You are on page 1of 12

ATVB in Focus

New Targets of Antiplatelet Drug Development


Series Editor: Xiaoping Du

Current State and Novel Approaches of Antiplatelet Therapy


Pat Metharom, Michael C. Berndt, Ross I. Baker, Robert K. Andrews

Abstract—An unresolved problem with clinical use of antiplatelet therapy is that a significant number of individuals either
still get thrombosis or run the risk of life-threatening bleeding. Antiplatelet drugs are widely used clinically, either
chronically for people at risk of athero/thrombotic disease or to prevent thrombus formation during surgery. However, a
subpopulation may be resistant to standard doses, while the platelet targets of these drugs are also critical for the normal
hemostatic function of platelets. In this review, we will briefly examine current antiplatelet therapy and existing targets
while focusing on new potential approaches for antiplatelet therapy and improved monitoring of effects on platelet
reactivity in individuals, ultimately to improve antithrombosis with minimal bleeding. Primary platelet adhesion-signaling
receptors, glycoprotein (GP)Ib-IX-V and GPVI, that bind von Willebrand factor/collagen and other prothrombotic factors
are not targeted by drugs in clinical use, but they are of particular interest because of their key role in thrombus formation
at pathological shear.   (Arterioscler Thromb Vasc Biol. 2015;35:1327-1338. DOI: 10.1161/ATVBAHA.114.303413.)
Key Words: anticoagulants ◼ blood coagulation ◼ blood platelets ◼ drug therapy ◼ platelet adhesion inhibitor

A ntiplatelet and anticoagulant drugs, used alone or in com-


bination, are a cornerstone of clinical treatment for human
diseases associated with increased athero/thrombotic risks, such
exist for the foreseeable future, emphasizing the clear need for
improved approaches for therapy and monitoring.
There are several classes of antiplatelet drugs, either cur-
as heart attack and stroke, and in preventing thrombosis during rently in clinical use (Table 1) or in preclinical or early clini-
surgery or other medical procedures involving vascular disrup- cal stages of development (Table 2), which act at a variety
Downloaded from http://ahajournals.org by on February 11, 2024

tion or perturbed blood flow.1–6 The confounding issue is that of targets. These include platelet receptors, their binding
rapid platelet activation and aggregation leading to thrombus partners, signaling proteins, or soluble mediators of plate-
formation in response to vascular injury is also a critical physi- let function (Figure 1). In this review, we will discuss these
ological mechanism,7 and blocking platelet function can there- therapeutic targets and their known pathological and physi-
fore elevate bleeding risk. Individuals within a population or a ological roles in healthy or diseased vasculature, the scientific
disease cohort will also vary in their relative platelet reactivity basis for their selection and inhibition by existing agents, and
and thrombotic propensity because of variations in platelet phe- new approaches for modulating their pathological function.
notype or genotype coincident with other thrombotic risk fac- Importantly, mechanisms of platelet activation and thrombus
tors, thereby complicating the development of practical clinical formation at the vessel wall need to be considered in the con-
guidelines for antiplatelet drugs, as well as dosing requirements text of shear stress and normal or abnormal blood flow.
and monitoring. For the latter, there is currently a lack of reli-
able platelet analytic, genotyping or functional testing that con- Platelet Activation and Thrombus Formation at
fidently predicts either thrombotic/bleeding risk or efficacy of the Vessel Wall
antiplatelet treatment in individuals. Of further importance, the The capacity for human platelets circulating in the blood-
availability of drugs (oral or intravenous) and their pharmaco- stream to attach to a specific region of the vasculature and
logical half-life in blood can potentially affect user compliance activate and aggregate within seconds to minutes is a sophis-
or reversibility, respectively. Thus, the problem of controlling ticated biological accomplishment, in particular at arterial
bleeding versus thrombosis by antiplatelet therapy is likely to flow rates where fluid shear forces act powerfully against
cell adhesion. Adherent activated platelets promote localized
Please see http://atvb.ahajournals.org/site/misc/ coagulation, release proinflammatory mediators, and recruit
ATVB_in_Focus.xhtml for all articles published leukocytes to initiate wound healing.7 At arterial shear rates,
in this series. this is enabled principally by a specialized platelet-specific

Received on: November 10, 2014; final version accepted on: March 19, 2015.
From the Faculty of Health Sciences, Curtin University, Perth, Western Australia, Australia (P.M., M.C.B); Western Australian Centre for Thrombosis
and Haemostasis, Murdoch University, Perth, Western Australia, Australia (R.I.B.); and Australian Centre for Blood Diseases, Department of Clinical
Haematology, Monash University, Melbourne, Victoria, Australia (R.K.A.).
Correspondence to Michael C. Berndt, PhD, Curtin University, PVC Health Sciences Suite, Bldg 400, Level 4, Room 408, Kent St, Bentley, Western
Australia 6102, Australia. E-mail michael.berndt@curtin.edu.au
© 2015 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol is available at http://atvb.ahajournals.org DOI: 10.1161/ATVBAHA.114.303413

1327
1328   Arterioscler Thromb Vasc Biol   June 2015

phosphatidylserine expression that accelerate coagulation


Nonstandard Abbreviations and Acronyms
leading to activation of factor Xa and thrombin. GPIbα binds
GP glycoprotein coagulation factors, XII, XI, high-molecular weight kininogen
ITAM immunoreceptor tyrosine–based activation motif (of the intrinsic coagulation pathway), and thrombin, local-
PCI percutaneous coronary intervention izing coagulation at the platelet surface, whereas increased
ROS reactive oxygen species phosphatidylserine facilitates formation of procoagulant com-
TxA2 thromboxane A2 plexes of factor Xa to increase thrombin generation. Active
vWF von Willebrand factor thrombin bound to GPIbα activates platelets and facilitates
stimulation of the G-protein–coupled 7-transmembrane recep-
tor, protease-activated receptor-1; thrombin also activates
adhesion-signaling system, made up of glycoprotein (GP)
human platelets via protease-activated receptor-4.25,26 In acti-
Ib-IX-V that binds von Willebrand factor (vWF; via the GPIbα
vated platelets, secretion of the agonist, ADP, and activation
subunit)8 and collagen (via GPV)9 and GPVI that binds colla-
of the cyclooxygenase pathway leading to synthesis and secre-
gen and the collagen-associated matrix protein, laminin.10–14
tion of thromboxane A2 (TxA2) reinforce platelet activation
Thus, on exposure of collagenous subendothelial matrix in
by autocrine stimulation of G-protein–coupled receptors for
the vasculature, platelet GPIb-IX-V/GPVI can rapidly induce
ADP (P2Y1 and P2Y12)27 or TxA2, respectively (Figure 1).
platelet adhesion and activation. Furthermore, in human
In this way, activation via primary platelet receptors, GPIb-
platelets, these receptor–ligand systems are highly evolved to
IX-V/GPVI, or secondary receptors for ADP, TxA2 or, throm-
mediate thrombus formation as the shear rate increases to high
bin leads to activation of the platelet integrin, αIIbβ3 (GPIIb/
physiological or pathological,15 as found in occluded coronary
IIIa) that binds fibrinogen or vWF and mediates platelet aggre-
arteries with atherosclerotic plaque or where normal blood
gation. This receptor is expressed in a low-affinity form that
flows are disrupted, for example, in ventricular assist devices.
GPIbα, disulfide-linked to GPIbβ, GPIX, and GPV are all does not bind ligand, but after intracellular signaling, it forms
members of the transmembrane leucine-rich repeat family.8 a high-affinity ligand-binding site, enabling platelet aggrega-
Analogous leucine-rich repeat proteins are involved in innate tion and outside-in-signaling to reinforce platelet activation.28
immunity in premammalian species.16 GPVI is a member of In addition to αIIbβ3, other platelet integrins, α2β1, α5β1, and
the immunoreceptor family, with 2 extracellular immuno- α6β1 that bind collagen, fibronectin, or laminin, respectively,
globulin domains, and forms a noncovalent complex with also become functional on activated platelets and promote firm
the Fc receptor γ-chain required for GPVI surface expres- platelet adhesion to collagenous subendothelial matrix.29,30
This amplification of platelet activation through release of sec-
Downloaded from http://ahajournals.org by on February 11, 2024

sion and ligand-induced signaling on human platelets.10–14


Signaling immediately downstream of GPIb-IX-V/GPVI ondary agonists is critical in stabilizing the thrombus and aug-
involves activation of Src and Syk kinase–dependent tyrosine menting the thrombus volume. In cases of unrestrained growth,
phosphorylation pathways and Src/Syk-dependent and Src/ which can result in occlusion and ischemia, inhibition of sec-
Syk-independent generation of intracellular reactive oxygen ondary messengers, such as TxA2, by aspirin or platelet adhe-
species (ROS).17–20 The cytoplasmic domains of GPIb-IX-V sion integrins assists in controlling thrombus size on the vessel
and GPVI directly bind to signaling or adaptor proteins, includ- wall. Figure 2 demonstrates how targeting of specific steps,
ing phosphatidylinositol 3-kinase (via the p85 subunit)21,22 and using αIIbβ3 as an example, modulates thrombus formation.
constitutively phosphorylated Lyn (via a proline-rich sequence In addition to activation pathways, there are at least 2
of GPVI),23 respectively. In the case of GPVI, ligand-induced inhibitory pathways that can control platelet reactivity. First,
receptor cross-linking enables active Lyn to phosphorylate inhibitory receptors bearing an immunoreceptor tyrosine–
Syk associated with the immunoreceptor tyrosine–based acti- based inhibitory motif domain, such as platelet/endothelial
vation motif (ITAM) in the cytoplasmic domain of Fc receptor cell adhesion molecule 1 or carcinoembryonic antigen-related
γ-chain, initiating canonical Syk–dependent signaling path- cell adhesion molecule-1/2,31 can attenuate ITAM-mediated
ways. Other ITAM-bearing receptors in platelets include the signaling by GPVI/Fc receptor γ-chain or other ITAM-
low-affinity IgG receptor, FcγRIIa, and the C-type lectin fam- bearing receptors, via recruitment of phosphatases or other
ily receptor-2.24 Positively charged conserved sequences of mechanisms, although how immunoreceptor tyrosine–based
both GPIbβ and GPVI also directly bind to tumor necrosis fac- inhibitory motif–containing receptors on circulating human
tor receptor–associated factor 4,18 a receptor-localizing subunit platelets are activated under physiological or pathological con-
of the nicotinamide adenine dinucleotide phosphate-oxidase ditions is uncertain. Second, elevation of intracellular cAMP/
oxidase complex (Nox-1/2), which generates intracellular cGMP inhibits platelet activation by activating cAMP/cGMP-
ROS in platelets.17,19,20 Rapid elevation of intracellular ROS dependent protein kinases, protein kinase A/protein kinase
promotes platelet activation and supports platelet adhesion to G,32 although the role of cGMP effects on platelet function
collagen, by a mechanism that may involve ROS-dependent remains controversial with several studies demonstrating that
inactivation of protein tyrosine phosphatases that inhibit sig- cGMP is stimulatory.33,34 Protein kinase A substrates include
naling involving Lyn and associated phosphorylated proteins. cytoplasmic serine residues within conserved sequences of
Engagement of GPIb-IX-V/GPVI by vWF/collagen or GPIbβ, regulating the interaction with intracellular bind-
other ligands leads to rapid platelet activation, morphological ing partners and attenuating vWF-dependent platelet activa-
changes, in platelet shape and rearrangement of the cytoskel- tion.35 Inhibitory prostacyclin (also known as prostaglandin)
etal actin filaments, and changes in platelet plasma membrane leads to activation of adenylyl or guanylyl cyclases to increase
Metharom et al   Current State of Antiplatelet Therapy   1329

Table 1. Antiplatelet Therapy: Current Drugs98–100


Route of
Type/Name (Drug Name) Marketing Status Administration Indication References
Cyclooxygenase inhibitors
 ASA and aspirin Over-the-counter Oral To reduce the risk of nonfatal 41, 101–103
thrombotic events
 Triflusal Available in some Oral Prevention of cardiovascular 104–106
European countries events and acute treatment of
infarction
ADP P2Y12 inhibitor
 Clopidogrel (Plavix) Prescription Oral For ACS NSTEMI, STEMI, 107–109
and reduction and prevent of
ischemic events
 Prasugrel (Effient) Prescription Oral Use in combination with aspirin 107, 109–111
in patients with ACS undergoing
PCI, also NSTEMI and STEMI
patients undergoing PCI
 Ticagrelor (Brilinta) Prescription Oral For prevention of thrombotic 111–113
events. Recommend in addition
to ASA for ACS patients
αIIbβ3 (GPIIb/IIIa) inhibitors
 Abciximab (Reopro) Prescription Injection As an adjunct to PCI for 42, 114–116
prevention of ischemic
complications
 Eptifibatide (Integrilin) Prescription Injection Treatment of MI and ACS 115, 116
 Tirofiban (Aggrastat) Prescription Injection For reduction of thrombotic 42, 115–117
cardiovascular events in patients
with NSTEMI ACS
Prostacyclin and analogues
Downloaded from http://ahajournals.org by on February 11, 2024

 PGI2 (Epoprostenol) Prescription Injection Long-term intravenous 118, 119


treatment of primary pulmonary
hypertension
 Iloprost (Ventavis) Prescription Oral inhalation Treatment of PAH 118, 119
 Treprostinil (Remodulin) Prescription Oral Treatment of PAH 119–121
Direct thrombin inhibitors
 Bivalirudin (Angiomax) Prescription IV For use in prevention of 122, 123
thrombosis in patients with ACS
undergoing PCI
 Dabigatran (Pradaxa) Prescription Oral Prevention of thrombosis post 124, 125
joint replacement and atrial
fibrillation
Phosphodiesterase inhibitor
 Dipyridamole (Persantine) Prescription Oral Use in adjunct to coumarin 126, 127
anticoagulants in the prevention
of thrombotic complications post
cardiac valve replacement
 Cilostazol (Pletal) Prescription Oral Reduction of symptoms of 126, 128, 129
intermittent claudication
ACS indicates acute coronary syndrome; ASA, acetylsalicylic acid; MI, myocardial infarction; NSTEMI, non–ST-segment–
elevation myocardial infarction; PAH, pulmonary arterial hypertension; PCI, percutaneous coronary intervention; PGI2,
prostaglandin; and STEMI, ST-segment–elevation myocardial infarction.

intracellular levels of cAMP/cGMP. Stimulation of the ADP promising new antithrombotic strategies and may have advan-
receptor, P2Y1, leads to inhibition of adenylyl cyclase and tages compared with current modes of antiplatelet therapy.36
reduced production of cAMP to facilitate platelet activa-
tion.27,32 Alternatively, phosphodiesterases can directly reduce Platelet Targets of Current Antithrombotics
the levels of cAMP to increase platelet reactivity, and phos- Current antiplatelet agents used clinically (Table 1) target sev-
phodiesterase inhibitors can thereby be used to inhibit platelet eral key components of platelet activation and aggregation
activation.36 These types of inhibitors are among a range of (Figure 1). The 3 major targets are cyclooxygenase of the TxA2
1330   Arterioscler Thromb Vasc Biol   June 2015

Table 2. Antiplatelet Therapy: Drugs in Clinical Trials and Clinical Development


Route of
Type/Name Developmental Status Administration Indication References
Prostacyclin analogue
 Beraprost Ongoing clinical trials Oral Treatment of PAH and diabetic 130, 131
nephropathy
GPIbα antagonist
 Anfibatide Phase I completed; phase II IV For treatment of thrombotic 71–73
listed but not yet active events MI
vWF-targeting (inhibit GPIbα binding)
 Anti-vWF aptamers (ARC1779 … IV For treatment of TTP, VWD 2b 132–134
and ARC15105)
 Anti-vWF nanobody (ALX- Phase I, phase II TITAN trial IV For treatment of TTP 132, 135, 136
0081, caplacizumab)
GPVI bivalent soluble form
 Revacept (PR-15), humanized Phase I completed; phase II IV For treatment of carotid artery 74, 75
Fc fusion of GPVI ectodomain recruiting stenosis, TIAs, and stroke
Thromboxane receptor antagonists
 Terutroban Clinical trials indicate no better Oral For secondary prevention 137–139
outcome than aspirin of thrombotic events in
cardiovascular disease
 Ifetroban Phase II Injection Treatment of hepatorenal 140
syndrome
PAR-1 inhibitors
 Vorapaxar (SCH530348) Recommended for approval Oral Reduction of atherothrombotic 141–143
by FDA’s Cardiovascular events in patients with history
and Renal Drugs Advisory of MI
Committee
Downloaded from http://ahajournals.org by on February 11, 2024

 Atopaxar (E5555) Phase II Oral CAD 141, 144, 145


ADP P2Y12 inhibitor
 Cangrelor FDA denied approval and IV For patients with ACS undergoing 146–148
requested further clinical data PCI; reduces MI and stent
thrombosis events compared
with other P2Y12 inhibitor drugs;
associates with bleeding risks
and lacks mortality benefit
ACS indicates acute coronary syndrome; CAD, coronary artery disease; FDA, Food and Drug Administration; IV, intravenous;
MI, myocardial infarction; PAH, pulmonary arterial hypertension; PAR-1, protease-activated receptor-1; PCI, percutaneous coronary
intervention; TIAs, transient ischemic attacks; TTP, thrombotic thrombocytopenic purpura; vWF, von Willebrand factor; and VWD 2b,
type 2b von Willebrand’s disease.

synthesis pathway (aspirin), the G-protein–coupled ADP recep- activation via TxA2 and ADP, respectively. The αIIbβ3 inhibitors
tor P2Y12 (clopidogrel, prasugrel, or ticagrelor), and inhibitors block thrombus formation to all agonists because αIIbβ3 is essen-
of αIIbβ3 (abciximab, eptifibatide, and tirofiban). Other targets tial for platelet aggregation. For these most widely used drugs, it
are the prostaglandin receptor that can be blocked by prosta- is interesting to consider the specific targets—cyclooxygenase,
cyclin mimetics and phosphodiesterase inhibitors that elevate P2Y12, and αIIbβ3—and the role of these targeted pathways in
intracellular cAMP/cGMP (Figure 1). Anticoagulants that are thrombus formation, in terms of antithrombotic protection versus
direct thrombin inhibitors3 can also suppress platelet activation bleeding risk, and resistance to dual antiplatelet therapy.
by inhibiting thrombin-dependent platelet activation via GPIbα
and protease-activated receptor-1/4. Notably, platelet adhesion Bleeding Risk Associated With Current
and aggregation induced at high shear stress are dependent on Antiplatelet Therapeutics
ADP and αIIbβ3, and they are inhibited by elevated platelet Although antiplatelet drugs are effective in reducing vascular
cAMP/cGMP.15,37 In contrast, cyclooxygenase inhibition by deaths and nonfatal events by ≤50% in high-risk cardiovascu-
aspirin has minimal effect.15,38 This may explain the relative lar patients, the main safety concern is excess hemorrhage.39
lack of efficacy of aspirin as treatment for arterial thrombosis Primary prevention of cardiovascular events with aspirin is
and the common use of aspirin in combination with other drugs. associated with a significant increased risk of extracranial
Dual antiplatelet therapy typically refers to a combination bleeding (relative risk, 1.55; 95% confidence interval, ­1.3–1.8;
of aspirin and a P2Y12 inhibitor, targeting secondary platelet P<0.001), which often negates the uncertain potential benefit
Metharom et al   Current State of Antiplatelet Therapy   1331

Figure 1. Platelet targets of antiplatelet therapy. Schematic showing platelet surface receptors and signaling pathways leading to plate-
let activation and activation of the integrin, αIIbβ3, which binds fibrinogen or Von Willebrand factor and mediates platelet aggregation.
­Targets of currently available (shaded boxes) and novel drugs in development or in clinical trial (white boxes) are indicated. COX indicates
Downloaded from http://ahajournals.org by on February 11, 2024

cyclooxygenase; GP, glycoprotein; and TxA2, thromboxane A2.

in otherwise healthy people.39 In patients with acute coronary therapy is associated with an annual incidence of 2% to 4%
syndromes, stable coronary artery disease, ischemic stroke or gastrointestinal bleeding and 0.5% rate of intracranial hemor-
peripheral vascular disease, single-agent aspirin, or clopidogrel rhage.40 A 2014 analysis of 8 trials involving 41 483 individuals

Figure 2. Antiplatelet therapy modulates thrombus formation on the vessel wall. The simplified flow chart indicates platelet activation
steps exploited in drug targeting to reduce platelet activation, aggregation, and ultimately thrombus formation. The figure focuses on
antiαIIbβ3 (GPIIb/IIIa) inhibition as an example illustrating reduction in thrombus volume as the result of blocking platelet aggregation
mediated by αIIbβ3 receptor. COX indicates cyclooxygenase; GP, glycoprotein; and TxA2, thromboxane A2.
1332   Arterioscler Thromb Vasc Biol   June 2015

showed daily aspirin treatment (160 or 300 mg) within 48 Aspirin and Clopidogrel Resistance
hours post ischemic stroke reduced the risk of early recurrent It is accepted that there is a wide interindividual variation
stroke, but it was connected with a small excess of intracra- in response to aspirin and clopidogrel, which can be defined
nial bleeding.41 With individuals undergoing percutaneous either through the patient experiencing recurrent platelet-
coronary revascularization, administration of αIIbβ3 inhibitors mediated clinical events (recurrent in stent thrombosis, acute
reduced the risk of death or myocardial infarction, however, myocardial infarction, stroke, and death) or pharmacody-
with an increased risk of severe bleeding. These meta-analyses namically with various ex vivo platelet inhibition measures
suggested the potential benefits of antiplatelet agents far out- (agonist-induced platelet aggregation, serum and urinary
weighed the risk of bleeding complications. On the other hand, thromboxane measurement, electric impedance aggregome-
a Cochrane review of αIIbβ3 inhibitors administered early after try, shear-induced platelet functional analysis [PFA100], flow
an ischemic stroke showed a link with a significant risk of intra- cytometry, and point of care agonist assessment [VerifyNow
cranial hemorrhage with no reduction in mortality.42 and VASP-P assay]).50 Controversy still exists as to whether
The risk of bleeding has been shown to be dose related any ex vivo test can simulate in vivo platelet response and
for all antiplatelet drugs, including aspirin (100 versus >325 there is uncertainty as to which test to preferentially perform.
mg daily),40 clopidogrel (300- versus 600-mg loading),43 and Observational studies suggest that at standard doses of aspi-
prasugrel (5 versus 10 mg daily).44,45 Combined antiplatelet rin (100 mg) or clopidogrel (75 mg), over a third of patients
therapy, with dual targeted inhibition of the platelet cyclooxy- have predefined ex vivo resistance.51 Reasons for antiplatelet
enase-1 pathway (aspirin) and the P2Y12 unit of the platelet resistance include poor compliance, insufficient dose, slow
ADP receptor (clopidogrel, prasugrel, and ticagrelor), is an pharmacodynamic effect, related gene polymorphisms that
effective strategy for reducing cardiovascular events, but it
effect antiplatelet drug metabolism (eg, cytochrome P450
is associated with an increased relative risk of major bleed-
2C19 loss of function allele for clopidogrel biotransformation
ing by ≤40%.46 Rates of bleeding increase with the prolonged
to active metabolite and ABCB1 polymorphisms, particularly
duration of dual antiplatelet therapy, percutaneous coronary
3435C>T, affecting efflux of clopidogrel via P-glycoprotein),40
intervention (PCI), or urgent coronary artery bypass surgery.46
and PEAR1 variants in patients exposed to aspirin,52 increased
More rapid and consistent P2Y12 platelet ADP receptor
baseline platelet reactivity such as that found in diabetes mel-
blockade by prasugrel and ticagrelor when compared with
litus and smoking, and increased platelet turnover.40 Whatever
clopidogrel generally produces better outcomes, particularly
the reason, predefined high on-treatment platelet reactivity
in patients with acute coronary syndromes treated with PCI.46
is consistently associated with 2- to 4-fold increased risk of
However, this benefit comes at the cost of an increase in major
acute myocardial infarction, stent thrombosis, and stroke.50
Downloaded from http://ahajournals.org by on February 11, 2024

life-threatening and fatal bleeding with prasugrel when com-


Clopidogrel leads to a variable platelet inhibition and has a
pared with clopidogrel (2.4% versus 1.8%; hazard ratio, 1.32;
P=0.03) as shown in the TRITON-TIMI 38 trial (TRial to delayed onset of action, mainly because of the need for bio-
assess Improvement in Therapeutic Outcomes by optimizing transformation in the liver via CYP450 isoenzymes (particu-
platelet InhibitioN with prasugrel Thrombolysis In Myocardial larly the CYP2C19 isoenzyme) from the inactive parent drug
Infarction 38).45 Patients with risk factors for bleeding were to the active metabolite. Prasugrel is also a prodrug requiring
elderly (>75 years) and with low body weight (<60 kg). Dose CYP450-mediated metabolic activation, but it is less affected
reduction of prasugrel to 5 mg may improve the benefit/risk by the CYP2C19 polymorphism and has a faster onset of
ratio in this susceptible group as shown in a subsequent study action. Ticagrelor is an orally active drug not requiring bio-
(TRILOGY ACS) where there was no difference in hemor- transformation and binds rapidly and reversibly to the P2Y12
rhage in a prespecified analysis of elderly patients (>75 years).44 receptor, making it an attractive direct P2Y12 inhibitor.46
Overall bleeding rates were similar with ticagrelor and clopido- Strategies for overcoming antiplatelet drug resistance
grel in the PLATelet inhibition and patient Outcomes (PLATO include increasing initial loading doses (eg, aspirin: 100–300
trial); however, when bleeding from coronary artery bypass sur- mg; clopidogrel: 75–600 mg; prasugrel: 10–60 mg; ticagre-
gery was removed from analysis, ticagrelor had a higher risk of lor: 90–180 mg)43,45,47 and personalized dose adjustment (eg,
major nonprocedure-related bleeding (4.5% versus 3.8%; haz- increase the maintenance dose of clopidogrel to 150 mg
ard ratio, 1.19; P=0.03).47 Comparison between up-front and daily53,54 or drug switching from clopidogrel to prasugrel53,55,56
delayed (after PCI) administration of either prasugrel or anti- or ticagrelor) that may include adjustment based on regular
platelet αIIbβ3 receptor antagonists (tirofiban and eptifibatide) testing platelet function monitoring.50
showed little difference in efficacy, but early administration The lack of effect of individualized antiplatelet therapy by
significantly increased bleeding and red-cell transfusion rates.48 platelet function testing in recent large randomized studies
Definitions of bleeding are poorly standardized and in PCI and medically managed patients with coronary artery
vary between different studies making direct comparison disease is disappointing.54–57 Studies that have not shown a
between various combinations of antiplatelet drugs more change in outcomes include the TRILOGY-ACS (TaRgeted
difficult. Dual oral antiplatelet therapy is now standard platelet Inhibition to cLarify the Optimal strateGy to medi-
practice for treatment of acute coronary syndromes with and callY manage Acute Coronary Syndromes) trial (n=2564;
without PCI; however, the individual assessment of bleed- prasugrel versus clopidogrel),56 GRAVITAS (Gauging
ing risk over time and risk of later thrombotic complica- Responsiveness With a VerifyNow P2Y12 Assay: Impact on
tions often determines the duration of combined antiplatelet Thrombosis and Safety) increased dose (n=2214; clopidogrel
therapy.46,49 150 mg daily),54 and ARCTIC (Double Randomization of a
Metharom et al   Current State of Antiplatelet Therapy   1333

Monitoring Adjusted Antiplatelet Treatment Versus a Common times are generally minimal. Overall, these findings support
Antiplatelet Treatment for DES Implantation, and Interruption further development of antiplatelet therapies targeting either
Versus Continuation of Double Antiplatelet Therapy) standard GPIbα–vWF, GPVI–collagen, or both.
versus monitoring antiplatelet drug/dose adjustment (n=2440 Currently used antiplatelet drugs are based on natural prod-
patients).55,57 Although ≈20% of patients did not achieve opti- ucts (aspirin), toxins (snake venom–based inhibitory peptides
mal reduction in platelet reactivity, the event rates were low and that blocked ligand binding to αIIbβ3), inhibitory antibodies
the studies were underpowered.50 However, a recent meta-anal- (derived from anti–αIIbβ3 monoclonal antibodies), or small
ysis of 9 pooled studies consisting of 12 048 subjects showed molecules (irreversible P2Y12 inhibitors developed as orally
significant benefit for tailored versus standard antiplatelet ther- available prodrugs). Inhibitory toxins and antibodies, ligand
apy with a risk reduction of ≈60% without an increase in bleed- mimetics, as well as nucleotide-based aptamers and soluble
ing.51 Smaller studies that demonstrated a consistent reduction recombinant forms of receptor, have already been substan-
in platelet reactivity in all patients below the threshold of high tially developed as means of modulating GPIbα–vWF, GPVI–
on-treatment platelet reactivity also showed significant clinical collagen, or other platelet targets, including the prostaglandin
benefit of reduction in cardiovascular events.50 receptor, TxA2 receptor, and protease-activated receptor-1
Observational studies have suggested that CYP2C19 poly- (Table 2; Figure 1). The GPIbα–vWF axis has been targeted
morphisms associated with reduced activation of clopidogrel using aptamers or antibody-based agents selective for the
increase the risk of thrombotic events by 2-fold in patients GPIbα-binding A1 domain of vWF that prevent vWF-binding
treated with clopidogrel.58 However, analysis of 3 large ran- platelet GPIbα. Snake venom proteins have been long known
domized control studies showed no effect of the CYP2C19 or to target human platelet GPIbα by metalloproteinase-medi-
ABCB1 genotype on clinical outcomes in patients on clopido- ated proteolysis of the vWF/ligand-binding domain (mocar-
grel.59–62 In an ongoing prospective post-PCI study assessing hagin, natural killer protease) or by binding of C-type lectin
CYP2C19 genotype, antiplatelet therapy was changed from family proteins to the leucine-rich repeat domain of GPIbα
clopidogrel to ticagrelor or prasugrel in those with a loss of (echicetin and alboaggregin-B).70 Anfibatide is a C-type lec-
function polymorphism.63 These data will clarify the role of tin-like protein that binds GPIbα and inhibits vWF-binding
CYP2C19 genotype in selecting the type of antiplatelet therapy GPIbα–dependent platelet adhesion and activation, impor-
and will either confirm or refute the smaller studies suggesting tantly with potent activity at elevated shear rates.71–73
an improvement in cardiovascular outcome with this strategy. The GPVI–collagen axis has been targeted using a bivalent
Recent clinical guidelines suggest that platelet function test- recombinant soluble form of the GPVI ectodomain expressed
ing may have a limited role in selecting the choice of P2Y12 as a humanized Fc fusion protein.11,74,75 The ≈55-kDa soluble
Downloaded from http://ahajournals.org by on February 11, 2024

inhibitors in high-risk patients treated with PCI (class IIb) ectodomain of GPVI is shed from the human platelet surface by
although routine testing is not recommended (class III).50 By the membrane-expressed sheddase, ADAM-10.76,77 This process
better personalizing the drug therapy, there is an opportunity is tightly regulated, with essentially all the GPVI on healthy cir-
to better define high and low platelet reactivity to address the culating platelets in an intact form, with shedding rapidly induced
balance for the individual between bleeding and thrombosis in by ligand binding to GPVI, by platelet activation, by platelet
the context of antiplatelet therapy. exposure to high shear stress,78 by coagulation/factor Xa,79 or by
other triggers, releasing plasma soluble ectodomain of GPVI,
More Recent Targets for Antiplatelet Therapy which is quantifiable by immunoassay as a platelet-specific
Interestingly, there are currently no inhibitors in routine clini- marker.80 Native soluble ectodomain of GPVI shed from plate-
cal use targeting the primary platelet receptors, GPIbα (or lets is monovalent and binds collagen weakly compared with the
GPIb-IX-V), GPVI, or their ligands (vWF/collagen), interac- bivalent form, which shows promise as a novel antithrombotic.11
tions that initiate platelet adhesion and activation and which Indeed, these new agents against GPIbα–vWF, GPVI–
become increasingly important as the shear rate increases. collagen, and other platelet targets have all shown merit as
The limited distribution of these receptors to megakaryocytes/ potential antiplatelet drugs experimentally or in clinical settings
platelets should also offer advantages compared with targets associated with pathological thrombosis (Table 2). The type and
with expression profiles that include other cells. On one hand, properties of these agents, in terms of oral availability, revers-
the rare congenital or acquired defects or deficiency of GPIb- ibility, bleeding risk, and antithrombotic or off-target effects,
IX-V (Bernard-Soulier syndrome) or GPVI results in vari- will prove critical to their clinical application in different patient
able bleeding risk, from relatively mild to more severe, which groups and will require longer term trials to establish viability.
could argue against directly targeting these receptors.64–67 On
the other hand, these cases often involve thrombocytopenia Future Targets for Antiplatelet Therapy
or concurrent hemostatic disorders that complicate attributing On the basis of recent research discoveries, there is far wider
bleeding to receptor dysfunction alone. Experimental stud- scope for therapeutic regulation of platelet targets, including
ies suggest that GPIbα has a far greater contribution to stable αIIbβ3,28,81 integrin α6β1,82 GPIb-IX-V, and GPVI adhesive
arterial thrombosis than vWF,67 whereas the GPVI-binding function and signaling. In addition to vWF- and GPIbα-
sites of collagen are not normally exposed to circulating plate- targeting inhibitors in development (Table 2), alternative strat-
lets. GPVI also regulates thrombus formation in experimen- egies could have advantages, for example, small molecules
tal models of arterial thrombosis or stroke,68,69 whereas the with increased scope for bioavailability. In this regard, alloste-
hemostatic consequences of GPVI deficiency on bleeding ric inhibitors can overcome the inherent difficulties of a small
1334   Arterioscler Thromb Vasc Biol   June 2015

molecule inhibiting a protein–protein interaction in which a by Nox-1/2 inhibitors19,20 raises the possibility that such inhibi-
large multisite interface occurs as for the interaction between tors could be antithrombotic. An inhibitor targeting NOX1
the ligand-binding domains of GPIbα and vWF. As proof of and NOX4 (GKT137831) is in clinical trial as a treatment for
this concept, a short cyclic peptide, CTERMALHNLC, which diabetic nephropathy,93 unrelated to any possible antiplatelet
binds GPIbα, potently inhibits vWF binding not through com- effects. Evidence for the value of antioxidants in treating car-
petitive inhibition but by preventing formation of a ligand- diovascular disease seems limited and requires further atten-
receptive structural conformation.83,84 tion. Although direct therapeutic targeting of Nox-1/294 is
An alternative strategy targeting the ADP-P2Y1/P2Y12 acti- viable as a general inhibitor of ROS and inflammation, it is
vation pathway is the use of a soluble form of CD39 (solCD39), not selective for platelet ROS pathways.95 However, an attrac-
an ectonucleoside triphosphate diphosphohydrolase.85 This tive possibility exists for these ROS inhibitors, as well as
approach uses the ability of CD39 to hydrolyze ADP, dissimi- nonplatelet-specific targets mentioned above, in drug-eluting
lar to the current ADP-P2Y12 drugs clopidogrel, prasugrel, stents where their actions will likely be more localized. Other
and ticagrelor that block ADP-mediated platelet activation by intracellular binding partners for G ­ PIb-IX-V in addition to the
binding to the receptor directly. An administration of a fusion p85 subunit of phosphatidylinositol 3-kinase21,22 include the
protein consisting of solCD39 and a single-chain Fv fragment regulatory protein, 14-3-3ζ, and the phosphorylation depen-
of an antibody specific for the αIIbβ3 active form showed dence, functional role, and potential for future targeting of this
promising antithrombotic effects with no prolonged bleeding interaction have been previously reviewed in detail.35
in an experimental mouse model. The drug is designed to allow Finally, antiplatelet strategies could focus on the expression
a layer of platelets to firmly adhere to the wound and αIIbβ3 to and function of platelet-specific GPVI. In addition to block-
become activated before exerting its function, thus minimizing ing ligand binding or inhibiting signaling, either the require-
the amplification phase of platelets in developing thrombus. ment for GPVI to dimerize to elicit signaling (by cross-linking
Innovative new approaches for targeting αIIbβ3 signaling FcRγ ITAM) or the controlled ectodomain shedding mediated
include the recent report of short myristoylated peptides based by ADAM10 could also potentially be targeted.10–12,96 Further
on cytoplasmic sequences of β3 that selectively regulate the understanding of how shear stress and membrane proper-
dynamic interaction between the receptor and intracellular ties control dimer formation and ADAM10 accessibility and
binding partners, including the G protein Gα13.81 Notably, cleavage could enable alternative therapeutic control of plate-
it seems feasible to dissect out precise signaling functions of let reactivity toward collagen.
αIIbβ3 involving Gα13 or talin associations, which control
platelet activation, adhesion, and aggregation/clot contrac- Conclusions
Downloaded from http://ahajournals.org by on February 11, 2024

tion, and to inhibit thrombus formation in mice comparable Current antiplatelet therapy has no doubt prevented incalcu-
with current αIIbβ3 inhibitors, yet without adverse bleeding.81 lable death and disability in people at risk of athero/throm-
Platelet integrin α6β1 also represents a novel antiplatelet tar- botic/cardiovascular disease by preventing thrombotic events
get. Recently, using platelet-specific knockout of integrin α6, and quelling the proinflammatory role of activated platelets.
the main vascular laminin receptor was indicated to play an Nevertheless, the lack of efficacy/resistance, reliable clini-
important part in platelet adhesion, activation, and in arterial cal guidelines, and increased bleeding risk in individuals
thrombosis without significant effect on bleeding risk.82 Future remains an ongoing problem. New experimental evidence
research will need to address possible detrimental effects of and increased understanding of platelet function and regula-
α6β1 blocking as the protein expression is not only limited tion are revealing new antiplatelet approaches to potentially
to platelets but it is also found on various cell types including improve clinical outcomes and monitoring.
glial cells, neuronal stem cells, and endothelial cells.86–88 The readers may also find additional information on anti-
Signaling protein targets downstream of GPIb-IX-V/GPVI platelet and antithrombosis therapy from other reviews
may also be worthwhile considering. First, Syk inhibitors have in the ATVB series New Targets of Antiplatelet Drug
been tested clinically as selective, reversible anti-inflammatory/ Development.14,28,36,97
cancer therapeutics, without inhibitory activity toward other
tyrosine kinases, such as Src, Lyn, or Btk.89 Syk inhibition Acknowledgments
also blocks GPVI-dependent activation of human platelets.14 We thank all our colleagues and collaborators for helpful discussions.
Second, Btk mediates signaling downstream of GPIb-IX-V
in platelets,90 and an irreversible Btk inhibitor (ibrutinib) is Sources of Funding
approved for treatment of the lymphoproliferative diseases, This work was supported by Curtin University, Murdoch University,
chronic lymphocytic leukemia, and mantle cell lymphoma. Monash University, and the National Health and Medical Research
Therapeutic concentrations of ibrutinib inhibit collagen/GPVI- Council of Australia.
dependent platelet aggregation,91 and although it is associated
with clinical bleeding in patients with chronic lymphocytic leu- Disclosures
kemia,91 this could be related to vulnerability resulting from None.
decreased GPVI expression in lymphoproliferative disease.92
Third, recent evidence that platelet Nox-1/2–dependent ROS References
production is important for GPIb-IX-V/GPVI-dependent plate- 1. Bhatt DL, Topol EJ. Scientific and therapeutic advances in antiplatelet
let function, including adhesion to collagen, and is inhibited therapy. Nat Rev Drug Discov. 2003;2:15–28. doi: 10.1038/nrd985.
Metharom et al   Current State of Antiplatelet Therapy   1335

2. Ungerer M, Münch G. Novel antiplatelet drugs in clinical development. 23. Schmaier AA, Zou Z, Kazlauskas A, Emert-Sedlak L, Fong KP, Neeves
Thromb Haemost. 2013;110:868–875. doi: 10.1160/TH13-02-0084. KB, Maloney SF, Diamond SL, Kunapuli SP, Ware J, Brass LF, Smithgall
3. Bhatt DL, Hulot JS, Moliterno DJ, Harrington RA. Antiplatelet and TE, Saksela K, Kahn ML. Molecular priming of Lyn by GPVI enables an
anticoagulation therapy for acute coronary syndromes. Circ Res. immune receptor to adopt a hemostatic role. Proc Natl Acad Sci U S A.
2014;114:1929–1943. doi: 10.1161/CIRCRESAHA.114.302737. 2009;106:21167–21172. doi: 10.1073/pnas.0906436106.
4. Lussana F, Squizzato A, Permunian ET, Cattaneo M. A systematic review 24. Ozaki Y, Suzuki-Inoue K, Inoue O. Platelet receptors activated via
on the effect of aspirin in the prevention of post-operative arterial throm- mulitmerization: glycoprotein VI, GPIb-IX-V, and CLEC-2. J Thromb
bosis in patients undergoing total hip and total knee arthroplasty. Thromb Haemost. 2013;11(suppl 1):330–339. doi: 10.1111/jth.12235.
Res. 2014;134:599–603. doi: 10.1016/j.thromres.2014.06.027. 25. De Candia E, Hall SW, Rutella S, Landolfi R, Andrews RK, De Cristofaro
5. Nairooz R, Sardar P, Amin H, Swaminathan RV, Kim LK, Chatterjee S, R. Binding of thrombin to glycoprotein Ib accelerates the hydrolysis of
Feldman DN. Meta-analysis of randomized clinical trials comparing bivali- Par-1 on intact platelets. J Biol Chem. 2001;276:4692–4698. doi: 10.1074/
rudin versus heparin plus glycoprotein IIb/IIIa inhibitors in patients under- jbc.M008160200.
going percutaneous coronary intervention and in patients with ST-segment 26. Wallace EL, Smyth SS. Targeting platelet thrombin receptor signaling
elevation myocardial infarction. Am J Cardiol. 2014;114:250–259. doi: to prevent thrombosis. Pharmaceuticals (Basel). 2013;6:915–928. doi:
10.1016/j.amjcard.2014.04.033. 10.3390/ph6080915.
6. Carreras ET, Mega JL. Dual antiplatelet therapy for heart disease. Circulation. 27. Gachet C. P2 receptors, platelet function and pharmacological implica-
2014;129:e506–e508. doi: 10.1161/CIRCULATIONAHA.113.004305. tions. Thromb Haemost. 2008;99:466–472. doi: 10.1160/TH07-11-0673.
7. Berndt MC, Metharom P, Andrews RK. Primary haemostasis: newer 28. Estevez B, Shen B, Du X. Targeting integrin and integrin signaling in
insights. Haemophilia. 2014;20(suppl 4):15–22. doi: 10.1111/hae.12427. treating thrombosis. Arterioscler Thromb Vasc Biol. 2015;35:24–29. doi:
8. Andrews RK, Berndt MC. Chapter 10 - The gpib-ix-v complex. In: 10.1161/ATVBAHA.114.303411.
Michelson AD, ed. Platelets. 3rd ed. San Diego: Academic Press; 29. Nuyttens BP, Thijs T, Deckmyn H, Broos K. Platelet adhesion to
2013:195–213. collagen. Thromb Res. 2011;127(suppl 2):S26–S29. doi: 10.1016/
9. Moog S, Mangin P, Lenain N, Strassel C, Ravanat C, Schuhler S, Freund S0049-3848(10)70151-1.
M, Santer M, Kahn M, Nieswandt B, Gachet C, Cazenave JP, Lanza F. 30. Deckmyn H, De Meyer S, Broos K, Vanhoorelbeke K. Inhibitors of the
Platelet glycoprotein V binds to collagen and participates in platelet adhe- interactions between collagen and its receptors on platelets. In: Gresele
sion and aggregation. Blood. 2001;98:1038–1046. P, Born GVR, Patrono C, Page CP, eds. Antiplatelet agents, Handbook of
10. Andrews RK, Arthur JF, Gardiner EE. Targeting GPVI as a novel anti- Experimental Pharmacology. Springer: Berlin Heidelberg; 2012:311–337.
thrombotic strategy. J Blood Med. 2014;5:59–68. doi: 10.2147/JBM. 31. Alshahrani MM, Yang E, Yip J, Ghanem SS, Abdallah SL, deAngelis AM,
S39220. O’Malley CJ, Moheimani F, Najjar SM, Jackson DE. CEACAM2 nega-
11. Gardiner EE, Andrews RK. Platelet receptor expression and shedding: gly- tively regulates hemi (ITAM-bearing) GPVI and CLEC-2 pathways and
thrombus growth in vitro and in vivo. Blood. 2014;124:2431–2441. doi:
coprotein Ib-IX-V and glycoprotein VI. Transfus Med Rev. 2014;28:56–
10.1182/blood-2014-04-569707.
60. doi: 10.1016/j.tmrv.2014.03.001.
32. Smolenski A. Novel roles of cAMP/cGMP-dependent signal-
12. Gawaz M, Vogel S, Pfannenberg C, Pichler B, Langer H, Bigalke B.
ing in platelets. J Thromb Haemost. 2012;10:167–176. doi:
Implications of glycoprotein VI for theranostics. Thromb Haemost.
10.1111/j.1538-7836.2011.04576.x.
2014;112:26–31. doi: 10.1160/TH13-09-0756.
33. Zhang G, Xiang B, Dong A, Skoda RC, Daugherty A, Smyth SS, Du X, Li
13. Zahid M, Mangin P, Loyau S, Hechler B, Billiald P, Gachet C, Jandrot-
Z. Biphasic roles for soluble guanylyl cyclase (sGC) in platelet activation.
Perrus M. The future of glycoprotein VI as an antithrombotic target.
Downloaded from http://ahajournals.org by on February 11, 2024

Blood. 2011;118:3670–3679. doi: 10.1182/blood-2011-03-341107.


J Thromb Haemost. 2012;10:2418–2427. doi: 10.1111/jth.12009.
34. Li Z, Xi X, Gu M, Feil R, Ye RD, Eigenthaler M, Hofmann F, Du X. A
14. Stegner D, Haining EJ, Nieswandt B. Targeting glycoprotein VI and
stimulatory role for cGMP-dependent protein kinase in platelet activation.
the immunoreceptor tyrosine-based activation motif signaling path-
Cell. 2003;112:77–86.
way. Arterioscler Thromb Vasc Biol. 2014;34:1615–1620. doi: 10.1161/
35. Andrews RK, Du X, Berndt MC. The 14-3-3zeta-GPIb-IX-V complex
ATVBAHA.114.303408.
as an antiplatelet target. Drug News Perspect. 2007;20:285–292. doi:
15. Kroll MH, Hellums JD, McIntire LV, Schafer AI, Moake JL. Platelets and
10.1358/dnp.2007.20.5.1120215.
shear stress. Blood. 1996;88:1525–1541. 36. Flaumenhaft R, Furie B, Zwicker JI. Therapeutic implications of protein
16. Han BW, Herrin BR, Cooper MD, Wilson IA. Antigen recognition by vari- disulfide isomerase inhibition in thrombotic disease. Arterioscler Thromb
able lymphocyte receptors. Science. 2008;321:1834–1837. doi: 10.1126/ Vasc Biol. 2015;35:16–23. doi: 10.1161/ATVBAHA.114.303410.
science.1162484. 37. Durante W, Schafer AI, Hrbolich JK, Claure RA, Mendelsohn
17. Arthur JF, Qiao J, Shen Y, Davis AK, Dunne E, Berndt MC, Gardiner ME, Kroll MH. Endothelium-derived relaxing factor inhibits shear
EE, Andrews RK. ITAM receptor-mediated generation of reactive oxy- stress-induced platelet aggregation. Platelets. 1993;4:135–140. doi:
gen species in human platelets occurs via Syk-dependent and Syk- 10.3109/09537109309013209.
independent pathways. J Thromb Haemost. 2012;10:1133–1141. doi: 38. Ikeda Y, Handa M, Kawano K, Kamata T, Murata M, Araki Y, Anbo H,
10.1111/j.1538-7836.2012.04734.x. Kawai Y, Watanabe K, Itagaki I. The role of von Willebrand factor and
18. Arthur JF, Shen Y, Gardiner EE, Coleman L, Murphy D, Kenny fibrinogen in platelet aggregation under varying shear stress. J Clin Invest.
D, Andrews RK, Berndt MC. TNF receptor-associated factor 4 1991;87:1234–1240. doi: 10.1172/JCI115124.
(TRAF4) is a novel binding partner of glycoprotein Ib and glycopro- 39. Baigent C, Blackwell L, Collins R, Emberson J, Godwin J, Peto R,
tein VI in human platelets. J Thromb Haemost. 2011;9:163–172. doi: Buring J, Hennekens C, Kearney P, Meade T, Patrono C, Roncaglioni MC,
10.1111/j.1538-7836.2010.04091.x. Zanchetti A; Antithrombotic Trialists’ (ATT) Collaboration. Aspirin in the
19. Vara D, Campanella M, Pula G. The novel NOX inhibitor 2-acetyl- primary and secondary prevention of vascular disease: collaborative meta-
phenothiazine impairs collagen-dependent thrombus formation in a analysis of individual participant data from randomised trials. Lancet.
GPVI-dependent manner. Br J Pharmacol. 2013;168:212–224. doi: 2009;373:1849–1860. doi: 10.1016/S0140-6736(09)60503-1.
10.1111/j.1476-5381.2012.02130.x. 40. Eikelboom JW, Hirsh J, Spencer FA, Baglin TP, Weitz JI. Antiplatelet drugs:
20. Walsh TG, Berndt MC, Carrim N, Cowman J, Kenny D, Metharom Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American
P. The role of Nox1 and Nox2 in GPVI-dependent platelet activation College of Chest Physicians Evidence-Based Clinical Practice Guidelines.
and thrombus formation. Redox Biol. 2014;2:178–186. doi: 10.1016/j. Chest. 2012;141(2 suppl):e89S–119S. doi: 10.1378/chest.11-2293.
redox.2013.12.023. 41. Sandercock PA, Counsell C, Tseng MC, Cecconi E. Oral antiplate-
21. Mu FT, Andrews RK, Arthur JF, Munday AD, Cranmer SL, Jackson SP, let therapy for acute ischaemic stroke. Cochrane Database Syst Rev.
Stomski FC, Lopez AF, Berndt MC. A functional 14-3-3zeta-independent 2014;3:CD000029. doi: 10.1002/14651858.CD000029.pub3.
association of PI3-kinase with glycoprotein Ib alpha, the major ligand- 42. Ciccone A, Motto C, Abraha I, Cozzolino F, Santilli I. Glycoprotein IIb-
binding subunit of the platelet glycoprotein Ib-IX-V complex. Blood. IIIa inhibitors for acute ischaemic stroke. Cochrane Database Syst Rev.
2008;111:4580–4587. doi: 10.1182/blood-2007-09-111096. 2014;3:CD005208. doi: 10.1002/14651858.CD005208.pub3.
22. Mu FT, Cranmer SL, Andrews RK, Berndt MC. Functional association of 43. Mehta SR, Tanguay JF, Eikelboom JW, et al; CURRENT-OASIS 7 Trial
phosphoinositide-3-kinase with platelet glycoprotein Ibalpha, the major Investigators. Double-dose versus standard-dose clopidogrel and high-
ligand-binding subunit of the glycoprotein Ib-IX-V complex. J Thromb dose versus low-dose aspirin in individuals undergoing percutaneous
Haemost. 2010;8:324–330. doi: 10.1111/j.1538-7836.2009.03672.x. coronary intervention for acute coronary syndromes (current-oasis 7): a
1336   Arterioscler Thromb Vasc Biol   June 2015

randomised factorial trial. Lancet. 2010;376:1233–1243. doi: 10.1016/ outcomes of clopidogrel treatment. N Engl J Med. 2010;363:1704–1714.
S0140-6736(10)61088-4. doi: 10.1056/NEJMoa1008410.
44. Roe MT, Armstrong PW, Fox KA, et al; TRILOGY ACS Investigators. 62. Wallentin L, James S, Storey RF, Armstrong M, Barratt BJ, Horrow J,
Prasugrel versus clopidogrel for acute coronary syndromes without Husted S, Katus H, Steg PG, Shah SH, Becker RC; PLATO Investigators.
revascularization. N Engl J Med. 2012;367:1297–1309. doi: 10.1056/ Effect of CYP2C19 and ABCB1 single nucleotide polymorphisms
NEJMoa1205512. on outcomes of treatment with ticagrelor versus clopidogrel for acute
45. Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo coronary syndromes: a genetic substudy of the PLATO trial. Lancet.
W, Gottlieb S, Neumann FJ, Ardissino D, De Servi S, Murphy SA, 2010;376:1320–1328. doi: 10.1016/S0140-6736(10)61274-3.
Riesmeyer J, Weerakkody G, Gibson CM, Antman EM; TRITON-TIMI 63. Bergmeijer TO, Janssen PW, Schipper JC, et al. CYP2C19 genotype-
38 Investigators. Prasugrel versus clopidogrel in patients with acute guided antiplatelet therapy in ST-segment elevation myocardial infarc-
coronary syndromes. N Engl J Med. 2007;357:2001–2015. doi: 10.1056/ tion patients-rationale and design of the Patient Outcome after primary
NEJMoa0706482. PCI (POPular) Genetics study. Am Heart J. 2014;168:16–22.e1. doi:
46. Pilgrim T, Windecker S. Antiplatelet therapy for secondary prevention of 10.1016/j.ahj.2014.03.006.
coronary artery disease. Heart. 2014;100(22):1750–1756. doi: 10.1136/ 64. López JA, Andrews RK, Afshar-Kharghan V, Berndt MC. Bernard-Soulier
heartjnl-2013-305399. syndrome. Blood. 1998;91:4397–4418.
47. Becker RC, Bassand JP, Budaj A, et al. Bleeding complications with 65. Andrews RK, Berndt MC. Bernard-Soulier syndrome: an update. Semin
the P2Y12 receptor antagonists clopidogrel and ticagrelor in the Thromb Hemost. 2013;39:656–662. doi: 10.1055/s-0033-1353390.
PLATelet inhibition and patient Outcomes (PLATO) trial. Eur Heart J. 66. Arthur JF, Dunkley S, Andrews RK. Platelet glycoprotein
2011;32:2933–2944. doi: 10.1093/eurheartj/ehr422. VI-related clinical defects. Br J Haematol. 2007;139:363–372. doi:
48. Stone GW, Bertrand ME, Moses JW, et al; ACUITY Investigators. Routine 10.1111/j.1365-2141.2007.06799.x.
upstream initiation vs deferred selective use of glycoprotein IIb/IIIa 67. Bergmeier W, Piffath CL, Goerge T, Cifuni SM, Ruggeri ZM, Ware
inhibitors in acute coronary syndromes: the ACUITY timing trial. JAMA. J, Wagner DD. The role of platelet adhesion receptor GPIbalpha far
2007;297:591–602. doi: 10.1001/jama.297.6.591. exceeds that of its main ligand, von Willebrand factor, in arterial throm-
49. Habib A, Finn AV. Endothelialization of drug eluting stents and its impact bosis. Proc Natl Acad Sci U S A. 2006;103:16900–16905. doi: 10.1073/
on dual anti-platelet therapy duration. Pharmacol Res. 2015;93:22–27. pnas.0608207103.
doi: 10.1016/j.phrs.2014.12.003. 68. Massberg S, Gawaz M, Grüner S, Schulte V, Konrad I, Zohlnhöfer D,
50. Tantry US, Bonello L, Aradi D, et al; Working Group on On-Treatment Heinzmann U, Nieswandt B. A crucial role of glycoprotein VI for platelet
Platelet Reactivity. Consensus and update on the definition of on-treat- recruitment to the injured arterial wall in vivo. J Exp Med. 2003;197:41–49.
ment platelet reactivity to adenosine diphosphate associated with ischemia 69. Kleinschnitz C, Pozgajova M, Pham M, Bendszus M, Nieswandt B, Stoll
and bleeding. J Am Coll Cardiol. 2013;62:2261–2273. doi: 10.1016/j. G. Targeting platelets in acute experimental stroke: impact of glycopro-
jacc.2013.07.101. tein Ib, VI, and IIb/IIIa blockade on infarct size, functional outcome, and
51. Li J, Jian Z, Song M, Guo W, Chen G, Lu W, Qian D, Ouyang J, Yu J, Hu intracranial bleeding. Circulation. 2007;115:2323–2330. doi: 10.1161/
H, Jin J, Wu X, Huang L. Tailored antiplatelet therapy and clinical adverse CIRCULATIONAHA.107.691279.
outcomes. Heart. 2014;100:41–46. doi: 10.1136/heartjnl-2013-304461. 70. Wijeyewickrema LC, Berndt MC, Andrews RK. Snake venom probes
52. Lewis JP, Ryan K, O’Connell JR, et al. Genetic variation in PEAR1 of platelet adhesion receptors and their ligands. Toxicon. 2005;45:1051–
is associated with platelet aggregation and cardiovascular out- 1061. doi: 10.1016/j.toxicon.2005.02.025.
comes. Circ Cardiovasc Genet. 2013;6:184–192. doi: 10.1161/ 71. Lee’s Pharmaceutical Limited. Anfibatide phase 1 clinical trial in healthy
Downloaded from http://ahajournals.org by on February 11, 2024

CIRCGENETICS.111.964627. volunteers. http://clinicaltrials.gov/ct2/show/NCT01588132. 2011.


53. Aradi D, Tornyos A, Pintér T, Vorobcsuk A, Kónyi A, Faluközy J, Veress 72. Lee’s Pharmaceutical Limited. Anfibatide phase ib-iia clinical trial. http://
G, Magyari B, Horváth IG, Komócsi A. Optimizing P2Y12 receptor inhi- www.clinicaltrials.gov/ct2/show/NCT01585259. 2012.
bition in patients with acute coronary syndrome on the basis of platelet 73. Hou Y, Li BX, Dai X, Yang Z, Qian F, Zhang G, Xu Z, Liu J, Liang C,
function testing: impact of prasugrel and high-dose clopidogrel. J Am Coll Zhao Y, Shen J, Zhou H, Wang Y, Marshall AH, Ni H. The first in vitro
Cardiol. 2014;63:1061–1070. doi: 10.1016/j.jacc.2013.12.023. and in vivo assessment of anfibatide, a novel glycoprotein ib antagonist, in
54. Price MJ, Berger PB, Teirstein PS, et al; GRAVITAS Investigators. mice and in a phase i human clinical trial. Blood. 2013;122:577.
Standard- vs high-dose clopidogrel based on platelet function testing after 74. Ungerer M, Rosport K, Bültmann A, Piechatzek R, Uhland K, Schlieper P,
percutaneous coronary intervention: the GRAVITAS randomized trial. Gawaz M, Münch G. Novel antiplatelet drug revacept (dimeric glycopro-
JAMA. 2011;305:1097–1105. doi: 10.1001/jama.2011.290. tein VI-Fc) specifically and efficiently inhibited collagen-induced platelet
55. Collet JP, Cuisset T, Rangé G, et al; ARCTIC Investigators. Bedside moni- aggregation without affecting general hemostasis in humans. Circulation.
toring to adjust antiplatelet therapy for coronary stenting. N Engl J Med. 2011;123:1891–1899. doi: 10.1161/CIRCULATIONAHA.110.980623.
2012;367:2100–2109. doi: 10.1056/NEJMoa1209979. 75. AdvanceCor GmbH. Revacept in symptomatic carotid stenosis (revacept/
56. Gurbel PA, Erlinge D, Ohman EM, et al; TRILOGY ACS Platelet cs/02). http://clinicaltrials.gov/ct2/show/NCT01645306. 2014.
Function Substudy Investigators. Platelet function during extended pra- 76. Gardiner EE, Arthur JF, Kahn ML, Berndt MC, Andrews RK.
sugrel and clopidogrel therapy for patients with ACS treated without Regulation of platelet membrane levels of glycoprotein VI by a platelet-
revascularization: the TRILOGY ACS platelet function substudy. JAMA. derived metalloproteinase. Blood. 2004;104:3611–3617. doi: 10.1182/
2012;308:1785–1794. doi: 10.1001/jama.2012.17312. blood-2004-04-1549.
57. Montalescot G, Rangé G, Silvain J, et al; ARCTIC Investigators. High 77. Gardiner EE, Karunakaran D, Shen Y, Arthur JF, Andrews RK, Berndt
on-treatment platelet reactivity as a risk factor for secondary preven- MC. Controlled shedding of platelet glycoprotein (GP)VI and GPIb-IX-V
tion after coronary stent revascularization: A landmark analysis of by ADAM family metalloproteinases. J Thromb Haemost. 2007;5:1530–
the ARCTIC study. Circulation. 2014;129:2136–2143. doi: 10.1161/ 1537. doi: 10.1111/j.1538-7836.2007.02590.x.
CIRCULATIONAHA.113.007524. 78. Al-Tamimi M, Tan CW, Qiao J, et al. Pathologic shear triggers shedding of
58. Mega JL, Simon T, Collet JP, et al. Reduced-function CYP2C19 genotype vascular receptors: a novel mechanism for down-regulation of platelet gly-
and risk of adverse clinical outcomes among patients treated with clopi- coprotein VI in stenosed coronary vessels. Blood. 2012;119:4311–4320.
dogrel predominantly for PCI: a meta-analysis. JAMA. 2010;304:1821– doi: 10.1182/blood-2011-10-386607.
1830. doi: 10.1001/jama.2010.1543. 79. Al-Tamimi M, Grigoriadis G, Tran H, Paul E, Servadei P, Berndt MC,
59. Fuster V, Sweeny JM. Clopidogrel and the reduced-function CYP2C19 Gardiner EE, Andrews RK. Coagulation-induced shedding of platelet
genetic variant: a limited piece of the overall therapeutic puzzle. JAMA. glycoprotein VI mediated by factor Xa. Blood. 2011;117:3912–3920. doi:
2010;304:1839–1840. doi: 10.1001/jama.2010.1566. 10.1182/blood-2010-08-301523.
60. Mega JL, Close SL, Wiviott SD, Shen L, Walker JR, Simon T, Antman EM, 80. Gardiner EE, Andrews RK. Plasma sGPVI: changing levels in
Braunwald E, Sabatine MS. Genetic variants in ABCB1 and CYP2C19 human disease. Thromb Res. 2014;133:306–307. doi: 10.1016/j.
and cardiovascular outcomes after treatment with clopidogrel and prasu- thromres.2013.12.028.
grel in the TRITON-TIMI 38 trial: a pharmacogenetic analysis. Lancet. 81. Shen B, Zhao X, O’Brien KA, Stojanovic-Terpo A, Delaney MK, Kim
2010;376:1312–1319. doi: 10.1016/S0140-6736(10)61273-1. K, Cho J, Lam SC, Du X. A directional switch of integrin signalling and
61. Paré G, Mehta SR, Yusuf S, Anand SS, Connolly SJ, Hirsh J, Simonsen a new anti-thrombotic strategy. Nature. 2013;503:131–135. doi: 10.1038/
K, Bhatt DL, Fox KA, Eikelboom JW. Effects of CYP2C19 genotype on nature12613.
Metharom et al   Current State of Antiplatelet Therapy   1337

82. Schaff M, Tang C, Maurer E, et al. Integrin α6β1 is the main receptor comparison meta-analysis. Clin Ther. 2013;35:1490–1500.e7. doi:
for vascular laminins and plays a role in platelet adhesion, activation, 10.1016/j.clinthera.2013.09.004.
and arterial thrombosis. Circulation. 2013;128:541–552. doi: 10.1161/ 103. Gouya G, Arrich J, Wolzt M, Huber K, Verheugt FW, Gurbel PA,
CIRCULATIONAHA.112.000799. Pirker-Kees A, Siller-Matula JM. Antiplatelet treatment for prevention
83. McEwan PA, Andrews RK, Emsley J. Glycoprotein Ibalpha inhibitor com- of cerebrovascular events in patients with vascular diseases: a system-
plex structure reveals a combined steric and allosteric mechanism of von atic review and meta-analysis. Stroke. 2014;45:492–503. doi: 10.1161/
Willebrand factor antagonism. Blood. 2009;114:4883–4885. doi: 10.1182/ STROKEAHA.113.002590.
blood-2009-05-224170. 104. Alvarez-Sabín J, Quintana M, Santamarina E, Maisterra O. Triflusal and
84. Benard SA, Smith TM, Cunningham K, Jacob J, DeSilva T, Lin L, Shaw aspirin in the secondary prevention of atherothrombotic ischemic stroke:
GD, Kriz R, Kelleher KS. Identification of peptide antagonists to glyco- a very long-term follow-up. Cerebrovasc Dis. 2014;37:181–187. doi:
protein Ibalpha that selectively inhibit von Willebrand factor dependent 10.1159/000357662.
platelet aggregation. Biochemistry. 2008;47:4674–4682. doi: 10.1021/ 105. Anninos H, Andrikopoulos G, Pastromas S, Sakellariou D, Theodorakis G,
bi702428q. Vardas P. Triflusal: an old drug in modern antiplatelet therapy. Review of its
85. Hohmann JD, Wang X, Krajewski S, Selan C, Haller CA, Straub A, action, use, safety and effectiveness. Hellenic J Cardiol. 2009;50:199–207.
Chaikof EL, Nandurkar HH, Hagemeyer CE, Peter K. Delayed target- 106. The European Stroke Organisation Executive C, the ESOWC. Guidelines
ing of CD39 to activated platelet GPIIb/IIIa via a single-chain antibody: for management of ischaemic stroke and transient ischaemic attack 2008.
breaking the link between antithrombotic potency and bleeding? Blood. Cerebrovascular Diseases. 2008;25:457–507.
2013;121:3067–3075. doi: 10.1182/blood-2012-08-449694. 107. Piccolo R, Galasso G, De Luca G, Parodi G, Antoniucci D, Esposito G,
86. Pellegatta M, De Arcangelis A, D’Urso A, et al. α6β1 and α7β1 integrins Trimarco B, Piscione F. Relationship between changes in platelet reactiv-
are required in Schwann cells to sort axons. J Neurosci. 2013;33:17995– ity and ischemic events following percutaneous coronary intervention:
18007. doi: 10.1523/JNEUROSCI.3179-13.2013. a meta-regression analysis of 30 randomized trials. Atherosclerosis.
87. Staquicini FI, Dias-Neto E, Li J, Snyder EY, Sidman RL, Pasqualini R, 2014;234:176–184. doi: 10.1016/j.atherosclerosis.2014.02.024.
Arap W. Discovery of a functional protein complex of netrin-4, lam- 108. Wang X, Gong X, Zhu T, Zhang Q, Zhang Y, Wang X, Yang Z, Li C.
inin gamma1 chain, and integrin alpha6beta1 in mouse neural stem Clopidogrel improves aspirin response after off-pump coronary ar-
cells. Proc Natl Acad Sci U S A. 2009;106:2903–2908. doi: 10.1073/ tery bypass surgery. J Biomed Res. 2014;28:108–113. doi: 10.7555/
pnas.0813286106. JBR.28.20120139.
88. Larrieu-Lahargue F, Welm AL, Thomas KR, Li DY. Netrin-4 activates 109. Schulz S, Richardt G, Laugwitz KL, Mehran R, Gershlick AH, Morath
endothelial integrin {alpha}6{beta}1. Circ Res. 2011;109:770–774. doi: T, Mayer K, Neudecker J, Tolg R, Ibrahim T, Hauschke D, Braun D,
10.1161/CIRCRESAHA.111.247239. Schunkert H, Kastrati A, Mehilli J. Comparison of prasugrel and bi-
89. Witzig TE, Gupta M. Signal transduction inhibitor therapy for lymphoma. valirudin vs clopidogrel and heparin in patients with st-segment el-
Hematology Am Soc Hematol Educ Program. 2010;2010:265–270. doi: evation myocardial infarction: design and rationale of the bavarian
10.1182/asheducation-2010.1.265. reperfusion alternatives evaluation (brave) 4 trial. Clinical cardiology.
90. Liu J, Fitzgerald ME, Berndt MC, Jackson CW, Gartner TK. Bruton tyro- 2014;37(5):270–276. doi: 10.1002/clc.22268.
sine kinase is essential for botrocetin/VWF-induced signaling and GPIb- 110. Nanau RM, Delzor F, Neuman MG. Efficacy and safety of prasugrel in
dependent thrombus formation in vivo. Blood. 2006;108:2596–2603. doi: acute coronary syndrome patients. Clin Biochem. 2014;47:516–528. doi:
10.1182/blood-2006-01-011817. 10.1016/j.clinbiochem.2014.03.005.
91. Kamel S, Horton L, Ysebaert L, Levade M, Burbury K, Tan S, Cole- 111. Hibbert B, Maze R, Pourdjabbar A, Simard T, Ramirez FD, Moudgil R,
Downloaded from http://ahajournals.org by on February 11, 2024

Sinclair M, Reynolds J, Filshie R, Schischka S, Khot A, Sandhu S, Keating Blondeau M, Labinaz M, Dick A, Glover C, Froeschl M, Marquis JF, So
MJ, Nandurkar H, Tam CS. Ibrutinib inhibits collagen-mediated but not DY, Le May MR. A comparative pharmacodynamic study of ticagrelor
adp-mediated platelet aggregation [published online ahead of print August versus clopidogrel and ticagrelor in patients undergoing primary percuta-
20, 2014]. Leukemia. 10.1038/leu.2014.247. neous coronary intervention: the CAPITAL RELOAD study. PLoS One.
92. Qiao J, Schoenwaelder SM, Mason KD, Tran H, Davis AK, Kaplan 2014;9:e92078. doi: 10.1371/journal.pone.0092078.
ZS, Jackson SP, Kile BT, Andrews RK, Roberts AW, Gardiner EE. Low 112. Henriksson M, Nikolic E, Ohna A, Wallentin L, Janzon M. Ticagrelor
adhesion receptor levels on circulating platelets in patients with lympho- treatment in patients with acute coronary syndrome is cost-effective
proliferative diseases before receiving Navitoclax (ABT-263). Blood. in Sweden and Denmark. Scand Cardiovasc J. 2014;48:138–147. doi:
2013;121:1479–1481. doi: 10.1182/blood-2012-12-467415. 10.3109/14017431.2014.902494.
93. Gray SP, Di Marco E, Okabe J, et al. NADPH oxidase 1 plays a key 113. Bonaca MP, Bhatt DL, Braunwald E, Cohen M, Steg PG, Storey
role in diabetes mellitus-accelerated atherosclerosis. Circulation. RF, Held P, Jensen EC, Sabatine MS. Design and rationale for the
2013;127:1888–1902. doi: 10.1161/CIRCULATIONAHA.112.132159. Prevention of Cardiovascular Events in Patients With Prior Heart Attack
94. Violi F, Pignatelli P. Platelet NOX, a novel target for anti-thrombotic treat- Using Ticagrelor Compared to Placebo on a Background of Aspirin-
ment. Thromb Haemost. 2014;111:817–823. doi: 10.1160/TH13-10-0818. Thrombolysis in Myocardial Infarction 54 (PEGASUS-TIMI 54) trial.
95. Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Am Heart J. 2014;167:437–444.e5. doi: 10.1016/j.ahj.2013.12.020.
Immunol. 2004;4:181–189. doi: 10.1038/nri1312. 114. Parikh D, Juergens CP. Abciximab as an adjunctive therapy for patients
96. Loyau S, Dumont B, Ollivier V, Boulaftali Y, Feldman L, Ajzenberg N, undergoing percutaneous coronary interventions. Expert Opin Biol Ther.
Jandrot-Perrus M. Platelet glycoprotein VI dimerization, an active pro- 2011;11:235–246. doi: 10.1517/14712598.2011.551113.
cess inducing receptor competence, is an indicator of platelet reactiv- 115. Xu Q, Yin J, Si LY. Efficacy and safety of early versus late glycopro-
ity. Arterioscler Thromb Vasc Biol. 2012;32:778–785. doi: 10.1161/ tein IIb/IIIa inhibitors for PCI. Int J Cardiol. 2013;162:210–219. doi:
ATVBAHA.111.241067. 10.1016/j.ijcard.2012.06.001.
97. Gurbel PA, Kuliopulos A, Tantry US. G-protein-coupled receptors signal- 116. Bledzka K, Smyth SS, Plow EF. Integrin αIIbβ3: from discovery to effi-
ing pathways in new antiplatelet drug development. Arterioscler Thromb cacious therapeutic target. Circ Res. 2013;112:1189–1200. doi: 10.1161/
Vasc Biol. 2015;35:500–512. doi: 10.1161/ATVBAHA.114.303412. CIRCRESAHA.112.300570.
98. US Food and Drug Administration. Drugs@fda. http://www.accessdata. 117. Lang SH, Manning N, Armstrong N, Misso K, Allen A, Di Nisio M,
fda.gov/Scripts/cder/drugsatfda/index.cfm. Accessed April 4, 2014. Kleijnen J. Treatment with tirofiban for acute coronary syndrome
99. US National Library of Medicine. Dailymed. http://dailymed.nlm.nih. (ACS): a systematic review and network analysis. Curr Med Res Opin.
gov/dailymed/about.cfm. Accessed April 4, 2014. 2012;28:351–370. doi: 10.1185/03007995.2012.657299.
100. Law V, Knox C, Djoumbou Y, et al. DrugBank 4.0: shedding new light on 118. Waxman AB, Zamanian RT. Pulmonary arterial hypertension: new
drug metabolism. Nucleic Acids Res. 2014;42(database issue):D1091– insights into the optimal role of current and emerging prostacyclin
D1097. doi: 10.1093/nar/gkt1068. therapies. Am J Cardiol. 2013;111(5 suppl):1A–16A; quiz 17A. doi:
101. Cavalca V, Rocca B, Squellerio I, Dragani A, Veglia F, Pagliaccia F, Porro 10.1016/j.amjcard.2012.12.002.
B, Barbieri SS, Tremoli E, Patrono C. In vivo prostacyclin biosynthe- 119. McLaughlin VV, Palevsky HI. Parenteral and inhaled prostanoid therapy
sis and effects of different aspirin regimens in patients with essential in the treatment of pulmonary arterial hypertension. Clin Chest Med.
thrombocythaemia. Thromb Haemost. 2014;112:118–127. doi: 10.1160/ 2013;34:825–840. doi: 10.1016/j.ccm.2013.09.003.
TH13-10-0844. 120. Torres F, Rubin LJ. Treprostinil for the treatment of pulmonary arte-
102. Malloy RJ, Kanaan AO, Silva MA, Donovan JL. Evaluation of anti- rial hypertension. Expert Rev Cardiovasc Ther. 2013;11:13–25. doi:
platelet agents for secondary prevention of stroke using mixed treatment 10.1586/erc.12.160.
1338   Arterioscler Thromb Vasc Biol   June 2015

121. Channick RN, Voswinckel R, Rubin LJ. Inhaled treprostinil: a therapeutic K, Stanssens P, Baumeister J, Holz JB. Antithrombotic drug candidate
review. Drug Des Devel Ther. 2012;6:19–28. doi: 10.2147/DDDT.S19281. ALX-0081 shows superior preclinical efficacy and safety compared with
122. Ndrepepa G, Neumann FJ, Deliargyris EN, Mehran R, Mehilli J, currently marketed antiplatelet drugs. Blood. 2011;118:757–765. doi:
Ferenc M, Schulz S, Schömig A, Kastrati A, Stone GW. Bivalirudin 10.1182/blood-2010-11-317859.
versus heparin plus a glycoprotein IIb/IIIa inhibitor in patients 136. Bartunek J, Barbato E, Heyndrickx G, Vanderheyden M, Wijns W, Holz
with non-ST-segment elevation myocardial infarction undergo- JB. Novel antiplatelet agents: ALX-0081, a Nanobody directed towards
ing percutaneous coronary intervention after clopidogrel pre- von Willebrand factor. J Cardiovasc Transl Res. 2013;6:355–363. doi:
treatment: pooled analysis from the ACUITY and ISAR-REACT 10.1007/s12265-012-9435-y.
4 trials. Circ Cardiovasc Interv. 2012;5:705–712. doi: 10.1161/ 137. Bousser MG, Amarenco P, Chamorro A, Fisher M, Ford I, Fox KM,
CIRCINTERVENTIONS.112.972869. Hennerici MG, Mattle HP, Rothwell PM, de Cordoüe A, Fratacci MD;
123. Shah A, Feldman DN. Outcome of the HORIZONS-AMI trial: bivali- PERFORM Study Investigators. Terutroban versus aspirin in patients
rudin enhances long-term survival in patients with ST-elevation myo- with cerebral ischaemic events (PERFORM): a randomised, double-
cardial infarction undergoing angioplasty. Vasc Health Risk Manag. blind, parallel-group trial. Lancet. 2011;377:2013–2022. doi: 10.1016/
2012;8:115–123. doi: 10.2147/VHRM.S23491. S0140-6736(11)60600-4.
124. Bloom BJ, Filion KB, Atallah R, Eisenberg MJ. Meta-analysis of ran- 138. Mearns BM. Antiplatelet therapy: terutroban does not out-PERFORM
domized controlled trials on the risk of bleeding with dabigatran. Am J aspirin in secondary prevention. Nat Rev Cardiol. 2011;8:423. doi:
Cardiol. 2014;113:1066–1074. doi: 10.1016/j.amjcard.2013.11.049. 10.1038/nrcardio.2011.93.
125. Providência R, Albenque JP, Combes S, Bouzeman A, Casteigt B, 139. Davì G, Santilli F, Vazzana N. Thromboxane receptors antagonists and/
Combes N, Narayanan K, Marijon E, Boveda S. Safety and efficacy or synthase inhibitors. In: Gresele P, Born GVR, Patrono C, Page CP, eds.
of dabigatran versus warfarin in patients undergoing catheter ablation Antiplatelet agents, Handbook of Experimental Pharmacology. Springer:
of atrial fibrillation: a systematic review and meta-analysis. Heart. Berlin Heidelberg; 2012:261–286.
2014;100:324–335. doi: 10.1136/heartjnl-2013-304386. 140. US National Institutes of Health. Safety and pharmacokinetics of if-
126. Gresele P, Momi S, Falcinelli E. Anti-platelet therapy: phosphodi- etroban in hepatorenal syndrome patients. http://clinicaltrials.gov/show/
esterase inhibitors. Br J Clin Pharmacol. 2011;72:634–646. doi: NCT01436500. Accessed April 4, 2014.
10.1111/j.1365-2125.2011.04034.x. 141. de Souza Brito F, Tricoci P. Novel anti-platelet agents: focus on throm-
127. De Schryver EL, Algra A, van Gijn J. Dipyridamole for prevent- bin receptor antagonists. J Cardiovasc Transl Res. 2013;6:415–424. doi:
ing stroke and other vascular events in patients with vascular disease. 10.1007/s12265-013-9454-3.
Cochrane Database Syst Rev. 2007; Issue 3. Art. No.: CD001820. doi: 142. Rosser G, Tricoci P, Morrow D, Christopoulos C, Niespialowska-Steuden
10.1002/14651858.CD001820.pub3. MN, Kozarski R, Wilcox R, Gorog DA. Par-1 antagonist vorapaxar fa-
128. Lee BK, Lee SW, Park SW, Lee SW, Park DW, Kim YH, Lee CW, Hong vorably improves global thrombotic status in patients with coronary
MK, Kim JJ, Jang S, Chi HS, Park SJ. Effects of triple antiplatelet therapy disease. J Thromb Thrombolysis. 2014;38(4):423–429. doi: 10.1007/
(aspirin, clopidogrel, and cilostazol) on platelet aggregation and P-selectin s11239-014-1075-4.
expression in patients undergoing coronary artery stent implantation. Am 143. Garde D. Merck clears an fda panel with its clot-fighting vorapaxar.
J Cardiol. 2007;100:610–614. doi: 10.1016/j.amjcard.2007.03.070. http://www.fiercebiotech.com/story/merck-clears-fda-panel-its-clot-
129. Stevens JW, Simpson E, Harnan S, Squires H, Meng Y, Thomas S, fighting-vorapaxar/2014-01-16.
Michaels J, Stansby G. Systematic review of the efficacy of cilostazol, 144. O’Donoghue ML, Bhatt DL, Flather MD, Goto S, Angiolillo DJ,
naftidrofuryl oxalate and pentoxifylline for the treatment of intermittent Goodman SG, Zeymer U, Aylward PE, Montalescot G, Ziecina R,
Downloaded from http://ahajournals.org by on February 11, 2024

claudication. Br J Surg. 2012;99:1630–1638. doi: 10.1002/bjs.8895. Kobayashi H, Ren F, Wiviott SD. Atopaxar and its effects on markers
130. Kunieda T, Nakanishi N, Matsubara H, Ohe T, Okano Y, Kondo H, of platelet activation and inflammation: results from the LANCELOT
Nishimura M, Shirato K, Tanabe N, Homma S, Yoshida S, Inokuma S, CAD program. J Thromb Thrombolysis. 2012;34:36–43. doi: 10.1007/
Kodama M, Koike T, Hishida H. Effects of long-acting beraprost sodium s11239-012-0750-6.
(TRK-100STP) in Japanese patients with pulmonary arterial hyperten- 145. Wiviott SD, Flather MD, O’Donoghue ML, Goto S, Fitzgerald DJ,
sion. Int Heart J. 2009;50:513–529. Cura F, Aylward P, Guetta V, Dudek D, Contant CF, Angiolillo DJ,
131. Na KY, Kim DK, Kim SG, Lee YK, Lim CS. Effect of beraprost sodium Bhatt DL; LANCELOT-CAD Investigators. Randomized trial of
on arterial stiffness in patients with type 2 diabetic nephropathy. Trials. atopaxar in the treatment of patients with coronary artery disease: the
2013;14:275. doi: 10.1186/1745-6215-14-275. lessons from antagonizing the cellular effect of Thrombin–Coronary
132. Firbas C, Siller-Matula JM, Jilma B. Targeting von Willebrand fac- Artery Disease Trial. Circulation. 2011;123:1854–1863. doi: 10.1161/
tor and platelet glycoprotein Ib receptor. Expert Rev Cardiovasc Ther. CIRCULATIONAHA.110.001404.
2010;8:1689–1701. doi: 10.1586/erc.10.154. 146. Waite LH, Phan YL, Spinler SA. Cangrelor: a novel intravenous antiplate-
133. Jilma-Stohlawetz P, Gorczyca ME, Jilma B, Siller-Matula J, Gilbert JC, let agent with a questionable future. Pharmacotherapy. 2014;34:1061–
Knöbl P. Inhibition of von Willebrand factor by ARC1779 in patients 1076. doi: 10.1002/phar.1471.
with acute thrombotic thrombocytopenic purpura. Thromb Haemost. 147. Serebruany VL, Pokov AN, Fortmann SD, DiNicolantonio JJ. Disbalance
2011;105:545–552. doi: 10.1160/TH10-08-0520. between mortality and non-fatal vascular events in the CHAMPION-
134. Jilma B, Paulinska P, Jilma-Stohlawetz P, Gilbert JC, Hutabarat R, PHOENIX trial: the cangrelor efficacy challenge. Thromb Haemost.
Knöbl P. A randomised pilot trial of the anti-von Willebrand factor ap- 2014;111:3–7. doi: 10.1160/TH13-08-0631.
tamer ARC1779 in patients with type 2b von Willebrand disease. Thromb 148. Serebruany VL, Aradi D, Kim MH, Sibbing D. Cangrelor infusion
Haemost. 2010;104:563–570. doi: 10.1160/TH10-01-0027. is associated with an increased risk for bleeding: meta-analysis of
135. Ulrichts H, Silence K, Schoolmeester A, de Jaegere P, Rossenu S, Roodt randomized trials. Int J Cardiol. 2013;169:225–228. doi: 10.1016/j.
J, Priem S, Lauwereys M, Casteels P, Van Bockstaele F, Verschueren ijcard.2013.08.133.

Significance
Arterial thrombosis precipitating heart attack and stroke is a major global cause of morbidity and mortality. Current antiplatelet therapies to
limit thrombosis in a variety of clinical settings primarily focus on the combined use of a P2Y12 receptor antagonist and aspirin, although this
is associated with an increased risk of a major bleeding event. An increasing body of evidence has identified several potential therapeutic
targets that would more specifically target arterial thrombosis with more limited or no potential bleeding risk. This review provides an over-
arching state of the art commentary on current and potential antiplatelet therapeutic approaches, which are individually covered in greater
detail in other reviews in this series.

You might also like