Abstract
Atrial fibrillation is the commonest arrhythmia worldwide and is a growing problem. AF is responsible for 25% of all strokes, and these patients suffer greater mortality and disability. Warfarin has traditionally been the only successful therapy for stroke prevention, but its limitations have resulted in underutilisation. Major progress has been made in AF research, leading to improved management strategies. Better risk stratification permits identification of truly low-risk patients who do not require anticoagulation and we are able to simplify ourevaluation of a patient’s bleeding risk.
The advent of novel anticoagulants means warfarin is no longer the only choice for stroke prophylaxis. These drugs circumvent many of warfarin’s inconveniences, but onlylong-term study and use will conclusively demonstrate how they compare to warfarin. The landscape of stroke prevention in AF has changed with effective alternatives to warfarin available for the first time in 60 years—but each new option brings new considerations.
Keywords: atrial fibrillation, warfarin, stroke prevention, oral anticoagulants
Introduction
Atrial fibrillation (AF) is the most common arrhythmia encountered worldwide, affecting 1% of the general population1 with a lifetime risk of 25% for adults.2 The prevalence of AF increases with age,3,4 with almost one in ten patients over 80 affected.1 AF is a growing problem and it is estimated that between 6 million and 16 million people in the US will suffer with the rhythm disorder by 2050.1,5 These projected increases are owed largely to the ageing population, and the increased survival of patients with chronic cardiac disorders which predispose to AF—such as ischaemic heart disease and heart failure.6,7 The presence of AF in these patients with underlying heart disease is associated with a worse outcome,8 and increased healthcare costs: AF is responsible for an estimated 1% of the United Kingdom National Health Service expenditure in the UK.9
The most feared and devastating complication of AF is stroke. The presence of AF is an independent risk factor for stroke, conferring a five-fold increase in the stroke risk.10 AF is held responsible for up to a quarter of all strokes,11 and these patients have more severe strokes: greater mortality, longer hospital stay, increased disability.12–14 AF is also a significant risk factor for stroke recurrence.15 There is substantial evidence to suggest that most thromboembolic complications could be prevented with adequate pharmacological anticoagulation.16,17
Traditionally, vitamin K antagonists (eg, warfarin) were the only available therapeutic option for oral anticoagulation. Warfarin provides highly effective prophylaxis against thromembolism.18 Unfortunately, warfarin has many limitations and inconveniences19 which have contributed to reluctance amongst both patients and physicians to use this therapy.20 This variability in the management of stroke prevention in AF has seen renewed efforts in the past decade to establish validated risk stratification models and novel alternative antithrombotic therapies. The progress made in this sphere has offered increased guidance and choice to both clinicians and patients to improve the prevention of stroke in atrial fibrillation.
This article reviews the current treatment options available for stroke prevention in AF, their evidence base, safety and efficacy. Attention is given to the novel oral anticoagulants which are available now or soon to become available, and their potential place in therapy.
Risk Stratification
The risk of stroke in AF is dependent upon the presence or absence of several risk factors.21,22 Traditionally these risk factors were used to stratify patients into “low”, “intermediate”, or “high” risk for stroke. Older guidelines used this grouping to recommend oral anticoagulation to high-risk patients, aspirin for low-risk patients, and a choice of either anticoagulation or aspirin for the intermediate grouping. This had the potential of introducing confusion (for example, for intermediate-risk patients no definitive guidance was given as to whether to offer anticoagulation or aspirin) and also undertreating a cohort of patients at substantial risk of stroke.
There is evidence that aspirin does not reduce the risk of stroke in low-risk patients,23 and that warfarin is superior to aspirin for patients at intermediate risk of stroke.24,25 The CHADS2 score26 also classified a large number of patients into the intermediate group. These limitations spurred on the development of a risk stratification system that more reliably identifies truly low-risk patients, and minimises patients being denied oral anticoagulation when they would derive significant benefit from it.
The CHA2DS2VASc score [Table 1] was suggested as such a scheme to improve risk stratification for stroke, to focus more on the identification of such ‘truly low risk’ patients.27 The CHA2DS2VASc scoreis better at identifying truly low-risk patients, and categorises fewer patients as intermediate risk.28 It has now been validated in various large real-world cohort of patients29 and may even performbetter than CHADS2 in identifying patients at high-risk of stroke. The CHA2DS2VASc score is now included in European guidelines on the management of atrial fibrillation.30
Table 1.
Risk factor | Score |
---|---|
Congestive cardiac failure | 1 |
Hypertension | 1 |
Age ≥75 | 2 |
Diabetes mellitus | 1 |
Stroke/TIA/thromboembolism | 1 |
Vascular disease | 1 |
Age 65–74 | 1 |
Female sex | 1 |
Maximum score | 9 |
Notes: A score of 0 indicates low risk; 1 indicates moderate risk; ≥2 indicates high risk. Congestive cardiac failure is defined as left ventricular ejection fraction ≤40%. Hypertension is defined as blood pressure consistently above 140/90 mmHg or treated hypertension on medication. Vascular disease is defined as previous myocardial infarction, peripheral arterial disease or aortic plaque.
Bleeding is the most important and feared complication of anticoagulant therapy among clinicians and patients. Bleeding risk is a limiting factor in the prescription of antithrombotic therapy, and leaves a substantial number of patients untreated when they have clear indications for anticoagulation.31 Clinicians should undertake an assessment of a patient’s risk for bleeding before initiating anticoagulant therapy.32
The novel HAS-BLED score33 was developed to allow clinicians to assess simply and practically assess the individual risk of bleeding in their patients before initiating antithrombotic therapy, and makes clinicians think about the common correctable risk factors for bleeding, for example, uncontrolled blood pressure, concomitant aspirin/NSAID use with oral anticoagulation, labile INRs, etc [Table 2]. It allows periodic reassessment of a patient’s bleeding risk considers the quality of the anticoagulation control.34 This risk score has been validated in a large cohort of real-world patients,35 and performs favourably when compared to other scoring schemes.36 The HAS-BLED score has also been included in European guidelines,30 and when used in conjunction with the CHA2DS2VASc score it allows clinicians to make a simple and informed judgment as to the relative benefits and risks of anticoagulation.
Table 2.
Risk factor | Score |
---|---|
Hypertension | 1 |
Abnormal renal/liver function | 1 or 2 |
Stroke | 1 |
Bleeding tendency | 1 |
Labile INR | 1 |
Age (eg. >65) | 1 |
Drugs (eg. concomitant aspirin, NSAIDs, etc) or alcohol | 1 or 2 |
Maximum score | 9 |
Notes: A score of 0–2 indicates low risk of bleeding; a score of ≥3 indicates high risk of bleeding. Hypertension is defined as a systolic blood pressure >160 mmHg. 1 point is awarded for each of abnormal renal or liver function, and drugs or alcohol.
The Ideal Anticoagulant
The efficacy of warfarin as prophylaxis against stroke is established and unequivocal.18,37 Unfortunately, there are many limitations associated with warfarin [Table 3]: its narrow therapeutic window, slow onset and offset of action, unpredictable pharmacokinetics and pharmacodynamics leading to variability in dose response amongst individuals and multiple drug and food interactions. Due to these factors, warfarin requires close laboratory monitoring of coagulation via the INR and subsequent dose adjustments. These regular clinic attendances bring an increased financial burden and inconvenience to patients. Thus many patients who are eligible for warfarin choose not to use it.38
Table 3.
|
A clinically viable alternative to warfarin will need to possess several key characteristics.39,40 Novel agents [Table 4] need to be proven to be predictably at least as effective as warfarin in clinical trials. Other key features include: oral administration, fixed dose regimens, wide therapeutic windows, low propensity for food and drug interactions, predictable pharmacokinetics and pharmacodynamics with little inter and intra patient variability [Table 5]. New therapies would of course need to be safe and well-tolerated, with low frequency and severity of adverse effects. They should also obviate the need for regular coagulation monitoring.
Table 4.
Dabigatran | Rivaroxaban | Apixaban | |
---|---|---|---|
Mechanism of action | Direct thrombin inhibitor | Direct factor Xa inhibitor | Direct factor Xa inhibitor |
Prodrug | Double prodrug | No | No |
Dosing frequency | Twice daily | Once daily | Twice daily |
Bioavailability % | 6.5 | 50 | 80 |
Tmax | 2 hours | 2–4 hours | 3 hours |
Half-life | 17 hours with multiple doses, 7–9 hours with single doses | 9 hours in healthy subjects, 12 hours in elderly subjects | 12 hours |
Mode of excretion | 80% cleared renally | One-third cleared renally, two-thirds metabolised by the liver | 70% cleared in faeces, 25% cleared renally |
Age effect | Affects pharmacokinetic parameters | No | No |
Drug interactions | Interaction with aspirin at high doses | None reported | None reported |
Table 5.
|
Mechanism of Action and Pharmacokinetic Profile
Warfarin
Warfarin is a vitamin-K antagonist that produces its anticoagulant effect by interfering with the cyclic interconversion of vitamin K and its epoxide. Vitamin K is a cofactor for the posttranslational carboxylation of glutamate residues of vitamin K-dependent clotting factors(II, VII, IX, X).41,42 These coagulation factors require carboxylation to be biologically active, therefore when warfarin inhibits the vitamin K conversion cycle it leads to hepatic synthesis of decarboxylated (or partially carboxylated) proteins with reduced coagulant activity.43 The effect of warfarin can be counteracted by vitamin K1 (either ingested in food or administered therapeutically) and this effect may persist for up to a week as vitamin K accumulates in the liver.
Warfarin has a high bioavailability,44 is absorbed quickly and reaches maximal plasma concentrations within 90 minutes.45 Warfarin has a half-life of 36-hours and predominantly circulates bound to albumin. Warfarin accumulates in the liver where it is metabolised by two pathways. The dose-response of warfarin is impacted on by environmental and genetic factors. Polymorphisms of genes that encode for the vitamin-K epoxide reductase enzyme and CYP2C9 enzyme have been identified as the most important contributors to the wide inter-individual variations in dose requirements.46–48 Drugs may influence the pharmacokinetics of warfarin by reducing GI absorption or interfering with metabolic clearance;49 drugs may also disrupt the pharmacodynamics of warfarin by inhibiting synthesis or increasing clearance of vitamin K-dependent clotting factors. Dietary intake of vitamin K can also impact on the anticoagulant effect of warfarin.50
Direct Thrombin Inhibitors
The final step of the coagulation pathway requires thrombin to convert fibrinogen to fibrin. Direct thrombin inhibitors bind to thrombin and prevent its interaction with substrates; this inhibits fibrin production.51 The effect of this class of drugs also prevents thrombin-mediated activation of activation of Factors V, VIII, XI, and XIII, and thrombin-induced platelet-aggregation.52 Direct thrombin inhibitors can inhibit clot-bound and free thrombin, owing to the fact they bind directly to the active catalytic site.53 Numerous parenteral direct thrombin inhibitors are available (such as bivalirudin, hirudin and lepirudin) but the lack of an oral preparation does not lend them to use in lifelong stroke prevention for patients with AF.
Ximelegatran was the first available oral direct thrombin inhibitor.54 It is a prodrug that is rapidly converted to melegatran (its active form).55 Ximelegatran had twice daily fixed dosing with a fast onset and offset of action. There were no food interactions,56 little potential for drug interactions,57 and low variability in the dose-response relationship.58 Ximelegatran was withdrawn from the market in 2004 due to its potential to cause raised liver enzymes and some reported cases of fulminant hepatic failure.59
Dabigatran etexilate is an oral prodrug which is converted in the liver to its active compound, dabigatran.60 Dabigatran is a competitive, direct and reversible inhibitor of thrombin.52 As detailed above, dabigatran exerts an effect on both clot-bound and free thrombin. Dabigatran has a fast onset of action (peak 0.5–4 hours), a half-life of 17 hours with multiple doses (7–9 hours with single doses),62 and reaches clinical steady state within 2.5 days of initiation.61 Dabigatran is predominantly (80%) cleared by the kidneys.61 Neither the prodrug nor its metabolite exert an effect on the CYP system, thus dabigatran lends itself to fewer food and drug interactions than warfarin. Absorption of dabigatran may be delayed by food,63 and there is an age effect on pharmacokinetic parameters64 although no reported gender effect.65
Factor Xa Inhibitors
Factor Xa plays a key role in the common pathway of the clotting cascade. This protein is located at the junction of the intrinsic and extrinsic pathway, leading to the conversion of prothrombin to factor IIa (thrombin). Thrombin then converts fibrinogen to fibrin, leading to clot formation. Parenterally administered heparin-based anticoagulants act by binding to antithrombin and producing a conformational change to inhibit both thrombin and factor X.66 Fondaparinux inhibits factor Xa indirectly, utilising antithrombin as a cofactor and producing a conformational change similar to heparin to inhibit factor Xa—but it can only inhibit factor Xa in its free form.67 Trials have demonstrated the efficacy of heparin-based anticoagulants improves with greater selectivity for FXa.68,69
The emerging direct factor Xa inhibitors do not require antithrombin as a cofactor, and bind directly to the active site of factor Xa.70 Factor Xa has fewer effects outside of the clotting cascade, so may cause fewer side-effects than direct thrombin inhibitors71 (although direct thrombin inhibitors may also have beneficial effects outside the coagulation pathway35).
Apixaban is an oral, selective, reversible72 direct factor Xa inhibitor.73 It has high oral bioavailability74 and onset of action is within three hours.60 The drug has a half-life of 12 hours75 and is cleared via multiple pathways: 25% renal, 75% in faeces.74 Data indicate that apixaban does not inhibit or induce CYP enzymes.77 Its absorption is not impacted by food.77
Rivaroxaban is an oral, reversible, direct factor Xa inhibitor.78 It inhibits prothrombinase activity, as well as free and clot-associated FXa activity in plasma.79 It has high oral bioavailability80 and is rapidly absorbed with a half-life of 9 hours in healthy subjects81 and 12 hours in those aged over 75.82 It has a rapid onset of action with maximal concentrations reached between 2 and 4 hours.80 Rivaroxaban has a dual mode of elimination with no identified active circulating metabolite: one-third of the dose is renally cleared, the remainder being metabolized by the liver.83 The pharmacokinetics of rivaroxaban are dose-proportional84 and generally unaffected by gender, body weight or extremes of age.85 Although rivaroxaban can theoretically be affectedby drugs that interact with CYP3A4,76 a low potential for clinically significant drug or food interactions has been reported.86–88
Clinical Studies: Efficacy and Safety
Warfarin
It was conclusively proved in a series of randomised control trials in the late eighties and early nineties that warfarin was effective prophylaxis for stroke prevention in atrial fibrillation when compared to placebo.88–93 In 1994 pooled data from five studies demonstrated that warfarin conferred a 68% risk reduction in stroke compared with no therapy, and with no increased risk of bleeding.94 A meta-analysis in 2002 showed that warfarin significantly reduced ischaemic stroke when compared to aspirin,95 and this was confirmed in 2007 when a further analysis of almost 30,000 patients revealed that warfarin reduced strokes by 40% compared to aspirin.96 The benefit of warfarin over aspirin was preserved when trials were confined to an elderly population.97
The ACTIVE-W trial98 in 2006 sought to identify whether a combination of aspirin and clopidogrel would be non-inferior when compared to warfarin. The study showed clearly that warfarin was superior to aspirin and clopidogrel, and was stopped early due to the clear benefit of oral anticoagulation. The rates of major bleeding in the two study groups were comparable (2.4% per annum for clopidogrel and aspirin vs. 2.2% per annum for warfarin). In the ACTIVE-A trial99 patients deemed unsuitable for warfarin were given either aspirin alone or a combination of aspirin and clopidogrel. Aspirin plus clopidogrel reduced the rate of ischaemic stroke by 28% compared with aspirin alone. However, it is worth noting that the rates of major bleeding with aspirin and clopidogrel were 2.0% in the ACTIVE-A study group. This figure is similar to the major bleeding rate of dual antiplatelet therapy observed in ACTIVE-W, and also to the rate of major bleeding with warfarin. Therefore, aspirin and clopidogrel in combination would not be a suitable alternative to warfarin in patients deemed high-risk for haemorrhage. Dual antiplatelet therapy may, however, be a therapeutic option for patients truly ineligible for warfarin for other reasons100 (inability or unwillingness to undergo regular monitoring, for example).
Dabigatran
Dabigatran was initially evaluated in 2007 in the phase II trial PETRO:101 in this trial, 502 patients with nonvalvular AF were randomized to dabigatran 50, 150, or 300 mg twice/day alone or combined with aspirin 81 or 325 mg/day versus open-label warfarin in patients with a CHADS2 score of 1 or higher. Major bleeding was more common in patients taking dabigatran 300 mg with aspirin (6.25%) compared with dabigatran 300 mg alone (0%, P < 0.02). Thromboembolism was only observed in patients randomised to dabigatran 50 mg.
The RE-LY trial was a large randomised controlled trial comparing dabigatran with warfarin.102 It was a phase III, blinded, noninferiority trial in 18,113 patients with nonvalvular AF with a CHADS2 score of 1 or higher or who were older than 65 years with coronary artery disease.103 Patients were randomised to either dabigatran, at a dosage of 110 or 150 mg twice daily or warfarin titrated to a goal INR of 2–3. The primary efficacy outcomes of the study included stroke or systemic embolism. Efficacy outcomes occurred at 1.69% per year in patients assigned to warfarin compared with 1.53% in the dabigatran 110-mg group (RR 0.91, CI 0.74–1.11, P < 0.001 for noninferiority, P = 0.34 for superiority compared with warfarin) and 1.11% in the dabigatran 150-mg group (RR 0.66, 95% CI 0.53–0.82, P < 0.001 for noninferiority and superiority compared with warfarin). This difference in effect between dabigatran 150 mg and warfarin was found to occur at 2 months into the trial and was carried throughout until trial completion. Thus low-dose dabigatran was shown to be non-inferior to warfarin and high-dose dabigatran was shown to be superior to warfarin. No statistically significant difference was demonstrated between the groups for the secondary outcome of all-cause mortality (4.13% for warfarin vs. 3.75% for dabigatran 110 mg; RR 0.91, 95% CI 0.80–1.03). There was, however, a numeric decrease in both dabigatran groups that approached significance for those receiving dabigatran 150 mg.
Major bleeding was the primary safety outcome, defined as a reduction in haemoglobin level of 2 g/dL, transfusion requiring at least 2 units of blood, or symptomatic bleeding in a critical area or organ. Major haemorrhage occurred in 3.36% per year in patients taking warfarin, 2.71% in low-dose dabigatran (RR 0.8, 95% CI 0.69–0.93, P = 0.003 vs. warfarin), and 3.11%/year in high-dose dabigatran 150-mg group (RR 0.93, 95% CI 0.81–1.07, P = 0.031 vs warfarin). Thus major bleeding was less with 110 mg of dabigatran when compared to warfarin, and rates of major haemorrhage are similar with 150 mg dabigatran and warfarin. High-dose dabigatran was associated with a significantly increased risk of major gastrointestinal haemorrhage (1.51%) compared with dabigatran 110 mg (1.12%) or warfarin (1.02%). However, all composite major bleeding rates were found to be similar between dabigatran 150 mg and warfarin.
Discontinuation rates were 15% for dabigatran 110 mg, 16% for dabigatran 150 mg, and 10% for warfarin after the first year of the trial; and 21% for dabigatran 110 mg, 21% for dabigatran 150 mg, and 17% for warfarin at the end of the second year of the trial (P < 0.001 for dabigatran vs warfarin). The primary driver for this increased discontinuation of dabigatran was its propensity to cause dyspepsia: 11.8% for 110 mg and 11.3% for 150 mg compared to 5.8% for warfarin (P < 0.001). Thus, warfarin was better tolerated than dabigatran.
Dabigatran 150-mg was found to have an increased rate of myocardial infarction (0.74%) when compared with warfarin (0.53%/year). This effect that trended towards, but did not reach, statistical significance (RR 1.38, 95% CI 1–1.91, P = 0.04). It is possible that the increased occurrence of myocardial infarction observed in patients taking dabigatran in this trial owes more to the protective effects of warfarin rather than an inherent risk associated with dabigatran treatment. A meta-analysis comparing warfarin and other treatment regimes showed that warfarin was associated with significant reduction in myocardial infarction (relative risk, 0.77; 95% CI, 0.63–0.95; P < 0.01).104
A subgroup analysis of the RE-LY trial investigated the safety and efficacy of dabigatran compared to warfarin with differing achievements in INR control.105 The study found that the time in therapeutic range did not impact on the original trial’s findings with regard to efficacy or intracranial haemorrhage. A further subgroup analysis was undertaken in patients with a history of previous stroke or TIA.106 The effects of dabigatran compared with warfarin were not significantly different in patients with a previous stroke or TIA in any other outcomes compared with other patients—confirming dabigatran’s role in secondary prevention and supporting the findings of the original RE-LY trial. An analysis of patients undergoing cardioversion107 showed the risk of stroke and major haemorrhage on dabigatran was similar to warfarin.
A network meta-analysis compared dabigatran favourably to antiplatelet therapy:108 dabigatran 150 mg reduced stroke risk by 63% compared to aspirin alone and 61% compared to dual antiplatelet therapy, as well as 77% when compared to placebo.
Rivaroxaban
The oral direct factor Xa inhibitor rivaroxaban was compared to warfarin in the ROCKET-AF study.109 This trial was a phase III, randomised, double-blind, event-driven noninferiority trial with over 14,000 patients comparing rivaroxaban with warfarin in nonvalvular AF (at least two documented episodes) and a history of stroke, TIA, or non-CNS embolism or at least two independent risk factors for future stroke. Enrolment of patients without stroke, TIA, or systemic embolism and only two risk factors was capped at 10% of the overall study population; all subsequently enrolled patients were required to have at least three stroke risk factors or a history of stroke, TIA, or systemic embolism. 86% of the total population had a CHADS2 score of 3 or higher.
Patients were randomised to rivaroxaban 20 mg once daily (or 15 mg once daily in patients with moderate renal impairment), or dose-adjusted warfarin titrated to a target INR of 2.5. The per-protocol, as-treated primary analysis was designed to determine whether rivaroxaban was noninferior to warfarin for the primary end point of stroke or systemic embolism; if the noninferiority criteria were satisfied, then superiority was analysed in the intent-to-treat population.
Rivaroxaban was similar to warfarin for the primary efficacy endpoint of prevention of stroke and systemic embolism (event rate 1.71 versus 2.16 per 100 patient years for rivaroxaban versus warfarin; hazard ratio [HR] 0.79, 95% confidence interval [CI] 0.66–0.96, P < 0.001 for noninferiority). The stricter intention-to-treat analysis also showed rivaroxaban was similar to warfarin but did not reach statistical significance for superiority: event rate 2.12 versus 2.42 per 100 patient years for rivaroxaban versus warfarin; HR 0.88, 95% CI 0.74–1.03, P = 0.117 for superiority. Superiority was only demonstrated in the per-protocol analysis of patients who continued to receive treatment for the 40-month trial period: event rate 1.70 versus 2.15 per 100 patient years for rivaroxaban versus warfarin; HR 0.79, 95% CI 0.65–0.95, P = 0.015 for superiority.
Major and nonmajor clinically relevant bleeding was similar with rivaroxaban and warfarin: event rate 14.91 versus 14.52 per 100 patient years for rivaroxaban versus warfarin; HR 1.03, 95% CI 0.96–1.11, P = 0.442. The rivaroxaban group demonstrated significantly less fatal bleeding (0.2 versus 0.5 per 100 patient years, HR 0.50, 95% CI 0.31–0.79, P = 0.003), intracranial haemorrhage (0.5 versus 0.7 per 100 patient years; P = 0.02). However, significantly more patients receiving rivaroxaban had a haemoglobin decrease of 2 g/dL or more (event rate 2.77 versus 2.26 per 100 patient years, HR 1.22, 95% CI 1.03–1.44, P = 0.019) and required a blood transfusion (1.65 versus 1.32 per 100 patient years, HR 1.25, 95% CI 1.01–1.55, P = 0.044).
The number of patients experiencing a serious adverse event was similar in the two groups (rivaroxaban 37.3% versus warfarin 38.2%) as was the documentation of an adverse event requiring discontinuation of the study drug (rivaroxaban 15.7% versus warfarin 15.2%). Premature discontinuation rates were also comparable, at approximately 23%. A higher percentage of patients taking rivaroxaban experienced epistaxis (10.1% versus 8.6%), and the rates of ALT elevation were the same in both groups (2.9%).
Apixaban
The AVERROES study was designed to evaluate the use of apixaban for stroke prophylaxis by comparing it to aspirin in patients unsuitable for warfarin.111 The study enrolled 5600 patients with AF who were either intolerant of or unsuitable for warfarin and compared apixaban 5 mg twice daily (2.5 mg twice daily for patients aged over 80 years, weighing under 60 kg, or with renal impairment) with aspirin 81–324 mg/day. The study was prematurely because of an acceptable safety profile and benefit in favour of apixaban. After a year, patients taking apixaban were found to have a 55% reduction in the primary endpoint of stroke or systemic embolism (1.6% versus 3.7% per year, HR 0.45, 95% CI 0.32–0.62, P < 0.001). The rate of major bleeding was similar in both groups: 1.4% per year for apixaban and 1.2% per year for aspirin (HR 1.13, 95% CI 0.74–1.75, P = 0.57). Aspirin was the less well-tolerated therapy.112
The ARISTOTLE trial has compared apixaban to warfarin in patients with atrial fibrillation.113 It is a randomised phase III, double-blind, international trial comparing apixaban 5 mg twice/day versus warfarin titrated to an INR between 2 and 3 in over 18,000 patients.114 The primary outcome was stroke (either ischaemic or haemorrhagic) or systemic embolism, and the trial was designed to test for noninferiority. Secondary objectives included an analysis for superiority with respect to the primary outcome and to the rates of major bleeding and all-cause mortality. The follow-up period was 1.8 years.
The rate of the primary outcome in ARISTOTLE was 1.27% per year in the apixaban group versus 1.60% per year in the warfarin group (hazard ratio with apixaban, 0.79; 95% confidence interval [CI], 0.66 to 0.95; P < 0.001 for noninferiority; P = 0.01 for superiority). This was primarily driven by a reduction in haemorrhagic stroke, as the rates of ischaemic stroke were comparable with warfarin: 0.97% per year in the apixaban group versus 1.05% per year in the warfarin group (hazard ratio, 0.92; 95% CI, 0.74 to 1.13; P = 0.42). Conversely, rate of haemorrhagic stroke was 0.24% per year in the apixaban group versus 0.47% per year in the warfarin group (hazard ratio, 0.51; 95% CI, 0.35 to 0.75; P < 0.001). Apixaban demonstrated a benefit with regards to all-cause mortality compared to warfarin: rates of death from any cause were 3.52% in the apixaban group versus 3.94% in the warfarin group (hazard ratio, 0.89; 95% CI, 0.80 to 0.99; P = 0.047). Apixaban was found to be safer than warfarin in regard to major bleeding: 2.13% per year in the apixaban group versus 3.09% per year in the warfarin group (hazard ratio, 0.69; 95% CI, 0.60 to 0.80; P < 0.001). Drug discontinuation occurred less frequently with apixaban compared to warfarin: 25.3% versus 27.5% (P = 0.001). The average time spent in therapeutic INR was 62.2% for the warfarin-treated patients. The reported adverse and serious adverse effects were similar in both groups of patients.
Patient Values and Preferences
An important consideration when deciding on a therapeutic strategy for stroke prophylaxis in patients with AF is that of patient preference. Patients will, generally speaking, be taking the prescribed therapies for the duration of their lives so it is crucial that they are adequately informed. Evidence suggests that well-informed patients are more compliant with therapy115 and have better outcomes.116 The predominant concern of patients is that of stroke,117 and many are willing to accept slightly increased bleeding risks to avoid a stroke. Physicians tend to be more concerned with hospital admissions, whereas patients are ultimately worried about death.118 The AF-AWARE study also found that physicians tended to overestimate the burden of anticoagulant treatment.118 By and large, patients are willing to accept the inconveniences of anticoagulation to avoid serious adverse outcomes.119 However, the use of decision-making aids leads to fewer patients opting for anticoagulation.120
The advent of novel anticoagulant therapies is changing the landscape of stroke prevention in atrial fibrillation, and will significantly impact on patient preference. The new agents circumvent many of the inconveniences of warfarin: regular INR checks, dietary restrictions, drug interactions. They also, however, bring with them their own considerations and caveats [Table 6].
Table 6.
|
There are no known antidotes currently available for dabigatran, rivaroxaban or apixaban (although preliminary work is being done on a potential factor Xa inhibitor antidote,121 and a phase IV study is investigating various reversal strategies for dabigatran).122 The benefit of not requiring regular INR monitoring is offset by the fact that there is no validated way to assess the anticoagulant effect or level of the drug. We are also yet to establish how successful anticoagulant bridging prior to surgery can be achieved with the new agents [Table 7].
Table 7.
|
Dabigatran and apixaban require twice daily dosing, which is not an issue for rivaroxaban. Patients with GI dysfunction must be counselled regarding dabigatran’s propensity to cause dyspepsia and increased rates of gastrointestinal bleeding. Dabigatran and rivaroxaban must be used with caution in patients with renal insufficiency, and the dose of dabigatran recommended by the FDA for renal impairment123 was not studied in the RE-LY trial.124 Concerns were raised following RE-LY of the increased (but small excess) number of myocardial infarction events in the dabigatran-treated group, but this finding has not been seen in the trials for apixaban or rivaroxaban. Furthermore, supplementary findings from the RE-LY trial125 reporting newly identified events in the dabigatran group found the difference in the myocardial infarction rates was less pronounced.
The efficacy and safety of warfarin has been established over the last two decades, and it is readily reversed by vitamin K. Patients must be fully aware that, by definition, little is known regarding the long-term safety and efficacy profiles of novel agents. Further research ought to enhance our knowledge of and confidence in the new agents available for stroke prophylaxis in AF (such as the RELY-ABLE trial,126 an extension of the RE-LY trial focussing on patient safety), and future work must emphasise patient preference.
Place in Therapy
Warfarin has a clearly defined place in therapy, as the established gold standard antithrombotic for stroke prevention in atrial fibrillation. The optimal INR for AF patients is 2.0–3.0,127 with increased risk of thromboembolism and haemorrhage outside this range at either end. The benefit of warfarin is strongly linked to the proportion of time spent in the therapeutic INR range (time in therapeutic range, TTR).128 A string of outcome measures in AF are all linked to the quality of the INR control: stroke and systemic embolism, myocardial infarction, major bleeding and death.129 Even modest TTR improvements of 5%–10% have profound beneficial effects on clinical outcomes.130 TTR in clinical trials is typically 60%–65%, but this exceeds that routinely achieved in clinical practice.131 Very low TTR may completely obliterate the potential benefit of warfarin. It has been demonstrated that self-monitoring improves the quality of INR control and therefore outcome measures.132 Despite its efficacy, the limitations of warfarin mean that a large group of patients with AF are not receiving effective prophylaxis against stroke.
The ultimate place in therapy of the novel oral anticoagulants is yet to be established. Currently, only dabigatran has been improved by the FDA and incorporated into guidelines. The US guidelines133 recommend dabigatran 150 mg BD as an alternative to warfarin (or 75 mg for patients with renal impairment). The European guidelines30 currently recommend 150 mg dabigatran twice a day for patients at low bleeding risk (HAS-BLED score 0–2) and 110 mg dabigatran twice a day for those at high risk of bleeding (HAS-BLED score 3 or greater). The Canadian guidelines134 also recommend dabigatran as an alternative to warfarin.
Rivaroxaban and apixaban have completed phase III trials and will now undergo analysis and approval before their inclusion in guidelines. These two factor Xa inhibitors have not been shown to cause significant GI upset, so may represent an appealing treatment option for those patients unsuited to warfarin and unable to tolerate dabigatran due to dyspepsia. It is difficult to offer speculative comparisons between the new agents based on their study designs. For example, it may be tempting to infer that rivaroxaban is has more proven efficacy in high-risk patients as ROCKET-AF included few low-risk patients whereas RE-LY had significantly more (32% of patients in RE-LY had a CHADS2 score of 0–1, compared to <1% for ROCKET-AF). Given the results of the ATLAS-ACS2 trial138 (where rivaroxaban reduced the risk of the composite end point of death from cardiovascular causes, myocardial infarction, or stroke in patients with a recent acute coronary syndrome), rivaroxaban may find favour with clinicians treating patients following acute coronary syndromes. Conclusive comparisons between the new and emerging agents cannot be made until they have been evaluated against each other in trials.
As new agents are becoming available to clinicians for prevention of stroke in AF, new considerations must be undertaken [Table 8]. Patients who are well-established on warfarin with good quality INR control are unlikely to derive sufficient benefit to warrant switching to a new drug. The safety data available for the novel anticoagulants is reassuring, but long-term data is mandatory as patients will mostly be maintained on thromboprophylaxis for the duration of their lives. Emphasis must be given to individual patient characteristics, and patient preferences.
Table 8.
|
Conclusions
For six decades, warfarin has been the only available therapeutic strategy for prophylaxis against stroke in patients with AF. Its limitations have led to its underutilisation and wide variability in AF management. Major progress has been made in AF research, providing clinicians with improved management strategies. Better risk stratification schemes permits accurate identification of truly low-risk patients who do not require anticoagulation, and those patients who ought to be receiving antithrombotic therapy. We are also able to simply and practically evaluate a patient’s risk in relation to bleeding, enabling risk-benefit decisions to be made in a more straightforward manner.
The advent of novel anticoagulants means that warfarin is no longer the only choice for effective stroke prophylaxis. Clinicians will be tasked with coming to terms with the strengths and weaknesses of each new therapeutic option and employing them in appropriate settings. Only long-term study and use of novel anticoagulants will conclusively demonstrate how these drugs measure up to warfarin in terms of efficacy, safety and cost-effectiveness (see Table 8). The nature of AF thromboprophylaxis means that patients will likely be receiving a drug lifelong, so the importance of long-term data for new agents and an emphasis on patient values and preference cannot be overstated. The landscape of stroke prevention in atrial fibrillation has changed and will continue evolve as research continues into newer drugs135—and with each new management option comes new considerations.
Footnotes
Disclosures
GL has served as a consultant for Bayer, Astellas, Merck, AstraZeneca, Sanofi, BMS/Pfizer, Daiichi-Sankyo, Biotronik, Portola and BoehringerIngelheim and has been on the speakers bureau for Bayer, BMS/Pfizer, BoehringerIngelheim, and Sanofi Aventis. YA-none declared.
Author(s) have provided signed confirmations to the publisher of their compliance with all applicable legal and ethical obligations in respect to declaration of conflicts of interest, funding, authorship and contributorship, and compliance with ethical requirements in respect to treatment of human and animal test subjects. If this article contains identifiable human subject(s) author(s) were required to supply signed patient consent prior to publication. Author(s) have confirmed that the published article is unique and not under consideration nor published by any other publication and that they have consent to reproduce any copyrighted material. The peer reviewers declared no conflicts of interest.
References
- 1.Go AS, Hylek EM, Phillips KA, et al. Prevalence of diagnosed atrial fibrillation in adults: national implications for rhythm management and stroke prevention: the AnTicoagulation and Risk Factors In Atrial Fibrillation (ATRIA) study. JAMA. 2001;285:2370–5. doi: 10.1001/jama.285.18.2370. [DOI] [PubMed] [Google Scholar]
- 2.Lloyd-Jones DM, Wang TJ, Leip EP, et al. Lifetime risk for development of atrial fibrillation: the Framingham Heart Study. Circulation. 2004;110:1042–6. doi: 10.1161/01.CIR.0000140263.20897.42. [DOI] [PubMed] [Google Scholar]
- 3.Benjamin EJ, Levy D, Vaziri SM, D’Agostino RB, Belanger AJ, Wolf PA. Independent risk factors for atrial fibrillation in a population-based cohort. The Framingham Heart Study. JAMA. 1994;271:840–4. [PubMed] [Google Scholar]
- 4.Heeringa J, van der Kuip DA, Hofman A, et al. Prevalence, incidence and lifetime risk of atrial fibrillation: the Rotterdam Study. Eur Heart J. 2006;27:949–53. doi: 10.1093/eurheartj/ehi825. [DOI] [PubMed] [Google Scholar]
- 5.Miyasaka Y, Barnes ME, Gersh BJ, et al. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation. 2006;114:119–25. doi: 10.1161/CIRCULATIONAHA.105.595140. [DOI] [PubMed] [Google Scholar]
- 6.Wolf PA, Mitchell JB, Baker CS, Kannel WB, D’Agostino RB. Impact of atrial fibrillation on mortality, stroke, and medical costs. Arch Intern Med. 1998;158:229–34. doi: 10.1001/archinte.158.3.229. [DOI] [PubMed] [Google Scholar]
- 7.Tsang TS, Gersh BJ. Atrial fibrillation: an old disease, a new epidemic. Am J Med. 2002;113:432–5. doi: 10.1016/s0002-9343(02)01245-7. [DOI] [PubMed] [Google Scholar]
- 8.Benjamin EJ, Wolf PA, D’Agostino RB, Silbershatz H, Kannel WB, Levy D. Impact of atrial fibrillation on the risk of death: the Framingham Heart Study. Circulation. 1998;98:946–52. doi: 10.1161/01.cir.98.10.946. [DOI] [PubMed] [Google Scholar]
- 9.Stewart S, Murphy NF, Walker A, McGuire A, McMurray JJ. Cost of an emerging epidemic: an economic analysis of atrial fibrillation in the UK. Heart. 2004;90:286–92. doi: 10.1136/hrt.2002.008748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independentrisk factor for stroke: the Framingham Study. Stroke. 1991;22:983–8. doi: 10.1161/01.str.22.8.983. [DOI] [PubMed] [Google Scholar]
- 11.Lip GY, Edwards SJ. Stroke prevention with aspirin, warfarin and ximelagatran in patients with non-valvular atrial fibrillation: A systematic review and meta-analysis. Thromb Res. 2006;118:321–33. doi: 10.1016/j.thromres.2005.08.007. [DOI] [PubMed] [Google Scholar]
- 12.Lin HJ, Wolf PA, Kelly-Hayes M, et al. Stroke severity in atrial fibrillation. The Framingham Study. Stroke. 1996;27:1760–4. doi: 10.1161/01.str.27.10.1760. [DOI] [PubMed] [Google Scholar]
- 13.Dulli DA, Stanko H, Levine RL. Atrial fibrillation is associated with severe acute ischemic stroke. Neuroepidemiology. 2003;22:118–23. doi: 10.1159/000068743. [DOI] [PubMed] [Google Scholar]
- 14.Lamassa M, Di Carlo A, Pracucci G, et al. Characteristics, outcome, and care of stroke associated with atrial fibrillation in Europe: data from a multicentermultinational hospital-based registry (The European Community Stroke Project) Stroke. 2001;32:392–8. doi: 10.1161/01.str.32.2.392. [DOI] [PubMed] [Google Scholar]
- 15.Penado S, Cano M, Acha O, Hernandez JL, Riancho JA. Atrial fibrillation as a risk factor for stroke recurrence. Am J Med. 2003;114:206–10. doi: 10.1016/s0002-9343(02)01479-1. [DOI] [PubMed] [Google Scholar]
- 16.Hart RG, Halperin JL. Atrial fibrillation and thromboembolism: a decade of progress in stroke prevention. Ann Intern Med. 1999;131:688–95. doi: 10.7326/0003-4819-131-9-199911020-00010. [DOI] [PubMed] [Google Scholar]
- 17.vanWalraven C, Hart RG, Singer DE, et al. Oral anticoagulants vs aspirin in nonvalvular atrial fibrillation: an individual patient meta-analysis. JAMA. 2002;288:2441–8. doi: 10.1001/jama.288.19.2441. [DOI] [PubMed] [Google Scholar]
- 18.Hart RG, Benavente O, McBride R, Pearce LA. Antithrombotic therapy to prevent stroke in patients with atrial fibrillation: A meta-analysis. Ann Intern Med. 1999;131:492–501. doi: 10.7326/0003-4819-131-7-199910050-00003. [DOI] [PubMed] [Google Scholar]
- 19.Lin P. Reviewing the reality: why we need to change. Eur Heart J Suppl. 2005;7:E15–E20. [Google Scholar]
- 20.White RH, McBurnie MA, Manolio T, et al. Oral anticoagulation in patients with atrial fibrillation: adherence with guidelines in an elderly cohort. Am J Med. 1999;106:165–71. doi: 10.1016/s0002-9343(98)00389-1. [DOI] [PubMed] [Google Scholar]
- 21.Hughes M, Lip GY. Stroke and thromboembolism in atrial fibrillation: a systematic review of stroke risk factors, risk stratification schema and cost effectiveness data. Thromb Haemost. 2008;99:295–304. doi: 10.1160/TH07-08-0508. [DOI] [PubMed] [Google Scholar]
- 22.Stroke Risk in Atrial Fibrillation Working Group Independent predictors of stroke in patients with atrial fibrillation: a systematic review. Neurology. 2007;69:546–54. doi: 10.1212/01.wnl.0000267275.68538.8d. [DOI] [PubMed] [Google Scholar]
- 23.Sato H, Ishikawa K, Kitabatake A, et al. Low-dose aspirin for prevention of stroke in low-risk patients with atrial fibrillation: Japan Atrial Fibrillation Stroke Trial. Stroke. 2006;37:447–51. doi: 10.1161/01.STR.0000198839.61112.ee. [DOI] [PubMed] [Google Scholar]
- 24.Lee BH, Park JS, Park JH, et al. The effect and safety of the antithrombotic therapies in patients with atrial fibrillation and CHADS score 1. J Cardiovasc Electrophysiol. 2010;21:501–7. doi: 10.1111/j.1540-8167.2009.01661.x. [DOI] [PubMed] [Google Scholar]
- 25.Gorin L, Fauchier L, Nonin E, et al. Antithrombotic treatment and the risk of death and stroke in patients with atrial fibrillation and a CHADS2 score = 1. Thromb Haemost. 2010;103:833–40. doi: 10.1160/TH09-10-0746. [DOI] [PubMed] [Google Scholar]
- 26.Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Validation of clinical classification schemes for predicting stroke: results from the national registry of atrial fibrillation. JAMA. 2001;285:2864–70. doi: 10.1001/jama.285.22.2864. [DOI] [PubMed] [Google Scholar]
- 27.Lip GY, Halperin JL. Improving stroke risk stratification in atrial fibrillation. Am J Med. 2010;123:484–8. doi: 10.1016/j.amjmed.2009.12.013. [DOI] [PubMed] [Google Scholar]
- 28.Lip GY, Nieuwlaat R, Pisters R, Lane DA, Crijns HJ. Refining clinical risk stratification for predicting stroke and thromboembolism in atrial fibrillation using a novel risk factor-based approach: the euro heart survey on atrial fibrillation. Chest. 2010;137:263–72. doi: 10.1378/chest.09-1584. [DOI] [PubMed] [Google Scholar]
- 29.Olesen JB, Lip GY, Hansen ML, et al. Validation of risk stratification schemes for predicting stroke and thromboembolism in patients with atrial fibrillation: nationwide cohortstudy. BMJ. 2011;342:d124. doi: 10.1136/bmj.d124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.European Heart Rhythm Association. European Association for Cardio-Thoracic Surgery. Camm AJ, Kirchhof P, Lip GY, et al. Guidelines for the management of atrial fibrillation: the Task Force forthe Management of Atrial Fibrillation of the European Society of Cardiology(ESC) Europace. 2010;12:1360–420. doi: 10.1093/europace/euq350. [DOI] [PubMed] [Google Scholar]
- 31.Hylek EM, D’Antonio J, Evans-Molina C, Shea C, Henault LE, Regan S. Translating the results of randomized trials into clinical practice: the challenge of warfarin candidacy among hospitalized elderly patients with atrial fibrillation. Stroke. 2006 Apr;37(4):1075–80. doi: 10.1161/01.STR.0000209239.71702.ce. [DOI] [PubMed] [Google Scholar]
- 32.Palareti G, Cosmi B. Bleeding with anticoagulation therapy—who is at risk, and how best to identify such patients. Thromb Haemost. 2009;102:268–78. doi: 10.1160/TH08-11-0730. [DOI] [PubMed] [Google Scholar]
- 33.Pisters R, Lane DA, Nieuwlaat R, de Vos CB, Crijns HJ, Lip GY. A novel user-friendly score (HAS-BLED) to assess 1-year risk of major bleeding in patients with atrial fibrillation: the Euro Heart Survey. Chest. 2010;138:1093–100. doi: 10.1378/chest.10-0134. [DOI] [PubMed] [Google Scholar]
- 34.Palareti G. The risk of bleeding with anticoagulant treatments. Phlebologie. 2011;40(4):203–9. doi: 10.5482/ha-1151. [DOI] [PubMed] [Google Scholar]
- 35.Olesen JB, Lip GY, Hansen PR, et al. Bleeding risk in ‘real world’ patients with atrial fibrillation: comparison of two established bleeding prediction schemes in anationwide cohort. J Thromb Haemost. 2011;9:1460–7. doi: 10.1111/j.1538-7836.2011.04378.x. [DOI] [PubMed] [Google Scholar]
- 36.Lip GY, Frison L, Halperin JL, Lane DA. Comparative validation of a novel risk score for predicting bleeding risk in anticoagulated patients with atrial fibrillation: the HAS-BLED (Hypertension, AbnormalRenal/Liver Function, Stroke, Bleeding History or Predisposition, Labile INR, Elderly, Drugs/Alcohol Concomitantly) score. J Am Coll Cardiol. 2011;57:173–80. doi: 10.1016/j.jacc.2010.09.024. [DOI] [PubMed] [Google Scholar]
- 37.Atrial Fibrillation Investigators Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation. Analysis of pooled data from five randomized controlled trials. Arch Intern Med. 1994;154:1449–57. [PubMed] [Google Scholar]
- 38.Bungard TJ, Ghali WA, Teo KK, McAlister FA, Tsuyuki RT. Why do patients with atrial fibrillation not receive warfarin? Arch Intern Med. 2000;160:41–6. doi: 10.1001/archinte.160.1.41. [DOI] [PubMed] [Google Scholar]
- 39.Lip GY. Stroke prevention in atrial fibrillation: the case for ximelagatran. Eur Heart J Suppl. 2005 Jun;7(Suppl E):E21–25. [Google Scholar]
- 40.Turpie AG. New oral anticoagulants in atrial fibrillation. Eur Heart J. 2008;29:155–65. doi: 10.1093/eurheartj/ehm575. [DOI] [PubMed] [Google Scholar]
- 41.Nelsestuen GL, Zytkovicz TH, Howard JB. The mode of action of vitamin K: identification of γ-carboxyglutamic acid as a component of prothrombin. J Biol Chem. 1974;249:6347–50. [PubMed] [Google Scholar]
- 42.Whitlon DS, Sadowski JA, Suttie JW. Mechanisms of coumarin action: significance of vitamin K epoxide reductase inhibition. Biochemistry. 1978 Apr 18;17:1371–7. doi: 10.1021/bi00601a003. [DOI] [PubMed] [Google Scholar]
- 43.Malhotra OP, Nesheim ME, Mann KG. The kinetics of activation of normal and gamma carboxy glutamic acid deficient prothrombins. J Biol Chem. 1985;260:279–87. [PubMed] [Google Scholar]
- 44.Breckenridge AM. Oral anticoagulant drugs: pharmacokinetic aspects. Semin Hematol. 1978;15:19–26. [PubMed] [Google Scholar]
- 45.Kelly JG, O’Malley K. Clinical pharmacokinetics of oral anticoagulants. Clin Pharmacokinet. 1979;4:1–15. doi: 10.2165/00003088-197904010-00001. [DOI] [PubMed] [Google Scholar]
- 46.Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G, American College of Chest Physicians Pharmacology and management of the vitamin K antagonists: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition) Chest. 2008 Jun;133(6 Suppl):160S–198S. doi: 10.1378/chest.08-0670. [DOI] [PubMed] [Google Scholar]
- 47.O’Reilly RA. Vitamin K and other oral anticoagulant drugs. Annu Rev Med. 1976;27:245–61. doi: 10.1146/annurev.me.27.020176.001333. [DOI] [PubMed] [Google Scholar]
- 48.Vecsler M, Loebstein R, Almog S, et al. Combined genetic profiles of components and regulators of the vitamin K-dependent gamma-carboxylation system affect individual sensitivity to warfarin. Thromb Haemost. 2006;95:205–11. doi: 10.1160/TH05-06-0446. [DOI] [PubMed] [Google Scholar]
- 49.O’Reilly RA, Aggeler PM. Determinants of the response to oral anticoagulant drug in man. Pharmacol Rev. 1970;22:35–96. [PubMed] [Google Scholar]
- 50.O’Reilly R, Rytand D. Resistance to warfarin due to unrecognized vitamin K supplementation. N Engl J Med. 1980;303:160–1. doi: 10.1056/nejm198007173030317. [DOI] [PubMed] [Google Scholar]
- 51.Di Nisio M, Middeldorp S, Buller HR. Direct thrombin inhibitors. N Engl J Med. 2005;353:1028–40. doi: 10.1056/NEJMra044440. [DOI] [PubMed] [Google Scholar]
- 52.Weitz JI, Bates SM. New anticoagulants. J Thromb Haemost. 2005;3:1843–53. doi: 10.1111/j.1538-7836.2005.01374.x. [DOI] [PubMed] [Google Scholar]
- 53.Kaplan KL, Francis CW. Direct thrombin inhibitors. Semin Hematol. 2002;39:187–96. doi: 10.1053/shem.2002.34092. [DOI] [PubMed] [Google Scholar]
- 54.Gustafsson D, Bylund R, Antonsson T, et al. A new oral anticoagulant: the 50-year challenge. Nat Rev Drug Discov. 2004;3:649–59. doi: 10.1038/nrd1466. [DOI] [PubMed] [Google Scholar]
- 55.Gustafsson D, Nyström J, Carlsson S, et al. The direct thrombin inhibitor melagatran and its oral prodrug H 376/95: intestinal absorption properties, biochemical and pharmacodynamic effects. Thromb Res. 2001;101:171–81. doi: 10.1016/s0049-3848(00)00399-6. [DOI] [PubMed] [Google Scholar]
- 56.Johansson LC, Frison L, Logren U, Fager G, Gustafsson D, Eriksson UG. Influence of age on the pharmacokinetics and pharmacodynamics of ximelagatran, an oral direct thrombin inhibitor. Clin Pharmacokinet. 2003;42:381–92. doi: 10.2165/00003088-200342040-00006. [DOI] [PubMed] [Google Scholar]
- 57.Bredberg E, Andersson TB, Frison L, et al. Ximelagatran, an oral direct thrombin inhibitor, has a low potential for cytochrome P450-mediated drug-drug interactions. Clin Pharmacokinet. 2003;42:765–77. doi: 10.2165/00003088-200342080-00005. [DOI] [PubMed] [Google Scholar]
- 58.Hauptmann J. Pharmacokinetics of an emerging new class of anticoagulant/antithrombotic drugs. A review of small-molecule thrombin inhibitors. Eur J Clin Pharmacol. 2002;57:751–8. doi: 10.1007/s00228-001-0392-7. [DOI] [PubMed] [Google Scholar]
- 59.Ahrens I, Lip GY, Peter K. New oral anticoagulant drugs in cardiovascular-disease. Thromb Haemost. 2010;104:49–60. doi: 10.1160/TH09-05-0327. [DOI] [PubMed] [Google Scholar]
- 60.Weitz JI. Emerging anticoagulants for the treatment of venous thromboembolism. Thromb Haemost. 2006;96:274–84. doi: 10.1160/TH06-05-0234. [DOI] [PubMed] [Google Scholar]
- 61.Stangier J, Rathgen K, Stahle H, Gansser D, Roth W. The pharmacokinetics, pharmacodynamics and tolerability of dabigatran etexilate, a new oral direct thrombin inhibitor, in healthy male subjects. Br J Clin Pharmacol. 2007;64:292–303. doi: 10.1111/j.1365-2125.2007.02899.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Stangier J, Rathgen K, Gansser D, Kohlbrenner V, Stassen JM. Pharmacokinetics of BIBR 953 ZW, a novel low molecular weight direct thrombin inhibitor in healthy volunteers. Thromb Haemost. 2001;86:OC2347. [Google Scholar]
- 63.Stangier J, Eriksson BI, Dahl OE, et al. Pharmacokinetic profile of the oral direct thrombin inhibitor dabigatran etexilate in healthy volunteers and patients undergoing total hip replacement. J Clin Pharmacol. 2005;45:555–63. doi: 10.1177/0091270005274550. [DOI] [PubMed] [Google Scholar]
- 64.Troconiz IF, Tillmann C, Liesenfeld KH, Schafer HG, Stangier J. Population pharmacokinetic analysis of the new oral thrombin inhibitor dabigatran etexilate (BIBR 1048) in patients undergoing primary elective total hip replacement surgery. J Clin Pharmacol. 2007;47:371–82. doi: 10.1177/0091270006297228. [DOI] [PubMed] [Google Scholar]
- 65.Stangier J, Stahle E, Rathgen K, Seitz CH. Pharmacokinetic and pharmacodynamics of the direct thrombin inhibitor diaigatran in elderly healthy subjects. J Thromb Haemost. 2005;3:P-2207. [Google Scholar]
- 66.Pham SV, Pham PC, Pham PM, Miller JM, Pham PT, Pham PA. Antithrombotic strategies in patients undergoing percutaneous coronary intervention for acute coronary syndrome. Drug Des Devel Ther. 2010;7:203–20. doi: 10.2147/DDDT.S12056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Samama MM, Gerotziafas GT. Newer anticoagulants in 2009. J Thromb Thrombolysis. 2010;29:92–104. doi: 10.1007/s11239-009-0392-5. [DOI] [PubMed] [Google Scholar]
- 68.Planes A, Vochelle N, Mazas F, et al. Prevention of postoperative venous thrombosis: a randomized trial comparing unfractionated heparin with low molecular weight heparin in patients undergoing total hip replacement. Thromb Haemost. 1988;60:407–10. [PubMed] [Google Scholar]
- 69.Turpie AG, Bauer KA, Eriksson BI, Lassen MR. Fondaparinux vs. enoxaparin for the prevention of venous thromboembolism in major orthopedic surgery: a meta-analysis of 4 randomized double-blind studies. Arch Intern Med. 2002;162:1833–40. doi: 10.1001/archinte.162.16.1833. [DOI] [PubMed] [Google Scholar]
- 70.Weitz JI, Hirsh J, Samama MM, for the American College of Chest Physicians New antithrombotic drugs: American College of Chest Physicians evidence-based clinical practice guidelines, 8th ed. Chest. 2008;133(6 suppl):S234–56. doi: 10.1378/chest.08-0673. [DOI] [PubMed] [Google Scholar]
- 71.Rai R, Sprengeler PA, Elrod KC, Young WB. Perspectives on factor Xa inhibition. Curr Med Chem. 2001;8:101–19. doi: 10.2174/0929867013373822. [DOI] [PubMed] [Google Scholar]
- 72.Wong PC, Watson CA, Crain EJ, et al. Effects of the Factor Xa inhibitor apixaban on venous thrombosis and hemostasis in rabbits. (Abstract 917) Blood. 2006;108 [Google Scholar]
- 73.Roser-Jones C, Becker RC. Apixaban: an emerging oral factor Xa inhibitor. J Thromb Thrombolysis. 2010;29:141–6. doi: 10.1007/s11239-009-0421-4. [DOI] [PubMed] [Google Scholar]
- 74.He K, He B, Grace JE, et al. Preclinical pharmacokinetic and metabolism of apixaban, a potent and selective Factor Xa inhibitor. (Abstract 910) Blood. 2006;108 doi: 10.1007/s13318-011-0037-x. [DOI] [PubMed] [Google Scholar]
- 75.Frost C, Nepal S, Mosqueda-Garcia R, Shenker A. Apixaban, an oral direct Factor Xa inhibitor: single-dose safety, pharmacokinetics and pharmacodynamics in healthy volunteers. J Thromb Haemost. 2007;5:P-M-665. [Google Scholar]
- 76.Nutescu E, Chuatrisorn I, Hellenbart E. Drug and dietary interactions of warfarin and novel oral anticoagulants: an update. J Thromb Thrombolysis. 2011;31:326–43. doi: 10.1007/s11239-011-0561-1. [DOI] [PubMed] [Google Scholar]
- 77.Samama MM, Gerotziafas GT. Newer anticoagulants in 2009. J Thromb Thrombolysis. 2010;29:92–104. doi: 10.1007/s11239-009-0392-5. [DOI] [PubMed] [Google Scholar]
- 78.Perzborn E, Strassburger J, Wilmen A, et al. In vitro and in vivo studies of the novel antithrombotic agent BAY 59-7939-an oral direct Factor Xa inhibitor. J Thromb Haemost. 2005;3:514–52. doi: 10.1111/j.1538-7836.2005.01166.x. [DOI] [PubMed] [Google Scholar]
- 79.Perzborn E, Strassburger J, Wilmen A, et al. Biochemical and pharmacologic properties of BAY 59-7939, an oral, direct Factor Xa inhibitor. (Abstract PO079) Pathophysiol Haemost Thromb. 2004;33(Suppl 2) [Google Scholar]
- 80.Kubitza D, Becka M, Voith B, Zuehlsdorf M, Wensing G. Safety, pharmacodynamics, and pharmacokinetics of single doses of BAY 59-7939, an oral, direct factor Xa inhibitor. Clin Pharmacol Ther. 2005;78:412–21. doi: 10.1016/j.clpt.2005.06.011. [DOI] [PubMed] [Google Scholar]
- 81.Eriksson BI, Quinlan DJ, Weitz JI. Comparative pharmacodynamics and pharmacokinetics of oral direct thrombin and factor Xa inhibitors in development. Clin Pharmacokinet. 2009;48:1–22. doi: 10.2165/0003088-200948010-00001. [DOI] [PubMed] [Google Scholar]
- 82.Kubitza D, Becka M, Mueck W, Zuehlsdorf M. The effect of extreme age, and gender, on the pharmacology and tolerability of rivaroxaban—an oral, direct Factor Xa inhibitor. (Abstract 905) Blood. 2006;108 [Google Scholar]
- 83.Weinz C, Schwartz T, Pleiss U, et al. Metabolism and distribution of [14C] BAY 59–7939—an oral, direct Factor Xa inhibitor—in rat, dog and human. (Abstract 196) Drug Metab Rev. 2004;36(Suppl 1):98. [Google Scholar]
- 84.Kubitza D, Becka M, Wensing G, Voith B, Zuehlsdorf M. Safety, pharmacodynamics, and pharmacokinetics of BAY 59-7939-an oral, direct Factor Xa inhibitor-after multiple dosing in healthy male subjects. Eur J Clin Pharmacol. 2005;61:873–80. doi: 10.1007/s00228-005-0043-5. [DOI] [PubMed] [Google Scholar]
- 85.Galanis T, Thomson L, Palladino M, Merli GJ. New oral anticoagulants. J Thromb Thrombolysis. 2011;31:310–20. doi: 10.1007/s11239-011-0559-8. [DOI] [PubMed] [Google Scholar]
- 86.Kubitza D, Becka M, Mueck W, Zuehlsdorf M. Safety, tolerability, pharmacodynamics, and pharmacokinetics of rivaroxaban-an oral, direct Factor Xa inhibitor-are not affected by aspirin. J Clin Pharmacol. 2006;46:981–90. doi: 10.1177/0091270006292127. [DOI] [PubMed] [Google Scholar]
- 87.Kubitza D, Becka M, Zuehlsdorf M, Mueck W. No interaction between the novel, oral direct Factor Xa inhibitor BAY 59-7939 and digoxin. (Abstract 11) J Clin Pharmacol. 2006;46:702. [Google Scholar]
- 88.Petersen P, Boysen G, Godtfredsen J, Andersen ED, Andersen B. Placebo-controlled, randomised trial of warfarin and aspirin for prevention of thromboembolic complications in chronic atrial fibrillation. The Copenhagen AFASAK study, Lancet. 1989;1:175–9. doi: 10.1016/s0140-6736(89)91200-2. [DOI] [PubMed] [Google Scholar]
- 89.Stroke Prevention in Atrial Fibrillation Study, Final results. Circulation. 1991;84:527–39. doi: 10.1161/01.cir.84.2.527. [DOI] [PubMed] [Google Scholar]
- 90.The Boston Area Anticoagulation Trial for Atrial Fibrillation Investigators The effect of low-dose warfarin on the risk of stroke in patients with nonrheumatic atrial fibrillation. N Engl J Med. 1990;323:1505–11. doi: 10.1056/NEJM199011293232201. [DOI] [PubMed] [Google Scholar]
- 91.Connolly SJ, Laupacis A, Gent M, Roberts RS, Cairns JA, Joyner C. Canadian atrial fibrillation anticoagulation (CAFA) study. J Am Coll Cardiol. 1991;18:349–55. doi: 10.1016/0735-1097(91)90585-w. [DOI] [PubMed] [Google Scholar]
- 92.Ezekowitz MD, Bridgers SL, James KE, et al. for the Veterans Affairs Stroke Prevention in Non-rheumatic Atrial Fibrillation Investigators Warfarin in the prevention of stroke associated with non-rheumatic atrial fibrillation. N Engl J Med. 1992;327:1406–12. doi: 10.1056/NEJM199211123272002. [DOI] [PubMed] [Google Scholar]
- 93.EAFT (European Atrial Fibrillation Trial) Study Group Secondary prevention in non-rheumatic atrial fibrillation after transient ischaemic attack or minor stroke. Lancet. 1993;342:1255–62. [PubMed] [Google Scholar]
- 94.Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation. Analysis of pooled data from five randomized controlled trials. Arch Intern Med. 1994;154:1449–57. [PubMed] [Google Scholar]
- 95.van Walraven C, Hart RG, Singer DE, et al. Oral anticoagulants vs. aspirin in nonvalvular atrial fibrillation: an individual patient meta-analysis. JAMA. 2002;288:2441–8. doi: 10.1001/jama.288.19.2441. [DOI] [PubMed] [Google Scholar]
- 96.Hart RG, Pearce LA, Aguilar MI. Meta-analysis: antithrombotic therapy to prevent stroke in patients who have nonvalvular atrial fibrillation. Ann Intern Med. 2007;146:857–67. doi: 10.7326/0003-4819-146-12-200706190-00007. [DOI] [PubMed] [Google Scholar]
- 97.Mant J, Hobbs FD, Fletcher K, et al. BAFTA investigators Midland Research Practices Network (MidReC): Warfarin versus aspirin for stroke prevention in an elderly community population with atrial fibrillation (the Birmingham Atrial Fibrillation Treatment of the Aged Study, BAFTA): a randomised controlled trial. Lancet. 2007;370:493–503. doi: 10.1016/S0140-6736(07)61233-1. [DOI] [PubMed] [Google Scholar]
- 98.Connolly S, Pogue J, Hart R, et al. Clopidogrel plus aspirin versus oral anticoagulation for atrial fibrillation in the Atrial fibrillation Clopidogrel Trial with Irbesartan for Prevention of Vascular Events (ACTIVE W): A randomised controlled trial. Lancet. 2006;367:1903–12. doi: 10.1016/S0140-6736(06)68845-4. [DOI] [PubMed] [Google Scholar]
- 99.Connolly SJ, Pogue J, Hart RG, et al. Effect of clopidogrel added to aspirin in patients with atrial fibrillation. N Engl J Med. 2009;360:2066–78. doi: 10.1056/NEJMoa0901301. [DOI] [PubMed] [Google Scholar]
- 100.Verheugt FW. Who is ineligible for warfarin in atrial fibrillation? Lancet. 2009;374:510–1. doi: 10.1016/S0140-6736(09)61471-9. [DOI] [PubMed] [Google Scholar]
- 101.Ezekowitz MD, Reilly PA, Nehmiz G, et al. Dabigatran with or without concomitant aspirin compared with warfarin alone in patients with non-valvular atrial fibrillation (PETRO study) Am J Cardiol. 2007;100:1419–26. doi: 10.1016/j.amjcard.2007.06.034. [DOI] [PubMed] [Google Scholar]
- 102.Connolly SJ, Ezekowitz MD, Yusuf S, et al. RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361:1139–51. doi: 10.1056/NEJMoa0905561. [DOI] [PubMed] [Google Scholar]
- 103.Ezekowitz MD, Connolly S, Parekh A, et al. Rationale and design of RE-LY: randomized evaluation of long-term anticoagulant therapy, warfarin, compared with dabigatran. Am Heart J. 2009;157:805–10. 810.e1–2. doi: 10.1016/j.ahj.2009.02.005. [DOI] [PubMed] [Google Scholar]
- 104.Lip GY, Lane DA. Does warfarin for stroke thromboprophylaxis protectagainst MI in atrial fibrillation patients? Am J Med. 2010;123:785–9. doi: 10.1016/j.amjmed.2010.01.031. [DOI] [PubMed] [Google Scholar]
- 105.Wallentin L, Yusuf S, Ezekowitz MD, et al. RE-LY investigators Efficacy and safety of dabigatran compared with warfarin at different levels of international normalized ratio control for stroke prevention in atrial fibrillation: an analysis of the RE-LY trial. Lancet. 2010;376:975–83. doi: 10.1016/S0140-6736(10)61194-4. [DOI] [PubMed] [Google Scholar]
- 106.Diener HC, Connolly SJ, Ezekowitz MD, et al. RE-LY study group Dabigatran compared with warfarin in patients with atrial fibrillation and previous transient ischemic attack or stroke: a subgroup analysis of the RE-LY trial. Lancet Neurol. 2010;9:1157–63. doi: 10.1016/S1474-4422(10)70274-X. [DOI] [PubMed] [Google Scholar]
- 107.Nagarakanti R, Ezekowitz MD, Oldgren J, et al. Dabigatran versus warfarin in patients with atrial fibrillation: an analysis of patients undergoing cardioversion. Circulation. 2011;123:131–6. doi: 10.1161/CIRCULATIONAHA.110.977546. [DOI] [PubMed] [Google Scholar]
- 108.Roskell NS, Lip GY, Noack H, Clemens A, Plumb JM. Treatments for stroke prevention in atrial fibrillation: a network meta-analysis and indirect comparisons versus dabigatran etexilate. Thromb Haemost. 2010;104:1106–15. doi: 10.1160/TH10-10-0642. [DOI] [PubMed] [Google Scholar]
- 109.Patel MR, Mahaffey KW, Garg J, et al. ROCKET AF Investigators Rivaroxaban versus warfarin in nonvalvularatrial fibrillation. N Engl J Med. 2011;365:883–91. doi: 10.1056/NEJMoa1009638. [DOI] [PubMed] [Google Scholar]
- 110.ROCKET AF Study Investigators Rivaroxaban: once daily, oral, direct factor Xa inhibition compared with vitamin K antagonism for prevention of stroke and embolism trial in atrial fibrillation: rationale and design of the ROCKET AF study. Am Heart J. 2010;159:340–7.e1. doi: 10.1016/j.ahj.2009.11.025. [DOI] [PubMed] [Google Scholar]
- 111.Connolly SJ, Eikelboom J, Joyner C, et al. AVERROES Steering Committee and Investigators Apixaban in patients with atrial fibrillation. N Engl J Med. 2011;364:806–17. doi: 10.1056/NEJMoa1007432. [DOI] [PubMed] [Google Scholar]
- 112.Connolly SJ, Eikelboom J, Joyner C, et al. AVERROES Steering Committee and Investigators Apixaban in patients with atrial fibrillation. N Engl J Med. 2011;364:806–17. doi: 10.1056/NEJMoa1007432. [DOI] [PubMed] [Google Scholar]
- 113.Granger CB, Alexander JH, McMurray JJ, et al. ARISTOTLE Committees and Investigators Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365:981–92. doi: 10.1056/NEJMoa1107039. [DOI] [PubMed] [Google Scholar]
- 114.Lopes RD, Alexander JH, Al-Khatib SM, et al. ARISTOTLE Investigators Apixaban for reduction in stroke and other thromboembolic events in atrial fibrillation (ARISTOTLE) trial: design and rationale. Am Heart J. 2010;159:331–9. doi: 10.1016/j.ahj.2009.07.035. [DOI] [PubMed] [Google Scholar]
- 115.Roter DL, Hall JA, Merisca R, Nordstrom B, Cretin D, Svarstad B. Effectiveness of interventions to improve patient compliance: a metaanalysis. Med Care. 1998;36:1138–61. doi: 10.1097/00005650-199808000-00004. [DOI] [PubMed] [Google Scholar]
- 116.Kaplan S, Greenfield S, Ware JE. Assessing the effects of physicianpatient interactions on the outcomes of chronicdisease. Med Care. 1989;27:S110–27. doi: 10.1097/00005650-198903001-00010. [DOI] [PubMed] [Google Scholar]
- 117.Fuller R, Dudley N, Blacktop J. Avoidance hierarchies and preferences for anticoagulation- semi-qualitative analysis of older patients’ views about stroke prevention and the use of warfarin. Age Ageing. 2004;33:608–11. doi: 10.1093/ageing/afh202. [DOI] [PubMed] [Google Scholar]
- 118.Aliot E, Breithardt G, Brugada J, Camm J, Lip GY, Vardas PE, Wagner M, Atrial Fibrillation AWareness And Risk Education group. Atrial Fibrillation Association. European Heart Rhythm Association. Stroke Alliance for Europe. World Heart Federation An international survey of physician and patient understanding, perception, and attitudes to atrial fibrillation and its contribution to cardiovascular disease morbidity and mortality. Europace. 2010;12:626–33. doi: 10.1093/europace/euq109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lip GYH, Agnelli G, Thach AA, Knight E, Rost D, Tangelder MJ. Oral anticoagulation in atrial fibrillation: a pan-European patient survey. Eur J Intern Med. 2007;18:202–8. doi: 10.1016/j.ejim.2006.11.005. CrossRef|PubMed. [DOI] [PubMed] [Google Scholar]
- 120.Holbrook A, Labiris R, Goldsmith CH, Ota K, Harb S, Sebaldt RJ. Influence of decision aids on patient preferences for anticoagulant therapy: a randomised trial. Can Med Assoc J. 2007;176:1583–7. doi: 10.1503/cmaj.060837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Merck and Portola Pharmaceuticals Phase 2 study showed investigational factor Xa inhibitor, betrixaban, reduced incidence of bleeding compared to warfarin in patients with atrial fibrillation. Press release March 15, 2010. Available from http://www.portola.com/pdfs/Betrixaban_EXPLORE_ACC_032010.pdf.
- 122.University Hospital, Grenoble . Study in healthy volunteers of the reversion by haemostatic drugs of the anticoagulant effect of new anti-thrombotics (REVNEWANTICO) ClinicalTrials.gov. Bethesda, MD: National Library of Medicine; Available from http://clinicaltrials.gov/ct2/show/NCT01210755?term=NCT01210755&rank=1. [Google Scholar]
- 123.Wann LS, Curtis AB, Ellenbogen KA, et al. ACCF/AHA/HRS Focused Update on the management of patients with atrial fibrillation (Update on Dabigatran): a report of the American College of Cardiology Foundation Foundation/American Heart Association Task Force on practice guidelines. J Am Coll Cardiol. 2011;57:1330–1337. doi: 10.1016/j.jacc.2011.01.010. [DOI] [PubMed] [Google Scholar]
- 124.Beasley BN, Unger EF, Temple R. Anticoagulant options—why the FDA approved a higher but not a lower dose of dabigatran. N Engl J Med. 2011;364:1788–90. doi: 10.1056/NEJMp1103050. [DOI] [PubMed] [Google Scholar]
- 125.Connolly SJ, Ezekowitz MD, Yusuf S, Reilly PA, Wallentin L. Newly identified events in the RE-LY Trial [letter] New Engl J Med. 2010;363:1875–87. doi: 10.1056/NEJMc1007378. [DOI] [PubMed] [Google Scholar]
- 126.BoehringerIngelheim Pharmaceuticals . RELY-ABLE long term multi-center extension of dabigatran treatment in patients with atrial fibrillation who completed RE-LY trial Updated 2011 ClinicalTrials.gov. Bethesda, MD: National Library of Medicine; Available from http://clinicaltrials.gov/ct2/show/NCT00808067?term=NCT00808067&rank=1. [Google Scholar]
- 127.Singer DE, Albers GW, Dalen JE, et al. American College of Chest Physicians Antithrombotic therapy in atrial fibrillation: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th Edition) Chest. 2008;133(6 Suppl):546S–92S. doi: 10.1378/chest.08-0678. [DOI] [PubMed] [Google Scholar]
- 128.Connolly SJ, Pogue J, Eikelboom J, et al. ACTIVE W Investigators Benefit of oral anticoagulant over antiplatelet therapy in atrial fibrillation depends on the quality of international normalized ratio control achieved by centers and countries as measured by time in therapeutic range. Circulation. 2008;118:2029–37. doi: 10.1161/CIRCULATIONAHA.107.750000. [DOI] [PubMed] [Google Scholar]
- 129.White HD, Gruber M, Feyzi J, et al. Comparison of outcomes among patients randomized to warfarin therapy accprding to anticoagulant control. Arch Intern Med. 2007;167:239–45. doi: 10.1001/archinte.167.3.239. [DOI] [PubMed] [Google Scholar]
- 130.Samsa GP, Matchar DB. Relationship between test frequency and outcomes of anticoagulation: A literature review and commentary with implications for the design of randomized trials of patient self-management. J Thromb Thrombolysis. 2000;9:283–92. doi: 10.1023/a:1018778914477. [DOI] [PubMed] [Google Scholar]
- 131.vanWalraven C, Jennings A, Oake N, Fergusson D, Forster AJ. Effect of study setting on anticoagulation control: A systematic review and metaregression. Chest. 2006;129:1155–66. doi: 10.1378/chest.129.5.1155. [DOI] [PubMed] [Google Scholar]
- 132.Garcia-Alamino JM, Ward AM, Alonso-Coello P, et al. Self-monitoring and self-management of oral anticoagulation. Cochrane Database Syst Rev. 2010;4:CD003839. doi: 10.1002/14651858.CD003839.pub2. [DOI] [PubMed] [Google Scholar]
- 133.Wann LS, Curtis AB, Ellenbogen KA, et al. 2011 ACCF/AHA/HRS Focused Update on the management of patients with atrial fibrillation (Update on Dabigatran): a report of the American College of Cardiology Foundation Foundation/American Heart Association Task Force on practice guidelines. J Am Coll Cardiol. 2011;57:1330–7. doi: 10.1016/j.jacc.2011.01.010. [DOI] [PubMed] [Google Scholar]
- 134.Cairns JA, Connolly S, McMurtry S, Stephenson M, Talajic M, CCS Atrial Fibrillation Guidelines Committee Canadian Cardiovascular Society atrial fibrillation guidelines 2010: prevention of stroke and systemic thromboembolism in atrial fibrillation and flutter. Can J Cardiol. 2011;27:74–90. doi: 10.1016/j.cjca.2010.11.007. [DOI] [PubMed] [Google Scholar]
- 135.Ruff CT, Giugliano RP, Antman EM, et al. Evaluation of the novel factor Xa inhibitor edoxaban compared with warfarin in patients with atrial fibrillation: design and rationale for the Effective aNticoaGulation with factor xA next GEneration in Atrial Fibrillation-Thrombolysis In Myocardial Infarction study 48 (ENGAGEAF-TIMI 48) Am Heart J. 2010;160:635–41. doi: 10.1016/j.ahj.2010.06.042. [DOI] [PubMed] [Google Scholar]
- 136.Freeman JV, Zhu RP, Owens DK, et al. Cost-effectiveness of dabigatran compared with warfarin for stroke prevention in atrial fibrillation. Ann Intern Med. 2011 Jan 4;154(1):1–11. doi: 10.7326/0003-4819-154-1-201101040-00289. [DOI] [PubMed] [Google Scholar]
- 137.Shah SV, Gage BF. Cost-effectiveness of dabigatran for stroke prophylaxis in atrial fibrillation. Circulation. 2011 Jun 7;123(22):2562–70. doi: 10.1161/CIRCULATIONAHA.110.985655. [DOI] [PubMed] [Google Scholar]
- 138.Mega JL, Braunwald E, Wiviott SD, et al. ATLAS ACS 2–TIMI 51 Investigators Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J Med. 2012 Jan 5;366(1):9–19. doi: 10.1056/NEJMoa1112277. [DOI] [PubMed] [Google Scholar]
- 139.van Ryn J, Stangier J, Haertter S, et al. Dabigatran etexilate—a novel, reversible, oral direct thrombin inhibitor: interpretation of coagulation assays and reversal of anticoagulant activity. Thromb Haemost. 2010 Jun;103(6):1116–27. doi: 10.1160/TH09-11-0758. [DOI] [PubMed] [Google Scholar]