153
Views
15
CrossRef citations to date
0
Altmetric
Review

Newer agents in antiplatelet therapy: a review

&
Pages 33-42 | Published online: 25 Jun 2012

Abstract

Antiplatelet therapy remains the mainstay in preventing aberrant platelet activation in pathophysiological conditions such as myocardial infarction, ischemia, and stroke. Although there has been significant advancement in antiplatelet therapeutic approaches, aspirin still remains the gold standard treatment in the clinical setting. Limitations in safety, efficacy, and tolerability have precluded many of the antiplatelet inhibitors from use in patients. Unforeseen incidences of increased bleeding risk and recurrent arterial thrombosis observed in patients have hampered the development of superior next generation antiplatelet therapies. The pharmacokinetic and pharmacodynamic profiles have also limited the effectiveness of a number of antiplatelet inhibitors currently in use due to variability in metabolism, time to onset, and reversibility. A focused effort in the development of newer antiplatelet therapies to address some of these shortcomings has resulted in a significant number of potential antiplatelet drugs which target enzymes (phosphodiesterase, cyclooxygenase), receptors (purinergic, prostaglandins, protease-activated receptors, thromboxane), and glycoproteins (αIIbβ3, GPVI, vWF, GPIb) in the platelet. The validation and search for newer antiplatelet therapeutic approaches proven to be superior to aspirin is still ongoing and should yield a better pharmacodynamic profile with fewer untoward side-effects to what is currently in use today.

View correction statement:
Erratum

Introduction

Antiplatelet drugs are the cornerstone in treatment of cardiovascular diseases. Despite the significant decrease in morbidity and mortality due to the currently approved anti-platelet drugs, recurrent ischemia, myocardial infarction (MI), and unwanted bleeding still occur. The majority of drugs in development have focused on targeting either surface receptors or enzymes in the platelet in order to protect against unwanted clot formation following initial platelet activation. The first target for antiplatelet therapy was cyclooxygenase-1 by aspirin. While newer approaches for containing platelet activity have been developed, the pharmacodynamics and pharmacoeconomics suggest that aspirin will continue to be a mainstay for platelet therapy in the years to come. Currently, a combination regimen of aspirin and clopidogrel is the standard of care for prevention of platelet activation, thrombosis, and stroke. Unfortunately many of the current antiplatelet drugs face limitations in their utility due to genetic differences in the ability to metabolize pro-drugs, such as is the case with clopidogrel, acquired allergic responses such as is seen with heparin and aspirin, and resistance as has been reported with aspirin (see ). Additional limitations observed in the application of currently approved antiplatelet drugs include a narrow therapeutic window and limited efficacy. An overview of the current Food and Drug Administration (FDA)-approved antiplatelet therapies as well as those in development will be discussed in this review.

Table 1 Approved antiplatelet drugs

P2Y receptor antagonists

The P2Y receptors are G-protein-coupled (GPCR) purinergic receptors belonging to the P2 family. Two receptors, P2Y1 and P2Y12, are present in the platelet. P2Y1 is a Gq coupled GPCR, while P2Y12 is coupled to Gαi2. Activation of P2Y1 signals phospholipase β, leading to DAG formation, calcium mobilization, and eventually PKC and CalDAG-GEF activation. Citation1 In contrast, P2Y12 activation inhibits adenylyl cyclase, activates phosphoinositide 3-kinase,Citation2 the small GTPase Rap1,Citation3 and activation of αIIbβ3.Citation4

Ticlopidine (Ticlid®; Roche, Basel, Switzerland) is a first generation thienopyridine that requires cytochrome P450 (CYP) 1 A metabolism prior to exerting its irreversible antagonistic effects on platelet reactivity via the P2Y12 receptor.Citation5 Early experimental observations showed agonist-induced platelet aggregation was intermittently inhibited by ticlopidine. Citation6,Citation7 Studies with ticlopidine, however, exhibit off-target effects mediated by the inhibition of intracellular calcium mobilization.Citation8 Maximal inhibition of platelet aggregation is observed 3–5 days post administration of ticlopidine.Citation9 The delayed onset of antiplatelet effects is a consequence of metabolism of the pro-drug.Citation6 Clinical trials (CATS and TASS studies) have shown ticlopidine to be more effective than aspirin alone,Citation10,Citation11 but exhibiting significant off-target effects including minor bleeding with hemorrhagic events observed in less than 1% of subjects studied. Additionally, ticlopidine-treated patients typically discontinue treatment due to a variety of secondary adverse events including diarrhea, skin rash, and neutropenia.Citation10,Citation12

Clopidogrel (Plavix®, Bristol-Myers Squibb, New York City, NY), a second generation oral thienopyridine, also requires metabolism of a pro-drug by the CYP2C19. The active metabolite, which is a highly labile compound, irreversibly binds to and inhibits the P2Y12 receptor through a disulfide bridge. The CURE trial has shown the clinical benefit of the dual clopidogrel-aspirin therapy compared with aspirin alone by significantly reducing mortality and nonfatal MI or stroke in patients with unstable angina; however, the dual regimen was associated with an increase in bleeding compared with placebo.Citation13,Citation14 The CAPRIE trial, which evaluated the efficacy of clopidogrel monotherapy compared with dual therapy of clopidogrel plus aspirin, showed clopidogrel treatment results in a reduction of primary endpoints.Citation15 Evidence of poor metabolizers for clopidogrel has helped to explain the reduced function in patients with an altered CYP2C19 allele.Citation16 Poor metabolizers of clopidogrel have diminished platelet inhibition resulting in a higher rate of adverse cardiovascular events than noncarriers.Citation17

Prasugrel (Effient®, Eli Lilly and Company, Indianapolis, IN) is a third-generation thienopyridine, chemically distinct from clopidogrel. In-vivo and in-vitro pharmacological studies have demonstrated that this adenosine triphosphate (ATP) analog selectively and irreversibly inhibits adenosine diphosphate (ADP)-induced aggregation to a greater degree than clopidogrel.Citation18 The irreversible binding is thought to be due to the disulfide binding between the reactive thiol group of the active metabolite and the cysteine residue of the P2Y12 receptor.Citation19Citation21 Prasugrel is an orally available pro-drug that requires active transformation via the CYP450 along with esterases.Citation22 Activation of the pro-drug requires CYP3 A4 and CYP2B6.Citation23 Clinical studies have verified that inhibition of platelet aggregation is more effective with prasugrel compared with clopidogrel after a single dose in healthy subjects.Citation23 Furthermore, subjects who responded poorly to clopidogrel showed greater platelet-induced inhibition in response to prasugrel.Citation23,Citation24 Further, assessment of secondary endpoints favors prasugrel due to lower incidences of cardiovascular death, nonfatal MI, and rehospitalization due to recurrent ischemia.

Ticagrelor (Brillinta®, AstraZeneca, London, UK), an oral cyclopentyl-triazolo-pyrimidine analog, unlike thienopyridines, is a direct and reversible inhibitor of the P2Y12 receptor that is activated from its pro-drug by CYP3 A.Citation25 Ticagrelor exerts its action via binding to the P2Y12 receptor at a site distinct from the ADP binding site, thus making it an allosteric inhibitor.Citation25 As a consequence of P2Y12 inhibition, ATP is converted to cyclic monophosphate, vasodilator-stimulated phospoprotein is dephosphorylated, and activation of PI3-K is inhibited.Citation26 The PLATO trial compared ticagrelor with clopidogrel in which the primary composite endpoints, stroke, MI, cardiovascular death, and stent thrombosis, were reduced in patients with acute coronary syndromes (ACS) (with or without ST-elevation MI).Citation27 The benefit of ticagrelor appears to be attenuated in patients with lower bodyweight and those not taking lipid-lowering drugs in North American groups relative to comparative studies elsewhere.Citation28 There is no significant difference in major bleeding between the two agents; however, spontaneous (noncoronary artery bypass grafts) or nonprocedural-related bleeding is increased with ticagrelor. Additionally, off-target effects of dyspnea and asymptomatic ventricular pauses are associated with ticagrelor use.Citation28,Citation29 In general, ticagrelor has so far proven superior to current treatment regimens, including a rapid onset of action, acceptable safety profile, and effectiveness in reducing the primary endpoints in ACS patients.

Elinogrel (PRT060128, Novartis, Basel, Switzerland/Portola Pharmaceuticals, South San Francisco, CA) is a direct-acting reversible P2Y12 receptor inhibitor that is currently undergoing clinical investigation (INNOVATE-PCI) for efficacy and safety in patients undergoing percutaneous coronary intervention (PCI) (see ).Citation30 Preclinical data show that intravenous or orally administered elinogrel is superior to clopidogrel and has minimal effect on bleeding times.Citation31 In addition, a single dose of elinogrel has been shown to overcome high platelet reactivity in patients undergoing PCI who were nonresponsive to clopidogrel.Citation31 Elinogrel, while still in clinical development for safety and efficacy assessment in patients, shows promise as a next generation P2Y12 antagonist.

Table 2 Antiplatelet drugs under development

Cangrelor (ARC-69931MX, The Medicines Company, Parsippany, NJ) is an intravenous nontheinopyridine and reversible P2Y12 inhibitor. Like prasugrel and ticagrelor, cangrelor showed a more rapid onset of action and greater degree of platelet inhibition than clopidogrel. Recent evaluations of the inhibitor in the CHAMPION-PCI and CHAMPION-PLATFORM trials were stopped early due to its lack of apparent differences in the primary endpoint of death, MI, or ischemia-driven revascularization 48 hours after PCI.Citation32 Also, the rate of major bleeding in patients undergoing PCI was higher with cangrelor compared with clopidogrel in both studies.Citation33

BX 667 is an orally active reversible P2Y12 receptor antagonist that is metabolized by esterases to form the carboxylic active form, BX 048.Citation34 In-vitro, ADP-induced aggregation is potently inhibited by BX 667. Additionally, administration of BX 667 results in a rapid and sustained inhibition aggregation.Citation35 This observation is also supported by the intravenous BX 048 and oral BX 667 administration in rat arteriovenous-shunt model which showed a similar pharmacodynamic relationship between the plasma concentration of BX 048 and thrombus inhibition.Citation34 This antagonist has yet to be evaluated in healthy human subjects.

Glycoprotein antagonists

αIIbβ3 antagonists

Glycoprotein GPIIbIIIa (αIIbβ3) is the most abundant integrin on the platelet surface.Citation36 αIIbβ3 is known to be involved in both inside-out or outside-in platelet signaling. The inside-out signaling in platelet activation involves the various signaling pathways that converge into a common signaling endpoint that leads to the activation of integrin αIIbβ3.Citation37 Ligand binding of fibrinogen or von Willebrand factor (Vwf) to αIIbβ3 mediates platelet adhesion and aggregation, triggers outside-in integrin activation and results in additional granule secretion, stabilization of platelet adhesion, aggregation, and clot retraction.Citation37

Abciximab (ReoPro®; Eli Lilly) is an antibody developed from the murine human chimera c7E3 Fab, which targets the integrin αIIbβ3,Citation38 preventing integrin binding to fibrinogen and Vwf. Abciximab rapidly binds with high affinity and has a slow rate of dissociation from its target.Citation39 In addition, abciximab binds with high affinity to αvβ3 (vitronectin receptor)Citation40 and low affinity to the leukocyte MAC-1 receptor.Citation41 Initial intravenous administration enables rapid onset of platelet inhibition.Citation42 As abciximab has an extremely short half-life,Citation43 platelet aggregation returns to baseline levels within 12–24 hours following discontinuation of therapy.Citation44,Citation45 Interestingly, the ISAR-REACT trial demonstrated no additional benefit of abciximab over placebo in the reduction of ischemic complications or mortality.Citation43,Citation46 Similarly, among diabetic patients without elevated troponin levels undergoing elective PCI, no difference was observed in primary endpoint events between abciximab and placebo/clopidogrel groups.Citation47 Conversely, in patients with elevated troponin levels, the incidence of mortality and recurrent ischemic complications was significantly reduced with abciximab.Citation48 Careful monitoring must be accompanied with the administration of abciximab as bleeding and thrombocytopenia have been observed.Citation49Citation51

Eptifibatide (Integrilin®, Millenium Pharmaceuticals, Cambridge, MA/Schering-Plough, Kenilworth, NJ) is a cyclic heptapeptide derived from snake venom that contains a KGD (lysine-glycine-aspartic acid) sequence which selectively recognizes αIIβ3.Citation52 The IMPACT-II study showed that a single loading dose followed by continuous infusion for 20–24 hours only resulted in 50% αIIbβ3 receptor blockade; thus, limited benefits and efficacy through eptifibatide were observed.Citation53 The ESPRIT trial, however, which utilized intravenous administration of a double bolusCitation54 followed by maintenance infusion, significantly reduced the 30 days incidence of death, MI, and target vessel revascularization,Citation55 establishing the clinical efficacy for this drug. These observations were confirmed in the PURSUIT trial, which showed an absolute reduction in the 30-day incidence of death and MI on eptifibatide.Citation56 Despite the reduction in mortality, the ACUITY trial also showed an increase incidence of major bleeding in patients with ACS undergoing PCI.Citation57,Citation58

Tirofiban (Aggrastat®, Merck, Whitehouse Station, NJ) is a tyrosine-derivative nonpeptide mimetic reversible inhibitor of αIIbβ3 that specifically and competitively binds to the receptor. Treatment with tirofiban in combination with aspirin and heparin in patients with ACS significantly reduced the 30-day post-treatment incidence of death, MI, or recurrent ischemia.Citation59 Further, tirofiban was superior for ACS patients recovering from invasive coronary angiography.Citation59 As for the use of tirofiban as an adjunct to PCI, tirofiban was shown to be inferior to abciximab in the RESTORE and TARGET trials where the incidence of composite death, nonfatal MI, and urgent target vessel revascularization were higher with tirofiban or abciximab at 30 days.Citation60

Z4 A5 is a novel αIIbβ3 peptide antagonist that is currently in development. This antagonist has been shown to inhibit platelet-induced aggregation and thrombi formation. Additionally, when Z4 A5 was examined along with heparin and/or aspirin in the rabbit arteriovenous shunt thrombosis model, it was shown to be an effective antithrombotic agent when administered with aspirin.Citation61 The pharmacodynamics and pharmacokinetics in humans are currently under investigation.Citation61

Additional glycoprotein antagonists

Additional glycoprotein targets have received a fair amount of attention in the drive to develop novel approaches for antiplatelet intervention. The Vwf, a multimeric glycoprotein that acts as a bridging element between damaged endothelial sites and the glycoprotein receptors on platelets, is one such target. The A1 and A3 domains of Vwf bind to collagen, while the A1 domain is bound to the GPIb-IX-V platelet receptor complex.Citation62,Citation63 Vwf also binds to active αIIbβ3 on the platelet surface. Interactions between αIIbβ3 and Vwf contribute to the final, irreversible binding of platelets to the subendothelium and play a leading role in platelet aggregation.Citation64 A second target receiving attention as a potential site for antiplatelet therapy is the collagen receptor glycoprotein VI (GPVI). The collagen-GPVI interaction triggers subsequent tyrosine phosphorylation of the immunoreceptor tyrosine-based activation motif of the Fc receptor γ chain, activating the Syk kinases pathway, LAT, SLP-76, and phospholipase Cγ2,Citation65 resulting in platelet activation or aggregation.

Vwf antagonists

AJW200 is an IgG4 humanized monoclonal antibody to Vwf which has been shown to specifically inhibit high-shear-stress-induced platelet aggregation in a concentration-dependent manner in vitro in blood from human volunteers.Citation66

ARC1779 (Archemix Corp, San Francisco, CA) is an aptamer-based antagonist. This second generation nuclease-resistant aptamer is conjugated to a 20-Kda polyethylene glycol and binds with high affinity to the active Vwf A1-domain and inhibits Vwf-dependent platelet aggregation.Citation67 A Phase II trial demonstrated that continuous infusion of ARC1779 effectively increased platelet counts in critically ill thrombotic thrombocytopenic purpura patients by preventing platelet aggregation and loss of platelets. Cessation of ARC1779 infusion resulted in platelet count reduction and progression of thrombotic thrombocytopenic purpura-related organ damage. Citation68 This drug is currently under clinical investigation.

Other Vwf antagonists in clinical development or investigations include ARC15105, ALX-0081 (Ablynx), ALX-0681, and 82D6 A3. ARC15105 is a chemically advanced aptamer with assumed higher affinity to Vwf, but less specific inhibitor of Vwf-dependent platelet aggregation than ARC1779, based on ex-vivo trials.Citation69 The preclinical and clinical trials have shown that ALX-0081, a bivalent humanized nanobody that recognizes the Glycoprotein Ib (GPIb) binding site of Vwf, is a potent and safe inhibitor of Vwf-mediate platelet aggregation over a wide range of doses when administered in combination with aspirin, heparin, and clopidogrel. ALX-0081 is currently under investigation in PCI patients in a Phase II trial. 82D6 A3, a monoclonal antibody directed against amino acids Arg-963, Pro-981, Asp-1009, Arg-1016, Ser-1020, Met-1022, and His-1023 of the Vwf A3 domain,Citation70 was shown to result in complete inhibition of Vwf binding to collagen during the first 3 days after stent implantation in baboons.Citation71 Further trials will need to follow to verify 82D6 A3 efficacy, safety, and tolerability.

GPVI receptor antagonists

PR-15 (Revacept®, ABX-CRO/Medifacts GmbH, Goerlitz, Saxony, Germany) is a soluble, dimeric glycoprotein (GPVI)-Fc that has been shown to adhere to exposed collagen in endothelial lesions preventing the binding to platelet GPVI receptors. Collagen-induced human platelet adhesion or plaque formation were significantly reduced with pre-treatment of soluble GPVI-Fc.Citation72,Citation73 Similarly, infusion of GPVI-Fc was shown to virtually abolish stable arrest and aggregation of platelets following vascular injury in mice.Citation65 Subsequently, a Phase I clinical trial demonstrated that intravenous administration of PR-15 is safe and well tolerated by healthy volunteers.Citation74

DZ-697b is an orally active collagen and ristocetin inhibitor. Safety and efficacy have been assessed in a Phase I trial which showed potential benefits such that bleeding time was substantially shortened compared with clopidogrel treatment.Citation75 DZ-697b is currently under clinical investigation.

GPIb receptor antagonists

Novel targets still under investigation include h6B4-Fab, GPGP-290, and SZ2. h6B4-Fab is a murine monoclonal antibody, derived from the humanized Fab fragment of 6B4 targeting GPIbα and neutralizes the binding site of the Vwf A1 domain.Citation76,Citation77 6B4 has been shown to inhibit platelet adhesion by competing with Vwf for binding to GPIbα under high-shear conditions. Moreover, preliminary data show 6B4 has no effect on platelet count or bleeding times in vivo in baboons, but dose- and time-dependently inhibited ristocetin-induced platelet aggregation.Citation78 GPG-290 is a recombinant, chimeric antibody purified from Chinese hamster ovary cell culture that contains the amino-terminal 290 amino acids of GPIbα linked to the human IgG1. GPG-290 treated dogs were shown to exhibit prolonged bleeding compared with the clopidogrel-treated control, despite the prevention of coronary artery thrombosis.Citation79 SZ2, a monoclonal antibody developed against GPIbα, has also been shown to inhibit both ristocetin- and botrocetin-induced platelet aggregation in vitro.Citation80 Preclinical investigations are still underway to determine the in-vivo efficacy of SZ2.

Phosphodiesterase antagonists

Platelets express three phosphodiesterase (PDE) isoenzymes, PDE 2, 3, and 5. PDEs regulate the levels of 3,5′-cyclic adenosine monophosphate (Camp) and 3′,5′-cyclic guanosine monophosphate (Cgmp) by catalyzing the hydrolysis of Camp and Cgmp to inactive 5′-AMP and 5′-GMP, respectively. Citation81 Platelet activation relies on degradation of Camp and Cgmp; hence regulating these secondary messengers is fundamental in regulating platelet activation and thrombosis.

Cilostazol (Pletal®, Otsuka Pharmaceutical Co, Tokyo, Kapan) is a type III PDE (PDE3) selective oral inhibitor.Citation82 Liu and colleagues have shown that cilostazol enhances the interstitial concentration of adenosine in several in-vitro and in-vivo modelsCitation83 by inhibiting adenosine uptake. This in turn stimulates A2 receptors, which further increases Camp levels. As a result, platelet-induced aggregation is reversibly inhibited by cilostazol.Citation84 Cilostazol is extensively metabolized by CYP3 A4, while CYP2C19 is also shown to have a minor role in cilostazol metabolism.Citation85 Cilostazol is safe and effective in reducing the incidence of repeated revascularization after PCI and risk of restenosis; however, this drug does not show superiority in reducing the primary composite endpoints of adverse cardiovascular events after drug-elution stent implantation.Citation86 Despite the functional implications of adjunctive treatment with cilostazol compared with standard aspirin and clopidogrel treatment, as shown in the OPTIMUS-2 study, the accompanied side effects (headaches, gastrointestinal symptoms, and skin rash) often lead to the discontinuation of the drug.Citation87

Dipyridamole (Aggrenox®, Boehringer Ingelheim, Ingelheim, Germany) is a pyridopyrimidine derivative with both antiplatelet and vasodilator properties.Citation88 Similar to cilostazol, dipyridamole inhibits cyclic nucleotide phosphodiesterase and blocks adenosine uptake, which results in increased Camp.Citation89 The ESPS-2 and ESPRIT trials showed that dual treatment of dipyridamole and aspirin reduced risk of stroke or death by 37% compared with aspirin alone.Citation90,Citation91 Based on the ESPRIT and ESPS-2 trials, dipyridamole has been FDA approved for stroke prevention.Citation91

Thromboxane A2 receptor antagonists

Platelets express the thromboxane receptor α (TPα), a GPCR that is coupled to Gq and G12/13 and signals platelet activation through a number of intracellular pathways which converge to reinforce primary platelet activation through thrombin or collagen.Citation92

S18886 (terutroban) is an oral reversible inhibitor of TPα. In preclinical studies, S18886 dose-dependently prolonged occlusive thrombus formation in animal models, but did not alter the size of the myocardial infarct size in the ischemia-perfusion model. S18886 and clopidogrel were effective in preventing occlusive thrombus formation with a moderate increase in bleeding time.Citation93 Subsequently, however, in the Phase III clinical trial (PERFORM), S18886 did not meet the predefined criteria for noninferiority since S18886 and aspirin had similar rates of protection without safety advantages for S18886.Citation94

Z-335 ((+/−)-sodium[2-(4-chlorophenylsulfonyl-aminomethyl) indan-5-yl]acetate monohydrate) is an oral TPα antagonist that has previously been shown to dose-dependently inhibit the specific binding of [3H]SQ-29548 (TPα inhibitor) to human and guinea pig platelet membranes.Citation95 In healthy male Japanese volunteers, Z-335 inhibited U46619-induced platelet aggregation within 2 hours of administration.Citation96

BM-573, another investigational inhibitor that targets TPα, has been shown to halt the progression of atherosclerosis in low-density lipoprotein receptor deficient mice.Citation97 Preclinical models have shown that arachidonic acid-induced aggregation is completely inhibited in the presence of BM-573Citation98 and clinical studies on this compound are currently ongoing.

Thrombin receptor antagonists

Thrombin activates human platelets via two protease activated receptors (PARs), PAR1 and PAR4. PAR activation leads to a diverse range of pro-thrombotic signaling events mediated through Gq, G12/13, and possibly Gi, resulting in phospholipase β activation, Rho activation, and adenyl cyclase inhibition, respectively. PAR activation requires thrombin cleavage of the amino terminus of the receptor, revealing a tethered ligand. While it has been challenging to develop an inhibitor that can directly compete with the endogenous tethered ligand, development of PAR1 inhibitors as a therapeutic target to minimize uncontrolled platelet activation has recently been investigated.

SCH 530348 (Vorapaxar®, Merck and Co, Whitehouse Station, NJ) is an orally active synthetic analog of himbacineCitation99 that competitively binds with high affinity to the PAR1. Previous in-vitro assays show SCH 530348 inhibited thrombin- and thrombin receptor activating peptide-induced platelet aggregation, without affecting the aggregation induced by ADP, U46619, or collagen. In addition, SCH 530348 did not affect the prothrombin and activated partial thromboplastin time, suggesting that bleeding time may not be increased. Pre-clinically, cynomolgus monkeys treated with SCH 530348 alone or in addition with aspirin and clopidogrel, showed no increase in bleeding times.Citation100 The TRA-PCI study verified that addition of SCH 530348 to standard antiplatelet therapy (aspirin and clopidogrel) was not associated with increases in thrombolysis in MI (TIMI) or bleeding compared with the control group.Citation101 The Phase III clinical trials TRA-CER and TRA 2ºP-TIMI, which sought to assess the impact of vorapaxar on cardiovascular death, MI, stroke, and recurrent vascular events in patients with established coronary, cerebral, or peripheral atherosclerosisCitation102 failed due to unforeseen intracranial bleeding.Citation102

Preclinical trials showed that oral administration of the PAR1 antagonist, E5555 (Atopaxar®, Eisai Co Ltd, Tokyo, Japan), significantly prolonged bleeding times in guinea pigs.Citation103 Further, PECAM-1, active αIIbβ3, GPIb, thrombospondin, and vitronectin expression were significantly reduced by E5555 in whole blood flow cytometry. Clinical studies have shown that E5555 attenuated thrombin-induced but not ADP-induced platelet aggregation.Citation104,Citation105

Additional PAR-1 antagonists SCH 205831 and SCH 602539 are still under investigation. Preliminary data show SCH 205831 derived from himbacine inhibited platelet deposition in baboons with arteriovenous-shunt thrombosis. Similarly, SCH 602539 inhibited thrombosis in a dose-dependent manner in the Folts model of thrombosis in anesthetized cynomolgus monkeys.Citation106 These compounds continue to be developed in preclinical models.

Conclusion

Significant progress has been made in advancing our understanding of how platelet activation directly regulates thrombus formation in the vessel leading to occlusive thrombi and stroke. However, a continued need for the development of new antiplatelet therapies exists as the risk for MI, stroke, and death, remain a persistent problem for individuals suffering from cardiovascular disease. Further, while aspirin continues to be the first line of pharmacological intervention in antiplatelet therapy, the risk of bleeding is significantly exacerbated by its irreversible action coupled to the additional regimen of dual therapy often employed to minimize thrombotic events. In hopes of reducing prolonged bleeding or myocardial infarct events, newer compounds continue to be developed to target alternative sites in the platelet. The successful implementation of these strategies may significantly reduce the morbidity and mortality in cardiovascular disease due to unwanted platelet activation as well as excessive bleeding due to traditional approaches. Even with the newer antiplatelet drugs entering the market in the near future, we are faced with the realization that activation of the platelet involves an increasingly complex signaling network. Hence, new frontiers will need to be explored which will take advantage of this signaling to reveal novel therapeutic targets with diminished off-target effects.

Acknowledgments

This work was supported in part by the National Institutes of Health National Heart, Lung, and Blood Institutes grant HL089457 (MH).

Disclosure

The authors report no conflicts of interest in this work.

References

  • CohenSBraimanAShubinskyGIsakovNProtein kinase C-theta in platelet activationFEBS Lett2011585203208321521944869
  • TrumelCPayrastreBPlantavidMA key role of adenosine diphosphate in the irreversible platelet aggregation induced by the PAR1-activating peptide through the late activation of phosphoinositide 3-kinaseBlood199994124156416510590060
  • JungSMOhnumaMWatanabeNSonodaMHandaMMoroiMAnalyzing the mechanism of Rap1 activation in platelets: Rap1 activation is related to the release reaction mediated through the collagen receptor GPVIThromb Res2006118450952116380154
  • KimSKunapuliSPP2Y12 receptor in platelet activationPlatelets2011221545821231822
  • SchrorKThe basic pharmacology of ticlopidine and clopidogrelPlatelets19934525226121043748
  • McTavishDFauldsDGoaKLTiclopidine. An updated review of its pharmacology and therapeutic use in platelet-dependent disordersDrugs19904022382592226215
  • AlbersGWRole of ticlopidine for prevention of strokeStroke19922369129161595115
  • DerianCKFriedmanPAEffect of ticlopidine ex vivo on platelet intracellular calcium mobilizationThromb Res198850165763400086
  • CattaneoMNew P2Y(12) inhibitorsCirculation2010121117117920048234
  • GentMBlakelyJAEastonJDThe Canadian American Ticlopidine Study (CATS) in thromboembolic strokeLancet198918649121512202566778
  • HassWKEastonJDAdamsHPJrA randomized trial comparing ticlopidine hydrochloride with aspirin for the prevention of stroke in high-risk patients. Ticlopidine Aspirin Stroke Study GroupN Engl J Med198932185015072761587
  • QuinnMJFitzgeraldDJTiclopidine and clopidogrelCirculation1999100151667167210517740
  • MehtaSRYusufSPetersRJEffects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: the PCI-CURE studyLancet2001358928152753311520521
  • YusufSZhaoFMehtaSRChrolaviciusSTognoniGFoxKKEffects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevationN Engl J Med2001345749450211519503
  • A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). CAPRIE Steering CommitteeLancet19963489038132913398918275
  • MegaJLCloseSLWiviottSDCytochrome p-450 polymorphisms and response to clopidogrelN Engl J Med2009360435436219106084
  • SimonTVerstuyftCMary-KrauseMGenetic determinants of response to clopidogrel and cardiovascular eventsN Engl J Med2009360436337519106083
  • SugidachiAAsaiFOgawaTInoueTKoikeHThe in vivo pharmacological profile of CS-747, a novel antiplatelet agent with platelet ADP receptor antagonist propertiesBr J Pharmacol200012971439144610742300
  • SugidachiAAsaiFYonedaKAntiplatelet action of R-99224, an active metabolite of a novel thienopyridine-type G(i)-linked P2T antagonist, CS−747Br J Pharmacol20011321475411156560
  • HasegawaMSugidachiAOgawaTIsobeTJakubowskiJAAsaiFStereoselective inhibition of human platelet aggregation by R-138727, the active metabolite of CS-747 (prasugrel, LY640315), a novel P2Y12 receptor inhibitorThromb Haemost200594359359816268477
  • SaviPPereilloJMUzabiagaMFIdentification and biological activity of the active metabolite of clopidogrelThromb Haemost200084589189611127873
  • FaridNAPayneCDSmallDSCytochrome P450 3 A inhibition by ketoconazole affects prasugrel and clopidogrel pharmacokinetics and pharmacodynamics differentlyClin Pharmacol Ther200781573574117361128
  • BrandtJTPayneCDWiviottSDA comparison of prasugrel and clopidogrel loading doses on platelet function: magnitude of platelet inhibition is related to active metabolite formationAm Heart J2007153166.e91617174640
  • MontalescotGWiviottSDBraunwaldEPrasugrel compared with clopidogrel in patients undergoing percutaneous coronary intervention for ST-elevation myocardial infarction (TRITON-TIMI 38): double-blind, randomised controlled trialLancet2009373966572373119249633
  • BirkelandKParraDRosensteinRAntiplatelet therapy in acute coronary syndromes: focus on ticagrelorJ Blood Med2010119721922282698
  • AndersonSDShahNKYimJEpsteinBJEfficacy and safety of ticagrelor: a reversible P2Y12 receptor antagonistAnn Pharmacother201044352453720124464
  • MahaffeyKWWojdylaDMCarrollKTicagrelor compared with clopidogrel by geographic region in the Platelet Inhibition and Patient Outcomes (PLATO) trialCirculation2011124554455421709065
  • WallentinLBeckerRCBudajATicagrelor versus clopidogrel in patients with acute coronary syndromesN Engl J Med2009361111045105719717846
  • CannonCPHustedSHarringtonRASafety, 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: primary results of the DISPERSE-2 trialJ Am Coll Cardiol200750191844185117980250
  • LeonardiSRaoSVHarringtonRARationale and design of the randomized, double-blind trial testing IntraveNous and Oral administration of elinogrel, a selective and reversible P2Y(12)-receptor inhibitor, versus clopidogrel to eVAluate Tolerability and Efficacy in nonurgent Percutaneous Coronary Interventions patients (INNOVATE-PCI)Am Heart J20101601657220598974
  • GurbelPABlidenKPAntoninoMJThe effect of elinogrel on high platelet reactivity during dual antiplatelet therapy and the relation to CYP2C19*2 genotype: first experience in patientsJ Thromb Haemost201081435319817997
  • HarringtonRAStoneGWMcNultySPlatelet inhibition with cangrelor in patients undergoing PCIN Engl J Med2009361242318232919915221
  • BhattDLLincoffAMGibsonCMIntravenous platelet blockade with cangrelor during PCIN Engl J Med2009361242330234119915222
  • WangYXVinceletteJda CunhaVA novel P2Y(12) adenosine diphosphate receptor antagonist that inhibits platelet aggregation and thrombus formation in rat and dog modelsThromb Haemost200797584785517479197
  • PostJMAlexanderSWangYXNovel P2Y12 adenosine diphosphate receptor antagonists for inhibition of platelet aggregation (II): pharmacodynamic and pharmacokinetic characterizationThromb Res2008122453354018539312
  • GogstadGOBrosstadFKrutnesMBHagenISolumNOFibrinogen-binding properties of the human platelet glycoprotein Iib- = IIIa complex: a study using crossed-radioimmunoelectrophoresisBlood19826036636716286013
  • LiZDelaneyMKO’BrienKADuXSignaling during platelet adhesion and activationArterioscler Thromb Vasc Biol201030122341234921071698
  • KristensenSDWurtzMGroveELContemporary use of glycoprotein Iib/IIIa inhibitorsThromb Haemost2012107221522422234385
  • WagnerCLMascelliMANeblockDSWeismanHFCollerBSJordanREAnalysis of GPIIb/IIIa receptor number by quantification of 7E3 binding to human plateletsBlood19968839079148704248
  • TamSHSassoliPMJordanRENakadaMTAbciximab (ReoPro, chimeric 7E3 Fab) demonstrates equivalent affinity and functional blockade of glycoprotein Iib/IIIa and alpha(v)beta3 integrinsCirculation19989811108510919736595
  • SimonDIXuHOrtleppSRogersCRaoNK7E3 monoclonal antibody directed against the platelet glycoprotein Iib/IIIa cross-reacts with the leukocyte integrin Mac-1 and blocks adhesion to fibrinogen and ICAM-1Arterioscler Thromb Vasc Biol19971735285359102172
  • KleimanNSRaiznerAEJordanRDifferential inhibition of platelet aggregation induced by adenosine diphosphate or a thrombin receptor-activating peptide in patients treated with bolus chimeric 7E3 Fab: implications for inhibition of the internal pool of GPIIb/IIIa receptorsJ Am Coll Cardiol1995267166516717594101
  • KastratiAMehilliJSchuhlenHA clinical trial of abciximab in elective percutaneous coronary intervention after pretreatment with clopidogrelN Engl J Med2004350323223814724302
  • LimGBAntiplatelet therapy: intracoronary abciximab during PCI for STEMINat Rev Cardiol20119525822411290
  • Van de WerfFArdissinoDBuenoHAcute coronary syndromes: considerations for improved acceptance and implementation of management guidelinesExpert Rev Cardiovasc Ther201210448950322458581
  • SchomigASchmittCDibraAOne year outcomes with abciximab vs. placebo during percutaneous coronary intervention after pretreatment with clopidogrelEur Heart J200526141379138415734767
  • KastratiAMehilliJNeumannFJAbciximab in patients with acute coronary syndromes undergoing percutaneous coronary intervention after clopidogrel pretreatment: the ISAR-REACT 2 randomized trialJAMA2006295131531153816533938
  • BhattDLTopolEJCurrent role of platelet glycoprotein Iib/IIIa inhibitors in acute coronary syndromesJAMA2000284121549155811000650
  • CollerBSPlatelet GPIIb/IIIa antagonists: the first anti-integrin receptor therapeuticsJ Clin Invest199710011 SupplS57S609413403
  • AmmarTScudderLECollerBSIn vitro effects of the platelet glycoprotein Iib/IIIa receptor antagonist c7E3 Fab on the activated clotting timeCirculation19979536146179024148
  • MichelsonADAntiplatelet therapies for the treatment of cardiovascular diseaseNat Rev Drug Discov20109215416920118963
  • ScarboroughRMRoseJWHsuMABarbourin. A GPIIb-IIIa-specific integrin antagonist from the venom of Sistrurus m. barbouriJ Biol Chem199126615935993622033037
  • Randomised placebo-controlled trial of effect of eptifibatide on complications of percutaneous coronary intervention: IMPACT-II. Integrilin to Minimise Platelet Aggregation and Coronary Thrombosis-IILancet19973499063142214289164315
  • GilchristICO’SheaJCKosoglouTPharmacodynamics and pharmacokinetics of higher-dose, double-bolus eptifibatide in percutaneous coronary interventionCirculation2001104440641111468201
  • Novel dosing regimen of eptifibatide in planned coronary stent implantation (ESPRIT): a randomised, placebo-controlled trialLancet200035692472037204411145489
  • KleimanNSLincoffAMFlakerGCEarly percutaneous coronary intervention, platelet inhibition with eptifibatide, and clinical outcomes in patients with acute coronary syndromes. PURSUIT InvestigatorsCirculation2000101775175710683348
  • StoneGWBertrandMEMosesJWRoutine upstream initiation vs deferred selective use of glycoprotein Iib/IIIa inhibitors in acute coronary syndromes: the ACUITY Timing trialJAMA2007297659160217299194
  • GiuglianoRPWhiteJABodeCEarly versus delayed, provisional eptifibatide in acute coronary syndromesN Engl J Med2009360212176219019332455
  • CannonCPWeintraubWSDemopoulosLAComparison of early invasive and conservative strategies in patients with unstable coronary syndromes treated with the glycoprotein Iib/IIIa inhibitor tirofibanN Engl J Med2001344251879188711419424
  • TopolEJMoliternoDJHerrmannHCComparison of two platelet glycoprotein Iib/IIIa inhibitors, tirofiban and abciximab, for the prevention of ischemic events with percutaneous coronary revascularizationN Engl J Med2001344251888189411419425
  • JingBBLiYXZhangHAntithrombotic activity of Z4 A5, a new platelet glycoprotein Iib/IIIa receptor antagonist evaluated in a rabbit arteriovenous shunt thrombosis modelThromb Res2011128546346921924458
  • HoylaertsMFPlatelet-vessel wall interactions in thrombosis and restenosis role of von Willebrand factorVerh K Acad Geneeskd Belg19975931611839490916
  • ParetiFIFujimuraYDentJAHollandLZZimmermanTSRuggeriZMIsolation and characterization of a collagen binding domain in human von Willebrand factorJ Biol Chem19862613215310153153490481
  • NicholsWLHultinMBJamesAHvon Willebrand disease (VWD): evidence-based diagnosis and management guidelines, the National Heart, Lung, and Blood Institute (NHLBI) Expert Panel report (USA)Haemophilia200814217123218315614
  • GibbinsJMOkumaMFarndaleRBarnesMWatsonSPGlycoprotein VI is the collagen receptor in platelets which underlies tyrosine phosphorylation of the Fc receptor gamma-chainFEBS Lett199741322552599280292
  • KageyamaSYamamotoHNakazawaHPharmacokinetics and pharmacodynamics of AJW200, a humanized monoclonal antibody to von Willebrand factor, in monkeysArterioscler Thromb Vasc Biol200222118719211788481
  • GilbertJCDeFeo-FrauliniTHutabaratRMFirst-in-human evaluation of anti von Willebrand factor therapeutic aptamer ARC1779 in healthy volunteersCirculation2007116232678268618025536
  • KnoblPJilmaBGilbertJCHutabaratRMWagnerPGJilma-StohlawetzPAnti-von Willebrand factor aptamer ARC1779 for refractory thrombotic thrombocytopenic purpuraTransfusion200949102181218519538431
  • Siller-MatulaJMMerhiYTanguayJFARC15105 is a potent antagonist of von Willebrand factor mediated platelet activation and adhesionArterioscler Thromb Vasc Biol201232490290922282355
  • VanhoorelbekeKDepraetereHRomijnRAHuizingaEGDe MaeyerMDeckmynHA consensus tetrapeptide selected by phage display adopts the conformation of a dominant discontinuous epitope of a monoclonal anti-VWF antibody that inhibits the von Willebrand factor-collagen interactionJ Biol Chem200327839378153782112855711
  • De MeyerSFStaelensSBadenhorstPNCoronary artery in-stent stenosis persists despite inhibition of the von Willebrand factor – collagen interaction in baboonsThromb Haemost20079861343134918064334
  • MassbergSKonradIBultmannASoluble glycoprotein VI dimer inhibits platelet adhesion and aggregation to the injured vessel wall in vivoFASEB J200418239739914656994
  • SchulzCPenzSHoffmannCPlatelet GPVI binds to collagenous structures in the core region of human atheromatous plaque and is critical for atheroprogression in vivoBasic Res Cardiol2008103435636718431526
  • UngererMRosportKBultmannANovel antiplatelet drug revacept (Dimeric Glycoprotein VI-Fc) specifically and efficiently inhibited collagen-induced platelet aggregation without affecting general hemostasis in humansCirculation2011123171891189921502572
  • ZafarMUIbanezBChoiBGA new oral antiplatelet agent with potent antithrombotic properties: comparison of DZ-697b with clopidogrel a randomised phase I studyThromb Haemost2010103120521220062928
  • HuizingaEGTsujiSRomijnRAStructures of glycoprotein Ibalpha and its complex with von Willebrand factor A1 domainScience200229755841176117912183630
  • UffSClemetsonJMHarrisonTClemetsonKJEmsleyJCrystal structure of the platelet glycoprotein Ib(alpha) N-terminal domain reveals an unmasking mechanism for receptor activationJ Biol Chem200227738356573566312087105
  • WuDVanhoorelbekeKCauwenberghsNInhibition of the von Willebrand (VWF)-collagen interaction by an antihuman VWF monoclonal antibody results in abolition of in vivo arterial platelet thrombus formation in baboonsBlood200299103623362811986216
  • HennanJKSwilloREMorganGAPharmacologic inhibition of platelet Vwf-GPIb alpha interaction prevents coronary artery thrombosisThromb Haemost200695346947516525575
  • YangJJiSDongNZhaoYRuanCEngineering and characterization of a chimeric anti-platelet glycoprotein Ibalpha monoclonal antibody and preparation of its Fab fragmentHybridoma (Larchmt)201029212513220443704
  • BenderATBeavoJACyclic nucleotide phosphodiesterases: molecular regulation to clinical usePharmacol Rev200658348852016968949
  • SchrorKThe pharmacology of cilostazolDiabetes Obes Metab20024Suppl 2S14S1912180353
  • LiuYFongMConeJWangSYoshitakeMKambayashiJInhibition of adenosine uptake and augmentation of ischemia-induced increase of interstitial adenosine by cilostazol, an agent to treat intermittent claudicationJ Cardiovasc Pharmacol200036335136010975593
  • UeharaSHirayamaAEffects of cilostazol on platelet functionArzneimittel-Forschung19893912153115342487508
  • SuriAForbesWPBramerSLEffects of CYP3 A inhibition on the metabolism of cilostazolClin Pharmacokinet199937 Suppl 2616810702888
  • SuhJWLeeSPParkKWMulticenter randomized trial evaluating the efficacy of cilostazol on ischemic vascular complications after drug-eluting stent implantation for coronary heart disease: results of the CILON-T (influence of CILostazol-based triple antiplatelet therapy ON ischemic complication after drug-eluting stenT implantation) trialJ Am Coll Cardiol201157328028921232664
  • AngiolilloDJCapranzanoPGotoSA randomized study assessing the impact of cilostazol on platelet function profiles in patients with diabetes mellitus and coronary artery disease on dual antiplatelet therapy: results of the OPTIMUS-2 studyEur Heart J200829182202221118567918
  • AktasBUtzAHoenig-LiedlPWalterUGeigerJDipyridamole enhances NO/Cgmp-mediated vasodilator-stimulated phosphoprotein phosphorylation and signaling in human platelets: in vitro and in vivo/ex vivo studiesStroke200334376476912624305
  • GamboaAAbrahamRDiedrichARole of adenosine and nitric oxide on the mechanisms of action of dipyridamoleStroke200536102170217516141426
  • DienerHCCunhaLForbesCSiveniusJSmetsPLowenthalAEuropean Stroke Prevention Study2. Dipyridamole and acetyl-salicylic acid in the secondary prevention of strokeJ Neurol Sci19961431–21138981292
  • HalkesPHvan GijnJKappelleLJKoudstaalPJAlgraAAspirin plus dipyridamole versus aspirin alone after cerebral ischaemia of arterial origin (ESPRIT): randomised controlled trialLancet200636795231665167316714187
  • GiannarelliCZafarMUBadimonJJProstanoid and TP-receptors in atherothrombosis: is there a role for their antagonism?Thromb Haemost2010104594995420886180
  • HongTTHuangJDriscollELucchesiBRPreclinical evaluation of S18886 in an experimental model of coronary arterial thrombosisJ Cardiovasc Pharmacol200648523924817110806
  • BousserMGAmarencoPChamorroATerutroban versus aspirin in patients with cerebral ischaemic events (PERFORM): a randomised, double-blind, parallel-group trialLancet201137797822013202221616527
  • TanakaTFukutaYHigashinoRAntiplatelet effect of Z-335, a new orally active and long-lasting thromboxane receptor antagonistEur J Pharmacol1998357153609788773
  • MatsunoHUematsuTNiwaMPharmacokinetic and pharmacodynamic properties of a new thromboxane receptor antagonist (Z-335) after single and multiple oral administrations to healthy volunteersJ Clin Pharmacol200242778279012092745
  • CyrusTYaoYDingTDogneJMPraticoDA novel thromboxane receptor antagonist and synthase inhibitor, BM-573, reduces development and progression of atherosclerosis in LDL receptor deficient miceEur J Pharmacol20075611–310511117306791
  • Tchana-SatoVDogneJMLambermontBEffects of BM-573, a thromboxane A2 modulator on systemic hemodynamics perturbations induced by U-46619 in the pigProstaglandins Other Lipid Mediat2005781–4829516303607
  • ChackalamannilSWangYGreenleeWJDiscovery of a novel, orally active himbacine-based thrombin receptor antagonist (SCH 530348) with potent antiplatelet activityJ Med Chem200851113061306418447380
  • ChintalaMStronyJYangBKurowskiSLiQSCH 602539, a protease-activated receptor-1 antagonist, inhibits thrombosis alone and in combination with cangrelor in a Folts model of arterial thrombosis in cynomolgus monkeysArterioscler Thromb Vasc Biol201030112143214920798382
  • BeckerRCMoliternoDJJenningsLKSafety and tolerability of SCH 530348 in patients undergoing non-urgent percutaneous coronary intervention: a randomised, double-blind, placebo-controlled phase II studyLancet2009373966791992819286091
  • MorrowDASciricaBMFoxKAEvaluation of a novel anti-platelet agent for secondary prevention in patients with a history of atherosclerotic disease: design and rationale for the Thrombin-Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events (TRA 2 degrees P)-TIMI 50 trialAm Heart J20091583335341e319699854
  • KogushiMMatsuokaTKawataTThe novel and orally active thrombin receptor antagonist E5555 (Atopaxar) inhibits arterial thrombosis without affecting bleeding time in guinea pigsEur J Pharmacol20116571–313113721300059
  • SerebruanyVLKogushiMDastros-PiteiDFlatherMBhattDLThe in-vitro effects of E5555, a protease-activated receptor (PAR)-1 antagonist, on platelet biomarkers in healthy volunteers and patients with coronary artery diseaseThromb Haemost2009102111111919572075
  • GotoSOgawaHTakeuchiMFlatherMDBhattDLInvestigatorsJLDouble-blind, placebo-controlled Phase II studies of the protease-activated receptor 1 antagonist E5555 (atopaxar) in Japanese patients with acute coronary syndrome or high-risk coronary artery diseaseEur Heart J201031212601261320805115
  • ChintalaMShimizuKOgawaMYamaguchiHDoiMJensenPBasic and translational research on proteinase-activated receptors: antagonism of the proteinase-activated receptor 1 for thrombin, a novel approach to antiplatelet therapy for atherothrombotic diseaseJ Pharmacol Sci2008108443343819098390