BAY 59-7939

Novel (Oral) Anticoagulant Challenges in Surgery
Richard Blennerhassett, BAppSci, MBBS1,2 Emmanuel J. Favaloro, PhD, FFSc (RCPA)1,3 Leonardo Pasalic, PhD, MBBS, FRCPA, FRACP1,3

1 Department of Laboratory Haematology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, NSW Health Pathology, Westmead, New South Wales, Australia
2 Sydney Medical School, University of Sydney, Westmead, New South
Wales, Australia
3 Sydney Centres for Thrombosis and Haemostasis, Westmead, New South Wales Australia

Summary Non-vitamin K oral anticoagulants (NOACs) are a relatively recent therapeutic modality for the prevention of systemic thromboembolic complications of atrial fibrillation and the prevention and management of venous thromboembolic disease. Approved indications for this class of anticoagulants are likely to further expand as the results of ongoing and new clinical trials are published and clinical experience grows. Despite their convenience compared with traditional methods of anticoagulation, there remain

Keywords
► rivaroxaban
► apixaban
► dabigatran
► perioperative
► surgery
► NOACs
► anticoagulation

a few potential pitfalls associated with their use in the perioperative setting. In particular, there is limited experience and evidence base regarding anticoagulant management in the perioperative setting, especially given the different NOACs available, which in turn are different from the classical anticoagulants. This narrative review synthesizes the recent advances in development of specific NOAC reversal agents and the understanding of the complexities of laboratory measurement of NOAC anticoagulant effects, and aims to provide practicing clinicians with basic evidence- based tools for perioperative management of patients taking NOACs.

The term “NOACs” is one in a minefield of terminology referring to oral anticoagulants that directly target and inhibit specific components of the coagulation pathway. NOACs, as an abbreviation, may refer to “‘new” oral antic- oagulants; novel oral anticoagulants; or non-vitamin K an- tagonist oral anticoagulants; other related abbreviations and terms include DOAC (direct oral anticoagulants), and TSOAC (target specific oral anticoagulant). For the purpose of this review, we will use the term NOACs to refer to the drugs rivaroxaban, apixaban, edoxaban (anti-Xa agents), and da- bigatran (antithrombin agent).
The NOACs are utilized in prophylaxis, treatment, and secondary prevention of venous thromboembolic (VTE) dis-

ease, nonvalvular atrial fibrillation (AF), and other cardiac indications.1–8 Most notably, in patients with AF, NOACs are noninferior with regard to prevention of stroke and systemic thromboembolic events.1,2,9 NOACs are also currently re- commended by international guidelines as the first choice for anticoagulation in non-cancer-related VTE.10 Over the last decade, NOACs have been increasingly prescribed for a variety of indications that traditionally utilized vitamin K antagonists (VKAs) such as warfarin and/or low-molecular- weight heparin (LMWH).11 The usage patterns show geo-
graphic variability dependent on locally approved indica- tions, availability, cost, and health care funding models. Currently, around one in three patients with AF use a

Issue Theme Thrombosis and Hemostasis in Surgery; Guest Editors: Anne-Mette Hvas, MD, PhD, Julie Brogaard Larsen, MD, and Leonardo Pasalic, MD, PhD.

Copyright © by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA.
Tel: +1(212) 584-4662.

DOI https://doi.org/ 10.1055/s-0037-1602667. ISSN 0094-6176.

NOAC for stroke prophylaxis and the number of patients taking NOACs is expected to increase further as the list of recommended indications grows.11
Compared with VKAs, NOACs are associated with compar- able or reduced rates of bleeding; specifically, a lower occurrence of intracranial hemorrhage.1,2,9,12 Additional benefits compared with VKAs include rapid anticoagulant effect without an initial prothrombotic risk period, owing to their direct/target specific effect. Unlike VKAs, NOACs are administered in a standard, indication-specific fixed dose,
and do not require routine laboratory monitoring. Patient preference is another driving force behind increasing use of NOACs.13 Many patients find the requirements for monitor- ing VKA anticoagulant effect and food–drug interactions burdensome and increasingly request their doctor a change to a NOAC.
Recent estimates suggest that annually, 10% of patients taking oral anticoagulation will undergo an interventional procedure.14 This is expected only to increase further due to an aging population and growing prevalence of indications for anticoagulation. Therefore, health practitioners will be expected to manage an ever-increasing number of patients, presenting with a more diverse array of anticoagulants, including NOACs in the perioperative setting.

Perioperative Management
The current practice of NOAC management in the periopera- tive setting is primarily based on expert opinion derived from knowledge of drug pharmacokinetics and pharmaco- dynamics, with associated experience derived from large registration studies and post hoc subgroup analyses15–19 as well as emerging retrospective trials in specific popula- tions.20 Management of inevitable clinical scenarios such
as urgent surgery or surgical bleeding is based on consensus opinion derived from preclinical studies, laboratory or in vitro studies, through to the occasional phase 3 study. The following narrative review aims to provide an integrated summary of our own practice, the expert opinion-based guidelines, and the associated data relevant to NOACs and the perioperative period.

Factors to Consider
The main factors to consider in patients taking NOACs, when approaching the perioperative period, are outlined in ►Fig. 1 and are discussed in detail below.

Pharmacokinetics and Pharmacodynamics
The mechanism of action of rivaroxaban, apixaban, and edoxaban is via direct inhibition of Xa, while dabigatran directly inhibits the action of thrombin. The pharmacoki- netics of each drug is summarized in ►Table 1.21–42
The number of NOAC drug–drug interactions is signifi- cantly less compared with VKAs.43 The medications that may alter the NOAC anticoagulant effect and thus the risk of intra- and/or postoperative bleeding are specific to the type of NOAC and are listed in ►Table 2.38,39,44–47 Clearly, the concurrent use of NOACs with other antithrombotic medica-

tions will further compromise hemostasis.48 Drugs that increase bleeding risk via their antiplatelet effect, commonly aspirin or thienopyridine agents (clopidogrel, prasugrel, and ticagrelor), dipyridamole, as well as nonsteroidal anti-in- flammatory drugs (NSAIDs) are relevant. There is an increas- ing interest in combining anticoagulation with a NOAC and dual antiplatelet therapy to create a triple therapy ap- proach.49 Thrombolytic drugs may occasionally be used in patients with acute stroke prior to a surgical intervention and would further impede normal hemostasis.50

Clinically Significant Drug Levels
One of the advantages of the NOAC class over VKAs is the absence of requirement for regular laboratory monitoring. Conversely, when needed, it becomes more problematic to quantitate the anticoagulant effect of NOACs than with classical anticoagulants.51 Methods to measure drug levels directly or indirectly are available but usually not in routine use 24 hours a day in most laboratories. Even when it is possible to measure the drug concentration, interpretation is difficult due to scarcity of data correlating drug levels with clinical outcomes, including bleeding risk. The vast majority of clinicians caring for patients requiring surgery will have a timely and rapid access to routine coagulation tests such as the prothrombin time/international normalized ratio (PT/ INR) and the activated partial thromboplastin time (APTT), while the thrombin time (TT) is somewhat less widely available. Notwithstanding their inadequacies for measuring the effect of NOACs,52 there are some circumstances where standard coagulation studies (PT, APTT, TT) may assist in the management of NOACs in the perioperative setting. Overall, these assays may have some ability to exclude the presence of therapeutic or supratherapeutic NOAC levels; this ability is greatest with regard to dabigatran, and it may be possible in the case of rivaroxaban, but is not reliable in the case of apixaban.52
Dabigatran results in significant elevations of the TT, even when the drug is present in low concentrations.53 Therefore, a normal TT in a patient taking dabigatran generally indicates that the patient is not anticoagulated; however, occasionally normal TTs may be reported with certain instruments/re- agents in samples containing dabigatran.53 Therapeutic le- vels of dabigatran result in a prolonged APTT, and therefore a normal APTT suggests that the dabigatran level is unlikely to be high, and the bleeding risk is probably not significantly increased above the baseline. Therefore, to exclude clinically relevant anticoagulant effect of dabigatran, laboratories should provide rapid access to the TT and APTT. Clinicians should, however, also be aware that the TT is very sensitive, resulting in prolonged times at dabigatran concentrations well below observed trough levels, and it is thus useful to interpret the TT and APTT together. Rivaroxaban results in elevation of the PT, but the relationship between rivaroxaban levels and prolongation of the PT is dependent on the specific thromboplastin reagents used in the assay. Nevertheless, the presence of a normal PT in a patient taking rivaroxaban
usually suggests that a significant anticoagulation effect is unlikely.52,54

Fig. 1 Proposed management pathway of urgent surgery in patients taking non-vitamin K oral anticoagulants (NOACs). Notes: Edoxaban has not been included in view of limited clinical experience. Standard coagulation results may vary in different laboratories; suggest liaison with local laboratory regarding test/reagents in use and their sensitivity to the presence of NOAC. ωConsider the patient anticoagulated at the longer timeframe if bleeding risk of required operation is high and/or impaired renal function (especially if dabigatran). ^A normal activated partial thromboplastin time is associated with insignificant drug plasma levels in the majority, but not 100%, of cases. þIn the event of unacceptable risk of postoperative bleeding (e.g., neurosurgery), until further data are available, it may be reasonable to administer a further dose of idarucizumab. In other cases, further idarucizumab should be considered in the event of clinically relevant bleeding with evidence of rebound anticoagulation.

Table 1 Select NOAC pharmacokinetic data pertinent to perioperative management

Dabigatran Rivaroxaban Apixaban Edoxaban
Half-life
ClCr > 80 mL/min 12–14 h 7–8 h 11 h 9 h
50–80 mL/min 17 h 9 h 14 h 13 h
30–50 mL/min 19 h 9 h 18 h 17 h
Time to peak level 1.5 h 2 h 1–3 h 1.3 h
Peak levelsa (ng/mL)b Results expressed as mediana 133 (110 mg BD)89
183 (150 mg BD)89 249–274 μg/mL
(20 mg daily)23,90 67–123d (2.5 mg BD)24
132–171 (5 mg BD)24
251 (10 mg BD)24 85 (30 mg daily)22
175 (60 mg daily)22
Trough levelsa (ng/mL)b Results expressed as medianc 65 (110 mg BD)89
93 (150 mg BD)89 25–44 μg/mL (20 mg daily)23,90 32–79d (2.5 mg BD)24
63–103 (5 mg BD)24
120 (10 mg BD)24 18–27 (30 mg daily)25
36 (60 mg daily)25
Renal elimination 80% 66% 27% 50%
Dialysis effect Removed by dialysis Not removed by dialysis Not removed by dialysis Not removed by dialysis
Abbreviation: BD, twice daily.
aDaibgatran 1–3 hours post dose, rivaroxaban 2–4 hours post dose, apixaban 1–3 hours post dose, edoxaban 1–2 hours post dose. Levels expressed
derived from liquid chromatography and tandem mass spectrometry in dabigatran, apixaban, edoxaban.
bng/mL, except in the case of rivaroxaban: μg/mL
cA range of results represents a range of medians found either within a given study as a result of different populations assessed, or across different studies.
dWide variation of apixaban median ranges in context of dose-adjusted population in the ARISTOTLE study.24 Sources: Adapted from various studies.21–42,89,90

Table 2 Drug–drug interactions associated with the NOACs

Dabigatran Rivaroxaban Apixaban Edoxaban
Drugs that increase anticoagulant level Ketoconazolea Quinidineb Amiodarone Verapamil Ketoconazoled Clarithromycind Ritonavird Erythromycin Fluconazole Ketoconazolee Diltiazem Naproxen Ketoconazole Quinidine Verapamil Erythromycin Cyclosporine Amiodarone
Drugs that reduce anticoagulant level Rifampicin Carbamazepine Rifampicin Rifampicinf Atorvastatin Esomeprazole
Unknown effect Itraconazolec Tacrolimusc Cyclosporinec
aContraindicated.
bDose change to 150 mg daily dabigatran.
cUse not advised with dabigatran.
dAvoid concurrent use with rivaroxaban.
eAvoid concurrent use in apixaban if two of the following are present: age > 80, weight < 60 kg, serum creatinine > 1.5 mg/dL; else reduce to
2.5 mg BD.
fAvoid concurrent use in apixaban.
Source: Adapted from various studies.38,39,44–47

Apixaban, even at high levels, does not reliably elevate the PT, and there are no standard coagulation effects evidenced with routine assays that can help guide whether there is likely to be any significant anticoagulation effect in a patient taking apixaban.52–54 Given that the sensitivity to NOACs of commercial PT and APTT reagents varies, coagulation labora- tories should assess the sensitivity of their particular meth-
ods and disseminate this information among the clinicians who they service.
There are specialized coagulation tests that can measure NOAC levels indirectly. These largely comprise dilute throm- bin time (dTT) for dabigatran and NOAC-calibrated anti-Xa activity assays.52–54 A commercially available dTT (Hemo- clot) is the assay often recommended to determine the dabigatran drug level51; however, it is not approved for clinical use in all jurisdictions. In-house developed dTT assays and Ecarin-based clotting or chromogenic assays provide suitable alternatives if properly validated.52,53 The Russel’s viper venom time (RVVT) assay, routinely used in the assessment of the lupus anticoagulant, demonstrates a near- linear relationship to the dabigatran level.52,53 However, further evaluation is required to demonstrate its utility, perhaps in a modified form, in the assessment of dabigatran level.
Chromogenic anti-Xa assays specific for rivaroxaban and apixaban can be performed to quantify these drug plasma levels,52,54 but require the use of drug-appropriate calibra- tors, which may not be available in routine use in all laboratories. It may be tempting to use more widely available anti-Xa assays intended for LMWH and calibrated against a LMWH standard to measure rivaroxaban or apixaban levels in urgent situations; however, the measured drug levels may
be overestimated by a factor of 10,54 and thus this practice is not recommended.
A main benefit of knowing quantitative drug levels may arise in the setting of major trauma-related bleeding, or in

patients requiring urgent surgery. Elevated plasma levels do little more than provide confirmation that the patient is indeed anticoagulated, as drug level ranges have not been correlated to clinical outcomes and further estimation of extent of anticoagulation is not possible. There are currently no empiric data that evaluate a strategy incorporating either standard coagulation testing or specific coagulation testing into a pre-procedural management protocol. Given these practical limitations, these nonroutine coagulation tests are unlikely to significantly impact the management of NOACs in the perioperative setting.
Finally, whole blood viscoelastic point-of-care assays of hemostasis, such as thromboelastography (TEG) and rota- tional thromboelastometry (ROTEM), have been evaluated for measuring NOAC anticoagulant effects, although the majority of studies are small and/or used blood samples spiked with NOACs. While there is some evidence to suggest that these methods can differentiate between different NOACs and can indicate the magnitude of anticoagulant effect,55 a normal ROTEM/TEG does not exclude the presence of a NOAC. Other limiting issues are similar to the specific coagulation studies; the equipment may not be universally available, trained personnel are required to interpret find- ings, and test findings do not necessarily reflect clinical utility. However, these point-of-care methods are increas- ingly being used in the setting of major trauma, or critical surgical bleeding, and so further work in this area is anticipated.

Type and Urgency of Surgery
The very fact that a patient is treated with a NOAC indicates that the patient has an underlying thrombotic tendency. The crux of perioperative management of anticoagulants is bal- ancing the risk of bleeding versus the risk of thrombotic complications, both of which are influenced by several complex and potentially related factors. Each of these

requires a careful evaluation on a case-by-case basis when a patient presents with a surgical issue.
Based on the type, complexity, and duration of surgery, surgical procedures can be classified according to the asso- ciated bleeding risk. The most widely cited classification systems relating to NOACs are the European Heart Rhythm Association (EHRA),56 the Working Group on Perioperative Haemostasis and the French Study Group on Thrombosis and Haemostasis.57 The EHRA guidelines divide surgical bleeding risk into three groups: insignificant bleeding risk, low bleed- ing risk, and high bleeding risk, while the latter group divides surgical bleeding risk into low bleeding risk and moderate/ high bleeding risk. Examples of such classification systems
have previously been described in detail elsewhere.56,58 The
associated bleeding risk of the proposed surgery will ob- viously affect the clinical decision-making process in the setting of emergent or urgent surgery.

Availability of Reversal Agent In contrast to VKAs, which produce their anticoagulant effect by suppressing production of vitamin K–dependent coagula- tion factors, NOACs, with the exception of dabigatran,59 do not yet have reliable and clinically evaluated agents to reverse their anticoagulant effect. NOACs act as inhibitors of either factor Xa or thrombin; therefore, to reverse their anticoagulant effect is not as simple as adding exogenous coagulation factors to correct a deficiency. A more targeted and biologically plausible approach would be to neutralize the NOAC drug itself. Idarucizumab is now available as an antidote to dabigatran (and will be discussed below); how- ever, the drugs inhibiting factor Xa do not yet have an available antidote, although two pertinent NOAC-specific
antidotes are currently being investigated: ciraparantag and andexanet alfa.60,61
Somewhat counterintuitively, there is consistent evidence that despite the absence of NOAC antidotes (at the time of the studies), patients on NOACs do not appear to fare worse than those taking VKAs in the context of planned or unplanned surgical procedures, as well as in those with major bleed- ing.12,16,17 Regardless, the availability of a specific antidote would change the landscape when considering urgent sur- gery. Currently, consensus guidelines urge deferring surgical
procedures, if possible, for an additional 12 to 24 hours, to reduce the anticoagulation effect via continued drug elim- ination.19,56,57,62–64 Access to a specific antidote will help eliminate the often delicate balancing act facing the surgical team between the risks of delaying the surgery versus increased bleeding; however, the financial cost and future
implications of reexposure to antidotes would need to be considered.
Idarucizumab was approved in the United States and European Union in 2015, and Canada and Australia in 2016, with a wholesale cost of approximately 3,500 USD per dose. In humans, time to plasma peak is immediately after intravenous infusion. In doses of at least 5 g, it com- pletely reverses abnormal standard coagulation tests for at least 72 hours.65 It is equally effective in those with impaired renal function.65 So far there does not appear to be any

concerns with regard to development of inactivating anti- bodies in those exposed to idarucizumab, and in general, its safety profile appears adequate, including no evidence to suggest independent contribution to a prothrombotic state.65 In an interim analysis of 90 patients of the RE-VERSE AD trial, idarucizumab demonstrated complete reversal of laboratory parameters as well as improved clinical hemo- static outcomes, with clinician-determined hemostasis (e.g., cessation of bleeding) achieved at a median of 11.4 hours and normal intraoperative hemostasis reported in 92% of pa- tients.59 Surprisingly, despite the pharmacokinetic study findings, 18% of patients developed a “rebound,” with an increase of dabigatran levels (with associated standard coa- gulation study abnormalities) at 24 hours post idarucizumab administration. Putatively, some degree of rebound may be due to dabigatran’s volume of distribution, which is larger than that of idarucizumab.65 However, the idarucizumab dose of 5 g was specifically chosen to bind the 99th percen- tile of the total body dabigatran (based on the RE-LY study65) despite the practical implications of significant cost. These discordant findings are of interest, and the final analysis of the RE-VERSE AD trial is eagerly awaited.
In patients taking dabigatran, 5 g of idarucizumab should be administered prior to urgent surgery. The pharmacoki- netic data indicate that the timing of idarucizumab admin- istration prior to surgery is flexible; it appears reasonable to give the dose any time prior to induction. Subsequent routine coagulation testing and monitoring of laboratory hematological parameters should occur at least 12 hourly for the first 48 hours postoperatively, and liaison with the hematology laboratory service should be considered for evaluation of dabigatran level testing during this time. There may be clinical circumstances such as brain or spinal trau- matic injury, where it would be reasonable to consider repeat dosing of idarucizumab in the event of rebound anticoagulation effect, regardless of clinically relevant bleed- ing; however, we would recommend seeking an expert hematology consult.

Current Guidelines and the Quality of Evidence
The real-world data of NOAC-related perioperative manage- ment is scarce in comparison to VKAs. Management guides, created by independent groups/bodies, contain very similar suggestions for NOAC management in the perioperative period.19,56,57,62,63,66 ►Table 3 highlights pertinent informa- tion from some of the more recent management guides.56,57,62,67 The most recent guideline by EHRA56 pub- lished in 2015 incorporates the findings from a prospective study17 as well as subgroup analysis from a large phase 3 trial.15 The available published data for interruption of NOACs in the perioperative setting is essentially limited to patients with AF.17,68 Recently, a retrospective analysis of 190 consecutive patients treated with NOACs for VTE under- going surgery was presented at the American Society of Hematology 2016.20 A high bleeding risk operation was performed in 24% of cases with a low rate of recurrent VTE (1%) and major bleeding (0.5%), providing real-world evi-
dence that the perioperative strategy employed for patients

Table 3 Summary of some of the recent guides on preoperative cessation of NOACs based on bleeding risk and renal function

Guide Renal function CrCl mL/min Dabigatran Rivaroxaban Apixaban Edoxaban
Surgical bleeding risk
Low High Low High Low High
Heidbuchel et al (EHRA 2015)56 CrCl > 80 24 h 48 h 24 h 48 h 24 h 48 h As per rivaroxaban and apixaban
CrCl 50–80 36 h 72 h 24 h 48 h 24 h 48 h
CrCl 30–50 48 h 96 h 24 h 48 h 24 h 48 h
ClCr 15–30 N/a N/a 36 h 48 h 36 h 48 h
Sié et al (2013)57 Not considered Low bleeding risk: 24 h No guide
Moderate/high bleeding risk: 5 d
Liew and Douketis (2013)67 CrCl > 50 Low bleeding risk: 2 d High bleeding risk:3 d No guide
CrCl 30–50 Low bleeding risk: 3 d High bleeding risk: 4–5 d
Tran et al (2014)62 CrCl >50 Low bleeding risk: 24 h High bleeding risk: 48–72 h No guide
CrCl 30–50 Low bleeding risk: 48 h High bleeding risk: 72 h (96 for dabigatran)
Abbreviation: CrCl, creatinine clearance.

with AF is likely to be suitable for patients taking NOACs for prevention and management of VTE.

Bridging
Bridging is a term originally used to describe cessation of a long-acting anticoagulant, such as, VKAs, allowing the drug effect to wane, with administration of a short-acting alter- native anticoagulation (i.e., heparin or LMWH) prior to and immediately after an invasive procedure.69 It is becoming increasingly apparent that, in patients on VKAs, bridging is not necessary in certain well-defined patient groups (throm- bogenic cardiac arrhythmias with low to intermediate CHADS2 scores), and indeed it may be harmful.70 It should be noted that the principle of bridging is even less applicable to anticoagulation with NOACs due to the nature of their short half-lives.
The subgroup analysis of the RE-LY study demonstrated that bridging, when used in patients on dabigatran, resulted in significantly increased major bleeding (6.5 vs. 1.8%) with- out any improvement in rates of thromboembolic events.68 While these findings are reassuring, they are not automati- cally applicable to the majority of patients encountered in the perioperative settings, as the following patient groups were excluded: patients with high CHADS2 scores, patients with metallic cardiac prostheses, patients with recent major bleeding event, as well as patients with thromboembolic disease within the past 12 weeks. Nevertheless, as demon- strated by registry data from 2013 to 2015, the use of bridging is in decline.17 Data from the RE-LY trial demon- strate that although there is significant geographical practice variation, 85% of patients receiving dabigatran did not re- ceive perioperative bridging therapy. Given their rapid onset and cessation of anticoagulant effect (provided oral intake,

absorption, and renal function are not impaired), we agree with the most recent publications that, in general, bridging anticoagulation should not be used in patients on NOAC therapy (meaning dabigatran, apixaban, and rivaroxaban) for approved indications.56,67,71

Resumption of Anticoagulation To reduce the risk of devastating and potentially fatal throm- boembolic events, a return to full anticoagulation should be established as soon as possible in the postoperative period. However, this needs to be weighed against the risk of post- operative bleeding, which will vary according to the type of procedure. The evidence base supporting recommendations on NOAC resumption after invasive procedures is derived predominantly from expert opinion.19,56,62,67,69,72–74 After
low bleeding risk operations and nontraumatic spinal punc- ture or epidural anesthesia, full-dose NOAC therapy can be recommenced 6 to 8 hours postprocedure. If the postopera- tive bleeding risk is deemed to be high, such that it would outweigh the risk of thromboembolic events, then thera- peutic anticoagulation can be withheld for 48 to 72 hours. However, during this period, low-dose medical thrombopro- phylaxis (usually using LMWH) should be considered. The risk–benefit analysis needs to take into account the under- lying thromboembolic disorder and the risk of recurrence. There is currently no evidence to support the use of modified dose NOACs in this context and clinicians should refer to their local institution policies regarding medical thromboprophy- laxis dosing.

Emergency Surgery
First, there are no known clinical or laboratory-based pre- operative tools that adequately predict perioperative

bleeding, or perioperative thromboembolic events in any given individual.75,76 Although it may seem self-evident, in patients on anticoagulants, the strongest predictors of major bleeding and thromboembolic events is the need for urgent surgery.68 If a patient on a NOAC requires urgent surgery, the factors highlighted in ►Fig. 1 need to be considered. Where available, it is recommended to seek expert consultation. Unless the surgery can proceed regardless of anticoagulant
effect, then the anticoagulant should be stopped. An attempt to assess anticoagulant effect should be performed with standard and, if available, specific coagulation studies. If there is evidence that the NOAC is absent, or likely to be present at a clinically insignificant level, then the patient can proceed to surgery.
If there is evidence that the NOAC is present either by measuring drug levels or on the basis of prolongations of routine coagulation tests, then the patient should be con- sidered anticoagulated. In case of dabigatran, if idarucizu- mab is available, it should be administered as per the product information (5 g in two 2.5 g /50 mL vials intravenously over 5–10 minutes each) and the patient may then proceed to surgery. If idarucizumab is not available, hemodialysis may be considered in those with impaired renal function (GFR
< 50 mL/min) and/or evidence of supratherapeutic levels (i.e., plasma level >500 ng/mL or APTT >80).34 However, the patient should be monitored for rebound anticoagulant
effect once renal replacement therapy is discontinued.77 If the patient is taking a direct factor Xa inhibitor, then the surgery should be deferred if safe to do so, while maintaining normal physiology (to ensure ongoing renal NOAC elimina- tion), obtaining bleeding control via local hemostatic mea- sures, providing transfusion support to maintain normal physiology, and avoiding dilutional coagulopathy.
If the surgery cannot be deferred in patients on direct anti-Xa inhibitors or dabigatran where idarucizumab is not available, seek expert advice with regard to the use of prohemostatic factors such as prothrombin complex con- centrates (PCCs) and activated PCCs (aPCCs). The evidence for effectiveness of recombinant factor VIIa (rFVIIa) in this setting is even weaker, and thus should only be considered as a last resort in select cases.
However, clinicians should be aware that the prohemo- static products are not specific “reversal” agents when used in the context of NOACs. There is no clinical evidence to prove any reversal of clinical bleeding measures. Perhaps more importantly, owing to their prohemostatic nature, these products are known to cause thromboembolic events.78–80

Management of Intraoperative/Postoperative Bleeding
Management of intra- and postoperative bleeding in the context of recent NOAC therapy will depend on the NOAC type, presence of antiplatelet medications, and presence of conventional indicators such as dilutional coagulopathy from massive transfusion or concurrent disseminated intravascu- lar coagulation. In patients with critical bleeding, massive transfusion protocols should be implemented as per local institutional policy regardless of the availability of reversal

agents. Supportive care is crucial to successful resuscitation and it includes maintenance of adequate renal perfusion, control of blood loss where possible, and transfusion sup- port, including fresh frozen plasma (FFP), cryoprecipitate or fibrinogen concentrate, and platelet transfusion as recom- mended by local policy. Notwithstanding the study design limitations, findings from experimental animal models and in vitro studies suggest that aPCC and PCC appear to restore
laboratory parameters toward normal baseline, although the effects seem to be NOAC and animal model dependent.80–85 The human studies are less extensive, and are limited to evaluation of in vitro parameters,81,86,87 and are difficult to compare due to the use of a variety of PCC formulations containing either three or four factors. There is limited data to guide the use of FFP or fibrinogen concentrate; however, based on knowledge of the drug pharmacokinetics and pharmacodynamics, exceedingly large volumes of FFP would be required to absorb the unbound drug if present and would confer unnecessary risk (e.g., of fluid overload). As such, we
agree with existing recommendations against the use of FFP unless it is used in prevention of associated dilutional coagulopathy.56 Similarly, there are little data on the use of fibrinogen concentrate; accordingly, it should be reserved as a part of maintenance of physiologic fibrinogen levels. Current recommendations are PCC (three or four factors) 25 to 50 IU/kg and aPPC 30 to 50 IU/kg.56 In the event of ongoing bleeding despite the supportive measures and the administration of PCC or aPCC, discussion with the hematol- ogy service is recommended; rVIIa may be considered at this stage. There is very little evidence to support the use of antifibrinolytics in patients taking NOACs.88

Conclusion
Managing patients on NOACs in the perioperative period remains a challenging clinical task. Patients taking dabiga- tran with life-threatening or uncontrolled bleeding or urgent surgery now have access to a specific reversal agent: idar- ucizumab. Although further phase 3 trial results are pending, this may obviate the need for nonspecific reversal agents such as PCC, aPCC, and rVIIa in this patient group. Specific reversal agents for the anti-Xa oral anticoagulants are cur- rently undergoing evaluation in preclinical studies. In the meantime, there are no specific reversal agents available for the anti-Xa oral anticoagulants and the use of nonspecific prohemostatic agents (PCC, aPCC, and rVIIa as a last resort) should only be considered in the context of life-threatening bleeding or requirement for immediate surgery, while being mindful of the potential prothrombotic risk profile as well as uncertain impact on clinical bleeding outcomes.
Regardless of the availability of specific reversal agents, liaison with the hematology service is strongly recom- mended in patients presenting with clinically significant bleeding or requirements for urgent surgery. Additionally, there are laboratory parameters that will affect evaluation of anticoagulation status that will vary depending on local laboratory factors. We suggest the laboratories engage with the clinicians to increase their awareness of the

availability and characteristics of laboratory assays per- formed within their local facilities to measure the antic- oagulant effect of NOACs used in their jurisdiction.

References
1 Connolly SJ, Ezekowitz MD, Yusuf S, et al; RE-LY Steering Com- mittee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 2009;361(12):1139–1151
2 Patel MR, Mahaffey KW, Garg J, et al; ROCKET AF Investigators. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011;365(10):883–891
3 Granger CB, Alexander JH, McMurray JJ, et al; ARISTOTLE Com- mittees and Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2011;365(11):981–992
4 Schulman S, Kearon C, Kakkar AK, et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med 2009;361(24):2342–2352
5 Bauersachs R, Berkowitz SD, Brenner B, et al; EINSTEIN Investi- gators. Oral rivaroxaban for symptomatic venous thromboembo- lism. N Engl J Med 2010;363(26):2499–2510
6 Büller HR, Prins MH, Lensin AW, et al; EINSTEIN–PE Investigators. Oral rivaroxaban for the treatment of symptomatic pulmonary embolism. N Engl J Med 2012;366(14):1287–1297
7 Agnelli G, Buller HR, Cohen A, et al; AMPLIFY Investigators. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med 2013;369(09):799–808
8 Mega JL, Braunwald E, Wiviott SD, et al; ATLAS ACS 2–TIMI 51 Investigators. Rivaroxaban in patients with a recent acute cor- onary syndrome. N Engl J Med 2012;366(01):9–19
9 Connolly SJ, Eikelboom J, Joyner C, et al; AVERROES Steering Committee and Investigators. Apixaban in patients with atrial fibrillation. N Engl J Med 2011;364(09):806–817
10 Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: chest guideline and expert panel report. Chest 2016; 149(02):315–352
11 Lauffenburger JC, Farley JF, Gehi AK, Rhoney DH, Brookhart MA, Fang G. Factors driving anticoagulant selection in patients with atrial fibrillation in the United States. Am J Cardiol 2015; 115(08):1095–1101
12 Beyer-Westendorf J, Forster K, Pannach S, Ebertz F, Gelbricht V, Thieme C. Rates, management and outcome of bleeding compli- cations during rivaroxaban therapy in daily care: results from the Dresden NOAC registry. Blood 2014;124(06):955–962
13 Zolfaghari S, Harenberg J, Frölich L, et al. Development of recom- mendations to continue anticoagulation with one of the two types of oral anticoagulants based on the identification of pa- tients’ preference. Semin Thromb Hemost 2015;41(02):166–177
14 Wysokinski WE, McBane RD II. Periprocedural bridging manage- ment of anticoagulation. Circulation 2012;126(04):486–490
15 Healey JS, Eikelboom J, Douketis J, et al; 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) rando- mized trial. Circulation 2012;126(03):343–348
16 Douketis JD, Healey JS, Brueckmann M, et al. Urgent surgery or procedures in patients taking dabigatran or warfarin: analysis of perioperative outcomes from the RE-LY trial. Thromb Res 2016; 139:77–81
17 Beyer-Westendorf J, Gelbricht V, Förster K, et al. Peri-interven- tional management of novel oral anticoagulants in daily care: results from the prospective Dresden NOAC registry. Eur Heart J 2014;35(28):1888–1896
18 Deharo JC, Sciaraffia E, Leclercq C, et al; Coordinated by the Scientific Initiatives Committee of the European Heart Rhythm Association. Perioperative management of antithrombotic treat-

ment during implantation or revision of cardiac implantable electronic devices: the European Snapshot Survey on Procedural Routines for Electronic Device Implantation (ESS-PREDI). Euro- pace 2016;18(05):778–784
19 Faraoni D, Levy JH, Albaladejo P, Samama C-M; Groupe d’Intérêt en Hémostase Périopératoire. Updates in the perioperative and emergency management of non-vitamin K antagonist oral antic- oagulants. Crit Care 2015;19(01):203
20 Shaw J, de Wit C, Le Gal G, Carrier M. Bleeding and Thrombotic Outcomes Following Perioperative Interruption of Direct Oral Anticoagulants in Patients with Prior Venous Thromboembolic Disease. Poster presented at the Annual Meeting/of American Society of Hematology; December 3–6, 2016; San Diego, CA
21 Mueck W, Borris LC, Dahl OE, et al. Population pharmacokinetics and pharmacodynamics of once- and twice-daily rivaroxaban for the prevention of venous thromboembolism in patients under- going total hip replacement. Thromb Haemost 2008; 100(03):453–461
22 Weitz JI, Connolly SJ, Patel I, et al. Randomised, parallel-group, multicentre, multinational phase 2 study comparing edoxaban, an oral factor Xa inhibitor, with warfarin for stroke prevention in patients with atrial fibrillation. Thromb Haemost 2010; 104(03):633–641
23 Mueck W, Lensing AW, Agnelli G, Decousus H, Prandoni P, Misselwitz F. Rivaroxaban: population pharmacokinetic analyses in patients treated for acute deep-vein thrombosis and exposure simulations in patients with atrial fibrillation treated for stroke prevention. [Erratum appears in Clin Pharmacokinet. 2012 Feb; 51(2):136]Clin Pharmacokinet 2011;50(10):675–686
24 Bristol-Myers-Squibb. Summary of product information—Eliquis. 2011. Available at: http://www.ema.europa.eu/docs/en_GB/doc- ument_library/EPAR_-_Product_Information/human/002148/ WC500107728.pdf. Accessed January 9, 2017
25 Ruff CT, Giugliano RP, Braunwald E, et al. Association between edoxaban dose, concentration, anti-Factor Xa activity, and out- comes: an analysis of data from the randomised, double-blind ENGAGE AF-TIMI 48 trial. Lancet 2015;385(9984):2288–2295
26 Skeppholm M, Al-Aieshy F, Berndtsson M, et al. Clinical evaluation of laboratory methods to monitor apixaban treatment in patients with atrial fibrillation. Thromb Res 2015;136(01):148–153
27 Kubitza D, Becka M, Voith B, Zuehlsdorf M, Wensing G. Safety, pharmacodynamics, and pharmacokinetics of single doses of BAY 59-7939, an oral, direct factor Xa inhibitor. Clin Pharmacol Ther 2005;78(04):412–421
28 Kubitza D, Becka M, Wensing G, Voith B, Zuehlsdorf M. Safety, pharmacodynamics, and pharmacokinetics of BAY 59-7939–an oral, direct Factor Xa inhibitor–after multiple dosing in healthy male subjects. Eur J Clin Pharmacol 2005;61(12):873–880
29 Stangier J, Rathgen K, Stähle H, Gansser D, Roth W. The pharma- cokinetics, pharmacodynamics and tolerability of dabigatran etexilate, a new oral direct thrombin inhibitor, in healthy male subjects. Br J Clin Pharmacol 2007;64(03):292–303
30 Stangier J, Stähle H, Rathgen K, Fuhr R. Pharmacokinetics and pharmacodynamics of the direct oral thrombin inhibitor dabiga- tran in healthy elderly subjects. Clin Pharmacokinet 2008; 47(01):47–59
31 Kubitza D, Becka M, Mueck W, et al. Effects of renal impairment on the pharmacokinetics, pharmacodynamics and safety of rivarox- aban, an oral, direct Factor Xa inhibitor. Br J Clin Pharmacol 2010; 70(05):703–712
32 Leil TA, Feng Y, Zhang L, Paccaly A, Mohan P, Pfister M. Quantifica- tion of apixaban’s therapeutic utility in prevention of venous thromboembolism: selection of phase III trial dose. Clin Pharma- col Ther 2010;88(03):375–382
33 Ogata K, Mendell-Harary J, Tachibana M, et al. Clinical safety, tolerability, pharmacokinetics, and pharmacodynamics of the novel factor Xa inhibitor edoxaban in healthy volunteers. J Clin Pharmacol 2010;50(07):743–753

34 Stangier J, Rathgen K, Stähle H, Mazur D. Influence of renal impair- ment on the pharmacokinetics and pharmacodynamics of oral dabigatran etexilate: an open-label, parallel-group, single-centre study. Clin Pharmacokinet 2010;49(04):259–268
35 Janssen-Pharmaceuticals. Xarelto Highlights of Prescribing Infor- mation. 2011. Available at: http://www.xareltohcp.com/shared/ product/xarelto/prescribing-information.pdf. Accessed January 9, 2017
36 Frost C, Wang J, Nepal S, et al. Apixaban, an oral, direct factor Xa inhibitor: single dose safety, pharmacokinetics, pharmacody- namics and food effect in healthy subjects. Br J Clin Pharmacol 2013;75(02):476–487
37 Yin OQP, Tetsuya K, Miller R. Edoxaban population pharmacoki- netics and exposure-response analysis in patients with non- valvular atrial fibrillation. Eur J Clin Pharmacol 2014; 70(11):1339–1351
38 Boehringer-Ingelheim-Pharmaceuticals. Pradaxa – Highlights of Prescribing Information. 2015. Available at: http://docs.boehrin- ger-ingelheim.com/Prescribing%20Information/PIs/Pradaxa/Pra- daxa.pdf. Accessed January 9, 2017
39 Daiichi-Sankyo. Savaysa Highlights of Prescribing Information. 2015. Available at: http://www.accessdata.fda.gov/drugsatfda_ docs/label/2015/206316lbl.pdf. Accessed January 9, 2017
40 Parasrampuria DA, Marbury T, Matsushima N, et al. Pharmaco- kinetics, safety, and tolerability of edoxaban in end-stage renal disease subjects undergoing haemodialysis. Thromb Haemost 2015;113(04):719–727
41 Bristol-Myers-Squibb. Eliquis Highlights of Prescribing Informa- tion. 2016. Available at: http://packageinserts.bms.com/pi/pi_ eliquis.pdf. Accessed January 9, 2017
42 Husted S, Verheugt FW, Comuth WJ. Reversal strategies for NOACs: state of development, possible clinical applications and future perspectives. Drug Saf 2016;39(01):5–13
43 Douketis JD. Pharmacologic properties of the new oral antic- oagulants: a clinician-oriented review with a focus on periopera- tive management. Curr Pharm Des 2010;16(31):3436–3441
44 European Medicines Agency. Pradaxa – Procedural steps taken and scientific information after the authorisation. 2016. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/ EPAR_-_Procedural_steps_taken_and_scientific_information_ after_authorisation/human/000829/WC500041063.pdf. Accessed January 9, 2017
45 Bristol-Myers-Squibb. Eliquis – Highlights of prescribing informa- tion. 2012. Available at: https://www.accessdata.fda.gov/drugsatf- da_docs/label/2012/202155s000lbl.pdf. Accessed January 9, 2017
46 Liesenfeld KH, Lehr T, Dansirikul C, et al. Population pharmaco- kinetic analysis of the oral thrombin inhibitor dabigatran etex- ilate in patients with non-valvular atrial fibrillation from the RE-LY trial. J Thromb Haemost 2011;9(11):2168–2175
47 Janssen Pharmaceuticals. Xarelto – Highlights of prescribing infor- mation. 2016. Available at: https://www.xareltohcp.com/shared/ product/xarelto/prescribing-information.pdf. Accessed January 9, 2017
48 Alfredsson J, Roe MT. Risks and benefits of triple oral anti- thrombotic therapies after acute coronary syndromes and percu- taneous coronary intervention. Drug Saf 2015;38(05):481–491
49 Elewa H, Ahmed D, Barnes GD. Triple oral antithrombotic therapy in atrial fibrillation and coronary artery stenting: searching for the best combination. Semin Thromb Hemost 2016;42(06):662–670
50 Kepplinger J, Prakapenia A, Barlinn K, et al. Standardized use of novel oral anticoagulants plasma level thresholds in a new thrombolysis decision making protocol. J Thromb Thrombolysis 2016;41(02):293–300
51 Lippi G, Favaloro EJ. Recent guidelines and recommendations for laboratory assessment of the direct oral anticoagulants (DOACs): is there consensus? Clin Chem Lab Med 2015;53(02):185–197
52 Favaloro EJ, Lippi G. Laboratory testing in the era of direct or non- vitamin K antagonist oral anticoagulants: a practical guide to

measuring their activity and avoiding diagnostic errors. Semin Thromb Hemost 2015;41(02):208–227
53 Bonar R, Favaloro EJ, Mohammed S, Pasalic L, Sioufi J, Marsden K. The effect of dabigatran on haemostasis tests: a comprehensive assessment using in vitro and ex vivo samples. Pathology 2015; 47(04):355–364
54 Bonar R, Favaloro EJ, Mohammed S, et al. The effect of the direct factor Xa inhibitors apixaban and rivaroxaban on haemostasis tests: a comprehensive assessment using in vitro and ex vivo samples. Pathology 2016;48(01):60–71
55 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(05):665–673
56 Heidbuchel H, Verhamme P, Alings M, et al. Updated European Heart Rhythm Association Practical Guide on the use of non- vitamin K antagonist anticoagulants in patients with non-valvu- lar atrial fibrillation. Europace 2015;17(10):1467–1507
57 Sié P, Samama CM, Godier A, et al; Working Group on Periopera- tive Haemostasis; French Study Group on Thrombosis and Haemostasis. Surgery and invasive procedures in patients on long-term treatment with direct oral anticoagulants: thrombin or factor-Xa inhibitors. Recommendations of the Working Group on Perioperative Haemostasis and the French Study Group on Thrombosis and Haemostasis. Arch Cardiovasc Dis 2011; 104(12):669–676
58 Burnett AE, Mahan CE, Vazquez SR, Oertel LB, Garcia DA, Ansell J. Guidance for the practical management of the direct oral antic- oagulants (DOACs) in VTE treatment. J Thromb Thrombolysis 2016;41(01):206–232
59 Pollack CVJ Jr, Reilly PA, Eikelboom J, et al. Idarucizumab for dabigatran reversal. N Engl J Med 2015;373(06):511–520
60 Siegal DM, Curnutte JT, Connolly SJ, et al. Andexanet alfa for the reversal of factor Xa inhibitor activity. N Engl J Med 2015; 373(25):2413–2424
61 Arbit B, Nishimura M, Hsu JC. Reversal agents for direct oral anticoagulants: a focused review. Int J Cardiol 2016;223:244–250
62 Tran H, Joseph J, Young L, et al; Australasian Society of Thrombosis and Haemostasis. New oral anticoagulants: a practical guide on prescription, laboratory testing and peri-procedural/bleeding management. Intern Med J 2014;44(06):525–536
63 Heidbuchel H, Verhamme P, Alings M, et al; 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(05):625–651
64 Munson CF, Reid AJ. Novel oral anticoagulants in plastic surgery. J Plast Reconstr Aesthet Surg 2016;69(05):585–593
65 Reilly PA, van Ryn J, Grottke O, Glund S, Stangier J. Idarucizumab, a specific reversal agent for dabigatran: mode of action, pharma- cokinetics and pharmacodynamics, and safety and efficacy in phase 1 subjects. Am J Emerg Med 2016;34(11S, 11s):26–32
66 Weitz JI, Pollack CV Jr. Practical management of bleeding in patients receiving non-vitamin K antagonist oral anticoagulants. Thromb Haemost 2015;114(06):1113–1126
67 Liew A, Douketis J. Perioperative management of patients who are receiving a novel oral anticoagulant. Intern Emerg Med 2013; 8(06):477–484
68 Douketis JD, Healey JS, Brueckmann M, et al. Perioperative brid- ging anticoagulation during dabigatran or warfarin interruption among patients who had an elective surgery or procedure. Sub- study of the RE-LY trial. Thromb Haemost 2015;113(03):625–632
69 Ortel TL. Perioperative management of patients on chronic an- tithrombotic therapy. Blood 2012;120(24):4699–4705
70 Douketis JD, Spyropoulos AC, Kaatz S, et al; BRIDGE Investigators. Perioperative bridging anticoagulation in patients with atrial fibrillation. N Engl J Med 2015;373(09):823–833
71 Mar PL, Familtsev D, Ezekowitz MD, Lakkireddy D, Gopinathan- nair R. Periprocedural management of anticoagulation in patients

taking novel oral anticoagulants: review of the literature and recommendations for specific populations and procedures. Int J Cardiol 2016;202:578–585
72 Ward C, Conner G, Donnan G, Gallus A, McRae S. Practical management of patients on apixaban: a consensus guide. Thromb J 2013;11(01):27
73 Untereiner O, Seince P-F, Chterev V, et al. Management of direct oral anticoagulants in the perioperative setting. J Cardiothorac Vasc Anesth 2015;29(03):741–748
74 Breuer G, Weiss DR, Ringwald J. ‘New’ direct oral anticoagulants in the perioperative setting. Curr Opin Anaesthesiol 2014; 27(04):409–419
75 Gombotz H, Knotzer H. Preoperative identification of patients with increased risk for perioperative bleeding. Curr Opin Anaes- thesiol 2013;26(01):82–90
76 Douketis JD. Perioperative management of patients who are receiving warfarin therapy: an evidence-based and practical approach. Blood 2011;117(19):5044–5049
77 Chai-Adisaksopha C, Hillis C, Lim W, Boonyawat K, Moffat K, Crowther M. Hemodialysis for the treatment of dabigatran-asso- ciated bleeding: a case report and systematic review. J Thromb Haemost 2015;13(10):1790–1798
78 Levi M, Levy JH, Andersen HF, Truloff D. Safety of recombinant activated factor VII in randomized clinical trials. N Engl J Med 2010;363(19):1791–1800
79 Dentali F, Marchesi C, Giorgi Pierfranceschi M, et al. Safety of prothrombin complex concentrates for rapid anticoagulation reversal of vitamin K antagonists. A meta-analysis. Thromb Hae- most 2011;106(03):429–438
80 Dickneite G, Hoffman M. Reversing the new oral anticoagulants with prothrombin complex concentrates (PCCs): what is the evidence? Thromb Haemost 2014;111(02):189–198
81 Lee FM, Chan AK, Lau KK, Chan HH. Reversal of new, factor-specific oral anticoagulants by rFVIIa, prothrombin complex concentrate and activated prothrombin complex concentrate: a review of animal and human studies. Thromb Res 2014;133(05):705–713
82 Grottke O, van Ryn J, Spronk HM, Rossaint R. Prothrombin complex concentrates and a specific antidote to dabigatran are

effective ex-vivo in reversing the effects of dabigatran in an anticoagulation/liver trauma experimental model. Crit Care 2014;18(01):R27
83 Martin AC, Le Bonniec B, Fischer AM, et al. Evaluation of recom- binant activated factor VII, prothrombin complex concentrate, and fibrinogen concentrate to reverse apixaban in a rabbit model of bleeding and thrombosis. Int J Cardiol 2013;168(04): 4228–4233
84 Pragst I, Zeitler SH, Doerr B, Kaspereit FJ, Herzog E, Dickneite G. Reversal of dabigatran anticoagulation by prothrombin complex concentrate (Beriplex P/N) in a rabbit model. J Thrombos Haemost 2012;10(09):1841–1848
85 Godier A, Miclot A, Le Bonniec B, et al. Evaluation of prothrombin complex concentrate and recombinant activated factor VII to reverse rivaroxaban in a rabbit model. Anesthesiology 2012; 116(01):94–102
86 Escolar G, Arellano-Rodrigo E, Reverter JC, et al. Reversal of apixaban induced alterations of hemostasis by different coagula- tion factor concentrates: studies in vitro with circulating human blood. Abstracts from the emerging science series. Circulation 2012;126(04):520–521
87 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(14): 1573–1579
88 Shih A, Crowther MA. Reversal of direct oral anticoagulants: a prac- tical approach. Presented at the Annual Meeting/ of the American Society of Hematology; December 3–6, 2016; San Diego, CA
89 Reilly PA, Lehr T, Haertter S, et al; RE-LY Investigators. The effect of dabigatran plasma concentrations and patient characteristics on the frequency of ischemic stroke and major bleeding in atrial fibrillation patients: the RE-LY Trial (Randomized Evaluation of Long-Term Anticoagulation Therapy). J Am Coll Cardiol 2014; 63(04):321–328
90 Mueck W, Stampfuss J, Kubitza D, Becka M. Clinical pharmacoki- netic and pharmacodynamic profile of rivaroxaban. Clin Pharma- cokinet 2014;53(01):1–16