Open access peer-reviewed chapter

An Overview of the Anticoagulant Drugs Used in Routine Clinical Practice

Written By

Nina Vene and Alenka Mavri

Submitted: 17 November 2017 Reviewed: 05 March 2018 Published: 29 August 2018

DOI: 10.5772/intechopen.76206

From the Edited Volume

Anticoagulant Drugs

Edited by Mojca Božič-Mijovski

Chapter metrics overview

1,494 Chapter Downloads

View Full Metrics

Abstract

Anticoagulant drugs directly or indirectly influence coagulation factors preventing fibrin formation thus preventing blot clotting. They are classified into two groups according to the mode of application, namely parenteral and oral drugs. Among the latter, vitamin K antagonists (most often warfarin) were the only available oral drugs and were widely used for almost a century. In the recent years, new oral anticoagulant drugs became available that directly target either factor IIa or Xa. This chapter provides an overview of both parenteral and oral anticoagulant drugs used in clinical practice with description of the mode of action and management of therapy in different clinical settings.

Keywords

  • anticoagulant drugs
  • indications
  • therapy

1. Introduction

Anticoagulant drugs directly or indirectly influence coagulation factors and thus inhibit the initiation and progress of coagulation and fibrin-clot formation. They are classified into two groups according to the mode of application, namely parenteral and oral drugs. Among the latter, vitamin K antagonists (most often warfarin) were the only available oral anticoagulants and were widely used for almost a century. In recent years, new oral anticoagulant drugs became available that directly target either factor IIa or Xa [1].

This chapter provides an overview of both parenteral and oral anticoagulant drugs used in clinical practice with a description of the mode of action and management of therapy in different clinical settings.

Advertisement

2. Parenteral anticoagulant drugs

2.1. Unfractionated heparin

Unfractionated heparin (UFH) binds antithrombin—a physiological inhibitor of coagulation—and accelerates its inhibitory action against coagulation factors II and X and in minor degrees also factors IX, XI and XII [2, 3]. UFH is active in a parenteral form only and therefore administered by intravenous (i.v.) infusion [2]. It is used for the treatment of acute thromboembolic events. One of the major disadvantages of UFH is its binding to plasma proteins and endothelial cells making its anticoagulant effect unpredictable [2, 3]. Treatment with UFH must, therefore, be regularly monitored with activated partial thromboplastin time (APTT). Due to different sensitivities of APTT reagents the therapeutic APTT range must be determined by each laboratory and must correspond to heparin anti-factor Xa activity between 0.3 and 0.7 IU/mL [4, 5, 6]. Treatment is initiated with UFH bolus of 80 U/kg i.v. and continued with continuous infusion of 18 U/kg body mass/h [7]. Dosage must be adjusted according to the APTT result. At the beginning of treatment, laboratory monitoring is needed several times a day, the first one 6 h after UFH initiation. The two most important non-hemorrhagic side effects of UFH treatment are osteoporosis and thrombocytopenia [2].

2.2. Low-molecular weight heparin

Low-molecular weight heparin (LMWH) is obtained by various methods of fractionation or depolymerization of polymeric UFH [8]. Because LMWHs differ in molecular mass, they also differ in pharmacological characteristics and anticoagulant effects [9]. All LMWHs inhibit coagulation factors II and X. Among the most commonly used LMWHs for treatment and the prevention of acute thromboembolic events are dalteparin (Fragmin®), enoxaparin (Clexane®) and nadroparin (Fraxiparine® and Fraxiparine forte®) in the form of subcutaneous injections. They can also be used as a bridging therapy in patients with high thromboembolic risk during a period when these patients cannot receive oral anticoagulants. Therapeutic dose is determined according to the patient’s body weight [2] (Table 1).

Therapeutic dose
LMWHTwice dailyOnce daily
Dalteparin (Fragmin®)100 IU/kg BW/12 h sc200 IU/kg BW/24 h sc
46–56 kg5.000 IU/12 h sc10.000 IU/24 h sc
57–68 kg6.000 IU/12 h sc12.500 IU/24 h sc
69–82 kg7.500 IU/12 h sc15.000 IU/24 h sc
82–120 kg100 IU/kgBW/12 h sc18.000 IU/24 h sc
Enoksaparin (Clexane®)1 mg/kg BW/12 h sc1.5 mg/kg BW/24 h sc
45–54 kg50 mg/12 h sc80 mg/24 h sc
55–64 kg60 mg/12 h sc90 mg/24 h sc
65–74 kg70 mg/12 h sc100 mg/24 h sc
75–84 kg80 mg/12 h sc120 mg/24 h sc
85–94 kg90 mg/12 h sc135 mg/24 h sc
94–120 kg100 mg/12 h sc150 mg/24 h sc
Nadroparin(Fraxiparine®) 0.1 ml/10 kg BW /12 h sc(Fraxiparine FORTE®) 0.1 ml/10 kg BW/24 h sc
50–59 kg0.5 ml/12 h sc0.5 ml/24 h sc
60–69 kg0.6 ml/12 h sc0.6 ml/24 h sc
70–79 kg0.7 ml/12 h sc0.7 ml/24 h sc
80–89 kg0.8 ml/12 h sc0.8 ml/24 h sc
90–120 kg0.9 ml/12 h sc0.9 ml/24 h sc

Table 1.

LMWH therapeutic doses according to body weight.

IU: International Units, BW: body weight, sc: subcutaneously.

For prevention of venous thromboembolism (VTE), lower (prophylactic) doses of LMWH are used [2] (Table 2). The adequate LMWH dose is selected according to the risk. Prophylactic doses are used in some patients during the interim cessation of oral anticoagulant therapy above all in the first days after large interventions.

LMWHLow prophylactic dose (moderate VTE risk)High prophylactic dose (high VTE risk)
Dalteparin2500 IU/24 h sc5000 IU/24 h sc
Enoxaparin20 mg/24 h sc40 mg/24 h sc
Nadroparin0.3 ml/24 h sc0.4 ml/24 h sc at BW ≤ 70 kg
0.6 ml/24 h sc at BW > 70 kg

Table 2.

Prophylactic doses of LMWH.

IU: International Units, BW: body weight, sc: subcutaneously, VTE: venous thromboembolism.

The most important advantage of LMWH over UFH is the lower degree of binding to plasma proteins and endothelial cells making their pharmacokinetics and anticoagulant effects predictable [10, 11]. Regular laboratory monitoring with coagulation tests is therefore not needed, except in patients with kidney disease and patients with very low (under 45 kg) or very high (above 120 kg) body weight [2]. Although APTT may be mildly prolonged during LMWH therapy it cannot be used for monitoring. The chromogenic anti-Xa is the test of choice for the determination of plasma LMWH concentration [12]. The LMWH dose should be adjusted to 0.5–1.0 IU/mL 4 h after the last LMWH dose when administered twice daily or to 1.0–2.0 IU/mL 5–6 h after the last dose when administered once daily [13, 14]. The two main non-hemorrhagic side effects of LMWH therapy are osteopenia and thrombocytopenia; however, both these side effects are considerably rarer compared to UFH therapy [2].

2.3. Fondaparin

Fondaparin (Arixtra®) is a synthetic pentasaccharide that closely resembles the pentasaccharide naturally occurring in the UFH and LMWH. It is an antithrombin-mediated factor Xa inhibitor that is devoid of any anti-factor IIa (thrombin) activity [15]. It is used for treating patients with acute coronary syndrome and heparin-induced thrombocytopenia. It is indicated also for certain patients with thrombophlebitis in a fixed dose of 2.5 mg daily s.c. Laboratory monitoring is not needed; however, if necessary fondaparin levels should only be determined using assays that use known fondaparin concentrations to generate their calibration curve. The use of fondaparin in patients with creatinine clearance below 30 mL/min is contraindicated [2].

2.4. Hirudin

Hirudin is a naturally occurring peptide in the salivary glands of medicinal leeches that irreversibly inhibits thrombin. Lepirudin, a recombinant hirudin derived from yeast cells, was used in clinical practice but is no longer available. Instead, the synthetic analog—bivalirudin (Angiox®)—with a short half-life is used at percutaneous coronary interventions and for treating patients with heparin-induced thrombocytopenia. The use of bivalirudin in patients with creatinine clearance below 30 mL/min is contraindicated [16].

2.5. Argatroban

Argatroban (Argatra®) is a synthetic reversible direct thrombin inhibitor. It is metabolized solely in the liver and is, therefore, suitable for patients with renal failure. It is used in patients with heparin-induced thrombocytopenia. Treatment with argatroban requires laboratory monitoring with activated partial thromboplastin time (APTT) and the dose adjusted to reach 1.5–3.0 times prolonged baseline APTT, but should not exceed 100 s [17].

Advertisement

3. Oral anticoagulants

3.1. Vitamin K antagonists

The vitamin K-dependent coagulation factors II, VII, IX and X require γ-carboxylation for their procoagulant activity. Treatment with vitamin K antagonists results in the hepatic production of partially carboxylated and decarboxylated proteins with reduced coagulant activity. Among the most commonly used vitamin K antagonists are warfarin and acenocoumarol. Although vitamin K antagonists are absorbed quickly their full effect develops after about 5 days when the activity of all vitamin K-dependent coagulation factors is reduced [1].

Warfarin therapy requires regular laboratory monitoring with prothrombin time (PT). Due to different sensitivities of thromboplastin reagents used for PT measurement the results are expressed as the International Normalized Ratio (INR). For the majority of indications the target INR range falls between 2.0 and 3.0. In certain patient populations, for example, in patients with mechanical heart valves, the target range is 2.5–3.5 INR. A rare non-hemorrhagic side effect of vitamin K antagonist therapy is skin necrosis that develops at therapy initiation and is a consequence of acute thrombosis of subcutaneous venules and capillaries [1].

3.2. Direct oral anticoagulants

3.2.1. Dabigatran

Dabigatran etexilate (Pradaxa®) is a low-molecular weight prodrug that exhibits no pharmacological activity. After oral administration, dabigatran etexilate is converted to its active form, dabigatran, a potent, competitive and reversible direct thrombin inhibitor [18]. The binding of dabigatran to thrombin is specific and selective and includes both free and thrombus-bound thrombin. Maximal blood concentration of dabigatran is reached after 1–3 h after the intake [18]. About 35% of the drug is bound to plasma proteins. Eighty percent of dabigatran is excreted through the kidneys [18]. Dabigatran half-life is 14–17 h [18]. It is given in fixed doses of either 150 or 110 mg twice daily in patients with atrial fibrillation and 150 mg twice daily in patients with VTE [19, 20]. Prophylactic doses after total hip or knee replacement are 220 or 150 mg once daily with only half the dose given as the first dose after surgery [21].

The anticoagulant effect of dabigatran is predictive and, therefore, requires no regular laboratory monitoring. During dabigatran therapy, APTT and thrombin time (TT) are prolonged, but these two tests can only offer a rough approximation of dabigatran blood concentration. In certain situations when dabigatran concentration needs to be assessed, a specific test must be used, such as modified thrombin time or a chromogenic assay [22, 23].

3.2.2. Rivaroxaban

Rivaroxaban (Xarelto®) directly inhibits factor Xa. It selectively binds both free and prothrombin complex bound factor Xa and in this way inhibits thrombin and clot formation. Peak blood concentration is achieved after 1–3 h after drug ingestion. As much as 95% of the drug is bound to plasma protein. One-third of the drug is excreted through kidneys, the other two-thirds are metabolized in the liver. The drug half-life is 8–13 h [1, 24]. Therapeutic doses are 20 and 15 mg once daily for patients with atrial fibrillation [25]. Patients with VTE are treated with 15 mg twice daily for the first 3 weeks, followed by 20 mg once daily [26, 27]. The drug must always be ingested with food. The prophylactic dose for patients with total hip or knee replacement is 10 mg once daily [28, 29].

No laboratory monitoring of therapy is needed due to the predictive effect of the drug. Rivaroxaban prolongs PT; however, when an assessment of the drug blood level is needed, an anti-Xa test calibrated to rivaroxaban should be used [30].

3.2.3. Apixaban

Apixaban (Eliquis®) directly and reversibly inhibits factor Xa. Maximal blood concentration of the drug is achieved 3–4 h after ingestion. As much as 87% of the drug is bound to blood protein. Twenty-seven percent of the drug is excreted through kidneys and the remainder through the liver. The drug half-life is 12 h [31]. Patients with atrial fibrillation are treated with 5 or 2.5 mg twice daily [32]. Patients with VTE are treated with 10 mg twice daily for the first 7 days followed by 5 mg daily [33]. The prophylactic dose for patients with total hip or knee replacement is 2.5 mg twice daily [34].

No laboratory monitoring of therapy is needed due to the predictive effect of the drug. Apixaban unreliably prolongs APTT and PT. When an assessment of the drug blood level is needed, an anti-Xa test calibrated to apixaban should be used [35].

3.2.4. Edoxaban

Edoxaban directly inhibits factor Xa. Maximal blood concentration of the drug is achieved 1–2 h after the ingestion. About 40–59% of the drug is bound to plasma protein. Roughly 35% of the drug is excreted through the kidneys and the remainder through the liver. The drug half-life is 9–14 h. Therapeutic doses are 60 and 30 mg daily for patients with atrial fibrillation and VTE. The prophylactic dose for patients with total hip or knee replacement is 30 mg once daily. Edoxaban prolongs APTT and PT, but for a quantitative assessment of the drug level, an anti-Xa test calibrated to edoxaban must be utilized [36, 37].

References

  1. 1. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Oral anticoagulant therapy antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(2):E44s-E88s
  2. 2. Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(2):E24s-E43s
  3. 3. Hirsh J, Raschke R, Warkentin TE, Dalen JE, Deykin D, Poller L. Heparin: Mechanism of action, pharmacokinetics, dosing considerations, monitoring, efficacy, and safety. Chest. 1995;108(4):258S-275S
  4. 4. Kitchen S, Jennings I, Woods TAL, Preston FE. Wide variability in the sensitivity of APTT reagents for monitoring of heparin dosage. Journal of Clinical Pathology. 1996;49(1):10-14
  5. 5. Manzato F, Mengoni A, Grilenzoni A, Lippi G. Evaluation of the activated partial thromboplastin time (APTT) sensitivity to heparin using five commercial reagents: Implications for therapeutic monitoring. Clinical Chemistry and Laboratory Medicine. 1998;36(12):975-980
  6. 6. Toulon P, Boutiere B, Horellou M-H, Trzeciak MC. Monitoring heparin therapy using activated partial thromboplastin time—Results of a multicenter trial establishing the therapeutic range for SILIMAT, a reagent with high sensitivity to heparin. Thrombosis and Haemostasis. 1998;80:104-108
  7. 7. Hirsh J, Warkentin TE, Shaughnessy SG, Anand SS, Halperin JL, Raschke R, Granger C, Ohman EM, Dalen JE. Heparin and low-molecular-weight heparin mechanisms of action, pharmacokinetics, dosing, monitoring, efficacy, and safety. Chest. 2001;119(1):64S-94S
  8. 8. Heit JA. Low-molecular-weight heparin: Biochemistry, pharmacology, and concurrent drug precautions. Regional Anesthesia and Pain Medicine. 1998;23(6 Suppl 2):135-139
  9. 9. Collignon F, Frydman A, Caplain H, Ozoux ML, Le Roux Y, Bouthier J, Thébault JJ. Comparison of the pharmacokinetic profiles of three low molecular mass heparins—dalteparin, enoxaparin and nadroparin—administered subcutaneously in healthy volunteers (doses for prevention of thromboembolism). Thrombosis and Haemostasis. 1995;73(4):630-640
  10. 10. Young E, Wells P, Holloway S, Weitz J, Hirsh J. Ex-vivo and in-vitro evidence that low molecular weight heparins exhibit less binding to plasma proteins than unfractionated heparin. Thrombosis and Haemostasis. 1994;71(3):300-304
  11. 11. Boneu B. Low molecular weight heparin therapy: Is monitoring needed? Thrombosis and Haemostasis. 1994;72(3):330-334
  12. 12. Douxfils J, Tamigniau A, Chatelain B, Goffinet C, Dogne JM, Mullier F. Measurement of non-VKA oral anticoagulants versus classic ones: The appropriate use of hemostasis assays. Thrombosis Journal. 2014;12:24
  13. 13. Bates SM, Weitz JI. Coagulation assays. Circulation. 2005;112(4):e53-e60
  14. 14. Peternel P, Terbižan M, Tratar G, Božič M, Horvat D, Salobir B, Stegnar M. Markers of hemostatic system activation during treatment of deep vein thrombosis with subcutaneous unfractionated or low-molecular weight heparin. Thrombosis Research. 2002;105:241-245
  15. 15. Bauer KA, Hawkins DW, Peters PC, Petitou M, Herbert JM, van Boeckel CAA, Meuleman DG. Fondaparinux, a synthetic pentasaccharide: The first in a new class of antithrombotic agents—the selective factor Xa inhibitors. Cardiovascular Drug Reviews. 2002;20(1):37-52
  16. 16. Hirsh J, Bauer KA, Donati MB, Gould M, Samama MM, Weitz JI. Parenteral anticoagulants: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (8th edition). Chest. 2008;133(6 Suppl):141S-159S
  17. 17. Lewis BE, Wallis DE, Berkowitz SD, Matthai WH, Fareed J, Walenga JM, Bartholomew J, Sham R, Lerner RG, Zeigler ZR, Rustagi PK, Jang IK, Rifkin SD, Moran J, Hursting MJ, Kelton JG, Investigators ARGS. Argatroban anticoagulant therapy in patients with heparin-induced thrombocytopenia. Circulation. 2001;103(14):1838-1843
  18. 18. Hankey GJ, Eikelboom JW. Dabigatran etexilate: A new oral thrombin inhibitor. Circulation. 2011;123(13):1436-1450
  19. 19. Schulman S, Kearon C, Kakkar AK, Mismetti P, Schellong S, Eriksson H, Baanstra D, Schnee J, Goldhaber SZ, R.-C.S. Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. The New England Journal of Medicine. 2009;361(24):2342-2352
  20. 20. Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A, Pogue J, Reilly PA, Themeles E, Varrone J, Wang S, Alings M, Xavier D, Zhu J, Diaz R, Lewis BS, Darius H, Diener HC, Joyner CD, Wallentin L, R.-L.S. Committee, Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. The New England Journal of Medicine. 2009;361(12):1139-1151
  21. 21. Eriksson BI, Dahl OE, Rosencher N, Kurth AA, van Dijk CN, Frostick SP, Kalebo P, Christiansen AV, Hantel S, Hettiarachchi R, Schnee J, Buller HR, R.-M.S. Group. Oral dabigatran etexilate vs. subcutaneous enoxaparin for the prevention of venous thromboembolism after total knee replacement: The RE-MODEL randomized trial. Journal of Thrombosis and Haemostasis. 2007;5(11):2178-2185
  22. 22. Bozic-Mijovski M, Malmstrom RE, Malovrh P, Antovic JP, Vene N, Sinigoj P, Mavri A. Diluted thrombin time reliably measures low to intermediate plasma dabigatran concentrations. Annals of Clinical Biochemistry. 2015;53(4):446-451
  23. 23. Gosselin R, Hawes E, Moll S, Adcock D. Performance of various laboratory assays in the measurement of dabigatran in patients receiving therapeutic doses: A prospective study based on peak and trough plasma levels. American Journal of Clinical Pathology. 2014;141(2):262-267
  24. 24. Abdulsattar Y, Bhambri R, Nogid A. Rivaroxaban (Xarelto) for the prevention of thromboembolic disease: An inside look at the oral direct factor Xa inhibitor. P T. 2009;34(5):238-244
  25. 25. Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, Breithardt G, Halperin JL, Hankey GJ, Piccini JP, Becker RC, Nessel CC, Paolini JF, Berkowitz SD, Fox KA, Califf RM, Investigators RA. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. The New England Journal of Medicine. 2011;365(10):883-891
  26. 26. Investigators E, Bauersachs R, Berkowitz SD, Brenner B, Buller HR, Decousus H, Gallus AS, Lensing AW, Misselwitz F, Prins MH, Raskob GE, Segers A, Verhamme P, Wells P, Agnelli G, Bounameaux H, Cohen A, Davidson BL, Piovella F, Schellong S. Oral rivaroxaban for symptomatic venous thromboembolism. The New England Journal of Medicine. 2010;363(26):2499-2510
  27. 27. Investigators E-P, Buller HR, Prins MH, Lensin AW, Decousus H, Jacobson BF, Minar E, Chlumsky J, Verhamme P, Wells P, Agnelli G, Cohen A, Berkowitz SD, Bounameaux H, Davidson BL, Misselwitz F, Gallus AS, Raskob GE, Schellong S, Segers A. Oral rivaroxaban for the treatment of symptomatic pulmonary embolism. The New England Journal of Medicine. 2012;366(14):1287-1297
  28. 28. Eriksson BI, Borris L, Dahl OE, Haas S, Huisman MV, Kakkar AK, Misselwitz F, Kalebo P, Investigators OD-HS. Oral, direct factor Xa inhibition with BAY 59-7939 for the prevention of venous thromboembolism after total hip replacement. Journal of Thrombosis and Haemostasis. 2006;4(1):121-128
  29. 29. Turpie AG, Fisher WD, Bauer KA, Kwong LM, Irwin MW, Kalebo P, Misselwitz F, Gent M, OdiXa-Knee Study G. BAY 59-7939: An oral, direct factor Xa inhibitor for the prevention of venous thromboembolism in patients after total knee replacement. A phase II dose-ranging study. Journal of Thrombosis and Haemostasis. 2005;3(11):2479-2486
  30. 30. Lindhoff-Last E, Samama MM, Ortel TL, Weitz JI, Spiro TE. Assays for measuring rivaroxaban: Their suitability and limitations. Therapeutic Drug Monitoring. 2010;32(6):673-679
  31. 31. Byon W, Sweeney K, Frost C, Boyd RA. Population pharmacokinetics, pharmacodynamics, and exploratory exposure-response analyses of apixaban in subjects treated for venous thromboembolism. CPT: Pharmacometrics & Systems Pharmacology. 2017;6(5):340-349
  32. 32. Connolly SJ, Eikelboom J, Joyner C, Diener HC, Hart R, Golitsyn S, Flaker G, Avezum A, Hohnloser SH, Diaz R, Talajic M, Zhu J, Pais P, Budaj A, Parkhomenko A, Jansky P, Commerford P, Tan RS, Sim KH, Lewis BS, Van Mieghem W, Lip GY, Kim JH, Lanas-Zanetti F, Gonzalez-Hermosillo A, Dans AL, Munawar M, O'Donnell M, Lawrence J, Lewis G, Afzal R, Yusuf S, A.S. Committee, Investigators. Apixaban in patients with atrial fibrillation. The New England Journal of Medicine. 2011;364(9):806-817
  33. 33. Agnelli G, Buller HR, Cohen A, Curto M, Gallus AS, Johnson M, Masiukiewicz U, Pak R, Thompson J, Raskob GE, Weitz JI, Investigators A. Oral apixaban for the treatment of acute venous thromboembolism. The New England Journal of Medicine. 2013;369(9):799-808
  34. 34. Raskob GE, Gallus AS, Pineo GF, Chen D, Ramirez LM, Wright RT, Lassen MR. Apixaban versus enoxaparin for thromboprophylaxis after hip or knee replacement: Pooled analysis of major venous thromboembolism and bleeding in 8464 patients from the ADVANCE-2 and ADVANCE-3 trials. Journal of Bone and Joint Surgery. British Volume (London). 2012;94(2):257-264
  35. 35. Gouin-Thibault I, Flaujac C, Delavenne X, Quenet S, Horellou MH, Laporte S, Siguret V, Lecompte T. Assessment of apixaban plasma levels by laboratory tests: Suitability of three anti-Xa assays. A multicentre French GEHT study. Thrombosis and Haemostasis. 2014;111(2):240-248
  36. 36. Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL, Waldo AL, Ezekowitz MD, Weitz JI, Spinar J, Ruzyllo W, Ruda M, Koretsune Y, Betcher J, Shi M, Grip LT, Patel SP, Patel I, Hanyok JJ, Mercuri M, Antman EM, Investigators EA-T. Edoxaban versus warfarin in patients with atrial fibrillation. The New England Journal of Medicine. 2013;369(22):2093-2104
  37. 37. Ruff CT, Giugliano RP, Braunwald E, Morrow DA, Murphy SA, Kuder JF, Deenadayalu N, Jarolim P, Betcher J, Shi MG, Brown K, Patel I, Mercuri M, Antman EM. Association between edoxaban dose, concentration, anti-factor Xa activity, and outcomes: An analysis of data from the randomised, double-blind ENGAGE AF-TIMI 48 trial. Lancet. 2015;385(9984):2288-2295

Written By

Nina Vene and Alenka Mavri

Submitted: 17 November 2017 Reviewed: 05 March 2018 Published: 29 August 2018