Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Circulation

IN DEPTH
Combination Antiplatelet and Oral
Anticoagulant Therapy in Patients With
Coronary and Peripheral Artery Disease
Focus on the COMPASS Trial

ABSTRACT: Antiplatelet therapy is the mainstay for the treatment of Paul A. Gurbel, MD
acute and chronic arterial disease involving the coronary and peripheral Keith A.A. Fox, MB ChB
beds. However, questions remain about optimal antithrombotic therapy Udaya S. Tantry, PhD
for long-term treatment of chronic vascular disease. The observation Hugo ten Cate, MD, PhD
that dual antiplatelet therapy with acetylsalicylic acid and clopidogrel Jeffrey I. Weitz, MD
was associated with lower thrombotic event rates than acetylsalicylic
acid monotherapy in patients with acute coronary syndromes and those
undergoing percutaneous coronary intervention changed the treatment
paradigm. Moreover, the demonstration that more pharmacodynamically
potent P2Y12 inhibitors than clopidogrel were associated with fewer
thrombotic event occurrences further solidified the dual antiplatelet
therapy approach. However, recurrent thrombotic events occur in ≈1 in
Downloaded from http://ahajournals.org by on June 29, 2023

10 patients in the first year following an acute coronary syndrome event,


despite treatment with the most potent P2Y12 inhibitors, a limitation that
has stimulated interest in exploring the efficacy and safety of approaches
using anticoagulants on top of antiplatelet therapy. These investigations
have included treatment with very-low-dose oral anticoagulation, and
even its replacement of acetylsalicylic acid in the presence of a P2Y12
inhibitor, in patients stabilized after an acute coronary syndrome event.
Recent basic and translational studies have suggested noncanonical
effects of coagulation factor inhibition that may further modulate
clinical benefits. This in-depth review will discuss developments in our
understanding of the roles that platelets and coagulation factors play
in atherothrombosis and review the rationale and clinical evidence for
combining antiplatelet and oral anticoagulant therapy in patients with
coronary and peripheral artery disease.

Key Words: anticoagulants ◼ aspirin


◼ blood coagulation ◼ blood platelets
◼ coronary artery disease ◼ factor Xa
inhibitors◼ peripheral arterial disease
◼ rivaroxaban

© 2019 American Heart Association, Inc.

https://www.ahajournals.org/journal/circ

2170 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

P
atients with coronary artery disease (CAD) and pe- inflammation, a thromboinflammatory syndrome.12,13
ripheral artery disease (PAD) are at high risk of fu- Vascular injury exposes the subendothelial matrix, and

STATE OF THE ART


ture cardiovascular events.1 Despite implementa- platelets adhere to newly exposed collagen and von Wil-
tion of contemporary secondary prevention strategies, lebrand factor, interactions mediated by specific platelet
there was an ≈25% 3-year cumulative risk of cardio- receptors. The von Willebrand factor-glycoprotein Ib/
vascular death, myocardial infarction (MI), or stroke in IX/V interactions play a major role in initial platelet ad-
patients with prior MI.2 Among patients with symptom- hesion and activation in the high-shear environment of
atic PAD and no history of stroke or transient ischemic coronary arteries, whereas this process has been less ex-
attack, the rate of cardiovascular death, nonfatal MI, plored in peripheral arteries. Following activation, ADP
or stroke was 17.6% by year 4 and was driven mainly is released from platelet dense granules, and Throm-
by cardiovascular mortality (11.1%). Systemic (com- boxane A2 is generated by the activity of thromboxane
posite of cardiovascular death, MI, or stroke) and limb synthase on prostaglandin H2, an intermediate formed
ischemic event (composite of lower limb amputation, by cyclooxygenase-1 (COX-1). Although both ADP and
peripheral bypass graft, and percutaneous intervention thromboxane A2 amplify platelet activation and aggre-
for PAD) occurrence was 11.9% at 2 years.3 Consistent gation, continuous ADP-P2Y12 receptor signaling is es-
with the systemic atherosclerotic process, atherothrom- sential for sustained activation of the glycoprotein IIb/
botic event rates involving coronary or cerebral arteries IIIa receptor and stable platelet thrombus generation.5
are higher in those with polyvascular bed involvement The contribution of coagulation in the initiation of
than patients with PAD alone.4 acute arterial thrombosis is less well established than
A critical component of the treatment strategy for platelet function. Only femtomolar (1×10–15) levels of
patients with CAD and PAD is antiplatelet therapy. De- thrombin are generated during the initial stages of clot
spite evidence of clinical event reduction associated formation, whereas the major amount of thrombin is
with potent P2Y12 inhibitors plus acetylsalicylic acid generated after clot formation, suggesting that throm-
(ASA) in large-scale trials, residual ischemic risk persists bin is primarily important for clot stability but not initia-
along with risks for bleeding that are proportional to tion.14 Nonetheless, elevated procoagulant activity and
the intensity of the antiplatelet regimen.5–8 Therefore, thrombin generation were reported during the acute
there is a need for better therapies. Recent attention has phases of unstable angina and MI and persisted for
focused on the efficacy and safety of combination an- months after presentation.15–20 Thrombin generation,
Downloaded from http://ahajournals.org by on June 29, 2023

tiplatelet and anticoagulant therapy with non–vitamin marked by prothrombin fragment 1+2, was heightened
K antagonist direct oral anticoagulants. In this review, in the first 12 hours of acute MI, and was higher in
we will discuss the developments in our understand- patients with acute coronary syndrome (ACS) who de-
ing of the roles that platelets and coagulation factors veloped in-hospital recurrent ischemia despite nearly 72
play in atherothrombosis with a focus on the results of hours of intravenous heparin infusion.16 In addition, an
the recent COMPASS trial (Cardiovascular Outcomes for association between in vivo thrombin generation and
People Using Anticoagulation Strategies) that imple- severity of coronary vessel disease defined by angiog-
mented a very low dose of rivaroxaban on top of ASA in raphy21 or coronary calcification by computed tomogra-
patients with chronic CAD and PAD.9 Mechanisms that phy has been reported.22
may underlie the results of COMPASS are described. The latter fundamental construct provides the ratio-
nale for pharmacological strategies that have focused
on platelet inhibition and anticoagulation to prevent
MECHANISTIC RATIONALE FOR ischemic events in patients with cardiovascular disease.
COMBINING ANTIPLATELETS AND Targets for inhibition include COX-1, P2Y12, and the
ANTICOAGULANTS thrombin pathway (Figure 1).5 At this time, the relative
importance of these individual pathways in the initial
Atherosclerosis is the common pathophysiological pro-
genesis of thrombus formation in the coronary and pe-
cess underlying both CAD and PAD. As initially described
ripheral arterial vascular beds remains elusive.
in the injury-repair hypothesis by Ross10, platelet adhe-
sion, activation, and aggregation (both platelet-platelet
and platelet-monocyte) play pivotal roles in the initia- CROSS-TALK BETWEEN PLATELETS
tion and progression of atherosclerotic lesions and the
final manifestation of thrombotic complications (athero- AND COAGULATION
thrombosis) following atherosclerotic plaque erosion or Exposed tissue factor (TF), derived from macrophages,
rupture.10,11 Coronary atherosclerotic disease progression vascular smooth muscle cells, and microvesicles at the
from a stable to an unstable state has been character- site of vascular injury,23 initiates the coagulation cas-
ized as a pathophysiological condition associated with cade, triggering thrombin generation. Thrombin ac-
heightened platelet activation, hypercoagulability, and tivates platelet protease-activated receptor (PAR)–1

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2171


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy
STATE OF THE ART

Figure 1. Canonical mechanistic concept of atherothrombosis and potential therapeutic targets.


Platelet activation and coagulation are stimulated by the interaction of flowing blood with injured vessel wall. There are 3 major pathways amplifying platelet
activation that are inhibited by currently available oral agents: 1, the COX-1 pathway; 2, the ADP-P2Y12 pathway; and 3, the thrombin pathway. Activity of the
thrombin pathway can be inhibited by direct inhibition of thrombin, inhibition of thrombin generation by targeting FXa, and inhibition of the platelet protease
activated receptor (PAR)-1, the thrombin receptor. ASA indicates acetylsalicylic acid; EC, endothelial cell; FVIIa, factor VIIa; FXa, factor Xa; GPIIb/IIIa, glycoprotein IIb/
IIIa; TF, tissue factor; and TxA2, thromboxane A2. Modified from Gurbel and Tantry5 with permission. © 2010, the American Heart Association.

and PAR-4, the latter in a dynamic manner and prob- platelets is in a partially activated state, and factor Xa
ably mostly during thromboinflammatory conditions,24 is 50 to 100 times more effective than thrombin in ac-
causing further platelet activation. Concurrently, the tivating platelet-released factor V. The activated factor
phosphatidylserine-rich surface of activated platelets V on the platelet surface is well positioned to combine
supports the assembly of clotting factor complexes that with factor Xa and form the potent prothrombin acti-
Downloaded from http://ahajournals.org by on June 29, 2023

amplify thrombin generation. In addition to activating vation complex. The activity of this complex is attenu-
platelets, thrombin converts fibrinogen to fibrin and ated by direct factor Xa inhibitors but not by throm-
activates factor XIII, and enhances its own generation bin inhibitors. Platelet prothrombinase may produce
through feedback activation of factors V, VIII, and XI. too much thrombin to be effectively controlled with
Therefore, activated platelets and hypercoagulability thrombin inhibitors.31,32 Therefore, platelet activation
play important roles in occlusive thrombus formation at and the formation of the initial prothrombinase com-
the site of vascular injury, and atherothrombosis under- plex play important roles in the early genesis of arterial
lies the ultimate clinical manifestations of MI, ischemic thrombosis.
stroke, and a substantial proportion of acute limb isch- In support of the cross-talk between platelets and
emic events in PAD.11,12,25 Multiple studies have dem- coagulation and the modulation of this cross-talk by
onstrated elevated markers of platelet activation and platelet inhibitors, ex vivo inhibition of the P2Y12 recep-
coagulation, and evidence of inflammation and en- tor by clopidogrel diminished the rapid exposure of TF in
dothelial activation, as well, in patients with CAD and a mouse model and attenuated TF-dependent factor Xa
PAD.26–29 activity of platelet-neutrophil suspensions.33 Clopidogrel
A recent murine transgenic model suggested that markedly and rapidly inhibited platelet annexin-binding
platelet-derived factor V is a critical mediator of in activity34 and prolonged the time to platelet-fibrin clot
vivo arterial thrombosis involving platelet activation formation measured by thromboelastography, a marker
and coagulation. Platelet-derived factor V significantly of the rate of thrombin generation.35 Other studies sup-
contributed to localized assembly of the prothrombi- port the attenuation of platelet procoagulant activity by
nase complex with factor Xa at the site of plaque rup- the in vitro addition of P2Y12 inhibitors.36,37
ture. The latter process leads to the generation of sig- However, coronary thrombosis may still occur despite
nificant amounts of thrombin on the activated platelet the presence of potent P2Y12 blockade, suggesting the
surface and early feedback loop stimulation of coagu- importance of mechanisms beyond platelet function
lation pathways. The direct thrombin inhibitors may driving thrombus occurrence.5 Thromboelastography
not have the same influence in attenuating thrombin studies in stented patients who were pharmacodynam-
generation in this setting as do factor Xa inhibitors.30 ically responsive to dual antiplatelet therapy (DAPT),
In support of this concept, it has been shown that hu- and who had recurrent ischemic events, revealed rapid
man factor V released from α-granules of activated times to platelet-fibrin clot generation and formation of

2172 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

STATE OF THE ART


Figure 2. Potential noncanonical pathways involved in atherothrombosis and their inhibition by FXa inhibitors and ASA.
The role of FXa in atherosclerosis has been attributed to direct activation of PAR-2 signaling or indirect activation of PAR-1 through the generation of thrombin
with resultant release of proinflammatory cytokines and expression of cell adhesion molecules. Resultant vascular effects include modulation of inflammation,
plaque progression, and fibrosis. ASA has been reported to modulate clot porosity through non–COX-1–mediated effects. Activated platelets generate thrombin
on their surface, an event that is attenuated by antiplatelet therapy. ASA indicates acetylsalicylic acid; EC, endothelial cell; FXa, factor Xa; PAR, protease activated
receptor; and WBC, white blood cell.

clots with high tensile strength, findings that support a thrombotic effects beyond the inhibition of aggrega-
hypercoagulable phenotype.38 tion (Figure 2). In a human microvascular injury model,
Another potentially relevant thromboinflammatory in addition to reducing thrombin generation and atten-
network involves a platelet-localized factor XI–depen- uating factor XIII activation, ASA also enhanced fibrin
dent mechanism of thrombin generation that impacts clot permeability and clot lysis by acetylating lysine resi-
vascular tone, blood pressure, and endothelial func- dues in fibrin.41,42 Therefore, very-low-dose rivaroxaban
tion in an angiotensin-dependent manner.39 Factor XI combined with ASA may act synergistically to inhibit
Downloaded from http://ahajournals.org by on June 29, 2023

is a component of the intrinsic pathway of coagulation, thrombosis through multiple pathways, including direct
which is activated by factor XIIa or by thrombin. It has effects of ASA on the clot and the effects mediated by
been shown that activated factor XI can activate coagu- the inhibition of platelet activation and resultant inhibi-
lation factors IX, V, and VIII, and inhibit the anticoagu- tion of activated coagulation factor generation. In an
lant TF pathway inhibitor. In addition, the role of factor in vitro study, enhanced inhibition of thrombin gen-
XI as an independent risk factor for deep vein throm- eration was observed when ticagrelor and ticagrelor
bosis, ischemic stroke, and MI has been demonstrat- plus ASA were combined with rivaroxaban in com-
ed.40 Taken together, the above data provide evidence parison with when the agents were not combined.43
of substantial cross-talk between platelet function and Also, in an arteriovenous shunt model, low doses of
coagulation and provide the supportive mechanistic ra- rivaroxaban combined with ASA or ASA plus clopi-
tionale for potential synergistic antithrombotic effects dogrel resulted in an increased antithrombotic effect
derived from the inhibition of platelet activation and in comparison with the effects observed when these
thrombin generation by combination therapy with an agents were not combined.43 The above potentiation
antiplatelet agent and a factor Xa inhibitor. Finally, non- of the inhibition of thrombin generation and throm-
canonical effects of factor Xa inhibition together with bus formation by the combination of rivaroxaban with
noncanonical effects of ASA and P2Y12 receptor inhibi- antiplatelet agents suggests potential mechanisms for
tors may also modulate clinical outcomes in patients the reduction of ischemic effects that were observed in
with arterial diseases. the COMPASS trial.9

NONCANONICAL EFFECTS OF P2Y12 Inhibitors


ANTITHROMBOTIC AGENTS P2Y12 is a G-protein–coupled receptor consisting of
a single polypeptide chain with 7 transmembrane α-
Acetylsalicylic Acid helices associated with multiple functionally different
In addition to the antithrombotic properties of ASA G proteins that elicit specific intracellular responses to
classically attributed to platelet COX-1 inhibition and ADP resulting in the activation of the glycoprotein IIb/
resultant inhibition of aggregation, ASA exhibits anti- IIIa receptor.44 Active metabolites of thienopyridines (ti-

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2173


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

clopidine, clopidogrel, and prasugrel) irreversibly bind of the platelet-fibrin clot that marks the robustness of,
to the ADP binding site and thereby prevent intracel- and the response to, thrombin generation.
STATE OF THE ART

lular signaling and ADP-induced platelet aggregation. Rivaroxaban has been shown to decrease proinflam-
In addition, P2Y12 receptor mRNA expression and ADP- matory cytokine expression in primary cultures of hu-
induced vasoconstriction have been reported in human man umbilical vein endothelial cells, and its effect was
vascular smooth muscle cells.45 Animal studies have comparable to PAR-1 and direct thrombin inhibition.61
also shown that the P2Y12 receptor may participate In apolipoprotein E–deficient mice with atherosclerotic
in atherogenesis by promoting the proliferation and lesions, rivaroxaban reduced the expression of pro-
migration of vascular smooth muscle cells and by in- inflammatory cytokines such as interleukin-6, tumor
ducing endothelial dysfunction. In an apolipoprotein necrosis factor-α, and monocyte chemoattractant pro-
E–deficient mice model, clopidogrel reduced levels of tein-1, and was suggested to enhance plaque stability.62
p-selectin, e-selectin, monocyte chemoattractant pro- Thrombin increases the activation of leukocytes and
tein-1, and platelet-derived growth factor β, and mac- surface expression of adhesion molecules. The pleiotro-
rophage and T-cell infiltration in atherosclerotic lesions, pic effects of factor Xa and thrombin are described in
and delayed the development and progression of de detail elsewhere.50,63 In summary, the data above sug-
novo atherosclerosis.46 In addition to potent inhibition gest that noncanonical mechanisms underlying the po-
of the P2Y12 receptor, ticagrelor, a cyclopentyl-triazolo- tential vascular protective effects of very-low-dose riva-
pyrimidine, inhibits adenosine reuptake in erythrocytes roxaban include modulation of cellular function either
and other cells. The latter effect has been attributed to directly by inhibiting the effects of factor Xa on PAR-2,
improved platelet inhibition and coronary blood flow or indirectly by inhibiting thrombin generation and its
and reduced infarct size.47–49 These noncanonical ef- effects on PAR-1.
fects of ticagrelor may account for some of the clini-
cal benefits associated with ticagrelor observed in the
PLATO trial (Platelet Inhibition and Patient Outcomes).7 CLINICAL RATIONALE
COMBINING ANTIPLATELETS AND
Factor Xa Inhibitors ANTICOAGULANTS
There is growing evidence that factor Xa and throm- DAPT with ASA and a P2Y12 receptor blocker is indicat-
bin play critical roles in various cellular processes50 (Fig- ed acutely and for up to 12 months in the high-risk pa-
Downloaded from http://ahajournals.org by on June 29, 2023

ure 2). In addition to its main role in triggering thrombin tient with ACS and in those undergoing coronary stent-
generation, factor Xa is involved in inflammation, vas- ing. For long-term secondary prevention, ASA remains
cular remodeling, atherosclerotic plaque progression, the cornerstone of therapy.5 Various strategies have
and tissue fibrosis.51,52 The role of factor Xa in the ath- been investigated to improve the long-term outcomes
erosclerosis process has been attributed to direct activa- of ASA therapy but with limited success (Table 18,64–70).
tion of PAR-2 signaling, or indirect activation of PAR-1 In patients with stable cardiovascular disease, there
via thrombin generation with resultant release of proin- was only a marginal benefit of clopidogrel over ASA
flammatory cytokines and the expression of cell adhe- in major adverse cardiovascular events (MACE) reduc-
sion molecules.50 Both PAR-1 and PAR-2 have been im- tion and no difference in cardiovascular or all-cause
plicated in atherosclerosis development.53 In addition to mortality.64 Among patients with symptomatic stable
factor Xa and thrombin, other agonists including metal- cardiovascular disease or multiple risk factors, DAPT
loproteinases have been implicated in PAR activation.54 with clopidogrel added to ASA did not lower MACE
In in vitro studies, factor Xa induced interleukin-6, in- or all-cause mortality in comparison with ASA alone.65
terleukin-8, and monocyte chemoattractant protein-1 In patients with prior MI, the combination of ticagre-
expression in endothelial cells and leukocytes,51,52,55 re- lor and ASA modestly reduced MACE but increased
sponses that are more efficient when factor Xa forms a bleeding in comparison with ASA monotherapy, with-
complex with TF-VIIa.50,56 out an effect on cardiovascular or all-cause mortality.8
Factor Xa inhibition attenuates some of the pleiotro- However, in patients with stable PAD, ticagrelor alone
pic effects of factor Xa, reduces proinflammatory cyto- did not lower MACE in comparison with the weaker
kine expression, and attenuates restenosis after balloon P2Y12 inhibitor, clopidogrel.66 Blockade of PAR-1 with
angioplasty in rabbits.57,58 Moreover, ex vivo–measured vorapaxar in patients with stable cardiovascular disease
high-tensile platelet fibrin-clot strength occurring after on top of single or dual antiplatelet therapy reduced
the generation of thrombin was associated with the MACE, but increased bleeding and did not reduce mor-
development of 6-month ischemic events after elective tality.67 However, a benefit of vorapaxar on reducing the
coronary stenting and in-stent restenosis.59,60 These ob- rates of hospitalization for acute limb ischemia and pe-
servations suggest that prothrombotic and mitogenic ripheral artery revascularization in patients with stable
phenotypes may be identified by the tensile property cardiovascular disease has been reported.68

2174 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

Table 1. Oral Antiplatelet and Anticoagulant Therapy Trials in Patients With Stable Coronary and Peripheral Artery Diseases

STATE OF THE ART


Clinical Trial (n)Ref Patient Population (n) Treatment End Points Outcomes
PEGASUS-TIMI 54 Patients with MI 1–3 y 90 mg twice daily TIG vs Primary efficacy end Primary efficacy end point=7.85% vs 7.77%
(n=21 162)8 earlier 60 mg twice daily TIG point=composite of vs 9.04%.
vs ASA for a median of cardiovascular mortality, MI, HR for 60 mg TIG vs placebo=0.85; P=0.008
33 mo or stroke.
HR for 60 mg TIG vs placebo=0.84; P=0.004
Primary safety end point=TIMI
TIMI major bleeding=2.60% with 90 mg and
major bleeding
2.30% with 60 mg TIG vs 1.06% placebo
(P<0.001 for each dose vs placebo)
CAPRIE Patients with 75 mg CLP vs 325 mg First occurrence of ischemic Primary end point=5.32% vs 5.83%
(n=19 185)64 atherosclerotic vascular ASA for median of 1.9 y stroke, MI, or vascular RRR=8.7%; P=0.043
disease manifested as mortality
Safety=no significant difference in bleeding
either recent ischemic
(9.28% vs 9.27%); ASA group greater
stroke, recent MI, or
gastrointestinal bleeding (2.66% vs 1.9%;
symptomatic PAD
P=0.05
No difference in cardiovascular or all-cause
mortality
CHARISMA CVD, or PAD or ≥3 75–162 mg ASA+75 mg Primary end point=composite Primary end point: RR=0.93; P=0.22;
(n=15 603)65 atherothrombotic risk CLP vs 75–162 mg/d of MI, stroke, or death from Secondary end point: RR=0.92; P=0.004
factors ASA+ placebo median for cardiovascular causes;
Increase in GUSTO severe bleeding
28 mo Secondary end
(RR=1.25; P=0.09) and moderate bleeding
point=hospitalization for UA,
(RR=1.62; P=0.001)
TIA, or revascularization
No difference in all-cause mortality
procedure
EUCLID (n=13 885) 66
Patients with 90 mg twice daily TIG vs Primary efficacy end Primary efficacy end point= 0.8% vs 10.6%,
symptomatic PAD 75 mg once daily CLP for point=composite of adjudicated HR=1.02; P=0.65
median of 30 mo cardiovascular mortality, MI,
Primary safety end point=1.6% each,
or ischemic stroke
HR=1.10; P=0.49
Primary safety end point =
major bleeding
TRAP2P-TI MI 50 Patients with a history 2.5 mg once daily Primary efficacy end Primary efficacy end point=9.3% vs 10.5%,
(n=26 449)67 of MI, ischemic stroke, vorapaxar or matching point=composite of HR=0.87; P<0.001
Downloaded from http://ahajournals.org by on June 29, 2023

or PAD placebo for a median of cardiovascular mortality, MI,


Primary safety end point=4.2% vs 2.5%,
30 mo or stroke
HR=1.66; P<0.001
Primary safety end
point=GUSTO moderate or
severe bleeding
Meta-analysis of Patients with established VKAs with and without Total mortality, MI, and stroke VKAs with INR>2.8 significantly reduced
randomized trials CAD antiplatelet therapy cardiovascular complications and increased
(n=20 000)69 bleeding in comparison with controls
VKAs with INR 2–3 and ASA is more effective
and as equally as safe as ASA alone
WAVE Patients with PAD Antiplatelet agent+oral First coprimary outcome=MI, First coprimary outcome=12.2% vs 13.3%,
(n=2161)70 anticoagulant agent stroke, cardiovascular RR=0.92; P=0.48
(target INR=2.0–3.0) or to mortality; Second coprimary outcome=15.9% vs 7.4%.
antiplatelet therapy alone Second coprimary RR=0.91; P=0.37
outcome=MI, stroke, severe Life-threatening bleeding=4.0% vs 1.2%,
ischemia of the peripheral RR=3.41; P<0.001
or coronary arteries leading
to urgent intervention, or
cardiovascular mortality

ASA indicates acetylsalicylic acid; CAD, coronary artery disease; CAPRIE, A Randomised, Blinded, Trial of Clopidogrel Versus Aspirin in Patients at Risk of Ischemic
Events; CHARISMA, Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management, and Avoidance; CLP, clopidogrel; EUCLID, Examining Use of
Ticagrelor in Peripheral Artery Disease; GUSTO, Global Utilization Of Streptokinase And Tpa For Occluded Arteries; HR, hazard ratio; INR, international normalization
ratio; MI, myocardial infarction; PAD, peripheral arterial disease; PEGASUS-TIMI 54, Prevention of Cardiovascular Events in Patients with Prior Heart Attack Using
Ticagrelor Compared to Placebo on a Background of Aspirin–Thrombolysis in Myocardial Infarction 54; RR, relative risk; TIG, ticagrelor; TIMI, thrombolysis in
myocardial infarction; TRAP2P-TI MI 50, Thrombin Receptor Antagonist in Secondary Prevention of Atherothrombotic Ischemic Events–Thrombolysis in Myocardial
Infarction 50; VKA, vitamin K antagonist; and WAVE, The Warfarin Antiplatelet Vascular Evaluation.

Earlier studies used vitamin K antagonists as an anti- tional normalized ratio, vitamin K antagonists reduced
coagulant in an attempt to improve clinical outcomes in cardiovascular events in patients with stable CAD but
patients with stable CAD and PAD. When administered increased bleeding.69 In contrast, no benefit was ob-
in doses sufficient to produce a therapeutic interna- served for low-dose vitamin K antagonists that did not

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2175


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

Table 2. Clinical Trials of Rivaroxaban in Patients With Coronary and Peripheral Artery Diseases
STATE OF THE ART

Patients
Population (Days
Clinical Trial (n)Ref From Index Event) Treatment End Points Outcomes
COMPASS Stable CAD and After a 30-day run in period, Primary efficacy end point: Primary efficacy end point=
(n=27 395)9 PAD, RIV 2.5 mg twice daily+ASA MI, stroke, or cardiovascular 2.5 mg RIV+ASA vs 5 mg RIV vs ASA alone; 4.1%
MI in the past 20 y (100 mg once daily), RIV death vs 4.9% vs 5.4%
alone (5 mg twice daily), or Primary safety end point: 2.5 mg RIV+ASA vs ASA alone: HR=0.76; P<0.001
ASA alone (100 mg once
major bleeding 5 mg RIV+ASA alone: HR=0.90, P=0.12
daily) for 1.95 y
Primary safety end point=2.5 mg RIV+ASA vs 5
mg RIV vs ASA alone: 3.1% vs 2.8% vs 1.9%.
2.5 mg RIV+ASA vs ASA alone: HR=1.70; P<0.001
5 mg RIV+ASA vs ASA alone: HR=1.51; P<0.001
ATLAS ACS-TIMI 46 Stabilized ACS RIV (5–20 mg) once or twice Primary safety end point: Primary safety end point: 5 mg HR=2.21, 10 mg
(n=3491)76 (2–7) daily vs placebo; ASA only clinically significant bleeding RIV HR=3.35, 15 mg RIV HR=3.60, and 20 mg RIV
vs ASA plus thienopyridine (TIMI major, TIMI minor, or HR=5.06; P<0.0001.
for 6 mo requiring medical attention). Primary efficacy end point: HR=0.79, P=0.$1$2s
Primary efficacy end Secondary efficacy end point: HR=0.69; P=0.027
point=death, MI, stroke, or
severe recurrent ischemia
requiring revascularization
Secondary efficacy end
point=death, MI, or stroke
ATLAS ACS-2 TIMI Stabilized ACS Rivaroxaban (2.5 or 5 mg Primary efficacy end Primary efficacy end point=2.5 mg RIV HR=0.84;
51 (n=15 526)77 (1–7) twice daily) vs placebo for a point=cardiovascular death, P=0.02 and 5 mg RIV HR=0.85; P=0.03
mean of 13 mo MI, or stroke Non-CABG TIMI bleeding=2.5 mg RIV HR=3.46;
P<0.001, 5 mg RIV HR=4.47; P<0.001
ICH=2.5 mg RIV HR=2.83, P=0.04; 5 mg RIV
HR=3.47, P=0.005
Fatal bleeding=2.5 mg RIV HR=0.67, P=0.45; 5
mg RIV HR=1.72, P=0.20
COMPASS-Stable Stable CAD, After a 30-day run in period, Primary efficacy end point: Primary efficacy end point=2.5 mg RIV +ASA vs 5
Downloaded from http://ahajournals.org by on June 29, 2023

CAD MI in the past 20 y RIV (2.5 mg twice daily)+ASA MI, stroke, or cardiovascular mg RIV vs ASA alone; 4% vs 5% vs 6%
(n=24 824)78 (100 mg once daily), death 2.5 mg RIV+ASA vs ASA alone: HR=0.74;
rivaroxaban alone (5 mg twice P<0.0001
Primary safety end point:
daily), or ASA alone (100 mg
major bleeding 5 mg RIV+ASA alone: HR=0.89; P=0.094
once daily) for 1.95 y
Primary safety end point: 2.5 mg RIV +ASA vs 5
mg RIV vs ASA alone; 3% vs 3% vs 2%
2.5 mg RIV+ASA vs ASA alone: HR=1.66;
P<0.0001
5 mg RIV vs ASA alone: HR=1.51; P<0.0001
COMPASS-Stable Stable PAD After a 30-day run in period, Primary efficacy end point: Primary efficacy end point: 2.5 mg RIV+ASA vs 5
PAD (n= 7470)79 RIV 2.5 mg twice daily + ASA MI, stroke, or cardiovascular mg RIV vs ASA alone: 5% vs 6% vs 7%
100 mg once daily, RIV alone death 2.5 mg RIV+ASA vs ASA alone: HR=0.72; P=0.0047
(5 mg twice daily), or ASA
Primary safety end point: 5 mg RIV+ASA alone: HR=0.86; P=0.19
alone (100 mg once daily)
for 1.95 y major bleeding Primary safety end point=2.5 mg RIV +ASA vs 5
mg RIV vs ASA alone: 3% vs 3% vs 2%
2.5 mg RIV+ASA vs ASA alone: HR=1.61; P=.0089
5 mg RIV+ASA vs ASA alone: HR=1.68; P=0.0043
GEMINI-ACS-1 ACS (<10) RIV 2.5 mg twice daily vs Primary end point= TIMI non- TIMI non-CABG clinically significant bleeding=5%
(n=3037)80 100 mg ASA once daily, in CABG clinically significant vs 5%, HR=1.09; P=0.58
addition to clopidogrel (44%) bleeding Cardiovascular death, MI, stroke, or definite stent
or ticagrelor (56%) (chosen at CV death, MI, stroke, or thrombosis=5% vs 5%, HR=1.06; P=0.73
investigator discretion before definite stent thrombosis
randomization), continued for
median of 291 days

ACS indicates acute coronary syndrome; ASA, acetylsalicylic acid; ATLAS, Anti-Xa Therapy to Lower Cardiovascular Events in Addition to Standard Therapy in Subjects
with Acute Coronary Syndrome; ATLAS ACS-TIMI 46, Anti-Xa Therapy to Lower Cardiovascular Events in Addition to Standard Therapy in Subjects with Acute Coronary
Syndrome- thrombolysis in myocardial infarction 46; 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; CAD, coronary artery disease; COMPASS, Cardiovascular Outcomes for People Using
Anticoagulation Strategies; COMPASS-Stable CAD, Cardiovascular Outcomes for People Using Anticoagulation Strategies- Stable Coronary Artery Disease; COMPASS-
Stable PAD, Cardiovascular Outcomes for People Using Anticoagulation Strategies- Stable Peripheral Artery Disease; CV, cardiovascular; GEMINI-ACS 1, Clinically significant
bleeding with low-dose rivaroxaban versus aspirin, in addition to P2Y12 inhibition, in acute coronary syndromes; HR, hazard ratio; ICH, intracranial hemorrhage; MI,
myocardial infraction; non-CABG, non–coronary artery bypass grafting; PAD, peripheral arterial disease; RIV, rivaroxaban; and TIMI, thrombolysis in myocardial infarction.

2176 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

prolong the international normalized ratio to therapeu- 0.55; P=0.09, respectively) in comparison with placebo.
tic levels, but increased bleeding in comparison with Among patients treated with the lowest doses of riva-

STATE OF THE ART


ASA alone.69 There is no documented evidence of the roxaban, the increase in the risk of thrombolysis in MI
long-term benefit of therapeutic levels of vitamin K an- major bleeding was more pronounced with ASA plus
tagonist therapy in patients with PAD.70 Smaller studies clopidogrel than with ASA monotherapy.76
have demonstrated the potential benefits of warfarin d-Dimer levels may reflect both increased thrombotic
plus ASA versus ASA in patients with ACS.71,72 A meta- activity as well as inflammation. d-Dimer was reported
analysis including 25 307 patients with ACS reported to be a predictor of cardiovascular events in patients
that combination therapy of ASA plus warfarin therapy with PAD, and in patients with acute MI in comparison
(irrespective of international normalized ratio) versus with unstable angina.81,82 In a hypothesis-generating
ASA therapy alone was not associated with less MACE substudy of the ATLAS ACS-TIMI 46 trial, d-dimer levels
(all-cause death, nonfatal MI, and nonfatal thrombo- were measured in 53% of patients enrolled. Elevated
embolic stroke), but was associated with more major baseline d-dimer level was associated with an increased
bleeding (P<0.00001).73 In an analysis of studies with risk of the composite outcome occurrence within 6
international normalized ratio values between 2 and 3, months of the index event, and the administration of
long-term warfarin plus ASA therapy was associated rivaroxaban was persistently associated with lower d-
with fewer major adverse events (P=0.0001) and an dimer levels in comparison with placebo over 6 months,
increased risk of major bleeding (P<0.00001) in com- suggesting the attenuation of in vivo thrombus forma-
parison with ASA alone.73 The unfavorable net clinical tion and inflammation.83
outcome (balancing ischemic events and bleeding) of Based on the outcomes observed in the ATLAS ACS-
full-dose warfarin in combination with ASA has led to TIMI 46 trial, rivaroxaban administered at the low dose of
its disfavor in the long-term treatment of CAD and PAD. 5.0 mg twice daily, and a very low dose of 2.5 mg twice
The inhibition of a single downstream coagulation daily, was therefore chosen and compared with place-
factor, such as factor Xa, rather than multiple factors bo in the phase 3 ATLAS ACS 2-TIMI 51 trial (Anti-Xa
(for example, warfarin) is a new therapeutic strategy Therapy to Lower Cardiovascular Events in Addition to
for long-term therapy for patients with arterial diseas- Standard Therapy in Subjects With Acute Coronary Syn-
es. Some of the advantages of factor Xa inhibitors over drome ACS 2–Thrombolysis In Myocardial Infarction) of
warfarin include a lower risk of intracranial bleeding, patients (n=15 526) with recently stabilized ACS treated
the absence of any significant reported interactions with standard antiplatelet therapy.76,77 Major exclusion
Downloaded from http://ahajournals.org by on June 29, 2023

with food, fewer interactions with medications, and the criteria included history of intracranial hemorrhage or
absence of the need for frequent laboratory monitoring history of either ischemic stroke or transient ischemic
and dose adjustments. However, administration of the attack among patients receiving ASA plus thienopyri-
factor Xa inhibitors, darexaban at ascending doses and dine. All patients were treated with low-dose ASA, and
apixaban at standard doses, was associated with an el- patients were stratified by the administration of a thi-
evated risk for bleeding without a reduction in compos- enopyridine (clopidogrel or ticlopidine). Overall, rivar-
ite ischemic end points when added on top of standard oxaban therapy was associated with a lower rate of the
antiplatelet therapy in patients with ACS.74,75 primary composite end point of cardiovascular death,
The initial clinical evidence to support the benefit of MI, or stroke versus placebo (HR, 0.84; P=0.008), and
a very low factor Xa inhibitor dose on top of antiplate- the primary efficacy end point was lower in both the
let therapy came from the ATLAS ACS studies (Anti-Xa 2.5 mg twice-daily dose (9.1% versus 10.7%; P=0.02)
Therapy to Lower Cardiovascular Events in Addition to and the 5 mg twice-daily dose groups (8.8% versus
ASA With or Without Thienopyridine Therapy in Sub- 10.7%; P=0.03). A decreased incidence of cardiovascu-
jects with Acute Coronary Syndrome) (Table 2).9,76–80 In lar death (2.7% versus 4.1%; P=0.002), death from any
the phase 2 ATLAS ACS-TIMI 46 trial (Rivaroxaban in cause (2.9% versus 4.5%; P=0.002), and also fewer fa-
Combination With Aspirin Alone and With Aspirin and tal bleeding events were observed in the 2.5 mg twice-
a Thienopyridine in Patients With Acute Coronary Syn- daily group in comparison with the 5 mg twice-daily
dromes), patients with stabilized ACS (n=3491) were dose (0.1% versus 0.4%; P=0.04). However, increased
treated with escalating doses of rivaroxaban (5–20 rates of non–coronary artery bypass grafting (CABG)
mg/d) to select the most favorable dose in combination major bleeding (2.1% versus 0.6%; P<0.001) and in-
with ASA or ASA plus a thienopyridine. Clinically sig- tracranial hemorrhage (0.6% versus 0.2%; P=0.009)
nificant bleeding increased in a dose-dependent man- were also associated with rivaroxaban without a sig-
ner (P<0.0001).76 Among patients treated with ASA nificant increase in fatal bleeding (0.3% versus 0.2%;
alone and ASA plus a thienopyridine, the lowest doses P=0.66) or other adverse events. Both doses of rivar-
of rivaroxaban (2.5 mg and 5 mg twice daily) were as- oxaban were associated with a significant reduction in
sociated with a trend toward a reduced risk of death, stent thrombosis. The latter observation may indicate
MI, or stroke (hazard ratio [HR], 0.54; P=0.08 and HR, that stent thrombosis events are not exclusively modu-

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2177


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

lated by platelet P2Y12 and COX-1 pathways and that COMPASS TRIAL DESIGN
thrombin, with its influence on platelet activation, fibrin
STATE OF THE ART

COMPASS was the first randomized clinical trial of a


formation, and stabilization of the platelet-fibrin clot,
non–vitamin K antagonist direct oral anticoagulant in
plays an important role in selected patients. To further
patients with chronic CAD, PAD, or chronic CAD plus
support this observation, apixaban was associated with
PAD (n=27 395). It was planned as an event-driven trial
numerically fewer stent thromboses in the prematurely
designed to continue until at least 2200 patients ex-
terminated APPRAISE-2 trial (Apixaban for Prevention
perienced a primary end point. Chronic CAD was de-
of Acute Ischemic Events 2), and vorapaxar was also
fined as MI within 20 years or multivessel CAD with
demonstrated to reduce stent thrombosis in patients
symptoms or history of stable or unstable angina, or
with ACS.67,75
multivessel percutaneous coronary intervention (PCI)
It is important to note that ATLAS was the first study
or CABG. PAD was defined as percutaneous or surgi-
to demonstrate that the addition of a very low dose
cal revascularization or amputation for arterial insuffi-
of rivaroxaban on top of antiplatelet therapy was as-
ciency, or claudication with either ankle-brachial index
sociated with a reduction in cardiovascular death in
<0.90, stenosis >50%, or carotid revascularization or
stabilized patients with a recent ACS event.77 Thus, in
stenosis >50%. Approximately 18% of the population
patients with stabilized ACS with low risk for bleeding,
had polyvascular disease (CAD+PAD); 73% had CAD
the ATLAS ACS 2 study results suggest that simultane-
alone and 9% had PAD alone. Patients were eligible
ous inhibition of platelet function with ASA and a thi-
for randomization if they completed a 30-day run-in
enopyridine and thrombin generation with a very low
period with rivaroxaban-matched placebo plus ASA
dose of rivaroxaban is effective in reducing cardiovas-
and did not have adverse events, or if they were 4 to
cular events at the cost of significant excess in nonfatal
14 days after CABG surgery. Eligible patients were ran-
bleeding, including intracranial hemorrhage. Patients
domly assigned to 2.5 mg twice-daily rivaroxaban plus
who were taking both ASA and a thienopyridine with
ASA, 5 mg twice-daily rivaroxaban alone, or 100 mg
a previous ischemic stroke or transient ischemic attack
once-daily ASA for a median of 1.95 years.9 Similar
were excluded in ATLAS ACS 2. The latter patients may
to ATLAS, the COMPASS trial excluded patients with
not derive a greater net benefit from more potent an-
stroke within 1 month and also patients with any his-
tithrombotic therapy as previously observed.6,67,84 For
tory of hemorrhagic or lacunar stroke. The primary ef-
example, in the TRITON-TIMI 38 trial (Trial to Assess
ficacy outcome was the composite of cardiovascular
Improvement in Therapeutic Outcomes by Optimiz-
Downloaded from http://ahajournals.org by on June 29, 2023

death, stroke, or MI. The main safety outcome was a


ing Platelet Inhibition with Prasugrel–Thrombolysis in
modification of the International Society on Thrombo-
Myocardial Infarction), patients who had a previous
sis and Haemostasis criteria for major bleeding that
stroke or transient ischemic attack had net harm from
included fatal bleeding, symptomatic bleeding into a
the more potent antithrombotic regimen of prasugrel
critical organ, bleeding into a surgical site requiring
plus ASA versus clopidogrel plus ASA (HR, 1.54; 95%
reoperation, and bleeding that led to hospitalization.
CI, 1.02–2.32; P=0.04).6 The findings of ATLAS ACS 2
Patients were well managed with respect to secondary
stand in contrast with those of the APPRAISE trial that
prevention strategies (eg, 90% received lipid-lowering
implemented full-dose apixaban on top of standard an-
therapies, 71% received angiotensin-converting en-
tiplatelet therapy.75,77
zyme inhibitors or angiotensin receptor blockers, and
The safety of replacing ASA with very-low-dose riva-
70% received β-blockers).9
roxaban on top of clopidogrel or ticagrelor was studied
in patients with stabilized ACS in the GEMINI-ACS-1
trial (A Study to Compare the Safety of Rivaroxaban COMPASS RESULTS
Versus Acetylsalicylic Acid in Addition to Either Clopi-
dogrel or Ticagrelor Therapy in Participants With Acute Overall Population
Coronary Syndrome) (n=3037).80 Non-CABG–related Because of efficacy observed in the very-low-dose riva-
thrombolysis in MI clinically significant bleeding was roxaban plus ASA arm at the first interim analysis, the
similar between groups. Although undersized to de- Data Safety Committee recommended stopping the trial
finitively assess the relation of therapeutic strategy to early with a mean follow-up of 23 months after 1323
ischemic events, there was no efficacy signal for rivar- primary outcome events among 27 395 patients. Very-
oxaban in the overall population or within subgroups. low-dose rivaroxaban plus ASA was associated with a
In a post hoc analysis, the ASA arm had a numerically 24% reduction in MACE in comparison with ASA alone
lower occurrence of the ischemic composite end point (4.1% versus 5.4%; HR, 0.76; P<0.001), whereas riva-
in the first 30 days, suggesting the strong influence roxaban 5 mg twice daily alone did not reduce MACE
of platelet function on thrombosis in this acute time more than ASA alone (4.9% versus 5.4%; HR, 0.90;
period and the importance of inhibiting both COX-1 P=0.12). In comparison with ASA monotherapy, very-
and P2Y12.80,85 low-dose rivaroxaban plus ASA was associated with

2178 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

STATE OF THE ART


HR= 0.76
6 ASA + 2.5mg Rivaroxaban ASA Alone
p<0.001
5.4

4.1
4 HR= 0.58 HR= 0.78 HR= 0.86 HR= 1.70
p<0.001 p=0.02 p=0.14 p<0.001
Events (%)

3.1
3

2.2
1.9 2.2 1.9
2 1.7
1.6

1 0.9

0
MACE Stroke Cardiovascular Myocardial Major
Death Infarction Bleeding
B
7 HR= 0.74
p<0.0001
6
6

5 Figure 3. Clinical outcomes in the COMPASS


trial.
4 HR= 0.50 HR= 0.75 HR= 0.86 HR= 1.66 A, Overall COMPASS cohort. B, COMPASS
Events (%)

4 p<0.0001 p=0.010 p=0.15 p<0.0001 stable coronary artery disease cohort. C,


3 COMPASS peripheral artery disease cohort. ASA
3 indicates acetylsalicylic acid; COMPASS, Cardio-
vascular Outcomes for People Using Anticoagu-
2 2 2 2 2 2 lation Strategies; HR, hazard ratio; and MACE,
2
major adverse cardiovascular events.
Downloaded from http://ahajournals.org by on June 29, 2023

1
1

0
MACE Stroke Cardiovascular Myocardial Major
Death Infarction Bleeding
C HR= 0.72
8
p=0.0047
7
7

6
5
5
Events (%)

HR= 0.54 HR= 0.54 HR= 0.56 HR= 1.61


4 p=0.0054 p=0.0037 p=0.042 p=0.0089

3
3
2 2 2
2
1 1 1 1
1

0
MACE Major Adverse Major Adverse Acute Limb Major
Limb Events Limb Events + Ischemia Bleeding
Major Amputation

lower incidences of ischemic stroke (0.7% versus 1.4%; components of MACE indicated that the risk reduction
HR, 0.51; P<0.001), overall stroke (0.9% versus 1.6%; with very-low-dose rivaroxaban plus ASA was largely
HR, 0.58; P<0.00), all-cause death (3.4% versus 4.1%; driven by a significant reduction in the risk of overall
HR, 0.82; P=0.01), and cardiovascular death (1.7% ver- stroke and cardiovascular death, whereas the rates of MI
sus 2.2%; HR, 0.78; P=0.02). Analysis of the individual were similar among the 3 treatment arms (Figure 3A).9

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2179


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

Overall, major bleeding was higher with very-low- 0.48–1.01), 2 to 5 years (HR, 0.81; 95% CI, 0.59–1.10),
dose rivaroxaban plus ASA versus ASA alone (3.1% ver- and >5 years (HR, 0.72; 95% CI, 0.57–0.91) before en-
STATE OF THE ART

sus 1.9%; HR, 1.70; P<0.001) and was mainly driven rollment. Results were also independent of history of
by more gastrointestinal bleeding events (1.5% versus PCI, although in the subgroup of patients with a his-
0.7%; HR, 2.15; P<0.001); the 1.3% absolute reduction tory of CABG, combination therapy was not beneficial
in MACE by combination therapy was equally offset by (HR, 0.99; 95% CI, 0.77–1.28; versus HR, 0.66; 95%
a 1.2% absolute increase in major bleeding. Rates of CI, 0.56–0.78 in the subgroup without prior CABG; P
fatal bleeding and intracranial hemorrhage were simi- for interaction=0.01). Patients with stable clinical symp-
lar between the combination and ASA treatment arms. toms, ie, in the absence of MI events, PCI, or CABG ≥2
Treatment with 5.0 mg twice-daily rivaroxaban versus years before enrollment, had a benefit with low-dose
ASA alone was associated with a higher rate of major rivaroxaban plus ASA similar to those who had MI, PCI,
bleeding (2.8% versus 1.9%; HR, 1.5; P<0.001), intra- or CABG <2 years before enrollment.78
cranial bleeding (0.5% versus 0.3%; HR, 1.8; P=0.02),
fatal bleeding or symptomatic intracranial hemorrhage
(0.5% versus 0.3%; HR, 1.59; P=0.05), and fatal bleed- PAD Group
ing or symptomatic bleeding into a critical organ (1.0% The COMPASS study included 7470 patients with
versus 0.6%; HR, 1.58; P=0.006). chronic PAD. In addition, ≈65% of these patients had
Very-low-dose rivaroxaban plus ASA resulted in a a history of CAD and ≈7% had a history of stroke. In
20% relative reduction in the risk of the composite net the PAD subgroup, very-low-dose rivaroxaban plus ASA
clinical benefit outcome, which was defined as a com- reduced MACE by 28% versus ASA alone (5.0% versus
posite of cardiovascular death, stroke, MI, fatal bleed- 7.0%; HR, 0.72; P=0.0047), major adverse limb events
ing, or symptomatic bleeding into a critical organ versus (MALE) by 46% (1% versus 2%; HR, 0.54; P=0.0054),
ASA alone (4.7% versus 5.9%; HR, 0.80; P<0.001).9 and MALE plus major amputation by 46% (1.0% ver-
This definition of net clinical outcome did not account sus 2.0%; HR, 0.54; P=0.0037). With respect to safety
for other major bleeding that was approximately twice outcomes, very-low-dose rivaroxaban plus ASA versus
as high in the combination therapy arm as in the ASA ASA alone was associated with a 61% increase in ma-
arm (2.3% versus 1.2%; HR, 1.88; P<0.001). jor bleeding (3.0% versus 2.0%; HR, 1.61; P=0.0089)
that was again mainly driven by a significant increase in
Downloaded from http://ahajournals.org by on June 29, 2023

Stable CAD Group gastrointestinal bleeding (2.0% versus 1.0%; HR, 2.28;
P=0.0027). The composite net clinical benefit outcome
Among 24 842 patients with stable CAD, 69% had a (defined per protocol as MACE, MALE including major
history of MI, with 55% occurring ≥2 years previously amputation, fatal bleeding, and bleeding into a critical
with a mean of 7.1 years. Most patients had undergone organ) occurred in 7% of patients treated with very-
revascularization (CABG 32%; PCI 60%). Very-low-dose low-dose rivaroxaban plus ASA and 9% of patients
rivaroxaban plus ASA reduced MACE by 26% (4.0% ver- treated with ASA alone (HR, 0.72; 95% CI, 0.59–0.87;
sus 6.0%; HR, 0.74; P<0.0001), an effect that was driven P=0.0008), a prevention of 27 MACE or MALE events
by significant reductions in stroke and death without an including major amputations for every 1000 patients
influence on MI. Low-dose rivaroxaban alone versus ASA treated over a 21-month period at the expense of 1
alone was associated with a nonsignificant reduction in fatal and 1 critical organ bleeding event (Figure 3C).79
MACE (5.0% versus 6.0%; HR, 0.89, P=0.094). Similar
to the overall trial population, the 66% increase in the
major bleeding associated with very-low-dose rivaroxa- INTERPRETATION OF THE COMPASS
ban plus ASA versus ASA alone (3.0% versus 2.0%; HR,
1.66; P<0.0001) was mainly driven by gastrointestinal
TRIAL
bleeding (2.0% versus 1.0%; HR, 2.13; P<0.0001) (Fig- The COMPASS study demonstrated favorable anti-
ure 3B). The net clinical outcome (defined as cardiovas- thrombotic effects from the combination of low-dose
cular death, stroke, MI, fatal bleeding, or symptomatic ASA and very-low-dose rivaroxaban. In contrast, rivar-
bleeding into a critical organ) occurred in 5% of patients oxaban alone at 5 mg twice daily had similar effects on
receiving very-low-dose rivaroxaban plus ASA versus the primary outcome as ASA but with more bleeding.
6.0% with ASA alone (HR, 0.78; P=0.0003).78 Absolute reduction in MACE occurrence was numeri-
In an exploratory analysis, the reduction in MACE cally greatest in those with polyvascular disease: abso-
with very-low-dose rivaroxaban plus ASA versus ASA lute risk reduction, 2.7% versus 1.0% versus 0.4%, and
alone was consistent in patients with and without relative risk reduction, 33% versus 23% versus 11%
prior MI (HR, 0.74; 95% CI, 0.63–0.88; and HR, 0.76; for the CAD+PAD, CAD alone, and PAD alone groups,
95%% CI, 0.58–0.98; P for interaction=0.91) and respectively.9,78,86 The relative reduction in MACE and
patients with a prior MI <2 years (HR, 0.70; 95% CI, MALE events was also numerically greatest in patients

2180 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

with CAD+PAD versus PAD alone: 35% versus 18%.79 alone and is supported by a comparison of the rivar-
These data should be considered hypothesis-generating oxaban alone arm versus ASA, where there were fewer

STATE OF THE ART


in support of a stronger benefit in patients with polyvas- ischemic strokes with rivaroxaban (HR, 0.69; P=0.006)
cular disease. Similar benefits were observed in patients but at the cost of more hemorrhagic strokes (HR, 2.7;
with polyvascular disease from ticagrelor plus ASA ther- P=0.005). Low-dose ASA may attenuate platelet acti-
apy in a subanalysis of the PEGASUS-TIMI 54 trial (Pre- vation and resultant formation of the prothrombinase
vention of Cardiovascular Events in Patients with Prior complex. Very-low-dose rivaroxaban may synergize the
Heart Attack Using Ticagrelor Compared to Placebo on attenuation of initial thrombin generation as described
a Background of Aspirin–Thrombolysis in Myocardial earlier.
Infarction 54); ticagrelor plus ASA significantly reduced Thus, after vessel wall injury, the simultaneous in-
MACE and MALE in comparison with ASA monothera- hibition of platelet activation by low-dose aspirin that
py.87 At this time, the optimal antithrombotic treatment attenuates prothrombinase complex formation and
strategy for patients with PAD remains unresolved. low-dose rivaroxaban that inhibits factor Xa within the
The COMPASS findings are particularly noteworthy complex may effectively blunt the initial amplification
because they apply to a population already receiving a of thrombin generation required for thrombus forma-
high level of adjunctive secondary prevention strategies. tion while at the same time preserve endogenous he-
An approximate halving of the rate of ischemic stroke mostasis pathways. Finally, noncanonical effects of fac-
by dual pathway inhibition versus ASA monotherapy tor Xa inhibition on inflammation, vascular remodeling,
was the major event driving the significant reduction atherosclerotic plaque progression, and tissue fibrosis
in MACE (see below). It is interesting to note that a and ASA may also modulate the occurrence of ischemic
definite effect on reduction in MI was not observed by stroke (see above). At this time, the type of stroke (ie,
combination therapy, suggesting potential differences lacunar, large vessel, cardioembolic) has not yet been
in the pathways modulating these clinical events. The reported by the COMPASS investigators. The stroke
benefit of combination therapy was present irrespective findings of COMPASS contrast with the effects of DAPT
of prior MI, the time of enrollment in relation to prior versus ASA,8,9,88 clopidogrel monotherapy versus ASA
MI occurrence, and the performance of PCI. Although plus clopidogrel,64 or potent DAPT versus weak DAPT
underpowered to make a firm conclusion, patients who strategies6,7 on ischemic stroke, where no differences
underwent CABG did not appear to benefit from dual were observed.
pathway inhibition. It is notable that the efficacy ad- In the recently published COMMANDER-HF trial (A
Downloaded from http://ahajournals.org by on June 29, 2023

vantage comes at a cost of increased major bleeding, Study to Assess the Effectiveness and Safety of Riva-
but not fatal bleeding or intracranial hemorrhage, and roxaban in Reducing the Risk of Death, Myocardial
bleeding is mostly in the gastrointestinal tract. It should Infarction, or Stroke in Participants with Heart Failure
be noted that patients with a high risk for bleeding and and Coronary Artery Disease Following an Episode of
stroke within 1 month or any history of hemorrhagic Decompensated Heart Failure) of patients with recent
or lacunar stroke were excluded from the COMPASS worsening of chronic heart failure, reduced ejection
trial. Therefore, the optimal patients for dual pathway fraction, and coronary artery disease, a strategy of very-
inhibition are likely those with chronic disease who are low-dose rivaroxaban on top of standard therapy (93%
at highest risk for recurrent ischemic events. Such pa- ASA and 35% ASA+thienopyridine) was not associated
tients would include those with polyvascular disease or with a reduction in MACE (death from any cause, MI,
symptomatic PAD, and patients with CAD who had ad- or stroke) in comparison with placebo, but, similar to
ditional risk factors. At the same time, patients at high the results of COMPASS, was associated with a lower
risk for bleeding such as the elderly, and patients with stroke rate.9,89 Most outcomes in COMMANDER-HF
prior bleeding, advanced kidney disease, anemia, and were death events, and, in this heart failure population,
low body weight may not be suitable candidates. the mechanism for death may not be strongly influ-
It is important to note that the clinical benefit of enced by thrombin generation and platelet activation.
adding very-low-dose rivaroxaban to ASA was present In summary, the results of COMPASS support the
irrespective of the length of the history of CAD in the concept of the enhancement of antithrombotic effects
COMPASS trial. This is in contrast to results from the by combining blockade of COX-1 and a very low dose
PEGASUS-TIMI 54 trial, where ischemic event reduction of a factor Xa inhibitor in patients with stable ath-
with ticagrelor plus low-dose ASA was not observed in erosclerotic vascular disease, in particular, those with
patients who had been stable for ≥1 years on single polyvascular disease. Very-low-dose rivaroxaban still
antiplatelet therapy.87 provides substantial inhibition of ex vivo thrombin gen-
The antithrombotic benefit of combination therapy eration.90 This potentiation may be based on the com-
on ischemic stroke reduction may, in part, be explained bined antithrombotic action of the drugs: inhibition of
by the benefit of an anticoagulant to reduce cardio- platelet function and coagulation. However, other po-
embolic stroke in comparison with antiplatelet therapy tential effects of factor Xa blockade and COX-1 block-

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2181


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

ade mediated through noncanonical pathways should Challenges in the uptake of dual pathway inhibition
be considered, and are discussed below. as administered in COMPASS into clinical practice will
STATE OF THE ART

The European Medicines Agency has approved (1) focus on the careful selection of patients and imple-
rivaroxaban 2.5 mg twice daily with ASA alone or with mentation of a multidisciplinary approach involving
DAPT for the secondary prevention of atherothrombotic stratification of competing risks of thrombosis and
events in adult patients after an ACS event with elevat- bleeding. Those most likely to benefit would include
ed cardiac biomarkers and (2) rivaroxaban 2.5 mg twice patients at high risk for recurrent ischemia with the
daily with ASA for the prevention of atherothrombotic following characteristics: polyvascular disease, symp-
events in adult patients with CAD or symptomatic PAD tomatic PAD, and additional risk factors. The ongoing
at high risk of ischemic events.91 In addition, rivaroxa- VOYAGER study (Efficacy and Safety of Rivaroxaban in
ban 2.5 mg twice daily in combination with low-dose Reducing the Risk of Major Thrombotic Vascular Events
ASA was approved to reduce the risk of MACE in pa- in Subjects With Symptomatic Peripheral Artery Disease
tients with CAD/PAD by the US Food and Drug Admin- Undergoing Peripheral Revascularization Procedures of
istration in 2018.86 the Lower Extremities) will provide further information
in the PAD population.92 Finally, a remaining challenge
is when to transition patients with high-risk ACS from
CONCLUSIONS DAPT to dual pathway inhibition. Because guidelines
A basic pathophysiological construct is that thrombus recommend DAPT for at least 1 year in patients with
formation at the site of atherosclerotic plaque rupture is ACS, this may be an appropriate time to make the
mediated by the function of platelets and coagulation. switch, but trials are needed to examine this possibil-
The totality of clinical evidence is directly in line with the ity. Still unknown is whether biomarkers of platelet-
basic data and demonstrates that, in the setting of early fibrin clot generation properties can help to identify
administration for secondary prevention, the blockade patients at high thrombotic risk who would derive the
of 2 key pathways that amplify platelet activation, COX- greatest net clinical benefit from dual pathway inhibi-
1 and the P2Y12 receptor, provides enhanced inhibition tion or whether new anticoagulant strategies such as
of coronary arterial thrombosis and reduced MACE. The those targeting factor XI will provide a safer platform
benefit of a very-low-dose factor Xa inhibitor in this on which to add antiplatelet therapy.93
time period, as in ATLAS, was observed when added to
Downloaded from http://ahajournals.org by on June 29, 2023

clopidogrel, a weak P2Y12 inhibitor, and ASA. This ad-


ARTICLE INFORMATION
ditional benefit likely reflects the combined attenuation
of platelet activation and thrombin generation, 2 major Correspondence
modulators of atherothrombosis. In contrast, when full- Paul A. Gurbel, MD, FAHA, Director, Interventional Cardiology and Cardiovas-
cular Medicine Research, Inova Center for Thrombosis Research and Drug De-
dose apixaban was added on top of DAPT in ACS, there velopment, Inova Heart and Vascular Institute, 3300 Gallows Rd, Falls Church,
was no significant reduction in MACE, whereas there VA 22042. Email Paul.Gurbel@inova.org
was a significant increase in major bleeding. Therefore,
the selection of the appropriate anticoagulant and the Affiliations
appropriate dose requires careful balancing of benefits Inova Center for Thrombosis Research and Drug Development, Inova Heart and
and risks. Vascular Institute, Falls Church, VA (P.A.G.). Sinai Center for Thrombosis Re-
search, Sinai Hospital of Baltimore, MD (U.S.T.). British Heart Foundation Centre
The success of combined antiplatelet and antico-
for Cardiovascular Sciences, University of Edinburgh, United Kingdom (K.A.A.F.).
agulant therapy is critically dependent on the safety Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal
profile of the anticoagulant. For example, therapeutic Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maas-
tricht University Medical Center, Netherlands (H.t.C.). Thrombosis and Athero-
doses of warfarin reduced recurrent ischemic events
sclerosis Research Institute and McMaster University, Hamilton, Canada (J.I.W.).
after MI, but this effect was offset by an increase in se-
rious bleeding. In the chronic disease phase, the COM- Acknowledgments
PASS trial revealed a reduction in MACE with a com-
We are grateful to Mr Patrick Lane for Figures 1 and 2.
bination of very-low-dose rivaroxaban plus ASA, dual
pathway inhibition, in patients with CAD and PAD that Disclosures
was largely driven by a reduction in ischemic stroke fol-
Dr Gurbel reports receiving grants from the National Institutes of Health, Bayer,
lowed by cardiovascular death. The enhanced clinical Medicure, Instrumentation Labs, Haemonetics, Amgen, Idorsia, Ionis, Haemon-
effect of the latter strategy, which includes a reduction etics, Janssen, and Merck; receiving honoraria and payment for lectures, con-
in MALE in the PAD population, may be explained by sultations including service on speakers’ bureaus from Bayer, Janssen, Merck,
UptoDate, and Medicure; and holding patents in the area of personalized an-
direct potentiation of antithrombotic effects. In addi- tiplatelet therapy and interventional cardiology. Dr Fox received grants from
tion, noncanonical effects modulating inflammation Bayer, Janssen, and AstraZeneca and is a consultant for Bayer, Janssen, Astra-
and reparative processes may play a role. Increased Zeneca, Sanofi/Regeneron, and Verseon. Dr Tantry reports receiving honoraria
from AstraZeneca, UptoDate, and Medicure. Dr Ten Cate received research sup-
bleeding may also be explained by potentiation of an- port from Bayer, Pfizer, and Leo Farma; performed consultancy for Stago; and
tithrombotic effects. was on the advisory board for Bayer and Pfizer. Dr Ten Cate received noncom-

2182 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

mercial support from the Dutch Heart Foundation (RACE-5 and CONTRAST). Dr 15. Ardissino D, Merlini PA, Bauer KA, Galvani M, Ottani F, Franchi F,
Ten Cate is the chair of the board of the Dutch Federation of Anticoagulation Bertocchi F, Rosenberg RD, Mannucci PM. Coagulation activation and

STATE OF THE ART


Clinics. Dr Weitz is a consultant for and has received honoraria from Bayer, long-term outcome in acute coronary syndromes. Blood. 2003;102:2731–
Boehringer Ingelheim, Bristol-Myers Squibb, Daiichi-Sankyo, Ionis, Janssen, 2735. doi: 10.1182/blood-2002-03-0954
Merck, Novartis, Pfizer, and Portola. 16. Merlini PA, Bauer KA, Oltrona L, Ardissino D, Cattaneo M, Belli C,
Mannucci PM, Rosenberg RD. Persistent activation of coagulation
mechanism in unstable angina and myocardial infarction. Circulation.
1994;90:61–68.
REFERENCES 17. Undas A, Szułdrzyński K, Brummel-Ziedins KE, Tracz W, Zmudka K, Mann KG.
1. communitymedicine4asses. WHO updates fact sheet on Cardiovascular Systemic blood coagulation activation in acute coronary syndromes.
diseases. May 18, 2017. https://communitymedicine4asses.wordpress. Blood. 2009;113:2070–2078. doi: 10.1182/blood-2008-07-167411
com/2017/05/19/who-updates-fact-sheet-on-cardiovascular-diseases- 18. Becker RC, Bovill EG, Corrao JM, Ball SP, Ault K, Mann KG, Tracy RP.
18-may-2017/. Accessed on December 20, 2018. Dynamic nature of thrombin generation, fibrin formation, and platelet
2. Rapsomaniki E, Thuresson M, Yang E, Blin P, Hunt P, Chung SC, activation in unstable angina and non-q-wave myocardial infarction.
Stogiannis D, Pujades-Rodriguez M, Timmis A, Denaxas SC, Danchin N, J Thromb Thrombolysis. 1995;2:57–64.
Stokes M, Thomas-Delecourt F, Emmas C, Hasvold P, Jennings E, Johansson S, 19. Ten Cate H, Hemker HC. Thrombin generation and atherothrombosis:
Cohen DJ, Jernberg T, Moore N, Janzon M, Hemingway H. Using big data what does the evidence indicate? J Am Heart Assoc. 2016;5:e003553.
from health records from four countries to evaluate chronic disease out- doi: 10.1161/JAHA.116.003553.
comes: a study in 114 364 survivors of myocardial infarction. Eur Heart J 20. Merlini PA, Ardissino D, Bauer KA, Oltrona L, Pezzano A, Bottasso B,
Qual Care Clin Outcomes. 2016;2:172–183. doi: 10.1093/ehjqcco/qcw004 Rosenberg RD, Mannucci PM. Persistent thrombin generation during hep-
3. Abtan J, Bhatt DL, Elbez Y, Sorbets E, Eagle K, Reid CM, Baumgartner I, arin therapy in patients with acute coronary syndromes. Arterioscler
Wu D, Hanson ME, Hannachi H, Singhal PK, Steg PG, Ducrocq G; REACH Thromb Vasc Biol. 1997;17:1325–1330.
Registry Investigators. Geographic variation and risk factors for systemic 21. Rho R, Tracy RP, Bovill EG, Ball SP, Becker RC. Plasma markers of proco-
and limb ischemic events in patients with symptomatic peripheral artery agulant activity among individuals with coronary artery disease. J Thromb
disease: insights from the REACH Registry. Clin Cardiol. 2017;40:710– Thrombolysis. 1995;2:239–243.
718. doi: 10.1002/clc.22721 22. Borissoff JI, Joosen IA, Versteylen MO, Spronk HM, ten Cate H, Hofstra L.
4. Sigvant B, Hasvold P, Kragsterman B, Falkenberg M, Johansson S, Accelerated in vivo thrombin formation independently predicts the pres-
Thuresson M, Nordanstig J. Cardiovascular outcomes in patients with pe- ence and severity of CT angiographic coronary atherosclerosis. JACC Car-
ripheral arterial disease as an initial or subsequent manifestation of ath- diovasc Imaging. 2012;5:1201–1210. doi: 10.1016/j.jcmg.2012.01.023
erosclerotic disease: results from a Swedish nationwide study. J Vasc Surg. 23. Tatsumi K, Mackman N. Tissue factor and atherothrombosis. J Atheroscler
2017;66:507–514.e1. doi: 10.1016/j.jvs.2017.01.067 Thromb. 2015;22:543–549. doi: 10.5551/jat.30940
5. Gurbel PA, Tantry US. Combination antithrombotic therapies. Circulation. 24. Fender AC, Rauch BH, Geisler T, Schrör K. Protease-activated receptor PAR-
2010;121:569–583. doi: 10.1161/CIRCULATIONAHA.109.853085 4: an inducible switch between thrombosis and vascular inflammation?
6. Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Thromb Haemost. 2017;117:2013–2025. doi: 10.1160/TH17-03-0219
Gottlieb S, Neumann FJ, Ardissino D, De Servi S, Murphy SA, Riesmeyer J, 25. Narula N, Dannenberg AJ, Olin JW, Bhatt DL, Johnson KW, Nadkarni G,
Weerakkody G, Gibson CM, Antman EM; TRITON-TIMI 38 Investigators. Min J, Torii S, Poojary P, Anand SS, Bax JJ, Yusuf S, Virmani R, Narula J.
Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Pathology of peripheral artery disease in critical limb ischemia. J Am Coll
Engl J Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482 Cardiol. 2018;72:2152–2163. doi: 10.1016/j.jacc.2018.08.002.
Downloaded from http://ahajournals.org by on June 29, 2023

7. Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, 26. Rajagopalan S, Mckay I, Ford I, Bachoo P, Greaves M, Brittenden J. Platelet
Horrow J, Husted S, James S, Katus H, Mahaffey KW, Scirica BM, Skene A, activation increases with the severity of peripheral arterial disease: im-
Steg PG, Storey RF, Harrington RA, Freij A, Thorsén M; PLATO Investigators. plications for clinical management. J Vasc Surg. 2007;46:485–490. doi:
Ticagrelor versus clopidogrel in patients with acute coronary syndromes. 10.1016/j.jvs.2007.05.039
N Engl J Med. 2009;361:1045–1057. doi: 10.1056/NEJMoa0904327 27. van Geffen JP, Kleinegris MC, Verdoold R, Baaten CC, Cosemans JM,
8. Bonaca MP, Bhatt DL, Cohen M, Steg PG, Storey RF, Jensen EC, Magnani G, Clemetson KJ, Ten Cate H, Roest M, de Laat B, Heemskerk JW. Normal plate-
Bansilal S, Fish MP, Im K, Bengtsson O, Oude Ophuis T, Budaj A, Theroux P, let activation profile in patients with peripheral arterial disease on aspirin.
Ruda M, Hamm C, Goto S, Spinar J, Nicolau JC, Kiss RG, Murphy SA, Thromb Res. 2015;135:513–520. doi: 10.1016/j.thromres.2014.12.029
Wiviott SD, Held P, Braunwald E, Sabatine MS; PEGASUS-TIMI 54 Steer- 28. Tan KT, Tayebjee MH, Lynd C, Blann AD, Lip GY. Platelet microparticles
ing Committee and Investigators. Long-term use of ticagrelor in patients and soluble P selectin in peripheral artery disease: relationship to extent of
with prior myocardial infarction. N Engl J Med. 2015;372:1791–1800. doi: disease and platelet activation markers. Ann Med. 2005;37:61–66.
10.1056/NEJMoa1500857 29. Cassar K, Bachoo P, Ford I, Greaves M, Brittenden J. Markers of coagula-
9. Eikelboom JW, Connolly SJ, Bosch J, Dagenais GR, Hart RG, Shestakovska O, tion activation, endothelial stimulation and inflammation in patients with
Diaz R, Alings M, Lonn EM, Anand SS, Widimsky P, Hori M, Avezum A, peripheral arterial disease. Eur J Vasc Endovasc Surg. 2005;29:171–176.
Piegas LS, Branch KRH, Probstfield J, Bhatt DL, Zhu J, Liang Y, Maggioni AP, doi: 10.1016/j.ejvs.2004.11.001
Lopez-Jaramillo P, O’Donnell M, Kakkar AK, Fox KAA, Parkhomenko AN, 30. Ren M, Li R, Chen N, Pang N, Li Y, Deng X, Wang L, Luo M,
Ertl G, Störk S, Keltai M, Ryden L, Pogosova N, Dans AL, Lanas F, Commerford PJ, Liu Y, Hao H, Liu Y, Sun H, Wu J. Platelet-derived factor v is a critical
Torp-Pedersen C, Guzik TJ, Verhamme PB, Vinereanu D, Kim JH, Tonkin AM, mediator of arterial thrombosis. J Am Heart Assoc. 2017;6:e006345. doi:
Lewis BS, Felix C, Yusoff K, Steg PG, Metsarinne KP, Cook Bruns N, 10.1161/JAHA.117.006345.
Misselwitz F, Chen E, Leong D, Yusuf S; COMPASS Investigators. Rivaroxa- 31. Monković DD, Tracy PB. Functional characterization of human platelet-re-
ban with or without aspirin in stable cardiovascular disease. N Engl J Med. leased factor V and its activation by factor Xa and thrombin. J Biol Chem.
2017;377:1319–1330. doi: 10.1056/NEJMoa1709118 1990;265:17132–17140.
10. Ross R, Glomset J, Harker L. Response to injury and atherogenesis. Am J 32. Esmon CT. What did we learn from new oral anticoagulant treat-
Pathol. 1977;86:675–684. ment? Thromb Res. 2012;130(suppl 1):S41–S43. doi: 10.1016/j.
11. Borissoff JI, Spronk HM, ten Cate H. The hemostatic system as a mod- thromres.2012.08.271
ulator of atherosclerosis. N Engl J Med. 2011;364:1746–1760. doi: 33. Leon C, Alex M, Klocke A, Morgenstern E, Moosbauer C, Eckly A,
10.1056/NEJMra1011670 Spannagl M, Gachet C, Engelmann B. Platelet ADP receptors contribute
12. Tantry US, Bliden KP, Suarez TA, Kreutz RP, Dichiara J, Gurbel PA. Hyper- to the initiation of intravascular coagulation. Blood. 2004;103:594–600.
coagulability, platelet function, inflammation and coronary artery disease doi: 10.1182/blood-2003-05-1385
acuity: results of the Thrombotic RIsk Progression (TRIP) study. Platelets. 34. Gurbel PA, Bliden KP, Antonino MJ, Gesheff T, Cummings CC, Dubois BV,
2010;21:360–367. doi: 10.3109/09537100903548903 Herzog WR, Tantry US. Time dependence of clopidogrel loading effect:
13. Weitz JI. Insights into the role of thrombin in the pathogenesis of re- platelet activation versus platelet aggregation. Thromb Res. 2012;129:1–
current ischaemia after acute coronary syndrome. Thromb Haemost. 2. doi: 10.1016/j.thromres.2011.07.048
2014;112:924–931. doi: 10.1160/TH14-03-0265 35. Gurbel PA, Bliden KP, Guyer K, Aggarwal N, Tantry US. Delayed throm-
14. Mann KG, Brummel K, Butenas S. What is all that thrombin for? J Thromb bin-induced platelet-fibrin clot generation by clopidogrel: a new dose-
Haemost. 2003;1:1504–1514. related effect demonstrated by thrombelastography in patients under-

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2183


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

going coronary artery stenting. Thromb Res. 2007;119:563–570. doi: Mackman N, van der Meijden P, Meijers JCM, Heestermans M, Renné T,
10.1016/j.thromres.2006.05.006 Dólleman S, Chayouâ W, Ariëns RAS, Baaten CC, Nagy M, Kuliopulos A,
STATE OF THE ART

36. Judge HM, Buckland RJ, Sugidachi A, Jakubowski JA, Storey RF. The active Posma JJ, Harrison P, Vries MJ, Crijns HJGM, Dudink EAMP, Buller HR,
metabolite of prasugrel effectively blocks the platelet P2Y12 receptor and Henskens YMC, Själander A, Zwaveling S, Erküner O, Eikelboom JW,
inhibits procoagulant and pro-inflammatory platelet responses. Platelets. Gulpen A, Peeters FECM, Douxfils J, Olie RH, Baglin T, Leader A, Schotten U,
2008;19:125–133. doi: 10.1080/09537100701694144 Scaf B, van Beusekom HMM, Mosnier LO, van der Vorm L, Declerck P,
37. Behan MW, Fox SC, Heptinstall S, Storey RF. Inhibitory effects of P2Y12 Visser M, Dippel DWJ, Strijbis VJ, Pertiwi K, Ten Cate-Hoek AJ, Ten Cate H.
receptor antagonists on TRAP-induced platelet aggregation, procoagu- Atherothrombosis and thromboembolism: position paper from the Sec-
lant activity, microparticle formation and intracellular calcium responses ond Maastricht Consensus Conference on Thrombosis. Thromb Haemost.
in patients with acute coronary syndromes. Platelets. 2005;16:73–80. doi: 2018;118:229–250.
10.1080/09537100400005634 54. Rana R, Huang T, Koukos G, Fletcher EK, Turner SE, Shearer A, Gurbel PA,
38. Gurbel PA, Bliden KP, Navickas I, Cohen E, Tantry US. Post-coro- Rade JJ, Kimmelstiel CD, Bliden KP, Covic L, Kuliopulos A. Noncanoni-
nary intervention recurrent ischemia in the presence of adequate cal matrix metalloprotease 1-protease-activated receptor 1 signaling
platelet inhibition by dual antiplatelet therapy: what are we over- drives progression of atherosclerosis. Arterioscler Thromb Vasc Biol.
looking? J Thromb Haemost. 2007;5:2300–2301. doi: 10.1111/j. 2018;38:1368–1380. doi: 10.1161/ATVBAHA.118.310967
1538-7836.2007.02749.x 55. Borissoff JI, Heeneman S, Kilinç E, Kassák P, Van Oerle R, Winckers K,
39. Kossmann S, Lagrange J, Jäckel S, Jurk K, Ehlken M, Schönfelder T, Govers-Riemslag JW, Hamulyák K, Hackeng TM, Daemen MJ, ten Cate H,
Weihert Y, Knorr M, Brandt M, Xia N, Li H, Daiber A, Oelze M, Reinhardt C, Spronk HM. Early atherosclerosis exhibits an enhanced procoagulant
Lackner K, Gruber A, Monia B, Karbach SH, Walter U, Ruggeri ZM, Renné T, state. Circulation. 2010;122:821–830. doi: 10.1161/CIRCULATIONAHA.
Ruf W, Münzel T, Wenzel P. Platelet-localized FXI promotes a vascular 109.907121
coagulation-inflammatory circuit in arterial hypertension. Sci Transl Med. 56. Riewald M, Ruf W. Mechanistic coupling of protease signaling and
2017;9:eaah4923. initiation of coagulation by tissue factor. Proc Natl Acad Sci U S A.
40. Puy C, Rigg RA, McCarty OJ. The hemostatic role of factor XI. Thromb Res. 2001;98:7742–7747. doi: 10.1073/pnas.141126698
2016;141(suppl 2):S8–S11. doi: 10.1016/S0049-3848(16)30354-1 57. Joo SS, Won TJ, Kim JS, Yoo YM, Tak ES, Park SY, Park HY, Hwang KW,
41. Undas A, Brummel K, Musial J, Mann KG, Szczeklik A. Blood coagula- Park SC, Lee DI. Inhibition of coagulation activation and inflammation by
tion at the site of microvascular injury: effects of low-dose aspirin. Blood. a novel factor Xa inhibitor synthesized from the earthworm Eisenia andrei.
2001;98:2423–2431. Biol Pharm Bull. 2009;32:253–258.
42. Undas A, Brummel-Ziedins KE, Mann KG. Antithrombotic properties of as- 58. Ragosta M, Gimple LW, Gertz SD, Dunwiddie CT, Vlasuk GP, Haber HL,
pirin and resistance to aspirin: beyond strictly antiplatelet actions. Blood. Powers ER, Roberts WC, Sarembock IJ. Specific factor Xa inhibition reduc-
2007;109:2285–2292. doi: 10.1182/blood-2006-01-010645 es restenosis after balloon angioplasty of atherosclerotic femoral arteries
43. Perzborn E, Heitmeier S, Laux V. Effects of rivaroxaban on platelet ac- in rabbits. Circulation. 1994;89:1262–1271.
tivation and platelet-coagulation pathway interaction: in vitro and 59. Bliden KP, Tantry US, Gesheff MG, Franzese CJ, Pandya S, Toth PP,
in vivo studies. J Cardiovasc Pharmacol Ther. 2015;20:554–562. doi: Mathew DP, Chaudhary R, Gurbel PA. Thrombin-induced platelet-fibrin
10.1177/1074248415578172 clot strength identified by thrombelastography: a novel prothrombotic
44. Gurbel PA, Kuliopulos A, Tantry US. G-protein-coupled receptors signaling marker of coronary artery stent restenosis. J Interv Cardiol. 2016;29:168–
pathways in new antiplatelet drug development. Arterioscler Thromb Vasc 178. doi: 10.1111/joic.12277
Biol. 2015;35:500–512. doi: 10.1161/ATVBAHA.114.303412. 60. Gurbel PA, Bliden KP, Guyer K, Cho PW, Zaman KA, Kreutz RP, Bassi AK,
45. Wihlborg AK, Wang L, Braun OO, Eyjolfsson A, Gustafsson R, Tantry US. Platelet reactivity in patients and recurrent events post-stent-
Downloaded from http://ahajournals.org by on June 29, 2023

Gudbjartsson T, Erlinge D. ADP receptor P2Y12 is expressed in vascu- ing: results of the PREPARE POST-STENTING Study. J Am Coll Cardiol.
lar smooth muscle cells and stimulates contraction in human blood 2005;46:1820–1826. doi: 10.1016/j.jacc.2005.07.041
vessels. Arterioscler Thromb Vasc Biol. 2004;24:1810–1815. doi: 61. Ellinghaus P, Laux V, Perzborn E. Effect of rivaroxaban on thrombin-in-
10.1161/01.ATV.0000142376.30582.ed duced pro-inflammatory gene expression in human umbilical vein endo-
46. Heim C, Gebhardt J, Ramsperger-Gleixner M, Jacobi J, Weyand M, thelial cells. J Thromb Haemost. 2011;9:491.
Ensminger SM. Clopidogrel significantly lowers the development of ath- 62. Zhou Q, Bea F, Preusch M, Wang H, Isermann B, Shahzad K, Katus HA,
erosclerosis in ApoE-deficient mice in vivo. Heart Vessels. 2016;31:783– Blessing E. Evaluation of plaque stability of advanced atherosclerotic
794. doi: 10.1007/s00380-015-0696-7 lesions in apo E-deficient mice after treatment with the oral factor Xa
47. Nylander S, Femia EA, Scavone M, Berntsson P, Asztély AK, Nelander K, inhibitor rivaroxaban. Mediators Inflamm. 2011;2011:432080. doi:
Löfgren L, Nilsson RG, Cattaneo M. Ticagrelor inhibits human platelet 10.1155/2011/432080
aggregation via adenosine in addition to P2Y12 antagonism. J Thromb 63. Esmon CT. Targeting factor Xa and thrombin: impact on coagulation
Haemost. 2013;11:1867–1876. doi: 10.1111/jth.12360 and beyond. Thromb Haemost. 2014;111:625–633. doi: 10.1160/
48. Bonello L, Laine M, Kipson N, Mancini J, Helal O, Fromonot J, Gariboldi V, TH13-09-0730
Condo J, Thuny F, Frere C, Camoin-Jau L, Paganelli F, Dignat-George F, 64. CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel
Guieu R. Ticagrelor increases adenosine plasma concentration in patients versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet.
with an acute coronary syndrome. J Am Coll Cardiol. 2014;63:872–877. 1996;348:1329–1339.
doi: 10.1016/j.jacc.2013.09.067 65. Bhatt DL, Fox KA, Hacke W, Berger PB, Black HR, Boden WE, Cacoub P,
49. van Giezen JJ, Sidaway J, Glaves P, Kirk I, Björkman JA. Ticagrelor inhibits Cohen EA, Creager MA, Easton JD, Flather MD, Haffner SM, Hamm CW,
adenosine uptake in vitro and enhances adenosine-mediated hyperemia Hankey GJ, Johnston SC, Mak KH, Mas JL, Montalescot G, Pearson TA,
responses in a canine model. J Cardiovasc Pharmacol Ther. 2012;17:164– Steg PG, Steinhubl SR, Weber MA, Brennan DM, Fabry-Ribaudo L, Booth J,
172. doi: 10.1177/1074248411410883 Topol EJ; CHARISMA Investigators. Clopidogrel and aspirin versus aspi-
50. Spronk HM, de Jong AM, Crijns HJ, Schotten U, Van Gelder IC, rin alone for the prevention of atherothrombotic events. N Engl J Med.
Ten Cate H. Pleiotropic effects of factor Xa and thrombin: what to ex- 2006;354:1706–1717. doi: 10.1056/NEJMoa060989
pect from novel anticoagulants. Cardiovasc Res. 2014;101:344–351. doi: 66. Hiatt WR, Fowkes FG, Heizer G, Berger JS, Baumgartner I, Held P,
10.1093/cvr/cvt343 Katona BG, Mahaffey KW, Norgren L, Jones WS, Blomster J, Millegård M,
51. Borensztajn K, Peppelenbosch MP, Spek CA. Factor Xa: at the crossroads Reist C, Patel MR; EUCLID Trial Steering Committee and Investigators. Ti-
between coagulation and signaling in physiology and disease. Trends Mol cagrelor versus clopidogrel in symptomatic peripheral artery disease. N
Med. 2008;14:429–440. doi: 10.1016/j.molmed.2008.08.001 Engl J Med. 2017;376:32–40. doi: 10.1056/NEJMoa1611688
52. Shimizu T, Nishihira J, Watanabe H, Abe R, Honda A, Ishibashi T, Shimizu H. 67. Morrow DA, Braunwald E, Bonaca MP, Ameriso SF, Dalby AJ, Fish MP,
Macrophage migration inhibitory factor is induced by thrombin and fac- Fox KA, Lipka LJ, Liu X, Nicolau JC, Ophuis AJ, Paolasso E, Scirica BM,
tor Xa in endothelial cells. J Biol Chem. 2004;279:13729–13737. doi: Spinar J, Theroux P, Wiviott SD, Strony J, Murphy SA; TRA 2P–TIMI 50
10.1074/jbc.M400150200 Steering Committee and Investigators. Vorapaxar in the secondary pre-
53. Spronk HMH, Padro T, Siland JE, Prochaska JH, Winters J, van der Wal AC, vention of atherothrombotic events. N Engl J Med. 2012;366:1404–1413.
Posthuma JJ, Lowe G, d’Alessandro E, Wenzel P, Coenen DM, doi: 10.1056/NEJMoa1200933
Reitsma PH, Ruf W, van Gorp RH, Koenen RR, Vajen T, Alshaikh NA, 68. Bonaca MP, Scirica BM, Creager MA, Olin J, Bounameaux H,
Wolberg AS, Macrae FL, Asquith N, Heemskerk J, Heinzmann A, Moorlag M, Dellborg M, Lamp JM, Murphy SA, Braunwald E, Morrow DA. Vorapaxar

2184 April 30, 2019 Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580


Gurbel et al Antiplatelet and Oral Anticoagulant Therapy

in patients with peripheral artery disease: results from TRA2{degrees} Bahit MC, Ardissino D, Bueno H, Claeys MJ, Nicolau JC, Cornel JH, Goto S,
P-TIMI 50. Circulation. 2013;127:1522–1529, 1529e1–1529e6. doi: Kiss RG, Güray Ü, Park DW, Bode C, Welsh RC, Gibson CM. Clinically sig-

STATE OF THE ART


10.1161/CIRCULATIONAHA.112.000679 nificant bleeding with low-dose rivaroxaban versus aspirin, in addition to
69. Anand SS, Yusuf S. Oral anticoagulants in patients with coronary artery P2Y12 inhibition, in acute coronary syndromes (GEMINI-ACS-1): a double-
disease. J Am Coll Cardiol. 2003;41(4 suppl S):62S–69S. blind, multicentre, randomised trial. Lancet. 2017;389:1799–1808. doi:
70. The Warfarin Antiplatelet Vascular Evaluation Trial Investigators. Oral anti- 10.1016/S0140-6736(17)30751-1
coagulant and antiplatelet therapy and peripheral arterial disease. N Engl 81. Reihani H, Sepehri Shamloo A, Keshmiri A. Diagnostic value of D-dimer
J Med. 2007;357:217–227. in acute myocardial infarction among patients with suspected acute coro-
71. Cohen M, Adams PC, Parry G, Xiong J, Chamberlain D, Wieczorek I, nary syndrome. Cardiol Res. 2018;9:17–21. doi: 10.14740/cr620w
Fox KA, Chesebro JH, Strain J, Keller C. Combination antithrombotic ther- 82. Kleinegris MC, ten Cate H, ten Cate-Hoek AJ. D-dimer as a marker for car-
apy in unstable rest angina and non-Q-wave infarction in nonprior aspi- diovascular and arterial thrombotic events in patients with peripheral arte-
rin users. Primary end points analysis from the ATACS trial. Antithrom- rial disease. A systematic review. Thromb Haemost. 2013;110:233–243.
botic Therapy in Acute Coronary Syndromes Research Group. Circulation. doi: 10.1160/TH13-01-0032
1994;89:81–88. 83. AlKhalfan F, Kerneis M, Nafee T, Yee MK, Chi G, Plotnikov A, Braunwald E,
72. Anand SS, Yusuf S, Pogue J, Weitz JI, Flather M. Long-term oral antico- Gibson CM. D-Dimer levels and effect of rivaroxaban on those levels and
agulant therapy in patients with unstable angina or suspected non-Q- outcomes in patients with acute coronary syndrome (an ATLAS ACS-
wave myocardial infarction: organization to assess strategies for ischemic TIMI 46 Trial Substudy). Am J Cardiol. 2018;122:1459–1464. doi:
syndromes (OASIS) pilot study results. Circulation. 1998;98:1064–1070. 10.1016/j.amjcard.2018.07.032
73. Andreotti F, Testa L, Biondi-Zoccai GG, Crea F. Aspirin plus warfarin 84. Diener HC, Bogousslavsky J, Brass LM, Cimminiello C, Csiba L, Kaste M,
compared to aspirin alone after acute coronary syndromes: an updat- Leys D, Matias-Guiu J, Rupprecht HJ; MATCH investigators. Aspirin and
ed and comprehensive meta-analysis of 25,307 patients. Eur Heart J. clopidogrel compared with clopidogrel alone after recent ischaemic stroke
2006;27:519–526. doi: 10.1093/eurheartj/ehi485 or transient ischaemic attack in high-risk patients (MATCH): randomised,
74. Steg PG, Mehta SR, Jukema JW, Lip GY, Gibson CM, Kovar F, Kala P, double-blind, placebo-controlled trial. Lancet. 2004;364:331–337. doi:
Garcia-Hernandez A, Renfurm RW, Granger CB; RUBY-1 Investigators. 10.1016/S0140-6736(04)16721-4
RUBY-1: a randomized, double-blind, placebo-controlled trial of the safety 85. Gurbel PA, Tantry US. GEMINI-ACS-1: toward unearthing the anti-
and tolerability of the novel oral factor Xa inhibitor darexaban (YM150) thrombotic therapy cornerstone for acute coronary syndromes. Lancet.
following acute coronary syndrome. Eur Heart J. 2011;32:2541–2554. 2017;389:1773–1775. doi: 10.1016/S0140-6736(17)30760-2
doi: 10.1093/eurheartj/ehr334 86. XARELTO. FDA Highlights of Prescribing Information. https://www.access-
75. Alexander JH, Lopes RD, James S, Kilaru R, He Y, Mohan P, data.fda.gov/drugsatfda_docs/label/2018/202439s025lbl.pdf. Accessed
Bhatt DL, Goodman S, Verheugt FW, Flather M, Huber K, Liaw D, Husted SE, December 19, 2018.
Lopez-Sendon J, De Caterina R, Jansky P, Darius H, Vinereanu D, Cornel JH, 87. Bonaca MP, Bhatt DL, Storey RF, Steg PG, Cohen M, Kuder J,
Cools F, Atar D, Leiva-Pons JL, Keltai M, Ogawa H, Pais P, Parkhomenko A, Goodrich E, Nicolau JC, Parkhomenko A, López-Sendón J, Dellborg M,
Ruzyllo W, Diaz R, White H, Ruda M, Geraldes M, Lawrence J, Harrington RA, Dalby A, Špinar J, Aylward P, Corbalán R, Abola MTB, Jensen EC,
Wallentin L; APPRAISE-2 Investigators. Apixaban with antiplatelet therapy Held P, Braunwald E, Sabatine MS. Ticagrelor for prevention of ischemic
after acute coronary syndrome. N Engl J Med. 2011;365:699–708. doi: events after myocardial infarction in patients with peripheral artery
10.1056/NEJMoa1105819 disease. J Am Coll Cardiol. 2016;67:2719–2728. doi: 10.1016/j.jacc.
76. Mega JL, Braunwald E, Mohanavelu S, Burton P, Poulter R, Misselwitz F, 2016.03.524
Hricak V, Barnathan ES, Bordes P, Witkowski A, Markov V, Oppenheimer L, 88. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK; Clopidogrel
Downloaded from http://ahajournals.org by on June 29, 2023

Gibson CM; ATLAS ACS-TIMI 46 study group. Rivaroxaban versus pla- in Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects
cebo in patients with acute coronary syndromes (ATLAS ACS-TIMI 46): of clopidogrel in addition to aspirin in patients with acute coronary syn-
a randomised, double-blind, phase II trial. Lancet. 2009;374:29–38. doi: dromes without ST-segment elevation. N Engl J Med. 2001;345:494–502.
10.1016/S0140-6736(09)60738-8 doi: 10.1056/NEJMoa010746
77. Mega JL, Braunwald E, Wiviott SD, Bassand JP, Bhatt DL, Bode C, Burton P, 89. Zannad F, Anker SD, Byra WM, Cleland JGF, Fu M, Gheorghiade M,
Cohen M, Cook-Bruns N, Fox KA, Goto S, Murphy SA, Plotnikov AN, Lam CSP, Mehra MR, Neaton JD, Nessel CC, Spiro TE, van Veldhuisen DJ,
Schneider D, Sun X, Verheugt FW, Gibson CM; ATLAS ACS 2–TIMI 51 Greenberg B; COMMANDER HF Investigators. Rivaroxaban in patients
Investigators. Rivaroxaban in patients with a recent acute coronary syn- with heart failure, sinus rhythm, and coronary disease. N Engl J Med.
drome. N Engl J Med. 2012;366:9–19. doi: 10.1056/NEJMoa1112277 2018;379:1332–1342. doi: 10.1056/NEJMoa1808848
78. Connolly SJ, Eikelboom JW, Bosch J, Dagenais G, Dyal L, 90. Borst O, Münzer P, Alnaggar N, Geue S, Tegtmeyer R, Rath D, Droppa M,
Lanas F, Metsarinne K, O’Donnell M, Dans AL, Ha JW, Parkhomenko AN, Seizer P, Heitmeier S, Heemskerk JWM, Jennings LK, Storey RF, Angiolillo
Avezum AA, Lonn E, Lisheng L, Torp-Pedersen C, Widimsky P, Maggioni AP, DJ, Rocca B, Spronk H, Ten Cate H, Gawaz M, Geisler T. Inhibitory mech-
Felix C, Keltai K, Hori M, Yusoff K, Guzik TJ, Bhatt DL, Branch KRH, anisms of very low-dose rivaroxaban in non-ST-elevation myocardial
Cook Bruns N, Berkowitz SD, Anand SS, Varigos JD, Fox KAA, Yusuf S; infarction. Blood Adv. 2018;2:715–730. doi: 10.1182/bloodadvances.
COMPASS investigators. Rivaroxaban with or without aspirin in patients 2017013573
with stable coronary artery disease: an international, randomised, dou- 91. Xarelto, INN-rivaroxaban. European Medicines Agency. europa.eu. http://
ble-blind, placebo-controlled trial. Lancet. 2018;391:205–218. doi: www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_In-
10.1016/S0140-6736(17)32458-3 formation/human/000944/WC500057108.pdf. Accessed August 12,
79. Anand SS, Bosch J, Eikelboom JW, Connolly SJ, Diaz R, Widimsky P, 2018.
Aboyans V, Alings M, Kakkar AK, Keltai K, Maggioni AP, Lewis BS, Störk S, 92. Capell WH, Bonaca MP, Nehler MR, Chen E, Kittelson JM, Anand SS,
Zhu J, Lopez-Jaramillo P, O’Donnell M, Commerford PJ, Vinereanu D, Berkowitz SD, Debus ES, Fanelli F, Haskell L, Patel MR, Bauersachs R,
Pogosova N, Ryden L, Fox KAA, Bhatt DL, Misselwitz F, Varigos JD, Vanassche T, Hiatt WR. Rationale and design for the Vascular Outcomes study of ASA
Avezum AA, Chen E, Branch K, Leong DP, Bangdiwala SI, Hart RG, Yusuf S; along with rivaroxaban in endovascular or surgical limb revascularization
COMPASS Investigators. Rivaroxaban with or without aspirin in patients for peripheral artery disease (VOYAGER PAD). Am Heart J. 2018;199:83–
with stable peripheral or carotid artery disease: an international, ran- 91. doi: 10.1016/j.ahj.2018.01.011
domised, double-blind, placebo-controlled trial. Lancet. 2018;391:219– 93. Weitz JI, Fredenburgh JC. 2017 Scientific sessions Sol Sherry Distin-
229. doi: 10.1016/S0140-6736(17)32409-1 guished Lecture in Thrombosis: Factor XI as a Target for New Anti-
80. Ohman EM, Roe MT, Steg PG, James SK, Povsic TJ, White J, Rockhold F, coagulants. Arterioscler Thromb Vasc Biol. 2018;38:304–310. doi:
Plotnikov A, Mundl H, Strony J, Sun X, Husted S, Tendera M, Montalescot G, 10.1161/ATVBAHA.117.309664

Circulation. 2019;139:2170–2185. DOI: 10.1161/CIRCULATIONAHA.118.033580 April 30, 2019 2185

You might also like