Sei sulla pagina 1di 10

REVIEWS

Oral factor Xa inhibitors for the long-term


management of ACS
James W. Wisler and Richard C. Becker
Abstract | Despite considerable reductions in cardiovascular events in patients with an acute coronary
syndrome (ACS) receiving dual antiplatelet therapy (DAPT), substantial residual risk persists. This unmet
need has stimulated the development of anticoagulant drugs that target specific coagulation factors involved
in the pathogenesis of thrombosis after atheromatous plaque disruption. Factor Xa is an attractive target
for inhibition because of both its integral role in coagulation and its recognized participation in cellular
proliferation and inflammation. Several oral, direct factor Xa inhibitors are undergoing investigation and large,
phase III clinical trials of two agents, apixaban and rivaroxaban, in patients with an ACS have been completed.
On the basis of the known pathobiology of ACS, one might anticipate that drugs in this class of anticoagulant
would beneficially reduce ischemic and thrombotic events; however, a strategy of combined anticoagulant
therapy and DAPT is likely to increase concomitant bleeding complications. The balance of benefit and risk will
ultimately determine uptake in clinical practice. We review the available data on factor Xa inhibitors in the
long-term management of patients with an ACS.
Wisler, J. W. & Becker, R. C. Nat. Rev. Cardiol. 9, 392–401 (2012); published online 21 February 2012; doi:10.1038/nrcardio.2012.18

Introduction
The development and clinical investigation of platelet Novel anticoagulants that target specific coagula-
antagonists and their combined administration, referred tion factors, such as thrombin and factor Xa, have been
to as dual antiplatelet therapy (DAPT), has successfully developed in an attempt to overcome the limitations of
reduced ischemic and thrombotic events in patients oral vitamin K antagonists. Factor Xa has emerged as an
with an ST-segment or non-ST-segment elevation acute attractive target for inhibition because of its prominent
coronary syndrome (ACS). Despite the benefit of DAPT, role in the initiation, amplification, and propagation
substantial residual risk of ischemic complications per- phases of coagulation,10 as well as its participation in cel-
sists over time.1–3 One potential strategy to reduce this lular proliferation,11 apoptosis,12 matrix-metalloprotein
risk is the addition of an anticoagulant to aspirin mono- integrity,13 and inflammation.14
therapy or DAPT after initial hospital-based manage- Preclinical studies of several oral, direct factor Xa
ment. Randomized clinical trials, have shown that the inhibitors have identified favorable characteristics of
addition of a vitamin K antagonist, such as warfarin, to these agents, which include a predictable dose–response
aspirin monotherapy reduces recurrent ischemic and effect, rapid onset and relatively rapid offset of action, low
thrombotic events.4–6 To date, no clear benefit has been potential for interactions with other drugs and with food,
identified from the addition of an oral anticoagulant to and a low risk of off-target effects.15 In addition, several of
DAPT, and increased bleeding seems likely with multi- these agents have demonstrated clinical efficacy to reduce
target antithrombotic strategies—both warfarin added the incidence of various thrombotic disorders such as
to aspirin monotherapy,4,6 and warfarin combined with venous thromboembolism (VTE) in patients undergoing
DAPT (aspirin and clopidogrel).7,8 Furthermore, the hip-replacement 16,17 or knee-replacement surgery,18,19 and
use of vitamin K antagonists in patients with an ACS is stroke or systemic embolism in patients with nonvalvu-
compli­cated by various inherent and well-recognized lar atrial fibrillation20–22 (the latter is separately reviewed
limita­tions of this class of drugs—their narrow thera- in this focus issue on anticoagulation therapies23). These
Division of Cardiology peutic index, variable dose–response relationships, inter­ results, together with the recognized role of factor Xa
(J. W. Wisler), Duke
Clinical Research actions with other drugs and food, and high propensity in the pathobiology of ACS, have sparked consider-
Institute (R. C. Becker), for bleeding complications.9 able interest in the potential use of factor Xa inhibitors
Duke University
Medical Center,
as an adjunct to monotherapy or DAPT for secondary
Box 3850, 2400 Pratt Competing interests prevention after an ACS.
Street, Durham, R. C. Becker declares associations with the following
NC 27705, USA.
companies: AstraZeneca, Bayer Pharmaceuticals, Bristol–Myers Cell-based model of coagulation
Squibb, Daiichi Sankyo, Eli Lilly, Johnson & Johnson, The
Correspondence to: According to the cell-based model, coagulation occurs
R. C. Becker Medicines Company, Merck, Momenta, Regado Biosciences,
richard.becker@ and Schering–Plough. See the article online for full details of the in three overlapping phases.10 The first phase, ‘initia-
duke.edu relationships. J. W. Wisler declares no competing interests. tion’ (Figure 1a), begins with vascular injury, such as

392  |  JULY 2012  |  VOLUME 9  www.nature.com/nrcardio


© 2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON ANTICOAGULATION THERAPIES

disruption of an atherosclerotic plaque, which exposes Key points


the subendothelial proteins collagen, tissue factor (Tf),
■■ Factor Xa is an attractive target for inhibition in patients with an acute coronary
and collagen-bound von Willebrand factor (vWf ) to
syndrome (ACS) because of its roles in coagulation, cellular proliferation,
circulating blood and Tf-bearing extravascular cells.10,24 apoptosis, matrix-metalloprotein stability, and inflammation
Exposed collagen subsequently triggers adhesion and ■■ Four oral, competitive factor Xa inhibitors (apixaban, darexaban, letaxaban,
activation of platelets, whereas exposed Tf forms a and rivaroxaban) have been investigated in patients with an ACS; only apixaban
complex with factor VIIa.25 The Tf–factor VIIa complex and rivaroxaban have progressed to phase III clinical trials
activates both factor IX and factor X. If it leaves the cell ■■ In the APPRAISE‑2 trial, addition of apixaban to standard therapy in patients
surface, factor Xa is rapidly inhibited by Tf pathway with an ACS showed no clinical benefit, and increased the risk of major and
inhibitor. However, any factor Xa molecules that remain intracranial bleeding
■■ In ATLAS ACS 2–TIMI 51, addition of rivaroxaban to dual antiplatelet therapy
on the cell surface can combine with factor Va to produce
(aspirin plus clopidogrel) reduced cardiovascular events and mortality, which
a small, but sufficient, amount of thrombin for platelet outweighed the increased bleeding risk
activation.26 The process of platelet binding to exposed
extracellular-matrix proteins at the site of vascular injury
activates platelets and brings them into proximity with a factor VIIIa and factor IXa, is formed when factor IX
site of Tf exposure.10 reaches the cell surface. The tenase complex activates
In the second phase of coagulation, termed ‘ampli- factor X on the platelet surface. Factor Xa then binds
fication’ or ‘priming’ (Figure 1b), the initial proco- to factor Va, and forms the prothrombinase complex.
agulant signal is amplified by the small amount of This complex subsequently initiates the conversion of pro-
thrombin generated on Tf-bearing cells via enhanced thrombin to thrombin, and produces the burst of throm-
platelet adhesion,27 full activation of platelets, and acti- bin necessary for fibrin-clot formation. Factor Xa thus
vation of factors V, VII, and XI. 26 During activation, serves as a participant and key mediator in each stage of
platelets release factor V from alpha granules onto their coagulation, and ultimately leads to thrombin generation.
surfaces in a partially active form.28 Factor V is then acti-
vated by either thrombin or factor Xa.29 Thrombin also Targeting factor Xa in ACS
activates factor VIII and releases it from vWf by cleaving In addition to having a vital role in each phase of coagu-
platelet-bound vWf–factor VIII.30 Factor VIIIa remains lation, factor Xa also exhibits a variety of cellular effects.
bound to the cell surface. Once platelets have been acti- Binding of factor Xa to vascular endothelial cells is
vated and factors Va and VIIIa have bound to the platelet associated with several events, including Tf expression,
surface, assembly of coagulation proteins, formation of release of plasminogen activator inhibitor 1, expression of
enzymatically active protein complexes, and thrombin E‑selectin, intercellular adhesion molecule 1, and vascular
generation begins. cell adhesion protein 1, and release of several inflamma-
The final phase of coagulation, ‘propagation’ tory cytokines, such as IL‑6 and IL‑8.11,31 Furthermore,
(Figure 1c), begins with the assembly of the proco- macrophages localized within atheromatous plaques can
agulant ‘tenase’ and ‘prothrombinase’ complexes on the synthesize factor Xa,32 which in turn stimulates leukocyte
cell surface.10 The tenase complex, which consists of chemotaxis and proliferation.33

a b c
Endothelial cell
Fibrin
Tf
Xa Va Xa Va
VIIa
Prothrombin
IX X Fibrinogen

X Thrombin Thrombin
IXa
VIII XIa Xa Va
V
VIIIa
Activated
VIIIa IXa platelet Prothrombin
Va IXa
Platelet Activated
platelet IX

Figure 1 | Cell-based model of coagulation, in which coagulation occurs in three overlapping phases. a | ‘Initiation’ begins
with endothelial injury. Exposed collagen and subendothelial proteins trigger adhesion and activation of platelets. Exposed Tf
forms a complex with factor VIIa. Tf–factor VIIa activates factor IX and factor X. Factor V is slowly activated by factor Xa.
Factor Xa then binds to factor Va and forms the prothrombinase complex, which generates small amounts of thrombin.
b | In the second phase, ‘amplification’ or ‘priming’, the initial procoagulant signal is amplified by small amounts of thrombin
generated on Tf-bearing cells. During activation, platelets release factor V, which is activated by either thrombin or factor Xa.
Thrombin also activates factor VIII. Upon platelet activation, factors Va and VIIIa bind to the platelet surface. c | The final
phase of coagulation, ‘propagation’, begins with assembly of procoagulant complexes on the cell surface. The tenase
complex (factor VIIIa and factor IXa) activates factor X on the platelet surface. Factor Xa then rapidly binds to factor Va
(generated by thrombin in the amplification phase), forming the prothrombinase complex, which initiates conversion of
prothrombin to thrombin, producing the thrombin burst necessary for fibrin-clot formation. Abbreviation: Tf, tissue factor.

NATURE REVIEWS | CARDIOLOGY VOLUME 9  |  JULY 2012  |  393


© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Pharmacokinetic and pharmacodynamic profiles of oral factor Xa inhibitors under development
Drug Dosing Time to Ki (nmol/l) Half-life (h) CYP Clearance Possible drug–drug Antidote
Cmax (h) metabolism interactions
Apixaban15,53,78 Twice daily 1–3  0.08 Once daily: 14  CYP3A4, 25% fecal Potent CYP3A4 Possibly recombinant
Twice daily: 9  CYP3A5 25% renal inhibitors* factor Xa derivative
(PRT 064445)
Betrixaban15,78,79 Once daily NR 0.12 19–20  None Almost entirely Low potential Possibly recombinant
unchanged in bile reported factor Xa derivative
<5% renal (PRT 064445)
Darexaban15,46,80,81 Once daily 1–1.5  0.03 14–18  NR Equally via fecal Minimal food NR
and renal routes interactions and no
reported drug–drug
interaction
Edoxaban15 Once daily 1–2  0.56 9–11  NR 65% fecal Potent CYP3A4 NR
35% renal inhibitors
P‑glycoprotein
inhibitors‡
Eribixaban48 Once daily NR 0.32 NR NR NR NR NR
Letaxaban Once daily 1–2  1.8 9–13  NR 30% renal NR NR
(TAK‑442)85,86
LY515711749,82 Once daily 0.5–4  4.6–6.6 25  NR Gastrointestinal NR NR
tract
Rivaroxaban83,84 Twice daily 2–4  0.4 Healthy: 5–9  CYP3A4, 66% renal Potent CYP3A4 Possibly recombinant
Elderly: 9–13  CYP2J2 34% fecal inhibitors factor Xa derivative
P‑glycoprotein (PRT 064445)
inhibitors‡
*Potent CYP3A4 inhibitors include ketoconazole, macrolides, verapamil, and protease inhibitors. 15 ‡Inhibitors of CYP3A4 and P‑glycoprotein include ketoconazole, itraconazole, voriconazole,
posaconazole, and ritonavir.15 Abbreviations: Cmax, peak plasma concentration; CYP, cytochrome P450; Ki, inhibitor dissociation constant; NR, not reported.

Factor Xa might be a more-attractive target for inhi- Clinical trials of factor Xa inhibitors in ACS
bition than thrombin for several reasons. Coagulation Several oral, direct factor Xa inhibitors (apixaban, 44
occurs in an amplified manner,34 with factor Xa func- betrixaban, 45 darexaban, 46 edoxaban, 47 eribixaban, 48
tioning at the confluence of several early steps (the initi­ letaxaban,49 LY5157117,50 and rivaroxaban51) have entered
ation and amplifi­cation phases) of coagulation on both clinical development for various indications, including
Tf-bearing cells and platelets.10 In addition, factor Xa is prevention of VTE after orthopedic surgery, treatment of
more thrombo­genic than thrombin on a molar basis.35 established VTE, stroke prevention in patients with non-
Consequently inhibition of factor Xa might be a more- valvular atrial fibrillation, and secondary prevention after
efficient means to reduce fibrin formation than inhibi­ an ACS. The pharmacokinetic and pharma­codynamic
tion of thrombin. By contrast with direct thrombin characteristics of these drugs are summarized in Table 1.
inhibitors, factor Xa inhibitors decrease endogenous Of these agents, apixaban, darexaban, letaxaban, and riva-
thrombin potential 36–38 and prolong the lag phase roxaban have been evaluated in phase II clinical trials in
of thrombin generation in a dose-dependent fashion.37 patients with an ACS, but only apixaban and riva­roxaban
Direct inhibition of factor Xa has also been shown to have progressed to phase III trials in these patients
allow small amounts of thrombin generation and acti- (Table 2). The other agents have not been assessed in
vation of vitamin K-dependent protein C, an important patients with an ACS.
contribution to vascular thromboresistance.38,39 This dif-
ferential effect might be at least partially responsible for Apixaban
the rebound thrombophilia observed with some direct Apixaban is an oral, highly selective, direct-acting, revers-
thrombin inhibitors.40,41 ible inhibitor of factor Xa.52 The drug is rapidly absorbed
For these reasons, targeting factor Xa in patients with after oral administration, with a peak plasma concentra-
an ACS offers the potential to attenuate or modulate the tion achieved after approximately 1 h.53 The half-life of
pluripotent effects of factor Xa. An illustration of benefit apixaban is 8–14 h,53 with approximately 25% of the drug
from factor Xa inhibition, albeit in the acute management excreted in urine, 25% excreted in feces, and the remain-
of ACS, is found in the Organization to Assess Strategies ing portion metabolized by liver enzymes, including
in Ischemic Syndromes trials, OASIS‑542 and OASIS‑6.43 cytochrome P450 3A4 and cytochrome P450 3A5.53–55
The indirect, parenteral factor Xa inhibitor fondaparinux The phase  II Apixaban for Prevention of Acute
demonstrated a significant reduction in bleeding compli- Ischemic and Safety Events (APPRAISE) trial44 was
cations, but no improvement in efficacy, compared with an international, multicenter, double-blind, random-
enoxaparin in OASIS‑5,42 and superior efficacy with a con- ized, placebo-controlled, dose-finding study of apixa-
comitant reduction in bleeding complications compared ban in patients with an ACS. Patients were eligible for
with unfractionated heparin in OASIS‑6.43 enrollment if they presented with an ACS (myocardial

394  |  JULY 2012  |  VOLUME 9  www.nature.com/nrcardio


© 2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON ANTICOAGULATION THERAPIES

Table 2 | Features of the phase III trials of oral factor Xa inhibitors in patients with an ACS
Characteristics ATLAS ACS 2–TIMI 51 trial62,87 APPRAISE‑2 trial57
Inclusion criteria Patients aged ≥18 years with symptoms of ACS ACS in past 7 days
(ST-segment MI, non-ST-segment MI, or unstable Clinically stable
angina) in past 7 days Receiving standard medical therapy with either aspirin
Receiving standard medical therapy with either or aspirin plus any thienopyridine
aspirin or aspirin plus any thienopyridine Two or more of the following high-risk characteristics:
Patients aged 18–54 years also had to have age ≥65 years, diabetes mellitus, MI in past 5 years,
diabetes mellitus or previous MI cerebrovascular disease, peripheral vascular disease,
clinical heart failure or ejection fraction <40% in
association with index event, creatinine clearance
<60 ml/min, no revascularization after index event
Key exclusion criteria Active bleeding Active bleeding
Platelet count <90,000/mm3 Platelet count <100,000/mm3
Hemoglobin <10 g/dl Hemoglobin <9 mg/dl
Creatinine clearance <30 ml/min Creatinine clearance <20 ml/min
History of intracranial bleeding History of intracranial bleeding
Clinically significant gastrointestinal bleeding in past Persistent, severe hypertension
12 months
Ischemic stroke or TIA in patients who were taking
both aspirin and a thienopyridine
Key patient Mean age: 61 years Median age: 69 years
characteristics Aged ≥65 years: 36.5% Aged ≥65 years: 58.9%
Previous MI: 26.9% Previous MI (in past 5 years): 26.2%
Diabetes mellitus: 32.0% Diabetes mellitus: 47.8%
History of stroke or TIA: 2.7% History of stroke or TIA: 10.0%
Concomitant aspirin only: 6.8% Concomitant aspirin only: 16%
Concomitant aspirin plus a thienopyridine: 93.2% Concomitant aspirin plus a thienopyridine: 81%
Treatment duration Rivaroxaban, mean 13.1 months Apixaban, mean 175 days
Abbreviations: ACS, acute coronary syndrome; APPRAISE‑2, Apixaban for Prevention of Acute Ischemic and Safety Events 2; ATLAS ACS 2–TIMI 51, Anti‑Xa
Therapy to Lower Cardiovascular Events in Addition to Standard Therapy in Subjects with Acute Coronary Syndrome 2—Thrombolysis in Myocardial Infarction 51;
MI, myocardial infarction; TIA, transient ischemic attack.

infarction [MI] with or without ST-segment elevation, or 2.5 mg twice daily (5.7% versus 3.0%; HR 1.78, 95% CI
unstable angina) in the past 7 days, were clinically stable, 0.91–3.48, P = 0.09) and 10 mg once daily (7.9% versus
were receiving standard treatment for ACS (including 3.0%; HR 2.45, 95% CI 1.31–4.61, P = 0.05). Substantially
aspirin alone or DAPT with aspirin and a thieno­pyridine), higher rates of ISTH major bleeding and total bleeding
and had at least one additional risk factor for recurrent were also observed in patients taking apixaban 10 mg
ischemic events (such as diabetes mellitus, cerebral or twice daily and 20 mg once daily compared with patients
peripheral vascular disease, congestive heart failure, taking either of the lower doses. The rates of ISTH major
or a left ventricular ejection fraction <40%). The study or CRNM bleeding were also more pronounced in
was conducted in two phases. In the first phase, patients patients on DAPT plus apixaban compared with those
were assigned in a 1:1:1 fashion to placebo, apixaban on DAPT who received a placebo.
2.5 mg twice daily, or apixaban 10 mg once daily. After The incidence of cardiovascular death, MI, severe
approximately 25% of the patients had received the study recurrent ischemia, or ischemic stroke was numerically,
drug for 30 days, an independent data and safety moni- but not significantly, decreased in patients who received
toring committee recommended the inclusion of two apixaban 2.5 mg daily (7.6% versus 8.7%; HR 0.73, 95% CI
planned higher doses, 10 mg twice daily and 20 mg once 0.44–1.19, P = 0.21) or 10 mg daily (6.0% versus 8.7%;
daily, in an attempt to gain more information on safety at HR 0.61, 95% CI 0.35–1.04, P = 0.07) compared with
those doses. In the second phase, patients were assigned placebo. The dose-related reduction in ischemic events
in a 3:1:1:2:2 ratio to placebo, and apixaban 2.5 mg twice in patients taking apixaban versus placebo was less pro-
daily, 10 mg once daily, 10 mg twice daily, and 20 mg nounced in those patients receiving concomitant DAPT.
once daily, respectively. The investigators concluded that, when comparing ISTH
The primary study outcome was occurrence of major major bleeding with the reduction in clinically important
bleeding and clinically relevant nonmajor (CRNM) bleed- ischemic events, apixaban 2.5 mg twice daily and apixa-
ing as defined by the International Society on Thrombosis ban 10 mg once daily resulted in absolute net reductions
and Haemostasis (ISTH).56 A secondary end point was the in clinical events in the overall study population of 0.3%
composite of cardiovascular death, MI, severe recurrent and 1.6%, respectively.
ischemia, or ischemic stroke. In total, 99.7% of patients On the basis of the results of the APPRAISE trial,44
enrolled were treated with aspirin and 75.6% were receiv- apixaban was investigated in the phase III APPRAISE‑2
ing DAPT with aspirin and clopidogrel. Compared with study.57 APPRAISE‑2 was a multicenter, prospective,
placebo, apixaban showed a significant, dose-dependent randomized, double-blind, placebo-controlled trial in
increase in ISTH major and CRNM bleeding at both which the efficacy and safety of apixaban in patients with

NATURE REVIEWS | CARDIOLOGY VOLUME 9  |  JULY 2012  |  395


© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

an ACS were investigated. The key inclusion and exclu- physician. Patients were then randomly allocated to
sion criteria are listed (Table 2). Patients were randomly receive either rivaroxaban or a matching placebo for
assigned in a 1:1 ratio to apixaban 5 mg twice daily or 6 months. Escalating dose regimens from 5 mg to 20 mg
a matched placebo. Those with an estimated creati- per day were administered once or twice daily. The
nine clearance <40 ml/min were randomly assigned to primary efficacy outcome was the composite of death,
apixaban 2.5 mg twice daily or a matched placebo. MI, stroke, or severe recurrent ischemia that required
The primary efficacy outcome was the composite of revasculari­zation. The primary safety end point was clini-
cardiovascular death, MI, or ischemic stroke, and the cally significant bleeding (TIMI major, TIMI minor, or
primary safety outcome was major bleeding according requiring medical attention).
to the Thrombolysis in Myocardial Infarction (TIMI) Overall, treatment with rivaroxaban was associated
definition.58 Of 7,392 patients, 97% were taking aspirin with a nonsignificant reduction in the primary compos-
and 81% were receiving DAPT at the time of random- ite efficacy outcome compared with placebo (5.6% versus
ization. The trial was discontinued prematurely by the 7.0%; HR 0.79, 95% CI 0.60–1.05, P = 0.10). A significant
data and safety monitoring committee because of an reduction was observed for the main secondary efficacy
increase in major bleeding events with apixaban in the outcome, a composite of death, MI, or stroke, compared
absence of a significant reduction in recurrent ischemic with placebo (3.9% versus 5.5%; HR 0.69, 95% CI 0.50–
events. The primary efficacy outcome was observed in 0.96, P = 0.027). A dose-dependent increase in the primary
7.5% and 7.9% of patients receiving apixaban or placebo, safety end point was observed with rivaroxaban compared
respectively (HR 0.95, 95% CI 0.80–1.11, P = 0.51). The with placebo (HR 2.21, 95% CI 1.25–3.91; HR 3.35, 95% 
primary safety outcome occurred in 1.3% of patients CI 2.31–4.87; HR 3.60, 95% CI 2.32–5.58; HR 5.06, 95% CI
who received at least one dose of apixaban, and 0.5% 3.45–7.42, for rivaroxaban 5 mg, 10 mg, 15 mg, and 20 mg
of patients who received at least one dose of placebo given once daily or the same total dose given twice daily,
(HR 2.59, 95% CI 1.50–4.46, P = 0.001). A greater respectively; P <0.0001).
number of intracranial (0.3% versus 0.1%; HR 4.06, Because of the favorable efficacy rates observed with
95% CI 1.15–14.38, P = 0.03) and fatal (0.1% versus 0.0%) rivaroxaban and the dose-dependent incidence in bleed-
bleeding events occurred with apixaban than placebo. ing, doses of 2.5 mg twice daily and 5 mg twice daily
These findings were consistent across all subgroups, were chosen for further assessment in the phase III
including those defined by antiplatelet strategy (aspirin ATLAS ACS 2–TIMI 51 trial (Table 2).62 Researchers
monotherapy versus DAPT) and by ACS-management investigated the use of rivaroxaban in addition to stan-
strategy (revascularization versus noninvasive manage­ dard antiplatelet therapy (either aspirin monotherapy or
ment). The investigators concluded that the addition of DAPT with aspirin and either clopidogrel or ticlopidine)
apixaban at a dose of 5 mg daily to antiplatelet therapy for the prevention of long-term adverse cardiovascular
after an ACS event increased the number of bleed- events after an ACS. In total, 15,526 patients with an ACS
ing events without a significant reduction in recurrent <7 days before enrollment were stratified according to
ischemic events. background antiplatelet therapy (aspirin or DAPT) and
randomly allocated within those strata in a 1:1:1 ratio to
Rivaroxaban receive rivaroxaban 2.5 mg twice daily, rivaroxaban 5 mg
Rivaroxaban is an oral, reversible, direct-acting factor Xa twice daily, or a matched placebo. The primary efficacy
inhibitor that is rapidly absorbed and reaches a maximum end point was a composite of cardiovascular death, MI,
plasma concentration in 2–4 h.59 In healthy and elderly or stroke. The primary safety end point was TIMI major
patients, the half-life is approximately 5–13 h.15 Rivaroxaban bleeding not associated with CABG surgery.
has a dual mode of elimination. Approximately two-thirds Rivaroxaban significantly reduced the rate of the
is degraded metabolically in the liver by pathways cata- primary efficacy end point compared with placebo (8.9%
lyzed by cytochrome P450 enzymes (predominately cyto- versus 10.7%; HR 0.84, 95% CI 0.74–0.96, P = 0.008;
chrome P450 3A4 and cytochrome P450 2J2), with half Figure 2). The 2.5 mg twice-daily dose also resulted in
of the degradation products eliminated renally and half via a significant reduction in death from cardiovascular
the fecal route.59–61 The other one-third is excreted renally causes (2.7% versus 4.1%; HR 0.66, 95% CI 0.51–0.86,
as the unchanged, active substance. P = 0.002) and death from any cause (2.9% versus 4.5%;
The phase II Anti‑Xa Therapy to Lower Cardiovascular HR 0.68, 95% CI 0.53–0.87, P = 0.002) compared with
Events in Addition to Standard Therapy in Subjects with placebo. No survival benefit was observed with the 5 mg
Acute Coronary Syndrome—Thrombolysis in Myocardial twice-daily dose compared with placebo. Rivaroxaban
Infarction 46 (ATLAS ACS–TIMI 46) study 51 was a multi- was associated with an increase in the primary safety
center, prospective, randomized, double-blind, placebo- end point (2.1% versus 0.6%; HR 3.96, 95% CI 2.46–6.38,
controlled, dose-escalation trial in which the efficacy P <0.001) and intracranial hemorrhage (0.6% versus
and safety of rivaroxaban were evaluated in approxi- 0.2%; HR 3.28, 95% CI 1.28–8.42, P = 0.009), without an
mately 3,500 patients with an ACS (ST-segment or non- increase in fatal bleeding (0.3% versus 0.2%; HR 1.19,
ST-­segment elevation MI, or unstable angina) event in 95% CI 0.54–2.59, P = 0.66) compared with placebo.
the past 7 days. In this study, all patients received stan- The 2.5 mg twice-daily dose was associated with fewer
dard antiplatelet therapy of either aspirin monotherapy episodes of TIMI bleeding requiring medical atten-
or aspirin and a thienopyridine at the discretion of the tion (12.9% versus 16.2%; P <0.001) and fatal bleeding

396  |  JULY 2012  |  VOLUME 9  www.nature.com/nrcardio


© 2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON ANTICOAGULATION THERAPIES

(0.1% versus 0.4%; P = 0.04) than rivaroxaban 5 mg 12


HR 0.84, 95% Cl 0.74–0.96, P = 0.008
twice daily. The investigators concluded that in patients

Death from cardiovascular causes,


myocardial infarction, or stroke (%)
Placebo
with a recent ACS event, treatment with rivaroxaban 10
reduced the risk of the composite end point of cardio­
8
vascular death, MI, or stroke, but resulted in a signifi-
cant increase in the risk of intracranial hemorrhage
6
without increasing fatal bleeding. Rivaroxaban

4
Darexaban
Darexaban (formerly YM‑150) is an oral, direct factor Xa 2
inhibitor with a rapid onset of action and a plasma half-
life of approximately 14–20 h.46,63 Upon absorption, the 0
drug is extensively metabolized to its main active metabo­ 0 90 180 270 360 450 540 630 720
lite, YM‑222714, and reaches peak plasma concentrations Time (days)
within 2 h of an oral dose.46 The Study Evaluating Safety, Number at risk
Tolerability, and Efficacy of YM‑150 in Subjects with Acute Rivaroxaban 10,229 8,817 7,797 6,324 5,137 3,967 2,830 1,747 831
Coronary Syndromes (RUBY‑1)46 was a phase II, dose- Placebo 5,113 4,437 3,974 3,253 2,664 2,059 1,460 878 421
finding, randomized, double-blind, placebo-­controlled
Figure 2 | Reduction in adverse cardiovascular events associated with rivaroxaban
trial of the safety and tolerability of darexaban in patients treatment. When added to standard (aspirin or dual) antiplatelet therapy after an
with an ACS. In this study, 1,279 patients with a high- acute coronary syndrome, rivaroxaban significantly reduced the risk of the primary
risk non-ST-segment or ST-segment elevation ACS event efficacy end point (a composite of death from cardiovascular causes, myocardial
within 7 days of presentation were randomly allocated infarction, or stroke) in the ATLAS ACS 2–TIMI 51 trial. Reproduced with permission
in a 1:1:1:1:1:1:2 ratio to one of six darexaban regimens from Mega, J. L. et al. Rivaroxaban in patients with a recent acute coronary
(5 mg twice daily, 10 mg once daily, 15 mg twice daily, syndrome. N. Engl. J. Med. 366 (1), 9–19 © 2012, with permission from the
30 mg once daily, 30 mg twice daily, or 60 mg once Massachusetts Medical Society.
daily) or a matched placebo, respectively, in addition to
DAPT with aspirin and clopidogrel. The primary outcome patients were randomly allocated to one of three doses of
was the incidence of ISTH-defined major or CRNM letaxaban (40 mg twice daily, 80 mg once daily, or 80 mg
bleeding. The primary efficacy end point was a compos- twice daily) or a matched placebo for up to 24 weeks. In
ite of death, stroke, MI, systemic thromboembolism, or stage 3, patients were randomly allocated to one of two
severe recurrent ischemia. doses of letaxaban (160 mg once daily or 120 mg twice
Overall, bleeding rates were significantly higher in daily) or a matched placebo for up to 24 weeks. At the
all darexaban arms compared with placebo (pooled end of stage 1, an independent data and safety moni-
HR 2.275, 95% CI 1.13–4.60, P = 0.022). No decrease was toring board assessed the incidence of major bleeding
observed in the primary efficacy event rates with darexa- and other safety findings before random allocation into
ban; however, the study was primarily designed to evalu- stage 2 or 3. The primary end point was the incidence of
ate safety, and was underpowered to assess efficacy. The TIMI major bleeding. Secondary end points included a
investigators concluded that darexaban, when added to more-sensitive bleeding scale consisting of a composite
standard DAPT after an ACS, produced a twofold to four- of major and clinically significant nonmajor bleeding
fold increase in bleeding with no other safety concerns, and the incidence of expanded major adverse cardiac
but with no signal for efficacy. Citing the increased bleed- events (defined in this study as cardiovascular death,
ing risk and lack of efficacy observed with darexaban, nonfatal MI, nonfatal stroke, or myocardial ischemia
the manufacturer announced the discontinuation of the that required hospitalization).
development of darexaban for all indications.64 The overall incidence of TIMI-major bleeding
observed with letaxaban was low and was numerically,
Letaxaban but not significantly, higher at larger doses compared
Letaxaban (formerly TAK‑442) is an oral, direct with placebo. A significant, dose-dependent increase
factor Xa inhibitor with a bioavailability of approxi- in the secondary bleeding scale was observed with
mately 50% 65 and a plasma half-life of 9–13 h. 66,67 letaxaban compared with placebo. No positive trend for
Letaxaban was investi­gated in patients with an ACS in a reduction in expanded major adverse cardiac events
the phase II Safety and Efficacy of TAK‑442 in Subjects was observed. Importantly, the investigators reported
with Acute Coronary Syndromes (AXIOM‑ACS) trial.49 an increase in adverse events (although details were
In this prospective, multicenter, randomized, double- not provided) in stages 2 and 3 compared with placebo,
blind, three-stage adaptive trial, 2,753 patients were which resulted in discontinuation of the trial; however,
randomly allocated to receive either placebo or letaxa- no hepatotoxicity was observed with letaxaban.
ban in addition to usual care (including aspirin and a
thienopyridine) after an ACS. In stage 1, patients were Why might factor Xa inhibitors fail in ACS?
randomly allocated to one of three doses of letaxaban The role of factor Xa in coagulation, as described above,
(10 mg twice daily, 20 mg twice daily, or 40 mg once is incontrovertible. Why then might factor Xa inhibition
daily) or a matched placebo for up to 24 weeks. In stage 2, not yield generally favorable results, including efficacy,

NATURE REVIEWS | CARDIOLOGY VOLUME 9  |  JULY 2012  |  397


© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

in patients with an ACS? Several biology-based hypo­ its effects on inflammation, apoptosis, cellular prolif-
theses are worthy of consideration. Arterial thrombosis eration, and matrix-metalloprotein stability might be
is highly platelet-dependent, occurs after the presen- demonstrable at relatively low levels of inhibition, where
tation of Tf predominantly from local, intravascular the overriding negative effects of bleeding are also less
sites,68 and is characterized by flow under high shear likely to be operative.
stress. Tf-induced blood coagulation yields approxi-
mately 2 nmol/l thrombin during the initiation phase, Future role of factor Xa inhibitors in ACS
with the majority (96%) of the thrombin being gener- To date, only two oral factor Xa inhibitors, apixaban44,57
ated long after clotting occurs through bioamplification and rivaroxaban,51,62 have been evaluated in phase III
and resupply of coagulation subtrates.68,69 Platelet activa- clinical trials for secondary prevention in patients with
tion occurs with approximately 0.5 nmol/l thrombin.69 In an ACS (Table 2). In both the ATLAS ACS–TIMI 4651
high-shear-stress environments, aspirin reduces throm- and APPRAISE‑1 studies,44 a trend towards a reduction
bin generation and reactions catalyzed by this enzyme.70 in cardiovascular events was observed. However, in the
Under conditions of flow, the cleavage of prothrombin APPRAISE‑2 trial,57 the addition of apixaban 5 mg twice
yields meizothrombin—a protein intermediate with daily to standard DAPT resulted in an increase in major
anticoagulant properties achieved through activated and fatal bleeding events, without a reduction in recurrent
vitamin K-dependent protein C. Therefore, a significant ischemic events. Whereas in the ATLAS ACS 2–TIMI 51
proportion of thrombin generated at the vessel-wall trial, 62 significant reductions in recurrent ischemic
surface under flow conditions is, in fact, anticoagulant events, cardiovascular death, and all-cause mortality were
in nature. observed with rivaroxaban 2.5 mg twice daily, in addition
The impact of flow on thrombin generation from to increases in both bleeding unrelated to CABG surgery
prothrombinase (factor Xa and factor Va) has been and intracranial bleeding.
summarized previously.71 Prothrombin arrives at pro- The exact reasons for the discrepancies in efficacy and
thrombinase anchored to the vessel wall primarily by safety observed in the APPRAISE‑2 and ATLAS ACS 2–
diffusion from adjacent blood. Under flow conditions, TIMI 51 trials are unclear; however, differences between
vessel-wall thrombin is diluted and neutralized by the the patient populations enrolled in the trials might
arrival of antithrombin. The boundary-layer concen- have contributed. For example, patients with a history
trations of substrates required for thrombin generation of stroke or transient ischemic attack who were treated
under flow conditions can be predicted by computational with aspirin in addition to a thienopyridine were
fluid dynamics.72 The importance of platelets in throm- excluded from the ATLAS ACS 2–TIMI 51 trial, but
bus growth under flow conditions is well illustrated in not the APPRAISE‑2 study. This group of patients in
hypothetical estimates of vessel occlusion in proximity to particular does not seem to benefit from an increased
anchored prothrombinase—in coronary arteries at high intensity of antithrombotic therapy.2,73 Fundamentally,
shear rates, platelet accumulation is required to alter flow one could argue that some very high-risk patients, with
sufficiently for thrombin generation, thrombus growth, multiple comorbid characteristics, might be beyond
and vessel occlusion. a threshold to derive benefit from increased intensity
Conceptually, the role of prothrombinase activity and antithrombotic therapy, and receive only an increased
thrombin generation in the natural history of an ACS risk of bleeding.
would decrease with platelet inhibition, particularly in Another possible explanation for the differing results
the setting of increasingly potent DAPT strategies and between drugs and trials might be related to dose selec-
nonocclusive thrombus, as is commonly encountered tion. In both the Apixaban for Reduction in Stroke and
with unstable angina and non-ST-segment elevation MI. Other Thromboembolic Events in Atrial Fibrillation
By contrast, prothrombinase and thrombin are always (ARISTOTLE) study 21 and the APPRAISE‑2 trial,57 a
vital for hemostasis. This might not be the case with a dose of 5 mg twice daily was selected. In trials of riva-
more-moderate strategy of inhibition with aspirin and roxaban for stroke prevention in patients with atrial
clopidogrel. Indeed, stent thrombosis was reduced with fibrillation or treatment of VTE, most patients received
the addition of rivaroxaban in the ATLAS ACS 2–TIMI 51 20 mg per day.22,74 In the ATLAS ACS 2–TIMI 51 study,62
trial,62 which underscores the dynamic nature of coronary a dose of one-quarter to one-half of the 20 mg dose was
arterial thrombosis in contemporary ACS. tested, and survival benefits were observed with the
Some patients with an ACS might have thrombin- lower dose of rivaroxaban (2.5 mg twice daily). This
dependent (or factor  Xa-dependent), rather than inverse dose–response relationship with cardiovascular
platelet-­dependent, arterial thrombosis. Similarly, in events was also observed in the ATLAS ACS–TIMI 46
speci­fic populations, the response to either aspirin mono- trial44 and RUBY‑1,46 which suggests that low doses of a
therapy or DAPT with clopidogrel can be sub­optimal factor Xa inhibitor might be preferred in this particular
because of highly prevalent loss-of-function genotypes patient population. Whether even lower doses of rivar-
or drug–drug interactions. Further investi­gation employ- oxaban than those tested in the ATLAS ACS 2–TIMI 51
ing novel biomarkers, including the coagu­lation pro- trial would be equally efficacious and cause less bleed-
teome and pathways-based analyses, will be required to ing than higher doses is unclear. In preclinical studies of
test these important hypotheses. The potential impact of rivaroxaban, single doses of 1.25 mg in healthy volun-
attenuating the pluripotent activity of factor Xa, including teers produced no significant inhibition of factor Xa.75

398  |  JULY 2012  |  VOLUME 9  www.nature.com/nrcardio


© 2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON ANTICOAGULATION THERAPIES

These results suggest that 2.5 mg of rivaroxaban might Conclusions


represent the lowest efficacious dose. Factor Xa has an important role in coagulation and exhi­
We are, therefore, left with the question of whether bits several cellular effects thought to contribute to the
the results of the ATLAS ACS 2–TIMI 51 trial represent progression and clinical expression of athero­thrombosis.
a drug-specific effect or are an example of selecting the Targeting factor Xa in patients with an ACS has numer-
appropriate dose for the right patient population—a tenet ous biologically supported benefits over current anti­
of drug development that underscores the importance of coagulation strategies; however, the available data suggest
early phase clinical trials and a critical need for parallel that the risk-to-benefit relationship might be prohibi-
investigations designed to provide signals for both on- tive with some agents, at least at the doses tested and in
target and off-target drug effects. On the basis of a cumu- very high-risk patients, when used in combination with
latively large experience from clinical trials, one might DAPT. The results of the ATLAS ACS 2–TIMI 51 trial62
conclude that the therapeutic window for factor Xa inhi- support the ‘factor Xa hypothesis’ in ACS and underscore
bition in patients with an ACS is narrow, particularly in the delicate balance between risk and benefit with triple
the context of ‘triple therapy’, and in patients at high risk anti­t­hrombotic therapy. This narrow therapeutic window
of bleeding complications, such as the elderly or those might, in turn, attenuate enthusiasm for the develop-
with multiple medical comorbidities.76 The propensity ment of oral, competitive, active-site factor Xa inhibitors
for intracranial hemorrhage in this high-risk patient designed specifically for secondary prevention in patients
population when treated with combined anti­coagulant with an ACS. Existing agents need to be tested in combi-
and antiplatelet therapy or triple antithrombotic therapy nation with novel antiplatelet regimens, such as prasugrel
is striking and requires further investigation.77 This or ticagrelor in addition to aspirin. Rivaroxaban seems to
undertaking must proceed without delay and take full represent a novel addition to DAPT with aspirin and clopi-
advantage of the large repositories of clinical data and dogrel after an ACS event, but its place in clinical prac-
collections of blood samples. tice, if approved for this indication, will depend on very
Importantly, oral factor Xa inhibitors have not been careful consideration of all the available treatment options,
studied in combination with new-generation antiplatelet risk–benefit relationships, patient-specific needs, and cost.
agents, such as prasugrel or ticagrelor. Given the increased
rates of major bleeding observed with prasugrel 2 and Review criteria
major bleeding unrelated to CABG surgery observed with The initial literature search was performed using
ticagrelor,1 one would anticipate the therapeutic window the PubMed database and the following key terms:
to be even narrower for factor Xa inhibition in combi- “factor Xa acute coronary syndrome”, “factor Xa
nation with these potent antiplatelet agents than with inhibitors”, “rivaroxaban”, “ATLAS ACS”, “apixaban”, and
clopidogrel. Whether the addition of a factor Xa inhibi- “APPRAISE apixaban”. All manuscripts identified were
tor, such as rivaroxaban, to ticagrelor could improve upon English-language, full-text articles. The reference lists
of comprehensive Review articles were examined for
the mortality benefits previously observed with ticagrelor
additional references.
and aspirin in the PLATO study 1 is unclear.

1. Wallentin, L. et al. Ticagrelor versus clopidogrel risk of bleeding. J. Inv. Cardiol. 18, 162–164 effector cell protease receptor‑1 induces acute
in patients with acute coronary syndromes. (2006). inflammation in vivo. J. Clin. Invest. 99,
N. Engl. J. Med. 361, 1045–1057 (2009). 8. Rossini, R. et al. Long-term outcomes in patients 2446–2451 (1997).
2. Wiviott, S. D. et al. Prasugrel versus clopidogrel undergoing coronary stenting on dual oral 15. Eriksson, B. I., Quinlan, D. J. & Weitz, J. I.
in patients with acute coronary syndromes. antiplatelet treatment requiring oral anticoagulant Comparative pharmacodynamics and
N. Engl. J. Med. 357, 2001–2015 (2007). therapy. Am. J. Cardiol. 102, 1618–1623 (2008). pharmacokinetics of oral direct thrombin
3. Yusuf, S. et al. Effects of clopidogrel in addition 9. Anand, S. S. & Yusuf, S. Oral anticoagulants and factor Xa inhibitors in development.
to aspirin in patients with acute coronary in patients with coronary artery disease. J. Am. Clin. Pharmacokinet. 48, 1–22 (2009).
syndromes without ST‑segment elevation. Coll. Cardiol. 41 (Suppl. S), 62S–69S (2003). 16. Eriksson, B. I. et al. Rivaroxaban versus
N. Engl. J. Med. 345, 494–502 (2001). 10. Hoffman, M. & Monroe, D. M. 3rd. A cell-based enoxaparin for thromboprophylaxis after hip
4. Rothberg, M. B., Celestin, C., Fiore, L. D., model of hemostasis. Thromb. Haemost. arthroplasty. N. Engl. J. Med. 358, 2765–2775
Lawler, E. & Cook, J. R. Warfarin plus aspirin 85, 958–965 (2001). (2008).
after myocardial infarction or the acute coronary 11. Herbert, J. et al. Effector protease receptor 1 17. Lassen, M. R. et al. The efficacy and safety of
syndrome: meta-analysis with estimates of risk mediates the mitogenic activity of factor Xa for apixaban, an oral, direct factor Xa inhibitor, as
and benefit. Ann. Intern. Med. 143, 241–250 vascular smooth muscle cells in vitro and in vivo. thromboprophylaxis in patients following total
(2005). J. Clin. Invest. 101, 993–1000 (1998). knee replacement. J. Thromb. Haemost. 5,
5. Bertrand, M. E. et al. Randomized multicenter 12. Versteeg, H. H., Spek, C. A., Richel, D. J. & 2368–2375 (2007).
comparison of conventional anticoagulation Peppelenbosch, M. P. Coagulation factors VIIa 18. Lassen, M. R. et al. Rivaroxaban versus
versus antiplatelet therapy in unplanned and Xa inhibit apoptosis and anoikis. Oncogene enoxaparin for thromboprophylaxis after total
and elective coronary stenting. The full 23, 410–417 (2004). knee arthroplasty. N. Engl. J. Med. 358,
anticoagulation versus aspirin and ticlopidine 13. Rauch, B. H., Bretschneider, E., Braun, M. 2776–2786 (2008).
(FANTASTIC) study. Circulation 98, 1597–1603 & Schror, K. Factor Xa releases matrix 19. Lassen, M. R. et al. Apixaban versus enoxaparin
(1998). metalloproteinase‑2 (MMP‑2) from human for thromboprophylaxis after hip replacement.
6. Hurlen, M., Abdelnoor, M., Smith, P., Erikssen, J. vascular smooth muscle cells and stimulates the N. Engl. J. Med. 363, 2487–2498 (2010).
& Arnesen, H. Warfarin, aspirin, or both after conversion of pro‑MMP‑2 to MMP‑2: role of MMP‑2 20. Connolly, S. J. et al. Apixaban in patients with
myocardial infarction. N. Engl. J. Med. 347, in factor Xa-induced DNA synthesis and matrix atrial fibrillation. N. Engl. J. Med. 364, 806–817
969–974 (2002). invasion. Circ. Res. 90, 1122–1127 (2002). (2011).
7. Khurram, Z. et al. Combination therapy with 14. Cirino, G. et al. Factor Xa as an interface 21. Granger, C. B. et al. Apixaban versus warfarin
aspirin, clopidogrel and warfarin following between coagulation and inflammation. in patients with atrial fibrillation. N. Engl. J. Med.
coronary stenting is associated with a significant Molecular mimicry of factor Xa association with 365, 981–992 (2011).

NATURE REVIEWS | CARDIOLOGY VOLUME 9  |  JULY 2012  |  399


© 2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

22. Patel, M. R. et al. Rivaroxaban versus warfarin of thrombin‑thrombomodulin–protein C system 56. Schulman, S. & Kearon, C. Definition of major
in nonvalvular atrial fibrillation. N. Engl. J. Med. [abstract]. Blood 108, a914 (2006). bleeding in clinical investigations of
365, 883–891 (2011). 40. Food and Drug Administration, Cardiovascular antihemostatic medicinal products in
23. Cabral, K. P. & Ansell, J. Oral direct factor Xa and Renal Drugs Advisory Committee. Briefing non-surgical patients. J. Thromb. Haemost.
inhibitors for stroke prevention in atrial information [online], http://www.fda.gov/ohrms/ 3, 692–694 (2005).
fibrillation. Nat. Rev. Cardiol. http://dx.doi.org/ dockets/ac/04/briefing/2004–4069b1.htm 57. Alexander, J. H. et al. Apixaban with antiplatelet
10.1038/nrcardio.2012.19 (2004). therapy after acute coronary syndrome. N. Engl.
24. Furie, B. & Furie, B. C. Mechanisms of thrombus 41. Ansell, J. Factor Xa or thrombin: is factor Xa a J. Med. 365, 699–708 (2011).
formation. N. Engl. J. Med. 359, 938–949 (2008). better target? J. Thromb. Haemost. 5 (Suppl. 1), 58. Chesebro, J. H. et al. Thrombolysis in Myocardial
25. Morrissey, J. H., Macik, B. G., 60–64 (2007). Infarction (TIMI) Trial, phase I: a comparison
Neuenschwander, P. F. & Comp, P. C. 42. Yusuf, S. et al. Comparison of fondaparinux between intravenous tissue plasminogen
Quantitation of activated factor VII levels in and enoxaparin in acute coronary syndromes. activator and intravenous streptokinase. Clinical
plasma using a tissue factor mutant selectively N. Engl. J. Med. 354, 1464–1476 (2006). findings through hospital discharge. Circulation
deficient in promoting factor VII activation. Blood 43. Yusuf, S. et al. Effects of fondaparinux on 76, 142–154 (1987).
81, 734–744 (1993). mortality and reinfarction in patients with acute 59. Davis, E. M., Packard, K. A., Knezevich, J. T. &
26. Monroe, D. M., Hoffman, M. & Roberts, H. R. ST-segment elevation myocardial infarction: Campbell, J. A. New and emerging anticoagulant
Transmission of a procoagulant signal from the OASIS‑6 randomized trial. JAMA 295, therapy for atrial fibrillation and acute coronary
tissue factor-bearing cell to platelets. Blood 1519–1530 (2006). syndrome. Pharmacotherapy 31, 975–1016
Coagul. Fibrinolysis 7, 459–464 (1996). 44. Alexander, J. H. et al. Apixaban, an oral, direct, (2011).
27. Diaz-Ricart, M. et al. Thrombin facilitates primary selective factor Xa inhibitor, in combination with 60. Weinz, C., Radtke, M., Schmeer, K., Kern, A.
platelet adhesion onto vascular surfaces in the antiplatelet therapy after acute coronary & Pleiss, U. In vitro metabolism of BAY 59‑7939
absence of plasma adhesive proteins: studies syndrome: results of the Apixaban for Prevention —an oral, direct factor Xa inhibitor. Drug Metab.
under flow conditions. Haematologica 85, of Acute Ischemic and Safety Events (APPRAISE) Rev. 36, 98–98 (2004).
280–288 (2000). trial. Circulation 119, 2877–2885 (2009). 61. Weinz, C. et al. Metabolism and distribution
28. Monkovic, D. D. & Tracy, P. B. Functional 45. Turpie, A. G. et al. A randomized evaluation of [C14]BAY 59‑7939—an oral, direct factor Xa
characterization of human platelet-released of betrixaban, an oral factor Xa inhibitor, for inhibitor—in rat, dog and human. Drug Metab.
factor V and its activation by factor Xa and prevention of thromboembolic events after total Rev. 36, 98–98 (2004).
thrombin. J. Biol. Chem. 265, 17132–17140 knee replacement (EXPERT). Thromb. Haemost. 62. Mega, J. L. et al. Rivaroxaban in patients with a
(1990). 101, 68–76 (2009). recent acute coronary syndrome. N. Engl. J. Med.
29. Monkovic, D. D. & Tracy, P. B. Activation of human 46. Steg, P. G. et al. RUBY‑1: a randomized, double- 366, 9–19 (2012).
factor V by factor Xa and thrombin. Biochemistry blind, placebo-controlled trial of the safety and 63. Mehta, R. S. Novel oral anticoagulants
29, 1118–1128 (1990). tolerability of the novel oral factor Xa inhibitor for prophylaxis and treatment of venous
30. Hultin, M. B. Modulation of thrombin-mediated darexaban (YM150) following acute coronary thromboembolism: part I (factor Xa inhibitors).
activation of factor VIII:C by calcium ions, syndrome. Eur. Heart J. 32, 2541–2554 (2011). Expert Rev. Hematol. 3, 227–241 (2010).
phospholipid, and platelets. Blood 66, 53–58 47. Ogata, K. et al. Clinical safety, tolerability, 64. Astellas Pharma. Astellas Pharma Inc.
(1985). pharmacokinetics, and pharmacodynamics of discontinues development of darexaban
31. Bono, F. et al. Human umbilical vein endothelial the novel factor Xa inhibitor edoxaban in healthy (YM150), an oral direct factor Xa inhibitor
cells express high affinity receptors for factor Xa. volunteers. J. Clin. Pharmacol. 50, 743–753 [online], http://www.astellas.com/en/
J. Cell. Physiol. 172, 36–43 (1997). (2010). corporate/news/detail/astellas‑pharma‑inc-
32. Pejler, G., Lunderius, C. & Tomasini-Johansson, B. 48. Kohrt, J. T. et al. The discovery of (2R,4R)-N- discontinu.html (2011).
Macrophages synthesize factor X and secrete (4-chlorophenyl)-N-(2-fluoro-4-(2-oxopyridin- 65. Fujimoto, T. et al. Discovery of a
factor X/Xa-containing prothrombinase activity 1(2H)-yl)phenyl)-4-methoxypyrrolidine-1,2- tetrahydropyrimidin-2(1H)-one derivative
into the surrounding medium. Thromb. Haemost. dicarboxamide (PD 0348292), an orally (TAK‑442) as a potent, selective, and orally
84, 429–435 (2000). efficacious factor Xa inhibitor. Chem. Biol. Drug active factor Xa inhibitor. J. Med. Chem.
33. Altieri, D. C. & Edgington, T. S. Identification Des. 70, 100–112 (2007). 53, 3517–3531 (2010).
of effector cell protease receptor‑1. A 49. Goldstein, S. et al. Safety evaluation of the 66. Weitz, J. I. New oral anticoagulants in
leukocyte-distributed receptor for the serine factor Xa inhibitor TAK‑442 in subjects with development. Thromb. Haemost. 103, 62–70
protease factor Xa. J. Immunol. 145, 246–253 acute coronary syndromes: phase 2 AXIOM-ACS (2010).
(1990). trial results [abstract]. Eur. Heart J. 67. Weitz, J. I. et al. A dose-finding study with
34. Macfarlane, R. G. An enzyme cascade in the 32 (Abstract Suppl.), 414 (2011). TAK‑442, an oral factor Xa inhibitor, in patients
blood clotting mechanism, and its function as 50. Agnelli, G. et al. A phase II study of the oral undergoing elective total knee replacement
a biochemical amplifier. Nature 202, 498–499 factor Xa inhibitor LY517717 for the prevention surgery. Thromb. Haemost. 104, 1150–1157
(1964). of venous thromboembolism after hip or knee (2010).
35. Yin, E. T. & Wessler, S. Investigation of the replacement. J. Thromb. Haemost. 5, 746–753 68. Lee, C. W. et al. Comparison of ruptured
apparent thrombogenicity of thrombin. Thromb. (2007). coronary plaques in patients with unstable
Diath. Haemorrh. 20, 465–468 (1968). 51. Mega, J. L. et al. Rivaroxaban versus placebo and stable clinical presentation. J. Thromb.
36. Ieko, M. et al. Synthetic selective inhibitors of in patients with acute coronary syndromes Thrombolysis 32, 150–157 (2011).
coagulation factor Xa strongly inhibit thrombin (ATLAS ACS–TIMI 46): a randomised, double- 69. Undas, A., Brummel, K., Musial, J., Mann, K. G.
generation without affecting initial thrombin blind, phase II trial. Lancet 374, 29–38 (2009). & Szczeklik, A. Blood coagulation at the site of
forming time necessary for platelet activation 52. Pinto, D. J. et al. Discovery of microvascular injury: effects of low-dose aspirin.
in hemostasis. J. Thromb. Haemost. 2, 612–618 1-(4-methoxyphenyl)-7-oxo-6-(4-(2-oxopiperidin- Blood 98, 2423–2431 (2001).
(2004). 1-yl)phenyl)-4,5,6,7-tetrahydro-1H-pyrazolo[3,4-c] 70. Brummel, K. E., Paradis, S. G., Butenas, S.
37. Petros, S., Siegemund, T., Siegemund, A. pyridine-3-carboxamide (apixaban, & Mann, K. G. Thrombin functions during tissue
& Engelmann, L. The effect of different BMS‑562247), a highly potent, selective, factor-induced blood coagulation. Blood
anticoagulants on thrombin generation. efficacious, and orally bioavailable inhibitor 100, 148–152 (2002).
Blood Coagul. Fibrinolysis 17, 131–137 (2006). of blood coagulation factor Xa. J. Med. Chem. 71. Mann, K. G. Thrombin generation in hemorrhage
38. Furugohri, T., Sugiyama, N., Morishima, Y. & 50, 5339–5356 (2007). control and vascular occlusion. Circulation
Shibano, T. Antithrombin-independent thrombin 53. Raghavan, N. et al. Apixaban metabolism and 124, 225–235 (2011).
inhibitors, but not direct factor Xa inhibitors, pharmacokinetics after oral administration to 72. Haynes, L. M., Dubief, Y. C., Orfeo, T. &
enhance thrombin generation in plasma through humans. Drug Metab. Dispos. 37, 74–81 (2009). Mann, K. G. Dilutional control of prothrombin
inhibition of thrombin–thrombomodulin– 54. Karthikeyan, G. & Eikelboom, J. W. Apixaban activation at physiologically relevant shear rates.
protein C system. Thromb. Haemost. 106, in acute coronary syndromes. Cardiovasc. Ther. Biophys. J. 100, 765–773 (2011).
1076–1083 (2011). 29, 285–290 (2011). 73. Diener, H. C. et al. Aspirin and clopidogrel
39. Morishima, Y., Furugohri, T., Shiozaki, Y., 55. Wang, L. et al. In vitro assessment of metabolic compared with clopidogrel alone after recent
Sugiyama, N. & Shibano, T. Antithrombin- drug–drug interaction potential of apixaban ischaemic stroke or transient ischaemic attack
independent thrombin inhibitors, but not through cytochrome P450 phenotyping, in high-risk patients (MATCH): randomised,
factor Xa inhibitors, enhance thrombin inhibition, and induction studies. Drug Metab. double-blind, placebo-controlled trial. Lancet
generation in human plasma via inhibition Dispos. 38, 448–458 (2010). 364, 331–337 (2004).

400  |  JULY 2012  |  VOLUME 9  www.nature.com/nrcardio


© 2012 Macmillan Publishers Limited. All rights reserved
FOCUS ON ANTICOAGULATION THERAPIES

74. Bauersachs, R. et al. Oral rivaroxaban for direct factor Xa inhibitor [abstract]. Blood thrombosis. J. Cardiovasc. Pharmacol.
symptomatic venous thromboembolism. N. Engl. 108, a273 (2006). 56, 156–161 (2010).
J. Med. 363, 2499–2510 (2010). 81. Groenendaal-van de Meent, D. et al. YM150, 86. Nakamura, K. et al. Comparison of the
75. Kubitza, D., Becka, M., Voith, B., Zuehlsdorf, M. an oral direct inhibitor of factor Xa, pharmacokinetics and pharmacodynamics
& Wensing, G. Safety, pharmacodynamics, demonstrated a predictable and of TAK‑442, a novel factor Xa inhibitor, between
and pharmacokinetics of single doses of dose-proportional pharmacokinetic/ Japanese acute coronary syndrome patients
BAY 59‑7939, an oral, direct factor Xa inhibitor. pharmacodynamic profile after single and and healthy Japanese males. J. Thromb.
Clin. Pharmacol. Ther. 78, 412–421 (2005). multiple dosing: results from three studies Haemost. 9, 107–107 (2011).
76. Roe, M. T. & Ohman, E. M. A new era in [abstract]. Blood 116, a3323 (2010). 87. Gibson, C. M. et al. Rationale and design of the
secondary prevention after acute coronary 82. Saiah, E. & Soares, C. Small molecule Anti-Xa therapy to lower cardiovascular events
syndrome. N. Engl. J. Med. 366, 85–87(2011). coagulation cascade inhibitors in the clinic. in addition to standard therapy in subjects with
77. Tricoci, P. et al. Thrombin-receptor antagonist Curr. Top. Med. Chem. 5, 1677–1695 (2005). acute coronary syndrome—thrombolysis in
vorapaxar in acute coronary syndromes. N. Engl. 83. Perzborn, E. et al. In vitro and in vivo studies myocardial infarction 51 (ATLAS ACS 2–TIMI 51)
J. Med. 366, 20–33 (2011). of the novel antithrombotic agent BAY 59‑7939 trial: a randomized, double-blind, placebo-
78. Lu, G. et al. Recombinant antidote for reversal —an oral, direct factor Xa inhibitor. J. Thromb. controlled study to evaluate the efficacy and
of anticoagulation by factor Xa inhibitors Haemost. 3, 514–521 (2005). safety of rivaroxaban in subjects with acute
[abstract]. Blood 112, a983 (2008). 84. Lu, G. et al. Reversal of rivaroxaban mediated coronary syndrome. Am. Heart J. 161,
79. Turpie, A. G. Oral, direct factor Xa inhibitors anticoagulation in animal models by a 815–821e6 (2011).
in development for the prevention and recombinant antidote protein (r‑Antidote,
treatment of thromboembolic diseases. PRT064445) [abstract]. Eur. Heart J. Author contributions
Arterioscler. Thromb. Vasc. Biol. 27, 1238–1247 32 (Abstract Suppl.), 640–641 (2011). J. W. Wisler researched the data for this article. Both
(2007). 85. Kawamura, M. et al. Antithrombotic and authors contributed substantially to discussion of its
80. Iwatsuki, Y. et al. Biochemical and anticoagulant profiles of TAK‑442, a novel contents and to writing, reviewing, and editing the
pharmacological profiles of YM150, an oral factor Xa inhibitor, in a rabbit model of venous manuscript before submission.

NATURE REVIEWS | CARDIOLOGY VOLUME 9  |  JULY 2012  |  401


© 2012 Macmillan Publishers Limited. All rights reserved

Potrebbero piacerti anche