Skip to main content
Clinical Cardiology logoLink to Clinical Cardiology
. 2015 Jul 14;38(11):684–691. doi: 10.1002/clc.22434

Non–Vitamin K Antagonist Oral Anticoagulant Use in Patients With Atrial Fibrillation and Associated Intracranial Hemorrhage: A Focused Review

Boris Arbit 1, Jonathan C Hsu 1,
PMCID: PMC6490851  PMID: 26173428

ABSTRACT

Atrial fibrillation (AF) is the most common cardiac arrhythmia and predisposes patients to an increased risk of embolic stroke. After nearly 60 years, warfarin is no longer the only effective therapeutic option for patients with AF. Large randomized trials have consistently shown that non–vitamin K oral anticoagulants (NOACs) including dabigatran, rivaroxaban, apixaban, and edoxaban significantly reduce from the risk of intracranial hemorrhage (ICH) compared with warfarin. We provide a focused review regarding the NOACs and ICH in AF patients by summarizing findings of these large clinical trials, mechanisms of lower ICH, reversal strategies with specific agents, and monitoring strategies.

Background

Atrial fibrillation (AF) is the most common cardiac arrhythmia,1 afflicting approximately 3 million people in the United States alone,2 and its prevalence is expected to rise to 15.9 million by 2050.3 Atrial fibrillation increases the risk of ischemic stroke by as much as 5‐fold and accounts for approximately 15% of all strokes nationally.4 Oral anticoagulation is the standard of care for the prevention of thromboembolism in at‐risk patients with AF.5

Warfarin, a vitamin K antagonist (VKA), has long remained the most widely prescribed oral anticoagulant in the United States since being approved for use in 1954.6 Warfarin decreases the risk of stroke and systemic thromboembolism in patients with AF by as much as 68%.7 However, the clinical management of patients taking warfarin can be complex due to multiple drug‐drug interactions, dietary interactions, and routine monitoring required to attain a therapeutic window of effect. In recent years, several non–vitamin K oral anticoagulants (NOACs)—dabigatran, rivaroxaban, apixaban, and edoxaban—have been approved by the US Food and Drug Administration (FDA) for prevention of embolic stroke in at‐risk patients with nonvalvular AF. The NOACs8 work by selectively inhibiting factor IIa (dabigatran) or factor Xa (rivaroxaban, apixaban, and edoxaban). These agents do not require routine laboratory monitoring for therapeutic effect and have been studied against warfarin in randomized controlled trials with regard to safety and efficacy.9, 10, 11, 12 In these trials, it has been shown that the NOACs' efficacy is at least noninferior to warfarin,9, 10, 11, 12 with pooled analyses revealing overall superiority of the NOACs compared with warfarin in regard to reduction of thromboembolism.13

The benefit of oral anticoagulation in reducing thromboembolism risk in AF patients needs to be weighed against the increased risk of bleeding with all agents.14 Intracranial hemorrhage (ICH) is the most feared bleeding event in terms of location and severity, due to its significantly increased morbidity and mortality.14, 15 Use of anticoagulant therapy with warfarin increases the risk of developing ICH by 7‐ to 10‐fold from baseline, and anticoagulation‐associated ICH accounts for up to 19% of all ICH cases.16, 17 The anticoagulant effect of the NOACs is currently not reversible and monitoring of effect remains difficult. However, in general, the NOACs appear to be consistently less likely to cause ICH compared with warfarin.13, 18 The goal of this review is to provide an examination of the current literature in regard to the NOACs vs warfarin and the risk of ICH.

Clinical Trials of Non–Vitamin K Oral Anticoagulants vs Warfarin

The first of the NOACs to be approved by the FDA was dabigatran etexilate, which is the only FDA‐approved oral direct thrombin inhibitor. The active form, dabigatran, works by directly inhibiting both free and clot‐bound thrombin. The Randomized Evaluation of Long‐term Anticoagulation Therapy (RE‐LY) trial was a noninferiority study that compared the long‐term efficacy and safety of dabigatran vs warfarin in patients with AF.9 The trial randomly assigned 18 113 patients to one of 3 cohorts: adjustable‐dose warfarin to a goal international normalized ratio (INR) of 2–3, dabigatran 150 mg twice daily, and dabigatran 110 mg twice daily. Over a median follow‐up period of 2.0 years, dabigatran 110 mg showed noninferiority to adjusted‐dose warfarin regarding the outcome of ischemic stroke (relative risk [RR]: 1.11, 95% confidence interval [CI]: 0.89‐1.40, P = 0.35), whereas dabigatran 150 mg showed superiority compared with warfarin (RR: 0.76, 95% CI: 0.60‐0.98, P = 0.03). Both doses of dabigatran were associated with significantly lower rates of ICH: With the 110‐mg dose, the rate of ICH was 0.23% per year (RR: 0.31, 95% CI: 0.20‐0.47, P < 0.001 compared with warfarin), and with the 150‐mg dose, the rate of ICH was 0.30% per year (RR: 0.40, 95% CI: 0.27‐0.60, P < 0.001 compared with warfarin).

Rivaroxaban, apixaban, and edoxaban are 3 other NOACs that have been FDA approved, with a mechanism of action of inhibiting factor Xa. Factor Xa has a key role in the clotting cascade by selectively and competitively inhibiting free and prothrombinase/clot‐associated factor Xa through reversible interactions, thereby inhibiting thrombin formation and decreasing fibrin clot formation.19, 20, 21

The Rivaroxaban Once Daily Oral Direct Factor Xa Inhibitor Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Arial Fibrillation (ROCKET‐AF) trial was a phase 3, randomized, double‐blind, event‐driven noninferiority trial with 14 264 patients comparing rivaroxaban with warfarin in nonvalvular AF (≥2 documented episodes) and a history of stroke, transient ischemic attack, or non–central nervous system embolism or ≥2 independent risk factors for stroke.10 The median duration of treatment exposure was 1.6 years; the median follow‐up period was 1.9 years. The rate of stroke or systemic embolism in the per‐protocol population was 1.7% per year vs 2.2% per year in the warfarin group (hazard ratio [HR]: 0.79, 95% CI: 0.66‐0.96, P < 0.001). Patients assigned to rivaroxaban had a significantly lower rate of ICH compared with the warfarin group (0.5% per year vs 0.7% per year; P = 0.02).

Apixaban is an orally active selective inhibitor of factor Xa.22 The Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial was a randomized controlled trial of 18 201 patients randomized to either apixaban or warfarin.11 The median follow‐up period was 1.8 years, and the primary outcome of stroke or systemic embolism occurred 1.27% per year in the apixaban group compared with 1.60% per year in the warfarin group (HR: 0.79, 95% CI: 0.66‐0.95, P < 0.001). The rate of ICH was significantly lower for the apixaban group at 0.33% per year vs 0.80% per year in the warfarin group (HR: 0.42, 95% CI: 0.30‐0.58, P < 0.001).

Edoxaban is the latest oral, direct factor Xa inhibitor to be approved for reduction of stroke risk in patients with nonvalvular AF. Its FDA approval was based on the Effective Anticoagulation With Factor Xa Next Generation in Atrial Fibrillation–Thrombolysis in Myocardial Infarction 48 (ENGAGE AF‐TIMI 48) trial,12 which was a 3‐group, randomized, double‐blind, double‐dummy trial comparing 2 dose regimens of edoxaban with warfarin. The trial included 21 105 patients with a median duration of treatment exposure of 2.5 years; the median follow‐up was 2.8 years. The rate of stroke in the intention‐to‐treat cohort was 1.49% per year (high‐dose edoxaban) vs 1.69% per year (warfarin group; HR: 0.88, 95% CI: 0.75‐1.03, P = 0.11). The rate of ICH was significantly lower for the edoxaban group at 0.39% per year (high‐dose edoxaban) vs 0.85% per year in the warfarin group (HR: 0.47, 95% CI: 0.34‐0.63, P < 0.001). A summary of characteristics and outcomes in each of the pivotal NOAC trials is provided in Table 1.

Table 1.

Characteristics and Outcomes of Randomized Controlled Trials Comparing NOACs vs Warfarin in Patients With AF

Characteristics and Outcomes Dabigatran 150 mg Dabigatran 110 mg Rivaroxaban 20 mg Apixaban 5 mg Edoxaban 60 mg Edoxaban 30 mg
RE‐LY9 ROCKET AF10 ARISTOTLE11 ENGAGE AF12
Randomized patients, n 6076 6015 7131 9120 7035 7034
Study design Open‐label warfarin vs double‐blind; D 150 vs D 110 Double‐blind vs warfarin Double‐blind vs warfarin Double‐blind vs warfarin
Primary efficacy outcome Stroke or SE Stroke or SE Stroke or SE Stroke or SE Stroke or SE Stroke or SE
Primary safety outcome Major bleeding Major bleeding Major bleeding and clinically relevant bleeding Major bleeding Major bleeding Major bleeding
No. of patients lost to follow‐up 20 32 69 1
Median duration of follow‐up, y 2 1.9 1.9 2.8
VKA, % 49.6 62.4 57.2 58.9
% TTR in the warfarin group 64 55 62 68
Outcome, HR (95% CI)
Stroke/SE 0.66 (0.53‐0.82) 0.91 (0.74‐1.11) 0.79 (0.65‐0.95) 0.79 (0.66‐0.95) 0.79 (0.63‐0.99) 1.07 (0.87‐1.31)
Hemorrhagic stroke 0.26 (0.14‐0.49) 0.31 (0.17‐0.56) 0.59 (0.37‐0.93) 0.51 (0.35‐0.75) 0.54 (0.38‐0.77) 0.33 (0.22‐0.50)
Intracranial bleeding 0.41 (0.28‐0.60) 0.30 (0.19‐0.45) 0.67 (0.47‐0.93) 0.42 (0.38‐0.58) 0.47 (0.34‐0.63) 0.30 (0.21‐0.43)
0.30%/y 0.23%/y 0.5%/y 0.33%/y 0.39%/y 0.26%/y
Ischemic stroke 0.76 (0.59‐0.97) 1.11 (0.89‐1.40) 0.94 (0.75‐1.17) 0.92 (0.74‐1.13) 1.00 (0.83‐1.19) 1.41 (1.19‐1.67)
Total mortality 0.88 (0.77‐1.00) 0.91 (0.80‐1.03) 0.85 (0.70‐1.02) 0.89 (0.80‐0.998) 0.92 (0.83‐1.01) 0.87 (0.79‐0.96)
Vascular mortality 0.85 (0.72‐0.99) 0.90 (0.77‐1.06) 0.89 (0.73‐1.10) 0.89 (0.76‐1.04) 0.86 (0.77‐0.97) 0.85 (0.76‐0.96)
Major bleeding 0.93 (0.81‐1.07) 0.80 (0.69‐0.93) 1.04 (0.90‐1.20) 0.57 (0.46‐0.70) 0.80 (0.71‐0.91) 0.47 (0.41‐0.55)
Fatal bleeding 0.70 (0.43‐1.14) 0.58 (0.35‐0.97) 0.50 (0.31‐0.79) 0.50 (0.33‐0.74) 0.55 (0.36‐0.84) 0.35 (0.21‐0.57)
GI bleeding 1.50 (1.19‐1.89) 1.10 (0.86‐1.41) 1.39 (1.19‐1.61) 0.89 14;(0.70‐1.15) 1.23 (1.02‐1.50) 0.67 (0.53‐0.83)

Abbreviations: AF, atrial fibrillation; ARISTOTLE, Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation; CI, confidence interval; ENGAGE AF, Effective Anticoagulation With Factor Xa Next Generation in Atrial Fibrillation; GI, gastrointestinal; HR, hazard ratio; NOAC, non–vitamin K antagonist oral anticoagulant; NR, not reported; RE‐LY, Randomized Evaluation of Long‐term Anticoagulation Therapy; ROCKET AF, Rivaroxaban Once Daily Oral Direct Factor Xa Inhibitor Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Arial Fibrillation; SE, systemic embolism; TTR, time in the therapeutic range; VKA, vitamin K antagonist.

In terms of real‐word data, the effectiveness and relative safety of dabigatran and rivaroxaban have been published in the Dresden NOAC Registry23 and the Swedish national quality registry for atrial fibrillation and anticoagulation (AuriculA).24 These smaller real‐world studies appear to corroborate the safety profile of the NOACs in clinical trials. Active registries include the Global Registry on Long‐term Oral Antithrombotic Treatment in Patients With Atrial Fibrillation (GLORIA‐AF)25 and the Outcomes Registry for Better Informed Treatment of Atrial Fibrillation II (ORBIT‐AF II).26 GLORIA‐AF is a large, international, observational registry involving patients with newly diagnosed nonvalvular AF at risk for stroke. The goal of the registry is to enroll 56 000 patients to study the effectiveness and safety of the NOACs and VKA in routine clinical care. ORBIT‐AF II is a multicenter, national registry of patients with AF that is enrolling up to 15 000 newly diagnosed patients with AF. Data regarding safety and efficacy from these registries are forthcoming but have not yet been fully published.

Recent research has focused on defining the risks of ICH with NOAC use in various populations. Elderly patients are more likely to experience fluctuations in renal function, experience polypharmacy, undergo hospitalization, and suffer falls. In a recent meta‐analysis of participants of clinical trials age ≥75 years, NOACs did not cause excess bleeding and were associated with equal or greater efficacy than conventional therapy.27 Altered metabolism of NOACs in the setting of impaired kidney function may subject patients with chronic kidney disease (CKD) to alterations in their efficacy and a higher risk of bleeding. A recent meta‐analysis examined the efficacy and safety of the NOACs vs warfarin in patients with CKD and found no significant difference in the primary efficacy outcomes of stroke with NOACs vs VKAs. The risk of major bleeding was similar between the groups as well.28 It is difficult to provide definitive recommendations on which NOAC is to be used in a particular patient. A recent review of considerations for using NOACs provides guidance based on expert opinion.29 Despite significant limitations of combining the results of trials of different agents, the NOACs have been shown to be associated with a significant reduction in rates of stroke or major bleeding when compared with warfarin in patients with previous stroke or transient ischemic attack.30, 31

Mechanisms of Lower Intracranial Hemorrhage Risk in the Non–Vitamin K Oral Anticoagulants vs Warfarin

The exact mechanism of brain‐protective effect of NOACs compared with warfarin remains to be fully elucidated, but potential mechanisms are summarized in Table 2. A critical difference may be the fact that warfarin inhibits vitamin K–dependent synthesis of coagulation factors II, VII, IX, and X and regulatory factor proteins C and S, whereas the NOACs target a single factor. Cell‐surface tissue factor (TF), found in high concentrations in the brain,32 forms TF‐VIIa complexes that initiate coagulation.33 TF‐VIIa complexes are suppressed by warfarin but not by the NOACs with their more highly selective targets.

Table 2.

Potential Mechanisms of Reduced ICH With NOACs

Target Mechanism
Tissue factor–factor VIIa complexes Found in high concentrations in the brain,31 these complexes initiate coagulation. They are suppressed by warfarin but not by target‐specific NOACs.32
P‐gp efflux transporters Dabigatran is a P‐gp substrate, and P‐gp efflux transporters at the BBB may actively pump molecules across the barrier.33
Animal studies show increased levels of rivaroxaban and apixaban in P‐gp double knockouts.34, 35

Abbreviations: BBB, blood–brain barrier; ICH, intracranial hemorrhage; NOAC, non–vitamin K antagonist oral anticoagulant; P‐gp, permeability glycoprotein.

Dabigatran is a zwitterion and thus a highly polar molecule, which may limit its transfer across the blood‐brain barrier (BBB).34 Another way in which the actions of dabigatran may be mitigated is via permeability glycoprotein (P‐gp) efflux transporters. These transporters exist in the BBB and provide protection against entry of potential noxious endogenous and exogenous compounds. Because dabigatran is a P‐gp substrate, P‐gp efflux transporters at the BBB may actively pump molecules across the barrier.34

Rivaroxaban (in mice)35 and apixaban (in rats)36 are found in much lower concentrations in the brain compared with plasma. These concentration differences were postulated to be due in part to P‐gp. In experimental P‐gp double knockout mice, brain‐to‐blood concentration ratios of rivaroxaban 15 and 60 minutes after oral administration were 1.6× and 3.2× higher, respectively, compared with wild‐type mice.35

Reversal Strategies

Treatment strategies for patients with anticoagulation‐associated ICH remains a fruitful area of research, given the severe morbidity and mortality associated with ICH. Therapies are divided into specific agents and coagulation factors. Specific antidotes to the NOACs are not commercially available but are under study. Strategies for reversal of the anticoagulant effect of NOACs are summarized in Table 3.

Table 3.

Strategies for Anticoagulation Reversal in Bleeding Associated With NOACs

Dabigatran Rivaroxaban, Apixaban, and Edoxaban
General measures Drug discontinuation, mechanical compression, transfusional support Drug discontinuation, mechanical compression, transfusional support
Activated charcoal Consider if last dose <2–3 h63 Consider if last dose <2 h64
Hemodialysis Removes 50%–60%65 Not effective66
Coagulation factors
PCC Mixed results in human trials; does not correct coagulation parameters67, 68 Improvement of laboratory parameters with rivaroxaban in human ex vivo trials68
rFVIIa Single case report in real‐life patient; improves clotting time69 rFVIIa was more effective than aPCC70
aPCC, FEIBA Rare case reports in humans; may be more potent than PCC; does not correct coagulation parameters71 More effective than PCC in reversing prolongations of PT, CT, and TG70
Specific inhibitors
Idarucizumab Humanized antibody fragment; noncompetitive binding to dabigatran72
Phase 1: immediate, complete and sustained reversal of dabigatran‐induced anticoagulation in healthy humans
Phase 3: ongoing
Ciraparantag (PER977) Small, synthetic, water‐soluble, cationic molecule; binds to heparins and oral FXa and IIa inhibitors through hydrogen bonding73
Complete reversal of anti‐Xa activity of rivaroxaban, apixaban, edoxaban, and dabigatran
Phase 3: rapid and sustained reversal of edoxaban
Andexanet alfa (PRT064445) Recombinant human FXa; binds competitively to direct FXa inhibitors.74 Phase 3: rapid reversal of apixaban. Ongoing trial with rivaroxaban and planned trial with edoxaban.
aDabi‐Fab Humanized antibody fragment that binds dabigatran stronger than its affinity for thrombin and rapidly reverses its anticoagulant effects in vitro and in vivo

Abbreviations: aPCC, activated prothrombin complex concentrate; CT, clotting time; FEIBA, factor eight inhibitor bypass activity; FXa, factor Xa; IIa, factor IIa; PCC, prothrombin complex concentrate; PT, prothrombin time; rFVIIa, recombinant activated factor VII; TG, thrombin generation.

Currently, there is a lack of consensus among board‐certified vascular neurologists regarding the treatment of a patient with NOAC‐related ICH. For example, in dabigatran‐associated ICH, 73% reported that they would attempt reversal of dabigatran with the following agents: prothrombin complex concentrate (61%), fresh frozen plasma (53%), factor VIIa (24%), hemodialysis (24%), and platelet transfusion (7%).37 Randomized controlled trials are required to compare the effect of different reversal agents on bleeding rate and volume, bleeding complications, and thrombotic complications.

Monitoring Anticoagulant Effect of the Non–Vitamin K Oral Anticoagulants

The standard of care for warfarin‐related ICH is to measure and correct the INR with the goal of reversing anticoagulation and limit hemorrhage expansion using vitamin K and/or blood products such as fresh frozen plasma. However, there have been no randomized controlled trials to determine if correction of INR is associated with a reduction in hematoma expansion or results in improved outcome and decreased mortality. The coagulation assays that are used to monitor heparin derivatives or VKAs may not accurately reflect the anticoagulant activity of NOACs.38 The standard laboratory coagulation tests of prothrombin time (PT), activated partial thromboplastin time (aPTT), and thrombin time (TT) cannot establish the precise anticoagulant status of a particular patient. Further, these tests are unsuitable to monitor the effects of NOACs, which affect specific targets within the coagulation cascade.

Dabigatran prolongs the aPTT in a concentration‐dependent manner in both ex vivo and in vitro experiments. However, the dose response is linear up to a concentration of 200 to 300 ng/mL and flattens out at higher drug levels.39 This test has high interindividual and reagent variability.40 In a study of patients taking dabigatran 150 mg twice daily, 18% of participants had a normal aPTT at trough.41 Prothrombin time is less sensitive to dabigatran than the aPTT. In an ex vivo study of patients treated with dabigatran, an INR ≥1.2 was only observed with dabigatran concentrations >400 ng/mL.42 Dabigatran prolongs TT, even at low concentrations.43 Thrombin time increases in a linear dose‐dependent relationship with dabigatran concentration but shows dramatic prolongation with increasing concentration.44 For this reason, a prolonged TT can be used to indicate the presence of dabigatran, but not its concentration. Excessive sensitivity of this test may be overcome by diluting the plasma sample.45 Ecarin‐based assays, ecarin clotting time, and ecarin chromogenic assay have shown a linear relationship with dabigatran concentrations, with R2 values of 0.94 to 0.99 in in vitro and ex vivo samples. All 3 factor Xa inhibitors also prolong PT and aPTT but at low sensitivity. Although PT is usually prolonged with clinically relevant concentrations of rivaroxaban, its utility is limited by interindividual variability, reagent‐dependent variability, and poor correlation between the degree of PT prolongation and the plasma concentration of rivaroxaban.46, 47 Edoxaban has also showed considerable reagent‐dependent variability in the magnitude of PT prolongation.48 Across several studies, PT was inadequately sensitive to apixaban, even above the expected trough concentration.49 A recent systematic review recommended chromogenic anti‐Xa assay to assess plasma rivaroxaban levels.49 This method can accurately measure a wide range of rivaroxaban concentrations in plasma, provided that a standard calibration curve is generated with rivaroxaban calibrators and controls.50 Accurate and reliable quantification of apixaban anticoagulant activity in plasma is possible using chromogenic anti‐Xa assays.51 Further, anti‐Xa activity correlates linearly with edoxaban over a broad range of concentrations52 and may be the best coagulation test for measuring factor Xa inhibitor levels. Despite the shortcomings of traditional tests, in the emergency setting, expert opinion favors checking aPTT for dabigatran and PT for rivaroxaban and apixaban when a readily available laboratory measurement is necessary to determine drug presence.53

The ability to assess exact anticoagulation status would be useful in specific settings, including a new‐onset vascular event like an embolic stroke, particularly for management options including use of thrombolysis. Tissue‐type plasminogen activator (tPA; Alteplase) is contraindicated in patients on treatment with warfarin with a PT/INR >1.7 because of an increased risk of bleeding.54 Warfarin use is not associated with ICH after tPA treatment in patients with acute ischemic stroke with a PT/INR <1.7.55 Although this approach cannot be used for NOACs, it suggests that tPA may be safe in patients with low levels of anticoagulant activity. Case reports of patients treated with a NOAC who underwent revascularization are sparse.56 A recent study showed that pretreatment with rivaroxaban and apixaban greatly reduced ICH compared with warfarin‐pretreated rats after thrombolytic therapy with tPA.57 Use of thrombolytic agents in patients already on a NOAC who present with thrombotic events is an area requiring further investigation.

Future Directions

A recent systematic review of 6 studies involving 57 491 patients concluded that NOACs are uniformly associated with an overall reduced risk of ICH when used for stroke prevention in AF.18 Any of the currently available NOACs can be considered first‐line therapy for patients at higher risk for ICH.18 Bleeding risk scores to quantify hemorrhage risk include HAS‐BLED (hypertension, abnormal renal/liver function, stroke, bleeding history or predisposition, labile INR, elderly, drugs/alcohol concomitantly), Computerized Registry of Patients With Venous Thromboembolism (RIETE), and Anticoagulation and Risk Factors in Atrial Fibrillation (ATRIA).58, 59, 60 Although these scores have been helpful in defining patients at elevated bleeding risk, none are specifically supported in the 2014 American Heart Association/American College of Cardiology/Heart Rhythm Society Guideline for the Management of Patients With Atrial Fibrillation.5 The European Heart Rhythm Association Practical Guide on the use of new oral anticoagulants in patients with nonvalvular AF uses HAS‐BLED to define patients at higher risk of bleeding.61 Scores are based on the presence of hypertension (systolic blood pressure >160 mm Hg), abnormal liver or renal function, history of stroke or bleeding, labile INRs, elderly age (>65 years), use of drugs that promote bleeding, or alcohol excess.62 A score ≥3 indicates potentially high risk for bleeding and may require closer observation. Because this formula was modeled using a population taking warfarin, and, further, labile INR is one of the variables used, further research is necessary as to whether similar models predict ICH bleeding risk in patients taking NOACs.

Conclusion

After nearly 60 years, warfarin is no longer the only effective therapeutic option for patients with AF. The NOACs are associated with an overall reduced risk of ICH when used for stroke prevention in AF across all of the NOACs tested against warfarin. Although questions persist regarding reversal and monitoring of anticoagulant effect, new insight into mechanisms of action continues to expand the role of these novel agents, particularly with their improved bleeding profile compared with warfarin in the intracranial space.

Dr. Hsu reports receiving honoraria from Medtronic and St. Jude Medical, and consulting fees from Celladon.

References

  • 1. Chugh SS, Roth GA, Gillum RF, et al. Global burden of atrial fibrillation in developed and developing nations. Glob Heart. 2014;9:113–119. [DOI] [PubMed] [Google Scholar]
  • 2. Naccarelli GV, Varker H, Lin J, et al. Increasing prevalence of atrial fibrillation and flutter in the United States. Am J Cardiol. 2009;104:1534–1539. [DOI] [PubMed] [Google Scholar]
  • 3. 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]
  • 4. Benjamin EJ, Wolf PA, D'Agostino RB, et al. Impact of atrial fibrillation on the risk of death: the Framingham Heart Study. Circulation. 1998;98:946–952. [DOI] [PubMed] [Google Scholar]
  • 5. January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society [published correction appears in J Am Coll Cardiol. 2014;64:2305–2307]. J Am Coll Cardiol. 2014;64:e1–e76. [DOI] [PubMed] [Google Scholar]
  • 6. Ansell J, Hirsh J, Poller L, et al. The pharmacology and management of the vitamin K antagonists: the Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy [published correction appears in Chest. 2005;127:415–416]. Chest. 2004;126(3 suppl):204S–233S. [DOI] [PubMed] [Google Scholar]
  • 7. Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation: analysis of pooled data from five randomized controlled trials [published correction appears in Arch Intern Med. 1994;154:2254]. Arch Intern Med. 1994;154:1449–1457. [PubMed] [Google Scholar]
  • 8. Husted S, de Caterina R, Andreotti F, et al; ESC Working Group on Thrombosis Task Force on Anticoagulants in Heart Disease. Non–vitamin K antagonist oral anticoagulants (NOACs): no longer new or novel. Thromb Haemost. 2014;111:781–782. [DOI] [PubMed] [Google Scholar]
  • 9. 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]
  • 10. 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]
  • 11. 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]
  • 12. 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]
  • 13. Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta‐analysis of randomised trials. Lancet. 2014;383:955–962. [DOI] [PubMed] [Google Scholar]
  • 14. van Walraven C, Hart RG, Singer DE, et al. Oral anticoagulants vs aspirin in nonvalvular atrial fibrillation: an individual patient meta‐analysis. JAMA. 2002;288:2441–2448. [DOI] [PubMed] [Google Scholar]
  • 15. Hart RG, Boop BS, Anderson DC. Oral anticoagulants and intracranial hemorrhage: facts and hypotheses. Stroke. 1995;26:1471–1477. [DOI] [PubMed] [Google Scholar]
  • 16. Bechtel BF, Nunez TC, Lyon JA, et al. Treatments for reversing warfarin anticoagulation in patients with acute intracranial hemorrhage: a structured literature review. Int J Emerg Med. 2011;4:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Flaherty ML, Adeoye O, Sekar P, et al. The challenge of designing a treatment trial for warfarin‐associated intracerebral hemorrhage. Stroke. 2009;40:1738–1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Chatterjee S, Sardar P, Biondi‐Zoccai G, et al. New oral anticoagulants and the risk of intracranial hemorrhage: traditional and Bayesian meta‐analysis and mixed treatment comparison of randomized trials of new oral anticoagulants in atrial fibrillation. JAMA Neurol. 2013;70:1486–1490. [DOI] [PubMed] [Google Scholar]
  • 19. Laux V, Perzborn E, Kubitza D, et al. Preclinical and clinical characteristics of rivaroxaban: a novel, oral, direct factor Xa inhibitor. Semin Thromb Hemost. 2007;33:515–523. [DOI] [PubMed] [Google Scholar]
  • 20. 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;3:514–521. [DOI] [PubMed] [Google Scholar]
  • 21. Pinto DJ, Orwat MJ, Quan ML, et al. 1‐[3‐Aminobenzisoxazol‐5′‐yl]‐3‐trifluoromethyl‐6‐[2′‐(3‐(R)‐hydroxy‐N‐pyrrolidinyl)methyl‐[1,1′]‐biphen‐4‐yl]‐1,4,5,6‐tetrahydropyrazolo‐[3,4‐c]‐pyridin‐7‐one (BMS‐740808): a highly potent, selective, efficacious, and orally bioavailable inhibitor of blood coagulation factor Xa. Bioorg Med Chem Lett. 2006;16:4141–4147. [DOI] [PubMed] [Google Scholar]
  • 22. Wong PC, Crain EJ, Xin B, et al. Apixaban, an oral, direct and highly selective factor Xa inhibitor: in vitro, antithrombotic and antihemostatic studies. J Thromb Haemost. 2008;6:820–829. [DOI] [PubMed] [Google Scholar]
  • 23. Beyer‐Westendorf J, Gelbricht V, Förster K, et al. Safety of switching from vitamin K antagonists to dabigatran or rivaroxaban in daily care—results from the Dresden NOAC registry. Br J Clin Pharmacol. 2014;78:908–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Labaf A, Carlwe M, Svensson PJ. Efficacy and safety of novel oral anticoagulants in clinical practice: a report from three centers in Sweden. Thromb J. 2014;12:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Huisman MV, Lip GY, Diener HC, et al. Design and rationale of Global Registry on Long‐Term Oral Antithrombotic Treatment in Patients with Atrial Fibrillation: a global registry program on long‐term oral antithrombotic treatment in patients with atrial fibrillation. Am Heart J. 2014;167:329–334. [DOI] [PubMed] [Google Scholar]
  • 26. Steinberg BA, Blanco RG, Ollis D, et al. Outcomes Registry for Better Informed Treatment of Atrial Fibrillation II: rationale and design of the ORBIT‐AF II registry. Am Heart J. 2014;168:160–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Sardar P, Chatterjee S, Chaudhari S, et al. New oral anticoagulants in elderly adults: evidence from a meta‐analysis of randomized trials. J Am Geriatr Soc. 2014;62:857–864. [DOI] [PubMed] [Google Scholar]
  • 28. Harel Z, Sholzberg M, Shah PS, et al. Comparisons between novel oral anticoagulants and vitamin K antagonists in patients with CKD. J Am Soc Nephrol. 2014;25:431–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Savelieva I, Camm AJ. Practical considerations for using novel oral anticoagulants in patients with atrial fibrillation. Clin Cardiol. 2014;37:32–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Ntaios G, Papavasileiou V, Diener HC, et al. Nonvitamin‐K‐antagonist oral anticoagulants in patients with atrial fibrillation and previous stroke or transient ischemic attack: a systematic review and meta‐analysis of randomized controlled trials. Stroke. 2012;43:3298–3304. [DOI] [PubMed] [Google Scholar]
  • 31. Dogliotti A, Paolasso E, Giugliano RP. Novel oral anticoagulants in atrial fibrillation: a meta‐analysis of large, randomized, controlled trials vs warfarin. Clin Cardiol. 2013;36:61–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Mackman N. The role of tissue factor and factor VIIa in hemostasis. Anesth Analg. 2009;108:1447–1452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Furie B, Furie BC. Molecular and cellular biology of blood coagulation. N Engl J Med. 1992;326:800–806. [DOI] [PubMed] [Google Scholar]
  • 34. Bovio JA, Smith SM, Gums JG. Dabigatran etexilate: a novel oral thrombin inhibitor for thromboembolic disease. Ann Pharmacother. 2011;45:603–614. [DOI] [PubMed] [Google Scholar]
  • 35. Gnoth MJ, Buetehorn U, Muenster U, et al. In vitro and in vivo P‐glycoprotein transport characteristics of rivaroxaban. J Pharmacol Exp Ther. 2011;338:372–380. [DOI] [PubMed] [Google Scholar]
  • 36. Wang L, He K, Maxwell B, et al. Tissue distribution and elimination of [14C]apixaban in rats. Drug Metab Dispos. 2011;39:256–264. [DOI] [PubMed] [Google Scholar]
  • 37. Rybinnik I, Mullen MT, Messe S, et al. Treatment of acute stroke in patients on dabigatran: a survey of US stroke specialists. J Stroke Cerebrovasc Dis. 2013;22:1312–1316. [DOI] [PubMed] [Google Scholar]
  • 38. Garcia D, Barrett YC, Ramacciotti E, et al. Laboratory assessment of the anticoagulant effects of the next generation of oral anticoagulants. J Thromb Haemost. 2013;11:245–252. [DOI] [PubMed] [Google Scholar]
  • 39. Hapgood G, Butler J, Malan E, et al. The effect of dabigatran on the activated partial thromboplastin time and thrombin time as determined by the Hemoclot thrombin inhibitor assay in patient plasma samples. Thromb Haemost. 2013;110:308–315. [DOI] [PubMed] [Google Scholar]
  • 40. Helin TA, Pakkanen A, Lassila R, et al. Laboratory assessment of novel oral anticoagulants: method suitability and variability between coagulation laboratories. Clin Chem. 2013;59:807–814. [DOI] [PubMed] [Google Scholar]
  • 41. Hawes EM, Deal AM, Funk‐Adcock D, et al. Performance of coagulation tests in patients on therapeutic doses of dabigatran: a cross‐sectional pharmacodynamic study based on peak and trough plasma levels. J Thromb Haemost. 2013;11:1493–1502. [DOI] [PubMed] [Google Scholar]
  • 42. Antovic JP, Skeppholm M, Eintrei J, et al. Evaluation of coagulation assays versus LC‐MS/MS for determinations of dabigatran concentrations in plasma. Eur J Clin Pharmacol. 2013;69:1875–1881. [DOI] [PubMed] [Google Scholar]
  • 43. 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;103:1116–1127. [DOI] [PubMed] [Google Scholar]
  • 44. Stangier J, Rathgen K, Stähle H, et al. 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] [PMC free article] [PubMed] [Google Scholar]
  • 45. Stangier J, Feuring M. Using the HEMOCLOT direct thrombin inhibitor assay to determine plasma concentrations of dabigatran. Blood Coagul Fibrinol. 2012;23:138–143. [DOI] [PubMed] [Google Scholar]
  • 46. Douxfils J, Tamigniau A, Chatelain B, et al. Comparison of calibrated chromogenic anti‐Xa assay and PT tests with LC‐MS/MS for the therapeutic monitoring of patients treated with rivaroxaban. Thromb Haemost. 2013;110:723–731. [DOI] [PubMed] [Google Scholar]
  • 47. van Veen JJ, Smith J, Kitchen S, et al. Normal prothrombin time in the presence of therapeutic levels of rivaroxaban. Br J Haematol. 2013;160:859–861. [DOI] [PubMed] [Google Scholar]
  • 48. Morishima Y, Kamisato C. Laboratory measurements of the oral direct factor Xa inhibitor edoxaban: comparison of prothrombin time, activated partial thromboplastin time, and thrombin generation assay. Am J Clin Pathol. 2015;143:241–247. [DOI] [PubMed] [Google Scholar]
  • 49. Cuker A, Siegal DM, Crowther MA, et al. Laboratory measurement of the anticoagulant activity of the non–vitamin K oral anticoagulants. J Am Coll Cardiol. 2014;64:1128–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Samama MM, Contant G, Spiro TE, et al. Evaluation of the anti‐factor Xa chromogenic assay for the measurement of rivaroxaban plasma concentrations using calibrators and controls. Thromb Haemost. 2012;107:379–387. [DOI] [PubMed] [Google Scholar]
  • 51. Gouin‐Thibault I, Flaujac C, Delavenne X, et al. Assessment of apixaban plasma levels by laboratory tests: suitability of three anti‐Xa assays. A multicentre French GEHT study. Thromb Haemost. 2014;111:240–248. [DOI] [PubMed] [Google Scholar]
  • 52. Cuker A, Husseinzadeh H. Laboratory measurement of the anticoagulant activity of edoxaban: a systematic review. J Thromb Thrombolysis. 2015;39:288–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Blann AD, Lip GY. Laboratory monitoring of the non–vitamin K oral anticoagulants. J Am Coll Cardiol. 2014;64:1140–1142. [DOI] [PubMed] [Google Scholar]
  • 54. Jauch EC, Saver JL, Adams HP Jr, et al. Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2013;44:870–947. [DOI] [PubMed] [Google Scholar]
  • 55. Xian Y, Liang L, Smith EE, et al. Risks of intracranial hemorrhage among patients with acute ischemic stroke receiving warfarin and treated with intravenous tissue plasminogen activator. JAMA. 2012;307:2600–2608. [DOI] [PubMed] [Google Scholar]
  • 56. Kimura S, Ogata T, Fukae J, et al. Revascularization for acute ischemic stroke is safe for rivaroxaban users. J Stroke Cerebrovasc Dis. 2014;23:e427–e431. [DOI] [PubMed] [Google Scholar]
  • 57. Kono S, Yamashita T, Deguchi K, et al. Rivaroxaban and apixaban reduce hemorrhagic transformation after thrombolysis by protection of neurovascular unit in rat. Stroke. 2014;45:2404–2410. [DOI] [PubMed] [Google Scholar]
  • 58. Camm AJ, Lip GY, De Caterina R, et al. 2012 focused update of the ESC Guidelines for the management of atrial fibrillation: an update of the 2010 ESC Guidelines for the management of atrial fibrillation. Developed with the special contribution of the European Heart Rhythm Association [published corrections appear in Eur Heart J. 2013;34:790 and Eur Heart J. 2013;34:2850–‐2851]. Eur Heart J. 2012;33:2719–2747. [DOI] [PubMed] [Google Scholar]
  • 59. Fang MC, Go AS, Chang Y, et al. A new risk scheme to predict warfarin‐associated hemorrhage: the ATRIA (Anticoagulation and Risk Factors in Atrial Fibrillation) Study. J Am Coll Cardiol. 2011;58:395–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Nieto JA, De Tuesta AD, Marchena PJ, et al. Clinical outcome of patients with venous thromboembolism and recent major bleeding: findings from a prospective registry (RIETE). J Thromb Haemost. 2005;3:703–709. [DOI] [PubMed] [Google Scholar]
  • 61. Heidbuchel H, Verhamme P, Alings M, et al. European Heart Rhythm Association Practical Guide on the use of new oral anticoagulants in patients with non‐valvular atrial fibrillation. Europace. 2013;15:625–651. [DOI] [PubMed] [Google Scholar]
  • 62. Pisters R, Lane DA, Nieuwlaat R, et al. 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;138:1093–1100. [DOI] [PubMed] [Google Scholar]
  • 63. Suryanarayan D, Schulman S. Potential antidotes for reversal of old and new oral anticoagulants. Thromb Res. 2014;133(suppl 2):S158–S166. [DOI] [PubMed] [Google Scholar]
  • 64. Wang X, Mondal S, Wang J, et al. Effect of activated charcoal on apixaban pharmacokinetics in healthy subjects. Am J Cardiovasc Drugs. 2014;14:147–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Warkentin TE, Margetts P, Connolly SJ, et al. Recombinant factor VIIa (rFVIIa) and hemodialysis to manage massive dabigatran‐associated postcardiac surgery bleeding. Blood. 2012;119:2172–2174. [DOI] [PubMed] [Google Scholar]
  • 66. De Vriese AS, Caluwé R, Bailleul E, et al. Dose‐finding study of rivaroxaban in hemodialysis patients. Am J Kidney Dis. doi: 10.1053/j.ajkd.2015.01.022. [DOI] [PubMed] [Google Scholar]
  • 67. Marlu 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]
  • 68. 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]
  • 69. Aron JL, Gosselin R, Moll S, et al. Effects of recombinant factor VIIa on thrombin generation and thromboelastography in a patient with dabigatran‐associated intracranial hemorrhage. J Thromb Thrombolysis. 2014;37:76–79. [DOI] [PubMed] [Google Scholar]
  • 70. Perzborn E, Heitmeier S, Laux V, et al. Reversal of rivaroxaban‐induced anticoagulation with prothrombin complex concentrate, activated prothrombin complex concentrate and recombinant activated factor VII in vitro. Thromb Res. 2014;133:671–681. [DOI] [PubMed] [Google Scholar]
  • 71. Schulman S, Ritchie B, Goy JK, et al. Activated prothrombin complex concentrate for dabigatran‐associated bleeding. Br J Haematol. 2014;164:308–310. [DOI] [PubMed] [Google Scholar]
  • 72. 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]
  • 73. Ansell JE, Bakhru SH, Laulicht BE, et al. Use of PER977 to reverse the anticoagulant effect of edoxaban. N Engl J Med. 2014;371:2141–2142. [DOI] [PubMed] [Google Scholar]
  • 74. 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]

Articles from Clinical Cardiology are provided here courtesy of Wiley

RESOURCES