Canadian Cardiovascular Society
Guideline Toolbar

8. Stroke Prevention

8.1 Stroke risk assessment

Observations from the Framingham cohort and subsequent clinical trials revealed that NVAF is an independent risk factor for stroke (annual incidence of approximately 4.1%-4.5%) and combined stroke/systemic embolism (annual incidence of 50%).[70],[169] The risk was further refined by the delineation of various baseline characteristics that might affect the risk of the stroke.[169][173] The first widely adopted tool for stroke risk assessment was the Congestive Heart Failure, Hypertension, Age, Diabetes, Stroke/Transient Ischemic Attack (CHADS2) score, which assigns a single point for HF, hypertension, age 75 years or older, and diabetes, and 2 points for previous stroke/systemic embolism.[26],[172] Unfortunately, CHADS2 was unable to adequately differentiate very low risk individuals (ie, in whom OAC is associated with a greater risk than benefit) from those at low but still clinically important stroke risk. This led to the development of the expanded Congestive Heart Failure, Hypertension, Age (75 years), Diabetes, Stroke/Transient Ischemic Attack, Vascular Disease, Age (65-74 years), Sex (Female) (CHA2DS2-VASc) score. This score includes the additional risk factors of vascular disease (1 point), female sex (1 point), and age between 65 and 74 years (1 point), and also increases the risk weight to 2 points for age 75 years or older.[26],[171] The 2010 CCS guidelines recommended stroke prevention therapies[4] on the basis of the CHADS2 whereas the 2012 update[5] differentiated among patients at low CHADS2 risk on the basis of risk factors derived from the CHA2DS2-VASc.[174] The algorithm for antithrombotic prescription was further revised in 2014,[175] on the basis of data from a Danish national cohort study, which showed that the annual risk of “stroke” (defined as a thromboembolic event precipitating hospitalization or death) was 2.1% for patients aged 65-74 years and 4.4% for those 75 years of age or older.[174] Because age could be reliably determined in all patients, it was agreed that age 65 years or older should be the starting point for a revised algorithm.[175] Likewise, OAC was justified for younger patients with any CHADS2 risk factors (annual risk of stroke: 2.4% with HF, 1.6% with hypertension, 2.3% with diabetes, and 7.9% with previous stroke/systemic embolism). The CCS Guidelines Committee judged that antiplatelet therapy has no role in AF-related stroke prevention and, therefore, no antithrombotic therapy was recommended for patients younger than 65 years with none of the CHADS2 risk factors. However, in the presence of vascular disease daily antiplatelet therapy is indicated to prevent ischemic vascular events independent of the presence of AF.[176] As such, antiplatelet therapy is only indicated in the presence of established vascular disease in NVAF patients aged younger than 65 years with no CHADS2 risk factors (see section 8.3.2.1). The CCS algorithm (CHADS-65) is presented in (Fig. 8). Among several organizations that publish guidelines for antithrombotic therapies for patients with AF, there are variations in the definitions of the component risk factors, and in the selection and interpretation of data on the stroke risk associated with the individual risk factors according to their preferred risk schemes.[10],[26],[177] These differences in definitions are unlikely to affect the categorization of patient stroke risk, however, they might provide different point estimates of the annual risk of stroke. It has also become evident that the risk of stroke varies among cohorts reported from different countries.[178] For similar baseline characteristics, the Taiwanese[179] and Danish cohorts[174] appear to have particularly high stroke risk, whereas cohorts from Sweden[180] and the United States[181] appear to be lower risk. The variation might be attributable to the dissimilarities in the definitions of stroke, treatment of comorbidities, and the protocols for data collection.

Recommendation

16. We recommend that all patients with AF should undergo annual assessment of their risk of stroke/systemic embolism, irrespective of their clinical pattern of AF (Strong Recommendation; HighQuality Evidence).

17. We recommend that the “CCS Algorithm” (CHADS65) be used to guide the choice of appropriate antithrombotic therapy for the purpose of stroke/systemic embolism prevention in patients with NVAF (Strong Recommendation; High-Quality Evidence).

Practical Tip

NVAF is defined as AF in the absence of mechanical heart valves or moderate to severe mitral stenosis.

8.1.1 Atrial flutter

Atrial flutter (AFL) carries a significant risk of stroke/systemic embolism but, as opposed to AF, the relationship is somewhat less precisely established due to limited number of small and retrospective cohort studies, as well as significant heterogeneity of data.[182] Because many patients with AFL also experience episodes of AF it becomes difficult to know the exact risk of stroke/systemic embolism related to AFL alone.[183] Moreover, there are no randomized trials of the value of OAC specifically in the AFL population. However, there is direct and indirect evidence from mechanistic, observational, and prospective studies that AFL confers a significant thromboembolic risk. TEE evidence of atrial thrombi has been documented in patients with sustained AFL not receiving long-term anticoagulation (LA thrombus in 1.6% with at least moderate spontaneous LA echo contrast in 13%; median duration of 33 days).[184] OAC has been shown to reduce the incidence of stroke/systemic embolism after cardioversion, with an incidence similar to that of AF.[185],[186] In a metaregression analysis, the annual risk of stroke/systemic embolism was approximately 3% in patients with AFL.[187] Although the rate of stroke is higher than that in control participants (HR, approximately 1.4), the risk of stroke appears to be lower than for patients with AF (HR, approximately 0.70).[183],[188] For AFL patients, the risk of stroke is higher with increasing CHA2DS2-VASc scores[189][192] and also increased in patients who develop AF after their initial diagnosis of AFL.[192] As such, it is recommended that patients with AFL be stratified and treated in the same manner as patients with AF.[193]

8.2 Oral anticoagulation

The benefit of OAC must be weighed against the risk of hemorrhage. The relative importance of a stroke prevented, and a major bleed caused is a subjective judgement. There is considerable scope for physician-patient discussion (ie, shared decision-making) to ensure that patient values are concordant with the decision to prescribe OAC, particularly when the annual risk of stroke is < 2% per year.[194] The CCS rationale for recommending OAC for most patients with age 65 years or older or CHADS2 score 1 is on the basis of the effects of OAC on the absolute risk reduction of stroke compared with the increase in major hemorrhage. In patients aged 65 years or older and without other risk factors for stroke, use of VKAs decreased the annual risk of stroke from 2.1% to 0.7% while it increased the risk of major bleeding by approximately 0.5% per year to 1.0%.[51],[195] Although the risk of major bleeding increases with increasing CHADS2 scores, the rate of rise is not as steep as that for stroke; therefore, the benefit to risk ratio for OAC increases as stroke risk factors accumulate. Furthermore, 70% of strokes result in death or major disability, whereas most patients survive major hemorrhage without long-term effects.[196] Thus, these results favour use of OAC in patients with age 65 years or older or CHADS2 score 1. OACs with efficacy and safety evidence in AF include VKAs and DOACs.

Recommendation

18. We recommend that OAC be prescribed for most patients with AF and age 65 years or older or CHADS2 score 1 (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places relatively greater weight on the absolute reduction of stroke risk with OAC compared with antiplatelet agents in patients aged 65 years or older or with CHADS2 score 1 and less weight on the potentially increased risk of major hemorrhage with OACs compared with antiplatelet agents.

19. We suggest that no antithrombotic therapy be prescribed for stroke prevention for most patients with NVAF who are aged younger than 65 years with no CHADS2 risk factors (Weak Recommendation; Moderate-Quality Evidence).

Values and Preferences

For patients with NVAF who are aged younger than 65 years with no CHADS2 risk factors the current evidence does not support antiplatelet monotherapy for stroke prevention. For those patients, with concomitant coronary or peripheral arterial vascular disease, the antithrombotic treatment should be directed at the underlying arterial disease as outlined in the CCS/Canadian Association of Interventional Cardiology (CAIC) guidelines (see section 8.3.2.1).

20. We recommend that the longitudinal follow-up of patients receiving OAC include regular assessment of bleeding risk, potential drug-drug interactions, as well as adherence and persistence to pharmacotherapy (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

Consider the use of a standardized follow-up and monitoring tools to facilitate systematic review of efficacy and bleeding in patients receiving OAC, particularly for patients receiving DOACs who do not undergo regular laboratory monitoring.

8.2.1 Warfarin and vitamin K antagonists

Six studies (2900 patients) have compared VKAs with placebo in the setting of NVAF; 5 primary prevention trials (2 doubleblinded) and 1 secondary prevention trial. The mean achieved international normalized ratio (INR) ranged from 2.0 to 2.6 among patients who were assigned to VKAs in the primary prevention trials and was 2.9 in the secondary prevention trial. A meta-analysis of these 6 studies showed a RR reduction of 64% (95% CI, 49%-74%) in all stroke (ischemic or hemorrhagic)[51] with an absolute risk reduction of 2.7% per year for primary prevention trials and 8.4% per year for secondary prevention. The absolute risk increase in major extracranial hemorrhage was 0.3% per year with use of VKAs. However, there was an absolute risk reduction of 1.6% per year in overall mortality with use of VKAs. In a meta-analysis of 8 RCTs (3647 patients), the RR reduction with use of VKAs over aspirin was 39% (95% CI, 19%-53%) for all strokes, equivalent to an absolute risk reduction of approximately 0.7% per year for primary prevention and 7% per year for secondary prevention.[51] There was an excess of bleeding in the VKA-treated patients leading to an absolute risk increase of 0.2% per year in major extracranial and intracranial hemorrhage (ICH). However, the absolute risk reduction for all-cause mortality remained in favour of VKAs at 0.5% per year. In an update of this meta-analysis to include the largest study of elderly patients[197] the absolute risk reduction increased to 0.9%, with the risk of major extracranial hemorrhage being no different between use of VKA and aspirin.[51] A further meta-analysis showed that adjusted-dose VKA was associated with half the rate of stroke/systemic embolism events with no significant difference in the rate of major bleeding relative to adjusted-dose antiplatelet therapy.[198] The benefit of VKAs for stroke prevention in patients with NVAF is optimized at a target INR of 2-3, with the time spent in this therapeutic range (TTR) directly correlated with clinical outcomes. The relationship between TTR and clinical outcomes was shown in a post hoc analysis of the AF Clopidogrel Trial With Irbesartan for Prevention of Vascular Events (ACTIVE W) trial.[199] In ACTIVE W there was no difference in stroke reduction between VKA and clopidogrel with aspirin therapy (RR, 0.93; 95% CI, 0.70-1.24; P ¼ 0.61) at centres with a TTR below the median TTR (65%). However, VKAs had a marked benefit for patients at centres with a TTR above the median, reducing vascular events by more than twofold (RR, 2.14; 95% CI, 1.61- 2.85; P < 0.0001). A subsequent systematic review reinforced this concept, showing a significant correlation between TTR and adverse outcomes (major hemorrhage and thromboembolic rate) in an analysis of 33,976 AF patients included in 38 studies.[200] More recently, a large worldwide observational study of 9934 VKA-treated patients enrolled from 35 countries between 2010 and 2015 showed an adjusted 2.6-fold increased risk of stroke/ systemic embolism, a 1.5-fold increased risk of major bleeding, and a 2.4-fold increased risk of all-cause mortality with TTR < 65% vs 65%.[201] Further, the study highlighted the difficulty in managing VKAs because the mean TTR for the total study group was 55%, with more than half of the North American cohort having a TTR < 65%.[201] VKAs such as warfarin remain the preferred OAC in the presence of any mechanical prosthetic heart valve and in patients with moderate to severe mitral stenosis. With respect to the former, in the Randomized, Phase II Study to Evaluate the Safety and Pharmacokinetics of Oral Dabigatran Etexilate in Patients After Heart Valve Replacement (RE-ALIGN) study patients with mechanical heart valves were randomized to VKA or dabigatran (150 mg, 220 mg, or 300 mg twice daily [BID], on the basis of renal function).[202] The trial was terminated early because of an excess of thromboembolic and bleeding events with dabigatran. Although RE-ALIGN is the only trial to evaluate DOACs in the setting of mechanical valve prostheses, VKA remains the treatment of choice in this population. With respect to patients with moderate to severe mitral stenosis, there are no randomized trials of OAC for prevention of thromboembolic events. Retrospective studies have suggested a 4- to 15-fold decrease in the incidence of embolic events with OAC therapy.[203] Because patients with severe mitral valve disease were excluded from the pivotal randomized DOAC trials,[21][25] VKAs remain the standard of care in this patient population until further evidence emerges. Ongoing trials of DOACs in patients with rheumatic valvular disease (Investigation of Rheumatic AF Treatment Using Vitamin K Antagonists, Rivaroxaban or Aspirin Studies [INVICTUS] studies; NCT02832531 and NCT02832544) will provide guidance.

8.2.2 Non-vitamin K direct oral anticoagulants

Four DOACs are currently approved for use in Canada: apixaban, dabigatran, edoxaban, and rivaroxaban. These four agents were developed to overcome the major limitations associated with VKAs and have been evaluated in large RCTs involving more than 70,000 patients.[21][25] Individually, DOACs were shown to be at least as effective as VKAs in decreasing the risk of NVAF-associated stroke/systemic embolism, with similar or less major bleeding. A meta-analysis of these four RCTs showed that the use of the higher approved DOAC dose resulted in a statistically significant reduction in stroke/systemic embolism (RR, 0.81; 95% CI, 0.73-0.91; P < 0.0001), ICH (RR, 0.48; 95% CI, 0.39-0.59; P < 0.0001), and all-cause mortality (RR, 0.90; 95% CI, 0.85-0.95; P ¼ 0.0003) with less major bleeding (RR, 0.85; 95% CI, 0.73-1.00; P ¼ 0.06), compared with VKAs.[52] Use of the lower-dose DOAC regimens resulted in similar rates of stroke/systemic embolism (RR, 1.03; 95% CI, 0.84-1.27; P ¼ 0.74), with less major (RR, 0.65; 95% CI, 0.43-1.00; P ¼ 0.05) and intracranial bleeding (RR, 0.31; 95% CI, 0.24-0.41; P < 0.0001), and lower mortality (RR, 0.89; 95% CI, 0.83-0.96; P ¼ 0.003), but significantly more ischemic strokes (RR, 1.28; 95% CI, 1.02-1.60; P ¼ 0.045) compared with VKAs. On the basis of these observations the CCS AF Guidelines Committee continues to recommend DOACs over VKAs for patients with NVAF. There have been no published RCTs directly comparing the four DOACs. Differences in the designs of the RCTs, including the population studied (and by extension their baseline stroke risk), preclude definitive comparisons between the agents. Although so-called “real world” comparisons are plentiful in the published literature, it is not possible to make reliable therapeutic inferences from observational associations because these types of studies are subject to significant biases and limitations.[204] Nevertheless, there are relevant differences in the pharmacological characteristics of the DOACs, which might influence their selection in individual patients such as: (1) bioavailability, with dabigatran being the least bioavailable, thereby requiring a special formulation for optimal absorption, and rivaroxaban requiring coadministration with food to optimize absorption of the 15- and 20-mg doses; (2) renal clearance, with dabigatran being predominantly (80%) renally cleared; (3) drug-drug interaction potential, with all of the DOACs influenced by P-glycoprotein interactions but only rivaroxaban and apixaban also being influenced by cytochrome P-450 isoenzyme function, specifically 3A4; (4) the elimination half-life and dosing schedule; and, (5) the presence and availability of an antidote. Regardless of the specific DOAC agent selected, it is important to ensure that the prescribed dose is consistent with Health Canada labelling and the product monograph (Table 7). Specifically, it has been observed that overtreatment (or the prescription of a standard dose in patients with an indication for dose reduction) occurs in approximately 4% of patients and is associated with an increased risk of major bleeding, hospitalization, and death, without a significant additional reduction in stroke.[205][207] Likewise, undertreatment (or prescription of a reduced dose without an indication to do so) occurs in 12%-15% of patients and is associated with a higher risk of thromboembolic events, hospitalization, and death, without a significant reduction in major bleeding.[205][207]

Recommendation

21. We recommend most patients should receive a DOAC (apixaban, dabigatran, edoxaban, or rivaroxaban) in preference to warfarin when OAC therapy is indicated for patients with NVAF (Strong Recommendation; High-Quality Evidence).

Values and Preferences

This recommendation places a relatively high value on the results of several large RCTs showing that the DOACs are either noninferior or superior to warfarin in preventing AF-related stroke; that they have no more or less major bleeding compared with warfarin; that they are associated with less ICH compared with warfarin; and on the greater ease of use of DOACs compared with dose-adjusted warfarin.

Practical Tip

Baseline renal function and complete blood counts should be measured before initiation of anticoagulation and at a regular intervals thereafter (see section 8.3.1).

Practical Tip

The dose of DOAC prescribed should follow the doses used in the RCTs and Health Canadaapproved prescribing information (see Table 7). Receipt of a higher than recommended dose is associated with increased bleeding events and overall mortality. Receipt of a lower than recommended dose is associated with increased rates of stroke/systemic embolism.

Practical Tip

Consideration should be given to switching eligible patients from warfarin to a DOAC, particularly if they are unable to maintain a therapeutic INR.

22. We recommend that warfarin be used for patients with a mechanical prosthetic valve and those with AF and moderate to severe mitral stenosis (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places high value on the evidence from 1 RCT of the inferiority of dabigatran compared with warfarin for the prevention of thromboembolism in patients with a mechanical prosthetic valve.

Values and Preferences

This recommendation places a relatively high value on the long experience and clinical reports of the use of warfarin in patientswith rheumatic mitral stenosis.

8.3 Anticoagulation in special populations

8.3.1 Anticoagulation in patients with chronic kidney disease and end-stage renal disease

AF patients with chronic kidney disease (CKD) represent a particularly high-risk subgroup because stroke, mortality, and major bleeding all increase as renal function deteriorates.[208],[209] RCTs demonstrate that OAC, including DOACs, are safe and effective for AF patients with mild to moderate CKD (stage 1-3 CKD or eGFR > 30 mL/min).[210] There are few randomized data for patients with severe renal impairment (stage 4-5 CKD or eGFR < 25 to 30 mL/min) as these patients were excluded from the large phase III RCTs. Nonrandomized and very limited randomized data support the use of OAC in patients with stage 4 CKD (eGFR 15-30 mL/min),[211],[329] however, the benefit of OAC for AF patients with severe CKD (eGFR < 15 mL/min) or end-stage renal disease (ESRD) who require dialysis remains unclear. There are no prospective randomized trials that have evaluated OACs vs placebo for dialysis-dependent AF patients, and results of the observational studies are conflicting.[209],[212][215] The ongoing randomized Study of the Benefit/Risk Ratio of Oral Anticoagulation in Hemodialysis Patients with Atrial Fibrillation (AVKDIAL) is studying VKA (target INR 2-3) compared with no OAC in hemodialysis patients with AF (NCT02886962). Until these results are available there is insufficient evidence to support or deny routine OAC use in the AF population with ESRD who require dialysis. When OAC is prescribed for AF patients with stage 1-4 CKD, the CCS AF guidelines panel recommends that a DOAC is preferred to a VKA. For those with moderate CKD in the landmark phase III trials, DOACs significantly reduced the risk of stroke/systemic embolism (RR, 0.79; 95% CI, 0.66-0.94) and major bleeding (RR, 0.80; 95% CI, 0.70-0.91), compared with VKAs.[216] Moreover, in patients with mild-moderate CKD the use of DOACs was associated with a lessened rate of adverse renal outcomes, including renal function decline, doubling in serum creatinine level, or acute kidney injury.[217] Although apixaban and rivaroxaban are approved for patients with stage 4 CKD (CrCl of > 15 mL/min), the evidence supporting DOAC use in preference to VKAs is limited.[218] In those with more severe stage 5 CKD or ESRD requiring dialysis the evidence supporting DOACs is incomplete, with the available observational data being subject to confounding and selection bias and the randomized studies being underpowered.[219][222],[885] The randomized Renal Hemodialysis Patients Allocated Apixaban versus VKA in AF trial (RENAL-AF; NCT02942407) was prematurely terminated following the enrollment of 154 patients with hemodialysisdependent ESRD (targeted enrollment, 760 patients). At study conclusion, the rates of bleeding and stroke/systemic embolism were similar between those randomized to apixaban 5 mg BID or VKAs. Ongoing studies include the Compare Apixaban and Vitamin-K Antagonists in Patients With Atrial Fibrillation (AF) and End-Stage Kidney Disease (ESKD) (AXADIA; NCT02933697), in which phenprocoumon vs is being compared to apixaban 2.5 mg twice daily (targeted enrollment 222 patients); and the Strategies for the Management of Atrial Fibrillation in Patients Receiving Dialysis (SAFE HD; NCT03987711) trial, in which VKA and apixaban 5 mg BID, and no anticoagulation are being compared (targeted enrollment 150 patients). Patients with clinically significant CKD require a reduction in DOAC dose in accordance with the approved prescribing information to avoid adverse clinical outcomes (Table 7).7,[205][207],[223] Although undertreatment (or prescription of a reduced dose without an indication to do so) is associated with a higher risk of thromboembolic events, hospitalization, and death,[205]-[207] appropriate dose reduction in those with moderate CKD can achieve clinical outcomes comparable with that in patients with preserved renal function (CrCl > 50 mL/min) who are receiving the higher DOAC dose.[224],[225] When medication dose adjustment is performed, CockcroftGault CrCl should be used. The rationale has been extensively outlined in the 2014 AF guidelines companion.[26] In brief, the Cockcroft-Gault CrCl was the formula used to assess eligibility in the landmark DOAC trials, and it is recommended to guide medication dose adjustment in the product monographs and approved prescribing information.[21][23],[25] Although the Modified Diet in Renal Disease (MDRD) or the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) eGFR equations are commonly reported by laboratories, use of these formulae fail to identify a significant proportion of patients with contraindications to DOACs or those who require dose adjustment.[226]

Recommendation

23. We recommend that patients with AF who are receiving OAC should have their renal function assessed at baseline and at least annually to detect latent kidney disease, determine OAC eligibility, and to support drug dosing (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

Because DOAC eligibility and dosing are dependent on renal function, we recommend at least annual assessment of renal function with calculation of CrCl. In stable patients with an eGFR of 30-60 mL/min renal function should be monitored every 6 months, and every 3 months for patients with an eGFR of 15-30 mL/min. In patients with fluctuating renal function or acute dehydrating illness renal function should be assessed more frequently. Follow-up monitoring should also include assessment of bleeding risk, drug interactions, and adherence and persistence.

24. We recommend that CrCl, as estimated using the Cockcroft-Gault method, be used to support dosing decisions of anticoagulant medications (Strong Recommendation; High-Quality Evidence).

Practical Tip

Multiple formulae have been developed to provide an estimate of renal function. The most commonly applied formulae estimate CrCl or filtration of creatinine by the glomerulus (glomerular filtration rate or eGFR). Although the eGFR equations (MDRD formula or the CKD-EPI formula) provide more accurate estimates of renal function, drug manufacturers have used the CrCl (Cockcroft-Gault formula) when recommending medication dosage adjustments for patients with renal dysfunction.

25. We recommend that antithrombotic therapy in AF patients with CKD be provided according to their risk of stroke/systemic embolism and the severity of renal dysfunction with selection of agent according to Table 7.

A. Stage 3 CKD or better (eGFR > 30 mL/min): we recommend that such patients receive antithrombotic therapy as determined by the “CCS algorithm” (Strong Recommendation; High-Quality Evidence).

B. Stage 4 CKD (eGFR 15-30 mL/min): we suggest that such patients receive antithrombotic therapy as determined by the “CCS algorithm” (Weak Recommendation; Low-Quality Evidence).

C. Stage 5 CKD (eGFR &lt; 15mL/min or dialysisdependent): we suggest that such patients not routinely receive antithrombotic therapy for stroke prevention in AF (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

These recommendations place a relatively higher value on prevention of ischemic stroke than on bleeding complications associated with antithrombotic therapy.

Practical Tip

Because of the lack of prospective data showing benefit in patients with a CrCl < 15 mL/min, the decision to use antithrombotic therapy should be individualized on the basis of physician and patient preference and considering the relative risks of stroke and bleeding. Therapy with antithrombotic therapy might be appropriate for some patients with AF and CrCl < 15 mL/min (or dialysis-dependent) in whom the benefit of preventing stroke outweighs the increased risk of bleeding.

8.3.2 Coronary artery disease

Up to 20%-30% of AF patients also have concomitant coronary artery disease (CAD), with a significant proportion who require percutaneous coronary intervention (PCI).[227],[228] OAC is indicated for the prevention of AF-related stroke/systemic embolism, which also provides benefit in preventing ischemic coronary events. Antiplatelet therapy is indicated for the prevention of coronary events after acute coronary syndromes (ACS) and/or PCI; however, it is inferior to OAC for the prevention of stroke/systemic embolism in an AF population at increased risk of AF-related stroke.[229] As such, management requires a careful and balanced assessment of the individual risks of bleeding vs the anticipated effect on thrombotic outcomes. To clarify potentially confusing terminology in this area, single-agent antiplatelet therapy (SAPT) refers to the use of a single antiplatelet drug (eg, acetylsalicylic acid [ASA]), dual antiplatelet therapy (DAPT) refers to the concomitant use of 2 antiplatelet agents (eg, ASA with P2Y12 inhibitor), dual pathway therapy refers to the concomitant use of a SAPT with an OAC agent (eg, VKA with P2Y12 inhibitor), and triple antithrombotic therapy (TT), the combination of DAPT with an OAC (eg, VKA with ASA and P2Y12 inhibitor). The comprehensive recommendations regarding antithrombotic treatment in AF patients indicated for OAC with concomitant coronary/arterial vascular disease are summarized in Figure 9.

Recommendation

26. We recommend that AF patients with coronary or arterial vascular disease (peripheral vascular disease or aortic plaque) receive an antithrombotic therapy regimen on the basis of a balanced assessment of their risk of AF-related stroke, ischemic coronary event, and clinically relevant bleeding associated with the use of antithrombotic agents (Strong Recommendation; High-Quality Evidence).

Practical Tip

For patients who require combinations of antiplatelet and OAC agents for concomitant AF and coronary/arterial vascular disease, measures should be used to reduce the risk of bleeding, including careful consideration of modifiable bleeding risk factors with vigorous efforts to mitigate them; consideration of proton pump inhibitor (PPI) use; avoidance of prasugrel and ticagrelor in conjunction with OACs; the use of the lower target INR range (eg, 2.0-2.5) when a VKA is used as part of combination therapy; specific measures during PCI to reduce bleeding outcomes (radial access or ultrasound-guided femoral access, use of small diameter sheaths if appropriate, early sheath removal if feasible, and minimized use of acute periprocedural antithrombotic therapies); delaying non-urgent procedures until dual or triple therapy is no longer required; use of walking aids for those with gait or balance disorders; avoidance of concomittant use of nonsteroidal anti-inflammatory drugs (NSAIDs) or other drugs that might increase bleeding risk; and, strict BP control.

27. We recommend a DOAC in preference to a VKA when an OAC is indicated for AF patients with coronary or arterial vascular disease (Strong Recommendation; High-Quality Evidence).

Values and Preferences

This recommendation places a relatively high value on the results of several large RCTs that showed that the DOACs are either noninferior or superior to VKAs in preventing AF-related stroke, that they cause no more or less major bleeding compared with VKAs, that they are associated with less ICH compared with VKAs, that they are associated with greater ease of use compared with dose-adjusted VKAs, and that DOACs are not associated with an increase in ischemic coronary outcomes.

8.3.2.1 Stable vascular disease and AF in patients at low risk of stroke/systemic embolism

SAPT (eg, ASA 81 mg/d) is recommended for patients with AF who are at low risk of stroke/systemic embolism (age younger than 65 years and CHADS2 score of 0) if coronary or peripheral arterial vascular disease is present (CAD, peripheral vascular disease, or aortic plaque). The SAPT recommendation is on the basis of the efficacy of ASA therapy for the prevention of coronary events among patients with stable CAD (1.5% per year absolute risk reduction for secondary prevention).[51],[230] Although there is extensive evidence for the efficacy of OAC for prevention of ischemic coronary events in patients with stable CAD,[231] the CCS AF guidelines recommend SAPT in preference to OAC in those at low risk of stroke because of the favourable safety profile and ease of use associated with antiplatelet therapy. In a non-AF population, the Cardiovascular Outcomes for People Using Anticoagulation Strategies (COMPASS) trial showed that dual pathway therapy with ASA and “vascular dose” rivaroxaban (2.5 mg BID) was associated with significant reduction in cardiovascular mortality and ischemic stroke, albeit with a significantly increased risk of major bleeding.[232] As such, the combination of ASA and rivaroxaban 2.5 mg BID may be considered a reasonable alternative to ASA alone for NVAF patients at low risk of stroke (age younger than 65 years and CHADS2 score of 0) who also have coronary or arterial vascular disease.

Recommendation

28. We suggest no oral anticoagulation for stroke prevention for most patients with NVAF aged younger than 65 years with no CHADS2 risk factors and stable coronary or arterial vascular disease (Weak Recommendation; Moderate-Quality Evidence).

Practical Tip

The risk of stroke associated with AF is not sufficiently elevated to justify routine OAC therapy for those patients with stable coronary or arterial vascular disease aged younger than 65 years with AF and no CHADS2 risk factors. Treatment should be directed at the underlying coronary/peripheral arterial disease as outlined in the CCS/CAIC guidelines. Therapeutic options include ASA 81 mg daily alone; or ASA 81 mg daily in combination with either clopidogrel 75 mg daily, ticagrelor 60 mg BID, or rivaroxaban 2.5 mg BID.

8.3.2.2 Stable vascular disease and AF in patients at high risk of stroke/systemic embolism

OAC is indicated for stroke prevention in patients with AF who are aged 65 years or older or with a CHADS2 score 1. When such a patient also has stable CAD (defined by the absence of ACS or PCI in the preceding 12 months), OAC provides protection against ischemic coronary events in addition to stroke and systemic embolism.[233][239] The CCS AF Guidelines Committee does not recommend that antiplatelet agents be routinely prescribed in combination with OAC for AF patients at high risk of stroke with stable CAD. The Warfarin-Aspirin Reinfarction Study (WARIS)-II showed that the additional use of antiplatelet therapy with OAC did not confer a beneficial effect (eg, combined end point of death, MI, and stroke; 16.7% with OAC alone vs 15.0% with combination OAC and ASA; P ¼ 0.18), but did increase the risk of adverse bleeding outcomes (overall bleeding 2.82% per year with OAC alone vs 3.27% per year with combination OAC and ASA).[236],[237] More recently, the Atrial Fibrillation and Ischemic Events With Rivaroxaban in Patients With Stable Coronary Artery Disease (AFIRE) trial[239] randomized 2236 patients with AF with stable CAD to receive rivaroxaban monotherapy or combination therapy with rivaroxaban and SAPT. The trial was prematurely terminated because of increased all-cause mortality in the combination therapy group (1.9% per patient-year with rivaroxaban vs 3.4% per patient-year with combination therapy; HR, 0.55; 95% CI, 0.38-0.81; P < 0.05). Rivaroxaban monotherapy was noninferior to combination therapy for the primary efficacy end point (stroke/systemic embolism, MI, unstable angina requiring revascularization, or death)with event rates of 4.14% and 5.75% per patient-year, respectively (HR, 0.72; 95% CI, 0.55-0.95; P < 0.001 for noninferiority). Rivaroxaban monotherapy was superior to combination therapy for the primary safety end point of International Society on Thrombosis and Haemostasis major bleeding (1.62% vs 2.76% per patient-year; HR, 0.59; 95% CI, 0.39-0.89; P ¼ 0.01 for superiority).

Recommendation

29. We recommend OAC alone for patients with AF aged 65 years or older or with a CHADS2 score 1 and stable coronary or arterial vascular disease (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

The use of combination antithrombotic therapy (eg, OAC with a single antiplatelet agent) is not routinely justified for patients with AF and stable coronary or arterial vascular disease (defined as the absence of ACS or revascularization procedure in the preceding 12 months) because of the observed increased risk of bleeding and all-cause mortality observed with combination therapy, without a significant reduction in ischemic coronary and cerebrovascular thrombotic events.

Practical Tip

A combination of an OAC and single antiplatelet therapy may be considered only in highly selected patients with high-risk features for ischemic coronary outcomes, and who are also at low risk of bleeding.

8.3.2.3 PCI or ACS in patients with Af at low risk of stroke/systemic embolism

OAC is not recommended for stroke prevention for patients with AF who are at low risk of stroke/systemic embolism (age younger than 65 years and CHADS2 score of 0). DAPT is generally prescribed for patients after elective PCI for a period of 6 months and for 12 months after ACS (with or without PCI), as outlined in the 2018 CCS/CAIC antiplatelet guidelines for non-AF patients.[240] Some patients with low thrombotic or high bleeding risk may appropriately receive shorter durations of DAPT to decrease the risk of major bleeding. Conversely, longer durations of DAPT might be appropriate for those at higher thrombotic but lower bleeding risk, because premature DAPT discontinuation might increase the risk of stent thrombosis and MI.

8.3.2.4 PCI or ACS in patients with AF at high risk of stroke/systemic embolism

Combination OAC and antiplatelet therapy is required for patients with AF who are aged 65 years or older or with CHADS2 score 1, who are undergoing PCI or treatment of ACS. In these patients the optimal therapeutic regimen should be individualized on the basis of a balanced assessment of their risk of AF-related stroke/systemic embolism (“CCS algorithm”, Fig. 8), ischemic coronary events, and clinically relevant bleeding (Fig. 10). The risk of ischemic coronary events is modulated by the clinical presentation (eg, ACS being higher risk than elective PCI), clinical characteristics (higher risk with comorbid diabetes mellitus or CKD, current tobacco use, or previous stent thrombosis), as well as PCI-related factors (higher risk with multivessel disease, multiple stent implantation, total stent length > 60 mm, bifurcation lesion, chronic total occlusion intervention, and stent type).[240],[241] The risk of bleeding can be estimated from clinical risk scores such as Hypertension, Abnormal Renal/Liver Function, Stroke, Bleeding History or Predisposition, Labile INR, Elderly (> 65 Years), Drugs/Alcohol Concomitantly (HASBLED), Predicting Bleeding Complications in Patients Undergoing Stent Implantation and Subsequent Dual Anti Platelet Therapy (PRECISE-DAPT), and Cardiovascular Disease Research Using Linked Bespoke Studies and Electronic Health Records (CALIBER), with the former validated in a VKA-treated population, and the latter 2 in a population with CAD treated with PCI and DAPT.[240]

Recommendation

30. For patients with AF aged 65 years or older or with a CHADS2 score 1 undergoing PCI without ACS or high-risk features, we recommend dual pathway therapy (OAC with P2Y12) (Strong Recommendation; High-Quality Evidence) for at least 1 month and up to 12 months after PCI (Weak Recommendation; Low-Quality Evidence).

Practical Tip

The OAC component evaluated as part of dual pathway therapy regimens include: warfarin daily, apixaban 5 mg BID (reduced to 2.5 mg if they met 2 or more of the following dose-reduction criteria: age older than 80 years, weight &lt; 60 kg, or creatinine &gt; 133 mmol/L), dabigatran 110 mg or 150 mg orally (PO) BID, edoxaban 60 mg PO daily (30 mg in patients with CrCl 15-50 mL/min, body weight  60 kg, or concomitant use of specified potent Pglycoprotein inhibitors), rivaroxaban 15 mg PO daily (10 mg in patients with CrCl 30-50 mL/min). A DOAC is preferred over warfarin, however, if warfarin is used the lower end of the recommended INR target range is preferred. Clopidogrel is the preferred P2Y12 inhibitor. All patients should receive a loading dose of ASA 160 mg at the time of PCI (if previously ASA-naive).

31. For patients with AF aged older than 65 years or with a CHADS2 score 1 undergoing PCI for ACS or elective PCI with high-risk features, we recommend an initial regimen of triple therapy (OAC with P2Y12 and ASA 81 mg/d) (Strong Recommendation; LowQuality Evidence). After ASA discontinuation, which may occur as early as the day after PCI, we recommend that dual pathway therapy (OAC with P2Y12) be continued for up to 12 months after PCI (Strong Recommendation; High-Quality Evidence).

Practical Tip

For some patients younger than 65 years of age with CHADS2 score of 1 at the lower end of the stroke risk spectrum (eg, isolated hypertension), DAPT (eg, aspirin and ticagrelor) may be considered in preference to triple therapy (an OAC with P2Y12 and ASA).

Practical Tip

A PCI is considered high-risk for ischemic coronary outcomes on the basis of the clinical presentation (eg, ACS), patient characteristics (comorbid diabetes mellitus treated with oral hypoglycemic agents or insulin, CKD with eGFR < 60 mL/min, current tobacco use, previous ACS, or previous stent thrombosis), as well as PCI-related factors (multivessel PCI, multiple [> 3] stents implanted, total stent length > 60 mm, complex bifurcation lesion, chronic total occlusion intervention, and stent type (eg, bioabsorbable vascular scaffold).

Practical Tip

The OAC component evaluated as part of a triple therapy regimens include: warfarin daily, rivaroxaban 2.5 mg PO BID, or apixaban 5 mg BID (reduced to 2.5 mg if they meet 2 or more of the following dose-reduction criteria: age older than 80 years, weight < 60 kg, or creatinine > 133 mmol/L). A DOAC is preferred over warfarin, however, if warfarin is used, the lower end of the recommended INR target is preferred. Clopidogrel is the preferred P2Y12 inhibitor. All patients should receive a loading dose of ASA 160 mg at the time of PCI (if previously ASA-naive). Thereafter, ASA may be discontinued as early as the day after PCI or it can be continued up to 30 days. The timing of when to discontinue ASA will depend on individual patient’s ischemic and bleeding risk.

The population of patients with ACS not needing revascularization (PCI or coronary artery bypass graft [CABG] surgery) represents a heterogenous group. This population includes patients with thrombotic plaque rupture (type I MI) as well as those with supply-demand mismatch (type II MI). For patients with type II MI, it is unclear if there is an advantage to routine use of combined OAC and antiplatelet therapy. For patients with true ACS (type I MI) who are not revascularized, management should take into consideration the relative risk and benefits of combination therapy. Recently, the AUGUSTUS trial affirmed the benefits of apixaban-based dual pathway therapy in medically managed ACS patients.[242]

Recommendation

32. For patients with AF aged 65 years or older or with a CHADS2 score 1, we suggest that dual pathway therapy (OAC with P2Y12) be given without concomitant ASA for up to 12 months after medically managed type I ACS (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

For patients with AF and type I MI who do not undergo revascularization, the CCS AF Guidelines Committee places relatively greater emphasis on the reduction in ischemic coronary and cerebrovascular thrombotic events, rather than the increase in bleeding observed with combination therapy.

8.3.2.5 Key trials of dual pathway therapy vs triple therapy in patients with AF and ACS/PCI

The key trials that have compared dual pathway therapy with VKA-based TT include: the What Is the Optimal Antiplatelet and Anticoagulation Therapy in Patients With Oral Anticoagulation and Coronary Stenting (WOEST) study, the Prevention Of Bleeding In Patients With AF Undergoing PCI (PIONEER AF-PCI) study, the Randomized Evaluation of Dual Antithrombotic Therapy With Dabigatran versus Triple Therapy With Warfarin in Patients With Nonvalvular Atrial Fibrillation Undergoing Percutaneous Coronary Intervention (RE-DUAL PCI) trial, the Edoxaban Treatment Versus Vitamin K Antagonist in Patients With Atrial Fibrillation Undergoing Percutaneous Coronary Intervention (ENTRUST-AF PCI) study, and the AUGUSTUS trial.[243][247] Relevant details of these trials are presented in Table 8. Collectively these studies showed that the use of DOACs with a P2Y12 inhibitor was associated with a significant reduction in major bleeding (HR, 0.62; 95% CI, 0.47-0.81), without a statistically significant excess in the occurrence of MI (HR, 1.18; 95% CI, 0.93-1.52), definite stent thrombosis (HR, 1.55; 95% CI, 0.99-2.41), and stroke (HR, 0.89; 95% CI, 0.58-1.36).[246] When considering these studies, it is important to recognize that a large proportion of patients were undergoing elective PCI for stable CAD (38%-72%), possibly affecting the absolute risk of thromboembolic complications relative to patients with ACS. Second, measures to decrease bleeding risk were underutilized, suggesting that the rates of bleeding in the TT arms might have been greater than in contemporary practice. Third, most of the patients who participated in these trials received clopidogrel as their P2Y12 inhibitor, meaning no conclusions can be drawn about the effects of prasugrel or ticagrelor.

Recommendation

33. We recommend that clopidogrel 75 mg daily be the preferred P2Y12 inhibitor when dual pathway or triple therapy is used (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation recognizes that clopidogrel was the predominant P2Y12 inhibitor used in the landmark randomized controlled trials of combination therapy in AF patients after PCI/ACS, and clopidogrel is associated with the lowest risk of bleeding among the P2Y12 inhibitors in non-AF studies.

8.3.2.6 Duration of triple therapy

The benefit of TT (reduction of recurrent MI and stent thrombosis) must be balanced against the increased bleeding risk with this therapeutic regimen. In the AF population, it is likely that shorter durations of DAPT after ACS or PCI are reasonable when concomitant OAC will be used for the prevention of stroke/systemic embolism. This concept was shown in the Intracoronary Stenting and Antithrombotic Regimen: Triple Therapy in Patients on Oral Anticoagulation After Drug Eluting Stent Implantation (ISAR-TRIPLE) study, which showed no significant difference in the primary end point of “net clinical benefit” (combination of death, MI, stent thrombosis, stroke, or Thrombolysis in Myocardial Infarction [TIMI] major bleeding) between those who received 6 weeks vs 6 months of TT with a significant reduction in any bleeding in the group that received 6 weeks of TT.[248] A post hoc analysis of the AUGUSTUS trial, which compared the risk of bleeding and ischemic outcomes from randomization to 30 days and from 30 days to 6 months showed that aspirin use (ie, TT) was associated with more severe bleeding (absolute risk difference 0.97%) but fewer ischemic events (absolute risk difference -0.91%) in the first 30 days following randomization, when compared to placebo (ie, dual pathway therapy). From 30 days to 6 months, the risk of severe bleeding was higher with aspirin than placebo (absolute risk difference 1.25%) whereas the risk of ischemic events was similar (absolute risk difference -0.17%). As such, it appears that the benefit of triple therapy on ischemic coronary outcomes is limited to the first 30 days of therapy, with continuation of triple therapy beyond 30 days resulting in increased rates of bleeding without reduction in ischemic outcomes. Likewise, the PIONEER AFPCI and RE-DUAL PCI trials showed no relationship between major adverse cardiovascular events and TT duration.[244],[245] On balance, these findings suggest that limiting the course of TT to 1 month, and thereafter continuing dual pathway therapy (OAC and a P2Y12) might achieve the optimal balance of ischemic events prevented and bleeding caused.

8.3.3 Liver disease

The optimal management of patients with AF and advanced liver disease is complex. Patients with advanced liver disease are at increased risk of bleeding because of perturbations in coagulation factor synthesis,[249],[250] which was affirmed in a large Swedish registry-based study of 182,678 AF patients.[249] In this study the presence of liver disease was associated with an increased risk of major bleeding (adjusted HR, 1.8; 95% CI, 1.45-2.23) even in the absence of concomitant OAC use.[249] In addition, there is a concern that advanced liver disease results in a prothrombotic state, which might increase the risk of stroke/systemic embolism.[249],[251],[252] Finally, liver disease results in altered metabolism of VKAs and DOACs, which further complicates OAC choice.[253][257] Although OACs should be considered in most AF patients with liver disease, the optimal treatment is more challenging because such patients were excluded from the landmark anticoagulation trials. Specifically, patients with significant active liver disease and those with persistent elevation of liver enzymes or bilirubin were excluded from the large RCTs of a DOAC vs VKA.[21][23],[25] Although randomized data are limited, observational studies have shown that VKA use in patients with AF and advanced liver disease resulted in reduction of ischemic stroke risk with a higher risk of bleeding complications.[250],[252],[256][258] Moreover, a post hoc analysis of a prospective observational multicentre study of AF patients showed that the presence of advanced liver fibrosis increased the risk of major bleeding in those who received a VKA, but not in those who received a DOAC.[259] More recently, a nationwide retrospective Taiwanese cohort study reported outcomes in 2428 cirrhotic patients with NVAF who were taking apixaban (n ¼ 171), dabigatran (n ¼ 535), rivaroxaban (n ¼ 732), or a VKA (n ¼ 990).[258] In this study a similar risk of ischemic stroke/systemic embolism between DOAC- and VKA-treated patients was observed, however, the rates of major bleeding (2.9% vs 5.4% per year; P ¼ 0.0003) and gastrointestinal (GI) bleeding (1.9% vs 3.6% per year; P ¼ 0.0030) were significantly lower with a DOAC. For patients with advanced cirrhosis the DOAC group had a lower risk of ICH (HR, 0.17; 95% CI, 0.03-0.96; P ¼ 0.04). The Child-Pugh score can be used to guide OAC choice in patients with AF and advanced liver disease.[260] This score is commonly used to express the severity of chronic liver disease on the basis of clinical (ascites and encephalopathy) and laboratory (INR, bilirubin, albumin) parameters. When OAC is being prescribed for AF patients with Child-Pugh grade C cirrhosis it is recommended that a VKA be the preferred agent, because such patients were excluded from the landmark DOAC vs VKA trials.[21][23],[25] For those with Child-Pugh grade B cirrhosis it is recommended that rivaroxaban be avoided because it undergoes substantial hepatic metabolism,[253],[254] whereas apixaban, dabigatran, and edoxaban may be used with caution in such patients.[256][258],[261] Large studies appear to confirm a very low risk of hepatotoxicity with the approved DOACs.[262] The use of the appropriate antithrombotic agent in AF patients with advanced liver disease requires individualized therapy and might benefit from the guidance of an expert multidisciplinary team including a hepatologist and hematologist.

Recommendation

34. We recommend that OAC not be routinely prescribed for patients with AF and advanced liver disease (Child-Pugh grade C or liver disease associated with significant coagulopathy) (Strong Recommendation; Low-Quality Evidence).

Practical Tip

In select patients, OAC might be appropriate even in light of advanced liver disease. In these patients, OAC treatment decisions should be made in collaboration with specialized expertise (eg, hepatologists). VKAs can be considered with careful and frequent monitoring but only if the baseline INR is < 1.7. There is no evidence regarding the safety and efficacy of DOACs in patients with advanced liver disease.

8.3.4 Cancer

AF and malignancy are relatively common conditions, with the prevalence of both increasing with age. Patients with cancer have an elevated risk of AF, possibly because of the presence of comorbid conditions, a local/direct tumour effect, systemic inflammation, altered sympathovagal balance, or as a complication of surgical or targeted therapies (eg, tyrosine kinase inhibitors such as ibrutinib).[263],[264] The management of coexisting AF and cancer is challenging, because of the increased rates of thrombosis and thromboembolism observed in association with malignancy, as well as the increased risk of bleeding associated with coagulation defects, blood vessel erosion, tumour vascularity, radiation injury, and the antiplatelet effects of chemotherapies.[265],[266] These factors increase the complexity of OAC decision-making. The evidence for efficacy of anticoagulation among AF patients with cancer comes from several sources. These include 1 population-based retrospective cohort study, 2 retrospective cohort studies, 1 case-control study, and 3 post hoc analyses of the landmark DOAC vs VKA RCTs.[267][273] Among patients where were receiving a DOAC, the annual incidence of thromboembolic events varied from 0.0% to 4.9% with cancer and from 1.3% to 5.1% without; the annual incidence of a major bleed during DOAC treatment varied from 1.2% to 4.4% with cancer and 1.2% to 3.1% without cancer.[274] A large registry using prescription-based analysis for AF patients receiving VKAs or DOACs, with and without cancer reported equivalence for bleeding and thromboembolic risk, although the rates of both were lower in the DOAC population.[270] Within the landmark randomized trials of DOACs vs VKAs, there did not appear to be any significant difference in relative efficacy and safety of DOACs compared with VKAs in patients with and without a history of cancer.[267],[271],[273] Likewise, RCTs evaluating the cancer associated venous thromboembolism population have shown that DOACs were noninferior to low molecular-weight heparin (LMWH) for efficacy as well as safety.[275] The use of the appropriate antithrombotic agent in a patient with a concomitant malignancy requires individualized therapy under the guidance of an expert multidisciplinary team. When myelosuppressive chemotherapy or radiation therapy is undertaken or intensified, further steps to reduce bleeding risk, such as dose reductions or temporary cessation of OAC might be necessary.

Recommendation

35. We suggest that OAC treatment decisions be individualized for patients with AF and active malignancy, in consideration of the goals of care, the risk of stroke/systemic embolism, the risk of bleeding, and the concomitant antineoplastic therapy(ies) (Weak Recommendation; Low-Quality Evidence).

36. When an OAC is indicated in the presence of active malignancy, we suggest a DOAC in preference to a VKA (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places relatively greater value on the difficulties in ensuring stable INRs and the extensive drug-drug interactions between VKAs and active cancer therapeutic agents.

Practical Tip

Although there are no randomized data on the use of DOACs in patients with active cancer and NVAF, this recommendation places a relatively high value on the recognition that DOACs cause no more or less major bleeding compared with VKAs; that they are associated with less ICH compared with VKAs; and on the greater ease of use of DOACs compared with dose adjusted VKAs. The specific choice of OAC should be tailored according to potential drug-drug interactions.

8.3.5 Congenital heart disease

8.3.5.1 Stratification for anticoagulation

Although the link between atrial arrhythmias and thromboembolic complications is less well established in patients with CHD compared with the general AF population, the existing data appear consistent. Absence of sinus rhythm was found to be the factor associated with the highest prevalence of stroke in a cohort of > 23,000 adults with CHD.[276] Moreover, a strong association was observed between atrial arrhythmias and stroke in a Quebec administrative database study of > 38,000 patients with CHD.[277] As such, questions regarding long-term anticoagulation for atrial arrhythmias frequently arise in the care of adults with CHD.[278] Two retrospective studies have assessed anticoagulation practices, thromboembolic event rates, and bleeding complications in adults with CHD and atrial arrhythmias. A single centre retrospective study derived from the Congenital Corvitia (CONCOR) registry in the Netherlands followed 229 adults with CHD and intra-atrial reentrant tachycardia (IART), AF, or atrial tachycardia for a median of 6 years.[279] Overall, 67% of patients received a VKA and 7% antiplatelet therapy. The thromboembolic event rate in patients without a mechanical valve was 1.4% per year, with a major bleeding rate of 4.4% per year during treatment with a VKA. In univariable analyses, a CHA2DS2-VASc score 2 was associated with a higher thromboembolic event rate (HR, 3.7; P ¼ 0.021). Similarly, a HAS-BLED score 2 was associated with a higher major bleeding rate in univariable analyses (HR, 2.9; P ¼ 0.017). The Anticoagulation Therapy in Congenital Heart Disease (TACTIC) study enrolled 482 patients (age 32.0  18.0 years) from 12 North American centres.[280] Although the study was retrospective, it adhered to clinical trial data management standards including blinded adjudication of arrhythmias and outcomes and multiple layers of data quality control. Patients were classified as having simple (18.5%), moderate (34.4%), or severe (47.1%) forms of CHD on the basis of an established classification scheme.[281] OAC, predominantly VKAs, were administered to 54% of participants and antiplatelet agents to 38% of participants. Freedom from thromboembolic events was 89% at 10 years and 85% at 15 years. Rates did not differ significantly according to whether patients had AF vs IART. In multivariable analyses, complexity of CHD was the only factor independently associated with thromboembolism. Corresponding thromboembolic event rates in patients with simple, moderate, and severe forms of CHD were 0.00%, 0.93%, and 1.95% per year, respectively (P < 0.001). Notably, no thromboembolic event occurred in patients with simple forms of CHD despite 44% not receiving OAC. In patients with moderate and severe forms of CHD, the thromboembolic event rate exceeded the major bleeding rate associated with a VKA (0.77% per year). Although 93% of the population had a HAS-BLED score of 0 or 1, a higher score was associated with a greater risk of bleeding in multivariable analyses (HR, 3.15; P ¼ 0.047). These results were consistent with the concept of incorporating complexity of CHD in stratification decisions for anticoagulation, as proposed by the Pediatric and Congenital Electrophysiology Society (PACES)/Heart Rhythm Society (HRS) expert consensus statement on the recognition and management of arrhythmias in adults with CHD.[282]

Recommendation

37. We recommend that patients who have CHD and concomitant AF or IART receive an antithrombotic therapy regimen on the basis of a balanced assessment of their risk of AF-related stroke, the complexity of CHD, and the risk of clinically relevant bleeding associated with the use of antithrombotic agents (Strong Recommendation; High-Quality Evidence).

Values and Preferences

This recommendation places a high value on thromboembolic and bleeding risk scores that are well established in the non-CHD population as well as observational studies specific to the CHD population. It recognizes that complexity of CHD was identified as the most powerful predictor of thromboembolism, with a thromboembolic event rate that exceeds the major bleeding rate during oral anticoagulant treatment in those with moderate or severe CHD, regardless of their stroke risk score.

Practical Tip

Moderate or severe CHD is defined in the PACES/HRS expert consensus statement for the management of arrhythmias in adults with CHD.[282]

38. We suggest OAC for most patients with AF or IART and age 65 years or older, CHADS2 score 1, or CHD of moderate or severe complexity (Weak Recommendation; Moderate-Quality Evidence).

8.3.5.2 Choice of anticoagulant

Evidence on the use of DOACs in adults with CHD and atrial arrhythmias has recently emerged in the form of observational studies, as featured in a recent review.[283] In the first series of 75 adults with CHD receiving DOACs, 57 (76%) were anticoagulated for atrial arrhythmias.[284] No thrombotic or major hemorrhagic event occurred over a mean follow-up of 12 months. Nevertheless, minor bleeds were observed in 47%. An international registry reported a 1-year experience on the use of DOACs in 530 adults with CHD, 482 of whom had atrial arrhythmias.285 The DOACs consisted of rivaroxaban in 43%, apixaban in 39%, dabigatran in 12%, and edoxaban in 7%. Overall, complexity of CHD was simple in 15%, moderate in 45%, and severe in 40%. Nearly half the population (46%) was considered to have a significant valve lesion. Bioprosthetic valves were present in 11%, with no patient having a mechanical valve. At 1-year of follow-up, the thromboembolic event rate was 1.1%, major bleeding rate 1.3%, and minor bleeding rate 6.3%. In light of this reassuring data and considering that valve disease is common in adults with CHD, it appears reasonable to select a DOAC instead of a VKA in CHD patients with IART or AF and forms of valve disease that are similar to those with reassuring efficacy and safety data from large DOAC trials.[283] However, thromboembolic and bleeding rates in the large international registry were disproportionately high in the 14% of patients with Fontan palliation, who experienced a remarkable 50% rate of thrombotic and bleeding complications.[285] In a separate series 10 minor bleeding events occurred in 21 patients with Fontan surgery who received DOACs (12 for atrial arrhythmias).[286] One patient with a right-to-left shunt through a fenestration developed deep vein thrombosis during treatment with dabigatran. Another patient receiving apixaban had progression of thrombosis within the Fontan circuit. A separate report described worsening thrombus in a patient with an intracardiac lateral tunnel Fontan and IART during treatment with apixaban.[287] Thus, rigourous comparative safety and efficacy data of DOACs vs VKAs in certain subgroups of patients with severe forms of, including Fontan palliation, are required before DOACs can be endorsed for routine use in this setting.[282]

Recommendation

39. When OAC is indicated for atrial arrhythmias in adults with simple or moderate forms of CHD, we suggest a DOAC in preference to a VKA in the absence of recent cardiac surgery (< 3 months), a mechanical valve, and atrioventricular (AV) valve stenosis with enlarged and diseased atria (Weak Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a relatively high value on the growing observational literature that suggests that DOACs appear to have a favourable safety and short-term efficacy (1 year) profile when prescribed for atrial arrhythmias in adults with heterogeneous forms of CHD. It places a high value on established contraindications for DOACs in the context of valvular heart disease and recent cardiac surgery. It also recognizes that there are certain subgroups of patients with severe forms of CHD, such as those with cyanotic heart disease and single-ventricle physiology, in whom there is currently insufficient safety and efficacy data to endorse routine DOAC use. For example, Fontan patients prescribed DOACs were > 6-fold more likely to experience a thromboembolic event than other CHD patients receiving DOACs. There is a need for further research on the safety and efficacy of DOACs in subgroups of patients with severe forms of CHD.

8.3.6 Secondary AF

Although secondary AF has been associated with an increased risk for stroke and mortality irrespective of the precipitant, it is unclear whether the benefits of OAC apply equally to secondary AF patients compared with those with primary AF.[288],[289] As such, it remains uncertain whether such patients should be treated with long-term OAC on the basis of the standard stroke risk stratification schema. Until randomized trials assessing OAC in the secondary AF population are available an individualized approach to OAC is warranted. One such strategy would evaluate the likelihood that the secondary AF is “reversible” (ie, AF that occurs solely secondary to an acute illness, with little to no abnormal underlying substrate and therefore limited risk of recurrence) or “provoked” (ie, AF that is unmasked by the acute illness, occurring in patients with significant abnormal underlying substrate and therefore significant risk for recurrence). In both instances OAC might be warranted in selected patients in the short-term (ie, until resolution of the acute AF episode) however the long-term need for OAC should take into consideration a given patient’s underlying risk of stroke (eg, CHADS-65), the specific secondary precipitant for AF, and the likelihood of recurrence. In some cases, such as sepsis, the acute administration of intravenous (I.V.) anticoagulation increases the risk of bleeding but does not appear to reduce the risk of ischemic events.[289][291] Conversely, OAC is required for most patients with thyrotoxicosis-related AF because hyperthyroidism shifts the balance of the coagulation fibrinolytic system toward a hypercoagulable state, acting as an independent risk factor for thromboembolic events.[292][294] Of note, frequent monitoring of INR is advisable when a VKA is used in patients with hyperthyroid-associated AF, because the potency of VKAs will change as the degree of hyperthyroidism changes.[295]

Recommendation

40. We suggest that patients with secondary AF, which has resolved, not be routinely anticoagulated in the absence of recurrence (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places high value on the recognition that secondary AF is often a self-limited process. In the absence of AF recurrence, the current evidence base is insufficient to recommend long-term OAC. However, some patients and providers might elect to pursue long-term OAC on the basis of assessment of the patients underlying risk of stroke and values and preferences.

41. We suggest that most patients with secondary AF due to thyrotoxicosis be anticoagulated until a euthyroid state is restored (Weak Recommendation; Low-Quality Evidence).

8.3.7 Hypertrophic cardiomyopathy

The risk of stroke/systemic embolism in patients with AF and hypertrophic cardiomyopathy (HCM) is substantial and is significantly greater than in the non-HCM population with AF.[296][301] Stroke/systemic embolism in the HCM population appears to be unrelated to the type of AF (paroxysmal vs persistent) or AF burden.[297] Moreover, traditional risk prediction scores (eg, CHADS2 or CHA2DS2-VASc) have not been validated in the HCM population, and do not reliably predict thromboembolic outcomes in the HCM population.[296],[297],[302] All subjects with HCM who develop AF should started treatment with OAC because there is currently insufficient evidence to support the use of stroke risk prediction tools in the HCM population.[296],[297],[301],[302] VKAs have been traditionally used as the preferential OAC in subjects with HCM and AF.[303] Observational data subject to selection and ascertainment bias suggest that DOACs are at least as effective as VKAs in preventing thromboembolic events with similar or lower bleeding and safety events, with some studies suggesting improved survival and greater treatment satisfaction with DOACs.[304][306] As such, it is suggested that either DOACs or VKAs can be used to reduce thromboembolic events in subjects with HCM and AF.

Recommendation

42. We recommend that OAC be prescribed for most patients with AF and HCM (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a high value on the knowledge that the annual rate of stroke/systemic embolism in patients with HCM is substantial, and that the mechanism of stroke in HCM patients differs from the general population, which precludes the use of stroke risk prediction algorithms.

Practical Tip

Although there are no randomized data on comparisons of DOACs with VKAs, observational studies suggest these agents might be safe and effective in the HCM population.

8.3.8 Cardiac amyloidosis

Patients with cardiac amyloidosis are at a particularly high risk for thromboembolism due to progressive atrial myopathy, electromechanical dissociation, and reduced LAA emptying velocity. LA thrombus has been reported in up to one-third of patients with cardiac amyloidosis in sinus rhythm, as well as in patients with AF who are receiving therapeutic anticoagulation.[307][309] Among patients with cardiac amyloidosis who undergo cardioversion, 22.4% had intracardiac thrombus identified on TEE, which was significantly more common than in matched controls. Of patients with cardiac amyloidosis with intracardiac thrombus, 4 of 13 had received therapeutic OAC for a period of 3 weeks and 2 of 13 had an AF episode duration of < 48 hours. Patients with amyloid light-chain (AL) amyloidosis appear to be at particularly high risk of intracardiac thrombosis.[310] There is limited information to inform a specific OAC strategy. Data regarding the safety and efficacy of warfarin and DOACs is limited, however, their use appears to be reasonably safe in patients with cardiac amyloidosis the absence of conventional contraindications.[311]

8.3.9 Frail elderly patients

Improvements in life expectancy have resulted in an increasing number of AF patients being cared for into advanced age.[36],[312] Advanced age is a well established risk factor for ischemic stroke and hemorrhagic events in subjects with AF.[313][316] Unfortunately, elderly patients with AF might present with multiple health conditions that might affect QOL (eg, anemia or cognitive impairment/dementia), and/or increase the risk of adverse drug events (eg, impaired renal or hepatic function). In addition, polypharmacy is common in the elderly, which increases the risk of adverse drug reactions due to drug-drug interactions. Further, these patients are often excluded from RCTs, leading to a relative paucity of data to guide treatment decisions. Unfortunately, despite clear benefit,[21][23],[25],[270],[317] OAC remains underutilized in this population.[317],[318] A recent study of older patients with AF noted 35% of OAC-eligible patients did not receive anticoagulation.[317] The major factors responsible for underutilization of anticoagulation in subjects with AF include older age, female sex, abnormal liver enzyme levels, history of falls, excessive alcohol consumption, and concomitant prescription of antiplatelet agents.[318][327] Despite these concerns, there is evidence that the net clinical benefit favours prescription of OAC in this population.[328],[329] Although there are limited RCTs specifically in the elderly population with AF, secondary analyses of the landmark DOAC vs VKA trials did not show a difference in efficacy among subjects aged 75 years or older and those aged younger than 75 years.[330][333] Specifically, a meta-analysis of 4 RCTs (20,165 AF participants 75 years of age or older) showed a significantly lower rate of stroke/systemic embolism in DOAC-treated elderly patients (3.3% vs 4.7%; OR, 0.65; 95% CI, 0.48-0.87), with no significant difference in the rates of major or clinically relevant nonmajor (CRNM) bleeding (6.2% vs 6.6%; OR, 0.82; 95% CI, 0.58-1.16).[334] A pooled analysis from 2 European registries showed that DOAC use was associated with net clinical benefit in AF patients 75 years of age or older; with significant reduction in the composite end point of major bleeding and ischemic cardiovascular events (6.6% per year with DOACs vs 9.1% per year with VKAs; OR, 0.71; 95% CI, 0.51-0.99; P ¼ 0.042), which was predominantly due to a lower rate of major bleeding (OR, 0.58; 95% CI, 0.38-0.90; P ¼ 0.013) and a trend to reduction in ischemic events (OR, 0.71; 95% CI, 0.51 1.00; P ¼ 0.05) with DOACs.[328] In those at risk of falls, consideration should be made to: provide walking aids and proper footwear; promote cardiovascular, resistance, and balance activities; and, avoid medications that result in hypotension.

Recommendation

43. We recommend that OAC be prescribed for most frail elderly patients with AF (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places relatively greater value on the observation that elderly AF patients are at higher risk of stroke and, therefore, are more likely to benefit from OAC than younger patients, and places less value on the perceived increased risk of adverse treatment-related events (eg, the risk of bleeding if the patient falls). In general, the net clinical benefit is in favour of anticoagulant therapy in older patients because of the high risk of ischemic stroke.

Practical Tip

Treatment decisions regarding specific OAC agents should carefully consider the patient’s comorbidity profile, the risk for drug-drug interactions, and the risk of drug-disease interactions.

8.3.10 Increased BMI, obesity, and morbid obesity

Obesity is an established risk factor for the development of AF.[120] Clinicians face challenges regarding dosing of OAC in obese patients, because increasing BMI is independently associated with a higher risk of bleeding but lower risk of stroke/systemic embolism.[335],[336] Recent publications regarding outcomes in obese patients enrolled in the landmark DOAC trials are reassuring. A post hoc analysis of the Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial revealed that the treatment effect of apixaban was consistent with the overall trial results across all of the weight categories (P for interaction ¼ 0.64).[337] Specifically, in 982 patients > 120 kg, there was a significant reduction in major and CRNM bleeding with apixaban (HR, 0.58; 95% CI, 0.35-0.95) with comparable efficacy for stroke/systemic embolism (HR, 0.39; 95% CI, 0.12-1.22), relative to warfarin. The Effective Anticoagulation With Factor Xa Next Generation in Atrial Fibrillation-Thrombolysis in Myocardial Infarction 48(ENGAGE AF-TIMI 48) trial included 2099 patients (10%) with a BMI of 35-39, 1149 with a BMI > 40 (5.5%), and 148 with a BMI > 50 (0.7%).[335] There was no significant difference in trough edoxaban plasma concentrations and anti-Factor Xa activity across BMI categories, and there was no significant interaction between BMI and clinical outcomes (stroke/systemic embolism, all-cause mortality, major bleeding, major or CRNM bleeding, or the net clinical outcome). The Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared With Vitamin K Antagonist for Prevention of Stroke and Embolism Trial in Atrial Fibrillation (ROCKET-AF) trial enrolled 5206 patients with a BMI > 30.[336] There was no significant treatment interaction by BMI category (< 25, 25-35, or > 35) for either the primary end point of stroke/systemic embolism, or major or CRNM bleeding.[22] Finally, the Randomized Evaluation of Longterm Anticoagulation Therapy (RE-LY) trial included 3099 patients> 100 kg. There was no significant treatment interaction by weight category on the rate of stroke/systemic embolism or major bleeding, with dabigatran 110 mg BID and 150 mg BID being at least as effective as warfarin in obese patients.[21],[338] Meta-analyses of these phase III trials suggest that DOACs are more effective than warfarin for overweight patients (BMI 25-30: OR, 0.87; 95% CI, 0.76-0.99 for stroke/systemic embolism; OR, 0.83; 95% CI, 0.71-0.96 for major bleeding), and similarly effective in obese patients (BMI > 30: OR, 0.87; 95% CI, 0.76-1.00 for stroke/systemic embolism; OR, 0.91; 95% CI, 0.81-1.03 for major bleeding).[338] Although it is important to note that severely obese patients were not well represented, these meta-analyses provide reassurance that DOACs are at least as effective as warfarin in overweight and obese patients.

8.4 Anticoagulation in special circumstances

8.4.1 Cardioversion

8.4.1.1 Increased thromboembolism risk at the time of cardioversion

The incidence of stroke/systemic embolism is increased after cardioversion. A pooled analysis of 16 studies published between 1960 and 1969 showed a 30-day postcardioversion incidence of thromboembolic events of 1.76% (2665 cardioversions) in patients not receiving OAC with AF of > 48 hours in duration.[339] Similarly, a more contemporary pooled analysis of 10 studies describing outcomes after 3564 cardioversions of AF of > 48 hours duration published between 1986 and 2003 showed a 30-day incidence of thromboembolic events of 2.39% in patients not receiving OAC.[339] These rates contrast with the monthly 0.5% background thromboembolic risk of patients with AF in the absence of OAC when cardioversion has not been performed.[51] The increased thromboembolic risk associated with cardioversion might be related to 3 phenomena. The first is generation of thrombi during the persistent AF episode, with subsequent embolization after restoration of organized atrial contraction.[340] The second relates to a period of transient atrial mechanical dysfunction after the restoration of sinus rhythm.[341][343] This “atrial stunning” might be responsible for development of new intracardiac thrombi post cardioversion despite the restoration of sinus rhythm.[344],[345] Such atrial mechanical dysfunction has been reported with pharmacologic, electrical, and spontaneous cardioversion and is maximal in the period immediately after cardioversion.[344][347] The duration and severity of atrial stunning varies depending on the duration of the atrial arrhythmia, atrial size, and presence of underlying structural heart disease.[344][347] The third possibility is that cardioversion does not directly cause stroke/systemic embolism but rather patients who require cardioversion have transient as well as permanent characteristics associated with a higher stroke/systemic embolism risk.[348]

8.4.1.2 Anticoagulation for cardioversion of AF of > 48 hours

In patients with continuous AF for > 48 hours, observational studies reported a lower 30-day incidence of post cardioversion thromboembolic events in patients who receive OAC than in patients who do not receive OAC (0.45% vs 1.76% in studies from 1960-1969; 0.20% vs 2.39% in studies from 1986-2003).[339] Although none of these studies were randomized, the results are compelling in that patients who received OAC were those at highest risk of stroke/systemic embolism (eg, those with rheumatic mitral stenosis and/or previous thromboembolic events). Accordingly, there is widespread agreement that OAC is required for 3 weeks before and 4 weeks after planned cardioversion in patients who present with either “valvular AF,” or NVAF of > 48 hours.

8.4.1.3 TEE instead of 3 weeks of OAC before cardioversion

When cardioversion is desired without 3 weeks of therapeutic OAC another approach is to establish immediate anticoagulation, then perform a TEE to exclude LA thrombi before immediate cardioversion. This practice is supported by the Assessment of Cardioversion Using Transesophageal Echocardiography (ACUTE) trial, which randomized 1222 patients with nonacute AF to TEE-guided cardioversion or to the conventional 3 weeks of OAC before cardioversion.[349] Although there was no significant difference in the rate of periprocedural thromboembolic events (0.8% in the TEE group vs 0.5% in the conventional group; P ¼ 0.50), patients in the TEE group had fewer hemorrhagic events (2.9% vs 5.5%; P ¼ 0.03), a shorter time to cardioversion (3.0 5.6 vs 30.6 10.6 days; P < 0.001), and more frequently had sinus rhythm restored (71.1% vs 65.2%; P ¼ 0.03). Although other imaging modalities such as cardiac computed tomography (CT), cardiac magnetic resonance (MR) imaging (MRI), and intracardiac ultrasound might have sensitivities and specificities for the detection of LA clot that are comparable with those of TEE,[350] only the use of TEE for screening patients for candidacy for cardioversion has been subjected to an RCT. Accordingly, TEE remains the goldstandard procedure for this purpose; nevertheless, the other procedures listed may be used for this purpose when TEE is contraindicated or is not available.

8.4.1.4 Anticoagulation for cardioversion of AF of version of AF of ≤ 48 hours

Patients who present with AF of ≤ 48 hours have long been considered, on a theoretical basis, to have a low risk of stroke/systemic embolism after cardioversion. This practice has been supported by observational reports of outcomes after cardioversion in patients with AF ≤ 48 hours, suggesting a low risk of stroke/systemic embolism in the 30 days after cardioversion (0.27% after 4836 cardioversions in 4380 patients).[339] Although this risk is comparable with that of planned cardioversion in patients with AF of > 48 hours who receive OAC, these reports were limited by selection bias; describing low-risk patients who presented in a stable state early in the 48-hour window, with a significant proportion receiving OAC.[339] Moreover, despite the significant selection bias the rate of stroke/systemic embolism substantially exceeded the established threshold for OAC initiation (1.5% per year or 0.12% per 30 days).[351] More recent data specifically focused on patients who underwent cardioversion for AF of < 48 hours in the absence of OAC provide a more disquieting viewpoint.[352][359] In the Cleveland Clinic Study[359] consecutive patients having cardioversion of AF of ≤ 48 hours duration were examined, and a significantly higher 30-day post cardioversion rate of stroke/systemic embolism was reported in patients with no or subtherapeutic OAC (0.88%; 6 events after 683 cardioversions) compared with those with therapeutic OAC (0.22%; 2 events after 898 cardioversions; OR, 4.8; P ¼ 0.03). In the ANTIKogulation Registry[352] 366 consecutive patients with AF of ≤ 48 hours were examined, and a significantly higher rate of LA thrombosis (4% vs 0%; P ¼ 0.02) and a nonsignificantly higher rate of 30-day post cardioversion stroke/TIA (1.3% vs 0.5%; P ¼ 0.58) was observed in those who did not receive OAC. Although the latter was not statistically significant, it is noteworthy that each of the patients with a stroke/TIA had a CHADS2 score of 0. The FinCV studies were retrospective, observational studies that determined outcomes after cardioversion of AF in catchment area patients of 8 hospitals in Finland between 2003 and 2016.[353][358] The program enrolled consecutive patients in whom electrical or pharmacological cardioversion was attempted for AF of ≤ 48 hours (FinCV: 3143 patients; 7660 cardioversions), consecutive patients in whom elective electrical cardioversion was attempted for AF of > 48 hours (FinCV2: 1271 patients; 1894 cardioversions), and patients with AF who received DOACs in whom electrical or pharmacological cardioversion was attempted (FinCV3: 1028 patients; 1298 cardioversions). Together these 6 reports, on the short-term outcomes after 10,852 pharmacologic or electrical cardioversions in 5441 patients, demonstrated the following: (1) in the absence of OAC, time to cardioversion is a very strong predictor of 30- day risk of stroke/systemic embolism[354]; (2) in the absence of OAC the incidence of stroke/systemic embolism after cardioversion is significantly higher in patients with AF duration of 12-48 hours than in patients with < 12 hours of AF (1.1% [30/ 2767 patients] vs 0.33% [8/2440 patients], respectively)[354]; (3) for patients who receive OAC, AF duration is not associated with the incidence of stroke/systemic embolism (0.1% for < 24 hours; 0% for 24-48 hours; 0% for 48 hours to 30 days; and 0.2% for > 30 days)[358]; (4) in the absence of periprocedural OAC, independent predictors of stroke/systemic embolism postcardioversion are older age (OR, 1.05 per year; 95% CI, 1.02-1.08; P < 0.001), female sex (OR, 2.1; 95% CI, 1.1-4.0; P ¼ 0.03), HF (OR, 2.9; 95% CI, 1.1-7.2; P ¼ 0.03), and diabetes mellitus (OR, 2.3; 95% CI, 1.1-4.9; P ¼ 0.03)[353],[355]; and (5) therapeutic OAC significantly decreases the risk of postcardioversion stroke/systemic embolism.[353][357],[360] Specifically, for patients with AF of≤ 48 hours the 30 day incidence of stroke/systemic embolism was significantly lower in the presence of OAC (0.13% [3 events in 2298 encounters] vs 0.71% [38 events in 5362 encounters]; P ¼ 0.001).[355] When parsed according to CHA2DS2-VASc score, OAC use significantly reduced the 30 day rate of definite stroke/systemic embolism in those with a CHA2DS2-VASc score of 2 (0.2% [3/1; 708] vs 1.1% [28/2590]; P ¼ 0.001), but the benefit of OAC was not statistically significant in patients with a CHA2DS2-VASc score of 0-1 (0.0% [0/590] vs 0.40% [10/2772]; P ¼ 0.23). Of note, 10 of the 38 (26%) definite thromboembolic events that occurred after successful cardioversion in patients without OAC occurred in patients with a CHA2DS2-VASc score of 0-1. In 2 other recent reports the thromboembolic risk associated with cardioversion of recent-onset AF in the absence of OAC were indirectly assessed: a report from the Danish National Patient Registry[361] and a report from the Swedish National Patient Registry.[362] Each used administrative databases to evaluate the 30-day rate of stroke/systemic embolism after cardioversion of AF as a function of the presence or absence of precardioversion OAC. Although these databases did not include the duration of AF, the authors proposed that it would be reasonable to assume that cardioversion would only have been performed without previous OAC in patients with recentonset AF of≤ 48 hours. In the Danish analysis,[361] the 30-day incidence of stroke/systemic embolism after cardioversion was 1.06% (54 events in 5084 patients) without precardioversion OAC and 0.29% (32 events in 11,190 patients) with precardioversion OAC. Stroke/systemic embolism did occur in patients with CHADS2 or CHA2DS2-VASc scores of 0 or 1 but the rates were not stated. In the Swedish analysis,[362] the crude 30-day incidence of stroke/systemic embolism in the absence of precardioversion OAC was significantly higher than in the presence of precardioversion OAC (0.86% [104 events in 12,152 patients] vs 0.33% [35 events in 10,722 patients]). After adjustment for unequal distributions of the CHA2DS2-VASc factors, the OR for stroke/systemic embolism in the 30 days after cardioversion was 2.54 (95% CI, 1.70-3.79; P < 0.001) without precardioversion OAC relative to patients with precardioversion OAC. Propensity analysis of 9500 patients who did not receive OAC matched with 9500 patients who did receive OAC showed that patients who did not receive OAC were more likely to have stroke/systemic embolism (OR, 2.51; 95% CI, 1.69-3.75; P < 0.001) with similar rates of major bleeding (OR, 1.00; 95% CI, 0.48-2.10) relative to those who received OAC.[362] Taken together, the authors concluded that cardioversion in patients with AF of < 48 hours without OAC conferred a greater risk of stroke/systemic embolism than cardioversion for patients with AF of > 48 hours who received OAC. In summary, the risk of stroke/systemic embolism after cardioversion is elevated, even in patients who present within 48 hours of AF onset. This risk does not differ on the basis of the method of cardioversion (30 day risk of cardioversion of 0.26% with electrical vs 0.39% with pharmacological cardioversion).[363] Immediate cardioversion without 3 weeks of therapeutic OAC appears to be associated with a low risk of stroke/systemic embolism only in patients who presenting within 12 hours of AF onset and in patients who presenting 12-48 hours after AF onset who have a low risk of stroke (eg, patients aged younger than 65 years with a CHADS2 score of 0-1). Other patients deemed to be at higher risk of stroke should receive at least 3 weeks of therapeutic OAC before cardioversion (or undergo a TEE to exclude LA thrombus). After cardioversion, all patients should receive at least 4 weeks of OAC in the absence of a strong contraindication, and such therapy should be initiated as soon as possible and preferably before the cardioversion.[364] Thereafter, the need for OAC should be on the basis of the risk of stroke/systemic embolism as determined by the “CCS Algorithm” (CHADS-65). The approach to anticoagulation at time of cardioversion is presented in Figure 11.

Recommendation

44. We recommend that, in addition to appropriate rate control, most hemodynamically stable patients with AF for whom elective electrical or pharmacological cardioversion is planned should receive therapeutic anticoagulation for at least 3 weeks before cardioversion (Strong Recommendation; Moderate-Quality Evidence).

45. We suggest that TEE may be used to exclude cardiac thrombus, as an alternative to at least 3 weeks of therapeutic anticoagulation before cardioversion (Weak Recommendation; Moderate-Quality Evidence).

46. We suggest that pharmacological or electrical cardioversion of symptomatic AF without at least 3 weeks of previous therapeutic anticoagulation (or TEE) be reserved for patients with the following characteristics (Weak Recommendation; Low-Quality Evidence):

A. patients with NVAF who present with a clear onset of AF within 12 hours in the absence of recent stroke or TIA; or,

B. patients with NVAF and a CHADS2 score of 0 or 1 who present after 12 hours but within 48 hours of AF onset.

47. When a decision has been reached that a patient will be undergoing unplanned pharmacological or electrical cardioversion of AF, we suggest that therapeutic anticoagulation therapy be initiated immediately (preferably before cardioversion) with either: (1) a DOAC; or (2) heparin followed by adjusted-dose VKA (Weak Recommendation; Low-Quality Evidence).

48. We suggest that, in the absence of a strong contraindication, all patients who undergo cardioversion of AF receive at least 4 weeks of therapeutic anticoagulation (adjusted-dose VKA or a DOAC) after cardioversion (Weak Recommendation; Low-Quality Evidence). Thereafter, we recommend that the need for ongoing antithrombotic therapy should be on the basis of the risk of stroke as determined by the CCS Algorithm (CHADS-65) (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places relatively greater emphasis on the benefits of stroke prevention and less emphasis on risk of bleeding with a short course of anticoagulation therapy.

Practical Tip

When OAC is to be used for only a short period (< 2 months) the use of a DOAC is preferred to adjusted dose VKA.

8.4.1.5 DOAC or VKA for pericardioversion OAC

Three randomized trials have compared a DOAC with adjusted-dose VKAs in the setting of planned cardioversion of AF.[365][367] A meta-analyses of these 3 RCTs[365][367] showed that DOAC compared with adjusted-dose VKA was associated with significant reduction in stroke/systemic embolism (0.2% vs 0.6%; RR, 0.33; 95% CI, 0.12-0.91; P ¼ 0.03),[368] but no significant differences in any bleeding (1.8% vs 2.5%; RR, 0.85; 95% CI, 0.158-1.23; P ¼ 0.38),[369] major bleeding (0.4% vs 0.7%; RR, 0.61; 95% CI, 0.28-1.34; P ¼ 0.22),[368] or mortality (0.3% vs 0.4%; RR, 0.70; 95% CI, 0.23-2.10; P ¼ 0.52).369 In a meta-analysis that combined these RCT[365][367] data with that from patients having cardioversion in the 4 pivotal trials that compared DOAC with VKA therapy,[370][373] it was reported that no significant differences were observed in the singular outcomes of ischemic stroke, hemorrhagic stroke, mortality, or major bleeding, or in the composite outcome of stroke/systemic embolism with DOACs compared with adjusted-dose VKAs.[374],[375] It should be noted that these trials, individually and in their aggregate, were not sufficiently powered to exclude a clinically meaningful difference in either safety or efficacy.

8.4.2 Catheter ablation of AF

AF ablation is an invasive procedure associated with a risk of stroke/systemic embolism as well as a risk of bleeding. There has been a tremendous amount of research in the past few years regarding periablation OAC management. The use of uninterrupted OAC with a VKA was associated with a lower risk of bleeding and a decreased risk of thromboembolic complications compared with interrupted VKAs with LMWH bridging.[376] More recently, several randomized trials have shown superior safety and efficacy of uninterrupted DOAC (apixaban, dabigatran, edoxaban, and rivaroxaban) compared with uninterrupted VKA.[377][381] For these reasons, uninterrupted OAC has been considered the standard of care for AF ablation. Uninterrupted OAC typically means initiating OAC 3-4 weeks before the procedure, continuing OAC right up to the time of procedure, and reinitiating OAC 6-8 hours after the procedure. More recently, 2 trials have assessed the safety of “minimal” DOAC interruption.[382],[383] On the basis of these trials, it might be reasonable to hold a DOAC for 1-2 doses before ablation with early reinitiation 6-12 hours post ablation. Intraprocedural anticoagulation is critical to avoid the risk of clinical and subclinical cerebral thromboembolism.[384],[385] Anticoagulation during ablation is typically given as I.V. boluses and/or infusion of heparin with frequent measurement of the activated clotting time. More recent guidelines have suggested more aggressive activated clotting time targets of 350-400 seconds to prevent cerebral microembolism.[384],[386] Post ablation management of OAC can be divided into 2 phases: the initial 2 months after the procedure and the period thereafter. Catheter ablation of AF transiently damages the LA endothelium, creating an early prothrombotic state, which might lead to thromboembolism. Such a prothrombotic state can occur even in patients considered to have a low risk of stroke/systemic embolism according to traditional risk schema. For this reason, OAC is recommended for at least 2 months after AF ablation. Thereafter, the need for ongoing OAC should be on the basis of the CCS Algorithm (CHADS-65), rather than the apparent success of the ablation procedure. Although limited retrospective data series have suggested a low risk of stroke after successful AF ablation in a wide variety of patient risk profiles, other studies, including the large randomized Catheter Ablation vs Anti-Arrhythmic Drug Therapy for Atrial Fibrillation (CABANA) trial, have not shown a significant reduction of stroke post ablation.[387][394] Furthermore, discontinuation of OAC is limited by asymptomatic episodes of AF (meaning patients would be unaware of AF recurrence), late AF recurrence (meaning short-term freedom from recurrent AF might not predict long-term success), as well as a lack of clear temporal association between AF recurrence and stroke/systemic embolism.[75],[95],[395] Because of the lack of large RCTs to provide a definitive answer on how to manage OAC late after ablation, at this point in time, AF ablation should not be considered as an alternative to OAC. If a patient has a high thromboembolic risk profile, then the patient should continue OAC even after successful AF ablation. If discontinuation of anticoagulation is being considered on the basis of patient values and preferences, regular and prolonged monitoring for AF screening is suggested. The Optimal Anticoagulation for Enhanced Risk Patients Post-Catheter Ablation for Atrial Fibrillation (OCEAN) trial will provide guidance in the future (NCT02168829).[396]

Recommendation

49. We recommend that catheter ablation procedures for AF be performed with uninterrupted OAC (Strong Recommendation; High-Quality Evidence).

50. We suggest that after successful catheter or surgical ablation of AF, the decision to continue OAC beyond 2 months after ablation should be determined on the basis of the patient’s risk of stroke (“CCS algorithm”) and not according to the apparent success of the procedure (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a high value on the use of predictive stroke risk indexes in determining long-term stroke risk in all AF and AFL patients, as well as the potential for ongoing risk even after successful ablation.

Practical Tip

Limited retrospective and large data series have suggested a low risk of stroke and, therefore, the ability to stop OAC after successful AF ablation. However, there are no randomized trials, or even large prospective studies, to confirm these findings.

8.4.2.1 Catheter ablation of AFL

Patients in sustained AFL need to be anticoagulated for at least 3 weeks before the ablation procedure and at least 1 month after ablation, on the basis of the same principles underlying the rationale for OAC after cardioversion. Similarly, a preprocedural TEE can be used to rule out atrial thrombus before AFL ablation if the arrhythmia has been continuous for more than 48 hours without appropriate anticoagulation.[397] Analogous to AF, it is reasonable to perform catheter ablation procedures for AFL with uninterrupted OAC. Continued anticoagulation after successful AFL ablation is generally warranted in light of recognition that patients with isolated typical right AFL have an increased risk of later developing AF. The risk of thromboembolic events remains high despite successful AFL ablation.[189],[398],[399] The main predictors for the occurrence of stroke after AFL ablation are older age, higher CHA2DS2-VASc score, recurrence of AFL, and subsequent development of AF.[189],[190],[399] Multivariate predictors of AF after ablation of AFL include AF before ablation, reduced LV ejection fraction (LVEF), hypertension, increased LA size, inducible AF during the electrophysiology study, endurance sports, and structural heart disease.[400] In patients without a preexisting history of AF up to 20%-40% will develop AF over the 18-36 months after AFL ablation.[189],[398],[400] In patients with a history of preexisting AF up to 40%-75% will continue to experience AF after AFL ablation.[400] Although there is no RCT on the benefits of continued anticoagulation after AFL ablation, observation data suggest that patients with AFL and a CHA2DS2-VASc score 2 have a higher risk of stroke.[189],[190]

8.4.3 Management of OAC for patients undergoing invasive procedures

It is estimated that one in six AF patients who are receiving OAC will require an elective surgery or invasive procedure annually.[401] For these patients, the periprocedural risk of a thromboembolic event with OAC interruption must be weighed against the risk of periprocedural bleeding. An OAC interruption interval that is longer than necessary might increase the thromboembolic risk, whereas an insufficient period of interruption might increase the bleeding risk, with consequent delays in OAC resumption.[402] As such, perioperative anticoagulation management necessitates an assessment of the patient’s thrombotic risk, procedural bleeding risk (eg, minimal, low/moderate, and high bleed risk), specific anticoagulant used, renal function, and procedural bleeding risk (Table 9 and Figs. 12 and 13). An electronic tool incorporating these parameters to provide an OAC dosing schedule is provided by Thrombosis Canada (thrombosiscanada.ca).

Recommendation

51. We recommend that the decision to interrupt antithrombotic therapy for an invasive procedure must balance the risks of a thromboembolic event with those of a periprocedural bleeding event (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

The Thrombosis Canada (thrombosiscanada.ca) Perioperative Anticoagulant Management Algorithm is a helpful tool to aid decisions regarding periprocedural anticoagulation.

52. We suggest that interruption of OAC is not necessary for most procedures with a minimal risk of bleeding (Weak Recommendation; Moderate-Quality Evidence).

53. We recommend interruption of OAC for most procedures with a low/moderate or high risk of bleeding, or those for which the bleeding risk associated with the procedure is uncertain (Strong Recommendation; Low-Quality Evidence).

8.4.3.1 No bridging for interrupted VKA in low thromboembolic risk patients

When a decision to interrupt VKA therapy has been made for an invasive procedure with a low/moderate or high risk of bleeding, interruption of VKA without LMWH or unfractionated heparin (UFH) bridging is recommended for most patients at low thromboembolic risk (Fig. 13). The rationale for the avoidance of bridging has come from several sources. A meta-analysis of 33 observational studies and 1 RCT involving 7118 predominantly non-AF patients showed that VKA interruption with bridging therapy was associated with increased overall bleeding (13.1% vs 3.4%; P < 0.0001) and increased major bleeding (4.2% vs 0.9%; P ¼ 0.004), but without reduction in thromboembolic events (0.9% vs 0.6%; P ¼ 0.50) compared with VKA interruption without bridging therapy.[403] As well, in the randomized double-blinded placebo-controlled trial, Bridging Anticoagulation in Patients Who Require Temporary Interruption of Warfarin Therapy for an Elective Invasive Procedure or Surgery (BRIDGE),[404] 1884 AF patients (most with CHADS2 score < 4) undergoing interruption of VKA for an elective surgery/invasive procedure were evaluated. Patients were randomized to placebo or LMWH from 3 days to 1 day before the procedure, and for 5- 10 days after the procedure. No bridging was noninferior to bridging for arterial thromboembolism (0.4% vs 0.3%; P ¼ 0.01 for noninferiority), but was associated with significantly less major bleeding (1.3% vs 3.2%; P ¼ 0.005 for superiority) and significantly less minor bleeding (12.0% vs 20.9%; P < 0.001). There were no significant differences for any other outcomes (all-cause mortality, MI, deep vein thrombosis, or pulmonary embolism).

Recommendation

54. When a decision to interrupt VKA therapy for an invasive procedure has been made, we suggest that the interruption begin 5 days before the procedure, that a procedure with a low bleeding risk may proceed when the INR is ≤ 1.5, and a procedure with an intermediate or high bleeding risk may proceed when the INR is ≤ 1.2 (Weak Recommendation; Low-Quality Evidence).

8.4.3.2 Bridging for interrupted VKA in high thromboembolic risk patients

Bridging should be considered only for patients at high risk of stroke/systemic embolism when a decision is made to interrupt VKA therapy for an invasive procedure with a low/moderate or high risk of bleeding. Such patients would include those with “valvular AF” (mechanical heart valves or moderate severe mitral valve stenosis), NVAF with a CHADS2 score of 5-6, and those with a recent stroke or TIA. Before the procedure, patients should receive 3 days of LMWH bridging, although the dose of LMWH bridging should be reduced by 50% on the day before the procedure (Fig. 13).

Recommendation

55. When a decision to interrupt VKA therapy for an invasive procedure has been made, we suggest that bridging therapy with LMWH or UFH should be started when the INR is below therapeutic level only in patients at high risk of thromboembolic events (mechanical heart valves, moderate-severe mitral valve stenosis, NVAF with a CHADS2 score of 5-6, and those with a recent stroke or TIA) (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a relatively higher value on prevention of stroke/systemic embolism, and a relatively lower value on the inconvenience and risk of major bleeding associated with heparin bridging.

8.4.3.3 No bridging for interrupted DOACs

Bridging is not necessary for DOAC-treated patients who need DOAC interruption. DOACs have a rapid onset and offset of action and relatively short elimination half-lives (ie, 10-14 hours), rendering bridging unnecessary from a pharmacological perspective.[405] In addition, the use of bridging with interrupted DOAC has been associated with more bleeding and limited efficacy. Increased bleeding with bridging was shown in the prospective observational Dresden Registry (76% rivaroxaban, 24% dabigatran) in which heparin bridging did not reduce major cardiovascular events (stroke, venous thromboembolism [VTE], or ACS; 0.8% with no bridging vs 1.6% with bridging; P ¼ 0.265), but led to significantly higher rates of major bleeding complications (2.7% vs 0.5%; P ¼ 0.010).[406] Post hoc analyses of the phase III DOAC trials[21]-[25] support no bridging for DOAC interruption. Interruption of dabigatran, rivaroxaban, and apixaban without bridging in these studies had comparable rates of bleeding and thromboembolic events compared with their respective VKA groups.[407][409] The Perioperative Anticoagulant Use for Surgery Evaluation for Patients on a Direct Oral AnticoagulantWho Need an Elective Surgery or Procedure (PAUSE) study was a large prospective cohort study of 3007 DOAC-treated AF patients scheduled to undergo an elective surgery or procedure.[410] This study demonstrated the safety with preserved efficacy of a simple standardized DOAC interruption and resumption protocol that did not involve perioperative heparin bridging or preoperative coagulation function testing. Specifically, interruption of DOAC was associated with low rates of major bleeding (1.35% in the apixaban cohort, 0.90% in the dabigatran cohort, and 1.85% in the rivaroxaban cohort) and low rates of stroke/systemic embolism (0.16% in the apixaban cohort, 0.60% in the dabigatran cohort, and 0.37% in the rivaroxaban cohort).

Recommendation

56. When a decision to interrupt DOAC for an invasive procedure has been made for a patient with AF, we suggest that duration of interruption be on the basis of the risk of bleeding associated with the procedure and the patient’s renal function (Weak Recommendation; Low-Quality Evidence).

Practical Tip

Duration of preprocedural interruption of a DOAC should be adjusted according to renal function.

57. We recommend no bridging (LMWH or UFH) for NVAF patients requiring DOAC interruption for elective surgery or invasive procedures (Strong Recommendation; Moderate-Quality Evidence).

8.4.3.4 OAC resumption

After the invasive procedure, DOAC is reintroduced after hemostasis has been achieved (usually 24 hours after a low/moderate bleed risk procedure and 48-72 hours after a high bleed risk procedure). VKA therapy may be reintroduced on the same evening after the procedure, recognizing that it will take several days for the INR to achieve therapeutic range. When bridging LMWH is used, we suggest using a therapeutic-dose regimen for 3 days before the procedure and to resume 24 hours after a low/moderate bleed risk procedure and 48-72 hours after a high bleed risk procedure. In selected patients, for example, those undergoing a cardiac, intracranial, or spinal procedure, it is suggested to forego postprocedure LMWH bridging altogether, although low-dose heparin regimens, typically used for VTE prophylaxis, can be considered post procedure. The recommended approach to OAC interruption is presented in Figures 12 and 13 and Table 9.

Recommendation

58. When OAC has been interrupted for an invasive procedure we suggest that such therapy be restarted when hemostasis is established (within 24 hours for a procedure with a low risk of bleeding and within 48-72 hours for a procedure with a high risk of bleeding) (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

Recommendations regarding the timing of postprocedural reintroduction of antithrombotic therapy are intended to promote a balanced approach to minimizing the combined outcome of postprocedural thromboembolic events and major bleeding.

8.5 Bleeding

8.5.1 Prevention

Bleeding is a known adverse effect of OAC. Despite the similar or lower risk of bleeding with DOACs vs VKAs, there remains a significant residual 2%-4% annual risk of major bleeding. Assessment of bleeding risk is important to understand the risk-benefit balance of OAC, to identify and optimize modifiable bleeding risk factors, to identify patients who might benefit from more frequent follow-up, and to provide a framework for ongoing monitoring. The use of a validated bleeding risk prediction algorithm appears to be better at predicting major bleeding than using individual modifiable bleeding risk factors.[411][413] Despite lack of supportive RCT evidence, the identification of modifiable bleeding risk factors before OAC initiation and at each point of contact should be routinely done. GI protection for patients at high risk of GI events and require concomitant ASA or NSAID therapy has been associated with a reduction in GI bleeding and ulcer complications. In one of the earliest studies, albeit a small study with limited number of events, patients with acute GI bleeding or obstruction due to ulcer who received Helicobacter pylori eradication therapy restarted ASA 100 mg daily treatment with lansoprazole 30 mg daily or placebo.[414] The trial was stopped early after the second of 2 planned interim analyses revealed a statistically significant reduction in recurrent bleeding in the patients who received lansoprazole (1.6% vs 15%; HR, 10.6; 95% CI, 1.3-86.1). Subsequently the benefit of using a PPI for patients with no baseline evidence of peptic ulcer disease who required daily ASA (in doses ranging from 75 to 325 mg daily) was shown in the Randomized, Double-Blind, Placebo Controlled Study to Assess the Prevention of Low Dose Acetylsalicylic Acid (ASA) Associated Gastroduodenal Lesions and Upper Gastrointestinal Symptoms in Patients Taking Esomeprazole 20 mg Once Daily for Two Weeks (ASTERIX) study.[415] In 991 patients 60 years of age or older, the additional use of esomeprazole 20 mg daily reduced the risk of endoscopically diagnosed gastric and duodenal ulcers (1.6% with esomeprazole vs 5.4% with placebo; P ¼ 0.0007). In addition, the use of PPIs lowered the risk of upper GI bleeding in patients who required DAPT with ASA and clopidogrel in the Clopidogrel and the Optimization of Gastrointestinal Events Trial (COGENT) trial, and decreased the occurrence of peptic ulcers in patients at high concomitant risk of cardiovascular and GI events who were taking ASA in the Randomized, DoubleBlind, Parallel Group, Multicentre, Phase III Study to Assess the Effect of Esomeprazole 20 or 40 mg od Versus Placebo on the Occurrence of Peptic Ulcers During 26 Weeks in Subjects on Continuous Low Dose Acetylsalicylic Acid (OBERON) trial.[416],[417] There is no published RCT in which PPI use was investigated in patients with AF who were receiving standard stroke prevention dose OAC alone or in combination with or. In a retrospective cohort study of > 1.6 million Medicare beneficiaries who were receiving anticoagulation, PPI co-therapy was associated with a lower rate of upper GI bleeding hospitalizations: incidence rate ratio, 0.66 (95% CI, 0.62-0.69).[418] However, more recently in the COMPASS – Proton Pump Inhibitor (COMPASS-PPI) study[419] the effect of PPI use in patients receiving ASA in combination with rivaroxaban 2.5 mg BID was investigated. In this study, patients with stable atherosclerotic vascular disease and no clinical need for a PPI who were receiving 1 of 3 regimens: rivaroxaban 2.5 mg BID with ASA 100 mg daily, rivaroxaban 5 mg BID, or ASA 100 mg daily without anticoagulation, were randomized to pantoprazole 40 mg daily or placebo. The main reason for exclusion from the PPI arm of this trial was physician judgement that their patients had a clinical need for a PPI; this accounted for more than 35% of eligible patients (n ¼ 9797). Of the 17,598 patients randomized, the mean age was 68 years, 78% were male, and 2.6% had baseline peptic ulcer disease. These patients were followed for a mean of 3 years. Clinically significant GI events were no different in the 2 arms: 1.2% in the PPI arm vs 1.3% in the placebo arm (HR, 0.88; 95% CI, 0.67-1.15). The event rate in the nonrandomized cohort was 0.6%. In an analysis of the individual GI events included in the primary outcome, there was a reduction in overt gastroduodenal bleeding with pantoprazole (0.4% per year) over placebo (1.2% per year; HR, 0.52; 95% CI, 0.28-0.94; P ¼ 0.03). In addition, the rate of discontinuation of pantoprazole was similar to that of placebo overall (21.4% vs 22.4%) and for serious adverse events (0.9% vs 0.75%). Of note, a Cochrane review is currently under way to assess the benefits and harms of PPIs vs histamine-2 receptor antagonists or placebo for the prevention of upper GI bleeding in adults receiving single-agent and combination antithrombotic therapy for cardiovascular conditions.[420]

Recommendation

59. We recommend initial and ongoing evaluation of bleeding risk for all patients with AF whose stroke risk warrants antithrombotic therapy, with the use of strategies to mitigate the increased risk of bleeding associated with OAC (Strong Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places greater emphasis on routinely and systematically evaluating bleeding risk factors in patients who are receiving antiplatelet or OAC with emphasis on modifying bleeding risk when possible and less emphasis on using a bleeding risk stratification score to determine whether to initiate antithrombotic therapy or not.

Practical Tip

Individual patient bleeding risk should be taken into account when determining the strategy for stroke risk management. The use of the HAS-BLED algorithm might be valuable to identify patients at high risk of major bleeding who might benefit from more frequent follow-up.

Practical Tip

Emphasis should be placed on the identification of modifiable risk factors to decrease the risk of major bleeding at each medical encounter. These include BP control, avoidance of unnecessary antiplatelet and NSAID therapy, management of anemia, limiting alcohol intake, improving INR control, and ensuring patients are receiving DOAC doses that align with the Health Canada-approved dosing recommendations.

Practical Tip

Patients receiving OAC should be educated regarding self-monitoring for bleeding, including when to seek medical help.

60. We suggest the additional use of a PPI to decrease the risk of GI adverse effects, for patients who require daily antithrombotic therapy that includes ASA (Weak Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places greater value on the published RCT evidence of decreased GI complications, including GI bleeding, with the additional use of a PPI with antithrombotic therapy, and lesser value on the short duration of PPI treatment used in the trials and the fact that the trials did not specifically enroll patients with AF.

Practical Tip

Gastroprotection with a PPI has been shown to decrease the risk of GI ulceration and other adverse bleeding events in patients receiving daily ASA who are older than 60 years or have at least 1 other risk factor for bleeding. It stands to reason that this benefit would be extended to patients receiving the combination of daily ASA and anticoagulation, despite the lack of published evidence in this patient group.

8.5.2 Management of a bleeding event

DOACs are the preferred agents for stroke prevention in NVAF patients who merit anticoagulation. Although less lifethreatening bleeding was shown with DOACs, annual rates of major bleeding were observed to be 2%-4%.[21][23],[25] Bleeding management protocols for DOACs have included supportive therapy alone, antifibrinolytic therapy, hemostatic factors such as fresh-frozen plasma, 3- or 4-factor prothrombin complex concentrates, and recombinant activated factor VII; activated charcoal for overdose or unintentional ingestion; dialysis or continuous renal replacement therapy; and, specific reversal agents such as idarucizumab and andexanet alfa. An approach to bleeding is outlined in Figure 14.

Recommendation

61. In patients experiencing a bleeding event with OAC treatment we recommend investigation into the cause of the bleeding (Strong Recommendation; LowQuality Evidence).

Values and Preferences

This recommendation places a relatively high value on the recognition that patients who experience a GI or genitourinary bleeding event after OAC initiation are at higher risk of having an underlying malignancy discovered as the cause of their bleeding.

62. We recommend that anticoagulant therapy should be recommenced in patients at high risk of stroke as soon as possible after the cause of bleeding has been identified and corrected (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a relatively high value on the recognition that OAC discontinuation after a bleeding event is associated with a significant increase in the risk of stroke and all-cause mortality. Idarucizumab is a humanized monoclonal antibody fragment that binds to protein-bound and unbound dabigatran with high affinity, neutralizing dabigatran and its active metabolites.[421] In phase I trials with > 200 volunteers, idarucizumab was well tolerated.[422][424] In the Reversal Effects of Idarucizumab on Active Dabigatran (REVERSE-AD) trial, adults with overt, life-threatening bleeding (n ¼ 301) or those requiring urgent invasive procedures (n ¼ 202) while receiving dabigatran received idarucizumab as two 2.5-g bolus infusions up to 15 minutes apart.[425] In those with overt bleeding, 137 (45.5%) presented with GI bleeding and 98 (32.6%) with ICH. Urgent surgery was defined as a procedure requiring normal hemostasis that could not be delayed for at least 8 hours. The median maximum percent reversal of the anticoagulant effect of dabigatran was 100% within 4 hours of idarucizumab administration in those who had a prolonged dilute thrombin time or ecarin clotting time at baseline (92%). Reversal was rapid and occurred independently of age, sex, renal function, and baseline dabigatran concentration. was 2.5 hours after idarucizumab administration in 134 patients (45%) with uncontrolled bleeding, and all had confirmed bleeding cessation within 24 hours. Of the 197 patients who underwent surgery, periprocedural hemostasis was assessed as normal or mildly abnormal in 98.5%. At 30 days, thrombotic events occurred in 4.8% and the mortality rate was 13.5% in those with uncontrolled bleeding and 12.6% in those requiring an urgent procedure. Postmarketing experience in large, practice-based cohorts of consecutive patients, has shown hemostatic effectiveness and clinical outcomes similar to those seen in REVERSE-AD.[426],[427]

Recommendation

63. We recommend administering idarucizumab for emergency reversal of dabigatran’s anticoagulant effect in patients with uncontrollable or potentially life-threatening bleeding and/or in patients who require urgent surgery for which normal hemostasis is necessary (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a relatively greater value on the ability of idarucizumab to reverse coagulation parameters indicative of dabigatran’s effect, its potential to decrease bleeding-related outcomes, and risks of urgent surgery, and its safety and tolerability profile, and less value on the absence of a control group in the REVERSE-AD trial and on the cost of the drug.

Practical Tip

Reversing OAC exposes patients to the thrombotic risk of their underlying disease. OAC should be reintroduced as soon as medically appropriate.

Andexanet alfa is a recombinant modified human factor Xa decoy protein with high affinity for the active site of factor Xa inhibitors, sequestering them within the vascular space.[428] In patients requiring anticoagulation that involves inhibition of Factor Xa, the presence of andexanet alfa will inhibit the anticoagulant effectiveness. The pharmacodynamic half-life is approximately 1 hour. The Andexanet Alfa, A Novel Antidote to the Anticoagulation Effects of Factor Xa Inhibitors (ANNEXA-4) was a single-group cohort study designed to assess the efficacy and safety of andexanet alfa in patients with acute major bleeding occurring while taking a factor Xa inhibitor. Patients who had received 1 of 4 factor Xa inhibitors (apixaban, edoxaban, enoxaparin, or rivaroxaban) within 18 hours were treated with a bolus and 2-hour infusion of andexanet alfa.[429] The dose of andexanet alfa was dependent on the timing of the last dose of factor Xa inhibitor. Patients were excluded if they required surgery within 12 hours, if they received other bleeding treatments such as prothrombin complex concentrate, recombinant factor VIIa, whole blood, or plasma, and if they had a Glascow Coma Scale of < 7, an estimated ICH volume of > 60 mL, or an expected survival of < 1 month. Patients enrolled (n ¼ 352) had a mean age of 77 years. Bleeding was predominantly ICH (64%) and GI (26%). Most of the patients were receiving apixaban (54%) or rivaroxaban (36%). Anti-factor Xa activity decreased by 92% for apixaban (149.7 ng/mL to 11.1 ng/mL) and rivaroxaban (211.8 ng/mL to 14.2 ng/mL). Of 249 patients who were eligible for the hemostatic efficacy analysis, 204 were adjudicated as having excellent or good hemostasis at 12 hours. Reduction in anti-factor Xa activity was not predictive of hemostatic efficacy overall but was modestly predictive in patients with ICH. At 30 days of follow-up of the overall study population 49 (14%) of the patients had died and 34 (10%) had a thrombotic event. All thrombotic events occurred before reinitiation of OAC. Andexanet alfa received accelerated Food and Drug Administration approval in the United States in 2018 for the management of life-threatening or uncontrolled bleeding in patients treated with rivaroxaban and apixaban.[430] A condition of this accelerated approval was the requirement to complete an RCT including patients with acute ICH, the results of which might affect the modification or withdrawal of approval. The Food and Drug Administration also mandated a black box warning that andexanet alfa has been associated with thromboembolic events, ischemic events, cardiac arrest, and sudden death. There appears to be some increase in endogenous thrombin generation potential upon bolus administration of andexanet alfa although the mechanism is not yet known[431] and did not appear to be associated with clinical thrombotic events in healthy volunteers.[428] The thrombosis rate in a single health system retrospective cohort study of 13 patients who received andexanet alfa was 30% (4 patients) and mortality was 15% (2 patients).[432] In another, the thrombosis rate was 0 and mortality was 40%.[433] Practical challenges reported with andexanet alfa include access to product, lack of availability of anti-factor Xa levels, and drug preparation requiring reconstitution of multiple vials.[433]

Recommendation

64. We recommend administering andexanet alfa (when available) for emergency reversal of the anticoagulant effect of apixaban, edoxaban, and rivaroxaban in patients who present with uncontrollable or potentially life-threatening bleeding who have received any of these agents within the preceding 18 hours (Strong Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places relatively greater value on the ability of andexanet alfa to reverse anti-factor Xa activity and its potential to facilitate hemostasis, and less value on the low risk of infusion reactions and the absence of a control group in the ANNEXA-4 trial.

Practical Tip

Patients who are anticipated to require unfractionated or low molecular-weight heparin within 12-24 hours should not receive andexanet alfa.

Practical Tip

The dose of andexanet alfa is on the basis of the specific FXa inhibitor, the dose the patient is taking, and the timing of the last dose before the bleed.

Practical Tip

Reversing OAC exposes patients to the thrombotic risk of their underlying disease. There is evidence of thrombin generation after administration of andexanet alfa. OAC should be reintroduced as soon as medically appropriate.

8.6 Secondary stroke prevention

8.6.1 Stroke during OAC treatment

Use of OAC is associated with a substantial risk reduction in the occurrence of ischemic stroke (see section 8.2), however, some patients treated with OAC will still experience ischemic stroke or TIA despite use of OAC.[51],[52] Studies have shown that strokes that occur in patients receiving OAC tend to be less severe than strokes that occur in AF patients not taking OAC; in VKA-treated patients, stroke severity has been inversely correlated with INR level at the time of the event.[434],[435] After confirmation of the stroke diagnosis, an on-treatment event should prompt an evaluation for potential causes (other than AF) and ensure optimization of OAC and risk factor management.

Recommendation

65. We recommend that patients with AF who experience an ischemic stroke while receiving OAC be managed acutely according to the secondary stroke prevention practice guidelines (eg, Canadian stroke best practice recommendations[118]), with emphasis on addressing OAC medication adherence, ensuring correct OAC dosing and avoidance of drug interactions, identifying and treating other potential causes for the stroke other than AF, and promoting general vascular risk factor modification and healthy lifestyle choices (Strong Recommendation; Moderate-Quality Evidence).

8.6.1.1 Potential causes of an ischemic stroke in a patient receiving OAC

I. Inadequate intensity of anticoagulation. In patients treated with VKAs, a subtherapeutic INR is associated with an increased risk of stroke as well as greater stroke severity.[434],[435] In those treated with DOACs, undertreatment (eg, low plasma levels) has been associated with higher risk of thromboembolic events, and increased stroke severity (eg, large vessel occlusion).[205][207],[436] For DOAC-treated patients, undertreatment might take the form of the prescription of a reduced dose without an indication to do so, failure to anticipate drug-drug interactions (eg, OAC levels might be decreased by interaction with anticonvulsants such as phenytoin) or, in the case of rivaroxaban, a failure to account for GI absorption. Specifically, rivaroxaban should be taken with food for maximum GI absorption.

II. Suboptimal medication adherence or persistence. Poor patient adherence is a common problem with many chronic medications, adherence and persistence to stroke prevention therapies being particularly problematic in the AF population. As outlined in section 7.2 suboptimal adherence and persistence with OAC has been associated with higher rates of all-cause mortality and stroke.[156],[157],[160][162] Missed doses is a particular concern for DOACs because of their short half-life (average 12 hours). Strategies to improve persistence and adherence are outlined in section 7.2 and Table 6.

III. Alternate stroke etiology. Patients with AF might have other etiologies for stroke, some of which require specific management. Examples include large artery atherosclerosis of the intra- or extracranial circulation, cerebral small-vessel disease, other cardiac sources of embolism eg, patent foramen ovale) or another determined cause (carotid or vertebral artery dissection, vasculitis, etc).

IV. Suboptimal risk factor management. Untreated or suboptimally controlled stroke risk factors (eg, hypertension, dyslipidemia, etc) might contribute to the occurrence of stroke, independent of the presence of AF. Of particular importance, reduction in BP reduces the risk of recurrence of ischemic stroke as well as incident hemorrhagic stroke.[118]

8.6.1.2 Practical clinical approach to patients with an ischemic stroke who are receiving OAC therapy

I. Ensure an accurate diagnosis of the event. Brain imaging with CT or MRI is recommended for the diagnostic evaluation of acute stroke to exclude ICH, hemorrhagic transformation of ischemic stroke, and other structural pathology (MRI has greater diagnostic sensitivity than CT for identifying acute ischemia and small lesions). TIAs are frequently overdiagnosed, with up to 60% of patients suspected to have had a TIA having nonischemic causes for their symptoms after systemic evaluation.[437],[438] Although TIA remains a clinical diagnosis mainly on the basis of a detailed history of the event, brain imaging is useful in assessing for mimics (eg, migraine aura, seizure, peripheral vertigo, presyncope, neuropathy, multiple sclerosis) including small subarachnoid hemorrhages.[439]

II. Investigate for the most likely etiology of the stroke event. A typical etiological stroke workup for a patient with known AF consists of brain imaging (CT or MRI); vascular imaging (carotid ultrasound at a minimum; ideally CT angiography or MR angiography from the aortic arch to vertex), echocardiography, and laboratory investigations.

III. Determine if any other treatments are indicated for stroke risk reduction. For example, symptomatic carotid artery stenosis that is moderate (50%-69%) or severe (70%-99%) might benefit from urgent revascularization, whereas nonsurgical management is usually recommended for asymptomatic carotid stenosis.[118]

IV. Assess medication adherence. Patients and/or family members should be questioned regarding missed doses or prolonged interruptions (eg, periprocedural or in association with a bleeding event). For VKA-treated patients, the INR record for the preceding months should be reviewed. Patients should be counselled about the importance of daily medication adherence, the dangers of missed doses, and avoidance of prolonged or unnecessary treatment interruptions. Encourage adherence- and persistence-enhancing strategies as outlined in section 7.2 and Table 6.

V. Determine whether or not any changes are needed to the patient’s current OAC (agent or dose). For VKA-treated patients with suboptimal INR control, either switch to a DOAC (if eligible) or aim for improved INR control (consider a higher target INR and more frequent INR monitoring). For DOAC-treated patients, it is reasonable to continue the same DOAC (after ensuring it is at the correct dose for the patient’s age, renal function, and body weight) or switch to a different DOAC. For patients unable to comply with DOACs, then a switch to a VKA might be reasonable because of the longer half-life and ability to monitor the INR. The additional use of an antiplatelet agent is sometimes considered, however, the additional use of an antiplatelet agent with therapeutic OAC is associated with a significant increase in bleeding events without an additional benefit for stroke prevention. LAA occlusion (LAAO) might be an option for some patients but the optimal candidates for this procedure remains unclear.

VI. Identify and address any untreated vascular risk factors. Optimize BP and lipids, recommend smoking cessation, and promote general secondary stroke prevention lifestyle recommendations (diet, exercise).

For further details on the etiological stroke workup and secondary stroke prevention treatment recommendations, see the Canadian stroke best practice recommendations[118] available at: strokebestpractices.ca.

8.6.2 Timing of OAC initiation after acute ischemic stroke in patients with AF

In the 2-week period immediately after a TIA or ischemic stroke, patients with AF have an increased risk of ischemic stroke that ranges from 0.5%-1.3% per day.[440],[441] Initiation of OAC during this phase must balance the benefit of OAC with the risk of hemorrhagic transformation of the acute brain infarct. Hemorrhagic transformation can range from asymptomatic petechiae to symptomatic parenchymal hematoma with mass effect.[442],[443] Hemorrhagic transformation is more frequent in large-volume infarcts and in the setting of thrombolysis or mechanical revascularization of the index stroke.[442],[443] The optimal timing for OAC initiation post stroke has not yet been clearly defined and practice patterns are highly variable.[444] RCTs are under way to help address this uncertainty. For now, the heterogeneity of strokes and the lack of randomized data (particularly for DOACs in the early poststroke period) preclude any specific recommendations regarding the timing of OAC initiation. In practice, treatment decisions are typically individualized on the basis of a benefit-risk assessment guided by brain imaging appearance and clinical context. Bridging with low-dose aspirin is often prescribed until OAC treatment is initiated. Serial head CT scans within the first days or weeks post stroke can be helpful to monitor the evolution of an acute infarct, especially for moderate or large infarcts, and to assess for the presence and extent of any hemorrhage, before OAC treatment initiation. Outcomes in relation to timing of OAC initiation have been studied in prospective cohort studies.[445],[446] In the Clinical Relevance of Microbleeds in Stroke-2 study 1490 participants with AF and stroke or TIA in whom anticoagulation was indicated were enrolled.[446] Of these, 1335 (90%) had a known date of OAC initiation post stroke that was dichotomized as early (≤ 4 days) or late ( ≥ 5 days or never). The groups differed significantly, with lower stroke severity, better premorbid functioning, and a lower probability of thrombolysis for the index event in those in the early OAC group. At 90 days of follow-up the combined end point of intracerebral hemorrhage, ischemic stroke, and death was similar between groups (2% with early and 5% with late initiation; adjusted OR, 1.17; 95% CI, 0.48-2.84; P ¼ 0.736). Recognizing that the event rate was low, the authors concluded that there did not appear to be a hazard associated with early anticoagulation in carefully selected patients. Results of a prospective cohort study consisting of 1029 individuals with ischemic stroke and AF suggested that initiation of anticoagulation between 4 and 14 days optimized the combined outcome of stroke, TIA, systemic embolus, symptomatic intracranial bleeding, and bleeding within 90 days of onset.[445] Most participants were treated with VKAs or mixed treatment protocols including heparin, with only 12% treated with DOACs. Results of 2 small RCTs have suggested the early use of DOACs is safe for small strokes using an MRI measure of hemorrhagic transformation.[447],[448] Participants (N ¼ 195) randomized to VKA or rivaroxaban with a median National Institutes of Health Stroke Scale (NIHSS) score of 2 (ie, very mild strokes) did not differ in the rate of hemorrhagic transformation detected using MRI, an imaging modality with high sensitivity for the detection of hemorrhage.[447] The Dabigatran Following Acute Transient Ischemic Attack and minor stroke trial (DATAS II) randomized 301 individuals with a median NIHSS score of 1 (ie, very mild strokes) to ASA or dabigatran within 72 hours of symptom onset. No symptomatic hemorrhagic transformation was observed and the rate of minor petechial hemorrhage within the acute infarct did not differ between the treatment arms. Infarct volume predicted the probability of hemorrhagic transformation.[448] These trials do not establish optimal time points for OAC initiation but suggest that an identifiable group of patients with very mild stroke severity and small volume infarcts can be safely anticoagulated early.

Recommendation

66. We recommend that the timing of initiation of anticoagulant therapy after an ischemic stroke should be individualized and take into account the competing risks of recurrent stroke against the risk of hemorrhagic transformation of infarction (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

The timing of OAC initiation or resumption might be as short as within 24 hours in patients with a TIA, 3 days in patients with a mild stroke (NIHSS score &lt; 8), 6 days in patients with a moderate stroke (NIHSS score 8-15), and 12-14 days in patients with a severe or large stroke (NIHSS score 16). Factors favouring delayed OAC initiation include: high NIHSS score (&gt; 8), moderate-large brain infarction on imaging, hemorrhagic transformation, neurological instability, advanced patient age, and uncontrolled hypertension. In these patients OAC treatment decisions should be made in collaboration with specialized expertise (eg, neurology).

In the absence of definitive data, the following is a pragmatic approach to the timing of OAC initiation or resumption after an ischemic stroke event.

I. It is reasonable to initiate a DOAC 1 day post event for patients with a TIA, 3 days for patients with a small infarct/mild stroke severity (NIHSS score < 8), 6 days in patients with a moderate-sized infarct/moderate severity stroke (NIHSS score 8-15), and 12-14 days in patients with a large infarct/severe stroke (NIHSS score >15). For a brief-duration TIA with no residual symptoms or deficits and no acute infarct or hemorrhage on head CT scan, it is reasonable to consider DOAC initiation on the day of the event provided timely follow-up is available (Figure 15).

II. It is reasonable to delay OAC initiation for more than 2 weeks post stroke if, in the judgement of the clinician the risk of bleeding is believed to be high (eg, for some patients with large infarcts and those with hemorrhagic transformation).

III. If OAC initiation is recommended after hospital discharge, coordination with the treating physicians and close followup is suggested because postdischarge recommendations are implemented in only two-thirds of patients.[449]

IV. Hypertension is a significant risk factor for intracerebral hemorrhage, and it is reasonable to delay OAC in the setting of uncontrolled hypertension.

V. In most instances, brain imaging should be repeated before initiating OAC to detect asymptomatic hemorrhagic transformation that might influence the timing for OAC initiation. Consideration should be given to delaying OAC in patients with hemorrhage other than minor petechial changes, because OAC in the presence of existing hemorrhage might increase bleeding. Delayed initiation in patients with hemorrhagic transformation was not associated with increased risk of ischemic stroke in an observational study of > 2000 patients with stroke and AF.[450]

VI. Neurological instability might suggest recurrent ischemia or hemorrhage and in either case, might require a repeat CT examination or reassessment of the timing of OAC initiation.

8.6.3 OAC initiation after hemorrhagic stroke

For patients with AF who have had an acute primary intracerebral hemorrhage, subdural, or subarachnoid hemorrhage, OAC is typically avoided in the acute subacute period, and decisions regarding OAC initiation should be made in consultation with a stroke/neurology specialist on the basis of careful risk stratification guided by brain MRI appearance, the presumed etiology for the hemorrhage, and the estimated risk of recurrence. Patients with a lobar (superficial) ICH attributed to cerebral amyloid angiopathy have a higher risk of recurrent ICH than patients with a deep hypertensive ICH. If OAC is initiated post-ICH, a DOAC is preferred over VKAs because of the lower rate of incidence of ICH with these agents.[52] Observational studies support the cautious use of OAC for selected patients post ICH, usually initiated weeks to months post ICH, but clinical equipoise exists.[451],[452] RCTs are currently under way to evaluate the safety and efficacy of DOAC vs aspirin in AF patients who have had an ICH.

8.7 Left atrial appendage occlusion

Imaging and postmortem studies suggest that most AFassociated ischemic strokes are cardioembolic, and that the majority arise from the LAA.[453],[454] In sinus rhythm the LAA has pulsatile flow, however in AF appendage emptying is reduced leading to stasis and clot formation. Physical elimination of the LAA (removal or occlusion) from the circulation is postulated to prevent thrombus formation and subsequent embolization, without suffering from the limitations of pharmacological therapy. Specifically, although OAC is effective at preventing AF-associated stroke/systemic embolism, its use is limited by short- and long term nonadherence, nonpersistence, and side effects such as the risk of major bleeding. Conversely, when performed, the effects of the physical elimination of the LAA are considered permanent and are not reliant on patient compliance.

8.7.1 Percutaneous LAAO

Two RCTs and a number of single and multicentre registries that have evaluated percutaneous device closure have been performed. The WATCHMAN LAA Closure Technology for Embolic Protection in Patients With Atrial Fibrillation (PROTECT AF) study enrolled 707 patients with AF and a CHADS2 score of ≥ 1, and the Prospective Randomized Evaluation of the WATCHMAN LAA Closure Device in Patients With Atrial Fibrillation vs Long-Term Warfarin Therapy (PREVAIL) study enrolled 407 patients with a CHADS2 score of 2.[455],[456] Both studies randomized patients to LAAO or VKA. The 5-year outcome data from these 2 studies were combined in a meta-analysis.[457] Although LAAO was deemed noninferior to VKAs for the combined end points, much of the benefit of LAAO is due to a reduction in intracranial bleeding (0.2% per year with LAAO vs 0.9% per year with VKA; HR, 0.20; 95% CI, 0.07-0.56; P ¼ 0.002).[457] However, the effect of LAAO on ischemic stroke remains to be determined, and is suggested to be inferior (1.6% per year with LAAO vs 0.95% per year with VKA; HR, 1.71; 95% CI, 0.94-3.11; P ¼ 0.08).[457] Higher risk of ischemic stroke might relate to device-related thrombosis, which occurs in 2%-4% of cases and is a known independent predictor of stroke (HR, 4.4; 95% CI, 1.05-18.43).[458] In the recently published randomized multicentre Left Atrial Appendage Closure Versus Novel Anticoagulation Agents in Atrial Fibrillation (PRAGUE-17) study the safety and efficacy of LAAO was compared with DOAC in high-risk AF patients with a history of significant bleeding or thromboembolic event while receiving OAC (CHA2DS2-VASc score 4.7 ±1.5; HAS-BLED score 3.1 ± 0.9). This study showed no significant difference between LAAO and DOAC, with similar rates of all-cause stroke/TIA (2.20% with LAAO vs 2.68% with DOACs), ischemic stroke/TIA (2.20% with LAAO vs 2.38% with DOACs), and major and CRNM bleeding (5.5% per year with LAAO vs 7.42% per year with DOACs). There was no difference in the end points of cardiovascular death, noncardiovascular death, or all-cause mortality. Major LAAO implant-related complications occurred in 4.5%. LAAO implantation was unsuccessful in 10.0% of the patients, however, the results of the study did not differ when analyzed according to treatment received (P ¼ 0.31) or per protocol (P ¼ 0.40).

Recommendation

67. We suggest that percutaneous LAAO be considered for stroke prevention in patients with NVAF who are at moderate to high risk of stroke and have absolute contraindications to OAC (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a high value on the evidence from multiple RCTs which showed significant benefit of VKAs and DOACs in the prevention of stroke/systemic embolism, relatively low risk of bleeding, and significant survival advantage, as well as the relative lack of data supporting LAAO for OACeligible patients.

Practical Tip

Patients should receive individualized counselling regarding the risks and benefits of stroke prevention therapy, with a focus on the evidence supporting long-term OAC as the preferred strategy.

Practical Tip

Contraindications to long-term OAC include but are not limited to: recurrent nontraumatic intracranial bleeding with high risk of recurrence, recurrent irreversible pulmonary bleed, recurrent irreversible urogenital bleed, recurrent irreversible GI bleed, recurrent irreversible retroperitoneal bleed, esophageal varices, intraocular bleeds, hereditary hemorrhagic telangiectasia.

8.7.2 Surgical LAA

The device-based trials provide proof of concept that LAAO might provide benefit, however they do not address the surgical AF population, do not inform on the combination of LAAO and OAC, and do not evaluate a surgical intervention that can be offered with little increase in risk. Results of existing observational studies suggest that standalone surgical LAAO can be safely and effectively performed, however, there are limited high-quality data to suggest that a stand-alone surgical LAAO approach is reasonable in patients eligible for a percutaneous approach.[459] In patients with AF who are already undergoing a thoracotomy, it is possible that concomitant surgical LAAO might offer significant benefit with minimal incremental risk. However, it is important to recognize that patients who undergo surgical LAAO often have higher CHA2DS2-VASc scores and more often have coexisting valvular heart disease (ie, are more likely to develop AF-associated thrombosis outside of the LAA).[454] To date the observational data of concomitant surgical LAAO are somewhat conflicting, with some observational series suggesting that surgical LAAO is associated with significantly greater freedom from stroke/systemic embolism, and others suggesting that LAAO did not reduce the incidence of stroke/systemic embolism in patients with AF (P ¼ 0.69), with the apparent stroke reduction being related to continued VKA use and not the LAAO procedure itself.[460],[461] However, these observational studies do not have sufficient power or freedom from bias to provide the level of evidence needed to clearly answer this important question. A large RCT of surgical exclusion of the LAA vs VKA therapy has completed recruitment of > 4800 patients and is in the follow-up phase.[462] The primary end point is stroke/systemic embolism over 4 years. Total mortality and safety end points will also be compared. Until this trial is completed, the only basis for recommendations regarding surgical LAA removal is consensus. As such, we have made a weak recommendation on the basis of the existing low-quality evidence.

Recommendation

68. We suggest surgical LAAO be considered for stroke prevention in patients with NVAF who are at moderate to high risk of stroke and have contraindications to OAC, and who are not suitable for percutaneous LAAO (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation is qualified by the patient’s stroke risk in relation to their perioperative risk.

Practical Tip

Patients should receive individualized counselling regarding the risks and benefits of stroke prevention therapy, with a focus on the evidence supporting long-term OAC treatment as the preferred prophylactic strategy.

69. We suggest concomitant surgical LAAO be considered in patients with AF who are undergoing an open chest cardiac surgical procedure and who are ineligible for long-term OAC (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation is qualified by the patient’s stroke risk in relation to their incremental surgical risk with concomitant LAAO, and that the contraindication for OAC is absolute.

Practical Tip

In patients with AF who are eligible for long-term OAC the evidence to date is insufficient to support a recommendation for or against concomitant surgical LAAO. In these patients it is recommended to continue OAC after surgical LAAO.

8.8 Cognitive function/dementia

An association between AF and cognitive impairment has been frequently reported over the past 30 years.[463],[464] Although the association has not been consistently observed,[465],[466] recent meta-analyses have shown a significant correlation between AF and cognitive impairment, irrespective of a history of previous stroke.[467][469] It has been estimated that the risk of dementia in individuals with AF and no overt cerebrovascular events is increased 30%-60% compared with patients without AF, even after adjustment for age, sex, multiple comorbidities, and cardiovascular medications.[470][472] An American study of community-dwelling individuals showed that AF was associated with a 50% increase in risk of developing Alzheimer disease compared with individuals without AF.[473] The findings are not just unique to North America and Europe, with a Taiwanese study involving 332,665 patients,[470] and a South Korean study of 262,611 patients[472] demonstrating that patients with AF had a 42% and a 63% higher risk of dementia, respectively, even after adjustment for age, sex, baseline differences, and medication. More recent data suggest that AF increases the risk of vascular and degenerative (eg, Alzheimer disease) dementia,[474],[475] and also that the relationship between AF and cognitive decline appears to be strongest when AF develops in middle age,[476],[477] when the AF is persistent and of long duration,[477],[478] and when the ventricular rate in AF is fast.[479],[480] Although AF is related to cognitive impairment and dementia, the etiological mechanisms remain a matter for conjecture. Shared risk factors have been proposed as part of the explanation. The CHADS2 and CHA2DS2-VASc scores have been shown to predict cognitive impairment as well as stroke in patients with AF. Nevertheless, the association between AF and cognitive decline persists even after controlling for age and these comorbidities.[467],[470],[481] Proposed mechanisms include silent cerebral infarcts due to microemboli, cerebral microbleeds, disruptions of the blood-brain barrier, cerebral vascular disease, cerebral hypoperfusion due to variability in the cardiac cycle and reduced cardiac output, oxidative stress, inflammation, and endothelial dysfunction.[471],[482][485] Of these, subclinical embolic cerebrovascular ischemia is thought to be the most likely mechanism.[485] Although white matter disease had been raised as another potential driver, recent work appears to refute this.[482],[486],[487] Treatments that have been proposed to reduce the risk of cognitive impairment include risk factor management,[488] sinus rhythm maintenance with ablation,[489] and anticoagulation.[490][492] A modest effect has been observed with specific risk factor treatments, such as statins or angiotensin conversion enzyme inhibitor and angiotensin receptor blocker therapy in some studies,[481],[488] but not in others.[493] There also remains controversy concerning the ability of anticoagulants to decrease the incidence of cognitive impairment and dementia in patients with AF,[494] with reviews of the available literature failing to find convincing evidence that they do so.[495],[496] Although positive data continue to accumulate, including from large population based studies,[497],[498] there have been no clinical trials to eliminate potential confounders. VKA use appears to be most effective when TTR is kept consistently very high (eg, > 75%).[499],[500] Data relating to the DOACs and dementia are sparse, with some studies showing relative benefit compared with VKAs,[491],[501],[502] whereas a nationwide Danish cohort study did not.503 Several clinical trials are being undertaken to allow greater clarity. In 2 of these studies, VKAs are being compared with dabigatran (Cognitive Impairment Related to Atrial Fibrillation Prevention Trial [GIRAF; NCT01994265] and Impact of Anticoagulation Therapy on the Cognitive Decline and Dementia in Patients With Non-Valvular Atrial Fibrillation [CAF; NCT03061006]) and in a third study, Blinded Randomized Trial of Anticoagulation to Prevent Ischemic Stroke and Neurocognitive Impairment in Atrial Fibrillation (BRAIN-AF; NCT02387229), rivaroxaban is being compared with standard of care. There are currently only 2 observational studies on the association between catheter ablation and cognitive dysfunction. The largest was a retrospective case control study from Intermountain Health in Utah, in which outcomes in 4212 patients who underwent AF ablation were compared with 16,848 age- and sex-matched patients with AF who did not undergo ablation. At 3 years, the incidence of dementia was low in both groups, but it was significantly less in the AF patients who had undergone ablation.[489] A small prospective case-control study was recently published, which involved 308 AF patients treated with ablation and 50 medically managed control participants.[504] Cognitive function was assessed at baseline, 3 months, and 1 year. Significant cognitive improvement was observed at 3 and 12 months in the ablation group compared with the control group, and no major adverse events were reported at either point in time. A larger prospective case-control study (Cognitive Impairment in Atrial Fibrillation [DIAL-F]; NCT01816308) is specifically addressing the effect of AF catheter ablation on dementia and is currently under way. Finally, LAAO might be expected to have a potential role on decreasing the incidence of cognitive impairment on the basis of subclinical strokes. However, whether the available devices can prevent microemboli remains to be seen.

References

169. Risk factors for stroke and efficacy of antithrombotic therapy in atrial fibrillation. Analysis of pooled data from five randomized controlled trials. Arch Intern Med 1994;154:1449-57.

170. Predictors of thromboembolism in atrial fibrillation: I. Clinical features of patients at risk. The Stroke Prevention in Atrial Fibrillation Investigators. Ann Intern Med 1992;116:1-5.

171. Olesen JB, Torp-Pedersen C, Hansen ML, Lip GY. The value of the CHA2DS2-VASc score for refining stroke risk stratification in patients with atrial fibrillation with a CHADS2 score 0-1: a nationwide cohort study. Thromb Haemost 2012;107:1172-9.

172. 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.

173. Lip GY, Nieuwlaat R, Pisters R, Lane DA, Crijns HJ. Refining clinical risk stratification for predicting stroke and thromboembolism in atrial fibrillation using a novel risk factor-based approach: the Euro Heart survey on atrial fibrillation. Chest 2010;137:263-72.

174. 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.

175. Verma A, Cairns JA, Mitchell LB, et al. 2014 Focused update of the Canadian Cardiovascular Society guidelines for the management of atrial fibrillation. Can J Cardiol 2014;30:1114-30.

176. Bell AD, Roussin A, Cartier R, et al. The use of antiplatelet therapy in the outpatient setting: Canadian Cardiovascular Society guidelines. Can J Cardiol 2011;27(suppl A):S1-59.

177. Proietti M, Lane DA, Boriani G, Lip GYH. Stroke prevention, evaluation of bleeding risk, and anticoagulant treatment management in atrial fibrillation contemporary international guidelines. Can J Cardiol 2019;35:619-33.

178. Quinn GR, Severdija ON, Chang Y, Singer DE. Wide variation in reported rates of stroke across cohorts of patients with atrial fibrillation. Circulation 2017;135:208-19.

179. Chao TF, Wang KL, Liu CJ, et al. Age threshold for increased stroke risk among patients with atrial fibrillation: a nationwide cohort study from Taiwan. J Am Coll Cardiol 2015;66:1339-47.

180. Friberg L, Skeppholm M, Terent A. Benefit of anticoagulation unlikely in patients with atrial fibrillation and a CHA2DS2-VASc score of 1. J Am Coll Cardiol 2015;65:225-32.

181. Singer DE, Chang Y, Borowsky LH, et al. A new risk scheme to predict ischemic stroke and other thromboembolism in atrial fibrillation: the ATRIA study stroke risk score. J Am Heart Assoc 2013;2:e000250.

182. Vadmann H, Nielsen PB, Hjortshoj SP, et al. Atrial flutter and thromboembolic risk: a systematic review. Heart 2015;101:1446-55.

183. Biblo LA, Yuan Z, Quan KJ, Mackall JA, Rimm AA. Risk of stroke in patients with atrial flutter. Am J Cardiol 2001;87:346-349, A349.

184. Corrado G, Sgalambro A, Mantero A, et al. Thromboembolic risk in atrial flutter. The FLASIEC (FLutter Atriale Societa Italiana di Ecografia Cardiovascolare) multicentre study. Eur Heart J 2001;22:1042-51.

185. Elhendy A, Gentile F, Khandheria BK, et al. Thromboembolic complications after electrical cardioversion in patients with atrial flutter. Am J Med 2001;111:433-8.

186. Gallagher MM, Hennessy BJ, Edvardsson N, et al. Embolic complications of direct current cardioversion of atrial arrhythmias: association with low intensity of anticoagulation at the time of cardioversion. J Am Coll Cardiol 2002;40:926-33.

187. Ghali WA, Wasil BI, Brant R, Exner DV, Cornuz J. Atrial flutter and the risk of thromboembolism: a systematic review and meta-analysis. Am J Med 2005;118:101-7.

188. Al-Kawaz M, Omran SS, Parikh NS, et al. Comparative risks of ischemic stroke in atrial flutter versus atrial fibrillation. J Stroke Cerebrovasc Dis 2018;27:839-44.

189. Gula LJ, Redfearn DP, Jenkyn KB, et al. Elevated incidence of atrial fibrillation and stroke in patients with atrial flutter-a population-based study. Can J Cardiol 2018;34:774-83.

190. Jin MN, Song C, Kim TH, et al. CHA(2)DS(2)-VASc score in the prediction of ischemic stroke in patients after radiofrequency catheter ablation of typical atrial flutter. Yonsei Med J 2018;59:236-42.

191. Lin YS, Chen YL, Chen TH, et al. Comparison of clinical outcomes among patients with atrial fibrillation or atrial flutter stratified by CHA2DS2-VASc score. JAMA Netw Open 2018;1:e180941.

192. Chen YL, Lin YS, Wang HT, et al. Clinical outcomes of solitary atrial flutter patients using anticoagulation therapy: a national cohort study. Europace 2019;21:313-21.

193. Parikh MG, Aziz Z, Krishnan K, Madias C, Trohman RG. Usefulness of transesophageal echocardiography to confirm clinical utility of CHA2DS2-VASc and CHADS2 scores in atrial flutter. Am J Cardiol 2012;109:550-5.

194. Eckman MH, Singer DE, Rosand J, Greenberg SM. Moving the tipping point: the decision to anticoagulate patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2011;4:14-21.

195. You JJ, Singer DE, Howard PA, et al. 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:e531S-75S.

196. Saposnik G, Gladstone D, Raptis R, et al. Atrial fibrillation in ischemic stroke: predicting response to thrombolysis and clinical outcomes. Stroke 2013;44:99-104.

197. Mant J, Hobbs FDR, Fletcher K, et al. 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.

198. Andersen LV, Vestergaard P, Deichgraeber P, et al. Warfarin for the prevention of systemic embolism in patients with non-valvular atrial fibrillation: a meta-analysis. Heart 2008;94:1607-13.

199. Connolly SJ, Pogue J, Eikelboom J, et al; ACTIVE W Investigators. Benefit of oral anticoagulant over antiplatelet therapy in atrial fibrillation depends on the quality of international normalized ratio control achieved by centers and countries as measured by time in therapeutic range. Circulation 2008;118:2029-37.

200. 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.

201. Haas S, Camm AJ, Bassand JP, et al. Predictors of NOAC versus VKA use for stroke prevention in patients with newly diagnosed atrial fibrillation: results from GARFIELD-AF. Am Heart J 2019;213:35-46.

202. Eikelboom JW, Connolly SJ, Brueckmann M, et al. Dabigatran versus warfarin in patients with mechanical heart valves. N Engl J Med 2013;369:1206-14.

203. Adams GF, Merrett JD, Hutchinson WM, Pollock AM. Cerebral embolism and mitral stenosis: survival with and without anticoagulants. J Neurol Neurosurg Psychiatry 1974;37:378-83.

204. Rush CJ, Campbell RT, Jhund PS, Petrie MC, McMurray JJV. Association is not causation: treatment effects cannot be estimated from observational data in heart failure. Eur Heart J 2018;39:3417-38.

205. Bell AD, Gross P, Heffernan M, et al. Appropriate use of antithrombotic medication in Canadian patients with nonvalvular atrial fibrillation. Am J Cardiol 2016;117:1107-11.

206. Steinberg BA, Shrader P, Thomas L, et al. Off-label dosing of nonvitamin K antagonist oral anticoagulants and adverse outcomes: the ORBIT-AF II Registry. J Am Coll Cardiol 2016;68:2597-604.

207. Yao X, Shah ND, Sangaralingham LR, Gersh BJ, Noseworthy PA. Nonvitamin K antagonist oral anticoagulant dosing in patients with atrial fibrillation and renal dysfunction. J Am Coll Cardiol 2017;69:2779-90.

208. Olesen JB, Lip GY, Kamper AL, et al. Stroke and bleeding in atrial fibrillation with chronic kidney disease. N Engl J Med 2012;367:625-35.

209. Bonde AN, Lip GY, Kamper AL, et al. Net clinical benefit of antithrombotic therapy in patients with atrial fibrillation and chronic kidney disease: a nationwide observational cohort study. J Am Coll Cardiol 2014;64:2471-82.

210. Nielsen PB, Lane DA, Rasmussen LH, Lip GY, Larsen TB. Renal function and non-vitamin K oral anticoagulants in comparison with warfarin on safety and efficacy outcomes in atrial fibrillation patients: a systemic review and meta-regression analysis. Clin Res Cardiol 2015;104:418-29.

211. Stanton BE, Barasch NS, Tellor KB. Comparison of the safety and effectiveness of apixaban versus warfarin in patients with severe renal impairment. Pharmacotherapy 2017;37:412-9.

212. Shah M, Avgil Tsadok M, Jackevicius CA, et al. Warfarin use and the risk for stroke and bleeding in patients with atrial fibrillation undergoing dialysis. Circulation 2014;129:1196-203.

213. Garg L, Chen C, Haines DE. Atrial fibrillation and chronic kidney disease requiring hemodialysis – does warfarin therapy improve the risks of this lethal combination? Int J Cardiol 2016;222:47-50.

214. Randhawa MS, Vishwanath R, Rai MP, et al. Association between use of warfarin for atrial fibrillation and outcomes among patients with endstage renal disease: a systematic review and meta-analysis. JAMA Netw Open 2020;3:e202175.

215. Pokorney SD, Black-Maier E, Hellkamp AS, et al. Oral anticoagulation and cardiovascular outcomes in patients with atrial fibrillation and endstage renal disease. J Am Coll Cardiol 2020;75:1299-308.

216. Del-Carpio Munoz F, Gharacholou SM, Munger TM, et al. Metaanalysis of renal function on the safety and efficacy of novel oral anticoagulants for atrial fibrillation. Am J Cardiol 2016;117:69-75.

217. Yao X, Tangri N, Gersh BJ, et al. Renal outcomes in anticoagulated patients with atrial fibrillation. J Am Coll Cardiol 2017;70:2621-32.

218. Stanifer JW, Pokorney SD, Chertow GM, et al. Apixaban versus warfarin in patients with atrial fibrillation and advanced chronic kidney disease. Circulation 2020;141:1384-92.

219. Chan KE, Edelman ER, Wenger JB, Thadhani RI, Maddux FW. Dabigatran and rivaroxaban use in atrial fibrillation patients on hemodialysis. Circulation 2015;131:972-9.

220. Chokesuwattanaskul R, Thongprayoon C, Tanawuttiwat T, et al. Safety and efficacy of apixaban versus warfarin in patients with end-stage renal disease: meta-analysis. Pacing Clin Electrophysiol 2018;41:627-34.

221. Reed D, Palkimas S, Hockman R, et al. Safety and effectiveness of apixaban compared to warfarin in dialysis patients. Res Pract Thromb Haemost 2018;2:291-8.

222. Siontis KC, Zhang X, Eckard A, et al. Outcomes associated with apixaban use in patients with end-stage kidney disease and atrial fibrillation in the United States. Circulation 2018;138:1519-29.

223. Gong IY, Kim RB. Importance of pharmacokinetic profile and variability as determinants of dose and response to dabigatran, rivaroxaban, and apixaban. Can J Cardiol 2013;29:S24-33.

224. Fox KA, Piccini JP, Wojdyla D, et al. Prevention of stroke and systemic embolism with rivaroxaban compared with warfarin in patients with non-valvular atrial fibrillation and moderate renal impairment. Eur Heart J 2011;32:2387-94.

225. Bohula EA, Giugliano RP, Ruff CT, et al. Impact of renal function on outcomes with edoxaban in the ENGAGE AF-TIMI 48 trial. Circulation 2016;134:24-36.

226. Andrade JG, Hawkins NM, Fordyce CB, et al. Variability in nonvitamin K antagonist oral anticoagulants dose adjustment in atrial fibrillation patients with renal dysfunction: the Influence of Renal Function Estimation Formulae. Can J Cardiol 2018;34:1010-8.

227. Kralev S, Schneider K, Lang S, Suselbeck T, Borggrefe M. Incidence and severity of coronary artery disease in patients with atrial fibrillation undergoing first-time coronary angiography. PLoS One 2011;6:e24964.

228. Lip GY, Laroche C, Dan GA, et al. A prospective survey in European Society of Cardiology member countries of atrial fibrillation management: baseline results of EURObservational Research Programme Atrial Fibrillation (EORP-AF) Pilot General Registry. Europace 2014;16:308-19.

229. Connolly S, Pogue J, Hart R, et al. Clopidogrel plus aspirin versus oral anticoagulation for atrial fibrillation in the Atrial fibrillation Clopidogrel Trial with Irbesartan for prevention of Vascular Events (ACTIVE W): a randomised controlled trial. Lancet 2006;367:1903-12.

230. Antithrombotic Trialists’ (ATT) Collaboration, Baigent C, Blackwell L, et al. Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials. Lancet 2009;373:1849-60.

231. Anand SS, Yusuf S. Oral anticoagulants in patients with coronary artery disease. J Am Coll Cardiol 2003;41. 62S-9S.

232. Eikelboom JW, Connolly SJ, Bosch J, et al. Rivaroxaban with or without aspirin in stable cardiovascular disease. N Engl J Med 2017;377:1319-30.

233. Randomised double-blind trial of fixed low-dose warfarin with aspirin after myocardial infarction. Coumadin Aspirin Reinfarction Study (CARS) Investigators. Lancet 1997;350:389-96.

234. Fiore LD, Ezekowitz MD, Brophy MT, et al. Department of Veterans Affairs Cooperative Studies Program Clinical Trial comparing combined warfarin and aspirin with aspirin alone in survivors of acute myocardial infarction: primary results of the CHAMP study. Circulation 2002;105:557-63.

235. Brouwer MA, van den Bergh PJ, Aengevaeren WR, et al. Aspirin plus coumarin versus aspirin alone in the prevention of reocclusion after fibrinolysis for acute myocardial infarction: results of the Antithrombotics in the Prevention of Reocclusion In Coronary Thrombolysis (APRICOT)-2 trial. Circulation 2002;106:659-65.

236. Hurlen M, Abdelnoor M, Smith P, Erikssen J, Arnesen H. Warfarin, aspirin, or both after myocardial infarction. N Engl J Med 2002;347:969-74.

237. van Es RF, Jonker JJ, Verheugt FW, et al. Aspirin and coumadin after acute coronary syndromes (the ASPECT-2 study): a randomised controlled trial. Lancet 2002;360:109-13.

238. Herlitz J, Holm J, Peterson M, et al. Effect of fixed low-dose warfarin added to aspirin in the long term after acute myocardial infarction; the LoWASA study. Eur Heart J 2004;25:232-9.

239. Yasuda S, Kaikita K, Ogawa H, et al. Atrial fibrillation and ischemic events with rivaroxaban in patients with stable coronary artery disease (AFIRE): protocol for a multicenter, prospective, randomized, openlabel, parallel group study. Int J Cardiol 2018;265:108-12.

240. Mehta SR, Bainey KR, Cantor WJ, et al. 2018 Canadian Cardiovascular Society/Canadian Association of Interventional Cardiology focused update of the guidelines for the use of antiplatelet therapy. Can J Cardiol 2018;34:214-33.

241. Angiolillo DJ, Goodman SG, Bhatt DL, et al. Antithrombotic therapy in patients with atrial fibrillation undergoing percutaneous coronary intervention: a North American perspective-2016 update. Circ Cardiovasc Interv 2016;9:e004395.

242. Windecker S, Lopes RD, Massaro T, et al. Antithrombotic therapy in patients with atrial fibrillation and acute coronary syndrome treated medically or with percutaneous coronary intervention or undergoing elective percutaneous coronary intervention: insights from the AUGUSTUS trial. Circulation 2019;140:1921-32.

243. Dewilde WJ, Oirbans T, Verheugt FW, et al. Use of clopidogrel with or without aspirin in patients taking oral anticoagulant therapy and undergoing percutaneous coronary intervention: an open-label, randomised, controlled trial. Lancet 2013;381:1107-15.

244. Gibson CM, Mehran R, Bode C, et al. Prevention of bleeding in patients with atrial fibrillation undergoing PCI. N Engl J Med 2016;375:2423-34.

245. Cannon CP, Bhatt DL, Oldgren J, et al. Dual antithrombotic therapy with dabigatran after PCI in atrial fibrillation. N Engl J Med 2017;377:1513-24.

246. Vranckx P, Valgimigli M, Eckardt L, et al. Edoxaban-based versus vitamin K antagonist-based antithrombotic regimen after successful coronary stenting in patients with atrial fibrillation (ENTRUST-AF PCI): a randomised, open-label, phase 3b trial. Lancet 2019;394:1335-43.

247. Lopes RD, Heizer G, Aronson R, et al. Antithrombotic therapy after acute coronary syndrome or PCI in atrial fibrillation. N Engl J Med 2019;380:1509-24.

248. Fiedler KA, Maeng M, Mehilli J, et al. Duration of triple therapy in patients requiring oral anticoagulation after drug-eluting stent implantation: the ISAR-TRIPLE trial. J Am Coll Cardiol 2015;65:1619-29.

249. Friberg L, Rosenqvist M, Lip GY. Evaluation of risk stratification schemes for ischaemic stroke and bleeding in 182 678 patients with atrial fibrillation: the Swedish Atrial Fibrillation cohort study. Eur Heart J 2012;33:1500-10.

250. Choi J, Kim J, Shim JH, Kim M, Nam GB. Risks versus benefits of anticoagulation for atrial fibrillation in cirrhotic patients. J Cardiovasc Pharmacol 2017;70:255-62.

251. Khoury T, Ayman AR, Cohen J, et al. The complex role of anticoagulation in cirrhosis: an updated review of where we are and where we are going. Digestion 2016;93:149-59.

252. Kuo L, Chao TF, Liu CJ, et al. Liver cirrhosis in patients with atrial fibrillation: would oral anticoagulation have a net clinical benefit for stroke prevention? J Am Heart Assoc 2017;6:e005307.

253. Kubitza D, Roth A, Becka M, et al. Effect of hepatic impairment on the pharmacokinetics and pharmacodynamics of a single dose of rivaroxaban, an oral, direct Factor Xa inhibitor. Br J Clin Pharmacol 2013;76:89-98.

254. Mueck W, Stampfuss J, Kubitza D, Becka M. Clinical pharmacokinetic and pharmacodynamic profile of rivaroxaban. Clin Pharmacokinet 2014;53:1-16.

255. Lauschke VM, Ingelman-Sundberg M. The importance of patient specific factors for hepatic drug response and toxicity. Int J Mol Sci 2016;17:1714.

256. Intagliata NM, Henry ZH, Maitland H, et al. Direct oral anticoagulants in cirrhosis patients pose similar risks of bleeding when compared to traditional anticoagulation. Dig Dis Sci 2016;61:1721-7.

257. Hum J, Shatzel JJ, Jou JH, Deloughery TG. The efficacy and safety of direct oral anticoagulants vs traditional anticoagulants in cirrhosis. Eur J Haematol 2017;98:393-7.

258. Lee HF, Chan YH, Chang SH, et al. Effectiveness and safety of nonvitamin K antagonist oral anticoagulant and warfarin in cirrhotic patients with nonvalvular atrial fibrillation. J Am Heart Assoc 2019;8:e011112.

259. Pastori D, Lip GYH, Farcomeni A, et al. Incidence of bleeding in patients with atrial fibrillation and advanced liver fibrosis on treatment with vitamin K or non-vitamin K antagonist oral anticoagulants. Int J Cardiol 2018;264:58-63.

260. Pugh RN, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg 1973;60:646-9.

261. Qamar A, Antman EM, Ruff CT, et al. Edoxaban versus warfarin in patients with atrial fibrillation and history of liver disease. J Am Coll Cardiol 2019;74:179-89.

262. Alonso A, MacLehose RF, Chen LY, et al. Prospective study of oral anticoagulants and risk of liver injury in patients with atrial fibrillation. Heart 2017;103:834-9.

263. Hu YF, Liu CJ, Chang PM, et al. Incident thromboembolism and heart failure associated with new-onset atrial fibrillation in cancer patients. Int J Cardiol 2013;165:355-7.

264. Farmakis D, Parissis J, Filippatos G. Insights into onco-cardiology: atrial fibrillation in cancer. J Am Coll Cardiol 2014;63:945-53.

265. Caine GJ, Stonelake PS, Lip GY, Kehoe ST. The hypercoagulable state of malignancy: pathogenesis and current debate. Neoplasia 2002;4:465-73.

266. Lenneman CG, Sawyer DB. Cardio-oncology: an update on cardiotoxicity of cancer-related treatment. Circ Res 2016;118:1008-20.

267. Melloni C, Dunning A, Granger CB, et al. Efficacy and safety of apixaban versus warfarin in patients with atrial fibrillation and a history of cancer: insights from the ARISTOTLE trial. Am J Med 2017;130:1440-1448.e1441.

268. Laube ES, Yu A, Gupta D, et al. Rivaroxaban for stroke prevention in patients with nonvalvular atrial fibrillation and active cancer. Am J Cardiol 2017;120:213-7.

269. Ianotto JC, Couturier MA, Galinat H, et al. Administration of direct oral anticoagulants in patients with myeloproliferative neoplasms. Int J Hematol 2017;106:517-21.

270. Ording AG, Horvath-Puho E, Adelborg K, et al. Thromboembolic and bleeding complications during oral anticoagulation therapy in cancer patients with atrial fibrillation: a Danish nationwide population-based cohort study. Cancer Med 2017;6:1165-72.

271. Flack KF, Desai J, Kolb JM, et al. Major gastrointestinal bleeding often is caused by occult malignancy in patients receiving warfarin or dabigatran to prevent stroke and systemic embolism from atrial fibrillation. Clin Gastroenterol Hepatol 2017;15:682-90.

272. Russo V, Rago A, Papa AA, et al. Use of non-vitamin K antagonist oral anticoagulants in atrial fibrillation patients with malignancy: clinical practice experience in a single institution and literature review. Semin Thromb Hemost 2018;44:370-6.

273. Chen ST, Hellkamp AS, Becker RC, et al. Efficacy and safety of rivaroxaban vs warfarin in patients with non-valvular atrial fibrillation and a history of cancer: observations from ROCKET AF. Eur Heart J Qual Care Clin Outcomes 2019;5:145-52.

274. Russo V, Bottino R, Rago A, et al. Atrial fibrillation and malignancy: the clinical performance of non-vitamin K oral anticoagulants-a systematic review. Semin Thromb Hemost 2019;45:205-14.

275. Lee AYY. Anticoagulant therapy for venous thromboembolism in cancer. N Engl J Med 2020;382:1650-2.

276. Hoffmann A, Chockalingam P, Balint OH, et al. Cerebrovascular accidents in adult patients with congenital heart disease. Heart 2010;96:1223-6.

277. Bouchardy J, Therrien J, Pilote L, et al. Atrial arrhythmias in adults with congenital heart disease. Circulation 2009;120:1679-86.

278. Khairy P. Arrhythmias in adults with congenital heart disease: what the practicing cardiologist needs to know. Can J Cardiol 2019;35:1698-707.

279. Heidendael JF, Bokma JP, de Groot JR, et al. Weighing the risks: thrombotic and bleeding events in adults with atrial arrhythmias and congenital heart disease. Int J Cardiol 2015;186:315-20.

280. Khairy P, Aboulhosn J, Broberg CS, et al. Thromboprophylaxis for atrial arrhythmias in congenital heart disease: a multicenter study. Int J Cardiol 2016;223:729-35.

281. Warnes CA, Williams RG, Bashore TM, et al. ACC/AHA 2008 guidelines for the management of adults with congenital heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (writing committee to develop guidelines on the management of adults with congenital heart disease). Circulation 2008;118:e714-833.

282. Khairy P, Van Hare GF, Balaji S, et al. PACES/HRS expert consensus statement on the recognition and management of arrhythmias in adult congenital heart disease: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Endorsed by the governing bodies of PACES, HRS, the American College of Cardiology (ACC), the American Heart Association (AHA), the European Heart Rhythm Association (EHRA), the Canadian Heart Rhythm Society (CHRS), and the International Society for Adult Congenital Heart Disease (ISACHD). Can J Cardiol 2014;30:e1-63.

283. Mongeon FP, Macle L, Beauchesne LM, et al. Non-vitamin K antagonist oral anticoagulants (NOACs) in adult congenital heart disease. Can J Cardiol 2019;35:1686-97.

284. Pujol C, Niesert AC, Engelhardt A, et al. Usefulness of direct oral anticoagulants in adult congenital heart disease. Am J Cardiol 2016;117:450-5.

285. Yang H, Bouma BJ, Dimopoulos K, et al. Non-vitamin K antagonist oral anticoagulants (NOACs) for thromboembolic prevention, are they safe in congenital heart disease? Results of a worldwide study. Int J Cardiol 2020;299:123-30.

286. Georgekutty J, Kazerouninia A, Wang Y, et al. Novel oral anticoagulant use in adult Fontan patients: a single center experience. Congenit Heart Dis 2018;13:541-7.

287. Pinto C, Samuel BP, Ratnasamy C, Vettukattil JJ. Thrombosis in Fontan patient on apixaban. Int J Cardiol 2015;182:66-7.

288. Lubitz SA, Yin X, Rienstra M, et al. Long-term outcomes of secondary atrial fibrillation in the community: the Framingham Heart Study. Circulation 2015;131:1648-55.

289. Quon MJ, Behlouli H, Pilote L. Anticoagulant use and risk of ischemic stroke and bleeding in patients with secondary atrial fibrillation associated with acute coronary syndromes, acute pulmonary disease, or sepsis. JACC Clin Electrophysiol 2018;4:386-93.

290. Walkey AJ, Quinn EK, Winter MR, McManus DD, Benjamin EJ. Practice patterns and outcomes associated with use of anticoagulation among patients with atrial fibrillation during sepsis. JAMA Cardiol 2016;1:682-90.

291. Sogaard M, Skjoth F, Kjaeldgaard JN, Lip GYH, Larsen TB. Bleeding complications in anticoagulated patients with atrial fibrillation and sepsis: a propensity-weighted cohort study. J Am Heart Assoc 2017;6:e007453.

292. Stuijver DJ, van Zaane B, Romualdi E, et al. The effect of hyperthyroidism on procoagulant, anticoagulant and fibrinolytic factors: a systematic review and meta-analysis. Thromb Haemost 2012;108:1077-88.

293. Presti CF, Hart RG. Thyrotoxicosis, atrial fibrillation, and embolism, revisited. Am Heart J 1989;117:976-7.

294. Petersen P, Hansen JM. Stroke in thyrotoxicosis with atrial fibrillation. Stroke 1988;19:15-8.

295. Chute JP, Ryan CP, Sladek G, Shakir KM. Exacerbation of warfarininduced anticoagulation by hyperthyroidism. Endocr Pract 1997;3:77-9.

296. Guttmann OP, Rahman MS, O’Mahony C, Anastasakis A, Elliott PM. Atrial fibrillation and thromboembolism in patients with hypertrophic cardiomyopathy: systematic review. Heart 2014;100:465-72.

297. Guttmann OP, Pavlou M, O’Mahony C, et al. Prediction of thromboembolic risk in patients with hypertrophic cardiomyopathy (HCM Risk-CVA). Eur J Heart Fail 2015;17:837-45.

298. Yang YJ, Yuan JQ, Fan CM, et al. Incidence of ischemic stroke and systemic embolism in patients with hypertrophic cardiomyopathy, nonvalvular atrial fibrillation, CHA2DS2-VASc score of 1 and without anticoagulant therapy. Heart Vessels 2016;31:1148-53.

299. Masri A, Kanj M, Thamilarasan M, et al. Outcomes in hypertrophic cardiomyopathy patients with and without atrial fibrillation: a survival meta-analysis. Cardiovasc Diagn Ther 2017;7:36-44.

300. Noubiap JJ, Bigna JJ, Agbor VN, et al. Meta-analysis of atrial fibrillation in patients with various cardiomyopathies. Am J Cardiol 2019;124:262-9.

301. Borer JS, Atar D, Marciniak T, Kim MH, Serebruany V. Atrial fibrillation and stroke in patients with hypertrophic cardiomyopathy: important new insights. Thromb Haemost 2019;119:355-7.

302. Guttmann OP. Atrial fibrillation and thromboembolism in hypertrophic cardiomyopathy – an underestimated risk. Int J Cardiol 2018;273:187-8.

303. Camm CF, Camm AJ. Atrial fibrillation and anticoagulation in hypertrophic cardiomyopathy. Arrhythm Electrophysiol Rev 2017;6:63-8.

304. Dominguez F, Climent V, Zorio E, et al. Direct oral anticoagulants in patients with hypertrophic cardiomyopathy and atrial fibrillation. Int J Cardiol 2017;248:232-8.

305. Jung H, Yang PS, Sung JH, et al. Hypertrophic cardiomyopathy in patients with atrial fibrillation: prevalence and associated stroke risks in a nationwide cohort study. Thromb Haemost 2019;119:285-93.

306. Lee HJ, Kim HK, Jung JH, et al. Novel oral anticoagulants for primary stroke prevention in hypertrophic cardiomyopathy patients with atrial fibrillation. Stroke 2019;50:2582-6.

307. Feng D, Syed IS, Martinez M, et al. Intracardiac thrombosis and anticoagulation therapy in cardiac amyloidosis. Circulation 2009;119:2490-7.

308. Feng D, Edwards WD, Oh JK, et al. Intracardiac thrombosis and embolism in patients with cardiac amyloidosis. Circulation 2007;116:2420-6.

309. El-Am EA, Dispenzieri A, Melduni RM, et al. Direct current cardioversion of atrial arrhythmias in adults with cardiac amyloidosis. J Am Coll Cardiol 2019;73:589-97.

310. Cheung CC, Roston TM, Andrade JG, Bennett MT, Davis MK. Arrhythmias in cardiac amyloidosis: challenges in risk stratification and treatment. Can J Cardiol 2020;36:416-23.

311. Mints YY, Doros G, Berk JL, Connors LH, Ruberg FL. Features of atrial fibrillation in wild-type transthyretin cardiac amyloidosis: a systematic review and clinical experience. ESC Heart Fail 2018;5:772-9.

312. Kamath S, Lip GY. Atrial fibrillation in the elderly: anticoagulation strategies and indications in the very elderly. Am J Geriatr Cardiol 2002;11:357-62. quiz 363-4.

313. Hylek EM, Evans-Molina C, Shea C, Henault LE, Regan S. Major hemorrhage and tolerability of warfarin in the first year of therapy among elderly patients with atrial fibrillation. Circulation 2007;115:2689-96.

314. Taggar JS, Lip GY. Anticoagulation for elderly patients with atrial fibrillation: not to be neglected. Europace 2008;10:1-2.

315. Xing YL, Ma Q, Ma XY, et al. Characteristics of non-valvular atrial fibrillation patients who benefit most from anticoagulation treatment. Int J Clin Exp Med 2015;8:18721-8.

316. Chao TF, Liu CJ, Lin YJ, et al. Oral anticoagulation in very elderly patients with atrial fibrillation: a nationwide cohort study. Circulation 2018;138:37-47.

317. Steinberg BA, Greiner MA, Hammill BG, et al. Contraindications to anticoagulation therapy and eligibility for novel anticoagulants in older patients with atrial fibrillation. Cardiovasc Ther 2015;33:177-83.

318. Ekerstad N, Karlsson T, Soderqvist S, Karlson BW. Hospitalized frail elderly patients – atrial fibrillation, anticoagulation and 12 months’ outcomes. Clin Interv Aging 2018;13:749-56.

319. Sanchez-Barba B, Navarrete-Reyes AP, Avila-Funes JA. Are geriatric syndromes associated with reluctance to initiate oral anticoagulation therapy in elderly adults with nonvalvular atrial fibrillation? J Am Geriatr Soc 2013;61:2236-7.

320. Inui TS, Parina R, Chang DC, Inui TS, Coimbra R. Mortality after ground-level fall in the elderly patient taking oral anticoagulation for atrial fibrillation/flutter: a long-term analysis of risk versus benefit. J Trauma Acute Care Surg 2014;76:642-9. discussion 649-50.

321. Wilke T, Groth A, Pfannkuche M, et al. Real life anticoagulation treatment of patients with atrial fibrillation in Germany: extent and causes of anticoagulant under-use. J Thromb Thrombolysis 2015;40:97-107.

322. Gumbinger C, Holstein T, Stock C, Rizos T, Horstmann S, Veltkamp R, et al. Reasons underlying non-adherence to and discontinuation of anticoagulation in secondary stroke prevention among patients with atrial fibrillation. Eur Neurol 2015;73:184-91.

323. Bahri O, Roca F, Lechani T, et al. Underuse of oral anticoagulation for individuals with atrial fibrillation in a nursing home setting in France: comparisons of resident characteristics and physician attitude. J Am Geriatr Soc 2015;63:71-6.

324. Palomaki A, Mustonen P, Hartikainen JE, et al. Underuse of anticoagulation in stroke patients with atrial fibrillation–the FibStroke study. Eur J Neurol 2016;23:133-9.

325. Rao MP, Vinereanu D, Wojdyla DM, et al. Clinical outcomes and history of fall in patients with atrial fibrillation treated with oral anticoagulation: insights from the ARISTOTLE trial. Am J Med 2018;131:269-275.e262.

326. Pappas MA, Vijan S, Rothberg MB, Singer DE. Reducing age bias in decision analyses of anticoagulation for patients with nonvalvular atrial fibrillation – a microsimulation study. PLoS One 2018;13:e0199593.

327. Oqab Z, Pournazari P, Sheldon RS. What is the impact of frailty on prescription of anticoagulation in elderly patients with atrial fibrillation? A systematic review and meta-analysis. J Atr Fibrillation 2018;10:1870.

328. Patti G, Pecen L, Lucerna M, et al. Net clinical benefit of non-vitamin K antagonist vs vitamin K antagonist anticoagulants in elderly patients with atrial fibrillation. Am J Med 2019;132:749-757.e745.

329. Okumura K, Akao M, Yoshida T, et al. Low-dose edoxaban in very elderly patients with atrial fibrillation. N Engl J Med 2020;383:1735-45.

330. Eikelboom JW, Wallentin L, Connolly SJ, et al. Risk of bleeding with 2 doses of dabigatran compared with warfarin in older and younger patients with atrial fibrillation: an analysis of the randomized evaluation of long-term anticoagulant therapy (RE-LY) trial. Circulation 2011;123:2363-72.

331. Halperin JL, Hankey GJ, Wojdyla DM, et al. Efficacy and safety of rivaroxaban compared with warfarin among elderly patients with nonvalvular atrial fibrillation in the 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). Circulation 2014;130:138-46.

332. Halvorsen S, Atar D, Yang H, et al. Efficacy and safety of apixaban compared with warfarin according to age for stroke prevention in atrial fibrillation: observations from the ARISTOTLE trial. Eur Heart J 2014;35:1864-72.

333. Kato ET, Giugliano RP, Ruff CT, et al. Efficacy and safety of edoxaban in elderly patients with atrial fibrillation in the ENGAGE AF-TIMI 48 trial. J Am Heart Assoc 2016;5:e003432.

334. Sardar P, Chatterjee S, Chaudhari S, Lip GY. New oral anticoagulants in elderly adults: evidence from a meta-analysis of randomized trials. J Am Geriatr Soc 2014;62:857-64.

335. Boriani G, Ruff CT, Kuder JF, et al. Relationship between body mass index and outcomes in patients with atrial fibrillation treated with edoxaban or warfarin in the ENGAGE AF-TIMI 48 trial. Eur Heart J 2019;40:1541-50.

336. Balla SR, Cyr DD, Lokhnygina Y, et al. Relation of risk of stroke in patients with atrial fibrillation to body mass index (from Patients Treated With Rivaroxaban and Warfarin in the Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared with Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation Trial). Am J Cardiol 2017;119:1989-96.

337. Hohnloser SH, Fudim M, Alexander JH, et al. Efficacy and safety of apixaban versus warfarin in patients with atrial fibrillation and extremes in body weight. Circulation 2019;139:2292-300.

338. Malik AH, Yandrapalli S, Shetty S, et al. Impact of weight on the efficacy and safety of direct-acting oral anticoagulants in patients with non-valvular atrial fibrillation: a meta-analysis. Europace 2020;22:361-7.

339. Andrade JG, Mitchell LB. Periprocedural anticoagulation for cardioversion of acute onset atrial fibrillation and flutter: evidence base for current guidelines. Can J Cardiol 2019;35:1301-10.

340. Roy D, Marchand E, Gagne P, Chabot M, Cartier R. Usefulness of anticoagulant therapy in the prevention of embolic complications of atrial fibrillation. Am Heart J 1986;112:1039-43.

341. Manning WJ, Leeman DE, Gotch PJ, Come PC. Pulsed Doppler evaluation of atrial mechanical function after electrical cardioversion of atrial fibrillation. J Am Coll Cardiol 1989;13:617-23.

342. Grimm RA, Stewart WJ, Maloney JD, et al. Impact of electrical cardioversion for atrial fibrillation on left atrial appendage function and spontaneous echo contrast: characterization by simultaneous transesophageal echocardiography. J Am Coll Cardiol 1993;22:1359-66.

343. Irani WN, Grayburn PA, Afridi I. Prevalence of thrombus, spontaneous echo contrast, and atrial stunning in patients undergoing cardioversion of atrial flutter. A prospective study using transesophageal echocardiography. Circulation 1997;95:962-6.

344. Manning WJ, Silverman DI, Katz SE, et al. Impaired left atrial mechanical function after cardioversion: relation to the duration of atrial fibrillation. J Am Coll Cardiol 1994;23:1535-40.

345. Fatkin D, Kuchar DL, Thorburn CW, Feneley MP. Transesophageal echocardiography before and during direct current cardioversion of atrial fibrillation: evidence for “atrial stunning” as a mechanism of thromboembolic complications. J Am Coll Cardiol 1994;23:307-16.

346. Mattioli AV, Tarabini Castellani E, et al. Restoration of atrial function after atrial fibrillation of different etiological origins. Cardiology 1996;87:205-11.

347. Sparks PB, Jayaprakash S, Mond HG, et al. Left atrial mechanical function after brief duration atrial fibrillation. J Am Coll Cardiol 1999;33:342-9.

348. McIntyre WF, Connolly SJ, Wang J, et al. Thromboembolic events around the time of cardioversion for atrial fibrillation in patients receiving antiplatelet treatment in the ACTIVE trials. Eur Heart J 2019;40:3026-32.

349. Klein AL, Grimm RA, Murray RD, et al. Use of transesophageal echocardiography to guide cardioversion in patients with atrial fibrillation. N Engl J Med 2001;344:1411-20.

350. Zhan Y, Joza J, Al Rawahi M, et al. Assessment and management of the left atrial appendage thrombus in patients with nonvalvular atrial fibrillation. Can J Cardiol 2018;34:252-61.

351. Cairns JA, Connolly S, McMurtry S, Stephenson M, Talajic M. Canadian Cardiovascular Society atrial fibrillation guidelines 2010: prevention of stroke and systemic thromboembolism in atrial fibrillation and flutter. Can J Cardiol 2011;27:74-90.

352. Kleemann T, Becker T, Strauss M, Schneider S, Seidl K. Prevalence of left atrial thrombus and dense spontaneous echo contrast in patients with short-term atrial fibrillation < 48 hours undergoing cardioversion: value of transesophageal echocardiography to guide cardioversion. J Am Soc Echocardiogr 2009;22:1403-8.

353. Airaksinen KE, Gronberg T, Nuotio I, et al. Thromboembolic complications after cardioversion of acute atrial fibrillation: the FinCV (Finnish CardioVersion) study. J Am Coll Cardiol 2013;62:1187-92.

354. Nuotio I, Hartikainen JE, Gronberg T, Biancari F, Airaksinen KE. Time to cardioversion for acute atrial fibrillation and thromboembolic complications. JAMA 2014;312:647-9.

355. Gronberg T, Hartikainen JE, Nuotio I, et al. Anticoagulation, CHA2DS2VASc score, and thromboembolic risk of cardioversion of acute atrial fibrillation (from the FinCV study). Am J Cardiol 2016;117:1294-8.

356. Bah A, Nuotio I, Gronberg T, et al. Sex, age, and time to cardioversion. Risk factors for cardioversion of acute atrial fibrillation from the FinCV study. Ann Med 2017;49:254-9.

357. Hellman T, Kiviniemi T, Nuotio I, et al. Intensity of anticoagulation and risk of thromboembolism after elective cardioversion of atrial fibrillation. Thromb Res 2017;156:163-7.

358. Hellman T, Kiviniemi T, Nuotio I, et al. Optimal timing for cardioversion in patients with atrial fibrillation. Clin Cardiol 2018;41:966-71.

359. Garg A, Khunger M, Seicean S, Chung MK, Tchou P. Incidence of thromboembolic complications within 30 days of electrical cardioversion performed within 48 hours of atrial fibrillation onset. JACC Clin Electrophysiol 2016;2:487-94.

360. Palomaki A, Mustonen P, Hartikainen JE, et al. Strokes after cardioversion of atrial fibrillation–the FibStroke study. Int J Cardiol 2016;203:269-73.

361. Hansen ML, Jepsen RM, Olesen JB, et al. Thromboembolic risk in 16 274 atrial fibrillation patients undergoing direct current cardioversion with and without oral anticoagulant therapy. Europace 2015;17:18-23.

362. Sjalander S, Svensson PJ, Friberg L. Atrial fibrillation patients with CHA2DS2-VASc >1 benefit from oral anticoagulation prior to cardioversion. Int J Cardiol 2016;215:360-3.

363. Crijns HJ, Weijs B, Fairley AM, et al. Contemporary real life cardioversion of atrial fibrillation: results from the multinational RHYTHMAF study. Int J Cardiol 2014;172:588-94.

364. Atzema CL, Jackevicius CA, Chong A, et al. Prescribing of oral anticoagulants in the emergency department and subsequent long-term use by older adults with atrial fibrillation. CMAJ 2019;191:E1345-54.

365. Cappato R, Ezekowitz MD, Klein AL, et al. Rivaroxaban vs vitamin K antagonists for cardioversion in atrial fibrillation. Eur Heart J 2014;35:3346-55.

366. Goette A, Merino JL, Ezekowitz MD, et al. Edoxaban versus enoxaparin-warfarin in patients undergoing cardioversion of atrial fibrillation (ENSURE-AF): a randomised, open-label, phase 3b trial. Lancet 2016;388:1995-2003.

367. Ezekowitz MD, Pollack CV Jr, Halperin JL, et al. Apixaban compared to heparin/vitamin K antagonist in patients with atrial fibrillation scheduled for cardioversion: the EMANATE trial. Eur Heart J 2018;39:2959-71.

368. Kotecha D, Pollack CV Jr, De Caterina R, Renda G, Kirchhof P. Direct oral anticoagulants halve thromboembolic events after cardioversion of AF compared with warfarin. J Am Coll Cardiol 2018;72:1984-6.

369. Gupta S, Um KJ, Pandey A, et al. Direct oral anticoagulants versus vitamin k antagonists in patients undergoing cardioversion for atrial fibrillation: a systematic review and meta-analysis. Cardiovasc Drugs Ther 2019;33:339-52.

370. Nagarakanti R, Ezekowitz MD, Oldgren J, et al. Dabigatran versus warfarin in patients with atrial fibrillation: an analysis of patients undergoing cardioversion. Circulation 2011;123:131-6.

371. Piccini JP, Stevens SR, Lokhnygina Y, et al. Outcomes after cardioversion and atrial fibrillation ablation in patients treated with rivaroxaban and warfarin in the ROCKET AF trial. J Am Coll Cardiol 2013;61:1998-2006.

372. Flaker G, Lopes RD, Al-Khatib SM, et al. Efficacy and safety of apixaban in patients after cardioversion for atrial fibrillation: insights from the ARISTOTLE Trial (Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation). J Am Coll Cardiol 2014;63:1082-7.

373. Plitt A, Ezekowitz MD, De Caterina R, et al. Cardioversion of atrial fibrillation in ENGAGE AF-TIMI 48. Clin Cardiol 2016;39:345-6.

374. Telles-Garcia N, Dahal K, Kocherla C, et al. Non-vitamin K antagonists oral anticoagulants are as safe and effective as warfarin for cardioversion of atrial fibrillation: a systematic review and meta-analysis. Int J Cardiol 2018;268:143-8.

375. Brunetti ND, Tarantino N, De Gennaro L, et al. Direct oral anticoagulants compared to vitamin-K antagonists in cardioversion of atrial fibrillation: an updated meta-analysis. J Thromb Thrombolysis 2018;45:550-6.

376. Di Biase L, Burkhardt JD, Santangeli P, et al. Periprocedural stroke and bleeding complications in patients undergoing catheter ablation of atrial fibrillation with different anticoagulation management: results from the Role of Coumadin in Preventing Thromboembolism in Atrial Fibrillation (AF) Patients Undergoing Catheter Ablation (COMPARE) randomized trial. Circulation 2014;129:2638-44.

377. Cappato R, Marchlinski FE, Hohnloser SH, et al. Uninterrupted rivaroxaban vs uninterrupted vitamin K antagonists for catheter ablation in non-valvular atrial fibrillation. Eur Heart J 2015;36:1805-11.

378. Calkins H, Willems S, Gerstenfeld EP, et al. Uninterrupted dabigatran versus warfarin for ablation in atrial fibrillation. N Engl J Med 2017;376:1627-36.

379. Kirchhof P, Haeusler KG, Blank B, et al. Apixaban in patients at risk of stroke undergoing atrial fibrillation ablation. Eur Heart J 2018;39:2942-55.

380. Reynolds MR, Allison JS, Natale A, et al. A prospective randomized trial of apixaban dosing during atrial fibrillation ablation: the AEIOU trial. JACC Clin Electrophysiol 2018;4:580-8.

381. Hohnloser SH, Camm J, Cappato R, et al. Uninterrupted edoxaban vs vitamin K antagonists for ablation of atrial fibrillation: the ELIMINATE-AF trial. Eur Heart J 2019;40:3013-21.

382. Nogami A, Harada T, Sekiguchi Y, et al. Safety and efficacy of minimally interrupted dabigatran vs uninterrupted warfarin therapy in adults undergoing atrial fibrillation catheter ablation: a randomized clinical trial. JAMA Netw Open 2019;2:e191994.

383. Nakamura K, Naito S, Sasaki T, et al. Uninterrupted vs interrupted periprocedural direct oral anticoagulants for catheter ablation of atrial fibrillation: a prospective randomized single-centre study on postablation thrombo-embolic and haemorrhagic events. Europace 2019;21:259-67.

384. Larbig R, Dittrich R, Kochhaeuser S, et al. Influence of clinical parameters and anticoagulation on intraprocedural cerebral microembolic signals during pulmonary vein isolation. PLoS One 2016;11:e0157886.

385. Martin AC, Godier A, Narayanan K, Smadja DM, Marijon E. Management of intraprocedural anticoagulation in patients on non-vitamin K antagonist oral anticoagulants undergoing catheter ablation for atrial fibrillation: understanding the gaps in evidence. Circulation 2018;138:627-33.

386. Wazni OM, Rossillo A, Marrouche NF, et al. Embolic events and char formation during pulmonary vein isolation in patients with atrial fibrillation: impact of different anticoagulation regimens and importance of intracardiac echo imaging. J Cardiovasc Electrophysiol 2005;16:576-81.

387. Oral H, Chugh A, Ozaydin M, et al. Risk of thromboembolic events after percutaneous left atrial radiofrequency ablation of atrial fibrillation. Circulation 2006;114:759-65.

388. Themistoclakis S, Corrado A, Marchlinski FE, et al. The risk of thromboembolism and need for oral anticoagulation after successful atrial fibrillation ablation. J Am Coll Cardiol 2010;55:735-43.

389. Hunter RJ, McCready J, Diab I, et al. Maintenance of sinus rhythm with an ablation strategy in patients with atrial fibrillation is associated with a lower risk of stroke and death. Heart 2012;98:48-53.

390. Bunch TJ, May HT, Bair TL, et al. Atrial fibrillation ablation patients have long-term stroke rates similar to patients without atrial fibrillation regardless of CHADS2 score. Heart Rhythm 2013;10:1272-7.

391. Saliba W, Schliamser JE, Lavi I, et al. Catheter ablation of atrial fibrillation is associated with reduced risk of stroke and mortality: a propensity score-matched analysis. Heart Rhythm 2017;14:635-42.

392. Srivatsa UN, Danielsen B, Amsterdam EA, et al. CAABL-AF (California Study of Ablation for Atrial Fibrillation): mortality and stroke, 2005 to Circ Arrhythm Electrophysiol 2018;11:e005739.

393. Joza J, Samuel M, Jackevicius CA, et al. Long-term risk of stroke and bleeding post-atrial fibrillation ablation. J Cardiovasc Electrophysiol 2018;29:1355-62.

394. Packer DL, Mark DB, Robb RA, et al. Effect of catheter ablation vs antiarrhythmic drug therapy on mortality, stroke, bleeding, and cardiac arrest among patients with atrial fibrillation: the CABANA randomized clinical trial. JAMA 2019;321:1261-74.

395. Ganesan AN, Shipp NJ, Brooks AG, et al. Long-term outcomes of catheter ablation of atrial fibrillation: a systematic review and meta analysis. J Am Heart Assoc 2013;2:e004549.

396. Verma A, Ha ACT, Kirchhof P, et al. The Optimal Anti-Coagulation for Enhanced-Risk Patients Post-Catheter Ablation for Atrial Fibrillation (OCEAN) trial. Am Heart J 2018;197:124-32.

397. Michael KA, Redfearn DP, Baranchuk A, et al. Transesophageal echocardiography for the prevention of embolic complications after catheter ablation for atrial fibrillation. J Cardiovasc Electrophysiol 2009;20:1217-22.

398. Voight J, Akkaya M, Somasundaram P, et al. Risk of new-onset atrial fibrillation and stroke after radiofrequency ablation of isolated, typical atrial flutter. Heart Rhythm 2014;11:1884-9.

399. Exposito V, Rodriguez-Entem F, Gonzalez-Enriquez S, et al. Stroke and systemic embolism after successful ablation of typical atrial flutter. Clin Cardiol 2016;39:347-51.

400. Perez FJ, Schubert CM, Parvez B, et al. Long-term outcomes after catheter ablation of cavo-tricuspid isthmus dependent atrial flutter: a meta-analysis. Circ Arrhythm Electrophysiol 2009;2:393-401.

401. Spyropoulos AC, Al-Badri A, Sherwood MW, Douketis JD. Periprocedural management of patients receiving a vitamin K antagonist or a direct oral anticoagulant requiring an elective procedure or surgery. J Thromb Haemost 2016;14:875-85.

402. Dunn AS, Spyropoulos AC, Turpie AG. Bridging therapy in patients on long-term oral anticoagulants who require surgery: the Prospective Perioperative Enoxaparin Cohort Trial (PROSPECT). J Thromb Haemost 2007;5:2211-8.

403. Siegal D, Yudin J, Kaatz S, et al. Periprocedural heparin bridging in patients receiving vitamin K antagonists: systematic review and meta analysis of bleeding and thromboembolic rates. Circulation 2012;126:1630-9.

404. Douketis JD, Spyropoulos AC, Kaatz S, et al. Perioperative bridging anticoagulation in patients with atrial fibrillation. N Engl J Med 2015;373:823-33.

405. Spyropoulos AC, Douketis JD. How I treat anticoagulated patients undergoing an elective procedure or surgery. Blood 2012;120:2954-62.

406. Beyer-Westendorf J, Gelbricht V, Forster K, et al. Peri-interventional management of novel oral anticoagulants in daily care: results from the prospective Dresden NOAC registry. Eur Heart J 2014;35:1888-96.

407. Healey JS, Eikelboom J, Douketis J, et al. Periprocedural bleeding and thromboembolic events with dabigatran compared with warfarin: results from the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) randomized trial. Circulation 2012;126:343-8.

408. Sherwood MW, Douketis JD, Patel MR, et al. Outcomes of temporary interruption of rivaroxaban compared with warfarin in patients with nonvalvular atrial fibrillation: results from the 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). Circulation 2014;129:1850-9.

409. Garcia D, Alexander JH, Wallentin L, et al. Management and clinical outcomes in patients treated with apixaban vs warfarin undergoing procedures. Blood 2014;124:3692-8.

410. Douketis JD, Spyropoulos AC, Duncan J, et al. Perioperative management of patients with atrial fibrillation receiving a direct oral anticoagulant. JAMA Intern Med 2019;33:551-61.

411. Esteve-Pastor MA, Rivera-Caravaca JM, Shantsila A, et al. Assessing bleeding risk in atrial fibrillation patients: comparing a bleeding risk score based only on modifiable bleeding risk factors against the HASBLED score. The AMADEUS trial. Thromb Haemost 2017;117:2261-6.

412. Chao TF, Lip GYH, Lin YJ, et al. Major bleeding and intracranial hemorrhage risk prediction in patients with atrial fibrillation: attention to modifiable bleeding risk factors or use of a bleeding risk stratification score? A nationwide cohort study. Int J Cardiol 2018;254:157-61.

413. Guo Y, Zhu H, Chen Y, Lip GYH. Comparing bleeding risk assessment focused on modifiable risk factors only versus validated bleeding risk scores in atrial fibrillation. Am J Med 2018;131:185-92.

414. Lai KC, Lam SK, Chu KM, et al. Lansoprazole for the prevention of recurrences of ulcer complications from long-term low-dose aspirin use. N Engl J Med 2002;346:2033-8.

415. Yeomans N, Lanas A, Labenz J, et al. Efficacy of esomeprazole (20 mg once daily) for reducing the risk of gastroduodenal ulcers associated with continuous use of low-dose aspirin. Am J Gastroenterol 2008;103:2465-73.

416. Bhatt DL, Cryer BL, Contant CF, et al. Clopidogrel with or without omeprazole in coronary artery disease. N Engl J Med 2010;363:1909-17.

417. Scheiman JM, Devereaux PJ, Herlitz J, et al. Prevention of peptic ulcers with esomeprazole in patients at risk of ulcer development treated with low-dose acetylsalicylic acid: a randomised, controlled trial (OBERON). Heart 2011;97:797-802.

418. Ray WA, Chung CP, Murray KT, et al. Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding. JAMA 2018;320:2221-30.

419. Moayyedi P, Eikelboom JW, Bosch J, et al. Pantoprazole to prevent gastroduodenal events in patients receiving rivaroxaban and/or aspirin in a randomized, double-blind, placebo-controlled trial. Gastroenterology 2019;157. 403-12.e405.

420. Causada-Calo N, Germini F, Yuan Y, Eikelboom JW, Moayyedi P. Proton-pump inhibitors for the prevention of upper gastrointestinal bleeding in adults receiving antithrombotic therapy. Cochrane Database Syst Rev 2019;8:CD013415. Available at: https://doi.org/10.1002/14651858.CD013415.

421. Eikelboom JW, Quinlan DJ, van Ryn J, Weitz JI. Idarucizumab: the antidote for reversal of dabigatran. Circulation 2015;132:2412-22.

422. Glund S, Stangier J, Schmohl M, et al. Idarucizumab, a specific antidote for dabigatran: immediate, complete and sustained reversal of dabigatran induced anticoagulation in elderly and renally impaired subjects [abstract 332]. Blood 2014;124:344.

423. Glund S, Moschetti V, Norris S, et al. A randomised study in healthy volunteers to investigate the safety, tolerability and pharmacokinetics of idarucizumab, a specific antidote to dabigatran. Thromb Haemost 2015;113:943-51.

424. Glund S, Stangier J, Schmohl M, et al. Safety, tolerability, and efficacy of idarucizumab for the reversal of the anticoagulant effect of dabigatran in healthy male volunteers: a randomised, placebo-controlled, doubleblind phase 1 trial. Lancet 2015;386:680-90.

425. Pollack CV Jr, Reilly PA, Weitz JI. Dabigatran reversal with idarucizumab. N Engl J Med 2017;377:1690-2.

426. Yasaka M, Yokota H, Suzuki M, et al. Idarucizumab for emergency reversal of anticoagulant effects of dabigatran: interim results of a japanese post-marketing surveillance study. Cardiol Ther 2020;9:167-88.

427. van der Wall SJ, van Rein N, van den Bemt B, et al. Performance of idarucizumab as antidote of dabigatran in daily clinical practice. Europace 2019;21:414-20.

428. Siegal DM, Curnutte JT, Connolly SJ, et al. Andexanet alfa for the reversal of Factor Xa inhibitor activity. N Engl J Med 2015;373:2413-24.

429. Connolly SJ, Crowther M, Eikelboom JW, et al. Full study report of andexanet alfa for bleeding associated with Factor Xa inhibitors. N Engl J Med 2019;380:1326-35.

430. FDA. US Food and Drug Administration. May 3, 2018 Accellerated Approval Letter – ANDEXXA. Available at: https://www.fda.gov/media/113285/download. Accessed November 11, 2020.

431. Kaatz S, Bhansali H, Gibbs J, et al. Reversing factor Xa inhibitors – clinical utility of andexanet alfa. J Blood Med 2017;8:141-9.

432. Stevens VM, Trujillo T, Mueller SW, et al. Coagulation Factor Xa (recombinant), inactivated-zhzo (andexanet alfa) hemostatic outcomes and thrombotic event incidence at an academic medical center. Clin Appl Thromb Hemost 2019;25. 1076029619896619.

433. Culbreth SE, Rimsans J, Sylvester K, Pallin DJ, Connors JM. Andexanet alfa-the first 150 days. Am J Hematol 2019;94:E21-4.

434. Hylek EM, Go AS, Chang Y, et al. Effect of intensity of oral anticoagulation on stroke severity and mortality in atrial fibrillation. N Engl J Med 2003;349:1019-26.

435. O’Donnell M, Oczkowski W, Fang J, et al. Preadmission antithrombotic treatment and stroke severity in patients with atrial fibrillation and acute ischaemic stroke: an observational study. Lancet Neurol 2006;5:749-54.

436. Macha K, Marsch A, Siedler G, et al. Cerebral ischemia in patients on direct oral anticoagulants. Stroke 2019;50:873-9.

437. Prabhakaran S, Silver AJ, Warrior L, McClenathan B, Lee VH. Misdiagnosis of transient ischemic attacks in the emergency room. Cerebrovasc Dis 2008;26:630-5.

438. Brazzelli M, Shuler K, Quayyum Z, et al. Clinical and imaging services for TIA and minor stroke: results of two surveys of practice across the UK. BMJ Open 2013;3:e003359.

439. Cooperman SS, Shah AK, Rajamani K. Amyloid spells. Neurol Clin Pract 2019;9:e17-8.

440. Hart RG, Coull BM, Hart D. Early recurrent embolism associated with nonvalvular atrial fibrillation: a retrospective study. Stroke 1983;14:688-93.

441. Paciaroni M, Agnelli G, Micheli S, Caso V. Efficacy and safety of anticoagulant treatment in acute cardioembolic stroke: a meta-analysis of randomized controlled trials. Stroke 2007;38:423-30.

442. Paciaroni M, Agnelli G, Corea F, et al. Early hemorrhagic transformation of brain infarction: rate, predictive factors, and influence on clinical outcome: results of a prospective multicenter study. Stroke 2008;39:2249-56.

443. D’Amelio M, Terruso V, Famoso G, et al. Early and late mortality of spontaneous hemorrhagic transformation of ischemic stroke. J Stroke Cerebrovasc Dis 2014;23:649-54.

444. Li YG, Lip GYH. Anticoagulation resumption after intracerebral hemorrhage. Curr Atheroscler Rep 2018;20:32.

445. Paciaroni M, Agnelli G, Falocci N, et al. Early recurrence and cerebral bleeding in patients with acute ischemic stroke and atrial fibrillation: effect of anticoagulation and its timing: the RAF study. Stroke 2015;46:2175-82.

446. Wilson D, Ambler G, Banerjee G, et al. Early versus late anticoagulation for ischaemic stroke associated with atrial fibrillation: multicentre cohort study. J Neurol Neurosurg Psychiatry 2019;90:320-5.

447. Hong KS, Kwon SU, Lee SH, et al. Rivaroxaban vs warfarin sodium in the ultra-early period after atrial fibrillation-related mild ischemic stroke: a randomized clinical trial. JAMA Neurol 2017;74:1206-15.

448. Butcher KS, Ng K, Sheridan P, et al. Dabigatran treatment of acute noncardioembolic ischemic stroke. Stroke 2020;51:1190-8.

449. Palm F, Kraus M, Safer A, et al. Management of oral anticoagulation after cardioembolic stroke and stroke survival data from a population based stroke registry (LuSSt). BMC Neurol 2014;14:199.

450. Paciaroni M, Bandini F, Agnelli G, et al. Hemorrhagic transformation in patients with acute ischemic stroke and atrial fibrillation: time to initiation of oral anticoagulant therapy and outcomes. J Am Heart Assoc 2018;7:e010133.

451. Murthy SB, Gupta A, Merkler AE, et al. Restarting anticoagulant therapy after intracranial hemorrhage: a systematic review and meta-analysis. Stroke 2017;48:1594-600.

452. Xu Y, Shoamanesh A, Schulman S, et al. Oral anticoagulant re-initiation following intracerebral hemorrhage in non-valvular atrial fibrillation: global survey of the practices of neurologists, neurosurgeons and thrombosis experts. PLoS One 2018;13:e0191137.

453. Stroke Prevention in Atrial Fibrillation Study. Final results. Circulation 1991;84:527-39.

454. Blackshear JL, Odell JA. Appendage obliteration to reduce stroke in cardiac surgical patients with atrial fibrillation. Ann Thorac Surg 1996;61:755-9.

455. Holmes DR, Reddy VY, Turi ZG, et al. Percutaneous closure of the left atrial appendage versus warfarin therapy for prevention of stroke in patients with atrial fibrillation: a randomised non-inferiority trial. Lancet 2009;374:534-42.

456. Holmes DR Jr, Kar S, Price MJ, et al. Prospective randomized evaluation of the Watchman Left Atrial Appendage Closure device in patients with atrial fibrillation versus long-term warfarin therapy: the PREVAIL trial. J Am Coll Cardiol 2014;64:1-12.

457. Reddy VY, Doshi SK, Kar S, et al. 5-Year outcomes after left atrial appendage closure: from the PREVAIL and PROTECT AF trials. J Am Coll Cardiol 2017;70:2964-75.

458. Glikson M, Wolff R, Hindricks G, et al. EHRA/EAPCI expert consensus statement on catheter-based left atrial appendage occlusion – an update. EuroIntervention 2020;15:1133-80.

459. Toale C, Fitzmaurice GJ, Eaton D, Lyne J, Redmond KC. Outcomes of left atrial appendage occlusion using the AtriClip device: a systematic review. Interact Cardiovasc Thorac Surg 2019;29:655-62.

460. Almahameed ST, Khan M, Zuzek RW, et al. Left atrial appendage exclusion and the risk of thromboembolic events following mitral valve surgery. J Cardiovasc Electrophysiol 2007;18:364-6.

461. Yao X, Gersh BJ, Holmes DR Jr, et al. Association of surgical left atrial appendage occlusion with subsequent stroke and mortality among patients undergoing cardiac surgery. JAMA 2018;319:2116-26.

462. Whitlock R, Healey J, Vincent J, et al. Rationale and design of the Left Atrial Appendage Occlusion Study (LAAOS) III. Ann Cardiothorac Surg 2014;3:45-54.

463. Ratcliffe PJ, Wilcock GK. Cerebrovascular disease in dementia: the importance of atrial fibrillation. Postgrad Med J 1985;61:201-4.

464. De Pedis G, Hedner K, Johansson BW, Steen B. Cardiac arrhythmia in geriatric patients with organic dementia. Compr Gerontol A 1987;1:115-7.

465. Mead GE, Keir S. Association between cognitive impairment and atrial fibrillation: a systematic review. J Stroke Cerebrovasc Dis 2001;10:35-43.

466. Udompanich S, Lip GY, Apostolakis S, Lane DA. Atrial fibrillation as a risk factor for cognitive impairment: a semi-systematic review. QJM 2013;106:795-802.

467. Santangeli P, Di Biase L, Bai R, et al. Atrial fibrillation and the risk of incident dementia: a meta-analysis. Heart Rhythm 2012;9:1761-8.

468. Kalantarian S, Stern TA, Mansour M, Ruskin JN. Cognitive impairment associated with atrial fibrillation: a meta-analysis. Ann Intern Med 2013;158:338-46.

469. Saglietto A, Matta M, Gaita F, et al. Stroke-independent contribution of atrial fibrillation to dementia: a meta-analysis. Open Heart 2019;6:e000984.

470. Liao JN, Chao TF, Liu CJ, et al. Risk and prediction of dementia in patients with atrial fibrillation–a nationwide population-based cohort study. Int J Cardiol 2015;199:25-30.

471. Alonso A, Arenas de Larriva AP. Atrial fibrillation, cognitive decline and dementia. Eur Cardiol 2016;11:49-53.

472. Kim D, Yang PS, Yu HT, et al. Risk of dementia in stroke-free patients diagnosed with atrial fibrillation: data from a population-based cohort. Eur Heart J 2019;40:2313-23.

473. Dublin S, Anderson ML, Haneuse SJ, et al. Atrial fibrillation and risk of dementia: a prospective cohort study. J Am Geriatr Soc 2011;59:1369-75.

474. Ihara M, Washida K. Linking atrial fibrillation with Alzheimer’s disease: epidemiological, pathological, and mechanistic evidence. J Alzheimers Dis 2018;62:61-72.

475. Bunch TJ, Weiss JP, Crandall BG, et al. Atrial fibrillation is independently associated with senile, vascular, and Alzheimer’s dementia. Heart Rhythm 2010;7:433-7.

476. Ott A, Breteler MM, de Bruyne MC, et al. Atrial fibrillation and dementia in a population-based study. The Rotterdam Study. Stroke 1997;28:316-21.

477. de Bruijn RF, Heeringa J, Wolters FJ, et al. Association between atrial fibrillation and dementia in the general population. JAMA Neurol 2015;72:1288-94.

478. Chen LY, Agarwal SK, Norby FL, et al. Persistent but not paroxysmal atrial fibrillation is independently associated with lower cognitive function: ARIC study. J Am Coll Cardiol 2016;67:1379-80.

479. Cacciatore F, Testa G, Langellotto A, et al. Role of ventricular rate response on dementia in cognitively impaired elderly subjects with atrial fibrillation: a 10-year study. Dement Geriatr Cogn Disord 2012;34:143-8.

480. Wang S, Fashanu OE, Zhao D, et al. Relation of elevated resting heart rate in mid-life to cognitive decline over 20 years (from the Atherosclerosis Risk in Communities [ARIC] study). Am J Cardiol 2019;123:334-40.

481. Marzona I, O’Donnell M, Teo K, et al. Increased risk of cognitive and functional decline in patients with atrial fibrillation: results of the ONTARGET and TRANSCEND studies. CMAJ 2012;184:E329-36.

482. Poggesi A, Inzitari D, Pantoni L. Atrial fibrillation and cognition: epidemiological data and possible mechanisms. Stroke 2015;46:3316-21.

483. Scarsoglio S, Saglietto A, Anselmino M, Gaita F, Ridolfi L. Alteration of cerebrovascular haemodynamic patterns due to atrial fibrillation: an in silico investigation. J R Soc Interface 2017;14:20170180.

484. Bunch TJ, Galenko O, Graves KG, Jacobs V, May HT. Atrial fibrillation and dementia: exploring the association, defining risks and improving outcomes. Arrhythm Electrophysiol Rev 2019;8:8-12.

485. Diener HC, Hart RG, Koudstaal PJ, Lane DA, Lip GYH. Atrial fibrillation and cognitive function: JACC review topic of the week. J Am Coll Cardiol 2019;73:612-9.

486. de Leeuw FE, de Groot JC, Oudkerk M, et al. Atrial fibrillation and the risk of cerebral white matter lesions. Neurology 2000;54:1795-801.

487. Shao IY, Power MC, Mosley T, et al. Association of atrial fibrillation with white matter disease. Stroke 2019;50:989-91.

488. Wandell P, Carlsson AC, Sundquist J, Sundquist K. Antihypertensive drugs and relevant cardiovascular pharmacotherapies and the risk of incident dementia in patients with atrial fibrillation. Int J Cardiol 2018;272:149-54.

489. Bunch TJ, Crandall BG, Weiss JP, et al. Patients treated with catheter ablation for atrial fibrillation have long-term rates of death, stroke, and dementia similar to patients without atrial fibrillation. J Cardiovasc Electrophysiol 2011;22:839-45.

490. Cheng W, Liu W, Li B, Li D. Relationship of anticoagulant therapy with cognitive impairment among patients with atrial fibrillation: a meta-analysis and systematic review. J Cardiovasc Pharmacol 2018;71:380-7.

491. Friberg L, Rosenqvist M. Less dementia with oral anticoagulation in atrial fibrillation. Eur Heart J 2018;39:453-60.

492. Friberg L, Andersson T, Rosenqvist M. Less dementia and stroke in low-risk patients with atrial fibrillation taking oral anticoagulation. Eur Heart J 2019;40:2327-35.

493. McGuinness B, Craig D, Bullock R, Passmore P. Statins for the prevention of dementia. Cochrane Database Syst Rev 2009;1:CD003160.

494. Silva R, Miranda CM, Liu T, Tse G, Roever L. Atrial fibrillation and risk of dementia: epidemiology, mechanisms, and effect of anticoagulation. Front Neurosci 2019;13:18.

495. Moffitt P, Lane DA, Park H, O’Connell J, Quinn TJ. Thromboprophylaxis in atrial fibrillation and association with cognitive decline: systematic review. Age Ageing 2016;45:767-75.

496. Dietzel J, Haeusler KG, Endres M. Does atrial fibrillation cause cognitive decline and dementia? Europace 2018;20:408-19.

497. Bunch TJ, May HT, Bair TL, et al. Atrial fibrillation patients treated with long-term warfarin anticoagulation have higher rates of all dementia types compared with patients receiving long-term warfarin for other indications. J Am Heart Assoc 2016;5:e003932.

498. Field TS, Weijs B, Curcio A, et al. Incident atrial fibrillation, dementia and the role of anticoagulation: a population-based cohort study. Thromb Haemost 2019;119:981-91.

499. Flaker GC, Pogue J, Yusuf S, et al. Cognitive function and anticoagulation control in patients with atrial fibrillation. Circ Cardiovasc Qual Outcomes 2010;3:277-83.

500. Jacobs V, Woller SC, Stevens S, et al. Time outside of therapeutic range in atrial fibrillation patients is associated with long-term risk of dementia. Heart Rhythm 2014;11:2206-13.

501. Jacobs V, May HT, Bair TL, et al. Long-term population-based cerebral ischemic event and cognitive outcomes of direct oral anticoagulants compared with warfarin among long-term anticoagulated patients for atrial fibrillation. Am J Cardiol 2016;118:210-4.

502. Chen N, Lutsey PL, MacLehose RF, et al. Association of oral anticoagulant type with risk of dementia among patients with nonvalvular atrial fibrillation. J Am Heart Assoc 2018;7:e009561.

503. Sogaard M, Skjoth F, Jensen M, et al. Nonvitamin K antagonist oral anticoagulants versus warfarin in atrial fibrillation patients and risk of dementia: a nationwide propensity-weighted cohort study. J Am Heart Assoc 2019;8:e011358.

504. Jin MN, Kim TH, Kang KW, et al. Atrial fibrillation catheter ablation improves 1-year follow-up cognitive function, especially in patients with impaired cognitive function. Circ Arrhythm Electrophysiol 2019;12:e007197.

Back to top