J Stroke Search

CLOSE


J Stroke > Volume 27(2); 2025 > Article
Choi and Kim: Anticoagulation Failure in Stroke: Causes, Risk Factors, and Treatment

Abstract

Anticoagulation is crucial to reducing the risk of cardioembolic strokes, particularly in vulnerable populations such as patients with atrial fibrillation, artificial heart valves, or left ventricular thrombus. Though successful, anticoagulation failure (the occurrence of an ischemic stroke or systemic embolism while receiving therapy) remains a major stroke-care issue. The reason for anticoagulation failure can be below the required threshold, inability to follow up, drug-drug interactions, preexisting hypercoagulable states, or anticoagulant resistance. This failure undermines stroke prevention and requires tailored management, often requiring more drastic or alternative interventions. This review examines what drives anticoagulation failure and explores predictors of this failure in clinical, imaging, and laboratory data. It also discusses current management techniques for improving control and points to new treatments and possible futures, such as high-resolution imaging and personalized medicine based on biomarkers, to help tackle this critical clinical problem.

Introduction

Worldwide, stroke is a leading cause of illness and death, and cardioembolic (CE) strokes account for 20%-30% of all ischemic strokes [1]. Atrial fibrillation (AF) is the most common cause of CE stroke, and anticoagulation can decrease stroke risk by 60%-70% in such patients [2]. Direct oral anticoagulants (DOACs) are safer and more effective than classic vitamin K antagonists (VKAs) due to their predictable pharmacokinetics, lower risk of intracranial hemorrhage, rapid onset and offset, lack of need for routine monitoring, and fewer drug and food interactions [3]. However, the relatively high frequency of recurrent strokes and systemic embolisms highlights the limitations of existing therapies [4]. Overcoming anticoagulation failure requires understanding stroke mechanisms, identifying high-risk patients, and making best-practice management recommendations. This review examines the spectrum of anticoagulation failure, identifies risk factors for early detection, and summarizes evidence-based interventions to resolve anticoagulation failure.

Causes of anticoagulation failure

Patient-related factors

Non-adherence

Studies indicate that up to 40% of patients report issues with adhering to their anticoagulant drug regimens due to unreliability. This inability to adhere is often caused by ignorance, exploitation, or fear of the side effects such as bleeding [5,6]. Regular monitoring and dietary restrictions make patients even more resistant to VKAs intake. Poor access to healthcare institutions for follow-up visits or international normalized ratio (INR) measurements leads to non-adherence. Inadequate education on the risks and advantages of anticoagulation is worrying [7]. Patients may not realize the severity of the disease or the necessity of treatment. Medical terminology or instructions that are difficult to understand can further confuse patients and hinder compliance [8].

Renal or hepatic dysfunction

Poor renal and liver function has critical consequences on the pharmacokinetic and pharmacodynamic profiles of anticoagulants, and their safety and efficacy can vary widely [9]. Severe renal impairment decreases DOAC excretion, which raises drug levels and increases the risk of toxicity. In addition, liver disease disrupts the metabolism of anticoagulants, leading to inconsistencies in their efficacy [10]. Renal or hepatic dysfunction further impacts the patient’s condition. Chronic fatigue, weakness, and systemic complications can reduce a patient’s ability to engage in their care actively [11].

Obesity and age-related factors

Obesity impacts the effectiveness and safety of anticoagulation therapy by altering drug pharmacokinetics and leading to relative underdosing [12]. As patients age, increased frailty, polypharmacy, and a lack of physiological reserve raise the likelihood of anticoagulation failure. Older patients may have difficulty in taking their medications regularly, which can dramatically reduce treatment effectiveness. For example, they may fall asleep before taking medications that are scheduled for the evening. This is a complex phenomenon—physiological, psychological, and social—unique to the elderly [13].

Malabsorption syndromes

Inflammatory bowel disease (IBD) or post-bariatric surgery can disrupt drug absorption and decrease the blood levels of anticoagulants. Patients with chronic diarrhea often face significant challenges in medication absorption, which can reduce the effectiveness of treatments and complicate disease management [14].

Food interaction

Warfarin function decreases when consumed with foods containing high levels of vitamin K, leading to INR changes. Vitamin K is found in various foods, including kale, spinach, broccoli, cabbage, romaine lettuce, and arugula [15].

Drug-related factors

Subtherapeutic dosing

The administration of inappropriately low medication doses is a common issue in clinical practice, potentially leading to suboptimal therapeutic effects. Low INR is a common problem with VKAs. A time in therapeutic range of less than 60% increases the risk of thromboembolic complications [16]. Physicians may reduce the dosage of medications, particularly in elderly or frail patients, to avoid potential adverse effects such as drug toxicity or organ dysfunction. In the case of DOACs, standardized doses may not consider individual patient variables such as body weight, renal function, or age [17]. When DOAC doses are reduced, they are sometimes based on misunderstandings or incorrect interpretations of clinical reasoning [18].

Drug-drug interactions

Drugs like rifampin and amiodarone, for example, work on the cytochrome enzymes or P-glycoprotein networks that can affect the effectiveness of anticoagulants. VKAs such as warfarin can be impacted by interactions with other drugs in several ways, primarily by altering the activity of cytochrome P450 enzymes. Enzyme stimulants such as rifampin and carbamazepine, for example, accelerate warfarin’s metabolism, resulting in reduced levels of the drug in the blood [19]. In contrast, enzyme inhibitors like amiodarone and fluconazole slow warfarin’s metabolism, which may result in excessive accumulation of the drug in the blood [20]. DOACs such as apixaban and rivaroxaban are also subject to interactions with P-glycoprotein and cytochrome P450 3A4 enzymes. Depending on the composition of the drug, these interactions can boost or decrease the drug’s activity. Drugs such as ketoconazole (an antifungal) and ritonavir (an antiretroviral), for instance, inhibit cytochrome P450 enzymes and can raise the levels of co-administered drugs. Inducers like phenytoin and St. John’s wort, in contrast, increase the activity of these enzymes, accelerating the breakdown of drugs and lowering the activity of anticoagulants [21]. Such interactions are more common in patients taking multiple medications [22]. Polypharmacy alters drug metabolism at an accelerated rate, especially in the elderly or those with chronic illness [23].

DOAC-specific limitations

DOACs such as apixaban, rivaroxaban, dabigatran, and edoxaban have become the preferred alternative to VKAs for stroke prevention and venous thromboembolism [24]. DOACs, with their benefits of fixed dosing, reduced drug-food interaction, and a lack of frequent monitoring in most contexts, are not free from problems. Absorption syndromes (IBD, celiac disease, bariatric surgery) can seriously impair DOAC absorption [25]. Further, because dabigatran depends on an acidic gastric environment to absorb it, it is sensitive to proton pump inhibitors that could impair its bioavailability and efficacy [26]. Fixed dosing makes DOAC administration easier but may not adequately address patient variation. Standard dosing thus stresses the need for tailored interventions in at-risk groups [27]. The lack of routine monitoring of DOAC levels makes it difficult to identify subtherapeutic or supratherapeutic levels, particularly in patients with unstable renal function or excessive body weight [28]. The fear of lack of an antidote or specific hemostatic agents to counter bleeding complications is a significant concern when using certain DOACs [29].

Disease-related factors

Hypercoagulable states in CE stroke

In some patients with CE stroke, underlying hypercoagulable states such as antiphospholipid syndrome (APS) may contribute to thromboembolic recurrence despite adequate anticoagulation [30]. APS is characterized by autoantibodies that enhance thrombin generation and impair natural anticoagulant pathways [31]. In patients with both AF and APS, anticoagulation failure may occur even under appropriate VKA therapy [32]. Although some early studies suggested that DOACs—particularly factor Xa inhibitors—might provide enhanced protection against arterial thrombosis due to platelet-rich clot formation, more recent evidence indicates that DOACs are inferior to VKAs in high-risk APS patients, especially those with triple-positive antibody profiles. Thus, warfarin remains the preferred agent in this subgroup [33].
Genetic prothrombotic conditions, such as factor V Leiden mutation or protein C/S deficiency, may also coexist in CE stroke patients and reduce the effectiveness of standard anticoagulation therapies. These inherited conditions promote persistent thrombin generation and platelet activation, possibly contributing to recurrent embolic events despite therapeutic anticoagulation levels [34].

Cancer-associated thrombosis in CE stroke patients

Cancer-associated thrombosis represents another significant challenge in CE stroke patients. Malignancy can amplify thromboembolic risk through tumor-derived procoagulants, endothelial injury, and inflammation [35]. In CE stroke patients with active cancer, anticoagulation may be less effective due to increased baseline coagulability and altered drug pharmacokinetics [36]. Furthermore, physical frailty and treatment-related toxicity in cancer patients may limit the tolerability of oral anticoagulants, necessitating individualized approaches such as low-molecular-weight heparin or close INR monitoring in VKA therapy [37]. In both APS and cancer, adjunctive strategies including reassessment of anticoagulant selection, dosing, and comorbidity-specific risk profiling may be necessary to prevent recurrence in CE stroke patients with coexisting hypercoagulable states [38].

Device-associated thrombosis

Patients face particular difficulties with mechanical heart valves, left ventricular assist devices, or left atrial appendage occlusion (LAAO) devices. Such devices serve as clot-provoking surfaces, even in the face of anticoagulation therapy. Warfarin is the anticoagulant of choice in such patients because it has proven effective in avoiding valve-associated thrombi. Heparinoids can be used in patients who require temporary interruption of warfarin therapy for surgery or invasive procedures. DOACs, by contrast, are found to be less effective in these situations [39].

Persistent intracardiac thrombus

Intracardiac thrombi, especially in the left atrium or ventricle, may remain resistant to anticoagulation, which may result in systemic embolization. Chronic thrombi could require increasing the dose of anticoagulation or other therapeutic interventions to decrease the embolic risk [40].

Chronic inflammatory conditions

Chronic inflammations, such as IBD or rheumatoid arthritis, creates a hypercoagulable landscape by activating platelets and ramping up thrombin production. Other common chronic inflammation, such as gingivitis, sinusitis, periodontitis, and chronic osteomyelitis, can elevate systemic inflammatory response. They also diminish the effectiveness of anticoagulants because chronic inflammation undermines their therapeutic effectiveness [41].
Key points summarizing the causes of anticoagulation failure are consolidated in Table 1.

Predictors of anticoagulation failure

Clinical predictors

CHA2DS2-VASc score

An elevated CHA2DS2-VASc score is an important stroke risk marker for AF, even in those receiving anticoagulation. Patients with a CHA2DS2-VASc score greater than 6 are at the highest risk, as such a high score indicates a significantly increased probability of recurrent thromboembolism [42].

Comorbidities

Renal failure significantly alters the pharmacokinetics of anticoagulants, especially DOACs [43]. In addition to contributing to increased thrombotic risk, diabetes mellitus contributes to chronic vascular inflammation and increases platelet activation, lowering the effectiveness of anticoagulants [44]. Chronic vascular injury caused by hypertension is a chronic condition of endothelial dysfunction and arterial stiffness [45]. Diabetes mellitus leads to increased levels of procoagulant factors like fibrinogen and plasminogen activator inhibitor-1, contributing to a hypercoagulable state [46]. Hypertension is associated with increased levels of oxidative stress and inflammatory mediators, both of which contribute to thrombogenesis [47]. Such changes increase atherothrombotic plaque generation and increase the risk of clot formation, making anticoagulation treatments less effective [48].

Lifestyle factors

Smoking and heavy drinking may increase vascular inflammation and interfere with anticoagulant metabolism. These issues, taken together, contribute to increased thrombotic risk and the difficulty of obtaining and maintaining therapeutic levels of anticoagulants [49].

Imaging predictors

Silent infarcts

On-screen silent cerebral infarction, especially cortical infarction, despite vigorous anticoagulant treatment, is an alarming result that points to anticoagulant failure. This is a clinical issue that brings important questions as to whether the current anticoagulation treatment is effective or requires reevaluation [50].

Atherosclerotic plaques in large arteries

Plaques of atherosclerosis in major arteries, particularly the carotid and aortic ones, pose major risk factors for embolization and stroke. Broken or weak plaques release thromboembolic fluid into the bloodstream, rendering anticoagulation less effective at preventing ischemic strokes [51].

Hemorrhagic transformation of ischemic stroke

The hemorrhagic transformation of an ischemic stroke can be a major challenge for anticoagulation therapy because it increases the risk of treatment failure and side effects [52].

Aortic atheromas

The presence of aortic atheromas necessitates an update on the current anticoagulation regimen. Aortic atheromas—mobile or irregular, and greater than 4 mm thick—constitute major sources of emboli in patients with AF [53]. The presence of significant aortic atheromas adds another layer of embolic risk, as emboli can originate from the aorta itself, independent of atrial thrombi. Identification of high-risk aortic atheromas may warrant more aggressive anticoagulation or antiplatelet therapy to mitigate embolic risks [54].

Laboratory predictors

D-dimer level

Chronically elevated D-dimer in patients receiving anticoagulation may indicate ongoing thrombotic activity and suggest a current anticoagulant regimen deficiency. This is particularly concerning in populations at high risk, including those with AF, venous thromboembolism, or hypercoagulable conditions such as cancer or APS [55]. Consistent D-dimer elevation also increases the risk of thromboembolic events and adverse clinical outcomes [56].

Drug levels

Anticoagulant doses below the recommended levels are a leading cause of treatment failure, whether due to adherence, altered metabolism, or misdosing. Underdosing—whether from physician misjudgment, patient characteristics, or adherence issues—is associated with increased risks of stroke and mortality, reflecting anticoagulation treatment failure [18].

Hypercoagulable state markers

Hypercoagulable disorders such as APS and inherited thrombophilia have the potential to diminish the efficacy of routine anticoagulation [57]. APS markers include lupus anticoagulant, anticardiolipin, and anti-2-glycoprotein I antibodies. Genetic tests can be utilized to detect the factor V Leiden mutation, the prothrombin G20210A mutation, as well as deficiencies in protein C, protein S, and antithrombin [58,59].
Table 2 summarizes the predictors of anticoagulation failure for ease of reference.

Management of anticoagulation failure

Optimizing anticoagulation therapy

Therapeutic drug monitoring

For patients on VKAs such as warfarin, routine INR monitoring is essential to ensure therapeutic efficacy and minimize bleeding risk. The target INR is typically 2.0-3.0 for most indications and 2.5-3.5 in high-risk conditions such as mechanical heart valves. INR levels are influenced by various factors, including diet, drug interactions, and liver function, necessitating frequent monitoring and dose adjustments [60].
In contrast, DOACs generally do not require routine therapeutic drug monitoring due to their predictable pharmacokinetics. However, plasma level assessment may be considered in special clinical settings such as major bleeding, renal impairment, urgent surgery, or suspected overdose. Drug-specific assays are available [61].
• Rivaroxaban and apixaban: Levels can be measured dusing anti-factor Xa activity assays, calibrated to the specific DOAC. These tests can quantify drug concentration, though they lack established therapeutic ranges [62].
• Dabigatran: Levels can be assessed using diluted thrombin time (dTT) or ecarin clotting time. dTT correlates linearly with plasma drug concentration [63].
• Edoxaban: Anti-factor Xa assays may be used but are less commonly available and not routinely employed [64].
Although DOAC level testing is available, it is infrequently used in everyday practice and is generally reserved for specialized or emergency scenarios.

Switching anticoagulants

Anticoagulant therapy is often adjusted when the current regimen presents concerns related to efficacy, safety, or patient preference. In clinical practice, DOACs are generally preferred due to their predictable pharmacokinetics, fixed dosing, and minimal monitoring requirements [65]. Switching from warfarin to a DOAC may be appropriate in patients who experience poor INR control (i.e., time-in-therapeutic range <60%) or who prefer a more convenient regimen that does not require frequent blood testing [66]. Conversely, there remain specific indications where warfarin is favored or necessary. These include patients with mechanical heart valves, those with APS, and clinical scenarios requiring high-intensity anticoagulation (target INR 3.0-4.0), for which DOACs are either ineffective or contraindicated [67].
Low molecular weight heparin is preferred over warfarin in certain clinical settings, such as cancer-associated thrombosis, pregnancy, and perioperative bridging, where studies have demonstrated superior efficacy and/or safety profiles [68]. However, we also acknowledge that warfarin remains the anticoagulant of choice in specific scenarios, such as in patients with mechanical heart valves, APS, or where cost and accessibility are key considerations [69].

Adjunctive therapies

Dual antithrombotic therapy

Dual antithrombotic therapy, which combines an anticoagulant with an antiplatelet agent such as aspirin, may be considered in high-risk patients who have overlapping indications—particularly those with AF and coexisting atherosclerotic cardiovascular disease, such as coronary artery disease or a recent stent placement [70,71]. However, the benefit must be weighed carefully against the increased risk of bleeding. Multiple randomized controlled trials —including the PIONEER AF-PCI (The Open-Label, Randomized, Controlled, Multicenter Study Exploring Two Treatment Strategies of Rivaroxaban and a Dose-Adjusted Oral Vitamin K Antagonist in Patients With Atrial Fibrillation Who Undergo Percutaneous Coronary Intervention), AUGUSTUS (An Open-label, 2x2 Factorial, Randomized Controlled Trial to Evaluate the Safety of Apixaban Compared to a Vitamin K Antagonist and Aspirin Compared to Placebo in Patients With Atrial Fibrillation and Acute Coronary Syndrome and/or Undergoing Percutaneous Coronary Intervention), and RE-DUAL PCI (The Randomized Evaluation of Dual Therapy With Dabigatran vs. Triple Therapy With Warfarin in Patients With Nonvalvular Atrial Fibrillation Undergoing Percutaneous Coronary Intervention)—have evaluated dual antithrombotic therapy in patients with atrial fibrillation undergoing percutaneous coronary intervention (PCI), and have generally favored short-term dual therapy (e.g., 1-6 months) followed by anticoagulant monotherapy for long-term management [72-74].
In general, indications include AF with recent PCI or acute coronary syndrome, as well as patients with high thromboembolic risk—such as those with a history of stroke and significant atherosclerotic disease.
The typical durations of therapy are as follows:
• Triple therapy (anticoagulant + dual antiplatelet agents) is typically limited to ≤1 week.
• Dual therapy (anticoagulant + a single antiplatelet agent) is generally recommended for 1 to 6 months.
• Anticoagulant monotherapy is continued thereafter, guided by a balance between ischemic and bleeding risk.
Careful monitoring is essential, and treatment decisions should be individualized based on factors such as the CHA2DS2-VASc score, HAS-BLED score, type of stent placed, and recent vascular events [70,72-74].

Left atrial appendage occlusion

LAAO is a catheter-based procedure that serves as an alternative stroke prevention strategy in patients with non-valvular AF (NVAF), particularly when oral anticoagulation is contraindicated or ineffective [75]. Left atrial appendage is the origin of over 90% of thrombi in patients with NVAF, and its occlusion significantly reduces the risk of CE stroke [76].
While LAAO is most often used in patients who are unsuitable for long-term anticoagulation, its role has expanded. The PRAGUE-17 trial, a randomized controlled study, demonstrated that LAAO was non-inferior to DOAC therapy in preventing a composite outcome of stroke, systemic embolism, cardiovascular death, and major bleeding in high-risk patients with AF. These findings support the broader use of LAAO, even in some patients eligible for anticoagulation, especially when bleeding risk is high [77]. Indications for LAAO include: absolute contraindication to anticoagulation (e.g., prior intracranial hemorrhage); high bleeding risk (e.g., HAS-BLED ≥3); recurrent thromboembolic events despite therapeutic anticoagulation; inability to maintain consistent anticoagulation due to adherence or tolerance issues; and patient preference, following shared decision-making [78]. LAAO is increasingly recognized as a viable option for selected patients at high thromboembolic and bleeding risk, following careful evaluation by a multidisciplinary team.

Treating underlying conditions

Cancer-associated thrombosis

When a tumor is eradicated by chemotherapy, radiation, or surgery, the production of procoagulant factors (both tissue factors and cancer procoagulants) is reduced. This retards the coagulation process, and thus prevents thrombus formation. Cancer therapy also minimizes the body’s inflammatory reaction that leads to excessive clotting. Lowering inflammatory mediators such as interleukin-6 and tumor necrosis factor-alpha can rebalance endothelial function and block clotting [79].

APS management

Management of APS typically requires VKA therapy, such as warfarin, with a target INR of 2.0-3.0. In refractory or high-risk cases, such as patients with recurrent thrombotic events despite adequate INR or those with triple-positive antibodies (i.e., positive lupus anticoagulant, anticardiolipin, and anti-β2-glycoprotein I), a higher-intensity anticoagulation target (INR 3.0-4.0) may be considered [21]. However, this is individualized and not standard for all APS patients [31-33]. Recent evidence has shown that DOACs such as rivaroxaban are less effective than warfarin in high-risk APS, particularly in patients with triple antibody positivity, and are therefore not recommended in this population. Conversely, in patients with non-APS-related atherosclerotic vascular disease, DOACs remain a suitable option due to their favorable safety profile and ease of use [33].
Table 3 provides a detailed summary of the management of anticoagulation failure for comparison.

Emerging therapies and future directions

Even with better anticoagulation, failure to respond to treatment is still an issue, especially in high-risk patients. Biologically, new drugs and treatments are designed to fill gaps in current treatments, enhance patient care, and decrease thromboembolic risk (Table 4).

Next-generation anticoagulants

Factor XI/XIa inhibitors, like asundexian and abelacimab, are a class of new anticoagulants that interfere with the fundamental coagulation mechanism. Because they selectively block factor XI, they prevent clot formation without risking bleeding, which is one major drawback of standard anticoagulants. These drugs are up-and-coming in patients who are predisposed to bleeding or with a history of repeated thromboembolisms [80]. However, recent randomized controlled trials of Factor XI/XIa inhibitors in AF populations have not demonstrated efficacy, and therefore, those programs have been discontinued or redirected [81,82]. Dual-target drugs are another promising avenue, targeting multiple pathways—including factor Xa and thrombin—to improve anticoagulation without massively lowering the risk of bleeding. These drugs seek to offer broader protection against thromboembolic complications while also providing a good safety profile [83].

Gene therapy

Gene therapy presents a promising avenue for the long-term correction of hereditary hypercoagulable conditions, such as antithrombin deficiency. This approach involves the delivery of functional copies of defective genes or direct editing of disease-causing mutations using tools such as adeno-associated viral vectors or clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology. By restoring normal expression and function of anticoagulant proteins like antithrombin, gene therapy aims to correct the underlying molecular defect, reduce thrombotic risk, and potentially eliminate the need for lifelong anticoagulation therapy [84].

Advanced imaging

Advanced imaging techniques have significantly enhanced anticoagulation management by providing detailed insights into hemodynamics, thrombus localization, and embolic activity. These modalities facilitate early risk assessment and enable more targeted anticoagulation strategies. Four-dimensional (4D) flow magnetic resonance imaging (MRI) allows for comprehensive visualization and quantification of blood flow dynamics. In patients with AF, 4D flow MRI has been instrumental in assessing left atrial blood flow patterns, identifying areas prone to thrombosis, and evaluating the risk of embolic events. This imaging modality offers detailed hemodynamic information that can guide anticoagulation therapy decisions [85]. Cardiac computed tomography (CT) is a non-invasive tool effective in detecting intracardiac thrombi, particularly in patients with acute ischemic stroke. Studies have shown that cardiac CT can identify thrombi that may necessitate anticoagulation therapy, thereby influencing patient management and potentially improving outcomes [86]. Transcranial Doppler ultrasonography is utilized to detect microembolic signals (MES) in the cerebral circulation, serving as biomarkers for embolic events and aiding in stroke risk assessment. The detection of MES can inform the effectiveness of anticoagulation therapy and guide adjustments to treatment plans [87]. Incorporating these advanced imaging modalities into clinical practice enhances the precision of anticoagulation management by enabling early detection of thrombotic risks and facilitating individualized treatment strategies.

Artificial intelligence

Integrating artificial intelligence (AI) with clinical, imaging, and laboratory data holds significant promise for identifying patients at risk of anticoagulation failure. AI algorithms can analyze complex datasets to uncover patterns and risk factors that may be overlooked by traditional methods, thereby enhancing patient management strategies [88].
Machine learning (ML) models have been developed to predict anticoagulation control in patients with AF. By processing variables such as demographics, comorbidities, and prior anticoagulation responses, these models can identify patients at risk of suboptimal anticoagulation control. This enables earlier interventions to optimize therapy and reduce the likelihood of thrombotic or bleeding complications [89]. Furthermore, AI has been utilized to predict the likelihood of developing AF by analyzing harmonized electronic health record data. ML approaches have shown promise in identifying individuals at risk, facilitating early interventions, and potentially preventing associated complications such as stroke [90]. By harnessing AI to integrate and analyze diverse data sources, healthcare providers can enhance the precision of anticoagulation management, leading to improved patient outcomes and personalized treatment strategies.

Personalized medicine

Personalized anticoagulation therapy utilizes pharmacogenomics and biomarker research to optimize efficacy and safety in anticoagulant use, particularly for warfarin, which has a narrow therapeutic index and variable individual response [91].
Pharmacogenomic differences, especially in the cytochrome P450 family 2 subfamily C member 9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) genes, can affect how people process and respond to warfarin. The Clinical Pharmacogenetics Implementation Consortium guideline gives doctors advice on how to adjust warfarin doses based on these genetic results. People with CYP2C9*2 or CYP2C9*3 variants break down warfarin more slowly, which can raise the risk of bleeding. Those with the VKORC1 -1639G>A variant (especially the A/A type) are more sensitive to warfarin and need lower doses [91]. For example, people with both poor CYP2C9 metabolism and the VKORC1 A/A type often need smaller starting doses (around 3 mg or less per day), while those without these changes can usually take standard doses (5-7.5 mg per day). Special dosing tools, like warfarinDosing.org and the Gage or International Warfarin Pharmacogenetics Consortium algorithms, help personalize warfarin therapy. These tools improve how well patients stay within the safe and effective range and help reduce side effects, especially when starting treatment [92].

Conclusion

In stroke care, failure of anticoagulation demands an intricate understanding of the patient, medication, and disease environment. Using the latest imaging and biomarkers, targeted care and novel treatments may improve patients’ outcomes. Combining evidence-based interventions with novel technologies helps clinicians better understand the issue of anticoagulation failure, making it more manageable in stroke prevention and care.

Notes

Funding statement
None
Conflicts of interest
The authors have no financial conflicts of interest.
Author contribution
Conceptualization: YC, JSK. Study design: YC, JSK. Methodology: YC. Data collection: YC. Investigation: YC. Statistical analysis: YC. Writing—original draft: YC. Writing—review & editing: YC, JSK. Funding acquisition: YC. Approval of final manuscript: YC, JSK.

Table 1.
Summary of causes of anticoagulation failure
Factor Description
Patient-related factors
 Non-adherence Up to 40% report difficulty following regimens due to fear, misunderstanding, or poor access to care.
 Renal or hepatic dysfunction Alters drug metabolism and clearance, increasing risk of toxicity or reduced efficacy.
 Obesity and age Affects pharmacokinetics; elderly patients face frailty, polypharmacy, and adherence issues.
 Malabsorption syndromes IBD or post-surgical changes impair absorption and reduce anticoagulant levels.
 Food interaction Vitamin K-rich foods (e.g., kale, spinach) interfere with warfarin, affecting INR.
Drug-related factors
 Subtherapeutic dosing Underdosing due to cautious prescribing or failure to adjust for renal function or body weight.
 Drug-drug interactions Enzyme inducers (e.g., rifampin) or inhibitors (e.g., amiodarone) impact anticoagulant levels.
 DOAC-specific limitations Fixed dosing, absorption issues (e.g., with PPIs), and lack of monitoring contribute to failure.
Disease-related factors
 Hypercoagulable states APS and inherited thrombophilia reduce efficacy even with appropriate therapy; warfarin preferred in high-risk APS.
 Cancer-associated thrombosis Malignancy alters coagulation and pharmacokinetics; requires tailored therapy.
 Device-associated thrombosis Mechanical valves or LAAO devices increase thrombotic risk; warfarin is preferred.
 Persistent intracardiac thrombus Thrombi may resist anticoagulation and need dose escalation or alternative treatments.
 Chronic inflammatory conditions Sustained inflammation increases thrombosis risk and reduces anticoagulant effectiveness.
IBD, inflammatory bowel disease; INR, international normalized ratio; DOAC: direct oral anticoagulant; PPI, proton pump inhibitor; APS, antiphospholipid syndrome; LAAO, left atrial appendage occlusion.
Table 2.
Summary of predictors of anticoagulation failure
Predictor Description
Clinical predictors
 CHA2DS2-VASc score Scores >6 indicate high stroke risk even with anticoagulation.
 Comorbidities Renal failure, diabetes, and hypertension affect drug efficacy and increase thrombotic risk.
 Lifestyle factors Smoking and heavy drinking raise vascular inflammation and affect drug metabolism.
Imaging predictors
 Silent infarcts Cortical infarctions despite treatment suggest failure of anticoagulation.
 Atherosclerotic plaques Plaques in major arteries increase embolic risk and reduce anticoagulant efficacy.
 Hemorrhagic transformation Raises risk of treatment failure and complications.
 Aortic atheromas Large, mobile plaques are significant embolic sources requiring tailored therapy.
Laboratory predictors
 D-dimer levels Chronic elevation indicates ongoing thrombosis and treatment insufficiency.
 Drug levels Underdosing due to various factors increases stroke and mortality risks.
 Hypercoagulable state markers APS markers and genetic mutations can undermine anticoagulation efficacy.
APS, antiphospholipid syndrome.
Table 3.
Summary of management of anticoagulation failure
Strategy / Therapy / Condition Description / Management
Optimizing anticoagulation therapy Therapeutic drug monitoring INR monitoring is essential for VKAs; DOAC levels assessed in special cases (e.g., bleeding, renal impairment).
Switching anticoagulants Consider switching to DOACs for convenience and safety; warfarin preferred in specific conditions like APS or mechanical valves.
Adjunctive therapies Dual antithrombotic therapy Short-term dual therapy may be used in AF with CAD or PCI, followed by anticoagulant monotherapy.
Left atrial appendage occlusion Indicated in NVAF patients with bleeding risk, anticoagulant contraindications, or recurrent thromboembolism.
Treating underlying conditions Cancer-associated thrombosis Treating the malignancy reduces procoagulants and inflammation, restoring coagulation balance.
APS management Warfarin is standard; high-risk cases may need INR 3.0-4.0. DOACs not recommended in triple-positive patients.
INR, international normalized ratio; VKA, vitamin K antagonist; DOAC, direct oral anticoagulant; AF, atrial fibrillation; CAD, coronary artery disease; PCI, percutaneous coronary intervention; NVAF, non-valvular atrial fibrillation; APS, antiphospholipid syndrome.
Table 4.
Summary of emerging therapies and future directions in anticoagulation
Therapy or approach Description
Next-generation anticoagulants Factor XI/XIa inhibitors (e.g., asundexian, abelacimab) prevent clotting with lower bleeding risk. Dual-target drugs aim for enhanced protection with good safety.
Gene therapy Targets genetic hypercoagulable disorders by restoring functional anticoagulant proteins, potentially eliminating lifelong therapy.
Advanced imaging 4D Flow MRI, Cardiac CT, and TCD improve thrombus detection, risk assessment, and guide tailored anticoagulation strategies.
Artificial intelligence AI analyzes clinical, imaging, and lab data to identify at-risk patients and optimize anticoagulation management.
Personalized medicine Pharmacogenomic tools adjust warfarin dosing based on CYP2C9 and VKORC1 variants to improve efficacy and minimize risk.
4D, four-dimensional; MRI, magnetic resonance imaging; CT, computed tomography; TCD, transcranial Doppler; CYP2C9, cytochrome P450 family 2 subfamily C member 9; VKORC1, vitamin K epoxide reductase complex, subunit 1.

References

1. Hart RG, Catanese L, Perera KS, Ntaios G, Connolly SJ. Embolic stroke of undetermined source: a systematic review and clinical update. Stroke 2017;48:867-872.
crossref pmid
2. Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent risk factor for stroke: the Framingham study. Stroke 1991;22:983-988.
crossref pmid
3. Granger CB, Alexander JH, McMurray JJ, Lopes RD, Hylek EM, Hanna M, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2011;365:981-992.
pmid
4. Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL, et al. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2013;369:2093-2104.
crossref pmid
5. O’Brien EC, Simon DN, Thomas LE, Hylek EM, Gersh BJ, Ansell JE, et al. The ORBIT bleeding score: a simple bedside score to assess bleeding risk in atrial fibrillation. Eur Heart J 2015;36:3258-3264.
pmid pmc
6. Ewen S, Rettig-Ewen V, Mahfoud F, Böhm M, Laufs U. Drug adherence in patients taking oral anticoagulation therapy. Clin Res Cardiol 2014;103:173-182.
crossref pmid pdf
7. Vazquez SR, Yates NY, Beavers CJ, Triller DM, McFarland MM. Differences in quality of anticoagulation care delivery according to ethnoracial group in the United States: a scoping review. J Thromb Thrombolysis 2024;57:1076-1091.
crossref pmid pmc pdf
8. Kobayashi LC, Wardle J, Wolf MS, von Wagner C. Aging and functional health literacy: a systematic review and metaanalysis. J Gerontol B Psychol Sci Soc Sci 2016;71:445-457.
crossref pmid pmc
9. Goel N, Jain D, Haddad DB, Shanbhogue D. Anticoagulation in patients with end-stage renal disease and atrial fibrillation: confusion, concerns and consequences. J Stroke 2020;22:306-316.
crossref pmid pmc pdf
10. Mar PL, Gopinathannair R, Gengler BE, Chung MK, Perez A, Dukes J, et al. Drug interactions affecting oral anticoagulant use. Circ Arrhythm Electrophysiol 2022;15:e007956.
crossref pmid pmc
11. Manns B, Hemmelgarn B, Lillie E, Dip SC, Cyr A, Gladish M, et al. Setting research priorities for patients on or nearing dialysis. Clin J Am Soc Nephrol 2014;9:1813-1821.
crossref pmid pmc
12. Kido K, Lee JC, Hellwig T, Gulseth MP. Use of direct oral anticoagulants in morbidly obese patients. Pharmacotherapy 2020;40:72-83.
crossref pmid pdf
13. White EM, Coons JC. Direct oral anticoagulant use in special populations: elderly, obesity, and renal failure. Curr Cardiol Rep 2021;23:27.
crossref pmid pdf
14. Sabol BJ, Basa RR, Wilkins CE. Malabsorption-associated warfarin resistance. Am J Health Syst Pharm 2009;66:1548-1553.
crossref pmid
15. Sconce E, Avery P, Wynne H, Kamali F. Vitamin K supplementation can improve stability of anticoagulation for patients with unexplained variability in response to warfarin. Blood 2007;109:2419-2423.
crossref pmid pdf
16. Witt DM, Clark NP, Kaatz S, Schnurr T, Ansell JE. Guidance for the practical management of warfarin therapy in the treatment of venous thromboembolism. J Thromb Thrombolysis 2016;41:187-205.
crossref pmid pmc pdf
17. Chen A, Stecker E, A Warden B. Direct oral anticoagulant use: a practical guide to common clinical challenges. J Am Heart Assoc 2020;9:e017559.
crossref pmid pmc
18. Steinberg BA, Shrader P, Thomas L, Ansell J, Fonarow GC, Gersh BJ, et al. Off-label dosing of non-vitamin K antagonist oral anticoagulants and adverse outcomes: the ORBIT-AF II registry. J Am Coll Cardiol 2016;68:2597-2604.
pmid
19. Holbrook AM, Pereira JA, Labiris R, McDonald H, Douketis JD, Crowther M, et al. Systematic overview of warfarin and its drug and food interactions. Arch Intern Med 2005;165:1095-1106.
crossref pmid
20. Vega AJ, Smith C, Matejowsky HG, Thornhill KJ, Borne GE, Mosieri CN, et al. Warfarin and antibiotics: drug interactions and clinical considerations. Life (Basel) 2023;13:1661.
crossref pmid pmc
21. Eun MY, Kim JY, Hwang YH, Park MS, Kim JT, Choi KH, et al. Initiation of guideline-matched oral anticoagulant in atrial fibrillation-related stroke. J Stroke 2021;23:113-123.
crossref pmid pmc pdf
22. Mueck W, Kubitza D, Becka M. Co-administration of rivaroxaban with drugs that share its elimination pathways: pharmacokinetic effects in healthy subjects. Br J Clin Pharmacol 2013;76:455-466.
crossref pmid pmc
23. Maher RL, Hanlon J, Hajjar ER. Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf 2014;13:57-65.
crossref pmid pmc
24. Lip GYH, Banerjee A, Boriani G, Chiang CE, Fargo R, Freedman B, et al. Antithrombotic therapy for atrial fibrillation: CHEST guideline and expert panel report. Chest 2018;154:1121-1201.
crossref pmid
25. Hakeam HA, Al-Sanea N. Effect of major gastrointestinal tract surgery on the absorption and efficacy of direct acting oral anticoagulants (DOACs). J Thromb Thrombolysis 2017;43:343-351.
crossref pmid pdf
26. Bolek T, Samoš M, Stancˇiaková L, Ivanková J, Škornˇ ová I, Staško J, et al. The impact of proton pump inhibition on dabigatran levels in patients with atrial fibrillation. Am J Ther 2019;26:e308-e313.
crossref pmid
27. Nwanosike EM, Merchant HA, Sunter W, Ansari MA, Conway BR, Hasan SS. Direct oral anticoagulants and the risk of adverse clinical outcomes among patients with different body weight categories: a large hospital-based study. Eur J Clin Pharmacol 2024;80:163-173.
crossref pmid pmc pdf
28. Seiffge DJ, Traenka C, Polymeris AA, Thilemann S, Wagner B, Hert L, et al. Intravenous thrombolysis in patients with stroke taking rivaroxaban using drug specific plasma levels: experience with a standard operation procedure in clinical practice. J Stroke 2017;19:347-355.
crossref pmid pmc pdf
29. Pollack CV Jr, Reilly PA, van Ryn J, Eikelboom JW, Glund S, Bernstein RA, et al. Idarucizumab for dabigatran reversal—full cohort analysis. N Engl J Med 2017;377:431-441.
crossref pmid
30. Cohen H, Hunt BJ, Efthymiou M, Arachchillage DR, Mackie IJ, Clawson S, et al. Rivaroxaban versus warfarin to treat patients with thrombotic antiphospholipid syndrome, with or without systemic lupus erythematosus (RAPS): a randomised, controlled, open-label, phase 2/3, non-inferiority trial. Lancet Haematol 2016;3:e426-e436.
crossref pmid pmc
31. Miyakis S, Lockshin MD, Atsumi T, Branch DW, Brey RL, Cervera R, et al. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost 2006;4:295-306.
crossref pmid
32. Dufrost V, Risse J, Reshetnyak T, Satybaldyeva M, Du Y, Yan XX, et al. Increased risk of thrombosis in antiphospholipid syndrome patients treated with direct oral anticoagulants. Results from an international patient-level data meta-analysis. Autoimmun Rev 2018;17:1011-1021.
crossref pmid
33. Pengo V, Denas G, Zoppellaro G, Jose SP, Hoxha A, Ruffatti A, et al. Rivaroxaban vs warfarin in high-risk patients with antiphospholipid syndrome. Blood 2018;132:1365-1371.
crossref pmid pdf
34. Moran J, Bauer KA. Managing thromboembolic risk in patients with hereditary and acquired thrombophilias. Blood 2020;135:344-350.
crossref pmid pdf
35. Khorana AA. Cancer-associated thrombosis: updates and controversies. Hematology Am Soc Hematol Educ Program 2012;2012:626-630.
crossref pmid pdf
36. Abdol Razak NB, Jones G, Bhandari M, Berndt MC, Metharom P. Cancer-associated thrombosis: an overview of mechanisms, risk factors, and treatment. Cancers (Basel) 2018;10:380.
crossref pmid pmc
37. Lee MJ, Chung JW, Ahn MJ, Kim S, Seok JM, Jang HM, et al. Hypercoagulability and mortality of patients with stroke and active cancer: the OASIS-CANCER study. J Stroke 2017;19:77-87.
crossref pmid pmc pdf
38. Connors JM. Thrombophilia testing and venous thrombosis. N Engl J Med 2017;377:1177-1187.
crossref pmid
39. Collado FMS, Lama von Buchwald CM, Anderson CK, Madan N, Suradi HS, Huang HD, et al. Left atrial appendage occlusion for stroke prevention in nonvalvular atrial fibrillation. J Am Heart Assoc 2021;10:e022274.
crossref pmid pmc
40. Zhan Y, Joza J, Al Rawahi M, Barbosa RS, Samuel M, Bernier M, et al. Assessment and management of the left atrial appendage thrombus in patients with nonvalvular atrial fibrillation. Can J Cardiol 2018;34:252-261.
crossref pmid
41. Iding AFJ, Limpens TMP, Ten Cate H, Ten Cate-Hoek AJ. Chronic inflammatory diseases increase the risk of post-thrombotic syndrome: a prospective cohort study. Eur J Intern Med 2024;120:85-91.
crossref pmid
42. Melgaard L, Gorst-Rasmussen A, Lane DA, Rasmussen LH, Larsen TB, Lip GY. Assessment of the CHA2DS2-VASc score in predicting ischemic stroke, thromboembolism, and death in patients with heart failure with and without atrial fibrillation. JAMA 2015;314:1030-1038.
crossref pmid
43. Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011;365:883-891.
crossref pmid
44. Angiolillo DJ, Ueno M, Goto S. Basic principles of platelet biology and clinical implications. Circ J 2010;74:597-607.
crossref pmid
45. Safar ME, Levy BI, Struijker-Boudier H. Current perspectives on arterial stiffness and pulse pressure in hypertension and cardiovascular diseases. Circulation 2003;107:2864-2869.
crossref pmid
46. Grant PJ. Diabetes mellitus as a prothrombotic condition. J Intern Med 2007;262:157-172.
crossref pmid
47. Rodriguez-Iturbe B, Pons H, Johnson RJ. Role of the immune system in hypertension. Physiol Rev 2017;97:1127-1164.
crossref pmid pmc
48. Boutouyrie P, Chowienczyk P, Humphrey JD, Mitchell GF. Arterial stiffness and cardiovascular risk in hypertension. Circ Res 2021;128:864-886.
crossref pmid
49. Messner B, Bernhard D. Smoking and cardiovascular disease: mechanisms of endothelial dysfunction and early atherogenesis. Arterioscler Thromb Vasc Biol 2014;34:509-515.
pmid
50. Bang OY. Silent brain infarction: a quiet predictor of future stroke. Precis Future Med 2018;2:167-174.
crossref pdf
51. Fishbein MC. The vulnerable and unstable atherosclerotic plaque. Cardiovasc Pathol 2010;19:6-11.
crossref pmid
52. Yaghi S, Willey JZ, Cucchiara B, Goldstein JN, Gonzales NR, Khatri P, et al. Treatment and outcome of hemorrhagic transformation after intravenous alteplase in acute ischemic stroke: a scientific statement for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2017;48:e343-e361.
crossref pmid
53. Tunick PA, Kronzon I. Atheromas of the thoracic aorta: clinical and therapeutic update. J Am Coll Cardiol 2000;35:545-554.
crossref pmid
54. Amarenco P, Cohen A, Tzourio C, Bertrand B, Hommel M, Besson G, et al. Atherosclerotic disease of the aortic arch and the risk of ischemic stroke. N Engl J Med 1994;331:1474-1479.
crossref pmid
55. Kumar V, Shaw JR, Key NS, Ilich A, Mallick R, Wells PS, et al. D-dimer enhances risk-targeted thromboprophylaxis in ambulatory patients with cancer. Oncologist 2020;25:1075-1083.
crossref pmid pmc pdf
56. Bang OY, Chung JW, Lee MJ, Seo WK, Kim GM, Ahn MJ; OASIS-Cancer Study Investigators. Cancer-related stroke: an emerging subtype of ischemic stroke with unique pathomechanisms. J Stroke 2020;22:1-10.
crossref pmid pmc pdf
57. Campello E, Spiezia L, Simion C, Tormene D, Camporese G, Dalla Valle F, et al. Direct oral anticoagulants in patients with inherited thrombophilia and venous thromboembolism: a prospective cohort study. J Am Heart Assoc 2020;9:e018917.
crossref pmid pmc
58. Pengo V, Tripodi A, Reber G, Rand JH, Ortel TL, Galli M, et al. Update of the guidelines for lupus anticoagulant detection. J Thromb Haemost 2009;7:1737-1740.
crossref pmid
59. Segal JB, Brotman DJ, Necochea AJ, Emadi A, Samal L, Wilson LM, et al. Predictive value of factor V Leiden and prothrombin G20210A in adults with venous thromboembolism and in family members of those with a mutation: a systematic review. JAMA 2009;301:2472-2485.
crossref pmid
60. Holbrook A, Schulman S, Witt DM, Vandvik PO, Fish J, Kovacs MJ, et al. Evidence-based management of anticoagulant therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2012;141(2 Suppl):e152S-e184S.
pmid pmc
61. Cuker A, Siegal DM, Crowther MA, Garcia DA. Laboratory measurement of the anticoagulant activity of the non-vitamin K oral anticoagulants. J Am Coll Cardiol 2014;64:1128-1139.
crossref pmid pmc
62. Samuelson BT, Cuker A, Siegal DM, Crowther M, Garcia DA. Laboratory assessment of the anticoagulant activity of direct oral anticoagulants: a systematic review. Chest 2017;151:127-138.
crossref pmid pmc
63. van Ryn J, Baruch L, Clemens A. Interpretation of point-of-care INR results in patients treated with dabigatran. Am J Med 2012;125:417-420.
crossref pmid
64. Douxfils J, Ageno W, Samama CM, Lessire S, Ten Cate H, Verhamme P, et al. Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians. J Thromb Haemost 2018;16:209-219.
crossref pmid pdf
65. Camm AJ, Lip GY, De Caterina R, Savelieva I, Atar D, Hohnloser SH, et al. 2012 Focused update of the ESC Guidelines for the management of atrial fibrillation: an update of the 2010 ESC Guidelines for the management of atrial fibrillation. Developed with the special contribution of the European Heart Rhythm Association. Eur Heart J 2012;33:2719-2747.
pmid
66. Kearon C, Akl EA, Ornelas J, Blaivas A, Jimenez D, Bounameaux H, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest 2016;149:315-352.
pmid
67. Agnelli G, Becattini C, Meyer G, Muñoz A, Huisman MV, Connors JM, et al. Apixaban for the treatment of venous thromboembolism associated with cancer. N Engl J Med 2020;382:1599-1607.
crossref pmid
68. Nishioka J, Goodin S. Low-molecular-weight heparin in cancer-associated thrombosis: treatment, secondary prevention, and survival. J Oncol Pharm Pract 2007;13:85-97.
crossref pmid pdf
69. Nishimura RA, Otto CM, Bonow RO, Carabello BA, Erwin JP 3rd, Fleisher LA, et al. 2017 AHA/ACC focused update of the 2014 AHA/ACC guideline for the management of patients with valvular heart disease: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines. J Am Coll Cardiol 2017;70:252-289.
pmid
70. Valgimigli M, Bueno H, Byrne RA, Collet JP, Costa F, Jeppsson A, et al. 2017 ESC focused update on dual antiplatelet therapy in coronary artery disease developed in collaboration with EACTS: The Task Force for dual antiplatelet therapy in coronary artery disease of the European Society of Cardiology (ESC) and of the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2018;39:213-260.
pmid
71. Hindricks G, Potpara T, Dagres N, Arbelo E, Bax JJ, Blomström-Lundqvist C, et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur Heart J 2021;42:373-498.
pmid
72. Gibson CM, Mehran R, Bode C, Halperin J, Verheugt FW, Wildgoose P, et al. Prevention of bleeding in patients with atrial fibrillation undergoing PCI. N Engl J Med 2016;375:2423-2434.
crossref pmid
73. Lopes RD, Heizer G, Aronson R, Vora AN, Massaro T, Mehran R, et al. Antithrombotic therapy after acute coronary syndrome or PCI in atrial fibrillation. N Engl J Med 2019;380:1509-1524.
pmid
74. Cannon CP, Bhatt DL, Oldgren J, Lip GYH, Ellis SG, Kimura T, et al. Dual antithrombotic therapy with dabigatran after PCI in atrial fibrillation. N Engl J Med 2017;377:1513-1524.
crossref pmid
75. Reddy VY, Doshi SK, Kar S, Gibson DN, Price MJ, Huber K, et al. 5-year outcomes after left atrial appendage closure: from the PREVAIL and PROTECT AF trials. J Am Coll Cardiol 2017;70:2964-2975.
pmid
76. Blackshear JL, Odell JA. Appendage obliteration to reduce stroke in cardiac surgical patients with atrial fibrillation. Ann Thorac Surg 1996;61:755-759.
crossref pmid
77. Osmancik P, Herman D, Neuzil P, Hala P, Taborsky M, Kala P, et al. Left atrial appendage closure versus direct oral anticoagulants in high-risk patients with atrial fibrillation. J Am Coll Cardiol 2020;75:3122-3135.
crossref pmid
78. January CT, Wann LS, Calkins H, Chen LY, Cigarroa JE, Cleveland JC Jr, et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines and the Heart Rhythm Society in collaboration with the society of thoracic surgeons. Circulation 2019;140:e125-e151.
crossref pmid
79. Farge D, Frere C, Connors JM, Ay C, Khorana AA, Munoz A, et al. 2019 international clinical practice guidelines for the treatment and prophylaxis of venous thromboembolism in patients with cancer. Lancet Oncol 2019;20:e566-e581.
pmid
80. Xie Z, Meng Z, Yang X, Duan Y, Wang Q, Liao C. Factor XIa inhibitors in anticoagulation therapy: recent advances and perspectives. J Med Chem 2023;66:5332-5363.
crossref pmid pdf
81. Ruff CT, Patel SM, Giugliano RP, Morrow DA, Hug B, Kuder JF, et al. Abelacimab versus rivaroxaban in patients with atrial fibrillation. N Engl J Med 2025;392:361-371.
crossref pmid
82. Piccini JP, Patel MR, Steffel J, Ferdinand K, Van Gelder IC, et al. Asundexian versus apixaban in patients with atrial fibrillation. N Engl J Med 2025;392:23-32.
pmid
83. Weitz JI, Eikelboom JW, Samama MM. New antithrombotic drugs: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2012;141(2 Suppl):e120Se151S.

84. Tang LV, Tao Y, Feng Y, Ma J, Lin W, Zhang Y, et al. Gene editing of human iPSCs rescues thrombophilia in hereditary antithrombin deficiency in mice. Sci Transl Med 2022;14:eabq3202.
crossref pmid
85. Markl M, Frydrychowicz A, Kozerke S, Hope M, Wieben O. 4D flow MRI. J Magn Reson Imaging 2012;36:1015-1036.
crossref pmid
86. Rinkel LA, Beemsterboer CF, Groeneveld NS, Lobé NH, Boekholdt SM, Bouma BJ, et al. Cardiac thrombi detected by CT in patients with acute ischemic stroke: a substudy of mind the heart. Eur Stroke J 2023;8:168-174.
crossref pmid pmc pdf
87. Choi Y, Saqqur M, Stewart E, Stephenson C, Roy J, Boulanger JM, et al. Relative energy index of microembolic signal can predict malignant microemboli. Stroke 2010;41:700-706.
crossref pmid
88. Liang H, Zhang H, Wang J, Shao X, Wu S, Lyu S, et al. The application of artificial intelligence in atrial fibrillation patients: from detection to treatment. Rev Cardiovasc Med 2024;25:257.
crossref pmid pmc
89. Kao YT, Huang CY, Fang YA, Liu JC, Chang TH. Machine learning-based prediction of atrial fibrillation risk using electronic medical records in older aged patients. Am J Cardiol 2023;198:56-63.
crossref pmid
90. Attia ZI, Noseworthy PA, Lopez-Jimenez F, Asirvatham SJ, Deshmukh AJ, Gersh BJ, et al. An artificial intelligence-enabled ECG algorithm for the identification of patients with atrial fibrillation during sinus rhythm: a retrospective analysis of outcome prediction. Lancet 2019;394:861-867.
crossref pmid
91. Johnson JA, Caudle KE, Gong L, Whirl-Carrillo M, Stein CM, Scott SA, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for pharmacogenetics-guided warfarin dosing: 2017 update. Clin Pharmacol Ther 2017;102:397-404.
crossref pmid pmc pdf
92. Klein TE, Altman RB, Eriksson N, Gage BF, Kimmel SE, Lee MT, et al. Estimation of the warfarin dose with clinical and pharmacogenetic data. N Engl J Med 2009;360:753-764.
crossref pmid pmc


ABOUT JoS
AUTHOR INFORMATION
ARTICLE CATEGORY

Browse all articles >

BROWSE ARTICLES
Editorial Office
Department of Neurology, Asan Medical Center,Ulsan University College of Medicine
88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea
Submission, status and progress, etc ⟫ E-mail: editor@j-stroke.org
Website and system ⟫ E-mail: journal@m2community.co.kr
Publishing company ⟫ E-mail: ka72sus@smileml.com
Developed in M2PI
Copyright © 2025 by Korean Stroke Society.
Close layer
prev next