Вы находитесь на странице: 1из 8

412 Review in depth

Novel oral anticoagulants in the management of coronary


artery disease
Sean R. McMahona, Kathleen Brummel-Ziedinsb and David J. Schneidera

Despite advances in interventional and pharmacologic an increased risk of bleeding. Future studies will need to
therapy, survivors of myocardial infarction remain at an identify optimal treatment combinations for selected
increased risk of subsequent cardiovascular events. Initial patients and conditions that address both the appropriate
pharmacological management includes both platelet combination of therapy and the appropriate dosage of each
inhibition and parenteral anticoagulation, whereas long- agent when used in combination. Coron Artery Dis
term pharmacological therapy relies on antiplatelet therapy 27:412–419 Copyright © 2016 Wolters Kluwer Health, Inc.
for prevention of thrombotic complications. Biomarkers All rights reserved.
showing ongoing thrombin generation after acute coronary Coronary Artery Disease 2016, 27:412–419
syndromes suggest that anticoagulants may provide
additional benefit in reducing cardiovascular events. We Keywords: coronary artery disease, novel oral anticoagulants, thrombosis

review the pharmacokinetics of novel anticoagulants, a


Cardiology Unit, Department of Medicine, Cardiovascular Research Institute and
b
clinical trial results, the role of monitoring, and future Department of Biochemistry, University of Vermont, Burlington, Vermont, USA

directions for the use of novel oral anticoagulants in the Correspondence to Sean R. McMahon, MD, Cardiology Unit, Department of
treatment of coronary artery disease. Clinical trials have Medicine, Cardiovascular Research Institute, University of Vermont, 111
Colchester Avenue, Burlington, VT 05401, USA
shown that long-term use of oral anticoagulants decreases Tel: + 1 802 847 3734; fax: + 1 802 847 8818;
the risk of cardiovascular events, but they do so at a cost of e-mail: sean.mcmahon@uvmhealth.org

Introduction Pharmacokinetics of selected novel oral


Despite advancements in cardiac revascularization and anticoagulants
pharmacologic therapy, survivors of myocardial infarction Clinical trials assessing the efficacy of novel antic-
(MI) remain at significant risk of subsequent cardiovas- oagulants in patients with CAD have been conducted
cular events. Pharmacological treatment of acute coronary with ximelagatran, darexaban, dabigatran, rivaroxaban,
syndromes (ACSs) includes an initial strategy that com- and apixaban. The mechanistic and pharmacokinetic
bines antiplatelet therapy and parenteral anticoagulation. characteristics of agents that are available for clinical use
This early strategy frequently includes revascularization. (dabigatran, rivaroxaban, edoxaban, and apixaban) will be
Subsequent thrombotic risk is limited most commonly reviewed.
with antiplatelet therapy alone. Despite advances in
antiplatelet therapy, the risk of cardiovascular death, Dabigatran is a prodrug whose metabolite is a reversible
stroke, or MI is ∼ 10% during the first year. Although an competitive direct-acting inhibitor of thrombin.
elevated risk of cardiovascular events is apparent through Dabigatran has a bioavailability of 3–7%. Peak plasma
12 months, the greatest burden of risk is observed during concentrations are observed within 2 h and the active
the first 30 days (∼50% of the events that will occur after metabolite has a clearance half-life of ∼ 12–17 h.
1 year are observed during the first month) [1,2]. The Dabigatran is excreted 80% unchanged in the urine.
beneficial effects of antiplatelet therapy [1,2] indicate the Clearance is facilitated by P-glycoprotein cell membrane
importance of platelets in thrombotic complications; pumps that are susceptible to inhibition by medications
however, evidence of ongoing thrombin generation and such as amiodarone and verapamil [6,7].
activity suggests that thrombotic complications are not
solely mediated by platelet activation [3,4]. Consistent Rivaroxaban is a highly selective direct-acting inhibitor of
with this observation, biochemical markers of ongoing factor Xa. The parent compound has anticoagulant
thrombin generation have been linked to an increased properties and peak plasma concentrations are observed
risk of subsequent cardiovascular events [5]. These 2–4 h after ingestion. The clearance half-life is 7–11 h in
observations suggest that anticoagulants may be effective the young and 11–13 h in elderly patients. Absorption is
in reducing subsequent risk after an ACS. This review not affected by food intake and no first-pass metabolism
will focus on our current understanding of the pharma- has been observed. The bioavailability is 80–100%.
cokinetics, recent trial results, the role of monitoring, and Clearance is mediated predominantly by metabolism
future directions for the use of novel oral anticoagulants (approximately two-thirds) in the liver by CYP3A4 and
in the treatment of coronary artery disease (CAD). the rest is excreted unchanged in the urine [8].
0954-6928 Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved. DOI: 10.1097/MCA.0000000000000387

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.


NOACs in the management of CAD McMahon et al. 413

Apixaban is another highly selective direct-acting inhi- major bleeding. The lack of considerable numbers of
bitor of factor Xa. The parent compound has antic- patients treated with percutaneous coronary intervention
oagulant properties and a bioavailability of ∼ 50%. Peak (PCI) prevents assessment of an effect on stent throm-
plasma concentrations are achieved in ∼ 3.5 h and the bosis or restenosis. Treatment with VKA carried a sig-
clearance half-life is ∼ 12 h. More than 50% of apixaban is nificant risk of bleeding complications, but this treatment
excreted unchanged in feces or urine. The rest is cleared reduced the incidence of ischemic events [22,23].
primarily by oxidative metabolism that is catalyzed by Treatment of CAD with coronary stents in patients with
CYP3A4/5 [9,10]. atrial fibrillation has led to the combination of VKA with
dual antiplatelet therapy. Observational studies have
Edoxaban is a factor Xa inhibitor that has not been
identified a marked increase in the risk of bleeding
evaluated in the treatment of patients with CAD. The
complications in this setting [24]. One large observational
parent compound has anticoagulant properties and peak
study found that the combination of VKA with dual
plasma concentrations are observed 1–2 h after ingestion.
antiplatelet therapy led to a four-fold increased risk of
Absorption is not affected by food intake and bioavail-
bleeding requiring hospitalization with a number needed
ability is ∼ 62%. Clearance half-life is 10–14 h.
to harm of 12.5 [25].
Elimination is mediated by the kidneys (50%) as well as
by biliary and intestinal excretion. Because an increased The use of clopidogrel with or without aspirin in patients
incidence of ischemic stroke was observed in patients receiving oral anticoagulant therapy and undergoing a
with a creatinine clearance greater than 95 ml/min, percutaneous coronary intervention trial (WOEST) was
edoxaban is not recommended in these patients [11,12]. the first prospective randomized trial to challenge the
necessity of ASA after coronary stenting in patients
Direct-acting anticoagulants exert more consistent antic-
treated with clopidogrel and VKA [26]. The WOEST trial
oagulant effects because they are not dependent on
was a single-center open-label multicenter randomized
endogenous cofactors for their anticoagulant properties.
control trial with 573 patients on VKA undergoing PCI.
Their direct effect on activated coagulation factors may
Patients were randomized to clopidogrel alone (double
enable them to exert pleiotropic effects [13] (Fig. 1).
therapy) or ASA and clopidogrel (triple therapy). After a
Direct-acting anticoagulants inhibit clot-bound factor Xa
1-year follow-up, the study showed a marked and highly
and prothrombinase more effectively than heparins.
significant reduction in the incidence of bleeding com-
Because thrombus may persist up to 30 days after plaque
plications (the primary endpoint). In the triple-therapy
rupture [14], the inhibition of clot-bound thrombin may
arm, there was a significant increase in all bleeding epi-
reduce cardiac events. Direct-acting anticoagulants may
sodes driven by thrombolysis in myocardial infarction
provide benefits independent of the prevention of
(TIMI) minor bleeding [14.4 vs. 44.4%, hazard ratio
thrombosis. Both thrombin and factor Xa activate inflam-
(HR): 0.36, confidence interval (CI): 0.26–0.50].
matory mediators and promote fibrosis. Proinflammatory
Surprisingly, the incidence of thrombotic complications
and fibrotic effects have been found in cardiac, renal, and
was lower with dual therapy than with triple therapy. The
lung tissue exposed to ischemia [15–17]. Direct thrombin
incidence of the combined secondary endpoint that
inhibition has been shown to improve organ survival in
included death, MI, target-vessel revascularization,
experimental renal preparations exposed to ischemia and
stroke, or stent thrombosis was lower in the double-
to reduce inflammation/fibrosis in bleomycin-induced
therapy group (11.1 vs. 17.6%, HR: 0.60, CI: 0.38–0.94;
pulmonary fibrosis [18,19]. Thus, treatment with direct-
number needed to treat 15).
acting anticoagulants after ACS may reduce postischemic
injury and fibrosis, and thereby enhance survival by pre- At this time, the FDA has not approved the use of novel
serving cardiac function. anticoagulants in the treatment of CAD. The optimal
treatment strategy for patients with atrial fibrillation who
Trials of long-term anticoagulation in the require dual antiplatelet therapy after coronary stenting
management of CAD has not been defined. The most common treatment
The use of long-term treatment with anticoagulants for strategy used in these patients is to combine VKA with
the prevention of thrombotic cardiovascular events in dual antiplatelet therapy (triple therapy) and attempt to
CAD was first studied with the use of vitamin K limit the duration of triple therapy. Several ongoing trials
antagonists (VKAs). Their use has been investigated with seek to evaluate the efficacy and risk of selected com-
mixed results in studies dating back to the 1980s [20]. A binations of antiplatelet therapies and anticoagulants in
meta-analysis of these trials suggested benefit in ACS patients with and without atrial arrhythmia.
patients with a low or an intermediate risk for bleeding.
However, because these trials were completed before Ximelagatran: the ESTEEM trial
coronary stents were commonly used, they do not include Ximelagatran, a direct thrombin inhibitor that was not
patients who underwent coronary stenting [21]. These approved for clinical use by the FDA, was the first novel
studies were plagued by variable international normal- oral anticoagulant studied in the prevention of cardio-
ized ratio (INR) values that likely increased the risk of vascular events after ACS. The Oral ximelagatran for

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.


414 Coronary Artery Disease 2016, Vol 27 No 5

Fig. 1

Intrinsic pathway

XII

XIIa

XI XIa

VIIIa Endothelial and


smooth muscle cell
IX IXa proliferation

X Platelet activation
TF VIIa

Extrinsic pathway Xa Inflammation


Exposure or expression
of tissue factor (TF) Xa Va
Coagulation promotion
II IIa

Fibrinogen Fibrin clot formation

The coagulation cascade and pleiotropic interactions.

secondary prophylaxis after myocardial infarction trial Consistent with the occurrence of thrombotic events, the
(ESTEEM trial) was a phase II multicenter, multi- extent of reduction was not greater with higher doses of
national, randomized-controlled trial assessing the effi- drug [28].
cacy and safety of four doses of ximelagatran in addition
Major bleeding was infrequent, but doubled in the
to ASA in patients with ACS [27]. The primary outcome ximelagatran group compared with placebo [23 patients
was a composite of death, MI, and recurrent ischemia. (1.8%) vs. six patients (0.9%), HR: 1.97, CI: 0.80–4.84].
Patients were treated with ASA 75–160 mg daily and Bleeding that led to the discontinuation of study medi-
excluded if they were treated with thienopyridines. Of cation was increased [83 (8%) vs. 13 (2%) HR: 3.35, CI:
the 1900 patients with biomarker evidence of MI enrol- 1.87–6.01] as was total bleeding [273 (22%) vs. 83 (13%)
led, 61% had ST-elevation myocardial infarction HR: 1.76, CI: 1.38–2.25] in those treated with ximela-
(STEMI) and 51% of those patients were treated with gatran. Unlike the effect on the risk of thrombosis, a
lytic therapy. greater incidence of bleeding was observed with higher
The results indicated that ximelagatran combined with doses of ximelagatran. Accordingly, the results from the
aspirin compared with placebo plus aspirin was associated ESTEEM trial were consistent with those found with
VKA, suggesting that greater intensity anticoagulation
with a reduction in the composite endpoint of death,
does not add benefit, but clearly increases the risk of
nonfatal MI, and severe recurrent ischemia. There was a
bleeding.
3.6% (12.7 vs. 16.3%, HR: 0.71, CI: 0.55–0.90) absolute
risk reduction and a 24% relative risk reduction. There
was no evidence of greater benefit with higher or lower Dabigatran: the RE-DEEM trial
doses among the four doses of ximelagatran studied. The Dabigatran was the second of the novel oral antic-
reduction in thrombotic risk was apparent during the first oagulants studied in patients with MI and the first in
month maintained through the treatment period. which all patients were treated with dual antiplatelet
Treatment with ximelagatran led to a consistent and therapy. The dabigatran vs. placebo in patients with
persistent reduction in prothrombin fragment 1.2 (F1.2, a ACSs on dual antiplatelet therapy trial (RE-DEEM trial)
marker released when thrombin is formed) and D-dimer was a phase II trial that assessed the safety and efficacy of
(a marker produced when plasmin cleaves fibrin). dabigatran for long-term (6 months) treatment after ACS

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.


NOACs in the management of CAD McMahon et al. 415

[29]. RE-DEEM was a double-blind, placebo-controlled, Fig. 2


dose-escalation trial that enrolled a total of 1861 patients
Placebo APPRAISE 2
who were treated, on average, 7.5 days after STEMI or
RE-DEEM ATLAS 2
non-ST-elevation myocardial infarction (NSTEMI) (60
9%
and 40%, respectively). Patients were randomized to
dabigatran (dose-escalation plan, randomized to 50, 75, 8%
110, or 150 mg) plus standard therapy of thienopyridine 7%
and aspirin (no dose limits) or placebo plus standard 6%
therapy. A total of 99% of patients were on dual anti- 5%
platelet treatment at randomization. The primary out-
4%
come was clinically relevant minor bleeding and major
3%
bleeding. The secondary outcomes included the con-
centration of D-dimer and the incidence of cardiovascular 2%
ischemic events. Cardiovascular ischemic events inclu- 1%
ded a composite of death, nonfatal MI, nonhemorrhagic 0%
stroke, MI, and recurrent ischemia. Of those enrolled, Dabigatran Apixaban Rivaroxaban Rivaroxaban
54.5% of patients underwent PCI for the index event. (combined doses) 5 mg BID 2.5 mg BID 5 mg BID

The study indicated that dabigatran compared with pla- Major bleeding among patients receiving novel oral anticoagulants in
cebo resulted in significantly increased rates of clinically addition to dual antiplatelet therapy. BID, twice daily.
relevant minor and major bleeding as defined by the
International Society of Thrombosis and Hemostasis
(ISTH) [30], with a four-fold increase [7.8 (110 mg), 7.9% Fig. 3
(150 mg)] in the high-dose group compared with placebo
(2.2%). Higher doses of dabigatran were associated with Placebo APPRAISE 2
more bleeding events. When the results from the two RE-DEEM ATLAS 2
lower dose groups were combined, the absolute risk of 12%
major bleeding was increased by 1%.
10%
Treatment with dabigatran decreased the concentration
of D-dimer in a non-dose-dependent manner by an 8%
average of 37 and 45% at weeks 1 and 4, respectively
(P < 0.001). Patients treated with dabigatran and showing 6%
a reduction in the D-dimer concentration at week 1 had a
significantly lower incidence of the composite endpoint 4%
of cardiovascular death, nonfatal MI, or nonhemorrhagic
2%
stroke (HR: 0.47, CI: 0.26–0.82). Cardiovascular ischemic
events were less common in patients treated with the 0%
higher doses of dabigatran (110 and 150 mg), but this was Dabigatran Apixaban Rivaroxaban Rivaroxaban
at the cost of an observed four-fold increased incidence of (combined doses) 5 mg BID 2.5 mg BID 5 mg BID
bleeding (Figs 2 and 3). Consistent with the results from
Combined endpoints of cardiovascular death, nonfatal myocardial
the ESTEEM trial, these results support lower intensity infarction, or stroke. BID, twice daily.
therapy to reduce the risk of bleeding. Further, these
results suggest that suppression of a biomarker of
thrombosis (D-dimer) may predict benefit.
clinically relevant nonmajor bleeding events as defined by
Darexaban: the RUBY-1 trial the modified version of the ISTH [30]. The secondary
The RUBY-1: a randomized, double-blind, placebo- efficacy outcome was a composite of death, stroke, MI,
controlled trial of the safety and tolerability of the severe recurrent ischemia, and systemic thromboembolism.
novel oral factor Xa inhibitor darexaban (YM150) fol-
lowing ACS trial (RUBY-1 trial) was a phase II rando- As anticipated, the results showed a higher incidence of
mized, double-blind, placebo-controlled trial of the safety bleeding in the treatment group that was dose dependent
and tolerability of the factor Xa inhibitor darexaban (HR: 2.28, CI: 1.13–4.60). Cumulative bleeding events
(YM150) after ACSs [31]. RUBY-1 enrolled 1279 patients were 6.2, 6.5, and 9.3% in the 10, 30, and 60 mg dose
with either high-risk NSTEMI (29%) or STEMI (71%) group, respectively, compared with 3.9% in the placebo
to receive one of six doses of darexaban or placebo in group. The range of major and clinically relevant minor
addition to clopidogrel plus ASA (ASA dose 75–325 daily) bleeding was 5.6% for the lowest dose (HR: 1.78, CI:
for 6 months. The primary outcome was major and 0.68–4.60) and 11.3% for the high-dose group (HR: 3.8,

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.


416 Coronary Artery Disease 2016, Vol 27 No 5

CI: 1.66–8.68) compared with placebo (2.8%). Consistent randomized to rivaroxaban 5 mg twice daily, rivaroxaban
with previous results, a decrease in composite efficiency 2.5 twice daily, or placebo. Patients were excluded if they
events was observed with the lowest dose tested. Rates had a history of intracranial hemorrhage, ischemic cere-
of instent thrombosis were low, but more common in brovascular accident, or transient ischemic attack. All
those receiving darexaban (1.1 vs. 0.4%). patients were treated with a proton pump inhibitor.
Patients were randomized within 7 days of admission for
Apixaban: the APPRAISE-2 trial ACS that included unstable angina (24%), NSTEMI
The apixaban with antiplatelet therapy after acute cor- (26%), or STEMI (50%).
onary syndrome trial (APPRAISE-2 trial) is a randomized, The results indicated a lower incidence of ischemic
double-blind, placebo-controlled trial that sought to events, but again at the cost of a higher rate of bleeding
evaluate the efficacy and safety of long-term treatment complications. The primary endpoint, a composite of
with apixaban after ACS for the prevention of acute cardiovascular death, MI, or stroke, was achieved in 8.9%
ischemic events [32]. The APPRAISE-2 trial enrolled of patients randomized to any dose of rivaroxaban com-
7392 patients and randomized them to received apixaban pared with 10.7% in the placebo group (relative risk
5 mg twice daily or placebo in addition to the P2Y12 reduction: 16%, HR: 0.84, CI: 0.74–0.96). A similar
receptor antagonist and low-dose aspirin. A dose of reduction in the incidence of ischemic events was
2.5 mg twice daily was used in patients with an estimated observed in patients treated with the 2.5 mg dose (9.1%,
creatinine clearance of less than 40 ml/min. Enrolled P = 0.02 vs. placebo) compared with the 5 mg dose (8.8%,
patients had ACS including NSTEMI (42%), STEMI P = 0.03 vs. placebo).
(40%), and unstable angina with the presence of dynamic
ST-segment changes (18%). Patients were enrolled at a The low-dose (2.5 mg twice daily) rivaroxaban group
median of 6 days after the index event and followed for showed a significant reduction in the incidence of car-
up to 240 days. The primary efficacy outcome was a diovascular death and overall death that was not observed
composite of cardiovascular death, MI, or ischemic in the high-dose group. Cardiovascular death occurred in
stroke. The primary safety outcomes were defined as 2.7% of those on 2.5 of rivaroxaban compared with 4.1%
TIMI major and minor bleeding, major, or clinically in the placebo group (HR: 0.66, CI: 0.51–0.86) and the
relevant nonmajor bleeding as defined by the ISTH and overall incidence of death was 2.9 and 4.5%, respectively
GUSTO [30,33,34]. Approximately 52% of the patients (HR: 0.68, CI: 0.53–0.87). The incidence of stent
enrolled underwent angiography and 44% had PCI. thrombosis was 21% lower in patients treated with riv-
aroxaban (2.3 vs. 2.9%, HR: 0.69, CI: 0.51–0.93).
The study was stopped because of an increase in major Rivaroxaban decreased the concentrations of biomarkers
bleeding without evidence of decreased ischemic events. that reflect ongoing thrombosis (D-dimer and PF1.2) that
After a median follow-up of 241 days, there was a similar were assessed 180 days after enrollment [4].
distribution of the primary outcome of cardiovascular
death, MI, or ischemic stroke between the apixaban TIMI major bleeding not related to coronary artery
group and placebo (7.5 vs. 7.9% respectively, HR: 0.95, bypass grafting and intracranial hemorrhage were
CI: 0.80–1.11). The primary safety outcome of TIMI increased in patients treated with rivaroxaban. When the
major bleeding occurred in 1.3% of the patients receiving results with both doses were combined, the rate of TIMI
apixaban and 0.5% of those receiving placebo (HR: 2.59, major bleeding was increased four-fold in those treated
CI: 1.50–4.46). In addition, in the treatment group, sig- with rivaroxaban (2.1 vs. 0.6%, HR: 3.96, CI: 2.46–6.38)
nificantly more patients had fatal bleeding, intracranial and the rate of intracranial bleeding was increased three-
hemorrhage, ISTH major or clinically relevant nonmajor fold (0.6 vs. 0.2%, P = 0.009). As would be expected on
bleeding, bleeding requiring transfusion, and total the basis of previous trials, a lower incidence of bleeding
bleeding. The results of this large trial were not con- was found with the 2.5 mg dose (Table 1). The rate of
sistent with previous results. An increased risk of TIMI major bleeding was nonsignificantly lower in the
bleeding was not offset by a reduction in the incidence of 2.5 mg dose group (1.8 vs. 2.4, P = 0.12). However, the
thrombotic complications. rates of TIMI minor bleeding (0.9 vs. 1.6%, P = 0.046),
TIMI bleeding requiring medical attention (12.9 vs.
16.2%, P < 0.001), and fatal bleeding (0.1 vs. 0.4%,
Rivaroxaban: the ATLAS ACS 2 TIMI 51 trial
P = 0.04) were all significantly lower in the 2.5 mg group
The rivaroxaban in patients with a recent acute coronary
compared with the 5 mg group.
syndrome trial (ATLAS-2 TIMI 51 trial) was a rando-
mized, double-blind, placebo-controlled phase 3 trial that The results from the ATLAS-2 trial and the APPRAISE
assessed thrombotic risk reduction with rivaroxaban in trial were quite different (Table 1). In particular, the
addition to standard medical therapy after ACS [35]. All ATLAS trial showed a significant reduction in events and
patients took low-dose aspirin (75–100 mg daily) and a the lower dose (2.5 mg twice daily) was associated with a
thienopyridine [clopidogrel (93%) or ticlopidine]. The significant reduction in mortality. The ATLAS trial
trial included three treatment arms; 15 526 patients were excluded patients with previous stroke, whereas this

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.


NOACs in the management of CAD McMahon et al. 417

Table 1 Clinical trials and major findings of oral anticoagulants studied in coronary artery disease with monitoring and reversal strategies
Medication Dose Antiplatelets Death, MI, stroke (placebo) Major bleeding (placebo) Monitoring Reversal
Dabigatran (mg) 50, 75, 110, or 150 DAPT 4.0% (3.8%) 7.9% (2.2%) Ecarin clotting time Idarucizumab
Apixaban (mg b.i.d.) 5.0 DAPT 7.5% (7.9%) 1.3% (0.5%) Factor Xa assay PCC
Rivaroxaban (mg b.i.d.) 2.5 DAPT 9.1% (10.7%) 1.8% (0.6%) Factor Xa assay PCC
5.0 DAPT 8.8% (10.7%) 2.4% (0.6%)
Coumadin (%) INR adjusted Plavix alone 11.1 vs. 17.6 19.4 vs. 44.4 PT/INR VK, PCC, FFP

b.i.d., twice daily; DAPT, dual antiplatelet therapy; FFP, fresh-frozen plasma; INR, international normalized ratio; MI, myocardial infarction; PCC, prothrombin complex
concentrate; PT, prothrombin time; VK, vitamin K.

exclusion was not applied to patients enrolled in the concentration of factor Xa inhibitor that led to doubling
APPRAISE trial. In addition, the ATLAS enrolled a of the PT [38]. Thus, the PT cannot be used to deter-
higher percentage of patients with STEMI than the mine the concentration of rivaroxaban or apixaban, but
APPRAISE trial (50 vs. 39%, respectively). Rivaroxaban this test may be used in emergent situations to qualita-
and apixaban have similar mechanisms of action. The tively assess hemostatic function. In contrast, when
lower dose of rivaroxaban yielded the greatest net benefit appropriate calibration is performed, chromogenic anti-
(combination of a lower risk of ischemic/thrombotic factor Xa assays can quantify the concentration of rivar-
events plus a lower risk of bleeding events). This dosage oxaban and apixaban [38,39].
would be expected to result in a less anticoagulant effect
Although there is no commercially available direct-acting
than the dosage of apixaban used in APPRAISE.
(specific) reversal agent for rivaroxaban and apixaban,
Accordingly, the ATLAS-2 trial suggests that the pre-
administration of the prothrombin complex concentrate
vention of cardiovascular events after MI in patients who
has been shown to exert a rapid reversal effect on bio-
are treated with dual antiplatelet therapy and low-dose
chemical tests [40]. Reversal was quantified by mea-
anticoagulation may be more effective than dual anti-
surement of the PT and endogenous thrombin potential
platelet therapy alone. In addition, the low-dose rivar-
and noted to be immediate, with an effect that persisted
oxaban (2.5 mg twice daily) was associated with a lower
for more than 24 h. Administration of prothrombin com-
risk of bleeding.
plex concentrate did not effectively reverse biochemical
tests in blood from patients treated with dabigatran.
Anticoagulation monitoring and reversal of
anticoagulant effects Two specific reversal agents have been evaluated.
The prothrombin time (PT) and the INR that was Andexanet-α has been shown to rapidly reduce antifactor
developed to standardize measurements have been used Xa activity by 97% [41,42]. Andexanet-α is a modified
to enhance prevention of thrombosis and limit the inactive factor Xa protein with a high affinity for factor Xa
occurrence of bleeding in patients treated with a VKA. inhibitors that has been shown to reverse the effects of
Because novel oral anticoagulants are direct acting and both apixaban and rivaroxaban as well as other Xa inhi-
have predictable pharmacokinetic and pharmacodynamic bitors. Antifactor Xa activity was reduced within 2–5 min
profiles, they were developed to be used without regular of administration. A continuous infusion was necessary to
monitoring. The predictable pharmacokinetic and phar- sustain the reversal of antifactor Xa activity. Ciraparantag
macodynamic profiles are likely to be offset, to some is another agent under evaluation for the reversal of
extent, by the considerable interindividual variability in anticoagulants [43,44]. Ciraparantag is a synthetic mole-
the concentration of coagulation factors and the stimulus cule with a high affinity for factor Xa inhibitors as well as
driving their activation. Accordingly, monitoring may be the oral thrombin inhibitor dabigatran. Ciraparantag has
useful under circumstances where substantial inter- been referred to as a universal reversal agent because of
individual variability would be expected. its ability to reverse unfractionated heparin, low-
molecular-weight heparin, and the direct-acting antic-
Inhibitors of factor Xa oagulants. After injection, ciraparantag binds rapidly and
Rivaroxaban and apixaban exert their anticoagulation inhibits anticoagulants. Ciraparantag decreases clotting
effects by inhibiting coagulation factor Xa. Tests of coa- time within 10 min and its effects are sustained for 24 h.
gulation that are affected by the inhibition of factor Xa
include PT, activated partial thromboplastin time Inhibitors of thrombin
(aPTT), and chromogenic assays that directly measure Similar to rivaroxaban and apixaban, the anticoagulant
the activity of factor Xa. Both factor Xa inhibitors prolong effect and hence the concentration of dabigatran cannot
the PT and have a direct linear correlation with the be reliably measured with the use of traditional assays.
plasma concentration of the anticoagulant [36,37]. The PT and its derived INR are insensitive to the effects
However, thromboplastin reagents are not specific to of dabigatran and provide an imprecise assessment of the
factor Xa and vary widely in their sensitivity. One effect of this agent on clotting. Similar to its effect on the
laboratory found a 2.6- to eight-fold difference in the PT, dabigatran exerts a nonlinear effect on the aPTT.

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.


418 Coronary Artery Disease 2016, Vol 27 No 5

Accordingly, the aPTT may be used as a qualitative Results from trials that used anticoagulants for long-term
measurement to inform clinicians of the presence or treatment after ACS suggest that when antiplatelet
absence of dabigatran, but not as a quantitative mea- therapy and anticoagulant therapy are combined, the
surement. Alternative methods that can assess the effect dosage of each agent may require adjustment. Results
of direct-acting antithrombins include the ecarin clotting from the ATLAS-2 trial showed that a reduction in
time (ECT) and the thrombin time (TT). Both the TT thrombotic risk can be achieved with low-dose antic-
and ECT show a linear response to dabigatran and pro- oagulation, allowing for a relatively lower risk of bleed-
vide an accurate quantitative assessment of thrombin ing. The effect on clinical events shown in ATLAS-2 was
inhibition. The ECT has been shown to be the most evident with biochemical marker testing when higher
accurate and specific [45]. A promising alternate assay doses of anticoagulant did not have greater effect on
under evaluation is the HEMOCLOT Thrombin thrombosis biomarkers in the RE-DEEM trial and
Inhibitor assay, which has been shown to be highly ESTEEM trial. Similarly, lower doses of darexaban
sensitive [46]. appeared to be more effective in the phase II RUBY-1
trial. The results from the ATLAS-2 trial were most
Recently, the FDA approved a specific reversal agent for
compelling, showing a survival benefit with the low-dose
dabigatran. Idarucizumab is a monoclonal antibody frag-
therapy that was not apparent with the high dose. A
ment that binds dabigatran with an affinity 350 times
difference in the intensity of anticoagulant effect could
higher than thrombin [47]. The idarucizumab for dabi-
be a determinant of the apparent discrepancy between
gatran reversal (RE-VERSE AD) clinical trial assessed
the results from ATLAS-2 compared with the
the efficacy of Idarucizumab [48]. Treatment with
APPRAISE-2 trial.
Idarucizumab produced rapid reversal of the dilute TT
and ECT – effects were observed within minutes and The results from the WOEST trial suggest a different
sustained for at least 24 h. Idarucizumab is administered paradigm. Rather than simply adding new therapy to
as two 2.5 g (50 ml) bolus infusions within 15 min and existing treatment strategies, the WOEST trial results
does not require repeated dosing or a continuous infu- suggest that treatment strategies may need to be tailored
sion. Idarucizumab has not found to exert a pro- to specific clinical situations. Identification of optimal
thrombotic effect [49]. treatment combinations and the ability to tailor therapy
to the needs of an individual would be supported greatly
by the availability of biochemical tests that have been
validated to predict clinical outcomes.
Future directions
Biochemical testing on blood from patients who have
experienced a MI has shown that patients have variable Conclusion
extents of ongoing thrombin generation. Greater ongoing Despite advances in therapies that reduce cardiovascular
thrombin generation and activity is associated with an events after ACS, continued opportunities for improve-
increased risk of subsequent cardiovascular events. ment remain. Novel oral anticoagulants may be an
Clinical trials have shown that long-term treatment with important adjunct in selected patients. Trials suggest that
oral anticoagulants decreases the risk of cardiovascular long-term anticoagulant therapy prevents ischemic com-
events, but the treatment does so at a cost of an increased plications at a cost of a higher risk of bleeding. Continued
risk of bleeding. Strategies that suppress thrombin gen- research to develop novel treatment strategies that use
eration without markedly increasing the risk of bleeding optimal combinations and dosages is necessary. Research
would improve patient care. The results from clinical in this area will be aided by the development of clinically
trials suggest two options. The ATLAS trial suggests that validated biochemical testing that can be used to tailor
the combination of very low doses of anticoagulants with therapies to the needs of individual patients.
antiplatelet agents may achieve this end. The WOEST
trial suggests that one component of therapy; aspirin in Acknowledgements
this case, may contribute a substantial component of the Conflicts of interest
increased risk of bleeding without markedly reducing the Dr Schneider reports grant funding from Bristol Myers
risk of thrombotic events when an anticoagulant is com- Squib and Janssen Pharmaceuticals as well as honorarium
bined with P2Y12 inhibition. Comparison of the ATLAS- from Janssen Pharmaceuticals. For the remaining authors
2 and APPRAISE trials also suggests that clinical char- there are no conflicts of interest.
acteristics may be useful to guide therapy. Specifically,
patients with a previous stroke appear to be at a higher
References
risk of intracranial hemorrhage when anticoagulants are 1 Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W,
combined with antiplatelet therapy. Because the Gottlieb S, et al. Prasugrel versus clopidogrel in patients with acute coronary
ATLAS-2 trial also included more patients with STEMI syndromes. N Engl J Med 2007; 357:2001–2015.
2 Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al.
than the APPRAISE trial, the efficacy of selected thera- Ticagrelor versus clopidogrel in patients with acute coronary syndromes.
pies may be impacted by the clinical syndrome. N Engl J Med 2009; 361:1045–1057.

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.


NOACs in the management of CAD McMahon et al. 419

3 Merlini PA, Bauer KA, Oltrona L, Ardissino D, Cattaneo M, Belli C, et al. anticoagulant therapy and undergoing percutaneous coronary intervention:
Persistent activation of coagulation mechanisms in unstable angina and an open-label, randomized, controlled trial. Lancet 2013; 381:1107–1115.
myocardial infarction. Circulation 1990; 90:61–68. 27 Wallentin PL, Wilcox RG, Weaver WD, Emanuelsson H, Goodvin A,
4 Gibson M, Mega J, Jennings LK, Mohanavelu S, Misselwitz F, Plotnikov A, Nyström P, et al. Oral ximelagatran for secondary prophylaxis after myocardial
et al. Effect of rivaroxaban on markers of coagulation in patients with acute infarction: the ESTEEM randomized controlled trial. Lancet 2003; 362:9386.
coronary syndromes: an ATLAS ACS-TIMI 46 substudy. Circulation 2009; 28 Christersson C, Oldgren J, Bylock A, Wallentin L, Siegbahn A. Long-term
120:S1035. treatment with ximelagatran, an oral direct thrombin. J Thromb Haemost
5 Christersson C, Oldgren J, Bylock A, Siegbahn A, Wallentin L. Early decrease 2005; 3:2245–2253.
in coagulation activity after myocardial infarction is associated with lower risk 29 Oldgren J, Budaj A, Granger C, Khder Y, Roberts J, Siegbahn A, et al.
of new ischaemic events: observations from the ESTEEM Trial. Eur Heart J Dabigatran vs. placebo in patients with acute coronary syndromes on dual
2007; 28:692–698. antiplatelet therapy: a randomized, double blind, phase II trial. Eur Heart J
6 Stangier J, Stahle H, Rathgen K, Fuhr R. Pharmacokinetics and 2011; 32:2781–2789.
pharmacodynamics of the direct oral thrombin inhibitor dabigatran in healthy 30 Schulman S, Kearon C. Definition of major bleeding in clinical investigations
elderly subjects. Clin Pharmacokinet 2008; 47:47–59. of antihemostatic medicinal products in non-surgical patients. J Thromb
7 Stangier J. Clinical pharmacokinetics and pharmacodynamics of the oral Haemost 2005; 3:692–694.
direct thrombin inhibitor dabigatran etexilate. Clin Pharmacokinet 2008; 31 Steg PG, Mehta SR, Jukema JW, Lip GY, Gibson CM, Kovar F, et al. RUBY-
47:285–295. 1: a randomized, double blind, placebo controlled trial of the safety and
8 Kreutz R. Pharmacodynamic and pharmacokinetic basics of rivaroxaban. tolerability of the novel oral factor Xa inhibitor darexaban (YM150) following
Fundam Clin Pharmacol 2012; 26:27–32. acute coronary syndrome. Eur Heart J 2011; 32:2541–2554.
9 Bhanwra S, Ahluwalia K. The new factor Xa inhibitor: apixaban. J Pharmacol 32 Alexander JH, Lopes RD, James S, Kilaru R, He Y, Mohan P, et al. Apixaban
Pharmacother 2014; 5:12–14. with antiplatelet therapy after acute coronary syndrome. N Engl J Med 2011;
10 Raghavan N, Frost CE, Yu Z, He K, Zhang H, Humphreys WG, et al. 635:699–708.
Apixaban metabolism and pharmacokinetics after oral administration 33 Rao SV, Eikelboom JA, Granger CB, Harrington RA, Califf RM, Bassand JP.
to humans. Drug Metab Dispos 2009; 37:74–81. Bleeding and blood transfusion issues in patients with non-ST-segment
11 Ogata K, Mendell-Harary J, Tachibana M, Masumoto H, Oguma T, Kojima M, elevation acute coronary syndromes. Eur Heart J 2007; 28:1193–1204.
et al. Clinical safety, tolerability, pharmacokinetics, and pharmacodynamics of 34 THE GUSTO INVESTIGATORS. An international randomized trial
the novel factor Xa inhibitor edoxaban in healthy volunteers. J Clin Pharmacol comparing four thrombolytic strategies for acute myocardial infarction.
2010; 50:743–753. N Engl J Med 1993; 329:673–682.
12 Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL, 35 Mega JL, Braunwald E, Wiviott SD, Bassand JP, Bhatt D, Bode C, et al.
et al. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J
Med 2012; 366:9–19.
2013; 369:2093–2104.
36 Samama MM, Contant G, Spiro TE, Perzborn E, le Flem L, Gourmelin Y, et al.
13 Schneider DJ. Potential contribution of pleiotropic effects of direct
Laboratory assessment of rivaroxaban: a review. Thromb J 2013; 11:11.
anticoagulants to clinical benefits. Drug Dev Res 2013; 74:472–477.
37 Samama MM, Martinoli JL, le Flem L, Guinet C, Plu-Bureau G, Depasse F,
14 Van Belle E, Lablanche JM, Bauters C, Renaud N, McFadden EP,
et al. Assessment of laboratory assays to measure rivaroxaban – an oral,
Bertrand ME. Coronary angioscopic findings in the infarct-related vessel
direct factor Xa inhibitor. Thromb Haemost 2010; 103:815–825.
within 1 month of acute myocardial infarction: natural history and effect of
38 Barrett YC, Wang Z, Frost C, Shenker A. Clinical laboratory measurement of
thrombolysis. Circulation 1998; 97:26–33.
direct factor Xa inhibitors: anti-Xa assay is preferable to prothrombin
15 Mackman N. The role of tissue factor-thrombin pathway in cardiac ischemia-
time assay. Thromb Haemost 2010; 104:1263–1271.
reperfusion injury. Semin Vasc Med 2003; 3:193–198.
39 Samama MM, Contant G, Spiro TE. Evaluation of the anti-factor Xa
16 Grandaliano G, Di Paolo S, Monno R, Stallone G, Ranieri E, Pontrelli P, et al.
chromogenic assay for the measurement of rivaroxaban plasma concentrations
Protease-activated receptor 1 and plasminogen activator inhibitor 1
using calibrators and controls. Thromb Haemost 2012; 107:379–387.
expression in chronic allograft nephropathy: the role of coagulation and
40 Eerenberg ES, Kamphuisen PW, Sijpkens MK. Reversal of rivaroxaban and
fibrinolysis in renal graft fibrosis. Transplantation 2001; 72:1437–1443.
dabigatran by prothrombin complex concentrate. Circulation 2011;
17 Ramachandran R, Hollenberg MD. Proteinases and signaling:
124:1573–1579.
pathophysiological and therapeutic implications via PARs and more. Br J
41 Crowther M, Gold A, Levy GG, Lu G, Leeds J, Wiens BL, et al. ANNEXA-A
Pharmacol 2008; 153:S263–S282. part 2: a phase 3 randomized, double-blind, placebo-controlled trial
18 Favreau F, Thuillier R, Cau J, Milin S, Manguy E, Mauco G, et al. Anti-thrombin demonstrating sustained reversal of apixaban-induced anticoagulation in
therapy during warm ischemia and cold preservation prevents chronic kidney older subjects by andexanet alfa, a universal antidote for factor Xa inhibitors.
graft fibrosis in a DCD model. Am J Transplant 2010; 10:30–39. J Thromb Hemost 2015; 13:84–85.
19 Bogatkevich GS, Ludwicka-Bradley A, Nietert PJ, Akter T, Van Ryn J, 42 Siegal DM, Curnette JT, Connolly SJ, Lu G, Conley P, Wiens BL, et al.
Silver RM. Anti-inflammatory and antifibrotic effects of the oral direct Andexanet alfa for the reversal of factor Xa inhibitor activity. N Engl J Med
thrombin inhibitor dabigatran etexilate in a murine model of interstitial lung 2015; 373:2413–2424.
disease. Arthritis Rheum 2011; 63:1416–1425. 43 Bakhru S, Laulicht B, Jiang X, Chen L, Grosso M, Morishima Y, et al. Reversal
20 EPSIM RESEARCH GROUP. A controlled comparison of aspirin and oral of anticoagulant-induced bleeding in external and internal bleeding models
antocoagulants in prevention of death after myocardial infarction. N Engl J by PER977, a small molecule anticoagulant antidote. Circulation 2014; 130:
Med 1982; 307:701–708. A19361.
21 Rothberg MB, Celestin C, Fiore LD, Lawler E, Cook JR. Warfarin plus aspirin 44 Ansell JE, Bakhru S, Laulicht BE, Steiner SS, Grosso M, Brown K, et al. Use
after myocardial infarction or the acute coronary syndrome: meta-analysis of PER977 to reverse the anticoagulant effect of edoxaban. N Engl J Med
with estimates of risk and benefit. Ann Intern Med 2005; 143:241–250. 2014; 339:2141–2142.
22 Van Es RF, Jonker JJ, Verheugt FW, Deckers JW, Grobbee DE. Aspirin and 45 Stangier J, Rathgen K, Stähle H, Gansser D, Roth W. The pharmacokinetics,
coumadin after acute coronary syndromes (the ASPECT-2 study): a pharmacodynamics, and tolerability of dabigatran etexilate, a new oral direct
randomized controlled trial. Lancet 2002; 360:109–113. thrombin inhibitor, in healthy male subjects. Br J Clin Pharmacol 2007;
23 Hurlen M, Abdelnoor M, Smith P, Erikssen J, Arnesen H. Warfarin, aspirin, or 64:292–303.
both after myocardial infarction. N Engl J Med 2002; 347:969–974. 46 Samoš M, Stančiaková L, Ivanková J, Staško J, Kovář F, Dobrotová M, et al.
24 Faxton DP, Eikelboom JW, Berger PB, Holmes DR, Bhatt DL, Moliterno DJ, Monitoring of dabigatran therapy using hemoclot thrombin inhibitor assay in
et al. Antithrombotic therapy in patients with atrial fibrillation undergoing patients with atrial fibrillation. J Thromb Thrombolysis 2015; 39:95–100.
coronary stenting: a North-American perspective. Circ Cardiovasc Interv 47 Schiele F, van Ryn J, Canada K, Newsome C, Sepulveda E, Park J, et al. A
2011; 4:522–534. specific antidote for dabigatran: functional and structural characterization.
25 Sørensen R, Hansen ML, Abildstrom SZ, Hvelplund A, Andersson C, Blood 2013; 121:3554–3562.
Jørgensen C, et al. Risk of bleeding in patients with acute myocardial 48 Pollack CV, Reilly PA, Eikelboom J, Glund S, Verhamme P, Bernstein RA, et al.
infarction treated with different combinations of aspirin, clopidogrel, and Idarucizumab for dabigatran reversal. N Engl J Med 2015; 373:511–520.
vitamin K antagonists in Denmark: a retrospective analysis of nationwide 49 Glund S, Stangier J, Schmohl M, Gansser D, Norris S, van Ryn J, et al.
registry data. Lancet 2009; 374:1967–1974. Safety, tolerability, and efficacy of idarucizumab for the reversal of the
26 Dewilde WJ, Oirbans T, Verheught FW, Kelder JC, de Smet BJ, Herrman JP, anticoagulant effect of dabigatran in healthy male volunteers: a randomized,
et al. Use of clopidogrel with or without aspirin in patients taking oral placebo-controlled, double-blind phase 1 trial. Lancet 2015; 386:680–690.

Copyright r 2016 Wolters Kluwer Health, Inc. All rights reserved.

Вам также может понравиться