Management of Patients on Non–Vitamin K Antagonist Oral Anticoagulants in the Acute Care and Periprocedural Setting: A Scientific Statement From the American Heart Association e604 March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 ABSTRACT: Non–vitamin K oral anticoagulants (NOACs) are now widely used as alternatives to warfarin for stroke prevention in atrial fibrillation and management of venous thromboembolism. In clinical practice, there is still widespread uncertainty on how to manage patients on NOACs who bleed or who are at risk for bleeding. Clinical trial data related to NOAC reversal for bleeding and perioperative management are sparse, and recommendations are largely derived from expert opinion. Knowledge of time of last ingestion of the NOAC and renal function is critical to managing these patients given that laboratory measurement is challenging because of the lack of commercially available assays in the United States. Idarucizumab is available as an antidote to rapidly reverse the effects of dabigatran. At present, there is no specific antidote available in the United States for the oral factor Xa inhibitors. Prothrombin concentrate may be considered in life-threatening bleeding. Healthcare institutions should adopt a NOAC reversal and perioperative management protocol developed with multidisciplinary input. A s the US population ages, the burden of atrial fibrillation (AF) and venous throm- boembolic disease is expected to increase, and prescriptions for long-term anticoagulation will climb. Anticoagulated patients are vulnerable to spontane- ous, traumatic and perioperative bleeding. Warfarin is a vitamin K antagonist (VKA) that has been used for decades to prevent and treat arterial and venous thromboem- bolism (VTE). More recently, 4 non–vitamin K antagonist oral anticoagulants (NOACs) have been approved in the United States as alternatives to warfarin for prevention of stroke resulting from nonvalvular AF (NVAF), and prevention and treatment of VTE. These are dabigatran etexilate (Pradaxa, Boehringer Ingelheim, Germany); rivaroxa- ban (Xarelto, Bayer HealthCare AG, Leverkusen, Germany), apixaban (Eliquis, Pfizer and Bristol-Myers Squibb, New York, NY) and edoxaban (Savaysa, Daiichi Sankyo, Tokyo, Japan). Direct oral anticoagulants has been proposed as alternative nomen- clature for these class of agents.1 NOACs are associated with comparable or lower risk of stroke, systemic embolism, major bleeding, and death compared with warfa- rin for NVAF.2–5 In contrast with warfarin, NOACs have a more predictable therapeutic effect, do not require routine monitoring, have fewer potential drug-drug interactions and no restriction on dietary consumption of vitamin K–containing food. However, universal adoption of NOACs has been stunted by the lack of specific antidotes and measurement assays. This scientific statement reviews the literature and offers practical suggestions for providers who manage patients who are actively bleeding and who are at risk for bleeding in the acute care and periprocedural setting. This statement focuses on interpreting available data rather than providing specific man- Amish N. Raval, MD, FAHA, Chair Joaquin E. Cigarroa, MD Mina K. Chung, MD, FAHA Larry J. Diaz-Sandoval, MD, FAHA Deborah Diercks, MD Jonathan P. Piccini, MD, MHS, FAHA Hee Soo Jung, MD Jeffrey B. Washam, PharmD, FAHA Babu G. Welch, MD Allyson R. Zazulia, MD, FAHA Sean P. Collins, MD, MSc, FAHA, Co-Chair On behalf of the Ameri- can Heart Association Clinical Pharmacology Subcommittee of the Acute Cardiac Care and General Cardiology Committee of the Coun- cil on Clinical Cardiol- ogy; Council on Car- diovascular Disease in the Young; and Council on Quality of Care and Outcomes Research Management of Patients on Non–Vitamin K Antagonist Oral Anticoagulants in the Acute Care and Periprocedural Setting A Scientific Statement From the American Heart Association © 2017 American Heart Association, Inc. Key Words: AHA Scientific Statements ◼ acute care ◼ anticoagulants ◼ non–vitamin K antagonist ◼ periprocedural AHA SCIENTIFIC STATEMENT D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e605 CLINICAL STATEM ENTS AND GUIDELINES agement recommendations in under-studied populations such as oncology patients. Members of this American Heart Association (AHA) writing group were selected for their diverse expertise in cardiovascular medicine, emergency medicine, critical care, neurology, surgery, and pharmacology. A system- atic search of the literature for each subtopic was per- formed in PubMed and Ovid and was supplemented by review of bibliographies as well as manual searches of key articles. Each of the following search terms were in- cluded individually and in combination: dabigatran, apix- aban, rivaroxaban, edoxaban, anticoagulation, reversal, antidote, atrial fibrillation, venous thromboembolism, bleeding, intracranial, cardioversion, catheterization, cardiac implantable devices, kidney injury, transition, switching, pharmacology, andexanet alfa, idarucizumab, ciraparantag, gastrointestinal, trauma, surgery, percu- taneous coronary intervention, neuraxial anesthesia, stroke, and overdose. Writing group members were instructed to write subtopic sections aligned with their experience. Members were instructed to cite contempo- rary guidelines and scientific statements where appropri- ate. The writing group did not assign formal classes of recommendation/level of evidence per the AHA Scien- tific Document Development Process recommendation that went into effect September 1, 2015. Sections were then reviewed by another writing group member. Sec- tion drafts were submitted to the writing group chair and co-chair and compiled into a single document. Web and teleconferences were convened to review and edit the full draft. The final document was submitted for indepen- dent peer review and approved for publication by the AHA Manuscript Oversight Committee on April 29, 2016. PHARMACOLOGY OF NOACS NOACs act through direct inhibition of thrombin or inhi- bition of factor Xa (Figure 1). Dabigatran etexilate me- sylate is a competitive direct thrombin inhibitor. Rivar- oxaban, apixaban, and edoxaban inhibit factor Xa and prothrombinase activity, thus inhibiting the conversion of prothrombin to thrombin. Thrombin catalyzes the con- version of fibrinogen to fibrin; activates factors V, VIII, XI, and XIII; and activates platelets. Therefore, inhibiting thrombin decreases thrombus formation. In contrast with warfarin, NOACs have a rapid onset of action, a shorter half-life, and more predictable pharmacokinet- ics. Routine therapeutic monitoring was not done in the major NOAC efficacy trials and is at present not recom- mended in usual clinical practice. Information pertaining to NOAC dose, time to peak effect, and time to offset of effect is outlined in Table 1. NOACs are substrates for P-glycoprotein (P-gp) trans- port and apixaban and rivaroxaban are substrates for CYP 3A4 metabolism. Therefore, concomitant medica- tions that are inducers or inhibitors of these pathways should be evaluated for the potential to interact (Table 2). Macrolides and nondihydropyridine calcium channel blockers are 2 commonly prescribed classes of medica- tions that impact therapeutic levels of NOACs, although a post hoc analysis of ROCKET AF (Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared with Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation) showed no evidence of differential outcomes between rivaroxaban and warfarin in patients treated with ≥1 combined P-gp and CYP 3A4 inhibitors.6 Edoxaban exists in a predominantly unchanged form Figure 1. Clotting cascade and anticoagulants. VKA indicates vitamin K antagonist. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e606 Table 1. Comparison Among NOACs Dabigatran Rivaroxaban Apixaban Edoxaban Approved indications Nonvalular AF ↓ Risk of stroke and systemic embolism Nonvalular AF ↓ Risk of stroke and systemic embolism Nonvalular AF ↓ Risk of stroke and systemic embolism Nonvalular AF ↓ Risk of stroke and systemic embolism. Limitation: should not use in patients with CrCl >95 mL/min as a result of ↑ risk of ischemic stroke compared with warfarin at 60 mg DVT, PE Treatment after 5–10 d parenteral AC ↓ Recurrence Prophylaxis after hip replacement DVT, PE Treatment ↓ Recurrence Prophylaxis after hip or knee replacement DVT, PE Treatment ↓ Recurrence Prophylaxis after hip replacement DVT, PE ↓ Recurrence Treatment after 5–10 d initial parenteral AC Mechanism of action Direct thrombin inhibitor Factor Xa inhibitor Factor Xa inhibitor Factor Xa inhibitor Time to peak 1 h; delayed to 2 h with food 2–4 h 3–4 h 1–2 h Bioavailability 3%–7% 10-mg dose: 80%–100% ~50% 62% 20-mg dose: 66% ↑ With food Plasma protein binding 35% 92%–95% ~87% 55% Volume of distribution 50–70 L 50 L 21 L 107 L Plasma t 1/2 12–17 h 5–9 h ~12 h (8–15 h) 10–14 h Elderly 14–17 h Elderly 11–13 h Mild to moderate renal impairment 15–18 h Severe renal impairment 28 h Metabolism Hepatic and plasma hydrolysis to active dabigatran Hepatic: oxidation by CYP3A4/5, CYP2J2; hydrolysis to inactive metabolites (51%) Hepatic: 25% mainly by CYP3A4/5; lesser by CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2J2; O-demethylation and hydroxylation Minimal CYP3A4 hydrolysis, conjugation, oxidation Hepatic glucuronidation to active metabolites (<10%) P-gp substrate No active circulating metabolites Active metabolite (M-4, <10% of parent) P-gp substrate No major or active circulating metabolites Substrate of CYP3A4, P-gp, BCRP P-gp substrate Substrate of P-gp and ABCG2 (BCRP) Excretion Renal (~80%) after IV administration Renal (66%): 36% active, 30% inactive metabolites Renal (27%) Renal (~50%): primarily as unchanged drug After oral, 7% recovered in urine, 86% in feces Feces (28%): 7% active, 21% inactive metabolites Biliary and direct intestinal excretion Metabolism and biliary/ intestinal excretion accounts for the rest (Continued ) D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e607 CLINICAL STATEM ENTS AND GUIDELINES Dosing Nonvalvular AF CrCl >30 mL/min: 150 mg BID CrCl >50 mL/min: 20 mg daily with evening meal 5 mg BID CrCl >50 to ≤95 mL/min: 60 mg daily CrCl 15–30 mL/min: 75 mg BID CrCl 15–50 mL/min: 15 mg daily with evening meal 2.5 mg BID, if 2 of 3 characteristics: Cr ≥1.5 mg/dL, age ≥80 y, weight ≤60 kg CrCl 15–50 mL/min: 30 mg daily CrCl <15 mL/min or on dialysis: Not recommended Not recommended for CrCl <15 mL/min or on dialysis in patients with AF NOT recommended for CrCl >95 mL/min CrCl 30–50 mL/min with concomitant P-gp inhibitors: 75 mg BID CrCl <30 mL/min with concomitant P-gp inhibitors: Avoid coadministration DVT or PE treatment CrCl >30 mL/min: 150 mg BID after 5-10 d parenteral anticoagulation 15 mg BID with food first 21 d for initial treatment, then 20 mg once daily with food 10 mg BID x 7 d, then 5 mg BID 60 mg once daily CrCl ≤30 mL/min or on dialysis: Not recommended Not recommended for CrCl <30 mL/min in patients with DVT or PE CrCl 15–50 mL/min or weight ≤60 kg or on certain P-gp inhibitors: 30 mg once daily ↓ in recurrent DVT/PE CrCl >30 mL/min: 150 mg BID after 5–10 d parenteral anticoagulation 20 mg daily with food 2.5 mg BID CrCl ≤30 mL/min or on dialysis: Not recommended DVT, PE prophylaxis after hip or knee replacement After hip replacement surgery: CrCl >30 mL/min after achievement of hemostasis: If given day of surgery, 110 mg 1–4 h postop; after day of surgery 220 mg once daily x 28–35 d CrCl ≤30 mL/min or on dialysis: Not recommended CrCl <50 mL/min with concomitant P-gp inhibitors: Avoid coadministration Initial dose 6–10 h after surgery provided hemostasis established 2.5 mg BID x 35 d after hip replacement surgery or x 12 d after knee replacement surgery 10 mg daily with or without food x 35 d for hip replacement, x 12 d for knee replacement Additional dosing comments Avoid use with patients with moderate-severe hepatic impairment (Child- Pugh class B/C) or hepatic disease with coagulopathy Not recommended in patients with severe hepatic impairment (Child-Pugh class C) Not recommended with CrCl <15 mL/min 15-20 mg taken with food; 10 mg with or without food Not recommended in patients with moderate-severe hepatic impairment (Child-Pugh class B/C) Table 1. Continued Dabigatran Rivaroxaban Apixaban Edoxaban (Continued ) D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e608 in plasma with minimal metabolism through hydrolysis, conjugation, and oxidation by CYP 3A4. LABORATORY MEASUREMENT OF NOAC EFFECT One advantage of NOACs over warfarin is more rapid on- set and offset of action with predictable pharmacokinet- ics and anticoagulant effect. This eliminates the necessi- ty for routine therapeutic monitoring except for periodic assessment of renal function.7 Laboratory measurement of NOAC level or effect may be necessary in certain acute care or perioperative settings, particularly when there is uncertainty about the timing of last ingestion, renal function, and gastrointestinal absorption. However, the lack of US Food and Drug Administration–approved NOAC laboratory assays complicates the management of NOAC overdose, NOAC-associated life-threatening bleeding, and the scheduling of urgent surgical proce- dures.8 All NOAC agents affect routine coagulation tests but not in a manner that allows for a predictable and quantitative measurement of anticoagulation effect. Spe- cific NOAC agents are subsequently discussed. Dabigatran This agent is known to prolong the activated partial thromboplastin time, prothrombin time, and thrombin time. The package insert recommends using partial thromboplastin time for measurement; however, there is no defined partial thromboplastin time therapeutic range for dabigatran and the assay is relatively insensitive to different plasma concentrations of direct thrombin inhibi- tors. Furthermore, the partial thromboplastin time can- not be used in patients with lupus anticoagulant or an intrinsic clotting factor deficiency because its prolonga- tion from these conditions would mask the anticoagulant effect of dabigatran. Thrombin time is far more sensi- tive, and prothrombin time is less sensitive to dabigatran. A normal partial thromboplastin time or thrombin time most likely excludes therapeutic levels of dabigatran, whereas a normal prothrombin time may not. Quantita- tive assessments of dabigatran levels can be obtained with the dilute thrombin time, the ecarin clotting time, or the ecarin chromogenic assay. Thrombin time and ecarin- based assays show excellent linearity across on-therapy drug concentrations and may be used for drug quantifica- tion. However, the US Food and Drug Administration has not approved these latter assays for measuring levels of dabigatran or other direct thrombin inhibitors.8 Rivaroxaban, Apixaban, Edoxaban At present, there are no US Food and Drug Administra- tion–approved assays or calibration reagents to measure the effect of direct oral factor Xa inhibitors. Rivaroxaban and apixaban affect activated clotting time and chro- mogenic anti–factor Xa assay; however, no therapeutic range exists. Prothrombin time is less sensitive (espe- cially for apixaban), and a normal prothrombin time may not exclude clinically relevant levels. Partial thromboplas- tin time demonstrates insufficient sensitivity and linearity for quantification.9 Studies using spiked plasma samples suggest using prothrombin time for a qualitative assess- ment of direct oral factor Xa inhibitors or chromogenic anti–factor Xa assay for a quantitative assessment of direct oral factor Xa inhibitors.8–10 Anti-Xa activity is linear over a wide range of drug levels and may be used for drug quantification. Undetectable anti-Xa activity likely excludes clinically relevant drug concentrations. In summary, although routine NOAC monitoring is unnecessary, measurement of NOAC effect may assist clinical management in certain acute care and peripro- cedural settings. In most situations, the time of last drug ingestion combined with a recent assessment of creati- nine clearance (CrCl) should enable appropriate clinical decision making. Therapeutic measurement Routine not required Routine not required Routine not required Routine not required To detect presence: aPTT, ECT (if available), TT To detect presence: PT, aPTT, antifactor Xa activity To detect presence: PT, aPTT, antifactor Xa activity Prolongs PT, aPTT, antifactor Xa activity aPTT >2.5 times control may indicate overanticoagulation Renal function, CBC periodically, at least annually; hepatic function Renal function, CBC periodically, at least annually Renal function, CBC periodically, at least annually Renal function, CBC periodically, at least annually AC indicates anticoagulant; AF, atrial fibrillation; aPTT, activated partial thromboplastin time; BID, twice daily; CBC, complete blood count; CrCl, creatinine clearance; DVT, deep vein thrombosis; ECT, ecarin clotting time; IV, intravenous; NOACs, non–vitamin K antagonist oral anticoagulants; PE, pulmonary embolism; P-gp, P-glycoprotein; PT, prothrombin time; and TT, thrombin time. Table 1. Continued Dabigatran Rivaroxaban Apixaban Edoxaban D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e609 CLINICAL STATEM ENTS AND GUIDELINES Table 2. NOAC Drug Interactions NOAC Interacting Medications Effect on NOAC Labeled Guidance; Comments Dabigatran P-gp inducer: rifampin ↓ Dabigatran exposure Concomitant use should generally be avoided. P-gp inhibitors: ketoconazole, dronedarone ↑ Dabigatran exposure if concomitant moderate renal impairment If moderate renal impairment (CrCl 30–50 mL/min) ↓ to 75 mg BID during concomitant use P-gp inhibitors: ketoconazole, dronedarone, verapamil, amiodarone, quinidine, clarithromycin, ticagrelor ↑ Dabigatran exposure if concomitant severe renal impairment If severe renal impairment (CrCl 15–30 mL/ min) avoid concomitant use Apixaban Strong dual P-gp and CYP3A4 inducers: rifampin, carbamazepine, phenytoin, St. John’s wort ↓ Apixaban exposure Avoid concomitant use Strong dual P-gp and CYP3A4 inhibitors: ketoconazole, itraconazole, ritonavir, clarithromycin ↑ Apixaban exposure In patients on 5 mg or 10 mg BID, ↓ dose by 50% when coadministered Avoid coadministration on 2.5 mg BID Rivaroxaban Combined P-gp and strong CYP3A4 inducers: rifampin, carbamazepine, phenytoin, St. John’s wort ↓ Rivaroxaban exposure Avoid concomitant use; may decrease rivaroxaban efficacy Combined P-gp and strong CYP3A4 inhibitors: ketoconazole, itraconazole, HIV protease inhibitors (ritonavir, lopinavir/ ritonavir, indinavir), conivaptan ↑ Rivaroxaban exposure Avoid concomitant use Combined P-gp and moderate CYP3A4 inhibitors: diltiazem, verapamil, amiodarone, dronedarone, erythromycin ↑ Rivaroxaban exposure in patients with renal impairment In patients with CrCl 15 to <80 mL/min, rivaroxaban should not be used concomitantly unless the potential benefit justifies the potential risks No evidence of interaction observed in ROCKET AF between treatment assignment and outcomes in patients using ≥1 combined P-gp and moderate 3A4 inhibitors (including amiodarone, diltiazem, and verapamil)6 Edoxaban P-gp inducer: rifampin ↓ Edoxaban exposure Avoid concomitant use Strong P-gp inhibitors: ritonavir, nelfinavir, saquinavir, indinavir, cyclosporine ↑ Edoxaban exposure Avoid concomitant use in patients taking edoxaban for treatment of VTE P-gp inhibitors: verapamil, quinidine, azithromycin, clarithromycin, itraconazole, ketoconazole ↑ Edoxaban exposure ↓ to 30 mg daily during concomitant administration for patients taking edoxaban for the treatment of VTE Dose reduction is not recommended for AF indications In ENGAGE AF, a ↓ dose of edoxaban as a result of concomitant P-gp inhibitor use (verapamil, quinidine, dronedarone) was associated with ↓ edoxaban exposure and a relative ↑ in risk of stroke or systemic embolism with edoxaban relative to warfarin176 AF, atrial fibrillation; BID, twice daily; CrCl, creatinine clearance; ENGAGE AF, Effective Anticoagulation With Factor Xa Next Generation in Atrial Fibrillation trial; NOAC, non–vitamin K antagonist oral anticoagulant; P-gp, P-glycoprotein; ROCKET AF, Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared with Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation; and VTE, venous thromboembolism. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e610 NOAC REVERSAL This AHA writing group suggests hospital systems adopt anticoagulation reversal protocols with multidisciplinary representation from emergency medicine, critical care, cardiology, hematology, gastroenterology, neurology, neurosurgery, trauma, acute care surgery, cardiotho- racic surgery, vascular surgery, pharmacy, and nursing. An example of a NOAC reversal protocol is shown in Figure 2. Dabigatran For minor bleeding, supportive care and careful obser- vation are suggested. For major bleeding, intravenous idarucizumab (Praxbind, Boehringer-Ingelheim, Germa- ny) at a dose of 5 grams (2 consecutive intravenous infu- sions of 2.5 g each) will reverse the anticoagulant effect of dabigatran within minutes.11 Idarucizumab is a mono- clonal antibody fragment that binds dabigatran with an affinity 350 times that of thrombin. The RE-VERSE AD (Reversal Effects of Idarucizumab on Active Dabigatran) was a prospective cohort study that showed that ida- rucizumab administration reversed anticoagulation as evidenced by the normalization of the dilute thrombin time and ecarin clotting time within minutes among sub- jects suffering a serious hemorrhage or who required an urgent procedure.11 Early hemostasis was achieved in bleeding subjects, and a low rate of perioperative bleeding events was observed in subjects undergoing urgent surgery. However, the strength of these clinical observations is limited by the nonrandomized nature of this study. Several studies have investigated the efficacy of pro- thrombin complex concentrates (PCCs), recombinant fac- tor VII activated, and fresh frozen plasma (FFP) in animal models; however, human data are mixed. One random- ized, placebo-controlled trial in healthy men treated with dabigatran showed that 4-factor PCC did not reverse the dabigatran effect on partial thromboplastin time, endoge- nous thrombin potential lag time, thrombin time, or ecarin clotting time.12 Case reports of patients with life-threat- ening bleeding associated with dabigatran therapy have demonstrated mixed results with the use of FFP, recombi- nant factor VII activated, PCCs, fibrinogen, and platelets.8 Hemodialysis may remove 49% to 57% of dabigatran within 4 hours given that the drug is only 35% bound to plasma proteins.13,14 Hemodialysis may be considered if the CrCl is chronically below 30 mL/min or in acute kidney injury. For major ingestion, there is some evi- dence to support the use of activated charcoal therapy if dabigatran was consumed within 1 to 2 hours; however, care must be taken to prevent aspiration in patients with decreased level of consciousness.7,8 Furthermore, acti- vated charcoal induced vomiting could have deleterious effects by increasing intracranial pressure in patients with intracranial hemorrhage (ICH). In summary, the AHA writing group suggests com- pression when possible, supportive measures, and up- front idarucizumab in the event of dabigatran-associated major bleeding. Rivaroxaban and Apixaban Similar to dabigatran, activated charcoal may prevent absorption of rivaroxaban and apixaban if administered Figure 2. Example of a “Serious Bleeding on a NOAC” protocol. BUN indicates blood urea nitrogen; CBC, complete blood count; IU, international units; IV, intravenous; NOAC, non–vitamin K antagonist oral anticoagulants; PCC, prothrombin complex concentrate; PRBC, packed red blood cells; PTT, partial thrombo- plastin time; and TT, thrombin time. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e611 CLINICAL STATEM ENTS AND GUIDELINES within 1 to 2 hours after NOAC ingestion. Rivaroxaban and apixaban are highly bound to plasma proteins; therefore, dialysis is ineffective in clearing these drugs. Andexanet alfa is a recombinant modified human fac- tor Xa decoy protein that serves as a specific reversal agent to neutralize the anticoagulant effects of direct and indirect factor Xa inhibitors. This drug is admin- istered as an initial intravenous bolus followed by an infusion for up to 2 hours. A recent study revealed that andexanet alpha reversed the laboratory assessed anticoagulant activity of rivaroxaban and apixaban in older healthy individuals within minutes of administra- tion.15 At present, the single arm, open-label ANNEXA-4 (Andexanet Alfa for Acute Major Bleeding Associated with Factor Xa Inhibitors) trial is under way to confirm the clinical benefit of this drug in patients on apixaban, rivaroxaban, edoxaban, or enoxaparin who present with an acute major hemorrhage. An interim analysis of 67 patients revealed an 89% and 93% reduction in anti– factor Xa activity for those on rivaroxaban and apixaban respectively.16 Of the entire cohort, 47 patients were followed for clinical hemostasis. Of these, 37 (79%; 95% confidence interval [CI], 64–89) were adjudicated as having excellent or good clinical hemostasis. The dosing protocol in this study was as follows: (1) for patients who had taken apixaban, or rivaroxaban >7 hours prior, andexanet alfa was given as a bolus dose of 400 mg followed by an infusion of 480 mg over 2 hours; and (2) for patients who had enoxaparin, edoxa- ban, or rivaroxaban <7 hours prior or at an unknown time, the bolus dose and infusion dose amount was doubled (800-mg bolus, 960-mg infusion over 2 hours). At present, andexanet alfa is not approved in the United States or elsewhere. A randomized placebo-controlled study of young, healthy volunteers treated with 20 mg of rivaroxaban dosed twice daily found that administration of a 4-factor PCC led to normalization of the prothrombin time and the endogenous thrombin potential.12 In contrast, an in vitro study using human plasma obtained from healthy donors found that recombinant factor VII activated was superior to a 4-factor PCC at normalizing laboratory co- agulation studies.17 Case reports of using FFP or PCC to treat excess rivaroxaban ingestion have shown mod- est success in improving laboratory coagulation param- eters.18–20 However, the correction of coagulation tests by PCC, FFP, or recombinant factor VII activated does not imply the reversal of the clinical anticoagulation ef- fect of the drug. There is no evidence that FFP or PCC controls NOAC-associated bleeding in humans. Edoxaban Four-factor PCC showed dose-dependent reversal of edoxaban effect with complete reversal of bleeding duration after skin punch biopsy in volunteers and par- tial reversal of prothrombin time after a 50-IU/kg dose administration.21 However, the clinical relevance of this finding is uncertain. Ciraparantag (PER977) is a small synthetic, water- soluble, cationic molecule designed to specifically bind to unfractionated heparin and low-molecular-weight hep- arin through noncovalent hydrogen bonding and charge- charge interactions. It also binds in a similar way to di- rect Xa inhibitors and direct thrombin inhibitors. It has been shown to normalize whole blood clot time within 10 to 30 minutes of administration.22,23 Ciraparantag is still being investigated in early clinical trials as an antidote for edoxaban associated bleeding. It remains unknown whether andexanet alfa will have greater, equal, or less- er clinical efficacy for edoxaban reversal compared with ciraparantag. MANAGEMENT OF LIFE-THREATENING BLEEDING All patients with life-threatening bleeding should be man- aged with similar basic resuscitation principals, irrespec- tive of what type of anticoagulant they may be on. Imme- diate management of the patient’s airway, breathing, and circulation with attempts to control hemorrhage is vital. When life-threatening bleeding occurs in a compressible area of the body, direct pressure along with selective use of tourniquets can be life-saving. Similarly, immedi- ate resuscitation and stabilization with intravenous flu- ids, packed red blood cells and plasma may be required in the unstable patient. NOAC reversal as indicated in NOAC Reversal should be considered. These concepts apply to blunt and penetrating trauma, massive gastro- intestinal, retroperitoneal, pericardial hemorrhage, and other forms of major bleeding. Specific Scenario: ICH A meta-analysis of studies that have tested NOACs for ischemic stroke prevention in NVAF have estimated a pooled incidence of hemorrhagic stroke of 0.4%.2–5,24 Overall, this represents a >50% relative reduction in ICH rate from the 0.9% observed with warfarin. Past VKA studies suggest that ICH is 11 times more likely to result in mortality compared with extracranial hemorrhage.25 The reduction in ICH rate coupled with consistent non- inferiority compared with VKAs in preventing thrombotic events has produced a steady increase in the use of NOACs to prevent stroke in patients with NVAF. Uniform recommendations do not exist regarding man- agement of patients on NOACs who suffer ICH primarily because no consistent approach to their management was undertaken in the NOAC trials. Factors to consider include availability of reversal agents, the timing of urgent neurosur- gery, risk of thromboembolic events during the period off D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e612 the anticoagulant, and reinstitution of anticoagulant therapy after the ICH event or after surgery. The presence of ICH creates a unique circumstance because of the noncom- pressible location of the hemorrhage and poor tolerance of the brain to continued bleeding. The AHA/American Stroke Association “Guidelines for the Management of Spontane- ous Intracerebral Hemorrhage” recommends prompt cre- ation of a hemostatic environment to limit extension of the hemorrhage and before surgical treatment.26 Any acute neurological change in a patient on NOAC therapy should be presumed to be vascular in origin. A baseline severity score should be performed as part of the initial evaluation.26,27 Computed tomography (CT) is widely available, detects acute hemorrhage with high sensitivity, and defines the extent of the injury on the sur- rounding parenchyma. Contrast-enhanced CT may iden- tify patients at high risk of ICH expansion on the basis of the presence of contrast within the hematoma, also known as the spot sign.28,29 Detailed vascular imaging may identify predisposing vascular lesions such as an- eurysm, arteriovenous malformation, and dural fistula. Concurrent with reversing the NOAC effect, blood pressure needs to be intensively managed. Many studies associate elevated systolic blood pressure with greater hematoma expansion, neurological deterioration, and death and dependency after ICH.30,31 The INTERACT2 tri- al (Intensive Blood Pressure Reduction in Acute Cerebral Hemorrhage Trial 2) showed that acute blood pressure reduction to <140 mm Hg systolic was safe and resulted in a trend toward improvement in functional recovery de- spite no significant reduction in the rate of hematoma growth.32 No patients with NOAC use were included in this trial. However, recent results from ATACH-2 (Antihy- pertensive Treatment of Acute Cerebral Hemorrhage-II) suggest aggressive lowering of systolic blood pressure to 110 to 139 mm Hg may not confer benefit.32a The safety of resuming a NOAC regimen after ICH is a common clinical dilemma. Decisions about whether to resume anticoagulation after ICH must take into account the patient’s underlying thromboembolic risk and the risk for ICH recurrence. Embolic stroke risk versus bleeding risk stratification schemes such as the CHA 2DS2-VASc and HAS-BLED scores may help guide treatment after ICH.30,31 The HAS-BLED score has been validated in a wide range of patients (AF and non-AF, VKA and non-VKA) and is the only bleeding risk scheme that is predictive of ICH.33 However, a high HAS-BLED score should not be the sole consideration in clinical management. The presence of a recent ICH should prompt closer evalua- tion of other factors related to ICH reoccurrence.24,34,35 Factors that are suggested to increase ICH risk include older age, poor blood pressure control, lobar ICH loca- tion, presence of microbleeds on gradient echo magnetic resonance imaging, concurrent aspirin use, and the pres- ence of apolipoprotein E ε2 or ε4 alleles. AHA/American Stroke Association guidelines provide a class IIb recom- mendation for anticoagulation to be considered only after nonlobar ICH; however, this recommendation is based on warfarin-associated ICH data.34 Whether NOACs can be safely administered in this population is still unknown. There is no clinical trial evidence to guide the man- agement of patients with traumatic brain injury while on anticoagulants. An initial head CT is typical; however, the role of repeated CT or inpatient observation with neuro- logical assessment remains controversial when the initial head CT is negative. Until further data become available, NOAC reversal for traumatic ICH should be considered similar to nontraumatic ICH. In summary, the AHA writing group suggests that trau- matic and nontraumatic ICH patients on dabigatran who re- quire NOAC reversal receive idarucizumab. ICH patients on rivaroxaban, apixaban, or edoxaban should receive PCC until more specific antidotes become available. Specific Scenario: Trauma The prevalence of NOAC use in the trauma population is unknown. To compare, the prevalence of warfarin use in the trauma population in 2006 was 4% with a 1.7% absolute rate increase over the previous 4 years.36 Pa- tients should be encouraged to carry information cards or bracelets that would alert emergency medical provid- ers regarding oral anticoagulation use. Apart from a few case reports, there are limited data to guide the management of NOACs in the setting of trauma.37–42 The American College of Surgeons Advance Trauma Life Support course43 recommends obtaining a brief, focused history during the initial evaluation of traumatically injured patients. This should include iden- tifying the specific NOAC, timing of last ingestion, and the underlying reason for NOAC use. Laboratory testing of renal function and coagulation parameters described in Laboratory Measurement of NOAC Effect may help with treatment decisions. Thromboelastography and rotational thromboelastometry to detect NOAC activity in isolated trauma cases has been reported; however, routine use cannot be recommended until further data becomes available.44,45 NOACs may be held during the period of clinic as- sessment or until hemostasis has been achieved in trauma patients without bleeding and with mild bleeding, or bleeding from easily controllable foci. Maintaining ade- quate urine output and specific NOAC reversal strategies (NOAC reversal) should be considered in trauma patients with moderate or severe bleeding, or suspected bleed- ing that requires further evaluation. Specific Scenario: Gastrointestinal Bleeding In major trials, dabigatran 150 mg twice daily, rivaroxa- ban, and edoxaban 60 mg once daily were associated with a 1.5-fold increased risk of gastrointestinal bleeding D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e613 CLINICAL STATEM ENTS AND GUIDELINES compared with warfarin; apixaban and dabigatran 110 mg twice daily had similar gastrointestinal bleeding risk; and edoxaban 30 mg once daily had significantly lower risk.2,3,5 Factors associated with gastrointestinal bleed- ing with NOAC use are anemia, previous gastrointestinal bleeding, long term aspirin use or baseline nonaspirin antiplatelet use, age, diastolic hypertension, smoking, sleep apnea, chronic obstructive pulmonary disease, pre- vious proton pump inhibitor use, renal dysfunction, and male sex.46 Although gastrointestinal bleeding accounts for nearly 90% of major extracranial hemorrhages in NVAF patients on therapeutic anticoagulation,47 clinical data specifically pertaining to NOAC reversal are lacking. Of the 3.3% of patients in the Dresden registry who expe- rienced major bleeding while on rivaroxaban, the majority of patients were managed conservatively without requir- ing surgery.48 As in the case of trauma, general resuscita- tion principles of airway, intravenous fluid, blood transfu- sion, and maintaining adequate urine output should be applied. A Blakemore tube for bleeding from esophageal varices may be considered. Immediate NOAC reversal should be considered in the unstable patient. Reinitiating NOAC therapy after gastrointestinal bleed- ing should take into account the patient’s underlying risk of bleeding and thrombosis risk. In a retrospective study of >4600 patients with NVAF who suffered gastrointes- tinal bleeding on anticoagulation (primarily warfarin), re- sumption of a single anticoagulant was associated with the lowest risk of mortality and thromboembolism com- pared with nonresumption of antithrombotic treatment. The risk of recurrent gastrointestinal bleeding was also low in the anticoagulated patients. Patients on NOACs comprised a very small subset of the entire cohort; therefore, it remains uncertain whether NOAC resump- tion after gastrointestinal bleeding would be similarly linked to these favorable outcomes.49 MANAGEMENT OF PATIENTS ON NOACS WHO ARE AT RISK FOR BLEEDING Management of Patients Who Overdose on NOACs Data regarding the prevalence of overdoses or unpre- scribed exposures to NOACs are largely based on ob- servational data from poison control centers and case reports.13,14,18,19,50–61 Stevenson et al reported that be- tween January 2011 and July 2013, there were 49 calls to a single poison control center regarding dabigatran and rivaroxaban.18 Of these, only 4 cases were a result of self-harm, and only mild bleeding was reported in 1 case. The majority of bleeding events were noted in pa- tients on long-term treatment and not acute ingestions, and there was no association with coagulation abnor- malities and risk of bleeding. Conway et al reported dab- igatran exposures from a national poison control center and noted that adverse outcomes occurred in only 5% of all calls, and only 1.3% were considered intentional.50 An observational study from poison control centers in 9 states showed that among 223 NOAC exposure calls related to rivaroxaban and apixaban ingestions, 42% had abnormal coagulation studies and no patient had bleeding.51 Unfortunately, there is limited information to guide management of patients with NOAC overdose with and without bleeding. Collection of information on the type of NOAC, the ingested dose, time of ingestion, concomitant renal/liver disease, and relevant medica- tion coingestion is critically important in the acute pe- riod. Therapeutic management strategies in the acute care setting have largely been developed on the basis of clinical experience and an understanding of the phar- macology rather than trial data. Management of Patients With Acute Kidney Injury on NOACs The risk of acute kidney injury is high in the patient pop- ulation who are frequently prescribed NOACs. Andreu- Cayuelas et al performed an observational study of 162 patients with NVAF after hospitalization for acute heart failure. Creatinine was measured during follow-up to de- termine the need for dose adjustment of the hypotheti- cal NOACs.62 The investigators reported 44% of patients would have needed dabigatran dosage adjustment, 35% would have needed rivaroxaban adjustment, and 29% would have needed apixaban dosage adjustment. The patients with a baseline CrCl of <60 mL/min or age >75 years were at greatest risk of needing a dose adjust- ment during follow-up.62 VKA-associated nephropathy has recently been de- scribed as acute kidney injury with supratherapeutic in- ternational normalized ratio (INR) values with and without hematuria.63–65 Alternatively, NOACs do not appear to be associated with kidney injury. In a meta-analysis conduct- ed by Caldeira et al, NOACs did not increase the risk of renal failure (relative risk [RR], 0.96; 95% CI, 0.87–1.07; I2= 17.8%; 6 randomized controlled trials) when com- pared with a VKA.63 A recent analysis of ROCKET AF re- vealed a small but statistically significant decline in mean CrCl± standard deviation among patients receiving war- farin (−4.3±14.6 mL/min) compared with patients re- ceiving rivaroxaban (−3.5±15.1 mL/min; P<0.001).66 A post hoc analysis of the RE-LY (Randomized Evaluation of Long-Term Anticoagulation Therapy) trial similarly re- vealed greater declines in CrCl with warfarin compared with dabigatran.67 Administering a NOAC in a patient with acute kidney injury increases the risk of bleeding. All NOACs should be used with caution in patients on hemodialysis, given the limited data available. Although a dosing recommen- dation for apixaban is provided for such patients in the product monograph, this recommendation is based on D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e614 pharmacokinetic data in fewer than 20 patients. There are no efficacy or safety data in this patient population. Until these data become available, close measurement or switching to an alternative anticoagulant is suggested for patients who develop acute kidney injury as a result of acute illness or injury. Management of Patients With Ischemic Stroke on NOACs Whereas NOACs represent a major advance in stroke prevention, it is still anticipated that acute ischemic stroke (AIS) will occur in 1% to 2% of individuals with NVAF treated with these agents each year.2–5 Their use presents a number of challenges for clinicians managing patients with AIS, including appropriate measurement of anticoagulant activity in neurovascular emergencies, the role of thrombolysis and endovascular therapy in AIS, and timing of reinstitution of oral anticoagulation after AIS. Thrombolytic therapy with intravenous recombinant tissue-type plasminogen activator within 4.5 hours of symptom onset is an established treatment in AIS68,69 but is associated with a >5-fold increase in the rate of ICH.70 Because of the danger of further increasing ICH, therapeutic anticoagulation is considered a contraindica- tion to thrombolytic therapy in AIS. AHA guidelines and observational data support intravenous thrombolysis in warfarin-treated patients provided the INR is no greater than 1.7.71,72 The data on safety of thrombolysis in the presence of low levels of anticoagulation with warfarin raises hope that the same may apply to NOACs. Determining appropriate treatment for AIS patients re- ceiving NOACs must balance the anticoagulant effect of these agents and the ICH risk associated with reperfusion strategies. As has been mentioned previously, routinely performed blood coagulation studies do not reliably ex- clude a significant plasma concentration of the NOACs. Another difficulty in a time-sensitive setting is that the more sensitive blood tests are either not routinely avail- able or have an unacceptably long delay to results. In experimental studies, pretreatment with dabigatran or rivaroxaban did not increase the rate of thrombolysis- associated ICH.72,73 Data on the safety and efficacy of intravenous thrombolysis in AIS patients receiving NO- ACs are limited to approximately 2 dozen case reports and a retrospective multicenter cohort study. Among the case reports, ICH and poor outcome were rarely report- ed when recombinant tissue-type plasminogen activator was administered minutes to 24 hours after the last an- ticoagulant dose.74,75 The cohort study76 comprised 78 NOAC-treated patients undergoing intravenous throm- bolysis or intra-arterial therapy a median of 13 hours after the last NOAC dose compared with 441 warfarin- treated patients and 8938 on no anticoagulants. After propensity score matching, there was no significant dif- ference in rate of any ICH, symptomatic ICH, or death among the groups. In the absence of immediately avail- able blood tests sensitive to the presence of NOACs, determining which patients taking these agents might be appropriate candidates for thrombolysis requires con- sideration of time from last dose, half-life of the agent used, and presence of impaired renal function that may reduce drug clearance. A new recommendation in the AHA “Guidelines for the Early Management of Patients With Acute Ischemic Stroke” is that recombinant tissue- type plasminogen activator should not be administered to patients who take NOACs unless sensitive laboratory tests are normal or the patient has not received a dose of these agents for >48 hours.71 Data guiding the use of endovascular therapy in AIS patients who take NOACs are even more limited. Among the pivotal trials that established the safety and efficacy of mechanical thrombectomy in patients with AIS and large vessel occlusion, patients receiving NOACs were either excluded77 or not specifically reported.78–81 A handful of case reports suggest safety of endovascular therapy in patients on dabigatran and rivaroxaban even in the setting of abnormal coagulation studies.82–87 In the previously described cohort study, none of the 33 pa- tients who underwent endovascular therapy with or with- out intravenous thrombolysis experienced a symptom- atic ICH. Reflecting the paucity of data in this area, the AHA’s guidelines provide no recommendations regarding mechanical thrombectomy in patients whose use of anti- coagulant medications excludes them from intravenous thrombolysis.88 The optimal timing of restarting anticoagulation after AIS presents another challenge to healthcare profession- als managing this population. Meta-analysis of 7 trials of parenteral anticoagulation started within 48 hours of cardioembolic ischemic stroke89 and systematic review of 24 trials involving 23 748 participants with AIS90 test- ing various parenteral and oral anticoagulants each con- cluded that while early anticoagulation is associated with a reduced risk of recurrent ischemic stroke, this benefit is entirely offset by an increased risk of symptomatic ICH with no reduction in risk of death or dependency. The decision of when to restart oral anticoagulation must balance the competing risks of recurrent thrombo- embolic events and of hemorrhagic transformation. Con- sideration is given to the type of event (transient isch- emic attack versus cerebral infarct), time from stroke onset, and presence of factors associated with increased hemorrhage risk (large infarct size, uncontrolled blood pressure, hyperglycemia, thrombocytopenia, previous hemorrhagic stroke, and thrombolytic treatment).70,91 Hemorrhagic transformation of ischemic brain tissue is a relatively common occurrence that is often asymp- tomatic or minimally symptomatic and uncommonly pro- gresses in extent in the absence of predisposing fac- tors.92,93 Assuming the hemorrhagic transformation is asymptomatic and remains stable, case series support D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e615 CLINICAL STATEM ENTS AND GUIDELINES the safety of starting or continuing warfarin in carefully selected patients with a compelling indication.94 Whether optimal timing of resumption of oral anticoagulation with NOACs should follow similar recommendations is un- known. Differences in the pharmacological properties of warfarin and the NOAC must be considered, notably the more rapid time to anticoagulant effect with the NOACs (a few hours compared with 4 to 5 days for warfarin). In experimental models of ischemic stroke, neither dabiga- tran pretreatment nor continued administration of dabi- gatran after stroke onset significantly increased the risk or volume of hemorrhagic transformation after middle cerebral artery occlusion.95,96 Clinical data are anecdotal only. The phase III trials establishing the role of NOACs for stroke prevention in NVAF excluded patients within 7 to 30 days of stroke.2–5,97 In general, guidelines support withholding oral anticoagulation until 1 to 2 weeks after stroke among individuals with NVAF, with shorter times for those with transient ischemic attack or small, nondis- abling strokes and longer times for moderate to severe strokes.98–100 In NOAC-treated patients who have an AIS, compliance with NOAC therapy should be established and alternative causes for the stroke investigated. There are no data to indicate that increasing the intensity of anticoagulation, adding an antiplatelet agent, or switch- ing to another oral anticoagulant provides additional pro- tection against future ischemic events. Because of the short half-lives of NOACs and rapid decline of protective anticoagulation that occurs with missed doses, patients with poor compliance might be more appropriately man- aged with the longer-acting warfarin. TRANSITIONING BETWEEN NOACS AND OTHER ANTICOAGULANTS IN THE ACUTE CARE SETTING Indications that require considerations for the transition- ing of anticoagulants in the acute care setting include the occurrence of a new clinical event (eg, myocardial infarction) in patients on established oral anticoagulant regimens, the development of a new or worsening co- morbid medical condition (eg, renal failure) that neces- sitates an anticoagulant transition and the need for an invasive procedure. In the United States, the current la- beled prescribing information for each NOAC provides guidance for the transition to and from NOAC agents to other anticoagulants; however, these suggestions are not specific for patients in the acute care setting (Table 3).33,43,101,102 Temporary interruptions in oral anticoagulation are commonly encountered in the acute care setting. On the basis of trial observations from NOAC agents in patients with AF, approximately one third of AF patients will ex- perience the need for a temporary interruption over the course of 2 years.103–105 The association of temporary interruptions in oral anticoagulant therapy with the risk for clinical events has been reported in 3 of the clinical trials comparing NOAC agents to VKAs in patients with AF.103–105 In addition, a meta-analysis using data from trials comparing the risk of thromboembolic events as- sociated with temporary discontinuation found no sta- tistically significant differences in the NOAC versus VKA randomized groups (RR, 1.01; 95% CI, 0.68–1.49).106 Whereas the majority of the temporary interruptions in the trials were around procedures, the use of peripro- cedural bridging regimens varied on the basis of pa- tient characteristics and trial protocols. Only 6% and 11.7% of patients with temporary oral anticoagulation interruption received bridging in ROCKET AF and the ARISTOTLE trial (Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation), re- spectively.104,105 Much of the clinical outcome data regarding the switching or transitioning between NOAC agents and other anticoagulants comes from the clinical trials in pa- tients with NVAF. Observations from trials reported the risk of embolic and bleeding events in the NOAC and VKA treatment groups associated with the transitions at the beginning and end of the trials have been published. A post hoc analysis of ROCKET AF reported an increased risk of stroke in the rivaroxaban treatment group during the end-of-study transition to the open-label therapy pe- riod.107 Patients who received rivaroxaban compared with those who received warfarin were observed to have an increased incidence of stroke during the period of transi- tion (3 to 30 days after the end of the study) to open- label therapy (n=22 versus n=6; hazard ratio [HR], 3.72; 95% CI, 1.51–9.16) as well as a greater proportion of major bleeding events (n=25 versus n=7; HR, 3.62; 95% CI, 1.56–8.36).107,108 In addition, during the end-of-study transition period, the median time to first therapeutic INR was 3 days in the warfarin treatment group compared with 13 days in the rivaroxaban treatment group.108 Simi- lar observations of an increased risk of clinical events in those assigned to NOAC therapy have been reported from the ARISTOTLE trial end-of-study open-label transi- tion period.109 At the end of ARISTOTLE, a 2-day bridging period with apixaban or apixaban placebo was recom- mended during the initiation of open-label VKAs. During the first 30 days after stopping blinded study drug, 21 stroke or systemic embolism events were noted in the apixaban group versus 5 in the warfarin group (adjust- ed HR, 4.10; 95% CI, 1.54–10.86). An excess in major bleeding events was also observed during this period in the apixaban versus warfarin groups (n=26 versus n=10; adjusted HR, 2.56; 95% CI, 1.23–5.30). On the basis of these observations, an end-of-study transition plan was designed for patients enrolled in the ENGAGE AF (Effec- tive Anticoagulation With Factor Xa Next Generation in Atrial Fibrillation) trial.110 In brief, for patients who were planned to transition to open-label NOAC therapy, mea- D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e616 surement of INR was conducted and the open-label NOAC was initiated when the INR was <2.0. For patients transi- tioning to a VKA, a 14-day kit was provided that included a VKA algorithm and a modified dose of edoxaban, which was to be continued until day 14 or an open-label INR ≥2.0, whichever occurred first. Within 30 days of study drug discontinuation, strokes were observed to occur in 7 patients in each of the 3 study treatment groups with major bleeding events noted in 11 patients in the warfarin group, 10 patients in the edoxaban high-dose Table 3. Overview of US-Labeled Guidance for NOAC Anticoagulant Transitions NOAC VKA Intravenous Anticoagulant LMWH/Other NOAC Apixaban Apixaban→warfarin: Discontinue apixaban and begin a parenteral anticoagulant and warfarin at the time the next scheduled apixaban dose would have been taken Apixaban→parenteral anticoagulant: Discontinue apixaban and begin the new anticoagulant at the usual time of the next dose of apixaban Apixaban→LMWH/other NOAC: Discontinue apixaban and begin the LMWH/other NOAC at the usual time of the next dose of apixaban Warfarin→apixaban: Discontinue warfarin and start apixaban when INR <2.0 LMWH/other NOAC→apixaban: Discontinue current NOAC/LMWH and begin apixaban at the usual time of the next dose of the other NOAC/LMWH Dabigatran Dabigatran→warfarin: For CrCl ≥50 mL/min, start warfarin 3 d before discontinuing dabigatran For CrCl 30–50 mL/min, start warfarin 2 d before discontinuing dabigatran For CrCl 15–30 mL/min, start warfarin 1 d before discontinuing dabigatran Dabigatran→parenteral anticoagulant: Wait 12 h (CrCl ≥30 mL/min) or 24 h (CrCl <30 mL/min) after last dabigatran dose before initiating a parenteral anticoagulant Dabigatran→LMWH: Wait 12 h (CrCl >30 mL/min) or 24 h (CrCl <30 mL/min) after last dabigatran dose before initiating a parenteral anticoagulant Warfarin→dabigatran: Discontinue warfarin and start dabigatran when INR <2.0 UFH→dabigatran: Start dabigatran at the time of continuous infusion discontinuation LMWH→dabigatran: Start dabigatran 0–2 h before the time that the next LMWH dose would have been given Edoxaban Edoxaban→warfarin: Oral option: Reduce daily edoxaban dose by 50% and begin taking warfarin concomitantly. Measure INR at least weekly just before daily edoxaban dose. Once a stable INR ≥2.0 is achieved, discontinue edoxaban and continue warfarin Parenteral option: Discontinue edoxaban and administer a parenteral anticoagulant and warfarin at the time of the next scheduled edoxaban dose. Edoxaban→parenteral anticoagulant: Discontinue edoxaban and start the parenteral anticoagulant at the time of the next scheduled dose of edoxaban Edoxaban→LMWH/other NOAC: Discontinue edoxaban and start the LMWH/other NOAC at the time of the next scheduled dose of edoxaban Warfarin→edoxaban: Discontinue warfarin and start edoxaban when the INR is <2.5 UFH→edoxaban: Discontinue UFH infusion and start edoxaban 4 h later LMWH/other NOAC→edoxaban: Discontinue current NOAC/LMWH and start edoxaban at the time of the next scheduled other NOAC/LMWH dose Rivaroxaban Rivaroxaban→warfarin: Discontinue rivaroxaban and begin a parenteral anticoagulant and warfarin at the time the next scheduled rivaroxaban dose would have been taken Rivaroxaban→UFH: Discontinue rivaroxaban and initiate the parenteral anticoagulant at the time the next rivaroxaban dose would have been taken Rivaroxaban→LMWH/other NOAC: Discontinue rivaroxaban and start the LMWH/other NOAC at the time of the next scheduled dose of rivaroxaban Warfarin→rivaroxaban: Discontinue warfarin and start rivaroxaban as soon as INR <3.0 UFH→rivaroxaban: Stop UFH infusion and administer rivaroxaban at the same time LMWH/other NOAC→rivaroxaban: Start rivaroxaban 0–2 h before the next scheduled evening LMWH/other NOAC dose and omit administration of the LMWH/other NOAC CrCl indicates creatinine clearance; INR, international normalized ratio; LMWH, low-molecular-weight heparin; NOAC, non–vitamin K antagonist oral anticoagulant; UFH, unfractionated heparin; and VKA, vitamin K antagonist. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e617 CLINICAL STATEM ENTS AND GUIDELINES group, and 18 patients in the edoxaban low-dose group. No statistically significant differences were observed in primary efficacy or safety events among the 3 treatment groups in patients transitioning to open-label VKAs or in those transitioning to open-label NOACs.110 It is notable that in patients transitioning to open-label VKAs, 85% had at least 1 INR ≥2 by day 14. Registry data on the outcomes of ambulatory AF pa- tients transitioning from a VKA to a NOAC (dabigatran or rivaroxaban) have also been published.111,112 In a matched-cohort study of AF patients, there was no as- sociation of transitioning from a VKA to either dabigatran or rivaroxaban compared with remaining on VKA therapy for embolic or bleeding events at a median follow-up of 10 months.112 Data from a large regional prospective registry showed clinical events were relatively infrequent in the 30-day period after VKA to NOAC transitions de- spite only 75% of patients having an INR measurement before NOAC initiation. Although clinical decisions regarding the transition between anticoagulants in the acute care setting are likely to be affected by a number of factors, careful consideration should be given to strategies that mini- mize prolonged durations of both subtherapeutic and excessive anticoagulation during the transition periods. Given the relatively infrequent use of periprocedural bridging strategies during temporary interruptions in the clinical trials, clinical consideration should be given to managing patients experiencing temporary inter- ruptions without bridging, as outlined in the individual NOAC trials. PERIPROCEDURAL MANAGEMENT OF PATIENTS WHO TAKE NOACS Each year, ≈10% of patients on any long-term oral antico- agulation require surgery or other invasive procedures.113 Approximately 20% of patients on warfarin undergo sur- gery that has an extremely low risk of bleeding such as mi- nor dental, dermatologic, or ophthalmologic procedures where anticoagulation may be safely continued without interruption.114 It is recommended that warfarin be held for 5 days before surgery when significant bleeding is anticipated and then reinitiated postoperatively when he- mostasis is secured.115 Pre- and postoperative bridging using low-molecular-weight heparin is recommended for those patients with high thrombosis risk, such as those with certain mechanical valve prostheses or recent pul- monary embolism. In patients at low to intermediate risk of thrombosis, bridging low-molecular-weight heparin does not prevent thrombotic events and increases bleed- ing events116 (Figure 3). Therefore, bridging anticoagula- tion is not necessary in this subgroup of patients. The limited data available pertaining to patients on NOAC therapy who require surgery suggest that the perioperative bleeding risk is low for nonurgent sur- gery. The Dresden NOAC registry prospectively evalu- ated 2179 patients taking NOACs, of which 595 pa- tients (27.3%) underwent 863 invasive procedures; most were not urgent.48 Invasive procedures were cat- egorized as major or minor, and a bleeding event was categorized as major, clinically relevant nonmajor, or minor per the International Society of Thrombosis and Figure 3. Periprocedural management of patients on NOACs (non–vitamin K antagonist oral anticoagulants). CrCl indicates creatinine clearance; ICD, implantable cardioverter-defibrillator; PT, prothrombin time; SVT, supraventricular tachy- cardia; TE, thromboembolic event; TIA, transient ischemic attack; and VTE, venous thromboembolism. *Bridging may be considered in patients with a history of systemic embolus in the last 6 weeks.110a D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e618 Haemostasis definition.117 Of the entire cohort, only 46 patients (5.3%) experienced any bleeding complication up to 30±5 days after the procedure. Major bleeding occurred in 10 of 863 (1.2%) procedures. Clinically relevant nonmajor bleeding occurred in 29 patients (3.4%) and minor bleeding occurred in only 7 patients (0.8%). Periprocedural bleeding was studied in a sub- group analysis of the RELY trial which compared war- farin to dabigatran for stroke prevention in NVAF.103 Procedures were classified as being associated with a low (coronary angiography, defibrillator implantation) or high risk of bleeding (cardiac, abdominal, and neu- rosurgery, or procedures requiring spinal anesthesia). There was no significant difference in the rates of peri- procedural major bleeding between patients who re- ceived dabigatran 110 mg (3.8%), dabigatran 150 mg (5.1%), or warfarin (4.6%); dabigatran 110 mg versus warfarin: RR, 0.83; 95% CI, 0.59 to 1.17; P=0.28; dabigatran 150 mg versus warfarin: RR, 1.09; 95% CI, 0.80 to 1.49; P=0.58. Among patients who had ur- gent surgery, major bleeding was increased, occurring in 17.8% with dabigatran 110 mg, 17.7% with dabi- gatran 150 mg, and 21.6% with warfarin: dabigatran 110 mg: RR, 0.82; 95% CI, 0.48 to 1.41; P=0.47; dabigatran 150 mg: RR, 0.82; 95% CI, 0.50 to 1.35; P=0.44. Tailoring periprocedural NOAC management to the type of invasive procedure may mitigate against bleeding. Common clinical scenarios are subsequently discussed. Cardiac Catheterization and Percutaneous Coronary Intervention Patients with AF commonly have coexisting coronary artery disease with an estimated 20% requiring per- cutaneous coronary intervention (PCI).118 The 2012 American College of Cardiology/Society for Cardovas- cular Angiography and Interventions consensus docu- ment recommends that elective coronary angiography for patients on long-term warfarin be deferred until the INR is 1.8 for femoral artery access or <2.2 for radial artery access.119 Unfortunately, there are very limited data that address the management of patients on a NOAC who require cardiac catheterization or PCI. Pre-, peri-, and postprocedural considerations are subse- quently discussed. Preprocedural Considerations Patients with stable ischemic heart disease with ischemic symptoms despite medical therapy or with intermediate- or high-risk features on stress testing are often referred for coronary angiography and possible PCI. Patients with stable ischemic heart disease on a NOAC and who are not at high thrombosis risk should have the NOAC held until the antico- agulation effect is dissipated before undergoing coronary angiography and PCI. From the prescribing information, dabigatran should be held for at least 24 hours if CrCl ≥50 mL/min; for at least 72 hours if CrCL <50 mL/min; rivar- oxaban, apixaban and edoxaban should be held for at least 24 hours.33,43,101,102 In the absence of high risk features, patients should not be bridged with a heparin before or after the proce- dure.116 The decision to resume antithrombotic therapy after the procedure should be guided by the throm- boembolic risk as assessed by the CHA 2DS2-VASc score.120 Clinicians need to consider which antithrom- botic and antiplatelet agents to resume and the du- ration of antiplatelet therapy, balancing ischemic and thrombotic events while minimizing the hemorrhagic complications. Patients presenting with an acute coronary syn- drome (ACS) often undergo coronary angiography and revascularization to reduce their risk of recurrent events, especially if they have an elevated Thromboly- sis in Myocardial Infarction (TIMI) risk score.121 Where- as patients with unstable angina or a non–ST-segment elevation myocardial infarction do not require im- mediate angiography, patients presenting with a ST- segment elevation myocardial infarction require emer- gency coronary angiography and revascularization of the infarct related artery.122 For the unstable angina/ non–ST-segment elevation myocardial infarction pa- tient, appropriate dual antiplatelet therapy (DAPT) and heparin therapy should be started upstream, the NOAC should be discontinued and the patient should be scheduled for an urgent catheterization. In the ab- sence of electrical or hemodynamic instability, it is reasonable to wait for the effects of the NOAC to dissi- pate and then perform the procedure through a radial artery approach. Periprocedural Considerations Patients on NOACs undergoing coronary angiography or PCI will have an increased risk of hemorrhagic com- plications, and therefore, careful attention should be made to choice of vascular access site and use of ad- junctive anticoagulants. Patients should undergo radial artery access, unless there is a contraindication, be- cause the risk of bleeding and vascular complications is reduced as compared with a femoral approach.123 If a femoral approach is required, one should consider using ultrasonography and fluoroscopy to guide vas- cular access. A micropuncture needle technique may decrease the probability of a retroperitoneal bleed. Although no data exist, it may be reasonable to use a vascular closure device to assist with postproce- dure hemostasis if the patient has amenable vascular anatomy. Venous access should be avoided unless absolutely required. All patients undergoing PCI re- quire antiplatelet therapy coupled with either heparin or bivalirudin to reduce the periprocedural thrombotic complication rates, irrespective of background use of D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e619 CLINICAL STATEM ENTS AND GUIDELINES VKAs or NOACs.124 The use of intravenous glycoprotein agents should be discouraged and reserved for bailout scenarios. For patients who receive intravenous hepa- rin, one should use low-dose heparin regimens with an activated clotting time goal of ≈250 seconds to re- duce hemorrhagic complications.125 Postprocedural Considerations The clinician should consider the patient’s risk of recur- rent myocardial infarction, stent thrombosis, thromboem- bolic risk, and hemorrhagic complications when selecting anticoagulants. It is helpful to use the CHA 2DS2-VASc risk score to estimate the thromboembolic risk and the HAS- BLED risk score to estimate the hemorrhagic risk and in- clude the patient in a shared decision regarding the selec- tion of DAPT versus triple therapy as well as the duration of therapy. Several themes have emerged. The standard of care to reduce coronary ischemic events post-PCI and post-ACS is DAPT. The duration of DAPT is directly impact- ed by the stent type (bare metal stent versus drug-eluting stent) and whether the patient underwent PCI for stable ischemic heart disease or ACS.126 However, oral anti- thrombotic agents (not antiplatelet agents) are required to prevent NVAF related stroke or VTE. Therefore, the clini- cian is faced with the consideration of DAPT, DAPT plus warfarin (triple therapy), DAPT plus a NOAC (triple therapy) or warfarin plus single antiplatelet therapy. In a phase II study, triple therapy with dabigatran in patients with ACS was associated with an increased risk of bleeding complications and planned phase III trials were not pursued.127 In a randomized clinical trial of patients with ACS, apixaban increased bleeding without reducing ischemic event in patients on either DAPT or aspirin alone. Intracranial bleed rates were in- creased in patients treated with apixaban. Because of concerns regarding safety without a signal of efficacy, the trial was terminated.128 Rivaroxaban was studied in the ATLAS ACS-TIMI 51 (Anti-Xa Therapy to Lower Cardiovascular Events in Addition to Aspirin With or Without Thienopyridine Therapy in Subjects With Acute Coronary Syndrome—Thrombolysis in Myocardial In- farction) trial, which compared rivaroxaban or placebo in addition to standard ACS therapies. Compared with placebo, rivaroxaban (2.5 mg twice daily and 5.0 mg twice daily) decreased the rates of the composite pri- mary end point including cardiovascular death, myo- cardial infarction or stroke (10.7% versus 8.9%) while increasing the rates of bleeding (non–coronary artery bypass graft surgery) and ICH.129 Only rivaroxaban cou- pled with DAPT has been demonstrated to reduce isch- emic events at a cost of increased bleeding. However, the studied doses of rivaroxaban are not the doses proven to reduce the risk of thromboembolic events secondary to AF. European and Canadian guidelines suggest NOACs are preferred over warfarin when it comes to triple ther- apy. However, these recommendations are based on observational data and post hoc analysis of warfarin vs. NOAC studies with limited number of patients. For ex- ample, in ROCKET AF, only 1% of patients underwent PCI during the trial.130 Until further prospective, randomized trial data become available on the subject, the AHA writ- ing group suggests that clinicians use good judgment, weighing the risk/benefits of NOACs in the context of triple therapy for their patients. Proton pump inhibitors decrease the rates of upper gastrointestinal bleeding in patients with DAPT and in pa- tients with DAPT and antithrombotic therapy.131 Patients should be advised to avoid nonsteroidal anti-inflammato- ry medications as the risks of myocardial infarction and hemorrhagic complications are increased. Ongoing ran- domized trials (Pioneer AF-PCI [Open-Label, Randomized, Controlled, Multicenter Study Exploring Two Treatment Strategies of Rivaroxaban and a Dose-Adjusted Oral Vi- tamin K Antagonist Treatment Strategy in Subjects with Atrial Fibrillation who Undergo Percutaneous Coronary Intervention],132 RE-DUAL PCI [Evaluation of Dual Therapy With Dabigatran vs. Triple Therapy With Warfarin in Pa- tients With AF That Undergo a PCI With Stenting], RT-AF [Rivaroxaban in Patients With Atrial Fibrillation and Coro- nary Artery Disease Undergoing Percutaneous Coronary Intervention],133 SAFE-A [Safety and Effectiveness Trial of Apixaban Use in Association With Dual Antiplatelet Thera- py in Patients With Atrial Fibrillation Undergoing Percuta- neous Coronary Intervention]134 and AUGUSTUS [A Study of Apixaban in Patients With Atrial Fibrillation, Not Caused by a Heart Valve Problem, Who Are at Risk for Thrombo- sis (Blood Clots) Due to Having Had a Recent Coronary Event, Such as a Heart Attack or a Procedure to Open the Vessels of the Heart]) will assess the efficacy of a NOAC coupled with antiplatelet therapy in patients undergoing PCI. Until these trials are completed and published, the writing group makes the following suggestions: 1. For patients with a CHA 2DS2-VASc score of 0 or 1 treated with PCI, it is reasonable to omit anticoagu- lant therapy and treat with DAPT. 2. For patients who require DAPT or triple therapy, use low-dose aspirin, 81 mg daily. 3. For stable ischemic heart disease patients who require anticoagulant therapy and treatment with PCI, discontinuation of P2Y 12 inhibitor therapy after 3 months may be reasonable.126 4. For ACS patients requiring anticoagulant therapy and treatment with PCI (bare metal stent or drug- eluting stent), continuation of aspirin 81 mg daily for 1 year and discontinuation of P2Y 12 therapy after 6 months may be reasonable.126 5. For patients with a moderate to high risk of bleeding, as assessed by the HAS-BLED score, a shortened duration of triple therapy or warfarin plus clopidogrel may be considered based on the exploratory WOEST (What Is the Optimal Antiplatelet D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e620 and Anticoagulant Therapy in Patients With Oral Anticoagulation and Coronary Stenting) trial.135 6. Prasugrel and ticagrelor should not be used in conjunction with NOACs, as a result of excessive bleeding risk. 7. At present, there are limited data to recommend the routine use of NOACs, coupled with clopidogrel alone or DAPT after PCI. Of note, in clinical prac- tice, it can be challenging to reach and maintain therapeutic warfarin levels in certain patients. In these patients, it may be reasonable to combine a NOAC and clopidogrel after PCI. Cardioversion of AF Post hoc analyses from pivotal NOAC clinical trials have not shown significant differences in outcomes after car- dioversion in those treated with NOACs compared with warfarin.136–138 Meta-analysis of events across random- ized trials appears to confirm these results, finding no significant difference in stroke/systemic embolism (odds ratio, 0.73; 95% CI, 0.31–1.72) or major/non- major clinical relevant International Society on Throm- bosis and Haemostasis bleeding events (odds ratio, 1.41; 95% CI, 0.87–2.28) after cardioversion.139,140 Moreover, there is 1 randomized clinical trial of car- dioversion in patients treated with a factor Xa inhibitor versus warfarin. More than 1500 patients undergoing early (target period of 1 to 5 days after randomiza- tion with transesophageal echocardiography [TEE]) or delayed (3 to 8 weeks) cardioversion were random- ized in a 2:1 fashion to rivaroxaban or warfarin. The primary efficacy end point (composite of stroke, tran- sient ischemic attack, peripheral embolism, myocardial infarction, and cardiovascular death) occurred in 0.51% of the rivaroxaban patients versus 1.02% of the VKA- treated patients (RR, 0.50; 95% CI, 0.15–1.73) with no significant difference in bleeding observed.141 Observational data from clinical practice demonstrate similar findings. Data from a large nationwide cohort study demonstrated no difference between outcomes in those treated with dabigatran versus warfarin. In 1230 patients un- dergoing cardioversion, the cumulative incidence of stroke, bleeding or death at 30 weeks was 2.0% in those treated with warfarin and 1.0% in those treated with dabigatran (ad- justed HR, 1.33; 95% CI, 0.33–5.42).142 High-volume single- center data (>4600 cardioversions) have also failed to iden- tify any difference in postcardioversion thromboembolic or bleeding events across warfarin and NOAC agents.143 The ENSURE-AF trial (Edoxaban Versus Enoxaparin- Warfarin in Patients Undergoing Cardioversion of Atrial Fibrillation) randomized 2199 patients to either edoxaban or enoxaparin/warfarin during TEE or non-TEE guided electrical cardioversion.144 For TEE-guided cardioversion, randomization occurred <3 days from cardioversion and study treatment was continued for at least 28 days. For non–TEE-guided cardioversion, study treatment was initiat- ed at least 21 days before cardioversion and extended for at least 28 days. The primary efficacy end point (compos- ite of stroke, systemic embolic event, myocardial infarc- tion, and cardiovascular mortality) and the primary safety end point (major and clinically relevant nonmajor bleeding) occurred at a statistically similar frequency. Edoxaban may be an effective and safe alternative to enoxaparin/warfarin for patients with NVAF requiring cardioversion. Several practical considerations must be weighed when cardioverting patients on NOAC therapy with AF duration >24 hours. Similar to recommendations with warfarin, pa- tients should be anticoagulated for a minimum of 3 weeks before elective cardioversion. If not, then a TEE should be performed to exclude the presence of left atrial append- age or left atrial thrombus. Similarly, if a given patient’s adherence to therapy is suboptimal (≥2 missed doses) or in question, then a TEE should be considered. If a pa- tient has been on a properly dosed NOAC with 3 weeks of therapy and is found to have left atrial appendage or left atrial thrombus, then consideration should be given to switching to an alternate anticoagulant with special atten- tion to consistent anticoagulant use during the transition. Catheter Ablation of AF Catheter ablation is an increasingly used treatment option for rhythm control in NVAF. Because of the risks of peri- procedural thromboembolism, anticoagulation is required during the procedure. However, the presence of anticoag- ulation can make the management of bleeding complica- tions more difficult. Before the advent of NOAC therapy, observational145,146 and randomized147 studies suggested that uninterrupted VKA therapy was associated with su- perior outcomes compared with VKA interruption with intraprocedural heparin. In particular, the COMPARE (Role of Coumadin in Preventing Thromboembolism in Atrial Fi- brillation [AF] Patients Undergoing Catheter Ablation) clini- cal trial randomized 1584 patients to interrupted warfarin with bridging anticoagulation (n=790) versus continuous warfarin (n=794). Bleeding events were less common in the continuous warfarin arm with no significant difference in stroke or transient ischemic attack (0.4% versus 0.8% major bleeding, 0.5% versus 0.9% pericardial effusion, and 4% versus 22% minor bleeding).147 How interrupted/continuous NOAC therapy compares to continuous warfarin has been the subject of intense study over the past 5 years. Multiple systematic assess- ments and meta-analyses have demonstrated similar outcomes in patients treated with NOACs (interrupted or continuous) versus continuous warfarin.148–152 One randomized study compared uninterrupted rivaroxaban and VKA in 248 patients. The occurrence of any throm- boembolic events (0 versus 2) and bleeding events (21 versus 18) was similar in the uninterrupted rivaroxa- ban and VKA arms. Although the study was relatively D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e621 CLINICAL STATEM ENTS AND GUIDELINES small with limited power, the results were largely in line with previous observational data that have suggested similar outcomes with NOAC and VKA therapy. Another randomized study compared uninterrupted apixaban versus continuous warfarin in 200 subjects with drug- refractory AF undergoing ablation and found no differ- ence in thromboembolic or bleeding outcomes.153 There are several ongoing larger randomized clinical trials of interrupted versus uninterrupted NOAC therapy and con- tinuous warfarin versus continuous NOAC therapy. The recommendation to use TEE to exclude the pres- ence of left atrial appendage/left atrial thrombus should be similar regardless of whether VKA or NOAC therapy is used.154 If the patient has not had 3 to 4 weeks of pre- procedural anticoagulation or if the patient is considered at increased risk for stroke, the use of TEE is mandatory. However, many laboratories conduct a TEE in all patients before ablation since thrombus can be observed even in low-risk patients with paroxysmal AF.155 Regardless of whether continuous or interrupted NOAC therapy is used, on the basis of current consensus recom- mendations, patients should be heparinized with 100-U/ kg bolus followed by an infusion of 10 U/kg/hour before or immediately after puncture. The activated clotting time should be checked every 10 to 15 minutes until target and then every 30 minutes thereafter. The activated clot- ting time target should be at least 300 to 350 seconds or 350 to 400 seconds in the case of spontaneous echocar- diographic contrast (“smoke”) or severe left atrial enlarge- ment.154 Heparinization before transeptal access may be associated with a lower risk of asymptomatic microembol- ic events as detected by brain magnetic resonance imag- ing.156 It is important to note that the use of NOAC therapy before and during the procedure results in the need for an increased dose of heparin to achieve target activated clotting times during the ablation procedure.157 After the procedure, NOAC therapy is generally reintroduced within 4 to 8 hours after sheath removal if access site hemo- stasis has been achieved. Consistent with consensus rec- ommendations, NOAC therapy should be continued for a minimum of 2 to 3 months after ablation. Thereafter, oral anticoagulation should be based on the patient’s underly- ing risk for stroke (CHA 2DS2-VASc score) rather than the current rhythm. Electronic Device Implantation Management of oral anticoagulation surrounding cardiac implantable electronic device insertion presents several challenges. Oral anticoagulation increases the risk of bleeding and hematoma formation after device implan- tation. Furthermore, hematoma formation increases the risk of postoperative infection. Based upon the results from randomized clinical trials, uninterrupted warfarin has been shown to lead to less bleeding and superior outcomes compared with interrupted warfarin and par- enteral bridging therapy.158,159 These findings are also consistent with the BRIDGE (Perioperative Bridging Anti- coagulation in Patients With Atrial Fibrillation) trial, which found no significant benefit to bridging for general inter- ruption of oral anticoagulation for invasive procedures in patients with NVAF.116 However, the optimal manage- ment of NOAC therapy surrounding cardiac implantable electronic device implantation remains unknown. In general, discontinuation of NOAC therapy before cardiac implantable electronic device implantation in a manner consistent with the elimination half-life is the most common practice pattern. For apixaban, edoxa- ban, and rivaroxaban, this would include discontinuation 24 hours in advance of the procedure. In the case of dabigatran, discontinuation is recommended 24 hours before in patients with a CrCl ≥80 mL/min, 36 hours be- fore in those with CrCl 50 to 79 mL/min, and 48 hours before in those with a CrCl <50 mL/min.160 Survey data from implanting physicians suggest wide variation in practice patterns reflecting the uncertainly over optimal management.161 However, the majority of physicians discontinue NOAC therapy at the time of implantation (82%).161 Although uninterrupted warfarin has the best evidence base (>1 randomized trial), an increasing num- ber of cardiac implantable electronic device patients are taking NOAC therapy. Whether NOAC therapy can be continued through cardiac implantable electronic de- vice implantation remains debated and is the subject of a large clinical trial (BRUISE CONTROL-2 [Strategy of Continued vs Interrupted Novel Oral Anticoagulant at Time of Device Surgery in Patients With Moderate to High Risk of Arterial Thromboembolic Events] study) in which perioperative management will be randomized to a strategy of continued versus interrupted NOAC thera- py. The few available observational data are limited by their small cohort size but have not identified significant risks of bleeding with uninterrupted NOAC therapy.162,163 When a decision is made to interrupt NOAC therapy for cardiac implantable electronic device implantation, the implanting physician must decide when the NOAC therapy should be restarted. This decision is often influ- enced by patient characteristics, including risk factors for bleeding and the postimplantation physical examina- tion (eg, hematoma). Similar to discontinuation, prac- tice patterns regarding resumption of NOAC therapy after implantation are highly variable.161 Typically, NOAC therapy was restarted 24 to 48 hours after surgical pro- cedures in the pivotal NOAC trials. Patients with multiple risk factors for bleeding, concomitant antiplatelet ther- apy, or evidence of hematoma on their postoperative examination may benefit from a greater delay to NOAC resumption (3 to 5 days). However, given the lack of evidence to guide these decisions, management should be approached on a patient-by-patient basis, weighing the risks and benefits of earlier versus later resumption of NOAC therapy. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e622 Cardiovascular Surgery There is limited information regarding the use of NOACs in coronary artery bypass grafting or valve replacement surgery. At present, information related to perioperative NOAC use in cardiac surgery is anecdotal or based on limited subset analyses.48,164,165 No significant bleeding event differences were observed between rivaroxaban and warfarin treated patients who underwent cardiac surgery in ROCKET AF.110a The ATLAS ACS-2-TIMI-51 trial tested rivaroxaban to lower cardiovascular events in patients with ACS and reported 10 patients undergoing coronary artery bypass grafting after ST-segment eleva- tion myocardial infarction.129,166 Per the trial protocol, the drug was stopped 12 hours before the procedure and resumed 12 hours after the postprocedural drains were removed or after the last dose of parenteral anticoagu- lant therapy had been administered. The results specific to this group were not reported; therefore, no conclu- sions regarding coronary artery bypass grafting–related care can be made. Established indications for NOACs in the pericardiac surgery setting include stroke prevention in preoperative AF, prolonged or frequent postoperative AF, and VTE treatment. NOAC use is contraindicated in patients with mechanical valves; as the RE-ALIGN (The Randomized, Phase II Study to Evaluate the Safety and Pharmacoki- netics of Oral Dabigatran Etexilate in Patients After Heart Valve Replacement) trial with dabigatran demonstrated, there is an increased rate of thromboembolic and bleed- ing complications compared with warfarin.167 There are case reports of dabigatran use after left ventricular assist device placement168 and of rivaroxaban use for heparin induced thrombocytopenia after coronary artery bypass grafting.169 However, these off-label uses are not supported by available clinical trial evidence. For cardiac surgery, NOACs should be stopped in the perioperative setting and restarted after clinical hemostasis has been established. As cardiac surgery is considered a high-bleeding-risk procedure, surgery should be postponed if at all possible until after the appropriate interruption period. Bleeding after car- diac surgery should be monitored via standard post- procedure drains. Life-threatening bleeding should be treated with supportive therapy, including transfusion of blood products and administration of antifibrinolyt- ics as indicated for hemorrhage resuscitation, and return to the operating room. If contributing to an on- going coagulopathy, administration of NOAC antidotes as previously described (Laboratory Measurement of NOAC Effect) could be considered. Mild bleeding may be monitored, but NOACs should not be reinitiated until there is bleeding control. Similarly, the published experience of NOAC manage- ment in patients undergoing vascular surgery is limited to case reports and very small trial subsets.48,164,165 In a subgroup analysis of ROCKET AF, patients with periph- eral artery disease on rivaroxaban had a higher risk of major bleeding and nonmajor clinically relevant bleeding compared with warfarin.170 Noncardiovascular Surgery Studies examining outcomes among NOAC users af- ter noncardiovascular surgery largely grouped patients into cohorts spanning minor to major high-risk sur- gery.48,103,164,171 NOACs do not increase the rate of post- operative bleeding events when compared with warfarin. A pooled analysis of dabigatran phase III trial bleeding data demonstrated no difference in postoperative bleed- ing events between patients on dabigatran and warfa- rin.171 In the ARISTOTLE trial, there was no difference in stroke, myocardial infarction, mortality, or bleeding for patients on apixaban versus warfarin for NVAF.105 How- ever, small differences may not have been detected as only 2.9% of procedures in this trial were considered emergent and only 10.2% of procedures were consid- ered major. Bridging therapy is not recommended during NOAC therapy interruption for patients undergoing surgery. The dabigatran RE-LY study demonstrated an increased risk for major bleeding with bridging therapy.165 Non- bridged patients had a thromboembolic event risk of 0.6%. Analysis of periprocedural dabigatran use in the RE-LY trial demonstrated no difference in major bleed- ing events between urgent versus elective surgery and major versus minor surgery.103 There was also no difference in fatal bleeding, reoperation as a result of bleeding, or transfusion requirements. There were few- er bleeding events in patients with shorter interruption periods, though this may not be a causal relationship given that shorter interruptions may indicate patients with characteristics of faster drug clearance. In con- trast, analysis of the Dresden NOAC registry demon- strated increased risk of bleeding in patients with major procedures.48 Heparin bridging still did not reduce car- diovascular events and did not statistically affect bleed- ing risk once the data were adjusted for major versus minor procedures.48 In the Canadian dabigatran cohort study, none of the 541 patients received preoperative bridging, and only 1.7% of patients received postopera- tive heparin or low-molecular-weight heparin. Despite this, there was only 1 transient ischemic attack event (0.2%) and no major arterial thromboembolic events. In the ARISTOTLE study, 37.5% of procedures did not re- quire NOAC interruption and 11.7% of patients received bridging anticoagulation.105 In phase III trials of NOAC use for VTE prevention in high-bleeding-risk orthopedic surgery, the first pro- phylactic dose was administered 6 to 12 hours post- operatively.172 Real-world registries of NOAC use after orthopedic surgery suggest higher rates of bleeding D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e623 CLINICAL STATEM ENTS AND GUIDELINES compared with those observed in the trials. In the Dres- den NOAC registry, 6 out of 42 patients undergoing major orthopedic surgery developed major cardiovas- cular (n=2) or bleeding events (n=4).48 In the Canadian dabigatran cohort, 5 out of 19 patients undergoing major orthopedic surgery developed major bleeding complications.164 Caution should be exercised in man- aging patients on NOACs who require major orthopedic interventions. Neuraxial Anesthesia Spinal or epidural hematoma can be a devastating com- plication of neuraxial anesthesia. There are limited data pertaining to the interval between the discontinuation of NOACs, the neuraxial anesthesia procedure itself, and subsequent resumption of the NOAC. Rivaroxaban to prevent VTE after total knee joint replacement or total hip arthroplasty with neuraxial anesthesia has also been examined. In an analysis of 4 trials, neuraxial hematoma occurred in only 1 of 4086 patients in the rivaroxaban group and this occurred before drug administration.173 Of the 2550 patients who underwent neuraxial anesthe- sia in the rivaroxaban group in a phase IV cohort study, 1 patient developed intraspinal/hemorrhagic puncture.174 These data suggest that the incidence of neuraxial he- matoma is low despite concurrent administration of therapeutic doses of a NOAC. There are no robust clinical outcomes data to address the timing and safety of NOAC discontinuation and rein- stitution. The American Society of Regional Anesthesia and European Society of Regional Anesthesia and Pain Therapy recommend stopping dabigatran 4 to 5 days before neuraxial block.175 For patients with end-stage renal disease, 6 days off dabigatran is recommended. For patients with high risk of VTE, dabigatran may be administered 12 hours after the pain intervention. This group recommends stopping apixaban and rivaroxaban 3 to 5 days before neuroaxial block, and resuming either drug 12 hours after the pain intervention if the risk of VTE is considered high. No guidance on edoxaban was considered in this document. These recommendations are controversial because discontinuation periods of ≥4 days are inconsistent with the return to hemostasis time of these agents, which may expose patients to excess thromboembolic risk. CONCLUSION NOACs are no longer novel and are now commonly used in day-to-day medical practice. Healthcare providers are encouraged to use well-defined protocols established in collaborations with multiple professional disciplines to address NOAC dose and continuation or cessation when invasive procedures are required. Such protocols should also be encouraged to assist acute care providers who manage bleeding while patients take NOACs. Simple to administer antidotes are either approved for use, such as idarucizumab for dabigatran, or are currently under investigation. Further studies that measure clinical out- comes after NOAC reversal are needed to optimize pro- tocols for NOAC-associated bleeding and periprocedural NOAC management. ACKNOWLEDGMENTS The AHA writing group thanks Taylor Klein for assistance in reference formatting and Anne Leonard and Connie Land for administrative assistance. FOOTNOTES The American Heart Association makes every effort to avoid any actual or potential conflicts of interest that may arise as a result of an outside relationship or a personal, professional, or business interest of a member of the writing panel. Specifically, all members of the writing group are required to complete and submit a Disclosure Questionnaire showing all such relationships that might be perceived as real or potential conflicts of interest. This statement was approved by the American Heart Asso- ciation Science Advisory and Coordinating Committee on Octo- ber 10, 2016, and the American Heart Association Executive Committee on October 25, 2016. A copy of the document is available at http://professional.heart.org/statements by using either “Search for Guidelines & Statements” or the “Browse by Topic” area. To purchase additional reprints, call 843-216- 2533 or e-mail kelle.ramsay@wolterskluwer.com. The American Heart Association requests that this docu- ment be cited as follows: Raval AN, Cigarroa JE, Chung MK, Diaz-Sandoval LJ, Diercks D, Piccini JP, Jung HS, Washam JB, Welch BG, Zazulia AR, Collins SP; on behalf of the American Heart Association Clinical Pharmacology Subcommittee of the Acute Cardiac Care and General Cardiology Committee of the Council on Clinical Cardiology; Council on Cardiovascu- lar Disease in the Young; and Council on Quality of Care and Outcomes Research. Management of patients on non–vitamin K antagonist oral anticoagulants in the acute care and peri- procedural setting: a scientific statement from the American Heart Association. Circulation. 2017;135:e604–e633. doi: 10.1161/CIR.0000000000000477. Expert peer review of AHA Scientific Statements is conducted by the AHA Office of Science Operations. For more on AHA state- ments and guidelines development, visit http://professional. heart.org/statements. Select the “Guidelines & Statements” drop-down menu, then click “Publication Development.” Permissions: Multiple copies, modification, alteration, en- hancement, and/or distribution of this document are not permit- ted without the express permission of the American Heart Asso- ciation. Instructions for obtaining permission are located at http:// www.heart.org/HEARTORG/General/Copyright-Permission- Guidelines_UCM_300404_Article.jsp. A link to the “Copyright Per- missions Request Form” appears on the right side of the page. Circulation is available at http://circ.ahajournals.org. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 mailto:kelle.ramsay@wolterskluwer.com Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e624 Writing Group Disclosures Writing Group Member Employment Research Grant Other Research Support Speakers’ Bureau/ Honoraria Expert Witness Ownership Interest Consultant/ Advisory Board Other Amish N. Raval University of Wisconsin None None None None None None None Sean P. Collins Vanderbilt University College of Medicine None None None None None None None Mina K. Chung Cleveland Clinic None None None None None None None Joaquin E. Cigarroa Oregon Health and Science University None None None None None None None Larry J. Diaz- Sandoval Michigan State University None None None None None None None Deborah Diercks UT Southwestern Emergency Medicine None Johnson & Johnson* (Steering Committee member on a study about the treatment of PE using NOACS) None None None None None Hee Soo Jung University of Wisconsin NIH(National Center for Advancing Translational Sciences)*; National Board of Medical Examiners* None None None None None None Jonathan P. Piccini Duke University Janssen Pharmaceuticals† None None None None Janssen Pharmaceuticals*; Medtronic*; BMS Pfizer* None Jeffrey B. Washam Duke University Heart Center None None None None None None None Babu G. Welch UT Southwestern Medical Center St. Paul Medical Foundation† None None None None Stryker Neurovascular*; Covidien* None Allyson R. Zazulia Washington University None None None None None None None This table represents the relationships of writing group members that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all members of the writing group are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition. *Modest. †Significant. DISCLOSURES D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e625 CLINICAL STATEM ENTS AND GUIDELINES REFERENCES 1. Barnes GD, Ageno W, Ansell J, Kaatz S; Subcommittee on the Control of Anticoagulation of the International Society on Throm- bosis and Haemostasis. Recommendation on the nomenclature for oral anticoagulants: communication from the SSC of the ISTH. J Thromb Haemost. 2015;13:1154–1156. doi: 10.1111/ jth.12969. 2. Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A, Pogue J, Reilly PA, Themeles E, Varrone J, Wang S, Alings M, Xavier D, Zhu J, Diaz R, Lewis BS, Darius H, Diener H-C, Joyner CD, Wallentin L; RE-LY Steering Committee and Investigators. Dabi- gatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361:1139–1151. doi: 10.1056/NEJMoa0905561. 3. Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, Breithardt G, Halperin JL, Hankey GJ, Piccini JP, Becker Reviewer Disclosures Reviewer Employment Research Grant Other Research Support Speakers’ Bureau/ Honoraria Expert Witness Ownership Interest Consultant/ Advisory Board Other Tyler W. Barrett Vanderbilt University NIH NHLBI (PI on a K23 studying A Fib that ended in Sept 2015)†; Janssen (Site PI for a multicenter international ORBIT- AF II registry)†; Boehringer Ingelheim (PI for a multicenter retrospective cohort that completed in early 2016)* None None None None Boehringer Ingelheim† None John W. Eikelboom McMaster University (Canada) BMS†; Boehringer Ingelheim†; Pfizer†; Bayer† None BMS†; Boehringer Ingelheim†; Pfizer†; Bayer†; Janssen*; Daiichi Sankyo* None None None None David A. Garcia University of Washington Daiichi Sankyo (Local PI and St Committee member for clinical trial involving edoxaban)*; Janssen (Local PI for clinical trial)* None None Pfizer*; BMS* None Pfizer*; BMS*; Boehringer Ingelheim* None Gregory Y.H. Lip City Hospital (United Kingdom) None None Bayer†; BMS/ Pfizer†; Medtronic†; Boehringer Ingelheim†; Microlife†; Roche†; Daiichi-Sankyo† None None Bayer/Janssen†; Astellas†; Merck†; Sanofi†; BMS/ Pfizer†; Biotronik†; Medtronic†; Portola†; Boehringer Ingelheim†; Microlife†; Daiichi- Sankyo† None Jeffrey I. Weitz Thrombosis & Atherosclerosis Research Institute (Canada) None None None None None None None This table represents the relationships of reviewers that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all reviewers are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition. *Modest. †Significant. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e626 RC, Nessel CC, Paolini JF, Berkowitz SD, Fox KA, Califf RM; ROCKET AF Steering Committee, for the ROCKET AF Inves- tigators. Rivaroxaban versus warfarin in nonvalvular atrial fi- brillation. N Engl J Med. 2011;365:883–891. doi: 10.1056/ NEJMoa1009638. 4. Granger CB, Alexander JH, McMurray JJ, Lopes RD, Hylek EM, Hanna M, Al-Khalidi HR, Ansell J, Atar D, Avezum A, Bahit MC, Diaz R, Easton JD, Ezekowitz JA, Flaker G, Garcia D, Geraldes M, Gersh BJ, Golitsyn S, Goto S, Hermosillo AG, Hohnloser SH, Horowitz J, Mohan P, Jansky P, Lewis BS, Lopez-Sendon JL, Pais P, Parkhomenko A, Verheugt FW, Zhu J, Wallentin L; ARISTOTLE Committees and Investigators. Apixaban versus warfarin in pa- tients with atrial fibrillation. N Engl J Med. 2011;365:981–992. doi: 10.1056/NEJMoa1107039. 5. Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL, Waldo AL, Ezekowitz MD, Weitz JI, Špinar J, Ruzyllo W, Ruda M, Koretsune Y, Betcher J, Shi M, Grip LT, Patel SP, Patel I, Hanyok JJ, Mercuri M, Antman EM; ENGAGE AF-TIMI 48 Investigators. Edoxa- ban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093–2104. doi: 10.1056/NEJMoa1310907. 6. Piccini JP, Hellkamp AS, Washam JB, Becker RC, Breithardt G, Berkowitz SD, Halperin JL, Hankey GJ, Hacke W, Mahaffey KW, Nessel CC, Singer DE, Fox KA, Patel MR. Polypharmacy and the efficacy and safety of rivaroxaban versus warfarin in the preven- tion of stroke in patients with nonvalvular atrial fibrillation. Cir- culation. 2016;133:352–360. doi: 10.1161/CIRCULATIONAHA. 115.018544. 7. Levy JH, Faraoni D, Spring JL, Douketis JD, Samama CM. Man- aging new oral anticoagulants in the perioperative and intensive care unit setting. Anesthesiology. 2013;118:1466–1474. doi: 10.1097/ALN.0b013e318289bcba. 8. Gehrie E, Tormey C. Novel oral anticoagulants: efficacy, labora- tory measurement, and approaches to emergent reversal. Arch Pathol Lab Med. 2015;139:687–692. doi: 10.5858/arpa.2013- 0677-RS. 9. Cuker A, Siegal DM, Crowther MA, Garcia DA. Laboratory mea- surement of the anticoagulant activity of the non-vitamin K oral anticoagulants. J Am Coll Cardiol. 2014;64:1128–1139. doi: 10.1016/j.jacc.2014.05.065. 10. Mar PL, Familtsev D, Ezekowitz MD, Lakkireddy D, Gopinathannair R. Periprocedural management of anticoagulation in patients tak- ing novel oral anticoagulants: review of the literature and recom- mendations for specific populations and procedures. Int J Cardiol. 2016;202:578–585. doi: 10.1016/j.ijcard.2015.09.035. 11. Pollack CV Jr, Reilly PA, Eikelboom J, Glund S, Verhamme P, Bern- stein RA, Dubiel R, Huisman MV, Hylek EM, Kamphuisen PW, Kreu- zer J, Levy JH, Sellke FW, Stangier J, Steiner T, Wang B, Kam CW, Weitz JI. Idarucizumab for dabigatran reversal. N Engl J Med. 2015;373:511–520. doi: 10.1056/NEJMoa1502000. 12. Eerenberg ES, Kamphuisen PW, Sijpkens MK, Meijers JC, Buller HR, Levi M. Reversal of rivaroxaban and dabigatran by prothrom- bin complex concentrate: a randomized, placebo-controlled, crossover study in healthy subjects. Circulation. 2011;124:1573– 1579. doi: 10.1161/CIRCULATIONAHA.111.029017. 13. Claisse G, Delavenne X, Masson I, Maillard N, Alamartine E, Mariat C. Venovenous haemodiafiltration for the management of dabiga- tran overdose in intensive care unit. Clin Kidney J. 2015;8:199– 201. doi: 10.1093/ckj/sfv001. 14. Chen BC, Sheth NR, Dadzie KA, Smith SW, Nelson LS, Hoffman RS, Winchester JF. Hemodialysis for the treatment of pulmo- nary hemorrhage from dabigatran overdose. Am J Kidney Dis. 2013;62:591–594. doi: 10.1053/j.ajkd.2013.02.361. 15. Siegal DM, Curnutte JT, Connolly SJ, Lu G, Conley PB, Wiens BL, Mathur VS, Castillo J, Bronson MD, Leeds JM, Mar FA, Gold A, Crowther MA. Andexanet alfa for the reversal of factor Xa inhibi- tor activity. N Engl J Med. 2015;373:2413–2424. doi: 10.1056/ NEJMoa1510991. 16. Connolly SJ, Milling TJ Jr, Eikelboom JW, Gibson CM, Curnutte JT, Gold A, Bronson MD, Lu G, Conley PB, Verhamme P, Schmidt J, Middeldorp S, Cohen AT, Beyer-Westendorf J, Albaladejo P, Lopez- Sendon J, Goodman S, Leeds J, Wiens BL, Siegal DM, Zotova E, Meeks B, Nakamya J, Lim WT, Crowther M; ANNEXA-4 Investi- gators. Andexanet alfa for acute major bleeding associated with factor Xa inhibitors. N Engl J Med. 2016;375:1131–1141. doi: 10.1056/NEJMoa1607887. 17. Perzborn E, Heitmeier S, Laux V, Buchmüller A. Reversal of rivaroxaban-induced anticoagulation with prothrombin com- plex concentrate, activated prothrombin complex concentrate and recombinant activated factor VII in vitro. Thromb Res. 2014;133:671–681. doi: 10.1016/j.thromres.2014.01.017. 18. Stevenson JW, Minns AB, Smollin C, Albertson TE, Cantrell FL, Tomaszewski C, Clark RF. An observational case series of dabi- gatran and rivaroxaban exposures reported to a poison control system. Am J Emerg Med. 2014;32:1077–1084. doi: 10.1016/j. ajem.2014.04.031. 19. Lehmann T, Hofer KE, Baumann M, Hasler K, Ceschi A, Kupfer- schmidt H, Rohde G, Korte W. Massive human rivaroxaban over- dose. Thromb Haemost. 2014;112:834–836. doi: 10.1160/ TH14-02-0138. 20. Dibu JR, Weimer JM, Ahrens C, Manno E, Frontera JA. The role of FEIBA in reversing novel oral anticoagulants in intracerebral hemorrhage. Neurocrit Care. 2015;24:413–419. doi: 10.1007/ s12028-015-0213-y. 21. Zahir H, Brown KS, Vandell AG, Desai M, Maa JF, Dishy V, Lom- eli B, Feussner A, Feng W, He L, Grosso MA, Lanz HJ, Antman EM. Edoxaban effects on bleeding following punch biopsy and reversal by a 4-factor prothrombin complex concentrate. Circulation. 2015;131:82–90. doi: 10.1161/CIRCULATIONAHA. 114.013445. 22. Ansell JE, Bakhru SH, Laulicht BE, Steiner SS, Grosso M, Brown K, Dishy V, Noveck RJ, Costin JC. Use of PER977 to reverse the an- ticoagulant effect of edoxaban. N Engl J Med. 2014;371:2141– 2142. doi: 10.1056/NEJMc1411800. 23. Ansell JE, Laulicht BE, Bakhru SH, Hoffman M, Steiner SS, Cos- tin JC. Ciraparantag safely and completely reverses the antico- agulant effects of low molecular weight heparin. Thromb Res. 2016;146:113–118. doi: 10.1016/j.thromres.2016.07.008. 24. Ruff CT, Giugliano RP, Braunwald E, Hoffman EB, Deenadayalu N, Ezekowitz MD, Camm AJ, Weitz JI, Lewis BS, Parkhomenko A, Yamashita T, Antman EM. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet. 2014;383:955–962. doi: 10.1016/S0140-6736(13)62343-0. 25. Linkins L, O’Donnell M, Julian JA, Kearon C. Intracranial and fatal bleeding according to indication for long-term oral antico- agulant therapy. J Thromb Haemost. 2010;8:2201–2207. doi: 10.1111/j.1538-7836.2010.04016.x. 26. Hemphill JC 3rd, Greenberg SM, Anderson CS, Becker K, Bendok BR, Cushman M, Fung GL, Goldstein JN, Macdonald RL, Mitch- ell PH, Scott PA, Selim MH, Woo D. Guidelines for the manage- ment of spontaneous intracerebral hemorrhage: a guideline for healthcare professionals from the American Heart Association/ American Stroke Association. Stroke. 2015;46:2032–2060. doi: 10.1161/STR.0000000000000069. 27. Hemphill JC 3rd, Farrant M, Neill TA Jr. Prospective validation of the ICH score for 12-month functional outcome. Neurology. 2009;73:1088–1094. doi: 10.1212/WNL.0b013e3181b8b332. 28. Wada R, Aviv RI, Fox AJ, Sahlas DJ, Gladstone DJ, Tomlinson G, Sy- mons SP. CT angiography “spot sign” predicts hematoma expan- sion in acute intracerebral hemorrhage. Stroke. 2007;38:1257– 1262. doi: 10.1161/01.STR.0000259633.59404.f3. 29. Chakraborty S, Blacquiere D, Lum C, Stotts G. Dynamic nature of the CT angiographic “spot sign”. Br J Radiol. 2010;83:e216– e219. doi: 10.1259/bjr/74416385. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e627 CLINICAL STATEM ENTS AND GUIDELINES 30. Sakamoto Y, Koga M, Yamagami H, Okuda S, Okada Y, Kimura K, Shiokawa Y, Nakagawara J, Furui E, Hasegawa Y, Kario K, Arihiro S, Sato S, Kobayashi J, Tanaka E, Nagatsuka K, Minematsu K, Toyoda K; SAMURAI Study Investigators. Systolic blood pressure after intravenous antihypertensive treatment and clinical outcomes in hyperacute intracerebral hemorrhage: the Stroke Acute Management with Urgent Risk-Factor Assessment and Improvement-Intracerebral Hemorrhage study. Stroke. 2013;44:1846–1851. doi: 10.1161/STROKEAHA.113.001212. 31. Rodriguez-Luna D, Piñeiro S, Rubiera M, Ribo M, Coscojuela P, Pagola J, Flores A, Muchada M, Ibarra B, Meler P, Sanjuan E, Hernandez-Guillamon M, Alvarez-Sabin J, Montaner J, Molina CA. Impact of blood pressure changes and course on hema- toma growth in acute intracerebral hemorrhage. Eur J Neurol. 2013;20:1277–1283. doi: 10.1111/ene.12180. 32. Anderson CS, Heeley E, Huang Y, Wang J, Stapf C, Delcourt C, Lindley R, Robinson T, Lavados P, Neal B, Hata J, Arima H, Parsons M, Li Y, Wang J, Heritier S, Li Q, Woodward M, Simes RJ, Davis SM, Chalmers J; INTERACT2 Investigators. Rapid blood-pressure lowering in patients with acute intracerebral hem- orrhage. N Engl J Med. 2013;368:2355–2365. doi: 10.1056/ NEJMoa1214609. 32a. Qureshi AI, Palesch YY, Barsan WG, Hanley DF, Hsu CY, Martin RL, Moy CS, Silbergleit R, Steiner T, Suarez JI, Toyoda K, Wang Y, Yamamoto H, Yoon B-W; for the ATACH-2 Trial Investigators and the Neurological Emergency Treatment Trials Network. Intensive blood- pressure lowering in patients with acute cerebral hemorrhage. N Engl J Med. 2016;375:1033–1043. doi: 10.1056/NEJMoa1603460. 33. Lip GY, Lane DA. Assessing bleeding risk in atrial fibrillation with the HAS-BLED and ORBIT scores: clinical application re- quires focus on the reversible bleeding risk factors. Eur Heart J. 2015;36:3265–3267. doi: 10.1093/eurheartj/ehv415. 34. Verheugt FW, Granger CB. Oral anticoagulants for stroke pre- vention in atrial fibrillation: current status, special situations, and unmet needs. Lancet. 2015;386:303–310. doi: 10.1016/ S0140-6736(15)60245-8. 35. Caldeira D, Barra M, Pinto FJ, Ferreira JJ, Costa J. Intracranial hemorrhage risk with the new oral anticoagulants: a systematic review and meta-analysis. J Neurol. 2015;262:516–522. doi: 10.1007/s00415-014-7462-0. 36. Dossett LA, Riesel JN, Griffin MR, Cotton BA. Prevalence and implications of preinjury warfarin use: an analysis of the National Trauma Databank. Arch Surg. 2011;146:565–570. doi: 10.1001/archsurg.2010.313. 37. Durie R, Kohute M, Fernandez C, Knight M. Prothrombin complex concentrate for the management of severe traumatic bleeding in a patient anticoagulated with apixaban. J Clin Pharm Ther. 2016;41:92–93. doi: 10.1111/jcpt.12339. 38. Cotton BA, McCarthy JJ, Holcomb JB. Acutely injured patients on dabigatran. N Engl J Med. 2011;365:2039–2040. doi: 10.1056/NEJMc1111095. 39. Croft PE, Cabral KP, Strout TD, Baumann MR, Gibbs MA, Delaney MC. Managing blunt trauma in patients receiving dabigatran etexilate: case study and review of the literature. J Emerg Nurs. 2013;39:302–308. doi: 10.1016/j.jen.2013.01.016. 40. Huang GS, Chance EA. When dabigatran and trauma collide. Am Surg. 2013;79:113–114. 41. Joseph B, Ditillo M, Pandit V, Aziz H, Sadoun M, Hays D, Davis K, Friese R, Rhee P. Dabigatran therapy: minor trauma injuries are no longer minor. Am Surg. 2014;80:E116–E118. 42. Obeng-Gyasi S, Loor MM, Samotowka MA, Moorman ML. Management of dabigatran-induced anticoagulation in trauma and acute care surgery patients. J Trauma Acute Care Surg. 2012;73:1064–1069. doi: 10.1097/TA.0b013e31827019c9. 43. Committee on Trauma, American College of Surgeons. Advanced Trauma Life Support Student Course Manual. Chicago, IL: American College of Surgeons; 2012. 44. Dias JD, Norem K, Doorneweerd DD, Thurer RL, Popovsky MA, Omert LA. Use of thromboelastography (TEG) for detection of new oral anticoagulants. Arch Pathol Lab Med. 2015;139:665–673. doi: 10.5858/arpa.2014-0170-OA. 45. Oswald E, Velik-Salchner C, Innerhofer P, Tauber H, Auckenthaler T, Ulmer H, Streif W. Results of rotational thromboelastometry, co- agulation activation markers and thrombin generation assays in or- thopedic patients during thromboprophylaxis with rivaroxaban and enoxaparin: a prospective cohort study. Blood Coagul Fibrinolysis. 2015;26:136–144. doi: 10.1097/MBC.0000000000000203. 46. Sherwood MW, Nessel CC, Hellkamp AS, Mahaffey KW, Piccini JP, Suh EY, Becker RC, Singer DE, Halperin JL, Hankey GJ, Berkowitz SD, Fox KA, Patel MR. Gastrointestinal bleeding in patients with atrial fibrilla- tion treated with rivaroxaban or warfarin: ROCKET AF Trial. J Am Coll Cardiol. 2015;66:2271–2281. doi: 10.1016/j.jacc.2015.09.024. 47. Fang MC, Go AS, Chang Y, Hylek EM, Henault LE, Jensvold NG, Singer DE. Death and disability from warfarin-associated intracra- nial and extracranial hemorrhages. Am J Med. 2007;120:700– 705. doi: 10.1016/j.amjmed.2006.07.034. 48. Beyer-Westendorf J, Gelbricht V, Förster K, Ebertz F, Köhler C, Werth S, Kuhlisch E, Stange T, Thieme C, Daschkow K, Weiss N. Peri-interventional management of novel oral anticoagulants in dai- ly care: results from the prospective Dresden NOAC registry. Eur Heart J. 2014;35:1888–1896. doi: 10.1093/eurheartj/eht557. 49. Staerk L, Lip GY, Olesen JB, Fosbøl EL, Pallisgaard JL, Bonde AN, Gundlund A, Lindhardt TB, Hansen ML, Torp-Pedersen C, Gislason GH. Stroke and recurrent haemorrhage associated with antithrom- botic treatment after gastrointestinal bleeding in patients with atrial fibrillation: nationwide cohort study. BMJ. 2015;351:h5876. doi: 10.1136/bmj.h5876. 50. Conway SE, Schaeffer SE, Harrison DL. Evaluation of dabigatran exposures reported to poison control centers. Ann Pharmacother. 2014;48:354–360. doi: 10.1177/1060028013513883. 51. Spiller HA, Mowry JB, Aleguas A Jr, Griffith JR, Goetz R, Ryan ML, Bangh S, Klein-Schwartz W, Schaeffer S, Casavant MJ. An observational study of the factor Xa inhibitors rivaroxaban and apixaban as reported to eight poison centers. Ann Emerg Med. 2016;67:189–195. doi: 10.1016/j.annemergmed.2015.07.014. 52. Alikhan R, Rayment R, Keeling D, Baglin T, Benson G, Green L, Marshall S, Patel R, Pavord S, Rose P, Tait C. The acute manage- ment of haemorrhage, surgery and overdose in patients receiv- ing dabigatran. Emerg Med J. 2014;31:163–168. doi: 10.1136/ emermed-2012-201976. 53. Chang DN, Dager WE, Chin AI. Removal of dabigatran by hemo- dialysis. Am J Kidney Dis. 2013;61:487–489. doi: 10.1053/j. ajkd.2012.08.047. 54. Woo JS, Kapadia N, Phanco SE, Lynch CA. Positive outcome after intentional overdose of dabigatran. J Med Toxicol. 2013;9:192– 195. doi: 10.1007/s13181-012-0276-5. 55. Sajkov D, Gallus A. Accidental rivaroxaban overdose in a patient with pulmonary embolism: some lessons for managing new oral anticoagulants. Clin Med Insights Case Rep. 2015;8:57–59. doi: 10.4137/CCRep.S27992. 56. Chiew AL, Khamoudes D, Chan BS. Use of continuous veno-venous haemodiafiltration therapy in dabigatran overdose. Clin Toxicol (Phi- la). 2014;52:283–287. doi: 10.3109/15563650.2014.900179. 57. Fountzilas C, George J, Levine R. Dabigatran overdose sec- ondary to acute kidney injury and amiodarone use. N Z Med J. 2013;126:110–112. 58. Linkins LA, Moffat K. Monitoring the anticoagulant effect af- ter a massive rivaroxaban overdose. J Thromb Haemost. 2014;12:1570–1571. doi: 10.1111/jth.12669. 59. Montaruli B, Erroi L, Vitale C, Berutti S, Cosseddu D, Sivera P, Co- glitore R, Marangella M, Migliardi M. Dabigatran overdose: case report of laboratory coagulation parameters and hemodialysis of an 85-year-old man. Blood Coagul Fibrinolysis. 2015;26:225– 229. doi: 10.1097/MBC.0000000000000221. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e628 60. Ratanapo S, Ungprasert P, Srivali N, Cheungpasitporn W, Bischof EF. Reversal of a dabigatran overdose: what are possible options? Am J Med Sci. 2013;346:259. doi: 10.1097/MAJ.0b013e318295cc72. 61. Mumoli N, Cei M, Fiorini M, Pennati P, Testa S, Dentali F. Conser- vative management of intentional massive dabigatran overdose. J Am Geriatr Soc. 2015;63:2205–2207. doi: 10.1111/jgs.13684. 62. Andreu-Cayuelas JM, Pastor-Perez FJ, Puche CM, Mateo-Martinez A, Garcia-Alberola A, Flores-Blanco PJ, Valdes M, Lip GY, Roldan V, Manzano-Fernandez S. Impact of variations in kidney function on nonvitamin K oral anticoagulant dosing in patients with atrial fibrillation and recent acute heart failure. Rev Esp Cardiol (Engl Ed). 2015;69:134–140. doi: 10.1016/j.rec.2015.06.021. 63. Caldeira D, Gonçalves N, Pinto FJ, Costa J, Ferreira JJ. Risk of renal failure with the non-vitamin K antagonist oral anticoagulants: systematic review and meta-analysis. Pharmacoepidemiol Drug Saf. 2015;24:757–764. doi: 10.1002/pds.3791. 64. Narasimha Krishna V, Warnock DG, Saxena N, Rizk DV. Oral an- ticoagulants and risk of nephropathy. Drug Saf. 2015;38:527– 533. doi: 10.1007/s40264-015-0290-z. 65. Brodsky SV, Nadasdy T, Rovin BH, Satoskar AA, Nadasdy GM, Wu HM, Bhatt UY, Hebert LA. Warfarin-related nephropathy occurs in patients with and without chronic kidney disease and is associated with an increased mortality rate. Kidney Int. 2011;80:181–189. doi: 10.1038/ki.2011.44. 66. Fordyce CB, Hellkamp AS, Lokhnygina Y, Lindner SM, Piccini JP, Becker RC, Berkowitz SD, Breithardt G, Fox KA, Mahaffey KW, Nessel CC, Singer DE, Patel MR; ROCKET AF Steering Commit- tee and Investigators. On-treatment outcomes in patients with worsening renal function with rivaroxaban compared with warfa- rin: insights from ROCKET AF. Circulation. 2016;134:37–47. doi: 10.1161/CIRCULATIONAHA.116.021890. 67. Böhm M, Ezekowitz MD, Connolly SJ, Eikelboom JW, Hohnloser SH, Reilly PA, Schumacher H, Brueckmann M, Schirmer SH, Kratz MT, Yu- suf S, Diener HC, Hijazi Z, Wallentin L. Changes in renal function in pa- tients with atrial fibrillation: an analysis from the RE-LY Trial. J Am Coll Cardiol. 2015;65:2481–2493. doi: 10.1016/j.jacc.2015.03.577. 68. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. Tissue plasminogen activator for acute isch- emic stroke. N Engl J Med. 1995;333:1581–1587. 69. Hacke W, Kaste M, Bluhmki E, Brozman M, Dávalos A, Guidetti D, Larrue V, Lees KR, Medeghri Z, Machnig T, Schneider D, von Kum- mer R, Wahlgren N, Toni D; ECASS Investigators. Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N Engl J Med. 2008;359:1317–1329. doi: 10.1056/NEJMoa0804656. 70. Lees KR, Bluhmki E, von Kummer R, Brott TG, Toni D, Grotta JC, Albers GW, Kaste M, Marler JR, Hamilton SA, Tilley BC, Davis SM, Donnan GA, Hacke W, Allen K, Mau J, Meier D, del Zoppo G, De Silva DA, Butcher KS, Parsons MW, Barber PA, Levi C, Bladin C, Byrnes G; ECASS, ATLANTIS, NINDS and EPITHET rt-PA Study Group. Time to treatment with intravenous alteplase and out- come in stroke: an updated pooled analysis of ECASS, ATLANTIS, NINDS, and EPITHET trials. Lancet. 2010;375:1695–1703. doi: 10.1016/S0140-6736(10)60491-6. 71. Jauch EC, Saver JL, Adams HP Jr, Bruno A, Connors JJ, Demaer- schalk BM, Khatri P, McMullan PW Jr, Qureshi AI, Rosenfield K, Scott PA, Summers DR, Wang DZ, Wintermark M, Yonas H; on behalf of the American Heart Association Stroke Council; Council on Cardiovascular Nursing; Council on Peripheral Vascular Dis- ease; Council on Clinical Cardiology. Guidelines for the early man- agement of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/ American Stroke Association. Stroke. 2013;44:870–947. doi: 10.1161/STR.0b013e318284056a. 72. Sun L, Zhou W, Ploen R, Zorn M, Veltkamp R. Anticoagulation with dabigatran does not increase secondary intracerebral haemorrhage after thrombolysis in experimental cerebral ischaemia. Thromb Hae- most. 2013;110:153–161. doi: 10.1160/TH12-12-0942. 73. Ploen R, Sun L, Zhou W, Heitmeier S, Zorn M, Jenetzky E, Velt- kamp R. Rivaroxaban does not increase hemorrhage after throm- bolysis in experimental ischemic stroke. J Cereb Blood Flow Metab. 2014;34:495–501. doi: 10.1038/jcbfm.2013.226. 74. Jayathissa S, Gommans J, Harper P. Stroke thrombolysis in pa- tients taking dabigatran. Intern Med J. 2013;43:826–828. doi: 10.1111/imj.12182. 75. Casado Naranjo I, Portilla-Cuenca JC, Jiménez Caballero PE, Calle Escobar ML, Romero Sevilla RM. Fatal intracerebral hemorrhage associated with administration of recombinant tissue plasminogen activator in a stroke patient on treatment with dabigatran. Cere- brovasc Dis. 2011;32:614–615. doi: 10.1159/000334578. 76. Seiffge DJ, Hooff RJ, Nolte CH, Béjot Y, Turc G, Ikenberg B, Berge E, Persike M, Dequatre-Ponchelle N, Strbian D, Pfeilschifter W, Zini A, Tveiten A, Næss H, Michel P, Sztajzel R, Luft A, Gensicke H, Traenka C, Hert L, Scheitz JF, De Marchis GM, Bonati LH, Peters N, Charidimou A, Werring DJ, Palm F, Reinhard M, Niesen WD, Nagao T, Pezzini A, Caso V, Nederkoorn PJ, Kägi G, von Hessling A, Padjen V, Cordonnier C, Erdur H, Lyrer PA, Brouns R, Steiner T, Tatlisumak T, Engelter ST; NOACISP Study Group. Recanalization therapies in acute ischemic stroke patients: impact of prior treat- ment with novel oral anticoagulants on bleeding complications and outcome. Circulation. 2015;132:1261–1269. doi: 10.1161/ CIRCULATIONAHA.115.015484. 77. Berkhemer OA, Fransen PS, Beumer D, van den Berg LA, Lings- ma HF, Yoo AJ, Schonewille WJ, Vos JA, Nederkoorn PJ, Wermer MJ, van Walderveen MA, Staals J, Hofmeijer J, van Oostayen JA, Lycklama à Nijeholt GJ, Boiten J, Brouwer PA, Emmer BJ, de Bruijn SF, van Dijk LC, Kappelle LJ, Lo RH, van Dijk EJ, de Vries J, de Kort PL, van Rooij WJ, van den Berg JS, van Hasselt BA, Aerden LA, Dal- linga RJ, Visser MC, Bot JC, Vroomen PC, Eshghi O, Schreuder TH, Heijboer RJ, Keizer K, Tielbeek AV, den Hertog HM, Gerrits DG, van den Berg-Vos RM, Karas GB, Steyerberg EW, Flach HZ, Marquering HA, Sprengers ME, Jenniskens SF, Beenen LF, van den Berg R, Koudstaal PJ, van Zwam WH, Roos YB, van der Lugt A, van Oosten- brugge RJ, Majoie CB, Dippel DW; MR CLEAN Investigators. A ran- domized trial of intraarterial treatment for acute ischemic stroke. N Engl J Med. 2015;372:11–20. doi: 10.1056/NEJMoa1411587. 78. Campbell BC, Mitchell PJ, Kleinig TJ, Dewey HM, Churilov L, Yassi N, Yan B, Dowling RJ, Parsons MW, Oxley TJ, Wu TY, Brooks M, Simpson MA, Miteff F, Levi CR, Krause M, Harrington TJ, Faulder KC, Steinfort BS, Priglinger M, Ang T, Scroop R, Barber PA, Mc- Guinness B, Wijeratne T, Phan TG, Chong W, Chandra RV, Bladin CF, Badve M, Rice H, de Villiers L, Ma H, Desmond PM, Donnan GA, Davis SM; EXTEND-IA Investigators. Endovascular therapy for ischemic stroke with perfusion-imaging selection. N Engl J Med. 2015;372:1009–1018. doi: 10.1056/NEJMoa1414792. 79. Saver JL, Goyal M, Bonafe A, Diener HC, Levy EI, Pereira VM, Albers GW, Cognard C, Cohen DJ, Hacke W, Jansen O, Jovin TG, Mattle HP, Nogueira RG, Siddiqui AH, Yavagal DR, Baxter BW, Dev- lin TG, Lopes DK, Reddy VK, du Mesnil de Rochemont R, Singer OC, Jahan R; SWIFT PRIME Investigators. Stent-retriever throm- bectomy after intravenous t-PA vs. t-PA alone in stroke. N Engl J Med. 2015;372:2285–2295. doi: 10.1056/NEJMoa1415061. 80. Goyal M, Demchuk AM, Menon BK, Eesa M, Rempel JL, Thornton J, Roy D, Jovin TG, Willinsky RA, Sapkota BL, Dowlatshahi D, Frei DF, Kamal NR, Montanera WJ, Poppe AY, Ryckborst KJ, Silver FL, Shuaib A, Tampieri D, Williams D, Bang OY, Baxter BW, Burns PA, Choe H, Heo JH, Holmstedt CA, Jankowitz B, Kelly M, Linares G, Mandzia JL, Shankar J, Sohn SI, Swartz RH, Barber PA, Coutts SB, Smith EE, Morrish WF, Weill A, Subramaniam S, Mitha AP, Wong JH, Lowerison MW, Sajobi TT, Hill MD; ESCAPE Trial Investiga- tors. Randomized assessment of rapid endovascular treatment of ischemic stroke. N Engl J Med. 2015;372:1019–1030. doi: 10.1056/NEJMoa1414905. 81. Jovin TG, Chamorro A, Cobo E, de Miquel MA, Molina CA, Rovira A, San Román L, Serena J, Abilleira S, Ribó M, Millán M, Urra X, D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e629 CLINICAL STATEM ENTS AND GUIDELINES Cardona P, López-Cancio E, Tomasello A, Castaño C, Blasco J, Aja L, Dorado L, Quesada H, Rubiera M, Hernandez-Pérez M, Goyal M, Demchuk AM, von Kummer R, Gallofré M, Dávalos A; REVASCAT Trial Investigators. Thrombectomy within 8 hours after symptom onset in ischemic stroke. N Engl J Med. 2015;372:2296–2306. doi: 10.1056/NEJMoa1503780. 82. Javedani PP, Horowitz BZ, Clark WM, Lutsep HL. Dabigatran etexilate: management in acute ischemic stroke. Am J Crit Care. 2013;22:169–176. doi: 10.4037/ajcc2013125. 83. Moey AW, Koblar SA, Chryssidis S, Robinson M, Jannes J. Endo- vascular therapy after stroke in a patient treated with dabigatran. Med J Aust. 2012;196:469–471. 84. Müller P, Topakian R, Sonnberger M, Nußbaumer K, Windpessl M, Eder V, Nesser HJ, Trenkler J, Haring HP. Endovascular throm- bectomy for acute ischemic stroke patients anticoagulated with dabigatran. Clin Neurol Neurosurg. 2013;115:2257–2259. doi: 10.1016/j.clineuro.2013.07.017. 85. Freeman WD, Kuo RS, Hanel RA. Letter by Freeman et al regard- ing article, “Using dabigatran in patients with stroke: a practical guide for clinicians.” Stroke. 2012;43:e48; author reply e49. doi: 10.1161/STROKEAHA.112.652313. 86. Mehta S, Dababneh H, Hussain M, Moussavi M, Kirmani JF. Endo- vascular treatment in acute ischemic stroke patient on factor Xa inhibitor. J Vasc Interv Neurol. 2014;7:5–7. 87. Kimura S, Ogata T, Fukae J, Okawa M, Higashi T, Iwaasa M, Inoue T, Tsuboi Y. Revascularization for acute ischemic stroke is safe for rivaroxaban users. J Stroke Cerebrovasc Dis. 2014;23:e427– e431. doi: 10.1016/j.jstrokecerebrovasdis.2014.05.015. 88. Powers WJ, Derdeyn CP, Biller J, Coffey CS, Hoh BL, Jauch EC, Johnston KC, Johnston SC, Khalessi AA, Kidwell CS, Meschia JF, Ovbiagele B, Yavagal DR; on behalf of the American Heart Association Stroke Council. 2015 American Heart Association/American Stroke Association focused update of the 2013 guidelines for the early management of patients with acute ischemic stroke regarding endo- vascular treatment: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2015;46:3020–3035. doi: 10.1161/STR.0000000000000074. 89. 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– 430. doi: 10.1161/01.STR.0000254600.92975.1f. 90. Sandercock PA, Counsell C, Kane EJ. Anticoagulants for acute isch- aemic stroke. Cochrane Database Syst Rev. 2015;3:CD000024. doi: 10.1002/14651858.CD000024.pub4. 91. Paciaroni M, Agnelli G, Corea F, Ageno W, Alberti A, Lanari A, Caso V, Micheli S, Bertolani L, Venti M, Palmerini F, Biagini S, Comi G, Previdi P, Silvestrelli G. Early hemorrhagic transformation of brain infarction: rate, predictive factors, and influence on clini- cal outcome: results of a prospective multicenter study. Stroke. 2008;39:2249–2256. doi: 10.1161/STROKEAHA.107.510321. 92. Fiorelli M, Bastianello S, von Kummer R, del Zoppo GJ, Larrue V, Lesaffre E, Ringleb AP, Lorenzano S, Manelfe C, Bozzao L. Hem- orrhagic transformation within 36 hours of a cerebral infarct: rela- tionships with early clinical deterioration and 3-month outcome in the European Cooperative Acute Stroke Study I (ECASS I) cohort. Stroke. 1999;30:2280–2284. doi: 10.1161/01.STR.30.11.2280. 93. Berger C, Fiorelli M, Steiner T, Schäbitz W-R, Bozzao L, Bluhm- ki E, Hacke W, von Kummer R. Hemorrhagic transformation of ischemic brain tissue: asymptomatic or symptomatic? Stroke. 2001;32:1330–1335. doi: 10.1161/01.STR.32.6.1330. 94. Pessin MS, Estol CJ, Lafranchise F, Caplan LR. Safety of anticoagula- tion after hemorrhagic infarction. Neurology. 1993;43:1298–1303. 95. Bohmann F, Mirceska A, Pfeilschifter J, Lindhoff-Last E, Steinmetz H, Foerch C, Pfeilschifter W. No influence of dabigatran anticoagu- lation on hemorrhagic transformation in an experimental model of ischemic stroke. PLoS One. 2012;7:e40804. doi: 10.1371/ journal.pone.0040804. 96. Gliem M, Hermsen D, van Rooijen N, Hartung HP, Jander S. Secondary intracerebral hemorrhage due to early initiation of oral anticoagula- tion after ischemic stroke: an experimental study in mice. Stroke. 2012;43:3352–3357. doi: 10.1161/STROKEAHA.112.666818. 97. Connolly SJ, Eikelboom J, Joyner C, Diener HC, Hart R, Golitsyn S, Flaker G, Avezum A, Hohnloser SH, Diaz R, Talajic M, Zhu J, Pais P, Budaj A, Parkhomenko A, Jansky P, Commerford P, Tan RS, Sim KH, Lewis BS, Van Mieghem W, Lip GY, Kim JH, Lanas- Zanetti F, Gonzalez-Hermosillo A, Dans AL, Munawar M, O’Donnell M, Lawrence J, Lewis G, Afzal R, Yusuf S; AVERROES Steering Committee and Investigators. Apixaban in patients with atrial fibrillation. N Engl J Med. 2011;364:806–817. doi: 10.1056/ NEJMoa1007432. 98. Lansberg MG, O’Donnell MJ, Khatri P, Lang ES, Nguyen-Huynh MN, Schwartz NE, Sonnenberg FA, Schulman S, Vandvik PO, Spencer FA, Alonso-Coello P, Guyatt GH, Akl EA. Antithrombotic and thrombolytic therapy for ischemic stroke: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141:e601S–636S. 99. Heidbuchel H, Verhamme P, Alings M, Antz M, Hacke W, Oldgren J, Sinnaeve P, Camm AJ, Kirchhof P; European Heart Rhythm Association. European Heart Rhythm Association Practical Guide on the use of new oral anticoagulants in patients with non-valvular atrial fibrillation. Europace. 2013;15:625–651. doi: 10.1093/ europace/eut083. 100. Kernan WN, Ovbiagele B, Black HR, Bravata DM, Chimowitz MI, Ezekowitz MD, Fang MC, Fisher M, Furie KL, Heck DV, Johnston SC, Kasner SE, Kittner SJ, Mitchell PH, Rich MW, Richardson D, Schwamm LH, Wilson JA; on behalf of the American Heart Association Stroke Council, Council on Cardiovascular and Stroke Nursing, Council on Clinical Cardiology, and Council on Peripheral Vascular Disease. Guidelines for the prevention of stroke in pa- tients with stroke and transient ischemic attack: a guideline for healthcare professionals from the American Heart Association/ American Stroke Association. Stroke. 2014;45:2160–2236. doi: 10.1161/STR.0000000000000024. 101. Lip GY. Assessing bleeding risk with the HAS-BLED score: balancing simplicity, practicality, and predictive value in bleeding-risk assess- ment. Clin Cardiol. 2015;38:562–564. doi: 10.1002/clc.22436. 102. Lip GY, Lane DA. Assessing eligibility for anticoagulation after diagnosis of atrial fibrillation–reply. JAMA. 2015;314:949–950. doi: 10.1001/jama.2015.8995. 103. Healey JS, Eikelboom J, Douketis J, Wallentin L, Oldgren J, Yang S, Themeles E, Heidbuchel H, Heidbuchle H, Avezum A, Reilly P, Connolly SJ, Yusuf S, Ezekowitz M; RE-LY Investigators. Periprocedural bleeding and thromboembolic events with dabi- gatran compared with warfarin: results from the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) ran- domized trial. Circulation. 2012;126:343–348. doi: 10.1161/ CIRCULATIONAHA.111.090464. 104. Sherwood MW, Douketis JD, Patel MR, Piccini JP, Hellkamp AS, Lokhnygina Y, Spyropoulos AC, Hankey GJ, Singer DE, Nessel CC, Mahaffey KW, Fox KA, Califf RM, Becker RC; ROCKET AF Investigators. 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– 1859. doi: 10.1161/CIRCULATIONAHA.113.005754. 105. Garcia D, Alexander JH, Wallentin L, Wojdyla DM, Thomas L, Hanna M, Al-Khatib SM, Dorian P, Ansell J, Commerford P, Flaker G, Lanas F, Vinereanu D, Xavier D, Hylek EM, Held C, Verheugt FW, Granger CB, Lopes RD. Management and clinical outcomes in patients treated with apixaban vs warfarin undergo- ing procedures. Blood. 2014;124:3692–3698. doi: 10.1182/ blood-2014-08-595496. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e630 106. Caldeira D, Costa J, Ferreira JJ, Pinto FJ. Thromboembolic risk in the initiation, switch and interruption/re-initiation of oral anti- coagulants: do newcomers improve outcomes? Insights from a meta-analysis of RCTs. Int J Cardiol. 2014;177:117–119. doi: 10.1016/j.ijcard.2014.09.099. 107. Patel MR, Hellkamp AS, Lokhnygina Y, Piccini JP, Zhang Z, Mohanty S, Singer DE, Hacke W, Breithardt G, Halperin JL, Hankey GJ, Becker RC, Nessel CC, Berkowitz SD, Califf RM, Fox KA, Mahaffey KW. Outcomes of discontinuing rivaroxaban compared with warfa- rin in patients with nonvalvular atrial fibrillation: analysis from the ROCKET AF trial (Rivaroxaban Once-Daily, Oral, Direct Factor Xa Inhibition Compared With Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation). J Am Coll Cardiol. 2013;61:651–658. doi: 10.1016/j.jacc.2012.09.057. 108. Mahaffey KW, Hellkamp AS, Patel MR, Hannan KL, Schwabe K, Nessel CC, Berkowitz SD, Halperin JL, Hankey GJ, Becker RC, Piccini JP, Breithardt G, Hacke W, Singer DE, Califf RM, Fox KA. End-of-study transition from study drug to open-label vitamin K antagonist therapy: the ROCKET AF experience. Circ Cardiovasc Qual Outcomes. 2013;6:470–478. doi: 10.1161/ CIRCOUTCOMES.113.000132. 109. Granger CB, Lopes RD, Hanna M, Ansell J, Hylek EM, Alexander JH, Thomas L, Wang J, Bahit MC, Verheugt F, Lawrence J, Xavier D, Wallentin L. Clinical events after transitioning from apixaban versus warfarin to warfarin at the end of the Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial. Am Heart J. 2015;169:25– 30. doi: 10.1016/j.ahj.2014.09.006. 110. Ruff CT, Giugliano RP, Braunwald E, Mercuri M, Curt V, Betcher J, Grip L, Cange AL, Crompton AE, Murphy SA, Deenadayalu N, Antman EM. Transition of patients from blinded study drug to open-label anticoagulation: the ENGAGE AF-TIMI 48 trial. J Am Coll Cardiol. 2014;64:576–584. doi: 10.1016/j.jacc.2014.05.028. 110a. Piccini JP, Garg J, Patel MR, Lokhnygina Y, Goodman SG, Becker RC, Berkowitz SD, Breithardt G, Hacke W, Halperin JL, Hankey GJ, Nessel CC, Mahaffey KW, Singer DE, Califf RM, Fox KA; ROCKET AF Investigators. Management of major bleeding events in patients treated with rivaroxaban vs. warfarin: results from the ROCKET AF trial. Eur Heart J. 2014;35:1873–1880. doi: 10.1093/eurheartj/ehu083. 111. Beyer-Westendorf J, Gelbricht V, Förster K, Ebertz F, Röllig D, Schreier T, Tittl L, Thieme C, Hänsel U, Köhler C, Werth S, Kuhlisch E, Stange T, Röder I, Weiss N. Safety of switching from vitamin K antagonists to dabigatran or rivaroxaban in daily care–results from the Dresden NOAC registry. Br J Clin Pharmacol. 2014;78:908–917. doi: 10.1111/bcp.12391. 112. Bouillon K, Bertrand M, Maura G, Blotière PO, Ricordeau P, Zureik M. Risk of bleeding and arterial thromboembolism in patients with non-valvular atrial fibrillation either maintained on a vitamin K an- tagonist or switched to a non-vitamin K-antagonist oral anticoagu- lant: a retrospective, matched-cohort study. Lancet Haematol. 2015;2:e150–e159. doi: 10.1016/S2352-3026(15)00027-7. 113. Douketis JD, Berger PB, Dunn AS, Jaffer AK, Spyropoulos AC, Becker RC, Ansell J. The perioperative management of anti- thrombotic therapy: American College of Chest Physicians evidence-based clinical practice guidelines (8th edition). Chest. 2008;133:299S–339S. 114. Douketis JD, Spyropoulos AC, Spencer FA, Mayr M, Jaffer AK, Eckman MH, Dunn AS, Kunz R. Perioperative manage- ment of antithrombotic therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141:e326S–350S 115. Gallego P, Apostolakis S, Lip GY. Bridging evidence-based practice and practice-based evidence in periprocedural antico- agulation. Circulation. 2012;126:1573–1576. doi: 10.1161/ CIRCULATIONAHA.112.135681. 116. Douketis JD, Spyropoulos AC, Kaatz S, Becker RC, Caprini JA, Dunn AS, Garcia DA, Jacobson A, Jaffer AK, Kong DF, Schulman S, Turpie AG, Hasselblad V, Ortel TL; BRIDGE Investigators. Perioperative bridging anticoagulation in patients with atrial fi- brillation. N Engl J Med. 2015;373:823–833. doi: 10.1056/ NEJMoa1501035. 117. Schulman S, Kearon C; Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost. 2005;3:692–694. doi: 10.1111/j.1538-7836.2005.01204.x. 118. Kralev S, Haag B, Spannenberger J, Lang S, Brockmann MA, Bartling S, Marx A, Haase KK, Borggrefe M, Süselbeck T. Expansion of the Multi-Link Frontier™ coronary bifurcation stent: micro-computed tomographic assessment in human autopsy and porcine heart samples. PLoS One. 2011;6:e21778. doi: 10.1371/journal.pone.0021778. 119. Bashore TM, Balter S, Barac A, Byrne JG, Cavendish JJ, Chambers CE, Hermiller JB Jr, Kinlay S, Landzberg JS, Laskey WK, McKay CR, Miller JM, Moliterno DJ, Moore JW, Oliver-McNeil SM, Popma JJ, Tommaso CL; ACCF Task Force Members. 2012 American College of Cardiology Foundation/Society for Cardiovascular Angiography and Interventions expert consensus document on cardiac catheterization laboratory standards up- date: a report of the American College of Cardiology Foundation Task Force on expert consensus documents. J Am Coll Cardiol. 2012;59:2221–2305. doi: 10.1016/j.jacc.2012.02.010. 120. Lip GY, Nieuwlaat R, Pisters R, Lane DA, Crijns HJ. Refining clini- cal 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– 272. doi: 10.1378/chest.09-1584. 121. Cannon CP, Weintraub WS, Demopoulos LA, Vicari R, Frey MJ, Lakkis N, Neumann FJ, Robertson DH, DeLucca PT, DiBattiste PM, Gibson CM, Braunwald E; TACTICS (Treat Angina with Aggrastat and Determine Cost of Therapy with an Invasive or Conservative Strategy)–Thrombolysis in Myocardial Infarction 18 Investigators. Comparison of early invasive and conservative strategies in patients with unstable coronary syndromes treated with the glycoprotein IIb/IIIa inhibitor tirofiban. N Engl J Med. 2001;344:1879–1887. doi: 10.1056/NEJM200106213442501. 122. O’Gara PT, Kushner FG, Ascheim DD, Casey DE Jr, Chung MK, de Lemos JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, Granger CB, Krumholz HM, Linderbaum JA, Morrow DA, Newby LK, Ornato JP, Ou N, Radford MJ, Tamis-Holland JE, Tommaso CL, Tracy CM, Woo YJ, Zhao DX, Anderson JL, Jacobs AK, Halperin JL, Albert NM, Brindis RG, Creager MA, DeMets D, Guyton RA, Hochman JS, Kovacs RJ, Kushner FG, Ohman EM, Stevenson WG, Yancy CW; American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation. 2013;127:e362–e425. doi: 10.1161/CIR.0b013e3182742cf6. 123. Jolly SS, Yusuf S, Cairns J, Niemelä K, Xavier D, Widimsky P, Budaj A, Niemelä M, Valentin V, Lewis BS, Avezum A, Steg PG, Rao SV, Gao P, Afzal R, Joyner CD, Chrolavicius S, Mehta SR; RIVAL trial group. Radial versus femoral access for coro- nary angiography and intervention in patients with acute coronary syndromes (RIVAL): a randomised, parallel group, multicentre trial. Lancet. 2011;377:1409–1420. doi: 10.1016/ S0140-6736(11)60404-2. 124. Levine GN, Bates ER, Blankenship JC, Bailey SR, Bittl JA, Cercek B, Chambers CE, Ellis SG, Guyton RA, Hollenberg SM, Khot UN, Lange RA, Mauri L, Mehran R, Moussa ID, Mukherjee D, Nallamothu BK, Ting HH. 2011 ACCF/AHA/SCAI guideline for percutaneous D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e631 CLINICAL STATEM ENTS AND GUIDELINES coronary intervention: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines and the Society for Cardiovascular Angiography and Interventions. Circulation. 2011;124:e574– e651. doi: 10.1161/CIR.0b013e31823ba622. 125. Gupta S, Cigarroa JE. Bivalirudin: an expensive heparin? Catheter Cardiovasc Interv. 2015;86:397–399. doi: 10.1002/ ccd.26124. 126. Levine GN, Bates ER, Bittl JA, Brindis RG, Fihn SD, Fleisher LA, Granger CB, Lange RA, Mack MJ, Mauri L, Mehran R, Mukherjee D, Newby LK, O’Gara PT, Sabatine MS, Smith PK, Smith SC, Jr. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines: an update of the 2011 ACCF/AHA/SCAI guideline for percutaneous coro- nary intervention, 2011 ACCF/AHA guideline for coronary artery bypass graft surgery, 2012 ACC/AHA/ACP/AATS/PCNA/SCAI/ STS guideline for the diagnosis and management of patients with stable ischemic heart disease, 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction, 2014 AHA/ACC guideline for the management of patients with non– ST-elevation acute coronary syndromes, and 2014 ACC/AHA guideline on perioperative cardiovascular evaluation and man- agement of patients undergoing noncardiac surgery. Circulation. 2016;134:1–33. doi: 10.1161/CIR.0000000000000404. 127. Oldgren J, Budaj A, Granger CB, Khder Y, Roberts J, Siegbahn A, Tijssen JG, Van de Werf F, Wallentin L; RE-DEEM Investigators. Dabigatran vs. placebo in patients with acute coronary syn- dromes on dual antiplatelet therapy: a randomized, double-blind, phase II trial. Eur Heart J. 2011;32:2781–2789. doi: 10.1093/ eurheartj/ehr113. 128. Alexander JH, Lopes RD, James S, Kilaru R, He Y, Mohan P, Bhatt DL, Goodman S, Verheugt FW, Flather M, Huber K, Liaw D, Husted SE, Lopez-Sendon J, De Caterina R, Jansky P, Darius H, Vinereanu D, Cornel JH, Cools F, Atar D, Leiva-Pons JL, Keltai M, Ogawa H, Pais P, Parkhomenko A, Ruzyllo W, Diaz R, White H, Ruda M, Geraldes M, Lawrence J, Harrington RA, Wallentin L; APPRAISE-2 Investigators. Apixaban with antiplatelet therapy after acute coronary syndrome. N Engl J Med. 2011;365:699– 708. doi: 10.1056/NEJMoa1105819. 129. Mega JL, Braunwald E, Wiviott SD, Bassand JP, Bhatt DL, Bode C, Burton P, Cohen M, Cook-Bruns N, Fox KA, Goto S, Murphy SA, Plotnikov AN, Schneider D, Sun X, Verheugt FW, Gibson CM; ATLAS ACS 2–TIMI 51 Investigators. Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J Med. 2012;366:9– 19. doi: 10.1056/NEJMoa1112277. 130. Sherwood MW, Cyr DD, Jones WS, Becker RC, Berkowitz SD, Washam JB, Breithardt G, Fox KA, Halperin JL, Hankey GJ, Singer DE, Piccini JP, Nessel CC, Mahaffey KW, Patel MR. Use of dual antiplatelet therapy and patient outcomes in those undergo- ing percutaneous coronary intervention: the ROCKET AF trial. JACC Cardiovasc Interv. 2016;9:1694–1702. doi: 10.1016/j. jcin.2016.05.039. 131. Bhatt DL, Cryer BL, Contant CF, Cohen M, Lanas A, Schnitzer TJ, Shook TL, Lapuerta P, Goldsmith MA, Laine L, Scirica BM, Murphy SA, Cannon CP; COGENT Investigators. Clopidogrel with or without omeprazole in coronary artery disease. N Engl J Med. 2010;363:1909–1917. doi: 10.1056/NEJMoa1007964. 132. Gibson CM, Mehran R, Bode C, Halperin J, Verheugt F, Wildgoose P, van Eickels M, Lip GY, Cohen M, Husted S, Peterson E, Fox K. An open-label, randomized, controlled, multicenter study explor- ing two treatment strategies of rivaroxaban and a dose-adjusted oral vitamin K antagonist treatment strategy in subjects with atrial fibrillation who undergo percutaneous coronary intervention (PIONEER AF-PCI). Am Heart J. 2015;169:472–478 e475 133. Gao F, Shen H, Wang ZJ, Yang SW, Liu XL, Zhou YJ. Rationale and design of the RT-AF study: combination of rivaroxaban and ticagre- lor in patients with atrial fibrillation and coronary artery disease undergoing percutaneous coronary intervention. Contemp Clin Trials. 2015;43:129–132. doi: 10.1016/j.cct.2015.05.012. 134. Hoshi T, Sato A, Nogami A, Gosho M, Aonuma K; SAFE-A Investigators. Rationale and design of the SAFE-A study: SAFety and Effectiveness trial of apixaban use in association with dual antiplatelet therapy in patients with atrial fibrillation undergoing percutaneous coronary intervention [published on- line ahead of print July 18, 2016]. J Cardiol. doi: 10.1016/j. jjcc.2016.06.007. http://www.sciencedirect.com/science/article/ pii/S0914508716301344. Accessed January 17, 2017. 135. Dewilde WJ, Oirbans T, Verheugt FW, Kelder JC, De Smet BJ, Herrman JP, Adriaenssens T, Vrolix M, Heestermans AA, Vis MM, Tijsen JG, van ‘t Hof AW, ten Berg JM; WOEST Study Investigators. 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;381:1107–1115. doi: 10.1016/S0140-6736(12)62177-1. 136. Nagarakanti R, Ezekowitz MD, Oldgren J, Yang S, Chernick M, Aikens TH, Flaker G, Brugada J, Kamensky G, Parekh A, Reilly PA, Yusuf S, Connolly SJ. Dabigatran versus warfarin in pa- tients with atrial fibrillation: an analysis of patients undergoing cardioversion. Circulation. 2011;123:131–136. doi: 10.1161/ CIRCULATIONAHA.110.977546. 137. Piccini JP, Stevens SR, Lokhnygina Y, Patel MR, Halperin JL, Singer DE, Hankey GJ, Hacke W, Becker RC, Nessel CC, Mahaffey KW, Fox KA, Califf RM, Breithardt G; ROCKET AF Steering Committee & Investigators. Outcomes after cardioversion and atrial fibrilla- tion ablation in patients treated with rivaroxaban and warfarin in the ROCKET AF trial. J Am Coll Cardiol. 2013;61:1998–2006. doi: 10.1016/j.jacc.2013.02.025. 138. Flaker G, Lopes RD, Al-Khatib SM, Hermosillo AG, Hohnloser SH, Tinga B, Zhu J, Mohan P, Garcia D, Bartunek J, Vinereanu D, Husted S, Harjola VP, Rosenqvist M, Alexander JH, Granger CB; ARISTOTLE Committees and Investigators. 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;63:1082–1087. doi: 10.1016/j.jacc.2013.09.062. 139. Sen P, Kundu A, Sardar P, Chatterjee S, Nairooz R, Amin H, Aronow WS. Outcomes after cardioversion in atrial fibrillation pa- tients treated with non-vitamin K antagonist oral anticoagulants (NOACs): insights from a meta-analysis. Am J Cardiovasc Drugs. 2015;16:33–41. doi: 10.1007/s40256-015-0136-1. 140. Caldeira D, Costa J, Ferreira JJ, Lip GY, Pinto FJ. Non-vitamin K antagonist oral anticoagulants in the cardioversion of pa- tients with atrial fibrillation: systematic review and meta-anal- ysis. Clin Res Cardiol. 2015;104:582–590. doi: 10.1007/ s00392-015-0821-8. 141. Cappato R, Ezekowitz MD, Klein AL, Camm AJ, Ma CS, Le Heuzey JY, Talajic M, Scanavacca M, Vardas PE, Kirchhof P, Hemmrich M, Lanius V, Meng IL, Wildgoose P, van Eickels M, Hohnloser SH; X-VeRT Investigators. Rivaroxaban vs. vitamin K antagonists for cardioversion in atrial fibrillation. Eur Heart J. 2014;35:3346– 3355. doi: 10.1093/eurheartj/ehu367. 142. Pallisgaard JL, Lindhardt TB, Hansen ML, Schjerning AM, Olesen JB, Staerk L, Torp-Pedersen C, Gislason GH. Cardioversion and risk of adverse events with dabigatran versus warfarin: a nationwide cohort study. PLoS One. 2015;10:e0141377. doi: 10.1371/journal.pone.0141377. 143. Coleman CM, Khalaf S, Mould S, Wazni O, Kanj M, Saliba W, Cantillon D. Novel oral anticoagulants for DC cardioversion proce- dures: utilization and clinical outcomes compared with warfarin. Pacing Clin Electrophysiol. 2015;38:731–737. doi: 10.1111/ pace.12618. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Raval et al March 7, 2017 Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477e632 144. Goette A, Merino JL, Ezekowitz MD, Zamoryakhin D, Melino M, Jin J, Mercuri MF, Grosso MA, Fernandez V, Al-Saady N, Pelekh N, Merkely B, Zenin S, Kushnir M, Spinar J, Batushkin V, de Groot JR, Lip GY; ENSURE-AF investigators. Edoxaban versus enoxaparin-warfarin in patients undergoing cardiover- sion of atrial fibrillation (ENSURE-AF): a randomised, open-label, phase 3b trial. Lancet. 2016;388:1995–2003. doi: 10.1016/ S0140-6736(16)31474-X. 145. Di Biase L, Burkhardt JD, Mohanty P, Sanchez J, Horton R, Gallinghouse GJ, Lakkireddy D, Verma A, Khaykin Y, Hongo R, Hao S, Beheiry S, Pelargonio G, Dello Russo A, Casella M, Santarelli P, Santangeli P, Wang P, Al-Ahmad A, Patel D, Themistoclakis S, Bonso A, Rossillo A, Corrado A, Raviele A, Cummings JE, Schweikert RA, Lewis WR, Natale A. Periprocedural stroke and management of major bleeding complications in patients undergoing catheter ablation of atrial fibrillation: the impact of periprocedural therapeu- tic international normalized ratio. Circulation. 2010;121:2550– 2556. doi: 10.1161/CIRCULATIONAHA.109.921320. 146. Santangeli P, Di Biase L, Horton R, Burkhardt JD, Sanchez J, Al-Ahmad A, Hongo R, Beheiry S, Bai R, Mohanty P, Lewis WR, Natale A. Ablation of atrial fibrillation under therapeutic warfarin reduces periprocedural complications: evidence from a meta- analysis. Circ Arrhythm Electrophysiol. 2012;5:302–311. doi: 10.1161/CIRCEP.111.964916. 147. Di Biase L, Burkhardt JD, Santangeli P, Mohanty P, Sanchez JE, Horton R, Gallinghouse GJ, Themistoclakis S, Rossillo A, Lakkireddy D, Reddy M, Hao S, Hongo R, Beheiry S, Zagrodzky J, Rong B, Mohanty S, Elayi CS, Forleo G, Pelargonio G, Narducci ML, Dello Russo A, Casella M, Fassini G, Tondo C, Schweikert RA, Natale A. 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) random- ized trial. Circulation. 2014;129:2638–2644. doi: 10.1161/ CIRCULATIONAHA.113.006426. 148. Shurrab M, Morillo CA, Schulman S, Kansal N, Danon A, Newman D, Lashevsky I, Healey JS, Crystal E. Safety and efficacy of dabigatran compared with warfarin for patients undergoing radiofrequency catheter ablation of atrial fibrillation: a meta- analysis. Can J Cardiol. 2013;29:1203–1210. doi: 10.1016/j. cjca.2013.07.005. 149. Providência R, Albenque JP, Combes S, Bouzeman A, Casteigt B, Combes N, Narayanan K, Marijon E, Boveda S. Safety and efficacy of dabigatran versus warfarin in patients undergo- ing catheter ablation of atrial fibrillation: a systematic review and meta-analysis. Heart. 2014;100:324–335. doi: 10.1136/ heartjnl-2013-304386. 150. Lu D, Zhang Q, Liu Q, Wang K, Wang S, Shan Q. Bleeding risks with novel oral anticoagulants during catheter ablation of atrial fibrillation: a systematic review and network meta-analysis. J Interv Card Electrophysiol. 2015;44:105–111. doi: 10.1007/ s10840-015-0053-x. 151 Nairooz R, Sardar P, Pino M, Aronow WS, Sewani A, Mukherjee D, Paydak H, Maskoun W. Meta-analysis of risk of stroke and thrombo-embolism with rivaroxaban versus vitamin K antagonists in ablation and cardioversion of atrial fibrillation. Int J Cardiol. 2015;187:345–353. doi: 10.1016/j.ijcard.2015.03.323. 152. Lu D, Liu Q, Wang K, Zhang QI, Shan QJ. Meta-analysis of efficacy and safety of apixaban in patients undergoing catheter ablation for atrial fibrillation. Pacing Clin Electrophysiol. 2016;39:54–59. doi: 10.1111/pace.12771. 153. Kuwahara T, Abe M, Yamaki M, Fujieda H, Abe Y, Hashimoto K, Ishiba M, Sakai H, Hishikari K, Takigawa M, Okubo K, Takagi K, Tanaka Y, Nakajima J, Takahashi A. Apixaban versus warfarin for the prevention of periprocedural cerebral thromboembolism in atrial fibrillation ablation: multicenter prospective randomized study. J Cardiovasc Electrophysiol. 2016;27:549–554. doi: 10.1111/jce.12928. 154. Calkins H, Kuck KH, Cappato R, Brugada J, Camm AJ, Chen SA, Crijns HJ, Damiano RJ Jr, Davies DW, DiMarco J, Edgerton J, Ellenbogen K, Ezekowitz MD, Haines DE, Haissaguerre M, Hindricks G, Iesaka Y, Jackman W, Jalife J, Jais P, Kalman J, Keane D, Kim YH, Kirchhof P, Klein G, Kottkamp H, Kumagai K, Lindsay BD, Mansour M, Marchlinski FE, McCarthy PM, Mont JL, Morady F, Nademanee K, Nakagawa H, Natale A, Nattel S, Packer DL, Pappone C, Prystowsky E, Raviele A, Reddy V, Ruskin JN, Shemin RJ, Tsao HM, Wilber D. 2012 HRS/EHRA/ECAS ex- pert consensus statement on catheter and surgical ablation of atrial fibrillation: recommendations for patient selection, proce- dural techniques, patient management and follow-up, definitions, endpoints, and research trial design. Europace. 2012;14:528– 606. doi: 10.1093/europace/eus027. 155. Wallace TW, Atwater BD, Daubert JP, Voora D, Crowley AL, Bahnson TD, Hranitzky PM. Prevalence and clinical characteris- tics associated with left atrial appendage thrombus in fully antico- agulated patients undergoing catheter-directed atrial fibrillation ablation. J Cardiovasc Electrophysiol. 2010;21:849–852. doi: 10.1111/j.1540-8167.2010.01729.x. 156. Di Biase L, Gaita F, Toso E, Santangeli P, Mohanty P, Rutledge N, Yan X, Mohanty S, Trivedi C, Bai R, Price J, Horton R, Gallinghouse GJ, Beheiry S, Zagrodzky J, Canby R, Leclercq JF, Halimi F, Scaglione M, Cesarani F, Faletti R, Sanchez J, Burkhardt JD, Natale A. Does periprocedural anticoagulation management of atrial fibrillation af- fect the prevalence of silent thromboembolic lesion detected by diffusion cerebral magnetic resonance imaging in patients under- going radiofrequency atrial fibrillation ablation with open irrigated catheters? Results from a prospective multicenter study. Heart Rhythm. 2014;11:791–798. doi: 10.1016/j.hrthm.2014.03.003. 157. Konduru SV, Cheema AA, Jones P, Li Y, Ramza B, Wimmer AP. Differences in intraprocedural acts with standardized heparin dosing during catheter ablation for atrial fibrillation in patients treated with dabigatran vs. patients on uninterrupted warfarin. J Interv Card Electrophysiol. 2012;35:277–284; discussion 284. 158. Cheng A, Nazarian S, Brinker JA, Tompkins C, Spragg DD, Leng CT, Halperin H, Tandri H, Sinha SK, Marine JE, Calkins H, Tomaselli GF, Berger RD, Henrikson CA. Continuation of warfarin during pacemaker or implantable cardioverter-defibrillator implantation: a randomized clinical trial. Heart Rhythm. 2011;8:536–540. doi: 10.1016/j.hrthm.2010.12.016. 159. Birnie DH, Healey JS, Wells GA, Verma A, Tang AS, Krahn AD, Simpson CS, Ayala-Paredes F, Coutu B, Leiria TL, Essebag V; BRUISE CONTROL Investigators. Pacemaker or defibrillator surgery without interruption of anticoagulation. N Engl J Med. 2013;368:2084–2093. doi: 10.1056/NEJMoa1302946. 160. Birnie DH, Healey JS, Essebag V. Management of anticoagula- tion around pacemaker and defibrillator surgery. Circulation. 2014;129:2062–2065. doi: 10.1161/CIRCULATIONAHA.113. 006027. 161. Nascimento T, Birnie DH, Healey JS, Verma A, Joza J, Bernier ML, Essebag V. Managing novel oral anticoagulants in patients with atrial fibrillation undergoing device surgery: Canadian survey. Can J Cardiol. 2014;30:231–236. doi: 10.1016/j. cjca.2013.11.027. 162. Jennings JM, Robichaux R, McElderry HT, Plumb VJ, Gunter A, Doppalapudi H, Osorio J, Yamada T, Kay GN. Cardiovascular implantable electronic device implantation with uninterrupted dabigatran: comparison to uninterrupted warfarin. J Cardiovasc Electrophysiol. 2013;24:1125–1129. doi: 10.1111/jce.12214. 163. Rowley CP, Bernard ML, Brabham WW, Netzler PC, Sidney DS, Cuoco F, Sturdivant JL, Leman RB, Wharton JM, Gold MR. Safety of continuous anticoagulation with dabigatran during implantation of cardiac rhythm devices. Am J Cardiol. 2013;111:1165–1168. doi: 10.1016/j.amjcard.2012.12.046. D ow nloaded from http://ahajournals.org by on A pril 5, 2021 Management of Patients on NOACs in the Acute Care and Periprocedural Setting Circulation. 2017;135:e604–e633. DOI: 10.1161/CIR.0000000000000477 March 7, 2017 e633 CLINICAL STATEM ENTS AND GUIDELINES 164. Schulman S, Carrier M, Lee AY, Shivakumar S, Blostein M, Spencer FA, Solymoss S, Barty R, Wang G, Heddle N, Douketis JD; Periop Dabigatran Study Group. Perioperative management of dabi- gatran: a prospective cohort study. Circulation. 2015;132:167– 173. doi: 10.1161/CIRCULATIONAHA.115.015688. 165. Douketis JD, Healey JS, Brueckmann M, Eikelboom JW, Ezekowitz MD, Fraessdorf M, Noack H, Oldgren J, Reilly P, Spyropoulos AC, Wallentin L, Connolly SJ. Perioperative bridging anticoagula- tion during dabigatran or warfarin interruption among patients who had an elective surgery or procedure: substudy of the RE- LY trial. Thromb Haemost. 2015;113:625–632. doi: 10.1160/ TH14-04-0305. 166. Mega JL, Braunwald E, Murphy SA, Plotnikov AN, Burton P, Kiss RG, Parkhomenko A, Tendera M, Widimsky P, Gibson CM. Rivaroxaban in patients stabilized after a ST-segment elevation myocardial infarction: results from the ATLAS ACS-2-TIMI-51 trial (Anti-Xa Therapy to Lower Cardiovascular Events in Addition to Standard Therapy in Subjects with Acute Coronary Syndrome- Thrombolysis In Myocardial Infarction-51). J Am Coll Cardiol. 2013;61:1853–1859. doi: 10.1016/j.jacc.2013.01.066. 167. Eikelboom JW, Connolly SJ, Brueckmann M, Granger CB, Kappetein AP, Mack MJ, Blatchford J, Devenny K, Friedman J, Guiver K, Harper R, Khder Y, Lobmeyer MT, Maas H, Voigt JU, Simoons ML, Van de Werf F; RE-ALIGN Investigators. Dabigatran versus warfarin in patients with mechanical heart valves. N Engl J Med. 2013;369:1206–1214. doi: 10.1056/ NEJMoa1300615. 168. Terrovitis JV, Ntalianis A, Kapelios CJ, Vakrou S, Diakos N, Katsaros L, Tsamatsoulis M, Kaldara E, Charitos C, Nanas JN. Dabigatran etexilate as second-line therapy in patients with a left ventricular assist device. Hellenic J Cardiol. 2015;56:20–25. 169. Abouchakra L, Khabbaz Z, Abouassi S, Badaoui G. Rivaroxaban for treatment of heparin-induced thrombocytopenia after cardiac surgery: a case report. J Thorac Cardiovasc Surg. 2015;150:e19– e20. doi: 10.1016/j.jtcvs.2015.04.054. 170. Jones WS, Hellkamp AS, Halperin J, Piccini JP, Breithardt G, Singer DE, Fox KA, Hankey GJ, Mahaffey KW, Califf RM, Patel MR. Efficacy and safety of rivaroxaban compared with warfarin in patients with peripheral artery disease and non-valvular atrial fibrillation: insights from ROCKET AF. Eur Heart J. 2014;35:242– 249. doi: 10.1093/eurheartj/eht492. 171. Majeed A, Hwang HG, Connolly SJ, Eikelboom JW, Ezekowitz MD, Wallentin L, Brueckmann M, Fraessdorf M, Yusuf S, Schulman S. Management and outcomes of major bleeding during treatment with dabigatran or warfarin. Circulation. 2013;128:2325–2332. doi: 10.1161/CIRCULATIONAHA.113.002332. 172. Paikin JS, Hirsh J, Chan NC, Ginsberg JS, Weitz JI, Eikelboom JW. Timing the first postoperative dose of anticoagulants: les- sons learned from clinical trials. Chest. 2015;148:587–595. doi: 10.1378/chest.14-2710. 173. Rosencher N, Llau JV, Mueck W, Loewe A, Berkowitz SD, Homering M. Incidence of neuraxial haematoma after total hip or knee surgery: RECORD programme (rivaroxaban vs. enoxaparin). Acta Anaesthesiol Scand. 2013;57:565–572. doi: 10.1111/ aas.12069. 174. Turpie AG, Haas S, Kreutz R, Mantovani LG, Pattanayak CW, Holberg G, Jamal W, Schmidt A, van Eickels M, Lassen MR. A non-interventional comparison of rivaroxaban with standard of care for thromboprophylaxis after major orthopaedic surgery in 17,701 patients with propensity score adjustment. Thromb Haemost. 2014;111:94–102. doi: 10.1160/TH13-08-0666. 175. Narouze S, Benzon HT, Provenzano DA, Buvanendran A, De Andres J, Deer TR, Rauck R, Huntoon MA. Interventional spine and pain procedures in patients on antiplatelet and anticoagulant medications: guidelines from the American Society of Regional Anesthesia and Pain Medicine, the European Society of Regional Anaesthesia and Pain Therapy, the American Academy of Pain Medicine, the International Neuromodulation Society, the North American Neuromodulation Society, and the World Institute of Pain. Reg Anesth Pain Med. 2015;40:182–212. doi: 10.1097/ AAP.0000000000000223. 176. Ruff CT, Giugliano RP, Braunwald E, Morrow DA, Murphy SA, Kuder JF, Deenadayalu N, Jarolim P, Betcher J, Shi M, Brown K, Patel I, Mercuri M, Antman EM. Association between edoxa- ban dose, concentration, anti-factor Xa activity, and outcomes: an analysis of data from the randomised, double-blind ENGAGE AF-TIMI 48 trial. Lancet. 2015;385:2288–2295. doi: 10.1016/ S0140-6736(14)61943-7. D ow nloaded from http://ahajournals.org by on A pril 5, 2021