You are on page 1of 14

Therapeutic Advances in Hematology Review

The role of oral anticoagulant therapy in


Ther Adv Hematol

2017, Vol. 8(12) 353–366

patients with acute coronary syndrome DOI: 10.1177/


2040620717733691
© The Author(s), 2017.
Reprints and permissions:
Jae Youn Moon, Deepa Nagaraju, Francesco Franchi, Fabiana Rollini http://www.sagepub.co.uk/
journalsPermissions.nav
and Dominick J. Angiolillo

Abstract: Dual antiplatelet therapy (DAPT) with aspirin and a P2Y12 receptor antagonist
represents the current standard of care to prevent atherothrombotic recurrences in patients
with acute coronary syndrome (ACS). However, despite the use of DAPT, the recurrence
rate of cardiovascular ischemic events still remains high. This persistent risk may be in part
attributed to the sustained activation of the coagulation cascade leading to generation of
thrombin, which may continue to play a key role in thrombus formation. The use of vitamin
K antagonists (VKAs) as a strategy to reduce atherothrombotic recurrences after an ACS
has been previously tested, leading to overall unfavorable outcomes due to the high risk of
bleeding complications. The recent introduction of non-VKA oral anticoagulants (NOACs),
characterized by a better safety profile and ease of use compared with VKA, has led to
a reappraisal of the use of oral anticoagulant therapy for secondary prevention in ACS
patients. The present article provides an overview of the rationale and prognostic role of oral
anticoagulant therapy in ACS patients as well as recent updated clinical data, in particular
with NOACs, in the field and future perspectives on this topic.

Keywords: acute coronary syndrome, coagulation cascade, non-vitamin K antagonist oral


anticoagulants, thrombin

Received: 23 May 2017; revised manuscript accepted: 4 September 2017.

Introduction patients with non-ST-segment elevation ACS


Cardiovascular disease in general and the devel- (NSTE-ACS) are more commonly characterized
opment of an acute coronary syndrome (ACS) in by partial vessel occlusion.2
Correspondence to:
particular is among the leading causes of morbid- Dominick J. Angiolillo
ity and mortality.1 The main pathophysiological These pathophysiological considerations under- University of Florida
College of Medicine –
mechanism leading to an ACS is represented by score how inhibition of both platelets and coagu- Jacksonville, 655 West 8th
thrombus formation in the coronary artery fol- lation factors are essential for the treatment and Street, Jacksonville, FL
32209, USA
lowing erosion or rupture of an atherosclerotic secondary prevention of ACS patients. Dual anti- dominick.angiolillo@jax.
plaque.2 After initial plaque erosion/rupture, a platelet therapy (DAPT) including aspirin and a ufl.edu

cascade of events driven by cellular (i.e. platelets) P2Y12 receptor antagonist represents the gold Jae Youn Moon
Division of Cardiology,
and plasma (i.e. coagulation factors) components standard antithrombotic treatment regimen to pre- University of Florida
leading to thrombus formation has a pivotal role vent atherothrombotic recurrences.5,6 Currently, College of Medicine
– Jacksonville, FL, USA
in this process. Thrombus formation in the coro- three oral P2Y12 receptor inhibitors including Department of Cardiology,
nary artery ultimately culminates in complete or clopidogrel, prasugrel and ticagrelor are approved CHA Bundang Medical
Center, CHA University,
partial vessel occlusion contributing to the con- for clinical use in ACS patients.7–9 Prasugrel and Seongnam, Korea
stellation of signs and symptoms that characterize ticagrelor are characterized by enhanced potency Deepa Nagaraju
Francesco Franchi
patients presenting with an ACS. 2–4 In particular, over clopidogrel and better net clinical outcomes Fabiana Rollini
most patients presenting with an ST-segment- in ACS patients.7,8 Accordingly, guidelines sup- Division of Cardiology,
elevation myocardial infarction (STEMI) have a port their preferential use in ACS settings.10–13 University of Florida
College of Medicine –
completely occluded coronary artery, while Anticoagulant therapy, typically parenteral Jacksonville, FL, USA

journals.sagepub.com/home/tah 353
Therapeutic Advances in Hematology 8(12)

agents, is commonly used in the acute phase of induces activation of the coagulation cascade
ACS presentations. However, following the intro- which leads to thrombin generation.4,19 Tissue
duction of DAPT, the use of oral anticoagulant factor (TF), locally exposed to the plasma at the
therapy, in particular vitamin K antagonists site of plaque rupture, binds to Factor VIIa
(VKAs), for long-term management of ACS (FVIIa) to form an activated TF/FVIIa complex.
patients has been largely abandoned. Nonetheless, This complex activates Factor IX and Factor X
the recent introduction of non-VKA oral antico- into Factor IXa and Factor Xa. The following
agulants (NOACs), characterized by a better combination of Factor IXa and Factor VIIIa form
safety profile and ease of use compared with the intrinsic tenase (FVIIIa/FIXa), which con-
VKAs, has led to a reappraisal of the option of tributes to the conversion of Factor X to Factor
using oral anticoagulant therapy for secondary Xa (FXa). FXa and Factor Va lead to formation
prevention in ACS patients.14 of the prothrombinase complex (FVa/FXa),
which cleaves Factor II (prothrombin) to gener-
The present manuscript will present an overview ate Factor IIa (thrombin).20,21 In the propagation
of the rationale and potential role of oral antico- phase of the coagulation cascade, both the FVIIIa/
agulant therapy in patients with ACS, as well as FIXa and FVa/FXa complexes gather on the acti-
the available clinical data in the field and future vated platelet surface and promote the burst in
perspectives on this topic. The use of oral anti- thrombin generation (Figure 2). Thrombin is a
coagulants for other indications, including atrial serine protease and has multifunctional proper-
fibrillation (AF), among patients with coronary ties in thrombus formation. Thrombin mediates
artery disease (CAD) will not elaborated in this the conversion of fibrinogen to fibrin, which is
manuscript and is described in details essential to form a thrombus. In particular, fibrin
elsewhere.15,16 monomers polymerize to form a fibrin-rich clot
and this clot is stabilized by crosslinking with
Factor XIIIa. In addition, thrombin plays an
Platelet activation and coagulation cascade essential role in platelet activation via binding to
Thrombus formation after rupture or erosion of protease activator receptors (PARs)-1 on the
an atherosclerotic plaque in the arterial wall is platelet membrane.22,23
known to be the main pathophysiological mecha-
nism leading to the development of an ACS. 2–4
After plaque rupture or superficial erosion, a cas- Rationale for anticoagulation therapy in ACS
cade of events consisting of platelet adhesion, patients
activation and aggregation in conjunction with Thrombin generation plays a key role in the
thrombin formation contributes to thrombus for- pathophysiology of ACS. In particular, thrombin
mation and vascular occlusion.2–4 In particular, contributes to platelet activation, enhancing acti-
von Willebrand factor (vWF) that binds to the vation of the coagulation cascade, overall favoring
collagen on the exposed extracellular matrix at thrombus formation and its associated complica-
the site of plaque rupture or erosion interacts with tions.22 Therefore, anticoagulant therapy is criti-
the glycoprotein (GP) Ib/V/IX receptor complex cal in the acute care of ACS patients. Currently,
on the platelet surface.3,4,17,18 Subsequently, col- various parenteral anticoagulant agents are avail-
lagens from the subendothelium at the site of vas- able for clinical use in the acute setting of ACS.
cular injury bind to GP VI on the platelet surface. These include heparin (unfractionated form and
This leads to conformational changes in platelet low molecular-weight heparin), bivalirudin (a
structure and release of various factors which pro- direct thrombin inhibitor) and fondaparinux (a
mote platelet activation, including thromboxane direct Factor Xa inhibitor).24 However, these
A2 (TxA2), adenosine diphosphate (ADP) and agents are used only for a short period of time,
thrombin.3,4,18 The final process of platelet activa- essentially for the in-hospital phase of care of
tion is represented by the conformational change ACS patients. Conversely, the use of oral antico-
of the platelet GP IIb/IIIa receptor to its active agulant drugs has not traditionally been part of
form, which allows it to bind to fibrinogen, pro- the long-term management of ACS patients, par-
moting platelet aggregation and contributing to ticularly after the introduction of current regi-
final thrombus formation (Figure 1). mens of standard DAPT.

In parallel with the platelet cascade described The rationale for supporting the use of DAPT in
above, the exposed extracellular matrix also patients experiencing an ACS is represented by

354 journals.sagepub.com/home/tah
J Y Moon, D Nagaraju et al.

Figure 1. The process of platelet adhesion and aggregation.


The interactions between the GP Ib/V/IX receptor complex in platelet surface and vWF that immobilized on the collagen of
exposed extracellular matrix lead to subsequent binding between GP VI and exposed collagen. This interaction contributes
to the conformational change in platelet structure and release of TxA2, ADP and thrombin. TF locally triggers thrombin
formation, which bind to the platelet PAR-1 and this leads to enhance platelet activation. The released mediators carry
out their role in the activation of platelets via their platelet activation pathway. The final process of platelet activation is
the conformal change of platelet GP IIb/IIIa receptor to its active form, which binds to fibrinogen and contributes to final
thrombus formation. Reproduced with permission from Circulation Journal.4
ADP, adenosine diphosphate; GP, glycoprotein; PAR-1, protease activated receptor-1; thromboxane prostanoid receptor; TF,
tissue factor; TP receptor; TxA2, thromboxane A2; vWF, von Willebrand factor.

Figure 2. The cell surface-based coagulation process.


In the initiation phase, TF exposed to plasma at the plaque rupture site binds to Factor VIIa. Platelets are activated by vessel injury
simultaneously. The TF/Factor VIIa (TF/FVIIa) complex activates Factor X and Factor IX into Factor Xa and Factor IXa, and small
amounts of thrombinare generated by Factor Xa. In the amplification phase, thrombin activates Factor V, Factor VIII and Factor
XI, located on the platelets’ surfaces. In the propagation phase of the coagulation cascade, both Factor VIIIa/Factor IXa (FVIIIa/
FIXa) complex and Factor Va/Factor Xa (FVa/FXa) complex gather on activated platelet surfaces and leads to the burst of thrombin,
which is sufficient for fibrin monomers to polymerize to form a fibrin-rich clot. Finally, this clot is stabilized by crosslinking with
Factor XIIIa; thereafter, a thrombus is formed. Reproduced with permission from Thrombosis and Haemostasis (2012).21
GP, glycoprotein; PAR-1, protease activated receptor-1; TF, tissue factor; vWF, von Willebrand factor.

journals.sagepub.com/home/tah 355
Therapeutic Advances in Hematology 8(12)

the fact that thrombotic complications after an the prevention of stroke in patients with AF for
ACS have been primarily considered a platelet- several decades, its role in the treatment of ath-
mediated process.5,6 Currently, three oral P2Y12 erosclerotic events such as ACS is not clear.31–33
receptor inhibitors – clopidogrel, prasugrel and The AFTER (aspirin and anticoagulation follow-
ticagrelor – are approved for clinical use in ACS ing thrombolysis with eminase in recurrent
patients.7–9 Prasugrel and ticagrelor are character- infarction) study did not show any benefit of war-
ized by enhanced potency over clopidogrel and farin monotherapy compared with aspirin 150
better net clinical outcomes in ACS patients.7,8 mg monotherapy in post-MI patients.34
Accordingly, guidelines support their preferential Thereafter, warfarin monotherapy without aspi-
use in ACS settings.10–13 However, regardless of rin in post-MI patients was largely abandoned.35
the use of optimal DAPT regimens including There were several large-scale randomized clini-
prasugrel and ticagrelor, recurrence rates of car- cal trials comparing aspirin monotherapy with
diovascular ischemic events still remain high. 1 dual therapy of aspirin plus warfarin. Among
This persistent risk may be in part attributed to these, the CARS (the Coumadin aspirin reinfarc-
the sustained generation of thrombin, which play tion study) and CHAMP (the combination
a key role in thrombus formation. Several studies hemotherapy and mortality prevention) studies
in fact support that thrombin generation is persis- were conducted in a relatively large number of
tently high in ACS patients after an acute epi- participants. The CARS study evaluated the role
sode.25–27 Biomarkers of persistent activation of of warfarin among 8803 patients with recent MI
the coagulation system, such as prothrombin (3–21 days after index MI). Patients were ran-
fragment 1 + 2, D-dimer have been shown to be domly assigned to three groups: 160 mg of aspi-
associated with worse clinical outcomes.28 These rin monotherapy, 80 mg of aspirin and 1 mg of
findings indicate that thrombotic complications warfarin, 80 mg of aspirin and 3 mg of warfarin. 31
may not be prevented by antiplatelet therapy After a median of 14 months, the addition of
alone and support the potential need for long- warfarin to aspirin did not provide any benefit in
term oral anticoagulant therapy to inhibit throm- the reduction of rates of cardiovascular events
bin generation. The use of anticoagulant therapy beyond that achieved from 160 mg of aspirin
for long-term management of ACS patients, in monotherapy (one year estimated event rate,
particular VKA, has been largely abandoned with 8.6% in 160 mg of aspirin monotherapy, 8.8% in
the use of DAPT. However, the recent introduc- 80 mg of aspirin + 1 mg of warfarin, and 8.4% in
tion of NOACs, characterized by a better safety 80 mg of aspirin + 3 mg of warfarin) and showed
profile and ease of use compared with VKA, has increased rates of major bleeding. The CHAMP
led to a reappraisal of the need for using oral anti- study also compared a combination therapy of
coagulant therapy for secondary prevention in warfarin and aspirin (81 mg) to aspirin mono-
ACS patients, as described below.14 therapy (162 mg) in 5059 patients with a recent
MI for a median follow up of 2.7 years. 32
Combination therapy did not demonstrate a
Vitamin K antagonists for the patients of reduction of cardiovascular events compared to
ACS aspirin monotherapy and the addition of warfarin
In the 1940s, intravenous unfractionated heparin was significantly associated with an increased risk
was used for the treatment of patients with myo- of bleeding (1.28 versus 0.72 events, per 100 per-
cardial infarction (MI). Then VKAs were used as son years, p < 0.001). In a meta-analysis includ-
oral anticoagulant therapy and had represented ing 14 studies enrolling 25,307 MI patients, as
the only oral agents for several decades.29 compared with aspirin monotherapy, combina-
Warfarin, a representing drug of the VKA class, tion of warfarin and aspirin was associated with a
inhibits multiple enzyme in the coagulation cas- 27% relative risk reduction in the rates of all-
cade, including Factor II (prothrombin), Factor cause death, non-fatal MI or stroke only when
VII, Factor IX, Factor X, protein C and protein INR targets were between 2.0 and 3.0; however,
S. However, warfarin has several limitations this occurred at the expense of increased major
including drug–drug and food–drug interactions, bleeding.36
need for frequent monitoring, delayed onset and
offset of action (due to indirect action and long Overall, the addition of warfarin to aspirin mono-
half-life), and genetic variability in liver enzyme therapy does not provide the clinical benefits
metabolism.30 Although warfarin has been the desired for the prevention of secondary events in
cornerstone of oral anticoagulation therapy for ACS patients, largely due to the increased risk of

356 journals.sagepub.com/home/tah
J Y Moon, D Nagaraju et al.

bleeding complications. There are no available Ximelagatran


data of combined therapy with warfarin and Ximelagatran, an oral direct thrombin inhibitor,
clopidogrel as well as with newer-generation is the first NOAC that has been studied for the
P2Y12 receptor inhibitors such as ticagrelor and reduction of ischemic recurrences in ACS
prasugrel. There is also limited evidence on the patients. After oral administration, ximelagatran
use of ‘triple therapy’ (combination of warfarin is rapidly converted to its active form, mela-
with DAPT), which, however, has shown to be gatran.42 The ESTEEM trial (The efficacy and
associated with a high risk of major bleeding in safety of the oral direct thrombin inhibitor ximel-
registry data and thus is not recommended for agatran in patients with recent myocardial dam-
secondary prevention of ACS.35 age trial) explored the safety and efficacy of
ximelagatran in ACS patients.43 A total of 1883
ACS patients (within 14 days of the index event)
Non-vitamin K antagonist oral were randomly assigned to ximelagatran [at twice
anticoagulants daily doses (BID) of 24 mg, 36 mg, 48 mg or 60 mg]
Since VKAs inhibit the synthesis of coagulation or placebo. Approximately, two-thirds of the total
factors which are dependent on vitamin K, VKAs study population were STEMI and about half of
are called indirect oral anticoagulants. On the these were treated with thrombolytic therapy.
other hand, NOACs act directly on their target Almost all patients received 160 mg of aspirin.
(i.e. FXa or thrombin) and are thus called direct However, this study does not reflect current prac-
oral anticoagulants.37 Because of these properties, tice. For example, percutaneous coronary inter-
their anticoagulation effect is more prompt and vention (PCI) performed in the past four months
they also have more rapid offset of action com- or planned within two months of randomization
pared with warfarin. Direct thrombin inhibitors were excluded. Patients needing adjunctive oral
directly bind to thrombin and prevent conversion antiplatelet therapy, including P2Y12 receptor
of fibrinogen into fibrin.38 Ximelagatran and inhibitors, and anticoagulants were excluded. In
dabigatran etexilate were tested in clinical trials, the trial, the combination of ximelagatran and
and currently dabigatran etexilate is the only oral aspirin significantly reduced the primary end-
direct thrombin inhibitor used in clinical practice point (defined by composite all-cause death, non-
for prevention of stoke in AF patients, as well as fatal MI and severe recurrent ischemia) compared
treatment and prevention of venous thromboem- with aspirin alone (HR 0.76, 95% CI 0.59–0.98,
bolism (VTE).39 The other group of NOACs is p = 0.036) with a 3.6% absolute risk reduction of
represented by the direct FXa inhibitors. FXa is the primary endpoint (12.7%, ximelagatran versus
upstream to prothrombin and its inhibition con- 16.3%, placebo). The frequency of major bleed-
tributes to the down-regulation of thrombin gen- ing was numerically higher in the ximelagatran
eration. The role of FXa inhibition in ACS group (1.8% versus 0.9%, HR 1.97, 95% CI
patients is supported by the result from paren- 0.80–4.84). However, elevation in liver enzymes
teral use of the indirect FXa inhibitor fonda- occurred with ximelagatran (about 12.2–13.0%
parinux in two large studies in the acute care on the higher dose of ximelagatran). Because of
setting.40,41 Contrary to indirect agents, cofac- liver toxicity, ximelagatran was withdrawn from
tors, like antithrombin III, are not required for commercialization.
the action of the direct FXa inhibitors which
directly target FXa activity.21 The oral direct
FXa inhibitors, including apixaban, rivaroxaban Dabigatran
and edoxaban, are currently used for the preven- Dabigatran is the only oral direct thrombin inhib-
tion of stoke in AF patients and prevention/treat- itor available for clinical use.39 After oral adminis-
ment of VTE.39 Other oral FXa inhibitors tration of the pro-drug of dabigatran etexilate, the
including darexaban, letaxaban and betrixaban serum esterase rapidly converts dabigatran etex-
have been investigated in phase II clinical trials. ilate into dabigatran. The efficacy and safety of
However, of all the clinically available NOACs, dabigatran in ACS patients was investigated in
only rivaroxaban has successfully completed the phase II, dose-ranging RE-DEEM (rand-
phase III investigation meeting its primary end- omized dabigatran etexilate dose finding study in
point in ACS patients and has received approval patients with acute coronary syndromes post
for clinical use for this indication. The individual index event with additional risk factors for cardio-
results of clinical trials for anticoagulation in vascular complications also receiving aspirin and
CAD are be described in detail below (Table 1). clopidogrel) trial.44 A total of 1861 patients with

journals.sagepub.com/home/tah 357
358

Therapeutic Advances in Hematology 8(12)


Table 1. Phase II dose-escalation, clinical trials for the safety and efficacy of NOAC in ACS patients.
Ximelagatran Dabigatran Apixaban Rivaroxaban Darexaban (YM150) Letaxaban

Study name ESTEEM RE-DEEM APPRAISE ATLAS ACS-TIMI 46 RUBY-1 AXIOM ACS
Number of 1883 1861 1715 3491 1279 2753
patients
Baseline Aspirin 160 mg monotherapy Dual antiplatelet: Dual antiplatelet: Stratum 1: aspirin (75–100 Dual antiplatelet: Dual antiplatelet:
antiplatelet aspirin (<100 mg) + aspirin (<162 mg) + mg) monotherapy aspirin (75–325 mg) + aspirin + clopidogrel in 75.6%
therapy clopidogrel in 99.2% clopidogrel in 75.6% stratum 2: aspirin (75–100 clopidogrel in 97%
mg) + clopidogrel
Study duration 6 months 6 months 6 months 6 months 6 months 6 months
Daily dosage 24 mg BID 50 mg BID 2.5 mg BID 2.5 mg BID, 5 mg BID, 7.5 5 mg BID Stage 1: 10 mg BID, 20 mg BID,
36 mg BID 75 mg BID 10 mg QD mg BID (stratum 2 only), 10 10 mg QD 40 mg QD;
48 mg BID 110 mg BID 10 mg BID mg BID, 15 mg BID Stage 2: 40 mg BID, 80 mg QD,
60 mg BID 150 mg BID 20 mg QD 5 mg QD, 10 mg QD, 15 mg 30 mg QD 80 mg BID;
QD (stratum 2 only), 20 mg 30 mg BID Stage 3: 160 mg QD, 120 mg BID
QD 60 mg QD
Safety endpoint Major bleeding (its own ISTH major and ISTH major and TIMI major, TIMI minor, or ISTH major and clinically TIMI major bleeding
and bleeding definition) or bleeding clinically relevant non- clinically relevant non- requiring medical attention relevant non-major bleeding
definition leading to discontinuation. major bleeding major bleeding
Safety 24 mg: 3.57 (1.80–7.09) 50 mg: 1.82 (0.77–4.29) 2.5 mg BID: 1.78 2.5 mg BID: 1.71 (0.76–3.85) 10 mg QD: 1.78 (0.68–4.60) *TIMI major + minor bleeding
outcome, HR 36 mg: 1.63 (0.71–3.72) 75 mg: 2.44 (1.05–5.65) (0.91–3.48) 5 mg BID: 3.36 (2.21–5.09) 30 mg QD: 1.83 (0.71–4.75) Placebo: 0.9%
(95% CI) 48 mg: 4.37 (2.25–8.47) 110 mg: 3.56 (1.60–7.91) 10 mg QD: 2.45 7.5 mg BID: 3.41 (1.97–5.89) 60 mg QD: 2.43 (0.98–5.97) 10 mg BID: 0.4%
60 mg: 3.80 (1.94–7.46) 150 mg: 3.88 (1.73–8.74) (1.31–4.61) 10 mg BID: 4.80 (3.09–7.45) 5 mg BID: 2.05 (0.81–5.15) 20 mg BID: 1.6%
5 mg QD: 2.73 (1.38–5.37) 15 mg BID: 2.27 (0.92–5.59) 40 mg QD: 0.4%
10 mg QD: 3.35 (2.21–5.09) 30 mg BID: 3.80 (1.66–8.68) 40 mg BID: 3.7% (p < 0.01)
15 mg QD: 3.69 (2.17–6.29) 80 mg QD: 3.6% (p < 0.01)
20 mg QD: 5.32 (3.46–8.18) 80 mg BID: 2.8%
160 mg QD: 1.2%
120 mg BID: 3.2% (p < 0.05)
Efficacy Ximelagatran reduced the No significant difference Showing the tendency Rivaroxaban non-significantly No significant difference in No significant differences in CV
outcome death, non-fatal MI and in cardiovascular in reduction of CV reduced death, MI, stroke, death, non-fatal MI, non-fatal death, non-fatal MI, non-fatal
HR (95% CI) severe recurrent ischemia: ischemic events death, MI, severe severe recurrent ischemia stroke or severe recurrent stroke or myocardial ischemia
0.76 (0.59–0.98, p = 0.036) recurrent ischemia or requiring revascularization: ischemia requiring hospitalization
ischemic stroke 0.79 (0.60–1.05, p = 0.10)
Other findings Liver enzyme elevation Reduced D-dimer 10 mg BID and 20 mg Rivaroxaban reduced death, The rate of efficacy endpoint was
(about 12.2–13.0% on the concentrations in QD arms were stopped MI or stroke: 0.69 (0.50–0.96, numerically higher than placebo
higher dose of ximelagatran) dabigatran groups because of bleeding p = 0.027) in daily 30 mg and 60 mg.
journals.sagepub.com/home/tah

*The incidences of the primary bleeding endpoint, TIMI major bleeding, in each dose were low, so we present the composite of TIMI major and minor bleeding.
ACS, acute coronary syndrome; BID, twice daily; CI, confidence intervals; CV, cardiovascular; HR, hazard ratio; ISTH, International Society of Thrombosis and Haemostasis; MI, myocardial infarction; NOAC, non-
vitamin K antagonist oral anticoagulants; QD, once daily.
J Y Moon, D Nagaraju et al.

recent STEMI or NSTEMI were randomly the reduction of ischemic events (rate of ischemic
assigned to different doses of dabigatran (dabi- event; 7.6% in 2.5 mg BID, 6.0% in 10 mg QD,
gatran 50 mg BID, 75 mg BID, 110 mg BID, 150 8.7 % in placebo).
mg BID) or placebo. At the time of randomiza-
tion, 99.2% of patients were already on DAPT Based on the result of the APPRAISE trial, a 5 mg
with aspirin plus clopidogrel and 83.8% of BID dose of apixaban was investigated in addition
patients continued DAPT until the end of the to DAPT after an ACS event in the large-scale,
study. The primary endpoint of this study was the phase III APPRAISE-2 trial.46 Among the base-
rate of major and clinically relevant minor bleed- line characteristics of study participants, 59% of
ing. The assessment of major bleeding was based patients were ⩾ 65 years old, 47.8% of patients
on the International Society of Thrombosis and had diabetes mellitus, 10.0% of patients had a
Haemostasis (ISTH) definition. There was a prior stroke and 28.9% of patients had impaired
dose-dependent increase in the rate of the pri- renal function. With a median follow up of
mary endpoint compared to placebo (2.2% in approximately 8 months, TIMI major bleeding
placebo group, 3.5% in 50 mg BID group, 4.3% (primary safety outcome) occurred in 1.3% and
in 75 mg BID, 7.9% in 110 mg BID and 7.8% in 0.5% of patients in the apixaban and placebo
150 mg BID). Dabigatran was not associated groups, respectively (HR 2.59, 95% CI 1.50–4.46,
with any ischemic benefit. It significantly reduced p = 0.001). In addition, apixaban was associated
coagulation activity, such as D-dimer concentra- with an increase in fatal bleeding events and
tion.44 Further investigations, including phase III, intracranial bleeding (ICH) compared to placebo.
of dabigatran in ACS patients have not been con- There was no reduction in ischemic events with
ducted after the RE-DEEM trial. the primary efficacy outcome, defined as a com-
posite of cardiovascular death, MI or ischemic
stroke, occurring in 7.5% and 7.9% in the apixa-
Apixaban ban and placebo groups, respectively. After
Apixaban is a selective direct-acting FXa inhibitor recruitment of 7392 participants, the APPRAISE-2
which affects both free and prothrombinase-bound trial terminated prematurely because of increased
FXa.39 The half-life of apixaban is approximately bleeding without clinical benefits.
12 h and it is eliminated through multiple path-
ways, including hepatic metabolism, renal clear-
ance and biliary secretion.39 The safety and efficacy Rivaroxaban
of apixaban in ACS patients were explored in the Rivaroxaban is a highly selective, direct FXa
phase II, dose-ranging APPRAISE trial.45 inhibitor.39 Similarly to apixaban, it inhibits both
APPRAISE enrolled 1715 patients with recent free FXa activity and prothrombinase-bound
STEMI or NSTE-ACS. Aspirin was used in almost FXa activity. Rivaroxaban has a half-life of 5–9 h,
all patients and clopidogrel was used in 76% of and is eliminated through the renal (66%) and
cases. Patients were randomly allocated to receive fecal/biliary systems (28%). The safety and effi-
apixaban [one of four different doses; 2.5 mg BID, cacy of rivaroxaban in ACS patients (1–7 days
10 mg QD (once daily), 10 mg BID, 20 mg QD] or after the index event) was explored in the phase
placebo. The primary outcome was major bleeding II, dose-escalation ATLAS ACS-TIMI 46 trial.47
(by ISTH definition) and clinically relevant non- This trial had two strata based on the antiplatelet
major bleeding during 6-month clinical follow up. treatment regimen used (stratum I: monotherapy
Apixaban 2.5 mg BID demonstrated a non-signifi- of aspirin 75–100 mg; stratum II: dual therapy of
cant increase in the primary outcome compared to aspirin 75–100 mg and clopidogrel). Patients (n =
placebo (HR 1.78, 95% CI 0.91–3.48, p = 0.09) 3491) were randomized to placebo or different
and the 10 mg QD dose resulted in a significant doses (5–20 mg) of rivaroxaban (2.5 mg BID, 5 mg
increase in the primary outcome (HR 2.45, 95% BID, 7.5 mg BID, 10 mg BID, 5 mg QD, 10 mg
CI 1.31–4.61, p = 0.005). Both the 10 mg BID QD, 15 mg QD, 20 mg QD). During the 6-month
and 20 mg QD arms were stopped due to excess follow-up period, rivaroxaban showed a dose-
bleeding. Although apixaban showed dose- dependent increase in clinically significant bleed-
dependent increased bleeding, a trend in reducing ing with a non-significant reduction in the primary
ischemic events was also demonstrated. Overall, efficacy endpoint (death, MI, stroke or severe
the addition of apixaban to ACS patients treated recurrent ischemia requiring revascularization).
with DAPT was associated with dose-related However, the secondary efficacy endpoints
increase in bleeding events, with modest benefit in (death, MI or stroke) was significantly decreased

journals.sagepub.com/home/tah 359
Therapeutic Advances in Hematology 8(12)

Figure 3. The survival benefit of rivaroxaban 2.5 mg BID from the ATLAS ACS2 TIMI 51 trial.
(a) The efficacy of rivaroxaban 2.5 mg BID in reduction of the cardiovascular death, MI or stroke (the primary efficacy
endpoints) compared with placebo is presented (mITT p = 0.02, ITT p = 0.007). (b) There is a significant reduction in the rate
of CV death by 34% compared with placebo (mITT p = 0.002, ITT p = 0.005). (c) All-cause death reduced by 32% compared
with placebo (mITT p = 0.002, ITT p = 0.004). Reproduced with permission from Thrombosis and Haemostasis (2016).35
CV, cardiovascular; mITT, modified intention to treat; NNT, number needed to treat.

with rivaroxaban (HR 0.69, 95% CI 0.50–0.96, p p = 0.03; 9.1% in 2.5 mg BID dose and 10.7% in
= 0.027). A dose of 2.5 mg BID was the only placebo, p = 0.02). Most importantly, a survival
regimen that did not show a statistically signifi- benefit was achieved with the 2.5 mg BID regi-
cant increase in bleeding risk (HR 1.71, 95% CI men; in particular, there was a significant reduc-
0.76–3.85), while this was significantly increased tion in cardiovascular death (2.7% in 2.5 mg BID
with all other doses. and 4.1% in placebo, p = 0.002) and death from
any cause (2.9% in 2.5 mg BID dose and 4.5% in
The findings from the ATLAS ACS-TIMI 46 trial placebo, p = 0.002) (Figure 3). However, no sur-
identified the most suitable dosing regimens of vival benefit compared to placebo was demon-
rivaroxaban to be used in combination with DAPT, strated with the 5 mg BID regimen. Rivaroxaban
which were applied in the phase III, ATLAS ACS was associated with a significant increase in non-
2-TIMI 51 study.48 In this trial, 15,526 patients coronary artery bypass grafting (CABG)-related
with a recent ACS were randomized to either rivar- major bleeding (2.1% in rivaroxaban and 0.6% in
oxaban (2.5 mg BID or 5 mg BID) or placebo. 46 placebo, p < 0.001) and ICH (0.6% in rivaroxa-
Among the baseline characteristics of study partici- ban and 0.2% in placebo, p = 0.009). The rate of
pants, 36.5% of patients were ⩾ 65 years old, TIMI bleeding requiring medical attention was also
32.0% had diabetes mellitus and 50.3% of patients increased with rivaroxaban (14.5% versus 7.5%, p <
were STEMI. Patients with a prior stroke were 0.001). Overall, the 5 mg BID dose was associated
excluded. The median time from the index event with more bleeding, including fatal bleeding (5 mg
to randomization was 4.7 days. Thienopyridines BID: 0.4% versus 2.5 mg BID: 0.1%, p = 0.004)
(clopidogrel or ticlopidine) were used as a back- compared with the 2.5 mg BID dose. The lower
ground antiplatelet therapy in 93% of the patients. rate of fatal bleeding with the 2.5 mg BID dose
The trial showed a significant reduction in the pri- may indeed have contributed to its survival bene-
mary efficacy endpoint (a composite of cardiovas- fit. Overall, the ATLAS ACS 2 trial demonstrated
cular death, MI or stroke) with rivaroxaban that the addition of low-dose rivaroxaban (2.5 mg
compared with placebo after mean treatment BID regimen) to DAPT with aspirin and clopi-
duration of 13.1 months (8.9%, rivaroxaban versus dogrel reduced recurrent cardiovascular events,
10.7%, placebo; HR 0.84, 95% CI 0.74–0.96, p = including mortality, in patients with a recent
0.008). In particular, both dosing regimens showed ACS at the expense of three-fold increased
a significant reduction in the primary efficacy end- major bleeding, but with no increase in fatal
point (8.8% in 5 mg BID and 10.7% in placebo, bleeding. To date, the only reported subgroup

360 journals.sagepub.com/home/tah
J Y Moon, D Nagaraju et al.

analysis of the ATLAS ACS 2 trial is that on discontinuation of parenteral anticoagulation


patients with STEMI (n = 7817). In this analy- (Class IIb, level of evidence B);12 in selected
sis, rivaroxaban reduced the incidence of the STEMI patients who receive aspirin and clopi-
primary efficacy endpoint consistently with the dogrel, low-dose rivaroxaban (2.5 mg BID) may
overall trial results (8.7% with 2.5 mg BID, be considered if the patient is at low bleeding risk
8.2% with 5 mg BID and 10.6% with placebo).49 (Class IIb, level of evidence B).11
Rivaroxaban was also associated with an increase
of major bleeding rate, but there was no signifi- The observations from the ATLAS ACS 2 trial
cant increase in fatal bleeding in the STEMI have led to questions on how to enhance the safety
subgroup. profile of rivaroxaban while maintaining efficacy.
Recently, there has been emerging interest in
The result of the two phase III trials, ATLAS exploring whether withdrawing aspirin from novel
ACS 2 and APPRAISE-2, which investigated the antithrombotic treatment regimens could be a
long-term use of NOACs in post-ACS patients potential approach to reaching this goal.51,52 The
are discordant. One probable reason for the con- rationale behind this is that in the presence of
flicting results may be attributed to the character- other antithrombotic agents, aspirin may not pro-
istics of the study population. The study vide further ischemic benefit, but simply enhance
participants of APPRAISE-2 were older and had the risk of bleeding complications, in particular
more comorbidities, including more diabetes and gastrointestinal. The bleeding risk of low-dose
renal dysfunction. Importantly, patients with rivaroxaban (in addition to a P2Y12 receptor
prior stroke who were excluded in ATLAS ACS 2 inhibitor) without aspirin was evaluated in the
were enrolled in the APPRAISE-2 trial. The phase II, GEMINI-ACS-1 (A study to compare
higher risk profile of patients enrolled in the the safety of rivaroxaban versus acetylsalicylic acid
APPRAISE-2 trial is also supported by the higher in addition to either clopidogrel or ticagrelor ther-
ischemic event rate. The second possible reason apy in participants with acute coronary syn-
is the level of anticoagulant effects achieved in the drome-1) trial.53 A total of 3037 ACS patients
two studies. The APPRAISE-2 trial tested 5 mg were randomly assigned to 2.5 mg BID rivaroxa-
BID of apixaban, which is the same dose used for ban versus 100 mg aspirin (1:1 ratio) in addition to
stroke prevention in AF patients. On the con- a P2Y12 receptor inhibitor in a double-blind fash-
trary, in ATLAS ACS 2 there was a survival ben- ion. The choice of the P2Y12 receptor inhibitor
efit with the 2.5 mg BID daily dose, which is (ticagrelor or clopidogrel) was not randomized,
one-quarter of the total daily dose of rivaroxaban and was left at the discretion of the investigator.
used in AF.14 Therefore, the high levels of antico- TIMI non-CABG-related clinically significant
agulant effect when added to DAPT in ACS bleeding, the primary endpoint of this trial, was
patients may not provide benefits due to the high similar in both groups at 12-month follow up
risk of bleeding complications. (rivaroxaban: 5% versus aspirin: 5%, p = 0.584).
The rates of TIMI major bleeding and ICH were
Given the survival benefit presented in the very low and not significantly different between
ATLAS ACS 2 trial, the 2.5 mg BID regimen of groups (TIMI major bleeding, rivaroxaban 1%
rivaroxaban with aspirin alone or with DAPT versus aspirin 1%; ICH, rivaroxaban <1% versus
obtained regulatory approval from the European aspirin 0%). The rate of the primary endpoint was
Medicines Agency (EMA) for secondary preven- similar between the two groups both in the tica-
tion in ACS patients with elevated cardiac bio- grelor stratum (rivaroxaban 7% versus aspirin 6%)
markers.50 However, rivaroxaban has not been and in the clopidogrel stratum (rivaroxaban 3%
approved by the Food and Drug Administration versus aspirin 3%). There was a significant increase
(FDA) for the ACS indication due to concerns in bleeding with ticagrelor compared to clopi-
regarding missing data in the pivotal trial. The dogrel (p = 0.0006). The exploratory ischemic
addition of low-dose rivaroxaban (2.5 mg BID for endpoint (CV death, MI, stroke or definite stent
one year) is now considered within the European thrombosis) was also similar between groups
Society of Cardiology guidelines on NSTE-ACS (rivaroxaban 5% versus aspirin 5%, p = 0.73).
and STEMI.11,12 In particular, in NSTEMI Overall, the findings from the GEMINI-ACS-1
patients without history of prior stroke/TIA and trial suggest that the use of low-dose rivaroxaban
at high ischemic risk as well as low bleeding risk with ticagrelor without aspirin was associated with
receiving aspirin and clopidogrel, low-dose rivar- an acceptable safety profile compared with DAPT
oxaban (2.5 mg BID) may be considered after with aspirin and ticagrelor.

journals.sagepub.com/home/tah 361
Therapeutic Advances in Hematology 8(12)

Darexaban (YM150) not selectively in patients with an ACS. The


The tolerability and safety of darexaban (YM150), COMPASS trial [Cardiovascular outcomes for
another direct FXa inhibitor, in ACS patients was people using anticoagulation strategies trial
tested in the phase II, RUBY-1 [A randomized, (ClinicalTrials.gov identifier: NCT01776424)]
double-blind, placebo controlled trial of the safety was designed to evaluate the efficacy and safety of
and tolerability of the novel oral Factor Xa inhibitor low-dose rivaroxaban plus aspirin compared to
darexaban (YM150) following ACS] trial.54 In this aspirin monotherapy for the prevention of
trial, a total of 1279 patients with recent NSTEMI ischemic events in patients with CAD or periph-
or STEMI were enrolled and randomly assigned to eral artery disease (PAD). Patients were randomly
different doses of darexaban (5 mg BID, 10 mg assigned to one of three groups (rivaroxaban
QD, 15 mg BID, 30 mg QD, 30 mg BID, 60 mg 2.5 mg BID plus aspirin 100 mg; rivaroxaban
QD) or placebo. There were higher rates of major 5 mg BID alone; or aspirin 100 mg alone). The
(by ISTH definition) and clinically relevant non- trial was stopped one year ahead of its scheduled
major bleeding in a dose-dependent manner with all completion date after enrollment of 27,402
dosing regimens of darexaban compared to placebo patients because rivaroxaban was shown to meet
without any improvement in efficacy endpoints (the its efficacy endpoint. In particular, rivaroxaban
composite of death, MI, stroke, systemic thrombo- plus aspirin showed better cardiovascular out-
embolism and severe recurrent ischemia). After the comes, including reduced mortality, than with
RUBY-1 trial, the development of darexaban for aspirin alone, with consistent findings in the
ACS patients was halted. CAD and PAD populations. Although the com-
bination group showed more major bleeding than
aspirin alone, there was no significant difference
Letaxaban (TAK-442) in rate of ICH or fatal bleeding between these
The safety and tolerability of letaxaban were tested two groups. Rivaroxaban monotherapy (5 mg
in the AXIOM ACS (Safety and efficacy of TAK- BID) did not result in better cardiovascular out-
442 in subjects with acute coronary syndromes) comes than aspirin alone and showed more major
trial.55 The AXIOM ACS trial was a dose-ranging, bleeding events.56 It is important to note that
phase II trial that enrolled 2753 ACS patients. only 62% of patients enrolled in COMPASS had
This trial explored a wide range of letaxaban doses a prior ACS, and the mean time from this event
(from 10 mg BID to 120 mg BID). The rate of to randomization was 7.1 years. Therefore, the
TIMI major bleeding, the primary endpoint of this observations from the COMPASS trial suggest
study, was not significantly different between that there may be a role for adjunctive use of a
groups (0.9% in letaxaban versus 0.5% in placebo, very low dose of rivaroxaban for secondary pre-
p = 0.47). However, the composite rate of TIMI vention of ischemic events in the patients with
major and minor bleeding was more frequent with stable atherosclerotic disease, and should not be
letaxaban (2.1% versus 0.9%, p = 0.025). The effi- considered as a post-ACS management strategy
cacy endpoint was similar between letaxaban and until further information is available. The
placebo. There has not been further testing for VOYAGER PAD trial [Efficacy and safety of
ACS patients with letaxaban. rivaroxaban in reducing the risk of major throm-
botic vascular events in subjects with sympto-
matic peripheral artery disease undergoing
Edoxaban peripheral revascularization procedures of the
Edoxaban is a direct oral Factor Xa inhibitor used lower extremities trial (ClinicalTrials.gov identi-
for the prevention of ischemic stroke in AF fier: NCT02504216)] is investigating the role of
patients as well as for the prevention/treatment of rivaroxaban in addition to the standard care of
VTE.39 However, there have not been any studies PAD. This ongoing trial will enroll 6500 partici-
assessing edoxaban in ACS patients. pants undergoing successful revascularization
procedure for symptomatic PAD, and rand-
omized to rivaroxaban 2.5 mg BID or placebo.
Clinical trial update The use of clopidogrel is kept to the minimum
Large randomized trials have either been recently necessary in accordance with international prac-
completed or are completing enrollment which are tice guidelines. The primary endpoint is a first
assessing the safety and efficacy of oral anticoagu- occurrence of MI, ischemic stroke, CV death,
lant therapy, in particular rivaroxaban, in patients acute limb ischemia and major amputation for 2
with atherosclerotic disease manifestations, albeit years; results are anticipated in 2019.

362 journals.sagepub.com/home/tah
J Y Moon, D Nagaraju et al.

Future perspectives Conclusions


Despite the use of standard of care DAPT with Over recent years there has been a reappraisal of
aspirin and a P2Y12 receptor antagonist, the risk of the use of oral anticoagulant therapy for secondary
ischemic recurrences in ACS patients remains prevention of ischemic recurrences in ACS
high. The persistence of thrombin generation after patients. This has been indeed facilitated by the
an ACS event may contribute to these observa- introduction of the NOACs, which have a favora-
tions, prompting investigators to reappraise the ble safety profile and ease of use compared with
role of long-term oral anticoagulant therapy in VKAs, which were tested in the same context a
these patients. Indeed, the introduction of the few decades ago, leading to disappointing out-
NOACs, which have a more favorable safety pro- comes due to the high risk of bleeding. Of the
file and ease of use compared with VKA, have clinically available NOACs, only rivaroxaban
stimulated interest in this field of investigation. A when used at a 2.5 mg BID regimen, which is sub-
multitude of NOACs have been tested in the con- stantially lower than that used for stroke preven-
text of ACS settings. However, only rivaroxaban tion in AF patients, has shown to be associated
successfully completed phase III clinical investiga- with an ischemic benefit, including a reduction in
tion meeting its primary endpoint and has received mortality and has received regulatory approval for
approval for clinical use in most parts of the world clinical use in most countries across the globe.
(not in the United States) for patients with ACS. However, risks of bleeding complications still
However, despite the survival benefit observed remain high when this regimen is added to stand-
with rivaroxaban when used at a 2.5 mg BID regi- ard DAPT, underscoring the need to define
men, in addition to standard of care therapy antithrombotic treatment regimens that maintain
including DAPT, the uptake of the drug for the efficacy but minimize bleeding complications. A
ACS indication has been very limited. Indeed, the current wave of investigations is evaluating
increased bleeding risk as well as the need to con- whether withdrawing aspirin therapy may repre-
sider a third antithrombotic agent contribute to sent a potential option to reach this goal. Indeed,
the lack of enthusiasm of physicians and patients a number of ongoing pharmacodynamic and clini-
embracing this approach in clinical practice. cal investigations will provide more insights on the
Encouraging data from recent trial results suggest role of using long-term oral anticoagulant therapy
that withdrawal of aspirin therapy may be a safe for secondary prevention in patients with cardio-
approach in patients treated with low-dose rivar- vascular disease manifestations.
oxaban plus ticagrelor. These investigations have
been prompted based on studies that have sug- Funding
gested that there is limited antithrombotic efficacy This research received no specific grant from any
associated with aspirin therapy in the presence of funding agency in the public, commercial or not-
potent P2Y12 blockade, which has been associated for-profit sectors.
with modulation of other key platelet signaling
pathways, including thromboxane generation.57–59 Conflict of interest statement
Indeed larger investigations are warranted to sup- Dr. Angiolillo reports receiving payments as an indi-
port the efficacy of this strategy. Moreover, a bet- vidual for: (a) consulting fee or honorarium from
ter understanding is needed to define the interplay Amgen, Aralez, AstraZeneca, Bayer, Biosensors,
between NOACs and antiplatelet agents. In fact, Bristol-Myers Squibb, Chiesi, Daiichi-Sankyo, Eli
to date there is very limited data on this topic and Lilly, Janssen, Merck, PLx Pharma, Pfizer, Sanofi,
it is also unclear how NOACs with different tar- and The Medicines Company; (b) participation
gets (anti-FII versus anti-FX) impact the effects of in review activities from CeloNova and St. Jude
different classes of antiplatelet agents (COX-1 Medical. Institutional payments for grants from
versus P2Y12 inhibitors).60,61 These studies will Amgen, AstraZeneca, Bayer, Biosensors, CeloNova,
indeed provide important insights on the safety CSL Behring, Daiichi-Sankyo, Eisai, Eli-Lilly,
and efficacy findings from clinical trial data associ- Gilead, Janssen, Matsutani Chemical Industry Co.,
ated with their combined use. Ultimately, how to Merck, Novartis, Osprey Medical, and Renal Guard
bridge from acute to long-term care with oral anti- Solutions; in addition, Dr. Angiolillo is recipient of
coagulant therapy among patients who experi- a funding from the Scott R. MacKenzie Foundation
enced an ACS is also needed. Indeed, the results and the NIH/NCATS Clinical and Translational
from patients with a more recent ACS who were Science Award to the University of Florida UL1
enrolled in the COMPASS trial will provide fur- TR000064 and NIH/NHGRI U01 HG007269,
ther insights on this topic. outside the submitted work.

journals.sagepub.com/home/tah 363
Therapeutic Advances in Hematology 8(12)

References 13. Amsterdam EA, Wenger NK, Brindis RG, et al.


1. Benjamin EJ, Blaha MJ, Chiuve SE, et al. Heart 2014 AHA/ACC guideline for the management
disease and stroke statistics – 2017 update: a of patients with non-ST-elevation acute coronary
report from the American Heart Association. syndromes: a report of the American College of
Circulation 2017; 135: e146–e603. Cardiology/American Heart Association Task
Force on Practice Guidelines. J Am Coll Cardiol
2. Libby P. Mechanisms of acute coronary 2014; 64: e139–e228.
syndromes and their implications for therapy. N
Engl J Med 2013; 368: 2004–2013. 14. De Caterina R, Husted S, Wallentin L, et al. New
oral anticoagulants in atrial fibrillation and acute
3. Davi G and Patrono C. Platelet activation and coronary syndromes: ESC Working Group on
atherothrombosis. N Engl J Med 2007; 357: Thrombosis-Task Force on Anticoagulants in
2482–2494. Heart Disease position paper. J Am Coll Cardiol
4. Angiolillo DJ, Ueno M and Goto S. Basic 2012; 59: 1413–1425.
principles of platelet biology and clinical 15. Angiolillo DJ, Goodman SG, Bhatt DL, et al.
implications. Circ J 2010; 74: 597–607. Antithrombotic therapy in patients with atrial
5. Franchi F and Angiolillo DJ. Novel antiplatelet fibrillation undergoing percutaneous coronary
agents in acute coronary syndrome. Nat Rev intervention: a North American perspective –
Cardiol 2015; 12: 30–47. 2016 update. Circ Cardiovasc Interv 2016; 9.

6. Franchi F, Rollini F and Angiolillo DJ. 16. Lip GY, Windecker S, Huber K, et al.
Antithrombotic therapy for patients with STEMI Management of antithrombotic therapy in
undergoing primary PCI. Nat Rev Cardiol 2017; atrial fibrillation patients presenting with
14: 361–379. acute coronary syndrome and/or undergoing
percutaneous coronary or valve interventions:
7. Wiviott SD, Braunwald E, McCabe CH, et al. a joint consensus document of the European
Prasugrel versus clopidogrel in patients with acute Society of Cardiology Working Group on
coronary syndromes. N Engl J Med 2007; 357: Thrombosis, European Heart Rhythm
2001–2015. Association (EHRA), European Association
8. Wallentin L, Becker RC, Budaj A, et al. of Percutaneous Cardiovascular Interventions
Ticagrelor versus clopidogrel in patients with (EAPCI) and European Association of Acute
acute coronary syndromes. N Engl J Med 2009; Cardiac Care (ACCA) endorsed by the Heart
361: 1045–1057. Rhythm Society (HRS) and Asia-Pacific Heart
Rhythm Society (APHRS). Eur Heart J 2014; 35:
9. Yusuf S, Zhao F, Mehta SR, et al. Effects of 3155–3179.
clopidogrel in addition to aspirin in patients with
acute coronary syndromes without ST-segment 17. Brass LF. Thrombin and platelet activation. Chest
elevation. N Engl J Med 2001; 345: 2003; 124: 18s–25s.
494–502. 18. Varga-Szabo D, Pleines I and Nieswandt B. Cell
10. O’Gara PT, Kushner FG, Ascheim DD, et al. adhesion mechanisms in platelets. Arterioscler
2013 ACCF/AHA guideline for the management Thromb Vasc Biol 2008; 28: 403–412.
of ST-elevation myocardial infarction: a report of 19. Monroe DM, Hoffman M and Roberts HR.
the American College of Cardiology Foundation/ Transmission of a procoagulant signal from tissue
American Heart Association Task Force on factor-bearing cell to platelets. Blood Coagul
Practice Guidelines. Circulation 2013; 127: Fibrinolysis 1996; 7: 459–464.
e362–e425.
20. Tanaka KA, Key NS and Levy JH. Blood
11. Steg PG, James SK Atar D, et al. ESC Guidelines coagulation: hemostasis and thrombin regulation.
for the management of acute myocardial Anesth Analg 2009; 108: 1433–1446.
infarction in patients presenting with ST-segment
elevation. Eur Heart J 2012; 33: 2569–2619. 21. De Caterina R, Husted S, Wallentin L, et al.
General mechanisms of coagulation and targets
12. Roffi M, Patrono C, Collet JP, et al. 2015 ESC
of anticoagulants (Section I). Position Paper
Guidelines for the management of acute coronary
of the ESC Working Group on Thrombosis –
syndromes in patients presenting without
Task Force on Anticoagulants in Heart Disease.
persistent ST-segment elevation: Task Force for
Thromb Haemost 2013; 109: 569–579.
the Management of Acute Coronary Syndromes
in Patients Presenting without Persistent 22. Angiolillo DJ, Capodanno D and Goto S.
ST-Segment Elevation of the European Society Platelet thrombin receptor antagonism and
of Cardiology (ESC). Eur Heart J 2016; 37: atherothrombosis. Eur Heart J 2010; 31:
267–315. 17–28.

364 journals.sagepub.com/home/tah
J Y Moon, D Nagaraju et al.

23. Coughlin SR. Protease-activated receptors in (the AFTER study): a multicentre unblinded
hemostasis, thrombosis and vascular biology. J randomised clinical trial. BMJ 1996; 313:
Thromb Haemost 2005; 3: 1800–1814. 1429–1431.
24. Angiolillo DJ and Ferreiro JL. Antiplatelet and 35. De Caterina R, Husted S, Wallentin L, et al. Oral
anticoagulant therapy for atherothrombotic anticoagulants in coronary heart disease (Section
disease: the role of current and emerging agents. IV). Position paper of the ESC Working Group
Am J Cardiovasc Drugs 2013; 13: 233–250. on Thrombosis – Task Force on Anticoagulants
in Heart Disease. Thromb Haemost 2016; 115:
25. Ardissino D, Merlini PA, Bauer KA, et al.
685–711.
Coagulation activation and long-term outcome
in acute coronary syndromes. Blood 2003; 102: 36. Andreotti F, Testa L, Biondi-Zoccai GG, et al.
2731–2735. Aspirin plus warfarin compared to aspirin alone
after acute coronary syndromes: an updated and
26. Merlini PA, Bauer KA, Oltrona L, et al.
comprehensive meta-analysis of 25,307 patients.
Persistent activation of coagulation mechanism
Eur Heart J 2006; 27: 519–526.
in unstable angina and myocardial infarction.
Circulation 1994; 90: 61–68. 37. Husted S, de Caterina R, Andreotti F, et al.
Non-vitamin K antagonist oral anticoagulants
27. Christersson C, Oldgren J, Bylock A, et al.
(NOACs): no longer new or novel. Thromb
Long-term treatment with ximelagatran, an oral
Haemost 2014; 111: 781–782.
direct thrombin inhibitor, persistently reduces the
coagulation activity after a myocardial infarction. 38. Eisert WG, Hauel N, Stangier J, et al.
J Thromb Haemost 2005; 3: 2245–2253. Dabigatran: an oral novel potent reversible
nonpeptide inhibitor of thrombin. Arterioscler
28. Christersson C, Oldgren J, Bylock A, et al. Early
Thromb Vasc Biol 2010; 30: 1885–1889.
decrease in coagulation activity after myocardial
infarction is associated with lower risk of 39. Chan NC, Eikelboom JW and Weitz JI. Evolving
new ischaemic events: observations from the treatments for arterial and venous thrombosis:
ESTEEM Trial. Eur Heart J 2007; 28: 692–698. role of the direct oral anticoagulants. Circ Res
2016; 118: 1409–1424.
29. Ebert RV. Use of anticoagulants in acute
myocardial infarction. Circulation 1972; 45: 40. Yusuf S, Mehta SR, Chrolavicius S, et al. Effects
903–910. of fondaparinux on mortality and reinfarction
in patients with acute ST-segment elevation
30. De Caterina R, Husted S, Wallentin L, et al.
myocardial infarction: the OASIS-6 randomized
Vitamin K antagonists in heart disease: current
trial. JAMA 2006; 295: 1519–1530.
status and perspectives (Section III). Position
paper of the ESC Working Group on Thrombosis 41. Yusuf S, Mehta SR, Fifth Organization to Assess
– Task Force on Anticoagulants in Heart Disease. Strategies in Acute Ischemic Syndromes, et al.
Thromb Haemost 2013; 110: 1087–1107. Comparison of fondaparinux and enoxaparin in
acute coronary syndromes. N Engl J Med 2006;
31. Coumadin Aspirin Reinfarction Study (CARS)
354: 1464–1476.
Investigators. Randomised double-blind trial
of fixed low-dose warfarin with aspirin after 42. Eriksson UG, Bredberg U, Hoffmann KJ, et al.
myocardial infarction. Lancet 1997; 350: 389– Absorption, distribution, metabolism, and
396. excretion of ximelagatran, an oral direct thrombin
inhibitor, in rats, dogs, and humans. Drug Metab
32. Fiore LD, Ezekowitz MD, Brophy MT, et al.
Dispos 2003; 31: 294–305.
Department of Veterans Affairs Cooperative
Studies Program Clinical Trial comparing 43. Wallentin L, Wilcox RG, Weaver WD, et al.
combined warfarin and aspirin with aspirin Oral ximelagatran for secondary prophylaxis
alone in survivors of acute myocardial infarction: after myocardial infarction: the ESTEEM
primary results of the CHAMP study. Circulation randomised controlled trial. Lancet 2003; 362:
2002; 105: 557–563. 789–797.
33. Rothberg MB, Celestin C, Fiore LD, et al. 44. Oldgren J, Budaj A, Granger CB, et al.
Warfarin plus aspirin after myocardial infarction or Dabigatran versus placebo in patients with acute
the acute coronary syndrome: meta-analysis with coronary syndromes on dual antiplatelet therapy:
estimates of risk and benefit. Ann Intern Med a randomized, double-blind, phase II trial. Eur
2005; 143: 241–250. Heart J 2011; 32: 2781–2789.
34. Julian DG, Chamberlain DA and Pocock SJ. 45. APPRAISE Steering Committee and
A comparison of aspirin and anticoagulation Investigators, Alexander JH, Becker RC, et al.
following thrombolysis for myocardial infarction Apixaban, an oral, direct, selective Factor Xa

journals.sagepub.com/home/tah 365
Therapeutic Advances in Hematology 8(12)

inhibitor, in combination with antiplatelet 54. Steg PG, Mehta SR, Jukema JW, et al. RUBY-1:
therapy after acute coronary syndrome: results of a randomized, double-blind, placebo-controlled
the Apixaban for Prevention of Acute Ischemic trial of the safety and tolerability of the novel oral
and Safety Events (APPRAISE) trial. Circulation factor Xa inhibitor darexaban (YM150) following
2009; 119: 2877–2885. acute coronary syndrome. Eur Heart J 2011; 32:
2541–2554.
46. Alexander JH, Lopes RD, James S, et al. Apixaban
with antiplatelet therapy after acute coronary 55. Goldstein S, Bates ER, Bhatt DL, et al. Phase 2
syndrome. N Engl J Med 2011; 365: 699–708. study of TAK-442, an oral factor Xa inhibitor,
in patients following acute coronary syndrome.
47. Mega JL, Braunwald E, Mohanavelu S, et al.
Thromb Haemost 2014; 111: 1141–1152.
Rivaroxaban versus placebo in patients with acute
coronary syndromes (ATLAS ACS-TIMI 46): a 56. Eikelboom JW, Connolly SJ, Bosch J, et al.
randomised, double-blind, phase II trial. Lancet Rivaroxaban with or without aspirin in stable
2009; 374: 29–38. cardiovascular disease. N Engl J Med. Epub
48. Mega JL, Braunwald E, Wiviott SD, et al. ahead of print 27 August 2017. DOI: 10.1056/
Rivaroxaban in patients with a recent acute NEJMoa1709118.
coronary syndrome. N Engl J Med 2012; 366: 9–19. 57. Warner TD, Armstrong PC, Curzen NP, et al.
49. Mega JL, Braunwald E, Murphy SA, et al. Dual antiplatelet therapy in cardiovascular
Rivaroxaban in patients stabilized after a disease: does aspirin increase clinical risk in the
ST-segment elevation myocardial infarction: presence of potent P2Y12 receptor antagonists?
results from the ATLAS ACS-2-TIMI-51 trial Heart 2010; 96: 1693–1694.
(Anti-Xa Therapy to Lower Cardiovascular 58. Armstrong PC, Dhanji AR, Tucker AT,
Events in Addition to Standard Therapy in et al. Reduction of platelet thromboxane A2
Subjects with Acute Coronary Syndrome – production ex vivo and in vivo by clopidogrel
Thrombolysis In Myocardial Infarction-51). J Am therapy. J Thromb Haemost 2010; 8: 613–615.
Coll Cardiol 2013; 61: 1853–1859.
59. Franchi F, Rollini F, Aggarwal N, et al.
50. Xarelto (rivaroxaban). Summary of product Pharmacodynamic comparison of prasugrel
characteristics. www.ema.europa.eu/docs/en_GB/ versus ticagrelor in patients with type 2 diabetes
document_library/EPAR_-_Product_Information/ mellitus and coronary artery disease: the
human/000944/WC500057108.pdf (accessed OPTIMUS (Optimizing Antiplatelet Therapy
April 2017). in Diabetes Mellitus)-4 study. Circulation 2016;
51. Miyazaki Y, Suwannasom P, Sotomi Y, et al. 134: 780–792.
Single or dual antiplatelet therapy after PCI. Nat 60. Franchi F, Rollini F, Cho JR, et al. Effects of
Rev Cardiol 2017; 14: 294–303.
dabigatran on the cellular and protein phase
52. Gargiulo G, Windecker S, Vranckx P, et al. of coagulation in patients with coronary artery
A critical appraisal of aspirin in secondary disease on dual antiplatelet therapy with aspirin
prevention: is less more? Circulation 2016; 134: and clopidogrel: results from a prospective,
1881–1906. randomised, double-blind, placebo-controlled
study. Thromb Haemost 2016; 115: 622–631.
53. Ohman EM, Roe MT, Steg PG, et al. Clinically
significant bleeding with low-dose rivaroxaban 61. Perzborn E, Heitmeier S and Laux V. Effects of
Visit SAGE journals online
versus aspirin, in addition to P2Y12 inhibition, rivaroxaban on platelet activation and platelet-
journals.sagepub.com/ in acute coronary syndromes (GEMINI-ACS-1): coagulation pathway interaction: in vitro and in
home/tah a double-blind, multicentre, randomised trial. vivo studies. J Cardiovasc Pharmacol Ther 2015;
SAGE journals Lancet 2017; 389: 1799–1808. 20: 554–562.

366 journals.sagepub.com/home/tah

You might also like