Anda di halaman 1dari 19

REVIEWS

Antithrombotic therapy for patients


with STEMI undergoing primary PCI
Francesco Franchi, Fabiana Rollini and Dominick J. Angiolillo
Abstract | Antithrombotic therapy, including antiplatelet and anticoagulant agents, is the
cornerstone of pharmacological treatment to optimize clinical outcomes in patients with
ST‑segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary
intervention (PPCI). Intravenous anticoagulant drugs available for PPCI include the indirect
thrombin inhibitors unfractionated heparin and low-molecular-weight heparin, and the
direct thrombin inhibitor bivalirudin. Intravenous antiplatelet drugs mainly include
glycoprotein IIb/IIIa inhibitors and the P2Y12-receptor inhibitor cangrelor. Dual antiplatelet
therapy with aspirin and an oral P2Y12-receptor inhibitor is pivotal for the acute and long-term
treatment of patients with STEMI undergoing PPCI. Prasugrel and ticagrelor provide a more
prompt, potent, and predictable antiplatelet effect compared with clopidogrel, which translates
into better clinical outcomes. Therefore, these agents are the first-line treatment in PPCI. However,
patients can still experience adverse ischaemic events, which might be in part attributed to
alternative pathways triggering thrombosis. Thrombin has an important role, suggesting the need
for strategies directly targeting circulating thrombin or other factors of the coagulation cascade,
such as oral anticoagulants (rivaroxaban), and the thrombin receptor on the platelet membrane
(vorapaxar). In this Review, we provide an overview of currently available antithrombotic therapies
used in patients with STEMI undergoing PPCI, results from pivotal clinical trials and their
implications for guidelines, as well as recommendations for clinical practice.

Acute ST‑segment elevation myocardial infarction (platelets) and plasma (coagulation) factors7–9. Therefore,
(STEMI) is a major cause of morbidity, mortality, and dis‑ antithrombotic therapy, including antiplatelet and anti‑
ability worldwide. Currently, STEMI comprises 25–40% coagulant agents, is the cornerstone of pharmacological
of myocardial infarction (MI) presentations1–4. The goal of treatment to optimize clinical outcomes in patients with
STEMI treatment is reperfusion, aimed to achieve an early STEMI undergoing PPCI1–4,9. In this Review, we provide
and complete recanalization of the infarct-related artery. an overview of currently available antithrombotic thera­
Indeed, timely reperfusion with primary percutaneous pies used in the setting of patients with STEMI under‑
coronary intervention (PPCI) is considered the treat‑ going PPCI, including describing the rationale for use,
ment of choice in this setting1–4. Over the past 30 years, pharmacological characteristics, pivotal clinical trial
the manage­ment of patients with STEMI has evolved data, and guidance for selecting the appropriate regimen
substantially in terms of reperfusion strategies, adjunc‑ in the early phase of management, primarily focusing on
tive antithrombotic therapy, technical approaches, and adjunctive therapies for PPCI.
University of Florida College development of coordinated systems of care, which have
of Medicine-Jacksonville, led to marked improvements in short-term and long-term Rationale for antithrombotic therapy in STEMI
Division of Cardiology-ACC outcomes1–4. Both in‑hospital and 1‑year mortality from Injury to the arterial vessel wall exposes the subendo­
Building 5th floor, 655 West
STEMI (5–6% and 7–18%, respectively) have decreased thelial layer and leads to recruitment and activation of
8th Street, Jacksonville,
Florida 32209, USA. significantly, but cardiovascular outcomes could still platelets, as well as generation of excessive levels of throm‑
Correspondence to D.J.A.
be improved1–4. bin. Following adhesion, platelets release many activat‑
dominick.angiolillo@ Atherosclerotic plaque rupture or erosion followed ing factors, mainly thromboxane A2, ADP, and thrombin,
jax.ufl.edu by arterial thrombosis and formation of an occlusive which activate intraplatelet signalling pathways leading to
doi:10.1038/nrcardio.2017.18 thrombus is the major determinant leading to STEMI5–7. enhanced interactions among adherent platelets and fur‑
Published online 23 Feb 2017 The main elements involved in this process are cellular ther recruitment and activation of circulating platelets7–9.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points unfractionated heparin (UFH) and low-molecular-­weight


heparin (LMWH); the direct thrombin inhibitor bivali‑
• Antithrombotic therapy is the cornerstone of pharmacological treatment to optimize rudin; and the direct factor Xa inhibitor fondaparinux.
clinical outcomes in patients with ST‑segment elevation myocardial infarction (STEMI) These agents have different mechanisms of action, bind‑
undergoing primary percutaneous coronary intervention (PPCI) ing specificity, pharmaco­kinetic and pharmacodynamic
• Anticoagulant therapy is mandatory in PPCI and includes heparins and bivalirudin, consistency, risks of heparin-induced thrombocytopenia
which have different pharmacological and clinical profiles; the choice between these (HIT), effects on platelet function, and half-lives (TABLE 1).
agents is mainly on the basis of bleeding risk profile
Because of the high rate of catheter thrombosis recorded
• Intravenous antiplatelet drugs mainly include glycoprotein IIb/IIIa inhibitors and the in the OASIS‑6 trial14, fondaparinux is not recommended
P2Y12-receptor inhibitor cangrelor, which can provide immediate platelet inhibition
as background a­ nticoagulant for PPCI and, therefore,
until the full antiplatelet effect of oral agents is achieved
is not discussed1–4.
• Dual antiplatelet therapy with a combination of aspirin and one of the new-generation
P2Y12-receptor inhibitors prasugrel or ticagrelor is the recommended first-line
treatment in patients undergoing PPCI
Unfractionated heparin. UFH is a sulfated polysacchar­
ide with a molecular weight range of 3,000–30,000 kDa
• In the absence of data supporting superiority of one agent over the other, the choice
between prasugrel and ticagrelor should take into consideration contraindications
(mean 15,000 kDa)15. Its major anticoagulant effect is
and patient characteristics; clopidogrel should be used when both prasugrel and based on inactivation of thrombin and activated factor X
ticagrelor are contraindicated or not available (factor Xa) through an antithrombin-­dependent mech‑
• Agents targeting thrombin-mediated effects on platelet activation, namely vorapaxar anism. The heparin–antithrombin complex inactiv­ates
and rivaroxaban, have been developed and can be used in selected patients with not only thrombin but also factors Xa, IXa, XIa, and
STEMI to reduce long-term atherothrombotic events XIIa. Major limitations of UFH use include its vari­able
pharmacokinetic profile with high intrapatient and
inter­patient variability, and the increased risk of HIT15.
The final step for all these signalling pathways is the HIT is a serious, potentially lethal, immuno­logically
conversion of the platelet glycoprotein IIb/IIIa receptor mediated adverse reaction to UFH and, less commonly,
into its active form, which binds to soluble adhesive sub‑ to LMWH, which occurs in 0.1–5.0% of patients receiv‑
strates, including fibrinogen and von Willebrand factor, ing heparin therapy, with variability according to patient
and leads to platelet aggregation and formation of an population and type of heparin exposure. Up to 50% of
occlusive thrombus mediated by platelet–fibrin inter­ patients with HIT develop thrombotic complications,
action7–9. Vascular injury also exposes subendothelial and mortality is from 5–10% up to 20–30% in those
tissue factor, which activates the coagulation cascade who develop thrombotic complications. The treatment
leading to thrombin generation7–9. Thrombin converts for this condition includes termination of heparin ther‑
fibrinogen to fibrin, an essential component of arterial apy (both UFH and LMWH), use of alternative anti‑
thrombus, and is one of the most potent platelet activ­ coagulants (the direct thrombin inhibitors bivalirudin
ators by binding to protease-activated receptors (PARs) or argatroban) to prevent thrombotic complications,
on the platelet membrane10–13. Notably, only a modest and platelet transfusion in severely thrombocytopenic
amount of thrombin is produced as a result of the coagu­ patients with bleeding16.
lation cascade, whereas the surface of activated platelets is Although no placebo-controlled trials have evalu‑
the main source of c­ irculating thrombin10–13. ated the use of UFH in PPCI, intravenous UFH titrated
Several classes of anticoagulant and antiplatelet to target-recommended activated clotting times is a
agents targeting mediators and receptors promoting famili­ar and extensively practised strategy for anti‑
thrombosis are currently approved for the early manage‑ coagulant therapy at the time of PCI for STEMI1–4.
ment of patients with acute STEMI, and are available for Dosage should follow standard recommendations for
either intravenous or oral administration1–4 (FIG. 1). The PCI, t­ aking into account the use of glycoprotein IIb/IIIa
following sections provide details on the pharmacology inhibitors (GPIs; 70–100 U/kg bolus with no GPI use
and clinical trial development of these different classes planned, and 50–70 U/kg bolus with GPI use planned)
of antithrombotic agents. to achieve therapeutic activated clotting times
(250–300 s without GPIs use, and 200–250 s with GPI
Intravenous antithrombotic therapies use)1–4. UFH ­currently has a class I indication for anti‑
The acute phase of STEMI is characterized by a high coagulation ­during PPCI in both the ACC/AHA and
prothrombotic milieu and an increased risk of throm‑ ESC guidelines1–4.
botic complications, which, in addition to the need to
reduce the time from hospital admission or first med‑ Low-molecular-weight heparin. LMWHs are derived
ical contact to coronary angiography, underscores the from UFH by chemical or enzymatic depolymerization
need for fast-acting, intravenous antithrombotic thera‑ and are about one-third the molecular weight of UFH
pies providing effective blockade of thrombin-mediated (mean molecular weight 4,000–5,000 kDa)15. The lower
effects and important platelet signalling pathways1–9. molecular weight compared with UFH translates into
a reduced capacity to bind to protein and cells and to a
Anticoagulant therapy more predictable dose–response profile, a longer plasma
Anticoagulant therapy is mandatory in PPCI. Anticoagu­ half-life, and a lower incidence of HIT15. Like UFH,
lant drugs available for parenteral use are ­indirect throm‑ LMWHs produce their anticoagulant effect by enhanc‑
bin (factor IIa) and factor Xa inhib­itors, which include ing antithrombin activity. Coagulation monitoring is

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

generally not needed, and dosing is weight-adjusted15. with STEMI were randomly assigned to receive an
The major limitation of LMWHs is that they have pre‑ intra­venous bolus of 0.5 mg/kg enoxaparin (followed
dominantly renal clearance, and their use is generally by an additional 0.25 mg/kg bolus in the event of a
not recommended in patients with creatinine clearance prolonged procedure or thrombotic complications)
of <30 ml/min, owing to increased risk of bleeding17,18. or UFH before PPCI19. Patients who received any anti‑
Different LMWHs are approved for use in Canada, coagulant before randomization and those requiring
Europe, and the USA. However, the most extensively crossover from one drug to the other were excluded.
tested in the setting of PCI is enoxaparin, which was Enoxaparin did not significantly reduce the incidence
also compared with UFH in patients undergoing PPCI of the primary end point (a composite of death, com‑
in the ATOLL trial19. In this trial, patients (n = 910) plication of MI, procedure failure, or major bleeding at

Endothelium
Necrotic core

Tissue Blood stream


factor

Plaque
rupture

Heparin

Activated Nonactivated platelets


XII XIIa platelets
• Rivaroxaban
XI XIa • Heparin
• Clopidogrel
IX IXa VIIa VII • Prasugrel
• Ticagrelor
Tissue ADP • Cangrelor
VIIIa factor • Bivalirudin TXA2
• Heparin Vorapaxar
X Xa X
P2Y12 TP Aspirin
Va receptor receptor
Thrombin COX1
Prothrombin (II) Thrombin (IIa) Arachidonic
PAR1 acid
receptor
Fibrinogen (I) Fibrin (Ia)
V
GP (IIb/IIIa) Platelet
receptor
Fibrinogen (I)

vWF vWF receptor


• Abciximab
Collagen Collagen receptor • Eptifibatide
Fibrin • Tirofiban

Figure 1 | Mechanism of thrombus formation during ST‑segment elevation myocardial infarction, and targets of
Nature Reviews | Cardiology
currently available antithrombotic agents. After plaque rupture, adhesion of platelets to the subendothelium during
the rolling phase is mediated by the interaction between the glycoprotein (GP) Ib/V/IX receptor complex located on the
platelet surface and von Willebrand factor (vWF), and between collagen exposed at the site of vascular injury and platelet
collagen receptors. Binding of collagen to these receptors triggers intracellular mechanisms that induce the release of
activating factors, mainly thromboxane A2 (TXA2), ADP, and thrombin. These factors enhance the interactions among
adherent platelets and promote further recruitment and activation of circulating platelets. Platelet activation by these
mediators has as the final pathway the conversion of the platelet GP IIb/IIIa receptor, the main receptor mediating platelet
aggregation, into its active form. Activated GP IIb/IIIa receptors bind to fibrinogen and vWF, leading to platelet aggregation
and thrombus formation mediated by platelet–platelet interactions. Vascular injury also exposes subendothelial tissue
factor, which forms a complex with factor VIIa and sets off a chain of events that culminates in formation of the
prothrombinase complex. Prothrombin is converted to thrombin, which subsequently converts fibrinogen to fibrin,
generating a fibrin-rich clot, and further activates platelets by binding to protease-activated receptors (PAR1) on the
platelet membrane. However, only a modest amount of thrombin is produced as a result of the coagulation cascade, and
the surface of activated platelets is the main source of circulating thrombin. Antiplatelet and anticoagulant agents work by
inhibiting key receptors and factors involved in this cascade of events. COX, cyclooxygenase; TP, thromboxane prostanoid.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Pharmacological properties of parenteral anticoagulants and indication in PPCI


Property Unfractionated Enoxaparin Fondaparinux Bivalirudin
heparin
Administration route Intravenous Intravenous, Subcutaneous Intravenous
subcutaneous
Factor Xa:IIa inhibition 1:1 3–4:1 Only Xa Only IIa
Action independent of No No No Yes
antithrombin
Nonspecific binding Yes Partial No No
Variable PK/PD measures Yes Yes (<unfractionated No No
heparin)
Inhibits fibrin-bound thrombin No No No Yes
Effect on platelets Activation Activation Activation Inhibition
Half-life ~60 min 90–120 min* 17 h 25 min
Risk of HIT Yes Yes Low No
(<unfractionated
heparin)
Indication in PPCI Class I Class IIa‡ Class III ACC/AHA: class I;
ESC: class IIa
Dose in PPCI 70–100 U/kg bolus 0.5 mg/kg NA 0.75 mg/kg
without GPIs§; 50–70 U/ intravenous bolus intravenous bolus;
kg bolus with GPIs§ 1.75 mg/kg/h infusion
Reversal agent Protamine sulfate No Recombinant No
factor VIIa
*After intravenous administration of 0.5 mg/kg. ‡Only in European guidelines; not recommended in US guidelines. §Further boluses
needed to maintain therapeutic activated clotting time. GPI, glycoprotein IIb/IIIa inhibitor; HIT, heparin-induced thrombocytopenia;
NA, not applicable; PD, pharmacodynamic; PK, pharmacokinetic; PPCI, primary percutaneous coronary intervention.

30 days) compared with UFH (28% versus 34%; relative Several clinical trials support the efficacy and safety
risk [RR] 0.83, 95% CI 0.68–1.01, P = 0.06). However, of bivalirudin compared with heparin plus routinely or
enoxaparin led to a significant 41% relative reduction provisionally administered intravenous GPIs in patients
in the composite main secondary end point of death, undergoing PCI, including for ACS20–22. However, the
recurrent acute coro­nary syndrome (ACS), or urgent benefit of bivalirudin use in patients with STEMI under‑
revasculariza­tion (7% versus 11%; P = 0.015), as well going PPCI has been the subject of debate. Clinical trials
as other secon­d ary end points, without increasing of bivalirudin versus heparin in PPCI are summarized
the risk of major bleeding (5% versus 5%; P = 0.79). in TABLE 2 and FIG. 2.
Of note, crossover between anticoagulants or simultan­ In the HORIZONS‑AMI trial23, patients (n = 3,602)
eous administration of both was associated with worst with STEMI undergoing PPCI were randomly assigned
outcomes19. On the basis of the neutral results of the to receive either bivalirudin with a provisional GPI, or
ATOLL trial, the ACC/AHA guidelines do not give any UFH plus routine GPIs. Compared with heparin plus
recommendation for the use of enoxaparin in PPCI1,2. GPIs, bivalirudin significantly reduced the risk of net
However, on the basis of a potential benefit in the adverse clinical events at 30 days (9.2% versus 12.1%;
secon­dary end point, the ESC guidelines give a class IIa RR 0.76, 95% CI 0.63–0.92, P = 0.005), mainly driven by
indication for enoxaparin in this setting3,4. a 40% reduction in major bleeding (4.9% versus 8.3%;
RR 0.60, 95% CI 0.46–0.77, P <0.001). Although major
Bivalirudin. The direct thrombin inhibitor bivalirudin adverse cardiovascular events (MACE) were similar
is a synthetic derivate of hirudin. The drug is a poly­ between the two treatments, bivalirudin use led to a four‑
peptide of 20 amino acids that binds to thrombin at both fold increase in the rate of acute (≤24 h) stent thrombosis
the active site and the fibrinogen-binding site (exosite 1) (1.3% versus 0.3%; P <0.001), with no differences in the
without the need for a cofactor15,20. Administered by rate of subacute (>24 h−30 days) stent thrombosis23. Of
intravenous infusion, bivalirudin has a linear dose– note, at 1‑year and 3‑year follow‑up, results were con‑
response profile, which translates into an immediate sistent with the 30‑day analysis, and bivalirudin use was
(2–15 min) and predictable anticoagulant effect, with no associated with a reduction in cardiac and all-cause mor‑
need for routine coagulation monitoring and rapid offset tality24,25. Interestingly, a post-hoc analysis of this trial sug‑
of anticoagulant effects after stopping the infusion15,20. gested that a UFH bolus administered before bivalirudin
Only 20% of bivalirudin clearance is dependent on the administration might be associated with an attenuated
kidneys, and its half-life increases from 25 min to 1 h in risk of acute stent thrombosis without increasing major
patients with severe kidney disease, and 3.5 h in patients bleeding26,27, which was supported by registry data on
undergoing haemodialysis15,20. almost 3,000 patients with STEMI undergoing PPCI28.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The EUROMAX trial29 was designed to investi‑ P = 0.008) or heparin plus tirofiban (difference −8.1%;
gate whether the benefit of bivalirudin over heparin RR 0.52, 95% CI, 0.39–0.69, P <0.001). These results
in PPCI persisted in the setting of a more contempor­ were primarily owing to a reduction in bleeding events
ary approach characterized by prehospital adminis‑ with bivalirudin compared with heparin and hep‑
tration of antithrombin therapy, optional use of GPIs, arin plus tirofiban (4.1% versus 7.5% versus 12.3%;
and use of radial access and of the more potent P2Y12- P = 0.001), without significant differences in MACE or
receptor inhibitors prasugrel and ticagrelor. In the trial, stent thrombosis. Moreover, acute stent thrombosis was
patients (n = 2,218) with STEMI transported for PPCI not increased with bivalirudin in this trial (0.3% in each
were randomly assigned to receive either bivalirudin or group), possibly owing to the routine use of a prolonged
hep­arin plus optional GPIs, initiated in the ambulance postprocedural bivalirudin infusion at PCI dose. In the
or in a hospital without PPCI capacity. Compared with subgroup of 1,925 patients with STEMI undergoing
heparin plus optional GPIs, bivalirudin significantly PPCI, results were consistent with the overall trial popu­
reduced the risk of death or non-CABG-related major lation for the primary end point, MACE, acute stent
bleeding at 30 days (5.1% versus 8.5%; RR 0.60, 95% CI thrombosis, and bleeding. Results were also consistent
0.43–0.82, P = 0.001) and the composite of death, re‑­ at the 1‑year follow-up34.
infarction, or non-CABG major bleeding (6.6% versus In the BRAVE‑4 trial35, patients with STEMI under‑
9.2%; P = 0.02), mainly driven by a 57% reduction in going planned PPCI were randomly assigned to a strat‑
major bleeding (2.6% versus 6.0%; P <0.001). Although egy based on prasugrel plus bivalirudin or clopidogrel
no differences in mortality were observed, bivalirudin plus UFH. The trial was terminated early owing to slow
use was associ­ated with a sixfold increase in the rate of recruitment after 548 patients (out of the 1,240 planned)
acute stent thrombosis (1.1% versus 0.2%; P = 0.007). were randomized. No significant difference in the pri‑
This increase occurred despite prolonged bivalirudin mary outcome (a composite of MACE or major bleed‑
infusion after PCI (in 93% of patients, of whom 22.5% ing at 30 days) was observed between the two strategies
received the PCI dose) and the use of prasu­g rel or (15.6% versus 14.5% in the bivalirudin and UFH groups,
ticagre­lor in half of the patients29. However, the risk of respectively), although the results should be interpreted
stent thrombosis seemed to be mitigated by prolong‑ with caution because the trial was underpowered.
ing the bivalirudin infusion at the PCI dose30. Of note, In the MATRIX trial36, patients (n = 7,213) with an
a prespecified, post-hoc analysis showed that bivaliru‑ ACS for whom PCI was anticipated were randomly
din reduced the composite end point of death or major assigned to receive either bivalirudin or UFH. The
bleeding compared with either heparin plus bailout rates of MACE and net adverse clinical events were not
GPIs or heparin plus routine upstream GPIs31. However, signifi­cantly lower with bivalirudin than with UFH,
these data should be considered as hypothesis-­ despite a significant reduction in major bleeding, all-
generating. A pooled analysis of 5,800 patients from cause death, and cardiac death. Although patients
the HORIZONS‑AMI and EUROMAX trials confirmed treated with bivalirudin had a modestly higher inci‑
a 26% reduction in net adverse outcomes and a 47% dence of definite 30‑day stent thrombosis, the rate of
reduction in major bleeding, with a significant sixfold acute (≤24 h) stent thrombosis was similar between
increase in the risk of acute stent thrombosis, mainly in treatment groups. Moreover, bivalirudin infusion after
the first 4 h after PPCI32. PCI did not significantly decrease the rate of urgent
In the HEAT‑PPCI trial33, patients (n = 1,812) with target-­vessel revascularization, definite stent thrombosis,
STEMI undergoing PPCI were randomly assigned to or net adverse clinical events compared with no infu‑
receive either bivalirudin or UFH. Only provisional sion36. In the subgroup of patients (n = 4,010, 56%) with
use of GPIs (abciximab) was allowed in both groups. STEMI, the use of bivalirudin did not reduce the rate
Bivalirudin use compared with UFH use significantly of MACE compared with the use of UFH (5.9% versus
increased the incidence of MACE (8.7% versus 5.7%; 6.5%; RR 0.90, 95% CI 0.70–1.16, P = 0.43), nor the rate
RR 1.52, 95% CI 1.09–2.13, P = 0.02), mainly driven by a of net adverse clinical events (7.0% versus 8.2%; RR 0.84,
significantly higher rate of re‑infarction, with no signifi­ 95% CI 0.67–1.05, P = 0.13). Consistent with the overall
cant difference in major bleeding (3.5% versus 3.1%; trial population, bivalirudin use was associated with a
P = 0.59). Bivalirudin use was associated with a more reduction in BARC 3 or 5 bleeding (1.7% versus 2.8%;
than threefold increase in the rate of acute stent throm‑ P = 0.019), and all-cause and cardiovascular death com‑
bosis (2.9% versus 0.9%; P = 0.007). Of note, the rate of pared with UFH, with no significant difference in acute
acute stent thrombosis with bivalirudin was twofold to stent thrombosis (0.9% versus 0.5%; P = 0.10). Of note,
threefold higher than in any previous study, possibly bivalirudin seemed to be safer and more effective than
related to the fairly short duration of infusion, which heparin in the subgroup of patients with STEMI and
was stopped immediately at the end of PCI33. chronic kidney disease and in those with previous use
In the BRIGHT trial34, patients (n = 2,194) with acute of UFH37. However, these results should be interpreted
MI undergoing emergency PCI (87.7% with STEMI with caution as they derive from a post-hoc analysis and
undergoing PPCI) were randomly assigned to receive need to be considered as hypothesis-generating.
either bivalirudin, heparin alone, or heparin plus GPI. Overall, results of these large-scale clinical trials,
Bivalirudin use resulted in a decrease in 30‑day net as well as data from several meta-analyses, show that
adverse clinical events compared with either heparin bivali­rudin compared with heparin yields similar rates
alone (difference −4.3%; RR 0.67, 95% CI 0.50–0.90, of MACE and net adverse clinical events at 30 days,

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 2 | Major clinical trials of bivalirudin versus heparin in PPCI


Feature HORIZONS-AMI23 EUROMAX29 HEAT-PPCI33 BRIGHT34 MATRIX-STEMI37
Trial design
Patient 3,602 STEMI undergoing 2,218 STEMI 1,812 STEMI 2,194 acute MI 4,010 STEMI
population PPCI transported for PPCI undergoing PPCI undergoing emergency PCI undergoing PPCI*
(87.7% STEMI)
Type of heparin UFH UFH or enoxaparin UFH UFH UFH
Heparin dose‡ 60 U/kg with subsequent UFH: 100 U/kg 70 U/kg 100 U/kg in heparin-only 70–100 U/kg without
boluses targeted to ACT without GPI or group; 60 U/kg in heparin GPI or 50–70 U/kg with
of 200–250 s 60 U/kg with GPI. plus tirofiban group GPI
Enoxaparin: 0.5 mg/kg
GPI use in Routine Routine or bailout Bailout Bailout (5.6% of patients) in Routine or bailout
heparin group (97.7% of patients) (69% of patients) (14% of patients) heparin-only group; routine (25.9% of patients)
(100%) in heparin plus
tirofiban group
GPI use in Bailout (7.5% of patients) Bailout Bailout Bailout Bailout
bivalirudin (11.5% of patients) (14% of patients) (4.4% of patients) (4.6% of patients)
group
Post-PCI None (but could be 4 h after PCI at None 30 min−4 h after PCI Patients randomized 1:1
bivalirudin continued at low doses if 0.25 mg/kg/h; (mean 180 min) at PCI dose to receive or not receive
infusion clinically indicated) continuation of post-PCI infusion
PCI dose was also (full dose for up to 4 h
permitted (22.5%) or reduced dose of
0.25 mg/kg/h for ≥6 h)
P2Y12-receptor Clopidogrel 300–600 mg Clopidogrel (50%), Clopidogrel (11%), Clopidogrel 300–600 mg Clopidogrel (29%),
inhibitor prasugrel (30%), prasugrel (27%), prasugrel (31%),
or ticagrelor (20%) or ticagrelor (62%) or ticagrelor (30%)
Radial access No 47% of patients 81% of patients 78% of patients 50% (randomized 1:1 to
radial versus femoral)
Primary end NACE (major bleeding Death or Efficacy: MACE NACE (MACE [all-cause MACE (death, MI, or
point or MACE [death, non-CABG-related (all-cause death, death, reinfarction, stroke) at 30 days;
reinfarction, TVR major bleeding at CVA, reinfarction, ischaemia-driven TVR, or NACE (MACE or major
for ischaemia, and 30 days or additional stroke] or any bleeding) at bleeding) at 30 days
stroke]) at 30 days; unplanned TLR) at 30 days
non-CABG-related major 28 days. Safety: major
bleeding at 30 days bleeding at 28 days
Bleeding Protocol-defined§ Protocol-defined|| BARC type 3–5 BARC BARC type 3 or 5
definition
Study results
Primary end NACE: bivalirudin 9.2% Bivalirudin 5.1% MACE: bivalirudin Bivalirudin 8.8% versus MACE: bivalirudin 5.9%
point(s) versus heparin 12.1% versus heparin 8.5% 8.7% versus heparin heparin 13.2% versus versus heparin 6.5%
(P = 0.005); non-CABG- (P = 0.001) 5.7% (P = 0.02); major heparin plus tirofiban 17.0% (P = 0.43); NACE:
related major bleeding: bleeding: bivalirudin (P <0.001) bivalirudin 7.0% versus
bivalirudin 4.9% versus 3.5% versus heparin heparin 8.2% (P = 0.13)
heparin 8.3% (P <0.001) 3.1% (P = 0.59)
MACE Bivalirudin 5.4% versus Bivalirudin 6.0% See primary end Bivalirudin 5.0% versus See primary end point
heparin 5.5% (P = 0.95) versus heparin 5.5% point heparin 5.8% versus heparin
(P = 0.64) plus tirofiban 4.9% (P = 0.83)
Major bleeding See primary end point Bivalirudin 2.6% See primary end BARC type 3–5: Bivalirudin 1.7% versus
versus heparin 6.0% point bivalirudin 0.5% versus heparin 2.8% (P = 0.019)
(P <0.001) heparin 1.5% versus heparin
plus tirofiban 2.1% (P = NA)
Acute stent Bivalirudin 1.3% versus Bivalirudin 1.1% Bivalirudin 2.9% Bivalirudin 0.3% versus Bivalirudin 0.9% versus
thrombosis heparin 0.3% (P <0.001) versus heparin 0.2% versus heparin 0.9% heparin 0.3% versus heparin heparin 0.5% (P = 0.10)
(≤24 h) (P = 0.007) (P = 0.007) plus tirofiban 0.3% (P = NA)
Bivalirudin dose was 0.75 mg/kg bolus followed by 1.75 mg/kg/h for the duration of PCI in all studies. *The MATRIX trial included 7,213 patients with ACS for whom PCI was
anticipated, of whom 4,010 had STEMI (56%) and 3,203 (44%) had non‑ST‑segment elevation ACS; results of the STEMI subgroup are reported here. ‡Initial bolus dose;
subsequent UFH boluses were to be administered to maintain target ACT. §Major bleeding: intracranial or intraocular haemorrhage; bleeding at the access site, with a
haematoma that was ≥5 cm or that required intervention; a decrease in the haemoglobin level of ≥4 g/dl without an overt bleeding source or ≥3 g/dl with an overt bleeding
source; reoperation for bleeding; or blood transfusion. ||Major bleeding: intracranial, retroperitoneal, or intraocular bleeding; access-site haemorrhage requiring
radiological or surgical intervention; a reduction in the haemoglobin level of >4 g/dl without an overt source of bleeding; a reduction in the haemoglobin level of >3 g/dl
with an overt source of bleeding; reintervention for bleeding; or use of any blood-product transfusion. ACS, acute coronary syndrome; ACT, activated clotting time;
BARC, Bleeding Academic Research Consortium; CVA, cardiovascular accident; GPI, glycoprotein IIb/IIIa inhibitor; MACE, major adverse cardiovascular events;
MI, myocardial infarction; NA, not available; NACE, net adverse clinical events; STEMI, ST‑segment-elevation myocardial infarction; PCI, percutaneous coronary
intervention; PPCI, primary percutaneous coronary intervention; TLR, target-lesion revascularization; TVR, target-vessel revascularization; UFH, unfractionated heparin.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

and reduces major bleeding, with a signal towards a a MACE


reduction in all-cause and cardiac mortality, at the price HORIZONS-AMI23
of an increased risk of acute stent thrombosis22,29,33–35,37–39.
Currently, bivalirudin, with or without previous hep­arin EUROMAX29
therapy, has a class I (level of evidence B) recommenda‑
HEAT-PPCI33
tion in the ACC/AHA guidelines1,2. The ESC guidelines
give a class IIa recommendation, also specify­ing that a BRIGHT34*
1.75 mg/kg/h infusion should be administered for up
to 4 h after the procedure3,4. In patients at high risk of MATRIX-STEMI37†
bleeding, preferential use of bivalirudin mono­therapy
compared with UFH plus GPIs is given a class  IIa b Major bleeding
recom­m endation in the ACC/AHA guidelines 1,2.
Of note, the different levels of recommendations in HORIZONS-AMI23
these guidelines might be owing to the timing of publi‑
EUROMAX29
cation relative to the clinical trials described above. The
ACC/AHA guidelines were published before the results HEAT-PPCI33
of the EUROMAX and HEAT‑PPCI trials were available,
whereas the ESC guidelines incorporated the results of BRIGHT34*
these trials, which led to a downgrading of bivalirudin
MATRIX-STEMI37†
from a class I recommendation to the current class IIa.
Whether the results of the BRIGHT and MATRIX trials 0.0 0.5 1.0 1.5 2.0
will lead to further changes in the guidelines is currently Bivalirudin better Heparin better
not known. Figure 2 | Ischaemic and bleeding outcomes in the
Nature Reviews | Cardiology
major clinical trials comparing bivalirudin versus
Antiplatelet therapy heparin in ST‑segment elevation myocardial infarction
Intravenous antiplatelet drugs include the cyclo­ (STEMI). The graphs show relative risk with the 95%
confidence intervals of bivalirudin versus heparin on
oxygenase (COX) 1 inhibitor aspirin (not approved by
a | major adverse cardiovascular events (MACE) and
the FDA for parenteral use); the GPIs abciximab, eptifi­ b | major bleeding. MACE and major bleeding are as
batide, and tirofiban; and the P2Y12-receptor inhibitor defined in each study protocol. Follow‑up was 30 days for
cangrelor. Given that aspirin is primarily used in an oral each trial, with the exception of HEAT-PPCI for which it was
formulation, details on this drug are given in the section 28 days. *Data are reported for the comparisons between
on oral antiplatelet therapy. bivalirudin versus heparin plus bailout glycoprotein IIb/IIIa
inhibitors. ‡The MATRIX trial included 7,213 patients with
Glycoprotein IIb/IIIa inhibitors. GPIs can be classified acute coronary syndrome for whom percutaneous
into two groups: small (eptifibatide, tirofiban) and non- coronary intervention was anticipated, of whom 4,010
small (abciximab) molecules, which are characterized (56%) had STEMI and 3,203 (44%) non‑ST‑segment
elevation acute coronary syndrome. The results of the
by different pharmacological properties (TABLE 3). They
STEMI subgroup are reported here.
target the final pathway of platelet aggregation, com‑
peting with von Willebrand factor and fibrinogen for
glycoprotein IIb/IIIa receptor binding and provide fast Intracoronary administration of GPIs, which could
and potent antiplatelet effects40. GPIs have been shown potentially allow a rapid achievement of local platelet
to improve ischaemic outcomes in patients with STEMI inhibition resulting in improved myocardial perfusion,
or non-STEMI undergoing PCI, and most clinical trials has been tested in several small studies and shown to be
of GPIs in STEMI have studied abciximab40–43. However, associated with some benefits, as compared with intra‑
recent trials also support the benefit of eptifibatide and venous administration, but these results have not been
tirofiban (used at high-bolus dose), which yield similar confirmed in large-scale clinical trials46–49. Currently, the
outcomes to abciximab44,45. Nevertheless, the clinical ACC/AHA guidelines give a class IIa recommendation
benefit of GPIs was mainly shown in the era of b ­ alloon for GPIs at the time of PPCI (abciximab, double-bolus
angioplasty, before routine use of dual antiplatelet ther‑ eptifibatide, or high-bolus-dose tirofiban) in selected
apy (DAPT), the advent of new stent technologies, radial patients with STEMI receiving UFH (with or without
artery interventions, thrombus aspiration, and direct stenting or clopidogrel pretreatment), and a class IIb
thrombin inhibitors. In these trials, GPI therapy was indication for intracoronary abciximab1,2. The ESC
associated with a reduction in MACE at the expense of guidelines instead give a class IIa recommendation for
increased bleeding and thrombocytopenia40–45. To date, GPIs for bailout use only (for example, in the event of
the administration of GPIs is an accepted treatment evidence of no‑reflow or thrombotic complications)3,4.
option in patients undergoing PPCI. However, the Several studies have also investigated whether early,
development of alternative treatment strategies with a upstream use of GPIs as a facilitating strategy to early
more favourable safety profile has reduced their role, reperfusion can improve clinical outcomes in STEMI50.
and available data support selective (that is, bail-out However, results were not consistent, and a clear benefit
in the event of large residual thrombus) rather than of this approach has not been demonstrated. A detailed
routine use40. description of pharmacoinvasive approaches for STEMI

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 3 | Pharmacology of glycoprotein IIb/IIIa inhibitors


Feature Abciximab Eptifibatide Tirofiban
Trade name ReoPro Integrilin Aggrastat
Molecule Fragment antigen binding (Fab)7E3 Synthetic peptide Nonpeptide mimetic
Molecular weight (Da) ~50,000 ~800 ~500
Stoichiometry (drug to ~1.5:1 >>100:1 >>100:1
glycoprotein IIb/IIIa)
Binding Noncompetitive Competitive Competitive
Half-life (h) • Plasma: 10–15 • Plasma: 2.0–2.5 • Plasma: 2.0–2.5
• Biologic: 12–24 • Biologic = plasma • Biologic = plasma
PCI dosing • Bolus: 250 µg/kg • Bolus: 180 µg/kg* plus • Bolus: 25 µg/kg
• Infusion: 0.125 µg/kg/min (12 h) 180 µg/kg (after 10 min) • Infusion: 0.15 µg/kg/
• Infusion: 2 µg/kg/min (12–24 h)‡ min (up to 18 h)
Renal adjustment No • Bolus: 180 µg/kg • Bolus: 25 µg/kg
• Infusion: 1 µg/kg/min (12–24 h) • Infusion: 0.075 µg/
kg/min (up to 18 h)
*Started immediately before PCI. ‡Started immediately after the first bolus. PCI, percutaneous coronary intervention. Reprinted
from Muñiz-Lozano, A. et al. Update on platelet glycoprotein IIb/IIIa inhibitors: recommendations for clinical practice. Ther. Adv.
Cardiovasc. Dis. 7 (4), 197–213 (2013).

goes beyond the scope of this Review, and is described After angiography, patients (n = 11,145) were randomly
elsewhere51. Both the ACC/AHA and ESC guidelines assigned to receive either cangrelor (30 μg/kg bolus fol‑
give a class IIb (level of evidence B) recommendation lowed by 4 μg/kg/min infusion for 2–4 h) or clopidogrel
for upstream use of GPIs1–4. In particular, upstream loading dose (300–600 mg before or immediately after
use of a GPI (versus in‑lab use) might be considered PCI, as per institutional standard). In the cangrelor
in high-risk patients undergoing transfer for PPCI, group, patients received 600 mg of clopidogrel at the
on the basis of the benefit shown in a post-hoc analysis end of infusion. Adjunctive cangrelor therapy signifi‑
of the FINESSE trial52. cantly reduced by 22% the composite of death from any
cause, MI, ischaemia-driven revascularization, and stent
Cangrelor. Cangrelor is an analogue of ATP and the only thrombosis at 48 h (4.7% versus 5.9%; P = 0.005), mainly
intravenous P2Y12-receptor inhibitor currently available driven by a 20% reduction in the rate of MI. Cangrelor
for clinical use. It directly and reversibly binds to the also led to a significant 38% reduction in the rate of the
P2Y12 receptor in a predominantly competitive ­manner main secondary efficacy end point of stent thrombosis55.
without the need to be metabolized53,54. Cangrelor is Intraprocedural stent thrombosis, which was shown to
characterized by a linear dose-dependent pharmaco­ be associated with an increase in adverse outcomes at
kinetic profile, which leads to very stable pharma‑ 48 h and 30 days, was also ­significantly reduced by the
codynamic effects, and achieves very potent platelet use of cangrelor56.
inhibition reaching steady-state concentrations within The rate of severe bleeding at 48 h was not signifi‑
a few minutes53,54. Its very short half-life (3–5 min) cantly increased by cangrelor according to the GUSTO
allows a fast offset of action, with platelet aggregation criteria, as well as with other definitions of bleeding.
returning to baseline levels within 30–60 min from However, the rate of major bleeding according to the
infusion termination53,54. The unique pharmacological more sensitive ACUITY criteria was significantly
properties of cangrelor, along with the central role of higher in patients treated with cangrelor, mainly owing
the ADP receptor in platelet activation and in amplifi‑ to higher incidence of haematoma at the site of vascu‑
cation of other platelet signalling pathways, make this lar access. Overall, the net rate of adverse clinical events
agent a very attractive option in the setting of PPCI. (ischaemic plus bleeding events) was significantly
In addition, cangrelor is not affected by renal function reduced by the use of cangrelor. The clinical benefit of
and thus, unlike small-molecule GPIs, does not require adjunctive cangrelor therapy was consistent at 30 days55.
dose adjustment in patients with kidney disease, which Of note, in the subgroup of patients receiving bivaliru‑
is often unknown at the time of presentation in patients din (19% of the trial population), cangrelor compared
with STEMI53,54. with clopidogrel significantly reduced ischaemic events
Cangrelor was approved for clinical use on the basis of as well as stent thrombosis to the same magnitude as in
the results of the CHAMPION PHOENIX trial55, which the main trial, with a trend for reduced stent thrombosis
tested the hypothesis that addition of cangrelor to DAPT in the first 2 h (REF. 57). An exploratory pooled analysis of
(aspirin plus clopidogrel) in patients under­going PCI patient-level data from the three phase III CHAMPION
could reduce the occurrence of acute ischaemic events in trials compared the ischaemic and bleeding risk of GPIs
P2Y12-receptor inhibitor-naive patients across the spec‑ versus cangrelor in patients undergoing PCI. Cangrelor
trum of coronary artery disease manifestations (stable was associated with a similar ischaemic risk and a trend
angina, non‑ST‑segment elevation ACS, and STEMI). towards a lower risk-adjusted risk of severe bleeding

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

compared with clopidogrel plus GPIs58. In the STEMI ADP-binding site on the P2Y12 receptor63–68. By con‑
subgroup of the CHAMPION PHOENIX trial55 (18% of trast, ticagrelor is a noncompetitive ADP antagonist
the overall trial population), cangrelor showed a con‑ and can, therefore, be administered before, during, or
sistent 25% reduction in the primary end point (2.8% after cangre­lor infusion63,69. No drug interaction has
versus 3.7%; OR 0.75, 95% CI 0.46–1.25, P = 0.98 for been described when transitioning from any oral P2Y12-
interaction) at the expense of an 84% increase in major receptor inhibitor to cangrelor, which can, therefore,
bleeding, which was also consistent with the overall trial be started at any time63–69.
population (P = 0.93 for interaction).
Cangrelor (30 μg/kg bolus plus 4 μg/kg/min infu‑ Oral antithrombotic therapies
sion initiated before PCI and continued for ≥2 h or for Oral antithrombotic therapy is the cornerstone of the
the duration of PCI, whichever is longer) is currently acute and long-term treatment of patients with STEMI.
approved for clinical use by the FDA and the European Several classes of antithrombotic, both antiplatelet and
Medicines Agency (EMA) as an adjunct to PCI for anticoagulant, agents available for oral administration
reducing the risk of periprocedural MI, repeat coro‑ are currently approved for clinical use for the manage‑
nary revascularization, and stent thrombosis in patients ment of patients with STEMI (TABLE 4). These agents
who have not been treated with an oral P2Y12-receptor include COX1 inhibitors (aspirin); P2Y12-receptor
antagonist and are not being given a GPI59,60. Although antagonists (clopidogrel, prasugrel, ticagrelor, and
no specific recommendations are available for patients ticlopidine); PAR1 inhibitors (vorapaxar); and fac‑
with STEMI, cangrelor has been endorsed in the ESC tor Xa inhibitors (rivaroxaban)1–4,9. The use of aspirin in
guidelines for patients with non‑ST‑segment elevation combin­ation with a P2Y12-receptor antagonist is com‑
ACS (class IIb, level of evidence A)61. To date, the clinical monly known as standard DAPT. Other oral therapies
profile of cangrelor compared with GPIs and in addi‑ have been tested in addition to standard DAPT and are,
tion to the more potent oral P2Y12 inhibitors prasugrel therefore, described as adjunctive therapies.
and ticagrelor has yet to be tested. Although switching
from cangrelor to clopidogrel has been tested in large- Standard dual antiplatelet therapy
scale clinical trials55,62, the transition between cangrelor Aspirin. Aspirin irreversibly inhibits COX1 and thereby
and other oral antiplatelet agents has been investigated blocks the production from arachidonic acid of throm‑
in a few small pharmacodynamic studies63–69. Data boxane A2, a vasoconstrictor and highly potent stimu‑
available so far suggest that clopidogrel and prasugrel lant of platelet activation70,71. Aspirin is rapidly absorbed
should be administered immediately after discontinu­ in the upper gastrointestinal tract and is associated
ation of cangre­lor infusion to avoid a pharmacodynamic with measurable platelet inhibition within 60 min.
interaction, because these agents compete for the same The plasma half-life of aspirin is approximately 20 min,

Table 4 | Pharmacological properties of oral antithrombotic therapy for STEMI


Feature Aspirin Clopidogrel Prasugrel Ticagrelor Vorapaxar Rivaroxaban*
Target COX1 P2Y12 receptor P2Y12 receptor P2Y12 receptor PAR1 Factor Xa
Type of blockade Irreversible Irreversible Irreversible Reversible Reversible Reversible
Dose 150–325 mg 600 mg 60 mg LD; 10 mg 180 mg LD; 90 mg 2.08 mg once-daily 2.5 mg twice-daily MD
LD; 81–100 mg LD; 75 mg once-daily MD twice-daily MD MD
once-daily MD once-daily MD
Prodrug No Yes Yes No‡ No No
Onset of action 60 min 2‑8 h 30 min−4 h§ 30 min−4 h§ >12 h|| 2–4 h
Offset of action 7–10 days 7–10 days 7–10 days 3–5 days 4–8 weeks 12 h
Drug interactions NSAIDs CYP2C19 No CYP3A inhibitors CYP3A inhibitors CYP3A4 inhibitors or
inhibitors or inducers, drugs or inducers, drugs inducers, P‑glycoprotein
using P‑glycoprotein using P‑glycoprotein transporter inhibitors
transporter transporter
Timing of Immediately At presentation At presentation At presentation or at After stabilization¶ After stabilization
administration after or at time or at time time of PPCI (>24 h after admission)
presentation of PPCI of PPCI
Contraindications Hypersensitivity Hypersensitivity, Prior CVA, high Prior ICH, high risk Prior ICH or Prior CVA, CrCl
active risk of bleeding, of bleeding, severe CVA, high risk <15 ml/min, high risk of
pathological hypersensitivity hepatic dysfunction, of bleeding, bleeding, severe hepatic
bleeding hypersensitivity hypersensitivity dysfunction, treatment
with other anticoagulant,
hypersensitivity
*Approved only by the European Medicines Agency; not approved by the FDA for this use. ‡Although most ticagrelor-mediated antiplatelet effects are direct,
approximately 30–40% are attributed to an active metabolite (AR‑C124910XX). §Depending on clinical setting. ||If administered without a loading dose. ¶2 weeks
after the acute event according to European Medicines Agency; no timing prespecified by the FDA. COX, cyclooxygenase; CrCl, creatinine clearance; CVA,
cerebrovascular accident; CYP, cytochrome P450; ICH, intracranial haemorrhage; LD, loading dose; MD, maintenance dose; NSAID, nonsteroidal anti-inflammatory
drug; PAR, protease-activated receptor; PPCI, primary percutaneous coronary intervention; STEMI, ST‑segment elevation myocardial infarction.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

and peak plasma levels are achieved 30–40 min after treatment of patients with STEMI1–4,9. The first avail‑
ingestion of uncoated aspirin70,71. In the ISIS‑2 trial72, able P2Y12-receptor inhibitor was the first-generation
aspirin therapy was associated with a significant thienopyridine ticlopidine, which in combination with
reduction in vascular mortality in patients with sus‑ aspirin was superior to both aspirin alone and aspirin
pected acute MI, either as a stand-alone therapy or plus warfarin for prevention of thrombotic events in
in combination with streptokinase. Aspirin is the patients undergoing PCI77–80. However, its use has been
established first-line therapy in patients with STEMI, largely abandoned owing to its frequent adverse effects,
and non-enteric-coated aspirin (150–325 mg) should including life-threatening haematological disorders,
be administered promptly after presentation (class I and because of the better clinical outcomes achieved
recommendation) and then continued indefinitely with clopidogrel87,88.
irrespective of the treatment strategy 1–4. The aspi‑
rin loading dose should preferably be given orally Clopidogrel. The second-generation thienopyridine
including chewing, to ensure complete inhibition clopidogrel is the most widely used P2Y12-receptor
of thromboxane A2-dependent platelet aggregation. inhibitor89–91. Clopidogrel is a prodrug that requires
An  intravenous formulation is available in some metabolic transformation to exert its antiplatelet
­countries, and can be used in patients who are unable to effect. After intestinal absorption, ~85% of clopidogrel
swallow. Current evidence suggests the use of low-dose is hydrolysed by carboxylase to an inactive metabo­
aspirin (≤100 mg) following the initial loading dose73,74. lite. The remaining ~15% is rapidly metabolized by
However, aspirin specifically targets COX1, and is not hepatic cytochrome P450 (CYP) isoenzymes, mainly
effective in reducing platelet activation mediated by CYP2C19, in a two-step oxidation process which leads
other pathways involved in arterial thrombosis, which to the gener­ation of a highly unstable active metabolite
exposes patients to residual thrombotic risk in both the that binds to the P2Y12 receptor in a competitive (irre‑
acute and long-term phases, underscoring the need for versible) ­manner9. Several clinical trials have shown
adjunctive therapies8,9. P2Y12-receptor inhibitors are the benefit of DAPT with aspirin and clopidogrel in a
the standard-of‑care therapy to be used in combination broad spectrum of coronary artery disease manifesta‑
with aspirin, also known as DAPT9. tions, including in patients with STEMI81–84 (TABLE 5).
The CLARITY trial83, conducted in patients (n = 3,491)
P2Y12-receptor inhibitors. ADP exerts its effects on with STEMI treated with fibrinolytic therapy and then
platelets through the purinergic G‑protein-coupled undergoing urgent angiography, showed that clopi‑
P2Y12 and P2Y1 receptors, with the former having the dogrel (300 mg loading dose, followed by 75 mg per
major role in platelet activation and aggregation9,75. The day) in addition to aspirin significantly reduced the
combination of a P2Y12-receptor antagonist and aspirin incidence of thrombotic events before angiography
can produce a degree of platelet inhibition higher than compared with placebo (21.7% versus 15.0%; OR 0.64,
that achieved by each single agent76, and this synergistic 95% CI 0.53–0.76, P <0.001). In the COMMIT trial84,
effect has been shown to be beneficial in several clin‑ patients (n = 45,852) with acute MI, mainly STEMI, were
ical trials that tested antiplatelet regimens in patients randomly assigned to receive either clopidogrel (75 mg
with ACS, including those with STEMI9,77–86 (TABLE 5). per day) or matching placebo, plus aspirin. Clopidogrel
Therefore, DAPT with aspirin and a P2Y12-receptor therapy significantly reduced thrombotic events (9.2%
inhibitor is the mainstay of the early and long-term versus 10.1%; OR 0.91, 95% CI 0.86–0.97, P = 0.002),

Table 5 | Major clinical trials of oral antithrombotic therapy in ACS including STEMI
Trial Setting Treatment groups Primary end point Results*
CLARITY 83
3,491 STEMI Aspirin plus clopidogrel plus FA Occluded infarct-related 15.0% vs 21.7%; OR 0.64,
vs aspirin plus FA artery, death, or recurrent MI 95% CI 0.53–0.76, P <0.001
before angiography
COMMIT84 45,852 acute MI (39,755 STEMI) Aspirin plus clopidogrel vs aspirin Death, reinfarction, or stroke 9.2% vs 10.1%; OR 0.91,
at 28 days 95% CI 0.86–0.97, P = 0.002
TRITON-TIMI 38 13,608 ACS undergoing PCI Aspirin plus prasugrel vs aspirin Cardiovascular death, 9.9% vs 12.1%; HR 0.81,
(REF. 85) (3,534 STEMI) plus clopidogrel nonfatal MI or nonfatal 95% CI 0.73–0.90, P <0.001
stroke at 14.5 months
PLATO86 18,624 ACS (7,544 STEMI) Aspirin plus ticagrelor vs aspirin Death from vascular causes, 9.8% vs 11.7%; HR 0.84,
plus clopidogrel MI, or stroke at 12 months 95% CI 0.77–0.92, P <0.001
TRA 2oP-TIMI 50 26,449 with prior MI, ischaemic Standard APT plus vorapaxar vs Cardiovascular death, MI, 9.3% vs 10.5%; HR 0.87,
(REF. 108) stroke, or PAD (9,248 with standard APT plus placebo or stroke at 36 months 95% CI 0.80–0.94, P <0.001
STEMI as qualifying event)
ATLAS ACS 2- 15,526 patients with recent DAPT plus rivaroxaban 5.0 mg vs Cardiovascular death, 8.8% vs 9.1% vs 10.7%;
TIMI 51 ACS (7,817 STEMI) DAPT plus rivaroxaban 2.5 mg vs nonfatal MI or nonfatal HR 0.84, 95% CI 0.74–0.96,
(REF. 115) DAPT plus placebo stroke at 24 months P = 0.008‡
*Results are reported for the main trial. ‡Combined rivaroxaban groups versus placebo. ACS, acute coronary syndrome; APT, antiplatelet therapy; DAPT, dual
antiplatelet therapy; FA, fibrinolytic agent; MI, myocardial infarction; PAD, peripheral artery disease; PCI, percutaneous coronary intervention; STEMI, ST‑segment
elevation myocardial infarction.

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

and death from any cause (7.5% versus 8.1%; OR 0.93,


TRITON96
95% CI 0.87–0.99, P = 0.03). On the basis of these data,
clopidogrel has been for many years the standard of TRITON PPCI97
care, in addition to aspirin, in patients with STEMI9. PLATO102
Clopidogrel (300–600 mg loading dose and 75 mg daily
maintenance dose) is currently recommended (class I) PLATO PPCI103
for the treatment of patients with STEMI. Available data TRA 2°P110
also suggest that a 600 mg loading dose should be pre‑
ferred in patients undergoing PPCI1–4. In patients with ATLAS-ACS115*
STEMI treated with fibrinolytics, clopidogrel is also the 0.0 0.5 1.0 1.5 2.0
only P2Y12-receptor inhibitor currently recommended Alternative Aspirin and
treatment better clopidogrel better
and should be administered as a 300 mg loading dose
followed by 75 mg per day in patients aged ≤75 years, Figure 3 | Ischaemic outcomes in theReviews
Nature ST‑segment
| Cardiology
and 75 mg per day without a loading dose in patients elevation myocardial infarction (STEMI) subgroups
aged >75 years1–4. of major clinical trials on novel oral antithrombotic
Concerns have grown about the heterogeneous agents. Risk reduction of novel antithrombotic agents
versus dual antiplatelet therapy with aspirin and
nature of individuals’ response to clopidogrel. Several
clopidogrel in post-hoc analyses on the STEMI subgroups
factors have been associated with clopidogrel response of clinical trials performed in patients with acute coronary
variability, including genetic (CYP polymorphisms), syndromes. Major adverse cardiovascular events are the
clinical (poor absorption, drug–drug interactions, ACS, primary end point of each trial. *Results are reported
diabetes mellitus, obesity, chronic kidney disease), and for the comparison between rivaroxaban 2.5 mg
cellular (accelerated platelet turnover or upregulation versus placebo.
of P2Y 12 pathway) factors 92,93. Importantly, several
pharmaco­dynamic studies have shown that approx­
imately 30–40% of patients have high platelet reactivity of non-CABG-related major bleeding, CABG-related
while on treatment with clopidogrel, which translates major bleeding, life-threatening bleeding, and fatal
into higher rates of cardiovascular events, including bleeding, the net clinical benefit was still in favour of
stent thrombosis92,93. These findings underscore the prasugrel-treated patients. Importantly, use of prasugrel
need for more potent antiplatelet agents, especially for had a neutral effect in patients with low body weight
patients with high prothrombotic milieu such as those (<60 kg) and elderly patients (aged ≥75 years), and a net
with STEMI, which has led to the clinical development harm in patients with a history of stroke or transient
of prasugrel and ticagrelor. ischaemic attack85.
In the subgroup of patients (n = 3,534) with STEMI
Prasugrel. Prasugrel is a third-generation thienopyri‑ undergoing PCI, prasugrel reduced the primary end
dine. Prasugrel is a prodrug and, after intestinal absorp‑ point at 30 days compared with clopidogrel (6.5% ver‑
tion, requires a single-step oxidation process through sus 9.5%; HR 0.68, 95% CI 0.54–0.87, P = 0.002), with
hepatic CYP to generate its active metabolite, which irre‑ an effect that was consistent with the main trial popu­
versibly binds to the P2Y12 receptor9. The active metabo‑ lation (P = 0.769 for interaction)96,97 (FIG. 3). The main
lites of prasugrel and clopidogrel have the same potency, secondary end point of cardiovascular death, MI, or
but the metabolic conversion of prasugrel is more effi‑ urgent target-vessel revascularization at 30 days, as well
cient, leading to higher bioavailability94. These pharma‑ as stent thrombosis, were also significantly reduced.
cological properties translate into faster onset of action, Non-CABG-related major bleeding was similar in the
enhanced platelet inhibition, and lower i­nterindividual clopidogrel and prasugrel groups, but no interaction was
variability compared with clopidogrel95. noted between presenting syndrome and the bene­fits of
In the TRITON-TIMI 38 trial85, patients (n = 13,608) prasugrel (P = 0.4091 for interaction). All these effects
with moderate-to‑high-risk ACS scheduled for PCI were persisted up to 15 months. Of note, patients with STEMI
randomly assigned to receive either prasugrel 60 mg were divided into two strata in the trial: those enrolled
loading dose followed by 10 mg daily maintenance dose, within 12 h of onset of symptoms (PPCI), and those
or clopidogrel 300 mg loading dose followed by 75 mg enrolled between 12 h and 14 days after symptom onset
daily maintenance dose, in addition to aspirin. In the (secondary PCI). No significant benefit with prasu­
trial, coronary anatomy had to be defined before ran‑ grel over clopidogrel was shown for patients receiving
domization in all patients, with the exception of those PPCI, and the effects seen in patients with STEMI as
with STEMI. Prasugrel significantly reduced the primary a whole were mainly driven by patients who received
efficacy end point (a composite of cardiovascular death, secondary PCI. However, the effects on the primary
nonfatal MI, or nonfatal stroke) by 19% compared with end point were consistent between primary and sec‑
clopidogrel over a median follow‑up of 14.5 months, ondary PCI at 15 months (P = 0.15 for interaction), with
mainly driven by a reduction in nonfatal MI. Prasugrel a tendency towards a difference in treatment effect at
also led to a significant 52% reduction in the rate of 30 days (P = 0.06 for interaction). This trend was attenu­
stent thrombosis, and a 34% decrease in the need for ated after periprocedural MI events were excluded from
urgent target-vessel revascularization. Although this the analysis. In addition, the magnitude of the risk
effect was hampered by significantly increased rates reduction achieved by prasugrel in patients treated with

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

PPCI for the primary end point at 30 days was similar In a post-hoc subgroup analysis of patients (n = 4,949)
to that achieved in the whole trial population (HR 0.80, with STEMI treated with PPCI within 12 h of admission
95% CI 0.60–1.08, P = 0.144), with a significant reduc‑ (excluding those initially medically managed), ticagrelor
tion in rates of stent thrombosis96,97. Prasugrel (60 mg led to a 9% relative reduction in the incidence of the
loading dose and 10 mg daily maintenance dose) is cur‑ primary end point compared with clopidogrel (7.9% ver‑
rently a class I recommendation in patients with STEMI sus 8.6%; HR 0.91, 95% CI 0.75–1.12, P = 0.38) (FIG. 3),
undergoing reperfusion with PPCI, and can be admin‑ which was consistent with the overall PLATO popula‑
istered in this setting even before coronary anatomy has tion (P = 0.40 for interaction). Ticagrelor also reduced
been established1–4. the rates of stent thrombosis in this subgroup, with‑
out increasing the risk of major bleeding103. This lack
Ticagrelor. The cyclopentyltriazolopyrimidine ticagre­ of increased rate of haemorrhagic events with ticagre‑
lor is not a prodrug and does not require metabolic lor, which is consistent with the STEMI subgroup of the
activation, although approximately 30–40% of its TRITON-TIMI 38 trial, could be owing to the younger
antiplatelet effects are attributed to an active metabo‑ age and low bleeding risk of this population. Ticagrelor
lite (AR‑C124910XX) generated through the hepatic (180 mg loading dose and 90 mg twice-daily mainten­ance
CYP3A system. Ticagrelor does not bind directly to the dose) is currently recommended (class I) for the treat‑
platelet ADP-binding site on the P2Y12 receptor. The ment and prevention of secondary ­atherothrombotic
drug reversibly binds in a noncompetitive manner to events in patients with STEMI u ­ ndergoing PPCI1–4.
a distinct site on the receptor, thereby preventing via
allosteric modulation ADP activation of the P2Y12 path‑ Adjunctive antithrombotic therapy
way9. Ticagrelor is rapidly absorbed after oral adminis‑ DAPT primarily targets pathways associated with
tration and, because of its half-life of 7–12 h, requires thromboxane A2 and ADP-mediated platelet activ­
twice-daily administration. Compared with clopidogrel, ation8,9. Therefore, other pathways, such as thrombin-­
ticagrelor achieves a more prompt, potent, and predict‑ mediated platelet activation remain unaffected, which
able antiplatelet effect, with a faster offset of action98. might account for the residual risk of atherothrombotic
Moreover, ticagrelor also has non‑P2Y 12-mediated events13,104. Given that levels of thrombin are known
effects; in particular, the drug increases plasma levels to be elevated after an ACS, and thrombin is the most
of adenosine owing to inhibition of cellular uptake99. potent platelet activator, targeting thrombin-mediated
These effects might contribute to the overall benefits of effects has been an important area of clinical investiga‑
ticagrelor, but have also been suggested to contribute to tion13,105. Two different approaches can be pursued to
its nonbleeding adverse effects, such as higher incidence block thrombin effects: indirect modulation by block‑
of dyspnoea (15–22% of ticagrelor-treated patients) and ade of the platelet PAR, and direct inhibition of either
ventricular pauses86,100,101. thrombin or other upstream coagulation factors13,104.
The efficacy and safety of ticagrelor in ACS were evalu­
ated in the PLATO trial86, in which patients (n = 18,624) Vorapaxar. PARs are a family of G‑protein-coupled
with ACS were randomly assigned to receive either tica‑ receptors, and PAR1 and PAR4 are expressed on human
grelor (180 mg loading dose followed by 90 mg twice- platelets. Of these, PAR1 has the principal role of mediat‑
daily maintenance dose) or clopidogrel (300–600 mg ing platelet activation at low concentrations of thrombin,
loading dose followed by 75 mg daily maintenance whereas PAR4 is activated only at high concentrations13.
dose) in addition to aspirin. Compared with clopi‑ The only PAR1 antagonist that has completed phase III
dogrel, ticagrelor significantly reduced the primary clinical investigations and is available for clinical use is
end point (a composite of death from vascular causes, vorapaxar (TABLE 4). Vorapaxar is a synthetic tricyclic
MI, or stroke) by 16% at 12 months, including a signifi­ 3‑phenylpyridine that is an analogue of himbacine. After
cant 21% reduction in cardiovascular death and a 16% oral administration, vorapaxar is rapidly absorbed with
reduction in MI. Ticagrelor treatment also significantly high bioavailability and a long half-life106. Vorapaxar
reduced the rate of definite or probable stent thrombosis. was tested in two large-scale, phase III clinical trials:
Although protocol-defined major bleeding was similar the TRACER trial107, which was conducted in patients
between groups, non-CABG-related major bleeding with non‑ST‑segment elevation ACS and did not show
and fatal intracranial haemorrhage were ­significantly a favourable balance between efficacy and bleeding with
increased with ticagrelor86. vorapaxar in acute management (and, therefore, will not
In the subgroup of patients (n = 7,544) with ST‑ be discussed further), and the TRA 2°P‑TIMI 50 trial108.
segment elevation ACS intended for reperfusion with The TRA 2°P‑TIMI 50 trial108 was a secondary pre‑
PPCI, ticagrelor reduced the incidence of the primary vention trial designed to investigate the efficacy and
end point at 12 months compared with clopidogrel safety of vorapaxar in reducing atherothrombotic events
(10.8% versus 9.4%; HR  0.87, 95%  CI 0.75–1.01, in patients with established atherosclerosis receiving
P = 0.07), with an effect that was consistent with the standard antiplatelet therapy, including aspirin and/or
overall trial results (P = 0.29 for interaction) (FIG. 3). a thienopyridine (mainly clopidogrel) (TABLE 5). Patients
Ticagrelor also reduced rates of MI, all-cause mortal‑ were eligible for the study if they had a history of MI
ity, and definite stent thrombosis, without increasing or ischaemic stroke (within the previous 2 weeks to
the risk of bleeding. However, this subgroup included 12 months), or symptomatic peripheral arterial dis‑
patients initially or ultimately treated conservatively102. ease. Patients (n = 26,449) were randomly assigned to

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

receive either vorapaxar 2.5 mg daily or placebo. After a evident as early as 30 days, with no significant difference
median follow‑up of 30 months, vorapaxar significantly between the two dosing regimens (8.7% with rivaroxa­
reduced the primary end point (death from cardio‑ ban 2.5 mg, 8.2% with rivaroxaban 5 mg, and 10.6% with
vascular causes, MI, or stroke) by 13% compared with placebo). Also in this subgroup, rivaroxaban 2.5 mg led
placebo, driven by a 17% reduction in the rate of MI, to a signifi­cant reduction in cardio­vascular and all-
at the expense of a significant increase in moderate or cause death compared with placebo (FIG. 3). However,
severe bleeding and a twofold increase in intracranial rivaroxaban treatment was still associated with a dose-­
bleeding108. In patients with previous stroke, vorapaxar dependent increase in major bleeding, including intra­
was associated with higher rates of major bleeding cranial haemorrhage, but with no significant increase in
and intracranial haemorrhage, with no improvement fatal bleeding115. The clinical profile of low-dose rivaroxa­
in ischaemic outcomes109. Patients with previous MI ban as an adjunctive therapy to prasugrel or ticagrelor
(n = 17,779) derived an increased benefit in the pri‑ is currently unknown. The phase II GEMINI ACS‑1
mary end point with vorapaxar compared with placebo trial116 is ongoing to test the safety of low-dose rivaroxa­
(8.1% versus 9.7%; P <0.0001), which was consistent ban in addition to P2Y12 inhib­ition with clopidogrel or
across timing and types of MI. In particular, in patients ticagrelor (without aspirin) as compared with standard
with previous STEMI (n = 9,248), vorapaxar led to a DAPT in patients with ACS, including STEMI. Although
significant 27% reduction in the risk of cardiovascular a discussion of the implications of dropping aspirin is
death, MI, or stroke (FIG. 3). Despite a higher incidence beyond the scope of this Review, this strategy has the
of bleeding events, intracranial and fatal bleeding were potential to reduce bleeding associated with the use of
not significantly increased by vorapaxar in patients with more potent antithrombotic therapy without affecting
previous MI, with an overall reduction in the net clin­ ischaemic protection, and is currently being investigated
ical outcome. The outcome improvement achieved with in several ongoing studies.
vorapaxar was even more pronounced after excluding Rivaroxaban is currently approved by the EMA at
patients at high risk of bleeding (previous stroke, body the dose of 2.5 mg twice daily in patients stabilized after
weight <60 kg, or aged >75 years)110. The efficacy and an ACS, co‑administered with either aspirin or DAPT,
safety of vorapaxar in combination with prasugrel and has received a class IIb recommendation in the
and ticagrelor has not been tested. ESC guidelines for selected patients with STEMI at low
Vorapaxar (2.08 mg once-daily maintenance dose) is risk of bleeding who are treated with aspirin and clopi‑
currently approved by the FDA111 and EMA112 for the dogrel3,4,117. Rivaroxaban is not approved by the FDA for
reduction of thrombotic events in patients with previ‑ this indication because of missing data in the pivotal
ous MI or peripheral arterial disease. In patients with trial. Rivaroxaban can also be considered as a therapeu‑
previous MI, the EMA specifies that the drug should tic option in patients requiring chronic oral anticoagu‑
be initiated 2 weeks after the acute event, in line with lant therapy in addition to antiplatelet therapy, such as
TRA 2°P‑TIMI 50 trial inclusion criteria, whereas no those with atrial fibrillation undergoing PCI118. However,
timing is specified by the FDA. a detailed discussion of this topic goes beyond the scope
of this Review, and is described elsewhere119.
Rivaroxaban. Several clinical trials have tested the effi‑
cacy and safety of various non-vitamin K antagonist Limitations and optimization strategies
oral anticoagulants in patients with ACS104,113. However, Given the clinical benefit shown in clinical trials in
only rivaroxaban met the primary end point in phase III patients with ACS, prasugrel and ticagrelor are now
clinical testing and is currently available for clinical use the recommended first-line P2Y12-receptor inhibitors
in this setting104,113. in the setting of PPCI1–4,9. However, subgroup analyses
Rivaroxaban is an oral factor Xa inhibitor that is of the STEMI populations in the TRITON-TIMI 38 and
­rapidly absorbed and reaches a maximum plasma con‑ PLATO trials97,103 showed a nonsignificant reduction in
centration in 2–4 h, with a half-life of approx­imately the primary end points for prasugrel or ticagrelor versus
5–13 h (REF. 104) (TABLE 4). The ATLAS ACS-TIMI 51 clopidogrel in the specific group of patients receiving
trial114 tested the clinical effects of rivaroxaban (2.5 mg PPCI. Arguably, the results in PPCI were consistent with
or 5.0 mg twice daily) compared with ­placebo in patients the overall study findings, and the reason for the absence
(n = 15,526) with a recent (>24 h but <7 days) ACS of significant benefit for prasugrel and ticagrelor in PPCI
receiving standard antiplatelet therapy with aspirin and a could be the insufficient numbers of patients in the sub‑
thienopyridine (mainly clopidogrel) (TABLE 5). Both doses groups to yield adequate statistical power97,103. However,
of rivaroxaban were similarly effective in reducing ischae‑ these findings could also be caused by limitations of oral
mic events after a median follow‑up of 13.1 months, but antiplatelet therapy. Indeed, several pharmacodynamic
this efficacy was offset by a dose-­dependent increase studies conducted in patients with STEMI undergoing
in non-CABG-related major bleeding, including intra­ PPCI have consistently demonstrated delayed antiplate‑
cranial haemorrhage, which was lower with the 2.5 mg let effects of oral P2Y12 inhibitors, including prasugrel
dose. Notably, the 2.5 mg dose was associ­ated with a and ticagrelor, which require >2 h to exert full antiplate‑
significant reduction in cardio­vascular death and all- let effects120–123. This finding might explain why the use
cause death compared with ­placebo. In the subgroup of of prasugrel or ticagrelor in clinical trials did not reduce
patients with STEMI (n = 7,817), rivaroxaban reduced the incidence of acute stent thrombosis associated with
the incidence of ischaemic events, with an effect that was bivalirudin administration29,33–36. The delayed onset and

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a CRUSH study127 (prasugrel) tablets was associated with faster drug absorption and
300 more prompt and potent antiplatelet effects compared
P = 0.053 Crushed
P <0.001 with whole-­tablet ingestion, with an effect observed as
Integral early as 30 min after loading-dose administration127,128
250
(FIG. 4). Although the clinical benefit of these findings
P = 0.022 should be tested in larger trials powered for efficacy and
P2Y12 reaction units (PRU)

200
safety, the use of a crushed loading dose of prasugrel or
ticagrelor seems a reasonable option in PPCI, given the
ANOVA P = 0.008
150 known association between peri-PCI platelet reactivity,
P = 0.023
­thrombotic events, and long-term outcomes56,93,129.
Pretreatment, defined as administration of the load‑
100 P = 0.102
ing dose before assessment of coronary anatomy, seems
P = 0.178 another appealing strategy to provide stronger platelet
50 inhibition at the time of PPCI, even if supporting data
are not strong130. In the STEMI subgroup of a large
meta-analysis on patients undergoing PCI, pretreatment
0
0 0.5 1 2 4 6 24 with clopidogrel was associated with an approximately
Time (h) 50% reduction in mortality131. In the TRITON-TIMI 38
b MOJITO study128 (ticagrelor) trial, only 32% of patients with STEMI received the
350 loading dose before PPCI and, therefore, the value of
pretreatment with prasugrel in STEMI is unknown.
300 In the PLATO trial, all patients were pretreated, so the
P2Y12 reaction units (PRU)

role of ticagrelor pretreatment in STEMI was tested in


250 the ATLANTIC trial132, in which patients (n = 1,862)
undergoing emergency angiography were randomly
200
assigned to receive either prehospital (in the ambulance)
or in‑hospital (in the cardiac catheterization lab­oratory)
150
treatment with a 180 mg loading dose of ticagrelor,
100 in addition to aspirin. Prehospital ticagrelor did not
*P = 0.006 *
improve pre-PCI markers of coronary reperfusion and
50 was not associated with significant differences in platelet
reactivity132,133. Although this strategy showed a signifi‑
0 cant reduction in the rate of definite acute (≤24 h) stent
Baseline 1 2 4 8
Time (h) thrombosis with no difference in major bleeding, the
Figure 4 | Pharmacodynamic comparison of crushed versus integral tablets trial was not powered for clinical end points, and results
Nature Reviews | Cardiology should be considered as exploratory only132,134. The
of P2Y12-receptor inhibitors in ST‑segment elevation myocardial infarction.
a | Comparison in P2Y12 reaction units (PRU) measured by VerifyNow P2Y12 between ACC/AHA guidelines recommend the administration of
crushed and whole tablets of 60 mg prasugrel loading dose. b | Comparison in PRU an oral P2Y12-receptor inhibitor as soon as possible after
between crushed and whole tablets of 180 mg ticagrelor loading dose. ANOVA P values first medical contact or at the time of PPCI, whereas the
are for the comparison between groups over time. Reprinted from Sardella, G., ESC guidelines recommend initiating therapy as early as
Calcagno, S. & Mancone, M. Different prasugrel administration in STEMI patients: possible before angiography1–4.
go faster and no fear to crush! J. Am. Coll. Cardiol. 67 (17), 2005–2007 © (2016), Inability to achieve adequate platelet inhibition with
with permission from Elsevier. oral P2Y12-receptor inhibitors in PPCI is further exacer‑
bated by other clinical conditions commonly associated
attenuated antiplatelet effects of P2Y12-receptor inhib­ with STEMI, such as inability to swallow (in patients who
itors have been attributed to impaired drug absorption are sedated, intubated, in shock, or those with ­nausea
resulting in reduced drug bioavailability in the early or vomiting), therapeutic hypothermia, or morphine
hours after the loading dose120,123. Given the higher administration120,132,133,135. Use of intravenous agents,
prothrombotic milieu of the STEMI setting, strategies such as GPIs or cangrelor, in the catheterization lab­
to increase bioavailability of orally administered P2Y12- ora­tory has the potential to provide immediate platelet
receptor inhibitors have been advocated for patients inhib­ition until the full antiplatelet effect of oral P2Y12-
undergoing PPCI120. Because the pharmacokinetic and receptor inhibitors is achieved48,55. In the FABOLUS PRO
pharmacodynamic profiles of P2Y12-receptor inhibitors study48, the addition of a bolus only of tirofiban led to
are dose-dependent, the use of higher loading-dose near-complete platelet inhibition, without the need for
regimens seems an intuitive option and has been tested an infusion, in patients undergoing PPCI who received a
in pharmacodynamic studies. However, this strategy 60 mg loading dose of prasugrel. However, the safety and
was associated with no or marginal enhancement of efficacy of this strategy is unknown. Although cangre‑
platelet inhibition123–126. The administration of crushed lor has been shown to enhance platelet inhibition when
prasugrel and ticagrelor in patients undergoing PPCI administered in addition to prasugrel or ticagrelor ther‑
has been tested in the CRUSH127 and MOJITO128 stud‑ apy, the clinical benefit of its use in addition to these
ies, respectively. These studies showed that crushing agents in PPCI is yet to be tested55.

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Practical recommendations of the agent should, therefore, be based on the throm‑


With the availability of several anticoagulant and botic and bleeding risk of the individual patient, and
antiplatelet agents with different safety and efficacy in some centres might also be dictated by economic
profiles, finding the best antithrombotic cocktail to considerations (FIG. 5).
help reperfusion during PPCI is a challenge for clin­ For patients at low risk of bleeding events, the use
icians. The ultimate goal of antithrombotic therapy is of heparin as an anticoagulant, in particular UFH,
to provide a rapid onset of potent pharmacodynamic seems to be the most cost-effective option. In many sys‑
effect to prevent stent thrombosis and periprocedural tems, a fixed bolus of UFH (generally 4,000–5,000 IU)
MI, without increasing serious bleeding. The choice is ­routinely administered upstream, at the time of clin­
ical presentation, as this strategy has been associated
with better clinical outcomes, particularly for those
STEMI intended for PPCI patients who then are treated with bivalirudin in the
catheteriza­tion laboratory. Importantly, such a strategy
is ­simple and not associated with potential time delays
At clinical presentation that might occur with bolus and infusion administration
Aspirin LD (325 mg oral or 80–150 mg intravenous) of other antithrombotic strategies. For patients who con‑
Consider UFH bolus (4,000–5,000 IU) tinue receiving UFH during PPCI, an additional bolus
should be administered based on activated clotting time
levels. For patients not receiving upstream UFH, enoxa‑
Consider pretreatment parin can be considered as an alternative anticoagulant,
Ticagrelor or prasugrel (clopidogrel if tricagrelor as endorsed by the ESC guidelines, although its use is
and prasugrel contraindicated) not recommended in the ACC/AHA guidelines1–4,136.
Conversely, bivalirudin should be considered in those
with a high risk of bleeding, such as elderly patients or
Intravenous anticoagulant Intravenous anticoagulant
patients with anaemia, thrombocytopenia, or chronic
Low risk of bleeding High risk of bleeding* kidney disease. If bivalirudin is used, strategies to
Heparin Bivalirudin (consider prolonged reduce the risk of acute stent thrombosis should be
(UFH if previous UFH bolus) infusion at PCI dose) employed, such as an initial bolus of UFH or prolonging
the infusion at PCI-dose for up to 4 h after reperfusion.
Cangrelor seems to be an attractive strategy to achieve
immediate and potent platelet inhibition. Cangrelor can
Consider cangrelor in P2Y12-naive patients be considered for all patients undergoing PPCI not pre‑
treated with oral P2Y12-receptor antagonists, especially
Consider GPI for bail-out use for those with clinical or angiographic factors of high
thrombotic risk. This strategy would also allow bridging
of the gap associated with the delayed onset of action of
P2Y12 inhibitor LD at time of PPCI oral P2Y12-receptor inhibitors, including prasugrel and
Consider crushed tablets
ticagrelor, which require several hours before they reach
Preferred agents Alternative agent their full platelet inhibitory effects. Although GPIs are
Prasugrel or ticagrelor Clopidogrel also an option to obtain immediate platelet inhibition,
Choice between agents is based on If both ticagrelor and prasugrel their routine use has fallen out of favour, and they are
contraindications and precautions contraindicated or not available
mostly reserved for bail-out use. Compared with GPIs,
cangrelor seems to be associated with fewer major
Continue DAPT for 12 months‡ bleeding events and has lower incidence of thrombo‑
cytopenia, a well-known and potentially lethal adverse
effect of GPIs54,58. However, a direct comparison between
Consider adding vorapaxar or low-dose rivaroxaban§ cangrelor and GPIs has not been tested in large-scale,
In selected patients at low risk of bleeding if receiving adequately powered clinical trials. Whereas overdosing
background therapy of aspirin and clopidogrel
of GPIs is not uncommon and has been associated with
Figure 5 | Proposed algorithm for the choice of antithrombotic Naturetherapy in| patients
Reviews Cardiology an increased risk of bleeding complications137, cangre‑
with ST‑segment elevation myocardial infarction (STEMI) undergoing primary lor overdosing is rare and was not associated with an
percutaneous coronary intervention (PPCI). *Clinical factors known to be associated increase in rates of bleeding or other adverse events138.
with increased risk of bleeding are: history of bleeding, oral anticoagulant therapy, Moreover, in patients with planned use of GPIs, the
female sex, advanced age, low body weight, chronic kidney disease, diabetes mellitus, UFH dose should be adjusted, whereas with cangre‑
anaemia, chronic steroid or nonsteroidal anti-inflammatory drug therapy151. ‡Shorter lor, the full dose of UFH should be used. Although the
dual antiplatelet therapy (DAPT; 6 months) can be considered in patients with overt
use of cangrelor in addition to bivalirudin seems to be
bleeding or who are at high risk of bleeding, whereas prolonging DAPT beyond
12 months is a reasonable option in selected patients at low risk of bleeding or the antithrombotic cocktail associated with the ideal
without overt bleeding while receiving DAPT. §Vorapaxar and low-dose rivaroxaban safety and efficacy profile, the clinical outcomes associ­
have not been tested as adjunctive therapy to prasugrel or ticagrelor. GPI, ated with this drug combination and how it compares
glycoprotein IIb/IIIa inhibitor; LD, loading dose; PCI, percutaneous coronary with other strategies has not been tested in adequately
intervention; UFH, unfractionated heparin. ­powered studies.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Among oral antiplatelet agents, aspirin at a loading-­ Indeed, the ability of a given health-care system or
dose regimen (325 mg) should be administered immedi‑ STEMI network to make an accurate diagnosis of STEMI
ately at time of clinical presentation, unless available in an might also define whether patients should be pretreated
intravenous formulation which can also be considered as with an oral P2Y12-receptor inhibitor. Indeed, this con‑
a route of administration. The P2Y12-receptor inhibitors sideration is particularly relevant in the USA, where
prasugrel and ticagrelor should be the preferred choice, almost one-third of STEMI activations to date have
with clopidogrel use reserved for patients in whom both been false activations148,149. Details on DAPT duration go
these more potent agents are contraindicated. On the beyond the scope of this manuscript and are described
basis of the results of pharmaco­kinetic and pharmaco‑ elsewhere150. In brief, regardless of the choice of P2Y12-
dynamic studies, the use of crushed tablets should be receptor inhibitor, current guidelines recommend that
considered to enhance bioavailability in the early hours DAPT should be continued for at least 12 months in
after the loading dose. Data from pharmaco­dynamic patients with STEMI. Discontinuation can be considered
studies comparing prasu­grel and ticagrelor do not sug‑ after 6 months in patients with overt bleeding or who
gest superiority of one agent over the other121,122,139–142. are at high risk of bleeding, whereas prolonging DAPT
In the randomized PRAGUE‑18 study143, the efficacy beyond 12 months is a reasonable option in selected
and safety of prasugrel and ticagrelor were compared in patients without overt bleeding or who are at low risk of
patients with acute MI (n = 1,230) treated with primary bleeding while receiving DAPT150,151.
or immediate PCI. Although no differ­ence between the
two agents in the 30‑day incidence of MACE (~4% in Conclusions
both groups) was shown, the study was terminated early Antithrombotic therapy has a pivotal role in the treat‑
for futility and was underpowered to detect differences in ment of patients with STEMI undergoing PPCI. Several
clinical events and, therefore, the results should be con‑ anticoagulant and antiplatelet agents are currently avail‑
sidered as hypothesis-­generating. Indeed, in the absence able, and finding the optimal cocktail is a clinical chal‑
of adequately powered clinical ­trials, the choice between lenge that needs to be tailored for each patient. The use
prasugrel and ticagrelor should be based on clinical of intravenous anticoagulant agents is mandatory during
grounds, taking into consideration patient characteris‑ PPCI, and the choice between a heparin and bivaliru‑
tics, presence of contraindications, and ease of access to din should be on the basis of the patient’s haemorrhagic
a specific med­ication. Ongoing clinical investigations, risk and, for some centres, financial resources given the
such as the ISAR-REACT 5 trial144, might provide further higher costs of bivalirudin. DAPT with a combination
insights into this topic. The use of tailored antiplatelet of aspirin and either prasugrel or ticagrelor is the anti‑
therapy on the basis of results of platelet-function and platelet treatment of choice, as these agents achieve more
genetic testing has also been investi­gated to optimize prompt and potent platelet inhibition compared with
outcomes in patients with coro­nary artery disease, clopidogrel, and improve clinical outcomes. Nonetheless,
including those with STEMI. Data available so far from patients continue to experience ischaemic events despite
randomized clinical trials do not support the routine use COX1 and P2Y12-receptor blockade, which has raised
of platelet-­function monitoring145,146, whereas the role of interest in alternative or adjunctive antithrombotic strat‑
genetic testing specifically in STEMI is being tested in an egies. The use of intravenous cangrelor is an attractive
­ongoing clinical trial147. strategy to achieve immediate and potent platelet inhib­
Despite the absence of strong evidence supporting ition during PPCI, whereas the use of agents target­
the ischaemic benefit of pretreatment, no data exist ing thrombin-mediated effects on platelet activation,
suggesting harm. However, pretreatment might expose namely vorapaxar and low-dose rivaroxaban, might be
patients to unnecessary platelet inhibition and related useful in selected patients in the secondary prevention
bleeding risk when PPCI is not performed, such as in of long-term adverse events. However, further studies
patients found not to have coronary artery disease pre‑ are warranted to explore the efficacy and safety profiles
senting with other causes of chest pain (such as a­ ortic of these strategies when used in addition to prasugrel
dissection) or patients requiring CABG surgery130. or ticagrelor.

1. O’Gara, P. T. et al. 2013 ACCF/AHA guideline for myocardial revascularization of the European Society 9. Franchi, F. & Angiolillo, D. J. Novel antiplatelet agents
the management of ST‑elevation myocardial infarction: of Cardiology (ESC) and the European Association for in acute coronary syndrome. Nat. Rev. Cardiol. 12,
a report of the American College of Cardiology Cardio-Thoracic Surgery (EACTS) developed with the 30–47 (2015).
Foundation/American Heart Association Task special contribution of the European Association of 10. Brass, L. F. Thrombin and platelet activation. Chest
Force on Practice Guidelines. J. Am. Coll. Cardiol. 61, Percutaneous Cardiovascular Interventions (EAPCI). 124, 18S–25S (2003).
e78–e140 (2013). Eur. Heart J. 35, 2541–2619 (2014). 11. Brummel, K. E., Paradis, S. G., Butenas, S.
2. Levine, G. N. et al. 2011 ACCF/AHA/SCAI guideline 5. Falk, E., Nakano, M., Bentzon, J. F., Finn, A. V. & Mann, K. G. Thrombin functions during tissue
for percutaneous coronary intervention. A report & Virmani, R. Update on acute coronary syndromes: factor-induced blood coagulation. Blood 100,
of the American College of Cardiology Foundation/ the pathologists’ view. Eur. Heart J. 34, 719–728 148–152 (2002).
American Heart Association Task Force on Practice (2013). 12. Coughlin, S. R. Protease-activated receptors in
Guidelines and the Society for Cardiovascular 6. Libby, P. Mechanisms of acute coronary syndromes hemostasis, thrombosis and vascular biology.
Angiography and Interventions. J. Am. Coll. Cardiol. and their implications for therapy. N. Engl. J. Med. J. Thromb. Haemost. 3, 1800–1814 (2005).
58, e44–e122 (2011). 368, 2004–2013 (2013). 13. Angiolillo, D. J., Capodanno, D. & Goto, S. Platelet
3. Steg, P. G. et al. ESC Guidelines for the management 7. Davì, G. & Patrono, C. Platelet activation and thrombin receptor antagonism and atherothrombosis.
of acute myocardial infarction in patients presenting atherothrombosis. N. Engl. J. Med. 357, 2482–2494 Eur. Heart J. 31, 17–28 (2010).
with ST‑segment elevation. Eur. Heart J. 33, (2007). 14. Yusuf, S. et al. Effects of fondaparinux on mortality
2569–2619 (2012). 8. Angiolillo, D. J., Ueno, M. & Goto, S. Basic principles and reinfarction in patients with acute ST‑segment
4. Windecker, S. et al. 2014 ESC/EACTS Guidelines of platelet biology and clinical implications. Circ. J. 74, elevation myocardial infarction: the OASIS‑6
on myocardial revascularization: the Task Force on 597–607 (2010). randomized trial. JAMA 295, 1519–1530 (2006).

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

15. Garcia, D. A., Baglin, T. P., Weitz, J. I. 35. Schulz, S. et al. Prasugrel plus bivalirudin versus presenting to nonpercutaneous coronary intervention
& Samama, M. M. Parenteral anticoagulants: clopidogrel plus heparin in patients with ST‑segment hospitals. JACC Cardiovasc. Interv. 2, 917–924
antithrombotic therapy and prevention of thrombosis, elevation myocardial infarction. Eur. Heart J. 35, (2009).
9th ed: American College of Chest Physicians 2285–2294 (2014). 53. Franchi, F., Rollini, F., Muñiz-Lozano, A., Cho, J. R.
Evidence-Based Clinical Practice Guidelines. Chest 36. Valgimigli, M. et al. Bivalirudin or unfractionated & Angiolillo, D. J. Cangrelor: a review on
141, e24S–e43S (2012). heparin in acute coronary syndromes. N. Engl. J. Med. pharmacology and clinical trial development.
16. Salter, B. S. et al. Heparin-induced thrombocytopenia: 373, 997–1009 (2015). Expert Rev. Cardiovasc. Ther. 11, 1279–1291 (2013).
a comprehensive clinical review. J. Am. Coll. Cardiol. 37. Leonardi, S. et al. Bivalirudin or unfractionated 54. Franchi, F., Rollini, F., Park, Y. & Angiolillo, D. J.
67, 2519–2532 (2016). heparin in patients with acute coronary syndromes A safety evaluation of cangrelor in patients undergoing
17. Boneu, B. et al. Pharmacokinetic studies of standard managed invasively with and without ST elevation PCI. Expert. Opin. Drug Saf. 15, 275–285 (2016).
unfractionated heparin, and low molecular weight (MATRIX): randomised controlled trial. BMJ 354, 55. Bhatt, D. L. et al. Effect of platelet inhibition with
heparins in the rabbit. Semin. Thromb. Hemost 14, i4935 (2016). cangrelor during PCI on ischemic events. N. Engl.
18–27 (1988). 38. Capodanno, D. et al. Bivalirudin versus heparin with J. Med. 368, 1303–1313 (2013).
18. Palm, M. & Mattsson, C. Pharmacokinetics of heparin or without glycoprotein IIb/IIIa inhibitors in patients 56. Généreux, P. et al. Impact of intraprocedural stent
and low molecular weight heparin fragment (Fragmin) with STEMI undergoing primary PCI: An updated thrombosis during percutaneous coronary
in rabbits with impaired renal or metabolic clearance. meta-analysis of 10,350 patients from five intervention: insights from the CHAMPION PHOENIX
Thromb. Res. 40, 129–133 (1985). randomized clinical trials. Eur. Heart J. Acute Trial (Clinical Trial Comparing Cangrelor to Clopidogrel
19. Montalescot, G. et al. Intravenous enoxaparin or Cardiovasc. Care 5, 253–262 (2016). Standard of Care Therapy in Subjects Who Require
unfractionated heparin in primary percutaneous 39. Shah, R., Rogers, K. C., Matin, K., Askari, R. Percutaneous Coronary Intervention). J. Am. Coll.
coronary intervention for ST‑elevation myocardial & Rao, S. V. An updated comprehensive meta-analysis Cardiol. 63, 619–629 (2014).
infarction: the international randomised open-label of bivalirudin versus heparin use in primary 57. White, H. D. et al. Outcomes with cangrelor versus
ATOLL trial. Lancet 378, 693–703 (2011). percutaneous coronary intervention. Am. Heart J. clopidogrel on a background of bivalirudin: insights
20. Arsenault, K. A. et al. Direct thrombin inhibitors in 171, 14–24 (2016). from the CHAMPION PHOENIX (a clinical trial
cardiovascular disease. Nat. Rev. Cardiol. 9, 402–414 40. Muñiz-Lozano, A., Rollini, F., Franchi, F. comparing cangrelor to clopidogrel standard therapy
(2012). & Angiolillo, D. J. Update on platelet glycoprotein IIb/ in subjects who require percutaneous coronary
21. Kastrati, A. et al. Abciximab and heparin versus IIIa inhibitors: recommendations for clinical practice. intervention [PCI]). JACC Cardiovasc. Interv. 8,
bivalirudin for non‑ST‑elevation myocardial Ther. Adv. Cardiovasc. Dis. 7, 197–213 (2013). 424–433 (2015).
infarction. N. Engl. J. Med. 365, 1980–1989 41. Stone, G. W. et al. Comparison of angioplasty 58. Vaduganathan, M. et al. Evaluation of ischemic
(2011). with stenting, with or without abciximab, in acute and bleeding risks associated with 2 parenteral
22. Stone, G. W. et al. Bivalirudin for patients with myocardial infarction. N. Engl. J. Med. 346, 957–966 antiplatelet strategies comparing cangrelor with
acute coronary syndromes. N. Engl. J. Med. 355, (2002). glycoprotein IIb/IIIa inhibitors: an exploratory analysis
2203–2216 (2006). 42. Mehilli, J. et al. Abciximab in patients with acute from the CHAMPION trials. JAMA Cardiol. http://
23. Stone, G. W. et al. Bivalirudin during primary PCI ST‑segment-elevation myocardial infarction undergoing dx.doi.org/10.1001/jamacardio.2016.4556 (2016).
in acute myocardial infarction. N. Engl. J. Med. 358, primary percutaneous coronary intervention after 59. US Food and Drug Administration. Highlights of
2218–2230 (2008). clopidogrel loading: a randomized double-blind trial. prescribing information (cangrelor). FDA
24. Mehran, R. et al. Bivalirudin in patients undergoing Circulation 119, 1933–1940 (2009). www.accessdata.fda.gov/drugsatfda_docs/
primary angioplasty for acute myocardial infarction 43. Antoniucci, D. et al. A randomized trial comparing label/2015/204958lbl.pdf (2016).
(HORIZONS-AMI): 1‑year results of a randomised primary infarct artery stenting with or without 60. European Medicines Agency. Summary of product
controlled trial. Lancet 374, 1149–1159 (2009). abciximab in acute myocardial infarction. J. Am. Coll. characteristics (cangrelor). EMA www.ema.europa.eu/
25. Stone, G. W. et al. Heparin plus a glycoprotein IIb/IIIa Cardiol. 42, 1879–1885 (2003). docs/en_GB/document_library/EPAR_-_Product_
inhibitor versus bivalirudin monotherapy and 44. Van’t Hof, A. W. et al. Prehospital initiation of tirofiban Information/human/003773/WC500188098.pdf
paclitaxel-eluting stents versus bare-metal stents in patients with ST‑elevation myocardial infarction (2016).
in acute myocardial infarction (HORIZONS‑AMI): undergoing primary angioplasty (On‑TIME 2): 61. Roffi, M. et al. 2015 ESC Guidelines for the
final 3‑year results from a multicentre, randomised a multicentre, double-blind, randomised controlled management of acute coronary syndromes in patients
controlled trial. Lancet 377, 2193–2204 (2011). trial. Lancet 372, 537–546 (2008). presenting without persistent ST‑segment elevation:
26. Dangas, G. D. et al. Effect of switching antithrombin 45. Zeymer, U. et al. Randomized comparison of Task Force for the management of acute coronary
agents for primary angioplasty in acute myocardial eptifibatide versus abciximab in primary syndromes in patients presenting without persistent
infarction: the HORIZONS-SWITCH analysis. percutaneous coronary intervention in patients with ST‑segment elevation of the European Society of
J. Am. Coll. Cardiol. 57, 2309–2316 (2011). acute ST‑segment elevation myocardial infarction: Cardiology (ESC). Eur. Heart J. 37, 267–315 (2016).
27. Dangas, G. D. et al. Frequency and predictors of stent results of the EVA-AMI Trial. J. Am. Coll. Cardiol. 56, 62. Angiolillo, D. J. et al. Pharmacodynamic effects of
thrombosis after percutaneous coronary intervention 463–469 (2010). cangrelor and clopidogrel: the platelet function
in acute myocardial infarction. Circulation 123, 46. Thiele, H. et al. Intracoronary compared with substudy from the cangrelor versus standard therapy
1745–1756 (2011). intravenous bolus abciximab application in patients to achieve optimal management of platelet inhibition
28. Koutouzis, M. et al. Unfractionated heparin with ST‑elevation myocardial infarction undergoing (CHAMPION) trials. J. Thromb. Thrombolysis 34,
administration in patients treated with bivalirudin primary percutaneous coronary intervention: the 44–55 (2012).
during primary percutaneous coronary intervention randomized Leipzig immediate percutaneous coronary 63. Rollini, F., Franchi, F. & Angiolillo, D. J. Switching
is associated lower mortality and target lesion intervention abciximab IV versus IC in ST‑elevation P2Y12‑receptor inhibitors in patients with coronary
thrombosis: a report from the Swedish Coronary myocardial infarction trial. Circulation 118, 49–57 artery disease. Nat. Rev. Cardiol. 13, 11–27 (2016).
Angiography and Angioplasty Registry (SCAAR). Heart (2008). 64. Steinhubl, S. R. et al. Transitioning patients from
97, 1484–1488 (2011). 47. Stone, G. W. et al. Intracoronary abciximab and cangrelor to clopidogrel: pharmacodynamic evidence
29. Steg, P. G. et al. Bivalirudin started during emergency aspiration thrombectomy in patients with large of a competitive effect. Thromb. Res. 121, 527–534
transport for primary PCI. N. Engl. J. Med. 369, anterior myocardial infarction: the INFUSE-AMI (2008).
2207–2217 (2013). randomized trial. JAMA 307, 1817–1826 (2012). 65. Dovlatova, N. L., Jakubowski, J. A., Sugidachi, A.
30. Clemmensen, P. et al. Acute stent thrombosis after 48. Valgimigli, M. et al. Prasugrel versus tirofiban bolus & Heptinstall, S. The reversible P2Y antagonist
primary percutaneous coronary intervention: insights with or without short post-bolus infusion with or cangrelor influences the ability of the active
from the EUROMAX trial (European Ambulance Acute without concomitant prasugrel administration in metabolites of clopidogrel and prasugrel to produce
Coronary Syndrome Angiography). JACC Cardiovasc. patients with myocardial infarction undergoing irreversible inhibition of platelet function. J. Thromb.
Interv. 8, 214–220 (2015). coronary stenting: the FABOLUS PRO (Facilitation Haemost. 6, 1153–1159 (2008).
31. Zeymer, U. et al. Bivalirudin is superior to heparins through Aggrastat By drOpping or shortening Infusion 66. Rollini, F. et al. Pharmacodynamic effects of cangrelor
alone with bailout GP IIb/IIIa inhibitors in patients Line in patients with ST‑segment elevation myocardial on platelet P2Y12 receptor-mediated signaling in
with ST‑segment elevation myocardial infarction infarction compared to or on top of PRasugrel given prasugrel-treated patients. JACC Cardiovasc. Interv. 7,
transported emergently for primary percutaneous at loading dOse) trial. JACC Cardiovasc. Interv. 5, 426–434 (2014).
coronary intervention: a pre-specified analysis from 268–277 (2012). 67. Schneider, D. J., Agarwal, Z., Seecheran, N. & Gogo, P.
the EUROMAX trial. Eur. Heart J. 35, 2460–2467 49. Thiele, H. et al. Intracoronary versus intravenous bolus Pharmacodynamic effects when clopidogrel is given
(2014). abciximab during primary percutaneous coronary before cangrelor discontinuation. J. Interv. Cardiol.
32. Stone, G. W. et al. Bivalirudin versus heparin with or intervention in patients with acute ST‑elevation 28, 415–419 (2015).
without glycoprotein IIb/IIIa inhibitors in patients with myocardial infarction: a randomised trial. Lancet 379, 68. Schneider, D. J., Seecheran, N., Raza, S. S.,
STEMI undergoing primary percutaneous coronary 923–931 (2012). Keating, F. K. & Gogo, P. Pharmacodynamic effects
intervention: pooled patient-level analysis from the 50. De Luca, G. et al. Early glycoprotein IIb-IIIa inhibitors during the transition between cangrelor and
HORIZONS-AMI and EUROMAX trials. J. Am. Coll. in primary angioplasty-abciximab long-term results prasugrel. Coron. Artery Dis. 26, 42–48 (2015).
Cardiol. 65, 27–38 (2015). (EGYPT-ALT) cooperation: individual patient’s data 69. Schneider, D. J., Agarwal, Z., Seecheran, N.,
33. Shahzad, A. et al. Unfractionated heparin versus meta-analysis. J. Thromb. Haemost. 9, 2361–2370 Keating, F. K. & Gogo, P. Pharmacodynamic effects
bivalirudin in primary percutaneous coronary (2011). during the transition between cangrelor and ticagrelor.
intervention (HEAT-PPCI): an open-label, single centre, 51. Gershlick, A. H., Banning, A. P., Myat, A., JACC Cardiovasc. Interv. 7, 435–442 (2014).
randomised controlled trial. Lancet 384, 1849–1858 Verheugt, F. W. & Gersh, B. J. Reperfusion therapy 70. Patrono, C., García Rodríguez, L. A., Landolfi, R.
(2014). for STEMI: is there still a role for thrombolysis in the & Baigent, C. Low-dose aspirin for the prevention of
34. Han, Y. et al. Bivalirudin versus heparin with or era of primary percutaneous coronary intervention? atherothrombosis. N. Engl. J. Med. 353, 2373–2383
without tirofiban during primary percutaneous Lancet 382, 624–632 (2013). (2005).
coronary intervention in acute myocardial infarction: 52. Herrmann, H. C. et al. Benefit of facilitated 71. Capodanno, D. & Angiolillo, D. J. Aspirin for primary
the BRIGHT randomized clinical trial. JAMA 313, percutaneous coronary intervention in high-risk cardiovascular risk prevention and beyond in diabetes
1336–1346 (2015). ST‑segment elevation myocardial infarction patients mellitus. Circulation 134, 1579–1594 (2016).

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 17


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

72. ISIS‑2 (Second International Study of Infarct Survival) adenosine diphosphate associated with ischemia 113. Oldgren, J. et al. New oral anticoagulants in addition
Collaborative Group. Randomised trial of intravenous and bleeding. J. Am. Coll. Cardiol. 62, 2261–2273 to single or dual antiplatelet therapy after an
streptokinase, oral aspirin, both, or neither among (2013). acute coronary syndrome: a systematic review and
17,187 cases of suspected acute myocardial 94. Sugidachi, A. et al. The greater in vivo antiplatelet meta‑analysis. Eur. Heart J. 34, 1670–1680 (2013).
infarction: ISIS‑2. Lancet 2, 349–360 (1988). effects of prasugrel as compared to clopidogrel 114. Mega, J. L. et al. Rivaroxaban in patients with a recent
73. Campbell, C. L., Smyth, S., Montalescot, G. reflect more efficient generation of its active acute coronary syndrome. N. Engl. J. Med. 366, 9–19
& Steinhubl, S. R. Aspirin dose for the prevention metabolite with similar antiplatelet activity to that (2012).
of cardiovascular disease: a systematic review. of clopidogrel’s active metabolite. J. Thromb. Haemost. 115. Mega, J. L. et al. Rivaroxaban in patients stabilized
JAMA 297, 2018–2024 (2007). 5, 1545–1551 (2007). after a ST‑segment elevation myocardial infarction:
74. Mehta, S. R. et al. Dose comparisons of clopidogrel 95. Wiviott, S. D. et al. Prasugrel compared with high results from the ATLAS ACS‑2‑TIMI‑51 trial (Anti‑Xa
and aspirin in acute coronary syndromes. N. Engl. loading- and maintenance-dose clopidogrel in patients therapy to lower cardiovascular events in addition to
J. Med. 363, 930–942 (2010). with planned percutaneous coronary intervention: standard therapy in subjects with acute coronary
75. Storey, R. F., Newby, L. J. & Heptinstall, S. Effects of the prasugrel in comparison to clopidogrel for syndrome-thrombolysis in myocardial infarction‑51).
P2Y 1 and P2Y 12 receptor antagonists on platelet inhibition of platelet activation and aggregation J. Am. Coll. Cardiol. 61, 1853–1859 (2013).
aggregation induced by different agonists in human thrombolysis in myocardial infarction 44 trial. 116. Povsic, T. J. et al. A randomized trial to compare the
whole blood. Platelets 12, 443–447 (2001). Circulation 116, 2923–2932 (2007). safety of rivaroxaban versus aspirin in addition to
76. Altman, R., Scazziota, A., Rouvier, J. & Gonzalez, C. 96. Montalescot, G. et al. Prasugrel compared with either clopidogrel or ticagrelor in acute coronary
Effects of ticlopidine or ticlopidine plus aspirin on platelet clopidogrel in patients undergoing percutaneous syndrome: the design of the GEMINI-ACS‑1 phase II
aggregation and ATP release in normal volunteers: coronary intervention for ST‑elevation myocardial study. Am. Heart J. 174, 120–128 (2016).
why aspirin improves ticlopidine antiplatelet activity. infarction (TRITON-TIMI 38): double-blind, 117. European Medicines Agency. Summary of product
Clin. Appl. Thromb. Hemost. 5, 243–246 (1999). randomised controlled trial. Lancet 373, 723–731 characteristics (rivaroxaban). EMA www.ema.europa.
77. Schömig, A. et al. A randomized comparison of (2009). eu/docs/en_GB/document_library/EPAR_-_Product_
antiplatelet and anticoagulant therapy after the 97. Udell, J. A. et al. Prasugrel versus clopidogrel in Information/human/000944/WC500057108.pdf
placement of coronary-artery stents. N. Engl. J. Med. patients with ST‑segment elevation myocardial (2016).
334, 1084–1089 (1996). infarction according to timing of percutaneous 118. Gibson, C. M. et al. Prevention of bleeding in patients
78. Bertrand, M. E. et al. Randomized multicenter coronary intervention: a TRITON-TIMI 38 subgroup with atrial fibrillation undergoing PCI. N. Engl. J. Med.
comparison of conventional anticoagulation versus analysis (Trial to Assess Improvement in Therapeutic 375, 1131–1141 (2016).
antiplatelet therapy in unplanned and elective Outcomes by Optimizing Platelet Inhibition with 119. Angiolillo, D. J. et al. Antithrombotic therapy
coronary stenting. The full anticoagulation versus Prasugrel-Thrombolysis In Myocardial Infarction 38). in patients with atrial fibrillation undergoing
aspirin and ticlopidine (FANTASTIC) study. Circulation JACC Cardiovasc. Interv. 7, 604–612 (2014). percutaneous coronary intervention:
98, 1597–1603 (1998). 98. Gurbel, P. A. et al. Randomized double-blind a North American perspective — 2016 update.
79. Leon, M. B. et al. A clinical trial comparing three assessment of the ONSET and OFFSET of the Circ. Cardiovasc. Interv. 9, e004395 (2016).
antithrombotic-drug regimens after coronary-artery antiplatelet effects of ticagrelor versus clopidogrel 120. Rollini, F. & Franchi, F. The conundrum of platelet
stenting. Stent Anticoagulation Restenosis Study in patients with stable coronary artery disease: the P2Y12 inhibition in ST‑segment elevation myocardial
Investigators. N. Engl. J. Med. 339, 1665–1671 ONSET/OFFSET study. Circulation 120, 2577–2585 infarction. Circ. J. 80, 2429–2431 (2016).
(1998). (2009). 121. Alexopoulos, D. et al. Randomized assessment
80. Urban, P. et al. Randomized evaluation of 99. Cattaneo, M., Schulz, R. & Nylander, S. of ticagrelor versus prasugrel antiplatelet effects
anticoagulation versus antiplatelet therapy after Adenosine‑mediated effects of ticagrelor: evidence in patients with ST‑segment-elevation myocardial
coronary stent implantation in high-risk patients: and potential clinical relevance. J. Am. Coll. Cardiol. infarction. Circ. Cardiovasc. Interv. 5, 797–804
the multicenter aspirin and ticlopidine trial after 63, 2503–2509 (2014). (2012).
intracoronary stenting (MATTIS). Circulation 98, 100. Bonaca, M. P. et al. Long-term use of ticagrelor in 122. Parodi, G. et al. Comparison of prasugrel and
2126–2132 (1998). patients with prior myocardial infarction. N. Engl. ticagrelor loading doses in ST‑segment elevation
81. Yusuf, S. et al. Effects of clopidogrel in addition to J. Med. 372, 1791–1800 (2015). myocardial infarction patients: RAPID (Rapid Activity
aspirin in patients with acute coronary syndromes 101. Bonaca, M. P. et al. Long-term tolerability of ticagrelor of Platelet Inhibitor Drugs) primary PCI study. J. Am.
without ST‑segment elevation. N. Engl. J. Med. 345, for the secondary prevention of major adverse Coll. Cardiol. 61, 1601–1606 (2013).
494–502 (2001). cardiovascular events: a secondary analysis of the 123. Franchi, F. et al. Impact of escalating loading dose
82. Steinhubl, S. R. et al. Early and sustained dual oral PEGASUS-TIMI 54 trial. JAMA Cardiol. 1, 425–432 regimens of ticagrelor in patients with ST‑segment
antiplatelet therapy following percutaneous coronary (2016). elevation myocardial infarction undergoing primary
intervention: a randomized controlled trial. JAMA 102. Steg, P. G. et al. Ticagrelor versus clopidogrel in percutaneous coronary intervention: results of
288, 2411–2420 (2002). patients with ST‑elevation acute coronary syndromes a prospective randomized pharmacokinetic and
83. Sabatine, M. S. et al. Addition of clopidogrel to aspirin intended for reperfusion with primary percutaneous pharmacodynamic investigation. JACC Cardiovasc.
and fibrinolytic therapy for myocardial infarction coronary intervention: a Platelet Inhibition and Patient Interv. 8, 1457–1467 (2015).
with ST‑segment elevation. N. Engl. J. Med. 352, Outcomes (PLATO) trial subgroup analysis. Circulation 124. Alexopoulos, D. et al. Double versus standard loading
1179–1189 (2005). 122, 2131–2141 (2010). dose of ticagrelor: onset of antiplatelet action in
84. Chen, Z. M. et al. Addition of clopidogrel to aspirin 103. Velders, M. A. et al. Safety and efficacy of ticagrelor patients with STEMI undergoing primary PCI. J. Am.
in 45 852 patients with acute myocardial infarction: and clopidogrel in primary percutaneous coronary Coll. Cardiol. 62, 940–941 (2013).
randomised placebo-controlled trial. Lancet 366, intervention. Heart 102, 617–625 (2016). 125. Alexopoulos, D. et al. Onset of antiplatelet action
1607–1621 (2005). 104. Wisler, J. W. & Becker, R. C. Oral factor Xa inhibitors with high (100 mg) versus standard (60 mg) loading
85. Wiviott, S. D. et al. Prasugrel versus clopidogrel for the long-term management of ACS. Nat. Rev. dose of prasugrel in patients with ST‑segment-
in patients with acute coronary syndromes. N. Engl. Cardiol. 9, 392–401 (2012). elevation myocardial infarction undergoing
J. Med. 357, 2001–2015 (2007). 105. Brummel-Ziedins, K. et al. Thrombin generation in primary percutaneous coronary intervention:
86. Wallentin, L. et al. Ticagrelor versus clopidogrel acute coronary syndrome and stable coronary artery pharmacodynamic study. Circ. Cardiovasc. Interv. 7,
in patients with acute coronary syndromes. N. Engl. disease: dependence on plasma factor composition. 233–239 (2014).
J. Med. 361, 1045–1057 (2009). J. Thromb. Haemost. 6, 104–110 (2008). 126. Parodi, G. et al. Comparison of double (360 mg)
87. Love, B., Biller, J. & Gent, M. Adverse haematological 106. Franchi, F., Rollini, F., Park, Y. & Angiolillo, D. J. ticagrelor loading dose with standard (60 mg)
effects of ticlopidine: prevention, recognition Platelet thrombin receptor antagonism with prasugrel loading dose in ST‑elevation myocardial
and management. Drug Saf. 19, 89–98 (1998). vorapaxar: pharmacology and clinical trial infarction patients: the Rapid Activity of Platelet
88. Bhatt, D. L. et al. Meta-analysis of randomized and development. Future Cardiol. 11, 547–564 (2015). Inhibitor Drugs (RAPID) primary PCI 2 study.
registry comparisons of ticlopidine with clopidogrel 107. Tricoci, P. et al. Thrombin-receptor antagonist Am. Heart J. 167, 909–914 (2014).
after stenting. J. Am. Coll. Cardiol. 39, 9–14 (2002). vorapaxar in acute coronary syndromes. N. Engl. 127. Rollini, F. et al. Crushed prasugrel tablets in patients
89. Fan, W., Plent, S., Prats, J. & Deliargyris, E. N. Trends J. Med. 366, 20–33 (2012). with STEMI undergoing primary percutaneous
in P2Y12 inhibitor use in patients referred for invasive 108. Morrow, D. A. et al. Vorapaxar in the secondary coronary intervention: the CRUSH study. J. Am. Coll.
evaluation of coronary artery disease in contemporary prevention of atherothrombotic events. N. Engl. Cardiol. 67, 1994–2004 (2016).
US practice. Am. J. Cardiol. 117, 1439–1443 (2016). J. Med. 366, 1404–1413 (2012). 128. Parodi, G. et al. Ticagrelor crushed tablets
90. Sherwood, M. W. et al. Early clopidogrel versus 109. Morrow, D. A. et al. Efficacy and safety of vorapaxar administration in STEMI patients: the MOJITO study.
prasugrel use among contemporary STEMI and in patients with prior ischemic stroke. Stroke 44, J. Am. Coll. Cardiol. 65, 511–512 (2015).
NSTEMI patients in the US: insights from the National 691–698 (2013). 129. Stone, G. W. et al. Platelet reactivity and clinical
Cardiovascular Data Registry. J. Am. Heart Assoc. 3, 110. Scirica, B. M. et al. Vorapaxar for secondary outcomes after coronary artery implantation of drug-
e000849 (2014). prevention of thrombotic events for patients with eluting stents (ADAPT-DES): a prospective multicenter
91. Bueno, H. et al. Opportunities for improvement in previous myocardial infarction: a prespecified registry study. Lancet 382, 614–623 (2013).
anti-thrombotic therapy and other strategies for the subgroup analysis of the TRA 2°P‑TIMI 50 trial. 130. Capodanno, D. & Angiolillo, D. J. Pretreatment with
management of acute coronary syndromes: Insights Lancet 380, 1317–1324 (2012). antiplatelet drugs in invasively managed patients
from EPICOR, an international study of current 111. US Food and Drug Administration. Highlights with coronary artery disease in the contemporary era:
practice patterns. Eur. Heart J. Acute Cardiovasc. Care of prescribing information (vorapaxar). FDA review of the evidence and practice guidelines.
5, 3–12 (2016). www.accessdata.fda.gov/drugsatfda_docs/ Circ. Cardiovasc. Interv. 8, e002301 (2015).
92. Angiolillo, D. J. et al. Variability in individual label/2014/204886s000lbl.pdf (2016). 131. Bellemain-Appaix, A. et al. Association of clopidogrel
responsiveness to clopidogrel: clinical implications, 112. European Medicines Agency. Summary of product pretreatment with mortality, cardiovascular events,
management, and future perspectives. J. Am. Coll. characteristics (vorapaxar). EMA www.ema.europa.eu/ and major bleeding among patients undergoing
Cardiol. 49, 1505–1516 (2007). docs/en_GB/document_library/EPAR_-_Product_ percutaneous coronary intervention: a systematic
93. Tantry, U. S. et al. Consensus and update on the Information/human/002814/WC500183329.pdf review and meta-analysis. JAMA 308, 2507–2516
definition of on‑treatment platelet reactivity to (2016). (2012).

18 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

132. Montalescot, G. et al. Prehospital ticagrelor in the OPTIMUS (Optimizing Antiplatelet Therapy in 148. Mixon, T. A. et al. Retrospective description and
ST‑segment elevation myocardial infarction. N. Engl. Diabetes Mellitus)-4 study. Circulation 134, 780–792 analysis of consecutive catheterization laboratory
J. Med. 371, 1016–1027 (2014). (2016). ST‑segment elevation myocardial infarction activations
133. Silvain, J. et al. P2Y12 receptor inhibition and effect 141. Rafique, A. M. et al. Optimal P2Y12 inhibitor with proposal, rationale, and use of a new
of morphine in patients undergoing primary PCI in patients with ST‑segment elevation myocardial classification scheme. Circ. Cardiovasc. Qual.
for ST‑segment elevation myocardial infarction. infarction undergoing primary percutaneous Outcomes 5, 62–69 (2012).
The PRIVATE-ATLANTIC study. Thromb. Haemost. coronary intervention: a network meta-analysis. 149. McDaniel, M. Pre-hospital ticagrelor in ST‑segment
116, 369–378 (2016). JACC Cardiovasc. Interv. 9, 1036–1046 (2016). elevation myocardial infarction? Probably not.
134. Montalescot, G. et al. Effect of pre-hospital ticagrelor 142. Cuisset, T., Capodanno, D. & Wijns, W. Optimal P2Y12 JACC Cardiovasc. Interv. 9, 657–659 (2016).
during the first 24 h after primary percutaneous inhibitor for primary percutaneous coronary 150. Park, Y., Franchi, F., Rollini, F. & Angiolillo, D. J.
coronary intervention in patients with ST‑segment intervention in ST‑segment elevation myocardial Dual antiplatelet therapy after coronary stenting.
elevation myocardial infarction: the ATLANTIC-H24 infarction: network meta-analysis in the data-free Expert Opin. Pharmacother. 17, 1775–1787 (2016).
analysis. JACC Cardiovasc. Interv. 9, 646–656 (2016). zone: do you believe in magic? JACC Cardiovasc. 151. Levine, G. N. et al. 2016 ACC/AHA guideline
135. Parodi, G. et al. Morphine is associated with a delayed Interv. 9, 1047–1050 (2016). focused update on duration of dual antiplatelet therapy
activity of oral antiplatelet agents in patients with 143. Motovska, Z. et al. Prasugrel versus ticagrelor in in patients with coronary artery disease: a report of the
ST‑elevation acute myocardial infarction undergoing patients with acute myocardial infarction treated with American College of Cardiology/American Heart
primary percutaneous coronary intervention. primary percutaneous coronary intervention: Association Task Force on Clinical Practice Guidelines.
Circ. Cardiovasc. Interv. 8, e001593 (2014). multicenter randomized PRAGUE‑18 study. Circulation J. Am. Coll. Cardiol. 68, 1082–1115 (2016).
136. Silvain, J. et al. Efficacy and safety of enoxaparin 134, 1603–1612 (2016). 152. Sardella, G., Calcagno, S. & Mancone, M. Different
versus unfractionated heparin during percutaneous 144. Schulz, S. et al. Randomized comparison of ticagrelor prasugrel administration in STEMI patients: go faster
coronary intervention: systematic review and versus prasugrel in patients with acute coronary and no fear to crush! J. Am. Coll. Cardiol. 67,
meta‑analysis. BMJ 344, e553 (2012). syndrome and planned invasive strategy — design 2005–2007 (2016).
137. Alexander, K. P. et al. Excess dosing of antiplatelet and rationale of the iNtracoronary Stenting and
and antithrombin agents in the treatment of Antithrombotic Regimen: Rapid Early Action for Author contributions
non‑ST‑segment elevation acute coronary syndromes. Coronary Treatment (ISAR-REACT) 5 trial. F.F. and D.J.A. researched data for the article, discussed its
JAMA 294, 3108–3116 (2005). J. Cardiovasc. Transl. Res. 7, 91–100 (2014). content, and wrote the manuscript. F.R. discussed the content
138. Angiolillo, D. J. et al. Impact of cangrelor overdosing 145. Gross, L. & Sibbing, D. Current role of platelet function of the manuscript and reviewed/edited it before submission.
on bleeding complications in patients undergoing testing in percutaneous coronary intervention and
percutaneous coronary intervention: insights from coronary artery bypass grafting. Interv. Cardiol. Clin. Competing interests statement
the CHAMPION trials. J. Thromb. Thrombolysis 40, 6, 151–166 (2017). D.J.A. has received payment as an individual for consulting
317–322 (2015). 146. Franchi, F. et al. Platelet function testing in fees or honoraria from Abbott Vascular, Amgen, AstraZeneca,
139. Rollini, F. et al. A head‑to‑head pharmacodynamic contemporary clinical and interventional practice. Bayer, Daiichi-Sankyo, Eli Lilly, Merck, Pfizer, PLx Pharma,
comparison of prasugrel versus ticagrelor after Curr. Treat. Options Cardiovasc. Med. 16, 300 Sanofi, and The Medicines Company; and for participation in
switching from clopidogrel in patients with coronary (2014). review activities from CeloNova, Johnson & Johnson, and
artery disease: results of a prospective randomized 147. Bergmeijer, T. O. et al. CYP2C19 genotype-guided St. Jude Medical. Institutional payments for grants have been
study. Eur. Heart J. 37, 2722–2730 (2016). antiplatelet therapy in ST‑segment elevation received from AstraZeneca, CSL Behring, Daiichi-Sankyo, Eli
140. Franchi, F. et al. Pharmacodynamic comparison of myocardial infarction patients — rationale and design Lilly, GlaxoSmithKline, Janssen Pharmaceuticals, Gilead,
prasugrel versus ticagrelor in patients with type 2 of the patient outcome after primary PCI (POPular) Novartis, Osprey Medical, and The Medicines Company.
diabetes mellitus and coronary artery disease: genetics study. Am. Heart J. 168, 16–22.e1 (2014). The other authors declare no competing interests.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 19


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Anda mungkin juga menyukai