Skip to main content
Clinical Cardiology logoLink to Clinical Cardiology
. 2016 Oct 7;39(12):739–746. doi: 10.1002/clc.22582

New Oral Anticoagulants in Nonvalvular Atrial Fibrillation

Fatima Urooj 1, Abhishek Kulkarni 2,3, Dwight Stapleton 2,3, Edo Kaluski 2,3,
PMCID: PMC6490750  PMID: 27716952

Abstract

The choice of an oral anticoagulant (OAC) for patients with nonvalvular atrial fibrillation (NVAF) is a major and complex clinical decision taking into account the individual risk‐benefit ratio and bearing in mind the chronicity of therapy. This review focuses on the safety and efficacy of new oral anticoagulants (NOACs) compared with conventional vitamin K antagonists (VKA) in patients with NVAF. Current data suggest that NOACs are at least as effective and safe as VKAs for most NVAF subjects. The NOACs do not mandate dietary restrictions and regular pharmacodynamic monitoring, and they seem to have lesser incidence of intracranial or fatal bleeding when compared with VKAs. However, both dabigatran 150 twice daily and rivaroxaban have a slightly higher incidence of gastrointestinal bleeding when compared with VKAs. The article will delineate the current knowledge as well as scientific gaps related to the choice and dosage of anticoagulation regimens for various NVAF subsets and will address certain common clinical scenarios requiring special considerations. The article also addresses the shortcomings of NOACs: lack of therapeutic pharmacokinetic and pharmacodynamic targets, absence of tools to assess compliance and efficacy, rigid and limited dosage options, and absence of effective and inexpensive reversal agents.

Keywords: Stroke prevention, Arrhythmia/all, General clinical cardiology/adult, Clinical trials

1. PREVALENCE OF ATRIAL FIBRILLATION

Atrial fibrillation (AF) is associated with 5‐fold increased risk of stroke1, 2, 3 and is the most common preventable cause of stroke. Strokes related to AF tend to be more disabling and have higher recurrence and case fatality rates. Nonvalvular AF (NVAF) affects approximately 5 million US residents, and that number is expected to at least double by 2050.4 Prevalence of AF increases with age, affecting approximately 5% of persons age >65 years and 10% of persons age >80 years.5, 6

Whether AF is paroxysmal, persistent, or permanent, and regardless of symptom severity, most patients with NVAF should receive OACs to prevent thromboembolic events. Sadly, a significant proportion of NVAF patients does not receive OACs at all or suffers from suboptimal oral anticoagulation.

2. RISK STRATIFICATION FOR STROKE IN NONVALVULAR ATRIAL FIBRILLATION

2.1. CHADS2 or CHA2DS2‐VASc Scores

Risk stratification for patients with NVAF is based on scoring systems, such as the CHADS2 or CHA2DS2‐VASc scores, and on the AF burden. A score of 0 or 1 is defined as low risk, and a score ≥2 is considered moderate to high risk. When compared with the CHADS2 score, the CHA2DS2‐VASc score has better discriminating power in identifying the low‐risk NVAF patients who may still benefit from anticoagulation.7 The clinical risk scores provide good sensitivity and negative predictive value for stroke; however, they are limited by poor specificity, positive predictive value, and overall accuracy.8 Therefore, in patients with intermediate and high scores, these scoring systems might not provide sufficient discrimination of the stroke risk.9

The greatest virtue of the stroke risk scores in NVAF is their simplicity and ease of use. However, this is also the drawback of this method, because:

  1. There is no accounting for severity or duration of the conditions (for instance, newly diagnosed borderline diabetes mellitus [DM] and insulin‐dependent DM of 20 years duration with considerable target‐organ damage will both receive a single point; similarly, the ages of 74 and 65 years will both qualify for 1 point).

  2. Certain parameters impose more risk than others (DM > heart failure).

  3. There is no accounting for other items associated with stroke risk (renal insufficiency, spontaneous echocardiographic contrast, hyperthyroidism, elevated D‐dimers, left atrial enlargement).

  4. The atrial fibrillation burden is not taken into account. The overall annual stroke risk associated with NVAF varies between 0% and 15.2% and correlates with the CHA2DS2‐VASc score.

Females with CHA2DS2‐VASc score of 1 or males with CHA2DS2‐VASc score of 0 are at low risk and mostly should not be treated, OAC. Among subjects with single risk factor, the reported annual thromboembolic event rates are variable, ranging from 0.5% to 3%. A recent retrospective analysis concluded that risk associated with score of 1 is much lower than initially thought; hence, these patients might not require anticoagulation.10 In this cohort, the single risk‐factor severity, other patient characteristics, and patient preference and means should be taken into account. There are no randomized phase 3 trials assessing the efficacy and safety of any NOAC in patients with a single risk factor; however, real‐world observational data are emerging.

2.1.1. AF Burden

The burden of NVAF seems to have some impact on both thromboembolism and cardiovascular mortality. Based on the Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE)11 and Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation (ROCKET‐AF) trials, the risk of systemic embolism and mortality, respectively, is somewhat higher in patients with permanent or persistent NVAF than with paroxysmal NVAF; however, this has been conflicted by other reports.

A current European consensus document suggests that first unprovoked episode of NVAF should be treated in the same way we treat repeated episodes of NVAF. The use of anticoagulant or the dosage should not be influenced by the pattern, frequency, or number of AF episodes.

2.1.2. Bleeding Risk on Anticoagulation

All NVAF should be assessed for bleeding propensity prior to the initiation of OAC. Scoring systems to identify the inherent risk of bleeding during OAC have been developed and subsequently validated. Some of the commonly used scoring systems are HEMORR2HAGES (hepatic or renal disease, ethanol abuse, malignancy, older age [>75 years], reduced platelet count or function, rebleeding risk, hypertension [uncontrolled], anemia, genetic factors [CYP2C9 variant], excessive fall risk, stroke), HAS‐BLED (hypertension, abnormal renal and liver function, stroke, bleeding history or predisposition, labile international normalized ratio [INR], elderly age >65 years), Registro Informatizado de la Enfermedad TromboEmbólica (RIETE), and Anticoagulation and Risk Factors in Atrial Fibrillation (ATRIA).

A HAS‐BLED score ≥3 indicates high risk of bleeding12; HAS‐BLED is currently endorsed by most guidelines.13, 14

2.1.3. Device‐Detected Atrial Fibrillation

The medical community is challenged to identify patients with asymptomatic AF to initiate primary stroke prevention. Long‐term Holter monitors, event monitors, and implanted devices enhance the detection of asymptomatic AF.

Certain trials provided data regarding device‐detected atrial high‐rate episodes (AHRE; >190–220 bpm), which could be AF, atrial flutter, or atrial or supraventricular tachycardia, and the risk of stroke.15 Most of these studies suggested that AHREs were associated with increased risk of thromboembolism,16, 17 stroke, AF,18 and even death.19, 20 A temporal relationship between AHREs and stroke could not be established. Additional studies are needed to further clarify the correlation between device‐detected AHREs and stroke and when OAC should be considered. The guidelines do not relate to device‐detected NVAF as a separate entity; hence, it should be treated similar to clinically detected NVAF.21

3. NEW ORAL ANTICOAGULANTS FOR NONVALVULAR ATRIAL FIBRILLATION

The commercially available NOACs in the United States are apixaban, dabigatran, rivaroxaban, and edoxaban. When compared with VKAs, some of their distinguishing features are rapid onset of action (1–3 hours), plasma half‐life of 7 to 15 hours, and most of them being partially excreted by the kidneys.22, 23, 24, 25, 26, 27 The characteristics of these agents are summarized in Table 1.

Table 1.

Commercially Available NOACs23, 24, 25, 26, 27

Dabigatran Rivaroxaban Apixaban Edoxaban
Mechanism Thrombin (Factor II) inhibitor Factor Xa inhibitor Factor Xa inhibitor Factor Xa inhibitor
Bioavailability 7% >80% 50% 62%
Peak level, h 2–3 3 3–4 1–2
Half‐life, h 12–17 5–13 10–14 10–14
Dosing 150 mg b.i.d.; 110 mg b.i.d.a (75 mg b.i.d. for CrCl 15–30 mL/m) 20 mg daily with food (15 mg for CrCl 15–50 mL/m) 5 mg b.i.d. (2.5 mg b.i.d. if 2 of: Cr >1.5 mg/dL, age >80 years, wt <60 kg) 60 mg daily; CrCl 50–95 mL/m (30 mg for CrCl 15–50 mL/m)
Renal cleared, % 80 36 25 50
Drug interactions P‐gp inhibitors CYP3A4 inhibitors and inducers P‐gp and CYP3A4 inhibitors and inducers P‐gp inhibitors
Pharmacodynamic monitoring Ecarin clotting time > thrombin time > aPTT and ACT Direct Xa activity, PTT mildly prolonged Direct Xa activity, PTT mildly prolonged Direct Xa activity, PTT mildly prolonged
When to stop presurgery ≥24 h ≥24 h ≥24 h ≥24 h
Reversal agent Idarucizumab Andexanet alfaa Andexanet alfaa Andexanet alfaa

Abbreviations: ACT, activated clotting time; aPTT, activated partial prothrombin time; b.i.d., twice daily; Cr, creatinine; CrCl, creatinine clearance; CYP3A4, cytochrome P450 3A4; NOAC, new oral anticoagulant; P‐gp, P‐glycoprotein; PTT, partial prothromboplastin time; wt, weight.

a

Not commercially available in the United States.

3.1. Rate and Rhythm Control With New Oral Anticoagulants

Special attention should be given to considerations of coexisting rhythm‐ and rate‐control agents because some of these agents interact with NOACs. The interactions among these agents are summarized in Table 2.

Table 2.

Interactions of Rate‐ and Rhythm‐Control Agents With NOACs

Drug NOAC Adjustment
Verapamil Dabigatran and edoxaban dose should be reduced, but no dose reduction is required for apixaban or rivaroxaban.
Diltiazem Dose adjustment required only for rivaroxaban in subjects with renal dysfunction.
Dronedarone Dabigatran is contraindicated, and edoxaban and rivaroxaban dose should be reduced.
Amiodarone Patients should receive a reduced rivaroxaban dose in the presence of renal dysfunction.

Abbreviations: NOAC, new oral anticoagulant.

4. THE EFFICACY AND SAFETY OF NEW ORAL ANTICOAGULANTS IN NONVALVULAR ATRIAL FIBRILLATION

Four major NVAF trials have compared the efficacy and safety of NOACs with VKAs (target INR, 2–3): Randomized Evaluation of Long‐Term Anticoagulation Therapy (RE‐LY) for dabigatran,28 ROCKET‐AF for rivaroxaban,29 ARISTOTLE for apixaban,30, 31 and the Global Study to Assess the Safety and Effectiveness of Edoxaban (DU‐176b) vs Standard Practice of Dosing With Warfarin in Patients With Atrial Fibrillation (ENGAGE‐AF) for edoxaban.32 These phase 3 trials resulted in the approval of these agents for commercial use for NVAF. Table 3 summarizes the differences in design, study population, and outcomes of these trials. When compared with VKAs, all NOACs reduced the risk of intracerebral, life‐threatening, and fatal bleeds. Certain NOAC regimens demonstrated superiority in safety (dabigatran 110 mg twice daily), efficacy (dabigatran mg 150 twice daily), or both (apixaban).

Table 3.

Clinical Trials of NVAF for Commercially Available NOACs28, 29, 30, 31, 32

Dabigatran Rivaroxaban Apixaban Edoxaban
Population/duration 18,113/24 mo 14,264/290 d 18,201/1.8 y 21,105/2.8 y
Study name RE‐LY28 ROCKET‐AF29 ARISTOTLE30, 31 ENGAGE‐AF32
Doses 110 mg/150 mg a b.i.d. 20 mg/d 5 mg b.i.d. 60 mg b /30 mg/d
CrCl for ↓dose None 30–50 mL/m, 15 mg Cr >1.5 mg/dL, age >80 y, wt <60 kg, 2.5 mg b.i.d. 30–50 mL/m
Means CHADS2 score 2.1 3.5 2.1 2.8
Prior stroke/emboli, % 20 55 19 28
Blinding Open Double Double Double
TTR, % 64 57.8 65.7 65
Noninferiority trial Yes Yes Yes Yes
Stroke/SE ↓ARR, % 0.58, P = 0.01 a 0.3, P = 0.26 0.33, P = 0.011 0.3, P = 0.017 b
Major bleed ↓ARR, % 0.3, P = 0.31 a −0.15, P = 0.5 0.96, P = 0.001 0.68, P = 0.017 b
ICH bleed ↓ARR, % 0.4 a 0.2 0.5 0.2 b
Fatal bleed ↓ARR, % N/A c 0.3 0.3 0.2 b
All death ↓ARR, % 0.5 a 0.4 0.4 0.36 b

Abbreviations: ARISTOTLE, Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation; ARR, absolute risk reduction; b.i.d., twice daily; CHADS2, congestive heart failure, HTN, age ≥75 y, DM, prior stroke/TIA/TE; Cr, creatinine; CrCl, creatinine clearance; DM, diabetes mellitus; ENGAGE‐AF, Global Study to Assess the Safety and Effectiveness of Edoxaban (DU‐176b) vs Standard Practice of Dosing With Warfarin in Patients With Atrial Fibrillation; HTN, hypertension; ICH, intracerebral hemorrhage; N/A, not applicable; NOAC, new oral anticoagulant; NVAF, nonvalvular atrial fibrillation; RE‐LY, Randomized Evaluation of Long‐Term Anticoagulation Therapy; ROCKET‐AF, Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation; SE, systemic embolism; TE, thromboembolism; TIA, transient ischemic attack; TTR, time in therapeutic range; wt, weight.

a

Dose of 150 mg b.i.d. (n = 6076) compared with warfarin (n = 6022).

b

Analysis of only 60‐mg dose (n = 7012) vs warfarin (n = 7012).

c

Life‐threatening bleeding reduced by 0.35%.

The Safety, Tolerability, and Pilot Efficacy of Oral Factor Xa Inhibitor Betrixaban Compared to Warfarin (EXPLORE‐Xa) was a phase 2 study that compared different doses of betrixaban (40, 60, and 80 mg daily) with warfarin. The study concluded that betrixaban is well tolerated, with similar or lower rates of bleeding compared with warfarin.27 The efficacy of the 60‐mg and 80‐mg doses was similar to warfarin.

When compared with VKAs, all NOACs are associated with reduced intracranial hemorrhage and fatal bleeding.33 Patients with well‐controlled INR on VKAs with time in therapeutic range >70% and no extreme outlying INR measurements (>4 or <1.8) could be still maintained on VKAs.

5. REVERSAL OF NEW ORAL ANTICOAGULANTS

In case of massive bleeding or emergent surgical procedure after standard resuscitation protocol, gastric lavage with activated charcoal can be used if ingestion occurred within ≤3 hours. Hemodialysis can be considered for dabigatran (due to low protein binding),34 but not for factor Xa inhibitors.

Trials demonstrated benefit from using prothrombin complex concentrates (PCCs) in the treatment of bleeding associated with rivaroxaban35; however, the role of PCCs in dabigatran reversal is less conclusive.36 Four‐factor PCC (50 IU/kg) was effective for treating bleeding caused by edoxaban.37

Recently, idarucizumab (humanized monoclonal antibody fragment targeting specifically to dabigatran) was approved by the US Food and Drug Administration for reversing dabigatran‐related anticoagulation in emergencies.38 Andexanet is a recombinant factor Xa protein that has been shown to be effective for neutralizing the anticoagulant effect of factor Xa inhibitors in healthy volunteers; however, its clinical efficacy and safety during Xa inhibitor–related bleeding remain to be proven.39

6. CARDIOVERSION, ABLATION, AND SURGERY ON NEW ORAL ANTICOAGULANTS

6.1.

6.0.1. Cardioversion

Thirty‐day post‐cardioversion stroke rates are approximately 1% with VKAs and 5% to 7% without VKAs. Comparative data regarding the safety of cardioversion with NOACs and VKAs emerges from RE‐LY (1983 cardioversions in 1270 patients), ROCKET‐AF (181 electrical cardioversions and 194 pharmacologic cardioversions), and ARISTOTLE (743 cardioversions in 540 patients), suggesting that dabigatran, rivaroxaban, and apixaban have similar safety as VKAs.40 The X‐VeRT study (Explore the Efficacy and Safety of Once‐daily Oral Rivaroxaban for the Prevention of Cardiovascular Events in Subjects With Nonvalvular Atrial Fibrillation Scheduled for Cardioversion) investigated the use of rivaroxaban in 1504 patients. The composite primary endpoint (stroke, transient ischemic attack, myocardial infarction, and cardiovascular death) occurred in 0.51% of the rivaroxaban arm and in 1.05% of the warfarin arm, whereas major bleeding occurred in 0.6% of the rivaroxaban arm and in 0.8% of the warfarin arm. Rivaroxaban was associated with shorter mean time to cardioversion (25 days vs 34 days; P < 0.001). These results added additional support for the safety of rivaroxaban‐based cardioversion. Both EMANATE (http://www.clinicaltrials.gov NCT02100228; phase 4 clinical trial) and ENSURE‐AF (http://www.clinicaltrials.gov NCT02072434; phase 3 clinical trial) are currently ongoing to assess the safety of cardioversion with apixaban and edoxaban, respectively, when compared with usual care.

6.0.2. Atrial Fibrillation Ablation Procedures

Atrial fibrillation ablation procedures mandate anticoagulation during the procedure and for ≥3 months post‐ablation. Periprocedural embolization still occurs in 1% to 5% of AF ablation patients, whereas asymptomatic silent new magnetic resonance imaging lesions occur in 10% to 15%. The mainstay of anticoagulation therapy has been uninterrupted warfarin or unfractionated heparin (with target activated clotting time of 300 seconds). The Study Exploring Two Treatment Strategies in Patients With Atrial Fibrillation Who Undergo Catheter Ablation Therapy (VENTURE‐AF) trial prospectively compared the safety of uninterrupted rivaroxaban 20 mg to that of uninterrupted warfarin in 250 subjects and found these 2 regimens to provide similar safety.41 Ongoing randomized clinical trials Apixaban During Atrial Fibrillation Catheter Ablation: Comparison to Vitamin K Antagonist Therapy (AFAXA) using apixaban (http://www.clinicaltrials.gov NCT02227550) and Uninterrupted Dabigatran Etexilate in Comparison to Uninterrupted Warfarin in Pulmonary Vein Ablation (RE‐CIRCUIT) for dabigatran (http://www.clinicaltrials.gov NCT02348723) may provide additional information regarding the safety of these agents during AF ablation. The most validated protocol for AF ablation is uninterrupted warfarin, which, according to the European Society of Cardiology guidelines and recent consensus paper, should be preferred over NOACs.

6.0.3. Surgical Interventions

In case of surgical interventions, preprocedural discontinuation of these agents should be based on procedural bleeding risks and the predicted pharmacokinetics of the drug in the specific patient. For procedures with low risk of bleeding, it is not necessary to hold NOACs and procedure can ideally be planned within 24 hours of drug discontinuation.42 For procedures that carry substantial bleeding risk, holding NOACs for ≥48 hours and resumption should be based on the surgical procedure and surgeon preference (bearing in mind that therapeutic anticoagulation occurs within hours of the initial dose).

7. ANTIPLATELET THERAPY AND NEW ORAL ANTICOAGULANTS

7.1. Stable Coronary Artery Disease and Stable Peripheral Arterial Occlusive Disease

In the RE‐LY trial, triple antithrombotic therapy (the addition of aspirin and clopidogrel to either dabigatran [150 mg twice daily or 110 mg twice daily] or warfarin) resulted in doubling of major bleeding events. Even the addition of either aspirin or clopidogrel to dabigatran or other NOACs resulted in excessive major bleeding.

For stable coronary artery disease (CAD), the meta‐analysis from the 4 NOAC clinical trials suggests that the event rates for patients treated only with NOAC or warfarin is <1.5% per year. A Joint European consensus document43 and FDA Medicare analysis refute the notion that excessive incidence of myocardial infarction occurs when using dabigatran as a single antithrombotic agent.

A recent European consensus paper44 suggests that for both stable CAD and peripheral artery disease, the preferred therapy is monotherapy with a NOAC (with no clear preference of a particular agent) with an addition of aspirin in rare cases in which individual risk assessment predicts exceedingly high atherothrombotic risk.

7.2. Acute Coronary Syndrome and Recent Coronary Intervention and Stenting

The role of anticoagulation in addition to standard dual antiplatelet therapy (triple therapy) with recent coronary stenting remains controversial. The results of the What Is the Optimal Antiplatelet and Anticoagulant Therapy in Patients With Oral Anticoagulation and Coronary Stenting (WOEST) trial support the notion that the addition of clopidogrel alone to warfarin is as effective and safer (significantly fewer bleeding events) than the addition of dual antiplatelet therapy (clopidogrel and aspirin; triple therapy). However, the study was not blinded and had an unusually high rate of bleeding events in the triple‐therapy arm. Contemporary guideline documents do not fully address this issue. Recently published European Society of Cardiology consensus documents have made recommendations in this and other specific challenging clinical scenarios.44, 45, 46

According to the current American College of Cardiology/American Heart Association/Heart Rhythm Society (ACC/AHA/HRS) guidelines, for patients undergoing percutaneous coronary intervention, OACs can be interrupted prior to the procedure to decrease risk of bleeding.47

The recent European consensus paper44 suggests that NVAF patients after coronary stenting can be treated by either warfarin (target INR, 2–2.5) or a lower‐dose NOAC (with no preference). The suggested dosage is apixaban 2.5 mg twice daily, rivaroxaban 15 mg daily, dabigatran 110 mg daily, and edoxaban 30 mg daily. These recommendations are not substantiated by any clinical trial and may not suit patients with excessively high stroke risk based on CHADS2 or CHA2DS2‐VASc score, especially those with a previous embolic event or stroke.

8. MECHANICAL AND BIOLOGIC PROSTHETIC VALVES

8.1. Mechanical Prosthetic Valves

The Dabigatran Etexilate in Patients With Mechanical Heart Valves (RE‐ALIGN) trial45 compared the safety and efficacy of high‐dose dabigatran (trough level, ≥50 ng/mL) with VKAs in 252 subjects with mechanical valves. The dabigatran arm suffered an excessive stroke rate (9 patients vs 0) and more incidents of major bleeding (7 patients vs 2) and a higher rate of valve thrombosis, resulting in study discontinuation. The only choice at this time for mechanical prosthetic valves is a VKA (with target INR based on valve location, type, and associated clinical conditions) and low‐dose aspirin. The use of NOACs is prohibited in these patients.

8.2. Biologic Valves or Post–Transcatheter Aortic Valve Replacement

Current trials show conflicting data regarding the safety and efficacy of prescribing warfarin during the initial 3 to 6 months after surgical biologic aortic valve replacement to reduce thromboembolic complications or cardiovascular mortality.

Biologic aortic or mitral valves do not mandate anticoagulation; so although VKAs are optional for the initial 3 months for both aortic and mitral valve replacement (class IIa recommendation), based on the Society of Thoracic Surgeons registry, VKAs are not routinely prescribed. Usually patients are given aspirin 75 to 100 mg daily for life after any surgical biologic valve replacement.

After transcatheter aortic valve replacement (TAVR), patients receive aspirin ≤100 mg/d for life and clopidogrel 75 mg/d for the initial 30 days. Patients with biologic prosthetic valves were included in ARISTOTLE and ENGAGE‐AF; however, the data regarding this subset of patients has not been published.

Although patients with biologic prosthetic valves have not been subject to large‐scale comparative studies with NOACs, the reason for anticoagulation is the presence of AF and not the prosthetic valve. Consequently, the authors believe that the prevailing practice of off‐label prescription of NOACs to patients with AF harboring biologic surgical valves or post‐TAVR should not be discouraged, even though these patients have not been extensively studied in NOAC trials.

A separate newly diagnosed entity is restricted valve mobility presumed to be related to some degree of leaflet thrombosis in biological aortic valve replacement or TAVR. This condition seems to favorably respond to anticoagulation, but neither warfarin nor NOACs have been extensively evaluated for efficacy and safety in this condition.

8.3. Valvular Heart Disease

Coexisting AF and ≥ moderate rheumatic mitral stenosis poses excessive thromboembolism risk; thus, it should not be evaluated by CHA2DS2‐VASc score. These patients were excluded from major NOAC trials, so safety and efficacy of NOACs in these patients have not been established. Most NOAC trials included other native valvular abnormalities and subset analysis of these patients had comparable outcomes with VKAs and NOACs.

9. PRACTICAL CONSIDERATIONS

9.1. Cost Analysis

Outcomes analysis based on NOAC fundamental trials suggests that NOACs may reduce overall medical costs (excluding drug costs) relative to VKAs.48 A subsequent analysis that incorporated NOAC drug costs suggested that NOACs are cost‐effective alternatives to VKAs.49 A recent analysis showed that average combined patient and insurer anticoagulant spending in the first 6 months after initiation was > $900 greater for patients initiating a NOAC.50

9.2. New Oral Anticoagulant Usage Distribution

A recent evaluation of anticoagulation practices in Europe shows a clear increase in the use of NOACs in AF. The overall usage in Europe is 6%, with considerable regional variability. Germany and Spain have the highest rates of prescription, at 11%, which is attributed to the availability of NOACs through their health scare systems. However, VKA remain the predominant choice.51 Usage of NOAC therapy in the United States is significantly influenced by cost; however, there has been a notable rise in their use. New oral anticoagulants accounted for 62% of all new prescriptions in the United States and 98% of all anticoagulant‐related costs between 2010 and 2013.50

10. SPECIAL TREATMENT GROUPS

The European consensus articles44, 47 on choosing particular anticoagulants and dosages for stroke prevention in NVAF discuss the choice of OAC and dosing for special treatment groups. These recommendations are summarized in Table 4. Most of these recommendations are based on consensus rather than randomized clinical trials.

Table 4.

OAC for AF in Specific Clinical Scenarios43, 44, 45, 46, 47

Category Recommendations
Stable CAD and PAD NOAC mostly without use of antiplatelet agents
After coronary stenting Triple therapy with VKA (target INR 2–2.5) or reduced‐dose NOACa, aspirin 75–100 mg/d, and clopidogrel 75 mg/d
Consider: BMS, abbreviated 6‐mo DAPT for DES, or omission of aspirin if bleeding propensity is high
Secondary stroke prevention NOACs preferred over VKAs unless TTR >70
No addition of antiplatelet agent to OAC is needed
Acute stroke r‐tPA only if anticoagulation by test or history is minimal
Mechanical thrombectomy for proximal intracranial occlusion
Acute ischemic stroke after neuroimaging (repeat imaging pre‐OAC for moderate to severe ischemic stroke) TIA: start OAC immediately
Mild ischemic stroke: start OAC after 3 days
Moderate ischemic stroke: start OAC at 5–7 days
Severe ischemic stroke: start OAC at 12–14 days
History of GI bleed Preference to apixaban and low‐dose dabigatran
End stage renal disease and dialysis VKA or Apixaban 5 mg b.i.d or Rivaroxaban 15 mg/d
Cardioversion VKAs and NOACs appear to be similarly effective
AF ablation Preferred VKA over NOACs (limited data on edoxaban)
Mechanical valves VKA target INR based on valve type, site, and associated conditions, along with aspirin 75–100 mg daily
Moderate/severe rheumatic mitral stenosis VKA target INR 2–3

Abbreviations: AF, atrial fibrillation; b.i.d., twice daily; BMS, bare‐metal stents; CAD, coronary artery disease; CrCl, creatinine clearance; DAPT, dual antiplatelet therapy; DES, drug‐eluting stent; GI, gastrointestinal; INR, international normalized ratio; NOAC, new oral anticoagulant; OAC, oral anticoagulants; PAD, peripheral artery disease; r‐tPA, recombinant tissue plasminogen activator; TIA, transient ischemic attack; TTR, time in therapeutic range; VKA, vitamin K antagonist.

a

Apixaban 2.5 mg b.i.d.; rivaroxaban 15 mg daily with food; dabigatran 110 mg b.i.d.

11. CONCLUSION

All subjects with NVAF should undergo initial and periodic risk‐benefit assessment to delineate their thromboembolic and bleeding risk. This assessment should be discussed and documented prior to therapy initiation. With very few exceptions, NOACs seem to be equally effective when compared with VKAs in NVAF and provide the benefits of rapid onset and offset, no pharmacodynamic monitoring or diet restrictions, fewer drug interactions, and predictable pharmacodynamics. NOACs are associated with decreased rates of intracranial and fatal bleeding, even without the use of reversal agents. New oral anticoagulants offer a reasonable option for patients undergoing cardioversion and AF ablation and facilitate access to surgical and invasive procedures. The relative safety and efficacy of NOACs in certain patient subsets is not established. For life‐threatening bleeding after initial resuscitation, activated charcoal and prothrombin complex concentrate can be used. Specific antidotes such as andexanet alfa (for Xa inhibitors) and idarucizumab (for dabigatran) can further improve outcomes of bleeding or emergency surgery. More dose flexibility and the ability to perform point‐of‐care pharmacodynamic assessments may provide additional safety and efficacy when committing to lifelong therapy.

Urooj F, Kulkarni A, Stapleton D, Kaluski E. New Oral Anticoagulants in Nonvalvular Atrial Fibrillation, Clin Cardiol 2016;39(12):739–746.

Dr. Stapleton is a paid consultant for Bristol‐Myers Squibb. Dr. Kaluski is a paid consultant and investigator for Janssen and Daiichi‐Sankyo.

The authors have no other funding, financial relationships, or conflicts of interest to disclose.

REFERENCES

  • 1. Lane DA, Lip GY. Use of the CHA2DS2‐VASc and HAS‐BLED scores to aid in decision‐making for thromboprophylaxis in nonvalvular atrial fibrillation. Circulation. 2012;126:860–865. [DOI] [PubMed] [Google Scholar]
  • 2. You JJ, Singer DE, Howard PA, et al; American College of Chest Physicians. Antithrombotic therapy for atrial fibrillation: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence‐Based Clinical Practice Guidelines. Chest. 2012;141(2 suppl):e531S–e575S. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Camm AJ, Kirchhof P, Lip GY, et al; European Heart Rhythm Association; European Association for Cardio‐Thoracic Surgery. Guidelines for the management of atrial fibrillation: the Task Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC) [published correction appears in Eur Heart J. 2011;32:1172]. Eur Heart J. 2010;31:2369–2429. [DOI] [PubMed] [Google Scholar]
  • 4. 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 [published correction appears in Circulation. 2006;114:e498]. Circulation. 2006;114:119–125. [DOI] [PubMed] [Google Scholar]
  • 5. Marinigh R, Lip GY, Fiotti N, et al. Age as a risk factor for stroke in atrial fibrillation patients: implications for thromboprophylaxis. J Am Coll Cardiol. 2010;56:827–837. [DOI] [PubMed] [Google Scholar]
  • 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;285:2370–2375. [DOI] [PubMed] [Google Scholar]
  • 7. Troughton RW, Crozier I. Fine tuning risk stratification for atrial fibrillation. J Am Coll Cardiol. 2013;61:2285–2287. [DOI] [PubMed] [Google Scholar]
  • 8. Olesen JB, Lip GY, Hansen ML, et al. Validation of risk stratification schemes for predicting stroke and thromboembolism in patients with atrial fibrillation: nationwide cohort study. BMJ. 2011;342:d124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Lip GY. Stroke and bleeding risk assessment in atrial fibrillation: when, how, and why? Eur Heart J. 2013;34:1041–1049. [DOI] [PubMed] [Google Scholar]
  • 10. Friberg L, Skeppholm M, Terént A. Benefit of anticoagulation unlikely in patients with atrial fibrillation and a CHA2DS2‐VASc score of 1. J Am Coll Cardiol. 2015;65:225–232. [DOI] [PubMed] [Google Scholar]
  • 11. Al‐Khatib SM, Thomas L, Wallentin L, et al. Outcomes of apixaban vs. warfarin by type and duration of atrial fibrillation: results from the ARISTOTLE trial. Eur Heart J. 2013;34:2464–2471. [DOI] [PubMed] [Google Scholar]
  • 12. Senoo K, Lane D, Lip GY. Stroke and bleeding risk in atrial fibrillation. Korean Circ J. 2014;44:281–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Apostolakis S, Lane DA, Guo Y, et al. Performance of the HEMORR2HAGES, ATRIA, and HAS‐BLED bleeding risk‐prediction scores in patients with atrial fibrillation undergoing anticoagulation: the AMADEUS (evaluating the use of SR34006 compared to warfarin or acenocoumarol in patients with atrial fibrillation) study. J Am Coll Cardiol. 2012;60:861–867. [DOI] [PubMed] [Google Scholar]
  • 14. Roldán V, Fernández H, Manzano‐Fernandez S, et al. Predictive value of the HAS‐BLED and ATRIA bleeding scores for the risk of serious bleeding in a real‐world population with atrial fibrillation receiving anticoagulant therapy. Chest. 2013;143:179–184. [DOI] [PubMed] [Google Scholar]
  • 15. Chen‐Scarabelli C, Scarabelli TM, Ellenbogen KA, et al. Device‐detected atrial fibrillation: what to do with asymptomatic patients? J Am Coll Cardiol. 2015;65:281–294. [DOI] [PubMed] [Google Scholar]
  • 16. Viles‐Gonzalez JF, Halperin JL. Everything counts in large amounts: device‐detected atrial high‐rate arrhythmias. Circ Arrhythm Electrophysiol. 2009;2:471–473. [DOI] [PubMed] [Google Scholar]
  • 17. Daoud EG, Glotzer TV, Wyse DG, et al. Temporal relationship of atrial tachyarrhythmias, cerebrovascular events, and systemic emboli based on stored device data: a subgroup analysis of TRENDS. Heart Rhythm. 2011;8:1416–1423. [DOI] [PubMed] [Google Scholar]
  • 18. Glotzer TV, Hellkamp AS, Zimmerman J, et al; MOST Investigators. Atrial high rate episodes detected by pacemaker diagnostics predict death and stroke: report of the Atrial Diagnostics Ancillary Study of the Mode Selection Trial (MOST). Circulation. 2003;107:1614–1619. [DOI] [PubMed] [Google Scholar]
  • 19. Healey JS, Connolly SJ, Gold MR, et al; ASSERT Investigators. Subclinical atrial fibrillation and the risk of stroke. N Engl J Med. 2012;366:120–129. [DOI] [PubMed] [Google Scholar]
  • 20. Brambatti M, Connolly SJ, Gold MR, et al; ASSERT Investigators. Temporal relationship between subclinical atrial fibrillation and embolic events. Circulation. 2014;129:2094–2099. [DOI] [PubMed] [Google Scholar]
  • 21. Ip J, Waldo AL, Lip GY, et al. Multicenter randomized study of anticoagulation guided by remote rhythm monitoring in patients with implantable cardioverter‐defibrillator and CRT‐D devices: rationale, design, and clinical characteristics of the initially enrolled cohort: the IMPACT study. Am Heart J. 2009;158:364.e1–370.e1. [DOI] [PubMed] [Google Scholar]
  • 22. Schaefer JK, McBane RD, Wysokinski WE. How to choose appropriate direct oral anticoagulant for patient with nonvalvular atrial fibrillation. Ann Hematol. 2016;95:437–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Stangier DJ. Clinical pharmacokinetics and pharmacodynamics of the oral direct thrombin inhibitor dabigatran etexilate. Clin Pharmacokinet. 2012;47:285–295. [DOI] [PubMed] [Google Scholar]
  • 24. Mueck W, Stampfuss J, Kubitza D, et al. Clinical pharmacokinetic and pharmacodynamic profile of rivaroxaban. Clin Pharmacokinet. 2014;53:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Wang L, Zhang D, Raghavan N, et al. In vitro assessment of metabolic drug‐drug interaction potential of apixaban through cytochrome P450 phenotyping, inhibition, and induction studies. Drug Metab Dispos. 2010;38:448–458. [DOI] [PubMed] [Google Scholar]
  • 26. Bounameaux H, Camm AJ. Edoxaban: an update on the new oral direct factor Xa inhibitor [published correction appears in Drugs. 2014;74:1455]. Drugs. 2014;74:1209–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Connolly SJ, Eikelboom J, Dorian P, et al. Betrixaban compared with warfarin in patients with atrial fibrillation: results of a phase 2, randomized, dose‐ranging study (EXPLORE‐Xa). Eur Heart J. 2013;34:1498–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE‐LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation [published correction appears in N Engl J Med. 2010;363:1877]. N Engl J Med. 2009;361:1139–1151. [DOI] [PubMed] [Google Scholar]
  • 29. Patel MR, Mahaffey KW, Garg J, et al; ROCKET‐AF Investigators. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883–891. [DOI] [PubMed] [Google Scholar]
  • 30. 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;365:981–992. [DOI] [PubMed] [Google Scholar]
  • 31. Ng KH, Shestakovska O, Connolly SJ, et al. Efficacy and safety of apixaban compared with aspirin in the elderly: a subgroup analysis from the AVERROES trial. Age Ageing. 2016;45:77–83. [DOI] [PubMed] [Google Scholar]
  • 32. Giugliano RP, Ruff CT, Braunwald E, et al; ENGAGE AF‐TIMI 48 Investigators. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093–2104. [DOI] [PubMed] [Google Scholar]
  • 33. Heidbuchel H, Verhamme P, Alings M, et al. Updated European Heart Rhythm Association Practical Guide on the use of non–vitamin K antagonist anticoagulants in patients with non‐valvular atrial fibrillation. Europace. 2015;17:1467–1507. [DOI] [PubMed] [Google Scholar]
  • 34. Ansell JE. Reversing the effect of oral anticoagulant drugs: established and newer options. Am J Cardiovasc Drugs. 2016;16:163–170. [DOI] [PubMed] [Google Scholar]
  • 35. Eerenberg ES, Kamphuisen PW, Sijpkens MK, et al. Reversal of rivaroxaban and dabigatran by prothrombin complex concentrate: a randomized, placebo‐controlled, crossover study in healthy subjects. Circulation. 2011;124:1573–1579. [DOI] [PubMed] [Google Scholar]
  • 36. Iu R, Hodaj E, Paris A, et al. Effect of non‐specific reversal agents on anticoagulant activity of dabigatran and rivaroxaban: a randomised crossover ex vivo study in healthy volunteers [published correction appears in Thromb Haemost. 2013;109:169]. Thromb Haemost. 2012;108:217–224. [DOI] [PubMed] [Google Scholar]
  • 37. Zahir H, Brown KS, Vandell AG, et al. Edoxaban effects on bleeding following punch biopsy and reversal by a 4‐factor prothrombin complex concentrate [published correction appears in Circulation. 2015;131:e10]. Circulation. 2015;131:82–90. [DOI] [PubMed] [Google Scholar]
  • 38. Schiele F, van Ryn J, Canada K, et al. A specific antidote for dabigatran: functional and structural characterization. Blood. 2013;121:3554–3562. [DOI] [PubMed] [Google Scholar]
  • 39. Lu G, DeGuzman FR, Hollenbach SJ, et al. A specific antidote for reversal of anticoagulation by direct and indirect inhibitors of coagulation factor Xa. Nat Med. 2013;19:446–451. [DOI] [PubMed] [Google Scholar]
  • 40. Reynolds MR. Cardioversion with oral anticoagulants: reconfirming a 50‐year‐old standard. J Am Coll Cardiol. 2014;63:1088–1089. [DOI] [PubMed] [Google Scholar]
  • 41. Cappato R, Marchlinski FE, Hohnloser SH, et al; VENTURE‐AF Investigators. Uninterrupted rivaroxaban vs. uninterrupted vitamin K antagonists for catheter ablation in non‐valvular atrial fibrillation. Eur Heart J. 2015;36:1805–1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Spyropoulos AC, Douketis JD. How I treat anticoagulated patients undergoing an elective procedure or surgery. Blood. 2012;120:2954–2962. [DOI] [PubMed] [Google Scholar]
  • 43. Lip GY, Windecker S, Huber K, et al. Management of antithrombotic therapy in atrial fibrillation patients presenting with acute coronary syndrome and/or undergoing percutaneous coronary or valve interventions: a joint consensus document of the European Society of Cardiology Working Group on Thrombosis, European Heart Rhythm Association (EHRA), European Association of Percutaneous Cardiovascular Interventions (EAPCI) and European Association of Acute Cardiac Care (ACCA) endorsed by the Heart Rhythm Society (HRS) and Asia‐Pacific Heart Rhythm Society (APHRS). Eur Heart J. 2014;35:3155–3179. [DOI] [PubMed] [Google Scholar]
  • 44. Diener HC, Aisenberg J, Ansell J, et al. Choosing a particular oral anticoagulant and dose for stroke prevention in individual patients with non‐valvular atrial fibrillation: part 1. Eur Heart J. 2016;pii:ehv643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Eikelboom JW, Connolly SJ, Brueckmann M, et al; RE‐ALIGN Investigators. Dabigatran versus warfarin in patients with mechanical heart valves. N Engl J Med. 2013;369:1206–1214. [DOI] [PubMed] [Google Scholar]
  • 46. Diener HC, Aisenberg J, Ansell J, et al. Choosing a particular oral anticoagulant and dose for stroke prevention in individual patients with non‐valvular atrial fibrillation: part 2. Eur Heart J. 2016;pii:ehw069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. January CT, Wann LS, Alpert JS, et al; ACC/AHA Task Force Members. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2014;64:2246–2280. [DOI] [PubMed] [Google Scholar]
  • 48. Deitelzweig S, Amin A, Jing Y, et al. Medical cost reductions associated with the usage of novel oral anticoagulants vs warfarin among atrial fibrillation patients, based on the RE‐LY, ROCKET‐AF, and ARISTOTLE trials. J Med Econ. 2012;15:776–785. [DOI] [PubMed] [Google Scholar]
  • 49. Harrington AR, Armstrong EP, Nolan PE Jr, et al. Cost‐effectiveness of apixaban, dabigatran, rivaroxaban, and warfarin for stroke prevention in atrial fibrillation. Stroke. 2013;44:1676–1681. [DOI] [PubMed] [Google Scholar]
  • 50. Desai NR, Krumme AA, Schneeweiss S, et al. Patterns of initiation of oral anticoagulants in patients with atrial fibrillation—quality and cost implications. Am J Med. 2014;127:1075.e1–1082.e1. [DOI] [PubMed] [Google Scholar]
  • 51. Kirchhof P, Ammentorp B, Darius H, et al. Management of atrial fibrillation in seven European countries after the publication of the 2010 ESC Guidelines on atrial fibrillation: primary results of the Prevention of Thromboembolic Events—European Registry in Atrial Fibrillation (PREFER in AF). Europace. 2014;16:6–14. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Cardiology are provided here courtesy of Wiley

RESOURCES