Skip to main content

An update on applications and limitations of direct oral anticoagulants

Abstract

A major advancement in the field of medicine has been the introduction and usage of direct oral anticoagulants (DOACs) such as dabigatran (Pradaxa), apixaban (Eliquis), and rivaroxaban (Xarelto). DOACs have been increasing in popularity for mainstay anticoagulation pharmacotherapy and are being preferred by physicians over warfarin due to their rapid onset, fewer drug and food interactions, and lack of frequent blood monitoring. DOACs have been indicated in the management of thromboembolic conditions and have been extensively researched in various medical trials and studies before the approval of dabigatran (Pradaxa) in 2010 by the FDA. DOACs, like warfarin, are associated with a risk of bleeding, requiring clearance of the drug from the bloodstream or administration of reversal agents. It is important for physicians to familiarize themselves with the various types of DOACs and their dosages, along with their advantages and disadvantages in comparison to other non-DAOC classes of medications before incorporating them into their patient management plans.

Introduction

History of the evolution of oral anticoagulants over decades included the discovery of warfarin in the 1940s, dabigatran in 2010, rivaroxaban in 2011, and apixaban in 2012 followed by edoxaban in 2014 [1]. Dabigatran was the first US FDA-approved oral anticoagulant for non-valvular atrial fibrillation and was subsequently studied for other indications [2]. Vitamin K antagonists such as coumadin were the only known oral anticoagulant for decades until the recent introduction of DOACs which target the specific steps of the coagulation cascade, are efficacious, and can be used without regular monitoring [3], but it has been found that DOACs have some interaction with food particles like St. John Worts and dietary fiber [4]. The widely used oral anticoagulants include warfarin, phenprocoumon, acenocoumarol which acts as a vitamin K antagonist, dabigatran, a thrombin inhibitor, and factor Xa inhibitor including apixaban, edoxaban, and rivaroxaban and commonly used parenteral anticoagulants include heparin, thrombin inhibitors (bivalirudin, argatroban) and fondaparinux [5]. DOACs like dabigatran, apixaban, edoxaban, and rivaroxaban are used for the prevention and treatment of arterial and venous thromboembolic diseases, prevention of stroke and systemic embolism in non-valvular atrial fibrillation (NVAF), prevention of recurrent VTE, thromboprophylaxis in major orthopedic surgeries including knee and hip arthroplasty, and treatment of left ventricular (LV) thrombus [6, 7]. A meta-analysis of 14 studies and 2498 patients found the use of DOACs as non-inferior to vitamin K inhibitors with a better safety profile and patient quality of life [8]. The indication associated dosage of DOACs is tabularized in (Table 1) [9]. A newly studied indication for use of DOACs is the role of low-dose rivaroxaban in the prophylaxis of LV thrombus post-PCI in anterior wall myocardial infarction [10]. Absolute contraindications to the use of DOACs include atrial fibrillation in mechanical valves as studied in the RE-ALIGN trial [11]. DOACS have been studied in inherited thrombophilia and are proven to be non-inferior to vitamin K antagonists but due to limited supporting data, the guidelines still do not recommend DOACs as the drug of choice for inherited thrombophilia, especially anti-phospholipid syndrome [12]. Use of DOACs has been associated with a lower risk of major bleed or a clinically relevant non-major bleed in patients with chronic kidney disease or end stage renal disease (on dialysis) as compared to warfarin [13]. Dose adjustment is warranted for apixaban in patients with Creatinine clearance (CrCl) < 30 ml/min but dabigatran is avoided in patients with CrCl < 30 ml/min and drugs like betrixaban, edoxaban, and rivaroxaban are avoided in CrCl < 15 ml/min. Although previously DOACS were not used in patients post bioprosthetic valves implantation and repair, ENVALE study prove that DOACs were non-inferior to warfarin and DOACs were also previously not indicated in cancer-associated VTE, but its use was proven to be non-inferior to low molecular weight heparin (LMWH) in ADAM VTE trial [14,15,16]. The ARISTOPHANES study showed that apixaban, rivaroxaban, and dibigatran were superior to vitamin K inhibitors and increased event free time after 12 months of initiation [17].

Table 1 Indications and the dose regimen for usage of DOACs

There are limited studies on the effect of obesity on the pharmacokinetics and dynamics of DOACs; however, available evidence suggests the standard dose use of certain DOACs such as apixaban and rivaroxaban for VTE and AF in patients with BMI > 40 kg/m2 and weight > 120 kg [15, 18, 19]. Apixaban and Endoxaban may be used in elderly patients (age > 75) for the treatment of NVAF [20]. Due to limited studies, the role of apixaban as an anticoagulant in pediatric patients, patients with cancer, low or high body BMI, identified thrombophilia, heparin-induced thrombocytopenia (HIT), and poor renal function is questionable [21]. A recent open-label, non-inferiority trial by Agnelli et al. concluded that apixaban was non-inferior to subcutaneous dalteparin for the treatment of cancer-associated venous thromboembolism without an increased risk of major bleeding. DOACs are relatively contra-indicated in patients with cancer due to increased risk of bleeding, but this trial showed comparable rates of bleeding including major gastrointestinal bleeding in both the patient arms. Patients with high thromboembolic risk like lung and colorectal cancer were well represented in this study and no anticancer therapy was excluded, which lead to the inclusion of patients receiving various cytotoxic and biologic therapies. Patients with a diagnosis of brain cancer with metastasis and acute leukemia were excluded from this study due to increased risk of bleeding and hence the results are not generalizable to this group [22].

The guidelines prohibit the use of DOACs in post-bariatric patients due to concerns regarding absorption. However, there are limited studies to validate this concern and a recent institutional retrospective review showed no recurrent VTE after apixaban use and only 1 case with rivaroxaban [23]. Another cohort of 110 patients showed safety in the use of rivaroxaban as a prophylactic agent in patients undergoing bariatric surgery at a 6 months follow-up [13].

The use of DOACs is associated with major side effects like fatal bleeding (0.06 to 0.30%) spontaneous hemorrhage including gastrointestinal bleeding (0.35 to 2.09%), intracranial hemorrhage (0.09 to 0.51%), and renal disease progression [6, 24].

The presence of gastrointestinal bleeding (GIB) does not preclude high-risk patients from receiving DOACs, but recognition of risk of GIB in order to take preventive measures or close monitoring and finally a benefit versus risk ratio discussion is of utmost importance [25]. The use of concomitant medications which increase the risk of bleeding in patients on DOACs and should be reviewed include NSAIDs, SSRIs or SNRIs, and norepinephrine. Studies regarding the reinstitution of DOACs in patients with ICH recommend holding anticoagulation for at least 4 weeks [26]. This article aims not only to highlight the various types of DOACs, their mechanisms of action, their indications, and advantages but also sheds light on the risks and disadvantages associated with them relative to other anticoagulant medications.

Pharmacokinetics and pharmacodynamics of DOACs

The common pathway of the coagulation cascade starts with factor Xa. Factor Xa is trypsin like serine protease, and is essential for conversion of prothrombin to thrombin, which in turn is needed for clot formation. Thrombin is an enzyme which activates platelets and catalyzes the conversion of fibrinogen to fibrin (Fig. 1). Antithrombin III is a serine protease inhibitor and functions as a natural anticoagulant, mainly inhibiting activity of factor Xa and II [27]. In an attempt to overcome the nuances associated with warfarin use, DOACs like apixaban, rivaroxaban, edoxaban, and dabigatran were developed, and it was hypothesized that inhibition of factor Xa or thrombin directly can prevent clot formation as shown in Fig. 1 below [28].

Fig. 1
figure 1

Simplified coagulation cascade. *Apixaban, Rivaroxaban, Edoxaban. **Dabigatran

Apixaban is a neutral bicyclic pyrazole with a molecular weight of 459.5 g/mol. It inhibits platelet activation and clot formation via direct, selective, and reversible inhibition of free as well as clot-bound factor Xa [29]. Additionally, antithrombin III is not needed for its antithrombotic activity [30]. Apixaban has no direct effect on platelet aggregation but by inhibiting thrombin activity, it indirectly inhibits thrombin-induced platelet aggregation [31]. Given that it has a half-life of approximately 8–15 h, and it takes 5 half-lives for anticoagulation to resolve, the timing of last dose can be used as a rough estimate to gauge the anticoagulation effect.

Rivaroxaban competitively inhibits free factor Xa, factor Xa bound to prothrombinase as well as factor Xa bound to clot [32], thus inhibiting thrombin and clot formation. Similar to apixaban, it has no direct effect on platelet aggregation and antithrombin III is not required to exert antithrombotic effects [33]. It has a half-life of 5–9 h and anticoagulation usually resolves 1–2 days after the last dose.

Edoxaban is a selective, competitive, dose-dependent, and direct inhibitor of free as well as prothrombinase complex bound factor Xa with a rapid onset of action around 1.5 h [34]. Similar to apixaban and rivaroxaban, it also inhibits thrombin-induced platelet aggregation.

Dabigatran is a direct and reversible thrombin inhibitor. It competitively and selectively binds to the active site of free, clot bound as well as fibrin-bound thrombin, thus inhibiting the conversion of soluble fibrinogen to insoluble fibrin [35]. Additionally, by binding to thrombin, dabigatran inhibits thrombin-induced platelet aggregation [36]. Dabigatran etexilate is a prodrug and is metabolized into the active compound dabigatran by enzymes in the plasma and liver [37]. It is a fast-acting drug with peak plasma concentration achieved in 1–2 h with a half-life of 12–17 h in comparison to the half-life of the other DOACs as mentioned in Table 2. Timing of the last dose aids to gauge the effect of anticoagulation as it is usually resolved after 5 half-lives. In patients with normal renal function, the following considerations apply [38].

Table 2 Pharmacokinetics of DOACs

For apixaban, rivaroxaban, and edoxaban, anti-factor Xa levels, though not commonly available, can be used to assess anticoagulation effects [39]. As the usage of DOACs is increasing, so are the life-threatening bleeds associated with them, it is imperative to be familiar with the current guidelines for the reversal of DOACs to prevent any major bleeds.

Treatment and role of DOAC

DOACs like dabigatran, rivaroxaban, apixaban, and edoxaban are used to lower stroke and embolism risk in NVAF, DVT, and PE treatments. Betrixaban is used for VTE prophylaxis in adults hospitalized for acute illnesses [15, 40]. Some limitations of DOAC include higher cost, shorter half-life, and limited specific reversal agents and monitoring tests. The cost of DOACs has increased from 200 to 400 USD from 2011 to 2020 [41]. DOAC reversal is indicated in life-threatening bleeds, trauma, emergency surgeries, or invasive procedures. Reversal can be accomplished by drug removal, non-specific reversal agents, and specific reversal agents. Currently, Idarucizumab and Andexanet are the only FDA approved specific reversal agents for DOACs. Other off-label management options include clotting factors such as prothrombin complex concentrate (PCC), activated platelet complex concentrate (aPCC), and recombinant factor VIIa [40]. Four factors’ PCC, also known as Kcentra, contains inactive forms of 4 coagulation factors II, VII, IX and X along with protein C and S, and has been studied extensively in vivo in humans. Randomized controlled trials have shown that a maximum dose of 50 U/kg can be used for emergency reversal of anticoagulation with factor Xa inhibitors [42, 43].

Idarucizumab

Idarucizumab is a humanized monoclonal antigen fragment, first approved in 2015, and is the only reversal agent that binds with direct thrombin inhibitor dabigatran to inhibit its effect [44]. The recommended dose of Idarucizumab is 5 g [45] and the cost for two 2.5 g/50 mL vials is approximately $3662 [46].

Idarucizumab has a high affinity for dabigatran, which is 350 times greater than thrombin. It does not bind thrombin substrates and thus does not enhance thrombin-mediated feedback of coagulation [47]. The initial half-life is 45 min and depending on the dose some amounts can be detected in the blood after 24 h [48]. Idarucizumab reversed the effects of dabigatran in the Reversal Effects of Idarucizumab on Active Dabigatran (RE-VERSE AD) study [49]. This study looked at the effects of 5 g of IV Idarucizumab in patients that required dabigatran reversal. Idarucizumab’s effect lasted for 24 h in most patients. Group A (n = 301) consisted of individuals who had uncontrolled bleeding (45.5% GI bleed and 32.6% intracranial hemorrhage). Group B (n = 202) consisted of individuals that required urgent surgery or an invasive procedure that could not be delayed by more than 8 h. dTT or ECT tests were used, and they showed a 100% median maximum reversal of dabigatran within 4 h after the idarucizumab use. Individuals assessed in group A took 2.5 h (median time) to stop their respective bleedings. In group B, the individuals had their procedures 1.6 h (median time) after the first infusion of idarucizumab. Additionally, the study showed that reversal was quick and occurred independently of age, sex, renal function, and dabigatran concentration at baseline. There were some side effects of the drug. At 90 days thrombotic events occurred in 6.8% of patients. The mortality rate was similar in both groups, 18.8% in group A and 18.9% in group B [49].

Andexanet

Andexanet is a modified recombinant inactive form of factor Xa protein. It binds specifically to and sequesters factor Xa inhibitors (rivaroxaban, apixaban, and betrixaban) to rapidly reverse their effect in a dose-dependent manner. It has a half-life of 1 h [50]. It was approved in 2018 for the reversal of apixaban and rivaroxaban in patients with uncontrolled or life-threatening bleeding or in the event of emergent surgeries [51].

Andexanet showed a reversal of apixaban and rivaroxaban in healthy individuals (mean age 57.9 years old) in the ANNEXA-A and ANNEXA-R trials. The participants in the ANNEXA-A trial received 5 mg of apixaban twice a day (n = 48) and the participants in the ANNEXA-R trial received 20 mg of rivaroxaban once a day (n = 53). This was a randomized, double-blind, placebo-controlled trial. All the patients were divided into groups that received either Andexanet or a placebo. Both trials showed that a bolus of Andexanet, with or without the addition of a 2-h infusion, reduced anti–factor Xa activity more than the placebo. Andexanet's effect lasted for 2 h with the bolus and continued for a longer period of time with the addition of the 2-h infusion. Additionally, Andexanet also restored thrombin generation within 2–5 min of treatment [52].

ANNEXA-4 looked at the effects of Andexanet bolus followed by 2-h infusion on patients (n = 352, mean age of 77 years old) who had major bleeding events within 18 h after taking apixaban, rivaroxaban, edoxaban, or enoxaparin. Most patients in this trial had cardiovascular disease and the bleeding was mostly intracranial (n = 227) or gastrointestinal (n = 90). In patients who had received apixaban and rivaroxaban, the median anti–factor Xa activity decreased 92% after the Andexanet bolus. In the 249 patients evaluated for hemostasis status, 82% (n = 204) had excellent or good hemostasis after 12 h of administration. At 30 days, 10% (n = 34) had a thrombotic event and 14% (= 49) died. This study did not show any relationship between reduction in anti-factor Xa activity and hemostatic efficacy overall [53]. The dose studied for off-label use in patients to reverse the effect of edoxaban or betrixaban is 800 mg IV bolus administered at a rate of 30 mg/min, followed by an IV infusion of 8 mg/min for up to 120 min. For patients who received apixaban or rivaroxaban, the dose for reversal of the anticoagulant effects may require either a low dose which is 400 mg IV bolus at a rate of 30 mg/min followed by 4 mg/min, or a high dose which is 800 mg IV bolus followed by an IV infusion of 8 mg/min. If the drug administration was more than 8 h or ≤ 5 mg for apixaban and ≤ 10 mg for rivaroxaban within an 8-h window, a low dose of andexanet is indicated. Administration of > 5 mg of apixaban or > 10 mg of rivaroxaban within the 8-h period is an indication for reversal with a high dose of andexanet [54].

New reversal agent in development

Ciraparantag (PER977) is a small, synthetic, water-soluble molecule that can reverse the anticoagulant effects of low-molecular-weight heparin, unfractionated heparin, and DOACs. It acts by inactivating heparin and DOAC through noncovalent hydrogen binding and charge interaction. This promising reversal agent has undergone phase 1 and 2 trials which have shown reversals of DOACs in healthy individuals. Additionally, Ciraparantag (100–300) mg IV injectable dosage appears safe and well tolerated [55]. In the ongoing trials, the only drug-related adverse effects reported are periorbital and facial flushing which occurred in 74% of the patients who were administered Ciraparantag, and no serious thrombotic episodes or death have been reported [56].

However, more prospective studies need to be conducted to test the use of these drugs in patients with acute indications like major bleeds or emergency surgeries [56].

Post reversal complications

Some complications that may present after reversal include thrombotic events and death. These complications were seen in a small percentage of individuals in the ANNEXA-4 and REVERSE AD trials. There is also the risk of incomplete reversal. Overall, Andexanet and Idarucizumab have been shown to be mostly well-tolerated [49, 53]. They are generally safe to use and their ability to rapidly reverse the effects of DOAC allows for better management of major bleeding episodes.

Conclusion

The introduction of DOACs into clinical medicine and subsequent approval by the FDA in 2010 has proven to be greatly beneficial to the progression of anticoagulation medication. They are preferable over other anticoagulant medications as they present a lower risk of bleeding, ability to act rapidly from the time of initiation, lack of frequent monitoring, and have predictive pharmacokinetic properties. They can be used for both prophylactic and therapeutic treatment of thromboembolic events. DOACs are not exempt from their share of limitations with the most striking concern with these drugs being the potential risk of causing life-threatening bleeding conditions such as ICH and GIB. This results in the need for immediate removal of the drug through hemodialysis or administration of reversal agents. Idarucizumab and andexanet are specific reversal agents that bind to dabigatran and factor Xa to cause inhibitory effects, respectively. The shorter half-life and higher cost of DOACs relative to warfarin also prove disadvantageous. Overall, the use of DOACs has significantly contributed to the advancement of anticoagulation therapy and eventual patient well-being, having an ultimately net positive effect despite their limitations.

Availability of data and materials

Not applicable.

Abbreviations

AF:

Atrial fibrillation

ANNEXA-4:

A Novel Antidote to the Anticoagulation Effects of Factor Xa Inhibitors

ANNEXA-A:

A Novel Antidote to the Anticoagulation Effects of Factor Xa Inhibitors-Apixaban

ANNEXA-R:

A Novel Antidote to the Anticoagulation Effects of Factor Xa Inhibitors—Rivaroxaban

aPCC:

Activated platelet complex concentrate

CrCl:

Creatinine clearance

DOAC:

Direct oral anticoagulant

dTT:

Diluted thrombin time

DVT:

Deep venous thrombosis

ECT:

Ecarin clotting time

ENAVLE:

Explore the Efficacy and Safety of Edoxaban in Patients After Heart Valve Repair or Bioprosthetic Valve Replacement

FDA:

Food and Drug Administration

GI:

Gastrointestinal

GIB:

Gastrointestinal bleeding

HIT:

Heparin induced thrombocytopenia

ICH:

Intracranial hemorrhage

LMWH:

Low molecular weight heparin

LV:

Left ventricular

NSAID:

Non-steroidal anti-inflammatory drug

NVAF:

Non-valvular atrial fibrillation

PER977:

Ciraparantag

PCC:

Prothrombin complex concentrate

PCI:

Percutaneous coronary intervention

PE:

Pulmonary embolism

rFVIIa:

Recombinant factor VIIa

RE-ALIGN:

A Randomised, Phase II Study to Evaluate the Safety and Pharmacokinetics of Oral Dabigatran Etexilate in Patients After Heart Valve Replacement

RE-VERSE AD:

Reversal Effects of Idarucizumab on Active Dabigatran

SNRI:

Serotonin and norepinephrine reuptake inhibitor

SSRI:

Selective serotonin reuptake inhibitor

USD:

United States dollar

VTE:

Venous thromboembolism

References

  1. Franchini M, Liumbruno GM, Bonfanti C, Lippi G (2016) The evolution of anticoagulant therapy. Blood Transfus 14(2):175–84. https://doi.org/10.2450/2015.0096-15. Epub 2015 Dec 16. PMID: 26710352; PMCID: PMC4781787

    Article  PubMed  PubMed Central  Google Scholar 

  2. Lee LH (2016) DOACs - advances and limitations in real world. Thromb J 14(Suppl 1):17. https://doi.org/10.1186/s12959-016-0111-3. PMID:27766043;PMCID:PMC5056491

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kustos SA, Fasinu PS (2019) Direct-acting oral anticoagulants and their reversal agents-an update. Medicines (Basel) 6(4):103. https://doi.org/10.3390/medicines6040103. PMID:31618893;PMCID:PMC6963825

    Article  CAS  PubMed  Google Scholar 

  4. Grześk G, Rogowicz D, Wołowiec Ł, Ratajczak A, Gilewski W, Chudzińska M, Sinkiewicz A, Banach J (2021) The clinical significance of drug-food interactions of direct oral anticoagulants. Int J Mol Sci 22(16):8531. https://doi.org/10.3390/ijms22168531. PMID:34445237;PMCID:PMC8395160

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hirsh J (1991) Oral anticoagulant drugs. N Engl J Med 324(26):1865–1875. https://doi.org/10.1056/NEJM199106273242606. PMID: 1801769

    Article  CAS  PubMed  Google Scholar 

  6. Gunasekaran K, Rajasurya V, Devasahayam J, Singh Rahi M, Chandran A, Elango K, Talari G (2020) A review of the incidence diagnosis and treatment of spontaneous hemorrhage in patients treated with direct oral anticoagulants. J Clin Med 9(9):2984. https://doi.org/10.3390/jcm9092984. PMID:32942757;PMCID:PMC7563837

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cheng JW, Barillari G (2014) Non-vitamin K antagonist oral anticoagulants in cardiovascular disease management: evidence and unanswered questions. J Clin Pharm Ther 39(2):118–135. https://doi.org/10.1111/jcpt.12122. Epub 2014 Jan 3 PMID: 24383983

    Article  CAS  PubMed  Google Scholar 

  8. da Silva Ferreira H, Lima Lopes J, Augusto J, Simões J, Roque D, Faria D, Ferreira J, Fialho I, Beringuilho M, Morais H, Ferreira AR, Morais J, Morais C (2023) Effect of direct oral anticoagulants versus vitamin K antagonists or warfarin in patients with left ventricular thrombus outcomes: a systematic review and meta-analysis. Rev Port Cardiol 42(1):63–70. English, Portuguese. https://doi.org/10.1016/j.repc.2021.11.013. Epub 2022 Nov 9. PMID: 36370988

  9. Stacy ZA, Richter SK (2018) Direct oral anticoagulants for stroke prevention in atrial fibrillation: treatment outcomes and dosing in special populations. Ther Adv Cardiovasc Dis 12(9):247–262. https://doi.org/10.1177/1753944718787384. PMID:30081727;PMCID:PMC6116754

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhang Z, Si D, Zhang Q, Jin L, Zheng H, Qu M, Yu M, Jiang Z, Li D, Li S, Yang P, He Y, Zhang W (2022) Prophylactic rivaroxaban therapy for left ventricular thrombus after anterior ST-segment elevation myocardial infarction. JACC Cardiovasc Interv 15(8):861–872. https://doi.org/10.1016/j.jcin.2022.01.285. Epub 2022 Mar 30 PMID: 35367170

    Article  PubMed  Google Scholar 

  11. Eikelboom JW, Connolly SJ, Brueckmann M, Granger CB, Kappetein AP, Mack MJ, Blatchford J, Devenny K, Friedman J, Guiver K, Harper R, Khder Y, Lobmeyer MT, Maas H, Voigt JU, Simoons ML, Van de Werf F, RE-ALIGN Investigators (2013) Dabigatran versus warfarin in patients with mechanical heart valves. N Engl J Med 369(13):1206–14. https://doi.org/10.1056/NEJMoa1300615. Epub 2013 Aug 31. PMID: 23991661

  12. Khider L, Gendron N, Mauge L (2022) Inherited thrombophilia in the era of direct oral anticoagulants. Int J Mol Sci 23(3):1821. https://doi.org/10.3390/ijms23031821. PMID:35163742;PMCID:PMC8837096

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tyselskyi V, Tryliskyy Y, Poylin V, Kebkalo A (2023) Prolonged thromboprophylaxis with rivaroxaban after bariatric interventions: single centre experience. Pol Przegl Chir 95(4):1–5. https://doi.org/10.5604/01.3001.0016.2729. PMID: 36808057

    Article  PubMed  Google Scholar 

  14. Shim CY, Seo J, Kim YJ, Lee SH, De Caterina R, Lee S, Hong GR (2021) Explore the efficacy and safety of edoxaban in patients after heart valve repair or bioprosthetic valve replacement (ENAVLE) study group. Efficacy and safety of edoxaban in patients early after surgical bioprosthetic valve implantation or valve repair: A randomized clinical trial. J Thorac Cardiovasc Surg S0022–5223(21)00228–2. https://doi.org/10.1016/j.jtcvs.2021.01.127. Epub ahead of print. PMID: 33726903

  15. Chen A, Stecker E, A Warden B (2020) Direct oral anticoagulant use: a practical guide to common clinical challenges. J Am Heart Assoc 9(13):e017559. https://doi.org/10.1161/JAHA.120.017559. Epub 2020 Jun 15. PMID: 32538234; PMCID: PMC7670541

  16. McBane RD 2nd, Wysokinski WE, Le-Rademacher JG, Zemla T, Ashrani A, Tafur A, Perepu U, Anderson D, Gundabolu K, Kuzma C, Perez Botero J, Leon Ferre RA, Henkin S, Lenz CJ, Houghton DE, Vishnu P, Loprinzi CL (2020) Apixaban and dalteparin in active malignancy-associated venous thromboembolism: the ADAM VTE trial. J Thromb Haemost 18(2):411–421. https://doi.org/10.1111/jth.14662. Epub 2019 Nov 28 PMID: 31630479

    Article  CAS  PubMed  Google Scholar 

  17. Deitelzweig S, Keshishian A, Kang A, Jenkins A, Atreja N, Schuler P, Jiang J, Yuce H, Sun X, Lip GYH (2023) Delaying clinical events among patients with non-valvular atrial fibrillation treated with oral anticoagulants: Insights from the ARISTOPHANES study. Eur J Intern Med 108:37–42. https://doi.org/10.1016/j.ejim.2022.10.021. Epub 2022 Nov 28 PMID: 36456387

    Article  CAS  PubMed  Google Scholar 

  18. Wiethorn EE, Bell CM, Wiggins BS (2021) Effectiveness and safety of direct oral anticoagulants in patients with nonvalvular atrial fibrillation and weighing ≥ 120 kilograms versus 60–120 kilograms. Am J Cardiovasc Drugs 21(5):545–551. https://doi.org/10.1007/s40256-021-00470-0. Epub 2021 Mar 30 PMID: 33782909

    Article  CAS  PubMed  Google Scholar 

  19. Zhao Y, Guo M, Li D, Xu W, Pan C, He C, Cui X (2023) Pharmacokinetics and dosing regimens of direct oral anticoagulants in morbidly obese patients: an updated literature review. Clin Appl Thromb Hemost 29:10760296231153638. https://doi.org/10.1177/10760296231153638. PMID: 36760080; PMCID: PMC9943962

  20. Ballestri S, Romagnoli E, Arioli D, Coluccio V, Marrazzo A, Athanasiou A, Di Girolamo M, Cappi C, Marietta M, Capitelli M (2023) Risk and management of bleeding complications with direct oral anticoagulants in patients with atrial fibrillation and venous thromboembolism: a narrative review. Adv Ther 40(1):41–66. https://doi.org/10.1007/s12325-022-02333-9. Epub 2022 Oct 16. PMID: 36244055; PMCID: PMC9569921

    Article  PubMed  Google Scholar 

  21. Koehl JL, Hayes BD, Al-Samkari H, Rosovsky R (2020) A comprehensive evaluation of apixaban in the treatment of venous thromboembolism. Expert Rev Hematol 13(2):155–173. https://doi.org/10.1080/17474086.2020.1711731. Epub 2020 Jan 23 PMID: 31958251

    Article  CAS  PubMed  Google Scholar 

  22. Agnelli G, Becattini C, Meyer G, Muñoz A, Huisman MV, Connors JM, Cohen A, Bauersachs R, Brenner B, Torbicki A, Sueiro MR, Lambert C, Gussoni G, Campanini M, Fontanella A, Vescovo G, Verso M, Caravaggio Investigators (2020) Apixaban for the treatment of venous thromboembolism associated with cancer. N Engl J Med 382(17):1599-1607. https://doi.org/10.1056/NEJMoa1915103. Epub 2020 Mar 29. PMID: 32223112

  23. Kushnir M, Gali R, Alexander M, Billett HH (2023) Direct oral Xa inhibitors for the treatment of venous thromboembolism after bariatric surgery. Blood Adv 7(2):224–226. https://doi.org/10.1182/bloodadvances.2021006696. PMID:35640233;PMCID:PMC9841032

    Article  CAS  PubMed  Google Scholar 

  24. Chai-Adisaksopha C, Crowther M, Isayama T, Lim W (2014) The impact of bleeding complications in patients receiving target-specific oral anticoagulants: a systematic review and meta-analysis. Blood 124(15):2450–2458. https://doi.org/10.1182/blood-2014-07-590323. Epub 2014 Aug 22 PMID: 25150296

    Article  CAS  PubMed  Google Scholar 

  25. Cheung KS, Leung WK (2017) Gastrointestinal bleeding in patients on novel oral anticoagulants: Risk, prevention and management. World J Gastroenterol 23(11):1954–1963. https://doi.org/10.3748/wjg.v23.i11.1954. PMID:28373761;PMCID:PMC5360636

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hemphill JC 3rd, Greenberg SM, Anderson CS, Becker K, Bendok BR, Cushman M, Fung GL, Goldstein JN, Macdonald RL, Mitchell PH, Scott PA, Selim MH, Woo D, American Heart Association Stroke Council; Council on Cardiovascular and Stroke Nursing; Council on Clinical Cardiology (2015) Guidelines for the management of spontaneous intracerebral hemorrhage: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 46(7):2032–60. https://doi.org/10.1161/STR.0000000000000069. Epub 2015 May 28. PMID: 26022637

  27. Loncar R, Kalina U, Stoldt V, Thomas V, Scharf RE, Vodovnik A (2006) Antithrombin significantly influences platelet adhesion onto immobilized fibrinogen in an in-vitro system simulating low flow. Thromb J 13(4):19. https://doi.org/10.1186/1477-9560-4-19. PMID:17040572;PMCID:PMC1618384

    Article  CAS  Google Scholar 

  28. Walenga JM, Jeske WP, Hoppensteadt D, Fareed J (2003) Factor Xa inhibitors: today and beyond. Curr Opin Investig Drugs 4(3):272–281 PMID: 12735228

    CAS  PubMed  Google Scholar 

  29. Luettgen JM, Knabb RM, He K, Pinto DJ, Rendina AR (2011) Apixaban inhibition of factor Xa: Microscopic rate constants and inhibition mechanism in purified protein systems and in human plasma. J Enzyme Inhib Med Chem 26(4):514–526. https://doi.org/10.3109/14756366.2010.535793. Epub 2010 Dec 20 PMID: 21171894

    Article  CAS  PubMed  Google Scholar 

  30. Ansell J (2007) Factor Xa or thrombin: is factor Xa a better target? J Thromb Haemost 5(Suppl 1):60–64. https://doi.org/10.1111/j.1538-7836.2007.02473.x. PMID: 17635710

    Article  CAS  PubMed  Google Scholar 

  31. Wong PC, Pinto DJ, Zhang D (2011) Preclinical discovery of apixaban, a direct and orally bioavailable factor Xa inhibitor. J Thromb Thrombolysis 31(4):478–492. https://doi.org/10.1007/s11239-011-0551-3. PMID:21318583;PMCID:PMC3090580

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Perzborn E, Roehrig S, Straub A, Kubitza D, Mueck W, Laux V (2010) Rivaroxaban: a new oral factor Xa inhibitor. Arterioscler Thromb Vasc Biol 30(3):376–381. https://doi.org/10.1161/ATVBAHA.110.202978. Epub 2010 Feb 5 PMID: 20139357

    Article  CAS  PubMed  Google Scholar 

  33. Kubitza D, Becka M, Voith B, Zuehlsdorf M, Wensing G (2005) Safety, pharmacodynamics, and pharmacokinetics of single doses of BAY 59–7939, an oral, direct factor Xa inhibitor. Clin Pharmacol Ther 78(4):412–421. https://doi.org/10.1016/j.clpt.2005.06.011. PMID: 16198660

    Article  CAS  PubMed  Google Scholar 

  34. Plitt A, Giugliano RP (2014) Edoxaban: review of pharmacology and key phase I to III clinical trials. J Cardiovasc Pharmacol Ther 19(5):409–416. https://doi.org/10.1177/1074248414523675. Epub 2014 Mar 7 PMID: 24607764

    Article  CAS  PubMed  Google Scholar 

  35. Ganetsky M, Babu KM, Salhanick SD, Brown RS, Boyer EW (2011) Dabigatran: review of pharmacology and management of bleeding complications of this novel oral anticoagulant. J Med Toxicol 7(4):281–287. https://doi.org/10.1007/s13181-011-0178-y. PMID:21887485;PMCID:PMC3550194

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Xiao Z, Théroux P (1998) Platelet activation with unfractionated heparin at therapeutic concentrations and comparisons with a low-molecular-weight heparin and with a direct thrombin inhibitor. Circulation 97(3):251–256. https://doi.org/10.1161/01.cir.97.3.251. PMID: 9462526

    Article  CAS  PubMed  Google Scholar 

  37. Stangier J, Rathgen K, Stähle H, Gansser D, Roth W (2007) The pharmacokinetics, pharmacodynamics and tolerability of dabigatran etexilate, a new oral direct thrombin inhibitor, in healthy male subjects. Br J Clin Pharmacol 64(3):292–303. https://doi.org/10.1111/j.1365-2125.2007.02899.x. Epub 2007 May 15. PMID: 17506785; PMCID: PMC2000643

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Scaglione F (2013) New oral anticoagulants: comparative pharmacology with vitamin K antagonists. Clin Pharmacokinet 52(2):69–82. https://doi.org/10.1007/s40262-012-0030-9. PMID: 23292752

    Article  CAS  PubMed  Google Scholar 

  39. Samuelson BT, Cuker A, Siegal DM, Crowther M, Garcia DA (2017) Laboratory assessment of the anticoagulant activity of direct oral anticoagulants: a systematic review. Chest 151(1):127–138. https://doi.org/10.1016/j.chest.2016.08.1462. Epub 2016 Sep 13. PMID: 27637548; PMCID: PMC5310120

    Article  PubMed  Google Scholar 

  40. Desai NR, Cornutt D (2019) Reversal agents for direct oral anticoagulants: considerations for hospital physicians and intensivists. Hosp Pract 47(3):113–122. https://doi.org/10.1080/21548331.2019.1643728

    Article  Google Scholar 

  41. Costa LS, Alsultan MM, Hincapie AL et al (2022) Trends in utilization, reimbursement, and price for DOACs and warfarin in the US Medicaid population from 2000 to 2020. J Thromb Thrombolysis  https://doi.org/10.1007/s11239-022-02727-0

  42. Zahir H, Brown KS, Vandell AG, Desai M, Maa JF, Dishy V, Lomeli B, Feussner A, Feng W, He L, Grosso MA, Lanz HJ, Antman EM (2015) Edoxaban effects on bleeding following punch biopsy and reversal by a 4-factor prothrombin complex concentrate. Circulation 131(1):82–90. https://doi.org/10.1161/CIRCULATIONAHA.114.013445

    Article  CAS  PubMed  Google Scholar 

  43. Tomaselli GF, Mahaffey KW, Cuker A, Dobesh PP, Doherty JU, Eikelboom JW, Florido R, Hucker W, Mehran R, Messé SR, Pollack CV Jr, Rodriguez F, Sarode R, Siegal D, Wiggins BS (2017) 2017 ACC expert consensus decision pathway on management of bleeding in patients on oral anticoagulants: a report of the American college of cardiology task force on expert consensus decision pathways. J Am Coll Cardiol 70(24):3042–3067. https://doi.org/10.1016/j.jacc.2017.09.1085

    Article  PubMed  Google Scholar 

  44. Yogaratnam D, Ditch K, Medeiros K, Doyno C, Fong JJ (2016) Idarucizumab for reversal of dabigatran-associated anticoagulation. Ann Pharmacother 50(10):847–854. https://doi.org/10.1177/1060028016659504. Epub 2016 Jul 7 PMID: 27389324

    Article  CAS  PubMed  Google Scholar 

  45. Jarrett JB, Gimbar RP (2017) Idarucizumab (Praxbind) for dabigatran (Pradaxa) anticoagulant Reversal. Am Fam Physician 95:798–800

    PubMed  Google Scholar 

  46. Boehringer Ingelheim. FDA provides full approval to Praxbind, specific reversal agent for Pradaxa. www.boehringer-ingelheim.us/press-release/fda-provides-full-approval-praxbind-specific-reversal-agent-pradaxa

  47. Schiele F, van Ryn J, Canada K et al (2013) A specific antidote for dabigatran: functional and structural characterization. Blood 121(18):3554–3562

    Article  CAS  PubMed  Google Scholar 

  48. Glund S, Moschetti V, Norris S et al (2015) A randomised study in healthy volunteers to investigate the safety, tolerability and pharmacokinetics of idarucizumab, a specific antidote to dabigatran. Thromb Haemost 113:943–951

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  50. Hafer A, McCann L (2021) Direct oral anticoagulant reversal: An update. Nursing 51(6):54–64. https://doi.org/10.1097/01.NURSE.0000743104.69943.67. PMID: 34014879

    Article  PubMed  Google Scholar 

  51. Lu G, DeGuzman FR, Hollenbach SJ et al (2013) A specific antidote for reversal of anticoagulation by direct and indirect inhibitors of coagulation factor Xa. Nat Med 19:446–451

    Article  CAS  PubMed  Google Scholar 

  52. Siegal DM, Curnutte JT, Connolly SJ et al (2015) Andexanet alfa for the reversal of factor Xa inhibitor activity. N Engl J Med 373(25):2413–2424

    Article  CAS  PubMed  Google Scholar 

  53. Connolly SJ, Crowther M, Eikelboom JW, ANNEXA-4 Investigators et al (2019) Full study report of andexanet alfa for bleeding associated with factor Xa inhibitors. N Engl J Med. 380(14):1326–1335

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Tomaselli GF, Mahaffey KW, Cuker A, Dobesh PP, Doherty JU, Eikelboom JW, Florido R, Gluckman TJ, Hucker WJ, Mehran R, Messé SR, Perino AC, Rodriguez F, Sarode R, Siegal DM, Wiggins BS (2020) 2020 ACC expert consensus decision pathway on management of bleeding in patients on oral anticoagulants: a report of the american college of cardiology solution set oversight committee. J Am Coll Cardiol 76(5):594–622. https://doi.org/10.1016/j.jacc.2020.04.053. Epub 2020 Jul 14. Erratum in: J Am Coll Cardiol. 2021 Jun 1;77(21):2760. PMID: 32680646

    Article  CAS  PubMed  Google Scholar 

  55. Ansell JE, Bakhru SH, Laulicht BE, Steiner SS, Grosso MA, Brown K, Dishy V, Lanz HJ, Mercuri MF, Noveck RJ, Costin JC (2017) Single-dose ciraparantag safely and completely reverses anticoagulant effects of edoxaban. Thromb Haemost 117(2):238–245. https://doi.org/10.1160/TH16-03-0224. Epub 2016 Nov 17. PMID: 27853809; PMCID: PMC6260118

    Article  PubMed  PubMed Central  Google Scholar 

  56. Leentjens J, Middeldorp S, Jung C (2022) A short review of ciraparantag in perspective of the currently available anticoagulant reversal agents. Drug Discov Today 27(10):103332. https://doi.org/10.1016/j.drudis.2022.07.017. Epub 2022 Aug 3. PMID: 35933085

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

No funding was obtained.

Author information

Authors and Affiliations

Authors

Contributions

The information on DOAC reversibility has been contributed by Sharon Wei, the introduction part has been written by Aanchal Sawhney, the pharmacokinetics has been written by Harshwardhan Khandait, the abstract and conclusion have been contributed by Amit Medha, figures and tables have been conceptualized by Vasu Gupta, and conceptualization of the idea and formatting has been done by Dr. Rohit Jain. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Aanchal Sawhney.

Ethics declarations

Ethics approval and consent to participate

No ethics approval needed.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wei, S., Sawhney, A., Khandait, H. et al. An update on applications and limitations of direct oral anticoagulants. Egypt J Intern Med 35, 26 (2023). https://doi.org/10.1186/s43162-023-00212-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43162-023-00212-5

Keywords