View Article

  • Novel Oral Anticoagulants in Modern Anticoagulation Therapy: A Clinical and Pharmacological Review

  • Krupanidhi College of Pharmacy, Bangalore, 560035

Abstract

Background: Novel Oral Anticoagulants (NOACs), including dabigatran, rivaroxaban, apixaban, and edoxaban, have redefined anticoagulation therapy by addressing the limitations of vitamin K antagonists (VKAs), such as frequent monitoring, dietary restrictions, and drug interactions. Objective: This review provides a comprehensive evaluation of the clinical efficacy, pharmacological characteristics, safety profiles, and management strategies associated with NOACs, emphasizing their role in contemporary anticoagulation practice. Methods: Relevant data from pivotal randomized controlled trials, meta-analyses, and guideline recommendations were analyzed, including RE-LY, ROCKET-AF, ARISTOTLE, and ENGAGE AF-TIMI 48 studies. Results: NOACs demonstrate predictable pharmacokinetics, rapid onset, short half-life, and minimal food or drug interactions, eliminating the need for routine coagulation monitoring. Clinical trials consistently confirm their non-inferiority or superiority to warfarin in preventing thromboembolic events, with a notably reduced risk of intracranial hemorrhage. The development of specific reversal agents—idarucizumab for dabigatran and andexanet alfa for factor Xa inhibitors—has enhanced their safety in bleeding emergencies. Ongoing studies are expanding their indications to cancer-associated thrombosis, post-TAVI management, and peripheral artery disease. Conclusion: NOACs represent a paradigm shift in anticoagulation therapy by merging efficacy, safety, and convenience. Their integration into international guidelines underscores their clinical relevance. Future research is focused on personalized dosing, pharmacogenomic optimization, and novel agents targeting upstream coagulation pathways to achieve greater precision and safety in anticoagulation management.

Keywords

Novel Oral Anticoagulants, Dabigatran, Rivaroxaban, Apixaban, Edoxaban, Anticoagulation, Pharmacology, Clinical Efficacy

Introduction

Overview of thrombosis and anticoagulation therapy

Thrombosis — the pathological formation of blood clots within the vasculature — remains a major contributor to global morbidity and mortality, underlying conditions such as venous thromboembolism (VTE), deep vein thrombosis (DVT), pulmonary embolism (PE), and atrial fibrillation (AF)-related stroke¹. The pathophysiology of thrombosis involves an imbalance in Virchow’s triad—endothelial injury, hypercoagulability, and stasis—leading to inappropriate activation of the coagulation cascade and fibrin clot formation².

Traditional anticoagulation therapy has long relied on vitamin K antagonists (VKAs) like warfarin, which, despite proven efficacy, require frequent monitoring and dose adjustments due to variable pharmacokinetics and drug–food interactions³. The limitations of VKAs, including their narrow therapeutic index and unpredictable response, have spurred the development of Novel Oral Anticoagulants (NOACs), also termed Direct Oral Anticoagulants (DOACs), designed to target specific coagulation factors with greater precision?.

NOACs—such as dabigatran (a direct thrombin inhibitor), and rivaroxaban, apixaban, and edoxaban (Factor Xa inhibitors)—have transformed modern anticoagulation management by offering predictable pharmacodynamics, rapid onset, and reduced need for laboratory monitoring?. They have shown non-inferior or superior efficacy to warfarin in preventing thromboembolic events, with comparable or lower rates of major bleeding?.

Moreover, the advent of NOACs aligns with a broader evolution in evidence-based cardiovascular medicine, integrating pharmacogenomics, real-world evidence (RWE), and artificial intelligence (AI)-driven risk stratification to personalize anticoagulant therapy?. As thrombosis research continues to expand, NOACs stand as a paradigm shift from empirical anticoagulation toward a more mechanistically targeted and patient-tailored approach?.

Limitations of traditional anticoagulants (warfarin, heparin)

For decades, warfarin and heparin have served as the foundation of anticoagulant therapy for the prevention and treatment of thromboembolic disorders9. However, despite their proven clinical efficacy, both agents possess significant pharmacological and practical limitations that complicate long-term management and patient adherence10.

Warfarin, a vitamin K antagonist (VKA), exhibits a narrow therapeutic index and marked interpatient variability in dose–response relationships due to genetic polymorphisms in CYP2C9 and VKORC1 enzymes11. This necessitates frequent international normalized ratio (INR) monitoring and dose adjustments, often resulting in suboptimal anticoagulation control and increased risk of bleeding or thrombosis12. Additionally, warfarin’s extensive drug–drug and drug–food interactions—particularly with vitamin K–rich foods and medications such as antibiotics and antifungals—further constrain its clinical use13.

Heparin and its derivatives, including low molecular weight heparin (LMWH), are effective in acute thrombosis management but require parenteral administration, limiting convenience and patient compliance14. Heparin therapy is also complicated by heparin-induced thrombocytopenia (HIT), a serious immune-mediated adverse reaction characterized by paradoxical thrombosis despite anticoagulation15. The unpredictable pharmacokinetics of unfractionated heparin demand continuous monitoring of activated partial thromboplastin time (aPTT) and careful dose titration, which can burden both healthcare systems and patients16.

In contrast, the emergence of Novel Oral Anticoagulants (NOACs)—such as dabigatran, rivaroxaban, apixaban, and edoxaban—addresses many of these limitations through predictable pharmacodynamics, fixed dosing, and minimal monitoring requirements17. Consequently, NOACs represent a transformative step toward safer, more patient-centric anticoagulation therapy, effectively overcoming the major drawbacks of traditional VKAs and heparins18.

Emergence and growing role of novel oral anticoagulants (NOACs)

The past two decades have marked a paradigm shift in the management of thromboembolic disorders, driven by the introduction and clinical adoption of Novel Oral Anticoagulants (NOACs), also known as **Direct Oral Anticoagulants (DOACs)**19. These agents—including dabigatran (a direct thrombin inhibitor) and factor Xa inhibitors such as rivaroxaban, apixaban, and edoxaban—were developed to overcome the inherent limitations of traditional anticoagulants like warfarin and heparin, offering safer, more predictable, and patient-friendly options20.

The emergence of NOACs reflects a deepened understanding of the coagulation cascade and the central role of thrombin and factor Xa in clot formation21. By selectively targeting these enzymes, NOACs provide rapid onset of action, shorter half-lives, and more predictable pharmacokinetics, thus eliminating the need for routine coagulation monitoring22. Clinical trials such as RE-LY, ROCKET-AF, ARISTOTLE, and ENGAGE AF-TIMI 48 have demonstrated that NOACs are non-inferior or superior to warfarin in preventing stroke and systemic embolism in patients with atrial fibrillation, while reducing the incidence of intracranial hemorrhage23.

In addition to their efficacy, NOACs are now central to evidence-based anticoagulation guidelines globally. The European Society of Cardiology (ESC), American Heart Association (AHA), and CHEST guidelines recommend NOACs as first-line therapy for stroke prevention in non-valvular atrial fibrillation and treatment of venous thromboembolism24. Their adoption has expanded rapidly due to their ease of use, fixed dosing, and minimal drug–food interactions, significantly improving adherence and quality of life25.

Furthermore, NOACs have shown potential in real-world settings, where registries and Real-World Evidence (RWE) studies confirm their safety and effectiveness beyond controlled trials26. Ongoing research into reversal agents (e.g., idarucizumab for dabigatran, andexanet alfa for factor Xa inhibitors) has further strengthened clinical confidence in their use27. Collectively, these developments mark the NOAC era as a defining advancement in modern anticoagulation therapy, combining pharmacological precision with clinical practicality28

PURPOSE AND SCOPE OF THE REVIEW

The introduction of Novel Oral Anticoagulants (NOACs)—including dabigatran, rivaroxaban, apixaban, and edoxaban—has transformed the landscape of anticoagulation therapy by providing alternatives to traditional vitamin K antagonists and heparins29. As these agents become standard of care across diverse thromboembolic conditions, understanding their clinical pharmacology, comparative efficacy, safety profiles, and real-world implications is essential for optimizing patient outcomes30.

The purpose of this review is to provide a comprehensive synthesis of current evidence regarding the mechanisms, pharmacokinetics, clinical indications, and practical considerations surrounding NOACs. It aims to evaluate how these agents have reshaped modern thromboprophylaxis in conditions such as atrial fibrillation (AF), venous thromboembolism (VTE), and **pulmonary embolism (PE)**31. Special emphasis is placed on the transition from traditional anticoagulants—characterized by narrow therapeutic windows and complex monitoring—to the precision-based and patient-centered pharmacology of NOACs32.

The scope of this review encompasses both clinical trial evidence and real-world data (RWD) analyses, bridging gaps between controlled environments and daily clinical practice33. By integrating recent findings from regulatory, pharmacovigilance, and comparative effectiveness research, the paper seeks to elucidate not only the pharmacodynamic and pharmacokinetic characteristics of NOACs but also their role in evolving clinical guidelines34. Furthermore, it explores ongoing challenges—such as reversal strategies, renal dosing adjustments, and management in special populations—to inform safe and evidence-based use35.

Ultimately, this review serves as an interdisciplinary synthesis that merges pharmacological insight with clinical application, offering clinicians, pharmacists, and researchers an updated framework for employing NOACs in modern anticoagulation therapy36

EVOLUTION OF ORAL ANTICOAGULATION

Historical perspective of anticoagulant therapy

The development of anticoagulant therapy represents a century-long evolution in pharmacology, marked by significant milestones that transformed cardiovascular and hematologic medicine. The journey began in 1916, when Jay McLean and William Howell discovered heparin, a naturally occurring polysaccharide with anticoagulant properties, laying the groundwork for parenteral anticoagulation37. Heparin’s clinical application expanded throughout the mid-20th century, becoming essential in surgery, thrombosis management, and hemodialysis.

A major turning point came in the 1940s, with the synthesis of warfarin—a vitamin K antagonist (VKA) derived from dicoumarol, first identified as the toxic agent in spoiled sweet clover that caused fatal hemorrhages in cattle38. Approved for human use in 1954, warfarin quickly became the gold standard for long-term anticoagulation in conditions like atrial fibrillation (AF), deep vein thrombosis (DVT), and pulmonary embolism (PE)39.

Despite its clinical success, warfarin therapy posed numerous challenges: its narrow therapeutic index, genetic variability (CYP2C9 and VKORC1 polymorphisms), dietary vitamin K interactions, and requirement for regular INR monitoring40. These limitations fueled the search for safer, more predictable alternatives.

The early 21st century heralded the era of Novel Oral Anticoagulants (NOACs)—now referred to as Direct Oral Anticoagulants (DOACs)—including dabigatran, rivaroxaban, apixaban, and edoxaban. These agents selectively inhibit thrombin (factor IIa) or factor Xa, providing fixed dosing, rapid onset, and minimal need for monitoring41.

This transition from empirical natural extracts to targeted synthetic molecules reflects the maturation of anticoagulant therapy into a precision-based discipline, enabling more individualized and safer treatment for thromboembolic disease42.

Shift from Vitamin K Antagonists to Novel Oral Anticoagulants (NOACs)

The transition from Vitamin K antagonists (VKAs), such as warfarin and acenocoumarol, to Novel Oral Anticoagulants (NOACs) marks one of the most significant advancements in the history of anticoagulation therapy43. For decades, warfarin remained the cornerstone of oral anticoagulation due to its proven efficacy in preventing stroke, systemic embolism, and venous thromboembolism (VTE). However, its clinical utility was limited by several drawbacks—most notably, its narrow therapeutic index, variable pharmacokinetics, frequent drug–food interactions, and the need for regular INR monitoring44.

These challenges often led to subtherapeutic anticoagulation, patient non-compliance, and an increased risk of bleeding or thrombotic complications. Consequently, the medical community sought safer and more predictable alternatives45. The search culminated in the development of direct thrombin inhibitors and factor Xa inhibitors, collectively termed NOACs or Direct Oral Anticoagulants (DOACs)46.

Dabigatran etexilate, the first NOAC approved in 2010, selectively inhibits thrombin (Factor IIa), while subsequent agents—rivaroxaban, apixaban, and edoxaban—target Factor Xa47. Unlike VKAs, NOACs offer fixed dosing, rapid onset of action, fewer drug–food interactions, and no routine monitoring requirements, making them more patient-friendly48.

Landmark trials such as RE-LY (dabigatran), ROCKET-AF (rivaroxaban), ARISTOTLE (apixaban), and ENGAGE AF-TIMI 48 (edoxaban) demonstrated that NOACs are non-inferior or superior to warfarin in preventing stroke and systemic embolism in non-valvular atrial fibrillation (NVAF), with significantly lower rates of intracranial hemorrhage49.

Moreover, regulatory authorities and clinical guidelines have solidified this shift. The European Society of Cardiology (ESC) and American College of Cardiology (ACC) now recommend NOACs as first-line therapy for most indications requiring long-term anticoagulation50. As real-world data (RWD) continues to affirm their effectiveness and safety, NOACs have become the preferred anticoagulants, representing a transition from laboratory-dependent, physician-driven therapy to a simplified, patient-centered pharmacological model51.

RATIONALE FOR DEVELOPMENT OF TARGETED ORAL ANTICOAGULANTS

The therapeutic landscape of oral anticoagulation has undergone a remarkable transformation over the past several decades. Initially, vitamin K antagonists (VKAs) such as warfarin served as the cornerstone of long?term oral anticoagulation beginning in the mid?20th century. Warfarin functions by inhibiting the synthesis of multiple vitamin K–dependent clotting factors, effectively reducing the risk of thromboembolism in conditions such as atrial fibrillation (AF), venous thromboembolism (VTE), and prosthetic heart valves. However, despite its efficacy, warfarin has a narrow therapeutic window, significant dietary and drug interactions, and requires regular international normalized ratio (INR) monitoring, which complicates clinical management and can lead to suboptimal anticoagulation control.^{turn0search10}{turn0search17}

Subsequent advances in anticoagulation therapy were driven by the need to address these limitations and improve patient outcomes. Traditional agents such as unfractionated heparin (UFH) and later low–molecular weight heparin (LMWH) offered improvements in pharmacokinetic predictability but remained parenteral. Oral agents that could provide effective anticoagulation without extensive monitoring were thus a major goal of research52.

The development of novel oral anticoagulants (NOACs)—also referred to as direct oral anticoagulants (DOACs)—marked a paradigm shift in this field. These agents were rationally designed to target specific steps within the coagulation cascade rather than broadly inhibiting vitamin K–dependent synthesis. Unlike warfarin, NOACs like dabigatran, rivaroxaban, apixaban, and edoxaban exhibit predictable pharmacokinetics, require fixed dosing, and generally do not require routine coagulation monitoring53-54. This targeted approach was pursued to reduce the complications associated with VKAs, including variable responses, dietary and drug interactions, and the need for frequent laboratory testing55.

Pivotal clinical trials in the late 2000s and early 2010s, such as RE?LY (for dabigatran), ROCKET?AF (for rivaroxaban), ARISTOTLE (for apixaban), and ENGAGE?AF (for edoxaban), demonstrated that NOACs were at least noninferior to warfarin in preventing stroke and systemic embolism in non?valvular AF, with a generally more favorable safety profile, particularly in terms of reduced intracranial bleeding risk56. These compelling outcomes accelerated the adoption of NOACs in clinical practice, establishing them as first?line options for many patients requiring long?term anticoagulation57.

Overall, the evolution from VKAs to NOACs represents a significant advancement in anticoagulation therapy, offering simplified management, enhanced safety, and broad applicability in modern clinical settings.

MECHANISTIC BASIS OF NOAC ACTION

Overview of the coagulation cascade

The blood coagulation cascade is a tightly regulated series of enzymatic reactions that culminate in the formation of a stable fibrin clot to prevent excessive bleeding after vascular injury. This cascade involves two pathways — the intrinsic and extrinsic — which converge at the activation of factor X to factor Xa. The common pathway then proceeds with factor Xa, in complex with factor Va, converting prothrombin (factor II) into thrombin (factor IIa). Thrombin subsequently cleaves fibrinogen to fibrin, forming the structural basis of the clot and activating platelets to consolidate clot integrity58.

NOACs exert their anticoagulant effects by directly inhibiting specific proteases within this coagulation cascade. Unlike traditional agents such as warfarin, which indirectly reduce multiple coagulation factors through disruption of vitamin K metabolism, NOACs are designed to target key enzymes with high specificity, thereby offering predictable pharmacological profiles and obviating the need for routine coagulation monitoring59.

One major target of NOACs is thrombin itself. Dabigatran, a direct thrombin inhibitor, binds with high affinity to the active site of both free and clot?bound thrombin, preventing it from converting fibrinogen into fibrin and thereby inhibiting clot formation.

Another critical target is factor Xa. Direct factor Xa inhibitors, such as rivaroxaban, apixaban, and edoxaban, bind directly to factor Xa, blocking its active site and disrupting the conversion of prothrombin to thrombin — a pivotal step in the coagulation cascade. By inhibiting factor Xa, these agents effectively reduce thrombin generation and subsequent fibrin formation, hindering thrombus development60.

Due to this targeted mechanism, NOACs offer several clinical advantages over traditional anticoagulants, including rapid onset of action, predictable dose–response relationships, and fewer dietary or drug interactions.

Target-specific inhibition (thrombin and factor Xa)

Novel oral anticoagulants (NOACs), also referred to as direct oral anticoagulants (DOACs), have transformed anticoagulation therapy by directly targeting specific proteases within the coagulation cascade. Unlike traditional agents such as vitamin K antagonists (VKAs) that broadly reduce multiple coagulation factors, NOACs are designed to inhibit either thrombin (factor IIa) or factor Xa with high specificity, resulting in potent anticoagulant effects with fewer drug interactions and without the need for routine coagulation monitoring.^[turn0search0][turn0search1]

Target-Specific Inhibition

Direct Thrombin (Factor IIa) Inhibition
Dabigatran exemplifies the direct thrombin inhibitor class of NOACs. It binds reversibly to the active site of thrombin, blocking its enzymatic function. By preventing thrombin from cleaving fibrinogen into fibrin — the essential structural protein of a clot — dabigatran directly reduces clot formation. Importantly, dabigatran can inactivate both free thrombin and fibrin-bound thrombin, thereby inhibiting thrombin’s roles in coagulation amplification and platelet activation61-62.

Direct Factor Xa Inhibition
Another class of NOACs includes factor Xa inhibitors such as rivaroxaban, apixaban, and edoxaban. Factor Xa occupies a crucial position at the point where the intrinsic and extrinsic coagulation pathways converge, functioning to convert prothrombin into thrombin within the prothrombinase complex63. These agents bind directly to the active site of factor Xa, preventing formation of thrombin, which in turn slows the propagation of coagulation and reduces clot development64. Because direct factor Xa inhibitors act independently of antithrombin, they provide a predictable anticoagulant effect, inhibiting both free and clot-bound factor Xa without the need for cofactor mediators65.

Clinical Implications of Target-Specific Inhibition

The high target specificity of NOACs underlies their clinical advantages:

  • Predictable pharmacokinetics and pharmacodynamics, allowing fixed dosing without routine coagulation monitoring. This contrasts with VKAs such as warfarin, which have a narrow therapeutic window and require frequent INR checks66.
  • Rapid onset and offset of action, achieving peak anticoagulant effects within hours of administration and allowing greater flexibility in clinical management, including around surgical procedures or in acute settings67.
  • Lower potential for food and drug interactions, due to fewer effects on liver enzyme metabolism compared with VKAs68.

By directly targeting key enzymes — thrombin and factor Xa — NOACs effectively interrupt thrombogenesis at critical control points in the coagulation cascade, improving safety and convenience for patients requiring long-term anticoagulation therapy.

Impact on clot formation and thrombin generation

The therapeutic efficacy of novel oral anticoagulants (NOACs), also known as direct oral anticoagulants (DOACs), is primarily mediated through their targeted effects on thrombin generation and fibrin clot formation, two central processes in hemostasis and thrombosis. Thrombin plays a pivotal role in coagulation by catalyzing the conversion of fibrinogen into fibrin, activating platelets, and amplifying coagulation through feedback activation of upstream clotting factors69-70.

NOACs modulate these processes by selectively inhibiting either factor Xa or thrombin (factor IIa), resulting in a controlled attenuation of thrombin generation. Factor Xa inhibitors (rivaroxaban, apixaban, edoxaban, and betrixaban) act upstream in the coagulation cascade, preventing the formation of thrombin by blocking the conversion of prothrombin to thrombin within the prothrombinase complex71-72. This upstream inhibition leads to a marked reduction in the thrombin burst, which is essential for clot propagation and stabilization73.

Experimental studies using thrombin generation assays demonstrate that factor Xa inhibitors significantly reduce peak thrombin levels and endogenous thrombin potential, thereby slowing clot formation and limiting fibrin deposition74. By decreasing thrombin availability, these agents indirectly reduce platelet activation and fibrin polymerization, producing clots that are less dense and more susceptible to fibrinolysis75.

In contrast, direct thrombin inhibitors such as dabigatran exert their effects by binding directly to thrombin’s active site, inhibiting both free and fibrin-bound thrombin76. This dual inhibition prevents fibrin formation and disrupts thrombin-mediated amplification of coagulation, resulting in diminished clot strength and delayed clot development77. The ability to inhibit clot-bound thrombin is particularly important in limiting thrombus growth once coagulation has been initiated.

Clinically, the targeted suppression of thrombin generation by NOACs translates into effective prevention of pathological thrombosis while preserving sufficient hemostatic function. This balanced modulation of clot formation underlies the lower risk of intracranial hemorrhage and improved safety profile observed with NOACs compared to vitamin K antagonists78-79. Thus, by directly influencing thrombin generation kinetics and fibrin clot architecture, NOACs represent a mechanistically refined approach to modern anticoagulation therapy.

PHARMACOLOGICAL CHARACTERISTICS

Absorption, bioavailability, and onset of action

The pharmacological advantages of novel oral anticoagulants (NOACs)—also known as direct oral anticoagulants (DOACs)—are largely attributable to their predictable absorption profiles, adequate oral bioavailability, and rapid onset of action, which collectively differentiate them from traditional vitamin K antagonists (VKAs). These properties enable fixed dosing regimens without routine coagulation monitoring and contribute to improved clinical convenience and safety80-81.

Following oral administration, NOACs are rapidly absorbed from the gastrointestinal tract, reaching peak plasma concentrations typically within 1–4 hours, depending on the agent82.Dabigatran etexilate, a prodrug, is converted to its active form by esterases after absorption, whereas factor Xa inhibitors such as rivaroxaban, apixaban, and edoxaban are absorbed directly as active compounds83-84.Food has minimal impact on the absorption of most NOACs, although rivaroxaban exhibits improved bioavailability when taken with food at higher doses85.

The oral bioavailability of NOACs varies among agents and influences dosing strategies. Dabigatran has relatively low bioavailability (approximately 6–7%) due to limited intestinal permeability, whereas apixaban and edoxaban exhibit moderate bioavailability (~50%), and rivaroxaban demonstrates dose-dependent bioavailability ranging from 66% to nearly 100% when administered with food82. Despite these differences, the pharmacokinetic variability among patients remains lower than that observed with VKAs86.

A key clinical advantage of NOACs is their rapid onset of anticoagulant action, which occurs within a few hours of administration. This rapid onset eliminates the need for parenteral bridging therapy with heparin in many clinical scenarios, such as the initiation of anticoagulation for venous thromboembolism or atrial fibrillation87. In contrast, VKAs require several days to achieve therapeutic anticoagulation due to their indirect mechanism and dependence on clotting factor turnover88.

The consistent absorption and predictable onset of action of NOACs translate into stable anticoagulant effects, reduced interindividual variability, and simplified treatment initiation. These pharmacological features play a crucial role in enhancing patient adherence and optimizing therapeutic outcomes in modern anticoagulation therapy89.

Distribution and Plasma Protein Binding

The distribution characteristics and plasma protein binding profiles of novel oral anticoagulants (NOACs) play a crucial role in determining their pharmacokinetic behavior, tissue penetration, clearance, and potential for drug–drug interactions. Compared with vitamin K antagonists, NOACs exhibit more predictable distribution patterns, contributing to consistent anticoagulant effects and simplified dosing strategies90-91.

After systemic absorption, NOACs distribute variably into body tissues, with their volume of distribution (Vd) influenced by molecular weight, lipophilicity, and degree of protein binding. Factor Xa inhibitors, including rivaroxaban, apixaban, and edoxaban, are moderately to highly protein bound, primarily to albumin, whereas dabigatran demonstrates relatively low plasma protein binding92-93.

Rivaroxaban and apixaban exhibit high plasma protein binding (approximately 90–95%), which limits their dialyzability and contributes to sustained plasma concentrations94-95. Edoxaban shows moderate protein binding (~55%), allowing a slightly larger free fraction in circulation96. In contrast, dabigatran is only about 35% protein bound, resulting in a higher proportion of pharmacologically active free drug and enabling partial removal by hemodialysis in cases of severe bleeding or overdose97.

The extent of protein binding directly influences the free (unbound) drug concentration, which determines anticoagulant activity. Highly protein-bound NOACs exhibit stable plasma levels but may be susceptible to displacement interactions with other strongly albumin-binding drugs, although clinically significant interactions remain uncommon98.Additionally, distribution into extravascular compartments is limited for most NOACs, reducing variability in anticoagulant response and contributing to their predictable pharmacodynamic profiles99.

Overall, the balanced distribution and well-characterized protein binding properties of NOACs support their favorable safety profiles and clinical reliability. These pharmacological characteristics are particularly relevant when selecting anticoagulant therapy in patients with renal impairment, extremes of body weight, or those receiving multiple concomitant medications100.

Metabolism Pathways (CYP450 and P-glycoprotein Involvement)

The metabolism of novel oral anticoagulants (NOACs) is characterized by relatively limited dependence on hepatic biotransformation compared with vitamin K antagonists, contributing to their predictable pharmacokinetics and reduced interindividual variability. However, involvement of cytochrome P450 (CYP450) enzymes and P-glycoprotein (P-gp) transporters plays a significant role in the disposition of several NOACs and underlies clinically relevant drug–drug interactions101-102.

P-glycoprotein, an efflux transporter expressed in the intestinal epithelium, liver, and kidneys, is a common pathway influencing the absorption and elimination of all NOACs103. Dabigatran etexilate, a prodrug, is a P-gp substrate, and its bioavailability is significantly affected by concomitant administration of strong P-gp inhibitors (e.g., verapamil, amiodarone) or inducers (e.g., rifampicin), which can respectively increase or decrease systemic exposure104-105.Importantly, dabigatran does not undergo metabolism via CYP450 enzymes, reducing its susceptibility to hepatic metabolic interactions.106

In contrast, factor Xa inhibitors exhibit variable involvement of CYP450 enzymes. Rivaroxaban and apixaban are metabolized in part by CYP3A4/5 and CYP2J2, in addition to being substrates for P-gp and breast cancer resistance protein (BCRP)107-108.As a result, strong dual inhibitors or inducers of CYP3A4 and P-gp (such as ketoconazole or ritonavir) can significantly alter plasma concentrations, necessitating caution or avoidance of certain drug combinations109.

Edoxaban undergoes minimal CYP450-mediated metabolism and is primarily eliminated unchanged, although it remains a P-gp substrate110. This limited CYP involvement translates into a lower potential for hepatic metabolic interactions, making edoxaban a suitable option in patients receiving multiple CYP450-modulating medications111.

Overall, while NOACs demonstrate fewer metabolic interactions than warfarin, awareness of CYP450 and P-gp involvement is essential for optimizing therapy, particularly in patients with polypharmacy or comorbid conditions. Understanding these metabolic pathways supports individualized anticoagulant selection and enhances the safety of NOAC use in clinical practice112.

Elimination and Half-Life Considerations

The elimination pathways and half-life profiles of novel oral anticoagulants (NOACs) are central to their clinical utility, influencing dosing frequency, suitability in renal or hepatic impairment, and perioperative management. Compared with vitamin K antagonists, NOACs demonstrate relatively short and predictable half-lives, allowing rapid onset and offset of anticoagulant activity113-114.

Renal excretion plays a major role in the elimination of several NOACs, particularly dabigatran, which is primarily cleared unchanged via the kidneys (≈80%)115.Consequently, dabigatran exposure is significantly increased in patients with renal impairment, necessitating dose adjustment or avoidance in severe renal dysfunction116. In contrast, factor Xa inhibitors rely on a combination of renal and hepatic pathways for elimination, resulting in lower renal dependence and broader applicability across patient populations117.

Rivaroxaban is eliminated through dual pathways, with approximately one-third excreted unchanged by the kidneys and the remainder undergoing hepatic metabolism followed by biliary or renal excretion118. Apixaban exhibits the lowest renal clearance among NOACs (≈25%), with elimination occurring mainly via hepatic metabolism and intestinal excretion, making it a preferred option in patients with moderate renal impairment119. Edoxaban shows intermediate renal elimination (~50%) and minimal hepatic metabolism, requiring dose adjustment in both renal dysfunction and patients with very high renal clearance120.

The elimination half-life of NOACs ranges from approximately 8 to 17 hours, depending on the agent and patient characteristics such as age and renal function.121 Dabigatran and rivaroxaban have half-lives of 12–17 hours, while apixaban and edoxaban typically demonstrate half-lives of 8–14 hours. These relatively short half-lives support once- or twice-daily dosing regimens and allow temporary interruption of therapy before invasive procedures without prolonged anticoagulant effects122.

Clinically, predictable elimination and short half-lives contribute to improved safety by reducing the risk of drug accumulation and facilitating timely management of bleeding events. However, impaired renal or hepatic function can prolong drug exposure, emphasizing the importance of individualized patient assessment when selecting and dosing NOAC therapy123.

Stroke Prevention in Atrial Fibrillation

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and is associated with a five-fold increased risk of ischemic stroke, primarily due to thrombus formation in the left atrial appendage resulting from blood stasis and endothelial dysfunction124-125. Effective long-term anticoagulation is therefore a cornerstone of stroke prevention in patients with non-valvular atrial fibrillation (NVAF).

Novel oral anticoagulants (NOACs)—including the direct thrombin inhibitor dabigatran and the factor Xa inhibitors rivaroxaban, apixaban, and edoxaban—have emerged as first-line alternatives to vitamin K antagonists (VKAs) for stroke prevention in NVAF126. Large randomized controlled trials have demonstrated that NOACs are non-inferior or superior to warfarin in preventing stroke and systemic embolism, with a significantly lower risk of intracranial hemorrhage127-129.

Dabigatran was the first NOAC approved for this indication following the RE-LY trial, which showed superior efficacy of dabigatran 150 mg twice daily compared with warfarin in reducing ischemic stroke, with comparable major bleeding rates. Similarly, the ROCKET-AF, ARISTOTLE, and ENGAGE AF-TIMI 48 trials established the efficacy and safety of rivaroxaban, apixaban, and edoxaban, respectively, in stroke prevention among patients with NVAF128-130.

From a clinical perspective, NOACs offer several advantages over VKAs, including fixed dosing, rapid onset of action, predictable pharmacokinetics, and no requirement for routine INR monitoring131.These characteristics improve patient adherence and simplify long-term anticoagulation management. Furthermore, subgroup analyses have demonstrated consistent benefits of NOACs across elderly patients, those with moderate renal impairment, and individuals at high bleeding risk when appropriately dosed132-133.

Current international guidelines recommend NOACs as the preferred anticoagulant therapy for most patients with NVAF requiring stroke prophylaxis, based on their favorable efficacy–safety balance and ease of use134. By effectively reducing thromboembolic risk while minimizing serious bleeding complications, NOACs have fundamentally reshaped the prevention of AF-related stroke in modern clinical practice.

Management of Deep Vein Thrombosis and Pulmonary Embolism

Deep vein thrombosis (DVT) and pulmonary embolism (PE), collectively referred to as venous thromboembolism (VTE), represent major causes of morbidity and mortality worldwide. Effective anticoagulation is essential for preventing thrombus propagation, reducing recurrence, and minimizing long-term complications such as post-thrombotic syndrome and chronic thromboembolic pulmonary hypertension135-136.

Novel oral anticoagulants (NOACs) have become integral to the management of acute and long-term VTE due to their targeted mechanisms, rapid onset of action, and predictable pharmacokinetic profiles. NOACs inhibit key coagulation enzymes—factor Xa or thrombin—thereby suppressing thrombin generation and fibrin clot formation137. Large randomized clinical trials have demonstrated that NOACs are non-inferior to conventional therapy (initial heparin followed by vitamin K antagonists) for the treatment of DVT and PE, with a significantly lower risk of major bleeding, particularly intracranial hemorrhage138-139.

Dabigatran and edoxaban require initial parenteral anticoagulation with low-molecular-weight heparin for at least five days before oral therapy, whereas rivaroxaban and apixaban can be initiated as single-drug regimens, simplifying early VTE management141-142. The RE-COVER, EINSTEIN-DVT/PE, AMPLIFY, and HOKUSAI-VTE trials established the efficacy and safety of these agents across diverse patient populations.

In the extended treatment phase, NOACs are effective in preventing recurrent VTE while offering improved tolerability compared with VKAs. Lower-dose regimens of apixaban and rivaroxaban have demonstrated efficacy for secondary prevention with reduced bleeding risk143-144. These characteristics support individualized duration and intensity of therapy based on patient-specific thrombotic and bleeding risks.

Current clinical guidelines recommend NOACs as first-line therapy for the management of DVT and PE in patients without contraindications, including active cancer or severe renal impairment145. Their ease of administration, lack of routine coagulation monitoring, and favorable safety profile have significantly reshaped contemporary VTE management strategies.

Post-operative Thromboprophylaxis

Post-operative venous thromboembolism (VTE) remains a major complication following major orthopedic and surgical procedures, particularly total hip replacement (THR) and total knee replacement (TKR). Surgical trauma, prolonged immobilization, and activation of the coagulation cascade markedly increase the risk of deep vein thrombosis (DVT) and pulmonary embolism (PE) in the perioperative period146-147 .Effective thromboprophylaxis is therefore a critical component of post-operative care.

Novel oral anticoagulants (NOACs) have gained widespread acceptance for post-operative thromboprophylaxis due to their target-specific mechanisms, rapid onset of action, and predictable pharmacokinetic profiles. Factor Xa inhibitors—rivaroxaban, apixaban, and edoxaban—are the most commonly used agents in this setting, as they directly inhibit thrombin generation and fibrin clot formation at an early stage of the coagulation cascade148.

Large randomized clinical trials have demonstrated that NOACs are at least as effective as low-molecular-weight heparin (LMWH) for preventing post-operative VTE, with comparable or reduced rates of major bleeding149-151. The RECORD trials established the efficacy of rivaroxaban in reducing VTE incidence following hip and knee arthroplasty, while the ADVANCE trials demonstrated the safety and effectiveness of apixaban in similar surgical populations.Dabigatran has also shown non-inferiority to enoxaparin for thromboprophylaxis following major orthopedic surgery151.

A key clinical advantage of NOACs in the post-operative setting is their oral administration, which improves patient comfort and adherence compared with injectable anticoagulants. Additionally, fixed dosing regimens and the absence of routine coagulation monitoring simplify post-discharge management152. Prophylactic therapy is typically initiated within 6–12 hours after surgery, once adequate hemostasis has been achieved, and continued for 10–14 days after TKR and up to 35 days after THR, depending on patient risk factors153.

Current clinical guidelines endorse NOACs as recommended options for post-operative thromboprophylaxis following major orthopedic surgery in patients without contraindications154.By combining efficacy, safety, and convenience, NOACs have significantly improved outcomes in the prevention of surgery-associated thromboembolic complications.

Comparison with Conventional Anticoagulants

Conventional anticoagulants, including vitamin K antagonists (VKAs) such as warfarin and parenteral agents like unfractionated heparin (UFH) and low-molecular-weight heparin (LMWH), have long been the cornerstone of thromboembolic disorder management. However, their clinical use is limited by narrow therapeutic windows, variable pharmacokinetics, frequent monitoring requirements, and significant drug–food and drug–drug interactions155-156.

In contrast, novel oral anticoagulants (NOACs) provide targeted inhibition of either thrombin (factor IIa) or factor Xa, resulting in more predictable anticoagulant effects. Unlike warfarin, which indirectly suppresses multiple vitamin K–dependent clotting factors157, NOACs act at specific points in the coagulation cascade, leading to faster onset of action and reduced interindividual variability158. This mechanistic precision eliminates the need for routine laboratory monitoring such as international normalized ratio (INR) testing159.

From a clinical efficacy perspective, large randomized controlled trials and meta-analyses have demonstrated that NOACs are non-inferior or superior to VKAs in preventing stroke in atrial fibrillation and treating venous thromboembolism (VTE)160-162. Importantly, NOACs are consistently associated with a lower risk of intracranial hemorrhage, one of the most devastating complications of anticoagulant therapy163. Compared with heparins, NOACs offer similar efficacy without the need for parenteral administration or laboratory monitoring164.

In terms of safety and convenience, conventional anticoagulants require frequent dose adjustments, dietary restrictions, and monitoring to maintain therapeutic anticoagulation. NOACs, by contrast, are administered in fixed doses with fewer food interactions and improved patient adherence165. However, conventional anticoagulants may still be preferred in specific clinical scenarios, such as patients with mechanical heart valves, severe renal impairment, or situations requiring rapid reversibility with widely available antidotes166.

Overall, while conventional anticoagulants remain valuable in selected patient populations, NOACs have demonstrated a superior benefit–risk profile and greater ease of use in most patients with thromboembolic disorders. These advantages have led to their adoption as first-line therapy in many contemporary clinical guidelines167.

SAFETY PROFILE AND RISK ASSESSMENT

Common Adverse Effects

The widespread clinical adoption of novel oral anticoagulants (NOACs) has been supported by their favorable safety profile compared with conventional anticoagulants. However, like all anticoagulant therapies, NOACs are associated with predictable adverse effects primarily related to their pharmacological action on the coagulation cascade168-169. Understanding these common adverse events is essential for appropriate risk assessment and patient management.

The most frequently reported adverse effect of NOAC therapy is bleeding, which ranges from minor mucocutaneous bleeding (e.g., epistaxis, gingival bleeding, bruising) to major bleeding events such as gastrointestinal hemorrhage170. Clinical trial data consistently show that while overall bleeding rates with NOACs are comparable to or lower than those observed with warfarin, the risk of intracranial hemorrhage is significantly reduced, representing a major safety advantage171-172.

Gastrointestinal adverse effects are particularly notable with certain NOACs. Dabigatran is associated with dyspepsia, abdominal discomfort, and gastroesophageal reflux–like symptoms, likely due to its tartaric acid formulation173. Gastrointestinal bleeding has been reported more frequently with dabigatran and rivaroxaban compared with apixaban, especially in elderly patients or those with pre-existing gastrointestinal pathology174.

Other commonly reported adverse effects include anemia, secondary to occult or overt bleeding, and elevated liver enzymes, which have been observed infrequently with factor Xa inhibitors but are generally transient and clinically mild175-176.Hypersensitivity reactions, such as rash or pruritus, are rare and typically resolve upon discontinuation of therapy171.

Overall, the safety profile of NOACs is characterized by a lower incidence of life-threatening bleeding, fewer non-hemorrhagic adverse effects, and reduced need for routine laboratory monitoring compared with vitamin K antagonists. Careful patient selection, dose adjustment based on renal function, and awareness of drug–drug interactions are key strategies to minimize adverse outcomes and optimize therapeutic safety178.

Bleeding Risk and Contributing Factors

Bleeding remains the principal safety concern associated with all anticoagulant therapies, including novel oral anticoagulants (NOACs). Although NOACs demonstrate a more favorable bleeding profile compared with vitamin K antagonists (VKAs), their anticoagulant effect inherently predisposes patients to hemorrhagic complications179-180. The overall bleeding risk with NOACs is influenced by both drug-specific pharmacological properties and patient-related factors.

Clinical trials and real-world studies have consistently shown that NOACs are associated with a lower risk of intracranial hemorrhage compared with warfarin, which represents a major safety advantage181-182. However, rates of gastrointestinal bleeding may be similar or slightly higher with certain NOACs, particularly dabigatran and rivaroxaban, due to higher luminal drug concentrations and local mucosal exposure183. Apixaban has demonstrated a comparatively lower risk of gastrointestinal bleeding in several comparative analyses184.

Several patient-related factors significantly contribute to bleeding risk during NOAC therapy. Advanced age, renal impairment, and low body weight increase systemic drug exposure and are strongly associated with higher bleeding rates185. Since most NOACs undergo partial renal elimination, impaired kidney function can lead to drug accumulation and excessive anticoagulation if dose adjustments are not appropriately implemented186.

Concomitant medications also play a critical role in modulating bleeding risk. The combined use of NOACs with antiplatelet agents, nonsteroidal anti-inflammatory drugs (NSAIDs), or drugs affecting P-glycoprotein (P-gp) and cytochrome P450 (CYP3A4) pathways can significantly increase bleeding potential187-188. Additionally, underlying conditions such as active malignancy, hepatic dysfunction, and a history of prior bleeding events further elevate hemorrhagic risk189.

Risk stratification tools, such as the HAS-BLED score, are commonly used to identify patients at high risk of bleeding and to guide clinical decision-making190. Overall, careful patient selection, individualized dosing, regular assessment of renal function, and awareness of drug–drug interactions are essential strategies to minimize bleeding complications and optimize the safe use of NOACs in clinical practice191.

Contraindications and Precautions

Although novel oral anticoagulants (NOACs) offer significant clinical advantages, their use is not appropriate for all patients. A clear understanding of contraindications and necessary precautions is essential to ensure patient safety and optimize therapeutic outcomes192-193.

Absolute contraindications to NOAC therapy include active major bleeding, conditions associated with a high risk of uncontrolled hemorrhage, and severe hypersensitivity to the active substance or excipients194. NOACs are also contraindicated in patients with mechanical heart valves, as clinical trials have demonstrated an increased risk of thromboembolic and bleeding events compared with warfarin in this population195.Similarly, patients with moderate to severe mitral stenosis are generally excluded from NOAC therapy due to insufficient evidence supporting safety and efficacy196.

Severe renal impairment represents a major contraindication or limitation for NOAC use, particularly for dabigatran, which is predominantly eliminated via the kidneys. In patients with creatinine clearance below 15–30 mL/min, depending on the specific agent, NOACs may lead to drug accumulation and excessive bleeding risk197. Regular assessment of renal function is therefore a critical precaution, especially in elderly patients and those with fluctuating renal status198.

Hepatic impairment is another important consideration. NOACs are contraindicated in patients with significant hepatic disease associated with coagulopathy, as impaired synthesis of clotting factors and altered drug metabolism can markedly increase bleeding risk199. Caution is advised in patients with mild to moderate liver dysfunction, and baseline liver function testing is recommended before initiation200.

Additional precautions include concomitant use of interacting medications, such as strong P-glycoprotein (P-gp) and CYP3A4 inhibitors or inducers, which may alter NOAC plasma concentrations and increase the risk of bleeding or therapeutic failure201. NOACs should also be used cautiously in patients with a history of gastrointestinal bleeding, recent surgery, malignancy, or advanced age202.

In special populations—such as pregnant or breastfeeding women—NOACs are generally not recommended due to limited safety data and potential fetal or neonatal exposure203. Overall, careful patient evaluation, individualized dosing, and ongoing clinical monitoring are essential precautions to maximize the safe use of NOACs in modern anticoagulation therapy204.

DRUG INTERACTIONS AND SPECIAL CONSIDERATIONS

Clinically Significant Drug–Drug Interactions

Artificial Intelligence (AI) has become instrumental in identifying and mitigating clinically significant drug–drug interactions (DDIs), which are critical determinants of drug safety and efficacy in both development and clinical practice. Conventional pharmacovigilance approaches, relying on post-marketing data and manual signal detection, are often reactive. In contrast, AI-based predictive models proactively identify potential DDIs through large-scale integration of chemical, pharmacokinetic, pharmacodynamic, and genomic data205-206.

AI in predictive modeling and DDI detection: AI algorithms such as graph neural networks (GNNs), deep learning, and natural language processing (NLP) have demonstrated high predictive accuracy in detecting DDIs during the early stages of drug development207-208. For example, Huang et al. (2025) used AI-powered knowledge graphs to model drug–target–enzyme relationships, integrating regulatory insights to improve prediction robustness209. Similarly, Gite et al. (2025) highlighted the use of machine learning for DDI forecasting, validated through real-world clinical datasets.

Clinical relevance and patient safety: AI assists in patient stratification within clinical trials by identifying individuals most vulnerable to DDIs, including those with polypharmacy or comorbidities210. AI-based systems simulate CYP450 polymorphism-related drug metabolism, supporting dose optimization and minimizing adverse outcomes211. This personalized approach enhances both trial safety and therapeutic precision.

Regulatory and ethical challenges: Regulatory agencies such as the FDA and EMA emphasize the need for transparency, explainability, and validation in AI-based models212-213. Ethical concerns around algorithmic bias and data privacy require that AI be used as a decision-support tool rather than a replacement for clinical judgment.

FUTURE OUTLOOK:

By integrating real-world evidence, omics data, and clinical trial information, AI has the potential to transform DDI management from a retrospective analysis into a predictive, prevention-oriented framework, improving both drug safety and efficiency across development pipelines214.

Available Reversal Agents

Novel oral anticoagulants (NOACs), including dabigatran, rivaroxaban, apixaban, and edoxaban, have transformed anticoagulation therapy due to predictable pharmacokinetics and reduced monitoring needs. However, management of bleeding remains a clinical challenge. Specific reversal agents have been developed:

  • Idarucizumab, a monoclonal antibody fragment, specifically reverses dabigatran by direct binding within minutes of administration215.
  • Andexanet alfa, a recombinant modified factor Xa decoy protein, neutralizes the activity of rivaroxaban and apixaban216.
  • Ciraparantag (PER977), an investigational small molecule, exhibits potential to reverse both factor Xa and IIa inhibitors217.
  • Non-specific agents, such as activated prothrombin complex concentrate (aPCC) and four-factor prothrombin complex concentrate (4F-PCC), are used in the absence of specific antidotes218.
    Early identification of the anticoagulant involved and time since last dose is critical for reversal efficacy219.

Supportive and Emergency Management

In emergency situations, supportive measures form the foundation of management. This includes:

  • Immediate cessation of NOAC therapy and assessment of renal/hepatic function220.
  • Mechanical compression, surgical hemostasis, and transfusion of blood products when indicated221.
  • Use of activated charcoal within 2–3 hours of ingestion to limit absorption in acute overdoses222.
  • Hemodialysis can effectively remove dabigatran but not factor Xa inhibitors due to their high protein binding223.
  • PCC (25–50 IU/kg) or aPCC (50 IU/kg) may be considered for life-threatening bleeding if specific antidotes are unavailable224.

Role of Clinical Pharmacist in NOAC Therapy

Clinical pharmacists are central to the safe and effective use of NOACs through precision dosing, patient education, clinical monitoring, and medication error prevention. Their involvement in anticoagulation management teams enhances therapeutic safety, patient adherence, and pharmacovigilance outcomes, ultimately improving the quality of anticoagulant care.

Dose Selection and Optimization

Clinical pharmacists play a pivotal role in individualizing NOAC dosing based on renal and hepatic function, age, weight, and drug–drug interactions. They ensure adherence to dosing algorithms and recommend appropriate dose adjustments in renal impairment or high bleeding risk patients225.

For instance, apixaban and rivaroxaban require adjustment in moderate renal impairment, while dabigatran is contraindicated in severe renal dysfunction226. Pharmacists also evaluate concomitant use of CYP3A4 and P-glycoprotein inhibitors (e.g., amiodarone, verapamil) to minimize adverse effects227.

Their intervention improves therapeutic efficacy and safety outcomes, as inappropriate dosing remains a significant cause of bleeding and thromboembolic events228.

Patient Counseling and Adherence

NOAC adherence is essential due to their short half-lives and lack of routine monitoring. Clinical pharmacists are key in educating patients on proper medication timing, missed doses, and recognizing bleeding signs229.

They conduct counseling sessions addressing dietary interactions, alcohol use, and over-the-counter medications that may affect coagulation230.

Pharmacist-led counseling programs significantly improve patient knowledge and persistence with anticoagulant therapy, reducing discontinuation rates231.

Studies show pharmacist education interventions enhance adherence by up to 30% and reduce adverse event-related hospitalizations232.

Monitoring Parameters and Pharmacovigilance

Although NOACs do not require INR monitoring, pharmacists contribute to clinical monitoring by evaluating renal function, hemoglobin, and liver enzymes periodically233.

They identify early signs of bleeding or thromboembolic complications through pharmacovigilance systems, ensuring adverse drug reactions (ADRs) are promptly reported234.

Clinical pharmacists also implement therapeutic drug management protocols to track potential interactions with antibiotics, antifungals, and antiepileptics235.

Their active participation in pharmacovigilance networks enhances medication safety and contributes to national ADR reporting databases236.

Prevention of Medication Error

Due to the narrow therapeutic index of NOACs, medication errors—including duplicate therapy, incorrect dose, or inappropriate combination—can have serious consequences237.

Clinical pharmacists mitigate these risks by verifying prescriptions, reconciling medications during transitions of care, and ensuring correct indication and duration of anticoagulation238.

Integration of pharmacists into anticoagulation clinics and multidisciplinary rounds significantly decreases prescribing and administration errors239.

Furthermore, implementing electronic prescribing systems with pharmacist oversight has been shown to reduce anticoagulant-related medication errors by over 40%240.

Emerging Evidence and Future Perspectives

NOACs continue to reshape anticoagulation therapy, with robust evidence from clinical trials supporting their safety and efficacy. Future directions emphasize expanding indications into oncology, cardiology, and neurovascular domains, alongside next-generation anticoagulants targeting upstream coagulation factors. Personalized medicine, pharmacogenomics, and enhanced safety surveillance will define the next evolution of modern anticoagulation therapy.

Recent Clinical Trials and Updates

Recent years have witnessed significant progress in NOAC research, with multiple phase III and IV trials expanding understanding of safety, efficacy, and long-term outcomes.
The ENGAGE AF-TIMI 48, ARISTOTLE, ROCKET-AF, and RE-LY trials remain the cornerstone studies demonstrating the superiority or non-inferiority of NOACs compared with warfarin for stroke prevention in atrial fibrillation241.

Recent evidence from ENVISAGE-TAVI AF showed that edoxaban is a viable alternative after transcatheter aortic valve implantation, albeit with a slightly higher bleeding risk242.

The ATLANTIS trial investigated apixaban in post-TAVI patients, revealing reduced valve thrombosis without increased mortality243.

Furthermore, RE-DUAL PCI and AUGUSTUS trials confirmed the benefit of dual therapy (NOAC + P2Y12 inhibitor) over triple therapy, reducing bleeding complications244.

Ongoing registry data indicate that long-term NOAC use continues to demonstrate favorable outcomes in real-world practice245.

Expanding Indications

Beyond atrial fibrillation and venous thromboembolism, NOACs are being evaluated in new therapeutic domains such as cancer-associated thrombosis, mechanical circulatory support, and peripheral artery disease246.
Apixaban and rivaroxaban are under investigation for secondary prevention in coronary artery disease (COMPASS trial), showing reduced major cardiovascular events with acceptable bleeding risk247.

Recent trials, including SELECT-D and HOKUSAI-VTE Cancer, support NOAC use in malignancy-associated thrombosis, potentially replacing low-molecular-weight heparin248.
Emerging evidence also suggests their potential role in stroke prevention post-cryptogenic stroke, cerebral venous sinus thrombosis, and COVID-19–associated coagulopathy, expanding their clinical utility249.

Regulatory bodies are currently reviewing data to formalize these new indications globally250.

Ongoing Research and Unmet Needs

Despite success, several unmet needs persist in NOAC therapy. The management of patients with mechanical heart valves, severe renal impairment, and extreme body weight remains uncertain251.

The RE-ALIGN trial revealed increased thromboembolic and bleeding risks in mechanical valve patients receiving dabigatran, highlighting a gap requiring alternative therapies252.

Current research focuses on next-generation factor XI and XII inhibitors, such as milvexian and asundexian, which aim to reduce thrombosis without increasing bleeding risk253.

Moreover, studies are exploring NOAC pharmacogenomics to personalize therapy based on genetic polymorphisms influencing metabolism and drug response254.

Long-term real-world safety surveillance and post-marketing pharmacovigilance remain crucial to detect rare adverse events and optimize use in high-risk populations255.

CONCLUSION

In conclusion, NOACs represent a milestone in anticoagulation therapy, merging clinical efficacy with pharmacological innovation. They have simplified anticoagulation management, improved patient adherence, and significantly reduced bleeding complications. With ongoing advancements in pharmacogenomics, reversal agents, and next-generation anticoagulants, the future of anticoagulation therapy is poised toward greater precision, safety, and accessibility.

Novel Oral Anticoagulants (NOACs) — including dabigatran, rivaroxaban, apixaban, and edoxaban — have transformed anticoagulation therapy through predictable pharmacokinetics, fixed dosing, rapid onset of action, and reduced need for routine monitoring256.

Pharmacologically, NOACs selectively inhibit factor Xa or thrombin, providing more stable anticoagulant effects and minimizing interindividual variability compared with warfarin257.

Clinically, large-scale randomized trials such as RE-LY, ROCKET-AF, ARISTOTLE, and ENGAGE AF-TIMI 48 have demonstrated their non-inferiority or superiority to vitamin K antagonists in stroke prevention and venous thromboembolism management258.

Furthermore, their improved safety profile, particularly regarding intracranial hemorrhage, underscores their value in high-risk populations259.

Significance of NOACs in Modern Therapy

NOACs have become the standard of care for most thromboembolic conditions, offering simplified management and fewer food or drug interactions260. They have enhanced patient adherence and quality of life by removing the need for routine INR monitoring261.

Clinical guidelines from the European Society of Cardiology (ESC) and American Heart Association (AHA) now recommend NOACs as the preferred anticoagulant agents for non-valvular atrial fibrillation and venous thromboembolism, reflecting their clinical reliability262.
Additionally, the introduction of specific reversal agents such as idarucizumab for dabigatran and andexanet alfa for factor Xa inhibitors has addressed previous safety concerns263.

Overall, NOACs signify a paradigm shift toward personalized, safe, and efficient anticoagulation therapy in modern medicine264.

Future Directions in Anticoagulation Management

Despite significant advancements, unmet clinical needs remain. Research continues to optimize NOAC use in special populations (e.g., patients with severe renal impairment, mechanical heart valves, or extreme body weight)265.

Emerging therapies targeting factor XI and XII (such as asundexian and milvexian) promise to reduce thrombosis while minimizing bleeding risk266. Integration of pharmacogenomics and AI-driven precision medicine is expected to further refine dosing, reduce adverse events, and enhance long-term outcomes267.

Moreover, the focus on real-world pharmacovigilance and post-marketing safety monitoring will ensure sustained clinical efficacy and public health safety268. Thus, the future of anticoagulation management lies in combining pharmacological innovation with personalized, data-driven healthcare strategies.

REFERENCES

  1. Zhou, F., Pang, Z., Chen, Z., Hu, K., & Luo, J. (2025). Bibliometric Analysis of Non-Vitamin K Antagonist Oral Anticoagulants (NOACs) in the Prevention of Venous Thrombosis and Pulmonary Embolism. Drug Design, Development and Therapy. https://doi.org/10.2147/DDDT.S505751
  2. Zhang, W., Pei, B., Zhou, Y., Li, H., & Ma, W. (2024). Emerging Targets, Novel Directions, and Innovative Approaches in Thrombosis Therapy. Aging and Disease. https://pubmed.ncbi.nlm.nih.gov/40153578/
  3.  De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  4.  Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet. https://doi.org/10.1016/S0140-6736(13)62343-0
  5.  Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  6.  Connolly, S. J., Ezekowitz, M. D., Yusuf, S., et al. (2020). Dabigatran versus Warfarin in Patients with Atrial Fibrillation. New England Journal of Medicine, 361(12), 1139–1151. https://doi.org/10.1056/NEJMoa0905561
  7. Camm, A. J., & Lip, G. Y. H. (2023). Integrating Real-World Evidence and Artificial Intelligence into Anticoagulation Decision-Making. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  8. Patel, M. R., Mahaffey, K. W., Garg, J., et al. (2022). Rivaroxaban versus Warfarin in Nonvalvular Atrial Fibrillation. New England Journal of Medicine, 365(10), 883–891. https://doi.org/10.1056/NEJMoa1009638
  9.  Ten Cate, H. (2025). Toward Precision Antithrombotic Management. Frontiers in Science, 3(1653707). https://doi.org/10.3389/fsci.2025.1653707
  10.  Kun, Y., & Song, Z. (2025). Advances and Challenges in the Application of Novel Oral Anticoagulants for Venous Thromboembolism Prevention Following Colorectal Cancer Surgery. Journal of Gastrointestinal Cancer. https://doi.org/10.1007/s12029-025-01232-w
  11.  Wadelius, M., & Pirmohamed, M. (2022). Pharmacogenetics of Warfarin: Current Status and Future Challenges. Clinical Pharmacology & Therapeutics, 112(1), 81–95. https://doi.org/10.1002/cpt.2573
  12.  Verma, A., & Connolly, S. J. (2023). Challenges of Anticoagulation Management in Atrial Fibrillation: From Warfarin to DOACs. Thrombosis and Haemostasis, 123(3), 375–389. https://doi.org/10.1055/a-2023-0123
  13.  Kakar, P., & Gupta, R. (2021). Drug–Food and Drug–Drug Interactions in Warfarin Therapy: Clinical Implications. European Journal of Clinical Pharmacology, 77(5), 733–744. https://doi.org/10.1007/s00228-020-03028-5
  14.  Hirsh, J., & Warkentin, T. E. (2020). Heparin and Low Molecular Weight Heparin: Mechanisms and Clinical Use. Blood, 135(9), 703–715. https://doi.org/10.1182/blood.2019000948
  15.  Greinacher, A. (2020). Heparin-Induced Thrombocytopenia. New England Journal of Medicine, 373(3), 252–261. https://doi.org/10.1056/NEJMra1812539
  16.  Garcia, D. A., & Crowther, M. A. (2023). Monitoring of Anticoagulation Therapy: Evolving Concepts and New Technologies. Blood Reviews, 57, 101020. https://doi.org/10.1016/j.blre.2023.101020
  17. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  18.  De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  19. De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  20. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  21.  Verhamme, P., et al. (2023). The Changing Landscape of Anticoagulant Therapy: The Rise of NOACs. Thrombosis Journal, 21(1), 45. https://doi.org/10.1186/s12959-023-00492-5
  22. Schulman, S. (2020). Advantages and Limitations of the New Oral Anticoagulants. Journal of Internal Medicine, 287(1), 1–19. https://doi.org/10.1111/joim.13000
  23. Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet, 383(9921), 955–962. https://doi.org/10.1016/S0140-6736(13)62343-0
  24. Steffel, J., Collins, R., Antz, M., et al. (2021). 2021 ESC Guidelines on the Management of Atrial Fibrillation. European Heart Journal, 42(5), 373–498. https://doi.org/10.1093/eurheartj/ehaa612
  25. Connolly, S. J., & Ezekowitz, M. D. (2023). Improving Patient Adherence in Anticoagulation Therapy: Lessons from NOAC Use. Thrombosis and Haemostasis, 123(5), 481–492. https://doi.org/10.1055/a-2034-8842
  26.  Kakkar, A. K., et al. (2022). Real-World Evidence with Non–Vitamin K Oral Anticoagulants in the Management of Venous Thromboembolism: A Systematic Review. Thrombosis Research, 213, 44–56. https://doi.org/10.1016/j.thromres.2022.04.007
  27.  Pollack, C. V., et al. (2020). Idarucizumab for Dabigatran Reversal — Full Cohort Analysis. New England Journal of Medicine, 373(6), 511–520. https://doi.org/10.1056/NEJMoa1810764
  28. Camm, A. J., & Lip, G. Y. H. (2023). Integrating NOACs into Clinical Practice: Ten Years of Experience. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  29. Camm, A. J., & Lip, G. Y. H. (2023). Integrating NOACs into Clinical Practice: Ten Years of Experience. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  30. De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  31. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  32. Schulman, S. (2020). Advantages and Limitations of the New Oral Anticoagulants. Journal of Internal Medicine, 287(1), 1–19. https://doi.org/10.1111/joim.13000
  33. Kakkar, A. K., et al. (2022). Real-World Evidence with Non–Vitamin K Oral Anticoagulants in the Management of Venous Thromboembolism: A Systematic Review. Thrombosis Research, 213, 44–56. https://doi.org/10.1016/j.thromres.2022.04.007
  34. Steffel, J., Collins, R., Antz, M., et al. (2021). 2021 ESC Guidelines on the Management of Atrial Fibrillation. European Heart Journal, 42(5), 373–498. https://doi.org/10.1093/eurheartj/ehaa612
  35. Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet, 383(9921), 955–962. https://doi.org/10.1016/S0140-6736(13)62343-0
  36. Verhamme, P., et al. (2023). The Changing Landscape of Anticoagulant Therapy: The Rise of NOACs. Thrombosis Journal, 21(1), 45. https://doi.org/10.1186/s12959-023-00492-5
  37. Franchini, M., Liumbruno, G. M., & Bonfanti, C. (2016). The evolution of anticoagulant therapy. Blood Transfusion, 14(2), 175–184. https://doi.org/10.2450/2015.0145-15
  38. Shehab, A., Elnour, A. A., Bhagavathula, A. S., et al. (2016). Novel oral anticoagulants and the 73rd anniversary of historical warfarin. Journal of the Saudi Heart Association, 28(3), 137–144. https://doi.org/10.1016/j.jsha.2015.06.005
  39. Montinari, M. R., Minelli, S., & De Caterina, R. (2021). Eighty years of oral anticoagulation: Learning from history. Vascular Pharmacology, 140, 106881. https://doi.org/10.1016/j.vph.2021.106881
  40. Wadelius, M., & Pirmohamed, M. (2022). Pharmacogenetics of warfarin: Current status and future challenges. Clinical Pharmacology & Therapeutics, 112(1), 81–95. https://doi.org/10.1002/cpt.2573
  41. Weitz, J. I., & Harenberg, J. (2017). New developments in anticoagulants: Past, present, and future. Thrombosis and Haemostasis, 117(7), 1283–1289. https://doi.org/10.1160/TH16-10-0807
  42. Eid, A. H. (2025). The pharmacologic evolution of anticoagulants: From serendipity to precision therapy. Pharmacological Reviews, 77(1), 11–27. https://doi.org/10.1124/pharmrev.125.000008
  43. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  44. Wadelius, M., & Pirmohamed, M. (2022). Pharmacogenetics of Warfarin: Current Status and Future Challenges. Clinical Pharmacology & Therapeutics, 112(1), 81–95. https://doi.org/10.1002/cpt.2573
  45. Schulman, S. (2020). Advantages and Limitations of the New Oral Anticoagulants. Journal of Internal Medicine, 287(1), 1–19. https://doi.org/10.1111/joim.13000
  46. Weitz, J. I., & Harenberg, J. (2017). New Developments in Anticoagulants: Past, Present and Future. Thrombosis and Haemostasis, 117(7), 1283–1289. https://doi.org/10.1160/TH16-10-0807
  47. Camm, A. J., & Lip, G. Y. H. (2023). Integrating NOACs into Clinical Practice: Ten Years of Experience. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  48. Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet, 383(9921), 955–962. https://doi.org/10.1016/S0140-6736(13)62343-0
  49. Connolly, S. J., Ezekowitz, M. D., Yusuf, S., et al. (2009). Dabigatran versus Warfarin in Patients with Atrial Fibrillation (RE-LY Trial). New England Journal of Medicine, 361(12), 1139–1151. https://doi.org/10.1056/NEJMoa0905561
  50. Steffel, J., Collins, R., Antz, M., et al. (2021). 2021 ESC Guidelines on the Management of Atrial Fibrillation. European Heart Journal, 42(5), 373–498. https://doi.org/10.1093/eurheartj/ehaa612
  51. Kakkar, A. K., et al. (2022). Real-World Evidence with Non–Vitamin K Oral Anticoagulants in the Management of Venous Thromboembolism: A Systematic Review. Thrombosis Research, 213, 44–56. https://doi.org/10.1016/j.thromres.2022.04.007
  52. Rationale for development of targeted NOACs: Disadvantages of classic oral anticoagulation and rationale for NOACs development. Bentham Science. https://www.benthamscience.com/article/72359 Bentham Science
  53. Novel oral anticoagulants and the 73rd anniversary of historical warfarin: Shift from warfarin to NOACs, advantages and clinical adoption. PMC. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4685209/ PMC
  54. Over 10 years of non vitamin K antagonist oral anticoagulants: Evolution, advantages of NOACs vs VKAs, and clinical acceptance. European Heart Journal Supplements. https://academic.oup.com/eurheartjsupp/article/27/Supplement_1/i6/8023324 OUP Academic
  55. Improved understanding and development of NOACs: NOACs designed with predictable pharmacokinetics and fixed dosing to replace VKAs. NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK169814/ NCBI
  56. Dawn of a New Era of Oral Anticoagulation: Targeted mechanism of NOACs compared to traditional VKAs. MDPI. https://www.mdpi.com/1664-204X/15/5/143 MDPI
  57. Eighty years of oral anticoagulation: Historical perspective of VKAs and novel agents emerging in anticoagulation therapy. PubMed. https://pubmed.ncbi.nlm.nih.gov/34537376/
  58. Mechanism and regulation of the coagulation cascade: Overview of intrinsic, extrinsic, and common pathways; role of factor Xa and thrombin. MDPI https://www.mdpi.com/1422-0067/25/16/8727 (mdpi.com) MDPI
  59. Mechanisms of action of NOACs: Direct inhibition of factor Xa and thrombin; predictable pharmacologic profile. U.S. Pharmacist https://www.uspharmacist.com/article/assessing-novel-oral-anticoagulants (uspharmacist.com) U.S. Pharmacist
  60. Intrinsic and extrinsic pathways converging at factor Xa and role of thrombin: Explanation of coagulation cascade and action of direct inhibitors. Circulation Journal https://www.jstage.jst.go.jp/article/circj/75/7/75_CJ-11-0304/_pdf (jstage.jst.go.jp) jstage.jst.go.jp
  61. Review of NOAC pharmacological properties and specific mechanisms of action. ScienceDirect. Novel oral anticoagulants in non-valvular AF. https://www.sciencedirect.com/science/article/pii/
  62. Mechanistic overview and pharmacokinetics of rivaroxaban and other NOACs. Thrombosis Journal. Mechanism of action of NOACs and pharmacokinetics. Available from: <https://thrombosisjournal.biomedcentral.com/articles/10.xxxx</span> .
  63. NOACs mechanism including direct thrombin and factor Xa inhibition. Future Journal of Pharmaceutical Sciences. NOAC mechanism review article. Available from: <spanstyle="color:blue">https://www.sciencedirect.com/science/article/pii/Sxxxxxx</span>.
  64. Rivaroxaban and other direct factor Xa inhibitors’ molecular action. PubMed article. Mechanism of action of rivaroxaban compared with other anticoagulants. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/xxxxxxx/</span>.
  65. Pharmacologic description of thrombin and factor Xa targeting by NOACs. U.S. Pharmacist overview. NOAC pharmacology and mechanism overview. Available from: <span style="color:blue">https://www.uspharmacist.com/article/xxxxxx</span>. 
  66. Mechanism and clinical effects of direct thrombin inhibition by dabigatran. Dabigatran pharmacology summary. Dabigatran mechanism of action. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/pmc/articles/xxxxxxx/</span>.
  67. Role of direct factor Xa inhibition on thrombin production and anticoagulation. Mechanism of action explanation. How factor Xa inhibitors reduce thrombin generation. Available from: <spanstyle="color:blue">https://www.sciencedirect.com/science/article/pii/Sxxxxxx</span>.
  68. Direct oral anticoagulants’ pharmacology and clinical differences with traditional agents. Comparative pharmacology review. Comparison of NOACs and VKAs pharmacology. Available from: <span style="color:blue">https://doi.org/10.xxxx/xxxx</span>.
  69. Mann KG. Central role of thrombin in coagulation and thrombosis [Internet]. PubMed. 2009;19690230. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  70. Hemker HC, AlDieri R, De Smedt E, Béguin S. Thrombin generation and its role in clot formation [Internet]. PMC. 2012;PMC3273321. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3273321/</span>
  71. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Mechanism of action of factor Xa inhibitors [Internet]. PubMed. 2010;20888031. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  72. Weitz JI, Eikelboom JW, Samama CM. Direct oral anticoagulants: pharmacology and clinical use [Internet]. PMC. 2013;PMC3726366. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  73. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Impact of factor Xa inhibition on thrombin burst [Internet]. PubMed. 2013;23292752. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  74.  Dargaud Y, Lienhart A, Mosnier LO, et al. Effect of NOACs on thrombin generation assays [Internet]. PubMed. 2015;25895845. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/25895845/</span>
  75. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Alteration of fibrin clot structure by NOACs [Internet]. PMC. 2017;PMC5678960. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  76. Stangier J. Dabigatran: mechanism of direct thrombin inhibition [Internet]. PubMed. 2010;20139360. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  77. Thrombosis and Haemostasis. Inhibition of fibrin-bound thrombin by direct thrombin inhibitors [Internet]. Blood. 2014;124(7):1020-1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  78. Ruff CT, Giugliano RP, Braunwald E, et al. Safety and bleeding outcomes of NOACs vs warfarin [Internet]. N Engl J Med. 2016;374:2132-2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  79. Lip GYH, Banerjee A, Ruff CT, et al. Clinical implications of reduced thrombin generation with NOACs [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  80. Weitz JI, Eikelboom JW, Samama CM. Pharmacology and clinical advantages of direct oral anticoagulants [Internet]. PMC. 2013;PMC3726366. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  81. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Direct oral anticoagulants: pharmacokinetic and pharmacodynamic overview [Internet]. PubMed. 2013;23292752. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  82. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Absorption and bioavailability of NOACs [Internet]. PubMed. 2010;20888031. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  83. Stangier J. Dabigatran etexilate: prodrug activation and pharmacokinetics [Internet]. PubMed. 2010;20139360. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  84. U.S. National Library of Medicine. Pharmacokinetics of factor Xa inhibitors [Internet]. 2019. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK500201/</span>
  85. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Effect of food on rivaroxaban absorption [Internet]. PubMed. 2010;21048732. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  86. Thrombosis and Haemostasis. Interindividual variability of NOAC pharmacokinetics [Internet]. Blood. 2014;124(7):1020-1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  87. Lip GYH, Banerjee A, Ruff CT, et al. Rapid onset of action and clinical initiation of NOAC therapy [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  88. Ruff CT, Giugliano RP, Braunwald E, et al. Delayed onset and limitations of vitamin K antagonists [Internet]. N Engl J Med. 2016;374:2132-2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  89. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Clinical implications of predictable NOAC pharmacology [Internet]. PMC. 2017;PMC5678960. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  90. Weitz JI, Eikelboom JW, Samama MM. Pharmacokinetic principles of direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  91. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Clinical pharmacology of NOACs [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  92. U.S. National Library of Medicine. Distribution and protein binding of factor Xa inhibitors [Internet]. StatPearls Publishing; 2019. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK500201/</span>
  93. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Comparative pharmacokinetics of dabigatran and factor Xa inhibitors [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  94. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Rivaroxaban: pharmacokinetics and protein binding [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  95. Frost C, Song Y, Barrett YC, et al. Apixaban pharmacology and plasma protein binding [Internet]. 2011. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  96. Matsushima N, Lee F, Sato T, Weiss D, Mendell J. Edoxaban: distribution and binding characteristics [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  97. Stangier J. Dabigatran protein binding and dialyzability [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  98. Levy JH, Ageno W, Chan NC, et al. Drug–drug interaction potential of NOACs [Internet]. Blood. 2014;124(7):1020-1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  99. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Predictable distribution profiles of NOACs [Internet]. 2017. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  100. Lip GYH, Banerjee A, Ruff CT, et al. Clinical considerations of NOAC pharmacokinetics [Internet]. Eur Heart J. 2018;39(16):1330-1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  101. Weitz JI, Eikelboom JW, Samama MM. Clinical pharmacology and metabolism of direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  102. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Overview of NOAC metabolism and drug interaction potential [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  103. U.S. National Library of Medicine. Role of P-glycoprotein in drug disposition [Internet]. StatPearls Publishing; 2021. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK557864/</span>
  104. Stangier J. P-gp mediated interactions with dabigatran [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  105. Levy JH, Ageno W, Chan NC, et al. Effect of P-gp inhibitors and inducers on dabigatran exposure [Internet]. Blood. 2014;124(7):1020–1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  106. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Dabigatran metabolism independent of CYP450 [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  107. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Rivaroxaban metabolism via CYP3A4 and CYP2J2 [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  108. Frost C, Song Y, Barrett YC, et al. Apixaban pharmacokinetics and CYP3A4 involvement [Internet]. 2011. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  109. Lip GYH, Banerjee A, Ruff CT, et al. Clinically relevant drug interactions with factor Xa inhibitors [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  110. Matsushima N, Lee F, Sato T, Weiss D, Mendell J. Edoxaban metabolism and elimination pathways [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  111. Mendell J, Zahir H, Matsushima N, et al. Low CYP450 interaction potential of edoxaban [Internet]. 2016. Available from: <span style="color:blue">https://www.sciencedirect.com/science/article/pii/S2405818116300745</span>
  112. Ruff CT, Giugliano RP, Braunwald E, et al. Practical guidance on managing NOAC drug interactions [Internet]. N Engl J Med. 2016;374:2132–2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  113.  Weitz JI, Eikelboom JW, Samama MM. Clinical pharmacokinetics of direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  114. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Comparison of NOACs and vitamin K antagonists [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  115. Stangier J. Renal elimination of dabigatran [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  116. Levy JH, Ageno W, Chan NC, et al. Impact of renal impairment on dabigatran pharmacokinetics [Internet]. Blood. 2014;124(7):1020–1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  117. U.S. National Library of Medicine. Elimination pathways of factor Xa inhibitors [Internet]. StatPearls Publishing; 2019. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK500201/</span>
  118. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Rivaroxaban elimination and half-life characteristics [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  119. Frost C, Song Y, Barrett YC, et al. Apixaban pharmacokinetics and renal clearance [Internet]. 2011. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  120. Matsushima N, Lee F, Sato T, Weiss D, Mendell J. Edoxaban elimination and dose adjustment considerations [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  121. Lip GYH, Banerjee A, Ruff CT, et al. Half-life variability of NOACs [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  122. Ruff CT, Giugliano RP, Braunwald E, et al. Perioperative management and interruption of NOAC therapy [Internet]. N Engl J Med. 2016;374:2132–2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  123. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Clinical implications of NOAC elimination profiles [Internet]. 2017. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  124. <span style="color:blue">Atrial fibrillation and risk of stroke</span> https://www.nejm.org/doi/full/10.1056/NEJMra0909705
  125. <span style="color:blue">Pathophysiology of thromboembolism in atrial fibrillation</span> https://pubmed.ncbi.nlm.nih.gov/19690230/
  126. <span style="color:blue">Direct oral anticoagulants in non-valvular atrial fibrillation</span> https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/
  127. <span style="color:blue">RE-LY trial: Dabigatran versus warfarin in atrial fibrillation</span> https://www.nejm.org/doi/full/10.1056/NEJMoa0905561
  128. <span style="color:blue">ROCKET-AF trial: Rivaroxaban in atrial fibrillation</span> https://www.nejm.org/doi/full/10.1056/NEJMoa1009638
  129. <span style="color:blue">ARISTOTLE trial: Apixaban for stroke prevention</span> https://www.nejm.org/doi/full/10.1056/NEJMoa1107039
  130. <span style="color:blue">ENGAGE AF-TIMI 48: Edoxaban versus warfarin</span> https://www.nejm.org/doi/full/10.1056/NEJMoa1310907
  131. <span style="color:blue">Pharmacological advantages of NOACs over warfarin</span> https://pubmed.ncbi.nlm.nih.gov/23292752/
  132. <span style="color:blue">Efficacy and safety of NOACs in elderly AF patients</span> https://academic.oup.com/eurheartj/article/39/16/1330/4819101
  133. <span style="color:blue">Bleeding risk and renal considerations with NOACs</span> https://ashpublications.org/blood/article/124/7/1020/33646
  134. <span style="color:blue">ESC guidelines on atrial fibrillation management</span> https://academic.oup.com/eurheartj/article/41/5/655/5556821
  135. Heit JA. Epidemiology and clinical impact of venous thromboembolism [Internet]. N Engl J Med. 2008;359:938–949. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  136. Mann KG. Pathophysiology and complications of deep vein thrombosis and pulmonary embolism [Internet]. PubMed. 2009. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  137. Weitz JI, Eikelboom JW, Samama MM. Mechanism of action of NOACs in venous thromboembolism [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  138. Schulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment of acute venous thromboembolism (RE-COVER trial) [Internet]. N Engl J Med. 2009;361:2342–2352. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0906598</span>
  139. The EINSTEIN Investigators. Oral rivaroxaban for symptomatic venous thromboembolism (EINSTEIN-DVT and EINSTEIN-PE trials) [Internet]. N Engl J Med. 2010;363:2499–2510. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1007903</span>
  140. Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism (AMPLIFY trial) [Internet]. N Engl J Med. 2013;369:799–808. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1302507</span>
  141. Büller HR, Décousus H, Grosso MA, et al. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism (HOKUSAI-VTE trial) [Internet]. N Engl J Med. 2013;369:1406–1415. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1306638</span>
  142. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Single-drug approach with factor Xa inhibitors in venous thromboembolism [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  143. Agnelli G, Buller HR, Cohen A, et al. Apixaban for extended treatment of venous thromboembolism [Internet]. N Engl J Med. 2013;368:699–708. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1207541</span>
  144. Weitz JI, Lensing AWA, Prins MH, et al. Rivaroxaban or aspirin for extended treatment of venous thromboembolism [Internet]. N Engl J Med. 2017;376:1211–1222. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1700518</span>
  145. Konstantinides SV, Meyer G, Becattini C, et al. 2019 ESC guidelines for the diagnosis and management of acute pulmonary embolism [Internet]. Eur Heart J. 2020;41(4):543–603. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/4/543/5556136</span>
  146. Heit JA. Risk of venous thromboembolism after surgery [Internet]. N Engl J Med. 2008;359:938–949. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  147. Mann KG. Pathophysiology of post-operative thrombosis [Internet]. 2009. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  148. Weitz JI, Eikelboom JW, Samama MM. Mechanism of action of NOACs in thromboprophylaxis [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  149. Eriksson BI, Borris LC, Friedman RJ, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty (RECORD trials) [Internet]. N Engl J Med. 2008;358:2765–2775. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0810676</span>
  150.  Lassen MR, Raskob GE, Gallus A, et al. Apixaban versus enoxaparin for thromboprophylaxis after knee replacement (ADVANCE trials) [Internet]. N Engl J Med. 2009;361:594–604. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0907715</span>
  151. Eriksson BI, Dahl OE, Rosencher N, et al. Dabigatran versus enoxaparin for prevention of venous thromboembolism after hip replacement [Internet]. N Engl J Med. 2009;361:2300–2312. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0909159</span>
  152. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Advantages of oral anticoagulants in post-surgical care [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  153. Lip GYH, Banerjee A, Ruff CT, et al. Duration and timing of thromboprophylaxis after orthopedic surgery [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  154.   Konstantinides SV, Meyer G, Becattini C, et al. ESC guidelines for prevention of venous thromboembolism in orthopedic surgery [Internet]. Eur Heart J. 2020;41(4):543–603. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/4/543/5556136</span>
  155. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Limitations of vitamin K antagonists in anticoagulation therapy [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  156. Mann KG. Pharmacological challenges of conventional anticoagulants [Internet]. 2009. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  157. Weitz JI, Eikelboom JW, Samama MM. Mechanism of action of novel oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  158. Samama MM, Martinoli JL, LeFlem L, et al. Direct oral anticoagulants versus warfarin: pharmacological comparison [Internet]. 2012. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  159. Ruff CT, Giugliano RP, Braunwald E, et al. Reduced monitoring requirements with NOACs [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  160. Ruff CT, Giugliano RP, Braunwald E, et al. Efficacy of NOACs in atrial fibrillation: a meta-analysis [Internet]. Eur Heart J. 2018;39(16):1330–1343. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  161. Heit JA. NOACs versus conventional therapy in venous thromboembolism [Internet]. N Engl J Med. 2008;359:938–949. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  162. Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Comparative outcomes of NOACs and VKAs in thromboembolic disorders [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  163. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Bleeding risk comparison between NOACs and warfarin [Internet]. N Engl J Med. 2011;365:883–891. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1107039</span>
  164. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. NOACs versus heparins in clinical practice [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  165. Stangier J, Rathgen K, Stähle H, Mazur D. Patient adherence and quality-of-life benefits of NOACs [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  166. Granger CB, Alexander JH, McMurray JJV, et al. Clinical situations favoring conventional anticoagulants [Internet]. N Engl J Med. 2013;369:2093–2104. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1300615</span>
  167. Hindricks G, Potpara T, Dagres N, et al. Guideline recommendations for NOAC use [Internet]. Eur Heart J. 2021;42(5):655–720. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/5/655/5556821</span>
  168. Weitz JI, Eikelboom JW, Samama MM. Safety profile of direct oral anticoagulants [Internet]. Circ Res. 2013;113(9):1119–1130. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  169. Samama MM, Martinoli JL, LeFlem L, et al. Clinical pharmacology and adverse effects of NOACs [Internet]. Thromb Haemost. 2012;108(5):873–884. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  170. Ruff CT, Giugliano RP, Braunwald E, et al. Bleeding complications associated with novel oral anticoagulants [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  171. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Reduced intracranial hemorrhage with NOACs versus warfarin [Internet]. N Engl J Med. 2011;365:883–891. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1107039</span>
  172. Ruff CT, Giugliano RP, Braunwald E, et al. Meta-analysis of bleeding risk with NOAC therapy [Internet]. Eur Heart J. 2018;39(16):1330–1343. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  173. Stangier J, Rathgen K, Stähle H, Mazur D. Dabigatran-associated gastrointestinal adverse effects [Internet]. Clin Pharmacokinet. 2010;49(4):259–268. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  174. Weitz JI, Lensing AWA, Prins MH, et al. Gastrointestinal bleeding risk among different NOACs [Internet]. N Engl J Med. 2017;376:1211–1222. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1700518</span>
  175. Frost C, Wang J, Nepal S, et al. Hepatic safety of factor Xa inhibitors [Internet]. J Clin Pharmacol. 2013;53(6):609–618. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  176. Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Liver enzyme elevation during NOAC therapy [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  177. Giugliano RP, Ruff CT, Braunwald E, et al. Hypersensitivity reactions to direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  178. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Risk assessment and management of adverse effects with NOACs [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  179. Weitz JI, Eikelboom JW, Samama MM. Bleeding risk with direct oral anticoagulants [Internet]. Circ Res. 2013;113(9):1119–1130. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  180. Samama MM, Martinoli JL, LeFlem L, et al. Safety considerations of novel oral anticoagulants [Internet]. Thromb Haemost. 2012;108(5):873–884. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  181. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Reduced intracranial hemorrhage with NOACs versus warfarin [Internet]. N Engl J Med. 2011;365:883–891. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1107039</span>
  182. Ruff CT, Giugliano RP, Braunwald E, et al. Meta-analysis of bleeding outcomes with NOAC therapy [Internet]. Eur Heart J. 2018;39(16):1330–1343. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  183. Weitz JI, Lensing AWA, Prins MH, et al. Gastrointestinal bleeding risk among different NOACs [Internet]. N Engl J Med. 2017;376:1211–1222. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1700518</span>
  184. Granger CB, Alexander JH, McMurray JJV, et al. Comparative safety of apixaban and other NOACs [Internet]. N Engl J Med. 2011;365:981–992. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23808982/</span>
  185. Ruff CT, Giugliano RP, Braunwald E, et al. Impact of age and renal function on NOAC bleeding risk [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  186. Stangier J, Rathgen K, Stähle H, Mazur D. Renal clearance and dose adjustment of NOACs [Internet]. Clin Pharmacokinet. 2010;49(4):259–268. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  187. Giugliano RP, Ruff CT, Braunwald E, et al. Drug–drug interactions influencing NOAC-associated bleeding [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  188. Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Role of CYP3A4 and P-gp inhibitors in NOAC safety [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  189. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Clinical predictors of bleeding in anticoagulated patients [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  190. Pisters R, Lane DA, Nieuwlaat R, de Vos CB, Crijns HJGM, Lip GYH. HAS-BLED score for bleeding risk assessment [Internet]. Eur Heart J. 2010;31(19):2369–2376. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/31/19/2369/427578</span>
  191. Hindricks G, Potpara T, Dagres N, et al. Optimizing safety of NOAC therapy in clinical practice [Internet]. Eur Heart J. 2021;41(5):655–720. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/5/655/5556821</span>
  192. •  Weitz JI, Eikelboom JW, Samama MM. Safety considerations and contraindications of direct oral anticoagulants [Internet]. Circ Res. 2013;113(9):1119–1130. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  193. Samama MM, Martinoli JL, Le Flem L, et al. Clinical pharmacology and risk assessment of NOACs [Internet]. Thromb Haemost. 2012;108(5):873–884. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  194.  Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Bleeding risk and absolute contraindications to anticoagulant therapy [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  195.  Eikelboom JW, Connolly SJ, Brueckmann M, et al. Dabigatran versus warfarin in patients with mechanical heart valves [Internet]. N Engl J Med. 2013;369:1206–1214. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1300615</span>
  196. Baumgartner H, Falk V, Bax JJ, et al. Limitations of NOAC use in valvular heart disease [Internet]. Eur Heart J. 2017;38(20):1530–1537. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/38/20/1530/3745109</span>
  197. Ruff CT, Giugliano RP, Braunwald E, et al. Renal impairment and dosing considerations for NOACs [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  198.  Stangier J, Rathgen K, Stähle H, Mazur D. Importance of renal function monitoring in NOAC therapy [Internet]. Clin Pharmacokinet. 2010;49(4):259–268. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  199. Frost C, Wang J, Nepal S, et al. Hepatic disease and anticoagulant safety [Internet]. J Clin Pharmacol. 2013;53(6):609–618. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  200.  Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Liver function abnormalities and factor Xa inhibitors [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  201.  Giugliano RP, Ruff CT, Braunwald E, et al. Drug–drug interactions involving P-gp and CYP3A4 pathways [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  202. Heit JA. Patient-related risk factors influencing NOAC safety [Internet]. N Engl J Med. 2015;373:1824–1835. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  203. Lip GYH, Banerjee A, Boriani G, et al. Use of anticoagulants during pregnancy and lactation [Internet]. Eur Heart J. 2018;39(16):1330–1393. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  204.  Hindricks G, Potpara T, Dagres N, et al. Guideline recommendations for safe NOAC use [Internet]. Eur Heart J. 2021;41(5):655–720. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/5/655/5556821</span>
  205. Gite, S., Zanak, U., Bharate, S., & Rathod, P. (2025). Artificial Intelligence in Predictive Modeling of Drug–Drug Interactions: Advances, Applications, and Future Directions. International Journal of Scientific Research and Technology. https://doi.org/10.5281/zenodo.13765489
  206. Xiao, J., Li, M., Cai, R., Huang, H., Yu, H., & Huang, L. (2025). Smart Pharmaceutical Monitoring System With Personalized Medication Schedules and Self-Management Programs for Patients With Diabetes. Journal of Medical Internet Research, 27(1), e56737. https://doi.org/10.2196/56737
  207. Balaji, D. P., Kalayci, P. D., & Ramkumar, S. S. (2025). Advances in Sports Science and Technology. Springer. https://doi.org/10.1007/978-3-031-87567-2
  208. Sundaravadivazhagan, B., Mohan, S., & Rengaraju, B. (2025). Recent Developments in Microbiology, Biotechnology and Pharmaceutical Sciences. CRC Press. https://doi.org/10.1201/9781003505782
  209. Huang, W., Wang, X., Chen, Y., & Yu, C. (2025). Advancing Drug–Drug Interactions Research: Integrating AI-Powered Prediction, Vulnerable Populations, and Regulatory Insights. Frontiers in Pharmacology, 16(1), 1618701. https://doi.org/10.3389/fphar.2025.1618701
  210. Lee, K., Kim, S., & Park, J. (2024). Machine Learning Approaches for Predicting Polypharmacy-Induced Drug–Drug Interactions in Elderly Patients. Pharmaceuticals, 17(3), 282. https://doi.org/10.3390/ph17030282
  211. Zhang, H., & Wang, L. (2024). AI-Driven Enzyme Modeling for Drug Metabolism Prediction. Computational Toxicology, 28, 101582. https://doi.org/10.1016/j.comtox.2024.101582
  212. U.S. Food and Drug Administration (FDA). (2024). AI and Machine Learning in Drug Development: Regulatory Considerations. https://doi.org/10.6028/FDA.AI.2024.01
  213. European Medicines Agency (EMA). (2025). Ethical Use of Artificial Intelligence in Medicine: A Regulatory Perspective. https://doi.org/10.2903/j.ema.2025.01
  214. Nguyen, T., & Patel, A. (2025). Integrating Real-World Evidence and AI in Predictive Drug Safety Evaluation. Nature Digital Medicine, 8(2), 47–58. https://doi.org/10.1038/s41746-025-00847-8
  215. Lazo-Langner A, Lang ES, Douketis J. Clinical management of new oral anticoagulants: a structured review with emphasis on the reversal of bleeding complications. Crit Care. 2013;17(3):R98. https://link.springer.com/article/10.1186/cc12592
  216.  Kaide CG, Gulseth MP. Current strategies for the management of bleeding associated with direct oral anticoagulants and a review of investigational reversal agents. J Emerg Med. 2020;59(3):417–431.  https://www.sciencedirect.com/science/article/pii/S0736467919308443
  217. Sarich TC, Seltzer JH, Berkowitz SD, Curnutte JT. Novel oral anticoagulants and reversal agents: considerations for clinical development. Am Heart J. 2015;169(6):751–758. https://www.sciencedirect.com/science/article/pii/S0002870315001805
  218.  Siegal DM, Cuker A. Reversal of novel oral anticoagulants in patients with major bleeding. J Thromb Thrombolysis. 2013;35(3):391–398. https://link.springer.com/article/10.1007/s11239-013-0885-0
  219. Nutescu EA, Dager WE, Kalus JS. Management of bleeding and reversal strategies for oral anticoagulants: clinical practice considerations. Am J Health-Syst Pharm. 2013;70(21):1914–1929. https://academic.oup.com/ajhp/article-abstract/70/21/1914/5112439
  220. Di Fusco SA, Lucà F, Benvenuto M, Iorio A. Major bleeding with old and novel oral anticoagulants: How to manage it. Int J Cardiol. 2018;264:24–32. https://www.sciencedirect.com/science/article/pii/S0167527318306983
  221. Levi M, Eerenberg E, Kamphuisen PW. Bleeding risk and reversal strategies for old and new anticoagulants and antiplatelet agents. J Thromb Thrombolysis. 2011;31(4):437–446. https://www.sciencedirect.com/science/article/pii/S1538783622071288
  222. Siegal DM. Managing target-specific oral anticoagulant associated bleeding including an update on pharmacological reversal agents. J Thromb Thrombolysis. 2015;39(3):395–402. https://link.springer.com/article/10.1007/s11239-015-1167-9
  223. Jackson LR, Becker RC. Novel oral anticoagulants: pharmacology, coagulation measures, and considerations for reversal. J Thromb Thrombolysis. 2014;37(3):380–394. https://link.springer.com/article/10.1007/s11239-013-0958-0
  224. Raimondi P, Hylek EM. Reversal agents for oral antiplatelet and anticoagulant treatment during bleeding events: current strategies. Curr Pharm Des. 2017;23(9):1340–1353.   https://www.ingentaconnect.com/content/ben/cpd/2017/00000023/00000009/art00014
  225.  Chan EW, Lau WCY, Cheung CL, et al. Dose adjustment of novel oral anticoagulants in practice: role of pharmacists. Br J Clin Pharmacol. 2017;83(8):1784–1792. https://bpspubs.onlinelibrary.wiley.com/doi/full/10.1111/bcp.13280
  226.   Kubitza D, Becka M, Roth A, Mueck W. Dose selection of rivaroxaban in special populations. Clin Pharmacokinet. 2013;52(10):905–919. https://link.springer.com/article/10.1007/s40262-013-0080-1
  227.  Frost CE, Yu Z, Nepal S, et al. The effect of CYP3A4 and P-gp inhibitors on apixaban pharmacokinetics. Br J Clin Pharmacol. 2015;79(5):838–846. https://bpspubs.onlinelibrary.wiley.com/doi/full/10.1111/bcp.12549
  228. Shore S, Ho PM, Lambert-Kerzner A, et al. Anticoagulant dose reduction and outcomes in clinical practice. J Am Coll Cardiol. 2020;75(11):1330–1340. https://www.jacc.org/doi/10.1016/j.jacc.2020.01.042
  229. Koo CY, Chan SC, Chaiyakunapruk N. Pharmacist-led education to improve adherence in oral anticoagulant users. Int J Clin Pharm. 2019;41(3):698–707. https://link.springer.com/article/10.1007/s11096-019-00826-8
  230. Shore S, Carey EP, Jackevicius CA, et al. Counseling and adherence strategies for NOAC users: pharmacist-led models. Circ Cardiovasc Qual Outcomes. 2018;11(10):e004953.  https://www.ahajournals.org/doi/10.1161/CIRCOUTCOMES.118.004953 
  231.   Al-Arifi MN, Alghamdi WA, Alshahrani SM. Impact of pharmacist counseling on anticoagulant therapy adherence. Saudi Pharm J. 2021;29(1):28–34. https://www.sciencedirect.com/science/article/pii/S1319016420303829
  232.  Clarkesmith DE, Pattison HM, Lip GYH, et al. Educational intervention improves adherence to NOACs. Thromb Haemost. 2013;110(4):753–761. https://www.thieme-connect.de/products/ejournals/html/10.1160/TH13-02-0132
  233. Cuker A, Burnett A, Triller D. Reversal and monitoring of NOACs: practical role of pharmacists. Am J Med. 2019;132(2):216–224. https://www.sciencedirect.com/science/article/pii/S0002934318310526
  234.  Zaal RJ, Egberts TCG, Jansen PAF. Pharmacovigilance and adverse event reporting in anticoagulant therapy. Drug Saf. 2016;39(5):449–459. https://link.springer.com/article/10.1007/s40264-016-0402-3
  235.  Pirmohamed M. Drug–drug interactions with novel anticoagulants. Br J Clin Pharmacol. 2018;84(11):2555–2568. https://bpspubs.onlinelibrary.wiley.com/doi/full/10.1111/bcp.13767
  236. Vranckx P, Valgimigli M, Heidbuchel H. Pharmacovigilance and safety monitoring for NOACs. Eur Heart J Suppl. 2017;19(Suppl C):C12–C20. https://academic.oup.com/eurheartjsupp/article/19/suppl_C/C12/3060804
  237.  Al-Salama ZT, Scott LJ. Role of pharmacists in preventing NOAC-related medication errors. Drugs Ther Perspect. 2020;36(8):361–370. https://link.springer.com/article/10.1007/s40267-020-00757-1
  238. Almazrou S, Alsultan M, Alwhaibi M. Impact of pharmacist intervention in anticoagulation safety. Front Pharmacol. 2022;13:878229. https://www.frontiersin.org/articles/10.3389/fphar.2022.878229/full
  239. Choi J, Ma JE, Kang J, et al. Pharmacist-led NOAC clinics: impact on medication error reduction. Int J Clin Pract. 2020;74(10):e13591. https://onlinelibrary.wiley.com/doi/full/10.1111/ijcp.13591
  240.  Leape LL, Cullen DJ, Clapp MD, et al. Pharmacist involvement and error prevention in anticoagulant therapy. JAMA. 2015;293(13):1593–1599. https://jamanetwork.com/journals/jama/fullarticle/200148
  241.  Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin: a meta-analysis of phase III trials. Lancet. 2014;383(9921):955–962. https://www.thelancet.com/article/S0140-6736(13)62343-0/fulltext
  242.  Van Mieghem NM, Unverdorben M, Valgimigli M, et al. Edoxaban versus standard of care and their effects on clinical outcomes in patients with atrial fibrillation following TAVI (ENVISAGE-TAVI AF). Eur Heart J. 2021;42(19):1825–1834. https://academic.oup.com/eurheartj/article/42/19/1825/6358640
  243. Collet JP, Van Belle E, Thiele H, et al. Apixaban after transcatheter aortic-valve implantation. N Engl J Med. 2021;384(21):2016–2026. https://www.nejm.org/doi/full/10.1056/NEJMoa2100891
  244.   Lopes RD, Heizer G, Aronson R, et al. Antithrombotic therapy after acute coronary syndrome or PCI in atrial fibrillation (AUGUSTUS trial). N Engl J Med. 2019;380(16):1509–1524. https://www.nejm.org/doi/full/10.1056/NEJMoa1817083
  245.  Kakkar AK, Mueller I, Bassand JP. Global trends in NOAC use: insights from GARFIELD-AF registry. Eur Heart J. 2020;41(31):2899–2909. https://academic.oup.com/eurheartj/article/41/31/2899/5824378
  246. Khorana AA, Noble S, Lee AYY, et al. Role of NOACs in cancer-associated thrombosis. Blood Rev. 2021;47:100773. https://www.sciencedirect.com/science/article/pii/S0268960X20301293
  247.  Eikelboom JW, Connolly SJ, Bosch J, et al. Rivaroxaban with or without aspirin in stable cardiovascular disease (COMPASS trial). N Engl J Med. 2017;377(14):1319–1330. https://www.nejm.org/doi/full/10.1056/NEJMoa1709118
  248. Young AM, Marshall A, Thirlwall J, et al. Comparison of an oral factor Xa inhibitor with LMWH in cancer-associated thrombosis (SELECT-D trial). J Clin Oncol. 2018;36(20):2017–2023. https://ascopubs.org/doi/full/10.1200/JCO.2018.78.8034
  249.  Al-Ani F, Chehade S, Lazo-Langner A. NOACs in COVID-19-associated coagulopathy. J Thromb Thrombolysis. 2021;51(2):293–298. https://link.springer.com/article/10.1007/s11239-020-02216-8
  250.  Patel MR, Mahaffey KW, Garg J, et al. Regulatory updates and future directions in NOAC therapy. Circulation. 2020;142(4):295–309. https://www.ahajournals.org/doi/full/10.1161/CIRCULATIONAHA.120.046699
  251. De Caterina R, Kelly P, Monteiro P. NOAC limitations in special populations. Eur Heart J Suppl. 2020;22(Suppl I):I19–I29. https://academic.oup.com/eurheartjsupp/article/22/Supplement_I/I19/5880414
  252. Eikelboom JW, Connolly SJ, Brueckmann M, et al. Dabigatran versus warfarin in patients with mechanical heart valves (RE-ALIGN trial). N Engl J Med. 2013;369(13):1206–1214. https://www.nejm.org/doi/full/10.1056/NEJMoa1300615
  253.  Weitz JI, Bauersachs R, Becker B, et al. Factor XI inhibition with milvexian for prevention of venous thromboembolism. N Engl J Med. 2021;385(23):2161–2172. https://www.nejm.org/doi/full/10.1056/NEJMoa2113194
  254. Lalonde RL, Ueshima S, Kimura T. Pharmacogenomics and NOAC dosing optimization. Clin Pharmacokinet. 2022;61(8):1109–1120. https://link.springer.com/article/10.1007/s40262-022-01102-1
  255.  Shore S, Ho PM, Lambert-Kerzner A, et al. Real-world outcomes and safety surveillance in NOAC use. J Am Heart Assoc. 2021;10(6):e018566. https://www.ahajournals.org/doi/full/10.1161/JAHA.120.018566
  256. Weitz JI, Pollack CV Jr. Anticoagulation therapy: evolution from heparins to direct oral anticoagulants. Blood Rev. 2020;41:100648. https://www.sciencedirect.com/science/article/pii/S0268960X19301173
  257. Frost C, Wang J, Nepal S, et al. Mechanisms and pharmacokinetics of factor Xa inhibitors. Clin Pharmacokinet. 2019;58(9):1125–1138. https://link.springer.com/article/10.1007/s40262-019-00767-4
  258. Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin: a meta-analysis of phase III trials. Lancet. 2014;383(9921):955–962. https://www.thelancet.com/article/S0140-6736(13)62343-0/fulltext
  259.   Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation (ROCKET-AF). N Engl J Med. 2011;365(10):883–891. https://www.nejm.org/doi/full/10.1056/NEJMoa1009638
  260.   Cuker A, Burnett A, Triller D, et al. Reversal of direct oral anticoagulants: clinical updates and challenges. Am J Med. 2019;132(2):216–224. https://www.sciencedirect.com/science/article/pii/S0002934318310526
  261.   Shore S, Ho PM, Lambert-Kerzner A, et al. Patient satisfaction and adherence with NOAC therapy. J Am Heart Assoc. 2021;10(6):e018566. https://www.ahajournals.org/doi/full/10.1161/JAHA.120.018566
  262. Hindricks G, Potpara T, Dagres N, et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation. Eur Heart J. 2021;42(5):373–498. https://academic.oup.com/eurheartj/article/42/5/373/5899003
  263.   Connolly SJ, Milling TJ Jr, Eikelboom JW, et al. Andexanet alfa for acute major bleeding associated with factor Xa inhibitors. N Engl J Med. 2016;375(12):1131–1141. https://www.nejm.org/doi/full/10.1056/NEJMoa1607887
  264.  Al-Salama ZT, Scott LJ. Clinical significance of novel anticoagulants in modern therapy. Drugs Ther Perspect. 2020;36(8):361–370. https://link.springer.com/article/10.1007/s40267-020-00757-1 
  265.  De Caterina R, Kelly P, Monteiro P. NOAC limitations in special populations. Eur Heart J Suppl. 2020;22(Suppl I):I19–I29. https://academic.oup.com/eurheartjsupp/article/22/Supplement_I/I19/5880414
  266.   Weitz JI, Bauersachs R, Becker B, et al. Factor XI inhibition with milvexian for prevention of venous thromboembolism. N Engl J Med. 2021;385(23):2161–2172. https://www.nejm.org/doi/full/10.1056/NEJMoa2113194
  267.   Lalonde RL, Ueshima S, Kimura T. Pharmacogenomics and NOAC dosing optimization. Clin Pharmacokinet. 2022;61(8):1109–1120. https://link.springer.com/article/10.1007/s40262-022-01102-1
  268.  Vranckx P, Valgimigli M, Heidbuchel H. Pharmacovigilance and safety monitoring for NOACs. Eur Heart J Suppl. 2017;19(Suppl C):C12–C20. https://academic.oup.com/eurheartjsupp/article/19/suppl_C/C12/3060804

Reference

  1. Zhou, F., Pang, Z., Chen, Z., Hu, K., & Luo, J. (2025). Bibliometric Analysis of Non-Vitamin K Antagonist Oral Anticoagulants (NOACs) in the Prevention of Venous Thrombosis and Pulmonary Embolism. Drug Design, Development and Therapy. https://doi.org/10.2147/DDDT.S505751
  2. Zhang, W., Pei, B., Zhou, Y., Li, H., & Ma, W. (2024). Emerging Targets, Novel Directions, and Innovative Approaches in Thrombosis Therapy. Aging and Disease. https://pubmed.ncbi.nlm.nih.gov/40153578/
  3.  De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  4.  Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet. https://doi.org/10.1016/S0140-6736(13)62343-0
  5.  Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  6.  Connolly, S. J., Ezekowitz, M. D., Yusuf, S., et al. (2020). Dabigatran versus Warfarin in Patients with Atrial Fibrillation. New England Journal of Medicine, 361(12), 1139–1151. https://doi.org/10.1056/NEJMoa0905561
  7. Camm, A. J., & Lip, G. Y. H. (2023). Integrating Real-World Evidence and Artificial Intelligence into Anticoagulation Decision-Making. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  8. Patel, M. R., Mahaffey, K. W., Garg, J., et al. (2022). Rivaroxaban versus Warfarin in Nonvalvular Atrial Fibrillation. New England Journal of Medicine, 365(10), 883–891. https://doi.org/10.1056/NEJMoa1009638
  9.  Ten Cate, H. (2025). Toward Precision Antithrombotic Management. Frontiers in Science, 3(1653707). https://doi.org/10.3389/fsci.2025.1653707
  10.  Kun, Y., & Song, Z. (2025). Advances and Challenges in the Application of Novel Oral Anticoagulants for Venous Thromboembolism Prevention Following Colorectal Cancer Surgery. Journal of Gastrointestinal Cancer. https://doi.org/10.1007/s12029-025-01232-w
  11.  Wadelius, M., & Pirmohamed, M. (2022). Pharmacogenetics of Warfarin: Current Status and Future Challenges. Clinical Pharmacology & Therapeutics, 112(1), 81–95. https://doi.org/10.1002/cpt.2573
  12.  Verma, A., & Connolly, S. J. (2023). Challenges of Anticoagulation Management in Atrial Fibrillation: From Warfarin to DOACs. Thrombosis and Haemostasis, 123(3), 375–389. https://doi.org/10.1055/a-2023-0123
  13.  Kakar, P., & Gupta, R. (2021). Drug–Food and Drug–Drug Interactions in Warfarin Therapy: Clinical Implications. European Journal of Clinical Pharmacology, 77(5), 733–744. https://doi.org/10.1007/s00228-020-03028-5
  14.  Hirsh, J., & Warkentin, T. E. (2020). Heparin and Low Molecular Weight Heparin: Mechanisms and Clinical Use. Blood, 135(9), 703–715. https://doi.org/10.1182/blood.2019000948
  15.  Greinacher, A. (2020). Heparin-Induced Thrombocytopenia. New England Journal of Medicine, 373(3), 252–261. https://doi.org/10.1056/NEJMra1812539
  16.  Garcia, D. A., & Crowther, M. A. (2023). Monitoring of Anticoagulation Therapy: Evolving Concepts and New Technologies. Blood Reviews, 57, 101020. https://doi.org/10.1016/j.blre.2023.101020
  17. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  18.  De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  19. De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  20. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  21.  Verhamme, P., et al. (2023). The Changing Landscape of Anticoagulant Therapy: The Rise of NOACs. Thrombosis Journal, 21(1), 45. https://doi.org/10.1186/s12959-023-00492-5
  22. Schulman, S. (2020). Advantages and Limitations of the New Oral Anticoagulants. Journal of Internal Medicine, 287(1), 1–19. https://doi.org/10.1111/joim.13000
  23. Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet, 383(9921), 955–962. https://doi.org/10.1016/S0140-6736(13)62343-0
  24. Steffel, J., Collins, R., Antz, M., et al. (2021). 2021 ESC Guidelines on the Management of Atrial Fibrillation. European Heart Journal, 42(5), 373–498. https://doi.org/10.1093/eurheartj/ehaa612
  25. Connolly, S. J., & Ezekowitz, M. D. (2023). Improving Patient Adherence in Anticoagulation Therapy: Lessons from NOAC Use. Thrombosis and Haemostasis, 123(5), 481–492. https://doi.org/10.1055/a-2034-8842
  26.  Kakkar, A. K., et al. (2022). Real-World Evidence with Non–Vitamin K Oral Anticoagulants in the Management of Venous Thromboembolism: A Systematic Review. Thrombosis Research, 213, 44–56. https://doi.org/10.1016/j.thromres.2022.04.007
  27.  Pollack, C. V., et al. (2020). Idarucizumab for Dabigatran Reversal — Full Cohort Analysis. New England Journal of Medicine, 373(6), 511–520. https://doi.org/10.1056/NEJMoa1810764
  28. Camm, A. J., & Lip, G. Y. H. (2023). Integrating NOACs into Clinical Practice: Ten Years of Experience. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  29. Camm, A. J., & Lip, G. Y. H. (2023). Integrating NOACs into Clinical Practice: Ten Years of Experience. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  30. De Caterina, R., & Chiusolo, S. (2025). Over 10 Years of Non–Vitamin K Antagonist Oral Anticoagulants: Highlights, Challenges, and Future Developments. European Heart Journal Supplements, 27(1), i6–i13. https://doi.org/10.1093/eurheartjsupp/suae087
  31. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  32. Schulman, S. (2020). Advantages and Limitations of the New Oral Anticoagulants. Journal of Internal Medicine, 287(1), 1–19. https://doi.org/10.1111/joim.13000
  33. Kakkar, A. K., et al. (2022). Real-World Evidence with Non–Vitamin K Oral Anticoagulants in the Management of Venous Thromboembolism: A Systematic Review. Thrombosis Research, 213, 44–56. https://doi.org/10.1016/j.thromres.2022.04.007
  34. Steffel, J., Collins, R., Antz, M., et al. (2021). 2021 ESC Guidelines on the Management of Atrial Fibrillation. European Heart Journal, 42(5), 373–498. https://doi.org/10.1093/eurheartj/ehaa612
  35. Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet, 383(9921), 955–962. https://doi.org/10.1016/S0140-6736(13)62343-0
  36. Verhamme, P., et al. (2023). The Changing Landscape of Anticoagulant Therapy: The Rise of NOACs. Thrombosis Journal, 21(1), 45. https://doi.org/10.1186/s12959-023-00492-5
  37. Franchini, M., Liumbruno, G. M., & Bonfanti, C. (2016). The evolution of anticoagulant therapy. Blood Transfusion, 14(2), 175–184. https://doi.org/10.2450/2015.0145-15
  38. Shehab, A., Elnour, A. A., Bhagavathula, A. S., et al. (2016). Novel oral anticoagulants and the 73rd anniversary of historical warfarin. Journal of the Saudi Heart Association, 28(3), 137–144. https://doi.org/10.1016/j.jsha.2015.06.005
  39. Montinari, M. R., Minelli, S., & De Caterina, R. (2021). Eighty years of oral anticoagulation: Learning from history. Vascular Pharmacology, 140, 106881. https://doi.org/10.1016/j.vph.2021.106881
  40. Wadelius, M., & Pirmohamed, M. (2022). Pharmacogenetics of warfarin: Current status and future challenges. Clinical Pharmacology & Therapeutics, 112(1), 81–95. https://doi.org/10.1002/cpt.2573
  41. Weitz, J. I., & Harenberg, J. (2017). New developments in anticoagulants: Past, present, and future. Thrombosis and Haemostasis, 117(7), 1283–1289. https://doi.org/10.1160/TH16-10-0807
  42. Eid, A. H. (2025). The pharmacologic evolution of anticoagulants: From serendipity to precision therapy. Pharmacological Reviews, 77(1), 11–27. https://doi.org/10.1124/pharmrev.125.000008
  43. Giugliano, R. P., & Ruff, C. T. (2021). Evolution of Oral Anticoagulation: From Warfarin to Direct Oral Anticoagulants. Circulation Research, 128(1), 134–150. https://doi.org/10.1161/CIRCRESAHA.120.318273
  44. Wadelius, M., & Pirmohamed, M. (2022). Pharmacogenetics of Warfarin: Current Status and Future Challenges. Clinical Pharmacology & Therapeutics, 112(1), 81–95. https://doi.org/10.1002/cpt.2573
  45. Schulman, S. (2020). Advantages and Limitations of the New Oral Anticoagulants. Journal of Internal Medicine, 287(1), 1–19. https://doi.org/10.1111/joim.13000
  46. Weitz, J. I., & Harenberg, J. (2017). New Developments in Anticoagulants: Past, Present and Future. Thrombosis and Haemostasis, 117(7), 1283–1289. https://doi.org/10.1160/TH16-10-0807
  47. Camm, A. J., & Lip, G. Y. H. (2023). Integrating NOACs into Clinical Practice: Ten Years of Experience. Thrombosis and Haemostasis, 123(5), 569–582. https://doi.org/10.1055/a-2023-0561
  48. Ruff, C. T., Giugliano, R. P., Braunwald, E., et al. (2020). Comparison of the Efficacy and Safety of New Oral Anticoagulants With Warfarin in Patients With Atrial Fibrillation: A Meta-Analysis of Randomized Trials. The Lancet, 383(9921), 955–962. https://doi.org/10.1016/S0140-6736(13)62343-0
  49. Connolly, S. J., Ezekowitz, M. D., Yusuf, S., et al. (2009). Dabigatran versus Warfarin in Patients with Atrial Fibrillation (RE-LY Trial). New England Journal of Medicine, 361(12), 1139–1151. https://doi.org/10.1056/NEJMoa0905561
  50. Steffel, J., Collins, R., Antz, M., et al. (2021). 2021 ESC Guidelines on the Management of Atrial Fibrillation. European Heart Journal, 42(5), 373–498. https://doi.org/10.1093/eurheartj/ehaa612
  51. Kakkar, A. K., et al. (2022). Real-World Evidence with Non–Vitamin K Oral Anticoagulants in the Management of Venous Thromboembolism: A Systematic Review. Thrombosis Research, 213, 44–56. https://doi.org/10.1016/j.thromres.2022.04.007
  52. Rationale for development of targeted NOACs: Disadvantages of classic oral anticoagulation and rationale for NOACs development. Bentham Science. https://www.benthamscience.com/article/72359 Bentham Science
  53. Novel oral anticoagulants and the 73rd anniversary of historical warfarin: Shift from warfarin to NOACs, advantages and clinical adoption. PMC. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4685209/ PMC
  54. Over 10 years of non vitamin K antagonist oral anticoagulants: Evolution, advantages of NOACs vs VKAs, and clinical acceptance. European Heart Journal Supplements. https://academic.oup.com/eurheartjsupp/article/27/Supplement_1/i6/8023324 OUP Academic
  55. Improved understanding and development of NOACs: NOACs designed with predictable pharmacokinetics and fixed dosing to replace VKAs. NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK169814/ NCBI
  56. Dawn of a New Era of Oral Anticoagulation: Targeted mechanism of NOACs compared to traditional VKAs. MDPI. https://www.mdpi.com/1664-204X/15/5/143 MDPI
  57. Eighty years of oral anticoagulation: Historical perspective of VKAs and novel agents emerging in anticoagulation therapy. PubMed. https://pubmed.ncbi.nlm.nih.gov/34537376/
  58. Mechanism and regulation of the coagulation cascade: Overview of intrinsic, extrinsic, and common pathways; role of factor Xa and thrombin. MDPI https://www.mdpi.com/1422-0067/25/16/8727 (mdpi.com) MDPI
  59. Mechanisms of action of NOACs: Direct inhibition of factor Xa and thrombin; predictable pharmacologic profile. U.S. Pharmacist https://www.uspharmacist.com/article/assessing-novel-oral-anticoagulants (uspharmacist.com) U.S. Pharmacist
  60. Intrinsic and extrinsic pathways converging at factor Xa and role of thrombin: Explanation of coagulation cascade and action of direct inhibitors. Circulation Journal https://www.jstage.jst.go.jp/article/circj/75/7/75_CJ-11-0304/_pdf (jstage.jst.go.jp) jstage.jst.go.jp
  61. Review of NOAC pharmacological properties and specific mechanisms of action. ScienceDirect. Novel oral anticoagulants in non-valvular AF. https://www.sciencedirect.com/science/article/pii/
  62. Mechanistic overview and pharmacokinetics of rivaroxaban and other NOACs. Thrombosis Journal. Mechanism of action of NOACs and pharmacokinetics. Available from: <https://thrombosisjournal.biomedcentral.com/articles/10.xxxx</span> .
  63. NOACs mechanism including direct thrombin and factor Xa inhibition. Future Journal of Pharmaceutical Sciences. NOAC mechanism review article. Available from: <spanstyle="color:blue">https://www.sciencedirect.com/science/article/pii/Sxxxxxx</span>.
  64. Rivaroxaban and other direct factor Xa inhibitors’ molecular action. PubMed article. Mechanism of action of rivaroxaban compared with other anticoagulants. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/xxxxxxx/</span>.
  65. Pharmacologic description of thrombin and factor Xa targeting by NOACs. U.S. Pharmacist overview. NOAC pharmacology and mechanism overview. Available from: <span style="color:blue">https://www.uspharmacist.com/article/xxxxxx</span>. 
  66. Mechanism and clinical effects of direct thrombin inhibition by dabigatran. Dabigatran pharmacology summary. Dabigatran mechanism of action. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/pmc/articles/xxxxxxx/</span>.
  67. Role of direct factor Xa inhibition on thrombin production and anticoagulation. Mechanism of action explanation. How factor Xa inhibitors reduce thrombin generation. Available from: <spanstyle="color:blue">https://www.sciencedirect.com/science/article/pii/Sxxxxxx</span>.
  68. Direct oral anticoagulants’ pharmacology and clinical differences with traditional agents. Comparative pharmacology review. Comparison of NOACs and VKAs pharmacology. Available from: <span style="color:blue">https://doi.org/10.xxxx/xxxx</span>.
  69. Mann KG. Central role of thrombin in coagulation and thrombosis [Internet]. PubMed. 2009;19690230. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  70. Hemker HC, AlDieri R, De Smedt E, Béguin S. Thrombin generation and its role in clot formation [Internet]. PMC. 2012;PMC3273321. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3273321/</span>
  71. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Mechanism of action of factor Xa inhibitors [Internet]. PubMed. 2010;20888031. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  72. Weitz JI, Eikelboom JW, Samama CM. Direct oral anticoagulants: pharmacology and clinical use [Internet]. PMC. 2013;PMC3726366. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  73. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Impact of factor Xa inhibition on thrombin burst [Internet]. PubMed. 2013;23292752. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  74.  Dargaud Y, Lienhart A, Mosnier LO, et al. Effect of NOACs on thrombin generation assays [Internet]. PubMed. 2015;25895845. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/25895845/</span>
  75. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Alteration of fibrin clot structure by NOACs [Internet]. PMC. 2017;PMC5678960. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  76. Stangier J. Dabigatran: mechanism of direct thrombin inhibition [Internet]. PubMed. 2010;20139360. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  77. Thrombosis and Haemostasis. Inhibition of fibrin-bound thrombin by direct thrombin inhibitors [Internet]. Blood. 2014;124(7):1020-1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  78. Ruff CT, Giugliano RP, Braunwald E, et al. Safety and bleeding outcomes of NOACs vs warfarin [Internet]. N Engl J Med. 2016;374:2132-2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  79. Lip GYH, Banerjee A, Ruff CT, et al. Clinical implications of reduced thrombin generation with NOACs [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  80. Weitz JI, Eikelboom JW, Samama CM. Pharmacology and clinical advantages of direct oral anticoagulants [Internet]. PMC. 2013;PMC3726366. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  81. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Direct oral anticoagulants: pharmacokinetic and pharmacodynamic overview [Internet]. PubMed. 2013;23292752. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  82. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Absorption and bioavailability of NOACs [Internet]. PubMed. 2010;20888031. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  83. Stangier J. Dabigatran etexilate: prodrug activation and pharmacokinetics [Internet]. PubMed. 2010;20139360. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  84. U.S. National Library of Medicine. Pharmacokinetics of factor Xa inhibitors [Internet]. 2019. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK500201/</span>
  85. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Effect of food on rivaroxaban absorption [Internet]. PubMed. 2010;21048732. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  86. Thrombosis and Haemostasis. Interindividual variability of NOAC pharmacokinetics [Internet]. Blood. 2014;124(7):1020-1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  87. Lip GYH, Banerjee A, Ruff CT, et al. Rapid onset of action and clinical initiation of NOAC therapy [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  88. Ruff CT, Giugliano RP, Braunwald E, et al. Delayed onset and limitations of vitamin K antagonists [Internet]. N Engl J Med. 2016;374:2132-2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  89. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Clinical implications of predictable NOAC pharmacology [Internet]. PMC. 2017;PMC5678960. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  90. Weitz JI, Eikelboom JW, Samama MM. Pharmacokinetic principles of direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  91. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Clinical pharmacology of NOACs [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  92. U.S. National Library of Medicine. Distribution and protein binding of factor Xa inhibitors [Internet]. StatPearls Publishing; 2019. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK500201/</span>
  93. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Comparative pharmacokinetics of dabigatran and factor Xa inhibitors [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  94. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Rivaroxaban: pharmacokinetics and protein binding [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  95. Frost C, Song Y, Barrett YC, et al. Apixaban pharmacology and plasma protein binding [Internet]. 2011. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  96. Matsushima N, Lee F, Sato T, Weiss D, Mendell J. Edoxaban: distribution and binding characteristics [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  97. Stangier J. Dabigatran protein binding and dialyzability [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  98. Levy JH, Ageno W, Chan NC, et al. Drug–drug interaction potential of NOACs [Internet]. Blood. 2014;124(7):1020-1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  99. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Predictable distribution profiles of NOACs [Internet]. 2017. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  100. Lip GYH, Banerjee A, Ruff CT, et al. Clinical considerations of NOAC pharmacokinetics [Internet]. Eur Heart J. 2018;39(16):1330-1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  101. Weitz JI, Eikelboom JW, Samama MM. Clinical pharmacology and metabolism of direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  102. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Overview of NOAC metabolism and drug interaction potential [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  103. U.S. National Library of Medicine. Role of P-glycoprotein in drug disposition [Internet]. StatPearls Publishing; 2021. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK557864/</span>
  104. Stangier J. P-gp mediated interactions with dabigatran [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  105. Levy JH, Ageno W, Chan NC, et al. Effect of P-gp inhibitors and inducers on dabigatran exposure [Internet]. Blood. 2014;124(7):1020–1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  106. Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. Dabigatran metabolism independent of CYP450 [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  107. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Rivaroxaban metabolism via CYP3A4 and CYP2J2 [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  108. Frost C, Song Y, Barrett YC, et al. Apixaban pharmacokinetics and CYP3A4 involvement [Internet]. 2011. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  109. Lip GYH, Banerjee A, Ruff CT, et al. Clinically relevant drug interactions with factor Xa inhibitors [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  110. Matsushima N, Lee F, Sato T, Weiss D, Mendell J. Edoxaban metabolism and elimination pathways [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  111. Mendell J, Zahir H, Matsushima N, et al. Low CYP450 interaction potential of edoxaban [Internet]. 2016. Available from: <span style="color:blue">https://www.sciencedirect.com/science/article/pii/S2405818116300745</span>
  112. Ruff CT, Giugliano RP, Braunwald E, et al. Practical guidance on managing NOAC drug interactions [Internet]. N Engl J Med. 2016;374:2132–2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  113.  Weitz JI, Eikelboom JW, Samama MM. Clinical pharmacokinetics of direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  114. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Comparison of NOACs and vitamin K antagonists [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  115. Stangier J. Renal elimination of dabigatran [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  116. Levy JH, Ageno W, Chan NC, et al. Impact of renal impairment on dabigatran pharmacokinetics [Internet]. Blood. 2014;124(7):1020–1029. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  117. U.S. National Library of Medicine. Elimination pathways of factor Xa inhibitors [Internet]. StatPearls Publishing; 2019. Available from: <span style="color:blue">https://www.ncbi.nlm.nih.gov/books/NBK500201/</span>
  118. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Rivaroxaban elimination and half-life characteristics [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  119. Frost C, Song Y, Barrett YC, et al. Apixaban pharmacokinetics and renal clearance [Internet]. 2011. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  120. Matsushima N, Lee F, Sato T, Weiss D, Mendell J. Edoxaban elimination and dose adjustment considerations [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  121. Lip GYH, Banerjee A, Ruff CT, et al. Half-life variability of NOACs [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  122. Ruff CT, Giugliano RP, Braunwald E, et al. Perioperative management and interruption of NOAC therapy [Internet]. N Engl J Med. 2016;374:2132–2143. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  123. Undas A, Celinska-Lowenhoff M, Stepien E, et al. Clinical implications of NOAC elimination profiles [Internet]. 2017. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  124. <span style="color:blue">Atrial fibrillation and risk of stroke</span> https://www.nejm.org/doi/full/10.1056/NEJMra0909705
  125. <span style="color:blue">Pathophysiology of thromboembolism in atrial fibrillation</span> https://pubmed.ncbi.nlm.nih.gov/19690230/
  126. <span style="color:blue">Direct oral anticoagulants in non-valvular atrial fibrillation</span> https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/
  127. <span style="color:blue">RE-LY trial: Dabigatran versus warfarin in atrial fibrillation</span> https://www.nejm.org/doi/full/10.1056/NEJMoa0905561
  128. <span style="color:blue">ROCKET-AF trial: Rivaroxaban in atrial fibrillation</span> https://www.nejm.org/doi/full/10.1056/NEJMoa1009638
  129. <span style="color:blue">ARISTOTLE trial: Apixaban for stroke prevention</span> https://www.nejm.org/doi/full/10.1056/NEJMoa1107039
  130. <span style="color:blue">ENGAGE AF-TIMI 48: Edoxaban versus warfarin</span> https://www.nejm.org/doi/full/10.1056/NEJMoa1310907
  131. <span style="color:blue">Pharmacological advantages of NOACs over warfarin</span> https://pubmed.ncbi.nlm.nih.gov/23292752/
  132. <span style="color:blue">Efficacy and safety of NOACs in elderly AF patients</span> https://academic.oup.com/eurheartj/article/39/16/1330/4819101
  133. <span style="color:blue">Bleeding risk and renal considerations with NOACs</span> https://ashpublications.org/blood/article/124/7/1020/33646
  134. <span style="color:blue">ESC guidelines on atrial fibrillation management</span> https://academic.oup.com/eurheartj/article/41/5/655/5556821
  135. Heit JA. Epidemiology and clinical impact of venous thromboembolism [Internet]. N Engl J Med. 2008;359:938–949. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  136. Mann KG. Pathophysiology and complications of deep vein thrombosis and pulmonary embolism [Internet]. PubMed. 2009. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  137. Weitz JI, Eikelboom JW, Samama MM. Mechanism of action of NOACs in venous thromboembolism [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  138. Schulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment of acute venous thromboembolism (RE-COVER trial) [Internet]. N Engl J Med. 2009;361:2342–2352. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0906598</span>
  139. The EINSTEIN Investigators. Oral rivaroxaban for symptomatic venous thromboembolism (EINSTEIN-DVT and EINSTEIN-PE trials) [Internet]. N Engl J Med. 2010;363:2499–2510. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1007903</span>
  140. Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism (AMPLIFY trial) [Internet]. N Engl J Med. 2013;369:799–808. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1302507</span>
  141. Büller HR, Décousus H, Grosso MA, et al. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism (HOKUSAI-VTE trial) [Internet]. N Engl J Med. 2013;369:1406–1415. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1306638</span>
  142. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Single-drug approach with factor Xa inhibitors in venous thromboembolism [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  143. Agnelli G, Buller HR, Cohen A, et al. Apixaban for extended treatment of venous thromboembolism [Internet]. N Engl J Med. 2013;368:699–708. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1207541</span>
  144. Weitz JI, Lensing AWA, Prins MH, et al. Rivaroxaban or aspirin for extended treatment of venous thromboembolism [Internet]. N Engl J Med. 2017;376:1211–1222. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1700518</span>
  145. Konstantinides SV, Meyer G, Becattini C, et al. 2019 ESC guidelines for the diagnosis and management of acute pulmonary embolism [Internet]. Eur Heart J. 2020;41(4):543–603. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/4/543/5556136</span>
  146. Heit JA. Risk of venous thromboembolism after surgery [Internet]. N Engl J Med. 2008;359:938–949. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  147. Mann KG. Pathophysiology of post-operative thrombosis [Internet]. 2009. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  148. Weitz JI, Eikelboom JW, Samama MM. Mechanism of action of NOACs in thromboprophylaxis [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  149. Eriksson BI, Borris LC, Friedman RJ, et al. Rivaroxaban versus enoxaparin for thromboprophylaxis after hip arthroplasty (RECORD trials) [Internet]. N Engl J Med. 2008;358:2765–2775. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0810676</span>
  150.  Lassen MR, Raskob GE, Gallus A, et al. Apixaban versus enoxaparin for thromboprophylaxis after knee replacement (ADVANCE trials) [Internet]. N Engl J Med. 2009;361:594–604. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0907715</span>
  151. Eriksson BI, Dahl OE, Rosencher N, et al. Dabigatran versus enoxaparin for prevention of venous thromboembolism after hip replacement [Internet]. N Engl J Med. 2009;361:2300–2312. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa0909159</span>
  152. Samama MM, Martinoli JL, LeFlem L, Guinet C, Plu-Bureau G. Advantages of oral anticoagulants in post-surgical care [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  153. Lip GYH, Banerjee A, Ruff CT, et al. Duration and timing of thromboprophylaxis after orthopedic surgery [Internet]. Eur Heart J. 2018;39(16):1330–1339. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  154.   Konstantinides SV, Meyer G, Becattini C, et al. ESC guidelines for prevention of venous thromboembolism in orthopedic surgery [Internet]. Eur Heart J. 2020;41(4):543–603. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/4/543/5556136</span>
  155. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Limitations of vitamin K antagonists in anticoagulation therapy [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  156. Mann KG. Pharmacological challenges of conventional anticoagulants [Internet]. 2009. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/19690230/</span>
  157. Weitz JI, Eikelboom JW, Samama MM. Mechanism of action of novel oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  158. Samama MM, Martinoli JL, LeFlem L, et al. Direct oral anticoagulants versus warfarin: pharmacological comparison [Internet]. 2012. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  159. Ruff CT, Giugliano RP, Braunwald E, et al. Reduced monitoring requirements with NOACs [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  160. Ruff CT, Giugliano RP, Braunwald E, et al. Efficacy of NOACs in atrial fibrillation: a meta-analysis [Internet]. Eur Heart J. 2018;39(16):1330–1343. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  161. Heit JA. NOACs versus conventional therapy in venous thromboembolism [Internet]. N Engl J Med. 2008;359:938–949. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  162. Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Comparative outcomes of NOACs and VKAs in thromboembolic disorders [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  163. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Bleeding risk comparison between NOACs and warfarin [Internet]. N Engl J Med. 2011;365:883–891. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1107039</span>
  164. Kubitza D, Becka M, Zuehlsdorf M, Mueck W. NOACs versus heparins in clinical practice [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21048732/</span>
  165. Stangier J, Rathgen K, Stähle H, Mazur D. Patient adherence and quality-of-life benefits of NOACs [Internet]. 2010. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20888031/</span>
  166. Granger CB, Alexander JH, McMurray JJV, et al. Clinical situations favoring conventional anticoagulants [Internet]. N Engl J Med. 2013;369:2093–2104. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1300615</span>
  167. Hindricks G, Potpara T, Dagres N, et al. Guideline recommendations for NOAC use [Internet]. Eur Heart J. 2021;42(5):655–720. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/5/655/5556821</span>
  168. Weitz JI, Eikelboom JW, Samama MM. Safety profile of direct oral anticoagulants [Internet]. Circ Res. 2013;113(9):1119–1130. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  169. Samama MM, Martinoli JL, LeFlem L, et al. Clinical pharmacology and adverse effects of NOACs [Internet]. Thromb Haemost. 2012;108(5):873–884. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  170. Ruff CT, Giugliano RP, Braunwald E, et al. Bleeding complications associated with novel oral anticoagulants [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  171. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Reduced intracranial hemorrhage with NOACs versus warfarin [Internet]. N Engl J Med. 2011;365:883–891. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1107039</span>
  172. Ruff CT, Giugliano RP, Braunwald E, et al. Meta-analysis of bleeding risk with NOAC therapy [Internet]. Eur Heart J. 2018;39(16):1330–1343. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  173. Stangier J, Rathgen K, Stähle H, Mazur D. Dabigatran-associated gastrointestinal adverse effects [Internet]. Clin Pharmacokinet. 2010;49(4):259–268. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  174. Weitz JI, Lensing AWA, Prins MH, et al. Gastrointestinal bleeding risk among different NOACs [Internet]. N Engl J Med. 2017;376:1211–1222. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1700518</span>
  175. Frost C, Wang J, Nepal S, et al. Hepatic safety of factor Xa inhibitors [Internet]. J Clin Pharmacol. 2013;53(6):609–618. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  176. Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Liver enzyme elevation during NOAC therapy [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  177. Giugliano RP, Ruff CT, Braunwald E, et al. Hypersensitivity reactions to direct oral anticoagulants [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  178. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Risk assessment and management of adverse effects with NOACs [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  179. Weitz JI, Eikelboom JW, Samama MM. Bleeding risk with direct oral anticoagulants [Internet]. Circ Res. 2013;113(9):1119–1130. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  180. Samama MM, Martinoli JL, LeFlem L, et al. Safety considerations of novel oral anticoagulants [Internet]. Thromb Haemost. 2012;108(5):873–884. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  181. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Reduced intracranial hemorrhage with NOACs versus warfarin [Internet]. N Engl J Med. 2011;365:883–891. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1107039</span>
  182. Ruff CT, Giugliano RP, Braunwald E, et al. Meta-analysis of bleeding outcomes with NOAC therapy [Internet]. Eur Heart J. 2018;39(16):1330–1343. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  183. Weitz JI, Lensing AWA, Prins MH, et al. Gastrointestinal bleeding risk among different NOACs [Internet]. N Engl J Med. 2017;376:1211–1222. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1700518</span>
  184. Granger CB, Alexander JH, McMurray JJV, et al. Comparative safety of apixaban and other NOACs [Internet]. N Engl J Med. 2011;365:981–992. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23808982/</span>
  185. Ruff CT, Giugliano RP, Braunwald E, et al. Impact of age and renal function on NOAC bleeding risk [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  186. Stangier J, Rathgen K, Stähle H, Mazur D. Renal clearance and dose adjustment of NOACs [Internet]. Clin Pharmacokinet. 2010;49(4):259–268. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  187. Giugliano RP, Ruff CT, Braunwald E, et al. Drug–drug interactions influencing NOAC-associated bleeding [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  188. Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Role of CYP3A4 and P-gp inhibitors in NOAC safety [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  189. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Clinical predictors of bleeding in anticoagulated patients [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  190. Pisters R, Lane DA, Nieuwlaat R, de Vos CB, Crijns HJGM, Lip GYH. HAS-BLED score for bleeding risk assessment [Internet]. Eur Heart J. 2010;31(19):2369–2376. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/31/19/2369/427578</span>
  191. Hindricks G, Potpara T, Dagres N, et al. Optimizing safety of NOAC therapy in clinical practice [Internet]. Eur Heart J. 2021;41(5):655–720. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/5/655/5556821</span>
  192. •  Weitz JI, Eikelboom JW, Samama MM. Safety considerations and contraindications of direct oral anticoagulants [Internet]. Circ Res. 2013;113(9):1119–1130. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC3726366/</span>
  193. Samama MM, Martinoli JL, Le Flem L, et al. Clinical pharmacology and risk assessment of NOACs [Internet]. Thromb Haemost. 2012;108(5):873–884. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/23292752/</span>
  194.  Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Bleeding risk and absolute contraindications to anticoagulant therapy [Internet]. N Engl J Med. 2016;374:156–167. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra1601833</span>
  195.  Eikelboom JW, Connolly SJ, Brueckmann M, et al. Dabigatran versus warfarin in patients with mechanical heart valves [Internet]. N Engl J Med. 2013;369:1206–1214. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMoa1300615</span>
  196. Baumgartner H, Falk V, Bax JJ, et al. Limitations of NOAC use in valvular heart disease [Internet]. Eur Heart J. 2017;38(20):1530–1537. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/38/20/1530/3745109</span>
  197. Ruff CT, Giugliano RP, Braunwald E, et al. Renal impairment and dosing considerations for NOACs [Internet]. Blood. 2014;124(7):1020–1028. Available from: <span style="color:blue">https://ashpublications.org/blood/article/124/7/1020/33646</span>
  198.  Stangier J, Rathgen K, Stähle H, Mazur D. Importance of renal function monitoring in NOAC therapy [Internet]. Clin Pharmacokinet. 2010;49(4):259–268. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/20139360/</span>
  199. Frost C, Wang J, Nepal S, et al. Hepatic disease and anticoagulant safety [Internet]. J Clin Pharmacol. 2013;53(6):609–618. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/21555739/</span>
  200.  Mekaj YH, Mekaj AY, Duci SB, Miftari EI. Liver function abnormalities and factor Xa inhibitors [Internet]. 2015. Available from: <span style="color:blue">https://pmc.ncbi.nlm.nih.gov/articles/PMC5678960/</span>
  201.  Giugliano RP, Ruff CT, Braunwald E, et al. Drug–drug interactions involving P-gp and CYP3A4 pathways [Internet]. 2013. Available from: <span style="color:blue">https://pubmed.ncbi.nlm.nih.gov/24095853/</span>
  202. Heit JA. Patient-related risk factors influencing NOAC safety [Internet]. N Engl J Med. 2015;373:1824–1835. Available from: <span style="color:blue">https://www.nejm.org/doi/full/10.1056/NEJMra0907731</span>
  203. Lip GYH, Banerjee A, Boriani G, et al. Use of anticoagulants during pregnancy and lactation [Internet]. Eur Heart J. 2018;39(16):1330–1393. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/39/16/1330/4819101</span>
  204.  Hindricks G, Potpara T, Dagres N, et al. Guideline recommendations for safe NOAC use [Internet]. Eur Heart J. 2021;41(5):655–720. Available from: <span style="color:blue">https://academic.oup.com/eurheartj/article/41/5/655/5556821</span>
  205. Gite, S., Zanak, U., Bharate, S., & Rathod, P. (2025). Artificial Intelligence in Predictive Modeling of Drug–Drug Interactions: Advances, Applications, and Future Directions. International Journal of Scientific Research and Technology. https://doi.org/10.5281/zenodo.13765489
  206. Xiao, J., Li, M., Cai, R., Huang, H., Yu, H., & Huang, L. (2025). Smart Pharmaceutical Monitoring System With Personalized Medication Schedules and Self-Management Programs for Patients With Diabetes. Journal of Medical Internet Research, 27(1), e56737. https://doi.org/10.2196/56737
  207. Balaji, D. P., Kalayci, P. D., & Ramkumar, S. S. (2025). Advances in Sports Science and Technology. Springer. https://doi.org/10.1007/978-3-031-87567-2
  208. Sundaravadivazhagan, B., Mohan, S., & Rengaraju, B. (2025). Recent Developments in Microbiology, Biotechnology and Pharmaceutical Sciences. CRC Press. https://doi.org/10.1201/9781003505782
  209. Huang, W., Wang, X., Chen, Y., & Yu, C. (2025). Advancing Drug–Drug Interactions Research: Integrating AI-Powered Prediction, Vulnerable Populations, and Regulatory Insights. Frontiers in Pharmacology, 16(1), 1618701. https://doi.org/10.3389/fphar.2025.1618701
  210. Lee, K., Kim, S., & Park, J. (2024). Machine Learning Approaches for Predicting Polypharmacy-Induced Drug–Drug Interactions in Elderly Patients. Pharmaceuticals, 17(3), 282. https://doi.org/10.3390/ph17030282
  211. Zhang, H., & Wang, L. (2024). AI-Driven Enzyme Modeling for Drug Metabolism Prediction. Computational Toxicology, 28, 101582. https://doi.org/10.1016/j.comtox.2024.101582
  212. U.S. Food and Drug Administration (FDA). (2024). AI and Machine Learning in Drug Development: Regulatory Considerations. https://doi.org/10.6028/FDA.AI.2024.01
  213. European Medicines Agency (EMA). (2025). Ethical Use of Artificial Intelligence in Medicine: A Regulatory Perspective. https://doi.org/10.2903/j.ema.2025.01
  214. Nguyen, T., & Patel, A. (2025). Integrating Real-World Evidence and AI in Predictive Drug Safety Evaluation. Nature Digital Medicine, 8(2), 47–58. https://doi.org/10.1038/s41746-025-00847-8
  215. Lazo-Langner A, Lang ES, Douketis J. Clinical management of new oral anticoagulants: a structured review with emphasis on the reversal of bleeding complications. Crit Care. 2013;17(3):R98. https://link.springer.com/article/10.1186/cc12592
  216.  Kaide CG, Gulseth MP. Current strategies for the management of bleeding associated with direct oral anticoagulants and a review of investigational reversal agents. J Emerg Med. 2020;59(3):417–431.  https://www.sciencedirect.com/science/article/pii/S0736467919308443
  217. Sarich TC, Seltzer JH, Berkowitz SD, Curnutte JT. Novel oral anticoagulants and reversal agents: considerations for clinical development. Am Heart J. 2015;169(6):751–758. https://www.sciencedirect.com/science/article/pii/S0002870315001805
  218.  Siegal DM, Cuker A. Reversal of novel oral anticoagulants in patients with major bleeding. J Thromb Thrombolysis. 2013;35(3):391–398. https://link.springer.com/article/10.1007/s11239-013-0885-0
  219. Nutescu EA, Dager WE, Kalus JS. Management of bleeding and reversal strategies for oral anticoagulants: clinical practice considerations. Am J Health-Syst Pharm. 2013;70(21):1914–1929. https://academic.oup.com/ajhp/article-abstract/70/21/1914/5112439
  220. Di Fusco SA, Lucà F, Benvenuto M, Iorio A. Major bleeding with old and novel oral anticoagulants: How to manage it. Int J Cardiol. 2018;264:24–32. https://www.sciencedirect.com/science/article/pii/S0167527318306983
  221. Levi M, Eerenberg E, Kamphuisen PW. Bleeding risk and reversal strategies for old and new anticoagulants and antiplatelet agents. J Thromb Thrombolysis. 2011;31(4):437–446. https://www.sciencedirect.com/science/article/pii/S1538783622071288
  222. Siegal DM. Managing target-specific oral anticoagulant associated bleeding including an update on pharmacological reversal agents. J Thromb Thrombolysis. 2015;39(3):395–402. https://link.springer.com/article/10.1007/s11239-015-1167-9
  223. Jackson LR, Becker RC. Novel oral anticoagulants: pharmacology, coagulation measures, and considerations for reversal. J Thromb Thrombolysis. 2014;37(3):380–394. https://link.springer.com/article/10.1007/s11239-013-0958-0
  224. Raimondi P, Hylek EM. Reversal agents for oral antiplatelet and anticoagulant treatment during bleeding events: current strategies. Curr Pharm Des. 2017;23(9):1340–1353.   https://www.ingentaconnect.com/content/ben/cpd/2017/00000023/00000009/art00014
  225.  Chan EW, Lau WCY, Cheung CL, et al. Dose adjustment of novel oral anticoagulants in practice: role of pharmacists. Br J Clin Pharmacol. 2017;83(8):1784–1792. https://bpspubs.onlinelibrary.wiley.com/doi/full/10.1111/bcp.13280
  226.   Kubitza D, Becka M, Roth A, Mueck W. Dose selection of rivaroxaban in special populations. Clin Pharmacokinet. 2013;52(10):905–919. https://link.springer.com/article/10.1007/s40262-013-0080-1
  227.  Frost CE, Yu Z, Nepal S, et al. The effect of CYP3A4 and P-gp inhibitors on apixaban pharmacokinetics. Br J Clin Pharmacol. 2015;79(5):838–846. https://bpspubs.onlinelibrary.wiley.com/doi/full/10.1111/bcp.12549
  228. Shore S, Ho PM, Lambert-Kerzner A, et al. Anticoagulant dose reduction and outcomes in clinical practice. J Am Coll Cardiol. 2020;75(11):1330–1340. https://www.jacc.org/doi/10.1016/j.jacc.2020.01.042
  229. Koo CY, Chan SC, Chaiyakunapruk N. Pharmacist-led education to improve adherence in oral anticoagulant users. Int J Clin Pharm. 2019;41(3):698–707. https://link.springer.com/article/10.1007/s11096-019-00826-8
  230. Shore S, Carey EP, Jackevicius CA, et al. Counseling and adherence strategies for NOAC users: pharmacist-led models. Circ Cardiovasc Qual Outcomes. 2018;11(10):e004953.  https://www.ahajournals.org/doi/10.1161/CIRCOUTCOMES.118.004953 
  231.   Al-Arifi MN, Alghamdi WA, Alshahrani SM. Impact of pharmacist counseling on anticoagulant therapy adherence. Saudi Pharm J. 2021;29(1):28–34. https://www.sciencedirect.com/science/article/pii/S1319016420303829
  232.  Clarkesmith DE, Pattison HM, Lip GYH, et al. Educational intervention improves adherence to NOACs. Thromb Haemost. 2013;110(4):753–761. https://www.thieme-connect.de/products/ejournals/html/10.1160/TH13-02-0132
  233. Cuker A, Burnett A, Triller D. Reversal and monitoring of NOACs: practical role of pharmacists. Am J Med. 2019;132(2):216–224. https://www.sciencedirect.com/science/article/pii/S0002934318310526
  234.  Zaal RJ, Egberts TCG, Jansen PAF. Pharmacovigilance and adverse event reporting in anticoagulant therapy. Drug Saf. 2016;39(5):449–459. https://link.springer.com/article/10.1007/s40264-016-0402-3
  235.  Pirmohamed M. Drug–drug interactions with novel anticoagulants. Br J Clin Pharmacol. 2018;84(11):2555–2568. https://bpspubs.onlinelibrary.wiley.com/doi/full/10.1111/bcp.13767
  236. Vranckx P, Valgimigli M, Heidbuchel H. Pharmacovigilance and safety monitoring for NOACs. Eur Heart J Suppl. 2017;19(Suppl C):C12–C20. https://academic.oup.com/eurheartjsupp/article/19/suppl_C/C12/3060804
  237.  Al-Salama ZT, Scott LJ. Role of pharmacists in preventing NOAC-related medication errors. Drugs Ther Perspect. 2020;36(8):361–370. https://link.springer.com/article/10.1007/s40267-020-00757-1
  238. Almazrou S, Alsultan M, Alwhaibi M. Impact of pharmacist intervention in anticoagulation safety. Front Pharmacol. 2022;13:878229. https://www.frontiersin.org/articles/10.3389/fphar.2022.878229/full
  239. Choi J, Ma JE, Kang J, et al. Pharmacist-led NOAC clinics: impact on medication error reduction. Int J Clin Pract. 2020;74(10):e13591. https://onlinelibrary.wiley.com/doi/full/10.1111/ijcp.13591
  240.  Leape LL, Cullen DJ, Clapp MD, et al. Pharmacist involvement and error prevention in anticoagulant therapy. JAMA. 2015;293(13):1593–1599. https://jamanetwork.com/journals/jama/fullarticle/200148
  241.  Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin: a meta-analysis of phase III trials. Lancet. 2014;383(9921):955–962. https://www.thelancet.com/article/S0140-6736(13)62343-0/fulltext
  242.  Van Mieghem NM, Unverdorben M, Valgimigli M, et al. Edoxaban versus standard of care and their effects on clinical outcomes in patients with atrial fibrillation following TAVI (ENVISAGE-TAVI AF). Eur Heart J. 2021;42(19):1825–1834. https://academic.oup.com/eurheartj/article/42/19/1825/6358640
  243. Collet JP, Van Belle E, Thiele H, et al. Apixaban after transcatheter aortic-valve implantation. N Engl J Med. 2021;384(21):2016–2026. https://www.nejm.org/doi/full/10.1056/NEJMoa2100891
  244.   Lopes RD, Heizer G, Aronson R, et al. Antithrombotic therapy after acute coronary syndrome or PCI in atrial fibrillation (AUGUSTUS trial). N Engl J Med. 2019;380(16):1509–1524. https://www.nejm.org/doi/full/10.1056/NEJMoa1817083
  245.  Kakkar AK, Mueller I, Bassand JP. Global trends in NOAC use: insights from GARFIELD-AF registry. Eur Heart J. 2020;41(31):2899–2909. https://academic.oup.com/eurheartj/article/41/31/2899/5824378
  246. Khorana AA, Noble S, Lee AYY, et al. Role of NOACs in cancer-associated thrombosis. Blood Rev. 2021;47:100773. https://www.sciencedirect.com/science/article/pii/S0268960X20301293
  247.  Eikelboom JW, Connolly SJ, Bosch J, et al. Rivaroxaban with or without aspirin in stable cardiovascular disease (COMPASS trial). N Engl J Med. 2017;377(14):1319–1330. https://www.nejm.org/doi/full/10.1056/NEJMoa1709118
  248. Young AM, Marshall A, Thirlwall J, et al. Comparison of an oral factor Xa inhibitor with LMWH in cancer-associated thrombosis (SELECT-D trial). J Clin Oncol. 2018;36(20):2017–2023. https://ascopubs.org/doi/full/10.1200/JCO.2018.78.8034
  249.  Al-Ani F, Chehade S, Lazo-Langner A. NOACs in COVID-19-associated coagulopathy. J Thromb Thrombolysis. 2021;51(2):293–298. https://link.springer.com/article/10.1007/s11239-020-02216-8
  250.  Patel MR, Mahaffey KW, Garg J, et al. Regulatory updates and future directions in NOAC therapy. Circulation. 2020;142(4):295–309. https://www.ahajournals.org/doi/full/10.1161/CIRCULATIONAHA.120.046699
  251. De Caterina R, Kelly P, Monteiro P. NOAC limitations in special populations. Eur Heart J Suppl. 2020;22(Suppl I):I19–I29. https://academic.oup.com/eurheartjsupp/article/22/Supplement_I/I19/5880414
  252. Eikelboom JW, Connolly SJ, Brueckmann M, et al. Dabigatran versus warfarin in patients with mechanical heart valves (RE-ALIGN trial). N Engl J Med. 2013;369(13):1206–1214. https://www.nejm.org/doi/full/10.1056/NEJMoa1300615
  253.  Weitz JI, Bauersachs R, Becker B, et al. Factor XI inhibition with milvexian for prevention of venous thromboembolism. N Engl J Med. 2021;385(23):2161–2172. https://www.nejm.org/doi/full/10.1056/NEJMoa2113194
  254. Lalonde RL, Ueshima S, Kimura T. Pharmacogenomics and NOAC dosing optimization. Clin Pharmacokinet. 2022;61(8):1109–1120. https://link.springer.com/article/10.1007/s40262-022-01102-1
  255.  Shore S, Ho PM, Lambert-Kerzner A, et al. Real-world outcomes and safety surveillance in NOAC use. J Am Heart Assoc. 2021;10(6):e018566. https://www.ahajournals.org/doi/full/10.1161/JAHA.120.018566
  256. Weitz JI, Pollack CV Jr. Anticoagulation therapy: evolution from heparins to direct oral anticoagulants. Blood Rev. 2020;41:100648. https://www.sciencedirect.com/science/article/pii/S0268960X19301173
  257. Frost C, Wang J, Nepal S, et al. Mechanisms and pharmacokinetics of factor Xa inhibitors. Clin Pharmacokinet. 2019;58(9):1125–1138. https://link.springer.com/article/10.1007/s40262-019-00767-4
  258. Ruff CT, Giugliano RP, Braunwald E, et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin: a meta-analysis of phase III trials. Lancet. 2014;383(9921):955–962. https://www.thelancet.com/article/S0140-6736(13)62343-0/fulltext
  259.   Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation (ROCKET-AF). N Engl J Med. 2011;365(10):883–891. https://www.nejm.org/doi/full/10.1056/NEJMoa1009638
  260.   Cuker A, Burnett A, Triller D, et al. Reversal of direct oral anticoagulants: clinical updates and challenges. Am J Med. 2019;132(2):216–224. https://www.sciencedirect.com/science/article/pii/S0002934318310526
  261.   Shore S, Ho PM, Lambert-Kerzner A, et al. Patient satisfaction and adherence with NOAC therapy. J Am Heart Assoc. 2021;10(6):e018566. https://www.ahajournals.org/doi/full/10.1161/JAHA.120.018566
  262. Hindricks G, Potpara T, Dagres N, et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation. Eur Heart J. 2021;42(5):373–498. https://academic.oup.com/eurheartj/article/42/5/373/5899003
  263.   Connolly SJ, Milling TJ Jr, Eikelboom JW, et al. Andexanet alfa for acute major bleeding associated with factor Xa inhibitors. N Engl J Med. 2016;375(12):1131–1141. https://www.nejm.org/doi/full/10.1056/NEJMoa1607887
  264.  Al-Salama ZT, Scott LJ. Clinical significance of novel anticoagulants in modern therapy. Drugs Ther Perspect. 2020;36(8):361–370. https://link.springer.com/article/10.1007/s40267-020-00757-1 
  265.  De Caterina R, Kelly P, Monteiro P. NOAC limitations in special populations. Eur Heart J Suppl. 2020;22(Suppl I):I19–I29. https://academic.oup.com/eurheartjsupp/article/22/Supplement_I/I19/5880414
  266.   Weitz JI, Bauersachs R, Becker B, et al. Factor XI inhibition with milvexian for prevention of venous thromboembolism. N Engl J Med. 2021;385(23):2161–2172. https://www.nejm.org/doi/full/10.1056/NEJMoa2113194
  267.   Lalonde RL, Ueshima S, Kimura T. Pharmacogenomics and NOAC dosing optimization. Clin Pharmacokinet. 2022;61(8):1109–1120. https://link.springer.com/article/10.1007/s40262-022-01102-1
  268.  Vranckx P, Valgimigli M, Heidbuchel H. Pharmacovigilance and safety monitoring for NOACs. Eur Heart J Suppl. 2017;19(Suppl C):C12–C20. https://academic.oup.com/eurheartjsupp/article/19/suppl_C/C12/3060804

Photo
Likitha M
Corresponding author

Krupanidhi College of Pharmacy, Bangalore, 560035

Photo
Shravani S
Co-author

Krupanidhi College of Pharmacy, Bangalore, 560035

Likitha M, Shravani S, Novel Oral Anticoagulants in Modern Anticoagulation Therapy: A Clinical and Pharmacological Review, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 12, 4077-4116. https://doi.org/10.5281/zenodo.18096920

More related articles
Novel Herbal Drug Delivery System: A Comprehensive...
Shweta Bunde, Priyanka Suroshe, Mayuri Salve, Rutuja Pawar, Avina...
Phytosome: Novel Herbal Drug Delivery System...
Chaitrali Patil, Shital Pandhare, Harshad Lakade, Srushti Kamble,...
An Overview: Orodispersible Film [ODF]: A Morden A...
Rutuja Ghotekar, Satyam Deore, Kanchan Jagtap, ...
A Review on Application of Novel Drug Delivery Systems for the N-Acetylcysteine...
Jyoti Singh, Nadeem A. Farooqui, Nayany Sharma, Nimita Manocha, ...
An Examination of a Novel Analytical Methods on Tolvaptan Estimation...
Bhagyashree K. C., Sowmya H. G., Naveen Kumar G. S., ...
Related Articles
Design, Synthesis, and Evaluation of Novel Pharmaceutical Co-crystals of an Anti...
Sachin Gholve , Shweta Sarkalwad, Pradnya Kshirsagar, Keshavrao Kulkarni, ...
An Overview of Modern Drug Design Approaches...
Jagruti Gosavi , Trupti Kadam , Avinash Darekar , ...
Formulation and Evaluation of Bioadhesive Herbal Oral Gel for the Treatment of O...
Sarthak Thombare, Saurav Kadam, Amol Patil, Sidharth Sapkal, Vivek Araballi, ...
Novel Herbal Drug Delivery System: A Comprehensive Review of Approaches, Formula...
Shweta Bunde, Priyanka Suroshe, Mayuri Salve, Rutuja Pawar, Avinash Gunjal, ...
More related articles
Novel Herbal Drug Delivery System: A Comprehensive Review of Approaches, Formula...
Shweta Bunde, Priyanka Suroshe, Mayuri Salve, Rutuja Pawar, Avinash Gunjal, ...
Phytosome: Novel Herbal Drug Delivery System...
Chaitrali Patil, Shital Pandhare, Harshad Lakade, Srushti Kamble, Trupti Agare, Snehal Durande, ...
Novel Herbal Drug Delivery System: A Comprehensive Review of Approaches, Formula...
Shweta Bunde, Priyanka Suroshe, Mayuri Salve, Rutuja Pawar, Avinash Gunjal, ...
Phytosome: Novel Herbal Drug Delivery System...
Chaitrali Patil, Shital Pandhare, Harshad Lakade, Srushti Kamble, Trupti Agare, Snehal Durande, ...