Vous êtes sur la page 1sur 14

JACC: Heart Failure Vol. 2, No.

1, 2014
 2014 by the American College of Cardiology Foundation ISSN 2213-1779/$36.00
Published by Elsevier Inc. http://dx.doi.org/10.1016/j.jchf.2013.07.007

STATE-OF-THE-ART PAPER

Antiplatelet and Anticoagulant Agents


in Heart Failure
Current Status and Future Perspectives
Paul A. Gurbel, MD, Udaya S. Tantry, PHD
Baltimore, Maryland

There has been a 3-fold increase in hospital discharges for heart failure (HF) and also significantly increased
mortality rate in HF patients in recent years. A major focus of HF research has been in the area of neurohormonal
control and resynchronization therapy. There is a great urgency in better understanding the pathophysiology
underlying the exceedingly high mortality and a need for exploration of therapeutic strategies beyond those that
influence neurohormonal pathways. The decision to treat patients with HF with antiplatelet therapy remains largely
influenced by the presence or absence of concomitant arterial disease. Antithrombotic therapy has been shown to
be effective in many forms of cardiac disease, including patients with HF and atrial fibrillation. Although it is clear
that platelet activation and hypercoagulability are present in HF, and there is evidence that stroke is reduced by
warfarin therapy in the HF patient, the available data suggest that the risk of major bleeding overshadows the
antithromboembolic benefit in HF patients in sinus rhythm. The utility of oral anticoagulant and/or antiplatelet
therapy has never been evaluated in an adequately powered dedicated clinical trial of HF patients in sinus rhythm.
In this state-of-the art paper we explore the evidence for targeting the inhibition of platelet function and coagulation
to improve outcomes in the HF patient. (J Am Coll Cardiol HF 2014;2:1–14) ª 2014 by the American College of
Cardiology Foundation

On the basis of data from National Health and Nutrition cardiac condition accounting for 9% of all strokes (4).
Examination Survey 2007 to 2010, an estimated 5.1 million Although AF significantly increases the risk of stroke, the
Americans 20 years of age have heart failure (HF) (National heightened risk of thromboembolism in HF patients is in-
Heart, Lung and Blood Institute tabulation). One in 9 death dependent of AF. These observations serve as evidence for
certificates (274,601 deaths) in the United States mentioned a potential “heart failure research paradox” where compara-
HF in 2009. There has been a 3-fold increase in hospital tively less information has been gained from adequately sized
discharges for HF since 1979 in the United States (1). prospective investigations of thrombogenicity and antith-
A major focus of HF research has been in the area of rombotic therapy in the HF patient. Despite the fact that
neurohormonal control and resynchronization therapy (2). treatment with angiotensin-converting enzyme (ACE) in-
However, venous thromboembolism (VTE), cardioembolic hibitors, angiotensin receptor blockers, beta adrenergic an-
stroke, and sudden death occur in 30% of HF patients and tagonists, and mineralocorticoid receptor antagonists in HF
contribute to the observed high overall mortality and patients has improved outcomes, morbidity or mortality re-
morbidity (3). After atrial fibrillation (AF), which accounts mains particularly high in patients with acute decompensated
for 15% of strokes, HF is the next most frequent associated HF (2). In patients hospitalized for HF, the mortality rate
is 4% at 1 month, 18% at 6 months, w30% at 12 months, and
w40% at 38 months (2). These dismal statistics suggest ur-
From the Sinai Center for Thrombosis Research, Baltimore, Maryland. Dr. Gurbel gency in better understanding the pathophysiology underly-
has served as a consultant for Daiichi Sankyo, Sankyo, Lilly, Pozen, Novartis, Bayer, ing the exceedingly high mortality and a need for exploration
AstraZeneca, Accumetrics, Nanosphere, Sanofi-Aventis, Boehringer Ingelheim,
of therapeutic strategies beyond those that influence neuro-
Merck, Medtronic, Iverson Genetics, CSL, and Haemonetics; has received grants
from the National Institutes of Health, Daiichi Sankyo, Lilly, Pozen, CSL, Astra- hormonal pathways. In this state-of-the art paper we explore
Zeneca, Sanofi-Aventis, Haemoscope, Medtronic, Harvard Clinical Research the evidence for targeting the inhibition of platelet function
Institute, and Duke Clinical Research Institute; has received payment for lectures,
and coagulation to improve outcomes in the HF patient.
including service on speakers’ bureaus from Lilly, Daiichi Sankyo, Nanosphere,
Sanofi-Aventis, Merck, and Iverson Genetics; has received payment for development
of educational presentations from Merck, the Discovery Channel, and Pri-Med; owns Pathophysiology of Arterial Thrombosis in HF
stock or stock options in Merck, Medtronic, and Pfizer; and holds patents in the area
of personalized antiplatelet therapy and interventional cardiology. Dr. Tantry has Approximately 70% of patients with HF and left ventricular
reported that he has no relationships relevant to the contents of this paper.
Manuscript received May 22, 2013; revised manuscript received July 25, 2013, systolic dysfunction will have ischemic heart disease (5).
accepted July 25, 2013. Nonischemic etiologies will be responsible for the remainder
2 Gurbel and Tantry JACC: Heart Failure Vol. 2, No. 1, 2014
Antiplatelet and Anticoagulant Agents in Heart Failure February 2014:1–14

Abbreviations of HF with systolic dysfunction. Occlusive thrombus generation at the site of plaque rupture
and Acronyms Platelets play a central role in the is dependent on vulnerable blood (hypercoagulability, en-
genesis of thrombosis at the site hanced platelet function, inflammation) and vulnerable vessel
ACE = angiotensin-
converting enzyme
of arterial wall injury (Fig. 1). (endothelial dysfunction, and ruptured plaque/denuded
Ruptured plaque or denuded endothelium/abnormal anatomy) (Fig. 2). Reduced platelet
ACS = acute coronary
syndromes arterial endothelium results in the survival time, increased mean platelet volume, and greater
ADP = adenosine
exposure of various prothrom- platelet activation and reactivity have been observed in
diphoshpate botic factors thereby promoting patients with HF (8,9). In addition, elevated levels of b-
AF = atrial fibrillation
platelet adhesion, activation, and thromboglobulin, p-selectin, platelet/endothelial cell adhe-
CAD = coronary artery
finally generation of a platelet- sion molecule-1, and osteonectin as well as increased platelet
disease rich thrombus generation. Expo- surface p-selectin and CD40L all serve as further evidence of
CI = confidence interval
sure of the subendothelial matrix heightened platelet activation in patients with HF (10–12).
following atherosclerotic plaque High levels of circulating fibrinogen, fibrinopeptide A, and
F = circulating coagulation
factor rupture and the subsequent release D-dimer have also been demonstrated in patients with HF.
GP = glycoprotein
of various factors facilitate platelet In addition, high levels of fibrinogen and D-dimer observed
adhesion and activation. Platelet in patients with HF have correlated with disease severity (13).
HF = heart failure
activation leads to the release Decreased circulating levels of ADAMTS-13 and increased von
HR = hazard ratio
of important secondary agonists, Willebrand factor (vWF) were found to be significant predictors
INR = international thromboxane A2 and adenosine of clinical events in patients with HF (14). In addition, a pro-
normalization ratio
diphosphate (ADP). Throm- thrombotic environment in the setting of HF is indicated by
LVEF = left ventricular
boxane A2 is produced from increased inflammatory cytokines such as tumor necrosis factor-
ejection fraction
membrane phospholipids, and a and interleukin-1. These factors in turn are associated with
MI = myocardial infarction
ADP is released from dense increased expression of tissue factor and thrombin generation
OAC = oral anticoagulant
granules. Through autocrine and and decreased thrombomodulin and protein C pathway acti-
OR = odds ratio paracrine mechanisms, these 2 vation (anticoagulant factors) (15–17). Vulnerable vessel is
RR = relative risk locally generated secondary ago- characterized by decreased antithrombotic factor expression
TF = tissue factor nists play a critical role in the such as nitric oxide, thrombomodulin and increased expression
VTE = venous sustained activation of aIIbb3 re- of TF, vWF, and elevated inflammation at the site of plaque
thromboembolism ceptors and stable platelet aggre- rupture. Moreover, a bifurcated and stenotic vessel affects
gation. Plaque rupture results in arterial shear and promotes platelet activation (18,19).
tissue factor (TF) exposure at the site of vascular injury and the
generation of femtomolar amounts of thrombin, the most Pathophysiology of Venous Thrombosis in HF
potent primary platelet agonist. Tissue factor bearing cells
come in contact with circulating coagulation factor (F) VII Heart failure has been recognized as a prothrombotic or hy-
resulting in the autoactivation of FVII to form the TF-FVIIa percoagulable state. The major components of Virchow’s triad
complex, which in turn activates FX to FXa and FIX to FIXa. are present in venous thrombosis-vessel wall abnormalities,
FXa initially activates small amounts of thrombin, which in vulnerable blood, and blood flow abnormalities (Fig. 3) (2).
turn activates platelets and also FV and FVIII. Subsequently, Venous stasis induced by high pressure, a low-flow state,
the intrinsic FXase (FVIIIa-FIXa) and prothrombinase (FVa- and immobility result in distention of the vessel wall local and
FXa) are formed on the activated platelet surface where large local hypoxia, causing ischemia and oxidative stress
amounts of thrombin are generated by the conversion of (18,20,21). The latter mechanisms in addition to neurohor-
prothrombin to thrombin. Plasminogen activator inhibitor-1, monal activation result in endothelial cell activation. Endo-
released from platelets, depresses fibrinolysis. Thrombin thelial activation is characterized by increased expression of
finally catalyzes the conversion of soluble fibrinogen to insol- adhesion molecules-selectins and decreased production of
uble strands of fibrin, thereby initiating clot formation (Fig. 1). antithrombotic factor-nitric oxide. These mechanisms pro-
Thrombin also activates FXIII to induce fibrin polymerization. mote platelet/monocyte adhesion and activation. Impor-
The final result is a platelet-fibrin clot that mediates normal tantly, TF-rich microparticles released from activated
hemostasis and pathologic thrombosis at the site of athero- endothelial cells and monocytes attach to the expressed
sclerotic plaque rupture or endothelial denudation. Thrombi adhesion molecules on the endothelium at the site of injured
generated on ruptured plaques may embolize downstream vessel wall (20,22,23). TF initiates the coagulation cascade
and occlude the microvasculature resulting in microinfarction. and, ultimately, fibrin-rich thrombus generation. In this line,
The latter mechanism may participate in the progression of increased circulating endothelial adhesion molecules,
left ventricular dysfunction in the HF patient. At present, it increased microparticles of endothelial origin, and leukocyte
remains unknown whether there is interindividual variability activation have been demonstrated in patients with VTE and
and time dependence in the importance of thromboxane A2, HF (22–24). Finally, venous stasis promotes thrombosis by
ADP, and thrombin in thrombus generation (6,7). decreased clearance of activated coagulation factors (23).
JACC: Heart Failure Vol. 2, No. 1, 2014 Gurbel and Tantry 3
February 2014:1–14 Antiplatelet and Anticoagulant Agents in Heart Failure

Figure 1 Antithtrombotic Therapy Targets in Heart Failure

During arterial thrombosis, at the site of “altered” vessel wall, exposure of the subendothelial matrix leads to adhesion and activation of platelets and subsequent release of
secondary agonists, thromboxane A2 (TxA2) and adenosine diphosphate (ADP). These 2 locally-generated secondary agonists play a critical role in the sustained activation of
glycoprotein (GP) IIb/IIIa receptors and stable platelet aggregation. During venous thrombosis, tissue factor (TF) delivered by microparticles to the site of vessel wall injury binds
to autoactivated factor (F) VII (VIIa) to form TF-FVIIa complex, which in turn activates factor X to Xa. Initial formation of small amounts of Xa initiates the coagulation process on
the surface of activated platelets and leukocytes, where large amounts of thrombin are generated. During arterial thrombosis, TF is exposed at the site of plaque rupture and
initiates the coagulation process. Xa along with Va converts FII to FIIa. Simultaneously, trace amounts of thrombin activate FVIII and FV, which dramatically enhances catalytic
activity of FIX and FX, respectively. Thrombin-activated FXIIIa catalyzes the formation of covalent cross-links between adjacent fibrin chains to form polymerized fibrin. Finally,
together with aggregated platelets, the polymerized fibrin network leads to the formation of a stable, occlusive, platelet-fibrin clot and subsequent ischemic events. In venous
thrombosis, the coagulation pathway plays a major role resulting in fibrin-rich clot formation, whereas in arterial thrombosis, platelet activation and aggregation play a major role
resulting in platelet-rich thrombus formation. Therefore, the management of venous thrombosis is mainly centered around anticoagulation strategies whereas management of
arterial thrombosis is focused on antiplatelet strategies. Vitamin K is a cofactor that promotes the biosynthesis of g-carboxyglutamic acid residues in coagulation factor proteins
that are essential for biological activity. Anticoagulation strategies include warfarin that acts by inhibiting the synthesis of vitamin K-dependent clotting factors, II, VII, IX, and X,
and the anticoagulant proteins C and S. Warfarin inhibits clotting factor synthesis by inhibition of vitamin K epoxide reductase (VKORC1), thereby reducing the regeneration of
vitamin K1 epoxide. Other anticoagulants include direct thrombin inhibitors such as bivalirudin and dabigatran; direct Xa inhibitors such as rivaroxaban and apixaban; LMWH,
heparin, and fondaparinux bind to antithrombin (AT), facilitating its inhibition of coagulation factors. Fondaparinux and LMWH preferentially inhibit Xa as compared to heparin,
which more greatly inhibits IIa. Antiplatelet strategies include inhibition of platelet cyclooxygenase-1 enzyme by aspirin resulting in the inhibition of generation of TxA2 and TxA2-
induced platelet aggregation; inhibition of ADP receptor, P2Y12, by clopidogrel, prasugrel, ticagrelor, and cangrelor; inhibition of activated GPIIb/IIIa receptor by abciximab,
eptifibatide, and tirofiban; and vorapaxar that inhibits thrombin receptor protease activated receptor (PAR)-1. Factor II ¼ prothrombin; Factor IIa ¼ thrombin; LMWH ¼ low
molecular weight heparin; TP ¼ thromboxane A2 receptor; vWF ¼ von Willebrand factor.

Myocardial Infarction and Stroke Risk in HF HF patients have a 1.0% to 3.5% annual rate of stroke, with
a possible relationship between low ventricular ejection
In patients with HF secondary to coronary artery disease fraction (LVEF) and stroke risk (28).
(CAD), ongoing silent or symptomatic ischemia has being Strokes associated with HF have been attributed to mul-
associated with HF progression (25). The elderly, women, tiple etiologies. Concomitant AF is associated with loss of
and those with prior myocardial infarction (MI) are at atrial contraction. In the setting of stasis, thrombosis is pro-
greatest risk for developing HF. However, a relatively low moted in the left appendage, and embolization may ensue. In
rate (2% to 4%) of fatal or non-fatal MI has been reported in addition, AF is associated with atherosclerosis. Therefore,
patients with HF (26,27). In contrast, compared with a 0.1% atheroembolism from nonatrial sources may also explain a
to 0.5% annual rate of stroke in patients 80 years of age, heightened risk for stroke. In HF patients in sinus rhythm,
4 Gurbel and Tantry JACC: Heart Failure Vol. 2, No. 1, 2014
Antiplatelet and Anticoagulant Agents in Heart Failure February 2014:1–14

Figure 2 Mechanisms of Coronary Arterial Thrombosis

Plaque rupture and endothelial erosion are the primary events associated with arterial thrombosis. The occlusive thrombus generation at the site of vessel injury is dependent on
the presence of vulnerable blood and vulnerable vessel. Platelet activation and aggregation play major roles in arterial thrombosis, leading to the formation of a platelet-rich
thrombus. NO ¼ nitric oxide; other abbreviations as in Figure 1.

underlying atherosclerosis may play a dominant mechanistic at 30 days to 6 months (HRs: 12.7 and 9.3, respectively) and
role in stroke occurrence. Among HF patients with stroke, >6 months (HRs: 4.9 and 4.0, respectively) (33). Warfarin
about one-half have been reported to have AF (29). The use was associated with a lower risk for death and the
significant impact of HF on stroke risk in AF patients was composite of death or stroke compared with no warfarin
suggested in the Stroke Prevention in Atrial Fibrillation In- therapy and this effect was more prominent in the first 30
vestigators studies (30). The risk may also depend on how HF days. Finally, independent predictors for death, stroke, and
is defined and whether it is decompensated versus stable (31). the composite of death or stroke were previous stroke/
Significantly more circulating platelet aggregates were transient ischemic attack/transient ischemia (33).
demonstrated in patients with HF compared with normal In the Rotterdam study, 7,546 participants 55 years old
volunteers by Mehta et al. (32). In the same study, the (female ¼ 61%) without a history of stroke were continu-
number of circulating platelet aggregates declined to normal ously monitored for major events including stroke and HF.
levels following sodium nitroprusside infusion, and there was At baseline, 233 (3.1%) patients had HF, whereas 1,014
significantly lower ex vivo epinephrine- and ADP-induced patients developed HF and 827 patients developed stroke
platelet aggregation, which was associated with a 30% during an average follow-up time of 9.7 years. There was a
decrease in systemic vascular resistance and a 28% increase in more than 5-fold increase in the risk of ischemic stroke in
cardiac output. The authors suggested that an increase in the first month after HF diagnosis (age- and sex-adjusted
vascular resistance in certain HF patients may cause an in- HR: 5.79, 95% confidence interval [CI]: 2.15 to 15.62),
crease in circulating platelet aggregates and that antiplatelet but the risk decreased over time after 1 to 6 months (age-
therapy may be beneficial in these patients. and sex-adjusted HR: 3.50, 95% CI: 1.96 to 6.25) and after
Insight into the heightened risk of stroke and death in HF 0.5 to 6 years (HR: 0.83, 95% CI: 0.53 to 1.29) (34).
was gained in the Diet, Cancer and Health study. Among In the SAVE (Survival and Ventricular Enlargement)
the 51,553 patients without prior HF, 1,239 were identified trial, 2,231 patients who had LV dysfunction after acute MI
with incident HF. There was a markedly increased risk of were followed for an average of 42 months. The annual rate
death, and death or stroke (ischemic or hemorrhagic or of fatal and nonfatal stroke was 1.5%, and the 5-year rate of
both) in the first 30 days after presentation with first HF stroke was 8.1%. There was a 2-fold increased risk for
relative to non-HF exposed patients (hazard ratios [HR]: stroke in patients with left ventricular ejection fraction
42.8 and 38.4, respectively). There was attenuation of risk (LVEF) 28% compared with patients with LVEF of
JACC: Heart Failure Vol. 2, No. 1, 2014 Gurbel and Tantry 5
February 2014:1–14 Antiplatelet and Anticoagulant Agents in Heart Failure

Figure 3 Mechanisms of Venous Thrombosis in Heart Failure

Venous thrombosis is dependent on vulnerable blood and vessel wall and blood flow abnormalities. Endothelial dysfunction results from hypoxia/distension/neurohormonal
activation, setting up a cascade of downstream prothrombotic events. CO ¼ cardiac output; IL ¼ interleukin; RBC ¼ red blood cell; TNF ¼ tumor necrosis factor; tPA ¼ tissue
plasminogen activator; other abbreviations as in Figures 1 and 2.

>35% (p ¼ 0.01). In this study, lower ejection fraction (for Antithrombotic Therapy in Patients With HF
every decrease of 5 percentage points in the ejection fraction
there was an 18% increase in the risk of stroke), older age, The previous evidence suggests that antithrombotic therapy
and the absence of aspirin or anticoagulant therapy were targeting the platelet and/or coagulation may provide
found to be independent risk factors for stroke (35). beneficial effects in patients with HF, particularly early after
presentation. Long-term oral anticoagulant (OAC) therapy
Venous Thromboembolism is an established major preventive treatment strategy against
stroke in patients with AF (5). However, unlike AF, the role
It has been reported that patients with HF have an increased of antithrombotic agents in the prevention of stroke in pa-
risk for developing deep vein thrombosis versus those without tients with HF and sinus rhythm has been much less
HF, with adjusted odds ratios (ORs) of 1.47 (95% CI: 1.47 to investigated. Personalized antithrombotic therapy has been
1.48) to 2.93 (95% CI: 1.55 to 5.56) (36). Moreover, there is a studied in patients treated with coronary stents with the goal
10% to 22% risk of venographically-proven VTE in HF pa- of determining a therapeutic window for on-treatment
tients in the absence of pharmacologic treatment (36,37). In platelet reactivity that optimally prevents ischemic events
several case-cohort studies, HF patients were found to have a 2- while avoiding medically-important bleeding (39). Labora-
to 3-fold increased risk for VTE compared with patients with tory tests are available that identify high intrinsic throm-
other medical conditions. The rate of VTE was 2.7 and 2.1 per bogenicity that have been associated with clinical thrombotic
100 patient-years in the V-HeFT (Veterans Affairs events after stenting (40). Similar investigations have never
Vasodilator-Congestive Heart Failure) I and II trials, respec- been undertaken to identify the prothrombotic HF patient.
tively, that included both patients in sinus rhythm and with AF Despite the extensive research in the PCI population, the
(38). In an analysis of the SCD-HeFT (Sudden Cardiac Death- goal of optimal antithrombotic therapy that balances
Heart Failure Trial), which excluded all patients with AF at reduction in ischemia and avoidance of bleeding remains
the time of randomization, the rate of thromboembolism was elusive in HF patients. Moreover, platelet activation and
3.4% over a median follow-up of 45.5 months. A trend toward aggregation, and thrombin formation, are highly interlinked.
an increased 4-year Kaplan-Meier rate of thromboembolism Optimal target(s) for the inhibition of the effects of
with lower LVEF was observed (3.5%, 3.6%, and 4.6% with thrombin remain unclear; proposed strategies include direct
LVEF 30% to 35%, 20% to 30%, and <20%, respectively) (28). thrombin inhibition, inhibition of the coagulation factors
6 Gurbel and Tantry JACC: Heart Failure Vol. 2, No. 1, 2014
Antiplatelet and Anticoagulant Agents in Heart Failure February 2014:1–14

upstream from thrombin formation including FXa, FIXa, (39). Moreover, it is also interesting to note that although
FVIIa, or FVa, or the platelet thrombin receptor-protease more potent P2Y12 receptor blockers are more effective than
activated receptor-1. clopidogrel in the overall patient population, in “very high-
risk patients” (diabetes mellitus, elderly patients, and those
Antiplatelet Therapy with renal failure, history of MI, stroke, and stent throm-
bosis), the overall rate of ischemic event occurrence remains
Aspirin is chosen in HF based on its role in secondary very high. In addition, some of these “very high risk” groups
prevention in patients with atherosclerotic vascular disease. are also associated with an increased bleeding risk. The
To this date there has been no dedicated randomized trial of previous limitations highlight the ceiling effect of current
antiplatelet therapy in the HF patient. All of the random- dual antiplatelet therapy targeting COX-1 and the P2Y12
ized trials have included an anticoagulant arm (see subse- receptor in high-risk CAD patients and suggest that the
quent discussion). In patients with high-risk coronary artery residual ischemic event occurrences are mediated by other
disease, it is well established that simultaneous inhibition of pathways that are unblocked by current antiplatelet therapy.
cyclooxygenase-1 with aspirin and the adenosine diphos- There is no information available on the outcomes of
phate-P2Y12 receptor with clopidogrel is associated with a HF patients treated with either prasugrel or ticagrelor.
significant reduction in MI (41). However, the efficacy of Thus, the indication for antiplatelet therapy in the HF patients
dual antiplatelet therapy on stroke reduction is uncertain. depends upon the presence of concomitant vascular disease.
Adding aspirin to clopidogrel in patients with recent
ischemic stroke or transient ischemic attack in the large- Warfarin
scale MATCH trial (Management of atherothrombosis
with clopidogrel in high-risk patients with recent transient Warfarin (vitamin K antagonist) blocks multiple steps of
ischaemic attack or ischaemic stroke) was not associated coagulation by reducing the synthesis of vitamin K depen-
with a benefit in reducing major vascular events (RRR dent coagulation factors. In a meta-analysis of trials
[relative risk reduction]: 6.4%, 95% CI: –4.6 to 16.3, p ¼ involving patients with AF, warfarin therapy was found to be
0.244). However, the risk of life-threatening or major associated with a 64% reduction in stroke and a 26%
bleeding was increased by the addition of aspirin to clopi- reduction in all-cause mortality when compared with pla-
dogrel (42). In the ESPRIT (European/Australasian Stroke cebo/control (46). As warfarin is metabolized by the cyto-
Prevention in Reversible Ischaemia Trial), treatment of chrome P450 isoenzymes, interactions with other drugs that
aspirin (30 to 325 mg/day) with dipyridamole (200 mg twice are metabolized by the same cytochrome P450 isoenzymes
daily) was associated with a 20% relative risk (RR) reduction influence the pharmacokinetics and pharmacodynamics of
in the composite end point of composite of death from all warfarin. The metabolism and efficacy of warfarin are
vascular causes, nonfatal stroke, nonfatal MI, or major influenced by liver function, genetic polymorphisms, and
bleeding complication in patients with transient ischemic alcohol and food consumption (47). The major concern of
attack or minor stroke of presumed arterial origin compared genetic variability lies within the CYP2C9 and VKORC1
to aspirin alone (43). Similarly, aspirin plus extended release genes that encode proteins involved in the metabolism of
dipyridamole versus clopidogrel was associated with similar warfarin (48,49). Warfarin therapy is associated with a
incidences of primary endpoint of first recurrence of stroke relatively narrow therapeutic window of international
(HR: 1.01, 95% CI: 0.92 to 1.11) in patients with a recent normalization ratio (INR) values between 2.0 and 3.0.
ischemic stroke in the PRoFESS (Prevention Regimen for Insufficient anticoagulation and a subsequent rise in the risk
Effectively Avoiding Second Strokes) trial (44). of ischemic strokes are associated with INR values below
A meta-analysis performed by Antithrombotic Trialists’ 2.0, and a higher risk of intracranial bleeding is associated
collaboration of secondary prevention trials demonstrated with an INR >4.0 (49,50).
that aspirin therapy was associated with an 1.5% absolute New Oral Anticoagulants
reduction in serious vascular events (6.7% vs. 8.2% per year,
p < 0.0001) and a 20% reduction in total stroke (2.08% vs. Currently known anticoagulants can be classified as
2.54% per year, p ¼ 0.002) and coronary events (4.3% vs. parenteral or oral (based on their administration route)
5.3% per year, p < 0.0001) but a nonsignificant increase in or direct or indirect inhibitors (based on their target of
hemorrhagic stroke (45). Treatment failure and pharmaco- action). New OACs have been developed to overcome
logic limitations associated with clopidogrel therapy fostered some of the limitations of warfarin therapy. The direct
the development of more potent P2Y12 receptor blockers thrombin inhibitors (“gatrans,” e.g., dabigatran) directly
such as, prasugrel, an irreversible agent, and ticagrelor, a bind to thrombin and block thrombin-induced conversion
reversibly binding agent. Treatment of patients with acute of fibrinogen to fibrin and the activation of FV, FVII, FIX,
coronary syndromes (ACS) with the latter agents was more and platelets. The latter functions are responsible for the
effective than clopidogrel; however, the observed degree of amplification of thrombin generation. Moreover, direct
adverse event reduction (w20% relative and w2% absolute), thrombin inhibitors inhibit the activity of fibrin-bound
and significantly greater bleeding remain major concerns thrombin (51,52).
JACC: Heart Failure Vol. 2, No. 1, 2014 Gurbel and Tantry 7
February 2014:1–14 Antiplatelet and Anticoagulant Agents in Heart Failure

The inhibition of coagulation protease enzymes respon- added support for the use of aspirin in HF patients on ACE
sible for the propagation phase attenuates the generation of inhibitor therapy. In over 2,000 patients with a history of
thrombin. Anticoagulants have been developed to inhibit HF and AF, there was no evidence of aspirin-related
FIXa (e.g., the DNA aptamer pegnivacogin), FVIIIa (TB- attenuation of ACE inhibitor benefit (60).
402), and FVa/FVIIIa (drotrecogin, a recombinant form of In a multivariate analysis of the SOLVD trial, warfarin
human activated protein C). Recomodulin and solulin are therapy (n ¼ 861) was associated with a significant reduction
soluble recombinant derivatives of human thrombomodulin. in all-cause mortality (adjusted HR: 0.76, 95% CI: 0.65 to
The most widely investigated FXa inhibitors have been 0.89, p ¼ 0.0006) and in the risk of death or hospital
rivaroxaban and apixaban. Other FXa inhibitors include admission for HF (HR: 0.82, 95% CI: 0.72 to 0.93,
edoxaban, otamixaban, darexaban, betrixaban, and TAK- p ¼ 0.0002) compared to warfarin nonuse. The benefit of
442. Among the new OACs, dabigatran and apixiban have warfarin therapy was not influenced by the presence of AF,
been shown to be more effective in preventing stroke than age, ejection fraction, New York Heart Association func-
warfarin in patients with nonvalvular AF. In addition, lower tional class, or etiology (57).
rates of intracranial bleeding have been reported with apix-
iban, rivaroxaban, and dabigatran in the latter group of pa- Randomized Trials of Antithrombotic Therapy in HF
tients. Additional advantages of these new OACs over
warfarin include the absence of any reported interactions with In the WASH (Warfarin/Aspirin Study in Heart Failure)
food, fewer interactions with other medications, and absence pilot study, 279 patients were randomized to receive no
of frequent laboratory monitoring and dose adjustments (51). antithrombotic treatment (26%), 300 mg/day aspirin (32%)
or warfarin (26%, target INR 2.5) for w27 months. In this
Evidence for Antithrombotic Efficacy in HF study, w75% of patients were male, w60% of patients had
ischemic heart disease, and only 4% to 7% of patients had
In a post hoc analysis of major HF trials associated with AF. No difference in the primary endpoint of first occur-
reduced ejection fraction and anticoagulant therapy, the rence of death, nonfatal MI, or nonfatal stroke was observed
annual rate of stroke was between 1.1% and 4.6%. Most of between the treatment groups. However, compared to
the trials included in the analysis had some patients with AF warfarin, aspirin treated patients were twice as likely to have
(53). Clinical trials conducted in the 1940s and 1950 have a cardiovascular hospitalization or death during the first 12
first assessed the utility of OAC in patients with HF. Many months follow-up. Aspirin treatment was also associated
of the patients in these studies also had accompanying AF with significantly more hospitalizations for cardiovascular
and valvular disease. Dicumarol therapy, the primary OAC reasons, especially worsening HF (p ¼ 0.044). Therapy with
used was associated with reduced thromboembolic event warfarin and aspirin therapy was associated with a higher
occurrence and death (54–56). However, the relevance of rate of minor bleeding, although few overall major hemor-
these trials in patients with sinus rhythm, which is more rhagic events were reported. The overall study results argued
prevalent in current practice, is limited. against the benefit of antiplatelet therapy in HF patients to
In the SOLVD (Studies of Left Ventricular Dysfunction) prevent thromboembolic events (Table 1) (61).
trial (n ¼ 6,797), antiplatelet therapy was used in 46.3% of The subsequent HELAS (HEart failure Long-term
patients and was associated with significantly reduced all Antithrombotic Study) trial was prematurely terminated af-
cause death (adjusted HR: 0.82, 95% CI: 0.73 to 0.92, p ¼ ter enrolling 197 of an intended 6,500 patients due to slow
0.0005) and reduced risk of death or hospital admission for enrollment. In this trial, 115 patients with ischemic cardio-
HF (adjusted HR: 0.81, 95% CI: 0.74 to 0.89, p < 0.0001) myopathy were randomized to 325 mg daily aspirin or warfarin
compared to patients not treated with antiplatelet therapy (INR 2.0 to 3.0), whereas 82 patients with nonischemic car-
(57). Interestingly, the observation that the addition of ena- diomyopathy were randomized to placebo or warfarin (INR
lapril did not decrease the mortality rate in patients receiving 2.5 to 3.0) for 18.5 to 21.9 months. Similar to the WASH
antiplatelet agents, and conversely neither did the addition of study, there was no difference in the occurrence of the primary
aspirin in patients receiving enalapril fueled great controversy composite endpoint between the groups and only warfarin
about an aspirin–ACE inhibitor interaction (58). Further therapy was associated with 7 major hemorrhagic events that
support for an aspirin-ACE inhibitor interaction arose from were predominantly due to high anticoagulation (62).
the CONSENSUS II (Co-operative New Scandinavian In the WATCH (Warfarin and Antiplatelet Therapy in
Enalapril Survival Study II) Study. Differential effects of the Chronic Heart Failure) trial, 1,587 of 4,500 planned patients
ACE inhibitor enalapril in subgroups defined by use of aspirin with symptomatic HF (New York Heart Association func-
at baseline were analyzed. Logistic regression demonstrated tional class II to IV) for at least 3 months were studied. The
that the enalapril–aspirin interaction term was a significant trial was terminated prematurely due to slow enrollment.
predictor of mortality and was a borderline significant pre- These patients were in sinus rhythm with documented
dictor of mortality 30 days after randomization (59). LVEF 35% and received 162 mg daily aspirin, 75 mg daily
The ACTIVE (Atrial fibrillation Clopidogrel Trial with clopidogrel (in a double-blind, double dummy fashion), or
Irbesartan for the prevention of Vascular Events) program open label warfarin (target INR 2.5 to 3.0) for a mean
8 Gurbel and Tantry JACC: Heart Failure Vol. 2, No. 1, 2014
Antiplatelet and Anticoagulant Agents in Heart Failure February 2014:1–14

duration of 1.9 years (63). There was no placebo arm in this of the secondary endpoints, throughout the follow-up period
study. Similar to previous studies, there was no difference in with warfarin therapy compared to aspirin therapy (0.72% vs.
the rate of the primary composite endpoint of first occur- 1.36%/year; HR: 0.52, 95% CI: 0.33 to 0.82, p ¼ 0.005) but
rence of death, nonfatal stroke, or nonfatal MI with warfarin with significantly greater major hemorrhagic events (5.8% vs.
versus aspirin therapy (HR: 0.98, 95% CI: 0.86 to 1.12, 2.7%; OR: 2.2, 95% CI: 1.42 to 2.47, p < 0.001) (63,64). The
p ¼ 0.77); with clopidogrel versus aspirin therapy (HR: reduction in stroke associated with warfarin therapy in pa-
1.08, 95% CI: 0.83 to 1.40, p ¼ 0.57); and with warfarin tients with HF appeared similar to the reduction in stroke
versus clopidogrel therapy (HR: 0.89, 95% CI: 0.68 to 1.16, observed in patients treated with warfarin for AF (53).
p ¼ 0.39). There was no difference in mortality between However, the overall stroke rate was lower in the HF popu-
therapies. Warfarin therapy was associated with a reduction lation in sinus rhythm. Furthermore, there was no difference
in the incidence of stroke (both fatal and disabling) compared in the combined rate of intracranial and intracerebral hem-
to aspirin and clopidogrel (p < 0.02 for both). But, there was orrhage between warfarin and aspirin therapy (0.8% vs. 0.8%)
significantly increased bleeding in the warfarin group but the rate of major gastrointestinal hemorrhage was 3 times
compared to clopidogrel group (n ¼ 30 vs. n ¼ 12, p < 0.01) higher with warfarin therapy compared to aspirin therapy
but not compared to the aspirin group (n ¼ 30 vs. n ¼ 19, (3.2% vs. 1.4%; HR: 3.0, 95% CI: 1.30 to 4.38, p ¼ 0.008).
p ¼ 0.22). A significant reduction in stroke was observed in In contrast to the WATCH and WASH trials, aspirin
patients with ischemic HF (73% of population) where the therapy was not associated with an increased rate of hospi-
stroke rates were 0% in warfarin group versus 2.7% in clo- talization (61,63). The lack of an effect of warfarin therapy
pidogrel group (p ¼ 0.009) and 1.6% in aspirin group (p ¼ on death in this HF population in sinus rhythm suggests that
0.01). Furthermore, significantly increased incidences of the mechanism of death in these patients is not due to
hospitalization were observed in patients receiving aspirin thromboembolism but may be mostly due to pump failure or
compared to warfarin (p < 0.001). The overall evidence from ventricular arrhythmias (65).
the primary endpoint occurrences and mortality did not In a recently reported subgroup analysis of the WARCEF
support that warfarin is superior to aspirin and that clopi- trial, it was demonstrated that among 32 variables explored
dogrel is superior to aspirin in HF patients with ejection for an interaction with treatment, patients <60 years of age
fraction 35% who are in sinus rhythm (63). had a 37% reduction in the occurrence of the primary
In the largest study to date, the WARCEF (Warfarin versus endpoint (HR: 0.63, 95% CI: 0.48 to 0.84, p ¼ 0.001) as
Aspirin in patients with Reduced Cardiac Ejection Fraction) compared with no benefit observed in patients the 60 years
study, the efficacy of 325 mg daily aspirin versus warfarin of age group. Again, this benefit in patients <60 years of age
therapy (target INR 2.5 to 3.0) was evaluated in a double- was mainly driven by a significantly lower mortality rate that
blind, double-dummy study design with sham INRs in was not observed in patients 60 years of age. In addition,
2,305 patients in sinus rhythm with LVEF  35% (64). This there was less major bleeding risk with warfarin therapy in
trial included a relatively young population (mean age ¼ 61 the younger cohort (66).
years) with HF and a mean LVEF of 25%. In this trial, 48% The previous 4 randomized trials have important limita-
of patients had MI, 43% had ischemic cardiomyopathy, 4% tions. Enrollment was largely incomplete and all studies were
had AF, w80% of patients were male, and 75% were a non- underpowered to test the proposed hypothesis. Second,
Hispanic white population. The mean duration of follow-up heterogeneous drug administration, a variable degree of un-
was 3.5 years. In 34% and 32% of the total follow-up time, derlying HF etiology, and varying definitions of the primary
patients did not receive the randomized warfarin and aspirin composite endpoint were observed. Finally, the WATCH
therapy, respectively. There was no control arm in this trial. and WARCEF trials did not have a placebo arm to evaluate
Similar to slow recruitment problems observed in previous the actual efficacy of antithrombotic therapy (63,64).
trials, in the WARCEF trial, the enrollment was stopped after
2,305 patients from 11 countries instead of a planned 2,860 Meta-Analyses of Randomized Trials
patients, reducing the power to test the primary hypothesis and Registry Data
from 89% to 69%. The maximum follow-up was extended to 6
years instead of 5 years. There was no significant difference In a meta-analysis that included 4,378 patients from the 4
between treatment groups for the occurrence of the primary randomized trials of chronic HF and reduced ejection
endpoint of death, ischemic stroke or intracerebral hemorrhage fraction in SR, there was no difference in all-cause mortality
(7.47% vs. 7.93% events per 100 patient-years, respectively; (RR: 1.00, 95% CI: 0.88 to 1.13, p ¼ 0.94), HF-related
HR: 0.93, 95% CI: 0.79 to 1.10, p ¼ 0.40). In a time-varying hospitalizations (RR: 1.16, 95% CI: 0.79 to 1.71, p ¼ 0.45),
analysis using a Cox model, there was a marginal trend in the and nonfatal MI (RR: 0.87, 95% CI: 0.63 to 1.21, p ¼ 0.40)
primary endpoint favoring a benefit with warfarin therapy between warfarin and aspirin treatments. However, warfarin
versus aspirin therapy over time with an HR of 0.89 per therapy compared to aspirin therapy was associated with
year (95% CI: 0.80 to 0.998, p ¼ 0.046) and HR of 0.76 fa- a 41% reduction in all strokes (RR: 0.59, 95% CI: 0.41
voring warfarin by year 4 (p ¼ 0.04) (64). As in the WATCH to 0.85, p ¼ 0.004) and 52% reduction in fatal and
trial, there was a significant reduction in ischemic stroke, 1 nonfatal ischemic strokes (RR: 0.48, 95% CI: 0.32 to 0.73,
JACC: Heart Failure Vol. 2, No. 1, 2014 Gurbel and Tantry 9
February 2014:1–14 Antiplatelet and Anticoagulant Agents in Heart Failure

Table 1 Randomized Antithrombotic Trials in Patients With Heart Failure

Patients Treatment and Follow-Up Primary Outcome Outcome Comments


WASH (Warfarin/Aspirin HF: 35% LVEF Randomized to no Death, nonfatal MI, 26% no antithrombotic More HF hospitalization
Study in Heart Failure) n ¼ 279 antithrombotic nonfatal stroke 32% aspirin with aspirin
therapy, 300 mg 26% warfarin More hemorrhage with
aspirin, or warfarin warfarin
(INR 2.5)
Mean follow-up:
27  1 month
HELAS (HEart failure HF: <35% LVEF and HF pts were randomized Nonfatal stroke, No significant difference Low n
Long-term NYHA functional class to 325 mg aspirin or peripheral or PE, MI, between groups
Antithrombotic II to IV warfarin (INR 2–3) re-hospitalization,
Study) n ¼ 197 n ¼ 115 exacerbation of HF
DCM pts were or death
randomized to
warfarin (INR 2.5)
or placebo
Mean followup:
19 to 21 months
WATCH (Warfarin and HF >3 months, in sinus Randomized to open- First occurrence of death, Warfarin vs. aspirin: Lower stroke rate with
Antiplatelet Therapy in rhythm + 35% LVEF label warfarin (INR nonfatal MI, or HR: 0.98, warfarin
Chronic Heart Failure) n ¼ 1,587 2.5–3.0) + double- nonfatal stroke 95% CI: 0.86–1.12; More HF hospitalization
blind treatment to p ¼ 0.77 with aspirin
162 mg aspirin or Clopidogrel vs. aspirin:
75 mg clopidogrel HR: 1.08,
Mean follow-up: 95% CI: 0.83–1.40;
>12 months p ¼ 0.57
Warfarin vs. clopidogrel:
HR: 0.89,
95% CI: 0.68–1.16,
p ¼ 0.39
WARCEF (Warfarin HF: normal sinus rhythm, Randomized to warfarin First occurrence of 26.4% vs. 27.5%. No significant difference
versus Aspirin in 35% LVEF (INR 2–3.5) or 325 mg ischemic stroke, HR: 0.93, between groups.
patients with Reduced n ¼ 2,305 aspirin intracerebral 95% CI: 0.79–1.10; Lower ischemic stroke
Cardiac Ejection Mean follow-up: hemorrhage, or death p ¼ 0.40 with warfarin offset by
Fraction) 42  18 months for any cause Ischemic stroke higher major
2.5% vs. 4.7%, hemorrhage.
HR: 0.52, 95% CI:
0.33–0.82;
p ¼ 0.005
Major hemorrhage
5.8% vs. 2.7%,
HR: 2.21,
95% CI: 1.42–3.47,
p < 0.001
Hospitalization for HF
20.9% vs. 17.5%,
HR: 1.12,
95% CI: 0.998–1.47;
p ¼ 0.053

CI ¼ confidence interval; DCM ¼ dilated cardiomyopathy; HF ¼ heart failure, HR ¼ hazard ratio, INR ¼ international normalization ratio; LVEF ¼ left ventricular ejection fraction; MI ¼ myocardial infarction;
NYHA ¼ New York Heart Association; PE ¼ pulmonary embolism.

p ¼ 0.0006), and a 2-fold increased risk of major hemor- anticoagulation indications and 26% of patients were
rhage (RR: 2.02, 95% CI: 1.45 to 2.80, p ¼ 0.0001). Thus, receiving anticoagulation therapy. Anticoagulation therapy
the overall benefit from warfarin in this population appears was associated with no significant differences in total mor-
counterbalanced by the increased risk of bleeding (67). tality (14% vs. 12.5%) or stroke (0.8% vs. 0.9%), but was
Another meta-analysis including the same patient popu- associated with a reduction in the combined endpoint
lation from the previous 4 randomized trials specifically occurrence of cardiac death, heart transplantation, coronary
addressed stroke prevention. The number needed to reduce revascularization, and cardiovascular hospitalization in a
1 stroke was 61 and that needed to harm (major hemor- propensity score–adjusted multivariate analysis (HR: 0.74,
rhage) was 34 for warfarin therapy (68). Warfarin did not 95% CI: 0.56 to 0.97, p ¼ 0.03) (69).
increase mortality or confer an increased risk of intracerebral
hemorrhage compared with aspirin. Potential Role of New Anticoagulants
The REDINSCOR (Red de investigación clínica y básica
en insuficiencia cardiaca) registry included 2,263 patients The new oral anticoagulants have been studied in compar-
with ejection fraction 35% and sinus rhythm without other ison to warfarin mainly in AF patients and for treatment
10 Gurbel and Tantry JACC: Heart Failure Vol. 2, No. 1, 2014
Antiplatelet and Anticoagulant Agents in Heart Failure February 2014:1–14

Table 2 Trials of New Oral Anticoagulants in the Subgroups of Patients With Heart Failure

Patients Treatment and Primary Outcome Results Comments


Dabigatran
RE-LY (Randomized Evaluation of Patients with AF Warfarin vs. dabigatran Overall ¼ 1.69% vs. No significant treatment
Long-term anticoagulant No sHF ¼ 13,203, sHF ¼ 4,904 110 mg vs. dabigatran 1.53% vs. 1.11% interaction (p ¼ 0.42
therapY) 150 mg 2-yr stroke or No sHF ¼ 1.63% vs. and 0.33)
systemic embolism 1.40% vs. 1.00%
sHF ¼ 1.85% vs.
1.90% vs. 1.44%
Apixaban
ARISTOTLE (Apixaban for Patients with AF and 1 additional Warfarin vs. apixaban Overall ¼ 1.6% vs. 1.27% No significant treatment
Reduction In STroke and Other risk for stroke 5 mg bid No HF ¼ 1.6% vs. 1.2% interaction (p ¼ 0.45)
ThromboemboLic Events in With HF ¼ 5,541 1.8 yr ischemic or HF ¼ 1.6% vs. 1.4%
atrial fibrillation) No HF ¼ 12,660 hemorrhagic or
systemic embolism
APPRAISE-2 (Apixaban for Patients with a recent ACS and at Placebo vs. apixaban Overall ¼ 7.9% vs. 7.5% No significant treatment
Prevention of Acute Ischemic least 2 additional risk factors for 5 mg bid + standard No HF, HR: 1.05 interaction (p ¼ 0.08)
Events 2) recurrent ischemic events antiplatelet therapy (95% CI: 0.86–1.29)
No HF ¼ 5,362 241 days CV death, MI, or HF, HR: 0.77
HF ¼ 2,028 ischemic stroke (95% CI: 058–1.02)
Rivaroxaban
ROCKET AF (Rivaroxaban Once Patients with nonvalvular AF Dose-adjusted warfarin vs. Overall ¼ 2.2% vs. 1.7% No significant treatment
Daily Oral Direct Factor Xa cHF ¼ 8,851 20 mg rivaroxaban cHF ¼ 3.9% vs. 3.6% interaction (p ¼ 0.419)
Inhibition Compared with No cHF ¼ 5,318 1.7 yr stroke or systemic no cHF ¼ 5.01% vs. 4.13%
Vitamin K Antagonism for embolism
Prevention of Stroke and
Embolism Trial in Atrial
Fibrillation)
ATLAS ACS TIMI 51 (Anti-Xa Recent ACS Placebo vs. 2.5 mg Overall ¼ 10.7% vs.
Therapy to Lower Cardiovascular No cHF¼ 4,282 rivaroxaban bid vs. 5 mg 9.1% vs. 8.8%
Events in Addition to Standard cHF ¼ 483 rivaroxaban No cHF¼ 6.0% vs. 5.5%
Therapy in Subjects with Acute 13 months CV death, MI, (2.5 mg rivaroxaban)
Coronary Syndrome– or stroke HF ¼ 16.8% vs. 10.1%
Thrombolysis In Myocardial (2.5 mg rivaroxaban)
Infarction 51)

ACS ¼ acute coronary syndromes; bid ¼ twice daily dose; cHF ¼ congestive heart failure; CV death ¼ cardiovascular death; sHF ¼ symptomatic heart failure; other abbreviations as in Table 1.

of and prophylaxis against VTE. In a recent meta-analysis mortality (p < 0.0001) was reported in patients with HF
of trials in patients with AF, new oral anticoagulants were who were treated with bivalirudin compared to heparin plus
22% more effective in the prevention of stroke compared a GPII/IIIa inhibitor (73). These hypothesis-generating
to warfarin (RR: 0.78, 95% CI: 0.67 to 0.92) and there was data suggest a potential benefit of direct thrombin inhibi-
a significantly reduced risk of intracranial hemorrhage tion in HF patients in the setting of ACS.
(RR: 0.49, 95% CI: 0.36 to 0.66) (70). Based on the data Dabigatran. In the RE-LY trial (Randomized Evaluation
from the Danish National Patient Registry, the new anti- of Long-term anticoagulant therapY), a fixed dose
coagulants were superior to warfarin for net clinical benefit (110 mg or 150 mg twice daily) of dabigatran was
regardless of the risk of bleeding in patients at high risk of compared for noninferiorty for the prevention of stroke or
strokedCHADS(2) score 1 or CHA(2)DS(2)-VASc 2. systemic embolism versus adjusted-dose warfarin in pa-
The benefit was more prevalent when stroke risk and tients with AF. This trial demonstrated that 110 mg twice
bleeding risk were high (71). daily dose of dabigatran was associated with similar rates
Bivalirudin. In a pooled analysis of randomized trials of of stroke and systemic embolism and lower rates of major
bivalirudin therapy that included 14,258 ACS patients hemorrhage compared with warfarin whereas 150 mg
treated with dual antiplatelet therapy, bivalirudin therapy twice daily dose of dabigatran was associated with lower
compared with heparin plus glycoprotein (GP) IIb/IIIa in- rates of stroke and systemic embolism but similar rates of
hibitor therapy was significantly associated with lower major hemorrhage compared to warfarin. Among 4,904
mortality risk in patients with <35% LVEF (random RR: patients enrolled with symptomatic HF, there was no
0.47, 95% CI: 0.30 to 0.72, p ¼ 0.0004), which was significant interaction with treatment effect with both 100
consistent across studies. However, the benefit of bivalirudin and 150 mg doses of dabigatran (p for interaction ¼ 0.42
therapy was attenuated in patients with 35% LVEF and 0.33, respectively) (74).
(pooled analysis RR: 0.92, 95% CI: 0.72 to 1.17, p ¼ 0.50) Apixaban. In the ARISTOTLE trial (Apixaban for
(72). Similarly, in the PREMIER (Prospective Registry Reduction In STroke and Other ThromboemboLic Events
Evaluating Outcomes After Myocardial Infarctions: Events in atrial fibrillation), 5 mg twice daily apixaban was superior
and Recovery) registry, a 37% RR reduction in in-hospital to warfarin in the prevention of stroke or systemic embolism
JACC: Heart Failure Vol. 2, No. 1, 2014 Gurbel and Tantry 11
February 2014:1–14 Antiplatelet and Anticoagulant Agents in Heart Failure

Table 3 Trials of Antiplatelet Agents and New Oral Anticoagulants in Patients With Heart Failure

Name of the Study Patients Treatment Arms and Duration Primary Endpoints
COMMANDER HF HF (EF 40%) þCAD Rivaroxaban 2.5 mg bid vs. Time to the first occurrence of any of the
NCT01877915 (A Study to n ¼ 5,000 placebo for 6 to 30 months following: death from any cause, MI, or stroke
Assess the Effectiveness Time to the first occurrence of either fatal
and Safety of Rivaroxaban bleeding or bleeding into a critical space
in Reducing the Risk of with potential for permanent disability
Death, Myocardial Infarction
or Stroke in Patients With
Heart Failure and Coronary
Artery Disease Following
Hospitalization for Heart Failure)
Platelet Inhibition in Patients New York Heart Association Prasugrel 10 mg qd vs. The change in platelet aggregation measured
With Systolic Heart Failure functional class III to IV (EF35%) clopidogrel 75 mg qd for 2 weeks by the VerifyNow P2Y12 assay between
NCT01765400 n ¼ 50 baseline and each antiplatelet medication

CAD ¼ coronary artery disease; EF ¼ ejection fraction; qd ¼ once daily; other abbreviations as in Tables 1 and 2.

and was associated with less major bleeding and lower 2.5 mg twice daily dose was associated with a lower rate of
mortality compared to warfarin (target INR 2.0 to 3.0) in occurrence of the primary endpoint compared with placebo
patients with AF. One-third of patients had HF or reduced (10.1% vs. 16.8%). Although the confidence intervals were
LVEF in this trial and similar to the RE-LY trial, there was wide in the HF group, there was no difference in the first
no significant interaction with treatment effect (p for occurrence of treatment-emergent non–coronary artery
interaction ¼ 0.50) (75). bypass graft related TIMI major bleeding events. However,
In the APPRAISE-2 trial (Apixaban for Prevention of in patients without HF, there was no apparent difference in
Acute Ischemic Events 2), a 5 mg twice daily dose of the primary endpoint occurrence between the rivaroxaban
apixaban in addition to standard antiplatelet therapy was group and placebo (5.5% vs. 6.0%). There was higher non–
associated with no significant reduction in recurrent coronary artery bypass graft related TIMI major bleeding
ischemic events but an increased incidence of major bleeding (1.4% vs. 0.4%) in the rivaroxaban group (78).
events in patients with a recent ACS and at least 2 additional In a small study of patients with acute decompensated
risk factors for recurrent ischemic events. The trial was HF and patients with New York Heart Association functional
terminated prematurely for the high bleeding rate in the class III/IV HF, the pharmacodynamics and pharmacoki-
apixaban arm. In this trial, 28% of patients had HF. There netics of rivaroxaban were similar. In patients with functional
was a 23% reduction in the occurrence of the primary class III/IV HF treated with placebo, prothrombin fragment
endpoint with apixaban versus placebo (HR: 0.77, 95% CI: 1.2 increased over 7 days whereas rivaroxaban therapy (10 mg
0.58 to 1.02) in patients with HF compared to 5% increase once daily) was associated with a reduction in prothrombin
in the occurrence of the primary endpoint in patients fragment 1.2. A nonsignificant reduction in the rate of
without HF (HR: 1.05, 95% CI: 0.86 to 1.29, p for D-dimer and thrombin-antithrombin complex increase over
interaction ¼ 0.08) (76). time was also associated with rivaroxaban therapy (79).
Rivaroxaban. In the ROCKET AF (Rivaroxaban Once There are only 2 trials undergoing involving treatment
Daily Oral Direct Factor Xa Inhibition Compared with with antiplatelet and new oral anticoagulants as given in
Vitamin K Antagonism for Prevention of Stroke and Em- Table 3.
bolism Trial in Atrial Fibrillation), rivaroxaban was
compared to warfarin for the prevention of ischemic stroke Meta-Analysis of New Oral Anticoagulants
in AF patients; w60% of the population had HF. Rivar-
oxaban therapy was noninferior to warfarin for the preven- In a recent semisystematic review and meta-analysis of phase
tion of stroke or systemic embolism. Major bleeding was III trials (ARISTOTLE, RE-LY, and ROCKET-AF)
similar between the groups and rivaroxaban was associated including 44,563 patients with AF, new OAC treatment
with less frequent intracranial and fatal bleeding. In a sub- was not associated with a significant reduction in stroke and
group analysis, there was no significant interaction between systemic embolism compared with warfarin among patients
treatment with rivaroxaban and the presence of HF (p for with HF (n ¼ 21,095, OR: 0.91, 95% CI: 0.78 to 1.06, p ¼
interaction ¼ 0.419) (77). 0.22) but was associated with 24% reduction in patients
In the ATLAS ACS–TIMI 51 trial (Anti-Xa Therapy without HF (OR: 0.76, 95% CI: 0.67 to 0.87, p < 0.0001).
to Lower Cardiovascular Events in Addition to Standard The definitions of HF in the 3 trials were not uniform. In
Therapy in Subjects with Acute Coronary Syndrome– the RE-LY trial, HF was defined as ejection fraction <40%
Thrombolysis In Myocardial Infarction 51), 2 dose regimens by echocardiogram, radionuclide, or contrast angiogram
of rivaroxaban (2.5 mg twice daily and 5.0 mg twice daily) within 6 months, or symptoms of New York Heart Asso-
were compared with placebo in patients with ACS. Nearly ciation functional class >II HF within 6 months. In the
10% of patients had HF and in this subgroup, rivaroxaban ROCKET AF trial, the definition included symptoms of
12 Gurbel and Tantry JACC: Heart Failure Vol. 2, No. 1, 2014
Antiplatelet and Anticoagulant Agents in Heart Failure February 2014:1–14

Table 4 Guidelines

A. Heart Failure Society of America (81)


 Treatment with warfarin (goal international normalized ratio [INR] 2.0–3.0) is recommended for all patients with HF and chronic or documented paroxysmal, persistent, or long-
standing atrial fibrillation (Strength of Evidence 5 A) or a history of systemic or pulmonary emboli, including stroke or transient ischemic attack (Strength of Evidence 5 C), unless
contraindicated.
 It is recommended that patients with symptomatic or asymptomatic ischemic cardiomyopathy and documented recent large anterior MI or recent MI with documented
LV thrombus be treated with warfarin (goal INR 2.0–3.0) for the initial 3 months post-MI (Strength of Evidence 5 B) unless contraindicated.
 Other patients with ischemic or nonischemic cardiomyopathy and LV thrombus should be considered for chronic anticoagulation, depending on the characteristics of the
thrombus, such as its size, mobility, and degree of calcification. (Strength of Evidence 5 C).
 Long-term treatment with an antiplatelet agent, generally aspirin in doses of 75 to 81 mg, is recommended for patients with HF due to ischemic cardiomyopathy, whether or
not they are receiving ACE inhibitors. (Strength of Evidence 5 B)
 Warfarin (goal INR 2.0–3.0) and clopidogrel (75 mg) also have prevented vascular events in post- MI patients and may be considered as alternatives to aspirin.
(Strength of Evidence 5 B)
 Routine use of aspirin is not recommended in patients with HF without atherosclerotic vascular disease. (Strength of Evidence 5 C)
B. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012 (80)
 Other than in patients with AF (both HF-REF and HF-PEF), there is no evidence that an oral anticoagulant reduces mortality–morbidity compared with placebo or aspirin.
C. 2012 Consensus Document from the ESC Heart Failure Association and the ESC Working Group on Thrombosis (53)
 In the absence of a specific indication, such as documented coronary artery disease, aspirin should not be initiated.
 Given no overall benefit of warfarin on rates of death and stroke, with an increase in major bleedingddespite the potential for a reduction in ischemic stroked
there is currently no compelling reason to use warfarin routinely for all HF patients in sinus rhythm.
 Until more evidence becomes available, clinical decisions to treat patients with HF in sinus rhythm with anticoagulants must be made on a patient-by-patient basis, balancing
the individual benefits against the risks of treatment, especially amongst high risk patients.
 Clinical trials are needed to see if the new oral anticoagulants (oral direct thrombin inhibitors, oral Factor Xa inhibitors) that may offer a different risk–benefit profile compared
with warfarin could offer the reduction in ischemic stroke with less risk of major bleeding.
 Anticoagulation may potentially be considered by some clinicians in the following HF patient groups: HFrEF with previous thrombo-embolism (stroke, transient ischemic
attack, VTE), newly diagnosed intracardiac thrombus, and right heart failure with pulmonary hypertension, but evidence is limited and more research is needed to
ascertain the long-term risk–benefit ratio.

ACE ¼ angiotensin-converting enzyme; AF ¼ atrial fibrillation; ESC ¼ European Society of Cardiology; HF-PEF ¼ heart failure with preserved ejection fraction; HF-REF ¼ heart failure with reduced ejection
fraction; LV ¼ left ventricular; VTE ¼ venous thromboembolism; other abbreviations as in Table 1.

HF or an ejection fraction of 35%. The definition in the paroxysmal, persistent, or long-standing AF or a history of
ARISTOTLE trial was symptomatic HF 3 months or systemic or pulmonary emboli, including stroke or transient
ejection fraction 40%. The analysis was uncorrected for ischemic attack, unless contraindicated,” and “Antiplatelet
confounding variables (31). therapy is recommended to reduce vascular events in patients
with HF and CAD unless contraindicated” (81).
Guidelines
Conclusions
The European and American guidelines for the treatment of
patients with AF are presented in Table 4 (80–82). Ac- Heart failure is a common clinical problem with a prevalence
cording to the 2012 European Society of Cardiology of 1% to 2% in the general population that is increasing due
guidelines, “other than in patients with AF (both HF with to the aging population. Despite the heightened risk for
reduced and preserved ejection fraction), there is no evi- stroke and thromboembolism in patients with HF and AF,
dence that an OAC reduces mortality and morbidity in patients with systolic HF in sinus rhythm, the level of
compared with placebo or aspirin” (80). A similar view also increased thromboembolic risk is less clear. The decision to
has been provided by the recent consensus document from treat patients with HF with antiplatelet therapy remains
the European Society of Cardiology Heart Failure Associ- largely influenced by the presence or absence of concomitant
ation and the European Society of Cardiology Working arterial disease. Antithrombotic therapy has been shown to
Group on Thrombosis as follows: “Given no overall benefit be effective in many forms of cardiac disease, including
of warfarin on rates of death and stroke, with an increase in patients with HF and AF. However, the utility of oral
major bleedingddespite the potential for a reduction in anticoagulant and/or antiplatelet therapy has never been
ischemic strokedthere is currently no compelling reason to evaluated in an adequately powered dedicated clinical trial of
routinely use warfarin for all HF patients in sinus rhythm” HF patients in sinus rhythm. Although it is clear that
(53). The 2009 updated American College of Cardiology/ platelet activation and hypercoagulability are present in HF,
American Heart Association guidelines acknowledged the and there is evidence that stroke is reduced by warfarin
availability of limited data for the recommendation of OAC therapy in the HF patient, the available data suggest that the
use in patients with HF, but it recommends OAC in HF risk of major bleeding overshadows the antithromboembolic
patients with a previous thromboembolic event (82). benefit in HF patients in sinus rhythm. These findings have
Finally, the 2010 Heart Failure Society of America been reflected in the guidelines.
guidelines for HF or systolic dysfunction recommend that The CHADS2 and HAS-BLED risk scores may be
“Treatment with warfarin (goal INR 2.0 to 3.0) is recom- helpful to select patients for future studies of antithrombotic
mended for all patients with HF and chronic or documented therapy in the setting of HF and sinus rhythm. The window
JACC: Heart Failure Vol. 2, No. 1, 2014 Gurbel and Tantry 13
February 2014:1–14 Antiplatelet and Anticoagulant Agents in Heart Failure

of greatest advantage for antithrombotic therapy may be 16. Levine B, Kalman J, Mayer L, Fillit HM, Packer M. Elevated circu-
lating levels of tumor necrosis factor in severe chronic heart failure.
early after initial presentation, when a markedly increased N Engl J Med 1990;323:236–41.
risk of death, and death or stroke, has been reported. 17. Rauchhaus M, Doehner W, Francis DP, et al. Plasma cytokine pa-
Selecting patients for enrollment may also be facilitated by rameters and mortality in patients with chronic heart failure. Circula-
tion 2000;102:3060–7.
objective assessments of thrombogenicity using laboratory 18. Lowe GD. Virchow’s triad revisited: abnormal flow. Pathophysiol
testing. Thromboelastography is a potential method that Haemost Thromb 2003;33:455–7.
determines platelet function in addition to thrombin gen- 19. Nesbitt WS, Mangin P, Salem HH, Jackson SP. The impact of blood
rheology on the molecular and cellular events underlying arterial
eration and the viscoelastic characteristics of the platelet- thrombosis. J Mol Med 2006;84:989–95.
fibrin clot. Large-scale, adequately, powered trials that 20. Lopez JA, Kearon C, Lee AY. Deep venous thrombosis. Hematology
have refined selection criteria, possible including laboratory Am Soc Hematol Educ Program 2004:439–56.
21. Kroegel C, Reissig A. Principle mechanisms underlying venous
testing, will be needed to establish the role of antithrombotic thromboembolism; epidemiology, risk factors, pathophysiology and
therapy in the HF patient in sinus rhythm. pathogenesis. Respiration 2003;70:7–30.
22. Chirinos JA, Heresi GA, Velasquez H, et al. Elevation of endothelial
microparticles, platelets, and leukocyte activation in patients with
Reprint requests and correspondence: Dr. Paul A. Gurbel, Sinai venous thromboembolism. J Am Coll Cardiol 2005;45:1467–71.
Center for Thrombosis Research, Cardiac Catheterization Labo- 23. Mackman N. New insights into the mechanisms of venous thrombosis.
ratory, 2401 West Belvedere Avenue, Baltimore, Maryland 21215. J Clin Invest 2012;122:2331–6.
E-mail: pgurbel@lifebridgelealth.org. 24. Carpio FD, Chirinos JA, Jimenez J. Endothelial, platelet and leukocyte acti-
vation in patients with congestive heart failure. J Card Fail 2004;10:S55–65.
25. Bourassa MG, Gurne O, Bangdiwala SI, et al. Natural history and
patterns of current practice in heart failure. J Am Coll Cardiol 1993;22:
14A–9A.
REFERENCES 26. Zannad F, McMurray JJ, Krum H, et al. EMPHASIS-HF Study
Group. Eplerenone in patients with systolic heart failure and mild
1. Go AS, Mozaffarian D, Roger VL, et al. American Heart Association symptoms. N Engl J Med 2011;364:11–21.
Statistics Committee and Stroke Statistics Subcommittee. Heart dis- 27. Tavazzi L, Maggioni AP, Marchioli R, et al. Effect of rosuvastatin in
ease and stroke statisticsd2013 update: a report from the American patients with chronic heart failure (the GISSI-HF trial): a randomised,
Heart Association. Circulation 2013;127:e6–245. double-blind, placebo-controlled trial. Lancet 2008;372:1231–9.
2. Zannad F, Stough WG, Regnault V, et al. Is thrombosis a contributor 28. Freudenberger RS, Hellkamp AS, Halperin JL, et al. SCD-HeFT
to heart failure pathophysiology? Possible mechanisms, therapeutic Investigators. Risk of thromboembolism in heart failure: an analysis
opportunities, and clinical investigation challenges. Int J Cardiol 2013; from the Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT).
167:1772–82. Circulation 2007;115:2637–41.
3. Witt BJ, Gami AS, Ballman KV, et al. The incidence of ischemic stroke 29. Vemmos K, Ntaios G, Savvari P, et al. Stroke aetiology and predictors
in chronic heart failure: a meta-analysis. J Card Fail 2007;13:489–96. of outcome in patients with heart failure and acute stroke: a 10-year
4. Pullicino PM, Halperin JL, Thompson JL. Stroke in patients with follow-up study. Eur J Heart Fail 2012;14:211–8.
heart failure and reduced left ventricular ejection fraction. Neurology 30. Stroke Prevention in Atrial Fibrillation Investigators. Risk factors for
2000;54:288–94. thromboembolism during aspirin therapy in patients with atrial fibril-
5. Vandvik PO, Lincoff AM, Gore JM, et al., American College of Chest lation: the Stroke Prevention in Atrial Fibrillation Study. J Stroke
Physicians. Primary and secondary prevention of cardiovascular disease: Cerebrovasc Dis 1995;5:147–57.
Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: 31. Ahmad Y, Lip GY, Apostolakis S. New oral anticoagulants for stroke
American College of Chest Physicians Evidence-Based Clinical Prac- prevention in atrial fibrillation: impact of gender, heart failure, diabetes
tice Guidelines. Chest 2012;141:e637S–68S. mellitus and paroxysmal atrial fibrillation. Expert Rev Cardiovasc Ther
6. Gurbel PA, Bliden KP, Hayes KM, Tantry U. Platelet activation in myo- 2012;10:1471–80.
cardial ischemic syndromes. Expert Rev Cardiovasc Ther 2004;2:535–45. 32. Mehta J, Mehta P. Platelet function studies in heart disease. VI.
7. Mann KG. Thrombin generation in hemorrhage control and vascular Enhanced platelet aggregate formation activity in congestive heart fail-
occlusion. Circulation 2011;124:225–35. ure: inhibition by sodium nitroprusside. Circulation 1979;60:497–503.
8. O’Connor CM, Gurbel PA, Serebruany VL. Usefulness of soluble and 33. Lip GY, Rasmussen LH, Skjøth F, et al. Stroke and mortality in pa-
surface bound P-selectin in detecting heightened platelet activity in tients with incident heart failure: the Diet, Cancer and Health (DCH)
patients with chronic heart failure. Am J Cardiol 1999;83:1345–9. cohort study. BMJ Open 2012;2(4).
9. Serebruany V, McKenzie M, Meister A, et al. Whole blood impedance 34. Alberts VP, Bos MJ, Koudstaal P, et al. Heart failure and the risk of
aggregometry for the assessment of platelet function in patients with stroke: the Rotterdam Study. Eur J Epidemiol 2010;25:807–12.
congestive heart failure (EPCOT Trial). Eur J Heart Fail 2002;4:461–7. 35. Loh E, Sutton MS, Wun CC, et al. Ventricular dysfunction and the risk
10. De Lorenzo F, Saba N, Kakkar VV. Blood coagulation in patients with of stroke after myocardial infarction. N Engl J Med 1997;336:251–7.
chronic heart failure: evidence for hypercoagulable state and potential 36. Piazza G, Goldhaber SZ, Lessard DM, et al. Venous thromboembo-
for pharmacological intervention. Drugs 2003;63:565–76. lism in heart failure: preventable deaths during and after hospitaliza-
11. O’Connor CM, Gurbel PA, Serebruany VL. Usefulness of soluble and tion. Am J Med 2011;124:252–9.
surface-bound P-selectin in detecting heightened platelet activity in 37. Khoury H, Welner S, Kubin M, et al. Disease burden and unmet needs
patients with congestive heart failure. Am J Cardiol 1999;83:1345–9. for prevention of venous thromboembolism in medically ill patients
12. Stumpf C, Lehner C, Eskafi S, et al. Enhanced levels of CD154 in Europe show underutilisation of preventive therapies. Thromb
(CD40 ligand) on platelets in patients with chronic heart failure. Eur J Haemost 2011;106:600–8.
Heart Fail 2003;5:629–37. 38. Dunkman WB, Johnson GR, Carson PE, Bhat G, Farrell L, Cohn JN, the
13. Alehagen U, Dahlstrom U, Lindahl TL. Elevated D-dimer level is an in- V-HeFT VA Cooperative Studies Group. Incidence of thromboembolic
dependent risk factor for cardiovascular death in out-patients with symp- events in congestive heart failure. Circulation 1993;87:VI94–VI101.
toms compatible with heart failure. Thromb Haemost 2004;92:1250–8. 39. Gurbel PA, Tantry US. Do platelet function testing and genotyping
14. Molvarec A, Rigó J Jr., Bõze T. Increased plasma von Willebrand factor improve outcome in patients treated with antithrombotic agents?:
antigen levels but normal von Willebrand factor cleaving protease Platelet function testing and genotyping improve outcome in patients
(ADAMTS13) activity in preeclampsia. Thromb Haemost 2009;101: treated with antithrombotic agents. Circulation 2012;125:1276–87.
305–11. 40. Gurbel PA, Bliden KP, Guyer K, et al. Platelet reactivity in patients
15. Nawroth PP, Stern DM. Modulation of endothelial cell hemostatic and recurrent events post-stenting: results of the PREPARE POST-
properties by tumor necrosis factor. J Exp Med 1986;163:740–5. STENTING Study. J Am Coll Cardiol 2005;46:1820–6.
14 Gurbel and Tantry JACC: Heart Failure Vol. 2, No. 1, 2014
Antiplatelet and Anticoagulant Agents in Heart Failure February 2014:1–14

41. Gurbel PA, Tantry US. Combination antithrombotic therapies. Cir- 63. Massie BM, Collins JF, Ammon SE, et al., WATCH Trial Investigators.
culation 2010;121:569–83. Randomized trial of warfarin, aspirin, and clopidogrel in patients with
42. Diener HC, Bogousslavsky J, Brass LM, et al. MATCH Investigators. chronic heart failure: the Warfarin and Antiplatelet Therapy in Chronic
Aspirin and clopidogrel compared with clopidogrel alone after recent Heart Failure (WATCH) trial. Circulation 2009;119:1616–24.
ischemic stroke or transient ischaemic attack in high-risk patients 64. Homma S, Thompson JLP, Pullicino PM, et al., WARCEF In-
(MATCH): randomized, double-blind, placebo-controlled trial. Lan- vestigators. Warfarin and aspirin in patients with heart failure and sinus
cet 2004;364:331–7. rhythm. N Engl J Med 2012;366:1859–69.
43. ESPRIT Study Group, Halkes PH, van Gijn J, Kappelle LJ, 65. Eikelboom JW, Connolly SJ. Warfarin in heart failure. N Engl J Med
Koudstaal PJ, Algra A. Aspirin plus dipyridamole versus aspirin alone 2012;366:1936–8.
after cerebral ischaemia of arterial origin (ESPRIT): randomised 66. Homma S. Warfarin versus aspirin in patients with reduced cardiac ejec-
controlled trial. Lancet 2006;367:1665–73. tion fraction (WARCEF) trial. Paper presented at: Heart Failure Society
44. Sacco RL, Diener HC, Yusuf S, et al. PRoFESS Study Group. Aspirin of America 2012 Scientific Meeting; September 10, 2012; Seattle, WA.
and extended-release dipyridamole versus clopidogrel for recurrent 67. Hopper I, Skiba M, Krum H. Updated meta-analysis on anti-
stroke. N Engl J Med 2008;359:1238–51. thrombotic therapy in patients with heart failure and sinus rhythm.
45. Baigent C, Blackwell L, Collins R, et al., Antithrombotic Trialists’ Eur J Heart Fail 2013;15:69–78.
(ATT) Collaboration. Aspirin in the primary and secondary prevention 68. Kumar G, Goyal MK. Warfarin versus aspirin for prevention of stroke
of vascular disease: collaborative meta-analysis of individual participant in heart failure: a meta-analysis of randomized controlled clinical trials.
data from randomised trials. Lancet 2009;373:1849–60. J Stroke Cerebrovasc Dis 2013;22:1279–87.
46. Hart RG, Pearce LA, Aguilar MI. Meta-analysis: antithrombotic 69. Avellana P, Segovia J, Ferrero A, Vázquez R, et al. Anticoagulation
therapy to prevent stroke in patients who have nonvalvular atrial therapy in patients with heart failure due to systolic dysfunction and
fibrillation. Ann Intern Med 2007;146:857–67. sinus rhythm: analysis of REDINSCOR registry. Rev Esp Cardiol
47. You JJ, Singer DE, Howard PA, Lane DA, et al. American College of 2012;65:705–12.
Chest Physicians. Antithrombotic therapy for atrial fibrillation: 70. Miller CS, Grandi SM, Shimony A, et al. Meta-analysis of efficacy and
Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: safety of new oral anticoagulants (dabigatran, rivaroxaban, apixaban)
American College of Chest Physicians Evidence-Based Clinical Prac- versus warfarin in patients with atrial fibrillation. Am J Cardiol 2012;
tice Guidelines. Chest 2012;141:e531S–75S. 110:453–60.
48. Eriksson N, Wadelius M. Prediction of warfarin dose: why, when and 71. Banerjee A, Lane DA, Torp-Pedersen C, Lip GY. Net clinical benefit of
how? Pharmacogenomics 2012;13:429–40. new oral anticoagulants (dabigatran, rivaroxaban, apixaban) versus no
49. Hylek E, Singer D. Risk factors for intracranial hemorrhage in out- treatment in a ’real world’ atrial fibrillation population: a modelling analysis
patients taking warfarin. Ann Intern Med 1994;120:897–902. based on a nationwide cohort study. Thromb Haemost 2012;107:584–9.
50. Oden A, Fahlen M, Hart RG. Optimal INR for prevention of stroke 72. Dangas G, Mehran R, Feit F, et al. Impact of bivalirudin therapy on
and death in atrial fibrillation: a critical appraisal. Thromb Res 2006; mortality in patients with high risk features undergoing PCI: a patient-
117:493–9. level pooled analysis from the REPLACE-2, ACUITY, and
51. De Caterina R, Husted S, Wallentin L, et al., Coordinating Com- HORIZONS-AMI trials (abstr). Eur Heart J 2011;32 Suppl:232–3.
mittee. New oral anticoagulants in atrial fibrillation and acute coronary 73. Kohli P, Chakrabarti A, Bansilal S, et al. on behalf of TIMI Study
syndromes: ESC Working Group on Thrombosis-Task Force on Group. Reduction in mortality from thrombin inhibition in CHF
Anticoagulants in Heart Disease position paper. J Am Coll Cardiol patients: results from the premier hospital database (abstr.). Eur Heart J
2012;59:1413–25. 2013;33 Suppl 1:393.
52. Weitz JI. Factor Xa and thrombin as targets for new oral anticoagu- 74. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin
lants. Thromb Res 2011;127:S5–12. in patients with atrial fibrillation. N Engl J Med 2009;361:1139–51.
53. Lip GY, Ponikowski P, Andreotti F, et al., ESC Task Force. 75. Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin
Thrombo-embolism and antithrombotic therapy for heart failure in in patients with atrial fibrillation. N Engl J Med 2011;365:981–92.
sinus rhythm. A joint consensus document from the ESC Heart Failure 76. Alexander JH, Lopes RD, James S, et al., APPRAISE-2 Investigators.
Association and the ESC Working Group on Thrombosis. Eur J Heart Apixaban with antiplatelet therapy after acute coronary syndrome.
Fail 2012;14:681–95. N Engl J Med 2011;365:699–708.
54. Wishart JH, Chapman CB. Dicumarol therapy in congestive heart 77. Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in
failure. N Engl J Med 1948;239:702–4. nonvalvular atrial fibrillation. N Engl J Med 2011;365:883–91.
55. Harvey WP, Finch CA. Dicumarol prophylaxis of thromboembolic 78. Mega JL, Braunwald E, Wiviott SD, et al., ATLAS ACS 2–TIMI 51
disease in congestive heart failure. N Engl J Med 1950;242:208–11. Investigators. Rivaroxaban in patients with a recent acute coronary
56. Griffith GC, Stragnell R, Levinson DC. A study of the beneficial ef- syndrome. N Engl J Med 2012;366:9–19.
fects of anticoagulant therapy in congestive heart failure. Ann Intern 79. Gheorghiade M, Thyssen A, Zolynas R, et al. Pharmacokinetics and
Med 1952;37:867–87. pharmacodynamics of rivaroxaban and its effect on biomarkers of hy-
57. Al-Khadra AS, Salem DN, Udelson JE, et al. Warfarin anticoagulation percoagulability in patients with chronic heart failure. J Heart Lung
and survival: a cohort analysis from the Studies of Left Ventricular Transplant 2011;30:218–26.
Dysfunction. J Am Coll Cardiol 1998;31:749–53. 80. McMurray JJ, Adamopoulos S, Anker SD, et al., ESC Committee for
58. Leor J, Reicher-Reiss H, Goldbourt U, et al. Aspirin and mortality in Practice Guidelines. ESC guidelines for the diagnosis and treatment of
patients treated with angiotensin-converting enzyme inhibitors: a acute and chronic heart failure 2012: the Task Force for the Diagnosis
cohort study of 11,575 patients with coronary artery disease. J Am Coll and Treatment of Acute and Chronic Heart Failure 2012 of the
Cardiol 1999;33:1920–5. European Society of Cardiology. Developed in collaboration with the
59. Nguyen KN, Aursnes I, Kjekshus J. Interaction between enalapril and Heart Failure Association (HFA) of the ESC. Eur J Heart Fail 2012;
aspirin on mortality after acute myocardial infarction: subgroup analysis 14:803–69.
of the Cooperative New Scandinavian Enalapril Survival Study II 81. Heart Failure Society of America, Lindenfeld J, Albert NM,
(CONSENSUS II). Am J Cardiol 1997;79:115–9. Boehmer JP, et al. HFSA 2010 comprehensive heart failure Practice
60. Connolly S, Pogue J, Hart R, et al., ACTIVE Writing Group of the guideline. J Card Fail 2010;16:e1–194.
ACTIVE Investigators. Clopidogrel plus aspirin versus oral anti- 82. Hunt SA, Abraham WT, Chin MH, et al. 2009 focused update
coagulation for atrial fibrillation in the Atrial fibrillation Clopidogrel incorporated into the ACC/AHA 2005 guidelines for the diagnosis
Trial with Irbesartan for prevention of Vascular Events (ACTIVE W): and management of heart failure in adults: a report of the American
a randomised controlled trial. Lancet 2006;367:1903–12. College of Cardiology Foundation/American Heart Association Task
61. Cleland JGF, Findlay I, Jafri S, et al. The Warfarin/Aspirin Study in Force on Practice Guidelines; developed in collaboration with the In-
Heart Failure (WASH): a randomized trial comparing antithrombotic ternational Society for Heart and Lung Transplantation. J Am Coll
therapies for patients with heart failure. Am Heart J 2004;148:157–64. Cardiol 2009;53:e1–90.
62. Cokkinos DV, Haralabopoulos GC, Kostis JB, Toutouzas PK. Efficacy
of antithrombotic therapy in chronic heart failure: the HELAS study.
Eur J Heart Fail 2006;8:428–32. Key Words: anticoagulant - antiplatelet - heart failure.

Vous aimerez peut-être aussi