Direct-Acting Oral Anticoagulants in Critically Ill Patients

Direct-Acting Oral Anticoagulants in Critically Ill Patients

[ 1 Contemporary Reviews in Critical Care Medicine ] 56 2 57 3 58 4 59 5 60 Direct-Acting Oral Anticoagulants in Critically Ill Patients ...

NAN Sizes 0 Downloads 45 Views

[

1

Contemporary Reviews in Critical Care Medicine

]

56

2

57

3

58

4

59

5

60

Direct-Acting Oral Anticoagulants in Critically Ill Patients

6 7 8 9 10 11

Q22

Q1

61 62 63 64 65

Parth Rali, MD; Andrew Gangemi, MD; Aimee Moores, MD; Kerry Mohrien, PharmD; and Lisa Moores, MD

66

12

67

13

68

The direct-acting oral anticoagulants (DOACs) have been increasingly used over vitamin K

14

69

15

antagonists in recent years because they do not require monitoring and have an immediate

16

anticoagulation effect. In general, DOACs have exhibited a better safety profile and non-

71

17

inferiority for prophylaxis and treatment of venous thromboembolism (VTE) and stroke pre-

72

18

vention in patients with atrial fibrillation compared with vitamin K antagonists in the non-ICU

73

19

population; whether this finding holds true in patients who are critically ill remains unknown.

74

20

70

The current review addresses the role of DOACs in special ICU populations, use of these agents

75

21

for VTE prophylaxis, perioperative management of DOACs, drug monitoring, and potential drug

76

22

interactions of DOACs in critically ill patients. Adverse events and available reversal agents for

23

DOACs are also discussed.

24 25

KEY WORDS:

CHEST 2019;

77 78

-(-):---

79 Q5

antithrombotic therapy; critical care; drugs

80

26

81

27

82

Increasing utilization of direct-acting oral anticoagulants (DOACs) for therapeutic and prophylactic indications warrants familiarity of these agents among critical care physicians. Despite perceived concerns regarding increased hemorrhagic risk, particularly GI bleeding (GIB), DOACs have an overall better safety profile and are noninferior to vitamin K antagonists (VKAs) for prophylaxis and treatment of venous thromboembolism (VTE) and stroke prevention in patients with atrial fibrillation (AF). Few trials have examined the use of DOACs in patients in the ICU, and most of the phase III trials examining their efficacy in the aforementioned

28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44

conditions excluded patients in the ICU. Given the availability of multiple agents, the ease of use, and availability of specific reversal agents, it is important for critical care physicians to understand the optimal management of patients receiving a DOAC. Managing anticoagulation in the perioperative period remains a significant challenge as well. The current review addresses the role of DOACs in special ICU populations, use of these agents for VTE prophylaxis, perioperative management of DOACs, appropriate drug monitoring, and potential drug interaction of DOACs in critically ill

45

48 49 50 51 52 53 54 55

85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100

46 47

83 84

101 ABBREVIATIONS:

ACS = acute coronary syndromes; AF = atrial fibrillation; aPTT = activated partial thromboplastin time; DOAC = direct-acting oral anticoagulant; FDA = US Food and Drug Administration; GIB = GI bleeding; HIT = heparin-induced thrombocytopenia; LMWH = low-molecular-weight heparin; NVAF = nonvalvular atrial fibrillation; PT = prothrombin time; TAVR = transcatheter aortic valve replacement; UFH = unfractionated heparin; VKA = vitamin K antagonist Q2 Q3 AFFILIATIONS: From the Division of Thoracic Medicine and Surgery (Drs Rali and Gangemi) and Department of Pharmacy (Dr Mohrien), Temple University Hospital, Philadelphia, PA; Department of Medicine (Dr A. Moores), Madigan Army Medical Center, Joint Base Lewis-

McChord, WA; and the Department of Medicine (Dr L. Moores), The Uniformed Services University of the Health Sciences, Bethesda, MD. The opinions are those of the authors and are not official statements of the Uniformed Services University or the Department of the Army. CORRESPONDENCE TO: Lisa Moores, MD, F. Edward Hebert School of Medicine, The Uniformed Services University of the Health Sciences, Bethesda, MD 20814; e-mail: [email protected] Q4 Copyright Ó 2019 American College of Chest Physicians. Published by Elsevier Inc. All rights reserved. DOI: https://doi.org/10.1016/j.chest.2019.05.025

1

chestjournal.org

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

102 103 104 105 106 107 108 109 110

111

patients. Adverse events and available reversal agents for DOACs are also covered.

112 113 114

Pharmacokinetic and Dynamic Interactions of DOACs in Critically Ill Patients

115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165

Q6

In contrast to VKA therapy, DOACs result in predictable pharmacokinetic and dynamic interactions in nonacutely ill patients. Key physiologic changes that are common in patients in the ICU which may lead to unpredictable anticoagulant effects of DOACs include impaired gastric perfusion and motility, increased volume of distribution, impaired hepatic clearance due to inhibition of enzymatic metabolism, and changes in renal function.1 Both apixaban and rivaroxaban undergo clinically significant biotransformation within the hepatic system.2 In patients with moderate hepatic function (Child-Turcotte-Pugh class B), a single 10-mg dose of rivaroxaban resulted in a 1.27-fold increase in maximum drug concentration and a 2.27-fold increase in area under the plasma concentration-time curve; there was no change in the maximum drug concentration and only a 1.09-fold increase in the area under the plasma concentration-time curve of apixaban.3,4 Despite recent in vitro studies reporting altered pharmacokinetic variables of the DOACs (mainly rivaroxaban and apixaban) in patients with cirrhosis, multiple small retrospective analyses from real-world patients with cirrhosis have reported safe and effective use of DOACs.5-9 In contrast to apixaban and rivaroxaban, the remaining DOACs have limited (edoxaban and betrixaban) or no (dabigatran) hepatic metabolism, and pharmacokinetic properties are minimally changed in the setting of liver dysfunction.10-12 However, DOACs should be avoided in the setting of severe liver dysfunction (Child-TurcottePugh class C) and acute liver failure. Renal impairment (acute and chronic) significantly alters the pharmacokinetic properties of all available DOACs. All DOACs require dose adjustment in the setting of renal failure, particularly dabigatran and rivaroxaban.2,13-16 US Food and Drug Administration (FDA) labeling recommends no change in apixaban dosing in patients undergoing hemodialysis based on an initial single-dose study.17 However, literature examining the effect of multidose pharmacokinetic changes revealed significant accumulation following 8 days of apixaban 5 mg bid.18 A reduction to 2.5 mg bid resulted in drug exposure comparable to standard dosing in patients with preserved renal function. In patients with acute kidney injury, the anticoagulant

2 Contemporary Reviews in Critical Care Medicine

effect of DOACs reportedly lasts for 4 days following the last dose.19 There is no literature evaluating the impact of acute injury on renal clearance of DOACs; such use is therefore not recommended.2 Drug exposure of DOACs increases in a stepwise manner as renal function worsens.13-16 Dabigatran can be significantly removed from plasma following hemodialysis.13 Due to their comparatively large volume of distribution and/or high plasma protein-binding, rivaroxaban, apixaban, and edoxaban are minimally removed following hemodialysis. Table 1 highlights the standard DOAC dosing for various clinical indications and adjusted dosing for patients with renal and liver impairment. Extremes of weight are also of concern. Current FDA labeling offers no recommendations for dose adjustments for either an abnormally high or low BMI. The impact of obesity (BMI > 30 kg/m2) on treatment with DOACs is variable and conflicting. Peak apixaban concentrations and total drug exposure were found to be 31% and 23% lower, respectively, in healthy patients weighing > 120 kg with a BMI $ 30 kg/m2 compared with those in a reference body weight group.20 In a pharmacokinetic analysis of the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) trial, dabigatran plasma concentrations were found to be 21% lower when total body weight was > 100 kg.21 The impact of obesity on the pharmacokinetic properties of rivaroxaban is minimal, likely due to a reduced lipophilicity compared with apixaban and dabigatran. Based on a subgroup analysis of the published literature, Martin et al22 observed that weight < 120 kg or BMI < 40 kg/m2 does not significantly alter the plasma concentrations, distribution, or elimination half-life of DOACs.

166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203

Emerging Role of DOACs in Special Clinical Scenarios in the ICU

204

Given the safety profile, ease of use, and reduction in drug interactions of DOACs compared with VKAs, it is likely that increasing numbers of patients admitted to the critical care setting will be receiving therapy with one of these agents. In addition, there will be situations in which the initiation of a DOAC in the ICU is considered. Specific patient populations discussed here include those with new-onset valvular or nonvalvular AF (NVAF); patients undergoing valve replacement; patients with acute coronary syndromes (ACS), stroke, or heparin-induced thrombocytopenia (HIT); and postoperative cardiac surgery patients. We also briefly discuss VTE treatment and prevention with DOACs in the ICU setting. We do not discuss critically ill patients

206

[

-#- CHEST - 2019

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

205

]

207 208 209 210 211 212 213 214 215 216 217 218 219 220

221

TABLE 1

223 224 225 226

] Standard DOAC Dosing for Different Clinical Indications and Recommended Adjusted Dosing for Renal and Liver Impairment

222 Variable

Recommended dosing

227 228 229 230 231 232 233 234 235 236

277

Apixaban

Betrixaban

Dabigatran

Edoxaban

NVAF: 5 mg bid VTE treatment: 10 mg bid  7 days, then 5 mg bid Prevention of VTE: 2.5 mg bid (following 6 mo standard therapy) VTE prophylaxis post-hip/knee surgery: 2.5 mg bid

VTE prophylaxis: 160 mg  1, then 80 mg once daily

NVAF: 150 mg bid VTE treatment: initial LMWH/ heparin, then 150 mg BID Prevention of VTE: 150 mg bid (following 6 mo standard therapy) VTE prophylaxis post-hip/knee surgery: 110 mg  1 dose, then 220 mg once daily

NVAF: 60 mg once daily Initial LMWH/ heparin, then 60 mg once daily

237 238 239 240 241 242

Recommended renal dose adjustments

243 244 245 246 247 248 249 250 251 252 253

Recommended hepatic dose adjustments

254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271

If 2 of the following: SCr $ 1.5 mg/ dL, age $ 80 y, weight # 60 kg: reduce dose to 2.5 mg bid CrCl < 15 mL/min or ESRD: use with caution; consider dose reduction to 2.5 mg bid

CrCl 1530 mL/min: reduce dose to 80 mg  1, then 40 mg once daily CrCl < 15 mL/ min or ESRD: avoid use

CTP class A: no adjustments necessary CTP class B: use with caution CTP class C: avoid use

No available data

CrCl 15-30 mL/min: reduce dose to 75 mg bida CrCl < 15 mL/min or ESRD: avoid use

278

Rivaroxaban

279

NVAF: 20 mg once daily VTE treatment: 15 mg BID  21 d, then 20 mg once daily Prevention of VTE: 10 mg once daily (following 6 mo standard therapy) VTE prophylaxis post-hip/knee surgery: 10 mg once daily

CrCl 15-50 mL/ min: reduce dose to 30 mg once daily CrCl < 15 mL/min or ESRD: avoid use CrCl > 95 mL/ min: avoid use (reduced efficacy in atrial fibrillation)

CrCl 15-50 mL/ min: reduce dose to 15 mg once dailyb CrCl < 15 mL/ min or ESRD: avoid use

CTP class A: no adjustments necessary CTP class B: avoid use CTP class C: avoid use

CTP class A: no adjustments CTP class B: avoid use CTP class C: avoid use

280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305

CTP class A: no adjustments necessary CTP class B: use with caution CTP class C: avoid use

306 307 308 309 310 311

CrCl ¼ creatinine clearance; CTP ¼ Child-Turcotte-Pugh; DOAC ¼ direct-acting oral anticoagulant; ESRD ¼ end-stage renal disease; LMWH ¼ lowmolecular-weight heparin; NVAF ¼ nonvalvular atrial fibrillation; SCr ¼ serum creatinine; VTE ¼ venous thromboembolism. a US Food and Drug Administration (FDA)-labeled dose reduction specific for NVAF. FDA labeling recommends avoiding the use of dabigatran if CrCl < 30 mL/min when used for the treatment of VTE or postorthopedic surgery. b FDA-labeled dose reduction specific for NVAF. FDA labeling recommends avoiding use of rivaroxaban if CrCl < 30 mL/min when used for the treatment/ prophylaxis of VTE or postorthopedic surgery.

admitted with sepsis or acute kidney injury. In our opinion, DOACs should be avoided in these conditions given the unpredictable pharmacokinetic profile and potential need for invasive procedures. For patients who are admitted to the ICU with sepsis or acute kidney injury and who are already receiving DOACs, a switch to unfractionated heparin (UFH) or low-molecular-weight heparin (LMWH) is warranted.

272 273

Atrial Fibrillation

274

Valvular AF is commonly referred to as AF that occurs in the setting of mechanical heart valves or moderate to

275

276

severe mitral stenosis. VKAs remain the agent of choice in this setting given the exclusion of such patients in major DOAC trials and evidence suggesting excessive thrombotic and bleeding risks with DOACs in this population.23-27 The term NVAF has been used when AF occurs outside of the aforementioned clinical scenarios. DOACs are noninferior to VKAs in stroke prevention in patients with NVAF.28 In patients with new-onset AF > 48 h, in which cardioversion is urgently needed, DOACs can be given 4 h prior to transesophageal echocardiogram and cardioversion.29 Local institutional protocols should be followed in terms

3

chestjournal.org

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330

331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369

of anticoagulation and need for transesophageal echocardiogram before cardioversion. In patients with AF > 48 h, in which cardioversion can be delayed, DOACs can be initiated in the ICU and cardioversion performed electively as an outpatient. Valve Replacement and ACS

In patients undergoing transcatheter aortic valve placement (TAVR), the incidence of radiographically apparent postvalve thrombosis ranges from 7% to 40%.28 Larger size valves and lack of postprocedure antithrombotic treatment seem to be associated with valvular thrombosis.30 It is unknown at this stage which regimen is ideal in patients with TAVR (dual antiplatelet therapy þ VKA vs VKA only vs dual antiplatelet therapy þ DOACs). The European Society of Cardiology allows the use of DOACs beginning 3 months’ post-TAVR, whereas the American Heart Association 2017 guidelines do not recommend the same given the lack of dedicated studies in this population.28,31 For bioprosthetic mitral valve and mitral valvuloplasty, a VKA remains the agent of choice over DOACs.28 For patients already taking DOACs who present with ACS, DOACs should be stopped on admission. At the time of the discharge following ACS or elective percutaneous coronary intervention, DOACs should be resumed, at least in the immediate period.29 When added to single platelet therapy following ACS, DOACs were well tolerated but did not lead to a reduction in major adverse cardiovascular events. When added to dual platelet therapy following ACS, DOACs resulted in a reduction in major adverse cardiovascular events but were associated with an increased risk of major bleeding.32

370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385

Patients With Stroke

In patients with acute ischemic stroke, American Heart Association/American Stroke Association 2018 expert guidelines caution against the use of IV thrombolytic therapy in patients who are receiving DOAC therapies at the time of presentation.33 If coagulation study results are completely normal and the last dose of DOAC was > 48 h prior to presentation (presuming normal renal function), thrombolysis may then be considered. A recent meta-analysis suggested that IV thrombolysis, along with pharmacomechanical neurologic intervention, is safe in patients presenting with acute stroke who are taking DOACs at presentation.34,35 There is uncertainty regarding the timing and safety of DOACs

4 Contemporary Reviews in Critical Care Medicine

in acute ischemic stroke postthrombolysis. Fortunately, Q7 several clinical trials are ongoing that will address whether thrombolysis is safe in patients who are on DOACs at presentation and the role of DOAC initiation for secondary prevention following acute ischemic stroke.36-40

386 387 388 389 390 391 392 393

Heparin-Induced Thrombocytopenia

HIT is traditionally treated with parenteral thrombin inhibitors until platelet recovery, after which patients are transitioned to VKAs. There is growing interest in the role of DOACs in treating patients with HIT both as an alternative to VKAs or as a first-line agent.41-45 Current data are limited to small retrospective and in vitro studies only. Of note, rivaroxaban did not cause platelet activation or aggregation in the presence of HIT antibodies in one in vitro study.46 A large prospective trial evaluating the role of rivaroxaban in HIT was stopped due to poor enrollment.47 Currently, DOACs are not approved for use in HIT. There is an ongoing trial in the enrollment phase assessing the use of apixaban in HIT.48

394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410

Patients With VTE

411

DOACs are the anticoagulation agents of choice for the long-term treatment of most patients with acute VTE. The use of DOACs for acute treatment of VTE in the ICU setting, however, has not been adequately studied. Patients presenting with submassive or massive pulmonary embolism and patients who are at high risk of decompensation should receive UFH over DOACs. Traditionally, patients are started on anticoagulation following thrombolysis when the activated partial thromboplastin time (aPTT) is less than one and onehalf times the normal limit. In a small retrospective study, DOAC use compared with warfarin following catheter-directed thrombolysis seemed safe and was associated with a reduced length of stay (4.0 vs 6.1 days; P ¼ .002).49 DOACs may be considered in patients with pulmonary embolism when they are stable for transfer out of the ICU, after taking into consideration bleeding risk, comorbid organ dysfunction, and potential drug interactions.

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 434

The incidence of VTE in critically ill patients who do not receive prophylaxis varies but has been reported to be as high as 40%.50 None of the current DOACs has been studied for the prevention of VTE events in patients in the ICU. Four phase III trials have examined the DOACs Q8 in VTE prevention in hospitalized medical patients:

-#- CHEST - 2019

413 414

433

Role of DOACs in VTE Prevention

[

412

]

435 436 437 438 439 440

441

Apixaban Dosing to Optimize Protection from Thrombosis (ADOPT), Venous Thromboembolic Event (VTE) Prophylaxis in Medically Ill Patients (MAGELLAN), Acute Medically Ill VTE (Venous Thromboembolism) Prevention with Extended Duration Betrixaban (APEX), and Medically Ill Patient Assessment of Rivaroxaban Versus Placebo in Reducing Post-Discharge Venous Thrombo-Embolism Risk (MARINER).51-54 Some of the information gathered may be used to inform care or to answer research questions in critically ill patients. Study design, outcomes, and patient characteristics from these trials are summarized in eTables 1 and 2. Importantly, only one trial (MARINER)54 specified the number of patients who were cared for in the ICU (54%), further limiting the extrapolation of results. Assessing the admission diagnoses of the patients in the other trials suggests that a proportion may have been cared for in the ICU. Importantly, all of these trials were designed to investigate extended vs short-term duration of chemical prophylaxis and were not designed to determine the best agent for chemical prophylaxis, particularly in the critically ill.

442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495

Q9

Al Yami et al55-57 published the most recent of three meta-analyses that have combined ADOPT, MAGELLAN, and APEX (all were published prior to the publication of the MARINER trial).51-53 Among other findings, this study advanced our understanding by reporting a quantitative approach to risk-benefit analysis. The authors reported that the trials failed to establish clear efficacy/safety signals for any of the DOACs. Although, as a class, DOACs extend a benefit against select efficacy outcomes (symptomatic VTE, all VTE), this achievement comes at the expense of a marked increase in the risk of bleeding events. Interestingly, when the authors examined the risk/ benefit ratio, there was a class effect in favor of prophylaxis with DOACs for all VTE events compared with major bleeding events. It should be noted, however, that the authors assumed that all DOACs have the same risk profile, whereas emerging evidence suggests that apixaban likely has a superior safety profile. Overall, there are few data regarding the use of DOACs in VTE prevention in the ICU. Currently, betrixaban is the only FDA-approved agent for VTE prevention in acutely ill medical patients, based on a favorable safety profile.58 Clinical trials in this patient population are warranted, especially because the characteristics of these oral agents may increase adherence to thromboprophylaxis guidelines. Clinicians need further guidance not on whether to provide prophylaxis but on which agent and

the nuances that identify the highest risk patients most likely to benefit.

500 501

Management of anticoagulation in the perioperative period has become more challenging since the introduction of DOACs. Current guidance largely comprises expert opinion, based on known pharmacologic properties of the drugs, and information derived from retrospective subgroup analysis of existing trials.59 For minor procedures (ophthalmologic, dermatologic, and dental) in which minimal blood loss is expected, it is considered safe to continue anticoagulation throughout the perioperative period as long as procedures are performed at trough drug concentration (usually at 24 h since the last dose), as local hemostasis can be easily achieved.60-63 The recently published Bridge or Continue Coumadin for Device Surgery Randomized Controlled Trial 2 (BRUISE CONTROL 2) reported no increase in clinically significant pocket hematoma with uninterrupted DOAC therapy during cardiac device implantation.64 However, interruption is generally recommended for all other interventions, with specific timing for preprocedural cessation based on the risk of bleeding and the clearance rate of the DOAC in question.60-63 Procedures deemed to be low risk of bleeding are those in which adequate hemostasis can be achieved and bleeding would not lead to significant morbidity or require a transfusion. Moderate bleeding risk procedures include ones in which it may be difficult to obtain hemostasis or might require a transfusion or lead to reoperation. High bleeding risk procedures include those in which bleeding would place the patient’s life at risk or affect the outcome of the procedure.65 DOACs should typically be withheld for 48 h in higher risk procedures; this window can be extended in cases of renal impairment to account for changes in drug clearance.66 Time to restart anticoagulation should be based on individual risk factors for bleeding, as well as bleeding risk inherent to the procedure itself. A simple rule is to withhold the anticoagulant for an identical number of days postprocedure as preprocedure.67 It is advisable to withhold anticoagulation at least 24 to 48 h for procedures with a high risk of bleeding. In instances in which anticoagulation is to be withheld for a prolonged period of time, one can consider using a prophylactic dose of LMWH or a DOAC. Unlike warfarin, the rapid onset of action and short half-lives of the DOACs largely negate the need for bridging anticoagulation regardless

5 REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

497 498 499

Management of DOACs in the Perioperative Period

chestjournal.org

496

502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550

551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586

Q10

of the duration of cessation.60-63,68 Routine use of bridging with LMWH is not recommended prior to procedures, as observational studies suggest a higher risk of bleeding in patients who undergo bridging.67 Dubois et al62 suggest considering a bridge in patients with high thromboembolic risk (CHA2DS2-VASc [congestive heart failure, hypertension, age $ 75 years, diabetes mellitus, stroke/transient ischemic attack, vascular disease, age 65-74 years, sex category] score > 5 in AF, VTE within 3 months, or VTE with severe thrombophilia) and prolonged interruption, such as in neuraxial anesthesia. This approach is based on the observation that in most trials showing no increased VTE risk in perioperative DOAC cessation, the cessation period was generally short (# 3 days), and patients at high risk of VTE were underrepresented. In 2015, Schulman et al69 conducted a multicenter, prospective cohort trial in 541 patients that reported the safety of their perioperative protocol for dabigatran, based on creatinine clearance and surgical bleeding risk; their findings subsequently influenced expert recommendations. To date, there are no completed trials pertaining to the remaining DOACs. However, the Perioperative Anticoagulant Use for Surgery Evaluation (PAUSE) trial70 is an ongoing randomized cohort study with an enrollment goal of 3,300 patients; the primary aim of the study is to show the safety of a standardized but patient-specific protocol for the perioperative management of apixaban, rivaroxaban, and dabigatran.62,71 Table 2 summarizes current expert opinion regarding stopping and re-starting DOACs in the perioperative period.65,67

606

Monitoring of DOACs in the ICU Although DOACs do not require routine laboratory monitoring, there may be select clinical scenarios, including obesity, renal failure, and potential drug interactions, in which monitoring is useful. Measuring plasma concentrations may also benefit those admitted to the ICU, especially in the setting of emergent procedures, potential therapeutic failure, or reversal of life-threatening bleeding. In studies examining the relationship between plasma drug concentrations and clinical outcomes, there seems to be a consistent predictable response among all agents.72 Having the ability to tailor plasma drug concentrations in patients prone to bleeding may allow clinicians to maximize the benefits, while minimizing the risk, associated with DOACs. Although no expert consensus exists regarding the optimal way to measure the anticoagulant effect of DOACs, various coagulation tests are affected during treatment. Prothrombin time (PT) and/or aPTT are prolonged in the setting of most DOACs. The relation to plasma drug concentration, however, is unpredictable and not suitable for quantitative monitoring.72-74 Elevations in PT are sensitive to rivaroxaban, but this association varies widely according to the reagent used. Prolongation of the PT during apixaban therapy is unpredictable and insensitive and should not be used either qualitatively or quantitatively. The direct thrombin inhibitor dabigatran preferentially prolongs aPTT; however, the relation is nonlinear and insensitive.73 More accurate representations of the level of factor Xa anticoagulants may be obtained by using calibrated chromogenic anti-Xa assays. Although highperformance liquid chromatography/tandem mass

587 588 589

TABLE 2

592

High or moderate

593

All DOACs Hold for 2 d preprocedure

595 Low

All DOACs Hold for 1 d preprocedure

598

30-50 mL/min

603 604 605

615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641

646 647

Dabigatrana Hold for 4 d preprocedure All oral Xa inhibitors Hold for 2 d preprocedureb

648

Dabigatran Hold for 2 d preprocedure All oral Xa inhibitors Hold for 1 d preprocedure

652

649 650 651 653

599

602

613 614

645

$ 50 mL/min

594

601

611 612

644

CrCl

591

600

610

643

] Perioperative Management of DOACs65,67

Procedure-Related Bleeding Risk

597

609

642

590

596

607 608

654 See Table 1 legend for expansion of abbreviations. a Spanish Guidelines (Vivas et al65) recommend a three-tiered approach to withholding dabigatran. In moderate or high bleeding risk procedures, guidelines recommend holding 3 days preprocedure for CrCl $ 80 mL/min, 4 days for CrCl 50 to 79 mL/min, and 5 days for CrCl < 50 mL/min. For low bleeding risk procedures, guidelines recommend holding for 2 days preprocedure for CrCl $ 80 mL/min, 3 days for CrCl 50 to 79 mL/min, and 4 days for CrCl < 50 mL/ min. b Spanish Guidelines (Vivas et al65) recommend a more conservative approach with the oral Xa inhibitors. For medium or high bleeding risk procedures, hold for 3 days preprocedure for CrCl > 30 mL/min and 4 days for CrCl 15 to 30 mL/min. For low bleeding risk procedures, hold for 2 days preprocedure for CrCl > 30 mL/min and 3 days for CrCl 15 to 30 mL/min.

6 Contemporary Reviews in Critical Care Medicine

[

-#- CHEST - 2019

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

]

655 656 657 658 659 660

661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683

spectroscopy is considered the most accurate method, chromogenic anti-Xa assays have shown good correlation.74,75 The availability of specific anti-Xa assays has increased in recent years, but it is still limited and can be associated with impractical turnaround times, particularly in patients admitted to the ICU. Accurate approximations of dabigatran can be achieved by using a dilute thrombin time or ecarin clotting time. Both laboratory tests have shown strong linear relationships at concentrations > 50 ng/mL, making them useful monitoring tools for dabigatran-treated patients.25,74,75 Table 3 summarizes the effect of DOACs on various nonspecific and drug-specific coagulation parameters. Although DOACs have fewer drug-to-drug interactions, in general, compared with VKAs, as their use becomes more prevalent in the ICU setting, clinicians should familiarize themselves with potential interactions with some of the most commonly used drugs in this situation (Table 4).29

684 685 686 687 688 689 690 691 692 693 694 695 696 697

716

Evolving real-world data are adding to our knowledge obtained from the pivotal phase III trials of DOACs. A large registry involving > 9,900 patients with AF reported similar major bleeding events, fewer intracranial hemorrhage events, and more GIB with DOACs compared with VKAs. Mortality, all-cause hospitalization, and recurrent major bleeding rates at 30 days did not differ compared with warfarin.89 The findings of higher GIB (mainly dabigatran) and fewer intracranial hemorrhage events with DOACs compared with VKAs are consistent across other multicenter retrospective studies.90 A more recent study of 146,871 Medicare patients, when stratified according to comorbidity burden, showed improved overall safety of dabigatran and rivaroxaban compared with VKAs. Rivaroxaban loses the advantage of reduced bleeding (particularly GIB) when comorbid scores are high.91 A summary of the major bleeding rates among main trials is provided in e-Table 3.24,51,53,77-88,92-108 It should be noted, however, that many of these trials excluded patients with preexisting renal or hepatic impairment, recent major bleeds, and thrombocytopenia, as well as patients receiving dual antiplatelet therapy or high-dose aspirin.

Adverse Side Effects of DOACs Major bleeding is the most concerning adverse effect of DOACs. Major bleeding is defined by the International Society on Thrombosis and Haemostasis as bleeding that is fatal, involves a critical organ (ie, intraspinal, intracerebral, intraocular, retroperitoneal, intramuscular), causes a 2 g/dL drop in hemoglobin level, requires transfusion of $ 2 units of blood, or leads to therapy cessation.76 Orthopedic literature also includes the criterion of bleeding that requires reoperation.77-88

717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742

Additional adverse effects of DOACs include transient transaminitis (1%-4%), GI intolerance (nausea, vomiting, dyspepsia, diarrhea, and constipation [up to 17%]), peripheral edema (2%-8% with apixaban), gross hematuria (12%-15%, particularly with edoxaban), and headaches and/or dizziness (2%-11%). Pyrexia has been reported in 6% to 12% of patients.52,77-88,92-96,100,101,103,105,106,109,110 Thrombocytopenia was only rarely reported with apixaban (< 2%) and would be low in the differential for

743 744 745 746 747 748 749 750 751 752

698

753

699

754

700

TABLE 3

] Effect of DOACs on Drug-Specific and Nonspecific Coagulation Parameters

701

Parameter

702

Approximate on-therapy peak concentrations

703

Dilute thrombin time/ecarin clotting assay, ng/mL

704

Chromogenic anti-factor-Xa, ng/mL

705 706 707 708 709

755

Apixaban

Dabigatran

Edoxaban

Rivaroxaban

NA

50-400

NA

NA

60-300

NA

100-300

180-350

756 757 Q17

759 760

Approximate on-therapy trough concentrations Dilute thrombin time/ecarin clotting assay, ng/mL Chromogenic anti-factor-Xa, ng/mL

758

NA

30-225

NA

NA

25-200

NA

10-40

10-140

761 762 763

Effect on routine coagulation testsa

764

710

Activated partial thromboplastin time

þ

þþþþ

þ

þ

765

711

Prothrombin time

þ

þ

þþ

þþþ

766

712

Thrombin time

NA

þþþþþ

NA

NA

767

713 714 715

768 See Table 1 legend for expansion of abbreviation. a The effect of DOACs on routine coagulation tests can be significantly influenced by variability in laboratory assays and reagents. Anti-Xa values can vary widely based on agent, indication, and dosing regimen. Values presented in this table are approximate values based on published trial data.

7

chestjournal.org

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

769 770

771 772

TABLE 4

] Major Drug Interaction Between DOACs and Commonly Used Drugs in the ICU Setting

773

Medication (Mechanism of Interaction)

774

Antiepileptic agents

775 776 777 778 779 780 781 782 783 784

Phenytoin (strong CYP450 3A4 inducer/ P-glycoprotein inducer)

791 792 793

Dabigatran

Avoid use; significant reduction in AUC predicted

800

Ketoconazole, itraconazole, and voriconazole (strong CYP450 3A4 inhibitor/ P-glycoprotein inhibitor)

Use with caution; theoretical increase in concentrations; consider alternative agent if additional interactions and/or risk factors presenta

No available data

Avoid use; 100% increase in AUC

Avoid use; 150% increase in AUC

Posaconazole (strong CYP450 3A4 inhibitor/ P-glycoprotein inhibitor)

Avoid use; significant increase in AUC predicted

Avoid use; significant increase in AUC predicted

802 804 805 806 807 808 809

Cardiovascular agent

815 816 817 818 819 820 821

Use with caution; 42% increase in AUC; consider alternative agent if additional interactions and/or risk factors presenta

Consider dose reduction or alternative agent; 95% increase in AUC; consider alternative agent if additional interactions and/or risk factors presenta

Avoid use; 160% increase in AUC

Use with caution; theoretical increase in concentrations; consider alternative agent if additional interactions and/or risk factors presenta

Avoid use; significant increase in AUC predicted

Avoid use; 35% reduction in AUC; increase in active metabolite

Avoid use; 50% reduction in AUC

837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858

Avoid use; 54% reduction in AUC

Avoid use; 66% reduction in AUC

860 861 862 863

Antiretroviral agent

814

812

No available data

859

Rifampin (strong CYP 3A4 inducer/ P-glycoprotein inducer)

813

811

833

Anti-infective agents

Ritonavir (strong CYP450 3A4 inhibitor/Pglycoprotein inhibitor)

810

831 832

Fluconazole (moderate CYP450 3A4 inhibitor)

801 803

Avoid use; significant reduction in AUC predicted

835

796

799

Avoid use; significant reduction in AUC predicted

830

836

795

798

Rivaroxaban

Antifungal agents

794

797

Edoxaban

834

787

790

Apixaban

Avoid use; significant reduction in AUC predicted

786

789

827 828 829

785

788

826

Amiodarone (P-glycoprotein inhibitor)

Avoid use; significant increase in AUC predicted

Avoid use; significant increase in AUC predicted

Avoid use; significant increase in AUC predicted

864

Avoid use; 153% increase in AUC

865 866 867 868 869

Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

Use with caution; 58% increase in AUC; consider alternative agent if additional interactions and/or risk factors presenta

Use with caution; 40% increase in AUC; consider alternative agent if additional interactions and/or risk factors presenta

870

Use with caution; 1.36-fold increase in AUC; consider alternative agent if additional interactions and/or risk factors presenta

822

871 872 873 874 875 876 877

(Continued)

823

878

824

879

825

880

8 Contemporary Reviews in Critical Care Medicine

[

-#- CHEST - 2019

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

]

881 882 883 884 885 886 887 888 889

TABLE 4

936

] (Continued)

Medication (Mechanism of Interaction) Diltiazem (mild to moderate CYP450 3A4 inhibitor/Pglycoprotein inhibitor)

890

937 Apixaban

Dabigatran

Use with caution; 40% increase in AUC; consider alternative agent if additional interactions and/or risk factors presenta

Use with caution; theoretical increase in concentrations; consider alternative agent if additional interactions and/or risk factors presenta

Edoxaban No available data

891 892 893 894 895 896 897

Verapamil (moderate CYP450 3A4 inhibitor/Pglycoprotein inhibitor)

Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914

921

Avoid use; significant reduction in AUC predicted

948

Avoid use; significant reduction in AUC predicted

Avoid use; significant reduction in AUC predicted

Avoid use; significant reduction in AUC predicted

958

Avoid use; significant increase in AUC predicted

Consider dose reduction or alternative agent; 73% increase in AUC

Tacrolimus (mild CYP450 3A4 inhibitor/Pglycoprotein inhibitor)

Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

Avoid use; significant increase in AUC predicted

Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

926

931 932 933 934 935

949 950 951 952 953 954 955 956

959 960 961 963

925

930

947

Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

962 Use with caution; increase in AUC by 120%; consider alternative agents if additional interactions and/or risk factors presenta

Cyclosporine (moderate CYP450 3A4 inhibitor/Pglycoprotein inhibitor)

924

929

945 946

Immunosuppressant agents

923

928

943 944

957

Dexamethasone (strong CYP450 3A4 inducer/Pglycoprotein inducer)

922

927

941 942

Consider dose reduction or alternative agent; 40% to 53% increase in AUC; consider alternative agent if additional interactions and/or risk factors presenta

917

920

939 940

Consider dose reduction or alternative agent; variable increase in AUC (12% to 180%); consider alternative agent if additional interactions and/or risk factors presenta

916

919

Rivaroxaban Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

Systemic steroid

915

918

938

964

Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

965 966 967 968 969 970 971 972 973

Use with caution; theoretical increase in concentration; consider alternative agent if additional interactions and/or risk factors presenta

974 975 976 977 978 979 980 981

AUC ¼ area under the plasma concentration-time curve; CYP ¼ cytochrome P450. See Table 1 legend for expansion of other abbreviation. a Additional factors for consideration include renal/liver impairment, age $ 75 years, weight # 60 kg, or concomitant medications that increase bleeding risk such as antiplatelet agents and/or nonsteroidal antiinflammatory agents.

drug-induced cytopenias.84,85 In most trials except those using dabigatran, major adverse cardiovascular events occur in < 2% of cases.99,102 QTc prolongation has not been shown with rivaroxaban,111 apixaban,112 or edoxaban.113

986

Initial concerns with the use of DOACs stemmed from the inability to reverse their anticoagulant effects. Newer specific and nonspecific agents, however, are now available to mitigate this concern. Reversal agents can be

9 REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

983 984 985

Reversal Agents for DOACs

chestjournal.org

982

987 988 989 990

991

992

993

994

995

996

997

998

999

1000

1001

1002

1003

1004

1005

1006

1007

1008

1009

1010

1011

1012

1013

1014

1015

1016

1017

1018

1019

1020

1021

1022

1023

1024

1025

1026

1027

1028

1029

1030

1031

1032

1033

1034

1035

1036

1037

1038

1039

1040

1041

1042

1043

1044

1045

] DOACs Reversal Agent Properties

Q18

Agent

Mechanism of Action

4-Factor PCCs (KCentra)

Replacement of coagulation factors II, VII, IX, and X, small amounts of proteins C and S

All

DOAC Target

25-50 U/kg Additional doses can be given as needed, 25 U/kg

Dosing

79% effective in controlling bleeding based on trials

7%, elevated risk when receiving other reversal agents within 4 h, undergoing surgery, or receiving massive transfusion protocol

Preferred over fresh frozen plasma according to CHEST and ASH guidelines Contraindicated with concomitant DIC and abnormal fibrinolysis Risk of contraction of HIV, viral hepatitis, and parvovirus B19 (all PCCs)

3-Factor PCC (Profilnine, Bebulin)

Replacement of coagulation factors II, IX, and X

All

25-50 U/kg

Reverses endogenous thrombin potentially within 3 h, PT normalization after 6 h

None reported

Data limited to phase I studies

Activated PCC (FEIBA)

Replacement of activated factor VII also inactivated II, IX, and X

All

25-50 U/kg Additional doses can be given as needed, 25 U/kg

Overall, 86% control of bleeding Control of ICH, 71% according to 2 small retrospective studies

4 per 100,000 (manufacturer safety data). 0%-12% (retrospective studies)

Contraindicated to use with DIC, acute thrombosis, or embolism (including myocardial infarction), and with history of anaphylactic or severe hypersensitivity reactions to FEIBA or any of its components

Idarucizumab (Praxbind)

Monoclonal antibody

Dabigatran

5 g dose (as two 2.5 g infusions 15 min apart)

68% control of bleeding 13.5% 30-day mortality 18% 90-day mortality

5% (0.4% thrombosisspecific 30-day mortality)

Rebound of dabigatran levels can occur

Andexanet alfa (Andexxa)

Inactivated factor Xa (decoy protein)

Rivaroxaban Apixaban

Low dose (for rivaroxaban # 10 mg, apixaban # 5 mg, or last confirmed dose > 8 h): 400-mg bolus (30 mg/min) followed by 4 mg/min (480 mg total) High dose (for rivaroxaban > 10 mg or apixaban > 5 mg confirmed within 8 h or unknown dose): 800mg bolus (30 mg/min) followed by 8 mg/min (960 mg total)

79% efficacy at 12 h

18% (10 deaths from CVD)

Data based on ANNEXA4124

[ -#- CHEST - 2019

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

10 Contemporary Reviews in Critical Care Medicine

TABLE 5

Efficacy

Thrombosis Risk

Comments

(Continued)

] 1047

1046

1049

1048

1051

1050

1052

1053

1055

1054

1057

1056

1059

1058

1061

1060

1062

1063

1065

1064

1067

1066

1069

1068

1071

1070

1072

1073

1075

1074

1077

1076

1079

1078

1081

1080

1082

1083

1085

1084

1087

1086

1089

1088

1091

1090

1092

1093

1095

1094

1097

1096

1098

1100

1099

1103 Comments

1104 1105 1106 1107 1108 1109 1110 1111

Thrombosis Risk

1113 1114 1115 1116 1117

NA

1112

1118 1120 1121 Efficacy

1122 1123 1124 1125 1126 1127

66% survival according to small case series

1119

1128 1130 1131 Dosing

1132 1133 1134 1135 1136 1137

Intermittent effective for rapid removal Continuous replacement methods may prevent rebound

1129

1143 1144 1145 1146

1149 1150 1151 1152 1153 1154 1155

TABLE 5

1148

] (Continued)

1147

Dabigatran

1142

Extracorporeal drug removal

1141

Hemodialysis

1140

Agent

1139

Mechanism of Action

DOAC Target

1138

ANNEXA-4 ¼ Andexanet Alfa, a Novel Antidote to the Anticoagulation Effects of Factor Xa Inhibitors; ASH ¼ American Society of Hematology; CHEST ¼ American College of Chest Physicians; CVD ¼ cardiovascular disease; DIC ¼ disseminated intravascular coagulation; FEIBA ¼ Factor Eight Inhibitor Bypassing Activity; ICH ¼ intracranial hemorrhage; PCC ¼ prothrombin protein complex; PT ¼ prothrombin time; PTT ¼ partial thromboplastin time. See Table 1 legend for expansion of other abbreviation.

Q19

1102

Variable normalization of PT and PTT seen

1101

considered when conservative measures have failed and the site of bleeding could be fatal, when there is organthreatening bleeding (eg, hemarthrosis, intraocular), or when there is an indication for urgent surgery. Nonmajor bleeds are less likely to require such aggressive measures in light of the short half-life of the DOACs, especially considering the high cost of reversal agents.114,115 Table 5 outlines the available agents, as well as their dosing, efficacy, and side effect profiles.19,114-126 We found no literature regarding the efficacy or safety of tranexamic acid, aminocaproic acid, or desmopressin in DOAC-related bleeding. Ciraparantag (PER977) is a synthetic molecule that binds heparinoids and DOACs and showed promise in phase I studies as a broad-spectrum reversal agent.126 A phase II clinical trial of ciraparantag is currently enrolling as of this publication.127

1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176

Conclusions It will be increasingly common for patients in the ICU to be receiving DOAC therapy or have an indication to begin such therapy during their hospital stay. Intensivists should be familiar with the properties, dosing adjustments, management of life-threatening bleeding, and common drug interactions related to DOACs. Following is a summary of the take-home points:  VKAs remain the agent of choice for valvular AF. In NVAF, DOACs are noninferior to VKAs in stroke prevention.  DOACs are not currently approved for the treatment of HIT.  Current guidelines caution against the use of IV thrombolysis in patients presenting with acute ischemic stroke while taking DOACs. Several studies are underway to answer this specific question.  Limited data exist regarding the use of DOACs postthrombolysis in patients with massive or submassive pulmonary embolism.  DOACs are not the anticoagulant agents of choice for the treatment of acute VTE in the ICU setting. Given the potential for hemodynamic decompensation and the need for thrombolysis, UFH should be used in the majority of patients in the ICU.  There are few data regarding the use of DOACs in VTE prevention in the ICU. Currently, betrixaban is the only FDA-approved agent for VTE prevention in acutely ill medical patients.  Minor procedures (eg, ophthalmologic, dermatologic, dental) should be performed at trough drug

11

chestjournal.org

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210

1211 1212 1213 1214 1215



1216 1217



1218 1219 1220 1221 1222



1223 1224 1225 1226



1227 1228 1229 1230 1231 1232 1233 1234 1235 1236

Acknowledgments Q21

Q11

1238 1239

1. Smith BS, Yogaratnam D, Levasseur-Franklin KE, Forni A, Fong J. Introduction to drug pharmacokinetics in the critically ill patient. Chest. 2012;141(5):1327-1336.

1241 1242 1243 1244 1245 1246 1247 1248 1249

1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265

Financial/nonfinancial disclosures: None declared.

References

1240

1251

Author contributions: All authors contributed equally to the drafting of the manuscript.

Additional information: The e-Tables can be found in the Supplemental Materials section of the online article.

1237

1250

concentration (usually 24 h since last dose). For highrisk procedures, DOACs should be held, with the duration depending on agent, renal clearance, and surgical procedure. Bridging with LMWH/UFH is usually not recommended in the periprocedural period. Dose adjustment of DOACs is recommended in patients with renal and liver failure. Apixaban has the most evidence for safety in both acute and chronic kidney disease; rivaroxaban and dabigatran should be avoided. Compared with VKAs, DOACs have an overall lower bleeding rate, although the rates of GIB vary according to agent. Development of specific reversal agents such as idarucizumab (for dabigatran) and andexanet alfa (for the factor Xa inhibitors apixaban, rivaroxaban, edoxaban, and betrixaban) have lessened clinician concerns for major bleeding with DOACs.

Q12

thrombin inhibitor dabigatran etexilate in patients with hepatic impairment. Clin Pharmacokinet. 2013;52(4):243-254.

1266

11. Mendell J, Johnson L, Chen S. An open-label, phase 1 study to evaluate the effects of hepatic impairment on edoxaban pharmacokinetics and pharmacodynamics. J Clin Pharmacol. 2015;55(12):1395-1405.

1268

1267 1269 1270

12. Lee K, Cham S, Lam S. Betrixaban: a novel factor Xa inhibitor for the prevention of venous thromboembolism in acutely ill medical patients. Cardiol Rev. 2018;26(6):331-338. 13. Stangier J, Rathgen K, Stähle H, Mazur D. Influence of renal impairment on the pharmacokinetics and pharmacodynamics of oral dabigatran etexilate: an open-label, parallel-group, singlecentre study. Clin Pharmacokinet. 2010;49(4):259-268.

1271 1272 1273 1274 1275

14. Chang M, Yu Z, Shenker A, et al. Effect of renal impairment on the pharmacokinetics, pharmacodynamics, and safety of apixaban. J Clin Pharmacol. 2016;56(5):637-645. 15. Jönsson S, Simonsson USH, Miller R, Karlsson MO. Population pharmacokinetics of edoxaban and its main metabolite in a dedicated renal impairment study. J Clin Pharmacol. 2015;55(11): 1268-1279. 16. Kubitza D, Becka M, Mueck W, et al. Effects of renal impairment on the pharmacokinetics, pharmacodynamics and safety of rivaroxaban, an oral, direct factor xa inhibitor. Br J Clin Pharmacol. 2010;70(5):703-712. 17. Wang X, Tirucherai G, Marbury TC, et al. Pharmacokinetics, pharmacodynamics, and safety of apixaban in subjects with endstage renal disease on hemodialysis. J Clin Pharmacol. 2016;56(5): 628-636. 18. Mavrakanas TA, Samer CF, Nessim SJ, Frisch G, Lipman ML. Apixaban pharmacokinetics at steady state in hemodialysis patients. J Am Soc Nephrol. 2017;28(7):2241-2248.

1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291

19. Singh T, Maw TT, Henry BL, et al. Extracorporeal therapy for dabigatran removal in the treatment of acute bleeding: a single center experience. Clin J Am Soc Nephrol. 2013;8(9):1533-1539.

1292 1293

20. Upreti VV, Wang J, Barrett YC, et al. Effect of extremes of body weight on the pharmacokinetics, pharmacodynamics, safety and tolerability of apixaban in healthy subjects. Br J Clin Pharmacol. 2013;76(6):908-916.

1294 1295 1296 1297

2. Steffel J, Verhamme P, Potpara TS, et al. The 2018 European Heart Rhythm Association practical guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation. Eur Heart J. 2018;39(16):1330-1393.

21. Reilly PA, Lehr T, Haertter S, et al. 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(4):321-328.

3. Kubitza D, Roth A, Becka M, et al. Effect of hepatic impairment on the pharmacokinetics and pharmacodynamics of a single dose of rivaroxaban, an oral, direct factor xa inhibitor. Br J Clin Pharmacol. 2013;76(1):89-98.

22. Martin K, Beyer-Westendorf J, Davidson BL, Huisman MV, Sandset PM, Moll S. Use of the direct oral anticoagulants in obese patients: guidance from the SSC of the ISTH. J Thromb Haemost. 2016;14(6):1308-1313.

4. Frost CEYZ, Wang J, Li C, Zeigler C, et al. Single-dose safety and pharmacokinetics of apixaban in subjects with mild or moderate hepatic impairment. Clin Pharmacol Ther. 2009;84.

23. Ezekowitz MD, Nagarakanti R, Noack H, et al. Comparison of dabigatran and warfarin in patients with atrial fibrillation and valvular heart disease: the RE-LY trial (randomized evaluation of long-term anticoagulant therapy). Circulation. 2016;134(8):589598.

1304

24. Diener H, Eikelboom J, Connolly SJ, et al. Apixaban versus aspirin in patients with atrial fibrillation and previous stroke or transient ischaemic attack: a predefined subgroup analysis from AVERROES, a randomised trial. Lancet Neurol. 2012;11(3):225-231.

1308

5. Hum J, Shatzel JJ, Jou JH, Deloughery TG. The efficacy and safety of direct oral anticoagulants vs traditional anticoagulants in cirrhosis. Eur J Haematol. 2017;98(4):393-397. 6. De Gottardi A, Trebicka J, Klinger C, et al. Antithrombotic treatment with direct-acting oral anticoagulants in patients with splanchnic vein thrombosis and cirrhosis. Liver Int. 2017;37(5): 694-699. 7. Hoolwerf EW, Kraaijpoel N, Büller HR, van Es N. Direct oral anticoagulants in patients with liver cirrhosis: a systematic review. Thromb Res. 2018;170:102-108.

25. Breithardt G, Baumgartner H, Berkowitz SD, et al. Clinical characteristics and outcomes with rivaroxaban vs. warfarin in patients with non-valvular atrial fibrillation but underlying native mitral and aortic valve disease participating in the ROCKET AF trial. Eur Heart J. 2014;35(47):3377-3385.

8. Potze W, Adelmeijer J, Lisman T. Decreased in vitro anticoagulant potency of rivaroxaban and apixaban in plasma from patients with cirrhosis. Hepatology. 2015;61(4):1435-1436.

26. De Caterina R, Renda G, Carnicelli AP, et al. Valvular heart disease patients on edoxaban or warfarin in the ENGAGE AF-TIMI 48 trial. J Am Coll Cardiol. 2017;69(11):1372-1382.

9. Potze W, Arshad F, Adelmeijer J, et al. Differential in vitro inhibition of thrombin generation by anticoagulant drugs in plasma from patients with cirrhosis. PLoS One. 2014;9(2):e88390. 10. Graff J, Harder S. Anticoagulant therapy with the oral direct factor xa inhibitors rivaroxaban, apixaban and edoxaban and the

12 Contemporary Reviews in Critical Care Medicine

27. Avezum A, Lopes RD, Schulte PJ, et al. Apixaban in comparison with warfarin in patients with atrial fibrillation and valvular heart disease: findings from the Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial. Circulation. 2015;132(8):624-632.

[

-#- CHEST - 2019

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

]

1298 1299 1300 1301 1302 1303 1305 1306 1307 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320

1321

28. Nishimura RA, Otto CM, Bonow RO, et al. 2017 AHA/ACC focused update of the 2014 AHA/ACC guideline for the management of patients with valvular heart disease: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines. J Am Coll Cardiol. 2017;70(2): 252-289.

1322 1323 1324 1325

29. Heidbuchel H, Verhamme P, Alings M, et al. Updated European Heart Rhythm Association practical guide on the use of nonvitamin-K antagonist anticoagulants in patients with non-valvular atrial fibrillation: executive summary. Eur Heart J. 2017;38(27): 2137-2149.

1326 1327 1328 1329

30. Hansson NC, Grove EL, Andersen HR, et al. Transcatheter aortic valve thrombosis: incidence, predisposing factors, and clinical implications. J Am Coll Cardiol. 2016;68(19):2059-2069.

1330 1331

31. Baumgartner H, Falk V, Bax JJ, et al. 2017 ESC/EACTS guidelines for the management of valvular heart disease. Rev Esp Cardiol (Engl Ed). 2018;71(2):110.

1332 1333 1334

32. Khan SU, Arshad A, Riaz IB, Talluri S, Nasir F, Kaluski E. Metaanalysis of the safety and efficacy of the oral anticoagulant agents (apixaban, rivaroxaban, dabigatran) in patients with acute coronary syndrome. Am J Cardiol. 2018;121(3):301-307.

1335 1336 1337

33. Powers WJ, Rabinstein AA, Ackerson T, et al. 2018 Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2018;49(3):e110.

1338 1339 1340

34. Liu M, Zheng Y, Li G. Safety of recanalization therapy in patients with acute ischemic stroke under anticoagulation: a systematic review and meta-analysis. J Stroke Cerebrovasc Dis. 2018;27(9): 2296-2305.

1341 1342 1343

35. Shahjouei S, Tsivgoulis G, Goyal N, Alexandrov AV, Zand R. Abstract 4: Safety of intravenous thrombolysis among patients taking direct oral anticoagulants: A systematic review and metaanalysis. Stroke. 2018;49(suppl_1):A4.

1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375

Q13

36. National Institutes of Health Clinical Center. Addressing realworld anticoagulant management issues in stroke (ARAMIS). NCT02478177. ClinicalTrials.gov. Bethesda, MD: National Institutes of Health; 2015. https://clinicaltrials.gov/ct2/show/ NCT02478177. Updated May 23, 2019. 37. National Institutes of Health Clinical Center. Novel oral anticoagulants in stroke patients (NOACISP). NCT02353585. ClinicalTrials.gov. Bethesda, MD: National Institutes of Health; 2015. https://clinicaltrials.gov/ct2/show/NCT02353585. Updated February 28, 2019. 38. National Institutes of Health Clinical Center. Rivaroxaban acute stroke safety study (RASS). NCT02279940. ClinicalTrials.gov. Bethesda, MD: National Institutes of Health; 2014. https:// clinicaltrials.gov/ct2/show/NCT02279940. Updated April 6, 2016. 39. National Institutes of Health Clinical Center. Rivaroxaban versus warfarin in acute ischemic stroke with atrial fibrillation (TripleAXEL). NCT02042534. ClinicalTrials.gov. Bethesda, MD: National Institutes of Health; 2014. https://clinicaltrials.gov/ct2/ show/NCT02042534. Updated February 8, 2017. 40. National Institutes of Health Clinical Center. Apixaban for early prevention of recurrent embolic stroke and hemorrhagic transformation (AREST). NCT02283294. ClinicalTrials.gov. Bethesda, MD: National Institutes of Health; 2014. https:// clinicaltrials.gov/ct2/show/NCT02283294. Updated March 19, 2019. 41. Sharifi M, Bay C, Vajo Z, Freeman W, Sharifi M, Schwartz F. New oral anticoagulants in the treatment of heparin-induced thrombocytopenia. Thromb Res. 2015;135(4):607-609. 42. Shatzel JJ, Crapster-Pregont M, Deloughery TG. Non-vitamin K antagonist oral anticoagulants for heparin-induced thrombocytopenia. A systematic review of 54 reported cases. Thromb Haemost. 2016;116(2):397-400. 43. Linkins LA, Warkentin TE, Pai M, et al. Rivaroxaban for treatment of suspected or confirmed heparin-induced thrombocytopenia study. J Thromb Haemost. 2016;14(6):1206-1210. 44. Davis KA, Davis DO. Direct acting oral anticoagulants for the treatment of suspected heparin-induced thrombocytopenia. Eur J Haematol. 2017;99(4):332-335.

1376

45. Warkentin TE, Pai M, Linkins L. Direct oral anticoagulants for treatment of HIT: update of Hamilton experience and literature review. Blood. 2017;130(9):1104-1113.

1377 1378

46. Walenga JM, Prechel M, Jeske WP, et al. Rivaroxaban—an oral, direct factor xa inhibitor—has potential for the management of patients with heparin-induced thrombocytopenia. Br J Haematol. 2008;143(1):92-99.

1379 1380 1381

47. Linkins L, Warkentin TE, Pai M, et al. Design of the rivaroxaban for heparin-induced thrombocytopenia study. J Thromb Thrombolysis. 2014;38(4):485-492.

1382

48. National Institutes of Health Clinical Center. Efficacy and safety of apixaban in the treatment of heparin induced thrombocytopenia (HIT). NCT03594045. ClinicalTrials.gov. Bethesda, MD: National Institutes of Health; 2018. https://clinicaltrials.gov/ct2/show/ NCT03594045. Updated January 25, 2019.

1384

49. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest. 2016;149(2):315-352.

1388

1383 1385 1386 1387 1389 1390

50. Groetzinger LM, Miller TJ, Rivosecchi RM, Smith RE, Gladwin MT, Rivera-Lebron BN. Apixaban or rivaroxaban versus warfarin for treatment of submassive pulmonary embolism after catheter-directed thrombolysis. Clin Appl Thromb Hemost. 2018;24(6):908-913.

1391 1392 1393 1394

51. McCullough M, Kholdani C, Zamanian RT. Prevention of deep vein thrombosis and pulmonary embolism in high-risk medical patients. Clin Chest Med. 2018;39(3):483-492.

1395 1396

52. Goldhaber SZ, Leizorovicz A, Kakkar AK, et al. Apixaban versus enoxaparin for thromboprophylaxis in medically ill patients. N Engl J Med. 2011;365(23):2167-2177.

1397 1398 1399

53. Cohen AT, Spiro TE, Büller HR, et al. Rivaroxaban for thromboprophylaxis in acutely ill medical patients. N Engl J Med. 2013;368(6):513-523. 54. Cohen AT, Harrington RA, Goldhaber SZ, et al. Extended thromboprophylaxis with betrixaban in acutely ill medical patients. N Engl J Med. 2016;375(6):534-544. 55. Spyropoulos AC, Ageno W, Albers GW, et al. Rivaroxaban for thromboprophylaxis after hospitalization for medical illness. N Engl J Med. 2018;379(12):1118-1127. 56. Al Yami MS, Kurdi S, Abraham I. Direct oral anticoagulants for extended thromboprophylaxis in medically ill patients: metaanalysis and risk/benefit assessment. J Blood Med. 2018;9:25-34.

1400 1401 1402 Q20

1403 1404 1405 1406 1407 1408

57. Tao DL, Bien JY, DeLoughery TG, Shatzel JJ. Extended thromboprophylaxis with direct oral anticoagulants for medical patients: a systematic review and meta-analysis. Blood. 2017;129(5): 653-655.

1409

58. Liew AYL, Piran S, Eikelboom JW, Douketis JD. Extended-duration versus short-duration pharmacological thromboprophylaxis in acutely ill hospitalized medical patients: a systematic review and meta-analysis of randomized controlled trials. J Thromb Thrombolysis. 2017;43(3):291-301.

1412

59. Shaw J, de Wit C, Le Gal G, Carrier M. Thrombotic and bleeding outcomes following perioperative interruption of direct oral anticoagulants in patients with venous thromboembolic disease. J Thromb Haemost. 2017;15(5):925-930.

1410 1411 1413 1414 1415 1416 1417 1418 1419

60. Lijfering WM, Tichelaar YIGV. Direct oral anticoagulant use and risk of perioperative bleeding: evidence of absence or absence of evidence? Res Pract Thromb Haemost. 2018;2(2):182-185.

1420

61. Barnes GD, Mouland E. Peri-procedural management of oral anticoagulants in the DOAC era. Prog Cardiovasc Dis. 2018;60(6): 600-606.

1422

62. Dubois V, Dincq A, Douxfils J, et al. Perioperative management of patients on direct oral anticoagulants. Thromb J. 2017;15:14.

1424

63. Hornor MA, Duane TM, Ehlers AP, et al. American College of Surgeons’ guidelines for the perioperative management of antithrombotic medication. J Am Coll Surg. 2018;227(5):536.e1.

1426

64. Birnie DH, Healey JS, Wells GA, et al. Continued vs. interrupted direct oral anticoagulants at the time of device surgery, in patients with moderate to high risk of arterial thrombo-embolic events (BRUISE CONTROL-2). Eur Heart J. 2018;39(44):3973-3979.

13

chestjournal.org

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

1421 1423 1425 1427 1428 1429 1430

1431

65. Vivas D, Roldán I, Ferrandis R, et al. Perioperative and periprocedural management of antithrombotic therapy: consensus document of SEC, SEDAR, SEACV, SECTCV, AEC, SECPRE, SEPD, SEGO, SEHH, SETH, SEMERGEN, SEMFYC, SEMG, SEMICYUC, SEMI, SEMES, SEPAR, SENEC, SEO, SEPA, SERVEI, SECOT and AEU. Rev Esp Cardiol (Engl Ed). 2018;71(7):553-564.

1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485

Q14

66. Weber J, Olyaei A, Shatzel J. The efficacy and safety of direct oral anticoagulants in patients with chronic renal insufficiency: a review of the literature. Eur J Haematol. 2019;102(4):312-318. 67. Tafur A, Douketis J. Perioperative management of anticoagulant and antiplatelet therapy. Heart. 2018;104(17):1461-1467. 68. Shaw JR, Woodfine JD, Douketis J, Schulman S, Carrier M. Perioperative interruption of direct oral anticoagulants in patients with atrial fibrillation: a systematic review and meta-analysis. Res Pract Thromb Haemost. 2018;2(2):282-290. 69. Schulman S, Carrier M, Lee AYY, et al. Perioperative management of dabigatran: a prospective cohort study. Circulation. 2015;132(3): 167-173. 70. ClinicalTrials.gov. NCT02228798. 71. Douketis JD, Spyropoulos AC, Anderson JM, et al. The perioperative Anticoagulant Use for Surgery Evaluation (PAUSE) study for patients on a direct oral anticoagulant who need an elective surgery or procedure: design and rationale. Thromb Haemost. 2017;117(12):2415-2424. 72. Chan N, Sager PT, Lawrence J, et al. Is there a role for pharmacokinetic/pharmacodynamic-guided dosing for novel oral anticoagulants? Am Heart J. 2018;199:59-67. 73. Cuker A, Siegal DM, Crowther MA, Garcia DA. Laboratory measurement of the anticoagulant activity of the non-vitamin K oral anticoagulants. J Am Coll Cardiol. 2014;64(11):1128-1139. 74. Eikelboom JW, Quinlan DJ, Hirsh J, Connolly SJ, Weitz JI. Laboratory monitoring of non-vitamin K antagonist oral anticoagulant use in patients with atrial fibrillation: a review. JAMA Cardiol. 2017;2(5):566-574. 75. Barrett YC, Wang Z, Frost C, Shenker A. Clinical laboratory measurement of direct factor xa inhibitors: anti-xa assay is preferable to prothrombin time assay. Thromb Haemost. 2010;104(6):1263-1271. 76. 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(4):692-694. 77. Eriksson BI, Dahl OE, Rosencher N, et al. Dabigatran etexilate versus enoxaparin for prevention of venous thromboembolism after total hip replacement: a randomised, double-blind, noninferiority trial. Lancet. 2007;370(9591):949-956. 78. Eriksson BI, Dahl OE, Rosencher N, et al. Oral dabigatran etexilate vs. subcutaneous enoxaparin for the prevention of venous thromboembolism after total knee replacement: the RE-MODEL randomized trial. J Thromb Haemost. 2007;5(11):2178-2185. 79. Eriksson BI, Borris LC, Friedman RJ, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty. N Engl J Med. 2008;358(26):2765-2775. 80. Fuji T, Wang CJ, Fujita S, et al. Safety and efficacy of edoxaban, an oral factor xa inhibitor, versus enoxaparin for thromboprophylaxis after total knee arthroplasty: the STARS E-3 trial. Thromb Res. 2014;134(6):1198-1204. 81. Fuji T, Fujita S, Kawai Y, et al. Efficacy and safety of edoxaban versus enoxaparin for the prevention of venous thromboembolism following total hip arthroplasty: STARS J-V. Thrombosis J. 2015;13: 27. 82. Kakkar AK, Brenner B, Dahl OE, et al. Extended duration rivaroxaban versus short-term enoxaparin for the prevention of venous thromboembolism after total hip arthroplasty: a doubleblind, randomised controlled trial. Lancet. 2008;372(9632):31-39. 83. Lassen MR, Raskob GE, Gallus A, Pineo G, Chen D, Portman RJ. Apixaban or enoxaparin for thromboprophylaxis after knee replacement. N Engl J Med. 2009;361(6):594-604.

14 Contemporary Reviews in Critical Care Medicine

84. Lassen MR, Gallus A, Raskob GE, et al. Apixaban versus enoxaparin for thromboprophylaxis after hip replacement. N Engl J Med. 2010;363(26):2487-2498. 85. Lassen MR, Raskob GE, Gallus A, et al. Apixaban versus enoxaparin for thromboprophylaxis after knee replacement (ADVANCE-2): a randomised double-blind trial. Lancet. 2010;375(9717):807-815.

1486 1487 1488 1489 1490 1491

86. Lassen MR, Ageno W, Borris LC, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty. N Engl J Med. 2008;358(26):2776-2786.

1492

87. Turpie AG, Lassen MR, Davidson BL, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after total knee arthroplasty (RECORD4): a randomised trial. Lancet. 2009;373(9676):1673-1680.

1494

1493 1495 1496

88. RE-MOBILIZE Writing Committee, Ginsberg JS, Davidson BL, et al. Oral thrombin inhibitor dabigatran etexilate vs North American enoxaparin regimen for prevention of venous thromboembolism after knee arthroplasty surgery. J Arthroplasty. 2009;24(1):1-9.

1498

89. Steinberg BA, Simon DN, Thomas L, et al. Management of major bleeding in patients with atrial fibrillation treated with non-vitamin K antagonist oral anticoagulants compared with warfarin in clinical practice (from phase II of the Outcomes Registry for Better Informed Treatment of Atrial Fibrillation ORBIT-AF II]). Am J Cardiol. 2017;119(10):1590-1595.

1502

90. Xu Y, Schulman S, Dowlatshahi D, et al. Direct oral anticoagulantor warfarin-related major bleeding: characteristics, reversal strategies, and outcomes from a multicenter observational study. Chest. 2017;152(1):81-91.

1497 1499 1500 1501 1503 1504 1505 1506 1507

91. Mentias A, Shantha G, Chaudhury P, Vaughan Sarrazin MS. Assessment of outcomes of treatment with oral anticoagulants in patients with atrial fibrillation and multiple chronic conditions: a comparative effectiveness analysis. JAMA Network Open. 2018;1(5): e182870.

1508

92. Einstein Investigators, Bauersachs R, Berkowitz SD, et al. Oral rivaroxaban for symptomatic venous thromboembolism. N Engl J Med. 2010;363(26):2499-2510.

1512

93. Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365(10):883891.

1509 1510 1511 1513 1514 1515 1516 1517

94. Hori M, Matsumoto M, Tanahashi N, et al. Rivaroxaban vs. warfarin in Japanese patients with atrial fibrillation—the J-ROCKET AF study. Circ J. 2012;76(9):2104-2111.

1518

95. Mega JL, Braunwald E, Wiviott SD, et al. Rivaroxaban in patients with a recent acute coronary syndrome. N Engl J Med. 2012;366(1): 9-19. 96. EINSTEIN-PE Investigators, Buller HR, Prins MH, et al. Oral rivaroxaban for the treatment of symptomatic pulmonary embolism. N Engl J Med. 2012;366(14):1287-1297.

1519 1520 1521 1522 1523

97. Cohen AT, Harrington RA, Goldhaber SZ, et al. Extended thromboprophylaxis with betrixaban in acutely ill medical patients. N Engl J Med. 2016;375(6):534-544. 98. Spyropoulos AC, Ageno W, Albers GW, et al. Rivaroxaban for thromboprophylaxis after hospitalization for medical illness. N Engl J Med. 2018;379(12):1118-1127.

1524 1525 1526 1527 1528 1529

99. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361(12):1139-1151.

1530

100. Schulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med. 2009;361(24):2342-2352.

1532

101. Schulman S, Kakkar AK, Goldhaber SZ, et al. Treatment of acute venous thromboembolism with dabigatran or warfarin and pooled analysis. Circulation. 2014;129(7):764-772.

1534

1531 1533 1535 1536

102. Schulman S, Kearon C, Kakkar AK, et al. Extended use of dabigatran, warfarin, or placebo in venous thromboembolism. N Engl J Med. 2013;368(8):709-718.

1538

103. Alexander JH, Lopes RD, James S, et al. Apixaban with antiplatelet therapy after acute coronary syndrome. N Engl J Med. 2011;365(8): 699-708.

1540

[

-#- CHEST - 2019

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296

1537

]

1539

1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574

104. Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365(11):981-992. 105. Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med. 2013;369(9):799-808. 106. Agnelli G, Buller HR, Cohen A, et al. Apixaban for extended treatment of venous thromboembolism. N Engl J Med. 2013;368(8): 699-708. 107. Hokusai-VTE Investigators, Buller HR, Decousus H, et al. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism. N Engl J Med. 2013;369(15):1406-1415. 108. Giugliano RP, Ruff CT, Braunwald E, et al. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369(22):2093-2104. 109. Connolly SJ, Ezekowitz MD, Yusuf S, Reilly PA, Wallentin L. Randomized Evaluation of Long-Term Anticoagulation Therapy Investigators. Newly identified events in the RE-LY trial. N Engl J Med. 2010;363(19):1875-1876. 110. Koretsune Y, Yamashita T, Kimura T, Fukuzawa M, Abe K, Yasaka M. Short-term safety and plasma concentrations of edoxaban in Japanese patients with non-valvular atrial fibrillation and severe renal impairment. Circ J. 2015;79(7):1486-1495. 111. Kubitza D, Mueck W, Becka M. Randomized, double-blind, crossover study to investigate the effect of rivaroxaban on QTinterval prolongation. Drug Saf. 2008;31(1):67-77. 112. Frost C, Nepal S, Byon W, et al. Randomized, blinded, placebo- and positive-controlled crossover study to determine the effect of multiple doses of apixaban on the QTc interval. J Clin Pharmacol. 2015;55(5):549-555. 113. Mendell J, Basavapathruni R, Swearingen D, Draves A, Zhang G, Morganroth J. A thorough electrocardiogram study of edoxaban, a novel factor xa inhibitor. J Clin Pharmacol. 2011;51(8):1241-1246. 114. Lip GYH, Banerjee A, Boriani G, et al. Antithrombotic therapy for atrial fibrillation: CHEST guideline and expert panel report. Chest. 2018;154(5):1121-1201. 115. Witt DM, Nieuwlaat R, Clark NP, et al. American Society of Hematology 2018 guidelines for management of venous thromboembolism: Optimal management of anticoagulation therapy. Blood Adv. 2018;2(22):3257. 116. Santibanez M, Lesch CA, Lin L, Berger K. Tolerability and effectiveness of 4-factor prothrombin complex concentrate (4F-

1579

PCC) for warfarin and non-warfarin reversals. J Crit Care. 2018;48: 183-190.

1580

117. Luu H, Ewenstein B. FEIBA safety profile in multiple modes of clinical and home-therapy application. Haemophilia. 2004;10(suppl 2):10-16.

1581 1582 1583

118. Brown KS, Wickremasingha P, Parasrampuria DA, et al. The impact of a three-factor prothrombin complex concentrate on the anticoagulatory effects of the factor xa inhibitor edoxaban. Thromb Res. 2015;136(4):825-831. 119. Engelbart JM, Zepeski A, Galet C, Policeni B, Skeete DA, Faine BA. Safety and effectiveness of Factor Eight Inhibitor Bypassing Activity for direct oral anticoagulant-related hemorrhage reversal. Am J Emerg Med. 2019;37(2):214-219.

1584 1585 1586 1587 Q15

120. Schulman S, Ritchie B, Nahirniak S, et al. Reversal of dabigatranassociated major bleeding with activated prothrombin concentrate: a prospective cohort study. Thromb Res. 2017;152:44-48.

1588 1589 1590 1591 1592

121. Mao G, King L, Young S, Kaplan R. Factor eight inhibitor bypassing agent (FEIBA) for reversal of target-specific oral anticoagulants in life-threatening intracranial bleeding. J Emerg Med. 2017;52(5):731-737.

1593 1594 1595

122. Yin EB, Tan B, Nguyen T, et al. Safety and effectiveness of factor VIII inhibitor bypassing activity (FEIBA) and fresh frozen plasma in oral anticoagulant-associated intracranial hemorrhage: a retrospective analysis. Neurocrit Care. 2017;27(1):51-59.

1596 1597 1598

123. Pollack CV, Reilly PA, van Ryn J, et al. Idarucizumab for dabigatran reversal—full cohort analysis. N Engl J Med. 2017;377(5):431-441.

1599 1600

124. Connolly SJ, Milling TJJ, Eikelboom JW, et al. Andexanet alfa for acute major bleeding associated with factor xa inhibitors. N Engl J Med. 2016;375(12):1131-1141.

1601 1602 1603

125. Bouchard J, Ghannoum M, Bernier-Jean A, et al. Comparison of intermittent and continuous extracorporeal treatments for the enhanced elimination of dabigatran. Clin Toxicol (Phila). 2015;53(3):156-163.

1604 1605 1606

126. Ansell JE, Bakhru SH, Laulicht BE, et al. Single-dose ciraparantag safely and completely reverses anticoagulant effects of edoxaban. Thromb Haemost. 2017;117(2):238-245. 127. National Institutes of Health Clinical Center. Phase 2 study of apixaban reversal by ciraparantag as measured by WBCT. NCT03288454. ClinicalTrials.gov. Bethesda, MD: National Institutes of Health; 2017. https://clinicaltrials.gov/ct2/show/ NCT03288454. Updated November 2, 2018.

1607 1608 1609 1610 Q16

1611 1612

1575

1613

1576

1614

1577

1615

1578

1616

15

chestjournal.org

REV 5.6.0 DTD  CHEST2390_proof  15 July 2019  3:19 pm  EO: CHEST-19-0296