ArticlePDF AvailableLiterature Review

Novel antiplatelet agents in the prevention of cardiovascular complications – focus on ticagrelor

Taylor & Francis
Vascular Health and Risk Management
Authors:

Abstract and Figures

Atherothrombosis, thrombus formation as a result of atherosclerotic plaque rupture, is a major modern health problem, often underlying coronary artery disease, stroke, and peripheral arterial disease. After the treatment of an acute thrombotic episode, long-term therapy is warranted as a secondary prophylaxis of such events and their complications. Because of the importance of platelets' involvement in the initiation and propagation of thrombosis, antiplatelet drugs have come to the forefront of atherothrombotic disease treatment. Dual antiplatelet therapy of aspirin plus clopidogrel--the current standard--has its benefits, but it also has its limitations with regard to its pharmacologic properties and adverse effects. For these reasons, within the last decade or so, the investigation of novel antiplatelet agents has prospered. Here, we review the main pathways through which platelets participate in acute thrombosis and the interruption of these pathways by using novel antiplatelet agents, including P2Y12 receptor antagonists (the recently approved prasugrel, the probable next-in-line ticagrelor, and others). The need for a more individualized patient therapy is evident; although most of the aforementioned pharmaceuticals have the potential to contribute to this, their clinical utility remains to be seen.
Content may be subject to copyright.
© 2010 Marczewski et al, publisher and licensee Dove Medical Press Ltd. This is an Open Access article
which permits unrestricted noncommercial use, provided the original work is properly cited.
Vascular Health and Risk Management 2010:6 419–429
Vascular Health and Risk Management
7054
Dovepress
submit your manuscript | www.dovepress.com
Dovepress 419
REVIEW
open access to scientific and medical research
Open Access Full Text Article
Novel antiplatelet agents in the prevention
of cardiovascular complications – focus
on ticagrelor
Margaret M Marczewski1
Marek Postula1,2
Dariusz Kosior1
11st Chair and Department of
Cardiology, Central University
Hospital, Medical University of
Warsaw, Warsaw, Poland; 2Department
of Experimental and Clinical
Pha rma col ogy, Medical Univer sit y
of Warsaw, Warsaw, Poland
Correspondence: Marek Postula
Department of Experimental and Clinical
Pharmacology, Medical University of
Warsaw, 26/28 Krakowskie Przedmieście
St, 00-927 Warsaw, Poland
Tel +48 22 826 2116
Fax +48 22 826 2116
Email mpostula@amwaw.edu.pl
Abstract: Atherothrombosis, thrombus formation as a result of atherosclerotic plaque
rupture, is a major modern health problem, often underlying coronary artery disease, stroke,
and peripheral arterial disease. After the treatment of an acute thrombotic episode, long-term
therapy is warranted as a secondary prophylaxis of such events and their complications. Because
of the importance of platelets’ involvement in the initiation and propagation of thrombosis,
antiplatelet drugs have come to the forefront of atherothrombotic disease treatment. Dual
antiplatelet therapy of aspirin plus clopidogrel the current standard – has its benefits, but
it also has its limitations with regard to its pharmacologic properties and adverse effects. For
these reasons, within the last decade or so, the investigation of novel antiplatelet agents has
prospered. Here, we review the main pathways through which platelets participate in acute
thrombosis and the interruption of these pathways by using novel antiplatelet agents, including
P2Y12 receptor antagonists (the recently approved prasugrel, the probable next-in-line
ticagrelor, and others). The need for a more individualized patient therapy is evident; although
most of the aforementioned pharmaceuticals have the potential to contribute to this, their
clinical utility remains to be seen.
Keywords: antiplatelet therapy, antagonist, P2Y12 receptor, ADP receptor
Different classes of platelet-inhibiting drugs
Platelets have an established role in the pathogenesis of atherosclerosis-related
diseases, including coronary artery disease (CAD), acute coronary syndromes
(ACSs), and stroke. Rupture of an atherosclerotic plaque promotes the activation
of platelets and initiates the coagulation cascade. Activation of platelets and their
subsequent aggregation, which is amplified by various pathways, lead to thrombus
formation at the site of vascular injury. Tissue hypoxia and irreversible damage in
conditions such as myocardial infarction (MI) and ischemic stroke are rather due
to an acute thrombus formed atop a ruptured atherosclerotic plaque than due to the
stenosis caused by atherosclerosis.1 Thus, antiplatelet agents have been used in acute
conditions of arterial thrombosis and as part of secondary prophylaxis to prevent
recurrent thromboembolic episodes.
Acetylsalicylic acid (aspirin) was the first antiplatelet agent, which irreversibly
inhibits the cyclooxygenase 1 enzyme in the arachidonic acid pathway through
acetylation, thereby preventing the conversion of arachidonic acid into thromboxane
A2 (TXA2) – a potent vasoconstrictor and platelet activator.2 Despite being used as a
drug for more than 100 years, aspirin continues to play a dominant role in the treatment
of cardiovascular diseases (CVDs) because of its ease of use and cost-effectiveness.
Number of times this article has been viewed
This article was published in the following Dove Press journal:
Vascular Health and Risk Management
24 May 2010
Vascular Health and Risk Management 2010:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
420
Marczewski et al
Long-term aspirin therapy brings about a 20%–25% odds
reduction in the risks of subsequent MI, stroke, or vascular death
among intermediate- to high-risk patients with atherothrombotic
disease.3,4 However, its relatively low potency [producing only
a partial inhibition of platelet aggregation (IPA)], some patients’
resistance to aspirin, and its gastrointestinal toxicity prompted
the search for more potent, more consistent, and more specific
platelet-inhibiting agents.1–3
Currently, two classes of antiplatelet drugs are used
alongside aspirin. Clopidogrel, a thienopyridine adenosine
diphosphate (ADP) receptor antagonist, on the basis of a
12,562-patient study, CURE (Clopidogrel in Unstable angina
to prevent Recurrent Events) trial in 2001, has become a
standard part of dual antiplatelet therapy with aspirin, which
is used for a longer term after an acute MI.5 Glycoprotein
IIb/IIIa (GP IIb/IIIa) antagonists, such as abciximab, are
used in high-risk patients before percutaneous coronary
intervention (PCI).3 The limitations of clopidogrel its
delayed onset of action, interpatient variability in response
to clopidogrel (including some patients’ outright resistance
to it), and the irreversibility of its action – have encouraged
the investigation of novel antiplatelet agents with satisfactory
safety profiles and adequate efficacy.3,6,7
The process of platelet activation and aggregation involves
multiple signaling molecules and their receptors, therefore
allowing for many therapeutic targets. First, platelets adhere
to the subendothelial proteins (ie, von Willebrand factor
and collagens) exposed at sites of vascular injury. This is
followed by intracellular signaling, with platelet activation
and secretion of further mediators, which amplify and sustain
the initial platelet response.8 In particular, activated platelets
release ADP, serotonin, and calcium via degranulation, as
well as synthesize thromboxane from arachidonic acid;
thrombin, another key substance, is locally generated
through the coagulation cascade.9,10 Platelet activation is
propagated through blood-soluble agonists acting upon
their respective platelet receptors: ADP via P2Y1 and P2Y12,
thrombin via protease-activated receptor 1 (PAR1) and
PAR4, and thromboxane via the thromboxane/prostanoid
(TP) receptor.11 The final common pathway for all these auto-
crine and paracrine activation signals is GP IIb/IIIa-mediated
platelet aggregation. Thus, in both physiologic hemostasis
and pathologic states, platelets are recruited into the mass of
a platelet – fibrin thrombus.9–11
The various classes of antiplatelet drugs act synergistically
through complementary yet independent mechanisms,
preventing platelet aggregation and thus acute thrombus
formation. Currently available drugs and those under
investigation target the thromboxane-induced (ie, aspirin and
terutroban), ADP-induced (such as ticlopidine, clopidogrel,
and prasugrel), and thrombin-induced (eg, SCH 530348
and E5555) pathways of platelet activation and their final
common pathway of GP IIb/IIIa (abciximab, eptifibatide, and
tirofiban)-induced platelet aggregation.11–14 The processes of
platelet adhesion, activation, and aggregation along with the
targets of platelet-inhibiting drugs are shown in Figure 1.
All antiplatelet drugs, in addition to inhibiting acute
arterial thrombosis, have the danger of interfering with the
physiologic role of platelets in hemostasis. Thus, the range
of adverse effects, particularly bleeding, is a major factor
in evaluating the utility of the available and upcoming
antiplatelet drugs and their combination regimens. In
addition to the safety profile and drug efficacy, other factors
to be taken into account when choosing from the available
and developing antiplatelet drugs include the specificity to
platelets, route of administration, reversibility of action,
the onset of action, peak of effect, and its duration or
wearing-off time (offset).
Blockade of ADP receptors
of subtype P2Y12 – from the
molecular level to the clinical
utility of thienopyridines
The key role of ADP in the processes of platelet aggregation and
thrombus formation has led to the development of antiplatelet
drugs targeting the P2Y12 receptor. Activated platelets release
ADP from their dense granules, which functions as a soluble
positive feedback mediator that binds to the receptors (P2Y1
and P2Y12) on the platelets’ surface. Both these purinoreceptors
belong to the group of G-protein-coupled receptors, with P2Y1
being coupled to Gq and P2Y12 to Gi; thus, each subtype of
ADP receptor has a distinct intracellular signaling pathway.9,10
Stimulation of the P2Y1 receptor and its Gq protein mobilizes
intracellular calcium and triggers a change in the platelet shape
and rapidly reversible aggregation.15 A signal through the
P2Y12 receptor and its Gi protein results in reduced levels of
cyclic adenosine monphosphate, amplification of the platelet
response, stabilization of the resulting aggregates, and secre-
tion of further mediators from the granules.16 Although the
coactivation of both purinoreceptors is necessary for normal
ADP-induced aggregation, P2Y12 is considered the major
platelet ADP receptor, and because of its more restricted
expression, it has become an attractive therapeutic target of
antithrombotic agents.9,13,17
The thienopyridine class of antiplatelets (ticlopidine,
clopidogrel, and prasugrel) selectively and irreversibly
Vascular Health and Risk Management 2010:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
421
Novel mechanisms of action of antiplatelet agents
inhibits the P2Y12 purinoreceptor throughout the lifespan of
the platelet. Although currently clopidogrel is the dominant
member, its modest platelet inhibition, delayed onset of
action, and significant interpatient variability in response to
clopidogrel (including some patients’ nonresponsiveness to
the drug) paved the way for more potent and stable drugs.18
Ticlopidine had been discovered before clopidogrel, but has
been eclipsed because of its adverse hematologic side effects,
including neutropenia and thrombotic thrombocytopenic
purpura.3 The opposite is true of the third-generation
thienopyridine, prasugrel previously known as CS-747,
LY-640315 – which has been approved by the United States
Food and Drug Administration recently and has found a
place in the latest recommendations of British and American
institutions.19,20
All the orally administered thienopyridines are prodrugs,
which require biotransformation into their active metabolites
by fixed sets of hepatic cytochrome P450 (CYP) enzymes, in
particular, the CYP3A4 isozyme. In contrast to clopidogrel’s
esterase inactivation and two-step CYP-dependent activation,
prasugrel has a more efficient and simpler metabolism,
which requires only one reaction by the liver enzymes
to yield its active metabolite.21,22 This difference in the
metabolism of these thienopyridines translates clinically
into different patient responses and drug interactions with
these antiplatelet agents. Certain common genetic variants of
CYP2C19 resulted in decreased levels of clopidogrel’s active
metabolite, resulting in a lower platelet inhibition and most
importantly a higher rate of major adverse cardiovascular
events; on the other hand, prasugrel’s pharmacology and
clinical efficacy were not found to be affected by CYP
polymorphisms or by the concomitant use of CYP-inhibitory
drugs. Other advantages of prasugrel include a faster onset
of action and an approximately tenfold higher potency than
clopidogrel.22–27
Prasugrel has been shown to be of particular benefit in
patients with diabetes, especially those on insulin (30%
relative risk reduction [RRR] in cardiovascular death, MI,
or stroke [P , 0.001] and 37% RRR, respectively). Another
look at the TRITON-TIMI 38 data with respect to diabetes
status showed the reduction by prasugrel of the composite
of CV death, MI and stroke noted in non-diabetic subjects
(n = 10,462, 9.2% vs 10.6%, P = 0.02), was even more
significant in those with diabetes mellitus (n = 3,146, 12.2%
vs 17.0%, P , 0.001), especially those treated with insulin
(n = 776, 14.3% vs 22.2%, P = 0.009).14,28 On the other
hand, its dose should be adjusted for low-weight patients
(,60 kg), and it should be rather avoided in the elderly
($75 years) and in those with a history of stroke or transient
ischemic attack because of an increased bleeding risk.29 Such
recommendations are largely based on the results of the
Phase 3 TRITON-TIMI 38 (TRial to assess Improvement
A) ADHESION
PLATELET
B) ACTIVATION C) AGGREGATION
PLATELET
collagen
ADP
P2Y2
P2Y1
PAR4
PAR1
Thrombin
α granule
AACOXTXA2
TP
Fibrinogen
GP IIb/IIa
GP IIb/IIa
vWF
endothelial
cell
TXA2
SCH 530348
E5555
ASA
NCX4016
terutroban
thienopyridines:
ticlopidine
clopidogrel
prasugrel
abciximab
eptifibatide
tirofiban
non-thienopyridines:
thrombin receptor
antagonists
ADP receptor
antagonists
GP IIb/IIa
antagonists
ticagrelor
cangrelor
elinogrel - PRT060128
BX667
BX048
Figure 1 Scheme of platelet adhesion, activation, and aggregation processes – showing the key players and sites of action of antiplatelet agents. Drugs at various stages of
investigation appear in parentheses.
Abbreviations: AA, arachidonic acid; ADP, adenosine diphosphate; ASA, aspirin; COX, cyclooxygenase; GP, glycoprotein; PAR, protease-activated receptor; PR, thromboxane/
prostanoid receptor; TXA2, thromboxane; vWF, von Willebrand Factor.
Vascular Health and Risk Management 2010:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
422
Marczewski et al
in Therapeutic Outcomes by optimizing platelet inhibition
with prasugrel – Thrombolysis In Myocardial Infarction 38)
on 13,608 patients with ACS undergoing planned PCI.30 It
demonstrated the composite rate of death, MI, or stroke to
be reduced by 19% and the rate of stent thrombosis to be
halved in patients receiving prasugrel compared with those
receiving clopidogrel. Although prasugrel had an increased
efficacy, it also increased the risk of bleeding, especially in
the aforementioned groups, including life-threatening hemor-
rhage and fatal bleeding.14,30
Although prasugrel is still under investigation, it has
already found an official place in antiplatelet therapy through
support from American and British institutions. In October
2009, the United Kingdom’s National Institute for Health
and Clinical Excellence recommended a limited use of
prasugrel, in combination with aspirin, in three specific but
sizeable subgroups of patients with ACS undergoing PCI:
those undergoing immediate primary PCI for myocardial
infarction with ST elevation (STEMI), those who had stent
thrombosis during clopidogrel treatment, and those with
diabetes mellitus.20 In November 2009, the American Col-
lege of Cardiology, the American Heart Association, and the
Society for Cardiovascular Angiography and Interventions
jointly released guidelines for the use of prasugrel in patients
with STEMI once the coronary anatomy is known and PCI
is planned.19
Ticagrelor – an overview of its
pharmacologic and clinical proles
Novel nonthienopyridine platelet P2Y12 receptor antago-
nists, including ticagrelor, cangrelor, and elinogrel, are also
being investigated. These are direct and reversible P2Y12
antagonists with various formulations but with rapid onsets
and short durations of action. Among these, ticagrelor has
come the furthest, having undergone a Phase 3 clinical
trial.31 As a prototype drug from the novel cyclopentyl-
triazolopyrimidine class of antiplatelets, ticagrelor (previ-
ously known as AZD6140) is chemically distinct from the
thienopyridines and can reversibly inhibit the P2Y12 ADP
receptor. It is highly selective and very specific for the P2Y12
receptor, and it exhibits a greater, more consistent IPA than
clopidogrel.32 Like the thienopyridines, ticagrelor is also
administered orally, but because of its direct action, ie, not
requiring metabolic activation, ticagrelor has a rapid onset,
peaking within 2–4 hours of dosing.33 The metabolism of
ticagrelor yields an active molecule (AR-C124910XX) that
has similar P2Y12-blocking activity as its parent molecule.
Ticagrelor’s plasma half-life is approximately 12 hours,
which corresponds to twice-daily dosing. In contrast to
clopidogrel and prasugrel, which should be discontinued
approximately 5 days before the restoration of normal
platelet-mediated hemostasis, ticagrelor’s short duration of
action and an offset of action in 1–2 days preclude the need
for delaying surgical intervention, such as coronary artery
bypass grafting (CABG). On the other hand, this potential
advantage also carries the risk of increased thrombotic events
if patients miss a ticagrelor dose.14,18,19 The pharmacologic
properties of some currently used and investigational anti-
platelet drugs are shown in Table 1.
Several Phase 2 clinical trials have been conducted using
ticagrelor, the first reversibly binding oral P2Y12 receptor
antagonist (Table 2). In an initial Phase 2a parallel-group
safety and efficacy trial (DISPERSE), 200 patients with
atherosclerosis were randomized to receive either ticagrelor
(doses of 50, 100, or 200 mg twice daily, or 400 mg once
daily) or clopidogrel (75 mg once daily) for 28 days, in addi-
tion to 75–100 mg of aspirin per day. Measuring the IPA
showed that the three higher doses of ticagrelor (ie, 100 and
200 mg twice daily and 400 mg once daily) provide a nearly
complete inhibition of ADP-induced platelet aggregation
after the initial dosing. Although investigators write that
in general ticagrelor was well tolerated, there was a higher
incidence of bleeding with the three higher doses, mostly
of mild to moderate severity, and there were also reports of
dyspnea.36
This trial was followed by the Phase 2b Dose confIrma-
tion Study assessing anti-Platelet Effects of AZD6140 vs
clopidogRel in non-ST-segment Elevation acute coronary
syndromes (DISPERSE-2) trial.37 In this trial, 990 patients
with non-ST-segment elevation ACS were randomized to
receive any one of the following dosage: ticagrelor 90 mg
twice daily, ticagrelor 180 mg twice daily, or clopidogrel
300 mg loading dose plus 75 mg once daily for up to 12
weeks. This study showed no difference in major bleeding but
an increase in minor bleeding at the higher dose of ticagrelor,
with encouraging although not statistically significant
results on the secondary end point of MI.37 For the possible
advantage of ticagrelor’s reversible inhibition, there was only
a numerically lower rate of bleeding (again nonsignificant) in
ticagrelor-treated patients undergoing CABG between 1 and
5 days after stopping ticagrelor, which would be consistent
with the recovery of platelet function. Another analysis of
the DISPERSE-2 data compared the antiplatelet effects of
ticagrelor with those of clopidogrel and assessed the effects
of ticagrelor on clopidogrel-pretreated patients.39 It demon-
strated that ticagrelor treatment resulted in further IPA in
Vascular Health and Risk Management 2010:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
423
Novel mechanisms of action of antiplatelet agents
Table 1 Pharmacologic properties and adverse effects of a selection of antiplatelet agents
Drug Action Aggregation
to ADP
Route of
administration
Metabolism Time to
peak effect
Offset of
action
Adverse effects
Clopidogrel
300 mg
IRR 30% Oral Esterase inactivation
and two-step hepatic
CYP-dependent
activation
4 hours 5 days Bleeding (major and
minor), interpatient
variability, and
dyspnea in
approximately
8% of patients34
Prasugrel
60 mg
IRR 75%–80% Oral Esterase activation
and one-step CYP-
dependent activation
(liver or gut)
1–2 hours 5 days Bleeding (major and
minor) and dyspnea in
approximately
5% of patients30
Ticagrelor R 75%–80% Oral None required 1–2 hours 1–2 days Bleeding, dyspnea in
approximately
14% of patients,
and ventricular pauses31
Cangrelor
4 μg/kg/min
R.90% Intravenous None required Minutes 20 minutes Bleeding (minor) and
dyspnea in
approximately
1% of patients35
Elinogrel
40 mg
R Oral/intravenous None required 20 minutes 1 day No increase in bleeding
SCH 530348 R (.90% to
TRAP)
Oral None required With
LD: hours;
without
LD: days
Weeks No increase in bleeding
Abbreviations: ADP, adenosine diphosphate; CYP, cytochrome P450; IRR, irreversible; LD, loading dose; R, reversible; TRAP, thrombin receptor antagonist peptide.
patients already receiving clopidogrel, including those with
the highest platelet aggregation response.
On the basis of previous dose-confirmation trials, the
Phase 3 PLATO (PLATelet inhibition and patient Outcomes)
trial was conducted to compare ticagrelor and clopidogrel
with respect to their efficacy in preventing cardiovascular
events and safety.31 In this trial, 18,624 patients with ACS
(both with and without ST-segment elevation), in addition
to the aspirin therapy, received loading doses of ticagrelor
180 mg or clopidogrel 300 mg (with an additional 300 mg
clopidogrel at PCI) and then ticagrelor 90 mg twice daily or
clopidogrel 75 mg once daily for 6–12 months. As compared
to treatment with clopidogrel, ticagrelor was found to
significantly reduce the rate of death from vascular causes,
MI, or stroke (primary end point: 11.7% vs 9.8%; P , 0.001),
without an increase in the rate of overall major bleeding.31
These findings are noteworthy in that ticagrelor is the first
investigational antiplatelet to demonstrate a reduction in
cardiovascular death when compared with clopidogrel in
patients with ACS. Ticagrelor reduced the risk of cardio-
vascular events early on, and this benefit over clopidogrel
increased over time. Among the subset of patients who
received a stent during the study, a 38% RRR of definite
stent thrombosis was achieved with ticagrelor (1.0% vs 1.6%;
P = 0.003).40 Patients weighing less than the median weight
for their sex, those who were not on lipid-lowering drugs at
randomization, and those from North America were found
to attain fewer benefits from ticagrelor treatment.31 Although
there was no increased risk of CABG-related bleeding, the
PLATO investigators did find an increase in the rate of
non-procedure-related bleeding (clopidogrel 3.8% vs ticagre-
lor 4.5%; P = 0.03), including more instances of fatal intrac-
ranial bleeding but fewer instances of fatal bleeding of other
types.31 The summation of major and minor bleeding events
also showed an increase with ticagrelor when compared
with clopidogrel (ticagrelor 16.1% vs clopidogrel 14.6%;
P = 0.008).40 In addition to the bleeding tendency, in line with
the findings of Phase 2 trials, ticagrelor was associated with
more ventricular pauses in the first week (ticagrelor 5.8% vs
clopidogrel 3.6%; P = 0.01), but not at day 30, and with more
reports of dyspnea (ticagrelor 13.8% vs clopidogrel 7.8%;
P , 0.001).31 The former occurred without clinical conse-
quences for the patient, whereas the latter was responsible
for 1 in 100 ticagrelor-treated patients stopping treatment.41
Thus, dyspnea was clinically limiting in around 1% of
ticagrelor-treated patients. This rare but clinically meaning-
ful and recurring finding awaits authoritative assessment and
explanation. Given the large database of the PLATO trial,
Vascular Health and Risk Management 2010:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
424
Marczewski et al
analyses continue to be made of the findings. A subanalysis
of patients with STEMI (PLATO STEMI) also showed that
compared with clopidogrel treatment, ticagrelor treatment
reduced cardiovascular events for up to a year (11.0% vs
9.3%; P = 0.02), a difference driven mainly by a statisti-
cally significant reduction in MIs (6.1% vs 4.7%; P = 0.01),
without an increase in major bleeding.42
In the meantime, results of further Phase 2 trials of
ticagrelor were released. The ONSET/OFFSET study
(n = 123) was designed to assess the rates of onset and offset
of the antiplatelet effects of ticagrelor versus clopidogrel
taken for 6 weeks by patients with stable CAD receiving
aspirin therapy.38 Using the same dosing scheme as that used
in PLATO trial, it showed that ticagrelor achieved faster
onset (41% IPA vs 8% at 30 minutes; P , 0.0001), greater
IPA, and faster offset in comparison with clopidogrel. The
RESPOND trial, through a two-way crossover, investigated
the antiplatelet effect of ticagrelor in 98 patients with stable
CAD, which included both clopidogrel responders and
clopidogrel nonresponders.43,44 The importance of this issue
is reflected in reports estimating the frequency of clopidogrel
nonresponders at about 10% and low responders at about
20%.44 Among patients identified as clopidogrel responders,
switching from clopidogrel to ticagrelor resulted in a mean
IPA increase of 26%, whereas switching from ticagrelor to
clopidogrel resulted in a mean IPA decrease of 24%.45 Thus,
the RESPOND trial found that patients could be switched
from ticagrelor treatment to clopidogrel treatment without
the interruption of antiplatelet effect. It was concluded that
clopidogrel nonresponders and responders exhibit superior
platelet inhibition during ticagrelor therapy. However, the
RESPOND trial findings also demonstrated that because of
the variability of the clopidogrel response, identification of
clopidogrel nonresponders is difficult.41 Finally, research has
shown ticagrelor to produce platelet inhibition, regardless
of the genotypic variations in the three genes that had been
associated with the variability in platelet reactivity.39
All these trials underline the potential of ticagrelor to
achieve a rapid and sustained antiplatelet effect, which
could be reversed and could overcome nonresponsiveness
and interpatient variability to clopidogrel, thus addressing
the main limitations of clopidogrel therapy.3 Nonetheless,
Table 2 Ticagrelor: results of clinical trials
Phase Clinical trial Patients Dosage Results
Phase 2 DISPERSE36 200 patients
with atherosclerosis
Randomization to ticagrelor
(doses of 50, 100, or 200 mg
TD, or 400 mg OD) or
clopidogrel (75 mg OD),
on top of aspirin (75–100 mg OD),
for 28 days
Ticagrelor TD produced a more rapid
and greater IPA than clopidogrel
(90% vs 60%)
DISPERSE-237 990 patients
with non-ST-segment
elevation ACS
Randomization to ticagrelor
(90 or 180 mg TD) or
clopidogrel (75 mg OD),
on top of aspirin
(75 and 100 mg OD),
for up to 12 weeks
No difference was observed in the total
bleeding rate between 90-mg
ticagrelor-treated, 180-mg
ticagrelor-treated, and clopidogrel-
treated groups (9.8%, 8.0%, and 8.1%,
respectively)
ONSET/OFFSET38 123 patients
with stable CAD
Randomization to ticagrelor
(90 mg TD), clopidogrel
(75 mg OD), or placebo,
on top of aspirin (75–100 mg OD),
for 6 weeks
Ticagrelor achieved more rapid and
greater platelet inhibition than high-LD
clopidogrel .50% IPA (98% vs 31%,
P , 0.0001) and .70% IPA (90% vs
16%, P , 0.0001) at 2-h post-LD; this
was sustained during the maintenance
phase and was faster in offset after
drug discontinuation
Phase 3 PLATO31 18,624 patients
with ACS
Randomization to LDs of
ticagrelor (180 mg) or
clopidogrel (300 mg; with an
additional 300 mg clopidogrel at
PCI), then ticagrelor (90 mg TD)
or clopidogrel (75 mg OD), on
top of aspirin (75–100 mg OD),
for 6–12 months
Compared with clopidogrel treatment,
ticagrelor treatment was found to
signicantly reduce the rate of death
from vascular causes, MI, or stroke
(primary end point: 11.7% vs 9.8%,
P , 0.001), without an increase in
the rate of overall major bleeding
Abbreviations: ACS, acute coronary syndrome; CAD, coronary artery disease; IPA, inhibition of platelet aggregation; LD, loading dose; MI, myocardial infarction; OD, once
daily; PCI, percutaneous coronary intervention; TD, twice daily.
Vascular Health and Risk Management 2010:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
425
Novel mechanisms of action of antiplatelet agents
its adverse effects such as dyspnea and bradycardia, its
relationship with patient weight, and the lack of its benefit in
North American patients require further investigation before
ticagrelor may advance further toward a place in antiplatelet
therapy guidelines.31
Other antiplatelet drugs in research
and development and their clinical
potential
Cangrelor, another novel reversible P2Y12 antagonist, is a
chemically modified version of ticagrelor, which can be
administered parenterally. It acts rapidly, producing profound
platelet inhibition within 15 minutes of initiating infusion
when compared with the 1- to 2-hour time frame for prasugrel
and ticagrelor. This intravenous (iv) adenosine triphosphate
analog has a plasma half-life of 5–9 minutes and is highly
reversible, so the platelet function returns to the baseline
level of activation 20 minutes after the discontinuation of
cangrelor.14,46,47 Because of such a rapid offset, cangrelor
was foreseen as a drug for the catheterization laboratory;
like GP IIb/IIIa antagonists, it has to be started right before
a PCI procedure and stopped immediately after it, while
other antiplatelets would provide long-term protection
from then onward.46,48 Phase 2 safety trials did not note
any deaths or serious events attributed to cangrelor.46,47
CHAMPION-PCI (A Clinical Trial Comparing Cangrelor
to Clopidogrel in Subjects Who Require Percutaneous
Coronary Intervention), a prospective Phase 3 trial of 9,000
patients with unstable angina, MI, or ACS awaiting PCI,
aimed to compare the efficacy of cangrelor with that of
clopidogrel in subjects requiring PCI.49 On the other hand,
the CHAMPION PLATFORM (A Clinical Trial Comparing
Treatment With Cangrelor [in Combination With Usual
Care] to Usual Care, in Subjects Who Require Percutaneous
Coronary Intervention), a trial on 6400 patients requiring
PCI, planned to test the potential superiority of cangrelor
(combined with usual care) over the placebo, as measured by
a composite of all-cause mortality, MI, and ischemia-driven
revascularization.50 Both studies have been terminated due to
insufficient evidence of cangrelor’s clinical efficacy.51
Although no Phase 3 data are available, another Phase 2
study is in progress, with the purpose of demonstrating that
patients receiving cangrelor infusion before CABG have an
acceptable safety profile and can undergo surgery without
excessive perioperative bleeding. This maintenance of
platelet inihiBition with cangRelor after dIscontinuation of
thienopyriDines in patients undergoing surGEry (BRIDGE)
trial plans to follow up 220 patients undergoing nonemergent
CABG, who had already received a thienopyridine antiplatelet
drug, and foresees completion in July 2010.52
Elinogrel (also known as PRT060128), the next addition
to the reversible, direct-acting nonthienopyridine P2Y12
receptor antagonists, is unique in that it can be administered
both orally and intravenously.53 This allows for an immedi-
ate and reversible effect of high-level platelet inhibition,
preferable in acute conditions, and an easy transition to
reversible platelet inhibition in the chronic setting.54 Its
simplified administration covers the full spectrum of care,
from acute onset to chronic care, and its reversibility allows
for surgery without significant delay. Thus far, elinogrel has
undergone Phase 2A ERASE MI (Early Rapid ReversAl of
platelet thromboSis with intravenous Elinogrel before PCI
to optimize reperfusion in acute Myocardial Infarction) pilot
trial.55 In this safety and efficacy dose-escalation trial, 70
patients with STEMI undergoing primary PCI were random-
ized to an iv bolus of placebo versus elinogrel (at doses of
10, 20, 40, and 60 mg), in addition to standard treatment,
prior to angiography. There appeared to be no difference
between the placebo and elinogrel (at doses of 10, 20, 40,
and 60 mg) with respect to the incidence of the infrequent
bleeding events. Neither were there any differences in serious
adverse events, laboratory values, and corrected thrombolysis
in MI (TIMI) frame count, nor was any ST resolution between
elinogrel and placebo.56 Although the dose-confirmation
phase was not started, as the trial was prematurely terminated
for administrative reasons, the ERASE MI findings provide
preliminary support for the use of elinogrel as an adjunctive
therapy in primary PCI for STEMI. Meanwhile, the results
of elinogrel’s Phase 2b INNOVATE-PCI (A randomized,
double-blind, active-controlled trial to evaluate intravenous
and oral PRT060128, a selective and reversible P2Y12
inhibitor, vs clopidogrel, as a novel antiplatelet therapy in
patients undergoing nonurgent PCI) are awaited. In this trial,
800 patients, who were randomized to the clopidogrel control
or to one of three experimental regimens of elinogrel (80-mg
bolus administered intravenously prior to PCI, followed by
twice-daily oral dosing of 50, 100, or 150 mg), are being
studied prior to nonurgent PCI and in the 60-day chronic
treatment phase.57 INNOVATE-PCI is designed not to exam-
ine a prespecified endpoint, but rather to explore a number
of analyses to understand the clinical efficacy, biological
activity, tolerability, and safety of elinogrel.
Two more investigational nonthienopyridine P2Y12
antagonists, BX 667 and its active metabolite BX 048, are in
the early stages of clinical development. Thus far, they have
undergone animal trials, performed on rat and dog models,
Vascular Health and Risk Management 2010:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
426
Marczewski et al
and pharmacodynamic and pharmacokinetic studies.58,59 They
are hoped to be useful in safely decreasing cardiovascular
events in patients undergoing PCI.
Besides the great variety of new ADP receptor antago-
nists, a new class of antiplatelet agents targeting the potent
thrombin-induced activation of platelets is emerging. The
first member of these thrombin receptor antagonists (TRAs)
SCH 530348 and the second member E5555 antagonize the
platelet PAR1. This receptor found in platelets and smooth
muscle cells possesses a high affinity to thrombin and
via coupling to Gp/Gi proteins, mediates the activation of
these cells by thrombin.11 The safety and efficacy of SCH
530348 have already been tested in various Phase 2 trials,
including the TRA-PCI study, and two Phase 3 trials are
presently being conducted.60 The TRA 2(°)P-TIMI 50 trial
(Thrombin Receptor Antagonist in Secondary Prevention
of Atherothrombotic Ischemic Events) on approximately
19,500 patients with atherosclerotic disease (prior MI or
stroke or with existing peripheral arterial disease), in the
course of at least 1-year follow-up, compares the safety
and efficacy of SCH 530348 against the existing standard
of care (aspirin and clopidogrel) in preventing MI, stroke,
and urgent coronary revascularization and in assessing its
bleeding risk.61 On the other hand, the TRA-CER (Thrombin
Receptor Antagonist for Clinical Event Reduction in Acute
Coronary Syndrome) study is designed to compare the
efficacy of a combination of SCH 530348 and the existing
standard of care (eg, aspirin and clopidogrel) with that of the
existing standard of care alone in preventing MI and stroke
in patients with ACS.62 By following up approximately
10,000 patients with non-STEMI for a minimum of 1 year,
the study will also assess the bleeding risk of both treatment
variants (ie, a combination of SCH 530348 and the existing
standard of care, and the existing standard of care alone). The
results of these large-scale trials, planned to be completed
in September 2010 and July 2011, respectively, are eagerly
awaited. The other member of the TRA group, E5555, is
currently being evaluated in two Phase 2 trials, LANCE-
LOT (Lessons from Antagonizing the Cellular Effects Of
Thrombin) trials 201 and 202.63,64 In each trial, approximately
600 patients with CAD will be followed up in order to assess
E5555’s safety and efficacy in inhibiting platelet aggregation
and endovascular inflammatory processes and to measure the
incidence of major adverse cardiovascular events.
There are also new up-and-coming drugs interfer-
ing with the thromboxane pathway, upon which aspirin
acts. NCX 4016 (nitroaspirin) is a nitric oxide-releasing
aspirin that combines all the benefits of aspirin with those
of nitric oxide.65 NCX 4016, with its antithrombotic,
antiatherogenic, and vasodilatory properties, is known to be
strongly cardioprotective. Also being evaluated is S18886
(terutroban), a TXA2 receptor inhibitor, currently undergo-
ing the PERFORM (Prevention of Cerebrovascular and
Cardiovascular Events of Ischemic Origin with Terutroban
in Patients with a History of Ischemic Stroke or Transient
Ischemic Attack) study in patients with recent stroke or
transient ischemic attack.12,66 This Phase 2 trial is designed to
compare this specific TP receptor antagonist with aspirin in
reducing the incidence of cerebrovascular and cardiovascular
events.67 Terutroban may be of particular interest because
studies in a murine model have shown the inhibition of the
TXA2 receptor to decrease atherosclerosis progression.68
Moreover, novel antithrombotic compounds discovered
inhibit thrombin while antagonizing GP IIb/IIIa. The future
of such dual-function pharmaceuticals in CVDs remains to be
seen. The same is true for PR-15, the investigational platelet
GP VI adhesion antagonist, and the drugs targeting platelet
endothelial aggregation receptor 1.69,70
Conclusions and future research
Given the pathologic process of intraarterial platelet activation
underlying CVDs, it is clear that antiplatelet therapeutic
options are needed for treating CVDs and preventing their
complications. Significant progress has already been made in
the field of antiplatelet therapy, as it aims to inhibit platelet
aggregation and acute thrombus formation, while preventing
intolerable bleeding. There exists a growing spectrum of
drugs modifying these pathologic processes, which continues
to expand in the hopes of overcoming the limitations of
current standard treatment. Novel platelet-inhibiting drugs
act through diverse mechanisms, mostly interfering with
ADP-induced platelet aggregation, but also targeting the
TXA2 pathway and thrombin-induced signaling.11–13 In con-
trast to the available GP IIb/IIIa antagonists, which block the
final common pathway of platelet aggregation, the possibility
emerges of disrupting initial mechanisms of adhesion by
targeting GP IV, a key receptor for collagen.69 As combina-
tion regimens are often required to achieve sufficient platelet
inhibition, new approaches to old mechanisms are explored,
such as the combination of nitric oxide and aspirin in the
investigational agent NCX 4016.65
As our understanding of the complex molecular and
genetic bases of atherothrombotic disease expands, modern
medicine moves toward more individualized patient treatment.
The growing stock of antiplatelet drugs at our disposal
can help us treat the full spectrum of and various stages
Vascular Health and Risk Management 2010:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
427
Novel mechanisms of action of antiplatelet agents
in atherothrombotic manifestations of atherosclerosis while
catering to the needs and risk factors of specific patients. This is
slowly becoming a reality, for example, prasugrel is especially
useful in patients with high risk of ischemic events as in those
with diabetes mellitus.19,28,29 Ticagrelor is possibly a useful
agent before CABG, as its more potent platelet inhibition did
not increase CABG-related bleeding, although its side effects
such as dyspnea and ventricular pauses could additionally
limit its patient base.31 On the other hand, both prasugrel
and ticagrelor seem less liable to decreased patient response
because of genetic polymorphism of receptors and enzymes;
thus, these agents potentially provide platelet inhibition in
patients who are clopidogrel nonresponders.22,38,39 Elinogrel,
even though it has not entered Phase 3 trials, is encouraging
because of its simplified administration scheme, allowing for
a smooth transition from acute to chronic treatment.
Progress with respect to the potency of platelet inhibition
has also brought increased bleeding tendency. To shift the
balance from causing excess bleeding toward preventing
vascular occlusion, dose adjustments are necessary in
low-weight patients for drugs such as prasugrel and
ticagrelor.29,31 As age is another factor found to increase bleed-
ing, elderly patients still await a registered drug to address
their needs. In contrast to the studies on clopidogrel and pra-
sugrel, ticagrelor, besides its stronger platelet inhibition, did
not increase the risk of major bleeding; thus, it may become
an antiplatelet option for older patients, although it may not be
possible for those with respiratory problems or symptomatic
bradycardia.30,31,34 Also, with regard to bleeding, the group of
thrombin receptor inhibitors is particularly promising, as it
is hypothesized that the pathway they block may play a role
only in pathologic thrombosis, leaving physiologic hemosta-
sis intact. Such an advantage with respect to bleeding risk is
also evidenced by animal studies, showing a decrease in the
progression of atherosclerosis.11,68
Dyspnea, another clinically relevant adverse effect,
was found to occur in up to 5%, 8%, 14%, and 1% of
patients receiving prasugrel, clopidogrel, ticagrelor, and
cangrelor, respectively.30,31,34,35 An analysis by Serebruany
et al71 considered the pathophysiology of the respiratory
distress and platelet inhibition. The authors found dyspnea
to be a very rare complication, mostly caused by underlying
diseases – cardiac, respiratory, or allergic – and concomitant
medications (high-dose angiotensin-converting enzyme
inhibitors) rather than by the antiplatelet therapy itself. It was
suggested that especially reversible agents (like ticagrelor)
could lead to the development of mild, asymptomatic
thrombotic thrombocytopenic purpura, fluid retention,
and dyspnea. Another proposed hypothesis is that certain
antiplatelets may be metabolized to adenosine (ticagrelor’s
molecular structure has components almost identical to those
of adenosine), which is an established bronchoprovocator.
Although the true reason for the statistically significant
increase in dyspnea – particularly with ticagrelor – remains to
be uncovered, it is clear that further studies should follow up
this finding to assess its clinical significance in the long-term
care of patients with atherothrombotic disease.71
In summary, the abundant novel antiplatelet agents, with
their higher potencies, higher platelet specificities, various
drug formulations, possible reversibility, less variability in
patient response, more rapid onsets and offsets of action,
and short time to peak, along with their attempts for better
safety, hold high potential. Some are in advanced stages of
development with assuring outcomes, and further clinical
evaluation is needed to determine which of them will find a
place within standard antiplatelet therapy.
Disclosures
The authors report no conflicts of interest. The authors alone
are responsible for the content and writing of this paper.
References
1. Jennings LK. Role of platelets in atherothrombosis. Am J Cardiol.
2009;103:4–10.
2. Awtry EA, Loscalzo J. Cardiovascular drugs – aspirin. Circulation.
2000;101:1206–1218.
3. Patrono C, Coller B, FitzGerald GA, Hirsh J, Roth G. Platelet-active
drugs: the relationships among dose, effectiveness, and side effects.
The Seventh ACCP Conference on Antithrombotic and Thrombolytic
Therapy. Chest. 2004;126 Suppl 3:S234–S264.
4. Antithrombotic Trialists’ Collaboration. Collaborative meta-analysis
of randomised trials of antiplatelet therapy for prevention of death,
myocardial infarction, and stroke in high risk patients. BMJ.
2002;324:71–86.
5. Yusuf S, Zhao F, Mehta SR, et al. Effects of clopidogrel in addition to
aspirin in patients with acute coronary syndromes without ST-segment
elevation. N Eng J Med. 2001;345:494–502.
6. Snoep JD, Hovens MM, Eikenboom JC, van der Bom JG, Jukema JW,
Huisman MV. Clopidogrel nonresponsiveness in patients undergoing
percutaneous coronary intervention with stenting: a systematic review
and meta-analysis. Am Heart J. 2007;154:221–231.
7. Gori AM, Marcucci R, Migliorini A. Incidence and clinical impact
of dual nonresponsiveness to aspirin and clopidogrel in patients with
drug-eluting stents. J Am Coll Cardiol. 2008;52:734–739.
8. Kamath S, Blannand AD, Lip GYH. Platelet activation: assessment and
quantification. Eur Heart J. 2001;22:1561–1571.
9. Shankar H, Kunapuli SP. Is the P2Y1 receptor a better target for
antithrombotic drugs? Drug Discov Today. 2005;2:285–290.
10. Quinton TM, Murugappan S, Kim S, Jin J, Kunapuli SP. Different
G protein-coupled signaling pathways are involved in alpha granule
release from human platelets. J Thromb Haemost. 2004;2:978–984.
11. Hamilton J. Protease-activated receptors as targets for antiplatelet
therapy. Blood Rev. 2009;23:61–65.
12. Kakkos SK, Nicolaides AN. S-18886 Servier. Curr Opin Investig Drugs.
2002;3:1324–1327.
Vascular Health and Risk Management 2010:6
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
428
Marczewski et al
13. Michelson AD. P2Y12 antagonism: promises and challenges. Arterioscler
Thromb Vasc Biol. 2008;28:33–38.
14. Sabatine MS. Novel antiplatelet strategies in acute coronary syndromes.
Cleve Clin J Med. 2009;76:8–14.
15. Mangin P, Ohlmann P, Eckly A, Cazenave JP, Lanza F, Gachet C.
The P2Y1 receptor plays an essential role in the platelet shape change
induced by collagen when TxA2 formation is prevented. J Thromb
Haemost. 2004;2:969–977.
16. Storey RF. Biology and pharmacology of the platelet P2Y12 receptor.
Curr Pharm Des. 2006;12:1255–1259.
17. Jin J, Quinton T, Zhang J, Rittenhouse SE, Kunapuli SP. Adenosine
diphosphate (ADP)-induced thromboxane A(2) generation in human
platelets requires coordinated signaling through integrin alpha(IIb)
beta(3) and ADP receptors. Blood. 2002;99:193–198.
18. Schomig A. Ticagrelor – is there need for a new player in the antiplatelet-
therapy field? N Engl J Med. 2009;11:1108–1111.
19. Kushner FG, Hand M, Smith SC Jr, et al. 2009 focused updates: ACC/
AHA guidelines for the management of patients with ST-elevation
myocardial infarction and ACC/AHA/SCAI guidelines on percutaneous
coronary intervention. J Am Coll Cardiol [serial on the Internet]. 2009
[cited 2009 Nov 18];54:2205-2241. http://content.onlinejacc.org/cgi/
content/full/j.jacc.2009.10.015. Accessed 2009 December 16.
20. National Institute for Health and Clinical Excellence. Prasugrel for the
treatment of acute coronary syndromes with percutaneous coronary
intervention [cited 2009 Oct]. http://www.nice.org.uk/TA182. Accessed
2009 December 16.
21. Wallentin L, Varenhorst C, James S, et al. Prasugrel achieves greater
and faster P2Y12 receptor mediated platelet inhibition than clopidogrel
due to more efficient generation of its active metabolite in aspirin-treated
patients with coronary artery disease. Eur Heart J. 2008;29:21–30.
22. Bran dt JT, Close SL, Itur ria SJ, et al. Common polymorphisms
of CYP2C19 and CYP2C9 affect the pharmacokinetic and
pharmacodynamic response to clopidogrel but not prasugrel. J Thromb
Haemost. 2007;5:2429–2436.
23. Varenhorst C, James S, Erlinge D, et al. Genetic variation of CYP2C19
affects both pharmacokinetic and pharmacodynamic responses to
clopidogrel but not prasugrel in aspirin-treated patients with coronary
artery disease. Eur Heart J. 2009;30:1744–1752.
24. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P450 genetic
polymorphisms and the response to prasugrel: relationship to
pharmacokinetic, pharmacodynamic, and clinical outcomes.
Circulation. 2009;119:2553–2560.
25. Small DS, Farid NA, Payne CD, et al. Effects of the proton pump
inhibitor lansoprazole on the pharmacokinetics and pharmacodynamics
of prasugrel and clopidogrel. J Clin Pharmacol. 2008;48:475–484.
26. Brandt JT, Payne CD, Wiviott SD, et al. A comparison of prasugrel
and clopidogrel loading doses on platelet function: magnitude of
platelet inhibition is related to active metabolite formation. Am Heart J.
2007;153:9–16.
27. Niitsu Y, Jakubowski JA, Sugidachi A, Asai F. Pharmacology of
CS-747 (prasugrel, LY640315), a novel, potent antiplatelet agent with
in vivo P2Y12 receptor antagonist activity. Semin Thromb Hemost.
2005;31:184–194.
28. Wiviott SD, Braunwald E, Angiolillo DJ, et al. Greater clinical benefit
of more intensive oral antiplatelet therapy with prasugrel in patients
with diabetes mellitus in the trial to assess improvement in therapeutic
outcomes by optimizing platelet inhibition with prasugrel thrombolysis
in myocardial infarction 38. Circulation. 2008;118:1626–1636.
29. Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus
clopidogrel in patients with acute coronary syndromes. N Engl J Med.
2007;357:2001–2015.
30. Antman EM, Wiviott SD, Murphy SA, et al. Early and late benefits
of prasugrel in patients with acute coronary syndromes undergoing
percutaneous coronary intervention: a TRITON-TIMI 38 (TRial to
Assess Improvement in Therapeutic Outcomes by Optimizing Platelet
InhibitioN with Prasugrel – Thrombolysis In Myocardial Infarction)
analysis. J Am Coll Cardiol. 2008;51:2028–2033.
31. Wallentin L, Becker RC, Budaj A, et al. Ticagrelor versus clopidogrel
in patients with acute coronary syndromes. N Engl J Med. 2009;361:
1045–1057.
32. Tantry US, Bliden KP, Gurbel PA. AZD6140. Expert Opin Investig
Drugs. 2007;16:225–229.
33. Peters GR, Butler KA, Winter HR. Multiple-dose pharmacokinetics
(PK) and pharmacodynamics (PD) of the reversible, orally active
ADP receptor antagonist AZD6140 [abstract]. Eur Heart J. 2006;27
Suppl 1:S758.
34. CAPRIE Steering Committee. A randomized, blinded, trial of clopidogrel
versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet.
1996;348:1329–1339.
35. Harrington RA, Stone GW, McNulty S, et al. Platelet inhibition with
cangrelor in patients undergoing PCI. N Engl J Med. 2009;361:
2318–2329.
36. Husted S, Emanuelsson H, Heptinstall S, Sandset PM, Wickens M,
Peters G. Pharmacodynamics, pharmacokinetics, and safety of the oral
reversible P2Y12 antagonist AZD6140 with aspirin in patients with
atherosclerosis: a double-blind comparison to clopidogrel with aspirin.
Eur Heart J. 2006;27:1038–1047.
37. Cannon CP, Husted S, Harrington RA, et al. Safety, tolerability,
and initial efficacy of AZD6140, the first reversible oral adenosine
diphosphate receptor antagonist, compared with clopidogrel, in
patients with non-ST-segment elevation acute coronary syndrome:
primary results of the DISPERSE-2 trial. J Am Coll Cardiol.
2007;50:1844–1851.
38. Gurbel PA, Bliden KP, Butler K, et al. Randomized double-blind
assessment of the ONSET and OFFSET of the antiplatelet effects of
ticagrelor versus clopidogrel in patients with stable coronary artery dis-
ease: The ONSET/OFFSET study. Circulation. 2009;120:2511–2522.
39. Storey RF, Thornton SM, Lawrance R, et al. Ticagrelor yields consistent
dose-dependent inhibition of ADP-induced platelet aggregation in
patients with atherosclerotic disease regardless of genotypic variations
in P2RY12, P2RY1, and ITGB3. Platelets. 2009;20:341–348.
40. AstraZeneca press release. Results from two phase II Trials add
to understanding of Brilinta (ticagrelor) and how it works in the
body [cited 2009 Nov 18]. http://www.astrazeneca.com/media/
latest-press-releases/2009/Brilinta_Phase_II_AHA?itemId = 7499699.
Accessed 2009 December 16.
41. AstraZeneca press release. Phase III head to head trial showed ticagrelor
reduced cardiovascular death and heart attacks over clopidogrel in
acute coronary syndromes patients [cited 2009 Aug 30]. http://www.
astrazeneca.com/media/latest-press-releases/2009/plato-esc?itemId =
6868332. Accessed 2009 December 16.
42. AstraZeneca press release. BRILINTA demonstrated greater efficacy
over clopidogrel in the most urgent clinical setting: patients with
ST-segment elevation myocardial infarction and planned percutaneous
coronary intervention (PCI) [cited 2009 Nov 16]. http://www.astrazen-
eca.com/media/latest-press releases/2009/PLATO_STEMI?itemId =
7476684. Accessed 2009 December 16.
43. Clinical Trials.gov [homepage on the Internet]. A study of the antiplatelet
effects comparing AZD6140 with clopidogrel responder and non-responders
(RESPOND) [updated 2009 Apr 23]. http://clinicaltrials.gov/ct2/show/
NCT00642811. Accessed 2009 December 16.
44. Müller I, Besta F, Schulz C, Massberg S, Schönig A, Gawaz M.
Prevalence of clopidogrel non-responders among patients with stable
angina pectoris scheduled for elective coronary stent placement. Thromb
Haemost. 2003;89:783–787.
45. Gurbel PA, Bliden KP, Antonino M, et al. Abstract 5901: the effect
of ticagrelor in stable coronary artery disease patients nonresponsive
to clopidogrel: the RESPOND study [abstract]. Circulation. 2009;120
Suppl:S1173.
46. Greenbaum AB, Grines CL, Bittl JA, et al. Initial experience with an
intravenous P2Y12 platelet receptor antagonist in patients undergoing
percutaneous coronary intervention: results from a 2-part, phase II,
multicenter, randomized, placebo- and active-controlled trial. Am
Heart J. 2006;151:689.e1–689.e10.
Vascular Health and Risk Management
Publish your work in this journal
Submit your manuscript here: http://www.dovepress.com/vascular-health-and-risk-management-journal
Vascular Health and Risk Management is an international, peer-
reviewed journal of therapeutics and risk management, focusing on
concise rapid reporting of clinical studies on the processes involved
in the maintenance of vascular health; the monitoring, prevention and
treatment of vascular disease and its sequelae; and the involvement of
metabolic disorders, particularly diabetes. This journal is indexed on
PubMed Central and MedLine. The manuscript management system
is completely online and includes a very quick and fair peer-review
system, which is all easy to use. Visit http://www.dovepress.com/
testimonials.php to read real quotes from published authors.
Vascular Health and Risk Management 2010:6 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Dovepress
429
Novel mechanisms of action of antiplatelet agents
47. Storey R, Oldroyd K, Wilcox R. Open multicentre study of the P2T
receptor antagonist AR-C69931MX assessing safety, tolerability and
activity in patients with acute coronary syndromes. Thromb Haemost.
2001;85:401–407.
48. Fugate SE, Cudd LA. Cangrelor for treatment of coronary thrombosis.
Ann Pharmacother. 2006;40:925–930.
49. Clinical Trials.gov [homepage on the Internet]. A clinical trial to
demonstrate the efficacy of cangrelor (PCI) [updated 2009 May 18].
http://clinicaltrials.gov/ct2/show/NCT00305162. Accessed 2009
December 16.
50. Clinical Trials.gov [homepage on the Internet]. Cangrelor versus
standard therapy to achieve optimal management of platelet inhibition
(PLATFORM) [updated 2009 May 18]. http://clinicaltrials.gov/ct2/
show/NCT00385138. Accessed 2009 December 16.
51. The Medicines Company press release. The medicines company
discontinues Phase 3 CHAMPION clinical trial program of cangrelor
[cited 2009 May 13]. http://ir.themedicinescompany.com/phoenix.
zhtml?c=122204&p=irol-newsArticle&ID=1287788&highlight.
Accessed 2009 December 16.
52. Clinical Trials.gov [homepage on the Internet]. Maintenance
of platelet inhibition with cangrelor (BRIDGE) [updated 2009
Dec 9]. http://clinicaltrials.gov/ct2/show/NCT00767507. Accessed
2009 December 16.
53. Gretler DD, Conley PB, Andre P, et al. “First in human” experience
with PRT060128, a new direct-acting, reversible, P2Y12 inhibitor for
IV and oral use [abstract]. J Am Coll Cardiol. 2007;9 Suppl 2:326.
54. Lieu HD, Conley PB, Andre P, et al. Initial intravenous experience
with PRT060128 (PRT128), an orally-available, direct-acting, and
reversible P2Y12 inhibitor [abstract]. J Thromb Haemost. 2007;5
Suppl 2:P-T-292.
55. Clinical Trials.gov [homepage on the Internet]. Safety and efficacy study
of adjunctive antiplatelet therapy prior to primary PCI in patients with
STEMI (ERASE-MI) [updated 2008 Jul 22]. http://clinicaltrials.gov/
ct2/show/NCT00546260. Accessed 2009 December 16.
56. Berger JS, Roe MT, Gibson CM, et al. Safety and feasibility of adjunctive
antiplatelet therapy with intravenous elinogrel, a direct-acting and
reversible P2Y12 ADP-receptor antagonist, before primary percutaneous
intervention in patients with ST-elevation myocardial infarction: the
early rapid reversal of platelet thrombosis with intravenous elinogrel
before PCI to optimize reperfusion in acute myocardial infarction
(ERASE MI) pilot trial. Am Heart J. 2009;158(6):998.e1–1004.e1.
57. Clinical Trials.gov [homepage on the Internet]. A Phase 2 safety and
efficacy study of PRT060128, a novel intravenous and oral P2Y12
inhibitor, in non-urgent PCI (INNOVATE-PCI) [updated 2009 Mar 29].
http://clinicaltrials.gov/ct2/show/NCT00751231. Accessed 2009
December 16.
58. Wang Y, Vincelette J, da Cunha V, et al. A novel P2Y(12) adenosine
diphosphate receptor antagonist that inhibits platelet aggregation
and thrombus formation in rat and dog models. Thromb Haemost.
2007;97(5):847–855.
59. Post, JM, Alexander S, Wang YX, et al. Novel P2Y12 adenosine
diphosphate receptor antagonists for inhibition of platelet aggregation
(II): pharmacodynamic and pharmacokinetic characterization. Thromb
Res. 2008;122:533–540.
60. Becker RC, Moliterno DJ, Jennings LK, et al. Safety and tolerability of
SCH 530348 in patients undergoing non-urgent percutaneous coronary
intervention: a randomised, double-blind, placebo-controlled phase II
study. Lancet. 2009;373:919–928.
61. Morrow DA, Scirica BM, Fox KA, et al; TRA 2°P-TIMI 50 Investigators.
Evaluation of a novel antiplatelet 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 trial.
Am Heart J. 2009;158:335–341.
62. Clinical Trials.gov [homepage on the Internet]. Trial to assess the
effects of SCH 530348 in preventing heart attack and stroke in patients
with acute coronary syndrome (TRA-CER) [updated 2009 Nov 23].
http://clinicaltrials.gov/ct2/show/NCT00527943. Accessed 2009
December 16.
63. Clinical Trials.gov [homepage on the Internet]. Safety and tolerability
of E5555 and its effects on markers of intravascular inflammation
in subjects with coronary artery disease [updated 2009 Sep 1].
http://clinicaltrials.gov/ct2/show/NCT00312052. Accessed 2009
December 16.
64. Clinical Trials.gov [homepage on the Internet]. Safety and tolerability of
E5555 and its effects on markers of intravascular inflammation in subjects
with acute coronary syndrome [updated 2009 Sep 2]. http://clinicaltrials.
gov/ct2/show/NCT00548587. Accessed 2009 December 16.
65. Napoli C, Aldini G, Wallace JL, et al. Efficacy and age-related effects
of nitric oxide-releasing aspirin on experimental restenosis. Proc Natl
Acad Sci U S A. 2002;99:1689–1694.
66. Bousser MG, Amarenco P, Chamorro A, et al; for PERFORM Study
Investigators. Rationale and design of a randomized, double-blind,
parallel-group study of terutroban 30 mg/day versus aspirin 100 mg/day
in stroke patients: the prevention of cerebrovascular and cardiovascular
events of ischemic origin with terutroban in patients with a history
of ischemic stroke or transient ischemic attack (PERFORM) study.
Cerebrovasc Dis. 2009;27:509–518.
67. Chamorro A. TP receptor antagonism: a new concept in atherothrombosis
and stroke prevention. Cerebrovasc Dis. 2009;27:20–27.
68. Cayatte AJ, Du Y, Oliver-Krasinski J, Lavielle G, Verbeuren TJ,
Cohen RA. The thromboxane receptor antagonist S18886 but not
aspirin inhibits atherogenesis in apo E-deficient mice: evidence that
eicosanoids other than thromboxane contribute to atherosclerosis.
Arterioscler Thromb Vasc Biol. 2000;20:1724–1728.
69. Shalito I, Kopyleva O, Serebruany V. Novel antiplatelet agents in
development: prasugrel, ticagrelor, and cangrelor and beyond. Am J
Ther. 2009;16:451–458.
70. Nanda N, Bao M, Lin H, et al. Platelet endothelial aggregation
receptor 1 (PEAR1), a novel epidermal growth factor repeat-containing
transmembrane receptor, participates in platelet contact-induced
activation. J Biol Chem. 2005;280:24680–24689.
71. Serebruany VL, Stebbing J, Atar D. Dyspnoea after antiplatelet agents:
the AZD6140 controversy. Int J Clin Pract. 2007;61:529–533.
... El tratamiento del SCA se fundamenta en reestablecer la circulación coronaria, ya sea por medios mecánicos (intervención coronaria percutánea (ICP), derivación coronaria) o farmacológicos (trombolisis), así como en impactar sobre las diversas vías fisiopatológicas implicadas en la génesis y progresión del SCA; la terapia dual antiagregante plaquetaria (ácido acetil salicílico sumado a un inhibidor del P2Y12, siendo el clopidogrel el más usado) es el pilar del tratamiento, tanto en la fase aguda como en la crónica (33,(47)(48)(49). ...
... La resistencia a los antiagregantes plaquetarios puede determinarse a nivel bioquímico a través de la prueba por transmisión de luz (ATL), o a nivel clínico cuando los pacientes presentan recurrencia de un evento coronario (48). Esta resistencia puede ser explicada por factores genéticos, epigenéticos y clínicos como la falta de adherencia al tratamiento, dosificación inapropiada, interacciones farmacológicas, comorbilidades asociadas (33), variantes en genes que codifican para las proteínas involucradas en la farmacocinética y farmacodinamia de los antiagregantes (48,(51)(52)(53)(54)(55)(56)(57), entre otras. ...
... La resistencia a los antiagregantes plaquetarios puede determinarse a nivel bioquímico a través de la prueba por transmisión de luz (ATL), o a nivel clínico cuando los pacientes presentan recurrencia de un evento coronario (48). Esta resistencia puede ser explicada por factores genéticos, epigenéticos y clínicos como la falta de adherencia al tratamiento, dosificación inapropiada, interacciones farmacológicas, comorbilidades asociadas (33), variantes en genes que codifican para las proteínas involucradas en la farmacocinética y farmacodinamia de los antiagregantes (48,(51)(52)(53)(54)(55)(56)(57), entre otras. ...
Article
Effect of hyperglycemia on acute coronary syndrome and the implications of antiplatelet therapy According to data from the World Health Organization, cardiovascular disease is the main cause of death associated with risk factors such as smoking, sedentary lifestyle, hypertension, dyslipidemia and diabetes mellitus. Precisely, this last disease is one of the most related to the appearance, progression, and complications of a coronary event. Hyperglycemia potentiates different biochemical and cellular pathways such as sorbitol, the nuclear factor kβ, the formation of advanced glycation end products, the protein kinase C pathway and oxidative stress, which end up favoring in the coronary patient a proinflammatory state and procoagulant, which is associated with a worse prognosis and aggravates myocardial injury; in addition, it inhibits and competes with the action of platelet antiaggregants, generating resistance not only to these but also to thrombolytic therapy. Therefore, it is necessary to generate an update of the topic, to sensitize the medical community about the importance of glycemic control, especially in patients with ischemic heart disease and thus improve control strategies. The bibliographic search was carried out in PubMed, in a structured, non-systematic way. Articles published in English and Spanish were included, without restriction by publication date.
... El tratamiento del SCA se fundamenta en reestablecer la circulación coronaria, ya sea por medios mecánicos (intervención coronaria percutánea (ICP), derivación coronaria) o farmacológicos (trombolisis), así como en impactar sobre las diversas vías fisiopatológicas implicadas en la génesis y progresión del SCA; la terapia dual antiagregante plaquetaria (ácido acetil salicílico sumado a un inhibidor del P2Y12, siendo el clopidogrel el más usado) es el pilar del tratamiento, tanto en la fase aguda como en la crónica (33,(47)(48)(49). ...
... La resistencia a los antiagregantes plaquetarios puede determinarse a nivel bioquímico a través de la prueba por transmisión de luz (ATL), o a nivel clínico cuando los pacientes presentan recurrencia de un evento coronario (48). Esta resistencia puede ser explicada por factores genéticos, epigenéticos y clínicos como la falta de adherencia al tratamiento, dosificación inapropiada, interacciones farmacológicas, comorbilidades asociadas (33), variantes en genes que codifican para las proteínas involucradas en la farmacocinética y farmacodinamia de los antiagregantes (48,(51)(52)(53)(54)(55)(56)(57), entre otras. ...
... La resistencia a los antiagregantes plaquetarios puede determinarse a nivel bioquímico a través de la prueba por transmisión de luz (ATL), o a nivel clínico cuando los pacientes presentan recurrencia de un evento coronario (48). Esta resistencia puede ser explicada por factores genéticos, epigenéticos y clínicos como la falta de adherencia al tratamiento, dosificación inapropiada, interacciones farmacológicas, comorbilidades asociadas (33), variantes en genes que codifican para las proteínas involucradas en la farmacocinética y farmacodinamia de los antiagregantes (48,(51)(52)(53)(54)(55)(56)(57), entre otras. ...
Article
Full-text available
La enfermedad cardiovascular representa, según los datos de la Organización Mundial de la Salud, la principal causa de muerte asociada con factores de riesgo como el tabaquismo, el sedentarismo, la hipertensión, la dislipidemia y la diabetes mellitus. Precisamente, esta última enfermedad es una de las que más se relaciona con la aparición, la progresión y las complicaciones de un evento coronario. La hiperglucemia potencia diferentes vías bioquímicas y celulares como la del sorbitol, el factor nuclear kβ, la formación de productos finales de glicación avanzada, la vía de la proteína cinasa C y el estrés oxidativo, que terminan favoreciendo en el paciente coronario un estado proinflamatorio y procoagulante, que se asocia con un peor pronóstico y agrava la lesión miocárdica; además, inhibe y compite con la acción de los antiagregantes plaquetarios, generando resistencia no solo a estos sino también a la terapia trombolítica. Por lo anterior, se hace necesario generar una actualización del tema, para sensibilizar a la comunidad médica sobre la importancia del control glucémico, sobre todo en acientes con cardiopatía isquémica, y así mejorar las estrategias de control. Se realizó la búsqueda bibliográfica en PubMed, de una forma estructurada, no sistemática. Se incluyeron artículos publicados en inglés y español, sin restricción por fecha de publicación.
... In addition, recent randomized clinical trials and the last Guidelines for stroke of AHA/ ASA, in 2018, also mention ticagrelor [8]. All of these antiplatelet therapies, besides inhibiting acute arterial thrombosis, also interfere with physiological hemostasis [9]. ...
Article
Full-text available
Platelets play an essential role in atherothrombosis and for this reason they are the primary target of antithrombotic therapy in ischemic stroke. We discussed here the evidence for efficacy and safety of current knowledge in antiplatelet therapy for stroke prevention after an acute ischemic stroke or transient ischemic attack. After an acute episode, long-term antithrombotic therapy is essential for the secondary prevention of stroke recurrence and complications. Antiplatelet therapy for acute ischemic stroke (non-cardioembolic) or ischemic stroke consists of three antiplatelet drugs, in accordance with Food and Drug Administration (FDA) from the USA and also with the Guidelines published by the American Heart Association (AHA) and nevertheless with the Guidelines of the American Stroke Association (ASA), in 2014, for preventing vascular events, such as stroke. These are aspirin, clopidogrel and dipyridamole. Moreover, recent randomized clinical trials and the last Guidelines for stroke of AHA∕ASA, in 2018, also mention ticagrelor. All of these antiplatelet therapies, besides inhibiting acute arterial thrombosis, also interfere with physiological hemostasis. In conclusion, we can say that current recommendations focused primarily on the therapy with aspirin for the secondary prevention of stroke in patients that presented vascular events, such as ischemic stroke of non-cardioembolic cause or transient ischemic attack and, as appropriate, aspirin plus dipyridamol or clopidogrel. The new therapy with ticagrelor in secondary stroke prevention seems to be promising, but more randomized clinical trials are needed to accurately assess the safety and efficacy of this new antiplatelet drug.
... Bleeding complications may occur, especially in patients who are concurrently treated with platelet inhibitors that target other pathways such as Ticagrelor and Clopidogrel, targets of the ADP receptor, P2Y12. GPVI-mediated platelet activation is highly dependent on secondary activation via ADP and suppression of both pathways may have important consequences in terms of bleeding [39][40][41]. ...
Article
Full-text available
Involvement of signal transducer and activator of transcription 3 (STAT3) in inflammation is well known. Recently, a role for STAT3 in platelet activation and platelet production has been suggested. Platelets exhibit important immune functions and engagement of STAT3 in platelet physiology may link inflammation and hemostasis. This study investigated the effects of STAT3 loss-of-function mutations and single nucleotide polymorphisms (SNPs) in STAT3 on glycoprotein VI (GPVI)-mediated platelet activation and platelet numbers in humans. Two cohorts were studied. The first cohort concerned patients with STAT3 loss-of-function mutations. Platelet numbers were investigated in eight patients and GPVI-mediated platelet activation was functionally tested in four patients. Additional experiments were performed to investigate underlying mechanisms. The second cohort concerned 334 healthy volunteers and investigated the consequences of SNPs in STAT3 on GPVI-mediated platelet activation and platelet numbers. Platelet activation was lower in STAT3 loss-of-function patients at baseline and after stimulation of the GPVI receptor, reflected by decreased P-selectin expression. This was independent of gene transcription. Blockade of the adenosine di-phosphate (ADP) pathway resulted in a further decrease of P-selectin expression, particularly in STAT3 loss-of-function patients. In contrast, the SNPs in STAT3 did not influence GPVI-mediated platelet activation. Also, platelet numbers were not affected by STAT3 loss-of-function mutations, nor was there an association with the SNPs. In conclusion, STAT3 signaling does not seem to play a major role in thrombopoiesis. We confirm that STAT3 is involved in GPVI-mediated platelet activation in humans, independent of gene transcription. GPVI-mediated platelet activation is highly dependent on secondary ADP release. Our findings suggest that STAT3 modulation may affect inflammation, hemostasis, and their interaction.
... 22 Ticagleror also reduced the relative risk of stent thrombosis among the patients who received a stent during the study by 38%. 23 In a retrospective analysis of the PLATO study, no significant differences were found between the Asian versus the non-Asian populations with respect to efficacy or bleeding rates. 24 Detailed bleeding rates have been analyzed in a post hoc analysis of the PLATO trial. ...
... While ASA reduces thrombotic events, stroke, and myocardial infarction by approximately 20% to 25% through decreased platelet activation, this drug results in only partial inhibition of platelet aggregation ( Figure 5). 8 In cases of coronary intervention with stent implantation, the additional administration of clopidogrel or newer P2Y12 antagonists decreases the risk for mature adverse cardiac events by significantly reducing the incidence of possibly fatal stent thrombosis. 9 Until the introduction of the 2 new-generation platelet inhibitors prasugrel and ticagrelor in 2009 and 2010, 2 classes of antiplatelet drugs were used along with ASA. ...
Article
Purpose To evaluate the prevalence of dual platelet inhibition in cases of severe retrobulbar hemorrhage following retrobulbar and peribulbar anesthesia. Setting Department of Ophthalmology, Ludwig-Maximilans Universität, München, Germany. Design Retrospective study. Methods Two groups of patients were screened retrospectively over a 5-year period for the inclusion criterion of severe retrobulbar hematoma after retrobulbar or parabulbar injection. The first group consisted of emergency cases referred to the clinic. A second group of patients had received retrobulbar block at the hospital. All cases were collected and screened for the presence of antiplatelet therapy. Results Among roughly 160 000 patient records screened, 3 patients with grade IV retrobulbar hematoma were identified. Two of these patients were taking dual antiplatelet medications and 2 were on anticoagulation therapy during the time of retrobulbar or peribulbar anesthesia. None of the cases showed single medication platelet inhibition. The visual acuity of all patients stayed low at the 6-month follow-up (1.2 logMAR in 1 patient and no light perception in 2 patients). Conclusions Retrobulbar hematoma is a rare but severe complication of retrobulbar anesthesia. With the high prevalence of dual platelet inhibition found in these cases, a prospective controlled trial seems unethical. In these high-risk patients, surgery should be performed under topical anesthesia if possible or general anesthesia if necessary. Financial Disclosure No author has a financial or proprietary interest in any material or method mentioned.
... Activation of platelets and their subsequent aggregation play a key role in thrombus formation at the site of vascular injury and atherothrombotic events [1,2]. Assessment of platelet activity and proper medical control are therefore mandatory for high-risk patients [3]; however, platelet aggregation after applying inducers, such as adenosine diphosphate (ADP) or 5-hydroxytryptamine, and collagen, is, in general, not measured in routine laboratory testing. ...
Article
Full-text available
Mean platelet volume (MPV) and platelet distribution width (PDW) are indices that reflect platelet activity. We investigated the association between these platelet indices and left ventricular hypertrophy and cardiac function. We analyzed the data of 1241 patients who were admitted to the Cardiology Department. Both MPV and PDW were selected as independent factors associated with left ventricular systolic and diastolic dysfunction, and left ventricular hypertrophy. The highest tertile of MPV and PDW was associated with left ventricular systolic dysfunction (left ventricular ejection fraction of <50 %) with an odds ratio of 1.53 and 2.03, respectively, when the respective lowest tertile was used as reference. The highest PDW tertile was associated with left ventricular hypertrophy with an odds ratio of 1.56 (95 % CI, 1.13-2.15) and with dysfunction with an odds ratio of 3.34 (95 % CI, 1.54-7.25). Indices of platelet activation (MPV and/or PDW) were independently associated positively with left ventricular hypertrophy and left ventricular systolic and diastolic dysfunction. Whether these platelet indices represent useful markers for identifying individuals at higher risk for thromboembolic disease and organ damage among cardiac patients awaits further investigation.
Article
Full-text available
BACKGROUND: Ticagrelor is an oral, reversible, direct-acting inhibitor of the adenosine diphosphate receptor P2Y12 that has a more rapid onset and more pronounced platelet inhibition than clopidogrel. METHODS: In this multicenter, double-blind, randomized trial, we compared ticagrelor (180-mg loading dose, 90 mg twice daily thereafter) and clopidogrel (300-to-600-mg loading dose, 75 mg daily thereafter) for the prevention of cardiovascular events in 18,624 patients admitted to the hospital with an acute coronary syndrome, with or without ST-segment elevation. RESULTS: At 12 months, the primary end point--a composite of death from vascular causes, myocardial infarction, or stroke--had occurred in 9.8% of patients receiving ticagrelor as compared with 11.7% of those receiving clopidogrel (hazard ratio, 0.84; 95% confidence interval [CI], 0.77 to 0.92; P<0.001). Predefined hierarchical testing of secondary end points showed significant differences in the rates of other composite end points, as well as myocardial infarction alone (5.8% in the ticagrelor group vs. 6.9% in the clopidogrel group, P=0.005) and death from vascular causes (4.0% vs. 5.1%, P=0.001) but not stroke alone (1.5% vs. 1.3%, P=0.22). The rate of death from any cause was also reduced with ticagrelor (4.5%, vs. 5.9% with clopidogrel; P<0.001). No significant difference in the rates of major bleeding was found between the ticagrelor and clopidogrel groups (11.6% and 11.2%, respectively; P=0.43), but ticagrelor was associated with a higher rate of major bleeding not related to coronary-artery bypass grafting (4.5% vs. 3.8%, P=0.03), including more instances of fatal intracranial bleeding and fewer of fatal bleeding of other types. CONCLUSIONS: In patients who have an acute coronary syndrome with or without ST-segment elevation, treatment with ticagrelor as compared with clopidogrel significantly reduced the rate of death from vascular causes, myocardial infarction, or stroke without an increase in the rate of overall major bleeding but with an increase in the rate of non-procedure-related bleeding. (ClinicalTrials.gov number, NCT00391872.)
Article
OBJECTIVE: To determine the effects of antiplatelet therapy among patients at high risk of occlusive vascular events. DESIGN: Collaborative meta-analyses (systematic overviews). INCLUSION CRITERIA: Randomised trials of an antiplatelet regimen versus control or of one antiplatelet regimen versus another in high risk patients (with acute or previous vascular disease or some other predisposing condition) from which results were available before September 1997. Trials had to use a method of randomisation that precluded prior knowledge of the next treatment to be allocated and comparisons had to be unconfounded-that is, have study groups that differed only in terms of antiplatelet regimen. STUDIES REVIEWED: 287 studies involving 135 000 patients in comparisons of antiplatelet therapy versus control and 77 000 in comparisons of different antiplatelet regimens. MAIN OUTCOME MEASURE: "Serious vascular event": non-fatal myocardial infarction, non-fatal stroke, or vascular death. RESULTS: Overall, among these high risk patients, allocation to antiplatelet therapy reduced the combined outcome of any serious vascular event by about one quarter; non-fatal myocardial infarction was reduced by one third, non-fatal stroke by one quarter, and vascular mortality by one sixth (with no apparent adverse effect on other deaths). Absolute reductions in the risk of having a serious vascular event were 36 (SE 5) per 1000 treated for two years among patients with previous myocardial infarction; 38 (5) per 1000 patients treated for one month among patients with acute myocardial infarction; 36 (6) per 1000 treated for two years among those with previous stroke or transient ischaemic attack; 9 (3) per 1000 treated for three weeks among those with acute stroke; and 22 (3) per 1000 treated for two years among other high risk patients (with separately significant results for those with stable angina (P=0.0005), peripheral arterial disease (P=0.004), and atrial fibrillation (P=0.01)). In each of these high risk categories, the absolute benefits substantially outweighed the absolute risks of major extracranial bleeding. Aspirin was the most widely studied antiplatelet drug, with doses of 75-150 mg daily at least as effective as higher daily doses. The effects of doses lower than 75 mg daily were less certain. Clopidogrel reduced serious vascular events by 10% (4%) compared with aspirin, which was similar to the 12% (7%) reduction observed with its analogue ticlopidine. Addition of dipyridamole to aspirin produced no significant further reduction in vascular events compared with aspirin alone. Among patients at high risk of immediate coronary occlusion, short term addition of an intravenous glycoprotein IIb/IIIa antagonist to aspirin prevented a further 20 (4) vascular events per 1000 (P<0.0001) but caused 23 major (but rarely fatal) extracranial bleeds per 1000. CONCLUSIONS: Aspirin (or another oral antiplatelet drug) is protective in most types of patient at increased risk of occlusive vascular events, including those with an acute myocardial infarction or ischaemic stroke, unstable or stable angina, previous myocardial infarction, stroke or cerebral ischaemia, peripheral arterial disease, or atrial fibrillation. Low dose aspirin (75-150 mg daily) is an effective antiplatelet regimen for long term use, but in acute settings an initial loading dose of at least 150 mg aspirin may be required. Adding a second antiplatelet drug to aspirin may produce additional benefits in some clinical circumstances, but more research into this strategy is needed.
Article
Background: Many clinical trials have evaluated the benefit of long-term use of antiplatelet drugs in reducing the risk of clinical thrombotic events. Aspirin and ticlopidine have been shown to be effective, but both have potentially serious adverse effects. Clopidogrel, a new thienopyridine derivative similar to ticlopidine, is an inhibitor of platelet aggregation induced by adenosine diphosphate. Methods: CAPRIE was a randomised, blinded, international trial designed to assess the relative efficacy of clopidogrel (75 mg once daily) and aspirin (325 mg once daily) in reducing the risk of a composite outcome cluster of ischaemic stroke, myocardial infarction, or vascular death; their relative safety was also assessed. The population studied comprised subgroups of patients with atherosclerotic vascular disease manifested as either recent ischaemic stroke, recent myocardial infarction, or symptomatic peripheral arterial disease. Patients were followed for 1 to 3 years. Findings: 19,185 patients, with more than 6300 in each of the clinical subgroups, were recruited over 3 years, with a mean follow-up of 1.91 years. There were 1960 first events included in the outcome cluster on which an intention-to-treat analysis showed that patients treated with clopidogrel had an annual 5.32% risk of ischaemic stroke, myocardial infarction, or vascular death compared with 5.83% with aspirin. These rates reflect a statistically significant (p = 0.043) relative-risk reduction of 8.7% in favour of clopidogrel (95% Cl 0.3-16.5). Corresponding on-treatment analysis yielded a relative-risk reduction of 9.4%. There were no major differences in terms of safety. Reported adverse experiences in the clopidogrel and aspirin groups judged to be severe included rash (0.26% vs 0.10%), diarrhoea (0.23% vs 0.11%), upper gastrointestinal discomfort (0.97% vs 1.22%), intracranial haemorrhage (0.33% vs 0.47%), and gastrointestinal haemorrhage (0.52% vs 0.72%), respectively. There were ten (0.10%) patients in the clopidogrel group with significant reductions in neutrophils (< 1.2 x 10(9)/L) and 16 (0.17%) in the aspirin group. Interpretation: Long-term administration of clopidogrel to patients with atherosclerotic vascular disease is more effective than aspirin in reducing the combined risk of ischaemic stroke, myocardial infarction, or vascular death. The overall safety profile of clopidogrel is at least as good as that of medium-dose aspirin.