key: cord-272060-o0wx0add authors: Li, Allen; Li, Ming K.; Crowther, Mark; Vazquez, Sara R. title: Drug-drug interactions affecting drug levels of direct oral anticoagulants in the real world: A systematic review() date: 2020-08-11 journal: Thromb Res DOI: 10.1016/j.thromres.2020.08.016 sha: doc_id: 272060 cord_uid: o0wx0add BACKGROUND: Direct oral anticoagulants (DOACs) have emerged as safe and effective alternatives to Vitamin-K antagonists for treatment and prevention of arterial and venous thrombosis. Due to their novelty, pharmacokinetic DOAC drug-drug interactions (DDIs) that result in clinical adverse events have not been well-documented. OBJECTIVE: This study aims to systematically review reported pharmacokinetic DDIs resulting in clinical adverse events through documented observational evidence to better inform clinicians in clinical practice. METHODS: A comprehensive literature review of EMBASE, MEDLINE, and Ovid HealthStar was conducted through March 10th, 2020. Two independent reviewers screened and extracted data from eligible articles according to pre-established inclusion and exclusion criteria. Articles reporting bleeding or thrombotic outcomes in non-controlled (observational) settings resulting from suggested pharmacokinetic DOAC DDIs were included. RESULTS: A total of 5567 citations were reviewed, of which 24 were included following data extraction. The majority were case reports (n = 21) documenting a single adverse event resulting from a suspected DOAC DDI, while the remaining papers were a case series (n = 1) and cohort studies (n = 2). The most commonly reported interacting drugs were amiodarone and ritonavir (bleeding), and phenobarbital, phenytoin, and carbamazepine (thrombosis). Bleeding events more often resulted from a combined mechanism (P-glycoprotein AND CYP3A4 inhibition), whereas thrombotic events resulted from either combined OR single P-glycoprotein/CYP3A4 induction. CONCLUSION: Current literature evaluating the real-world risk of DOAC DDIs is limited to few case reports and retrospective observational analyses. Clinicians are encouraged to continue to report suspected drug interactions resulting in adverse events. Warfarin has historically been a cornerstone for treating or preventing thrombosis in various settings. Warfarin, due to its variable interpatient response, requires systematic monitoring of the international normalized ratio (INR) to ensure safety and efficacy. Warfarin is subject to numerous food and drug interactions, and there is reasonable quality evidence that some of these interactions are associated with bleeding and/or thrombotic complications. 1, 2 However, the universal availability of international normalized ratio (INR) testing can mitigate the impact of these interactions by allowing real-time dose adjustment. In the last decade, direct oral anticoagulants (DOACs) have emerged as an effective and safe alternative to warfarin. [3] [4] [5] DOACs are administered in fixed doses and have fewer drug-drug interactions (DDIs) compared to warfarin but are still subject to DDI-induced alterations in plasma concentrations that may result in bleeding or thrombotic events. All DOACs (including dabigatran, rivaroxaban, apixaban, and edoxaban) are substrates of the efflux transporter permeability-glycoprotein (P-gp, also referred to as p-gp transportor, ABCB1, and p-gp multidrug transporter), which regulates the absorption of DOACs from the gastrointestinal lumen and is also involved in their hepatic and renal excretion. Additionally, rivaroxaban and apixaban have CYP450-mediated metabolism (rivaroxaban primarily by CYP3A4 with minor contribution by CYP2J2 and apixaban primarily by CYP3A4 with minor contributions by CYP2C19, CYP1A2, CYP2C8, and CYP2C9). 6, 7 Drugs that induce P-gp or CYP3A4 may reduce the plasma concentration of the DOAC; conversely, drugs that inhibit P-gp or CYP3A4 may increase the plasma concentration of the DOAC. Although fewer in number than warfarin, 1, 2, 8 the clinical importance of pharmacokinetic DOAC DDIs is increased as the lack of an "INR-equivalent" results in less testing and monitoringthus such interactions are likely to remain unnoticed until a complication occurs since DOAC levels are not routinely monitored. Currently, pharmacokinetic studies, case reports, and guidance documents based on these reports in the literature propose several pharmacokinetic DDIs for DOACs demonstrated by significant alterations in plasma DOAC concentrations 9-18 but some occur in the absence of adverse clinical events. [19] [20] [21] [22] While the ability of P-gp and/or CYP3A4 modifiers to alter DOAC drug levels is well-established, given the wider therapeutic index of the DOACs relative to a narrow therapeutic index drug like warfarin, it is unknown if these pharmacokinetic alterations are sufficient to contribute to clinical adverse events. Therefore, to aid clinicians in deciphering the clinical significance of DOAC DDIs, this study seeks to systematically evaluate the existing body of literature of pharmacokinetic DOAC DDIs that result in clinical adverse events (bleeding and thrombosis) in uncontrolled clinical settings. As the pharmacodynamic interactions between anticoagulants and antiplatelet medications, non-steroidal anti-inflammatory drugs (NSAIDs), and selective serotonin reuptake inhibitors (SSRIs) and selective norepinephrine reuptake inhibitors (SNRIs) are known to enhance bleeding risk [23] [24] [25] [26] [27] [28] [29] Studies to be included in the review had to report either a thrombotic or bleeding event due to potential pharmacokinetic drug-drug interaction between a DOAC an another drug in a non-controlled clinical setting. Studies that did not control for concurrent administration of SSRIs, NSAIDs, antiplatelets, thrombolytics, and other anticoagulants were excluded as these drugs can independently cause bleeding or as the result of pharmacodynamic interactions. For each study that was included, investigators evaluated the study quality (if applicable) using the NewCastle-Ottawa Scale, 31 and extracted data using precomposed data extraction templates on Microsoft Excel. Extracted data included outcome observed (bleeding or thrombosis), DOAC involved, interacting drug, study population, coagulation or DOAC plasma concentration measurements (if available), and a drug-interaction probability score (DIPS) (if available). Using a 10-item questionnaire, the DIPS tool considers a variety of factors including alternative causes, and previous documentation of the DDI. 32, 33 It is a widely used reference standard for determining the probability of drug interactions and has been shown through a 2015 systematic review to overcome limitations present in other assessment instruments. 32, 33 Data synthesis involved utilizing tables and descriptive statistics to summarize data. The mechanism of the interaction was objectively identified using Lexi-Drugs Wolters Kluwer Clinical Drug Information database. After our initial search among the databases, 6,580 articles were identified from our search. Of these studies, a total of 1,013 duplicates were removed with 5,567 proceeding to title and abstract screening. Of these, 5,453 articles were deemed ineligible, with 114 articles moved to full-text review. A total of 90 citations were excluded based on exclusion criteria, leaving 24 for data extraction (Figure 1 , online supplement). Of the 24 included publications, 21 were case reports, one was a case series, and the remaining two were large cohort studies. Of the case reports, just over half (n=11) reported DDIs resulting in thrombotic outcomes while the other half reported bleeding outcomes. The most frequently reported DDI for bleeding events in the case reports involved the combinations of amiodarone and dabigatran (via inhibition of P-gp) and ritonavir and rivaroxaban (via inhibition of both P-gp and CYP3A4). The most frequently reported DDI for thrombosis involved the antiepileptic drugs phenobarbital, phenytoin, and carbamazepine (via induction of P-gp and/or CYP3A4). Five case reports included a statement involving a DIPS (Drug Interaction Probability Scale) score (Table 1 , online supplement). In cases that also reported laboratory values (13 of 21 reports), seven cases reported only coagulation laboratory tests, three reported only DOAC plasma concentrations,and three reported a combination of coagulation and DOAC-specific laboratory tests. In the bleeding cases reporting coagulation laboratory results, five reported the prothrombin time (PT), four reported the aPTT, and two reported the INR; all results were above the reference range. For the thrombotic cases, one case reported an elevated PT but all J o u r n a l P r e -p r o o f other coagulation labs were within normal limits. Six of the 20 case reports reported DOAC-specific drug levels. All three bleeding cases reported above on-therapy range DOAC levels. One thrombotic case reported an on-therapy trough for apixaban in the presence of phenobarbital, yet a random plasma apixaban level quadrupled with the removal of phenobarbital. We found one case series by Bortz et al, which evaluated a series of patients treated with rivaroxaban and carbamazepine.Four of seven patients in this study developed some form of recurrent or extension of a thrombus during concurrent treatment ( Table 2 , online supplement). 34 Both cohort studies scored a perfect on the NewCastle-Ottawa Scale for Nonrandomized Studies and assessed bleeding outcomes. Chang, et al identified 91,330 Taiwanese atrial fibrillation patients taking DOACs during a 5-year period and analyzed them for bleeding events (defined as a hospitalization or an emergency department visit with a primary diagnosis of intracranial hemorrhage or gastrointestinal, urogenital, or other bleeding event) during DOAC therapy with and without the presence of CYP3A3 and P-gp modifying drugs. 35 Results demonstrated that after inverse probability of treatment weighting using the propensity score including adjustment for several confounders, the use of dabigatran or rivaroxaban with amiodarone, fluconazole, or phenytoin was associated with a higher rate of major bleeding; apixaban combined with fluconazole or cyclosporine was associated with a higher rate of bleeding. 35 (Table 3 , online supplement)Pham et al, also evaluated bleeding outcomes. 36 Using the IBM Watson MarketScan Databases, J o u r n a l P r e -p r o o f this study identified 9,886 patients that were prescribed a DOAC with concomitant verapamil, diltiazem, amlodipine, or metoprolol from Janurary 2019 to July 2019. Bleeding rates were compared between patients that were prescribed known CYP3A4 and P-gp transporter inhibitors (verapamil or diltiazem) with a DOAC to patients prescribed with non-interacting drugs (amlodipine or metoprolol) with a DOAC. Dabigatran had statistically significant increases in bleeding risk when paired with diltiazem than with amlodipine and when paired with verapamil than with metoprolol. The rest of the comparisons did not reach statistical significance (Table 4 , online supplement).Among both case reports and and the cohort study, 11 unique drugs were implicated as a contributor to bleeding events. The most commonly identified mechanism of interaction was a drug that was a combined inhibitor of CYP3A4 and Pgp. For the case reports of thrombotic adverse events, eight unique drugs were implicated, with only three having a combined CYP3A4/P-gp induction mechanism. Three reports of DDIs causing bleeding events had an unclear mechanism (topical miconazole, loperamide, and phenytoin) ( Only five of the 20 cases reported DOAC-specific drug levels in the setting of adverse events, which reflects the limited availability of these laboratory tests. Of note, DOAC plasma concentrations were above the published on-therapy reference ranges in all bleeding cases, as were the more widely available coagulation laboratory tests. Recently, one Italian center noted a six-fold increase in the trough DOAC concentration of patients hospitalized for COVID-19 pneumonia who were also receiving antiviral therapy with ritonavir. 62 No bleeding complications were reported in these patients; however, over half had the DOAC discontinued as a result of the concerning elevation in trough concentration. In our study, a DOAC-ritonavir DDI was responsible for two reported bleeding events, and given this risk, DOAC-specific labeling recommendations include avoiding this drug combination or employing a DOAC dose reduction. 55, 56, 58 Ritonavir is likely a contributor to the elevated DOAC concentrations in these COVID-19 patients, but the scenario is likely multifactorial. In addition to antiviral therapy with ritonavir, some patients in the Italian study were also receiving darunavir (a strong CYP3A4 inhibitor) and/or azithromycin (a p-gp inhibitor) which could represent an additive DDI effect on DOAC concentrations. Additionally, renal impairment is a common sequela of COVID-19 infection and could impair DOAC clearance. 63 In our study, all but one coagulation test was within normal limits for patients with thrombotic events. This reinforces the concept that standard coagulation tests may be helpful to qualitatively assess excess DOAC effect in the absence of DOAC-specific assays 64 but may have limited utility in suspected DOAC failures or thrombotic events. The included cohort studies also warrants discussion. The Chang, et al study's findings with amiodarone are consistent with the case reports in this analysis and with other literature finding no difference in adverse outcomes with the combination of apixaban and amiodarone. 65 However, a systematic review showed no difference in clinical outcomes in patients taking any DOAC and amiodarone. 66 The finding that phenytoin contributed to increased bleeding risk is paradoxical given the proposed however, this may be due to Pham et al, using an active comparator group instead of a non-user group. Overall, an interesting mechanistic finding in this study is that more DDI-related bleeding adverse events were likely resulting from a combination CYP3A4/P-gp inhibition mechanism, whereas thrombotic adverse events could have been either from a single CYP3A4 inducer or a combined CYP3A4/P-gp inducer. A sub-analysis of the apixaban atrial fibrillation clinical trial found no significant differences in bleeding outcomes in patients taking apixaban and amiodarone, 65 and as mentioned above, a systematic review showed no difference in clinical outcomes in patients taking any DOAC and amiodarone. 62 The Chang,et al study showed an increase in bleeding related to combinations of dabigatran-amiodarone and rivaroxaban-amiodarone, but not apixaban-amiodarone. 35 As previously noted, amiodarone-dabigatran-related bleeding was cited in two case reports 40, 41 . Interestingly, the manufacturer prescribing information for both rivaroxaban and apixaban have been recently updated to allow the use of clarithromycin with these DOACs, despite its being a strong inhibitor of both P-gp and CYP3A4. 55, 56 Pharmacokinetic data indicate that J o u r n a l P r e -p r o o f changes in DOAC exposure due to clarithromycin are not likely to be clinically relevant. 73, 74 This is consistent with the findings of the Chang, et al study. 35 As most of the body of literature in this area consists of case reports, reporting bias potentially limits the scope and findings of our review. Specifically, underreporting of clinical events has almost certainly occurred, making it difficult or impossible to identify all clinically relevant interactions resulting in adverse events. Finally, other reports may have been published and not fallen into the search criteria for this systematic review. The safety and efficacy profile of a drug is often well tested and documented in clinical trials prior to reaching the market, however, its profile will continue to evolve and build after reaching the market. 75 The topic of drug-drug interactions may be best addressed by a collaboration between already established governmental databases such as the United States Food and Drug Administration Adverse Event Reporting System, the pharmaceutical industry, and academia. One collaborative product could be the creation of a mandatory de-identified prospective registry. This could eliminate reporting bias by identifying clinically relevant DDIs associated with adverse events in addition to the absence of clinically relevant DDIs. Efforts to make DOAC-specific laboratory monitoring tests more widely available would allow for more quantitative data in the setting of DDIs. Publicizing existing internal data and/or funding of prospective studies to investigate how DOAC-specific laboratory tests correlate to clinical adverse events would provide useful clinical guidance. As such collaboration between these agencies and academia is called upon and can promote efficient and expansive research towards drug-drug interactions. 77, 78 J o u r n a l P r e -p r o o f The strengths of this study lie in the reporting of DDIs that resulted in adverse events. Many prior DDI studies have been conducted in highly controlled environments with healthy subjects, or drew conclusions from in vitro data. [23] [24] [25] [26] [27] Although our study had to utilize lower quality of evidence case reports and observational data, it offers insight toward the DDIs that would most likely be encountered in a real-world clinical scenario. Furthermore, we choose to look at studies comparing specific DOACs with specific CYP3A4/P-gp modifiers. Studies where DOACs and modifiers are grouped together and assessed for risks may over estimate the risk for bleeding or thrombotic outcomes. Current literature evaluating the real-world risk of DOAC DDIs is limited to case reports and retrospective observational analyses. The most commonly reported interacting drugs to cause bleeding events were amiodarone and ritonavir via inhibition of P-gp and CYP3A4; culprit drugs for thrombotic events were phenytoin and carbamazepine (via combined strong induction of P-gp and CYP3A4), and phenobarbital (via strong induction of CYP3A4). Clinicians are encouraged to recognize multiple pathways of interaction and other clinical factors, assess the DDI literature appropriately, and identify and report potential interactions they encounter and to determine the likelihood of the validity of the interaction using validated tools such as the DIPS score (Table 2 ). J o u r n a l P r e -p r o o f reporting, resulting in very poor quality data in this clinically important area."  Assess the relative contributions of drug absorption, metabolic and elimination pathways and the clinical significance of each. Recognize drug-drug interactions may involve multiple pathways and multiple individual patient characteristics and consider the net effect on the patient.  When assessing drug-drug interaction literature, prioritize reports of human in vivo data in actual patients, particularly those reporting on adverse clinical outcomes as a result of a proposed drug-drug interaction.  Utilize labeled recommendations for avoiding drug combinations that have been demonstrated to cause bleeding or thrombotic events and utilize recommended dose reductions according to product labeling.  Apply the Drug Interaction Probability Scale (DIPS) to retrospectively assess the likelihood of a drug-drug interaction.  Report drug-drug interactions that result in adverse events as per institutional protocol and to the Food and Drug Administration Adverse Event Reporting System (FAERS) as appropriate, and write up a report of the proposed DDI and adverse event for publication and contribution to the literature as a case report. J o u r n a l P r e -p r o o f Systematic overview of warfarin and its drug and food interactions Drug and dietary interactions of warfarin and novel oral anticoagulants: an update Assessment of Outcomes of Treatment With Oral Anticoagulants in Patients With Atrial Fibrillation and Multiple Chronic Conditions: A Comparative Effectiveness Analysis Risks and benefits of direct oral anticoagulants versus warfarin in a real world setting: cohort study in primary care Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials Guidance for the practical management of the direct oral anticoagulants (DOACs) in VTE treatment Drug-drug interactions in an era of multiple anticoagulants: a focus on clinically relevant drug interactions New oral anticoagulants: their advantages and disadvantages compared with vitamin K antagonists in the prevention and treatment of patients with thromboembolic events Drug-drug interaction studies of cardiovascular drugs involving P-glycoprotein, an efflux transporter, on the pharmacokinetics of edoxaban, an oral factor Xa inhibitor Decrease in the oral bioavailability of dabigatran etexilate after co-medication with rifampicin Oral bioavailability of dabigatran etexilate (Pradaxa((R)) ) after co-medication with verapamil in healthy subjects Effect of Rifampin on the Pharmacokinetics of Apixaban, an Oral Direct Inhibitor of Factor Xa The effect of rifampin on the pharmacokinetics of edoxaban in healthy adults Co-administration of rivaroxaban with drugs that share its elimination pathways: pharmacokinetic effects in healthy subjects Effect of ketoconazole and diltiazem on the pharmacokinetics of apixaban, an oral direct factor Xa inhibitor Edoxaban drugdrug interactions with ketoconazole, erythromycin, and cyclosporine An openlabel study to estimate the effect of steady-state erythromycin on the pharmacokinetics, pharmacodynamics, and safety of a single dose of rivaroxaban in subjects with renal impairment and normal renal function European Heart Rhythm Association Practical Guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation: executive summary Measurement of Dabigatran Drug Levels to Manage Patients Taking Interacting Drugs: A Case Report Reduced Anticoagulant Effect of Dabigatran in a Patient Receiving Concomitant Phenytoin Correlation between trough plasma dabigatran concentrations and estimates of glomerular filtration rate based on creatinine and cystatin C Drug interaction as a predictor of direct oral anticoagulant drug levels in atrial fibrillation patients Characteristics and Outcomes in Patients with Venous Thromboembolism Taking Concomitant Anti-Platelet Agents and Anticoagulants in the AMPLIFY Trial Prediction of major and clinically relevant bleeding in patients with VTE treated with edoxaban or vitamin K antagonists Rivaroxaban in patients stabilized after a STsegment 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) Apixaban with antiplatelet therapy after acute coronary syndrome Predictors of gastrointestinal bleeding in older persons taking nonsteroidal anti-inflammatory drugs: Results from the FDA adverse events reporting system Bleeding risk of patients with acute venous thromboembolism taking nonsteroidal anti-inflammatory drugs or aspirin Selective Serotonin Reuptake Inhibitor Use and Risk of Gastrointestinal and Intracranial Bleeding The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses Proposal for a new tool to evaluate drug interaction cases Consensus recommendations for systematic evaluation of drug-drug interaction evidence for clinical decision support Increasing Understanding Regarding the Risk of Concomitant Use of Carbamazepine and Direct Oral Anticoagulants Association Between Use of Non-Vitamin K Oral Anticoagulants With and Without Concurrent Medications and Risk of Major Bleeding in Nonvalvular Atrial Fibrillation Association of Oral Anticoagulants and Verapamil or Diltiazem With Adverse Bleeding Events in Patients With Nonvalvular Atrial Fibrillation and Normal Kidney Function Postoperative Bleeding After Administration of a Single Dose of Rivaroxaban to a Patient Receiving Antiretroviral Therapy. Drug safety -case reports Gastrointestinal bleeding associated with rivaroxaban administration in a treated patient infected with human immunodeficiency virus Dabigatran toxicity in a 65 year old male who failed other anticoagulation methods Dabigatran and amiodarone interaction: A serious hemorrhage case report. Fundamental and Clinical Pharmacology Drug interactions of dabigatran: Report of one case. Basic and Clinical Pharmacology and Toxicology Bleeding associated with coadministration of rivaroxaban and clarithromycin Increased bleeding tendency from interaction between rivaroxaban and topical miconazole: Case report Spontaneous gross hematuria during dabigatran therapy for secondary stroke prevention Probable drug-drug interaction between dabigatran and quinidine resulting in thrombin time rebound despite multiple idarucizumab doses Development of Left Atrial Thrombus After Coadministration of Dabigatran Etexilate and Phenytoin Clinical challenges related to novel oral anticoagulants: drug-drug interactions and monitoring Venous thromboembolism due to suspected interaction between rivaroxaban and nevirapine Tocilizumab and mesenteric arterial thrombosis: Effect per se and/or interaction with anticoagulants. Fundamental and Clinical Pharmacology Management of Phenobarbital and Apixaban Interaction in Recurrent Cardioembolic Stroke Pulmonary embolism due to interaction between rivaroxaban and carbamazepine When not to choose apixaban: A case of apixaban failure Xarelto(R) [Package Insert Eliquis(R) [Package Insert Bristol-Myers Squibb Company Induction of CYP3A4 by efavirenz in primary human hepatocytes: comparison with rifampin and phenobarbital Induction of drug-metabolizing enzymes by phenobarbital in layered co-culture of a human liver cell line and endothelial cells In vitro predictability of drug-drug interaction likelihood of P-glycoprotein-mediated efflux of J o u r n a l P r e -p r o o f dabigatran etexilate based on [I]2/IC50 threshold Direct oral anticoagulant plasma levels' striking increase in severe COVID-19 respiratory syndrome patients treated with antiviral agents: The Cremona experience Renal Involvement and Early Prognosis in Patients with COVID-19 Pneumonia Laboratory Assessment of the Anticoagulant Activity of Direct Oral Anticoagulants: A Systematic Review Amiodarone, anticoagulation, and clinical events in patients with atrial fibrillation: insights from the ARISTOTLE trial Efficacy and Safety Outcomes of Direct Oral Anticoagulants and Amiodarone in Patients with Atrial Fibrillation Drug Interactions With Non-Vitamin K Oral Anticoagulants-Reply Drug Interactions With Non-Vitamin K Oral Anticoagulants Drug Interactions With Non-Vitamin K Oral Anticoagulants Drug Interactions With Non-Vitamin K Oral Anticoagulants Efficacy and Safety of Rivaroxaban Versus Warfarin in Patients Taking Nondihydropyridine Calcium Channel Blockers for Atrial Fibrillation (from the ROCKET AF Trial) Clinical Safety Outcomes in Patients With Nonvalvular Atrial Fibrillation on Rivaroxaban and Diltiazem Application of physiologically based pharmacokinetic modeling to the prediction of drug-drug and drug-disease interactions for rivaroxaban The Effects of Clarithromycin on the Pharmacokinetics of Apixaban in Healthy Volunteers: A Single-Sequence Crossover Study Postmarketing safety surveillance : where does signal detection using electronic healthcare records fit into the big picture? Detection of drug-drug interactions through data mining studies using clinical sources, scientific literature and social media Unifying drug safety and clinical databases Playing well with others! Initiating and sustaining successful collaborations between industry, academia and government 1. Patients on direct oral anticoagulants (DOACs) can encounter drug interactions 2. Drug interactions can alter the amount of DOAC in a patient's system. 3 Health providers should continue to be cautious with potential DOAC interactions We would like to thank the librarians at McMaster University, Health Sciences Library for helping us conduct our literature search. No funding was provided for conduction of this study. AL has no conflicts to disclose. ML has no conflicts to disclose. SRV serves as an editorial consultant for UpToDate®, which is a product of Wolters-Kluwer who also produces Lexi-Comp® drug interaction database. She is also a member of the board of directors for the Anticoagulation Forum. Doctor Crowther discloses Data Safety Monitoring board work for Bayer, Advisory Board work for Servier Canada , Asahi Kasei, and Precision Biologicals and preparation of educational materials and/or presentations for Pfizer, CSL Behring and Diagnostica Stago. Doctor Crowther holds individual stocks in Alnylam. Doctor Crowther has ongoing relationships with a number of for profit, and not for profit agencies and has worked on medicolegal cases involving anticoagulant therapy.J o u r n a l P r e -p r o o f