–53.

            32. SmockKJ, SaundersPJ, RodgersGM, JohariV. Laboratory evaluation of clopidogrel responsiveness by platelet function and genetic methods. Am J Hematol 2011;86:1032–4.

            33. AlexopoulosD. Prasugrel resistance: fact or fiction. Platelets 2012;23:83–90.

            34. WiviottSD, AntmanEM. Clopidogrel resistance. Circulation 2004;109:3064–7.

            35. HodgesLM, MarkovaSM, ChinnLW, GowJM, KroetzDL, KleinTE, et al. Very important pharmacogene summary: ABCB1 (MDR1, P-glycoprotein). Pharmacogenet Genomics 2011;21:152–61.

            36. KubicaA, KozinskiM, GrzeskG, FabiszakT, NavareseEP, GochA. Genetic determinants of platelet response to clopidogrel. J Thromb Thrombolysis 2011;32:459–66.

            37. TaubertD, von BeckerathN, GrimbergG, LazarA, JungN, GoeserT, et al. Impact of P-glycoprotein on clopidogrel absorption. Clin Pharmacol Ther 2006;80:486–501.

            38. MegaJL, CloseSL, WiviottSD, ShenL, WalkerJR, SimonT, et al. Genetic variants in ABCB1 and CYP2C19 and cardiovascular outcomes after treatment with clopidogrel and prasugrel in the TRITON-TIMI 38 trial: a pharmacogenetic analysis. Lancet 2010;376:1312–9.

            39. WallentinL, JamesS, StoreyRF, ArmstrongM, BarrattBJ, HorrowJ, et al. 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. Lancet 2010;376:1320–8.

            40. JaitnerJ, MorathT, ByrneRA, BraunS, GebhardD, BernlochnerI, et al. No association of ABCB1 C3435T genotype with clopidogrel response or risk of stent thrombosis in patients undergoing coronary stenting. Circ Cardiovasc Interv 2012;5:82–8.

            41. Fricke-GalindoI, Céspedes-GarroC, Rodrigues-SoaresF, NaranjoME, DelgadoÁ, de AndrésF, et al. Interethnic variation of CYP2C19 alleles, “predicted” phenotypes and “measured” metabolic phenotypes across world populations. Pharmacogenomics J 2016;16:113–23.

            42. ShuldinerAR, O’ConnellJR, BlidenKP, GandhiA, RyanK, HorensteinRB, et al. Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. J Am Med Assoc 2009;302:849–57.

            43. MegaJL, CloseSL, WiviottSD, ShenL, HockettRD, BrandtJT, et al. Cytochrome P-450 polymorphisms and response to clopidogrel. N Engl J Med 2009;360:354–62.

            44. HulotJS, BuraA, VillardE, AziziM, RemonesV, GoyenvalleC, et al. Cytochrome P450 2C19 loss-of-function polymorphism is a major determinant of clopidogrel responsiveness in healthy subjects. Blood 2006;108:2244–7.

            45. WangX, LaiY, LuoY, ZhangX, ZhouH, YeZ, et al. Relationship between clopidogrel-related polymorphisms and variable platelet reactivity at 1 year: a cohort study from Han Chinese. J Res Med Sci 2016;21:111.

            46. ScottSA, SangkuhlK, SteinCM, HulotJS, MegaJL, RodenDM, et al. Clinical Pharmacogenetics Implementation Consortium guidelines for CYP2C19 genotype and clopidogrel therapy: 2013 update. Clin Pharmacol Ther 2013;94:317–23.

            47. MaoL, JianC, ChangzhiL, DanH, SuihuaH, WenyiT, et al. Cytochrome CYP2C19 polymorphism and risk of adverse clinical events in clopidogrel-treated patients: a meta-analysis based on 23,035 subjects. Arch Cardiovasc Dis 2013;106:517–7.

            48. OgboguPU, BochnerBS, ButterfieldJH, GleichGJ, Huss-MarpJ, KahnJE, et al. Hypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapy. J Allergy Clin Immunol 2009;124:1319–25.e3.

            49. HulotJS, ColletJP, SilvainJ, PenaA, Bellemain-AppaixA, BarthélémyO, et al. Cardiovascular risk in clopidogrel-treated patients according to cytochrome P450 2C19*2 loss-of-function allele or proton pump inhibitor coadministration. J Am Coll Cardiol 2010;56:134–43.

            50. GiustiB, GoriAM, MarcucciR, SaraciniC, SestiniI, PanicciaR, et al. Relation of cytochrome P450 2C19 loss-of-function polymorphism to occurrence of drug-eluting coronary stent thrombosis. Am J Cardiol 2009;103:806–11.

            51. SibbingD, StegherrJ, LatzW, KochW, MehilliJ, DörrlerK, et al. Cytochrome P450 2C19 loss-of-function polymorphism and stent thrombosis following percutaneous coronary intervention. Eur Heart J 2008;30:916–22.

            52. Li-Wan-PoA, GirardT, FarndonP, CooleyC, LithgowJ. Pharmacogenetics of CYP2C19: functional and clinical implications of a new variant CYP2C19*17. Br J Clin Pharmacol 2010;69:222–30.

            53. DeanL. Prasugrel therapy and CYP genotype. Bethesda: National Center for Biotechnology Information; 2012.

            54. ZabalzaM, SubiranaI, SalaJ, Lluis-GanellaC, LucasG, TomásM, et al. Meta-analyses of the association between cytochrome CYP2C19 loss- and gain-of-function polymorphisms and cardiovascular outcomes in patients with coronary artery disease treated with clopidogrel. Heart 2012;98:100–8.

            55. LauWC, GurbelPA, WatkinsPB, NeerCJ, HoppAS, CarvilleDG, et al. Contribution of hepatic cytochrome P450 3A4 metabolic activity to the phenomenon of clopidogrel resistance. Circulation 2004;109:166–71.

            56. SmithSM, JudgeHM, PetersG, ArmstrongM, FontanaP, GaussemP, et al. Common sequence variations in the P2Y 12 and CYP3A5 genes do not explain the variability in the inhibitory effects of clopidogrel therapy. Platelets 2006;17:250–8.

            57. LevineGN, BatesER, BlankenshipJC, BaileySR, BittlJA, CercekB, et al. 2011 ACCF/AHA/SCAI guideline for percutaneous coronary intervention. J Am Coll Cardiol 2011;58;2550–83.

            58. CapodannoD, FerreiroJL, AngiolilloDJ. Antiplatelet therapy: new pharmacological agents and changing paradigms. J Thromb Haemost 2013;11:316–29.

            59. TantryUS, BonelloL, AradiD, PriceMJ, JeongYH, AngiolilloDJ, et al. Consensus and update on the definition of on-treatment platelet reactivity to adenosine diphosphate associated with ischemia and bleeding. J Am Coll Cardiol 2013;62:2261–73.

            60. FDA, CDER. Highlights of prescribing information in proceedings of prescription drug labeling: challenges and issues. Venue: Pooks Hill, MD, USA, November 3–4, 2015. Washington, DC, USA: FDA.

            61. Antiplatelet drug compari-son chart. http://www.vhpharmsci.com/vhformulary/Tools/ANTIPLATELETDRUGCOMPARISONCHART.pdf. Accessed August 22, 2017.

            62. ShinJM, SachsG. Pharmacology of proton pump inhibitors. Curr Gastroenterol Rep 2008;10:528–34.

            63. BatesER, LauWC, AngiolilloDJ. Clopidogrel-drug interactions. J Am Coll Cardiol 2011;57:1251–63.

            64. DouglasIJ, EvansSJW, HingoraniAD, GrossoAM, TimmisA, HemingwayH, et al. Clopidogrel and interaction with proton pump inhibitors: comparison between cohort and within person study designs. Br Med J 2012;345:e4388.

            65. BundhunPK, TeeluckAR, BhurtuA, HuangW-Q. Is the concomitant use of clopidogrel and Proton Pump Inhibitors still associated with increased adverse cardiovascular outcomes following coronary angioplasty?: a systematic review and meta-analysis of recently published studies (2012–2016). BMC Cardiovasc Disord 2017;17:3.

            66. BhattDL, CryerBL, ContantCF, CohenM, LanasA, SchnitzerTJ, et al. Clopidogrel with or without omeprazole in coronary artery disease. N Engl J Med 2010;363:1909–17.

            67. SouthworthMR, TempleR. Interaction of clopidogrel and omeprazole. N Engl J Med 2010;363:1977.

            68. EtemadL. Statins and potentially interacting medications: a managed care perspective. Prev Medicine Managed Care 2004;4:S27–9.

            69. LauWC, WaskellLA, WatkinsPB, NeerCJ, HorowitzK, HoppAS, et al. Atorvastatin reduces the ability of clopidogrel to inhibit platelet aggregation: a new drug-drug interaction. Circulation 2003;107:32–7.

            70. MitsiosJV, PapathanasiouAI, RodisFI, ElisafM, GoudevenosJA, TselepisAD. Atorvastatin does not affect the antiplatelet potency of clopidogrel when it is administered concomitantly for 5 weeks in patients with acute coronary syndromes. Circulation 2004;109:1335–8.

            71. WenaweserP, WindeckerS, BillingerM, CookS, TogniM, MeierB, et al. Effect of atorvastatin and pravastatin on platelet inhibition by aspirin and clopidogrel treatment in patients with coronary stent thrombosis. Am J Cardiol 2007;99:353–6.

            72. BhindiR, OrmerodO, NewtonJ, BanningAP, TestaL. Interaction between statins and clopidogrel: is there anything clinically relevant? Quart J Med 2008;101:915–25.

            73. MalmströmRE, ÖstergrenJ, JørgensenL, HjemdahlP. Influence of statin treatment on platelet inhibition by clopidogrel – a randomized comparison of rosuvastatin, atorvastatin and simvastatin co-treatment. J Intern Med 2009;266:457–66.

            74. ZhangJR, WangDQ, DuJ, QuGS, DuJL, DengSB, et al. Efficacy of clopidogrel and clinical outcome when clopidogrel is coadministered with atorvastatin and lansoprazole: a prospective, randomized, controlled trial. Medicine (Baltimore) 2015;94:e2262.

            75. KreutzRP, BreallJA, SinhaA, von der LoheE, KovacsRJ, FlockhartDA. Simultaneous administration of high-dose atorvastatin and clopidogrel does not interfere with platelet inhibition during percutaneous coronary intervention. Clin Pharmacol 2016;8:45–50.

            76. GodinoC, PavonAG, MangieriA, SalernoA, CeraM, MonelloA, et al. Platelet reactivity in response to loading dose of atorvastatin or rosuvastatin in patients with stable coronary disease before percutaneous coronary intervention: the STATIPLAT randomized study. Clin Cardiol 2017;40:605–11.

            77. NorgardNB, DiNicolantonioJJ. Clopidogrel, prasugrel, or ticagrelor? A practical guide to use of antiplatelet agents in patients with acute coronary syndromes. Postgrad Med 2013;125(4):91–102.

            78. ChapmanN, SchachterM. Clopidogrel and calcium-channel blockers: a clinically important interaction? Heart 2010;96:179–80.

            79. HarmszeAM, RobijnsK, van WerkumJW, BreetNJ, HackengCM, Ten BergJM, et al. The use of amlodipine, but not of P-glycoprotein inhibiting calcium channel blockers is associated with clopidogrel poor-response. Thromb Haemost 2010;103:920–5.

            80. Siller-MatulaJM, LangI, ChristG, JilmaB. Calcium-channel blockers reduce the antiplatelet effect of clopidogrel. J Am Coll Cardiol 2008;52:1557–63.

            81. SeoK-D, KimYD, YoonY-W, KimJ-Y, LeeK-Y. Antiplatelet Effect of clopidogrel can be reduced by calcium-channel blockers. Yonsei Med J 2014;55:683.

            82. GremmelT, DurstbergerM, EichelbergerB, KoppensteinerR, PanzerS. Calcium-channel blockers attenuate the antiplatelet effect of clopidogrel. Cardiovasc Ther 2015;33:264–9.

            83. OlesenJB, GislasonGH, CharlotMG, FosbølEL, AnderssonC, WeekeP, et al. Calcium-channel blockers do not alter the clinical efficacy of clopidogrel after myocardial infarction. J Am Coll Cardiol 2011;57:409–17.

            84. GoodCW, SteinhublSR, BrennanDM, LincoffAM, TopolEJ, BergerPB. Is there a clinically significant interaction between calcium channel antagonists and clopidogrel? Circ Cardiovasc Interv 2012;5:77–81.

            85. LiAYW, NgF-H, ChanFKL, TunggalP, ChanK, LauY-K. Effect of amlodipine on platelet inhibition by clopidogrel in patients with ischaemic heart disease: a randomised, controlled trial. Heart 2013;99:468–73.

            86. DvorakZ. Drug-drug interactions by azole antifungals: beyond a dogma of CYP3A4 enzyme activity inhibition. Toxicol Lett 2011;202:129–32.

            87. FaridNA, PayneCD, SmallDS, WintersKJ, Ernest CS2nd, BrandtJT, et al. Cytochrome P450 3A Inhibition by ketoconazole affects prasugrel and clopidogrel pharmacokinetics and pharmacodynamics differently. Clin Pharmacol Ther 2007;81:735–41.

            88. CAPRIE SteeringCommittee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet 1996;348:1329–39.

            89. BertrandME, RupprechtHJ, UrbanP, GershlickAH. Double-blind study of the safety of clopidogrel with and without a loading dose in combination with aspirin compared with ticlopidine in combination with aspirin after coronary stenting?: the clopidogrel aspirin stent international cooperative study (CLASSICS). Circulation 2000;102:624–9.

            90. BergerPB, BellMR, HasdaiD, GrillDE, MelbyS, HolmesDR, et al. Safety and efficacy of ticlopidine for only 2 weeks after successful intracoronary stent placement. Circulation 1999;99:248–53.

            91. TaniuchiM, KurzH, SmithS. Ticlopidine or Plavix post-stent (TOPPS): randomization to 2-week treatment. Am J Cardiol 1999;84(6A):68P–69P.

            92. CURE StudyInvestigators. The Clopidogrel in Unstable angina to prevent Recurrent Events (CURE) trial programme. Rationale, design and baseline characteristics including a meta-analysis of the effects of thienopyridines in vascular disease. Eur Heart J 2000;21:2033–41.

            93. MehtaSR, YusufS, PetersRJ, BertrandME, LewisBS, NatarajanMK, 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;358:527–33.

            94. SteinhublSR, BergerPB, Mann JT3rd, FryET, DeLagoA, WilmerC, et al. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention: a randomized controlled trial. J Am Med Assoc 2002;288:2411–20.

            95. ChenZM, PanHC, ChenYP, PetoR, CollinsR, JiangLX, et al. Early intravenous then oral metoprolol in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005;366:1622–32.

            96. SabatineMS, CannonCP, GibsonCM, López-SendónJL, MontalescotG, TherouxP, et al. Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with ST-segment elevation. N Engl J Med 2005;352:1179–89.

            97. BhattDL, FoxKA, HackeW, BergerPB, BlackHR, BodenWE, et al. Clopidogrel and aspirin versus aspirin alone for the prevention of atherothrombotic events. N Engl J Med 2006;354:1706–17.

            98. PriceMJ, BergerPB, TeirsteinPS, TanguayJF, AngiolilloDJ, SpriggsD, et al. Standard- vs high-dose clopidogrel based on platelet function testing after percutaneous coronary intervention. J Am Med Assoc 2011;305:1097.

            99. Di SciascioG, PattiG, PasceriV, ColonnaG, MangiacapraF, MontinaroA, et al. Clopidogrel reloading in patients undergoing percutaneous coronary intervention on chronic clopidogrel therapy: results of the ARMYDA-4 RELOAD (Antiplatelet therapy for Reduction of MYocardial Damage during Angioplasty) randomized trial. Eur Heart J 2010;31:1337–43.

            100. Di SciascioG, PattiG, PasceriV, GattoL, ColonnaG, MontinaroA, et al. Effectiveness of in-laboratory high-dose clopidogrel loading versus routine pre-load in patients undergoing percutaneous coronary intervention. J Am Coll Cardiol 2010;56:550–7.

            101. PattiG, PasceriV, MangiacapraF, ColonnaG, VizziV, RicottiniE, et al. Efficacy of clopidogrel reloading in patients with acute coronary syndrome undergoing percutaneous coronary intervention during chronic clopidogrel therapy (from the Antiplatelet therapy for Reduction of MYocardial Damage during Angioplasty [ARMYDA-8 RELOAD-ACS] trial). Am J Cardiol 2013;112:162–8.

            102. PattiG, GriecoD, DicuonzoG, PasceriV, NuscaA, Di SciascioG. High versus standard clopidogrel maintenance dose after percutaneous coronary intervention and effects on platelet inhibition, endothelial function, and inflammation. J Am Coll Cardiol 2011;57:771–8.

            103. LevineGN, BatesER, BittlJA, BrindisRG, FihnSD, FleisherLA, et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease. J Am Coll Cardiol 2016;68:1082–115.

            104. WangY, WangY, ZhaoX, LiuL, WangD, WangC, et al. Clopidogrel with aspirin in acute minor stroke or transient ischemic attack. N Engl J Med 2013;369:11–9.

            105. MarkusHS, DrosteDW, KapsM, LarrueV, LeesKR, SieblerM, et al. Dual antiplatelet therapy with clopidogrel and aspirin in symptomatic carotid stenosis evaluated using Doppler embolic signal detection: the Clopidogrel and Aspirin for Reduction of Emboli in Symptomatic Carotid Stenosis (CARESS) trial. Circulation 2005;111:2233–40.

            106. WongKS, ChenC, FuJ, ChangHM, SuwanwelaNC, HuangYN, et al. Clopidogrel plus aspirin versus aspirin alone for reducing embolisation in patients with acute symptomatic cerebral or carotid artery stenosis (CLAIR study): a randomised, open-label, blinded-endpoint trial. Lancet Neurol 2010;9:489–97.

            107. ChimowitzMI, LynnMJ, DerdeynCP, TuranTN, FiorellaD, LaneBF, et al. Stenting versus aggressive medical therapy for intracranial arterial stenosis. N Engl J Med 2011;365:993–1003.

            108. DienerHC, BogousslavskyJ, BrassLM, CimminielloC, CsibaL, KasteM, et al. Aspirin and clopidogrel compared with clopidogrel alone after recent ischaemic stroke or transient ischaemic attack in high-risk patients (MATCH): randomised, double-blind, placebo-controlled trial. Lancet 2004;364:331–7.

            109. SPS3Investigators. Effects of clopidogrel added to aspirin in patients with recent lacunar stroke. N Engl J Med 2012;367:817–25.

            110. JohnstonSC, EastonJD, FarrantM, BarsanW, BattenhouseH, ConwitR, et al. Platelet-oriented inhibition in new TIA and minor ischemic stroke (POINT) trial: rationale and design. Int J Stroke 2013;8:479–83.

            111. KernanWN, OvbiageleB, BlackHR, BravataDM, ChimowitzMI, EzekowitzMD, et al. Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack. Stroke 2014;45;2160–236.

            112. ParikhSV, SayaS, DivanjiP, BanerjeeS, SelzerF, AbbottJD, et al. Risk of death and myocardial infarction in patients with peripheral arterial disease undergoing percutaneous coronary intervention (from the National Heart, Lung and Blood Institute Dynamic Registry). Am J Cardiol 2011;107:959–64.

            113. CacoubPP, BhattDL, StegPG, TopolEJ, CreagerMA. Patients with peripheral arterial disease in the CHARISMA trial. Eur Heart J 2008;30:192–201.

            114. RookeTW, HirschAT, MisraS, SidawyAN, BeckmanJA, FindeissL, et al. Management of patients with peripheral artery disease (compilation of 2005 and 2011 ACCF/AHA guideline recommendations). J Am Coll Cardiol 2013;61:1555–70.

            115. MoscucciM, FoxK, CannonC, KleinW. Predictors of major bleeding in acute coronary syndromes: the Global Registry of Acute Coronary Events (GRACE). Eur Heart J 2003;24(20):1815–23.

            116. DoyleBJ, RihalCS, GastineauDA, HolmesDR. Bleeding, blood transfusion, and increased mortality after percutaneous coronary intervention. J Am Coll Cardiol 2009;53:2019–27.

            117. DewildeWJM, JanssenPWA, VerheugtFWA, StoreyRF, AdriaenssensT, HansenML, et al. Triple therapy for atrial fibrillation and percutaneous coronary intervention. J Am Coll Cardiol 2014;64:1270–80.

            118. DewildeWJ, OirbansT, VerheugtFW, KelderJC, De SmetBJ, HerrmanJP, et al. Use of clopidogrel with or without aspirin in patients taking oral anticoagulant therapy and undergoing percutaneous coronary intervention: an open-label, randomised, controlled trial. Lancet 2013;381:1107–15.

            119. SeivaniY, Abdel-WahabM, GeistV, RichardtG, SulimovDS, El-MawardyM, et al. Long-term safety and efficacy of dual therapy with oral anticoagulation and clopidogrel in patients with atrial fibrillation treated with drug-eluting stents. Clin Res Cardiol 2013;102:799–806.

            120. LambertsM, GislasonGH, OlesenJB, KristensenSL, Schjerning OlsenAM, MikkelsenA, et al. Oral anticoagulation and antiplatelets in atrial fibrillation patients after myocardial infarction and coronary intervention. J Am Coll Cardiol 2013;62:981–9.

            121. AmsterdamEA, WengerNK, BrindisRG, CaseyDE, GaniatsTG, HolmesDR, et al. 2014 AHA/ACC guideline for the management of patients with non-ST-elevation acute coronary syndromes. J Am Coll Cardiol 2014;64:e139–e228.

            122. JanuaryCT, WannLS, AlpertJS, CalkinsH, CigarroaJE, Cleveland JCJr, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: executive summary. J Am Coll Cardiol 2014;139(23):2071–104.

            123. GibsonCM, MehranR, BodeC, HalperinJ, VerheugtFW, WildgooseP, et al. Prevention of bleeding in patients with atrial fibrillation undergoing PCI. N Engl J Med 2016;375:2423–34.

            124. CannonCP, BhattDL, OldgrenJ, LipGYH, EllisSG, KimuraT, et al. Dual antithrombotic therapy with dabigatran after pci in atrial fibrillation. N Engl J Med 2017;377:1513–24.

            125. ValgimigliM, BuenoH, ByrneRA, ColletJP, CostaF, JeppssonA, et al. 2017 ESC focused update on dual antiplatelet therapy in coronary artery disease developed in collaboration with EACTS. Eur Heart J 2018;39:213–60.

            126. OttoCM, KumbhaniDJ, AlexanderKP, CalhoonJH, DesaiMY, KaulS, et al. 2017 ACC expert consensus decision pathway for transcatheter aortic valve replacement in the management of adults with aortic stenosis. J Am Coll Cardiol 2017;69:1313–46.

            127. ReardonMJ, KleimanNS, AdamsDH, YakubovSJ, CoselliJS, DeebGM, et al. Outcomes in the randomized CoreValve US pivotal high risk trial in patients with a society of thoracic surgeons risk score of 7% or less. JAMA Cardiol 2016;1:945.

            128. DeebGM, ReardonMJ, ChetcutiS, PatelHJ, GrossmanPM, YakubovSJ, et al. 3-Year outcomes in high-risk patients who underwent surgical or transcatheter aortic valve replacement. J Am Coll Cardiol 2016;67:2565–74.

            129. AdamsDH, PopmaJJ, ReardonMJ, YakubovSJ, CoselliJS, DeebGM, et al. Transcatheter aortic-valve replacement with a self-expanding prosthesis. N Engl J Med 2014;370:1790–8.

            130. LeonMB, SmithCR, MackMJ, MakkarRR, SvenssonLG, KodaliSK, et al. Transcatheter or surgical aortic-valve replacement in intermediate-risk patients. N Engl J Med 2016;374:1609–20.

            131. LeonMB, SmithCR, MackM, MillerDC, MosesJW, SvenssonLG, et al. Transcatheter aortic-valve implantation for aortic stenosis in patients who cannot undergo surgery. N Engl J Med 2010;363:1597–607.

            132. SmithCR, LeonMB, MackMJ, MillerDC, MosesJW, SvenssonLG, et al. Transcatheter versus surgical aortic-valve replacement in high-risk patients. N Engl J Med 2011;364:2187–98.

            133. Rodés-CabauJ, MassonJ-B, WelshRC, Garcia Del BlancoB, PelletierM, WebbJG, et al. Aspirin versus aspirin plus clopidogrel as antithrombotic treatment following transcatheter aortic valve replacement with a balloon-expandable valve. JACC Cardiovasc Interv 2017;10:1357–65.

            134. NakataniS. Subclinical leaflet thrombosis after transcatheter aortic valve implantation. Heart 2017;103:1942–6.

            135. ChakravartyT, SøndergaardL, FriedmanJ, De BackerO, BermanD, KofoedKF, et al. Subclinical leaflet thrombosis in surgical and transcatheter bioprosthetic aortic valves: an observational study. Lancet 2017;389:2383–92.

            136. HafizAM, KalraA, RamadanR, PoulinMF, AndalibA, PhillipsCT, et al. Clinical or symptomatic leaflet thrombosis following transcatheter aortic valve replacement: insights from the U.S. FDA MAUDE database. Struct Heart 2017;1:256–64.

            137. NijenhuisVJ, BennaghmouchN, HassellM, Baan JJr, van KuijkJP, AgostoniP, et al. Rationale and design of POPular-TAVI: antiPlatelet therapy fOr Patients undergoing Transcatheter Aortic Valve Implantation. Am Heart J 2016;173:77–85.

            138. SinghP, HarperY, OliphantCS, MorsyM, SkeltonM, AskariR, et al. Peripheral interventions and antiplatelet therapy: role in current practice. World J Cardiol 2017;9:583–93.

            139. JeongY-H, HwangJ-Y, KimI-S, ParkY, HwangSJ, LeeSW, 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 2010;3:17–26.

            140. SuhJ-W, LeeS-P, ParkK-W, LeeHY, KangHJ, KooBK, et al. Multicenter randomized trial evaluating the efficacy of cilostazol on ischemic vascular complications after drug-eluting stent implantation for coronary heart disease. J Am Coll Cardiol 2011;57:280–9.

            141. PanchalHB, ShahT, PatelP, AlbalbissiK, MolnarJ, CoffeyB, et al. Comparison of on-treatment platelet reactivity between triple antiplatelet therapy with cilostazol and standard dual antiplatelet therapy in patients undergoing coronary interventions. J Cardiovasc Pharmacol Ther 2013;18:533–43.

            142. LeeS-W, ParkS-W, YunS-C, KimYH, ParkDW, KimWJ, et al. Triple antiplatelet therapy reduces ischemic events after drug-eluting stent implantation: drug-Eluting stenting followed by Cilostazol treatment REduces Adverse Serious cardiac Events (DECREASE registry). Am Heart J 2010;159:284–91.e1.

            143. IidaO, YokoiH, SogaY, InoueN, SuzukiK, YokoiY, et al. Cilostazol reduces angiographic restenosis after endovascular therapy for femoropopliteal lesions in the sufficient treatment of peripheral intervention by cilostazol study. Circulation 2013;127:2307–15.

            144. IidaO, NantoS, UematsuM, MorozumiT, KitakazeM, NagataS. Cilostazol reduces restenosis after endovascular therapy in patients with femoropopliteal lesions. J Vasc Surg 2008;48:144–9.

            145. NantoK, IidaO, TakaharaM, SogaY, SuzukiK, HiranoK, et al. Effect of cilostazol following endovascular intervention for peripheral artery disease. Angiology 2015;66:774–8.

            146. ParkKW, KangS-H, ParkJJ, YangHM, KangHJ, KooBK, et al. Adjunctive cilostazol versus double-dose clopidogrel after drug-eluting stent implantation. JACC Cardiovasc Interv 2013;6:932–42.

            147. SongPS, SongYB, YangJH, HahnJY, ChoiSH, ChoiJH, et al. Triple versus dual antiplatelet therapy after percutaneous coronary intervention for coronary bifurcation lesions: results from the COBIS (COronary BIfurcation Stent) II registry. Heart Vessels 2015;30:458–68.

            148. RileyAB, TafreshiMJ, HaberSL. Prasugrel: a novel antiplatelet agent. Am J Health Syst Pharm 2008;65:1019–28.

            149. JakubowskiJA, PayneCD, BrandtJT, WeerakkodyGJ, FaridNA, SmallDS, et al. The platelet inhibitory effects and pharmacokinetics of prasugrel after administration of loading and maintenance doses in healthy subjects. J Cardiovasc Pharmacol 2006;47:377–84.

            150. DansettePM, RosiJ, DebernardiJ, BerthoG, MansuyD. Metabolic activation of prasugrel: nature of the two competitive pathways resulting in the opening of its thiophene ring. Chem Res Toxicol 2012;25:1058–65.

            151. RehmelJLF, EcksteinJA, FaridNA, HeimJB, KasperSC, KuriharaA, et al. Interactions of two major metabolites of prasugrel, a thienopyridine antiplatelet agent, with the cytochromes P450. Drug Metab Dispos 2006;34:600–7.

            152. BeckerRC, GurbelPA. Platelet P2Y12 receptor antagonist pharmacokinetics and pharmacodynamics: a foundation for distinguishing mechanisms of bleeding and anticipated risk for platelet-directed therapies. Thromb Haemost 2010;103:535–44.

            153. AngiolilloDJ, CapranzanoP. Pharmacology of emerging novel platelet inhibitors. Am Heart J 2008;156:S10–5.

            154. AncrenazV, DéglonJ, SamerC, StaubC, DayerP, DaaliY, et al. Pharmacokinetic interaction between prasugrel and ritonavir in healthy volunteers. Basic Clin Pharmacol Toxicol 2013;112:132–7.

            155. DobeshPP. Pharmacokinetics and Pharmacodynamics of prasugrel, a thienopyridine P2Y12 inhibitor. Pharmacotherapy 2009;29:1089–102.

            156. NorgardNB, Abu-FadelM. Comparison of prasugrel and clopidogrel in patients with acute coronary syndrome undergoing percutaneous coronary intervention. Vasc Health Risk Manag 2009;5:873–82.

            157. YuK-S, ParkKW, KellyRP, GuN, PayneC, SmallDS, et al. Pharmacokinetic and pharmacodynamic effects of prasugrel in healthy Korean males. J Cardiovasc Pharmacol 2013;62:72–7.

            158. NiitsuY, JakubowskiJA, SugidachiA, AsaiF. Pharmacology of CS-747 (prasugrel, LY640315), a novel, potent antiplatelet agent with in vivo P2Y 12 receptor antagonist activity. Semin Thromb Hemost 2005;31:184–94.

            159. WiviottSD, TrenkD, FrelingerAL, O’DonoghueM, NeumannFJ, MichelsonAD, et al. Prasugrel compared with high loading- and maintenance-dose clopidogrel in patients with planned percutaneous coronary intervention. Circulation 2007;116:2923–32.

            160. AlexopoulosD, DimitropoulosG, DavlourosP, XanthopoulouI, KassimisG, StavrouEF, et al. Prasugrel Overcomes high on-clopidogrel platelet reactivity post-stenting more effectively than high-dose (150-mg) clopidogrel. JACC Cardiovasc Interv 2011;4:403–10.

            161. BrandtJT, CloseSL, IturriaSJ, PayneCD, FaridNA, Ernest CS2nd, et al. Common polymorphisms of CYP2C19 and CYP2C9 affect the pharmacokinetic and pharmacodynamic response to clopidogrel but not prasugrel. J Thromb Haemost 2007;5:2429–36.

            162. HolmbergMT, TornioA, HyvärinenH, NeuvonenM, NeuvonenPJ, BackmanJT, et al. Effect of grapefruit juice on the bioactivation of prasugrel. Br J Clin Pharmacol 2015;80:139–45.

            163. MichelsonAD, FrelingerAL, BraunwaldE, DowneyWE, AngiolilloDJ, XenopoulosNP, et al. Pharmacodynamic assessment of platelet inhibition by prasugrel vs. clopidogrel in the TRITON-TIMI 38 trial. Eur Heart J 2009;30:1753–63.

            164. VarenhorstC, JamesS, ErlingeD, BrandtJT, BraunOO, ManM, et al. Genetic variation of CYP2C19 affects both pharmacokinetic and pharmacodynamic responses to clopidogrel but not prasugrel in aspirin-treated patients with coronary artery disease. Eur Heart J 2009;30:1744–52.

            165. MegaJL, CloseSL, WiviottSD, ShenL, HockettRD, BrandtJT, et al. Cytochrome P450 genetic polymorphisms and the response to prasugrel relationship to pharmacokinetic, pharmacodynamic, and clinical outcomes. Circulation 2009;119(19):2553–60.

            166. JaureguiME, UmejeiOU, CruzMP. Prasugrel (Effient), an adenosine diphosphate receptor antagonist for the treatment of acute coronary syndrome. Pharm Ther 2009;34:417.

            167. AncrenazV, DaaliY, FontanaP, BessonM, SamerC, DayerP, et al. Impact of genetic polymorphisms and drug-drug interactions on clopidogrel and prasugrel response variability. Curr Drug Metab 2010;11:667–77.

            168. BadriPS, KingJR, PolepallyAR, McGovernBH, DuttaS, MenonRM. dosing recommendations for concomitant medications during 3D anti-HCV therapy. Clin Pharmacokinet 2016;55:275–95.

            169. DaaliY, AncrenazV, BosilkovskaM, DayerP, DesmeulesJ. Ritonavir inhibits the two main prasugrel bioactivation pathways in vitro: a potential drug-drug interaction in HIV patients. Metabolism 2011;60:1584–9.

            170. WiviottSD, AntmanEM, WintersKJ, WeerakkodyG, MurphySA, BehounekBD, et al. Randomized comparison of prasugrel (CS-747, LY640315), a novel thienopyridine P2Y12 antagonist, with clopidogrel in percutaneous coronary intervention: results of the Joint Utilization of Medications to Block Platelets Optimally (JUMBO)-TIMI 26 trial. Circulation 2005;111:3366–73.

            171. De ServiS, GoedickeJ, SchirmerA, WidimskyP. Clinical outcomes for prasugrel versus clopidogrel in patients with unstable angina or non-ST-elevation myocardial infarction: an analysis from the TRITON-TIMI 38 trial. Eur Heart J Acute Cardiovasc Care 2014;3:363–72.

            172. MontalescotG, SiderisG, CohenR, MeulemanC, Bal dit SollierC, BarthélémyO, et al. Prasugrel compared with high-dose clopidogrel in acute coronary syndrome. Thromb Haemost 2009;103:213–23.

            173. UdellJA, BraunwaldE, AntmanEM, MurphySA, MontalescotG, WiviottSD. Prasugrel versus clopidogrel in patients with ST-segment elevation myocardial infarction according to timing of percutaneous coronary intervention. JACC Cardiovasc Interv 2014;7:604–12.

            174. AntmanEM, WiviottSD, MurphySA, VoitkJ, HasinY, WidimskyP, et al. Early and late benefits of prasugrel in patients with acute coronary syndromes undergoing percutaneous coronary intervention. J Am Coll Cardiol 2008;51:2028–33.

            175. WiviottSD, BraunwaldE, McCabeCH, HorvathI, KeltaiM, HerrmanJP, et al. Intensive oral antiplatelet therapy for reduction of ischaemic 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 randomised trial. Lancet 2008;371:1353–63.

            176. O’DonoghueM, AntmanEM, BraunwaldE, MurphySA, StegPG, FinkelsteinA, et al. The efficacy and safety of prasugrel with and without a glycoprotein IIb/IIIa inhibitor in patients with acute coronary syndromes undergoing percutaneous intervention. J Am Coll Cardiol 2009;54:678–85.

            177. WiviottSD, BraunwaldE, AngiolilloDJ, MeiselS, DalbyAJ, VerheugtFW, et al. Greater clinical benefit of more intensive oral antiplatelet therapy with prasugrel in patients with diabetes mellitus in the Trial to Assess Improvement in Therapeutic Outcomes by Optimizing Platelet Inhibition with Prasugrel-Thrombolysis in Myocardial Infarction 38. Circulation 2008;118:1626–36.

            178. WiviottSD, BraunwaldE, McCabeCH, MontalescotG, RuzylloW, GottliebS, et al. Prasugrel versus Clopidogrel in patients with acute coronary syndromes. N Engl J Med 2007;357:2001–15.

            179. MarchiniJ, MorrowD, ResnicF, ManicaA, KirshenbaumJ, CannonC, et al. An algorithm for use of prasugrel (Effient) in patients undergoing cardiac catheterization and percutaneous coronary intervention. Crit Pathw Cardiol 2010;9:192–8.

            180. RiesmeyerJS, SalazarDE, WeerakkodyGJ, NiL, WrishkoRE, Ernest CS2nd, et al. Relationship between exposure to prasugrel active metabolite and clinical outcomes in the TRITON-TIMI 38 substudy. J Clin Pharmacol 2012;52:789–97.

            181. RoeMT, ArmstrongPW, FoxKAA, WhiteHD, PrabhakaranD, GoodmanSG, et al. Prasugrel versus clopidogrel for acute coronary syndromes without revascularization. N Engl J Med 2012;367:1297–309.

            182. GurbelPA, ErlingeD, OhmanEM, NeelyB, NeelyM, GoodmanSG, et al. Platelet Function during extended prasugrel and clopidogrel therapy for patients with ACS treated without revascularization. J Am Med Assoc 2012;308:1785.

            183. TrenkD, StoneGW, GawazM, KastratiA, AngiolilloDJ, MüllerU, et al. A randomized trial of prasugrel versus clopidogrel in patients with high platelet reactivity on clopidogrel after elective percutaneous coronary intervention with implantation of drug-eluting stents. J Am Coll Cardiol 2012;59:2159–64.

            184. MontalescotG, BologneseL, DudekD, GoldsteinP, HammC, TanguayJF, et al. Pretreatment with prasugrel in non-ST-segment elevation acute coronary syndromes. N Engl J Med 2013;369:999–1010.

            185. AngiolilloDJ, FranchiF. Defining a role for prasugrel in patients with stable coronary artery disease undergoing ad hoc percutaneous coronary intervention. JACC Cardiovasc Interv 2016;9:228–30.

            186. HochholzerW, AmannM, TitovA, YounasI, LöffelhardtN, RiedeF, et al. Randomized comparison of different thienopyridine loading strategies in patients undergoing elective coronary intervention. JACC Cardiovasc Interv 2016;9:219–27.

            187. SpringthorpeB, BaileyA, BartonP, BirkinshawTN, BonnertRV, BrownRC, et al. From ATP to AZD6140: the discovery of an orally active reversible P2Y12 receptor antagonist for the prevention of thrombosis. Bioorg Med Chem Lett 2007;17:6013–8.

            188. TengR, ButlerK. AZD6140, the first reversible oral platelet P2Y12 receptor antagonist, has linear pharmacokinetics and provides near complete inhibition of platelet. Can J Clin Pharmacol 2008;15:e426.

            189. TengR, ButlerK. Pharmacokinetics, pharmacodynamics, tolerability and safety of single ascending doses of ticagrelor, a reversibly binding oral P2Y12 receptor antagonist, in healthy subjects. Eur J Clin Pharmacol 2010;66:487–96.

            190. LiH, ButlerK, YangL, YangZ, TengR. Pharmacokinetics and tolerability of single and multiple doses of ticagrelor in healthy Chinese subjects. Clin Drug Investig 2012;32:87–97.

            191. TengR. Ticagrelor: pharmacokinetic, pharmacodynamic and pharmacogenetic profile: an update. Clin Pharmacokinet 2015;54:1125–38.

            192. TengR, OliverS, HayesMA, ButlerK. Absorption, distribution, metabolism, and excretion of ticagrelor in healthy subjects. Drug Metab Dispos 2010;38:1514–21.

            193. GurbelPA, BlidenKP, ButlerK, TantryUS, GesheffT, WeiC, et al. Randomized Double-blind assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coronary artery disease: the ONSET/OFFSET study. Circulation 2009;120:2577–85.

            194. TengR, MitchellPD, ButlerKA. Pharmacokinetic interaction studies of co-administration of ticagrelor and atorvastatin or simvastatin in healthy volunteers. Eur J Clin Pharmacol 2013;69:477–87.

            195. HustedS, EmanuelssonH, HeptinstallS, SandsetPM, WickensM, PetersG. Pharmacodynamics, pharmacokinetics, and safety of the oral reversible P2Y12 antagonist AZD6140 with aspirin in patients with atherosclerosis: a double-blind comparison to clopidogrel with aspirin. Eur Heart J 2005;27:1038–47.

            196. DobeshPP, OestreichJH. Ticagrelor: pharmacokinetics, pharmacodynamics, clinical efficacy, and safety. Pharmacotherapy 2014;34:1077–90.

            197. StoreyRF, HustedS, HarringtonRA, HeptinstallS, WilcoxRG, PetersG, et al. Inhibition of platelet aggregation by AZD6140, a reversible oral P2Y12 receptor antagonist, compared with clopidogrel in patients with acute coronary syndromes. J Am Coll Cardiol 2007;50:1852–6.

            198. van GiezenJJJ, NillsonL, BerntssonP, WissingBM, GiordanettoF, TomlinsonW, et al. Ticagrelor binds to human P2Y12 independently from ADP but antagonizes ADP-induced receptor signaling and platelet aggregation. J Thromb Haemost 2009;7:1556–65.

            199. TengR, MitchellP, ButlerK. Effect of age and gender on pharmacokinetics and pharmacodynamics of a single ticagrelor dose in healthy individuals. Eur J Clin Pharmacol 2012;68:1175–82.

            200. ButlerK, TengR. Pharmacokinetics, pharmacodynamics, and safety of ticagrelor in volunteers with severe renal impairment. J Clin Pharmacol 2012;52:1388–98.

            201. HustedS, van GiezenJJJ. Ticagrelor: the first reversibly binding oral P2Y 12 receptor antagonist. Cardiovasc Ther 2009;27:259–74.

            202. ChengJWM. Ticagrelor: oral reversible P2Y12 receptor antagonist for the management of acute coronary syndromes. Clin Ther 2012;34:1209–20.

            203. ArmstrongD, SummersC, EwartL, NylanderS, SidawayJE, van GiezenJJJ. Characterization of the adenosine pharmacology of ticagrelor reveals therapeutically relevant inhibition of equilibrative nucleoside transporter 1. J Cardiovasc Pharmacol Ther 2014;19:209–19.

            204. WittfeldtA, EmanuelssonH, Brandrup-WognsenG, van GiezenJJ, JonassonJ, NylanderS, et al. Ticagrelor enhances adenosine-induced coronary vasodilatory responses in humans. J Am Coll Cardiol 2013;61:723–7.

            205. van GiezenJJJ, SidawayJ, GlavesP, KirkI, BjörkmanJ-A. Ticagrelor inhibits adenosine uptake in vitro and enhances adenosine-mediated hyperemia responses in a canine model. J Cardiovasc Pharmacol Ther 2012;17:164–72.

            206. NanhwanMK, LingS, KodakandlaM, NylanderS, YeY, BirnbaumY. Chronic treatment with ticagrelor limits myocardial infarct size: an adenosine and cyclooxygenase-2-dependent effect. Arterioscler Thromb Vasc Biol 2014;34:2078–85.

            207. LiX, WangQ, XueY, ChenJ, LvQ. Ticagrelor compared with clopidogrel increased adenosine and cyclic adenosine monophosphate plasma concentration in acute coronary syndrome patients. Basic Clin Pharmacol Toxicol 2017;120(6):610–4.

            208. CattaneoM, SchulzR, NylanderS. Adenosine-mediated effects of ticagrelor. J Am Coll Cardiol 2014;63:2503–9.

            209. HustedSE, StoreyRF, BlidenK, TantryUS, HøimarkL, ButlerK, et al. Pharmacokinetics and pharmacodynamics of ticagrelor in patients with stable coronary artery disease. Clin Pharmacokinet 2012;51:397–409.

            210. TantryUS, BlidenKP, WeiC, StoreyRF, ArmstrongM, ButlerK, et al. First analysis of the relation between CYP2C19 genotype and pharmacodynamics in patients treated with ticagrelor versus clopidogrel: the ONSET/OFFSET and RESPOND genotype studies. Circ Cardiovasc Genet 2010;3:556–66.

            211. ÅkerblomA, ErikssonN, WallentinL, SiegbahnA, BarrattBJ, BeckerRC, et al. Polymorphism of the cystatin C gene in patients with acute coronary syndromes: results from the PLATelet inhibition and patient Outcomes study. Am Heart J 2014;168:96–102.e2.

            212. VarenhorstC, ErikssonN, JohanssonÅ, BarrattB, ÅkerblomA, HagströmE, et al. Ticagrelor plasma levels but not clinical outcomes are associated with transporter and metabolism enzyme genetic polymorphisms. J Am Coll Cardiol 2014;63:A25.

            213. ZhouD, AnderssonTB, GrimmSW. In vitro evaluation of potential drug-drug interactions with ticagrelor: cytochrome P450 reaction phenotyping, inhibition, induction, and differential kinetics. Drug Metab Dispos 2011;39:703–10.

            214. WigginsBS, SaseenJJ, PageRL, ReedBN, SneedK, KostisJB, et al. Recommendations for management of clinically significant drug-drug interactions with statins and select agents used in patients with cardiovascular disease: a scientific statement from the American Heart Association. Circulation 2016;134:e468–95.

            215. NavareseEP, BuffonA, KozinskiM, ObonskaK, RychterM, KunadianV, et al. A critical overview on ticagrelor in acute coronary syndromes. QJM 2013;106:105–15.

            216. TengR, ButlerK. Effect of the CYP3A inhibitors, diltiazem and ketoconazole, on ticagrelor pharmacokinetics in healthy volunteers. J Drug Assess 2013;2:30–9.

            217. CannonCP, HustedS, HarringtonRA, SciricaBM, EmanuelssonH, PetersG, et al. Safety, tolerability, and initial efficacy of AZD6140, the first reversible oral adenosine diphosphate receptor antagonist, compared with clopidogrel, in patients with non-ST-segment elevation acute coronary syndrome. J Am Coll Cardiol 2007;50:1844–51.

            218. ChoJR, RolliniF, FranchiF, DeGroatC, BhattiM, DunnEC, et al. Pharmacodynamic effects of ticagrelor dosing regimens in patients on maintenance ticagrelor therapy. JACC Cardiovasc Interv 2015;8:1075–83.

            219. WallentinL, BeckerRC, BudajA, CannonCP, EmanuelssonH, HeldC, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2009;361:1045–57.

            220. StegPG, JamesS, HarringtonRA, ArdissinoD, BeckerRC, CannonCP, et al. Ticagrelor versus clopidogrel in patients with ST-elevation acute coronary syndromes intended for reperfusion with primary percutaneous coronary intervention: a Platelet Inhibition and Patient Outcomes (PLATO) trial subgroup analysis. Circulation 2010;122:2131–41.

            221. LindholmD, VarenhorstC, CannonCP, HarringtonRA, HimmelmannA, MayaJ, et al. Ticagrelor vs. clopidogrel in patients with non-ST-elevation acute coronary syndrome with or without revascularization: results from the PLATO trial. Eur Heart J 2014;35:2083–93.

            222. CannonCP, HarringtonRA, JamesS, ArdissinoD, BeckerRC, EmanuelssonH, et al. Comparison of ticagrelor with clopidogrel in patients with a planned invasive strategy for acute coronary syndromes (PLATO): a randomised double-blind study. Lancet 2010;375:283–93.

            223. JamesSK, RoeMT, CannonCP, CornelJH, HorrowJ, HustedS, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes intended for non-invasive management: substudy from prospective randomised PLATelet inhibition and patient Outcomes (PLATO) trial. Br Med J 2011:d3257. doi: 10.1136/bmj.d3527.

            224. HeldC, ÅsenbladN, BassandJP, BeckerRC, CannonCP, ClaeysMJ, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes undergoing coronary artery bypass surgery. J Am Coll Cardiol 2011;57:672–84.

            225. JamesS, BudajA, AylwardP, BuckKK, CannonCP, CornelJH, et al. Ticagrelor versus clopidogrel in acute coronary syndromes in relation to renal function. Circulation 2010;122:1056–67.

            226. JamesS, AngiolilloDJ, CornelJH, ErlingeD, HustedS, KontnyF, et al. Ticagrelor vs. clopidogrel in patients with acute coronary syndromes and diabetes: a substudy from the PLATelet inhibition and patient Outcomes (PLATO) trial. Eur Heart J 2010;31:3006–16.

            227. CornelJH, BeckerRC, GoodmanSG, HustedS, KatusH, SantosoA, et al. Prior smoking status, clinical outcomes, and the comparison of ticagrelor with clopidogrel in acute coronary syndromes-insights from the PLATelet inhibition and patient Outcomes (PLATO) trial. Am Heart J 2012;164:334–42.e1.

            228. HustedS, JamesS, BeckerRC, HorrowJ, KatusH, StoreyRF, et al. Ticagrelor versus clopidogrel in elderly patients with acute coronary syndromes. Circ Cardiovasc Qual Outcomes 2012;5:680–8.

            229. KunadianV, JamesSK, WojdylaDM, ZorkunC, WuJ, StoreyRF, et al. Angiographic outcomes in the PLATO trial (Platelet Inhibition and Patient Outcomes). JACC Cardiovasc Interv 2013;6:671–83.

            230. KohliP, WallentinL, ReyesE, HorrowJ, HustedS, AngiolilloDJ, et al. Reduction in first and recurrent cardiovascular events with ticagrelor compared with clopidogrel in the PLATO study. Circulation 2013;127:673–80.

            231. MahaffeyKW, WojdylaDM, CarrollK, BeckerRC, StoreyRF, AngiolilloDJ, et al. Ticagrelor compared with clopidogrel by geographic region in the Platelet Inhibition and Patient Outcomes (PLATO) trial. Circulation 2011;124:544–54.

            232. GurbelPA, BlidenKP, ButlerK, AntoninoMJ, WeiC, TengR, et al. Response to ticagrelor in clopidogrel nonresponders and responders and effect of switching therapies: the RESPOND study. Circulation 2010;121:1188–99.

            233. BonacaMP, BhattDL, CohenM, StegPG, StoreyRF, JensenEC, et al. Long-Term use of ticagrelor in patients with prior myocardial infarction. N Engl J Med 2015;372:1791–800.

            234. MontalescotG, van’t HofAW, LapostolleF, SilvainJ, LassenJF, BologneseL, et al. Prehospital ticagrelor in st-segment elevation myocardial infarction. N Engl J Med 2014;371:1016–27.

            235. MontalescotG, van’t HofAW, BologneseL, CantorWJ, CequierA, ChettibiM, et al. Effect of pre-hospital ticagrelor during the first 24 h after primary percutaneous coronary intervention in patients with ST-segment elevation myocardial infarction. JACC Cardiovasc Interv 2016;9:646–56.

            236. BonacaMP, BhattDL, Oude OphuisT, StegPG, StoreyR, CohenM, et al. Long-term tolerability of ticagrelor for the secondary prevention of major adverse cardiovascular events. JAMA Cardiol 2016;1:425.

            237. DiNicolantonioJJ, SerebruanyVL. Comparing ticagrelor versus clopidogrel in patients with a history of cerebrovascular disease. Stroke 2012;43:3409–10.

            238. JohnstonSC, AmarencoP, AlbersGW, DenisonH, EastonJD, EvansSR, et al. Ticagrelor versus aspirin in acute stroke or transient ischemic attack. N Engl J Med 2016;375:35–43.

            239. AmarencoP, AlbersGW, DenisonH, EastonJD, EvansSR, HeldP, et al. Efficacy and safety of ticagrelor versus aspirin in acute stroke or transient ischaemic attack of atherosclerotic origin: a subgroup analysis of SOCRATES, a randomised, double-blind, controlled trial. Lancet Neurol 2017;16:301–10.

            240. HiattWR, FowkesFGR, HeizerG, BergerJS, BaumgartnerI, HeldP, et al. Ticagrelor versus clopidogrel in symptomatic peripheral artery disease. N Engl J Med 2017;376:32–40.

            241. JonesWS, BaumgartnerI, HiattWR, HeizerG, ConteMS, WhiteCJ, et al. Ticagrelor compared with clopidogrel in patients with prior lower extremity revascularization for peripheral artery disease. Circulation 2017;135:241–50.

            242. GreenbaumAB, GrinesCL, BittlJA, BeckerRC, KereiakesDJ, GilchristIC, et al. Initial experience with an intravenous P2Y12 platelet receptor antagonist in patients undergoing percutaneous coronary intervention: results from a 2-part, phase II, multicenter, randomized, placebo- and active-controlled trial. Am Heart J 2006;151:689.e1–10.

            243. LeonardiS, MahaffeyKW, WhiteHD, GibsonCM, StoneGW, StegGW, et al. Rationale and design of the Cangrelor versus standard therapy to acHieve optimal Management of Platelet InhibitiON PHOENIX trial. Am Heart J 2012;163:768–76.e2.

            244. AkersWS, OhJJ, OestreichJH, FerrarisS, WethingtonM, SteinhublSR. Pharmacokinetics and pharmacodynamics of a bolus and infusion of cangrelor: a direct, parenteral P2Y12 receptor antagonist. J Clin Pharmacol 2010;50:27–35.

            245. BhattDL, LincoffAM, GibsonCM, StoneGW, McNultyS, MontalescotG, et al. Intravenous Platelet blockade with cangrelor during PCI. N Engl J Med 2009;361:2330–41.

            246. HarringtonRA, StoneGW, McNultyS, WhiteHD, LincoffAM, GibsonCM, et al. Platelet Inhibition with cangrelor in patients undergoing PCI. N Engl J Med 2009;361:2318–29.

            247. BhattDL, StoneGW, MahaffeyKW, GibsonCM, StegPG, HammCW, et al. Effect of platelet inhibition with cangrelor during PCI on ischemic events. N Engl J Med 2013;368:1303–13.

            248. BakerDE, IngramKT. Cangrelor. Hosp Pharm 2015;50:922–9.

            249. ZoccaP, van der HeijdenL, KokM, LöwikMM, HartmannM, StoelMG, et al. Clopidogrel or ticagrelor in acute coronary syndrome patients treated with newer-generation drug-eluting stents: CHANGE DAPT. EuroIntervention 2017;13:1168–76.

            250. CuissetT, DeharoP, QuiliciJ, JohnsonTW, DeffargesS, BassezC, et al. Benefit of switching dual antiplatelet therapy after acute coronary syndrome: the TOPIC (timing of platelet inhibition after acute coronary syndrome) randomized study. Eur Heart J 2017;38:3070–8.

            251. KedhiE, FabrisE, van der EntM, KennedyMW, BuszmanP, von BirgelenC, et al. A prospective, randomized, open-label trial of 6-month versus 12-month dual antiplatelet therapy after drug-eluting stent implantation in ST-elevation myocardial infarction: rationale and design of the “DAPT-STEMI trial.” Am Heart J 2017;188:11–7.

            252. EVOLVE Short DAPT Study. Available at: https://clinicaltrials.gov/ct2/show/NCT02605447 Accessed December 22, 2017.

            253. A Randomized Controlled Trial with Resolute Onyx in One Month Dual Antiplatelet Therapy (DAPT) for high-bleeding risk patients (Onyx ONE). Available at: https://clinicaltrials.gov/ct2/show/NCT03344653 Accessed December 22, 2017.

            254. RolliniF, FranchiF, ChoJR, DeGroatC, BhattiM, Muniz-LozanoA, et al. A head-to-head pharmacodynamic comparison of prasugrel vs. ticagrelor after switching from clopidogrel in patients with coronary artery disease: results of a prospective randomized study. Eur Heart J 2016;37:2722–30.

            255. MotovskaZ, HlinomazO, MiklikR, HromadkaM, VarvarovskyI, DusekJ, et al. Prasugrel versus ticagrelor in patients with acute myocardial infarction treated with primary percutaneous coronary intervention: multicenter randomized PRAGUE-18 study. Circulation 2016;134:1603–12.

            256. CowperPA, PanW, AnstromKJ, KaulP, WallentinL, Davidson-RayL, et al. Economic analysis of ticagrelor therapy from a U.S. perspective. J Am Coll Cardiol 2015;65:465–76.

            257. KaziDS, HlatkyMA. A delicate balance the cost effectiveness of new antiplatelet agents. J. Am Coll Cardiol 2015;65:477–9.

            258. PaikinJS, EikelboomJW, CairnsJA, HirshJ. New antithrombotic agents –insights from clinical trials. Nat Rev Cardiol 2010;7:498–509.

            Author and article information

            Journal
            CVIA
            Cardiovascular Innovations and Applications
            CVIA
            Compuscript (Ireland )
            2009-8782
            2009-8618
            July 2018
            August 2018
            : 3
            : 2
            : 175-202
            Affiliations
            [1] 1The University of California San Francisco and The San Francisco Veteran Affairs Hospital, 505 Parnassus Ave, M1182, San Francisco, CA 94143-0124, USA
            Author notes
            Correspondence: Punag Divanji, MD, Division of Cardiology, The University of California San Francisco and The San Francisco Veteran Affairs Hospital, 505 Parnassus Ave, M1182 SF, San Francisco, CA 94143-0124, USA, Tel.: +1-832-8666794, Fax: +1-660-2480160, E-mail: punag.divanji@ 123456ucsf.edu
            Article
            cvia20170049
            10.15212/CVIA.2017.0049
            2d59a831-0ff8-4215-baf5-abe70353b75c
            Copyright © 2018 Cardiovascular Innovations and Applications

            This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 Unported License (CC BY-NC 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. See https://creativecommons.org/licenses/by-nc/4.0/.

            History
            : 2 January 2018
            : 2 March 2018
            : 6 March 2018
            Categories
            Reviews

            General medicine,Medicine,Geriatric medicine,Transplantation,Cardiovascular Medicine,Anesthesiology & Pain management
            P2Y12 inhibitor,acute coronary syndrome,thienopyridine,stable coronary artery disease,antiplatelet therapy

            Comments

            Comment on this article