Article

Anticoagulation in Atrial Fibrillation

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

Patients with atrial fibrillation (AF) are at increased thromboembolic risk, and they suffer more severe strokes with worse outcomes. Most thromboembolic complications of AF are eminently preventable with oral anticoagulation, and the increasing numbers of AF patients mean antithrombotic therapy is the most crucial management aspect of this common arrhythmia. Despite the proven efficacy of warfarin, a string of limitations have meant that it is underused by physicians and patients alike. This has prompted a search for new anticoagulants that could overcome many of the inconveniences of dose variability and anticoagulant monitoring associated with warfarin, but without sacrificing efficacy in thromboprophylaxis. The arrival of new oral anticoagulants has been complemented by improved risk stratification schemes, which permit clinicians to easily and reliably identify patients requiring anticoagulation and their bleeding risk. These advances in AF treatment will hopefully translate into improved outcomes for patients, especially as our experience with the new agents grows.

Disclosure:Yousif Ahmad has no conflits of interest to declare. Gregory YH Lip has served as a consultant for Bayer, Astellas, Merck, Astr aZeneca, Sanofi, BMS/Pfizer, Daiichi-Sankyo, Biotronik, Portola and Boehringer Ingelheim; he has been on the speakers bureau for Bayer, BMS/Pfizer, Boehring er Ingelheim and Sanofi Aventis.

Received:

Accepted:

Support:The publication of this article was supported by Daiichi Sankyo Europe GmbH. The views and opinions expressed are those of the authors and not necessarily those of Daiichi Sankyo Europe GmbH.

Correspondence Details:Gregory YH Lip, University of Birmingham Centre for Cardiovascular Sciences, City Hospital, Birmingham B18 7QH, UK. E : g.y.h.lip@bham.ac.uk

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Atrial fibrillation (AF) leads to a prothrombotic state1 and places patients at risk of thromboembolic disease. The most common and serious complication of thromboembolism is stroke, and AF is held responsible for 25 % of all strokes.2 Strokes in the context of AF are associated with a higher mortality, longer hospital stay and lower levels of independence at discharge.3 These factors combine to make antithrombotic therapy the most important management consideration for AF patients.

AF is a growing clinical problem owing to the ageing population, and the advances in treatment and improved survival of patients with cardiac disorders such as ischaemic heart disease and heart failure, which predispose to AF.4 It is estimated that, by 2050, up to 16 million people in the US will suffer from AF,5 which is already the commonest arrhythmia encountered in clinical practice.6 The healthcare costs are substantial and AF accounts for 1 % of the National Health Service expenditure in the UK.7

Most thromboembolic complications associated with AF can be prevented with anticoagulant therapy.8 For over fifty years, the only effective therapeutic option to prevent stroke in AF patients were vitamin K antagonists – i.e., warfarin. Although warfarin has been proved to be highly efficacious as thromboprophylaxis, its limitations have negatively affected physicians’ willingness to prescribe – and patients’ willingness to receive – this effective treatment.9 This variability in prescribing habits for thromboprophylaxis has seen large numbers of patients left unprotected against stroke or inadequately protected with antiplatelet therapy,10 despite evidence highlighting that aspirin is an inferior option and carries its own substantial bleeding risk.11 The last decade has therefore seen an intensive search for novel oral anticoagulant drugs that could overcome the limitations associated with warfarin and see more patients with AF properly protected against stroke.

In addition, there has been advancement in the risk stratification of patients with AF. Although AF increases the risk of stroke fivefold,12 this risk is not homogenous and depends on the presence or absence of specific stroke risk factors.13 These risk factors were incorporated into the simple CHADS2 score14 and used to artificially categorise patients as ‘low’, ’intermediate’ or ‘high’ risk. Traditionally, guidelines have recommended oral anticoagulation for high-risk patients, aspirin for low-risk patients, and either anticoagulation or aspirin for the intermediate-risk group. The CHADS2 score also classifies many patients as ‘intermediate-risk’,15 and there is evidence that warfarin is superior to aspirin for this group of patients.16 Similarly, the evidence suggests that aspirin is ineffective as stroke prevention for patients identified as ‘low-risk’ by CHADS2.17 Therefore the utilisation of the CHADS2 score in isolation would result in substantial numbers of patients at risk of stroke being undertreated.

These limitations of the CHADS2 score prompted efforts to establish a new schema that would be more inclusive of common stroke risk factors and would reliably identify ‘truly low-risk’ patients who do not require antithrombotic therapy, as well as reduce the number of patients at risk of stroke being denied oral anticoagulation.

The CHA2DS2-VASc score18 has been proposed and, in various independent validation cohorts, has been shown to reliably identify ‘truly low-risk’ patients (annual stroke rate <1 % per year) and classes fewer patients as ‘intermediate-risk’.19 It also seems to be at least as good as – or possibly better than – the CHADS2 score at identifying ‘high-risk’ patients.19 This score has now been included in international guidelines.20

Table 1 outlines the risk factors that make up the CHA2DS2-VASc score. Congestive heart 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 feared complication of antithrombotic therapy, and this can limit the prescription of oral anticoagulants.21 The HAS-BLED score22 is a simple tool to aid clinicians in undertaking a bleeding risk assessment and prompts them to consider the correctable risk factors for bleeding, such as labile international normalized ratios (INRs), uncontrolled hypertension and concomitant drugs. It can periodically be reassessed and has been validated in various large real-world cohorts,23 performing favourably compared with other bleeding risk scores.24 The HAS-BLED score has been incorporated into international guidelines.

Table 2 outlines the risk factors that make up the HAS-BLED score. Hypertension is defined as a systolic blood pressure >160 mmHg. One point is awarded for abnormal renal function and another point for abnormal liver function, and the same applies to drugs and/or alcohol. A score of 0–2 indicates a low risk of bleeding; a score ≥3 indicates a high risk of bleeding.

Many of the risk factors for stroke are also risk factors for bleeding and, at higher HAS-BLED scores, the net clinical benefit of warfarin is actually greater. This is because there is a greater and more dramatic reduction in the risk of ischaemic stroke and a comparatively smaller increase in the risk of bleeding when warfarin is given to patients with high HAS-BLED scores. Patients with CHA2DS2-VASc scores of 0 are truly low-risk and derive a negative net clinical benefit from warfarin.25

Warfarin

The efficacy of warfarin for preventing stroke and systemic embolism in AF patients was demonstrated in a string of randomised controlled trials (RCTs) over the preceding three decades, with a two-thirds risk reduction in ischaemic stroke compared with placebo. These data led to an increase in the use of warfarin and a consequent decrease in the number of strokes.26 As well as having a clear benefit over placebo, warfarin was also demonstrated to be superior to aspirin for stroke prevention in non-valvular AF.27 An RCT dedicated to evaluating stroke prevention in elderly patients with AF also showed the superiority of warfarin for the prevention of stroke, with no difference between warfarin and aspirin for major bleeding or intracranial haemorrhage.28

The CHA2DS2-VASc Score

Article image

Table 2: The HAS-BLED Score for Risk of Bleeding

Article image

Table 3: Limitations of Warfarin

Article image

The benefit of warfarin is dependent on the amount of time that patients spend with their INRs in therapeutic range (TTR, time in therapeutic range).29 The optimal INR for patients with AF on warfarin for protection against thromboembolism has been established as 2.0–3.0, with an increased risk of thromboembolic and bleeding complications outside of this range. An increased TTR is associated with less thromboembolism, less bleeding, fewer myocardial infarctions and fewer deaths.30,31 Small improvements in TTR translate into significant benefits,32 with a low TTR potentially obliterating the benefit of anticoagulation. Self-monitoring can improve the quality of INR control33 and may bring the TTR closer to that achieved in clinical trials.34 Despite its unequivocal efficacy when properly used, warfarin has well documented limitations (see Table 3).35

Alternatives to warfarin must be proven to reliably perform at least as well as warfarin in RCTs, with an acceptable safety profile (ximelegatran was withdrawn due to hepatotoxicity36). New drugs should circumvent many of the limitations associated with warfarin that necessitate regular coagulation monitoring. They should therefore feature fixed-dose regimens, oral formulations, wide therapeutic windows, low propensity for food and drug interactions, and predictable pharmacokinetics and pharmacodynamics with little inter- and intra-patient variability.

Antiplatelet Therapy

In 2006, the ACTIVE-W (Atrial fibrillation clopidogrel trial with irbesartan for prevention of vascular events) compared dual antiplatelet therapy with aspirin and clopidogrel to warfarin for the prevention of thromboembolism in AF.37 The trial was stopped early due to the clear superiority of warfarin over dual antiplatelet therapy. Furthermore, the rates of bleeding in the two groups were very similar (2.4 % per year for clopidogrel and aspirin versus 2.2 % per year for warfarin). The evidence proves conclusively that antiplatelet therapy is an inferior option when compared with warfarin for stroke prevention. The comparable rates of bleeding also mean that, in patients deemed at too great a haemorrhagic risk, oral anticoagulation would not be a suitable treatment option.

Pharmacokinetic &amp; Pharmacodynamic Properties of Novel Anticoagulants

Article image

Novel Oral Anticoagulants

There have been concerted efforts to unearth new drugs that would fulfil the criteria for the ideal anticoagulant and could represent a viable alternative to warfarin. The three new drugs currently available fall into two classes: oral direct thrombin inhibitors (dabigatran) and oral factor Xa inhibitors (rivaroxaban and apixaban).38 Their pharmacokinetic and pharmacodynamic properties are summarised in Table 4.

Dabigatran

Dabigatran is a competitive, direct and reversible inhibitor of thrombin. The RE-LY (Randomized evaluation of long term anticoagulant therapy with dabigatran etexilate) trial was a large RCT comparing dabigatran with warfarin.39 This Phase III, blinded, non-inferiority trial studied 18,113 patients, either with non-valvular AF with a CHADS2 score of 1 or higher, or aged over 65 years with coronary artery disease. Patients were randomised to either dabigatran at a dosage of 110 or 150 mg twice daily or warfarin (INR of 2–3). The primary efficacy outcomes were stroke or systemic embolism. They occurred at a rate of 1.69 % per year in patients assigned to warfarin, versus 1.53 % per year in patients assigned to dabigatran 110 mg (relative risk [RR] 0.91, 95 % confidence interval [CI] 0.74–1.11, p<0.001 for non-inferiority, p=0.34 for superiority compared with warfarin) and 1.11 % in patients assigned to dabigatran 150 mg (RR 0.66, 95 % CI 0.53–0.82, p<0.001 for non-inferiority and superiority compared with warfarin). Thus low-dose dabigatran was non-inferior to warfarin and high-dose dabigatran was superior to warfarin for preventing stroke.

The primary safety outcome was major bleeding (defined as a reduction of 2 g/dl in haemoglobin, transfusion requiring at least two units of blood, or symptomatic bleeding in a critical organ). Major bleeding occurred less frequently with dabigatran 110 mg, and rates of major bleeding were similar with dabigatran 150 mg and warfarin: major bleeding occurred at a rate of 3.36 % per year in patients taking warfarin, 3.11 % per year in patients taking high-dose dabigatran (RR 0.93, 95 % CI 0.81–1.07, p=0.031 versus warfarin) and 2.71 % in patients taking low-dose dabigatran (RR 0.8, 95 % CI 0.69–0.93, p=0.003 versus warfarin). Both doses of dabigatran were associated with a significantly reduced risk of haemorrhagic stroke and intracranial haemorrhage when compared with warfarin.

Warfarin was the better tolerated therapy, with dyspepsia the primary contributor to discontinuation. Discontinuation rates were 21 % in the dabigatran 110 mg group, 21 % in the dabigatran 150 mg group and 17 % in the warfarin group at the end of the second year (p<0.001 for dabigatran versus warfarin). High-dose dabigatran was also associated with an increased risk of gastrointestinal haemorrhage.

Compared with warfarin, dabigatran 150 mg was found to induce a small increase in the number of myocardial infarction events that was not statistically significant.40 Warfarin is protective against myocardial infarction,41 and the small numerical increase in myocardial infarction events with dabigatran must be interpreted in the context of the overall net clinical benefit of dabigatran over warfarin and the lack of any increase in new angina hospitalisations or revascularisations.42

Rivaroxaban

Rivaroxaban is an oral, reversible, direct factor Xa inhibitor.43 Although its half-life is 7–12 hours, factor Xa is inhibited for up to 24 hours. This permits once-daily dosing, which is in contrast with the other novel oral anticoagulants. ROCKET-AF (An efficacy and safety study of rivaroxaban with warfarin for the prevention of stroke and non-central nervous system systemic embolism in patients with non-valvular atrial fibrillation) was a Phase III, randomised, double-blind, non-inferiority trial comparing rivaroxaban (20 mg once daily or 15 mg once daily in patients with moderate renal impairment) with warfarin (INR 2.5) in over 14,000 patients with non-valvular AF and a history of stroke, transient ischaemic attack (TIA) or non-central nervous system embolism or at least two independent risk factors for future stroke.44 Rivaroxaban was similar to warfarin for the primary endpoint of stroke or systemic embolism (event rate 1.71 % per 100 patient years with rivaroxaban versus 2.16 % with warfarin; hazard ratio [HR] 0.79, 95 % CI 0.66–0.96, p<0.001 for non-inferiority).

Bleeding was similar in the two groups (event rate 14.91 % per 100 patient years with rivaroxaban versus 14.52 % with warfarin; HR 1.03, 95 % CI 0.96–1.11, p=0.442). Rivaroxaban induced significantly less fatal bleeding and intracranial haemorrhage, but paradoxically significantly more patients on rivaroxaban required a blood transfusion or suffered a haemoglobin decrease of 2 g/dl or more.

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 %). The trial’s population was generally high-risk, with 86 % of the total population possessing a CHADS2 score of 3 or higher. The quality of INR control, however, was less good, with an average TTR of 55 %.

Apixaban

Apixaban is an oral, selective and reversible direct factor Xa inhibitor.45 The ARISTOTLE (Apixaban for the prevention of stroke in subjects with atrial fibrillation) study was a randomised, Phase III, double-blind trial comparing apixaban 5 mg twice daily with warfarin titrated to an INR between 2 and 3 in over 18,000 patients.46 The primary outcome was stroke (either ischaemic or haemorrhagic) or systemic embolism, which occurred at a rate of 1.27 % per year in the apixaban group versus 1.60 % per year in the warfarin group (HR 0.79, 95 % CI 0.66–0.95, p<0.001 for non-inferiority, p=0.01 for superiority). This was primarily driven by a reduction in haemorrhagic stroke, which occurred at a rate of 0.24 % per year in the apixaban group versus 0.47 % per year in the warfarin group (HR 0.51, 95 % CI 0.35–0.75, p<0.001), as the rates of ischaemic stroke were similar (0.97 % per year in the apixaban group versus 1.05 % per year in the warfarin group [HR 0.92, 95 % CI 0.74–1.13, p=0.42]).

Apixaban demonstrated a benefit with regards to all-cause mortality compared with warfarin: rates of death from any cause were 3.52 % in the apixaban group versus 3.94 % in the warfarin group (HR 0.89, 95 % CI 0.80–0.99, p=0.047). Apixaban caused less major bleeding: 2.13 % per year, versus 3.09 % per year in the warfarin group (HR 0.69, 95 % CI 0.60–0.80, p<0.001). Drug discontinuation occurred less frequently: 25.3 % with apixaban versus 27.5 % with warfarin (p=0.001). Thus apixaban was superior to warfarin in preventing stroke or systemic embolism, caused less bleeding and resulted in lower mortality.

Limitations of Novel Oral Anticoagulants

The arrival of these new agents represents a significant advancement in the treatment of AF, but clinicians must also be aware of the potential drawbacks and limitations of the new drugs (see Table 5). Dabigatran and apixaban require twice-daily dosing, which may lead to missed doses and place the patient in a pro-thrombotic state with increased risk of thromboembolism. Furthermore, anticoagulation monitoring is not required, so it is difficult to objectively evaluate a patient’s compliance with treatment.

Dabigatran has the propensity to cause dyspepsia and gastrointestinal haemorrhage (likely caused by its acidic core), which may limit its use in patients with gastrointestinal upset. Dabigatran and rivaroxaban must be used with caution in patients with moderate renal dysfunction, and dabigatran is contraindicated in patients with severe renal insufficiency. The 75 mg twice daily dose of dabigatran approved by the US Food and Drug Administration in renal impairment was never actually studied in the RE-LY trial.47 A subgroup analysis of the RE-LY trial found that the rates of stroke and thromboembolism were increased in patients with impaired renal function. Renal function should be checked periodically or if a decline may be suspected (commencement of nephrotoxic drugs, dehydration, intercurrent illness).

Potential Limitations of Novel Anticoagulants

Article image

When managing a haemorrhage associated with the use of warfarin, or in other scenarios when prompt reversal of anticoagulation is necessary, there is an established antidote in the form of vitamin K. There are no known antidotes currently available in clinical practice for dabigatran, rivaroxaban or apixaban. Preliminary work is being done on a potential factor Xa inhibitor antidote,48 which works as a decoy type molecule. A Phase IV study is investigating various reversal strategies for dabigatran,49 and there is interest around an antibody against the fragment antigen-binding (FAB fragment) of dabigatran being used as a potential antidote: this may work in a similar manner to antibodies used against digoxin in cases of digoxin toxicity. It is hoped that antidotes will be available for use in the next three to five years.

The absence of an established antidote is important but must be kept in perspective: there are myriad antithrombotics used in cardiovascular medicine today – including aspirin, clopidogrel, bivalirudin and tirofiban – that do not have an absolute antidote available. The general management strategies of haemorrhage on a novel anticoagulant may include stopping the drug, local measures to cease bleeding, intravenous fluid and blood product support, maintaining a diuresis for renally excreted drugs and the use of haemostatic agents such as prothrombin complex where necessary.

Furthermore, there is no evidence base to guide management in certain specific challenging clinical scenarios, such as an intentional overdose of anticoagulants, switching between warfarin and new agents, major bleeding, and anticoagulant bridging in the peri-operative period. Clinicians can, however, draw on expert consensus on how to manage these situations with dabigatran,50,51 as well as on a position document from the Italian Federation of Thrombosis Centers.52

Conclusions

Clinicians now have an improved array of antithrombotic strategies to prevent stroke in AF patients, which should translate into improved outcomes for patients and a decline in the number of people suboptimally protected with antiplatelet therapy. New anticoagulants have already been incorporated into international guidelines, and physicians will therefore be tasked with choosing between a number of oral agents. While it is always difficult to infer comparisons between drugs that have not been tested against each other, there are some differences in the evidence, which clinicians may use to help determine the optimal antithrombotic strategy.

Rivaroxaban and apixaban have not been shown to cause dyspepsia, so may be the optimal option for patients unable to take dabigatran due to dyspepsia. When used in a low-dose twice-daily regimen, rivaroxaban also has a benefit for patients with a recent acute coronary syndrome (ACS),53 but the impact of the AF prophylaxis dose (20 mg once daily) plus dual antiplatelet therapy in the setting of an ACS has not been studied. Rivaroxaban is the only novel oral anticoagulant with once-daily dosing, which represents a small advantage over the other agents.

All three drugs also demonstrate positive bleeding profiles when compared with warfarin, especially intracranial haemorrhage and haemorrhagic stroke. Dabigatran is the only drug to significantly reduce the risk of ischaemic stroke.

Irrespective of the putative similarities and differences between the new antithrombotic agents, we are entering a new era for stroke prevention in AF patients which should result in improved outcomes, with more patients than ever receiving effective and safe thromboprophylaxis. Although there is limited evidence informing us on how to deal with specific scenarios, such as peri-operative bridging and anticoagulant overdose, all the safety data available are very reassuring. As our experience with these new drugs grows, we will better be able to deal with the challenges they present.

References

  1. Watson T, Shantsila E, Lip GY. Mechanisms of thrombogenesis in atrial fibrillation: Virchow’s triad revisited. Lancet 2009;373:155–66.
    Crossref | PubMed
  2. 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.
    Crossref | PubMed
  3. Steger C, Pratter A, Martinek-Bregel M, et al. Stroke patients with atrial fibrillation have a worse prognosis than patients without: data from the Austrian Stroke registry. Eur Heart J 2004;25:1734–40.
    Crossref | PubMed
  4. Tsang TS, Gersh BJ. Atrial fibrillation: an old disease, a new epidemic. Am J Med 2002;113:432–5.
    Crossref | PubMed
  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.
    Crossref | PubMed
  6. 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.
    Crossref | PubMed
  7. Stewart S, Murphy NF, Walker A, et al. Cost of an emerging epidemic: an economic analysis of atrial fibrillation in the UK. Heart 2004;90:286–92.
    Crossref | PubMed
  8. 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.
    Crossref | PubMed
  9. 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.
    Crossref | PubMed
  10. Ogilvie IM, Newton N, Welner SA, et al. Underuse of oral anticoagulants in atrial fibrillation: a systematic review. Am J Med 2010;123:638–45.
    Crossref | PubMed
  11. Lip GY. The role of aspirin for stroke prevention in atrial fibrillation. Nat Rev Cardiol 2011;8:602–6.
    Crossref | PubMed
  12. Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent risk factor for stroke: the Framingham Study. Stroke 1991;22:983–8.
    Crossref | PubMed
  13. 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.
    Crossref | PubMed
  14. Gage BF, Waterman AD, Shannon W, et al. Validation of clinical classification schemes for predicting stroke: Results from the National Registry of Atrial Fibrillation. JAMA 2001;285:2864–70.
    Crossref | PubMed
  15. VanStaa TP, Setakis E, Di Tanna GL, et al. A comparison of risk stratification schemes for stroke in 79,884 atrial fibrillation patients in general practice. J Thromb Haemost 2011;9:39–48.
    Crossref | PubMed
  16. 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.
    Crossref | PubMed
  17. 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 1996;37:447–51.
    Crossref | PubMed
  18. Lip GY, Nieuwlaat R, Pisters R, et al. 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.
    Crossref | PubMed
  19. Van Staa TP, Setakis E, Di Tanna GL, et al. A comparison of risk stratification schemes for stroke in 79,884 atrial fibrillation patients in general practice. J Thromb Haemost 2011;9:39–48.
    Crossref | PubMed
  20. 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 for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC). Europace 2010;12:1360–420.
    Crossref | PubMed
  21. Hylek EM, D’Antonio J, Evans-Molina C, et al. Translating the results of randomized trials into clinical practice: the challenge of warfarin candidacy among hospitalized elderly patients with atrial fibrillation. Stroke 2006;37:1075–80.
    Crossref | PubMed
  22. Lip GY, Andreotti F, Fauchier L, et al. European Heart Rhythm Association. Bleeding risk assessment and management in atrial fibrillation patients. Executive summary of a position document from the European Heart Rhythm Association [EHRA], endorsed by the European Society of Cardiology [ESC] Working Group on Thrombosis. Thromb Haemost 2011;106:997–1011.
    Crossref | PubMed
  23. 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 a nationwide cohort. J Thromb Haemost 2011;9:1460–7.
    Crossref | PubMed
  24. 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 HASBLED (Hypertension, Abnormal Renal/Liver Function, Stroke, Bleeding History or Predisposition, Labile INR, Elderly, Drugs/Alcohol Concomitantly) score. J Am Coll Cardiol 2011;57:173–80.
    Crossref | PubMed
  25. Olesen JB, Lip GY, Lindhardsen J, et al. Risks of thromboembolism and bleeding with thromboprophylaxis in patients with atrial fibrillation: A net clinical benefit analysis using a 'real world' nationwide cohort study. Thromb Haemost 2011;106:739–49.
    Crossref | PubMed
  26. Lakshminarayan K, Solid CA, Collins AJ, et al. Atrial fibrillation and stroke in the general Medicare population: a 10-year perspective (1992 to 2002). Stroke 2006;37:1969–74.
    Crossref | PubMed
  27. 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.
    Crossref | PubMed
  28. Mant J, Hobbs FD, Fletcher K, et al. 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.
    Crossref | PubMed
  29. Gallagher AM, Setakis E, Plumb JM, et al. Risks of stroke and mortality associated with suboptimal anticoagulation in atrial fibrillation patients. Thromb Haemost 2011;106:968–77.
    Crossref | PubMed
  30. Wan Y, Heneghan C, Perera R, et al. Anticoagulation control and prediction of adverse events in patients with atrial fibrillation: a systematic review. Circ Cardiovasc Qual Outcomes, 2008;1:84–91.
    Crossref | PubMed
  31. White HD, Gruber M, Feyzi J, et al. Comparison of outcomes among patients randomized to warfarin therapy according to anticoagulant control. Arch Intern Med 2007;167:239–245.
    Crossref | PubMed
  32. 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.
    Crossref | PubMed
  33. Garcia-Alamino JM, Ward AM, Alonso-Coello P, et al. Selfmonitoring and self-management of oral anticoagulation. Cochrane Database Syst Rev 2010;4:CD003839.
    Crossref
  34. Van Walraven C, Jennings A, Oake N, et al. Effect of study setting on anticoagulation control: A systematic review and metaregression. Chest 2006;129:1155–66.
    Crossref | PubMed
  35. Lin P. Reviewing the reality: why we need to change. Eur Heart J Suppl 2005;7:E15–E20.
    Crossref
  36. Ahrens I, Lip GY, Peter K. New oral anticoagulant drugs in cardiovascular disease. Thromb Haemost, 2010;104:49–60.
    Crossref | PubMed
  37. 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;367:1903–1912.
    Crossref | PubMed
  38. Phillips KW, Ansell J. The clinical implications of new oral anticoagulants: will the potential advantages be achieved? Thromb Haemost. 2010;103:34–9.
    Crossref | PubMed
  39. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 2009;361:1139–51.
    Crossref | PubMed
  40. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Newly identified events in the RE-LY Trial [letter]. New Engl J Med 2010;363:1875–87.
    Crossref | PubMed
  41. Lip GY, Lane DA. Does warfarin for stroke thromboprophylaxis protect against MI in atrial fibrillation patients? Am J Med 2010;123:785–9.
    Crossref | PubMed
  42. Hohnloser SH, Oldgren J, Yang S, et al. Myocardial ischemic events in patients with atrial fibrillation treated with dabigatran or warfarin in the RE-LY (Randomized Evaluation of Long-Term Anticoagulation Therapy) trial. Circulation 2012;125:669–76.
    Crossref | PubMed
  43. 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.
    Crossref | PubMed
  44. Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011;365:883–91.
    Crossref | PubMed
  45. Roser-Jones C, Becker RC. Apixaban: an emerging oral factor Xa inhibitor. J Thromb Thrombolysis 2010;29:141–6.
    Crossref | PubMed
  46. Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2011;365:981–92.
    Crossref | PubMed
  47. 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.
    Crossref | PubMed
  48. Portola Pharmaceuticals. Available at: www.portola.com/PRT064445-Fxa-Inhibitor-Antidote (accessed 5 September 2012).
  49. Study in Healthy Volunteers of the Reversion by Haemostatic Drugs of the Anticoagulant Effect of New Antithrombotics (REVNEWANTICO), Available at: http://clinicaltrials.gov/ct2/show/NCT01210755?term=NCT01210755&rank=1 (accessed 24 August 2012).
  50. 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;103:1116–27.
    Crossref | PubMed
  51. Huisman MV, Lip GY, Diener HC, et al. Dabigatran etexilate for stroke prevention in patients with atrial fibrillation: Resolving uncertainties in routine practice. Thromb Haemost 2012;107:838–47.
    Crossref | PubMed
  52. Pengo V, Crippa L, Falanga A, et al. Questions and answers on the use of dabigatran and perpectives on the use of other new oral anticoagulants in patients with atrial fibrillation. A consensus document of the Italian Federation of Thrombosis Centers (FCSA). Thromb Haemost 2011;106:868–76.
    Crossref | PubMed
  53. Mega JL, Braunwald E, Wiviott SD, et al. Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J Med 2012;366:9–19.
    Crossref | PubMed